Next Article in Journal
Revealing the Roles of Keratin 8/18-Associated Signaling Proteins Involved in the Development of Hepatocellular Carcinoma
Next Article in Special Issue
Keratin Profiling by Single-Cell RNA-Sequencing Identifies Human Prostate Stem Cell Lineage Hierarchy and Cancer Stem-Like Cells
Previous Article in Journal
Novel TTLL5 Variants Associated with Cone-Rod Dystrophy and Early-Onset Severe Retinal Dystrophy
Previous Article in Special Issue
The Role of lncRNAs in the Stem Phenotype of Pancreatic Ductal Adenocarcinoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genomic Analyses of Pediatric Acute Lymphoblastic Leukemia Ph+ and Ph-Like—Recent Progress in Treatment

by
Agnieszka Kaczmarska
1,
Patrycja Śliwa
1,
Joanna Zawitkowska
2,† and
Monika Lejman
3,*,†
1
Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
2
Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
3
Laboratory of Genetic Diagnostics, II Chair of Paediatrics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
*
Author to whom correspondence should be addressed.
These authors contributed equally as senior and supervisor authors.
Int. J. Mol. Sci. 2021, 22(12), 6411; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126411
Submission received: 29 April 2021 / Revised: 4 June 2021 / Accepted: 14 June 2021 / Published: 15 June 2021
(This article belongs to the Special Issue Cancer Stem Cells in Basic Science and in Translational Oncology)

Abstract

:
Pediatric acute lymphoblastic leukemia (ALL) with t(9;22)(q34;q11.2) is a very rare malignancy in children. Approximately 3–5% of pediatric ALL patients present with the Philadelphia chromosome. Previously, children with Ph+ had a poor prognosis, and were considered for allogeneic stem cell transplantation (allo-HSCT) in their first remission (CR1). Over the last few years, the treatment of childhood ALL has significantly improved due to standardized research protocols. Hematopoietic stem cell transplantation (HSCT) has been the gold standard therapy in ALL Ph+ patients, but recently first-generation tyrosine kinase inhibitor (TKI)-imatinib became a major milestone in increasing overall survival. Genomic analyses give the opportunity for the investigation of new fusions or mutations, which can be used to establish effective targeted therapies. Alterations of the IKZF1 gene are present in a large proportion of pediatric and adult ALL Ph+ cases. IKZF1 deletions are present in ~15% of patients without BCR-ABL1 rearrangements. In BCR-ABL1-negative cases, IKZF1 deletions have been shown to have an independent prognostic impact, carrying a three-fold increased risk of treatment failure. The prognostic significance of IKZF1 gene aberrations in pediatric ALL Ph+ is still under investigation. More research should focus on targeted therapies and immunotherapy, which is not associated with serious toxicity in the same way as classic chemotherapy, and on the improvement of patient outcomes. In this review, we provide a molecular analysis of childhood ALL with t(9;22)(q34;q11.2), including the Ph-like subtype, and of treatment strategies.

1. Introduction

Acute lymphoblastic leukemia (ALL) is the most common malignancy diagnosed in children according to American Cancer Society data, accounting for 26% of all cancers in children aged 1–14 and 8% in teenagers aged 15–19 in the USA [1]. Over the last few years, the treatment of childhood ALL has improved significantly due to standardized research protocols. The cure rate has increased dramatically and is now about 90% in many developed countries. Identification of chromosomal abnormalities and translocations is important to establish treatment protocols and predict the clinical outcome [2].
The Philadelphia chromosome (Ph), arising from a reciprocal translocation between chromosomes 9 and 22 t(9;22)(q34;q11.2), is present in 3% to 5% of children and 25% of adults with ALL [3]. The normal BCR gene encodes a 160-kD phosphoprotein associated with serine/threonine kinase activity. ABL1 is a proto-oncogene that encodes a cytoplasmatic and nuclear protein tyrosine kinase involved in a variety of cellular processes, including cell adhesion, cell division, cell differentiation, and the stress response [4]. BCR/ABL1 has a cytoplasmic localization and may have a carcinogenetic role, in contrast with ABL1, which has a mostly nuclear localization. The hybrid protein has an increased protein tyrosine kinase (TK) activity compared to ABL1. These activated kinases disturb downstream signaling pathways, causing enhanced proliferation, differentiation arrest, and resistance to cell apoptosis. The crucial event lies on der(22); the 5′ BCR/3′ ABL hybrid gene is pathogenic, while ABL/BCR may or may not be expressed. Expression of the fusion gene in ALL results in two types of chimeric mRNAs, dependent upon the position of the breakpoint within the breakpoint cluster region of the BCR gene. In a third of patients with ALL and the majority of those with CML, the breakpoint occurs within a 5.8 kb region spanning BCR exons 12–16 (exons b1–b5), known as the major bcr (M-bcr). The translocation involving this breakpoint results in the production of a p210 BCR–ABL protein. In the remaining two thirds of ALL patients and rarely in CML, the breakpoint arises further upstream, between exons e2′ and e2, known as the minor bcr (m-bcr). The translocation involving this breakpoint produces a p190 protein. The p190 group (typical for patients with ALL Ph+) tends to show an early response and complete remission at the end of the induction phase, which indicates that patients carrying a p190 transcript have a more favorable prognosis than the p210 group [5].
Historically, the outcome for children with ALL Ph+ was an exceptionally poor prognosis, and all children were classified into the high-risk group. According to the EsPhALL2010 protocol, the overall survival after treatment was 71.8% and event-free survival (EFS) was 57.0% at the five years follow-up. Although current treatment strategies are becoming increasingly successful, the most frequent cause of treatment failure is relapse. Cumulative relapse risk is 9.3–20.6%, and the outcome prognosis after a relapse is poor [6,7,8]. Several studies have clearly demonstrated that minimal residual disease (MRD) also has a crucial predictive role in ALL Ph+ [9,10,11,12]. Moreover, not only the presence of BCR/ABL1 fusion, but also the age at diagnosis and leukocyte count are adverse prognostic factors associated with a high risk of therapeutic failure [13]. Unfortunately, due to the small number of pediatric ALL Ph+ cases, randomized trials focusing on treatment are still lacking. Genomic analyses give opportunities for the detection of new mutations, which can be used to establish effective targeted therapies [14]. The use of high-throughput analytical techniques has allowed the description of novel high-risk ALL subtypes. One such novel subtype, Ph-like acute lymphoblastic leukemia (Ph-like ALL), is characterized by a spectrum of underlying genetic alterations that activate kinase or cytokine receptor signaling, while lacking the BCR-ABL1 fusion gene. ALL Ph+ and Ph-like show a high incidence of IKZF1 alterations. Approximately two-thirds of pediatric B-ALL Ph+ cases have an IKZF1 deletion, but IKZF1 point mutations have been identified in up to 10% of IKZF1 deletion-negative B-ALL Ph+ cases. The presence of IKZF1 deletions has been associated with an older age at diagnosis, higher presenting white blood cell counts, and higher levels of MRD after induction and consolidation [15]. In this review, we provide the genomic landscape of childhood ALL with t(9;22)(q34;q11.2) and present potential strategies for treatment, and also include consideration of the Ph-like subtype.

2. Treatment of Pediatric ALL Ph+

Phosphorylation is the main function of protein tyrosine kinases (PTKs), which are normally absent in the ABL protein. However, when BCR-ABL1 fusion occurs, PTK functions out of control, damaging the signaling pathways and potentially leading to leukemogenesis. The idea of using a special inhibitor for an exact gene mutation seems to be the most logical option. Imatinib mesylate binds the inactive moiety of BCR-ABL kinase, which completely blocks its ATP binding site, leading to the inhibition of tyrosine phosphorylation of proteins that are involved in the intracellular signal transduction that BCR-ABL mediates. This prevents a conformational switch to the active oncoprotein. Using tyrosine kinase inhibitors (TKIs), especially imatinib, increases the chance of an improved outcome in Ph-positive ALL patients. Studies conducted by the Children Oncology Group (COG) and the European EsPhALL consortium have provided evidence that pediatric ALL Ph+ patients can be effectively treated with a combination of chemotherapy and tyrosine kinase inhibitors (TKIs), without HSCT, in the first remission (CR1). To date, three trials have been conducted: EsPhALL2004, EsphALL2010, and EsphALL2017 [16,17]. The first study focused on using post-induction imatinib and chemotherapy in accordance with the Berlin–Frankfurt–Münster protocol. In EsPhALL2004, after induction, patients were classified into two groups: the good risk and poor risk groups. Patients who were classified into the good risk group were randomly allocated imatinib (300 mg/m2 per day) plus chemotherapy or chemotherapy alone, whereas those who were categorized into the poor risk group received post-induction imatinib plus chemotherapy. The authors suggested that imatinib can improve patient outcomes in the poor risk group, with minimal residual disease. Any adverse events were mainly related to myelosuppression, rather than to the use of imatinib per se. The effectiveness of HSCT and imatinib treatment remains unclear, however, because the majority of the study group (77%) had HSCT. The second trial, EsPhALL2010, investigated whether continuous administration of imatinib during chemotherapy could give better results compared to EsPhALL2004. In view of ALL Ph+ heterogeneity, patients were divided into two groups: the good risk group, which received HSCT, and the poor risk group, which received only chemotherapy treatment because of the lack of a genotype-matched donor. The major difference to the previous study was that in EsPhALL2010 all patients were continuously given imatinib, along with chemotherapy, from Day 15 of induction to the end of the treatment. EsPhALL2010 also produced opposing results concerning toxicity compared to the EsPhALL 2004 study. There were 14 reported serious events, as well as serious adverse events, which occurred in approximately 52% of all patients. This leads to the conclusion that earlier and longer exposure to imatinib in the course of chemotherapy might increase the toxicity. In the EsPhaLL2010 study, which consisted of administrating imatinib during induction, the results of the first complete remission were 97% compared to 78% in EsPhALL2004, where imatinib was administered after induction. The role of imatinib after HSCT has not been established yet. There were significant differences in exposure to imatinib between the trials. In the EsPhALL2004 study it was 3 or 5 months, depending on whether the patients had transplanted HSCT or not, while in EsPhALL2010, non-transplanted patients were treated for 24 months. The comparison of treatment results without the use of TKIs and the EsPhALL 2004 and 2010 protocols is presented in Table 1. The latest trial, EsPhALL2017, has attempted to balance treatment-related toxicity from intensive chemotherapy, without affecting the high rate of favorable outcomes. From Day 15 of induction, all children with ALL Ph+ received imatinib, and HSCT was performed. Due to MRD patients having been divided into a high-risk group and standard-risk group, MRD was the only factor used in this trial to risk-stratify patients with ALL Ph+. Current clinical trials suggest that chemotherapy plus imatinib could improve the treatment results for ALL Ph+ patients. The EsPhALL2017 study is ongoing, and hopefully its results will represent a breakthrough in ALL Ph+ treatment [18]. Therapeutic failure in ALL Ph+ could be the result of resistance to TKI-based therapies, caused by point mutations in the BCR-ABL1 kinase domain. The generally used Sanger sequencing (SS) for diagnostic BCR-ABL1 kinase domain mutations has some limitations—it has poor sensitivity and is unable to identify compound mutations. Soverini et al. reported the first attempt at defining indications for the use of Next-Generation Sequencing (NGS) for BCR-ABL1 kinase domain mutation screening in ALL Ph+. The authors suggested that NGS results in hopeless cases (relapsed or resistant to treatment ALL), which may have an impact on therapeutic decision-making [19]. However, some studies have indicated that the second-generation tyrosine kinase inhibitors, including dasatinib and nilotinib, should also be considered in ALL Ph+ treatment, because they appear to be more effective in the suppression of BCR-ABL1 kinase activity [20,21,22]. In addition, case reports have suggested that acquired T315l mutations can define resistance to dasatinib [23]. A few reports also failed to demonstrate TKI as having a positive impact on overall survival [24]. Nevertheless, the majority of patients in these studies were adults, which probably influenced the results [25,26]. Most of the reports on ALL Ph+ therapies have originated from Europe, which is a possible limitation. However, the Ma et al. study on 67 Southeast Asian patients (aged 8–62 years) provided convincing evidence that the biological response to TKI treatment in ALL Ph+ is comparable with that found in Caucasian patients. In that study, a major factor that impacted clinical outcome was the BCR–ABL1 fusion transcript. Among 67 patients, 63% (n = 42) harbored the p190 transcript and 18% (n = 12) the p210 transcript. In 13% (n = 9) of the patients both p210 and 190 showed changes, which is very rare in studies on Caucasian patients [27]. The Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG) investigated the role of imatinib in ALL Ph+ pediatric patients (median age 7 years old) immediately before hematopoietic stem cell transplantation (HSCT). The treatment protocol consisted of the induction phase, consolidation phase, reinduction phase, two weeks of imatinib monotherapy, and the HSCT phase. The 4-year OS rate among all patients was 78.1 ± 6.5% [28].

3. Future Directions in Treatment

Before the era of tyrosine kinase inhibitors, the treatment of ALL Ph+ patients often failed, and outcomes were frequently poor. Imatinib allowed patients to achieve greater overall survival, and there is hope now for new strategies, such as second and third generation TKIs, bi-specific monoclonal antibodies, retinoids, and BCL-2 inhibitors. Dasatinib and nilotinib are second-generation tyrosine kinase inhibitors that were developed to overcome resistance to imatinib. Dasatinib is 300 times more potent than imatinib at blocking ABL kinase activity; it accumulates in the central nervous system and can successfully be used in most patients with imatinib resistance. The Children’s Oncology Group trial tested the safety and feasibility of adding dasatinib to intensive chemotherapy from Day 15 of induction in patients with ALL Ph+, aged 1 to 30 years. The five-year overall survival in the dasatinib group was 86% ± 5% and 87% ± 5% for standard-risk patients [21]. The first randomized clinical study, comparing the efficacy of imatinib and dasatinib in children with ALL Ph+, was conducted by the Chinese Children’s Cancer Group (CCCG-ALL-2015). After starting dexamethasone, the ALL Ph+ patients began to receive dasatinib (80 mg/m2 per day) or imatinib mesylate (300 mg/m2 per day), and this continued until the end of therapy. The 4-year overall survival rate was 69.2% in the imatinib group vs. 88.4% in the dasatinib group [29]. Lastly, nilotinib is a highly specific TKI that can be considered as a therapeutic option for imatinib-resistant ALL Ph+ patients [30]. Kantarjian et al. suggested that nilotinib has limited efficiency in this subgroup, because only 2 of 13 patients in their study had a response to the drug [31]. However, there is no available treatment for patients with primary or secondary resistance to dasatinib or nilotinib, and a third generation TKI, ponatinib, could be effective in these cases. To our knowledge, there are no studies on ponatinib treatment in childhood Ph+ ALL, but in adults, ponatinib has shown significant clinical activity in a limited sample (n = 5). Therefore, more research is needed to more comprehensively evaluate the therapeutic value of ponatinib [32,33].
Blinatumomab is an anti-CD3 and anti-CD19 bispecific monoclonal antibody. Blinatumomab activates T cells with its anti-CD3 group and binds to tumor cells with its anti-CD19 group, to promote cellular cytotoxicity. Foà et al. conducted a Phase 2 single-group trial of first-line therapy in adults with newly diagnosed ALL Ph+. After induction with dasatinib plus glucocorticoids, two cycles of blinatumomab were administered. The results showed that at the end of dasatinib induction therapy, 29% of the patients had a molecular response; however, this percentage increased to 60% after two cycles of blinatumomab and increased even more after additional blinatumomab cycles. The OS after 18 months was 95% and the DFS was 88%; however, the DFS was lower among patients with IKZF1 or IKZF1plus alterations [34].
Focal adhesion kinase (FAK) is a non-receptor-type tyrosine kinase that is constitutively activated in ALL Ph+. Hu et al. suggested that FAK is critical for leukemogenesis, and FAK inhibitors might be interesting candidates for B cell acute lymphoblastic leukemia treatment [35]. In an experimental study on mouse models of BCR-ABL1 positive leukemia, detection of IKZF1 promoted the development of an aggressive lymphoid leukemia. Churchman et al. revealed that IKZF1 alterations are the result of the gain of stem cell-like features, such as self-renewal and increased bone marrow stromal adhesion. Retinoid receptor agonists reversed this phenotype by inducing the expression of IKZF1. This stimulated the abrogation of adhesion and self-renewal, stopped the cell cycle, and reduced proliferation, without direct cytotoxicity. Using retinoids potentiated the activity of dasatinib in mouse BCR-ABL1 ALL, representing a promising therapeutic option in IKZF1-mutated ALL [36]. The B-cell lymphoma 2 (BCL-2) protein family plays a significant role in the intrinsic mitochondrial apoptosis pathway, through interactions between pro- and antiapoptotic proteins. Direct targeting of BCL-2 constitutes a promising approach for treating leukemia. Venetoclax is an oral, highly selective BCL-2 inhibitor that has shown activity in BCL-2–dependent hematologic malignancies (BCR/ABL1-positive ALL, d T-ALL, KMT2A-rearranged ALL, hypodiploid) ALL. Preclinical study results suggested that ALL cells were dependent on both BCL-2 and BCL-XL. Adults with relapsed/refractory ALL or lymphoblastic lymphoma were treated with venetoclax and navitoclax (BCL-2 and BCL-XL inhibitor). Among 47 heavily pre-treated patients, the complete remission rate was 60%, which confirms that it could be an effective treatment option in the future [37,38,39].

4. IKZF1 Deletions

Over the last few years, the treatment of childhood ALL has significantly improved, but precise awareness of the risk factors remains key in order to estimate the prognosis [2]. Recent discoveries have helped us to increase our knowledge about ALL based on genetics. Nowadays, treatment of each patient begins with genetic diagnostics of the genomic alterations using karyotyping, fluorescent in situ hybridization (FISH), multiplex ligation probe-dependent amplification (MLPA), microarrays, or next generating sequencing (NGS) [39]. Gene expression profiling studies have indicated that ALL Ph+ patients frequently carry co-occurring mutations (e.g., IKZF1, PAX5, EBF1) with BCR-ABL1 fusion. Much effort in recent years has focused on screening for the gene that defines ALL patient’s outcomes. According to genetic changes, most subtypes of ALL have been identified to have mutations in the genes encoding epigenetic regulators and chromatin-modifying proteins. Several mutations acquired at relapse were detected in subclones at diagnosis, suggesting that the mutations may be responsible for resistance to therapy. The mutation of epigenetic modifiers, such as CREBBP, is detected in 18.3% of relapse ALL cases [40]. However, the most common finding is IKZF1 gene alterations. The report of Mullighan et al. added to the existing knowledge that IKZF1 mutations are often (87.5%) accompanied by BCR-ABL1 rearrangements [41]. According to the age range, IKZF1 alterations in pediatric ALL Ph+ occur in up to 50% of cases [41,42], and this increases to 84% among adult ALL Ph+ [43]. The IKZF1 gene (OMIM*603023) is located on the short arm of Chromosome 7 (region p12.2) and consists of eight exons. The IKZF1 (Ikaros Family Zinc Finger 1) gene encodes the transcription factor Ikaros, which is associated with chromatin remodeling and regulation of lymphocyte differentiation [44]. Ikaros has six zinc fingers, four of which are located in the DNA-binding domain encoded by Exons 4 to 6, and they are necessary to sustain Ikaros tumor-suppressor function. The rest of the zinc fingers are determined by Exon 8 and mediate the dimerization of Ikaros either as a homodimer or with other transcription factors of the Ikaros Zinc Finger family group [45]. Loss of Ikaros activity decreases sensitivity to tyrosine kinase inhibitors and deregulates the cascade of lymphoid genes in the hematopoietic stem cells, which normally stimulate differentiation into all three of the major hemolymphoid lineages [46]. There are three functional types of IKZF1 mutations. The most common alterations (55%) are haploinsufficiency, when one copy of the IKZF1 gene is inactivated or deleted, and the remaining allele of the gene is not adequate to produce the gene product, which is needed to preserve normal function. The second (33%) type is a dominant-negative form of Ikaros, made by an in-frame deletion of Exons 4 to 7 [47]. IKZF1 deletion is associated with higher levels of white blood cells, older age at diagnosis, and MRD [48]. The many defined genetic alternations and markers related to IKZF1 deletion have led to the need for a new subgroup, termed IKZF1plus. IKZF1plus emphasizes the importance of moving from prognostic factors to a prognostic profile. IKZF1plus is defined as an IKZF1 deletion co-occurring with deletions in CDKN2A or CDKN2B (only homozygous deletions) or the PAX5 or PAR1 region (P2RY8-CRLF2), in the absence of ERG deletion. IKZF1plus patients have a 5-year EFS of 53 ± 6%, compared with 79 ± 5% in patients with IKZF1 deletion or 87 ± 1% in patients who lack any IKZF1 deletion. IKZF1plus is a strong prognostic factor, but only in patients who are still carrying measurable MRD of a leukemic cell load exceeding 10−4 after induction treatment [49]. This relationship has already been underlined as a high-risk stratification criterion in the current frontline AIEOP-BFM ALL 2017 protocol for ALL treatment [50]. Moreover, research on the characteristics that unify IKZF1plus patients as a subgroup has found the GATA3 single-nucleotide variant rs382466245 to be enriched within IKZF1plus patients. This may be the background to future studies into IKZF1plus inheritance [51]. Inherited GATA3 variants are associated with Ph-like childhood acute lymphoblastic leukemia, and risk of relapse [52]. The Malaysia–Singapore ALL 2010 (MS2010) study was one of the first trials focusing on effective chemotherapy in ALL Ph+ pediatric patients with IKZF1 deletions. All patients with ALL Ph+ were treated with imatinib, starting from Day 15 of induction, for a total of 2 years. Likely because of imatinib administration, the 5-year cumulative incidence of relapse (CIR) decreased from 66.7% (n = 12) to 20% (n = 10) compared with MS2003, an earlier version of the study [53,54]. Interestingly, Palmi and colleagues put forward the hypothesis that the poor outcomes of the patients with IKZF1 deletions is evidence of genomic instability in aggressive leukemic clones, rather than a result of reduced IKZF1 function caused by IKZF1 haploinsufficiency, as earlier trials suggested [55]. Studies on monozygotic twins have suggested that ALL Ph+ can be an effect of prenatal BCR-ABL1 fusion. Additionally, in a pair where both children had BCR/ABL1 fusion, one twin with an IKZF1 mutation was diagnosed with ALL and died later, and the other remained healthy. Based on the research results, we assume that IKZF1 mutations may be associated with a poor prognosis. Moreover, this data suggests that BCR-ABL1 gene fusion can already exist prenatally [56]. The Children’s Oncology study confirmed that IKZF1 alteration is strongly associated with poor outcomes in BCR-ABL1-negative patients as well, which indicates that IKZF1 determines leukemogenesis and response to therapy [57,58,59]. Unfortunately, because of ALL Ph+ heterogeneity, IKZF1 mutation is not the only important aspect of outcome prediction. Research in various centers around the world may indicate genetic predisposition of various ethnic groups. A study from India on 132 patients (aged from 2 months to 67 years, median age 8.5 years) confirmed that IKZF1 is a commonly detected mutation at 26.5% (n = 43), and is associated with older age and higher induction failure rates [60]. Similar results were observed in a study from China, where patients (aged 14–70) with IKZF1 deletions had a higher relapse risk and worse survival rate. These data also suggested that transplants could overcome the adverse impact of IKZF1 deletions, although these results need to be confirmed in a randomized study in the future [61].

5. Molecular Background of Ph-Like

High-throughput genomic studies have enabled the identification of another genetic alteration, providing the rationale to test targeted therapies in genetically defined patient subsets. ALL Ph+ is defined by the t(9;22)(q34;q11) translocation that produces BCR-ABL1, a constitutively active tyrosine kinase. Based on gene expression profiling studies, a new subtype of ALL was described, named Philadelphia chromosome-like ALL [62]. Ph-like ALL is characterized by a gene expression profile that is extremely similar to ALL Ph+, but lacking the exact BCR-ABL1 fusion. Therefore, among ALL subtypes, Ph-like ALL is the most heterogeneous, which poses challenges in diagnosis and treatment. Copy number changes, chromosomal rearrangements, aneuploidy, and deregulated gene expression are only some types of genetic alterations. Male and Down Syndrome patients more often suffer from Ph-like ALL [63]. Ph-like ALL incidence increases with age and is indicated in 15% of B-ALL pediatric cases, rising to 21% in adolescents and up to 27% in young adults with B-ALL [64]. However, after young adulthood, the frequency of Ph-like ALL decreases [65]. This is the opposite to the frequency of Ph-positive ALL, which is known to continually increase with age [66]. Studies have confirmed that more than 80% of Ph-like cases are associated with the kinase activating pathway or cytokine receptor alteration [67,68]. The heterogeneity of genetic variants is enormous; however, the two main groups are based on activation of the ABL-class (ABL1, ABL2, PDGFRA, PDGFRB) or JAK-STAT (IL7R, JAK1, JAK3) alteration. Among Ph-like ALL patients, 10–14% have rearrangements of the ABL-class genes (ABL1, ABL2, PDGFRA, PDGFRB, LYN, CSF1R) other than BCR-ABL1 [63,69]. Knowledge of ABL-class fusions helps to better determine the treatment of these patients. Patients with the EBF1-PDGFRB fusion have a significantly higher rate of MRD and induction failure [70]. In addition, the EBF1 and PAX5 genes are frequently deleted, but their prognostic significance is not fully understood. Results of the Therapeutically Applicable Research to Generate Effective Treatments (TARGET) study undertaken by the National Cancer Institute identified new gene mutations, including CRLF2, IL-7R, JAK1, and JAK2, which assign ALL patients to a high-risk group [71]. Another similarity to ALL Ph+ is a high frequency of IKZF1 mutations, which is observed in 70–80% of Ph-like patients [72]. Additionally, half of Ph-like ALL patients have cytokine receptor-like factor 2 gene-CRLF2 over-expression, and it is more often indicated in older children [63,68,73]. Furthermore, up to 50% of CRLF2 cases harbor concomitant JAK mutations, the most popular of which is the R683G point mutation located in the pseudokinase domain of JAK2 [74,75]. In contrast, some studies have suggested that in Ph-like patients, ETV6-RUNX1 is more frequent in younger children compared with older children. Moreover, ETV6-RUNX1 is also associated with a favorable outcome [76]. Furthermore, in 4% of Ph-like patients, EPOR rearrangements are indicated, which normally stimulate red blood cells development [77,78]. In the most recent Children’s Oncology Group (COG) study, Ph-like ALL patients’ 5-year event-free survival rate was 63%, compared with 86% in non-Ph-like cases [71]. An observation made by Weston et al. indicated that BCR-ABL1-like ALL patients with EBF1-PDGFRB rearrangements (identified in 8% of patients) could be successfully treated with TKI in cases refractory to conventional four-drug induction therapy [79,80]. Various findings exist as to the occurrence of IKZF1 mutations in the BCR-ABL1 gene in ALL Ph-like patients. The St. Jude Children’s Research Hospital and Children’s Oncology Group found them in 80% to 90% of cases, but Van Der Veer identified IKZF1 mutations in 40% of patients [81,82]. Nevertheless, IKZF1 mutations may predict an unfavorable outcome in ALL Ph+, as well as in Ph-like ALL. The results of a study conducted in Taiwan suggested that the incidence of Ph-like ALL, as well as IKZF1 deletion in B-cell ALL patients, are lower in Taiwan than in Europe. More attention should be paid to the diversity of frequency of genetic alternations in different ethnicity groups [83]. It has long been known that some ploidy genomes or number of chromosomes impact on ALL outcome. We can classify Ph-like patients based on the number of chromosomes. Hyperdiploidy is characteristic of Chromosomes 51–65, high hypodiploidy of 40–44, low hypodiploidy of 30–39, and near-haploidy is typical for chromosomes <30. Hypodiploidy is rare and occurs in around 6% of ALL patients, of which the majority (80%) have 45 chromosomes [84,85]. High hyperdiploidy is associated with a better outcome; however, low hypodiploidy and near-haploidy are both linked with high rates of relapse [14,86,87]. Hypodiploidy, as well as near haploid ALL, demonstrate activation of the Ras and PI3K signaling pathways, and are sensitive to PI3K inhibitors, which may be the ultimate aim of targeted therapy.
The 2016 World Health Organization (WHO) classification of hematopoietic and lymphoid tissues highlights progress in the identification of unique biomarkers and gene expression, which has improved the diagnostic criteria [88]. In GMALL (the German Multicenter Study Group for Adult ALL), IGH-CRLF2 rearrangements and mutations in JAK2 were found exclusively in the Ph-like ALL subgroup. The authors suggested that screening gene expression analysis of these genes could be an easy way to identify Ph-like ALL patients. Ph-like ALL is a challenge to diagnose [89]. Boer et al. analyzed the gene expression of patients with newly diagnosed ALL and identified BCR-ABL1-like in 15% of precursor B-ALL cases. However, an analysis of the karyotype of BCR-ABL1-like patients did not reveal a common genetic feature. Patients did not present anomalies such as hyperdiploidy, ETV6–RUNX1 translocation, TCF3-rearrangement, KMT2A-rearrangement, or BCR–ABL1 translocation. However, array-CGH (comparative genomic hybridization) tests revealed the most common deletions in Chromosomes 9p and 20q, and local amplification of regions of Chromosomes 21q21–q22 [62].

6. Treatment of Ph-Like

The results of allogeneic stem cell transplants in Ph-like ALL in children, as well as adults, show it is often unsuccessful, and new therapies could be life changing [90]. Moreover, Boer et al. suggested that BCR-ABL1-like leukemic cells have special properties that make them 1.6 times more resistant to daunorubicin and 73 times more resistant to L-asparaginase than other precursor B-ALL cases. Interestingly, there were no differences in adverse cytotoxic effects of vincristine or prednisolone [62]. The role of TKI with intensive chemotherapy treatment in Ph-like ALL patients has not been established yet, but promising results in ALL Ph+ patients lead us to expect therapeutic success. Close to 20% of B-ALL cases showed gene rearrangements of CRL2, ABL1, JAK2, PDGFRB, JAK1, and JAK2, which activate kinase signaling [91]. The kinase fusion and partner genes identified in Ph-like ALL are presented in Table 2. Due to the high occurrence of kinase-activating mutations in Ph-like, there is a possibility that unique approaches in precision medicine could drive tailored treatment [82]. Data from various groups have clearly shown that the ABL1 alteration can be treated by TKI, and JAK-STAT mutations by JAK inhibitors [92]. Tanasi et al. found that patients with ABL-class kinase fusion treated with TKI frontline or at relapse demonstrated a better MRD response and overall survival. Moreover, the authors reported significantly improved outcomes among children and adolescents in comparison to adults [93]. The results of a multicenter worldwide retrospective study conducted by the Ponte di Lego group strengthens the evidence regarding the use of TKIs. A total of 122 patients with newly diagnosed ABL-class fusion B-cell ALL underwent treatment that did not contain tyrosine-kinase inhibitors. In this group, the 5-year event-free survival was 59.1%, and the 5-year overall survival was 76.1% [94]. By contrast, in Cario’s study, patients with ABL-class fusion were divided into two groups. The first group (n = 33) was treated according to AIEOP-BFM (Associazione Italiana di Ematologia-Oncologia Pediatrica–Berlin-Frankfurt-Münster) protocols, without TKI, and the second group (n = 13) received TKI (imatinib in eight and dasatinib in five cases) during different phases of treatment. Interestingly, their 5-year EFS and 5-year OS did not differ significantly compared with the no-TKI group [9]. However, the non-randomized UKALL2011 trial demonstrated a reduced risk of relapse for ABL-class fusion patients with MRD ≥ 1% treated with adjuvant TKI, without a significantly increased risk of severe toxicity [95]. Due to the diversity and small number of Ph-like patients, it was hard to plan a randomized clinical trial on the basis of developing treatment standards. There is a need for international, multicenter collaboration to achieve this [23]. In this context, successful treatment in Ph-like patients remains challenging. However, early indication of molecular treatment targets and future studies on TKI could lead to progress in the near future in developing treatment protocols.

7. Conclusions

The mechanisms of the detailed molecular changes in ALL Ph+, especially the sequence of changes, as well as the genetic risk factors, have not yet been established. It is expected that, in the next few years, the genomic landscape of ALL Ph+ will be completely described, which will provide the basis for further research into treatments. Molecular research allowed us to distinguish a new group of ALL, Ph-like ALL, which is characterized by broad-spectrum genetic alterations, but does not have an exact BCRL/ABL1 fusion. Identification of genetic mutations can help predict the prognosis, but classification to the appropriate risk group still remains a challenge. IKZF1 deletion is a predictor of adverse outcomes in pediatric ALLs. ALL Ph+ is characterized by poor outcomes, but more recently, tyrosine kinase inhibitors appear to have improved patient outcomes. The spectrum of drugs that hold promise as therapeutic options for treating ALL Ph+ is expanding. Although ALL Ph+ treatment is standardized by the EsPhALL protocol, we are still waiting for treatment recommendations for Ph-like ALL. Ph-like heterogeneity is a limiting factor in the conduction of randomized controlled trials, which are nevertheless required to establish treatment approaches.

Author Contributions

M.L. and J.Z. were responsible for the conception; M.L. was responsible for the design of the study; A.K. and P.Ś. were responsible for the acquisition of the literature for the manuscript; A.K. and P.Ś. wrote the original draft of manuscript; M.L. and J.Z. reviewed and edited; M.L. and J.Z. supervised the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Medical Research Agency, Poland (project number: 2019/ABM/01/00069-00; approval date 1 June 2020).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABL1V-abl Abelson murine leukemia viral oncogene homolog 1
ABL2V-abl Abelson murine leukemia viral oncogene homolog 2
AIEOP-BFMAssociazione Italiana di Ematologia-Oncologia Pediatrica–Berlin-Frankfurt-Münster
ALLacute lymphoblastic leukemia
ALLOhsct-allogenic hematopoietic stem cell transplantation
ATF1activating transcription factor 1
ATF71Pactivating transcription factor 7-interacting protein
B-ALLB-cells Acute Lymphoblastic Leukemia
BCRbreakpoint cluster region
BCR/ABL1BCR and ABL fusion gene
BLNKB-cell linker protein
CENPCcentromeric protein c1
CIRcumulative incidence of relapse
COGchildren oncology group
CR1first complete remission
CRLF2cytokine receptor-like factor 2
CSF1RColony Stimulating Factor 1 Receptor
DFSdisease-free survival
DGKHdiacylglycerol kinase, ETA, 130-KD
EBF1early B-cell factor 1
EFSevent-free survival
EPORerythropoietin receptor
ETV6ets variant transcription factor 6
ETV6–RUNX1ETV6 and RUNX1 fusion gene
FGFR1fibroblast growth factor receptor 1
FIP1L1actor interacting with PAPOLA and CPSF1
FISHfluorescence in situ hybridization
FLT3FMS-related tyrosine kinase 3
FOXP1forkhead box P1
FRSevent-free survival
GATAD2Agata zinc finger domain-containing protein A2
GNF2allosteric Bcr-abl inhibitors
GNF5allosteric inhibitor of Bcr-Abl
GOLGA5golgin A5
HSCThematopoietic sterm cell transplantation
IGHimmunoglobulin heavy
IGKimmunoglobulin kappa locus
IL2RBinterleukin 2 receptor, beta
IL7Rinterleukin 7 receptor alpha chain
IQGAP2IQ motif-containing GTPase-activating protein 2
JAK1Janus kinase 1
JAK2Janus kinase 2
JAK3Janus kinase 3
JAK-STATJanus kinase-signal transducer and activator of transcription
KDM6Alysine demethylase 6A
LAIR1leukocyte associated immunoglobulin like receptor 1
LSM14AmRNA processing body assembly factor
LYNLYN proto-oncogene
MRDminimal residual disease
MS2010Malaysia–Singapore ALL 2010 study
MYBprotooncogene, transcription factor
MYH9myosin, heavy chain 9, nonmuscle
NCOR1nuclear receptor corepressor 1
NGSnext generating sequencing
NTRK3neurotrophic tyrosine kinase, receptor, type 3
NUP153nucleoporin 153
NUP214nucleoporin 214
P2RY8pyrimidinergic receptor P2Y, G protein coupled 8
PAG1phosphoprotein associated with glycosphingolipid-enriched microdomains 1
PAX5paired box gene 5
PCM1pericentriolar material 1
PDGFRAPlatelet-derived growth factor receptor alpha
PDGFRBPlatelet-derived growth factor receptor beta
PhPhiladelphia chromosome
Ph+-Philadelphia chromosome positive
PPFIBP1protein-tyrosine phosphatase, receptor type, F polypeptide-interacting protein-binding protein 1
PTKprotein tyrosine kinase
PTK2Bprotein-tyrosine kinase 2, beta
RANBP2ran-binding protein 2
RCSD1RCSD domain containing 1
RFX3regulatory factor 3
SFPQsplicing factor proline- and glutamine-rich
SMARCA4SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily A, member 4
SMU1DNA replication regulator and spliceosomal factor
SNX1sorting nexin 1
SNX29sorting nexin 29
SPTAN1spectrin alpha, non-erythrocytic 1
SSBP2single-stranded DNA-binding protein 2
STAG2stromal antigen 2
STRN3striatin, calmodulin-binding protein 3
TARGETTherapeutically Applicable Research to Generate Effective Treatments
TBL1XR1transducin-beta-like 1 receptor 1
TERF2telomeric repeat-binding factor 2
THADAthada armadillo repeat-containing protein
TKItyrosine kinase inhibitors
TMEM2transmembrane protein 2
TNFAIP3interacting protein 1
TNIP1TNFAIP3 interacting protein 1
TPRtranslocated promoter region
TSLPthymic stroma lymphoprotein
TYK2tyrosine kinase 2
USAUnited States of America
USP25ubiquitin-specific protease 25
ZC3HAV1zinc finger CCCH-type containing, antiviral 1
ZEB2zinc finger E box-binding homeobox 2
ZFAND3zinc finger AN1 domain-containing protein 3
ZMIZ1zinc finger miz-domain containing 1
ZMYM2zinc finger, MYM-type 2
ZNF274zinc finger protein 274

References

  1. Ward, E.; DeSantis, C.; Robbins, A.; Kohler, B.; Jemal, A. Childhood and Adolescent Cancer Statistics, 2014. CA. Cancer J. Clin. 2014, 64, 83–103. [Google Scholar] [CrossRef] [PubMed]
  2. Pui, C.H.; Pei, D.; Campana, D.; Cheng, C.; Sandlund, J.T.; Bowman, W.P.; Hudson, M.M.; Ribeiro, R.C.; Raimondi, S.C.; Jeha, S.; et al. A Revised Definition for Cure of Childhood Acute Lymphoblastic Leukemia. Leukemia 2014, 28, 2336–2343. [Google Scholar] [CrossRef] [Green Version]
  3. Bernt, K.; Hunger, S. Current concepts in pediatric Philadelphia chromosome-positive acute lymphoblastic leukemia. Front. Oncol. 2014, 4, 54. [Google Scholar] [CrossRef] [PubMed]
  4. Sattler, M.; Griffin, J.D. Molecular mechanisms of transformation by the BCR-ABL oncogene. Semin. Hematol. 2003, 40, 4–10. [Google Scholar] [CrossRef] [PubMed]
  5. Pane, F.; Cimino, G.; Izzo, B.; Camera, A.; Vitale, A.; Quintarelli, C.; Picardi, M.; Specchia, G.; Mancini, M.; Cuneo, A.; et al. Significant reduction of the hybrid BCR/ABL transcripts after induction and consolidation therapy is a powerful predictor of treatment response in adult Philadelphia-positive acute lymphoblastic leukemia. Leukemia 2005, 19, 628–635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Aricò, M.; Valsecchi, M.G.; Camitta, B.; Schrappe, M.; Chessells, J.; Baruchel, A.; Gaynon, P.; Silverman, L.; Janka-Schaub, G.; Kamps, W.; et al. Outcome of Treatment in Children with Philadelphia Chromosome–Positive Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2000, 342, 998–1006. [Google Scholar] [CrossRef] [PubMed]
  7. Zawitkowska, J.; Lejman, M.; Romiszewski, M.; Matysiak, M.; Ćwiklińska, M.; Balwierz, W.; Owoc-Lempach, J.; Kazanowska, B.; Derwich, K.; Wachowiak, J.; et al. Results of Two Consecutive Treatment Protocols in Polish Children with Acute Lymphoblastic Leukemia. Sci. Rep. 2020, 10, 1–10. [Google Scholar]
  8. Oskarsson, T.; Söderhäll, S.; Arvidson, J.; Forestier, E.; Montgomery, S.; Bottai, M.; Lausen, B.; Carlsen, N.; Hellebostad, M.; Lähteenmäki, P.; et al. Relapsed Childhood Acute Lymphoblastic Leukemia in the Nordic Countries: Prognostic Factors, Treatment and Outcome. Haematologica 2016, 101, 68–76. [Google Scholar] [CrossRef] [Green Version]
  9. Cazzaniga, G.; de Lorenzo, P.; Alten, J.; Röttgers, S.; Hancock, J.; Saha, V.; Castor, A.; Madsen, H.O.; Gandemer, V.; Cavé, H.; et al. Predictive Value of Minimal Residual Disease in Philadelphia-Chromosome-Positive Acute Lymphoblastic Leukemia Treated with Imatinib in the European Intergroup Study of Post-Induction Treatment of Philadelphia-Chromosome-Positive Acute Lymphoblastic Leukemia, Based on Immunoglobulin/T-Cell Receptor and BCR/ABL1 Methodologies. Haematologica 2018, 103, 107–115. [Google Scholar]
  10. Borowitz, M.J.; Devidas, M.; Hunger, S.P.; Bowman, W.P.; Carroll, A.J.; Carroll, W.L.; Linda, S.; Martin, P.L.; Pullen, D.J.; Viswanatha, D.; et al. Clinical Significance of Minimal Residual Disease in Childhood Acute Lymphoblastic Leukemia and Its Relationship to Other Prognostic Factors: A Children’s Oncology Group Study. Blood 2008, 111, 5477–5485. [Google Scholar] [CrossRef] [Green Version]
  11. Cario, G.; Leoni, V.; Conter, V.; Attarbaschi, A.; Zaliova, M.; Sramkova, L.; Cazzaniga, G.; Fazio, G.; Sutton, R.; Elitzur, S.; 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, 1887–1894. [Google Scholar] [CrossRef] [PubMed]
  12. Pfeifer, H.; Cazzaniga, G.; van der Velden, V.H.J.; Cayuela, J.M.; Schäfer, B.; Spinelli, O.; Akiki, S.; Avigad, S.; Bendit, I.; Borg, K.; et al. Standardisation and Consensus Guidelines for Minimal Residual Disease Assessment in Philadelphia-Positive Acute Lymphoblastic Leukemia (Ph + ALL) by Real-Time Quantitative Reverse Transcriptase PCR of E1a2 BCR-ABL1. Leukemia 2019, 33, 1910–1922. [Google Scholar] [CrossRef] [PubMed]
  13. Inaba, H.; Mullighan, C.G. Pediatric Acute Lymphoblastic Leukemia. Haematologica 2020, 105, 2524–2539. [Google Scholar] [CrossRef]
  14. Moorman, A.V. The Clinical Relevance of Chromosomal and Genomic Abnormalities in B-Cell Precursor Acute Lymphoblastic Leukaemia. Blood Rev. 2012, 26, 123–135. [Google Scholar] [CrossRef]
  15. Tran, T.H.; Harris, M.H.; Nguyen, J.V.; Blonquis, T.M.; Stevenson, K.E.; Stonerock, E.; Asselin, B.L.; Athale, U.H.; Clavell, L.A.; Cole, P.D.; et al. Prognostic impact of kinase-activating fusions and IKZF1 deletions in pediatric high-risk B-lineage acute lymphoblastic leukemia. Blood Adv. 2018, 2, 529–533. [Google Scholar] [CrossRef] [PubMed]
  16. Biondi, A.; Schrappe, M.; De Lorenzo, P.; Castor, A.; Lucchini, G.; Gandemer, V.; Pieters, R.; Stary, J.; Escherich, G.; Campbell, M.; 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, 936–945. [Google Scholar] [CrossRef]
  17. Biondi, A.; Gandemer, V.; De Lorenzo, P.; Cario, G.; Campbell, M.; Castor, A.; Pieters, R.; Baruchel, A.; Vora, A.; Leoni, V.; et al. Imatinib Treatment of Paediatric Philadelphia Chromosome-Positive Acute Lymphoblastic Leukaemia (EsPhALL2010): A Prospective, Intergroup, Open-Label, Single-Arm Clinical Trial. Lancet Haematol. 2018, 5, e641–e652. [Google Scholar] [CrossRef] [Green Version]
  18. Ribera, J.M.; Oriol, A.; González, M.; Vidriales, B.; Brunet, S.; Esteve, J.; Del Potro, E.; Rivas, C.; Moreno, M.J.; Tormo, M.; et al. Concurrent Intensive Chemotherapy and Imatinib before and after Stem Cell Transplantation in Newly Diagnosed Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia. Final Results of the CSTIBES02 Trial. Haematologica 2010, 95, 87–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Soverini, S.; Albano, F.; Bassan, R.; Fabbiano, F.; Ferrara, F.; Foà, R.; Olivieri, A.; Rambaldi, A.; Rossi, G.; Sica, S.; et al. Next-Generation Sequencing for BCR-ABL1 Kinase Domain Mutations in Adult Patients with Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia: A Position Paper. Cancer Med. 2020, 9, 2960–2970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Zwaan, C.M.; Rizzari, C.; Mechinaud, F.; Lancaster, D.L.; Lehrnbecher, T.; van der Velden, V.H.J.; Beverloo, B.B.; den Boer, M.L.; Pieters, R.; Reinhardt, D.; et al. Dasatinib in Children and Adolescents with Relapsed or Refractory Leukemia: Results of the CA180-018 Phase I Dose-Escalation Study of the Innovative Therapies for Children with Cancer Consortium. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2013, 31, 2460–2468. [Google Scholar] [CrossRef] [PubMed]
  21. Slayton, W.B.; Schultz, K.R.; Kairalla, J.A.; Devidas, M.; Mi, X.; Pulsipher, M.A.; Chang, B.H.; Mullighan, C.; Iacobucci, I.; Silverman, L.B.; 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, 2306–2313. [Google Scholar] [CrossRef] [PubMed]
  22. Kobayashi, K.; Miyagawa, N.; Mitsui, K.; Matsuoka, M.; Kojima, Y.; Takahashi, H.; Ootsubo, K.; Nagai, J.; Ueno, H.; Ishibashi, T.; et al. TKI Dasatinib Monotherapy for a Patient with Ph-like ALL Bearing ATF7IP/PDGFRB Translocation. Pediatr. Blood Cancer 2015, 62, 1058–1060. [Google Scholar] [CrossRef] [PubMed]
  23. Fei, F.; Stoddart, S.; Müschen, M.; Kim, Y.M.; Groffen, J.; Heisterkamp, N. Development of resistance to dasatinib in Bcr/Abl-positive acute lymphoblastic leukemia. Leukemia 2010, 24, 813–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Tran, T.H.; Hunger, S.P. ABL-Class Fusion Positive Acute Lymphoblastic Leukemia: Can Targeting ABL Cure ALL? Haematologica 2020, 105, 1754–1757. [Google Scholar] [CrossRef]
  25. Kebriaei, P.; Saliba, R.; Rondon, G.; Chiattone, A.; Luthra, R.; Anderlini, P.; Andersson, B.; Shpall, E.; Popat, U.; Jones, R.; et al. Long-Term Follow-up of Allogeneic Hematopoietic Stem Cell Transplantation for Patients with Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia: Impact of Tyrosine Kinase Inhibitors on Treatment Outcomes. Biol. Blood Marrow Transplant. 2012, 18, 584–592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Doki, N.; Ohashi, K.; Oshikawa, G.; Kobayashi, T.; Kakihana, K.; Sakamaki, H. Clinical Outcome of Hematopoietic Stem Cell Transplantation for Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia (Ph + ALL): Experience from a Single Institution. Pathol. Oncol. Res. 2014, 20, 61–66. [Google Scholar] [CrossRef] [PubMed]
  27. Ma, L.; Hao, S.; Diong, C.; Goh, Y.T.; Gopalakrishnan, S.; Ho, A.; Hwang, W.; Koh, L.P.; Koh, M.; Lim, Z.Y.; et al. Pre-Transplant Achievement of Negativity in Minimal Residual Disease and French-American-British L1 Morphology Predict Superior Outcome after Allogeneic Transplant for Philadelphia Chromosome Positive Acute Lymphoblastic Leukemia: An Analysis of Southeast. Leuk. Lymphoma 2015, 56, 1362–1369. [Google Scholar] [CrossRef] [PubMed]
  28. Manabe, A.; Kawasaki, H.; Shimada, H.; Kato, I.; Kodama, Y.; Sato, A.; Matsumoto, K.; Kato, K.; Yabe, H.; Kudo, K.; et al. Imatinib Use Immediately before Stem Cell Transplantation in Children with Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia: Results from Japanese Pediatric Leukemia/Lymphoma Study Group (JPLSG) Study Ph+ALL04. Cancer Med. 2015, 4, 682–689. [Google Scholar] [CrossRef] [PubMed]
  29. Shen, S.; Chen, X.; Cai, J.; Yu, J.; Gao, J.; Hu, S.; Zhai, X.; Liang, C.; Ju, X.; Jiang, H.; 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, 6, 358–366. [Google Scholar] [CrossRef] [PubMed]
  30. Koo, H.H. Philadelphia Chromosome-Positive Acute Lymphoblastic Leukemia in Childhood. Korean J. Pediatr. 2011, 54, 106–110. [Google Scholar] [CrossRef]
  31. Kantarjian, H.; Giles, F.; Wunderle, L.; Bhalla, K.; O’Brien, S.; Wassmann, B.; Tanaka, C.; Manley, P.; Rae, P.; Mietlowski, W.; et al. Nilotinib in Imatinib-Resistant CML and Philadelphia Chromosome–Positive ALL. N. Engl. J. Med. 2006, 354, 2542–2551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Cortes, J.E.; Kantarjian, H.; Shah, N.P.; Bixby, D.; Mauro, M.J.; Flinn, I.; O’Hare, T.; Hu, S.; Narasimhan, N.I.; Rivera, V.M.; et al. Ponatinib in Refractory Philadelphia Chromosome–Positive Leukemias. N. Engl. J. Med. 2012, 367, 2075–2088. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Cortes, J.E.; Kim, D.W.; Pinilla-Ibarz, J.; le Coutre, P.D.; Paquette, R.; Chuah, C.; Nicolini, F.E.; Apperley, J.F.; Khoury, H.J.; Talpaz, M.; et al. Ponatinib Efficacy and Safety in Philadelphia Chromosome–Positive Leukemia: Final 5-Year Results of the Phase 2 PACE Trial. Blood 2018, 132, 393–404. [Google Scholar] [CrossRef]
  34. Foà, R.; Bassan, R.; Vitale, A.; Elia, L.; Piciocchi, A.; Puzzolo, M.-C.; Canichella, M.; Viero, P.; Ferrara, F.; Lunghi, M.; et al. Dasatinib–Blinatumomab for Ph-Positive Acute Lymphoblastic Leukemia in Adults. N. Engl. J. Med. 2020, 383, 1613–1623. [Google Scholar] [CrossRef] [PubMed]
  35. Hu, Z.; Slayton, W.B. Integrin VLA-5 and FAK Are Good Targets to Improve Treatment Response in the Philadelphia Chromosome Positive Acute Lymphoblastic Leukemia. Front. Oncol. 2014, 4, 112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Churchman, M.L.; Low, J.; Qu, C.; Paietta, E.M.; Lawryn, H.; Chang, Y.; Payne-turner, D.; Althoff, M.J.; Song, G.; Chen, S.; et al. Efficacy of Retinoids in IKZF1-Mutated BCR-ABL1 Acute Lymphoblastic Leukemia. Cancer Cell 2016, 28, 343–356. [Google Scholar] [CrossRef]
  37. Karol, S.E.; Cooper, T.M.; Bittencourt, H.; Gore, L.; O’Brien, M.M.; Fraser, C.; Gambart, M.; Cario, G.; Zwaan, C.M.; Bourquin, J.-P.; et al. Safety, efficacy, and PK of the BCL2 inhibitor venetoclax in combination with chemotherapy in pediatric and young adult patients with relapsed/refractory acute myeloid leukemia and acute lymphoblastic leukemia: Phase 1 study. Blood 2019, 134, 2649. [Google Scholar] [CrossRef]
  38. Pullarkat, V.A.; Lacayo, N.J.; Jabbour, E.; Rubnitz, J.E.; Bajel, A.; Laetsch, T.W.; Leonard, J.; Colace, S.I.; Khaw, S.L.; Fleming, S.A.; et al. Venetoclax and navitoclax in combination with chemotherapy in patients with relapsed or refractory acute lymphoblastic leukemia and lymphoblastic lymphoma. Cancer Discov. 2021, 11, 1–14. [Google Scholar] [CrossRef]
  39. Frisch, A.; Ofran, Y. How i Diagnose and Manage Philadelphia Chromosome-like Acute Lymphoblastic Leukemia. Haematologica 2019, 104, 2135–2143. [Google Scholar] [CrossRef] [Green Version]
  40. Joice, R.; Nilsson, S.K.; Montgomery, J.; Dankwa, S.; Morahan, B.; Seydel, K.B.; Bertuccini, L.; Alano, P.; Kim, C.; Duraisingh, M.T.; et al. CREBBP Mutations in Relapsed Acute Lymphoblastic Leukaemia Charles. Nature 2014, 6, 1–16. [Google Scholar]
  41. Mullighan, C.; Su, X.; Zhang, J.; Radtke, I.; Phillips, L.A.A.; Miller, C.B.; Ma, J.; Liu, W.; Cheng, C.; Schulman, B.A.; et al. Deletion of IKZF1 and Prognosis in Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2009, 360, 470–480. [Google Scholar] [CrossRef] [PubMed]
  42. Schwab, C.J.; Chilton, L.; Morrison, H.; Jones, L.; Al-Shehhi, H.; Erhorn, A.; Russell, L.J.; Moorman, A.V.; Harrison, C.J. Genes Commonly Deleted in Childhood B-Cell Precursor Acute Lymphoblastic Leukemia: Association with Cytogenetics and Clinical Features. Haematologica 2013, 98, 1081–1088. [Google Scholar] [CrossRef] [PubMed]
  43. Fedullo, A.L.; Messina, M.; Elia, L.; Piciocchi, A.; Gianfelici, V.; Lauretti, A.; Soddu, S.; Puzzolo, M.C.; Minotti, C.; Ferrara, F.; et al. Prognostic Implications of Additional Genomic Lesions in Adult Philadelphia Chromosomepositive Acute Lymphoblastic Leukemia. Haematologica 2019, 104, 312–318. [Google Scholar] [CrossRef]
  44. Vairy, S.; Tran, T.H. IKZF1 Alterations in Acute Lymphoblastic Leukemia: The Good, the Bad and the Ugly. Blood Rev. 2020, 100677. [Google Scholar] [CrossRef] [PubMed]
  45. Churchman, M.L.; Qian, M.; te Kronnie, G.; Zhang, R.; Yang, W.; Zhang, H.; Lana, T.; Tedrick, P.; Baskin, R.; Verbist, K.; et al. Germline Genetic IKZF1 Variation and Predisposition to Childhood Acute Lymphoblastic Leukemia. Cancer Cell 2018, 33, 937–948.e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Stanulla, M.; Eì Ene Cavé, H.; Cavé, C.; Moorman, A.V. Blood Spotlight IKZF1 Deletions in Pediatric Acute Lymphoblastic Leukemia: Still a Poor Prognostic Marker? Blood 2020, 135, 252–260. [Google Scholar] [CrossRef] [Green Version]
  47. Iacobucci, I.; Storlazzi, C.T.; Cilloni, D.; Lonetti, A.; Ottaviani, E.; Soverini, S.; Astolfi, A.; Chiaretti, S.; Vitale, A.; Messa, F.; 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 Leuke. Blood 2009, 114, 2159–2167. [Google Scholar] [CrossRef]
  48. Georgopoulos, K. Acute Lymphoblastic Leukemia—On the Wings of IKAROS. N. Engl. J. Med. 2009, 360, 524–526. [Google Scholar] [CrossRef]
  49. Stanulla, M.; Dagdan, E.; Zaliova, M.; Möricke, A.; Palmi, C.; Cazzaniga, G.; Eckert, C.; te Kronnie, G.; Bourquin, J.-P.; Bornhauser, B.; et al. 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, 1240–1249. [Google Scholar] [CrossRef] [Green Version]
  50. O’Connor, D.; Moorman, A.V.; Wade, R.; Hancock, J.; Tan, R.M.R.; Bartram, J.; Moppett, J.; Schwab, C.; Patrick, K.; Harrison, C.J.; et al. Use of Minimal Residual Disease Assessment to Redefine Induction Failure in Pediatric Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2017, 35, 660–667. [Google Scholar] [CrossRef] [Green Version]
  51. Perez-Andreu, V.; Roberts, K.G.; Harvey, R.C.; Yang, W.; Cheng, C.; Pei, D.; Xu, H.; Gastier-Foster, J.; Shuyu, E.; Lim, J.Y.; et al. Inherited GATA3 Variants Are Associated with Ph-like Childhood Acute Lymphoblastic Leukemia and Risk of Relapse. Nat. Genet. 2013, 45, 1494–1498. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Perez-Andreu, V.; Roberts, K.G.; Xu, H.; Smith, C.; Zhang, H.; Yang, W.; Harvey, R.C.; Payne-Turner, D.; Devidas, M.; Cheng, I.M.; et al. A Genome-Wide Association Study of Susceptibility to Acute Lymphoblastic Leukemia in Adolescents and Young Adults. Blood 2015, 125, 680–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Yeoh, A.E.J.; Tan, D.; Li, C.K.; Hori, H.; Tse, E.; Pui, C.H. Management of Adult and Paediatric Acute Lymphoblastic Leukaemia in Asia: Resource-Stratified Guidelines from the Asian Oncology Summit 2013. Lancet Oncol. 2013, 14, e508–e523. [Google Scholar] [CrossRef] [Green Version]
  54. Yeoh, A.E.J.; Ariffin, H.; Chai, E.L.L.; Kwok, C.S.N.; Chan, Y.H.; Ponnudurai, K.; Campana, D.; Tan, P.L.; Chan, M.Y.; Kham, S.K.Y.; et al. Minimal Residual Disease-Guided Treatment Deintensification for Children with Acute Lymphoblastic Leukemia: Results from the Malaysia-Singapore Acute Lymphoblastic Leukemia 2003 Study. J. Clin. Oncol. 2012, 30, 2384–2392. [Google Scholar] [CrossRef] [PubMed]
  55. Palmi, C.; Lana, T.; Silvestri, D.; Savino, A.; Kronnie, G.T.; Conter, V.; Basso, G.; Biondi, A.; Valsecchi, M.G.; Cazzaniga, G. Impact of IKZF1 Deletions on IKZF1 Expression and Outcome in Philadelphia Chromosome Negative Childhood BCP-ALL. Reply to “Incidence and Biological Significance of IKZF1/Ikaros Gene Deletions in Pediatric Philadelphia Chromosome Negative and Philadelphia chromosome positive B-cell precursor acute lymphoblastic leukemia”. Haematologica 2013, 98, e164–e165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Cazzaniga, G.; van Delft, F.W.; Lo Nigro, L.; Ford, A.M.; Score, J.; Iacobucci, I.; Mirabile, E.; Taj, M.; Colman, S.M.; Biondi, A.; et al. Developmental Origins and Impact of BCR-ABL1 Fusion and IKZF1 Deletions in Monozygotic Twins with Ph+ Acute Lymphoblastic Leukemia. Blood 2011, 118, 5559–5564. [Google Scholar] [CrossRef] [PubMed]
  57. Cario, G.; Leoni, V.; Conter, V.; Baruchel, A.; Schrappe, M.; Biondi, A. BCR-ABL1-like acute lymphoblastic leukemia in childhood and targeted therapy. Haematologica 2020, 105, 2200–2204. [Google Scholar] [CrossRef]
  58. Kuiper, R.P.; Waanders, E.; Van Der Velden, V.H.J.; Van Reijmersdal, S.V.; Venkatachalam, R.; Scheijen, B.; Sonneveld, E.; Van Dongen, J.J.M.; Veerman, A.J.P.; van Leeuwen, F.N.; et al. IKZF1 Deletions Predict Relapse in Uniformly Treated Pediatric Precursor B-ALL. Leukemia 2010, 24, 1258–1264. [Google Scholar] [CrossRef]
  59. Collins-Underwood, J.R.; Mullighan, C.G. Genomic Profiling of High-Risk Acute Lymphoblastic Leukemia. Leukemia 2010, 24, 1676–1685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Gupta, S.K.; Bakhshi, S.; Kumar, L.; Kamal, V.K.; Kumar, R. Gene Copy Number Alteration Profile and Its Clinical Correlation in B-Cell Acute Lymphoblastic Leukemia. Leuk. Lymphoma 2017, 58, 333–342. [Google Scholar] [CrossRef]
  61. Yao, Q.M.; Liu, K.Y.; Gale, R.P.; Jiang, B.; Liu, Y.R.; Jiang, Q.; Jiang, H.; Zhang, X.H.; Zhang, M.J.; Chen, S.-S.; et al. Prognostic Impact of IKZF1 Deletion in Adults with Common B-Cell Acute Lymphoblastic Leukemia. BMC Cancer 2016, 16, 4–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Den Boer, M.L.; van Slegtenhorst, M.; De Menezes, R.X.; Cheok, M.H.; Buijs-Gladdines, J.G.; Peters, S.T.; Van Zutven, L.J.; Beverloo, H.B.; Van der Spek, P.J.; Escherich, G.; et al. A Subtype of Childhood Acute Lymphoblastic Leukaemia with Poor Treatment Outcome: A Genome-Wide Classification Study. Lancet Oncol. 2009, 10, 125–134. [Google Scholar] [CrossRef] [Green Version]
  63. Roberts, K.G.; Pei, D.; Campana, D.; Payne-Turner, D.; Li, Y.; Cheng, C.; Sandlund, J.T.; Jeha, S.; Easton, J.; Becksfort, J.; 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, 3012–3020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Roberts, K.G.; Li, Y.; Payne-Turner, D.; Harvey, R.C.; Yang, Y.-L.; Pei, D.; McCastlain, K.; Ding, L.; Lu, C.; Song, G.; et al. Targetable Kinase-Activating Lesions in Ph-like Acute Lymphoblastic Leukemia. N. Engl. J. Med. 2014, 371, 1005–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Herold, T.; Baldus, C.D.; Gökbuget, N. Ph-like Acute Lymphoblastic Leukemia in Older Adults. N. Engl. J. Med. 2014, 371, 2235. [Google Scholar] [CrossRef] [PubMed]
  66. Burmeister, T.; Schwartz, S.; Bartram, C.R.; Gökbuget, N.; Hoelzer, D.; Dc, W. Patients’ Age and BCR-ABL Frequency in Adult B-Precursor ALL: A Retrospective Analysis from the GMALL Study Group. Blood 2008, 112, 918–919. [Google Scholar] [CrossRef] [PubMed]
  67. Rives, S.; Camós, M.; Estella, J.; Gómez, P.; Moreno, M.J.; Vivanco, J.L.; Melo, M.; Fernández-Delgado, R.; Verdeguer, A.; Fernández-Teijeiro, A.; et al. Longer Follow-up Confirms Major Improvement in Outcome in Children and Adolescents with Philadelphia Chromosome Acute Lymphoblastic Leukaemia Treated with Continuous Imatinib and Haematopoietic Stem Cell Transplantation. Results from the Spanish Cooperative Study SHOP/ALL-2005. Br. J. Haematol. 2013, 162, 419–421. [Google Scholar] [PubMed]
  68. Reshmi, S.C.; Harvey, R.C.; Roberts, K.G.; Stonerock, E.; Smith, A.; Jenkins, H.; Chen, I.M.; Valentine, M.; Liu, Y.; Li, Y.; et al. Targetable Kinase Gene Fusions in High-Risk B-ALL: A Study from the Children’s Oncology Group. Blood 2017, 129, 3352–3361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Lizcova, L.; Zemanova, Z.; Lhotska, H.; Zuna, J.; Hovorkova, L.; Mejstrikova, E.; Malinova, E.; Rabasova, J.; Raska, I.; Sramkova, L.; et al. An Unusual Case of High Hyperdiploid Childhood ALL with Cryptic BCR/ABL1 Rearrangement. Mol. Cytogenet. 2014, 7, 1–6. [Google Scholar] [CrossRef] [Green Version]
  70. Schwab, C.; Enshaei, A.; Roberts, K.G.; Russell, L.J.; Harvey, R.C.; Chen, I.-M.L.; Willman, C.L.; Mullighan, C.; Vora, A.J.; Moorman, A.V.; et al. The Frequency and Outcome of Ph-like ALL Associated Abnormalities in Childhood Acute Lymphoblastic Leukaemia Treated on MRC UKALL2003. Blood 2016, 128, 2914. [Google Scholar] [CrossRef]
  71. Loh, M.L.; Zhang, J.; Harvey, R.C.; Roberts, K.; Payne-Turner, D.; Kang, H.; Wu, G.; Chen, X.; Becksfort, J.; Edmonson, M.; et al. Tyrosine Kinome Sequencing of Pediatric Acute Lymphoblastic Leukemia: A Report from the Children’s Oncology Group TARGET Project. Blood 2013, 121, 485–488. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Roberts, K.G. Why and How to Treat Ph-like ALL? Best Pract Res Clin Haematol. 2018, 31, 351–356. [Google Scholar] [CrossRef]
  73. Chen, M.; Zhu, Y.; Lin, Y.; Tengwang, T.; Zhang, L. Use of Tyrosine Kinase Inhibitors for Paediatric Philadelphia Chromosome-Positive Acute Lymphoblastic Leukaemia: A Systematic Review and Meta-Analysis. BMJ Open 2021, 11, e042814. [Google Scholar] [CrossRef] [PubMed]
  74. Mullighan, C.G.; Collins-Underwood, J.R.; Phillips, L.A.A.; Loudin, M.G.; Liu, W.; Zhang, J.; Ma, J.; Coustan-Smith, E.; Harvey, R.C.; Willman, C.L.; et al. Rearrangement of CRLF2 in B-Progenitor- and Down Syndrome-Associated Acute Lymphoblastic Leukemia. Nat. Genet. 2009, 41, 1243–1246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Mullighan, C.G. New Strategies in Acute Lymphoblastic Leukemia: Translating Advances in Genomics into Clinical Practice. Clin. Cancer Res. 2011, 17, 396–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Gu, Z.; Churchman, M.; Roberts, K.; Li, Y.; Liu, Y.; Harvey, R.C.; McCastlain, K.; Reshmi, S.C.; Payne-Turner, D.; Iacobucci, I.; et al. Genomic Analyses Identify Recurrent MEF2D Fusions in Acute Lymphoblastic Leukaemia. Nat. Commun. 2016, 7, 1–10. [Google Scholar] [CrossRef] [Green Version]
  77. Iacobucci, I. Truncating Erythropoietin Receptor Rearrangements in ALL. Cancer Cell 2016, 29, 186–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Watowich, S.S. The Erythropoietin Receptor: Molecular Structure and Hematopoietic Signaling Pathways. J. Investig. Med. 2011, 59, 1067–1072. [Google Scholar] [CrossRef]
  79. Weston, B.W.; Hayden, M.A.; Roberts, K.G.; Bowyer, S.; Hsu, J.; Fedoriw, G.; Rao, K.W.; Mullighan, C.G. Tyrosine Kinase Inhibitor Therapy Induces Remission in a Patient With Refractory EBF1-PDGFRB–Positive Acute Lymphoblastic Leukemia. J. Clin. Oncol. 2013, 31, e413–e416. [Google Scholar] [CrossRef] [PubMed]
  80. 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, 146–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Van Der Veer, A.; Waanders, E.; Pieters, R.; Willemse, M.E.; Van Reijmersdal, S.V.; Russell, L.J.; Harrison, C.J.; Evans, W.E.; Van Der Velden, V.H.J.; Hoogerbrugge, P.M.; 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, 2622–2629. [Google Scholar] [CrossRef]
  82. Roberts, K.G.; Morin, R.D.; Zhang, J.; Hirst, M.; Zhao, Y.; Su, X.; Chen, S.-C.; Payne-Turner, D.; Churchman, M.L.; Harvey, R.C.; et al. Genetic Alterations Activating Kinase and Cytokine Receptor Signaling in High-Risk Acute Lymphoblastic Leukemia. Cancer Cell 2012, 22, 153–166. [Google Scholar] [CrossRef] [Green Version]
  83. Hsu, Y.C.; Yu, C.H.; Chen, Y.M.; Roberts, K.G.; Ni, Y.L.; Lin, K.-H.; Jou, S.T.; Lu, M.-Y.; Chen, S.-H.; Wu, K.H.; et al. Philadelphia Chromosome-Negative B-Cell Acute Lymphoblastic Leukaemia with Kinase Fusions in Taiwan. Sci. Rep. 2021, 11, 5802. [Google Scholar] [CrossRef]
  84. Safavi, S.; Olsson, L.; Biloglav, A.; Veerla, S.; Blendberg, M.; Tayebwa, J.; Behrendtz, M.; Castor, A.; Hansson, M.; Jonhansson, B.; et al. Genetic and Epigenetic Characterization of Hypodiploid Acute Lymphoblastic Leukemia. Oncotarget 2015, 6, 42793–42802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Pui, C.H.; Williams, D.L.; Raimondi, S.C.; Rivera, G.K.; Look, A.T.; Dodge, R.K.; George, S.L.; Behm, F.G.; Murphy, S.B. Hypodiploidy Is Associated with a Poor Prognosis in Childhood Acute Lymphoblastic Leukemia. Blood 1987, 70, 247–253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Harrison, C.J.; Moorman, A.V.; Broadfield, Z.J.; Cheung, K.L.; Harris, R.L.; Jalali, G.R.; Robinson, H.M.; Barber, K.E.; Richards, S.M.; Mitchell, C.D.; et al. Three Distinct Subgroups of Hypodiploidy in Lymphoblastic Leukaemia. Br. J. Haematol. 2004, 125, 552–559. [Google Scholar] [CrossRef] [PubMed]
  87. Holmfeldt, L.; Wei, L.; Diaz-Flores, E.; Walsh, M.; Zhang, J.; Ding, L.; Payne-Turner, D.; Churchman, M.; Andersson, A.; Chen, S.-C.; et al. The Genomic Landscape of Hypodiploid Acute. Nat. Genet. 2013, 45, 242–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Swerdlow, S.H.; Campo, E.; Pileri, S.A.; Lee Harris, N.; Stein, H.; Siebert, R.; Advani, R.; Ghielmini, M.; Salles, G.A.; Zelenetz, A.D.; et al. The 2016 Revision of the World Health Organization Classification of Lymphoid Neoplasms. Blood 2016, 127, 2375–2390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Herold, T.; Schneider, S.; Metzeler, K.H.; Neumann, M.; Hartmann, L.; Roberts, K.G.; Konstandin, N.P.; Greif, P.A.; Bräundl, K.; Ksienzyk, B.; 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, 130–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Shiraz, P.; Payne, K.J.; Muffly, L. The Current Genomic and Molecular Landscape of Philadelphia-like Acute Lymphoblastic Leukemia. Int. J. Mol. Sci. 2020, 21, 2193. [Google Scholar] [CrossRef] [Green Version]
  91. Mullighan, C.G. Molecular genetics of B-precursor acute lymphoblastic leukemia. J. Clin. Investig. 2012, 122, 3407–3415. [Google Scholar] [CrossRef] [Green Version]
  92. Tasian, S.K.; Doral, M.Y.; Borowitz, M.J.; Wood, B.L.; Chen, I.M.; Harvey, R.C.; Gastier-Foster, J.M.; Willman, C.L.; Hunger, S.P.; Mullighan, C.G.; et al. Aberrant STAT5 and PI3K/MTOR Pathway Signaling Occurs in Human CRLF2-Rearranged B-Precursor Acute Lymphoblastic Leukemia. Blood 2012, 120, 833–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Tanasi, I.; Ba, I.; Sirvent, N.; Braun, T.; Cuccuini, W.; Ballerini, P.; Duployez, N.; Tanguy-Schmidt, A.; Erômeerˆerôme Tamburini, J.; Ebastien Maury, S.; et al. Letters to Blood Efficacy of Tyrosine Kinase Inhibitors in Ph-like Acute Lymphoblastic Leukemia Harboring ABL-Class Rearrangements. Blood 2019, 134, 1351–1355. [Google Scholar] [CrossRef]
  94. Den Boer, M.L.; Cario, G.; Moorman, A.V.; Boer, J.M.; de Groot-Kruseman, H.A.; Fiocco, M.; Escherich, G.; Imamura, T.; Yeoh, J.; Sutton, R.; 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, e55–e66. [Google Scholar] [CrossRef]
  95. Moorman, A.V.; Schwab, C.; Winterman, E.; Hancock, J.; Castleton, A.; Cummins, M.; Gibson, B.; Goulden, N.; Kearns, P.; James, B.; 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, 191, 844–851. [Google Scholar] [CrossRef] [PubMed]
  96. Ernst, T.; Score, J.; Deininger, M.; Hidalgo-Curtis, C.; Lackie, P.; Ershler, W.B.; Goldman, J.M.; Cross, N.C.P.; Grand, F. Identification of FOXP1 and SNX2 as Novel ABL1 Fusion Partners in Acute Lymphoblastic Leukaemia. Br. J. Haematol. 2011, 153, 43–46. [Google Scholar] [CrossRef] [PubMed]
  97. Soler, G.; Radford-Weiss, I.; Ben-Abdelali, R.; Mahlaoui, N.; Ponceau, J.F.; Macintyre, E.A.; Vekemans, M.; Bernard, O.A.; Romana, S.P. Fusion of ZMIZ1 to ABL1 in a B-Cell Acute Lymphoblastic Leukaemia with a t(9;10)(Q34;Q22.3) Translocation. Leukemia 2008, 22, 1278–1280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Russell, L.J.; Capasso, M.; Vater, I.; Akasaka, T.; Bernard, O.A.; Calasanz, M.J.; Chandrasekaran, T.; Chapiro, E.; Gesk, S.; Griffiths, M.; et al. Deregulated Expression of Cytokine Receptor Gene, CRLF2, Is Involved in Lymphoid Transformation in B-Cell Precursor Acute Lymphoblastic Leukemia. Blood 2009, 114, 2688–2698. [Google Scholar] [CrossRef] [PubMed]
  99. Jain, N.; Roberts, K.G.; Jabbour, E.; Patel, K.; Eterovic, A.K.; Chen, K.; Zweidler-McKay, P.; Lu, X.; Fawcett, G.; Wang, S.A.; et al. Ph-like Acute Lymphoblastic Leukemia: A High-Risk Subtype in Adults. Blood 2017, 129, 572–581. [Google Scholar] [CrossRef] [Green Version]
  100. Lacronique, V.; Boureux, A.; Valle, V.D.; Poirel, H.; Quang, C.T.; Mauchauffé, M.; Berthou, C.; Lessard, M.; Berger, R.; Ghysdael, J.; et al. A TEL-JAK2 Fusion Protein with Constitutive Kinase Activity in Human Leukemia. Science 1997, 278, 1309–1312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Poitras, J.L.; Cin, P.D.; Aster, J.C.; DeAngelo, D.J.; Morton, C.C. Novel SSBP2-JAK2 Fusion Gene Resulting from a t(5;9)(Q14.1;P24.1) in Pre-B Acute Lymphocytic Leukemia. Genes Chromosom. Cancer 2008, 47, 884–889. [Google Scholar] [CrossRef]
  102. Roberts, K.G.; Yang, Y.L.; Payne-Turner, D.; Lin, W.; Files, J.K.; Dickerson, K.; Gu, Z.; Taunton, J.; Janke, L.J.; Chen, T.; et al. Oncogenic Role and Therapeutic Targeting of ABL-Class and JAK-STAT Activating Kinase Alterations in Ph-like ALL. Blood Adv. 2017, 1, 1657–1671. [Google Scholar] [PubMed]
  103. Knezevich, S.R.; Garnett, M.J.; Pysher, T.J.; Beckwith, J.B.; Grundy, P.E.; Sorensen, P.H. ETV6-NTRK3 Gene Fusions and Trisomy 11 Establish a Histogenetic Link between Mesoblastic Nephroma and Congenital Fibrosarcoma. Cancer Res. 1998, 58, 5046–5048. [Google Scholar] [PubMed]
  104. Munthe-Kaas, M.C.; Forthun, R.B.; Brendehaug, A.; Eek, A.K.; Høysæter, T.; Osnes, L.T.N.; Prescott, T.; Spetalen, S.; Hovland, R. Partial Response to Sorafenib in a Child with a Myeloid/Lymphoid Neoplasm, Eosinophilia, and a ZMYM2-FLT3 Fusion. J. Pediatr. Hematol. Oncol. 2021, 43, e508–e511. [Google Scholar] [CrossRef]
  105. Chase, A.; Bryant, C.; Score, J.; Haferlach, C.; Grossmann, V.; Schwaab, J.; Hofmann, W.K.; Reiter, A.; Cross, N.C.P. Ruxolitinib as Potential Targeted Therapy for Patients with JAK2 Rearrangements. Haematologica 2013, 98, 404–408. [Google Scholar] [CrossRef] [PubMed]
  106. Zhang, P.; Wang, Y.; Qin, M.; Li, D.; Odhiambo, W.O.; Yuan, M.; Lv, Z.; Liu, C.; Ma, Y.; Dong, Y.; et al. Involvement of Blnk and Foxo1 in Tumor Suppression in BCR-ABL1-Transformed pro-B Cells. Oncol. Rep. 2021, 45, 693–705. [Google Scholar] [CrossRef] [PubMed]
Table 1. Comparison of treatment results without the use of TKIs, and the EsPhALL 2004 and 2010 protocols.
Table 1. Comparison of treatment results without the use of TKIs, and the EsPhALL 2004 and 2010 protocols.
Treatment ProtocolsDFS 1 (%)EFS 2 (%)OS 3 (%)Age < 10 Years (%)Age ≥ 10 Years (%)Serious Adverse Events (%)p190 Transcript (%)p210 Transcript (%)MRD 4 (%)
Without using TKIs study 1986–1996 (n = 326)--28.340.36436----
EsPhALL2004Good risk imatinib group (n = 46)72.9-78.561392892863
Good risk no imatinib group (n = 44)61.7-644632901035
Poor risk group (n = 70)53.5-62.9415934782396
EsPhALL2010Good risk group (n = 102)-62.775.7653550782215
Poor risk group (n = 53)-46.363.651495598252
1 DFS, the 4-year disease free survival (%); 2 EFS, the 5-year event free survival; 3 OS, the 5-year overall survival; 4 MRD, minimal residual disease (at end of induction ≥ 5 × 10−4).
Table 2. Kinase fusion and partner genes identified in Ph-like ALL, and therapy strategies.
Table 2. Kinase fusion and partner genes identified in Ph-like ALL, and therapy strategies.
Kinase Fusion Identified in Ph-Like ALLFusion Partners GeneTreatmentReferences
ABL1CENPC, ETV6, LSM141, NUP153, NUP214, RANBP2, RSCD1, ZMIZ1, FOXP1, LSM14A, NUP153, NUP214, RANBP2, RCSD1, SFPQ, SNX1, SNX, SPTAN1, ZC3HAV1Imatinib, Dasatinib, GNF2 GNF5[82,96,97]
ABL2PAG1, RSCD1, ZC3HAV1,Imatinib/Dasatinib[64]
PDGFRBSSBP2, TBL1XR1, EBF1, TNIP1, ZEB2, ATF71P, ETV6, PAX5, PCM1, PPF1BP1, RFX3, SSBP2, STRN3Dasatinib[79,80,82]
PDGFRAFIP1L1Dasatinib[98]
CSF1RSSBP2Dasatinib[64]
CRLF2IGH, P2RY8JAK2 inhibitor[74,99]
LYNGATAD2A, NCOR1Imatinib/Dasatinib[98]
JAK2BCR, PAX5, PCM1, RFX3, USP25, ZNF274, ATF1, EBF1, ETV6, PAX5, PCM1, PPFIBP1, RFX3, SSBP2, STRN3, TERF2, TPR, USP25, ZNF274, GOLGA5, SMU1, SNX29, ZNF340JAK2 inhibitor[82,100,101,102]
EPORIGH, IGK, LAIR1, THADAJAK2 inhibitor[64,82,98]
TYK2MYB, SMARCA4,TYK2 inhibitor[64]
TSLPIQGAP2JAK2 inhibitor[64]
DGKHZFAND3Unknow[64]
IL2RBMYH9JAK1/JAK3 inhibitor or both[64]
NTRK3ETV6TRK inhibitor, Crizotinib[103]
PTK2BKDM6A, STAG2, TMEM2FAK inhibitor[64]
FLT3ZMYM2FLT3 inhibitor[104]
FGFR1BCRSorafenib, Dasatinib, Ponatinib[105]
BLNKDNNTUnknown[106]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kaczmarska, A.; Śliwa, P.; Zawitkowska, J.; Lejman, M. Genomic Analyses of Pediatric Acute Lymphoblastic Leukemia Ph+ and Ph-Like—Recent Progress in Treatment. Int. J. Mol. Sci. 2021, 22, 6411. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126411

AMA Style

Kaczmarska A, Śliwa P, Zawitkowska J, Lejman M. Genomic Analyses of Pediatric Acute Lymphoblastic Leukemia Ph+ and Ph-Like—Recent Progress in Treatment. International Journal of Molecular Sciences. 2021; 22(12):6411. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126411

Chicago/Turabian Style

Kaczmarska, Agnieszka, Patrycja Śliwa, Joanna Zawitkowska, and Monika Lejman. 2021. "Genomic Analyses of Pediatric Acute Lymphoblastic Leukemia Ph+ and Ph-Like—Recent Progress in Treatment" International Journal of Molecular Sciences 22, no. 12: 6411. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms22126411

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop