Improved personalized adjustment of primary therapy to the perceived risk of relapse by using new prognostic markers for treatment stratification may be beneficial to patients with acute lymphoblastic leukemia (ALL). Here, we review the advances that have shed light on the role of IKZF1 aberration as prognostic factor in pediatric ALL and summarize emerging concepts in this field. Continued research on the interplay of disease biology with exposure and response to treatment will be key to further improve treatment strategies.

Pediatric acute lymphoblastic leukemia (ALL), the most common malignancy observed in children and adolescents, is characterized by broad clinical and biological heterogeneity that is largely sustained by a diverse background of disease-initiating and -maintaining recurrent structural and/or numerical genetic aberrations acquired by the leukemic clone.1-5  Despite this heterogeneity, overall treatment results in pediatric ALL are one of the true success stories in clinical oncology with current cure rates exceeding 85%.1,4  On modern clinical protocols, this is achieved by the application of risk-adapted therapy, reflecting the probability of treatment failure. For this purpose, prognostic factors are used to estimate an individual patient’s risk of relapse and to adjust the required treatment intensity by patient stratification into different therapeutic risk groups, for example, standard, intermediate, high, and very high risk.1,4,5  Unfortunately, despite all of these efforts, a significant proportion of patients with ALL still experiences relapse. Thus, further improved personalized adjustment of primary therapy to the perceived risk of ALL relapse by using new prognostic markers for treatment stratification is likely to be beneficial to patients and, therefore, of great interest to those involved in the diagnosis and treatment of pediatric ALL and beyond.

Technical advances in the “omics” field fueled multiple comprehensive exploratory studies, and a number of candidate prognostic markers for ALL risk stratification have been identified and published during the last 10 years.1-5  Caregivers are thus faced with an ever-growing body of literature on new prognostic markers, unfortunately, mostly full of uncertainties about their clinical relevance. Therefore, it does not come as a surprise that the majority of newly described genetic or genomic markers are not regularly used to support decision-making procedures on current clinical protocols for ALL. General reasons for this lack of translation include (1) large heterogeneity across studies with differently sized, selected, and treated patient populations; (2) methodological differences in marker assessment and uncertainties regarding assay procedures; (3) differences in reported end points and conflicting outcomes; and (4) differences in the statistical analyses used, as well as other issues. One obvious difficulty in the practical implementation of new prognostic markers for ALL is the limited clinical significance conferred by many of the new high-risk markers, making it difficult to justify exposure to more intensive and toxic treatments for the potential benefit of a minority of patients among those identified by the marker.

The IKZF1 gene is located on chromosome band 7p12.2, consists of 8 exons, and codes for the transcription factor IKAROS with key regulatory functions in lymphopoiesis.6,7  IKAROS harbors 6 zinc fingers. Four of these are located in the DNA-binding domain encoded by exons 4 to 6 and are essential to maintain IKAROS’ tumor-suppressor function. The remaining 2 zinc fingers are encoded by exon 8 and mediate the dimerization of IKAROS either as a homodimer or with other transcription factors of its family (eg, AIOLOS and HELIOS).6-8 IKZF1 has remained in the spotlight of the leukemia field since 2008 when Mullighan et al first described that recurrent, mostly monoallelic, focal deletions affect the coding regions of IKZF1.9,10  These deletions involve either the whole gene or only parts of IKZF1 and are observed at an overall frequency of ∼15% in pediatric and 40% in adult ALL cases.2,11,12  Although some deletions (eg, those affecting the whole gene, intragenic deletions including exons 2 and/or 8, deletions of exon 1 and 5′ untranscribed regulatory regions) result in haploinsufficiency, other deletions, most commonly those affecting exons 4 to 7, lead to loss of the DNA-binding domain and generation of dominant-negative isoforms.2,9-12  The latter compromise the tumor-suppressor function of IKAROS translated from the remaining wild-type allele. The frequency of somatic point mutations in IKZF1 has been studied far less extensively. They are present to a much lower extent (Figure 1) and, as with deletions, their molecular consequence can be either haploinsufficiency (eg, truncating variants) or a dominant-negative effect (Figure 1).10,13-19  Recently, a new B-cell precursor (BCP) ALL subgroup characterized by the IKZF1 missense mutation p.Asn159Tyr (N159Y) affecting the DNA-binding domain was identified through a distinct gene-expression profile.17,18  Of interest, the specific gene-expression profile also differed from those BCP ALL cases with other known IKZF1 alterations. In addition, an increasing number of cases with fusion transcripts involving IKZF1 have been described (Figure 1).17-26  Besides the somatic alterations described earlier in this section, frequent intronic germline variants in IKZF1 (rs11978267 and rs4132601) have been identified in genome-wide association studies and consistently described to modestly modulate the risk of pediatric BCP ALL (Figure 1).27,28  Of major importance, Churchman et al recently characterized IKZF1 as a leukemia predisposition gene by reporting mostly adverse germline IKZF1 variation in familial pediatric ALL and 43 of 4963 (0.9%) unselected BCP ALL patients29  (Figure 1).

Figure 1.

Genetic alterations of the IKZF1 gene at chromosome band 7p12.2 in pediatric ALL. Red boxes indicate the observed approximate frequencies of the different types of genetic aberrations: deletions (bottom left), gene fusions (bottom right), and somatic as well as germline single-nucleotide variants (top). The 2 intronic germline risk variants identified in genome-wide association studies are indicated by their Reference SNP cluster ID. Frequencies (percentages) of the most common specific deletions (DEL) within the group of IKZF1-deleted ALL are indicated in black. Chr, chromosome; UTR, untranslated region.

Figure 1.

Genetic alterations of the IKZF1 gene at chromosome band 7p12.2 in pediatric ALL. Red boxes indicate the observed approximate frequencies of the different types of genetic aberrations: deletions (bottom left), gene fusions (bottom right), and somatic as well as germline single-nucleotide variants (top). The 2 intronic germline risk variants identified in genome-wide association studies are indicated by their Reference SNP cluster ID. Frequencies (percentages) of the most common specific deletions (DEL) within the group of IKZF1-deleted ALL are indicated in black. Chr, chromosome; UTR, untranslated region.

Close modal

Most of the published data on IKZF1 deletions in pediatric ALL have been generated by multiplex ligation probe-dependent amplification (MLPA) analysis (Table 1).13,30-44  Alternative techniques include polymerase chain reaction (PCR) or array techniques.10,14,15,30,39,40,45,46  MLPA detects virtually all deletions targeting the coding sequence, and most large studies evaluating the prognostic effect of IKZF1 deletion in patient cohorts relied on this technique (Table 1). However, like array techniques, it cannot reliably detect deletions present in <25% of cells. Thus, MLPA may fail to detect IKZF1 deletions in samples showing a low leukemia burden or deletions that are limited to a leukemic subclone only. For recurrent intragenic deletions, this limitation can be overcome by quantitative PCR assays. This technique allows detection of intragenic IKZF1 deletions with a higher sensitivity than classical techniques and, furthermore, allows minimal residual disease (MRD) monitoring.47  However, it has to be emphasized that the prognostic impact of subclonal IKZF1 deletions has not been specifically evaluated yet and may differ from that of full clone deletions. Another debated issue relates to isolated IKZF1 exon 1 deletions and whether they should be considered in routine diagnosis or not. Their accurate detection can be complicated due to false MLPA positivity favored by a high guanine-cytosine content in this region. Molecular mapping of exon 1 deletions together with an adapted MLPA protocol with a reinforced denaturation step recently confirmed the frequency and impact of exon 1 deletions on IKZF1 transcription48  and suggest that these deletions should be considered for diagnosis when properly controlled. It is noteworthy that additional IKZF1 deletions targeting the 5′ regulatory regions have been described and are not detected by current MLPA protocols.9,48 IKZF1 sequence mutations were mainly analyzed by Sanger sequencing and targeted or nontargeted next-generation sequencing strategies whereas the majority of fusion genes have been identified in transcriptomic studies.10,13-26,29  The different qualities and sensitivities of these techniques need to be considered when comparing frequency and type of IKZF1 aberrations in the literature.

Table 1.

Summary of studies on the prognostic impact of IKZF1 aberration in treatment trials of pediatric ALL

TrialRef.CountryTrial periodNo. of patientsType of ALLFrequency of aberrant IKZF1 (method of detection)EFS (DFS)OSCumulative incidence of relapseComments
COG P9906, St. Jude Total XI, XII, XIII, XIV, XV, and Interfant-99 10  US COG P9906: 2000-2003 COG P9906: 221 High-risk BCP 28.6% (SNP array, Sanger sequencing) 25% vs 73% (at 5 y), P < .0001  73% vs 25% (at 5 y), P < .0001 Groundbreaking first study demonstrating prognostic impact of IKZF1 deletion; includes 5 patients with IKZF1 point mutation in the COG cohort and 21 BCR-ABL1–rearranged cases in the St. Jude cohort 
St. Jude and Interfant: 1986-2007 St. Jude and Interfant: 258 BCP 18.6% (SNP array) 40% vs 72% (at 10 y), P < .001 48% vs 26% (at 10 y), P = .004 
DCOG ALL9 13, 48  The Netherlands 1997-2000 131 BCP 13.0% (MLPA, Sanger sequencing) 39% vs 89% (at 8 y), P < .001 56.0% vs 91.0% (at 8 y), P < .001  Exemplifies the importance of IKZF1 deletion as a prognostic factor, especially in nonhigh-risk patients; included 2 BCR-ABL1–rearranged cases; subsequent study in related population demonstrated the prognostic power of integrated use of aberrant IKZF1 and MRD levels 
TPOG-ALL-93, TPOG-97-VHR, TPOG-ALL-2002 14  Taiwan 1995-2009 242 BCP 10.7% (PCR) 15% vs 76% (at 10 y), P < .0001 38% vs 78% (at 10 y), P = .0016  First study indicating poor prognostic effect of IKZF1 deletion in an Asian population 
DCOG ALL8, ALL9, ALL10, UKALL 97, 97/99, 2003 30  The Netherlands, UK 1997-2006 DCOG: 34 Down syndrome ALL DCOG: 35.3% (CGH array, MLPA, Sanger sequencing) DCOG: 45% vs 95% (at 6 y), P = .002 DCOG: 66% vs 95% (at 6 y), P = .02 DCOG: 37% vs 5% (at 6 y), P = .044 Strong prognostic effect of IKZF1 deletion in Down syndrome ALL 
UKALL: 85 UKALL: 27.1% (MLPA) UKALL: 21% vs 58% (at 6 y), P = .002 UKALL: 15% vs 71% (at 6 y), P = .002 UKALL: 37% vs 18% (at 6 y), P = .06 
Japan Childhood Leukemia Study ALL02 31  Japan 2002-2008 202 BCP 9.4% (MLPA) 63% vs 89% (at 5 y),P = ..001 72% vs 90% (at 5 y),P = ..02  Particularly strong prognostic effect of IKZF1 deletion in the NCI high-risk group 
JCCLSG ALL 2004 32  Japan 2004-2008 177 BCP 12.0% (MLPA) 68% vs 85% (at 4 y), P = .04   Prognostic effect of IKZF1 deletion more pronounced in high-risk patients 
ALL-REZ BFM 2002 33  Germany, Austria, Switzerland 2002-2009 204 BCP relapse 33.3% (MLPA) 30% vs 51% (at 5 y), P = .002 36% vs 60% (at 5 y), P = .001 41% vs 23% (at 5 y), P = .006 Prognostic impact of IKZF1 deletion also in second-line treatment of relapsed ALL; one-quarter of IKZF1 deletions was acquired at relapse 
AIEOP-BFM ALL 2000 34  Germany 1999-2005 694 BCP and T ALL 12.0% (MLPA) 69% vs 85% (at 5 y), P < .0001 82% vs 92% (at 5 y), P = .003 21% vs 10% (at 5 y), P = .001 Pronounced prognostic effect of IKZF1 deletion in the intermediate-risk group 
AIEOP-BFM ALL 2000 35  Italy 2003-2005 410 BCP 13.2% (MLPA) 70% vs 85% (at 5 y), P = .007 87% vs 93% (at 5 y), P = .100 24% vs 13% (at 5 y), P = .049 Due to the modest effect of IKZF1 deletion observed, the relevance of IKZF1 deletion as a clinically useful stratification factor is debated 
DCOG ALL8, ALL9, ALL10, COALL-97, COALL-03 36  The Netherlands, Germany 1991-2012 857 BCP 15.9% (MLPA)   34% vs 13% (at 5 y), P = <.001 Confirms and extends the strong prognostic effect of IKZF1 deletion; IKZF1 deletion remained predictive in intermediate risk patients on the MRD-guided DCOG ALL10 trial 
EsPhALL and I-BFM SG studies (for pre-TKI assessment) 37  Austria, Czech Republic, France, Germany, The Netherlands, UK 1995-2005 Pre-TKI cohort: 84 EsPh-ALL cohort: 107 BCR-ABL1–rearranged BCP 66.0% (MLPA, CGH array, SNP array) DFS pre-TKI: 30% vs 58% (at 4 y), P = .013 Pre-TKI: 49% vs 75% (at 4 y), P = .075 Pre-TKI: 57% vs 21% (at 4 y), P = .026 IKZF1 deletion is demonstrated to be a poor prognostic marker in BCR-ABL1–rearranged pediatric ALL independent of imatinib treatment 
DFS EsPhALL: 54% vs 63% (at 4 y), P = .168 EsPhALL: 58% vs 83% (at 4 y), P = .070 EsPhALL: 28% vs 31% (at 4 y), P = .817 
DFS EsPhALL TKI-treated good-risk patients: 56% vs 75% (at 4 y), P = .051 EsPhALL TKI-treated good-risk patients: 66% vs 100% (at 4 y), P = .039 EsPhALL TKI-treated good-risk patients: 29% vs 25% (at 4 y), P = .249 
UKALL trials ALL 97/99 and ALL 2003 38  UK, Ireland UKALL 97/99: 1997-2002 UKALL 97/99: 864 BCP 13.4% (MLPA) 56% vs 80% (at 5 y), P = <.001 70% vs 89% (at 5 y), P = <.001 40% vs 18% (at 5 y), P = <.001 Integration of cytogenetic with genomic data including IKZF1 deletion refines risk groups in a clinically meaningful way 
UKALL 2003: 2003-2011 UKALL 2003: 782 BCP 11.1% (MLPA) 80% vs 92% (at 5 y), P = <.001 86% vs 95% (at 5 y), P = <.001 16% vs 6% (at 5 y), P = .002 
NOPHO ALL-1992, NOPHO ALL-2000, NOPHO ALL-2008 39  Denmark, Finland, Norway, Sweden 1992-2013 334 BCP 15.0% (SNP array, MLPA) 60% vs 83% (at 10 y), P < .001 73% vs 89% (at 10 y), P = .001 35% vs 12% (at 10 y), P < .001 Prognostic impact independent of WBC count and MRD; co-occurrence of PAR1 deletion increased prognostic impact of IKZF1 deletion 
EORTC Children’s Leukemia Group study 58951 40  Belgium, France, Portugal 1998-2008 1223 BCP 14.6% (PCR, MLPA) 68% vs 87% (at 5 y), P = <.001 87% vs 92% (at 5 y), P = .035  IKZF1-aberrant BCP ALL benefited from vincristine-steroid pulses during maintenance treatment 
ALLR3 41  UK, Ireland, The Netherlands, Australia, New Zealand 2002-2013 222 BCP relapse 23.0% (MLPA)  48% vs 53% (at 5 y), P = .30  No prognostic impact of IKZF1 deletion at relapse 
ANZCHOG ALL8 42  Australia, New Zealand 2002-2011 475 BCP standard and intermediate risk 10.5% (MLPA) 53% vs 83% (at 7 y), P < .0001 78% vs 94% (at 7 y), P < .0001 41% vs 15% (at 7 y), P < .0001 Strong prognostic effect of IKZF1 deletion in non–high-risk BCP ALL 
ALL-BFM 95 45  Germany 1995-2000 655 BCP intermediate risk 12.2% (PCR) 66% vs 82% (at 5 y), P = .001   Vincristine-dexamethasone pulses during maintenance were not of benefit to IKZF1-deleted intermediate-risk patients 
DFCI ALL Consortium Protocol 05-001 43  US 2005-2010 385 BCP 16.0% (MLPA) 63% vs 88% (at 5 y), P < .001 79% vs 94% (at 5 y), P < .001 29% vs 8% (at 5 y), P < .001 IKZF1 deletion was confirmed as an independent predictor of inferior outcome 
Malaysia-Singapore ALL 2003, 2010 44  Malaysia, Singapore 2002-2017 665 BCP 15.9% (MLPA)   Without IKZF1 as high-risk criterion: 30% vs 8% (at 5 y), P < .001 Demonstrates that intensifying treatment of IKZF1-deleted BCP ALL patients reduces risk of relapse 
With IKZF1 as high-risk criterion: 14% vs 5% (at 5 y), P = .030 
COG Trial AALL0622 46  US, Canada 2008-2012 44 BCR-ABL1–rearranged BCP 56.8% (SNP array) 52% vs 82% (at 5 y), P = .040 80% vs 100% (at 5 y), P = .040  Confirms poor outcome for IKZF1-deleted BCR-ABL1–rearranged ALL on dasatinib-containing treatment regimen 
AIEOP-BFM ALL 2000 61  Germany, Italy 1999-2009 1408 BCP 14.6% (MLPA) IKZF1-deleted, but not IKZF1plus: 77% vs 86% (at 5 y), P = .0005 IKZF1-deleted, but not IKZF1plus: 89% vs 94% (at 5 y), P = .023 IKZF1-deleted, but not IKZF1plus: 16% vs 11% (at 5 y), P = .045 Definition of a poor MRD-dependent prognostic pattern termed IKZF1plus with IKZF1 deletions co-occurring with deletions in CDKN2A, CDKN2B, PAX5, or PAR1 in the absence of ERG deletion 
IKZF1plus: 51% vs 86% (at 5 y), P < .0001 IKZF1plus: 75% vs 94% (at 5 y), P < .0001 IKZF1plus: 44% vs 11% (at 5 y), P < .0001 
TrialRef.CountryTrial periodNo. of patientsType of ALLFrequency of aberrant IKZF1 (method of detection)EFS (DFS)OSCumulative incidence of relapseComments
COG P9906, St. Jude Total XI, XII, XIII, XIV, XV, and Interfant-99 10  US COG P9906: 2000-2003 COG P9906: 221 High-risk BCP 28.6% (SNP array, Sanger sequencing) 25% vs 73% (at 5 y), P < .0001  73% vs 25% (at 5 y), P < .0001 Groundbreaking first study demonstrating prognostic impact of IKZF1 deletion; includes 5 patients with IKZF1 point mutation in the COG cohort and 21 BCR-ABL1–rearranged cases in the St. Jude cohort 
St. Jude and Interfant: 1986-2007 St. Jude and Interfant: 258 BCP 18.6% (SNP array) 40% vs 72% (at 10 y), P < .001 48% vs 26% (at 10 y), P = .004 
DCOG ALL9 13, 48  The Netherlands 1997-2000 131 BCP 13.0% (MLPA, Sanger sequencing) 39% vs 89% (at 8 y), P < .001 56.0% vs 91.0% (at 8 y), P < .001  Exemplifies the importance of IKZF1 deletion as a prognostic factor, especially in nonhigh-risk patients; included 2 BCR-ABL1–rearranged cases; subsequent study in related population demonstrated the prognostic power of integrated use of aberrant IKZF1 and MRD levels 
TPOG-ALL-93, TPOG-97-VHR, TPOG-ALL-2002 14  Taiwan 1995-2009 242 BCP 10.7% (PCR) 15% vs 76% (at 10 y), P < .0001 38% vs 78% (at 10 y), P = .0016  First study indicating poor prognostic effect of IKZF1 deletion in an Asian population 
DCOG ALL8, ALL9, ALL10, UKALL 97, 97/99, 2003 30  The Netherlands, UK 1997-2006 DCOG: 34 Down syndrome ALL DCOG: 35.3% (CGH array, MLPA, Sanger sequencing) DCOG: 45% vs 95% (at 6 y), P = .002 DCOG: 66% vs 95% (at 6 y), P = .02 DCOG: 37% vs 5% (at 6 y), P = .044 Strong prognostic effect of IKZF1 deletion in Down syndrome ALL 
UKALL: 85 UKALL: 27.1% (MLPA) UKALL: 21% vs 58% (at 6 y), P = .002 UKALL: 15% vs 71% (at 6 y), P = .002 UKALL: 37% vs 18% (at 6 y), P = .06 
Japan Childhood Leukemia Study ALL02 31  Japan 2002-2008 202 BCP 9.4% (MLPA) 63% vs 89% (at 5 y),P = ..001 72% vs 90% (at 5 y),P = ..02  Particularly strong prognostic effect of IKZF1 deletion in the NCI high-risk group 
JCCLSG ALL 2004 32  Japan 2004-2008 177 BCP 12.0% (MLPA) 68% vs 85% (at 4 y), P = .04   Prognostic effect of IKZF1 deletion more pronounced in high-risk patients 
ALL-REZ BFM 2002 33  Germany, Austria, Switzerland 2002-2009 204 BCP relapse 33.3% (MLPA) 30% vs 51% (at 5 y), P = .002 36% vs 60% (at 5 y), P = .001 41% vs 23% (at 5 y), P = .006 Prognostic impact of IKZF1 deletion also in second-line treatment of relapsed ALL; one-quarter of IKZF1 deletions was acquired at relapse 
AIEOP-BFM ALL 2000 34  Germany 1999-2005 694 BCP and T ALL 12.0% (MLPA) 69% vs 85% (at 5 y), P < .0001 82% vs 92% (at 5 y), P = .003 21% vs 10% (at 5 y), P = .001 Pronounced prognostic effect of IKZF1 deletion in the intermediate-risk group 
AIEOP-BFM ALL 2000 35  Italy 2003-2005 410 BCP 13.2% (MLPA) 70% vs 85% (at 5 y), P = .007 87% vs 93% (at 5 y), P = .100 24% vs 13% (at 5 y), P = .049 Due to the modest effect of IKZF1 deletion observed, the relevance of IKZF1 deletion as a clinically useful stratification factor is debated 
DCOG ALL8, ALL9, ALL10, COALL-97, COALL-03 36  The Netherlands, Germany 1991-2012 857 BCP 15.9% (MLPA)   34% vs 13% (at 5 y), P = <.001 Confirms and extends the strong prognostic effect of IKZF1 deletion; IKZF1 deletion remained predictive in intermediate risk patients on the MRD-guided DCOG ALL10 trial 
EsPhALL and I-BFM SG studies (for pre-TKI assessment) 37  Austria, Czech Republic, France, Germany, The Netherlands, UK 1995-2005 Pre-TKI cohort: 84 EsPh-ALL cohort: 107 BCR-ABL1–rearranged BCP 66.0% (MLPA, CGH array, SNP array) DFS pre-TKI: 30% vs 58% (at 4 y), P = .013 Pre-TKI: 49% vs 75% (at 4 y), P = .075 Pre-TKI: 57% vs 21% (at 4 y), P = .026 IKZF1 deletion is demonstrated to be a poor prognostic marker in BCR-ABL1–rearranged pediatric ALL independent of imatinib treatment 
DFS EsPhALL: 54% vs 63% (at 4 y), P = .168 EsPhALL: 58% vs 83% (at 4 y), P = .070 EsPhALL: 28% vs 31% (at 4 y), P = .817 
DFS EsPhALL TKI-treated good-risk patients: 56% vs 75% (at 4 y), P = .051 EsPhALL TKI-treated good-risk patients: 66% vs 100% (at 4 y), P = .039 EsPhALL TKI-treated good-risk patients: 29% vs 25% (at 4 y), P = .249 
UKALL trials ALL 97/99 and ALL 2003 38  UK, Ireland UKALL 97/99: 1997-2002 UKALL 97/99: 864 BCP 13.4% (MLPA) 56% vs 80% (at 5 y), P = <.001 70% vs 89% (at 5 y), P = <.001 40% vs 18% (at 5 y), P = <.001 Integration of cytogenetic with genomic data including IKZF1 deletion refines risk groups in a clinically meaningful way 
UKALL 2003: 2003-2011 UKALL 2003: 782 BCP 11.1% (MLPA) 80% vs 92% (at 5 y), P = <.001 86% vs 95% (at 5 y), P = <.001 16% vs 6% (at 5 y), P = .002 
NOPHO ALL-1992, NOPHO ALL-2000, NOPHO ALL-2008 39  Denmark, Finland, Norway, Sweden 1992-2013 334 BCP 15.0% (SNP array, MLPA) 60% vs 83% (at 10 y), P < .001 73% vs 89% (at 10 y), P = .001 35% vs 12% (at 10 y), P < .001 Prognostic impact independent of WBC count and MRD; co-occurrence of PAR1 deletion increased prognostic impact of IKZF1 deletion 
EORTC Children’s Leukemia Group study 58951 40  Belgium, France, Portugal 1998-2008 1223 BCP 14.6% (PCR, MLPA) 68% vs 87% (at 5 y), P = <.001 87% vs 92% (at 5 y), P = .035  IKZF1-aberrant BCP ALL benefited from vincristine-steroid pulses during maintenance treatment 
ALLR3 41  UK, Ireland, The Netherlands, Australia, New Zealand 2002-2013 222 BCP relapse 23.0% (MLPA)  48% vs 53% (at 5 y), P = .30  No prognostic impact of IKZF1 deletion at relapse 
ANZCHOG ALL8 42  Australia, New Zealand 2002-2011 475 BCP standard and intermediate risk 10.5% (MLPA) 53% vs 83% (at 7 y), P < .0001 78% vs 94% (at 7 y), P < .0001 41% vs 15% (at 7 y), P < .0001 Strong prognostic effect of IKZF1 deletion in non–high-risk BCP ALL 
ALL-BFM 95 45  Germany 1995-2000 655 BCP intermediate risk 12.2% (PCR) 66% vs 82% (at 5 y), P = .001   Vincristine-dexamethasone pulses during maintenance were not of benefit to IKZF1-deleted intermediate-risk patients 
DFCI ALL Consortium Protocol 05-001 43  US 2005-2010 385 BCP 16.0% (MLPA) 63% vs 88% (at 5 y), P < .001 79% vs 94% (at 5 y), P < .001 29% vs 8% (at 5 y), P < .001 IKZF1 deletion was confirmed as an independent predictor of inferior outcome 
Malaysia-Singapore ALL 2003, 2010 44  Malaysia, Singapore 2002-2017 665 BCP 15.9% (MLPA)   Without IKZF1 as high-risk criterion: 30% vs 8% (at 5 y), P < .001 Demonstrates that intensifying treatment of IKZF1-deleted BCP ALL patients reduces risk of relapse 
With IKZF1 as high-risk criterion: 14% vs 5% (at 5 y), P = .030 
COG Trial AALL0622 46  US, Canada 2008-2012 44 BCR-ABL1–rearranged BCP 56.8% (SNP array) 52% vs 82% (at 5 y), P = .040 80% vs 100% (at 5 y), P = .040  Confirms poor outcome for IKZF1-deleted BCR-ABL1–rearranged ALL on dasatinib-containing treatment regimen 
AIEOP-BFM ALL 2000 61  Germany, Italy 1999-2009 1408 BCP 14.6% (MLPA) IKZF1-deleted, but not IKZF1plus: 77% vs 86% (at 5 y), P = .0005 IKZF1-deleted, but not IKZF1plus: 89% vs 94% (at 5 y), P = .023 IKZF1-deleted, but not IKZF1plus: 16% vs 11% (at 5 y), P = .045 Definition of a poor MRD-dependent prognostic pattern termed IKZF1plus with IKZF1 deletions co-occurring with deletions in CDKN2A, CDKN2B, PAX5, or PAR1 in the absence of ERG deletion 
IKZF1plus: 51% vs 86% (at 5 y), P < .0001 IKZF1plus: 75% vs 94% (at 5 y), P < .0001 IKZF1plus: 44% vs 11% (at 5 y), P < .0001 

AIEOP, Associazione Italiana di Ematologia e Oncologia Pediatrica; ALL-REZ, Acute Lymphoblastic Leukemia-Relapse Study; BFM, Berlin-Frankfurt-Münster; ALLR3, An International Collaborative Trial for Relapsed and Refractory Acute Lymphoblastic Leukaemia Combination Chemotherapy in Treating Young Patients With Relapsed or Refractory Acute Lymphoblastic Leukemia; ANZCHOG, Australian & New Zealand Children’s Haematology/Oncology Group; CGH, comparative genomic hybridization; COG, Children’s Oncology Group; DCOG, Dutch Childhood Oncology Group; DFCI, Dana-Farber Cancer Institute; DFS, disease-free survival; EFS, event-free survival; EORTC, European Organisation for Research and Treatment of Cancer; EsPHALL, Safety and Efficacy of Imatinib Added to Chemotherapy in Treatment of Ph+ Acute Lymphoblastic Leukemia in Children; I-BFM SG, International Berlin-Frankfurt-Münster Study Group; JCCLSG, Japanese Children's Cancer and Leukemia Study Group; MLPA, multiplex ligation probe-dependent amplification; MRD, minimal residual disease; NCI, National Cancer Institute; NOPHO, Nordic Society of Pediatric Hematology and Oncology; OS, overall survival; PCR, polymerase chain reaction; Ref., reference(s); Sanger sequencing, dye-terminator sequencing; SNP, single-nucleotide polymorphism; TKI, tyrosine kinase inhibitor; TPOG, Taiwan Pediatric Oncology Group; UK, United Kingdom; US, United States; VHR, very high risk; WBC, white blood cell.

During the last 10 years, various studies have analyzed the clinical importance of IKZF1 aberration in pediatric ALL treated on different treatment protocols.10,13-16,30-46  The presence of IKZF1 deletions has been associated with older age at diagnosis, higher presenting white blood cell counts, and higher levels of MRD after induction and consolidation.10,31,32,34,35,43,44,49,50  Consequently, IKZF1 deletions are overrepresented in high-risk patients with pediatric ALL. Importantly, the distribution of IKZF1 aberrations within the subgroups of ALL is not homogeneous. Although approximately two-thirds of BCR-ABL1+ pediatric BCP ALL carry an IKZF1 deletion,9,37,44,46  the frequency in BCR-ABL1 BCP ALL subgroups is lower. It ranges from roughly 20% in the B-other group, which lacks the “classical” recurrent acquired genetic aberrations of ALL, but includes Philadelphia-like and BCR-ABL1–like BCP ALL characterized by an activated kinase signature, and 15% in high hyperdiploid ALL to <5% in ETV6-RUNX1-, TCF3- or KMT2A-rearranged ALL as well as T-cell ALL.10,31,34,35,38-40,44,51-53  Although very rare in BCR-ABL1 BCP ALL (∼1%), IKZF1 point mutations have been described in up to 10% of IKZF1 deletion-negative BCR-ABL1+ BCP ALL.10,13-16  One unifying and important feature of a majority of prognostic studies in pediatric BCP ALL is that the different types of IKZF1 deletions have been consistently linked to an unfavorable clinical outcome of frontline treatment (Table 1).10,13,14,30-46,54  It is worth mentioning that IKZF1 point mutations seem to have a similar impact on outcome.10,13-16  As a consequence, some international study groups on treatment of ALL early on included IKZF1 deletion status into their high-risk treatment-stratification strategies for BCP ALL patients whereas others did not follow this strategy because the prognostic effect of IKZF1 deletions alone was considered not sufficiently strong enough to justify exposure to the toxic side effects of high-risk or even very-high-risk therapy.38,53 

Of significance, although it has been clearly demonstrated that IKZF1 aberrations exert an independent prognostic impact, some studies early on suggested the presence of effect modification or confounding by different levels of MRD, activated JAK-STAT signaling, or co-occurring deletions of the lymphoid transcriptional regulator gene BTG1 or the ets family transcription factor ERG.15,32,34,35,49,55-57  For example, integrated use of MRD and IKZF1 deletion status predicted 79% of relapses in a Dutch study.49  Such an epistatic effect of other genetic aberrations has been particularly documented for ERG deletions because IKZF1 deletion surprisingly does not affect the prognosis of BCP ALL when co-ocurring with ERG deletion.56,57  It is now known that ERG deletions almost exclusively occur in the recently described DUX4-rearranged BCP ALL subtype, which also bears IKZF1 deletions in >20% of the cases.19,58,59  In this respect, ERG deletion can be considered a surrogate marker for DUX4 rearrangements, although approximately one-third of DUX4-rearranged BCP ALL lacks an ERG deletion.60  So far, it is unclear whether the previously described prognostic effect of the ERG deletion56,57  is mediated by itself or the association with the DUX4 rearrangement. Of interest, there are hints that positivity for an ERG deletion also seems to confer an advantageous outcome within DUX4-rearranged BCP ALL.60  Hence, the interrelationships of ERG deletion and DUX4 rearrangements with IKZF1 deletions and their impact on outcome are complex and not fully understood. Recently, a large study of the International Berlin-Frankfurt-Münster (I-BFM) Study Group refined the prognostic strength of IKZF1 deletions in BCR-ABL1 pediatric BCP ALL by describing an extremely poor prognostic IKZF1 deletion-associated genetic aberration profile termed IKZF1plus.61  It is defined by IKZF1 deletions co-occurring with deletions in CDKN2A, CDKN2B, PAX5, or PAR1 in the absence of ERG deletion. Of interest, but unfortunately not yet understood, IKZF1plus exerts its strong prognostic impact only in patients still carrying measurable MRD of a leukemic cell load exceeding 10−4 after induction treatment. This simple to assess profile in combination with MRD analyses has already been implemented as a high-risk stratification criterion in the current frontline AIEOP-BFM ALL 2017 trial for treatment of ALL, and its prognostic value has been confirmed by others.62 

Lastly, the prognostic importance of IKZF1 deletions for second-line treatment of BCP ALL is relatively underexamined in comparison with frontline protocols and, therefore, is less well understood. Despite differences in results of 2 published studies, 1 unifying conclusion is supportive of the idea that, similar to frontline therapy, the prognostic effect of IKZF1 deletions seem to be context-dependent.33,41 

Overall, these observations indicate that the complex interplay of exposure and response to treatment with the underlying disease biology is the key driver determining the prognostic impact of IKZF1 aberrations. Thus, a greater understanding of the interrelationships between treatment, treatment response, and cooperating molecular lesions in IKZF1-aberrant ALL could help to develop improved treatments tailored to the different scenarios.

The suggested molecular mechanisms involved in drug resistance mediated by IKZF1 alteration in BCP ALL are complex and not completely understood. However, there is evidence that loss of IKAROS in normal BCP cells or BCR-ABL1+ BCP ALL leads to acquisition of a stem cell–like phenotype with increases in self-renewal, upregulation of focal adhesion kinase (FAK), cell-adhesion molecules, and drug resistance through formation of a de novo superenhancer landscape of collaborating master transcription regulators and B-cell transcription factors.63-65  Of interest, Churchman et al demonstrated reversal of such a phenotype in IKZF1-aberrant BCR-ABL1+ BCP ALL by treatment with retinoid receptor agonists.65  Retinoids induced selective expression of wild-type IKZF1 and initiated expression of IKZF1 target genes. Similar results were obtained through FAK inhibition and both treatments, retinoid receptor agonists and FAK inhibition, improved the sensitivity of BCR-ABL1+ BCP ALL to tyrosine kinase inhibitor therapy.65,66  In addition, there are intriguing data demonstrating that IKZF1 controls energy metabolism in BCP ALL with a direct link to glucocorticoid response.67,68  These important insights may allow the development of new specific therapeutic approaches targeting IKZF1-altered signaling networks in BCP ALL.

Risk stratification and associated treatment strategies in trials on pediatric ALL are often not fully comparable. This may, at least in part, contribute to the partially different impact of aberrant IKZF1 on outcome observed in some of the published trials (Table 1). However, these complicating issues may also bear chances, if we engage into more detailed and careful comparisons of the different treatment backgrounds and their variant stratification strategies in context with IKZF1 aberrations and the relevant clinical end points. Following such strategies may allow us to nourish our knowledge on IKZF1 aberration with a better understanding of the differential response of affected patients to specific parts of the applied treatment protocols. Cooperative approaches incorporating well-characterized trial populations from large study groups will likely be key to success in this regard and should also answer many additional open questions such as the importance of a truly comprehensive assessment of the molecular spectrum of IKZF1 alterations (copy-number variation, nucleotide mutations, gene fusions), their prognostic importance in ALL subgroups, and the development of a full picture of the interplay of aberrant IKZF1 with cooperating genetic aberrations (eg, CDKN2A, PAX5, BTG1, JAK pathway aberrations, RAS pathway aberrations, other kinase activating lesions, ERG, subclonality of cooperating lesions).

Therefore, by continuing our research on IKZF1 in pediatric ALL, we will (1) be able to improve information for patients and/or their guardians regarding the risk of recurrence and final outcome of ALL; (2) very likely gain further insights into the biology of ALL; and (3) increase our ability to design and conduct clinical trials delivering optimized treatment strategies based on improved individual characterization of children and adolescents with ALL.

So, is IKZF1 aberration still a prognostic factor? As it turns out, the answer is: yes, more than ever!

The authors thank all participants and personnel involved in trials AIEOP-BFM ALL 2000, EORTC-CLG 58951, UKALL 97/99, and UKALL 2003. Particular thanks go to Gianni Cazzaniga for critical discussions. The authors also thank Anna Stengel and Stefanie Junk for their support regarding the development and adaptation of the Rscript for mutation plotting.

This work was supported by the ERA-NET TRANSCAN/European Commission under the 7th Framework Programme (FP7), Madeleine-Schickedanz-Kinderkrebsstiftung, Deutsche Krebshilfe, and Bloodwise blood cancer charity.

Contribution: M.S., H.C., and A.V.M. designed and performed research, analyzed and discussed data, and wrote the paper.

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

Correspondence: Martin Stanulla, Department of Pediatric Hematology and Oncology, Hannover Medical School, Carl-Neuberg-Str 1, D-30625 Hannover, Germany; e-mail: stanulla.martin@mh-hannover.de.

1.
Pui
CH
,
Yang
JJ
,
Hunger
SP
, et al
.
Childhood acute lymphoblastic leukemia: progress through collaboration
.
J Clin Oncol
.
2015
;
33
(
27
):
2938
-
2948
.
2.
Iacobucci
I
,
Mullighan
CG
.
Genetic basis of acute lymphoblastic leukemia
.
J Clin Oncol
.
2017
;
35
(
9
):
975
-
983
.
3.
Starý
J
,
Zuna
J
,
Zaliova
M
.
New biological and genetic classification and therapeutically relevant categories in childhood B-cell precursor acute lymphoblastic leukemia
.
F1000Res
.
2018
;
7
.
4.
Vrooman
LM
,
Silverman
LB
.
Treatment of childhood acute lymphoblastic leukemia: prognostic factors and clinical advances
.
Curr Hematol Malig Rep
.
2016
;
11
(
5
):
385
-
394
.
5.
Hunger
SP
,
Mullighan
CG
.
Redefining ALL classification: toward detecting high-risk ALL and implementing precision medicine
.
Blood
.
2015
;
125
(
26
):
3977
-
3987
.
6.
Georgopoulos
K
,
Bigby
M
,
Wang
JH
, et al
.
The Ikaros gene is required for the development of all lymphoid lineages
.
Cell
.
1994
;
79
(
1
):
143
-
156
.
7.
Molnár
A
,
Wu
P
,
Largespada
DA
, et al
.
The Ikaros gene encodes a family of lymphocyte-restricted zinc finger DNA binding proteins, highly conserved in human and mouse
.
J Immunol
.
1996
;
156
(
2
):
585
-
592
.
8.
Rebollo
A
,
Schmitt
C
.
Ikaros, Aiolos and Helios: transcription regulators and lymphoid malignancies
.
Immunol Cell Biol
.
2003
;
81
(
3
):
171
-
175
.
9.
Mullighan
CG
,
Miller
CB
,
Radtke
I
, et al
.
BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros
.
Nature
.
2008
;
453
(
7191
):
110
-
114
.
10.
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
;
360
(
5
):
470
-
480
.
11.
Chiaretti
S
,
Gianfelici
V
,
O’Brien
SM
,
Mullighan
CG
.
Advances in the genetics and therapy of acute lymphoblastic leukemia
.
Am Soc Clin Oncol Educ Book
.
2016
;
35
:
e314
-
e322
.
12.
Iacobucci
I
,
Storlazzi
CT
,
Cilloni
D
, et al
.
Identification and molecular characterization of recurrent genomic deletions on 7p12 in the IKZF1 gene in a large cohort of BCR-ABL1-positive acute lymphoblastic leukemia patients: on behalf of Gruppo Italiano Malattie Ematologiche dell’Adulto Acute Leukemia Working Party (GIMEMA AL WP)
.
Blood
.
2009
;
114
(
10
):
2159
-
2167
.
13.
Kuiper
RP
,
Waanders
E
,
van der Velden
VH
, et al
.
IKZF1 deletions predict relapse in uniformly treated pediatric precursor B-ALL
.
Leukemia
.
2010
;
24
(
7
):
1258
-
1264
.
14.
Yang
YL
,
Hung
CC
,
Chen
JS
, et al
.
IKZF1 deletions predict a poor prognosis in children with B-cell progenitor acute lymphoblastic leukemia: a multicenter analysis in Taiwan
.
Cancer Sci
.
2011
;
102
(
10
):
1874
-
1881
.
15.
Olsson
L
,
Albitar
F
,
Castor
A
, et al
.
Cooperative genetic changes in pediatric B-cell precursor acute lymphoblastic leukemia with deletions or mutations of IKZF1
.
Genes Chromosomes Cancer
.
2015
;
54
(
5
):
315
-
325
.
16.
Lana
T
,
de Lorenzo
P
,
Bresolin
S
, et al
.
Refinement of IKZF1 status in pediatric Philadelphia-positive acute lymphoblastic leukemia
.
Leukemia
.
2015
;
29
(
10
):
2107
-
2110
.
17.
Li
JF
,
Dai
YT
,
Lilljebjörn
H
, et al
.
Transcriptional landscape of B cell precursor acute lymphoblastic leukemia based on an international study of 1,223 cases
.
Proc Natl Acad Sci USA
.
2018
;
115
(
50
):
E11711
-
E11720
.
18.
Gu
Z
,
Churchman
ML
,
Roberts
KG
, et al
.
PAX5-driven subtypes of B-progenitor acute lymphoblastic leukemia
.
Nat Genet
.
2019
;
51
(
2
):
296
-
307
.
19.
Lilljebjörn
H
,
Henningsson
R
,
Hyrenius-Wittsten
A
, et al
.
Identification of ETV6-RUNX1-like and DUX4-rearranged subtypes in paediatric B-cell precursor acute lymphoblastic leukaemia
.
Nat Commun
.
2016
;
7
:
11790
.
20.
Atak
ZK
,
Gianfelici
V
,
Hulselmans
G
, et al
.
Comprehensive analysis of transcriptome variation uncovers known and novel driver events in T-cell acute lymphoblastic leukemia
.
PLoS Genet
.
2013
;
9
(
12
):
e1003997
.
21.
Gu
Z
,
Churchman
M
,
Roberts
K
, et al
.
Genomic analyses identify recurrent MEF2D fusions in acute lymphoblastic leukaemia
.
Nat Commun
.
2016
;
7
:
13331
.
22.
Qian
M
,
Zhang
H
,
Kham
SK
, et al
.
Whole-transcriptome sequencing identifies a distinct subtype of acute lymphoblastic leukemia with predominant genomic abnormalities of EP300 and CREBBP
.
Genome Res
.
2017
;
27
(
2
):
185
-
195
.
23.
Chen
B
,
Jiang
L
,
Zhong
ML
, et al
.
Identification of fusion genes and characterization of transcriptome features in T-cell acute lymphoblastic leukemia
.
Proc Natl Acad Sci USA
.
2018
;
115
(
2
):
373
-
378
.
24.
Panagopoulos
I
,
Brunetti
M
,
Stoltenberg
M
, et al
.
Novel GTF2I-PDGFRB and IKZF1-TYW1 fusions in pediatric leukemia with normal karyotype
.
Exp Hematol Oncol
.
2019
;
8
:
12
.
25.
Mata-Rocha
M
,
Rangel-López
A
,
Jiménez-Hernández
E
, et al
.
Identification and characterization of novel fusion genes with potential clinical applications in Mexican children with acute lymphoblastic leukemia
.
Int J Mol Sci
.
2019
;
20
(
10
).
26.
Lopes
BA
,
Meyer
C
,
Barbosa
TC
, et al
.
IKZF1 deletions with COBL breakpoints are not driven by RAG-mediated recombination events in acute lymphoblastic leukemia
.
Transl Oncol
.
2019
;
12
(
5
):
726
-
732
.
27.
Papaemmanuil
E
,
Hosking
FJ
,
Vijayakrishnan
J
, et al
.
Loci on 7p12.2, 10q21.2 and 14q11.2 are associated with risk of childhood acute lymphoblastic leukemia
.
Nat Genet
.
2009
;
41
(
9
):
1006
-
1010
.
28.
Treviño
LR
,
Yang
W
,
French
D
, et al
.
Germline genomic variants associated with childhood acute lymphoblastic leukemia
.
Nat Genet
.
2009
;
41
(
9
):
1001
-
1005
.
29.
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
.
30.
Buitenkamp
TD
,
Pieters
R
,
Gallimore
NE
, et al
.
Outcome in children with Down’s syndrome and acute lymphoblastic leukemia: role of IKZF1 deletions and CRLF2 aberrations
.
Leukemia
.
2012
;
26
(
10
):
2204
-
2211
.
31.
Asai
D
,
Imamura
T
,
Suenobu
S
, et al;
Japan Association of Childhood Leukemia Study (JACLS)
.
IKZF1 deletion is associated with a poor outcome in pediatric B-cell precursor acute lymphoblastic leukemia in Japan
.
Cancer Med
.
2013
;
2
(
3
):
412
-
419
.
32.
Yamashita
Y
,
Shimada
A
,
Yamada
T
, et al
.
IKZF1 and CRLF2 gene alterations correlate with poor prognosis in Japanese BCR-ABL1-negative high-risk B-cell precursor acute lymphoblastic leukemia
.
Pediatr Blood Cancer
.
2013
;
60
(
10
):
1587
-
1592
.
33.
Krentz
S
,
Hof
J
,
Mendioroz
A
, et al
.
Prognostic value of genetic alterations in children with first bone marrow relapse of childhood B-cell precursor acute lymphoblastic leukemia
.
Leukemia
.
2013
;
27
(
2
):
295
-
304
.
34.
Dörge
P
,
Meissner
B
,
Zimmermann
M
, et al
.
IKZF1 deletion is an independent predictor of outcome in pediatric acute lymphoblastic leukemia treated according to the ALL-BFM 2000 protocol
.
Haematologica
.
2013
;
98
(
3
):
428
-
432
.
35.
Palmi
C
,
Valsecchi
MG
,
Longinotti
G
, et al
.
What is the relevance of Ikaros gene deletions as a prognostic marker in pediatric Philadelphia-negative B-cell precursor acute lymphoblastic leukemia?
Haematologica
.
2013
;
98
(
8
):
1226
-
1231
.
36.
van der Veer
A
,
Waanders
E
,
Pieters
R
, et al
.
Independent prognostic value of BCR-ABL1-like signature and IKZF1 deletion, but not high CRLF2 expression, in children with B-cell precursor ALL
.
Blood
.
2013
;
122
(
15
):
2622
-
2629
.
37.
van der Veer
A
,
Zaliova
M
,
Mottadelli
F
, et al
.
IKZF1 status as a prognostic feature in BCR-ABL1-positive childhood ALL
.
Blood
.
2014
;
123
(
11
):
1691
-
1698
.
38.
Moorman
AV
,
Enshaei
A
,
Schwab
C
, et al
.
A novel integrated cytogenetic and genomic classification refines risk stratification in pediatric acute lymphoblastic leukemia
.
Blood
.
2014
;
124
(
9
):
1434
-
1444
.
39.
Olsson
L
,
Ivanov Öfverholm
I
,
Norén-Nyström
U
, et al
.
The clinical impact of IKZF1 deletions in paediatric B-cell precursor acute lymphoblastic leukaemia is independent of minimal residual disease stratification in Nordic Society for Paediatric Haematology and Oncology treatment protocols used between 1992 and 2013
.
Br J Haematol
.
2015
;
170
(
6
):
847
-
858
.
40.
Clappier
E
,
Grardel
N
,
Bakkus
M
, et al;
European Organisation for Research and Treatment of Cancer, Children’s Leukemia Group (EORTC-CLG)
.
IKZF1 deletion is an independent prognostic marker in childhood B-cell precursor acute lymphoblastic leukemia, and distinguishes patients benefiting from pulses during maintenance therapy: results of the EORTC Children’s Leukemia Group study 58951
.
Leukemia
.
2015
;
29
(
11
):
2154
-
2161
.
41.
Irving
JA
,
Enshaei
A
,
Parker
CA
, et al
.
Integration of genetic and clinical risk factors improves prognostication in relapsed childhood B-cell precursor acute lymphoblastic leukemia
.
Blood
.
2016
;
128
(
7
):
911
-
922
.
42.
Sutton
R
,
Venn
NC
,
Law
T
, et al
.
A risk score including microdeletions improves relapse prediction for standard and medium risk precursor B-cell acute lymphoblastic leukaemia in children
.
Br J Haematol
.
2018
;
180
(
4
):
550
-
562
.
43.
Vrooman
LM
,
Blonquist
TM
,
Harris
MH
, et al
.
Refining risk classification in childhood B acute lymphoblastic leukemia: results of DFCI ALL Consortium Protocol 05-001
.
Blood Adv
.
2018
;
2
(
12
):
1449
-
1458
.
44.
Yeoh
AEJ
,
Lu
Y
,
Chin
WHN
, et al
.
Intensifying treatment of childhood B-lymphoblastic leukemia with IKZF1 deletion reduces relapse and improves overall survival: results of Malaysia-Singapore ALL 2010 study
.
J Clin Oncol
.
2018
;
36
(
26
):
2726
-
2735
.
45.
Hinze
L
,
Möricke
A
,
Zimmermann
M
, et al
.
Prognostic impact of IKZF1 deletions in association with vincristine-dexamethasone pulses during maintenance treatment of childhood acute lymphoblastic leukemia on trial ALL-BFM 95
.
Leukemia
.
2017
;
31
(
8
):
1840
-
1842
.
46.
Slayton
WB
,
Schultz
KR
,
Kairalla
JA
, et al
.
Dasatinib plus intensive chemotherapy in children, adolescents, and young adults with Philadelphia chromosome-positive acute lymphoblastic leukemia: results of Children’s Oncology Group trial AALL0622
.
J Clin Oncol
.
2018
;
36
(
22
):
2306
-
2314
.
47.
Caye
A
,
Beldjord
K
,
Mass-Malo
K
, et al
.
Breakpoint-specific multiplex PCR allows the detection of IKZF1 intragenic deletions and minimal residual disease monitoring in B-cell precursor acute lymphoblastic leukaemia
.
Haematologica
.
2013
;
98
(
4
):
597
-
601
.
48.
Morel
G
,
Deau
MC
,
Simand
C
, et al
.
Large deletions of the 5′ region of IKZF1 lead to haploinsufficiency in B-cell precursor acute lymphoblastic leukaemia
.
Br J Haematol
.
2019
;
186
(
5
):
e155
-
e159
.
49.
Waanders
E
,
van der Velden
VH
,
van der Schoot
CE
, et al
.
Integrated use of minimal residual disease classification and IKZF1 alteration status accurately predicts 79% of relapses in pediatric acute lymphoblastic leukemia
.
Leukemia
.
2011
;
25
(
2
):
254
-
258
.
50.
Volejnikova
J
,
Mejstrikova
E
,
Dörge
P
, et al
.
Ikaros (IKZF1) alterations and minimal residual disease at day 15 assessed by flow cytometry predict prognosis of childhood BCR/ABL-negative acute lymphoblastic leukemia
.
Pediatr Blood Cancer
.
2013
;
60
(
3
):
420
-
427
.
51.
Tran
TH
,
Harris
MH
,
Nguyen
JV
, et al
.
Prognostic impact of kinase-activating fusions and IKZF1 deletions in pediatric high-risk B-lineage acute lymphoblastic leukemia
.
Blood Adv
.
2018
;
2
(
5
):
529
-
533
.
52.
Roberts
KG
,
Li
Y
,
Payne-Turner
D
, et al
.
Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia
.
N Engl J Med
.
2014
;
371
(
11
):
1005
-
1015
.
53.
Steeghs
EMP
,
Boer
JM
,
Hoogkamer
AQ
, et al
.
Copy number alterations in B-cell development genes, drug resistance, and clinical outcome in pediatric B-cell precursor acute lymphoblastic leukemia
.
Sci Rep
.
2019
;
9
(
1
):
4634
.
54.
Boer
JM
,
van der Veer
A
,
Rizopoulos
D
, et al
.
Prognostic value of rare IKZF1 deletion in childhood B-cell precursor acute lymphoblastic leukemia: an international collaborative study
.
Leukemia
.
2016
;
30
(
1
):
32
-
38
.
55.
Mullighan
CG
,
Zhang
J
,
Harvey
RC
, et al
.
JAK mutations in high-risk childhood acute lymphoblastic leukemia
.
Proc Natl Acad Sci USA
.
2009
;
106
(
23
):
9414
-
9418
.
56.
Clappier
E
,
Auclerc
MF
,
Rapion
J
, et al
.
An intragenic ERG deletion is a marker of an oncogenic subtype of B-cell precursor acute lymphoblastic leukemia with a favorable outcome despite frequent IKZF1 deletions
.
Leukemia
.
2014
;
28
(
1
):
70
-
77
.
57.
Zaliova
M
,
Zimmermannova
O
,
Dörge
P
, et al
.
ERG deletion is associated with CD2 and attenuates the negative impact of IKZF1 deletion in childhood acute lymphoblastic leukemia
.
Leukemia
.
2014
;
28
(
1
):
182
-
185
.
58.
Zhang
J
,
McCastlain
K
,
Yoshihara
H
, et al;
St. Jude Children’s Research Hospital–Washington University Pediatric Cancer Genome Project
.
Deregulation of DUX4 and ERG in acute lymphoblastic leukemia
.
Nat Genet
.
2016
;
48
(
12
):
1481
-
1489
.
59.
Yasuda
T
,
Tsuzuki
S
,
Kawazu
M
, et al
.
Recurrent DUX4 fusions in B cell acute lymphoblastic leukemia of adolescents and young adults [published correction appears in Nat Genet. 2016;48(12):1591]
.
Nat Genet
.
2016
;
48
(
5
):
569
-
574
.
60.
Zaliova
M
,
Potuckova
E
,
Hovorkova
L
, et al
.
ERG deletions in childhood acute lymphoblastic leukemia with DUX4 rearrangements are mostly polyclonal, prognostically relevant and their detection rate strongly depends on screening method sensitivity
.
Haematologica
.
2019
;
104
(
7
):
1407
-
1416
.
61.
Stanulla
M
,
Dagdan
E
,
Zaliova
M
, et al;
International BFM Study Group
.
IKZF1plus defines a new minimal residual disease-dependent very-poor prognostic profile in pediatric B-cell precursor acute lymphoblastic leukemia
.
J Clin Oncol
.
2018
;
36
(
12
):
1240
-
1249
.
62.
O’Connor
D
,
Moorman
AV
,
Wade
R
, et al
.
Use of minimal residual disease assessment to redefine induction failure in pediatric acute lymphoblastic leukemia
.
J Clin Oncol
.
2017
;
35
(
6
):
660
-
667
.
63.
Joshi
I
,
Yoshida
T
,
Jena
N
, et al
.
Loss of Ikaros DNA-binding function confers integrin-dependent survival on pre-B cells and progression to acute lymphoblastic leukemia
.
Nat Immunol
.
2014
;
15
(
3
):
294
-
304
.
64.
Hu
Y
,
Zhang
Z
,
Kashiwagi
M
, et al
.
Superenhancer reprogramming drives a B-cell-epithelial transition and high-risk leukemia
.
Genes Dev
.
2016
;
30
(
17
):
1971
-
1990
.
65.
Churchman
ML
,
Low
J
,
Qu
C
, et al
.
Efficacy of retinoids in IKZF1-mutated BCR-ABL1 acute lymphoblastic leukemia
.
Cancer Cell
.
2015
;
28
(
3
):
343
-
356
.
66.
Churchman
ML
,
Evans
K
,
Richmond
J
, et al
.
Synergism of FAK and tyrosine kinase inhibition in Ph+ B-ALL
.
JCI Insight
.
2016
;
1
(
4
).
67.
Chan
LN
,
Chen
Z
,
Braas
D
, et al
.
Metabolic gatekeeper function of B-lymphoid transcription factors [published correction appears in Nature. 2018;558(7711):E5]
.
Nature
.
2017
;
542
(
7642
):
479
-
483
.
68.
Marke
R
,
Havinga
J
,
Cloos
J
, et al
.
Tumor suppressor IKZF1 mediates glucocorticoid resistance in B-cell precursor acute lymphoblastic leukemia
.
Leukemia
.
2016
;
30
(
7
):
1599
-
1603
.
Sign in via your Institution