To the editor:

Hypodiploid acute lymphoblastic leukemia (ALL) with <45 chromosomes has been associated with a dismal prognosis.1-4  Near-haploid (24-31 chromosomes) and low-hypodiploid (32-39 chromosomes) ALL have particularly poor outcomes5,6  and are distinct entities.7  Near-haploid ALL is characterized by a younger age at diagnosis5  and genetic alterations targeting receptor tyrosine kinase signaling, Ras signaling, and the lymphoid transcription factor gene IKZF3.7  Low-hypodiploid patients are older5  and have genetic alterations of IKZF2, RB1, and TP53 that are often inherited.7 

Despite overall improved treatment outcome of childhood ALL,8  patients with hypodiploid ALL continued to fare poorly. In a Children’s Oncology Group study of 41 hypodiploid cases treated between 2002 and 2006, the 4-year overall survival was 54% ± 8%, and the study could not evaluate the efficacy of allogeneic transplantation.9  Because there are no known prognostic indicators in hypodiploid ALL, allogeneic transplantation continues to be recommended.10 

We recently showed that minimal residual disease (MRD) levels during remission induction treatment have important prognostic and therapeutic implications in the context of MRD-guided therapy of ALL.11  Notably, the adverse prognosis of pediatric Ph-like ALL can be significantly improved by this treatment approach.12  We therefore sought to determine if the MRD-guided treatment strategy, as applied in 2 consecutive clinical trials, could also improve the outcome of hypodiploid ALL. In parallel, we comprehensively examined genetic features and their prognostic importance.

From June 2000 to October 2007, 498 patients (1 to 18 years of age) with newly diagnosed ALL were consecutively enrolled in the St. Jude Total Therapy Study 15,13  and from October 2007 to June 2014, 410 patients (2 months to 18 years) in the Study 16.14  The protocols were approved by the institutional review boards and registered at www.clinicaltrials.gov as #NCT00137111 and #NCT00549848, respectively. Written informed consent was obtained from the parents or guardians, and assent from the patients, as appropriate. All hypodiploid patients received intensive chemotherapy including high-dose methotrexate, dexamethasone, vincristine, and asparaginase. Patients with MRD ≥1%, as determined by flow cytometry and/or polymerase chain reaction analysis15,16  after completion of induction therapy were offered the option of allogeneic transplantation.

Twenty (2.2%) patients had hypodiploid ALL. Eight patients had near-haploid ALL (median age 3.6 years, range 2.5-8.3), and 12 had low-hypodiploid ALL (median age 13.9 years, range 8.2-17.5 years, P < .01). There was no significant difference in presenting leukocyte count between near-haploid and low-hypodiploid ALL: median 8.1 × 109/L (range 3.0 to 52.0 × 109/L) vs median 5.6 × 109/L (range 1.5 to 36.9 × 109/L), P = .21.

All 20 patients achieved clinical remission. Negative MRD status (<0.01% leukemic cells among bone marrow mononucleated cells) was achieved in 6 of the 8 near-haploid and 8 of the 12 low-hypodiploid patients upon completion of remission induction (P = 1.0). Of the 6 patients with detectable MRD at the end of remission induction, 5 had MRD levels between 0.01% and 0.33%, and 1 had an MRD of 4.14%. Allogeneic transplantation was performed during initial remission in 1 MRD-negative patient with near haploidy because of the preference of the primary physician, and in the low-hypodiploid patient with MRD of 4.14% based on the protocol criterion.

The 5-year event-free survival was 73.6% (95% CI, 47.7-88.1) for all 20 patients (Figure 1) and was 68.2% (95% CI, 39.6-85.4) for the 16 treated in Study 15 as compared with 89.9% (86.5-92.5) for the other 409 B-cell ALL patients treated in Study 15 (P < .001). The result was not significantly different between the 8 patients with near-haploid and the 12 patients with low-hypodiploid ALL (72.9% [27.6-92.5] vs 75.0% [40.8-91.2], P = .80). However, it was significantly better for the 14 patients who achieved negative MRD status at the end of remission induction as compared with the 6 patients with detectable MRD: 85.1% (52.3-96.1) vs 44.4% (6.6-78.5), P = .03 (Figure 1). There were no other features associated with treatment outcome in this cohort. Twelve of the 13 hypodiploid patients with negative MRD status at the end of remission induction treated with chemotherapy only are alive in continuous complete remission for 0.7+ to 12.3+ years (median 7.0+ years); the 5-year event-free survival for this group was 91.7% (53.9-98.8). The near-haploid patient with negative MRD at the end of induction who was transplanted during initial remission died of transplant-related toxicity. Among the 6 patients with detectable MRD at the end of induction, the one with MRD of 4.14% remains in remission for 9.2+ years after transplantation, and 4 of the other 5 with lower levels of MRD (0.01% to 0.33%) treated with chemotherapy alone died of relapse with only 1 alive in remission for 1.4+ years.

Figure 1

Kaplan-Meier estimates of event-free survival for all 20 patients with hypodiploid ALL and according to the presence (n = 6) or absence (n = 14) of MRD.

Figure 1

Kaplan-Meier estimates of event-free survival for all 20 patients with hypodiploid ALL and according to the presence (n = 6) or absence (n = 14) of MRD.

Close modal

Sixteen hypodiploid patients had suitable tumor samples, and all patients had remission samples for genomic analysis and sequencing, performed as previously described (Table 1).17-21  The spectrum of genetic alterations was similar to those previously reported.7  Somatic Ras pathway alterations were identified in 7 of 8 near-haploid patients, with a higher proportion of loss-of-function alterations of NF1 than in our previous study (6 of 7 cases, with a KRAS mutation in 1 case). Four of 8 low-hypodiploid patients harbored TP53 mutations, with 1 identified in the germ line. We also observed alterations of IKZF2 in 2 low-hypodiploid patients and mutations of the histone acetyl-transferase genes CREBBP and EP300 in near-haploid ALL.

Table 1

Cytogenetic and molecular genetics of the 20 patients with near-haploid and low-hypodiploid ALL

PatientPCGP IDPloidy groupNext-generation sequencingKaryotypeGenomic lesion
SJHYPO122 Near haploid WES *48(24,X,+21)x2[20]/46,XY[1] NF1 Y2455*; RB1 L317fs; CDKN2A/B deletion 
SJBALL156 Near haploid WES *50(25,X,+14,+21)x2,del(1)(q42),add(11)(p15)[5]/46,XY[4] KRAS G12D and EP300 D1399N 
SJBALL014876 Near haploid WES *53(26,X,+10,+14,+21)x2,del(9)(p22),i(9)(q10),+mar[4]/53,idem,+9,-mar[16]/46,XX[1] CREBBP S1432fs, WHSC1 E1099K, USH2A I1326M, NF1 P678fs 
SJHYPO143 Near haploid WES 27,X,+X,+18,+21,+mar[6]/54,idemx2[3]/46,XX [15] No mutations in key pathways 
SJHYPO128 Near haploid WES 27,X,+X,+14,+21,+22[13]/54,idemx2,-X,+10,-14,+17,+18,-21,-22,+mar[7] NF1 T2600fs 
SJHYPO123 Near haploid, WGS, RNA-seq 27,X,+Y,+13,+18,+21[8]/35,idem,+1,+2,+3,+5,+6,+7,+11,+15[6]/46,XY[14] NF1 deletion of exons 33-41; histone cluster (6p22) deletion 
SJHYPO146 Near haploid WGS, WES 27,X,+Y,+17,+18,+21[5]/54,idemx2[6]/46,XY[8] NF1 T2133fs and deletion; CREBBP E1240K and deletion; PAX5 G25E 
SJHYPO124 Near haploid WES 30,X,+Y,+4,+8,+10,+14,+18,+21[4]/60,idemx2[14]/46,XY[2] NF1 E470* 
SJHYPO141 Low hypodiploid§ WES (germ line) 33,X,+1,+2,+6,+10,+11,+12,+18,+19,+21,+22[6]/46,XX [14] Not available for tumor 
10 SJHYPO22048 Low hypodiploid§ WES (germ line) 34,X,+Y,+1,+5,+6,+7,+10,+11,+15,+17,+19,+21[13]/62,idemx2,-X,-Y,-1,+3,-5,-6,-7,-10,-11,+14,-15,-17,+18,-19,+20,+22[4]/46,XY[3] Not available for tumor 
11 SJBALL014875 Low hypodiploid WES 35,X,+1,+5,+6,+9,+11,+12,+14,+18,+19,+20,+21,+22[11]/46,XY[9] MAPK mutation 
12 SJHYPO135 Low hypodiploid WES 35,X,+X,+1,+2,+4,+5,+6,+8,+11,+12,+16,+19,+21[10]/67,idemx2,-2,-5,-6,+10,+10,-11,-12,-12,+14,-16,-16,+18,-19,+20,+22[5]/46,XX[2] RB1 V654M; TP53 C283Y 
13 SJHYPO125 Low hypodiploid WES 35,X,+X,+1,der(1)t(1;7)(p36.1;q11.2)c,+5,+6,+8,+10,+11,+14,+18,+19,+21,+22[9]/67,idemx2,-5,-14,-19[9] No mutations in key pathways 
14 SJHYPO138 Low hypodiploid§ WES (germ line) 35,X,+Y,+1,+5,+6,+8,+9,+10,+11,+18,+19,+21,+22[20] Not available for tumor 
15 SJHYPO126 Low hypodiploid WES 35,X,+1,+5,+6,+8,+10,+11,+14,+15,+18,+19,+21,+22[8]/64,idemx2,-5,-10,-11,-14,-15,-19[9]/46,XX[3] IKZF2 exon 5 deletion; TP53 I162fs mutation (germ line) 
16 SJHYPO144 Low hypodiploid WES 36,X,+Y,+1,+5,+6,+8,+10,+11,+14,+15,+18,+19,+21,+22[16]/46,XY [4] TP53 L32_E4splice 
17 SJHYPO139 Low hypodiploid WES 36,X,+Y,+1,+2,+5,+6,+8,+9,+10,+11,+17,+19,+21,+22[5]/72,idemx2[2]/46,XY[13] TP53 R175H; IKZF2 deletion 
18 SJHYPO140 Low hypodiploid WES 37,X,+Y,+1,+5,+6,+8,+9,+10,+11,+14,+18,+19,+20,+21,+22[11]/74,idemx2[10]/46,XY[3] RB1 mutation 
19 SJBALL108 Low hypodiploid WES 38,X,+1,+2,+5,+6,+8,+9,del(9)(p13),ins(10;11)(q26;q21q24),+11,+14,+15,+16,+18,+19,+20,+21,+22[7]/38,idem,del(2)(p13p23)[4]/46,XX[9] PAX5, CDKN2A/B deletion 
20 SJHYPER22044 Low hypodiploid* NA 73(40,X,+Y,+1,+4,+5,+6,+8,+9,+10,+11,+13,+14,+15,+17,+18,+20,+21,+22)x2,-4,-5,-11,-13,-14,-15,-20[11]/46,XY[9] No focal deletions; LOH of chromosomes 2,3,4,7,11,12,13,15,16,17 
PatientPCGP IDPloidy groupNext-generation sequencingKaryotypeGenomic lesion
SJHYPO122 Near haploid WES *48(24,X,+21)x2[20]/46,XY[1] NF1 Y2455*; RB1 L317fs; CDKN2A/B deletion 
SJBALL156 Near haploid WES *50(25,X,+14,+21)x2,del(1)(q42),add(11)(p15)[5]/46,XY[4] KRAS G12D and EP300 D1399N 
SJBALL014876 Near haploid WES *53(26,X,+10,+14,+21)x2,del(9)(p22),i(9)(q10),+mar[4]/53,idem,+9,-mar[16]/46,XX[1] CREBBP S1432fs, WHSC1 E1099K, USH2A I1326M, NF1 P678fs 
SJHYPO143 Near haploid WES 27,X,+X,+18,+21,+mar[6]/54,idemx2[3]/46,XX [15] No mutations in key pathways 
SJHYPO128 Near haploid WES 27,X,+X,+14,+21,+22[13]/54,idemx2,-X,+10,-14,+17,+18,-21,-22,+mar[7] NF1 T2600fs 
SJHYPO123 Near haploid, WGS, RNA-seq 27,X,+Y,+13,+18,+21[8]/35,idem,+1,+2,+3,+5,+6,+7,+11,+15[6]/46,XY[14] NF1 deletion of exons 33-41; histone cluster (6p22) deletion 
SJHYPO146 Near haploid WGS, WES 27,X,+Y,+17,+18,+21[5]/54,idemx2[6]/46,XY[8] NF1 T2133fs and deletion; CREBBP E1240K and deletion; PAX5 G25E 
SJHYPO124 Near haploid WES 30,X,+Y,+4,+8,+10,+14,+18,+21[4]/60,idemx2[14]/46,XY[2] NF1 E470* 
SJHYPO141 Low hypodiploid§ WES (germ line) 33,X,+1,+2,+6,+10,+11,+12,+18,+19,+21,+22[6]/46,XX [14] Not available for tumor 
10 SJHYPO22048 Low hypodiploid§ WES (germ line) 34,X,+Y,+1,+5,+6,+7,+10,+11,+15,+17,+19,+21[13]/62,idemx2,-X,-Y,-1,+3,-5,-6,-7,-10,-11,+14,-15,-17,+18,-19,+20,+22[4]/46,XY[3] Not available for tumor 
11 SJBALL014875 Low hypodiploid WES 35,X,+1,+5,+6,+9,+11,+12,+14,+18,+19,+20,+21,+22[11]/46,XY[9] MAPK mutation 
12 SJHYPO135 Low hypodiploid WES 35,X,+X,+1,+2,+4,+5,+6,+8,+11,+12,+16,+19,+21[10]/67,idemx2,-2,-5,-6,+10,+10,-11,-12,-12,+14,-16,-16,+18,-19,+20,+22[5]/46,XX[2] RB1 V654M; TP53 C283Y 
13 SJHYPO125 Low hypodiploid WES 35,X,+X,+1,der(1)t(1;7)(p36.1;q11.2)c,+5,+6,+8,+10,+11,+14,+18,+19,+21,+22[9]/67,idemx2,-5,-14,-19[9] No mutations in key pathways 
14 SJHYPO138 Low hypodiploid§ WES (germ line) 35,X,+Y,+1,+5,+6,+8,+9,+10,+11,+18,+19,+21,+22[20] Not available for tumor 
15 SJHYPO126 Low hypodiploid WES 35,X,+1,+5,+6,+8,+10,+11,+14,+15,+18,+19,+21,+22[8]/64,idemx2,-5,-10,-11,-14,-15,-19[9]/46,XX[3] IKZF2 exon 5 deletion; TP53 I162fs mutation (germ line) 
16 SJHYPO144 Low hypodiploid WES 36,X,+Y,+1,+5,+6,+8,+10,+11,+14,+15,+18,+19,+21,+22[16]/46,XY [4] TP53 L32_E4splice 
17 SJHYPO139 Low hypodiploid WES 36,X,+Y,+1,+2,+5,+6,+8,+9,+10,+11,+17,+19,+21,+22[5]/72,idemx2[2]/46,XY[13] TP53 R175H; IKZF2 deletion 
18 SJHYPO140 Low hypodiploid WES 37,X,+Y,+1,+5,+6,+8,+9,+10,+11,+14,+18,+19,+20,+21,+22[11]/74,idemx2[10]/46,XY[3] RB1 mutation 
19 SJBALL108 Low hypodiploid WES 38,X,+1,+2,+5,+6,+8,+9,del(9)(p13),ins(10;11)(q26;q21q24),+11,+14,+15,+16,+18,+19,+20,+21,+22[7]/38,idem,del(2)(p13p23)[4]/46,XX[9] PAX5, CDKN2A/B deletion 
20 SJHYPER22044 Low hypodiploid* NA 73(40,X,+Y,+1,+4,+5,+6,+8,+9,+10,+11,+13,+14,+15,+17,+18,+20,+21,+22)x2,-4,-5,-11,-13,-14,-15,-20[11]/46,XY[9] No focal deletions; LOH of chromosomes 2,3,4,7,11,12,13,15,16,17 

LOH, loss of heterozygosity; NA, not available; PCGP, Pediatric Cancer Genome Project; WES, whole exome sequencing; WGS, whole genome sequencing.

*

Chromosome number reflected doubling of the hypodiploid clone.

Also Ph-like.

Genome sequencing previously reported.

§

No diagnostic material available for genomic analysis.

Five patients had hematologic relapse, including 2 with near haploidy (nos. 2 and 7) and 3 with low hypodiploidy (nos. 15, 18, and 19). Both near-haploid patients harbored Ras-activating mutations, 1 NF1 and 1 KRAS, and 1 also acquired IKZF1 and PAG1 deletions at relapse. Of the 3 low-hypodiploid patients that experienced relapse, 1 (no. 15) had alterations of IKZF2 and TP53, the second (no. 18) had a sequence mutation of RB1, and the third (no. 19) had deletions of PAX5 and CDKN2A/B. An additional 2 low-hypodiploid patients (nos. 9 and 16) had detectable MRD at the end of induction but did not experience relapse. Patient 16 had a somatic TP53 mutation, and patient 9 lacked tumor material for analysis but did not have a germ-line TP53 mutation. Three patients (nos. 6 and 7 with near haploidy and no. 18 with low hypodiploidy) also exhibited a Ph-like gene expression profile,21  2 of whom (nos. 7 and 18) relapsed. No associations between individual genetic alterations and outcome were observed.

In summary, our study demonstrates for the first time that MRD is the most important prognostic indicator for childhood hypodiploid ALL and that the outcome can be substantially improved by MRD-guided therapy. Importantly, hypodiploid patients with negative MRD status at the end of 6-week remission induction are highly curable with intensive chemotherapy alone. Studies are needed to determine if transplantation in first remission will improve the outcome of those with positive MRD at the end of remission induction. Genomic analysis confirmed the distinct genetic alterations characteristic of hypodiploid ALL7  but, because of the small number of cases, did not identify any genomic features associated with treatment response or outcome. However, 1 near-haploid patient acquired a deletion of PAG1 (also known as CBP, Csk-binding protein) at relapse. PAG1 encodes a putative modulator of Ras signaling, and PAG1 alterations were previously associated with treatment failure in hypodiploid ALL.7  Additional studies are needed to develop effective targeted therapy for hypodiplod ALL. In this regard, inhibitors of phosphatidylinositol-3-kinase signaling were identified as potentially effective drugs in our previous study7  and are being explored in preclinical studies as a new therapeutic strategy.

Acknowledgments: This work was supported in part by grants from the National Institutes of Health National Cancer Institute (CA21765, CA36401, CA176063, and U01 GM92666), the American Lebanese and Syrian Associated Charities, and the Leukemia and Lymphoma Society Special Fellowship and Alex’s Lemonade Stand Foundation Young Investigator Grant (K.G.R.). E.W. is a Koningin Wilhelmina Fonds fellow from the Dutch Cancer Society (KUN2012-5366), J.J.Y. is an American Society of Hematology Scholar, C.G.M. is a St. Baldrick’s Scholar, and C.-H.P. is an American Cancer Society professor.

Contribution: C.G.M. and C.-H.P. designed the study; S.J., H.I., W.H.L., W.P.B., and C.-H.P. participated in the clinical care of the patients; D.P.-T. performed statistical analysis; D.P.-T., E.C.-S., K.G.R., E.W., J.K.C., X.M., S.C.R., Y.F., G.S., J.J.Y., J.Z., and D.C. performed experiments and analyzed data; J.R.D., W.Y., M.V.R., W.E.E., and D.C. critically read the manuscript; and C.G.M. and C.-H.P. wrote the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Ching-Hon Pui, Department of Oncology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105; e-mail: ching-hon.pui@stjude.org; and Charles Mullighan, Department of Pathology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105; e-mail: charles.mullighan@stjude.org.

1
Pui
 
CH
Williams
 
DL
Raimondi
 
SC
, et al. 
Hypodiploidy is associated with a poor prognosis in childhood acute lymphoblastic leukemia.
Blood
1987
, vol. 
70
 
1
(pg. 
247
-
253
)
2
Pui
 
CH
Carroll
 
AJ
Raimondi
 
SC
, et al. 
Clinical presentation, karyotypic characterization, and treatment outcome of childhood acute lymphoblastic leukemia with a near-haploid or hypodiploid less than 45 line.
Blood
1990
, vol. 
75
 
5
(pg. 
1170
-
1177
)
3
Heerema
 
NA
Nachman
 
JB
Sather
 
HN
, et al. 
Hypodiploidy with less than 45 chromosomes confers adverse risk in childhood acute lymphoblastic leukemia: a report from the children’s cancer group.
Blood
1999
, vol. 
94
 
12
(pg. 
4036
-
4045
)
4
Raimondi
 
SC
Zhou
 
Y
Mathew
 
S
, et al. 
Reassessment of the prognostic significance of hypodiploidy in pediatric patients with acute lymphoblastic leukemia.
Cancer
2003
, vol. 
98
 
12
(pg. 
2715
-
2722
)
5
Harrison
 
CJ
Moorman
 
AV
Broadfield
 
ZJ
, et al. 
Childhood and Adult Leukaemia Working Parties
Three distinct subgroups of hypodiploidy in acute lymphoblastic leukaemia.
Br J Haematol
2004
, vol. 
125
 
5
(pg. 
552
-
559
)
6
Nachman
 
JB
Heerema
 
NA
Sather
 
H
, et al. 
Outcome of treatment in children with hypodiploid acute lymphoblastic leukemia.
Blood
2007
, vol. 
110
 
4
(pg. 
1112
-
1115
)
7
Holmfeldt
 
L
Wei
 
L
Diaz-Flores
 
E
, et al. 
The genomic landscape of hypodiploid acute lymphoblastic leukemia.
Nat Genet
2013
, vol. 
45
 
3
(pg. 
242
-
252
)
8
Pui
 
CH
Yang
 
JJ
Hunger
 
SP
, et al. 
Childhood acute lymphoblastic leukemia: progressive through collaboration.
J Clin Oncol
2015
, vol. 
33
 
27
(pg. 
2938
-
2948
)
9
Schultz
 
KR
Devidas
 
M
Bowman
 
WP
, et al. 
Children’s Oncology Group
Philadelphia chromosome-negative very high-risk acute lymphoblastic leukemia in children and adolescents: results from Children’s Oncology Group Study AALL0031.
Leukemia
2014
, vol. 
28
 
4
(pg. 
964
-
967
)
10
Oliansky
 
DM
Camitta
 
B
Gaynon
 
P
, et al. 
American Society for Blood and Marrow Transplantation
Role of cytotoxic therapy with hematopoietic stem cell transplantation in the treatment of pediatric acute lymphoblastic leukemia: update of the 2005 evidence-based review.
Biol Blood Marrow Transplant
2012
, vol. 
18
 
4
(pg. 
505
-
522
)
11
Pui
 
CH
Pei
 
D
Coustan-Smith
 
E
, et al. 
Clinical utility of sequential minimal residual disease measurements in the context of risk-based therapy in childhood acute lymphoblastic leukaemia: a prospective study.
Lancet Oncol
2015
, vol. 
16
 
4
(pg. 
465
-
474
)
12
Roberts
 
KG
Pei
 
D
Campana
 
D
, et al. 
Outcomes of children with BCR-ABL1–like acute lymphoblastic leukemia treated with risk-directed therapy based on the levels of minimal residual disease.
J Clin Oncol
2014
, vol. 
32
 
27
(pg. 
3012
-
3020
)
13
Pui
 
CH
Campana
 
D
Pei
 
D
, et al. 
Treating childhood acute lymphoblastic leukemia without cranial irradiation.
N Engl J Med
2009
, vol. 
360
 
26
(pg. 
2730
-
2741
)
14
Pui
 
CH
Campana
 
D
Sandlund
 
JT
, et al. 
Treatment of childhood acute lymphoblastic leukemia without cranial irradiation.
Ann Hematol
2011
, vol. 
90
 
suppl 1
(pg. 
S61
-
S63
)
15
Coustan-Smith
 
E
Sancho
 
J
Hancock
 
ML
, et al. 
Clinical importance of minimal residual disease in childhood acute lymphoblastic leukemia.
Blood
2000
, vol. 
96
 
8
(pg. 
2691
-
2696
)
16
Stow
 
P
Key
 
L
Chen
 
X
, et al. 
Clinical significance of low levels of minimal residual disease at the end of remission induction therapy in childhood acute lymphoblastic leukemia.
Blood
2010
, vol. 
115
 
23
(pg. 
4657
-
4663
)
17
Pounds
 
S
Cheng
 
C
Mullighan
 
C
Raimondi
 
SC
Shurtleff
 
S
Downing
 
JR
Reference alignment of SNP microarray signals for copy number analysis of tumors.
Bioinformatics
2009
, vol. 
25
 
3
(pg. 
315
-
321
)
18
Venkatraman
 
ES
Olshen
 
AB
A faster circular binary segmentation algorithm for the analysis of array CGH data.
Bioinformatics
2007
, vol. 
23
 
6
(pg. 
657
-
663
)
19
Mullighan
 
CG
Goorha
 
S
Radtke
 
I
, et al. 
Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia.
Nature
2007
, vol. 
446
 
7137
(pg. 
758
-
764
)
20
Mullighan
 
CG
Su
 
X
Zhang
 
J
, et al. 
Children’s Oncology Group
Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia.
N Engl J Med
2009
, vol. 
360
 
5
(pg. 
470
-
480
)
21
Roberts
 
KG
Li
 
Y
Payne-Turner
 
D
, et al. 
Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia.
N Engl J Med
2014
, vol. 
371
 
11
(pg. 
1005
-
1015
)
Sign in via your Institution