Watanabe 2018

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Pediatrics International (2019) 61, 222–229 doi: 10.1111/ped.

13776

Review Article

Recent advances in the understanding of transient abnormal


myelopoiesis in Down syndrome

Kenichiro Watanabe
Department of Hematology and Oncology, Shizuoka Children’s Hospital, Aoi-ku, Shizuoka, Japan

Abstract Neonates with Down syndrome (DS) have a propensity to develop the unique myeloproliferative disorder, transient
abnormal myelopoiesis (TAM). TAM usually resolves spontaneously in ≤3 months, but approximately 10% of
patients with TAM die from hepatic or multi-organ failure. After remission, 20% of patients with TAM develop
acute myeloid leukemia associated with Down syndrome (ML-DS). Blasts in both TAM and ML-DS have trisomy
21 and GATA binding protein 1 (GATA1) mutations. Recent studies have shown that infants with DS and no clinical
signs of TAM or increases in peripheral blood blasts can have minor clones carrying GATA1 mutations, referred to
as silent TAM. Low-dose cytarabine can improve the outcomes of patients with TAM and high white blood cell
count. A number of studies using fetal liver cells, mouse models, or induced pluripotent stem cells have elucidated
the roles of trisomy 21 and GATA1 mutations in the development of TAM. Next-generation sequencing of TAM
and ML-DS patient samples identified additional mutations in genes involved in epigenetic regulation. Xenograft
models of TAM demonstrate the genetic heterogeneity of TAM blasts and mimic the process of clonal selection
and expansion of TAM clones that leads to ML-DS. DNA methylation analysis suggests that epigenetic dysregula-
tion may be involved in the progression from TAM to ML-DS. Unraveling the mechanisms underlying leukemogen-
esis and identification of factors that predict progression to leukemia could assist in development of strategies to
prevent progression to ML-DS. Investigation of TAM, a unique pre-leukemic condition, will continue to strongly
influence basic and clinical research into the development of hematological malignancies.

Key words Down syndrome, GATA1, leukemia, transient abnormal myelopoiesis, trisomy 21.

Neonates with Down syndrome (DS) have a marked propen- next-generation sequencing (NGS) have detected subclones
sity to develop the unique myeloproliferative disorder referred that emerge during the TAM phase and appear to expand to
to as transient abnormal myelopoiesis (TAM), transient myelo- develop into overt leukemia through clonal selection and addi-
proliferative disorder, or transient leukemia.1 In the majority tional genetic alterations.18,19
of cases, TAM resolves spontaneously in ≤3 months, but This review outlines the clinical features of TAM and
approximately 10% of patients with TAM die from hepatic or focuses on the latest findings that may provide clues to the
multi-organ failure. Recent studies have identified risk factors understanding of this unique disease. I also consider future
for infant death due to TAM and indicate the efficacy of low- research directions in this field.
dose cytarabine for treatment of severe manifestations of
TAM.2–5
Definition and diagnosis of TAM
After remission, approximately 20% of patients with TAM
develop acute myeloid leukemia (AML) associated with Down Transient abnormal myelopoiesis is a unique myeloprolifera-
syndrome (ML-DS).2,3,5,6 Given that blasts in both TAM and tive disorder in newborns with DS who present with circulat-
ML-DS have common genetic abnormalities, including tri- ing blasts that are morphologically and phenotypically similar
somy 21 and GATA binding protein 1 (GATA1) mutation,7–13 to AML. The blasts have morphologic and immunologic fea-
TAM is considered a pre-leukemic disorder, and understanding tures of the megakaryocytic lineage.2,20–22 Clinical diagnosis
how TAM progresses to ML-DS may provide information cru- of TAM is based on the presence of characteristic blasts in
cial to clarifying the mechanism involved in multi-step leuke- peripheral blood smears. Given that TAM blasts carry muta-
mogenesis.14–17 Recent studies using a xenograft model and tions of GATA1 in addition to trisomy 21,8–13 positivity for
GATA1 mutation confirms the diagnosis of TAM, but the per-
Correspondence: Kenichiro Watanabe, MD PhD, Department of centage of TAM cells varies enormously, both temporally and
Hematology and Oncology, Shizuoka Children’s Hospital, 860 between patients, and there are no clear diagnostic criteria for
Urushiyama, Shizuoka 420-8660, Japan. Email: wataken@kuhp. TAM.23
kyoto-u.ac.jp
Diagnosis of TAM is important, particularly because
Received 10 May 2018; revised 8 December 2018; accepted 28
December 2018. infants with TAM have a higher likelihood of developing

© 2018 Japan Pediatric Society


Review of TAM 223

ML-DS; children with no clear history of TAM, however, also study screened for GATA1 mutations using Guthrie cards from
develop ML-DS. These patients may not have overt clinical 590 DS infants and detected them in 3.8% of samples.25 In
symptoms associated with TAM, or may present with normal contrast, the application of targeted NGS by OIDSCS found
blood cell count and low blast count in the neonatal period. GATA1 mutations in 20% of DS neonates who were GATA1
Thus, current clinical diagnosis, based on the presence of mutation negative on Sanger sequencing.23 Hence, the inci-
blasts in peripheral blood smears, is unsatisfactory for identifi- dence of TAM can vary depending on the clinical definition
cation of the group at high risk of ML-DS development. used and the sensitivity of the methods for detection of
The investigators of the Oxford–Imperial Down Syndrome GATA1 mutations.
Cohort Study Group (OIDSCS) analyzed clinical findings,
blood counts and smears, and GATA1 mutation status in 200
Clinical and hematological features of TAM
DS neonates.23 They reported that all DS neonates had multi-
ple blood count and morphological abnormalities and that The clinical and hematological features of TAM are listed in
97.5% had circulating blasts, indicating that all DS neonates Table 1. Clinical presentation of TAM may vary from asymp-
have at least some hematological abnormalities, regardless of tomatic to very severe disease. Previous studies report a med-
GATA1 mutation status. On conventional Sanger sequencing ian age at diagnosis of 3–7 days, with almost all cases
and denaturing high-performance liquid chromatography, diagnosed in the 2 months after birth. Asymptomatic babies
GATA1 mutations were detected in 17 of 200 (8.5%) enrolled with DS may be diagnosed with TAM on incidental detection
DS neonates.23 Furthermore, infrequent GATA1 mutant clones of blasts in peripheral blood smears with or without leukocyto-
were detected using the more sensitive method of NGS in 18 sis. In contrast, neonates with TAM may present with severe
of 88 (20.4%) DS neonates in whom GATA1 mutations were symptoms, such as marked hepatosplenomegaly, serous effu-
not detected on conventional sequencing methods.23 The clini- sions, and multi-organ failure, due to blast cell infiltration.
cal and hematologic features of these neonates were indistin- Patients with very severe disease are at high risk of early
guishable from those of DS neonates without GATA1 death.2–5 TAM can present as hydrops fetalis and hep-
mutations detectable on NGS. The OIDSCS proposed the term atosplenomegaly in fetuses with DS.26–29 The most severe
“silent TAM” for DS neonates with GATA1 mutation detect- form of TAM is diagnosed on post-mortem in stillborn DS
able only on NGS. Of note, one of 18 individuals with silent fetuses.30–32 Infants with TAM may also have delayed-onset
TAM developed ML-DS, while none of the 70 neonates with- hyperbilirubinemia, which is a sign of progressive liver fibro-
out NGS-detected GATA1 mutations were documented to sis that can result in fatal liver failure.33–35 Pathogenesis of
develop the condition.23 That indicates that peripheral blood liver fibrosis in TAM appears to be similar to that of marrow
smear and GATA1 mutation analysis should be performed for fibrosis that is associated with acute megakaryoblastic leuke-
all DS neonates, and that NGS-based methods can identify mia (AMKL). TAM blasts from patients with liver fibrosis
neonates with minor GATA1 mutant clones. can exhibit high platelet-derived growth factor and transform-
In the era of high-throughput genome sequencing, the defi- ing growth factor-b1 expression,36,37 leading to the hypothesis
nition of TAM needs to be reconsidered and revised to reflect that TAM blasts may arise from the fetal hepatic hematopoi-
the clinical behavior and the latest molecular insight into the etic environment, rather than from the bone marrow.
disease. It may be appropriate to define TAM according to the Patients with TAM present with various hematological
presence of GATA1 mutations detected using a more sensitive abnormalities.2–5 Leukocytosis and thrombocytopenia are com-
method such as NGS as well as conventional Sanger sequenc- mon, and white blood cell (WBC) count may exceed 100 9
ing. The incidence and clinical significance of silent TAM 109/L. TAM is characterized by a varying degree of increased
should be re-evaluated for separate cohorts, given that they blast cells in the peripheral blood. The typical morphologic
may vary depending on the sensitivity of the methods used to features of blasts are similar to those of AMKL,2 but the
detect GATA1 mutations.
Table 1 Clinical and hematological features of TAM

Incidence of TAM Hepatosplenomegaly


Skin rash
Transient abnormal myelopoiesis occurs in approximately 10% Pericardial/pleural effusion, ascites
of DS newborns. In Japan, during the 18 year period between Hydrops fetalis
1980 and 1997, the overall prevalence of DS was 5.82 per Jaundice
10 000 live births (8.3–9.7 per 10 000 after correction accord- Coagulopathy
Liver failure (may occur in late neonatal period)
ing to the estimated ascertainment ratio, 60–70%).24 Accord-
Thrombocytopenia
ing to these data, the incidence of TAM in Japan is estimated Leukocytosis
as approximately 0.58 per 10 000 live births, but, as afore- Anemia
mentioned, the diagnostic criteria for TAM differ between pre- Early death (approx. 10%)
vious studies, and not all DS neonates may have been Progression to ML-DS (approx. 20%)
carefully examined for TAM on thorough morphological MD-DS, myeloid leukemia associated with Down syndrome;
assessment of peripheral blood smears. One population-based TAM, transient abnormal myelopoiesis.

© 2018 Japan Pediatric Society


224 K Watanabe

morphology of TAM blasts is variable in practice. Increased It should be noted that low-dose cytarabine may cause sig-
neutrophils, myelocytes, monocytes, and basophils are fre- nificant myelosuppression. In the COG study, grade 3/4
quently observed in peripheral blood smears, and, interestingly, myelosuppression was detected in 96% of treated patients,
there are cases of TAM involving prominent eosinophilia.38,39 likely due to continuous infusion of relatively high-dose
Relatively small percentages of blasts are found in the bone cytarabine.5 The most common side-effect of cytarabine in a
marrow of patients with TAM.2,3,5 Thus, bone marrow aspira- recent study was neutropenia (grade 3–4, 59%).42 Although
tion or biopsy is not usually recommended for diagnosis or low-dose cytarabine is more frequently applied to TAM
evaluation of TAM. patients with potential risk factors, there has been no study
The immunophenotype of TAM blasts is the same as that that defines clear criteria for its application and treatment
of AMKL, with blast cells positive for stem cell markers schedule for TAM. A prospective intervention study is
(CD34, CD117), myeloid markers (CD13, CD33), platelet gly- planned, to establish a standard for treatment with cytarabine
coproteins (CD36, CD42, CD61), CD56, and CD7, with varia- for patients with TAM.
tion among cases.20–22 Low-dose cytarabine therapy has been hypothesized to pre-
Coagulopathy is a potential complication of TAM,3,5 and vent progression to ML-DS by eliminating pre-leukemic
considered a risk factor for early death. Disseminated intravas- clones, but no studies have shown that cytarabine reduces the
cular coagulation (DIC) is usually observed in the majority of risk of leukemia development.3,5 Most recently, a multicenter,
severe cases of TAM with fulminant liver dysfunction.40 non-randomized, historically controlled study, TMD 2007,
indicated that low-dose cytarabine was insufficient to prevent
progression to ML-DS.43
Management of TAM
It is recommended to follow up patients with TAM every
Most patients with TAM have spontaneous regression, usually 1–3 months to screen for the development of ML-DS. Throm-
within a few months, and do not need treatment, but some bocytopenia often proceeds to manifestation of overt acute
individuals develop progressive, life-threatening disease, and leukemia. At such an early phase, it may be difficult to detect
up to 20% of patients with TAM eventually die in early a significant percentage of blasts on smear slides obtained by
infancy. Risk factors for early death in patients with TAM bone marrow aspiration, while the identification of clusters of
include hyperleukocytosis, ascites, and prematurity, as well as leukemic cells in bone marrow biopsy specimens may be more
cholestasis, liver fibrosis, renal dysfunction, and DIC.2–5 feasible.
The DS myeloblasts are highly sensitive to cytarabine,41
and low-dose cytarabine is effective in decreasing blast cells
Pathogenesis of TAM
in patients with TAM. Thus, low-dose cytarabine was used in
previous large prospective studies to treat the severe form of A schematic diagram of the pathogenesis of TAM and pro-
TAM. In a Berlin-Frankfurt-M€ unster (BFM) group study, gression to ML-DS is shown in Fig. 1. TAM is a unique con-
low-dose cytarabine (0.5–1.5 mg/kg for 3–12 days) was rec- dition that occurs only in blood cells with trisomy 21. Blast
ommended for TAM patients with thrombocytopenia, liver cells of both TAM and subsequent ML-DS have acquired
dysfunction, or high white cell count (>50 9 109/L); 5 year GATA1 mutations, in addition to trisomy 21.8,10,12 In the
overall survival (OS) in the treated group was similar to that majority of cases, TAM resolves spontaneously after birth,2–5
in the untreated group (78%  8% vs 85%  3%; P = 0.44).3 seemingly along with the shift in the site of hematopoiesis
In a Children’s Oncology Group (COG) study, cytarabine from fetal liver44 to bone marrow. TAM can cause massive
(3.33 mg/kg/day continuous infusion for 7 days) was given to infiltration of the liver and hydrops fetalis in utero.26–29 Based
patients with life-threatening symptoms. The patient survival on these observations, it has been considered that acquisition
rate was 51%.14 Recently the Japan Children’s Cancer Group of an N-terminal GATA1 mutation in hematopoietic cells car-
(JCCG) TAM committee, conducted a large multi-institutional rying trisomy 21 gives rise to TAM clones in the fetal liver.
prospective study of patients with TAM in Japan.42 In that
study high WBC count and anasarca were confirmed as risk
Abnormal hematopoiesis associated with trisomy 21
factors for early death in patients with DS diagnosed with
TAM. Although low-dose cytarabine could significantly Several studies of primary fetal liver cells demonstrate abnor-
improve the survival rate in patients with a high WBC count, mal hematopoiesis in fetuses with trisomy 21 by the second
it failed to change the prognosis of patients with anasarca.42 trimester. Hematopoietic stem cells (HSC) and megakary-
Overall, low-dose cytarabine is effective in reducing blasts, ocyte–erythroid progenitors are markedly increased,45 whereas
thereby improving the outcome for a specific subset of granulocyte–macrophage progenitors are reduced in primary
patients with TAM. Nevertheless, patients with very severe fetal liver cells with trisomy 21 compared with primary dis-
disease, such as serous effusion, liver failure, and/or multi- omic controls.46,47 In clonogenic assays, HSC and megakary-
organ dysfunction, often fail to respond to low-dose cytarabine ocyte–erythroid progenitors with trisomy 21 have higher
and require other supportive treatment, including steroids, proliferation rates, with increased megakaryocyte, megakary-
exchange transfusion, and/or intensive care; the development ocyte–erythroid, and replatable blast colonies. Furthermore,
of novel treatment modalities for this group is imperative. megakaryocyte lineage differentiation is hampered in fetal

© 2018 Japan Pediatric Society


Review of TAM 225

Fetal liver mutated GATA1, resulting in the accelerated production of


Clonal selection/expansion
TAM Additional genetic events:
ML-DS aberrantly differentiated cells. These effects were mediated by
Trisomy 21→
Perturbed gene mutations affecting dosage alterations of RUNX1, ETS2, and ERG, which are
cohesin/CTCF, EZH2, etc.
hematopoiesis
Large clones Epigenetic dysregulation located in a critical 4 Mb region of chromosome 21.51
Acquired Regression
GATA1mutations
GATA1 mutations in TAM
GATA1mutant
clones Clonal extinction Remission GATA1 is a key hematopoietic transcription factor with an
essential role in megakaryocyte–erythroid differentiation.61,62
Silent ML-DS Both TAM and ML-DS cells harbor acquired N-terminal trun-
TAM cating GATA1 mutations.7–13 GATA1 mutations are primarily
Small clones detected in exon 2, and result in premature stop codons, lead-
Birth 6 months 1 year 5 years
ing to the production of a short form of the GATA1 protein,
referred to as GATA1s.7,10 Given that GATA1 is located on
Fig. 1 Schematic diagram of the pathogenesis of transient the X chromosome, cells with GATA1 mutations exclusively
abnormal myelopoiesis (TAM) and progression to myeloid leuke- express GATA1s and no full-length GATA1 protein.7 Individ-
mia associated with Down syndrome (ML-DS). CTCF, CCC uals without trisomy 21 who have constitutional GATA1 muta-
TC-binding factor; EZH2, enhancer of zeste homolog 2; GATA1,
GATA binding protein 1. tions that produce only GATA1s develop thrombocytopenia,
but not TAM or ML-DS.63,64 Thus, GATA1 mutation alone is
not sufficient to transform cells into pre-leukemic or leukemic
hematopoietic cells with trisomy 21. Notably, these character- clones.
istics of perturbed hematopoiesis are observed in the absence The roles of GATA1s in the development of TAM and
of GATA1 mutation.46,47 This suggests that trisomy 21 itself is ML-DS have not been fully elucidated. Forced expression of
an initial event of TAM, prior to acquisition of GATA1 muta- GATA1s in fetal hematopoietic progenitors from mice with
tions.48 Recent studies using embryonic stem and induced wild-type GATA1 causes marked expansion of megakary-
pluripotent stem cells (iPSC) with trisomy 21 also demon- oblastic progenitors.65 Toki et al. also found that GATA1s has
strated impaired erythro-megakaryopoiesis, consistent with the a more profound effect in promotion of megakaryocyte prolif-
observations in fetal liver cells.49–51 eration than that induced by GATA1 deficiency,66 supporting
The notion that trisomy 21 alone causes abnormal hemato- a gain-of-function mechanism for GATA1s. GATA1s is
poiesis prompted us to consider that the gene dosage effect of thought to perturb GATA1-mediated regulation of other tran-
trisomy 21 may be responsible for the TAM phenomenon. A scription factors, including GATA2, MYB, MYC, and
number of studies have attempted to identify genes or regions IKAROS family zinc finger 1 (IKZF1), in fetal megakary-
of chromosome 21 that could be responsible for hematological ocytes.67 Transduction of GATA1s into GATA1 knockdown
phenotypes in DS patients. Previous genotype–phenotype stud- mice results in abnormal accumulation of CD41+ megakary-
ies identified subregions in chromosome 21 necessary and suf- ocytic progenitors.68 Loss of expression of full-length GATA1
ficient to produce the clinical phenotype of DS, referred to as may also influence the development of TAM by impairing
the “Down syndrome critical region” (DSCR).52,53 The DSCR the growth and differentiation of murine fetal/embryonic
contains several genes involved in hematopoiesis, including hematopoietic progenitors.69 Overall, the expression of
BACH1, RUNX1, ETS2, ERG, and DYRK1A,11,54–57 but the GATA1s, along with GATA1 dysfunction, appears to be
role of increased doses of those genes in the development of essential for the development of TAM. A recent study using
TAM and ML-DS has not been fully determined. DS mouse genetically engineered human iPSC showed that GATA1s is
models, engineered to have extra chromosomal segments con- upregulated by trisomy 21, leading to impaired megakary-
taining almost all of murine chromosome 16, which shows opoiesis, which indicates that a synergistic interaction between
synteny with human chromosome 21, fail to develop condi- trisomy 21 and GATA1 mutation is important for development
tions similar to either TAM or ML-DS.58–60 A recent study of TAM.51
establishing systematic disease models using human iPSC and
genome/chromosome-editing technologies suggests that
Progression from TAM to ML-DS
RUNX1/ETS2/ERG, in association with GATA1 mutation, are
critical for the hematopoietic abnormalities observed in DS.51
Factors predictive of ML-DS
Using genome editing, the investigators generated a panel of
human iPSC with different combinations of complements of It is a fascinating idea that the development of overt leukemia
chromosome 21 (disomy, trisomy, or partial trisomy) and could be predicted in the pre-leukemic phase. If risk factors
GATA1 status (wild-type, short, knockout). Comparative analy- for ML-DS were identified in infants with DS, it could
sis of these engineered iPSC demonstrated that trisomy 21 become possible to design a more adequate follow-up sched-
affected hematopoietic development through the enhanced pro- ule and devise effective methods to prevent progression to
duction of early hematopoietic progenitors and upregulation of ML-DS. Previous large clinical trials by Pediatric Oncology

© 2018 Japan Pediatric Society


226 K Watanabe

Group (POG), BFM, and COG reported candidate risk factors It was recently reported that dysregulation of DNA methyla-
for ML-DS.2,3,5 The POG trial reported that additional chro- tion may be involved in progression from TAM to ML-DS.72
mosome abnormalities at the TAM phase were risk factors.2 Given that ML-DS is frequently associated with additional
The BFM group demonstrated that pleural effusion at diagno- mutations in epigenetic regulators, it is reasonable to speculate
sis of TAM was predictive,3 while the COG trial found that that epigenetic dysregulation could have a critical role in the
longer time to regression of TAM was prognostic.5 In these transformation of TAM to ML-DS.
studies, the use of cytarabine to treat TAM was not identified
as a significant factor predicting development of ML-DS.
Clonal evolution from TAM to ML-DS
Kanezaki et al. showed that the expression level of
GATA1s varied according to the type of GATA1 mutation.70 It has long been hypothesized that a linear sequential acquisi-
Moreover, patients with GATA1 mutations leading to low tion of genetic alterations induces disease progression in
GATA1s expression had significantly higher risk of progres- TAM/ML-DS.73 Recent studies using high-throughput geno-
sion to ML-DS.70 This suggests that the type of GATA1 muta- mic technology indicate that evolutionary trajectories are more
tion may be a predictive factor for ML-DS, and this potential complex and branching in other cancers and leukemias. In this
association is currently under investigation in a prospective schema, genomic instability in founder cells gives rise to
study conducted by the JCCG. heterogeneous mutant subclones, and, under selective pressure,
Technologies such as quantitative polymerase chain reac- some subclones expand, resulting in disease progression, while
tion (PCR) and NGS enable detection of minimal populations others become extinct or remain dormant. Thus, leukemic
of clones with GATA1 mutations.18,19,23 Tracing GATA1 clones may evolve and emerge through the complex interac-
mutant clones by such sensitive methods in infants with DS tion of selectively advantageous “driver” mutations, additional
would help to clarify the behavior of pre-leukemic clones and advantageous “cooperating” mutations, neutral “passenger”
assess clonal evolution to ML-DS, which would allow identifi- mutations, and deleterious mutations.74,75 Recent studies indi-
cation of patients at high risk of progression to ML-DS. cate that this schema may also be applicable to the progres-
sion from TAM to ML-DS.
Multiple distinct GATA1 mutant clones can occur in a sin-
Additional genetic events involved in progression from
gle patient with TAM. For example, Xu et al. reported a case
TAM to ML-DS
of ML-DS derived from a minor clone with a GATA1 muta-
The mechanism of progression from TAM to ML-DS is an tion that differed from the mutation in the major clone present
important research topic, because this unique condition is con- at the TAM phase.76 Moreover, a recent study using whole
sidered a useful model for investigation of how full-blown leu- exome/genome sequencing of paired TAM and ML/DS sam-
kemia evolves from the pre-leukemic phase. Investigators ples showed that ML-DS can arise from minor, as well as
have hypothesized that genetic events additional to trisomy 21 major, TAM clones.19 These observations suggest that hetero-
and GATA1 are responsible for the transformation of TAM geneous TAM clones harboring distinct GATA1 mutations are
cells to ML-DS blasts. present during the TAM phase and that subsequent ML-DS
The ML-DS blast cells often carry additional chromosomal originates from one of multiple TAM subclones through selec-
abnormalities, such as dup(1q), del(6q), del(7p), dup(7q), +8, tion.
+11, and del(16q).2,40,71 Moreover, several mutations in cancer- A robust xenograft model of TAM was recently estab-
related genes, including JAK1/2/3, TP53, FLT3, and MPL, have lished using highly immunodeficient NOG mice. This model
been identified in cells from patients with ML-DS. Recently, enabled the successful observation of clonal selection and
Yoshida et al. performed comprehensive genetic analysis on expansion of minor mutant TAM clones.18 In that study, pri-
samples from patients with TAM and ML-DS using massively mary TAM cell samples from three of 11 patients were
parallel sequencing.19 The mean number of non-silent mutations engrafted into NOG mice. Interestingly, serial engraftment to
per TAM sample was extremely small compared with that in subsequently transplanted recipients was successful using
other human cancers. Other than common GATA1 mutations, cells obtained from one patient, indicating that some TAM
no other recurrent mutations were identified in TAM samples, clones have long-term self-renewal capacity. Of note, that
suggesting that development of TAM does not require genetic patient later developed ML-DS at the age of 1 year. ML-DS
changes additional to those of GATA1 mutation and trisomy 21. cells often have additional chromosome abnormalities or
In the majority of cases, ML-DS was accompanied by newly copy number alterations (CNA), while TAM cells are rarely
acquired mutations not present in the original TAM samples. associated with such genetic changes. Extensive serial trans-
Mutations affecting cohesin/CCCTC-binding factor (65%), plantation was performed using TAM cells from that patient
other epigenetic regulators, including enhancer of zeste homo- and CNA analyzed in the propagating cells. The primary
log 2 (45%), and RAS/signal transduction molecules were fre- samples had no CNA, other than trisomy 21. Serial trans-
quently detected as additional mutations in ML-DS.19 Given plantation indicated the emergence of subclones with various
that these genes have important roles in human leukemogenesis additional CNA and diverse repopulating capacities as xeno-
or tumorigenesis, such mutations may be essential genetic grafts. The genomic structure of a del(16q22) rearrangement
events for transformation of TAM blasts. found in one of the engrafted cell samples was determined,

© 2018 Japan Pediatric Society


Review of TAM 227

and breakpoint-specific PCR was performed to detect the Next-generation sequencing of samples from TAM and
presence of cells carrying the del(16q22) change. Surpris- ML-DS patients identified additional mutations in genes
ingly, on breakpoint-specific PCR a minor clone with del involved in epigenetic regulation that potentially transform
(16q22) was already present in the primary samples. Exten- pre-leukemia to full-blown leukemia. The xenograft model
sive transplantation also indicated a distinct GATA1 mutant demonstrates the genetic heterogeneity of TAM blasts and
that was not a major clone in the primary sample. Yet, geno- mimics how TAM clones expand or become extinct through
mic analysis showed that the subclone with the distinct clonal selection, leading to progression to ML-DS.18 DNA
GATA1 mutation was present as a minor clone among the methylation analysis, using this model and clinical samples,
primary cells, consistent with clinical observations of the suggests that epigenetic dysregulation may be involved in pro-
development of ML-DS from a minor TAM clone.18 This gression from TAM to ML-DS.
xenograft model is likely to mimic the early phase of the Unraveling the mechanisms involved in leukemogenesis
leukemic evolutionary process, because the TAM blasts have and identification of factors predictive for the development of
striking genetic heterogeneity, and subclones with leukemia- ML-DS may lead to strategies to prevent progression to ML-
initiating potential may already reside among them during DS. Investigation of TAM, a unique pre-leukemic condition,
the pre-leukemic phase. These findings support the hypothe- will continue to result in deep impacts on basic research and
sis that ML-DS develops from a pool of heterogeneous the development of new methods for treatment of hematologi-
minor clones through clonal selection, illustrating the early cal malignancy.
evolutionary process of leukemia.77
As mentioned, it was recently shown that dysregulation of
Acknowledgments
DNA methylation may be involved in progression from TAM
to ML-DS.72 Analysis using primary blood samples indicated This article is based on research performed in collaboration with
distinct DNA methylation profiles in TAM and ML-DS Professor Etsuro Ito and Dr Akira Watanabe, who are supported
cells.72 Consistent with this finding, the global DNA methyla- by AMED under Grant Number JP17 cm0106407h0002. TAM-
tion profiles of engrafted cells from first-generation mice were 10 was conducted by JCCG as a multi-institutional prospective
similar to those of TAM patients, whereas those from the study.
fourth, fifth, and sixth generations approached the profiles of
ML-DS patients.72 Further investigation is ongoing to clarify
Author contribution
the precise mechanisms involved in the development of TAM
and ML-DS. K.W. conceived of this review and wrote the manuscript.

Future directions Disclosure


Recent developments in genomic technology have facilitated The author declares no conflict of interest.
the revelation that DS infants with neither apparent clinical
signs of TAM nor increased blasts in peripheral blood may
References
carry minor GATA1 mutant clones, a condition referred to as
silent TAM. The incidence of silent TAM may vary according 1 Zipursky A, Poon A, Doyle J. Leukemia in Down syndrome:
to the sensitivity of the method used to detect GATA1 mutant A review. Pediatr. Hematol. Oncol. 1992; 9: 139–49.
2 Massey GV, Zipursky A, Chang MN et al. A prospective
clones. It follows that the clinical significance, that is, the study of the natural history of transient leukemia (TL) in
potential risk of developing ML-DS from silent TAM, may neonates with Down syndrome (DS): Children’s Oncology
also depend on the methodology used for GATA1 mutation Group (COG) study POG-9481. Blood 2006; 107: 4606–13.
analysis. Thus, the incidence and significance of silent TAM 3 Klusmann JH, Creutzig U, Zimmermann M et al. Treatment
requires investigation in different cohorts. and prognostic impact of transient leukemia in neonates with
Down syndrome. Blood 2008; 111: 2991–8.
Low-dose cytarabine is effective in improving the outcome 4 Muramatsu H, Kato K, Watanabe N et al. Risk factors for
for TAM patients with high WBC count, but a standard dose early death in neonates with Down syndrome and transient
and treatment schedule need to be established to ensure safe leukaemia. Br. J. Haematol. 2008; 142: 610–5.
and effective use of the drug for neonates with DS. Moreover, 5 Gamis AS, Alonzo TA, Gerbing RB et al. Natural history of
development of novel therapies is required for successful treat- transient myeloproliferative disorder clinically diagnosed in Down
syndrome neonates: A report from the Children’s Oncology
ment of the most severe form of TAM, complicated with ser- Group Study A2971. Blood 2011; 118: 6752–9; quiz 996.
ous effusion, multi-organ dysfunction, and liver fibrosis, 6 Zipursky A, Brown E, Christensen H, Sutherland R, Doyle J.
which results in fulminant hepatic failure. Leukemia and/or myeloproliferative syndrome in neonates
A number of studies using fetal liver cells, mouse models, with Down syndrome. Semin. Perinatol. 1997; 21: 97–101.
or iPSC have elucidated the roles of trisomy 21 and GATA1 7 Wechsler J, Greene M, McDevitt MA et al. Acquired
mutations in GATA1 in the megakaryoblastic leukemia of
mutations in the development of TAM. Nevertheless, the pre- Down syndrome. Nat. Genet. 2002; 32: 148–52.
cise mechanisms underlying the expansion and regression of 8 Greene ME, Mundschau G, Wechsler J et al. Mutations in
TAM cells remain to be clarified. GATA1 in both transient myeloproliferative disorder and

© 2018 Japan Pediatric Society


228 K Watanabe

acute megakaryoblastic leukemia of Down syndrome. Blood 28 Hojo S, Tsukimori K, Kitade S et al. Prenatal sonographic
Cells Mol. Dis. 2003; 31: 351–6. findings and hematological abnormalities in fetuses with
9 Groet J, McElwaine S, Spinelli M et al. Acquired mutations transient abnormal myelopoiesis with Down syndrome.
in GATA1 in neonates with Down’s syndrome with transient Prenat. Diagn. 2007; 27: 507–11.
myeloid disorder. Lancet 2003; 361: 1617–20. 29 Tamblyn JA, Norton A, Spurgeon L et al. Prenatal therapy in
10 Hitzler JK, Cheung J, Li Y, Scherer SW, Zipursky A. GATA1 transient abnormal myelopoiesis: A systematic review. Arch.
mutations in transient leukemia and acute megakaryoblastic Dis. Child. Fetal Neonatal Ed. 2016; 101: F67–71.
leukemia of Down syndrome. Blood 2003; 101: 4301–4. 30 Heald B, Hilden JM, Zbuk K et al. Severe TMD/AMKL with
11 Rainis L, Bercovich D, Strehl S et al. Mutations in exon 2 of GATA1 mutation in a stillborn fetus with Down syndrome.
GATA1 are early events in megakaryocytic malignancies Nat. Clin. Pract. Oncol. 2007; 4: 433–8.
associated with trisomy 21. Blood 2003; 102: 981–6. 31 Ishigaki H, Miyauchi J, Yokoe A et al. Expression of
12 Xu G, Nagano M, Kanezaki R et al. Frequent mutations in the megakaryocytic and myeloid markers in blasts of transient
GATA-1 gene in the transient myeloproliferative disorder of abnormal myelopoiesis in a stillbirth with Down syndrome:
Down syndrome. Blood 2003; 102: 2960–8. Report of histopathological findings of an autopsy case. Hum.
13 Ahmed M, Sternberg A, Hall G et al. Natural history of Pathol. 2011; 42: 141–5.
GATA1 mutations in Down syndrome. Blood 2004; 103: 32 Federmann B, Fasan A, Kagan KO, Haen S, Fend F. Transient
2480–9. abnormal myelopoiesis/acute megakaryoblastic leukemia
14 Gamis AS, Smith FO. Transient myeloproliferative disorder in diagnosed in the placenta of a stillborn Down syndrome fetus
children with Down syndrome: Clarity to this enigmatic with targeted next-generation sequencing. Leukemia 2015; 29:
disorder. Br. J. Haematol. 2012; 159: 277–87. 232–3.
15 Roy A, Roberts I, Vyas P. Biology and management of 33 Yagihashi N, Watanabe K, Yagihashi S. Transient abnormal
transient abnormal myelopoiesis (TAM) in children with myelopoiesis accompanied by hepatic fibrosis in two infants
Down syndrome. Semin. Fetal Neonatal. Med. 2012; 17: 196– with Down syndrome. J. Clin. Pathol. 1995; 48: 973–5.
201. 34 Schwab M, Niemeyer C, Schwarzer U. Down syndrome,
16 Bhatnagar N, Nizery L, Tunstall O, Vyas P, Roberts I. transient myeloproliferative disorder, and infantile liver
Transient abnormal myelopoiesis and AML in Down fibrosis. Med. Pediatr. Oncol. 1998; 31: 159–65.
syndrome: An update. Curr. Hematol. Malig. Rep. 2016; 11: 35 Park MJ, Sotomatsu M, Ohki K et al. Liver disease is
333–41. frequently observed in Down syndrome patients with transient
17 Saida S. Evolution of myeloid leukemia in children with abnormal myelopoiesis. Int. J. Hematol. 2014; 99: 154–61.
Down syndrome. Int. J. Hematol. 2016; 103: 365–72. 36 Ogawa J, Kanegane H, Tsuneyama K et al. Platelet-derived
18 Saida S, Watanabe K, Sato-Otsubo A et al. Clonal selection in growth factor may be associated with fibrosis in a Down
xenografted TAM recapitulates the evolutionary process of syndrome patient with transient myeloproliferative disorder.
myeloid leukemia in Down syndrome. Blood 2013; 121: Eur. J. Haematol. 2008; 81: 58–64.
4377–87. 37 Hattori H, Matsuzaki A, Suminoe A, Ihara K, Nakayama H,
19 Yoshida K, Toki T, Okuno Y et al. The landscape of somatic Hara T. High expression of platelet-derived growth factor and
mutations in Down syndrome-related myeloid disorders. Nat. transforming growth factor-beta 1 in blast cells from patients
Genet. 2013; 45: 1293–9. with Down Syndrome suffering from transient
20 Girodon F, Favre B, Couillaud G, Carli PM, Parmeland C, myeloproliferative disorder and organ fibrosis. Br. J.
Maynadie M. Immunophenotype of a transient Haematol. 2001; 115: 472–5.
myeloproliferative disorder in a newborn with trisomy 21. 38 Maroz A, Stachorski L, Emmrich S et al. GATA1s induces
Cytometry 2000; 42: 118–22. hyperproliferation of eosinophil precursors in Down syndrome
21 Karandikar NJ, Aquino DB, McKenna RW, Kroft SH. transient leukemia. Leukemia 2014; 28: 1259–70.
Transient myeloproliferative disorder and acute myeloid 39 Minakata S, Sakata N, Wada N et al. Liver fibrosis with
leukemia in Down syndrome. An immunophenotypic analysis. hypereosinophilia causing transient abnormal myelopoiesis.
Am. J. Clin. Pathol.. 2001; 116: 204–10. Pediatr. Int. 2016; 58: 1222–5.
22 Langebrake C, Creutzig U, Reinhardt D. Immunophenotype of 40 Hayashi Y, Eguchi M, Sugita K et al. Cytogenetic findings
Down syndrome acute myeloid leukemia and transient and clinical features in acute leukemia and transient
myeloproliferative disease differs significantly from other myeloproliferative disorder in Down’s syndrome. Blood 1988;
diseases with morphologically identical or similar blasts. Klin. 72: 15–23.
Padiatr. 2005; 217: 126–34. 41 Ge Y, Jensen TL, Stout M et al. The role of cytidine
23 Roberts I, Alford K, Hall G et al. GATA1-mutant clones are deaminase and GATA1 mutations in the increased cytosine
frequent and often unsuspected in babies with Down arabinoside sensitivity of Down syndrome myeloblasts and
syndrome: Identification of a population at risk of leukemia. leukemia cell lines. Cancer Res. 2004; 64: 728–35.
Blood 2013; 122: 3908–17. 42 Muramatsu H, Watanabe T, Hasegawa D et al. Prospective
24 Hoshi N, Hattori R, Hanatani K et al. Recent trends in the study of 168 infants with transient abnormal myelopoiesis
prevalence of Down syndrome in Japan, 1980-1997. Am. J. with Down syndrome: Japan Pediatric Leukemia/Lymphoma
Med. Genet. 1999; 84: 340–5. Study Group, TAM-10 Study. Abstract 1311; 57th Annual
25 Pine SR, Guo Q, Yin C, Jayabose S, Druschel CM, Sandoval Meeting of the American Society of Hematology, 2015. Blood
C. Incidence and clinical implications of GATA1 mutations in 2015; 126: 1311.
newborns with Down syndrome. Blood 2007; 110: 2128–31. 43 Flasinski M, Scheibke K, Zimmermann M et al. Low-dose
26 Strobelt N, Ghidini A, Locatelli A, Vergani P, Mariani S, cytarabine to prevent myeloid leukemia in children with Down
Biondi A. Intrauterine diagnosis and management of transient syndrome: TMD Prevention 2007 study. Blood Adv. 2018; 2:
myeloproliferative disorder. Am. J. Perinatol. 1995; 12: 132–4. 1532–40.
27 Zipursky A, Rose T, Skidmore M, Thorner P, Doyle J. 44 Chou S, Lodish HF. Fetal liver hepatic progenitors are
Hydrops fetalis and neonatal leukemia in Down syndrome. supportive stromal cells for hematopoietic stem cells. Proc.
Pediatr. Hematol. Oncol. 1996; 13: 81–7. Natl Acad. Sci. USA 2010; 107: 7799–804.

© 2018 Japan Pediatric Society


Review of TAM 229

45 Chou ST, Opalinska JB, Yao Y et al. Trisomy 21 enhances 61 Weiss MJ, Orkin SH. Transcription factor GATA-1 permits
human fetal erythro-megakaryocytic development. Blood survival and maturation of erythroid precursors by preventing
2008; 112: 4503–6. apoptosis. Proc. Natl Acad. Sci. USA 1995; 92: 9623–7.
46 Tunstall-Pedoe O, Roy A, Karadimitris A et al. Abnormalities 62 Ferreira R, Ohneda K, Yamamoto M, Philipsen S. GATA1
in the myeloid progenitor compartment in Down syndrome function, a paradigm for transcription factors in hematopoiesis.
fetal liver precede acquisition of GATA1 mutations. Blood Mol. Cell. Biol. 2005; 25: 1215–27.
2008; 112: 4507–11. 63 Crispino JD, Horwitz MS. GATA factor mutations in
47 Roy A, Cowan G, Mead AJ et al. Perturbation of fetal liver hematologic disease. Blood 2017; 129: 2103–10.
hematopoietic stem and progenitor cell development by 64 Freson K, Wijgaerts A, Van Geet C. GATA1 gene variants
trisomy 21. Proc. Natl Acad. Sci. USA 2012; 109: 17579–84. associated with thrombocytopenia and anemia. Platelets 2017;
48 Liu B, Filippi S, Roy A, Roberts I. Stem and progenitor cell 28: 731–4.
dysfunction in human trisomies. EMBO Rep. 2015; 16: 44–62. 65 Salek-Ardakani S, Smooha G, de Boer J et al. ERG is a
49 Chou ST, Byrska-Bishop M, Tober JM et al. Trisomy 21- megakaryocytic oncogene. Cancer Res. 2009; 69: 4665–73.
associated defects in human primitive hematopoiesis revealed 66 Toki T, Kanezaki R, Kobayashi E et al. Naturally occurring
through induced pluripotent stem cells. Proc. Natl Acad. Sci. oncogenic GATA1 mutants with internal deletions in transient
USA 2012; 109: 17573–8. abnormal myelopoiesis in Down syndrome. Blood 2013; 121:
50 Maclean GA, Menne TF, Guo G et al. Altered hematopoiesis 3181–4.
in trisomy 21 as revealed through in vitro differentiation of 67 Li Z, Godinho FJ, Klusmann JH, Garriga-Canut M, Yu C,
isogenic human pluripotent cells. Proc. Natl Acad. Sci. USA Orkin SH. Developmental stage-selective effect of somatically
2012; 109: 17567–72. mutated leukemogenic transcription factor GATA1. Nat.
51 Banno K, Omori S, Hirata K et al. Systematic cellular disease Genet. 2005; 37: 613–9.
models reveal synergistic interaction of trisomy 21 and 68 Shimizu R, Kobayashi E, Engel JD, Yamamoto M. Induction
GATA1 mutations in hematopoietic abnormalities. Cell Rep. of hyperproliferative fetal megakaryopoiesis by an N-
2016; 15: 1228–41. terminally truncated GATA1 mutant. Genes Cells 2009; 14:
52 Delabar JM, Theophile D, Rahmani Z et al. Molecular 1119–31.
mapping of twenty-four features of Down syndrome on 69 Stachura DL, Chou ST, Weiss MJ. Early block to
chromosome 21. Eur. J. Hum. Genet. 1993; 1: 114–24. erythromegakaryocytic development conferred by loss of
53 Ronan A, Fagan K, Christie L, Conroy J, Nowak NJ, Turner G. transcription factor GATA-1. Blood 2006; 107: 87–97.
Familial 4.3 Mb duplication of 21q22 sheds new light on the 70 Kanezaki R, Toki T, Terui K et al. Down syndrome and
Down syndrome critical region. J. Med. Genet. 2007; 44: 448–51. GATA1 mutations in transient abnormal myeloproliferative
54 Bourquin JP, Subramanian A, Langebrake C et al. disorder: Mutation classes correlate with progression to
Identification of distinct molecular phenotypes in acute myeloid leukemia. Blood 2010; 116: 4631–8.
megakaryoblastic leukemia by gene expression profiling. Proc. 71 Forestier E, Izraeli S, Beverloo B et al. Cytogenetic features
Natl Acad. Sci. USA 2006; 103: 3339–44. of acute lymphoblastic and myeloid leukemias in pediatric
55 Stankiewicz MJ, Crispino JD. ETS2 and ERG promote patients with Down syndrome: An iBFM-SG study. Blood
megakaryopoiesis and synergize with alterations in GATA-1 2008; 111: 1575–83.
to immortalize hematopoietic progenitor cells. Blood 2009; 72 Saida S, Toki T, Nishinaka-Arai Y et al. Dysregulation of
113: 3337–47. DNA methylation involves in progression of myeloid
56 Malinge S, Bliss-Moreau M, Kirsammer G et al. Increased leukemia in Down syndrome. Abstract; 57th Annual Meeting
dosage of the chromosome 21 ortholog Dyrk1a promotes of the American Society of Hematology, 2015. Blood 2015;
megakaryoblastic leukemia in a murine model of Down 126: 89.
syndrome. J. Clin. Invest. 2012; 122: 948–62. 73 Khan I, Malinge S, Crispino J. Myeloid leukemia in Down
57 Ichikawa M, Yoshimi A, Nakagawa M, Nishimoto N, Watanabe- syndrome. Crit. Rev. Oncog. 2011; 16: 25–36.
Okochi N, Kurokawa M. A role for RUNX1 in hematopoiesis and 74 Nowell PC. The clonal evolution of tumor cell populations.
myeloid leukemia. Int. J. Hematol. 2013; 97: 726–34. Science 1976; 194: 23–8.
58 Kirsammer G, Jilani S, Liu H et al. Highly penetrant 75 Welch JS, Ley TJ, Link DC et al. The origin and evolution of
myeloproliferative disease in the Ts65Dn mouse model of mutations in acute myeloid leukemia. Cell 2012; 150: 264–78.
Down syndrome. Blood 2008; 111: 767–75. 76 Xu G, Kato K, Toki T, Takahashi Y, Terui K, Ito E.
59 Carmichael CL, Majewski IJ, Alexander WS et al. Development of acute megakaryoblastic leukemia from a
Hematopoietic defects in the Ts1Cje mouse model of Down minor clone in a Down syndrome patient with clinically overt
syndrome. Blood 2009; 113: 1929–37. transient myeloproliferative disorder. J. Pediatr. Hematol.
60 Alford KA, Slender A, Vanes L et al. Perturbed Oncol. 2006; 28: 696–8.
hematopoiesis in the Tc1 mouse model of Down syndrome. 77 Greaves M, Maley CC. Clonal evolution in cancer. Nature
Blood 2010; 115: 2928–37. 2012; 481: 306–13.

© 2018 Japan Pediatric Society

You might also like