Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Dec 1.
Published in final edited form as: Best Pract Res Clin Haematol. 2021 Oct 23;34(4):101331. doi: 10.1016/j.beha.2021.101331

Has Ph-like ALL Superseded Ph+ ALL as the Least Favorable Subtype?

Thai Hoa Tran 1,2, Sarah K Tasian 3,4,*
PMCID: PMC8649641  NIHMSID: NIHMS1753625  PMID: 34865703

Abstract

Philadelphia chromosome-like acute lymphoblastic leukemia (Ph-like ALL) is a subset of high-risk B-ALL associated with high relapse risk and inferior clinical outcomes across the pediatric-to-adult age spectrum. Ph-like ALL is characterized by frequent IKZF1 alterations and a kinase-activated gene expression profile similar to that of Philadelphia chromosome-positive (Ph+) ALL, yet lacks the canonical BCR-ABL1 rearrangement. Advances in high-throughput sequencing technologies during the past decade have unraveled the genomic landscape of Ph-like ALL, revealing a diverse array of kinase-activating translocations and mutations that may be amenable to targeted therapies that have set a remarkable precision medicine paradigm for patients with Ph+ ALL. Collaborative scientific efforts to identify and characterise Ph-like ALL during the past decade has directly informed current precision medicine trials investigating the therapeutic potential of tyrosine kinase inhibitor-based therapies for children, adolescents, and adults with Ph-like ALL, although the most optimal treatment paradigm for this high-risk group of patients has yet to be established. Herein, we describe the epidemiology, clinical features, and biology of Ph-like ALL, highlight challenges in implementing pragmatic and cost-effective diagnostic algorithms in the clinic, and describe the milieu of treatment strategies under active investigation that strive to decrease relapse risk and improve long-term survival for patients with Ph-like ALL as has been successfully achieved for those with Ph+ ALL.

Keywords: ABL, acute lymphoblastic leukemia, clinical trials, CRLF2, JAK/STAT, Philadelphia chromosome-like, precision medicine, tyrosine kinase inhibitor

Definition of Ph-like ALL

Ph-like ALL was identified in 2009 independently by two research groups using different gene expression profiling classifiers. Researchers in the Children’s Oncology Group (COG), University of New Mexico (UNM), and St Jude Children’s Research Hospital (SJCRH) via the National Cancer Institute (NCI) TARGET initiative (https://ocg.cancer.gov/programs/target/projects/acute-lymphoblastic-leukemia) utilized Affymetrix gene expression microarray data to identify 257 gene probe sets that defined a distinct gene expression signature of both Ph+ ALL and Ph-like ALL (1), while den Boer and colleagues at Erasmus Medical Center and the Dutch Children’s Oncology Group used hierarchical clustering of 110 probe sets to predict 6 major pediatric ALL subtypes (ETV6-RUNX1, high hyperdiploid, TCF3-PBX1, KMT2A-rearranged, BCR-ABL1, and TALL) (2). Despite sharing only 9 common probe sets of 7 genes (CCND2, SH3BP5, ABL1, SOCS2, DUSP6, LST1 and EGFL7), both gene classifiers identified a subset of high-risk B-ALL patients with poor survival who had frequent deletions of B-cell development and transcription factor genes, such as IKZF1 (3). These assays and additional advances in RNA-sequencing were further able to define new genetic alterations deregulating tyrosine kinase or cytokine receptor genes in Ph-like ALL, including CRLF2, JAK2, ABL1, PDGFRB, and others (4). The COG/UNM/SJCRH group subsequently developed a clinically-validated Ph-like ALL screening assay, which measures the expression of 8 or 15 genes in a 384-well low-density microarray (LDA) microfluidic card to detect the Ph-like ALL signature. This clinical assay is now capable of results return within 24–48 hours of sample submission (5). The LDA assay is currently used by the COG and other consortia to screen patients with newly-diagnosed high-risk B-ALL for the Ph-like signature and to allocate those with LDA positivity for downstream molecular testing to identify specific Ph-like-associated genetic alterations (6). The most clinically relevant endpoint in patients with Ph-like ALL remains identification of specific translocations and mutations that activate oncogenic kinase signaling and may be therapeutically targeted.

Biology and genomic landscape of Ph-like ALL

In their 2014 landmark paper, Roberts and colleagues described the genomic landscape of Ph-like ALL via a comprehensive genomic analysis of 154 high-risk B-ALL specimens from children and adolescents/young adults (AYAs) (7). The unifying molecular hallmark of Ph-like ALL resides in the heterogeneous spectrum of genetic alterations activating cytokine receptor genes and kinase-signaling pathways (7). These alterations can be subdivided in 4 distinct genomically-defined subsets based upon their underlying kinase-activating lesions: (1) alterations in JAK/STAT pathway genes (predominantly CRLF2, JAK2, EPOR, IL7R, SH2B3), (2) ABL class alterations (ABL1, ABL2, CSF1R, LYN, PDGFRA, and PDGFRB), (3) uncommon Ras pathway mutations (NRAS, KRAS, NF1, PTPN11, CBL), and (4) rare kinase fusions (NTRK3, PTK2B, BLNK) and are described in greater detail below (Table 1, Figure 1) (7, 8). While CRLF2 (cytokine receptor-like factor 2) rearrangements also occur with lower frequency in children with standard-risk (SR) B-ALL (911) and in >50% of trisomy 21/Down syndrome (DS)-associated B-ALL (12, 13) (and may or may not have the associated Ph-like gene expression signature), other Ph-like ALL-associated kinase fusions have only extremely rarely been discovered in patients with SR disease.

Table 1.

Current landscape of Ph-like ALL kinase rearrangements, therapeutic targets, and clinical trials.

Ph-like genetic subgroups 3’ kinase genes 5’ fusion partner genes Kinase inhibitors Clinical trials
JAK/STAT pathway alterations CRLF2 CSF2RA, IGH, P2RY8 ruxolitinib NCT02420717
(MDACC)

NCT02723994
(COG AALL1521)

NCT03117751
(SJCRH Total XVII)

NCT03571321
(University of Chicago)
JAK2 ATF7IP, BCR, EBF1, ETV6, GOLGA5, HMBOX1, OFD1, PAX5, PCM1, PPFIBP1, RFX3, SMU1, SNX29, SSBP2, STRN3, TERF2, TPR, USP25, ZBTB46, ZNF274, ZNF340 ruxolitinib
EPOR IGH, IGK, IGL, LAIR1, THADA ruxolitinib
TSLP IQGAP2 ruxolitinib
IL2RB MYH9 ruxolitinib
ABL class alterations ABL1 CENPC, ETV6, FOXP1, LSM14A, NUP153, NUP214, RANBP2, RCSD1, SFPQ, SHIP1, SNX1, SNX2, SPTNA1, ZMIZ1 dasatinib, imatinib, others NCT01406756
(COG AALL1131)

NCT02143414
(SWOG S1318)

NCT02420717
(MDACC)

NCT03007147
(COG AALL1631)

NCT03117751
(SJCRH Total XVII)
ABL2 PAG1, RCSD1, ZC3HAV1 dasatinib, imatinib
CSF1R MEF2D, SSBP2, TBL1XR1 dasatinib, imatinib
PDGFRA FIP1L1 dasatinib, imatinib
PDGFRB ATF7IP, EBF1, ETV6, NUMA1, SNX29, SSBP2, TERF2, TNIP1, ZEB2, ZMYND8, ZNF608 dasatinib, imatinib
LYN GATAD2A, NCOR1 dasatinib, imatinib
Other kinases NTRK3 ETV6 entrectinib
larotrectinib
NCT03066661
NCT03834961
PTK2B KDM6A, STAG2, TMEM2 FAK inhibitors
FGFR1 BCR ponatinib
FLT3 ZMYM2 FLT3 inhibitors
TYK2 MYB, SMARCA4, ZNF340 JAK1/3 inhibitor
BLNK DNTT
CBL KANK1
DGKH ZFAND3

COG = Children’s Oncology Group, SJCRH = St Jude Children’s Research Hospital, MDACC = MD Anderson Cancer Center, SWOG = Southwestern Oncology Group.

Figure 1. Frequency of Ph-like ALL and genetic subtypes by age group.

Figure 1.

NCI HR B-ALL = National Cancer Institute high risk B-acute lymphoblastic leukemia, NCI SR B-ALL = National Cancer Institute standard risk B-acute lymphoblastic leukemia.

Deletions of IKZF1 and other lymphoid transcription factor genes occur commonly in Ph-like ALL, as has been similarly seen in Ph+ ALL (7, 1417). IKZF1 encodes the zinc-finger DNA-binding Ikaros, a transcription factor essential for B-cell lymphoid development. Its alteration results in acquired stem cell-like properties, aberrant bone marrow stromal adhesion, and chemotherapy resistance (1820). The most common IKZF1 alteration is intragenic focal deletion of exons 4–7, which results in the dominant-negative Ik6 isoform (20). In one study, IKZF1 alterations were detected in 68% of Ph-like ALL compared to 16% of non-Ph-like ALL cases (7). Inferior clinical outcomes in patients with IKZF1-deleted Ph-like ALL have also been reported (7, 21). Recent studies from European consortia have further described inferior outcomes of patients with the new IKZF1plus molecular profile, which is defined by deletion of IKZF1 co-occurring with one or more deletions in CDKN2A (heterozygous or homozygous), CDKN2B (homozygous only), PAX5, or the pseudoautosomal region 1 (PAR1) region of the sex chromosomes where CRLF2 is located and in the absence of ERG deletion (2225). Stanulla and colleagues reported that the IKZF1plus signature conferred the highest hazard ratio for relapse in multivariate analysis and could be incorporated in clinical decision algorithms to refine risk stratification in addition to MRD response (22). The IKZF1plus subgroup in these studies likely contains a high proportion of Ph-like ALL patients, as both populations share adverse clinical and biologic features of higher white blood cell (WBC) at diagnosis, poor prednisone response, positive minimal residual disease (MRD) after induction therapy, and higher frequency of the germline GATA3 variant rs3824662 reported to predispose to developing Ph-like ALL (2224, 26).

JAK/STAT pathway gene alterations

Approximately 50% of children and AYAs with Ph-like ALL harbor CRLF2 rearrangements (10, 27, 28), which leads to CRLF2 overexpression and increased surface protein expression of the thymic stromal lymphopoietin receptor (TSLPR; encoded by CRLF2) detectable by flow cytometry (29). CRLF2 alterations occur via two major mechanisms: (1) focal deletion of PAR1 on chromosomes Xp22/Yp11 resulting in the P2RY8-CRLF2 fusion or as (2) translocation to the immunoglobulin heavy chain enhancer region on chromosome 14, resulting in IGH-CRLF2 rearrangement (12, 30). Both rearrangements result in overexpression of full-length CRLF2, which heterodimerizes with the IL7R-alpha subunit to form the TSLPR involved in early B-cell development (3133). P2RY8-CRLF2 fusions appear to occur more frequently in younger children and in those with DS-ALL, (13) whereas IGH-CRLF2 predominates among adolescents and young adults, particularly those of Hispanic/Latino or Native American ancestry (12, 30, 34, 35). Rarely, activating CRLF2 F232C point mutations, which typically coexist with CRLF2 rearrangements, also lead to CRLF2 deregulation (36). Half of CRLF2-rearranged cases harbor concomitant point mutations in JAK2 or, less commonly, JAK1. The most frequently occurring point mutation is R683G in the JAK2 pseudokinase domain. JAK1 V658F, which is analogous to the JAK2 V617F mutation seen in adult myeloproliferative neoplasms, occurs much less frequently in CRLF2-rearranged Ph-like ALL (7, 10). Sequence mutations in IL7R and SH2B3 have also been identified in a small number of CRLF2-rearranged cases that lack concomitant JAK mutations (7, 37). Less commonly (~10% of patients), some children with detected CRLF2 rearrangements leading to CRLF2 overexpression (usually P2RY8-CRLF2 fusions in children with SR B-ALL) do not have the Ph-like expression signature (37). CRLF2 overexpression in the absence of specific rearrangement detection has been reported in a small percentage of patients (38).

Kinase fusions involving JAK2 or EPOR rearrangements, each representing approximately 5–10% of Ph-like ALL cases, are also associated with activation of JAK/STAT signaling (7, 10). Some groups have reported an increased prevalence of JAK2 fusions among the young adult population compared to children, ~15% vs 5%, respectively (5, 39). JAK2 is a promiscuous 3’ gene in Ph-like ALL with at least 19 different 5’ partner genes reported to date. All JAK2 fusions are in-frame and disrupt the JAK2 pseudokinase domain, which releases the normal auto-inhibition of the kinase domain and results in constitutive JAK/STAT signaling activation (4, 7, 8). Several 5’ partner genes in EPOR rearrangements have been described, each involving the juxtaposition of the EPOR gene to the enhancer region of immunoglobulin heavy (IGH) (most common), κ (IGK), or l (IGL) loci and leading to deregulated expression of a truncated form of EPOR that has been shown to drive Ph-like leukemogenesis and activated JAK/STAT signaling (40). Insertion and truncation of EPOR into the upstream region of LAIR1 or the THADA loci have also been reported in a very small number of patients (40). As with JAK2 fusions, prevalence of EPOR rearrangements rises with increasing age with peak prevalence among young adults (7, 40).

Additional mechanisms leading to JAK/STAT pathway activation beyond the aforementioned kinase or cytokine receptor-activating rearrangements implicate a diverse range of sequence mutations and copy number variations in genes such as JAK1, JAK3, IL7R, SH2B3, IL2RB, and TYK2 (7). These lesions collectively comprise 14% of children compared to 5% and 7.3% of adolescents and older adults respectively (4, 7). Although they lack a kinase-activating rearrangement, these cases often harbor chromosomal rearrangements expressing fusion oncoproteins involving transcription factor genes (EBF1, PAX5, ETV6) and/or epigenetic regulators (CREBBP, SETD2, ASXL1) that merit further study (4).

ABL class alterations

The second most clinically relevant subgroup of Ph-like ALL is ABL class alterations, which account for approximately 10% of cases and are mutually exclusive from the CRLF2 rearrangements and other JAK pathway-activating alterations (7, 27). Prevalence peaks among children with NCI HIGH-RISK B-ALL at 17%, then decreases to about 10% in young and older adults (7, 27, 37). ABL class rearrangements involve 3’ ABL1, ABL2, CSF1R, LYN, PDGFRA, or PDGFRB fusing with multiple 5’ partner genes. Preclinical and clinical studies have shown that ABL class fusion leukemias are sensitive to tyrosine kinase inhibitors, such as imatinib, dasatinib, or ponatinib (7, 8, 10, 41).

Ras pathway mutations

Approximately 4% of Ph-like ALL patients have activating mutations in Ras pathway genes, including KRAS, NRAS, NF1, PTPN11, and CBL (7). Ras pathway mutations are usually subclonal and can occur as the sole detected anomaly or in conjunction with sentinel Ph-like translocations (e.g., CRLF2, ABL class, JAK2, or EPOR fusions) (7, 28). Ras-activating mutations are also commonly found in hyperdiploid, hypodiploid, KTM2A-rearranged, and relapsed ALL and are also often subclonal (4244). It is not currently known whether these mutations are pathogenic drivers in childhood ALL.

Rare kinase fusions

Other rare fusions involving NTRK3, BLNK, DGKH, FGFR1, PTK2B, FLT3, or TYK2 collectively account for 3% of Ph-like ALL cases (7, 37). The ETV6-NTRK3 fusion, which has been reported in other malignancies such as infantile fibrosarcoma and secretory breast carcinoma, can induce an aggressive ALL with in vitro and in vivo sensitivity to TRK inhibitors (45). Clinical responses to larotrectinib, a pan-TRK inhibitor, in patients with relapsed/refractory B-ALL harboring ETV6-NTRK3 have been described in single case reports (46, 47). Functional modeling of other kinase fusions is important to determine their oncogenic role and identify novel therapeutic targets. As an example, FGFR1 fusions may be targetable with ponatinib (48) or pazopanib (49).

Epidemiology and clinical picture of Ph-like ALL

The prevalence of Ph-like ALL rises with increasing age and varies by ethnicity, gender, and NCI-defined ALL risk group. A recent meta-analysis of 15 studies reported that the pooled prevalence of Ph-like ALL across the age spectrum was 15.4% (50). By age group, the Ph-like subtype comprised 15.6% of B-ALL cases in children 1 to 10 years old, 26.2% in adolescents aged 11 to 20 years old, 25.8% in young adults 21 to 40 years old, and 16.9% in adults older than 40 years (7, 27, 28, 50, 51). Among children and adolescents with B-ALL, Ph-like ALL accounts for 13.6% of NCI SR cases and 22.4% of NCI HIGH-RISK cases (11, 37). In comparison to Ph+ ALL, Ph-like ALL is 3 times more common in the pediatric age group (5). Males are more commonly affected than females across the age spectrum with a male-to-female ratio of 1.5:1 among children and adults (7, 27). Ph-like ALL also has a predilection for patients of Hispanic/Latino or Native American ethnicity, especially among those with CRLF2 rearrangements. This phenomenon has been attributed in part to the increased prevalence of germline Ph-like ALL risk variant in GATA3 (rs3824662) in Hispanic individuals with Native American genetic ancestry (26, 52, 53). Furthermore, Ph-like ALL patients frequently have adverse clinical features with significantly higher rates of hyperleukocytosis at diagnosis, end-induction MRD positivity, and increased risk of treatment failure and relapse (7, 27, 34, 37, 51, 54). The Ph-like ALL gene expression profile itself may independently confer an adverse risk, as shown in some multivariate analyses (27, 51, 55). Patients harboring PDGFRB, JAK2, or EPOR fusions are notoriously associated with more aggressive disease course and frequent high rates of end-induction MRD or even induction failure (7, 5660).

The inferior survival of patients with Ph-like ALL (compared to non-Ph-like ALL) occurs across the age spectrum, and differential outcomes may further exist within the based upon induction chemotherapy response. Children with NCI high-risk B-ALL treated on the COG AALL0232 phase 3 trial (NCT00075725) with a retrospectively-identified Ph-like expression signature had a 5-year event-free survival (EFS) of 63% versus 86% (p<0.0001) of those with non-Ph-like ALL (55). Importantly, this inferior outcome was detected for patients with Ph-like ALL regardless of the randomized treatment arm assigned, which was in contrast to patients with non-Ph-like ALL with superiority of high-dose methotrexate versus dose-escalating Capizzi-style methotrexate in the first interim maintenance phase (55). More recent analyses of children with NCI SR Ph-like ALL treated on the COG AALL0331 phase 3 trial (NCT00103285) (61) showed statistically inferior outcomes versus those with SR non-Ph-like ALL with 7-year EFS 82.4% and 90.7% (p=0.0022). However, these patients appear to be salvageable at relapse with no difference in overall survival (OS) (93.2% vs 95.8%, p=0.14) between the two groups (11). Recent comprehensive analysis of all children with identified CRLF2-overexpressing ALL treated on COG SR B-ALL trials AALL0331 and AALL0932 (n=77) or high-risk B-ALL trials AALL0232 or AALL1131 (n=244) confirmed reasonable clinical outcomes for children with Ph-like SR B-ALL with 5-year EFS 87.2% and OS 94.5%, although EFS was inferior to that of children with non-Ph-like ALL (38). However, outcomes were quite poor for patients with high-risk B-ALL with 5-year EFS and OS of 56.3% and 75.4%, respectively. Finally, a recent comprehensive retrospective analysis from the Ponte di Legno group comprised of 14 pediatric oncology cooperative groups studied 122 children with Ph-like ABL-class ALL treated with chemotherapy alone (without TKI) and reported high rates of end-induction MRD and poor outcomes despite intensive chemotherapy and often HSCT in first remission. This cohort serves to establish baseline outcomes of children with ABL-class fusions in the pre-TKI era with reported 5-year cumulative incidence of relapse, EFS, and OS of 31.0%, 59.1%, and 76.1%, respectively (60).

Data from the CALGB 10403 phase 2 trial (NCT00558519) have shown marked improvement in outcome for adolescents and young adults (AYA) less than 40 years of age when using a similarly intensive pediatric-inspired regimen, although outcomes for Ph-like ALL AYA patients in that trial remained unfavorable with estimated 3-year EFS of 42% compared to 69% (p=0.008) for those with non-Ph-like ALL (62). Clinical outcomes of patients with Ph-like ALL also worsen with increasing age. Five recent studies focused upon defining the incidence and characteristics of Ph-like ALL occurring in young and older adults with B-ALL. Among 49 adults with Ph-like ALL treated at the MD Anderson Cancer Center (MDACC), the 5-year OS was 23% for Ph-like ALL vs 59% (p=0.006) for patients with non-Ph-like ALL (51). Another study of 194 Ph-like ALL patients from 21 to 86 years old reported 5-year EFS for young adults (21–39 years old), adults (40–59 years), and older adults (60–86 years) of 40.4%, 29.8%, and 18.9%, respectively (27). Among 88 Ph-negative B-ALL adult patients enrolled on the GIMEMA LAL1913 frontline protocol, 28 (31.8%) patients were identified as Ph-like harboring various kinase fusions. These patients had a significantly lower CR (74.1% vs. 91.5%; p=0.044) and EFS (33.5% vs. 66.2%; p=0.005) compared to non-Ph-like patients despite being treated with a pediatric-inspired and MRD-stratified protocol (63). Two additional North American and German studies of adult Ph-like ALL cohorts confirmed the poor outcomes of this population (28, 34). Ph-like ALL patients with concomitant IKZF1 alterations may have further inferior outcomes compared to those without IKZF1 alterations, although these analyses have been limited by small patient numbers (7, 21) (Table 2).

Table 2.

Prevalence, clinical features, and treatment outcomes of pediatric and adult patients with Ph-like ALL reported by study groups.

Study group Study period Patient population Prevalence of Ph-like ALL n (%) Median age at diagnosis (CI) Median WBC at diagnosis (CI) Male,% Ph-like gene signature detection methods Kinase alteration detection methods Clinical trials Treatment outcome References
Ponte di Legno ALL Working Group 2000–2018 Pediatric B-ALL ABL-class (n=122) 9.7 (mean age) 97.7 (mean WBC) 62 LDA FISH RT-PCR RNA-seq Frontline ALL trials without TKI 5-year EFS: 59%
5-year OS: 76.1%
den Boer et al Lancet Haematol, 2021
UK ALL 2012–2018 Pediatric ALL with slow response to induction (n=126) ABL-class (n=21) 9 35 71 N/A FISH SNP RNA fusion panel UKALL2011 Chemotherapy (n=8)
Imatinib + chemotherapy (n=13)
4-year EFS: 83.9% (TKI)
4-year OS: 83.9% (TKI)
4-year EFS : 37.5% (no TKI)
4-year OS : 75% (no TKI)
Moorman et al BJH 2020
GIMEMA 2014–2018 Adult Ph-negative B-ALL (n=88) n=28, 31,8% 42 (18–65) 3.34 (0.23–347) 68 LDA FISH MLPA NGS GIMEMA LAL1913 2-year EFS: 33.5%
2-year OS: 48.5%
Chiaretti et al Haematologica 2020
MDACC 2010–2012 Adult R/R B-ALL (n=53) n=12, 22.6% 36 (20–57) 4.2 (1.0–9.3) 67 PAM WGS
RNA-seq
Inotuzumab monotherapy Phase 2 trials 1-year EFS: 33%
1-year OS: 33%
Jabbour et al Blood 2019
AIEOP-BFM 2000–2018 B-ALL (n=3854) ABL-class (n=46) NA NA 63 HC FISH RT-PCR CGH RNA-seq Chemotherapy (n=33)
Chemotherapy + TKI (n=13)
5-year EFS: 62.9% (TKI)
5-year OS: 75.5% (TKI)
5-year EFS: 47.7% (no TKI)
5-year OS: 70.9% (no TKI)
Cario et al Haematologica 2019
FRALLE
CAALL
GRAALL
EWALL
EORTC
NA Pediatric and adults with B-ALL ABL-class (n=24) 24 (5–72) 30 (4–570) 67 HC FISH RT-MLPA RQ-PCR BP-PCR RNA-seq Chemo + TKI (n=24) 3-year EFS: 55%
3-year OS: 77%
Tanasi et al Blood 2019
CALGB 2007–2012 AYA B-ALL (n=131) n=41, 31.3% NA NA NA LDA LDA for CRLF2 aberration Pediatric-based chemotherapy Regimen CALGB 10403 3-year EFS: 42%
3-year OS: 63%
Stock et al Blood 2019
COG 2006–2008 SR B-ALL (n=1023) n=139, 13.6% NA 15.9 ± 13.7 50 LDA FISH RT-PCR Sanger sequencing RNA-seq COG AALL0331 phase 3 for SR B-ALL 7-year EFS: 82%
7-year OS: 93%
Roberts et al Blood 2018
CALGB
ECOG
MDACC
NILG
PMCC
SWOG
City of Hope
NA Adult B-ALL (n=798) n=194, 24.3% 40 (21–86) 56.6 (0.2–434) 61 PAMLDA FISH PCR Sanger sequencing RNA-seq Multiple trials 5-year EFS: 22.5%
5-year OS:23.8%
Roberts et al J Clin Oncol 2017
GMALL 1999–2005 Adult B-ALL (n=207) n=26, 13% 31 (16–59) NA 74 PAM FISH Q-PCR Sanger sequencing Multiple trials 5-year DFS: 19%
5-year OS: 22%
Herold et al Haematologica 2017
MDACC 2000–2012 Adult B-ALL (n=148) n=49, 33.1% 33.5 (15–71) 17 (1–603) 66 PAM LDA FISH Targeted NGS Chemotherapy (hyper-CVAD or augmented BFM) 5-year OS: 23% Jain et al Blood 2017
University Pennsylvania University of Michigan NA Adult B-ALL (n=89) n=18, 20.2% 43 (19–63) 71.1 ± 31.8 (median: 28.6) 69 LDA FISH PCR RT-PCR Sanger sequencing Archer FusionPlex Multiple trials Median survival: 1.6 years Tasian et al Leukemia 2017
JACLS
TCCSG
CCLSG
KYCCSG
NA Pediatric B-ALL (n=373) n=29, 8% 8.8 (1.9–16) 94.3 (0.6–420) 76 GSEA Multiplex RT-PCR RNA-seq Multiple trials 5-year EFS: 48.6% 5-year OS: 73.5% Imamura et al Blood Cancer J 2016
HOVON 1993–2009 Adult B-ALL (n=127) n=21, 17% 25 (16–59) NA NA HC RT-PCR Multiple trials 3-year EFS: ~25% Boer et al Haematologica 2015
COG
SJCRH
ECOG
CALGB MDACC
2000–2007 Children, AYA with B-ALL (n=1725) n=264, 15.3% NA 106 cHR: 62 A: 73 YA: 82 PAM FISH RT-PCR WES RNA-seq WGS Multiple trials 5-year EFS: 58 % (cHR), 41% (A), 24% (YA)
5-year OS: 73% (cHR), 66% (ado), 26% (YA)
Roberts et al NEJM 2014
SJCRH 2000–2007 Pediatric B-ALL (n=344) n=40, 11.6% 5.3 (1.31–8.6) 7.1 (1.7–258.3) 68 PAM FISH RT-PCR Sanger sequencing RNA-seq WGS MRD-based risk intensification regimen 5-year EFS: 90%
5-year OS: 92.5%
Roberts et al J Clin Oncol 2014
DCOG COALL 1994–2015 Pediatric B-ALL (n=572) n=94, 16% 7 44.5 49 HC FISH CRLF2 mRNA expression by microarray Multiple trials 5-year CIR: 32% van der Veer et al Blood 2013
COG 2002–2011 HR B-ALL (n=572) n=81, 14% NA NA NA PAM Kinome sequencing COG AALL0232 phase 3 trial for HR B-ALL 5-year EFS: 62.6% Loh et al Blood 2013
COG 2000–2003 Pediatric B-ALL (n=221) n=68, 31% NA NA NA PAM SNP PCR COG P9906 5-year EFS: 26% Mullighan et al NEJM 2009
DCOG COALL 1990–2004 Pediatric B-ALL (n=154) n=30, 19% NA NA NA HC RT-PCR Multiple trials 5-year DFS: 60%
(COALL) 5-year DFS: 57% (DCOG)
Den Boer et al Lancet Oncol 2009

AIEOP-BFM = Associazione Italiana di Ematologia e Oncologia Pediatrica and Berlin-Frankfurt-Münster cooperative groups, AYA = adolescents and young adults, BP-PCR = breakpoint-specific multiplex polymerase chain reaction, CAALL = French Protocol for the Treatment of ALL in Children and Adolescents, CALGB = Cancer and Leukemia Group B, CCLSG = Children’s Cancer and Leukemia Study Group, CGH = comparative genomic hybridization, cHR = childhood high-risk, CIR = cumulative incidence of relapse, COALL = Childhood Oncology Acute Lymphoblastic Leukemia, COG = Children’s Oncology Group, CRLF2 = cytokine receptor like factor 2, DCOG = Dutch Childhood Oncology Group, DFS = disease-free survival, EFS = event-free survival, ECOG = Eastern Cooperative Oncology Group, EORTC = European Organisation for Research and Treatment of Cancer, EWALL = European Working Group on Adult ALL, FISH = fluorescence in situ hybridization, FRALLE = French Group for Childhood ALL, GMALL = German Multicenter Study Group for Adult ALL, GRAALL = French-Swiss-Belgian Group for Research on Adult Acute Lymphoblastic Leukemia, GSEA = gene-set enrichment analysis, HC = hierarchical clustering, HOVON = Hemato-Oncologie voor Volwassenen Nederland, JACLS = Japanese Association of Childhood Leukemia Study, KYCCSG = Kyushu-Yamaguchi Children’s Cancer Study Group, LDA = low-density array, MDACC = MD Anderson Cancer Center, MLPA = multiplex ligation-dependent probe amplification, MRD = minimal residual disease, NILG = Northern Italy Leukemia Group, NA = not available, OS = overall survival, PAM = prediction analysis of microarrays, PMCC = Princess Margaret Cancer Centre, RNA-seq = RNA sequencing, RQ-PCR = real-time quantitative polymerase chain reaction, R/R B-ALL = relapsed/refractory B-acute lymphoblastic leukemia, RT-PCR = reverse transcription-polymerase chain reaction, SJCRH = St Jude Children’s Research Hospital, SNP = single nucleotide polymorphism, SR = standard-risk, SWOG = Southwestern Oncology Group, TCCSG = Tokyo Children’s Cancer Study Group, TKI = tyrosine kinase inhibitor, WES = whole-exome sequencing, WGS = whole genome sequencing.

Clinical outcomes among Ph-like ALL patients may also vary according to their underlying kinase-activating alterations, although larger prospective studies are required to confirm these observations. Roberts and colleagues reported that there are significant differences in 5-year EFS among different Ph-like genomic subgroups, with JAK2/EPOR-rearranged cases (26.1%) and CRLF2-rearranged JAK2-mutant cases (38.8%) having dismal prognosis, in contrast to cases harboring other JAK/STAT (68.3%) or Ras pathway mutations (85.7%) having a more favorable outcome (p<0.0001)(7). The recent Ponte di Legno ALL working group study also highlights the variability in MRD response and clinical outcomes in their cohort of ABL-class fusions depending on the specific fusion subtype. Notably, children with PDGFRB and ABL2 fusions appear more likely to have MRD levels greater than 10−2 at the end of induction and inferior EFS compared to their counterparts with ABL1 or the uncommon CSF1R rearrangements (60).

Diagnostic modalities and clinical workflow algorithms for Ph-like ALL

The heterogeneous spectrum of kinase-activating alterations, cryptic nature of these genetic aberrations by conventional cytogenetic analysis, and complexity of required testing has rendered Ph-like ALL quite challenging to diagnose via clinical laboratory assays. Several cooperative oncology consortia have adopted different screening strategies largely based on the patient population size to be tested, development of more rapid next-generation sequencing platforms, and availability of therapeutic clinical trials (6).

Clinical diagnosis of Ph-like ALL has involved assessment of the pathognomonic gene expression signature (used by some, but not all, groups) and detection of targetable kinase-activating alterations. Gene expression profiling, the utilized discovery modality in initial European and North American studies (1, 2), is not readily available in the clinic and has now been largely replaced by the TaqMan LDA microfluidic card measuring the expression of 8 or 15-gene panels (IGJ, SPATS2L, MUC4, CRLF2, CA6, NRXN3, BMPR1B, GPR110, CHN2, SEMA6A, PON2, SLC2A5, S100Z, TP53INP1, IFITM1) now used by the COG to determine the Ph-like ALL signature (6, 55, 64). An integrated score between 0 and 1 is generated from the 8 or 15-gene assay with a score ≥0.5 considered positive for the Ph-like gene signature (37). Higher LDA score (e.g., >0.7) typically suggests an underlying kinase fusion (6). This LDA-based approach has provided a rapid and cost-effective screening modality for some groups to identify patients with probable Ph-like ALL who require further detailed genomic characterization to identify targetable kinase-activating alterations and to minimize unnecessary downstream testing for LDA-negative patients. Aside from direct detection of CRLF2 overexpression and P2RY8-CRLF2 fusions on the LDA card, this modality is intended only as a screening tool and does not detect other Ph-like kinase-activating lesions. The LDA card is also capable of identifying and ‘ruling out’ patients with Ph+ ALL and ETV6-RUNX1 ALL who do not require additional Ph-like testing (65).

To enable detection of kinase-activating alterations (the most clinically relevant endpoint in the Ph-like ALL diagnostic work-up), several commercial, research-level, and clinical next-generation sequencing (NGS) platforms have been developed and are replacing prior multiplexed clinical RT-PCR panels (6, 66, 67). The ArcherDx FusionPlex Heme panel uses anchored multiplex PCR-based enrichment with the ability to detect novel fusions involving 87 genes associated with hematologic malignancies (6). The FoundationOne Heme panel is a targeted combined DNA and RNA sequencing method capable of fusion and mutation detection in >400 cancer-related genes (68). Of particular interest, transcriptomic/RNA sequencing (RNA-seq) represents a powerful tool for comprehensive fusion and mutation detection in addition to identifying the Ph-like GEP by hierarchical clustering (6) and is becoming more clinically available. Indeed, RNA-seq is the only single platform capable of fulfilling the two essential aspects of Ph-like ALL’s diagnosis, but is currently not considered time- or cost-effective for routine analysis of all patients. Clinical fusion and NGS assays are relatively more cost-effective, but still require a relatively long turnaround time (between 2–4 weeks) prior to clinical result reporting.

More rapid testing assays using conventional methods such as fluorescence in situ hybridization (FISH) and flow cytometry still retain clinical relevance for the diagnosis of Ph-like ALL. For example, dual color break-apart FISH probes are now commercially available for the canonical Ph-like 3’ kinase genes ABL1, ABL2, PDGFRB (will also detect CSF1R disruption at 5q32), CRLF2, JAK2, and EPOR with results typically delivered within 48 hours. Since the vast majority of these kinase fusions are exclusively seen with the Ph-like phenotype, an abnormal FISH result might be sufficient to start tyrosine kinase inhibitor (TKI) therapy while waiting for confirmatory molecular testing. The COG has expanded its routine FISH panel to include ABL class FISH probes in its HIGH-RISK B-ALL AALL1732 phase 3 trial (NCT03959085) and its Ph+ ALL AALL1631 phase 3 trial (NCT03007147) in order to facilitate earlier introduction of TKI during induction for patients with identified ABL class alterations. Increased flow cytometric staining of TSLPR also suggests underlying CRLF2 rearrangement, which is known to occur in over half of Ph-like ALL cases (29). Flow cytometry availability in most institutions’ diagnostic laboratories and rapid result return within 24 hours makes TSLPR immunophenotyping a compelling additional Ph-like screening assay.

In summary, successful identification of patients with Ph-like ALL will most likely benefit from a combined approach of cytogenetic, FISH, and molecular analysis via fusion and NGS testing given the known genetic heterogeneity of this leukemia subtype and ongoing new discovery. Pragmatic clinical screening algorithms will need to be personalized on one’s available resources, patient volume, and clinical goals. A suggested diagnostic algorithm for Ph-like ALL is shown in Figure 2 (6).

Figure 2. Suggested clinical screening algorithm for Ph-like ALL.

Figure 2.

CRLF2 = cytokine receptor-like factor 2, LDA = low-density array, FISH = fluorescence in situ hybridization, NGS = next-generation sequencing, PCR = polymerase chain reaction, RNA = ribonucleic acid, RT-PCR = reverse-transcription polymerase chain reaction, TSLPR = thymic stromal lymphopoietin receptor. Adapted from Harvey & Tasian Blood Adv 2020.

Precision medicine trials in Ph-like ALL

Targeted therapies

The recently characterized genomic landscape of Ph-like ALL has uncovered a myriad of kinase-activating alterations that expand the treatment paradigm of molecularly-targeted therapies in ALL and leverage the success story of TKI incorporation for children and adults with Ph+ ALL (15, 6972) (Table 3). Despite their heterogeneity, Ph-like ALL-associated genetic alterations commonly converge to activate JAK/STAT, ABL, or Ras signaling pathways. Extensive in vitro and in vivo data have provided compelling evidence to incorporate relevant ABL/PDGFR- or JAK-directed TKIs in combination with chemotherapy to improve these patients’ poor outcomes. The efficacy of such precision medicine approaches is currently being prospectively assessed in several clinical trials for children and adults with Ph-like ALL harboring ABL class alterations or JAK/STAT pathway gene lesions.

Table 3.

Outcomes of children and adolescents/young adults with Ph+ ALL treated with chemotherapy and tyrosine kinase inhibitors treated on cooperative group clinical trials.

COG AALL0031 EsPhALL2004 EsPhALL2010 COG AALL0622 COG AALL1122 CCCG-ALL-2015
Phase 3 2 2 2 2 3
TKI imatinib 340 mg/m2 imatinib 300 mg/m2 imatinib 300 mg/m2 dasatinib 60 mg/m2 dasatinib 60 mg/m2 imatinib 300 mg/m2
vs
dasatinib 80 mg/m2
Years 2002–2006 2004–2009 2010–2014 2008–2012 2012–2014 2015–2018
Patients (n) 91 160 155 60 106 97 (imatinib)
92 (dasatinib)
HSCT in CR1 25% 83% 38% 32% 14% 2%
5y EFS 71% (cohort 5) 60% 57% 60% 55% 4y EFS49% (imatinib)
4y EFS: 71% (dasatinib)
p=0.005
5y OS 81% (cohort 5) 72% 72% 86% 82% 4y EFS: 69% (imatinib)
4y EFS: 88% (dasatinib)
p=0.04

CCCG = Chinese Children’s Cancer Group, COG = Children’s Oncology Group, CR1, complete first remission, EFS = event-free survival, EsPhALL = European Ph+ ALL working group, HSCT = hematopoietic stem cell transplantation (allogeneic), OS = overall survival, 4y = 4-year, 5y = 5-year.

ABL class alterations phenocopy BCR-ABL1 and exhibit exquisite sensitivity to imatinib and dasatinib in preclinical models (7, 8, 73). There is increasing anecdotal evidence demonstrating that the addition of imatinib or dasatinib monotherapy or in combination with chemotherapy can induce remission in patients with relapsed/refractory ALL with ABL class alterations (7, 5658, 74, 75). Durable remissions and favorable outcomes have been reported in recent series of Ph-like ABL-class patients treated with imatinib or dasatinib in combination with chemotherapy compared to historical controls (7678). Based on the increasing clinical experience in Ph-like ALL and the robust demonstration of safety and efficacy in children with Ph+ ALL, the COG AALL1131 phase 3 trial (NCT02883049) was amended in 2016 to add a dedicated treatment arm adding dasatinib to post-induction chemotherapy for Ph-like ALL patients with identified ABL class alterations. AALL1131 was closed to accrual in August 2019, and complete clinical results from this study are not yet available. The international phase 3 EsPhALL2017/COG AALL1631 phase 3 trial (NCT03007147), which is randomizing two different chemotherapy backbones in combination with imatinib for children with Ph+ ALL, will extend eligibility to include patients with Ph-like ABL class fusions in 2021. The St. Jude Children’s Research Hospital (SJCRH) Total Therapy XVII ALL (NCT03117751) protocol was activated in early 2017 and incorporates dasatinib beginning in induction therapy for those identified with an ABL class fusion by RNA-seq within 2 weeks of diagnosis (79). An MDACC phase 2 trial for adults with relapsed/refractory Ph-like ALL and ABL class fusions (NCT02420717) also combined dasatinib with the intensive hyper-CVAD chemotherapy backbone. Results have not yet been reported for these studies.

The largest class of Ph-like kinase-activating alterations constitutes those deregulating JAK/STAT signaling, making this pathway a major potential therapeutic vulnerability in Ph-like ALL, although JAK inhibitors have been less well studied in patients with ALL to date. Preclinical studies of engineered Ba/F3 cells and patient-derived xenograft (PDX) models harboring a diverse range of JAK/STAT pathway lesions (CRLF2 rearrangements with or without JAK mutations, JAK2 fusions, EPOR fusions, sequence mutations of IL7R and/or SH2B3) have shown in vitro and in vivo activity to different JAK inhibitors (7, 8, 29, 59, 73, 8084). Potent, but sometimes differential, preclinical activity of the JAK1/2 inhibitor ruxolitinib has been reported in Ph-like ALL models with CRLF2 fusions or JAK2 fusions, which may be influenced by the level of JAK pathway oncogene addiction or, potentially, by paradoxical JAK2 hyperphosphorylation with prolonged treatment (51, 80, 84). Subsequent preclinical studies further demonstrated enhanced activity of combinatorial treatment with JAK and PI3K/mTOR inhibitors in Ph-like ALL cell lines and PDX models (81, 82). Investigating dual pathway inhibition seems quite relevant for Ph-like ALL, as upregulation of alternative signaling pathways is a known mechanism of resistance to single-targeting agents (8587). To this end, a recent preclinical study showed that CRLF2-rearranged Ph-like ALL cells mediate a complex “BCR-like” signaling characterized by activated SRC family kinase and downstream signaling in the absence of surface μ-heavy chain expression, which may mediate resistance to ruxolitinib monotherapy, but could be overcome by multi-TKI and/or dexamethasone combination (87).

Currently, ruxolitinib is being assessed prospectively in several clinical trials for patients with JAK/STAT pathway-mutant Ph-like ALL. The COG AALL1521 phase 2 trial (NCT02723994) is investigating the efficacy of incorporating ruxolitinib with post-induction chemotherapy for children, adolescents, and young adults with HIGH-RISK Ph-like ALL harboring JAK/STAT pathway lesions (88). In this study, patients are stratified into 4 different cohorts based on their underlying Ph-like genetic lesions and by end-induction MRD status to delineate potential differential efficacy for each subset. A soon-to-open phase 1 trial will also assess the safety and tolerability of ruxolitinib addition to chemotherapy specifically in a Ph-like ALL AYA population ages 18–39 years (NCT03571321). The two previously discussed SJRCH and MDACC trials also have a ruxolitinib treatment arm in combination with chemotherapy for patients with de novo or relapsed JAK-mutant Ph-like ALL, respectively.

The enriched prevalence of IKZF1 deletions in Ph-like ALL opens up another potential therapeutic avenue for this HIGH-RISK patient population, although it is not yet known how these alterations might best be targeted. As above, IKZF1 alterations are known to mediate aberrant stromal adhesion and therapy resistance in murine models of Ph+ ALL, and it is plausible that such effects could be reversed by retinoic acid compounds or focal adhesion kinase (FAK) inhibitors when combined with TKIs (18, 19).

Targeted inhibitors of apoptotic proteins are promising therapeutic candidates for children and adults with relapsed/refractory acute leukemias and are under early-phase clinical investigation in pediatric trials. Venetoclax, an orally-administered selective inhibitor of the anti-apoptotic BCL-2 protein, has shown encouraging efficacy when combined with chemotherapy in 25 children with relapsed/refractory ALL with an overall response rate (ORR) of 56%, including 4 complete remission (CR), 4 CR with incomplete marrow recovery (CRi), and 1 CR without platelet recovery (CRp) (89). The combination of venetoclax with dasatinib or ponatinib appears highly synergistic in Ph+ ALL cell lines and PDX models with demonstrated inhibition of LYN signaling and prevention of downstream MCL-1 upregulation (90). A recent study demonstrated that dual BCL-2 and MCL-1 inhibition exhibits potent anti-leukemic activity in Ph+ ALL and CRLF2-rearranged Ph-like ALL PDX models (91), providing further rationale for potential investigation of BH3-mimetic inhibitors in clinical trials for patients with Ph+ and Ph-like ALL.

Hematopoietic stem cell transplantation

The role of hematopoietic stem cell transplantation (HSCT) in the care of patients with Ph-like ALL remains unclear in the TKI era (92). Earlier data demonstrated definitive improvement in EFS and OS of children with Ph+ ALL with imatinib or dasatinib addition to chemotherapy, which also eliminated need for HSCT in most patients (15, 69, 93, 94). Mirroring this Ph+ ALL experience, it is plausible that TKI addition to chemotherapy could be similarly successful for patients with Ph-like ABL-class ALL.

A single-center study recently reported comparable outcomes between children with Ph-like ALL and non-Ph-like ALL (5-year EFS 90.0% vs 88.4%, p=0.41, respectively) using MRD-directed therapy intensification for relevant patients (95). Consequently, a significant higher proportion of patients with Ph-like ALL underwent HSCT in first remission due to end-induction MRD positivity (95), which is known to occur in two-thirds of children with Ph-like ALL (55). These results demonstrate the therapeutic efficacy of HSCT in patients with Ph-like ALL and suggest that MRD is also an important outcome predictor in this patient population. Conversely, HSCT in first complete remission did not improve the EFS and OS of children with Ph-like ABL-class ALL, and HSCT-related mortality was particularly high (17%) (60). Another single-center study reported that adult patients with Ph-like ALL fare comparably poorly even when they achieve post-remission MRD negativity (median OS for MRD+ group 23.0 months vs MRD-group 26.2 months; p=0.318). Decisions for HSCT in first CR for adult patients may be challenged in the current era where pediatric-inspired chemotherapy regimens and access to frontline immunotherapy trials foster high hopes for inducing remission, deepening MRD response, and improving long-term survival (62, 9699).

Antibody-based and cellular immunotherapy

Major advances in immunotherapy during the past decade have revolutionized the landscape of relapsed leukemia therapy. The CD19xCD3 bispecific T-cell engager antibody blinatumomab, anti-CD22 antibody-drug conjugate inotuzumab ozogamicin, and CD19-redirected chimeric antigen receptor (CAR)-modified T-cell immunotherapy tisagenlecleucel have consecutively received FDA approval for patients with relapsed/refractory B-ALL based on several paradigm-shifting trials (98, 100102). Although the above trials did not specifically screen for the Ph-like ALL subtype, it is presumed that a reasonable proportion of these relapsed/refractory, heavily pretreated patients were Ph-like given their known high rates of chemoresistance and relapse. The randomized TOWER phase 3 trial (NCT02013167) showed that treatment with blinatumomab resulted in significantly higher remission rates and longer survival compared to standard chemotherapy amongst adults with relapsed/refractory Ph-negative B-ALL (98). Subsequent results from the BLAST phase 2 trial (NCT01207388) demonstrated that the majority (78%) of MRD-positive adult B-ALL patients in first or later CR achieved a complete MRD response following 1 cycle of blinatumomab, which was associated with better outcomes than MRD non-responders (100). Moreover, single-agent blinatumomab had strong anti-leukemic activity among adults with relapsed Ph+ ALL (103) and was associated with favorable treatment outcomes when compared to an external cohort receiving standard chemotherapy in a propensity score analysis (104). Favorable safety profiles and anti-leukemic activity with blinatumomab monotherapy were also observed in a heavily pre-treated relapsed/refractory pediatric B-ALL cohort (105, 106).

A recent retrospective analysis of 42 adults with R/R B-ALL who had available material for genomic analysis and were treated with blinatumomab monotherapy also showed that Ph-like ALL patients had a high response rate to blinatumomab (16/23; 70%), especially those with Ph-like ALL harboring CRLF2 rearrangements (12/16; 75%) and non-CRLF2 rearrangements (4/7; 57%) (107). Small cases series have also reported anecdotal safety and efficacy of combining blinatumomab and dasatinib in small numbers of patients with relapsed/refractory Ph+ ALL or Ph-like ABL-class ALL (108111). The SWOG S1318 phase 2 trial (NCT02143414) is currently comparing the efficacy of blinatumomab with combination chemotherapy versus blinatumomab and dasatinib in older adults with ABL-driven leukemias. The GIMEMA D-ALBA LAL2116 phase 2 trial (NCT02744768) is also currently assessing rates of molecular remission after 2 cycles of blinatumomab and dasatinib consolidation therapy in adult patients with newly-diagnosed Ph+ ALL. In an interim analysis with very early follow-up, 60% of study patients achieved a molecular response at the primary endpoint of approximately five months (three months of dasatinib and glucocorticoid chemotherapy followed by two cycles of blinatumomab). Additional improvement to 81% of molecular response after four cycles of blinatumomab was also reported with 1-year disease-free and overall survival of 88% and 95%, respectively, and 24 of the 63 enrolled patients received allogeneic HSCT (112). Longer-term follow-up is needed to determine if these early favorable outcomes will be sustained. Concerns have been raised with respect to potential antagonism of blinatumomab-dasatinib combination, as dasatinib has been shown to inhibit T cell function and could potentially abrogate the desired anti-leukemic activity of blinatumomab that requires endogenous T cell engagement (109, 113, 114). Correlative functional assays that comprehensively assess potential immunomodulatory effects of dasatinib upon immune cells will provide critical data for future trial design.

In a retrospective analysis of 53 adult patients with relapsed/refractory B-ALL treated with inotuzumab as salvage therapy, 12 patients identified as having the Ph-like subtype had an ORR of 58%, including 3 with CR (25%) and 4 with CR (33%) and partial hematologic recovery (CRh) (115). Five of the 7 (71%) Ph-like patients with inotuzumab-induced CR achieved MRD negativity (115). Inotuzumab at the FDA-approved fractionated adult dosing of 1.8 mg/m2 per cycle also induced impressive CR response rates among heavily-pretreated children with relapsed/refractory B-ALL (116, 117). In a retrospective study of 51 pediatric R/R B-ALL patients who received inotuzumab via a compassionate use program, the overall CR rate was 67%; 3 of 4 Ph-like ALL achieved CR/CRi, one of whom was MRD-negative (117).

Two recent studies showed that CD123 is more commonly expressed in adults with Ph+ ALL (118) and Ph-like ALL (119) compared to Ph-negative B-ALL and that CD123 may represent an additional therapeutic target for these high-risk leukemias. Anti-CD123 targeted immunotherapies (e.g., IMGN-632, flotetuzumab) have shown promising activity in preclinical models of acute myeloid leukemia (AML) (120122) and are under current clinical investigation in early-phase trials for patients with relapsed/refractory AML.

Finally, anecdotal reports of CD19 chimeric antigen receptor (CAR) T cell-induced remission in patients with relapsed/refractory Ph+ and Ph-like ALL have been reported (102). The current COG AALL1721 phase 2 trial (NCT03876769) assessing the efficacy of tisagenlecleucel in patients with newly-diagnosed HIGH-RISK B-ALL with end-consolidation MRD positivity excludes patients treated with kinase inhibitors, however. A planned phase 1 trial based upon promising preclinical data (123) will specifically investigate the clinical safety and preliminary efficacy of TSLPR-redirected CAR T cell immunotherapy in children, adolescents, and young adults with CRLF2-rearranged/overexpressing leukemias, including Ph-like ALL and Down Syndrome-associated ALL.

Conclusions and future perspective

Ph-like ALL is now known to be a prevalent subtype of B-ALL defined by its kinase-activated gene expression signature and associated genetic alterations. Children and adults with Ph-like ALL have historically experienced high relapse rates and inferior clinical outcomes despite best-available conventional chemotherapy, and their outcomes are currently far inferior to those of patients with Ph+ ALL with contemporary TKI-based therapy. Compelling evidence now exists from an extensive preclinical body of work for incorporation of relevant TKIs in combination with chemotherapy also for patients with Ph-like ALL, although results from clinical trials testing these strategies are not yet known. Although Ph-like ALL is thrice as common than Ph+ ALL in children and adolescents, its genetic heterogeneity with >70 fusions identified to date (6) represents a limiting factor in designing appropriately statistically-powered, randomized controlled trials to assess potential TKI efficacy within the major ABL class and CRLF2/JAK pathway-mutant subsets. As some patients with Ph-like ALL (particularly those with EBF1-PDGFRB or JAK2 fusions) are at high risk of induction chemotherapy failure, future efforts must focus upon swift identification of these genetic alterations and TKI addition early in induction therapy. Such strategy has proven successful in children with Ph+ ALL with superior CR rates and MRD negativity with TKI addition mid-induction versus at the beginning of consolidation (15, 69). Several immunotherapy modalities have now also demonstrated exciting efficacy in children with relapsed/refractory B-ALL. Ongoing and future clinical trials may help to elucidate the potential of such approaches (as monotherapy or combined with TKIs) more specifically in patients with Ph-like ALL.

International collaboration in designing the next generation of Ph-like ALL studies will expedite systematic study of novel treatment strategies that may improve clinical outcomes for this high risk patient population (124). Development and implementation of standardized clinical screening strategies among cooperative groups for rapid identification of patients with Ph-like ALL will also be essential for successful and efficient conduction of clinical trials. In parallel, future investigations in Ph-like ALL should also focus on investigating potential mechanisms of TKI resistance, as has been observed in patients with BCR-ABL1-driven chronic myeloid leukemia or Ph+ ALL with emergence of drug-resistant kinase domain mutations after long-term imatinib exposure (125129). Similar mutations have also been identified via in vitro saturation mutagenesis screens of Ph-like ALL with EBF1-PDGFRB fusions, and resistance mutations likely facilitating clinical relapse in patients with Ph-like ALL have now been reported (130, 131).

In summary, Ph-like ALL illustrates a paradigm of genomic discovery translation into targeted therapeutic approaches and presents an exciting opportunity for new precision medicine opportunities that aim to decrease relapse and improve long-term survival for patients with these high-risk leukemias across the age spectrum.

Acknowledgments

Dr Tasian receives/d research support from Incyte Corporation (Ph-like ALL studies) and Gilead Sciences (unrelated studies). She also serves on the scientific advisory boards of Aleta Biotherapeutics and Kura Oncology (unrelated studies).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures: Sarah Tasian: Consulting Fees: Aleta Biotherapeutics, Kura Oncology; Contracted Research: Incyte Corporation, Gilead Sciences

Disclosures: Thai Hoa Tran: No real or apparent conflicts of interest

Conflicts of interest: Dr Tran declares no conflicts of interest.

References

  • 1.Mullighan CG, Su X, Zhang J, Radtke I, Phillips LA, Miller CB, et al. Deletion of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med 2009;360(5):470–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Den Boer ML, van Slegtenhorst M, De Menezes RX, Cheok MH, Buijs-Gladdines JG, Peters ST, et al. A subtype of childhood acute lymphoblastic leukaemia with poor treatment outcome: a genome-wide classification study. Lancet Oncol 2009;10(2):125–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Boer JM, Marchante JR, Evans WE, Horstmann MA, Escherich G, Pieters R, et al. BCR-ABL1-like cases in pediatric acute lymphoblastic leukemia: a comparison between DCOG/Erasmus MC and COG/St. Jude signatures. Haematologica 2015;100(9):e354–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Pui CH, Roberts KG, Yang JJ, Mullighan CG. Philadelphia Chromosome-like Acute Lymphoblastic Leukemia. Clin Lymphoma Myeloma Leuk 2017;17(8):464–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Tran TH, Loh ML. Ph-like acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program 2016;2016(1):561–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Harvey RC, Tasian SK. Clinical diagnostics and treatment strategies for Philadelphia chromosome-like acute lymphoblastic leukemia. Blood Adv 2020;4(1):218–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Roberts KG, Li Y, Payne-Turner D, Harvey RC, Yang YL, Pei D, et al. Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med 2014;371(11):1005–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Roberts KG, Morin RD, Zhang J, Hirst M, Zhao Y, Su X, et al. Genetic alterations activating kinase and cytokine receptor signaling in high-risk acute lymphoblastic leukemia. Cancer Cell 2012;22(2):153–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.van der Veer A, Waanders E, Pieters R, Willemse ME, Van Reijmersdal SV, Russell LJ, 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):2622–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tasian SK, Loh ML, Hunger SP. Philadelphia chromosome-like acute lymphoblastic leukemia. Blood 2017;130(19):2064–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Roberts KG, Reshmi SC, Harvey RC, Chen IM, Patel K, Stonerock E, et al. Genomic and outcome analyses of Ph-like ALL in NCI standard-risk patients: a report from the Children’s Oncology Group. Blood 2018;132(8):815–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Russell LJ, Capasso M, Vater I, Akasaka T, Bernard OA, Calasanz MJ, et al. Deregulated expression of cytokine receptor gene, CRLF2, is involved in lymphoid transformation in B-cell precursor acute lymphoblastic leukemia. Blood 2009;114(13):2688–98. [DOI] [PubMed] [Google Scholar]
  • 13.Hertzberg L, Vendramini E, Ganmore I, Cazzaniga G, Schmitz M, Chalker J, 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):1006–17. [DOI] [PubMed] [Google Scholar]
  • 14.Mullighan CG, Miller CB, Radtke I, Phillips LA, Dalton J, Ma J, et al. BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros. Nature 2008;453(7191):110–4. [DOI] [PubMed] [Google Scholar]
  • 15.Slayton WB, Schultz KR, Kairalla JA, Devidas M, Mi X, Pulsipher MA, 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):2306–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.van der Veer A, Zaliova M, Mottadelli F, De Lorenzo P, Te Kronnie G, Harrison CJ, et al. IKZF1 status as a prognostic feature in BCR-ABL1-positive childhood ALL. Blood 2014;123(11):1691–8. [DOI] [PubMed] [Google Scholar]
  • 17.Martinelli G, Iacobucci I, Storlazzi CT, Vignetti M, Paoloni F, Cilloni D, et al. IKZF1 (Ikaros) deletions in BCR-ABL1-positive acute lymphoblastic leukemia are associated with short disease-free survival and high rate of cumulative incidence of relapse: a GIMEMA AL WP report. J Clin Oncol 2009;27(31):5202–7. [DOI] [PubMed] [Google Scholar]
  • 18.Churchman ML, Mullighan CG. Ikaros: Exploiting and targeting the hematopoietic stem cell niche in B-progenitor acute lymphoblastic leukemia. Exp Hematol 2017;46:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Churchman ML, Low J, Qu C, Paietta EM, Kasper LH, Chang Y, et al. Efficacy of Retinoids in IKZF1-Mutated BCR-ABL1 Acute Lymphoblastic Leukemia. Cancer Cell 2015;28(3):343–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Vairy S, Tran TH. IKZF1 alterations in acute lymphoblastic leukemia: the Good, the Bad and the Ugly. Blood Reviews 2020:Manuscript under revision [DOI] [PubMed] [Google Scholar]
  • 21.Tran TH, Harris MH, Nguyen JV, Blonquist TM, Stevenson KE, Stonerock E, 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):529–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Stanulla M, Dagdan E, Zaliova M, Moricke A, Palmi C, Cazzaniga G, et al. IKZF1(plus) 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):1240–9. [DOI] [PubMed] [Google Scholar]
  • 23.Steeghs EMP, Boer JM, Hoogkamer AQ, Boeree A, de Haas V, de Groot-Kruseman HA, 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zaliova M, Stuchly J, Winkowska L, Musilova A, Fiser K, Slamova M, et al. Genomic landscape of pediatric B-other acute lymphoblastic leukemia in a consecutive European cohort. Haematologica 2019;104(7):1396–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Fedullo AL, Messina M, Elia L, Piciocchi A, Gianfelici V, Lauretti A, et al. Prognostic implications of additional genomic lesions in adult Philadelphia chromosome-positive acute lymphoblastic leukemia. Haematologica 2019;104(2):312–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Perez-Andreu V, Roberts KG, Harvey RC, Yang W, Cheng C, Pei D, et al. Inherited GATA3 variants are associated with Ph-like childhood acute lymphoblastic leukemia and risk of relapse. Nat Genet 2013;45(12):1494–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Roberts KG, Gu Z, Payne-Turner D, McCastlain K, Harvey RC, Chen IM, et al. High Frequency and Poor Outcome of Philadelphia Chromosome-Like Acute Lymphoblastic Leukemia in Adults. J Clin Oncol 2017;35(4):394–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Tasian SK, Hurtz C, Wertheim GB, Bailey NG, Lim MS, Harvey RC, et al. High incidence of Philadelphia chromosome-like acute lymphoblastic leukemia in older adults with B-ALL. Leukemia 2017;31(4):981–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tasian SK, Doral MY, Borowitz MJ, Wood BL, Chen IM, Harvey RC, et al. Aberrant STAT5 and PI3K/mTOR pathway signaling occurs in human CRLF2-rearranged B-precursor acute lymphoblastic leukemia. Blood 2012;120(4):833–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Mullighan CG, Collins-Underwood JR, Phillips LA, Loudin MG, Liu W, Zhang J, et al. Rearrangement of CRLF2 in B-progenitor- and Down syndrome-associated acute lymphoblastic leukemia. Nat Genet 2009;41(11):1243–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Scheeren FA, van Lent AU, Nagasawa M, Weijer K, Spits H, Legrand N, et al. Thymic stromal lymphopoietin induces early human B-cell proliferation and differentiation. Eur J Immunol 2010;40(4):955–65. [DOI] [PubMed] [Google Scholar]
  • 32.Isaksen DE, Baumann H, Zhou B, Nivollet S, Farr AG, Levin SD, et al. Uncoupling of proliferation and Stat5 activation in thymic stromal lymphopoietin-mediated signal transduction. J Immunol 2002;168(7):3288–94. [DOI] [PubMed] [Google Scholar]
  • 33.Isaksen DE, Baumann H, Trobridge PA, Farr AG, Levin SD, Ziegler SF. Requirement for stat5 in thymic stromal lymphopoietin-mediated signal transduction. J Immunol 1999;163(11):5971–7. [PubMed] [Google Scholar]
  • 34.Herold T, Schneider S, Metzeler KH, Neumann M, Hartmann L, Roberts KG, et al. Adults with Philadelphia chromosome-like acute lymphoblastic leukemia frequently have IGH-CRLF2 and JAK2 mutations, persistence of minimal residual disease and poor prognosis. Haematologica 2017;102(1):130–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Moorman AV, Schwab C, Ensor HM, Russell LJ, Morrison H, Jones L, et al. IGH@ translocations, CRLF2 deregulation, and microdeletions in adolescents and adults with acute lymphoblastic leukemia. J Clin Oncol 2012;30(25):3100–8. [DOI] [PubMed] [Google Scholar]
  • 36.Yoda A, Yoda Y, Chiaretti S, Bar-Natan M, Mani K, Rodig SJ, et al. Functional screening identifies CRLF2 in precursor B-cell acute lymphoblastic leukemia. Proc Natl Acad Sci U S A 2010;107(1):252–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Reshmi SC, Harvey RC, Roberts KG, Stonerock E, Smith A, Jenkins H, et al. Targetable kinase gene fusions in high risk B-ALL: a study from the Children’s Oncology Group. Blood 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Tasian SK, Dai Y, Devidas M, Roberts KG, Harvey RC, Chen IL, Carroll AJ, Heerema NA,Reshmi SC, Gastier-Foster J, Borowitz MJ, Wood B, Mullighan CM, Willman CL, Maloney KW, Larsen EC, Angiolillo AL, Schore RS, Burke MJ, Salzer WL, Winick NJ, Carroll WL, Hunger SP, Raetz EA, Rabin KR, and Loh ML Outcomes of Patients with CRLF2-Overexpressing Acute Lymphoblastic Leukemia without Down Syndrome: A Report from the Children’s Oncology Group. Blood 2020:1095. [Google Scholar]
  • 39.Roberts KG, Mullighan CG. Genomics in acute lymphoblastic leukaemia: insights and treatment implications. Nat Rev Clin Oncol 2015;12(6):344–57. [DOI] [PubMed] [Google Scholar]
  • 40.Iacobucci I, Li Y, Roberts KG, Dobson SM, Kim JC, Payne-Turner D, et al. Truncating Erythropoietin Receptor Rearrangements in Acute Lymphoblastic Leukemia. Cancer Cell 2016;29(2):186–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Jabbour E, Short NJ, Ravandi F, Huang X, Daver N, DiNardo CD, et al. Combination of hyper-CVAD with ponatinib as first-line therapy for patients with Philadelphia chromosome-positive acute lymphoblastic leukaemia: long-term follow-up of a single-centre, phase 2 study. Lancet Haematol 2018;5(12):e618–e27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wiemels JL, Kang M, Chang JS, Zheng L, Kouyoumji C, Zhang L, et al. Backtracking RAS mutations in high hyperdiploid childhood acute lymphoblastic leukemia. Blood Cells Mol Dis 2010;45(3):186–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Holmfeldt L, Wei L, Diaz-Flores E, Walsh M, Zhang J, Ding L, et al. The genomic landscape of hypodiploid acute lymphoblastic leukemia. Nat Genet 2013;45(3):242–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Irving J, Matheson E, Minto L, Blair H, Case M, Halsey C, et al. Ras pathway mutations are prevalent in relapsed childhood acute lymphoblastic leukemia and confer sensitivity to MEK inhibition. Blood 2014;124(23):3420–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Roberts KG, Janke LJ, Zhao Y, Seth A, Ma J, Finkelstein D, et al. ETV6-NTRK3 induces aggressive acute lymphoblastic leukemia highly sensitive to selective TRK inhibition. Blood 2018;132(8):861–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Nardi V, Ku N, Frigault MJ, Dubuc AM, Tsai HK, Amrein PC, et al. Clinical response to larotrectinib in adult Philadelphia chromosome-like ALL with cryptic ETV6-NTRK3 rearrangement. Blood Adv 2020;4(1):106–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Schewe DM, Lenk L, Vogiatzi F, Winterberg D, Rademacher AV, Buchmann S, et al. Larotrectinib in TRK fusion-positive pediatric B-cell acute lymphoblastic leukemia. Blood Adv 2019;3(22):3499–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Brown LM, Bartolo RC, Davidson NM, Schmidt B, Brooks I, Challis J, et al. Targeted therapy and disease monitoring in CNTRL-FGFR1-driven leukaemia. Pediatr Blood Cancer 2019;66(10):e27897. [DOI] [PubMed] [Google Scholar]
  • 49.Helsten T, Schwaederle M, Kurzrock R. Fibroblast growth factor receptor signaling in hereditary and neoplastic disease: biologic and clinical implications. Cancer Metastasis Rev 2015;34(3):479–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Owattanapanich W, Rujirachun P, Ungprasert P, Buaboonnam J, Techavichit P. Prevalence and Clinical Outcome of Philadelphia-Like Acute Lymphoblastic Leukemia: Systematic Review and Meta-analysis. Clin Lymphoma Myeloma Leuk 2019. [DOI] [PubMed] [Google Scholar]
  • 51.Jain N, Roberts KG, Jabbour E, Patel K, Eterovic AK, Chen K, et al. Ph-like acute lymphoblastic leukemia: a high-risk subtype in adults. Blood 2017;129(5):572–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Harvey RC, Mullighan CG, Chen IM, Wharton W, Mikhail FM, Carroll AJ, et al. Rearrangement of CRLF2 is associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/Latino ethnicity, and a poor outcome in pediatric B-progenitor acute lymphoblastic leukemia. Blood 2010;115(26):5312–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Madzio J, Pastorczak A, Sedek L, Braun M, Taha J, Wypyszczak K, et al. GATA3 germline variant is associated with CRLF2 expression and predicts outcome in pediatric B-cell precursor acute lymphoblastic leukemia. Genes Chromosomes Cancer 2019;58(9):619–26. [DOI] [PubMed] [Google Scholar]
  • 54.Boer JM, Koenders JE, van der Holt B, Exalto C, Sanders MA, Cornelissen JJ, et al. Expression profiling of adult acute lymphoblastic leukemia identifies a BCR-ABL1-like subgroup characterized by high non-response and relapse rates. Haematologica 2015;100(7):e261–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Loh ML, Zhang J, Harvey RC, Roberts K, Payne-Turner D, Kang H, et al. Tyrosine kinome sequencing of pediatric acute lymphoblastic leukemia: a report from the Children’s Oncology Group TARGET Project. Blood 2013;121(3):485–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Weston BW, Hayden MA, Roberts KG, Bowyer S, Hsu J, Fedoriw G, et al. Tyrosine kinase inhibitor therapy induces remission in a patient with refractory EBF1-PDGFRB-positive acute lymphoblastic leukemia. J Clin Oncol 2013;31(25):e413–6. [DOI] [PubMed] [Google Scholar]
  • 57.Lengline E, Beldjord K, Dombret H, Soulier J, Boissel N, Clappier E. Successful tyrosine kinase inhibitor therapy in a refractory B-cell precursor acute lymphoblastic leukemia with EBF1-PDGFRB fusion. Haematologica 2013;98(11):e146–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Schwab C, Ryan SL, Chilton L, Elliott A, Murray J, Richardson S, et al. EBF1-PDGFRB fusion in pediatric B-cell precursor acute lymphoblastic leukemia (BCP-ALL): genetic profile and clinical implications. Blood 2016;127(18):2214–8. [DOI] [PubMed] [Google Scholar]
  • 59.Ding YY, Stern JW, Jubelirer TF, Wertheim GB, Li F, Chang F, et al. Clinical efficacy of ruxolitinib and chemotherapy in a child with Philadelphia chromosome-like acute lymphoblastic leukemia with GOLGA5-JAK2 fusion and induction failure. Haematologica 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.den Boer ML, Cario G, Moorman AV, Boer JM, de Groot-Kruseman HA, Fiocco M, et al. Outcomes of paediatric patients with B-cell acute lymphocytic leukaemia with ABL-class fusion in the pre-tyrosine-kinase inhibitor era: a multicentre, retrospective, cohort study. Lancet Haematol 2021;8(1):e55–e66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Maloney KW, Devidas M, Wang C, Mattano LA, Friedmann AM, Buckley P, et al. Outcome in Children With Standard-Risk B-Cell Acute Lymphoblastic Leukemia: Results of Children’s Oncology Group Trial AALL0331. J Clin Oncol 2020;38(6):602–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Stock W, Luger SM, Advani AS, Yin J, Harvey RC, Mullighan CG, et al. A pediatric regimen for older adolescents and young adults with acute lymphoblastic leukemia: results of CALGB 10403. Blood 2019;133(14):1548–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chiaretti S, Messina M, Della Starza I, Piciocchi A, Cafforio L, Cavalli M, et al. Philadelphia-like acute lymphoblastic leukemia is associated with minimal residual disease persistence and poor outcome. First report of the minimal residual disease-oriented GIMEMA LAL1913. Haematologica 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kang H, Chen IM, Wilson CS, Bedrick EJ, Harvey RC, Atlas SR, et al. Gene expression classifiers for relapse-free survival and minimal residual disease improve risk classification and outcome prediction in pediatric B-precursor acute lymphoblastic leukemia. Blood 2010;115(7):1394–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Harvey RC, Tasian SK. Clinical Diagnostics and Treatment Strategies for Philadelphia Chromosome-like Acute Lymphoblastic Leukemia. Blood Advances 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Surrey LF, MacFarland SP, Chang F, Cao K, Rathi KS, Akgumus GT, et al. Clinical utility of custom-designed NGS panel testing in pediatric tumors. Genome Med 2019;11(1):32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chang F, Lin F, Cao K, Surrey LF, Aplenc R, Bagatell R, et al. Development and Clinical Validation of a Large Fusion Gene Panel for Pediatric Cancers. J Mol Diagn 2019;21(5):873–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.He J, Abdel-Wahab O, Nahas MK, Wang K, Rampal RK, Intlekofer AM, et al. Integrated genomic DNA/RNA profiling of hematologic malignancies in the clinical setting. Blood 2016;127(24):3004–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Schultz KR, Carroll A, Heerema NA, Bowman WP, Aledo A, Slayton WB, 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):1467–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wassmann B, Pfeifer H, Goekbuget N, Beelen DW, Beck J, Stelljes M, et al. Alternating versus concurrent schedules of imatinib and chemotherapy as front-line therapy for Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL). Blood 2006;108(5):1469–77. [DOI] [PubMed] [Google Scholar]
  • 71.Shen S, Chen X, Cai J, Yu J, Gao J, Hu S, et al. Effect of Dasatinib vs Imatinib in the Treatment of Pediatric Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia: A Randomized Clinical Trial. JAMA Oncol 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Lilly MB, Ottmann OG, Shah NP, Larson RA, Reiffers JJ, Ehninger G, et al. Dasatinib 140 mg once daily versus 70 mg twice daily in patients with Ph-positive acute lymphoblastic leukemia who failed imatinib: Results from a phase 3 study. Am J Hematol 2010;85(3):164–70. [DOI] [PubMed] [Google Scholar]
  • 73.Roberts KG, Yang YL, Payne-Turner D, Lin W, Files JK, Dickerson K, et al. Oncogenic role and therapeutic targeting of ABL-class and JAK-STAT activating kinase alterations in Ph-like ALL. Blood Adv 2017;1(20):1657–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kobayashi K, Miyagawa N, Mitsui K, Matsuoka M, Kojima Y, Takahashi H, et al. TKI dasatinib monotherapy for a patient with Ph-like ALL bearing ATF7IP/PDGFRB translocation. Pediatr Blood Cancer 2015;62(6):1058–60. [DOI] [PubMed] [Google Scholar]
  • 75.Fazio F, Barberi W, Cazzaniga G, Fazio G, Messina M, Della Starza I, et al. Efficacy of imatinib and chemotherapy in a pediatric patient with Philadelphia-like acute lymphoblastic leukemia with Ebf1-Pdgfrb fusion transcript. Leuk Lymphoma 2019:1–4. [DOI] [PubMed] [Google Scholar]
  • 76.Cario G, Leoni V, Conter V, Attarbaschi A, Zaliova M, Sramkova L, et al. Relapses and treatment-related events contributed equally to poor prognosis in children with ABL-class fusion positive B-cell acute lymphoblastic leukemia treated according to AIEOP-BFM protocols. Haematologica 2020;105(7):1887–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Tanasi I, Ba I, Sirvent N, Braun T, Cuccuini W, Ballerini P, et al. Efficacy of tyrosine kinase inhibitors in Ph-like acute lymphoblastic leukemia harboring ABL-class rearrangements. Blood 2019;134(16):1351–5. [DOI] [PubMed] [Google Scholar]
  • 78.Moorman AV, Schwab C, Winterman E, Hancock J, Castleton A, Cummins M, et al. Adjuvant tyrosine kinase inhibitor therapy improves outcome for children and adolescents with acute lymphoblastic leukaemia who have an ABL-class fusion. Br J Haematol 2020. [DOI] [PubMed] [Google Scholar]
  • 79.Inaba H, Azzato EM, Mullighan CG. Integration of Next-Generation Sequencing to Treat Acute Lymphoblastic Leukemia with Targetable Lesions: The St. Jude Children’s Research Hospital Approach. Front Pediatr 2017;5:258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Maude SL, Tasian SK, Vincent T, Hall JW, Sheen C, Roberts KG, et al. Targeting JAK1/2 and mTOR in murine xenograft models of Ph-like acute lymphoblastic leukemia. Blood 2012;120(17):3510–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Tasian SK, Teachey DT, Li Y, Shen F, Harvey RC, Chen IM, et al. Potent efficacy of combined PI3K/mTOR and JAK or ABL inhibition in murine xenograft models of Ph-like acute lymphoblastic leukemia. Blood 2017;129(2):177–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Zhang Q, Shi C, Han L, Jain N, Roberts KG, Ma H, et al. Inhibition of mTORC1/C2 signaling improves anti-leukemia efficacy of JAK/STAT blockade in CRLF2 rearranged and/or JAK driven Philadelphia chromosome-like acute B-cell lymphoblastic leukemia. Oncotarget 2018;9(8):8027–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Khan M, Siddiqi R, Tran TH. Philadelphia chromosome-like acute lymphoblastic leukemia: A review of the genetic basis, clinical features, and therapeutic options. Semin Hematol 2018;55(4):235–41. [DOI] [PubMed] [Google Scholar]
  • 84.Wu SC, Li LS, Kopp N, Montero J, Chapuy B, Yoda A, et al. Activity of the Type II JAK2 Inhibitor CHZ868 in B Cell Acute Lymphoblastic Leukemia. Cancer Cell 2015;28(1):29–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Hurtz C, Tasian SK, Wertheim G, Astles R, Zebrowski A, Perl AE, et al. Adaptive Reactivation of Signaling Pathways As a Novel Mechanism of Resistance to JAK Inhibitors in Ph-like ALL. Blood 2016;128(22):755-. [Google Scholar]
  • 86.Gotesman M, Vo T-TT, Mallya S, Zhang Q, Shi C, Müschen M, et al. mTOR Kinase Inhibitors Enhance Efficacy of TKIs in Preclinical Models of Ph-like B-ALL. Blood 2016;128(22):2763-. [Google Scholar]
  • 87.Hurtz C, Wertheim GB, Loftus JP, Blumenthal D, Lehman A, Li Y, et al. Oncogene-independent BCR-like signaling adaptation confers drug resistance in Ph-like ALL. J Clin Invest 2020;130(7):3637–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Tasian SK, Assad A, Hunter DS, Du Y, Loh ML. A Phase 2 Study of Ruxolitinib with Chemotherapy in Children with Philadelphia Chromosome-like Acute Lymphoblastic Leukemia (INCB18424–269/AALL1521): Dose-Finding Results from the Part 1 Safety Phase. Blood 2018;132(Supplement 1):555.30093383 [Google Scholar]
  • 89.Place AE, Karol SE, Forlenza CJ, Gambart M, Cooper TM, Fraser C, et al. Pediatric Patients with Relapsed/Refractory Acute Lymphoblastic Leukemia Harboring Heterogeneous Genomic Profiles Respond to Venetoclax in Combination with Chemotherapy. Blood 2020:2793. [Google Scholar]
  • 90.Leonard JT, Rowley JS, Eide CA, Traer E, Hayes-Lattin B, Loriaux M, et al. Targeting BCL-2 and ABL/LYN in Philadelphia chromosome-positive acute lymphoblastic leukemia. Sci Transl Med 2016;8(354):354ra114. [DOI] [PubMed] [Google Scholar]
  • 91.Moujalled DM, Hanna DT, Hediyeh-Zadeh S, Pomilio G, Brown L, Litalien V, et al. Cotargeting BCL-2 and MCL-1 in high-risk B-ALL. Blood Adv 2020;4(12):2762–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.El Fakih R, Savani B, Mohty M, Aljurf M. Hematopoietic Cell Transplant Consideration for Philadelphia Chromosome-Like Acute Lymphoblastic Leukemia Patients. Biol Blood Marrow Transplant 2019. [DOI] [PubMed] [Google Scholar]
  • 93.Schultz KR, Bowman WP, Aledo A, Slayton WB, Sather H, Devidas M, 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):5175–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Biondi A, Schrappe M, De Lorenzo P, Castor A, Lucchini G, Gandemer V, et al. Imatinib after induction for treatment of children and adolescents with Philadelphia-chromosome-positive acute lymphoblastic leukaemia (EsPhALL): a randomised, open-label, intergroup study. Lancet Oncol 2012;13(9):936–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Roberts KG, Pei D, Campana D, Payne-Turner D, Li Y, Cheng C, 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;32(27):3012–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Siegel SE, Stock W, Johnson RH, Advani A, Muffly L, Douer D, et al. Pediatric-Inspired Treatment Regimens for Adolescents and Young Adults With Philadelphia Chromosome-Negative Acute Lymphoblastic Leukemia: A Review. JAMA Oncol 2018;4(5):725–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Kantarjian H, Ravandi F, Short NJ, Huang X, Jain N, Sasaki K, et al. Inotuzumab ozogamicin in combination with low-intensity chemotherapy for older patients with Philadelphia chromosome-negative acute lymphoblastic leukaemia: a single-arm, phase 2 study. Lancet Oncol 2018;19(2):240–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kantarjian H, Stein A, Gokbuget N, Fielding AK, Schuh AC, Ribera JM, et al. Blinatumomab versus Chemotherapy for Advanced Acute Lymphoblastic Leukemia. N Engl J Med 2017;376(9):836–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.DeFilipp Z, Advani AS, Bachanova V, Cassaday RD, Deangelo DJ, Kebriaei P, et al. Hematopoietic Cell Transplantation in the Treatment of Adult Acute Lymphoblastic Leukemia: Updated 2019 Evidence-Based Review from the American Society for Transplantation and Cellular Therapy. Biol Blood Marrow Transplant 2019;25(11):2113–23. [DOI] [PubMed] [Google Scholar]
  • 100.Gokbuget N, Dombret H, Bonifacio M, Reichle A, Graux C, Faul C, et al. Blinatumomab for minimal residual disease in adults with B-cell precursor acute lymphoblastic leukemia. Blood 2018;131(14):1522–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Kantarjian HM, DeAngelo DJ, Stelljes M, Martinelli G, Liedtke M, Stock W, et al. Inotuzumab Ozogamicin versus Standard Therapy for Acute Lymphoblastic Leukemia. N Engl J Med 2016;375(8):740–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, et al. Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med 2018;378(5):439–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Martinelli G, Boissel N, Chevallier P, Ottmann O, Gokbuget N, Topp MS, et al. Complete Hematologic and Molecular Response in Adult Patients With Relapsed/Refractory Philadelphia Chromosome-Positive B-Precursor Acute Lymphoblastic Leukemia Following Treatment With Blinatumomab: Results From a Phase II, Single-Arm, Multicenter Study. J Clin Oncol 2017;35(16):1795–802. [DOI] [PubMed] [Google Scholar]
  • 104.Rambaldi A, Ribera JM, Kantarjian HM, Dombret H, Ottmann OG, Stein AS, et al. Blinatumomab compared with standard of care for the treatment of adult patients with relapsed/refractory Philadelphia chromosome-positive B-precursor acute lymphoblastic leukemia. Cancer 2020;126(2):304–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.von Stackelberg A, Locatelli F, Zugmaier G, Handgretinger R, Trippett TM, Rizzari C, et al. Phase I/Phase II Study of Blinatumomab in Pediatric Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia. J Clin Oncol 2016;34(36):4381–9. [DOI] [PubMed] [Google Scholar]
  • 106.Gore L, Locatelli F, Zugmaier G, Handgretinger R, O’Brien MM, Bader P, et al. Survival after blinatumomab treatment in pediatric patients with relapsed/refractory B-cell precursor acute lymphoblastic leukemia. Blood Cancer J 2018;8(9):80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Zhao Y, Aldoss I, Qu C, Crawford JC, Gu Z, Allen EK, et al. Tumor intrinsic and extrinsic determinants of response to blinatumomab in adults with B-ALL. Blood 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Assi R, Kantarjian H, Short NJ, Daver N, Takahashi K, Garcia-Manero G, et al. Safety and Efficacy of Blinatumomab in Combination With a Tyrosine Kinase Inhibitor for the Treatment of Relapsed Philadelphia Chromosome-positive Leukemia. Clin Lymphoma Myeloma Leuk 2017;17(12):897–901. [DOI] [PubMed] [Google Scholar]
  • 109.King AC, Pappacena JJ, Tallman MS, Park JH, Geyer MB. Blinatumomab administered concurrently with oral tyrosine kinase inhibitor therapy is a well-tolerated consolidation strategy and eradicates measurable residual disease in adults with Philadelphia chromosome positive acute lymphoblastic leukemia. Leuk Res 2019;79:27–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Tanasi I, Ba I, Sirvent N, Braun T, Cuccuini W, Ballerini P, et al. Efficacy of Tyrosine Kinase Inhibitors in Ph-like Acute Lymphoblastic Leukemia harboring ABL-class Rearrangements. Blood 2019. [DOI] [PubMed] [Google Scholar]
  • 111.Petruzziello F, Giagnuolo G, Cazzaniga G, Beneduce G, Locatelli F, Stellato P, Fazio G, Mirabelli G, Menna G, & Parasole R Successful of Chemo-Free Treatment with Dasatinib and Blinatumomab in a Pediatric EBF1-PDGFRβ Positive Acute Lymphoblastic Leukemia. Blood 2018;132(Suppl 1):5213. [Google Scholar]
  • 112.Foa R, Bassan R, Vitale A, Elia L, Piciocchi A, Puzzolo MC, et al. Dasatinib-Blinatumomab for Ph-Positive Acute Lymphoblastic Leukemia in Adults. N Engl J Med 2020;383(17):1613–23. [DOI] [PubMed] [Google Scholar]
  • 113.Leonard J, Kosaka Y, Malla P, LaTocha D, Lamble A, Hayes-Lattin B, et al. Concomitant use of a dual ABL/Src kinase inhibitor eliminates the in vitroefficacy of blinatumomab against Ph+ ALL. Blood 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Weber EW, Lynn RC, Sotillo E, Lattin J, Xu P, Mackall CL. Pharmacologic control of CAR-T cell function using dasatinib. Blood Adv 2019;3(5):711–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Jabbour E Inotuzumab Ozogamicin May Overcome the Impact of Philadelphia Chromosome (Ph)-like Phenotype in Adult Patients with Relapsed/Refractory ALL. Blood 2019. [Google Scholar]
  • 116.O’Brien MM, Ji L, Shah NN, Rheingold SR, Bhojwani D, Yi JS, et al. A Phase 2 Trial of Inotuzumab Ozogamicin (InO) in Children and Young Adults with Relapsed or Refractory (R/R) CD22+ B-Acute Lymphoblastic Leukemia (B-ALL): Results from Children’s Oncology Group Protocol AALL1621. Blood 2019;134(Supplement 1):741.31243041 [Google Scholar]
  • 117.Bhojwani D, Sposto R, Shah NN, Rodriguez V, Yuan C, Stetler-Stevenson M, et al. Inotuzumab ozogamicin in pediatric patients with relapsed/refractory acute lymphoblastic leukemia. Leukemia 2019;33(4):884–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Angelova E, Audette C, Kovtun Y, Daver N, Wang SA, Pierce S, et al. CD123 expression patterns and selective targeting with a CD123-targeted antibody-drug conjugate (IMGN632) in acute lymphoblastic leukemia. Haematologica 2019;104(4):749–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Lyapichev KA, Sukswai N, Angelova E, Kersh MJ, Pierce S, Konopleva M, et al. CD123 Expression in Philadelphia Chromosome-like B Acute Lymphoblastic Leukemia/Lymphoma. Clin Lymphoma Myeloma Leuk 2020. [DOI] [PubMed] [Google Scholar]
  • 120.Han L, Jorgensen JL, Brooks C, Shi C, Zhang Q, Nogueras Gonzalez GM, et al. Antileukemia Efficacy and Mechanisms of Action of SL-101, a Novel Anti-CD123 Antibody Conjugate, in Acute Myeloid Leukemia. Clin Cancer Res 2017;23(13):3385–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kovtun Y, Jones GE, Adams S, Harvey L, Audette CA, Wilhelm A, et al. A CD123-targeting antibody-drug conjugate, IMGN632, designed to eradicate AML while sparing normal bone marrow cells. Blood Adv 2018;2(8):848–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Chichili GR, Huang L, Li H, Burke S, He L, Tang Q, et al. A CD3xCD123 bispecific DART for redirecting host T cells to myelogenous leukemia: preclinical activity and safety in nonhuman primates. Sci Transl Med 2015;7(289):289ra82. [DOI] [PubMed] [Google Scholar]
  • 123.Qin H, Cho M, Haso W, Zhang L, Tasian SK, Oo HZ, et al. Eradication of B-ALL using chimeric antigen receptor-expressing T cells targeting the TSLPR oncoprotein. Blood 2015;126(5):629–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Pui CH, Yang JJ, Hunger SP, Pieters R, Schrappe M, Biondi A, et al. Childhood Acute Lymphoblastic Leukemia: Progress Through Collaboration. J Clin Oncol 2015;33(27):2938–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Shah NP, Sawyers CL. Mechanisms of resistance to STI571 in Philadelphia chromosome-associated leukemias. Oncogene 2003;22(47):7389–95. [DOI] [PubMed] [Google Scholar]
  • 126.Soverini S, Branford S, Nicolini FE, Talpaz M, Deininger MW, Martinelli G, et al. Implications of BCR-ABL1 kinase domain-mediated resistance in chronic myeloid leukemia. Leuk Res 2014;38(1):10–20. [DOI] [PubMed] [Google Scholar]
  • 127.Soverini S, Hochhaus A, Nicolini FE, Gruber F, Lange T, Saglio G, et al. BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet. Blood 2011;118(5):1208–15. [DOI] [PubMed] [Google Scholar]
  • 128.Soverini S, Colarossi S, Gnani A, Rosti G, Castagnetti F, Poerio A, et al. Contribution of ABL kinase domain mutations to imatinib resistance in different subsets of Philadelphia-positive patients: by the GIMEMA Working Party on Chronic Myeloid Leukemia. Clin Cancer Res 2006;12(24):7374–9. [DOI] [PubMed] [Google Scholar]
  • 129.Druker BJ. Circumventing resistance to kinase-inhibitor therapy. N Engl J Med 2006;354(24):2594–6. [DOI] [PubMed] [Google Scholar]
  • 130.Yeung DT, Moulton DJ, Heatley SL, Nievergall E, Dang P, Braley J, et al. Relapse of BCR-ABL1-like ALL mediated by the ABL1 kinase domain mutation T315I following initial response to dasatinib treatment. Leukemia 2015;29(1):230–2. [DOI] [PubMed] [Google Scholar]
  • 131.Zhang Y, Gao Y, Zhang H, Zhang J, He F, Hnizda A, et al. PDGFRB mutation and tyrosine kinase inhibitor resistance in Ph-like acute lymphoblastic leukemia. Blood 2018;131(20):2256–61. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES