Patients with constitutional trisomy 21 (Down syndrome [DS]) are at high risk of developing both acute lymphoblastic leukemia and myeloid leukemia (DS-ALL and ML-DS, respectively). These malignant conditions are associated with distinctive epidemiology, genetic signatures, and clinical outcomes compared to those of non-DS leukemias.1-3  DS-ALL is nearly exclusively of the B-cell lineage and occurs in children and adolescents/young adults, with extremely few cases reported in infants younger than one year. Many DS-ALL cases are cytogenetically normal, suggesting a role for activating mutations as leukemic drivers.3  Almost all ML-DS cases are of the acute megakaryoblastic subtype and occur in children younger than four years. ML-DS commonly ensues following the canonical preleukemic transient myeloproliferative disorder (TMD) characterized by somatic GATA1 mutations and is hypothesized to arise from a residual TMD population that subsequently acquires additional driver mutation(s).1, 4, 5  Patients with DS-ALL have inferior clinical outcomes versus those with non-DS-ALL, which is attributed to higher rates of treatment-associated mortality (TRM) and relapse.6, 7  Conversely, children with ML-DS have excellent chemosensitivity and high overall survival (OS), although survival of those with relapsed ML-DS is dismal.5 

Attempts to reduce the treatment toxicities for pediatric patients with DS-associated leukemias have shown mixed success to date. TRM is typically due to infection and can occur during any phase of treatment, including maintenance.6  In addition to mortality resulting from the toxicity itself, individual treatment modifications in response to toxicities seem to be an important cause of lower event-free survival (EFS) in children with DS-ALL.7  Prior strategies in the Children’s Oncology Group (COG) AALL0932 and AALL1131 clinical trials to reduce anthracycline exposure, modify methotrexate dosing, decrease vincristine/glucocorticoid pulses, and intensify supportive care for patients with DS-ALL did not appreciably decrease TRM.8  The current COG AALL1731 trial further reduces chemotherapy toxicity by non-randomly assigning children with DS-ALL to three cycles of blinatumomab, a bispecific CD19×CD3 antibody immunotherapy, and omitting daunomycin in induction (all DS-ALL) and the cyclophosphamide/cytarabine-based second month of delayed intensification (high-risk DS-ALL only). Together, these two phases of treatment represent almost all of the TRM in high-risk DS-ALL.9  Preliminary analysis of interim AALL1731 data showed similar TRM rates (approximately 8%) among patients with DS-ALL on AALL1131 and AALL1731 however, and an increased blinatumomab-associated seizure risk that can be mitigated with antiepileptic drug prophylaxis.10 

The reasons for the significantly higher rate of relapse in DS-ALL are likely multifactorial, including underlying leukemia biology, decreased immune surveillance compared to non-DS patients, and under-reported treatment reductions in response to adverse events.3, 6  A specific association between the distinct genetics of DS-ALL and poorer outcomes has not been established. For example, CRLF2 rearrangements with frequent JAK2 co-mutations that are associated with poor outcomes in patients with non-DS-ALL11  occur in 50 to 60 percent of DS-ALL and seem to have limited prognostic significance.6, 12, 13  Conversely, favorable genetic alterations in non-DS-ALL, such as ETV6-RUNX1 fusions, are also associated with good prognosis in DS-ALL.6, 7 

Given the superior EFS of ML-DS compared to non-ML-DS, a major focus of recent clinical trials has been to reduce treatment intensity and toxicity.5, 14, 15  The COG AAML0431 trial aimed to improve EFS and reduce TRM by administering high-dose cytarabine (HiDAC) during induction II instead of intensification, decreasing total anthracycline exposure, and reducing prophylactic intrathecal chemotherapy.14  While the adjustments to anthracyclines and intrathecal chemotherapy did not affect EFS or OS, the earlier HiDAC exposure improved both metrics, though adverse events occurred more frequently during induction II.5, 14  The COG AAML1531 trial subsequently had the goal of omitting HiDAC in standard-risk patients with ML-DS who achieved measurable residual disease (MRD) less than 0.05 percent at the end of induction I.5  Unfortunately, children who did not receive HiDAC-based induction II had significantly higher relapse rates and worse EFS compared to MRD-negative patients treated on AAML0431, demonstrating that HiDAC inclusion is an essential component of ML-DS therapy.5  Interestingly, a Japanese Childhood AML Cooperative Study Group trial investigating a similar cytarabine dose-reduction reported much lower TRM and no significant differences in three-year EFS and OS,16  concordant with earlier studies.15, 17  Differences in anthracycline pharmacodynamics and other small differences among treatment protocols have been hypothesized to account for these discrepant results,18  but these effects remain difficult to isolate, and HiDAC remains essential in ML-DS therapy in the United States.

The small numbers of children with relapsed/refractory ML-DS have limited the ability to identify prognostic molecular characteristics and other biomarkers. In addition to MRD positivity, older age at diagnosis (≥4 years) has been suggested to portend an inferior prognosis,17, 19  though such ML-DS cases are usually more genetically similar to non-DS-associated AML.20  The genetic characteristics of ML-DS blasts are clearly distinct from non-ML-DS cells,5  which is consistent with the unique cellular origins of ML-DS, but the exact significance of the pathogenic GATA1 mutations remains unclear. In a phenomenon termed “silent TMD,” as many as 30 percent of neonates with DS have been reported to have GATA1 mutations without detectable TMD in peripheral blood.17, 21, 22  Interestingly, no association has been identified between specific GATA1 mutations and risk of ultimately developing ML-DS.23 

Patients with relapsed DS-ALL24, 25  or ML-DS5, 19  have poor prognoses, with successful salvage (including allogeneic hematopoietic stem cell transplantation) often limited by TRM and subsequent relapses.24, 25  Exclusion of children with DS-associated leukemias from many early-phase clinical trials has hampered both access to innovative new therapies and tailored protocols focused upon DS-specific toxicity reduction.26  Emerging data encouragingly suggest that patients with relapsed/refractory DS-ALL tolerate CD19-directed cellular therapy well, which can perhaps be used as definitive therapy without hematopoietic stem cell transplantation in some patients.27  Similar advances are still needed for children with relapsed/refractory ML-DS.

Dr. Takasaki and Dr. Tasian indicated no relevant conflicts of interest.

1
Gruber
TA
,
Downing
JR
.
The biology of pediatric acute megakaryoblastic leukemia
.
Blood
.
2015
;
126
(
8
):
943
949
.
2
Forestier
E
,
Izraeli
S
,
Beverloo
B
, et al
.
Cytogenetic features of acute lymphoblastic and myeloid leukemias in pediatric patients with Down syndrome: an iBFM-SG study
.
Blood
.
2008
;
111
(
3
):
1575
1583
.
3
Izraeli
S
,
Vora
A
,
Zwaan
CM
, et al
.
How I treat ALL in Down’s syndrome: pathobiology and management
.
Blood
.
2014
;
123
(
1
):
35
40
.
4
McNulty
M
,
Crispino
JD
.
Acute megakaryocytic leukemia
.
Cold Spring Harb Perspect Med
.
2020
;
10
(
2
):
a034884
.
5
Hitzler
J
,
Alonzo
T
,
Gerbing
R
, et al
.
High-dose AraC is essential for the treatment of ML-DS independent of postinduction MRD: results of the COG AAML1531 trial
.
Blood
.
2021
;
138
(
23
):
2337
2346
.
6
Buitenkamp
TD
,
Izraeli
S
,
Zimmermann
M
, et al
.
Acute lymphoblastic leukemia in children with Down syndrome: a retrospective analysis from the Ponte di Legno study group
.
Blood
.
2014
;
123
(
1
):
70
77
.
7
Matloub
Y
,
Rabin
KR
,
Ji
L
, et al
.
Excellent long-term survival of children with Down syndrome and standard-risk ALL: a report from the Children’s Oncology Group
.
Blood Adv
.
2019
;
3
(
11
):
1647
1656
.
8
Rabin
KR
,
Hitzler
J
,
Rodriguez
V
, et al
.
Treatment-related mortality (TRM) in children with Down syndrome (DS) and B-lymphoblastic leukemia (B-ALL): an interim report from the Children’s Oncology Group trials AALL0932 and AALL1131
.
Blood
.
2015
;
126
(
23
):
2502
2502
.
9
Children’s Oncology Group, AALL1731
.
A phase 3 trial investigating blinatumomab in combination with chemotherapy in patients with newly diagnosed standard risk or Down syndrome B-lymphoblastic leukemia (B-ALL) and the treatment of patients with localized B-lymphoblastic leukemia (B-LLy)
. Available at: https://clinicaltrials.gov/ct2/show/NCT03914625.
10
Li
AM
,
Rabin
KR
,
Kairalla
J
, et al
.
Blinatumomab associated seizure risk in patients with Down syndrome and B-lymphoblastic leukemia: an interim report from Children’s Oncology Group (COG) study AALL1731
.
Blood
.
2021
;
138
(
Suppl 1
):
2304
2304
.
11
Tasian
SK
,
Dai
Y
,
Devidas
M
, et al
.
Outcomes of patients with CRLF2 -overexpressing acute lymphoblastic leukemia without Down syndrome: a report from the Children’s Oncology Group
.
Blood
.
2020
;
136
(
Suppl 1
):
45
46
.
12
Hertzberg
L
,
Vendramini
E
,
Ganmore
I
, et al
.
Down syndrome acute lymphoblastic leukemia, a highly heterogeneous disease in which aberrant expression of CRLF2 is associated with mutated JAK2: a report from the International BFM Study Group
.
Blood
.
2010
;
115
(
5
):
1006
1017
.
13
Rabin
KR
,
Devidas
M
,
Chen
Z
, et al
.
Outcomes of patients with Down syndrome and CRLF2-overexpressing acute lymphoblastic leukemia (ALL): a report from the Children’s Oncology Group (COG)
.
Blood
.
2020
;
136
(
Supplement 1
):
44
45
.
14
Taub
JW
,
Berman
JN
,
Hitzler
JK
, et al
.
Improved outcomes for myeloid leukemia of Down syndrome: a report from the Children’s Oncology Group AAML0431 trial
.
Blood
.
2017
;
129
(
25
):
3304
3313
. doi:.
15
Uffmann
M
,
Rasche
M
,
Zimmermann
M
, et al
.
Therapy reduction in patients with Down syndrome and myeloid leukemia: the international ML-DS 2006 trial
.
Blood
.
2017
;
129
(
25
):
3314
3321
.
16
Kudo
K
,
Kojima
S
,
Tabuchi
K
, et al
.
Prospective Study of a pirarubicin, intermediate-dose cytarabine, and etoposide regimen in children with Down syndrome and acute myeloid leukemia: the Japanese Childhood AML Cooperative Study Group
.
J Clin Oncol
.
2007
;
25
(
34
):
5442
5447
.
17
Boucher
AC
,
Caldwell
KJ
,
Crispino
JD
, et al
.
Clinical and biological aspects of myeloid leukemia in Down syndrome
.
Leukemia
.
2021
;
35
(
12
):
3352
3360
.
18
Ravindranath
Y
,
O’Dwyer
KM
.
In response to back to HiDAC
.
The Hematologist
2022
;
19
(
4
):
8
.
19
Taga
T
,
Saito
AM
,
Kudo
K
, et al
.
Clinical characteristics and outcome of refractory/relapsed myeloid leukemia in children with Down syndrome
.
Blood
.
2012
;
120
(
9
):
1810
1815
.
20
Hasle
H
,
Abrahamsson
J
,
Arola
M
, et al
.
Myeloid leukemia in children 4 years or older with Down syndrome often lacks GATA1 mutation and cytogenetics and risk of relapse are more akin to sporadic AML
.
Leukemia
.
2008
;
22
(
7
):
1428
1430
.
21
Garnett
C
,
Cruz Hernandez
D
,
Vyas
P
.
GATA1 and cooperating mutations in myeloid leukaemia of Down syndrome
.
IUBMB Life
.
2020
;
72
(
1
):
119
130
.
22
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
.
23
Alford
KA
,
Reinhardt
K
,
Garnett
C
, et al
.
Analysis of GATA1 mutations in down syndrome transient myeloproliferative disorder and myeloid leukemia
.
Blood
.
Published online
2011
;
118
(
8
):
2222
2238
.
24
Meyr
F
,
Escherich
G
,
Mann
G
, et al
.
Outcomes of treatment for relapsed acute lymphoblastic leukaemia in children with Down syndrome
.
Br J Haematol
.
2013
;
162
(
1
):
98
106
.
25
Hitzler
JK
,
He
W
,
Doyle
J
, et al
.
Outcome of transplantation for acute myelogenous leukemia in children with Down syndrome
.
Biol Blood Marrow Transplant
.
2013
;
19
(
6
):
893
897
.
26
Rabin
K
,
Izraeli
S
,
Hijiya
N
, et al
.
Need for new thinking: treatment of relapsed leukemia in children with Down syndrome
.
Pediatr Blood Cancer
.
2019
;
66
(
6
):
e27644
.
27
Laetsch
TW
,
Maude
SL
,
Grupp
SA
, et al
.
CTL019 therapy appears safe and effective in pediatric patients with Down syndrome with relapsed/refractory (r/r) acute lymphoblastic leukemia
.
Blood
.
2017
;
130
(
Suppl 1
):
1280
.