Brady
SW
,
Roberts
KG
,
Gu
Z
, et al
.
The genomic landscape of pediatric acute lymphoblastic leukemia
.
Nat Genet
.
2022
;
54
(
9
):
1376
-
1389
.

Acute lymphoblastic leukemia (ALL) is the most common cancer occurring in children. Survival now exceeds 90 percent with multidrug frontline chemotherapy regimens tested and refined via cooperative group clinical trials during the past 40 years.1,2  Modern risk stratification and therapeutic selection for pediatric patients with ALL depends on leukemia immunophenotype, age, and white blood cell count at diagnosis; central nervous system and other extramedullary sites of leukemia involvement; cytomolecular characteristics; and response to initial chemotherapy via minimal/measurable residual disease (MRD) assessment.3,4  Patients with specific high-risk genetic alterations require postinduction chemotherapy intensification and, in some cases, additional drugs (e.g., tyrosine kinase inhibitors for Ph+ ALL) or immunotherapy/hematopoietic stem cell transplantation to maximize cure.5-7  Conversely, children with “low risk” ALL who have particularly favorable features (National Cancer Institute standard risk by age and white blood cell count, no central nervous system involvement [CNS1], ETV6::RUNX1 fusion or high hyperdiploidy with trisomies 4 and 10, and negative early- and end-induction MRD) have event-free and overall survival rates exceeding 95 percent and benefit from chemotherapy reduction that preserves these outstanding cure rates while reducing therapy-associated toxicity.8-10 

A recent landmark collaborative study, led by first authors Drs. Samuel Brady and Kathryn Roberts and senior authors Drs. Stephen Hunger, Jinghui Zhang, and Charles Mullighan, performed comprehensive paired tumor/normal whole-exome sequencing, whole-genome sequencing, single nucleotide polymorphism array, and/or tumor-only whole transcriptome RNA sequencing on 2,288 and 466 cases of newly diagnosed B-cell ALL (B-ALL) and T-cell ALL (T-ALL), respectively, occurring in children and adolescents/young adults treated on Children’s Oncology Group or St. Jude Children’s Research Hospital clinical trials. Results from this study validated the prognostic significance of more than 30 known genetic subtypes of pediatric ALL.11  The results also refined or newly defined the spectrum of clonality, co-occurrence, and acquisition timing of germline and somatic leukemia-associated primary and secondary genetic alterations. The investigators reported a median of four somatic oncogenic driver mutations per case and noted that nearly 3 percent of cases harbored pathogenic, or likely-pathogenic, germline variants associated with cancer predisposition. New findings from this study also shed further light on the probable sequence of fusion and mutation acquisition in specific childhood ALL subtypes and discovery of potential ultraviolet light–induced ALL-associated mutations that occur at different frequencies among patients of specific race/ethnicity/ancestry backgrounds. Importantly, the authors highlighted differences in timing acquisition of kinase-signaling gene mutations in between B-ALL and T-ALL cases, cautioning that clinical targeting with tyrosine kinase inhibitor–based therapies may have differential activity (i.e., lesser in B-ALL with later-gained mutations and greater in T-ALL with earlier-gained mutations).

Lastly, the investigators report detailed overall survival data for children with 22 B-ALL and 11 T-ALL genetic subtypes, as well as for “B-other” and “T-other” cases lacking known canonical genetic rearrangements or expression signatures. These clinical outcomes data increase our robustness of knowledge regarding newly identified B-ALL subtypes, such as those involving DUX4, MEF2D, ZNF384, and NUTM1 rearrangements. In total, the investigators identified 376 putative ALL-associated driver genes involved in perturbation of lymphoid maturation, transcriptional dysregulation, cell cycle regulation, chromatin modification, and kinase signaling, which further highlights the genetic complexity and heterogeneity of childhood ALL that often confers prognostic and/or therapeutic significance.

This important genomic analysis details the comprehensive landscape of childhood B-ALL and T-ALL in the largest cohort of newly-diagnosed cases to date and confers critical new biologic insight that will continue to refine risk stratification of children and adolescents/young adults with ALL and to enhance the application of relevant precision medicine therapeutic approaches.9,10 

Dr. Tasian indicated no relevant conflicts of interest.

1
Hunger
SP
,
Mullighan
CG
.
Acute lymphoblastic leukemia in children
.
N Engl J Med
.
2015
;
373
(
16
):
1541
1552
.
2
Inaba
H
,
Mullighan
CG
.
Pediatric acute lymphoblastic leukemia
.
Haematologica
.
2020
;
105
(
11
):
2524
2539
.
3
Hunger
SP
,
Mullighan
CG
.
Redefining ALL classification: toward detecting high-risk ALL and implementing precision medicine
.
Blood
.
2015
;
125
(
26
):
3977
3987
.
4
Smith
M
,
Arthur
D
,
Camitta
B
, et al
.
Uniform approach to risk classification and treatment assignment for children with acute lymphoblastic leukemia
.
J Clin Oncol
.
1996
;
14
(
1
):
18
24
.
5
Nachman
JB
,
Sather
HN
,
Sensel
MG
, et al
.
Augmented post-induction therapy for children with high-risk acute lymphoblastic leukemia and a slow response to initial therapy
.
N Engl J Med
.
1998
;
338
(
23
):
1663
1671
.
6
Schultz
KR
,
Bowman
WP
,
Aledo
A
, et al
.
Improved early event-free survival with imatinib in Philadelphia chromosome-positive acute lymphoblastic leukemia: a children’s oncology group study
.
J Clin Oncol
.
2009
;
27
(
31
):
5175
5181
.
7
Schultz
KR
,
Carroll
A
,
Heerema
NA
, et al
.
Long-term follow-up of imatinib in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia: Children’s Oncology Group study AALL0031
.
Leukemia
.
2014
;
28
(
7
):
1467
1471
.
8
Vora
A
,
Goulden
N
,
Wade
R
, et al
.
Treatment reduction for children and young adults with low-risk acute lymphoblastic leukaemia defined by minimal residual disease (UKALL 2003): a randomised controlled trial
.
Lancet Oncol
.
2013
;
14
(
3
):
199
-
209
.
9
Pieters
R
,
de Groot-Kruseman
H
,
Van der Velden
V
, et al
.
successful therapy reduction and intensification for childhood acute lymphoblastic leukemia based on minimal residual disease monitoring: study ALL10 From the Dutch Childhood Oncology Group
.
J Clin Oncol
.
2016
;
34
(
22
):
2591
2601
.
10
Schore
RJ
,
Angiolillo
AJ
,
Kairalla
JA
, et al
.
Outcomes with reduced intensity therapy in a low-risk subset of children with National Cancer Institute(NCI) standard-risk (SR) B-lymphoblastic leukemia (B-ALL): a report from Children’s Oncology Group (COG) AALL0932
.
J Clin Oncol
.
2020
;
38
(
15 suppl
):
10509
10509
.
11
Jeha
S
,
Choi
J
,
Roberts
KG
, et al
.
Clinical significance of novel subtypes of acute lymphoblastic leukemia in the context of minimal residual disease-directed therapy
.
Blood Cancer Discov
.
2021
;
2
(
4
):
326
337
.