Wagenblast
E
,
Araújo
J
,
Gan
OI
, et al
.
Mapping the cellular origin and early evolution of leukemia in Down syndrome
.
Science
.
2021
;
373
(
6551
):
eabf6202
. .

For patients with childhood malignancies, leukemia-initiating genetic mutations frequently occur antenatally, followed by the acquisition of additional mutations in the first few years of life.1  A classic example of this phenomenon occurs in children with trisomy 21 (T21) who have a 150-fold increased risk of developing acute myeloid leukemia (AML). This is recognized as a unique form of pediatric AML: myeloid leukemia associated with Down syndrome (ML-DS).25  Real-world observational studies in children born with T21 have identified somatic GATA1 mutations as the first molecular event leading to the development of a preleukemic clone, seeding the subsequent development of ML-DS.6 GATA1 mutations are found in approximately 30 percent of children born with T21, and have been detected as early as 21 weeks gestation,7  suggesting that the first mutations required for the development of ML-DS occur in utero. GATA1 mutations result in the production of a truncated isoform of GATA1 (GATA1-short or GATA1s),8  which functionally impacts normal hematopoiesis leading to thrombocytopenia, excess blasts in the peripheral blood film, and a pre-leukemic state known as “transient abnormal myelopoiesis” (TAM).6,911  TAM is only described in the context of T21 and GATA1 mutations; it has never been associated with leukemia in the absence of T21.12  In most cases, TAM spontaneously resolves in the neonatal period with only 10 to 20 percent of this patient group subsequently developing ML-DS, typically within the first four years of life.1315  ML-DS cases always carry GATA1 mutations, demonstrating that this is an absolute requirement for the development of ML-DS. However, additional molecular defects, predominantly in cohesin complex genes such as STAG2, are detected in patient samples following transformation to ML-DS.1618 

These observations raise several questions regarding the evolution of ML-DS: What is the cell of origin of ML-DS during fetal life? Are these ML-DS–initiating cells unique to fetal hematopoiesis? What is the molecular mechanism by which an extra copy of chromosome 21 predisposes to a pre-leukemic TAM phenotype when a GATA1s mutation is acquired? Does the cell of origin (or underlying molecular mechanisms) present possible therapeutic vulnerabilities that could be used to treat or even prevent leukemia development in high-risk children? To experimentally address these important questions, sophisticated model systems are required; however, modelling and characterization of aggressive childhood leukemias is extremely challenging due to early initiating events occurring during antenatal life in developmentally restricted hematopoietic stem and progenitor cell (HSPC) populations. Advances in genome editing technologies have created new opportunities to manipulate HSPCs at different stages of ontogeny to address these challenging problems.

Dr. Elvin Wagenblast and colleagues used CRISPR/Cas9 genome editing of HSPC from human fetal liver to induce TAM- and ML-DS–related mutations. The authors characterized the hematopoietic phenotype of purified HSPC derived from normal disomic fetal liver (N-FL) and T21 fetal liver (T21-FL). T21-FL HSPCs showed reduced proliferation in vitro, increased megakaryocyte-erythroid progenitors (MEP) as previously described,19  and a lower level of engraftment in xenotranplantation assays. A preleukemic TAM-like phenotype was initiated in T21-FL but not in N-FL through the introduction of GATA1 mutations into long-term hematopoietic stem cells (LT-HSC). This in turn induced a proliferation advantage in T21-FL LT-HSC, accompanied by an increased production of CD41+ megakaryocytes, higher level of engraftment in xenotransplant assays, and an increase in blasts, together indicating that T21-FL LT-HSC is likely the cell of origin for ML-DS. Importantly, serial transplantation studies demonstrated that this T21-FL–induced preleukemia underwent spontaneous resolution, recapitulating the phenotype of TAM. The authors explored the molecular mechanism of the T21-associated FL development, and through analysis of the distinct pattern of DNA-binding occupancy of GATA1s, which, combined with gene expression analysis and lentiviral transduction, identified that simultaneous overexpression of three chromosome 21 microRNAs (MiR-99a, miR-125b-2, and miR-155) could recapitulate a T21 phenotype. Furthermore, knockout of these micro-RNAs inhibited the development of GATA1s-induced preleukemia in T21-FL.

Introduction of GATA1s in combination with STAG2 deletion or other cohesin complex genes led to the development of a leukemic phenotype with reduced survival of engrafted mice and markedly increased blast cells. Intriguingly, this phenotype was largely independent of T21. Unlike the GATA1s T21-FL pre-leukemia, multiple different progenitor cell populations were shown to be capable of propagating the leukemia. Moreover, the leukemia was serially transplantable but intriguingly could only be induced using fetal and early postnatal, but not adult, HPSCs, mimicking the developmental restriction of ML-DS. The leukemia was characterized by aberrant CD117 expression in the leukemic HSPCs, which the authors demonstrated could be used as a marker for disease-propagating cells. This was used as a potential therapeutic target through demonstration of a promising effect of KIT inhibitors on the GATA1s-induced preleukemic state and primary TAM samples, making it worthy of future clinical evaluation as a potential therapy to prevent leukemic progression of TAM.

Taken together, this impressive work showcases how technological advances can be utilized to provide novel insights into longstanding questions in leukemia biology. The distinct cellular and molecular dependencies, across different developmental states, help to explain why childhood leukemias are often associated with unique genetic features. The molecular mechanism of this developmental restriction, however, remains unexplained and will be an important area for future study as it might reveal additional therapeutic vulnerabilities. A remarkable aspect of this study is the degree to which the phenotype of human disease was recapitulated through the elegant model systems used, and this work will serve as a paradigm for the study of other childhood leukemias with prenatal origin.

Dr. Louka and Dr. Mead indicated no relevant conflicts of interest.

1.
Greaves
M
.
A causal mechanism for childhood acute lymphoblastic leukaemia
.
Nat Rev Cancer
.
2018
;
18
(
8
):
471
484
.
2.
Hasle
H
,
Clemmensen
IH
,
Mikkelsen
M
.
Risks of leukaemia and solid tumours in individuals with Down's syndrome
.
Lancet
.
2000
;
355
(
9199
):
165
169
.
3.
Zipursky
A
,
Thorner
P
,
De Harven
E
, et al
.
Myelodysplasia and acute megakaryoblastic leukemia in Down's syndrome
.
Leuk Res
.
1994
;
18
(
3
):
163
171
.
4.
Ravindranath
Y
.
Down syndrome and acute myeloid leukemia: the paradox of increased risk for leukemia and heightened sensitivity to chemotherapy
.
J Clin Oncol
.
2003
;
21
(
18
):
3385
3387
.
5.
Hasle
H
,
Niemeyer
CM
,
Chessells
JM
, et al
.
A pediatric approach to the WHO classification of myelodysplastic and myeloproliferative diseases
.
Leukemia
.
2003
;
17
(
2
):
277
282
.
6.
Roberts
I
,
Alford
K
,
Hall
G
, et al
.
GATA1-mutant clones are frequent and often unsuspected in babies with Down syndrome: identification of a population at risk of leukemia
.
Blood
.
2013
;
122
(
24
):
3908
3917
.
7.
Taub
JW
,
Mundschau
G
,
Ge
Y
, et al
.
Prenatal origin of GATA1 mutations may be an initiating step in the development of megakaryocytic leukemia in Down syndrome
.
Blood
.
2004
;
104
(
5
):
1588
1589
.
8.
Halsey
C
,
Tunstall
O
,
Gibson
B
, et al
.
Role of GATA-1s in early hematopoiesis and differences between alternative splicing in human and murine GATA-1
.
Blood
.
2010
;
115
(
16
):
3415
3416
.
9.
Wechsler
J
,
Greene
M
,
McDevitt
MA
, et al
.
Acquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome
.
Nat Genet
.
2002
;
32
(
1
):
148
152
.
10.
Ahmed
M
,
Sternberg
A
,
Hall
G
, et al
.
Natural history of GATA1 mutations in Down syndrome
.
Blood
.
2004
;
103
(
7
):
2480
2489
.
11.
Alford
KA
,
Reinhardt
K
,
Garnett
C
, et al
.
Analysis of GATA1 mutations in Down syndrome transient myeloproliferative disorder and myeloid leukemia
.
Blood
.
2011
;
118
(
8
):
2222
2238
.
12.
Roberts
I
,
Vyas
P
.
Sowing the seeds of leukemia before birth
.
Science
.
2021
;
373
(
6551
):
155
156
.
13.
Gamis
AS
,
Alonzo
TA
,
Gerbing
RB
, et al
.
Natural history of transient myeloproliferative disorder clinically diagnosed in Down syndrome neonates: a report from the Children's Oncology Group Study A2971
.
Blood
.
2011
;
118
(
26
):
6752
6759
.
14.
Groet
J
,
McElwaine
S
,
Spinelli
M
, et al
.
Acquired mutations in GATA1 in neonates with Down's syndrome with transient myeloid disorder
.
Lancet
.
2003
;
361
(
9369
):
1617
1620
.
15.
Klusmann
JH
,
Creutzig
U
,
Zimmermann
M
, et al
.
Treatment and prognostic impact of transient leukemia in neonates with Down syndrome
.
Blood
.
2008
;
111
(
6
):
2991
2998
.
16.
Labuhn
M
,
Perkins
K
,
Matzk
S
, et al
.
Mechanisms of progression of myeloid preleukemia to transformed myeloid leukemia in children with Down syndrome
.
Cancer Cell
.
2019
;
36
(
3
):
340
.
17.
Yoshida
K
,
Toki
T
,
Okuno
Y
, et al
.
The landscape of somatic mutations in Down syndrome-related myeloid disorders
.
Nat Genet
.
2013
;
45
(
11
):
1293
1299
.
18.
Nikolaev
SI
,
Santoni
F
,
Vannier
A
, et al
.
Exome sequencing identifies putative drivers of progression of transient myeloproliferative disorder to AMKL in infants with Down syndrome
.
Blood
.
2013
;
122
(
4
):
554
561
.
19.
Roy
A
,
Cowan
G
,
Mead
AJ
, et al
.
Perturbation of fetal liver hematopoietic stem and progenitor cell development by trisomy 21
.
Proc Natl Acad Sci U S A
.
2012
;
109
(
43
):
17579
17584
.