In this issue of Blood, Escherich et al report that deleterious heterozygous germ line variants in the nibrin (NBN) gene result in a loss of protein stability and are associated with an increased risk of B-cell acute lymphoblastic leukemia (ALL) in children.1 

The role of germ line genetic variation in the development of childhood ALL is well established. Sequencing studies in familial ALL and in large numbers of sporadic cases have revealed the contribution of rare and pathogenic/likely pathogenic germ line variants in predisposition genes, including in the lymphoid transcription factor genes IKZF1, ETV6, and PAX5, and in the tumor suppressor gene TP53.2-5 In addition, several autosomal recessive chromosome instability disorders, including Fanconi anemia, ataxia telangiectasia, Bloom syndrome, and Nijmegen breakage syndrome (NBS), are characterized by varying clinical manifestations as well as an increased risk of childhood ALL.6 

NBS is caused by biallelic loss-of-function germ line variants in the DNA damage repair gene NBN, with most patients with NBS being of Eastern European origin and harboring the p.K219fs founder mutation. NBS is associated with a high risk of lymphoid malignancies, especially lymphomas, but also for ALL,7,8 whereas heterozygous carriers of the p.K219fs variant, although clinically asymptomatic, are reported to have an increased risk of adult-onset cancers.9 Although putatively deleterious heterozygous germ line variants have been identified in a handful of childhood patients with ALL,10 the role of NBN variants in ALL susceptibility has not been comprehensively studied.

To address this knowledge gap, Escherich et al describe the detection and characterization of germ line NBN variants in a cohort of ≈4300 patients with B-cell ALL (see figure). They identified rare variants that were computationally predicted to be deleterious in 1.2% of patients with B-cell ALL, a frequency significantly higher than that found in population-based controls. Employing the consistent summary counts–based rare variant burden test (CoCoRV), which to some extent helps to account for ancestry differences between cases and controls (more on this later), they estimated that rare and predicted deleterious NBN variants confer a 1.8-fold risk of B-cell ALL. Several variants, including the NBS founder mutation p.K219fs, were individually enriched in patients with ALL compared with controls. No biallelic NBN variants were detected among the patients, albeit this is not entirely surprising given the rare nature of NBS.

Detection and characterization of germ line NBN variants in childhood B-cell ALL (B-ALL). Targeted sequencing of NBN was performed in 4325 patients with B-ALL enrolled in the Children’s Oncology Group and St. Jude Children’s Research Hospital trials (left). Rare (allele frequency, <0.001) and predicted deleterious (Combined Annotation Dependent Depletion [CADD] score, >20) variants were identified in 1.2% of patients, and in 0.4% of controls (n = 118 479) using summary counts from whole-exome sequencing (WES) in the Genome Aggregation Database. Ancestry stratified analysis of enrichment of NBN variants in B-ALL was conducted using CoCoRV, with patients and controls split into 5 ancestry groups, defined as non-Finnish European, African American, admixed American (Hispanic/Latino), South Asian, and East Asian individuals (as represented by the different colors). Of the 25 rare and predicted deleterious NBN variants identified in patients, 14 were experimentally shown to be nonfunctional or partially functional based on protein stability and drug sensitivity assays. OR, odds ratio. Figure created with BioRender.com.

Detection and characterization of germ line NBN variants in childhood B-cell ALL (B-ALL). Targeted sequencing of NBN was performed in 4325 patients with B-ALL enrolled in the Children’s Oncology Group and St. Jude Children’s Research Hospital trials (left). Rare (allele frequency, <0.001) and predicted deleterious (Combined Annotation Dependent Depletion [CADD] score, >20) variants were identified in 1.2% of patients, and in 0.4% of controls (n = 118 479) using summary counts from whole-exome sequencing (WES) in the Genome Aggregation Database. Ancestry stratified analysis of enrichment of NBN variants in B-ALL was conducted using CoCoRV, with patients and controls split into 5 ancestry groups, defined as non-Finnish European, African American, admixed American (Hispanic/Latino), South Asian, and East Asian individuals (as represented by the different colors). Of the 25 rare and predicted deleterious NBN variants identified in patients, 14 were experimentally shown to be nonfunctional or partially functional based on protein stability and drug sensitivity assays. OR, odds ratio. Figure created with BioRender.com.

Close modal

Importantly, the authors went on to explore the functional effects of the 25 rare and predicted deleterious NBN variants identified in their patient cohort (see figure). First, they examined NBN protein stability in HEK293T cells, by knocking out the endogenous NBN gene and then introducing each variant of interest into the cell line model. Three protein truncating variants, including p.K219fs, led to a complete loss of protein stability, as expected. Of 22 missense variants identified, 4 resulted in complete loss of NBN protein expression and 6 led to partial loss of protein stability, with the remaining 12 variants showing mild or no loss of stability. Second, they investigated the sensitivity of cells harboring NBN variants to the radiomimetic drug mitomycin C (MMC). MMC sensitivity is an established clinical assay for the diagnosis of NBS because of the hypersensitivity of patient cells to irradiation. All 7 of the variants that resulted in a loss of protein stability also demonstrated a high sensitivity to MMC and were therefore classified as “nonfunctional.” On the basis of the results of both functional assays, an additional 7 missense variants were classified as “partially functional,” with the remaining 11 variants designated as “functional.” Of note, 13 of the 14 nonfunctional or partially functional variants were located in the N-terminal forkhead-associated and BRCA1 C-terminal (FHA-BRCT)-repeat domain of NBN, suggesting that variation in this region has a significant impact on protein function.

In examining the clinical implications of NBN variants, Escherich et al found no associations with patient minimal residual disease, nor with overall or event-free survival. Hyperdiploidy was enriched among patients with B-cell ALL with nonfunctional NBN variants, similar to the molecular subtype patterns found in patients with rare germ line variants in IKZF1 or ETV6.2,3 And when considering only the 33 patients harboring nonfunctional or partially functional NBN variants, this corresponds to ≈0.8% of their patient cohort, a frequency similar to those reported for predisposition genes IKZF1, ETV6, and TP53 in patients with sporadic ALL.2-4 

Altogether, this study provides strong evidence for the role of heterozygous deleterious germ line NBN variants in the development of B-cell ALL. It is important to highlight that the analyses were limited to B-cell ALL, and the association of germ line NBN variants with risk of T-cell ALL and other hematological malignancies should be assessed. Furthermore, results from their functional assays in NBN–/– cells represent the effects of homozygous NBN variants, and additional studies are required to understand the effects of heterozygous variants and to decipher the precise mechanisms through which loss of NBN protein function may lead to ALL development. Finally, although a significant enrichment of NBN variants was found in patients with B-cell ALL compared with population-based controls using summary counts data, there are limitations to the approach used. For one, it does not account for variation in genetic ancestry that exists within population groups, which is of particular importance for admixed populations, such as African American/Black and Hispanic/Latino individuals. More accurate estimates of the effects of NBN variants on ALL risk would be obtained by sequencing large numbers of patients and controls from diverse populations, along with adjustment for genetic ancestry in association tests. Sequencing in larger sample sets may also reveal the impact of individual NBN variants on ALL risk, as these are likely to differ based on their functional effects (eg, nonfunctional vs partially functional variants).

Results to date suggest that each childhood ALL predisposition gene individually contributes a small piece of the susceptibility puzzle, and collectively account for only 5% to 10% of patients with sporadic ALL. There are likely additional predisposition genes waiting to be discovered, and investigation of germ line variants in the genes underlying other inherited chromosome instability disorders besides NBS is warranted.

Conflict-of-interest disclosure: A.J.d.S. declares no competing financial interests.

1.
Escherich
CS
,
Chen
W
,
Li
Y
, et al
.
Germ line genetic NBN variation and predisposition to B-cell acute lymphoblastic leukemia in children
.
Blood
.
2024
;
143
(
22
):
2270
-
2283
.
2.
Churchman
ML
,
Qian
M
,
Te Kronnie
G
, et al
.
Germline genetic IKZF1 variation and predisposition to childhood acute lymphoblastic leukemia
.
Cancer Cell
.
2018
;
33
(
5
):
937
-
948.e8
.
3.
Moriyama
T
,
Metzger
ML
,
Wu
G
, et al
.
Germline genetic variation in ETV6 and risk of childhood acute lymphoblastic leukaemia: a systematic genetic study
.
Lancet Oncol
.
2015
;
16
(
16
):
1659
-
1666
.
4.
Qian
M
,
Cao
X
,
Devidas
M
, et al
.
TP53 germline variations influence the predisposition and prognosis of B-cell acute lymphoblastic leukemia in children
.
J Clin Oncol
.
2018
;
36
(
6
):
591
-
599
.
5.
Shah
S
,
Schrader
KA
,
Waanders
E
, et al
.
A recurrent germline PAX5 mutation confers susceptibility to pre-B cell acute lymphoblastic leukemia
.
Nat Genet
.
2013
;
45
(
10
):
1226
-
1231
.
6.
Stieglitz
E
,
Loh
ML
.
Genetic predispositions to childhood leukemia
.
Ther Adv Hematol
.
2013
;
4
(
4
):
270
-
290
.
7.
Sharapova
SO
,
Pashchenko
OE
,
Bondarenko
AV
, et al
.
Geographical distribution, incidence, malignancies, and outcome of 136 Eastern Slavic patients with Nijmegen breakage syndrome and NBN founder variant c.657_661del5
.
Front Immunol
.
2020
;
11
:
602482
.
8.
Wolska-Kuśnierz
B
,
Gregorek
H
,
Chrzanowska
K
, et al
.
Nijmegen breakage syndrome: clinical and immunological features, long-term outcome and treatment options–a retrospective analysis
.
J Clin Immunol
.
2015
;
35
(
6
):
538
-
549
.
9.
Belhadj
S
,
Khurram
A
,
Bandlamudi
C
, et al
.
NBN pathogenic germline variants are associated with pan-cancer susceptibility and in vitro DNA damage response defects
.
Clin Cancer Res
.
2023
;
29
(
2
):
422
-
431
.
10.
Varon
R
,
Reis
A
,
Henze
G
,
von Einsiedel
HG
,
Sperling
K
,
Seeger
K
.
Mutations in the Nijmegen breakage syndrome gene (NBS1) in childhood acute lymphoblastic leukemia (ALL)
.
Cancer Res
.
2001
;
61
(
9
):
3570
-
3572
.
Sign in via your Institution