International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Chronic myeloid leukemia (CML) is a life-threatening hematological malignancy driven by intricate and multifactorial pathogenic mechanisms that underlie its clinical heterogeneity and therapeutic challenges. The Philadelphia chromosome (Ph), arising from a reciprocal translocation t(9;22)(q34;q11.2), is detected in up to 95% of adult CML cases (1). This chromosomal aberration generates an abnormal BCR-ABL1 fusion transcript, with molecular detection of this transcript serving as a key component for genetic confirmation of CML diagnosis (2,3). Recent advancements in molecular biology have highlighted the emerging role of atypical fusion genes in CML pathogenesis and progression (4,5). In Ph-positive leukemia, the canonical BCR-ABL1 fusion gene functions as a pivotal oncogenic driver. Variable chromosomal breakpoints within BCR and ABL1 genes result in distinct BCR-ABL1 transcript variants and corresponding protein isoforms (6). The e13a2 and e14a2 subtypes represent the most prevalent BCR-ABL1 isoforms in patients with CML, both containing intact sequences encoding the Src homology 3 domain(SH3), SH2 and kinase domains of ABL1. Beyond these common variants, rare fusion genotypes including e13a3, e14a3 and e1a3 have been documented (7,8). The e13a3 and e14a3 subtypes, characterized by the absence of ABL1 exon 2, collectively account for <1% of CML cases (9). These atypical fusion proteins exhibit structural and functional alterations secondary to ABL1 truncation, potentially influencing leukemia biological behavior, therapeutic responsiveness and clinical outcomes. Recent study has revealed that these atypical variants may regulate cellular signaling pathways and gene expression, influencing the progression of leukemia (3,4). Simvastatin overcomes drug resistance in chronic myeloid leukemia cells to imatinib by inhibiting the PI3K/AKT survival signaling pathway and downregulating its controlled anti-apoptotic proteins (10). Simultaneously, in combination with imatinib, it interferes with Wnt/β-catenin signaling and increases suppressive histone modification to decrease expression of the oncogene. Through these multi-pathway effects, it ultimately induces mitochondrial pathway apoptosis, thereby effectively overcoming imatinib resistance (10). Furthermore, emerging targeted therapies for these atypical variants are under investigation, aiming to improve patient outcomes and overcome the limitations of current treatment strategies (11). Multicenter studies, including the European Treatment Outcome Study (EUTOS) collaborative network, are advancing understanding of atypical BCR-ABL1 fusion genes in CML (4,12). These studies have developed protocols for monitoring these variants using advanced techniques, such as reverse transcription-quantitative (RT-q)PCR, as standard methods are not applicable because its ‘standardized’ or ‘universal’ detection tools (primers and probes) are designed for ‘typical’ or ‘common’ fusion variants. When atypical variants are encountered, these tools cannot bind and recognize them effectively, leading to detection failure (false-negative results). Efforts by EUTOS are focused on refining treatment strategies and establishing guidelines for managing these rare variants (13).
Guidelines from organizations, including the National Comprehensive Cancer Network, emphasize the necessity of detecting specific recurrent genetic abnormalities in bone marrow nucleated cells or peripheral blood leukocytes for optimal risk stratification and treatment planning (14). Recommended methodologies include cytogenetic analysis (karyotyping), interphase fluorescence in situ hybridization (FISH) and RT-PCR for fusion gene detection. Previous investigations have implemented RT-qPCR for JAK2, Calreticulin and myeloproliferative leukemia proto-oncogene gene analysis (15), supplemented with specialized primer sets targeting BCR exon (e)1, 12 and 3 to identify BCR-ABL1 breakpoints, demonstrating comprehensive coverage of previously reported uncommon breakpoints (11,16). Bone marrow smear examination combined with FISH and karyotyping provides preliminary evidence of BCR-ABL1 fusion (17). Next-generation sequencing (NGS) enables genomic analysis through fragmentation of genomic DNA or transcriptomic RNA, library preparation and high-throughput sequencing via fluorescence signal detection during polymerase/ligase-mediated nucleotide incorporation (18). For non-IS standardized transcripts, quantitative calibration and reporting methods, such as relative ABL1 copy number analysis and laboratory-built reference curves, are recommended for improved quantification (19). Droplet digital (dd)PCR), with its defined detection limit and quantification limit, is a key tool for monitoring residual disease levels in these variants, with variant-specific primer design and stringent quality control procedures essential to ensure accuracy. Whole-genome sequencing (WGS) using exon capture techniques facilitates detection of BCR-ABL1 fusions through comprehensive genomic interrogation (20). Additional methodologies, such as nested PCR coupled with agarose gel electrophoresis, have utility in detecting these transcripts, offering enhanced sensitivity and specificity compared with conventional techniques while enabling amplification of extended DNA fragments (21). Recent guidelines from European LeukemiaNet 2023 and EUTOS suggest regular monitoring of measurable residual disease using advanced techniques and more frequent follow-up for patients with atypical transcripts to improve patient management (6,22). Ongoing multicenter collaborations, such as the EUTOS study, are key in providing robust data on the clinical outcomes of these variants. These studies aim to validate the prognostic value of atypical BCR-ABL1 fusion genes and refine treatment strategies for these rare subtypes. Atypical BCR-ABL1 testing should involve multiplex RT-PCR and NGS, followed by ddPCR for minimal residual disease monitoring, providing a structured approach to managing cases with atypical BCR-ABL1 fusion genes (Fig. 1), with follow-up frequency and therapeutic adjustments based on patient response.
Current research on atypical fusion genes in leukemia remains exploratory, with knowledge gaps persisting. The low incidence of these genetic variants in leukemia populations has resulted in limited case reports (23,24), posing diagnostic and therapeutic challenges for clinicians managing patients with atypical fusion-positive CML. The present study aimed to review the structural characteristics therapeutic management, and prognostic implications of the e13a3, e14a3, e1a3, e1a2, e6a2, e8a2, e19a2, e12a2 and e13a1 BCR-ABL1 fusion transcripts to delineate their clinical significance (Figs. 2 and 3).
The literature search was conducted using PubMed(pubmed.ncbi.nlm.nih.gov/), Embase (embase.com/landing?status=grey) and Web of Science(webofscience.com/wos/) from January 2000 to July 2025 using the following search strategy: ((BCR-ABL[Title/Abstract] OR BCR::ABL1[Title/Abstract]) AND (atypical[Title/Abstract] OR rare[Title/Abstract] OR e13a3 OR e14a3 OR e1a3 OR e1a2 OR e6a2 OR e8a2 OR e19a2 OR e12a2 OR e18a2 OR e13a1) AND (CML[Title/Abstract] OR ‘chronic myeloid leukemia’[MeSH Terms] OR Ph + ALL[Title/Abstract]). A two-step ‘include-then-exclude’ process was performed: All case reports, series or retrospective studies in which atypical BCR-ABL1 transcripts were confirmed at the RNA or DNA level, the diagnosis met World Health Organization(WHO) criteria for CML or acute lymphoblastic leukemia(Ph+ ALL) and both treatment details and evaluable follow-up outcomes were provided were eligible (25,26); conversely, reviews, editorials, animal studies lacking primary data and duplicate publications with overlapping cases were excluded, retaining only the most complete dataset for each patient. A total of two reviewers independently screened titles/abstracts, extracted data. Discrepancies resolved by a third reviewer. Because study designs varied widely, the present review conducted a descriptive synthesis. The PRISMA flowchart (Fig. 4) documents the systematic selection process. For each transcript subtype, the strength of evidence was graded hierarchically: Grade A (robust), ≥10 clinically annotated cases with a median follow-up ≥1 year; grade B (moderate), 3–9 cases or follow-up <1 year and grade C (limited), 1–2 cases or in vitro data only (Table I).
The e13a3 (b2a3) BCR-ABL1 transcript is generated through direct linkage of e13 of the BCR gene to e3 (a3) of the ABL1 gene, resulting in deletion of ABL1 e2 (a2). This fusion produces a truncated protein that retains constitutively activated TK activity (27). The lack of the Src homology 3 (SH3) domain in this variant is linked to its unique structural properties, contributing to the formation of an SH3-deficient isoform. SH3 deficiency can impact downstream signaling, enhancing kinase activity and potentially promoting leukemogenesis (28). SH3-deficient variants such as e13a3 are associated with altered protein interactions and subcellular localization, potentially affecting cell signaling pathways and contributing to disease progression. In murine models, e13a3, as an SH3-deficient variant, shows slower disease progression compared with canonical isoforms, though it retains leukemogenic potential, capable of inducing CML (29). This slower progression may be influenced by changes in cell adhesion, which alter the interaction between leukemic cells and the microenvironment, affecting disease dynamics. The SH3 domain normally serves as a negative regulator of ABL1 TK activity, and its deletion in the e13a3 variant enhances kinase activity (7). The e13a3 fusion breakpoint resides within the major breakpoint cluster region, resulting in the production of a 210 kDa (p210) fusion protein. Notably, the absence of the SH3 domain in the truncated ABL1 moiety induces structural alterations in the chimeric protein. This aberrant protein retains constitutively activated TK activity, which drives leukemic cell proliferation and inhibits differentiation (27). Compared with canonical fusion subtypes such as e14a2 or e13a2, this variant exhibits a unique genomic architecture (30). The e13a3 transcript is predominantly observed in patients with chronic phase CML, with rare case reports in Ph chromosome-positive ALL (31–33). Notably, a Chinese study initially failed to detect the e13a3 fusion using RT-qPCR, underscoring the risk of missing rare fusion subtypes when employing primer sets targeting conventional breakpoints, even in cases with confirmed t(9;22) translocation (17). Conversely, another study (34) documented a CML case with a normal karyotype and negative RT-PCR findings, where subsequent FISH analysis revealed BCR-ABL1 fusion. This highlights the importance of multimodal diagnostic approaches, particularly when conventional methods yield equivocal results.
Imatinib, a first-generation TKI is widely utilized in e13a3 variant CML. Most Ph-positive CML patients receiving 400 mg/day imatinib achieve complete cytogenetic remission (CCyR) within 6–12 months and maintain durable responses (35,36). McCarron et al (37) reported a 66-year-old male patient with Ph-positive CML who attained progressively deepening cytogenetic responses and declining BCR-ABL1/ABL1 ratios following sustained 400 mg/day imatinib therapy. In Ph-negative CML cohorts (representing 5–10% of cases, characterized by CR-ABL1 rearrangements undetectable by conventional cytogenetics) (38,39), studies (34,40) have evaluated second-generation TKIs including nilotinib. These agents demonstrate efficacy in achieving CCyR and major molecular response (MMR) in Ph-positive populations (17,27), which is consistent with the report by Zhou et al (41). Dasatinib, another second-generation TKI, has also been employed in this context. Mechanistic and in vitro studies indicate that the e13a3 variant may exhibit resistance to asciminib, with clinical evidence remaining limited (7). Resistance observed in e13a3 variant CML is largely based on laboratory-based research (7,42), and there is insufficient clinical data to support these findings.
Combination strategies integrating TKIs with chemotherapy have been explored. One notable example is the use of the ponatinib-fludarabine + Low-dose Cytarabine (Ara-C) + Granulocyte Colony-Stimulating Factor(G-CSF) + idarubicin regimen followed by allogeneic stem cell transplantation (ASCT), which resulted in molecular negativity and full donor chimerism at 19 months post-transplant in one case (43). Massimino et al (44) implemented a tailored approach in an 89-year-old male patient with mild renal impairment, including initial hydroxyurea (2,000 mg/day) for leukocytosis management, transitioning to dasatinib 100 mg/day, which achieved a deep molecular response (MR), characterized by a further reduction in transcript levels to undetectable. This strategy has also been used in other studies (17,30).
Most studies suggest that e13a3-positive patients exhibit a lower risk of progression to accelerated phase or blast crisis, with superior long-term event-free survival compared with rare variants such as e1a2 or e19a2 (12,45). Most patients attain deep, sustained responses following TKI monotherapy or combination regimens. A patient achieved complete hematological remission (CHR) at 2 months and MMR with RT-PCR negativity by 8 months, maintaining remission for 2 years (44). Another case demonstrated FISH-confirmed CCyR (0% BCR-ABL1 fusion) at 6 months, sustained beyond 24 months (34). While certain TKI-treated cases exhibit persistent low-level e13a3 transcripts despite CCyR (37), combination therapies have shown favorable results. In addition to treatment efficacy, it is key to evaluate how the treatment regimen affects quality of life. Long-term use of TKIs can lead to side effects such as chronic fatigue, nausea and musculoskeletal pain, which may limit the ability to perform daily tasks and participate in social activities (46,47). Balancing treatment effectiveness with the impact on physical and emotional wellbeing is essential for optimal clinical decision-making. Resistance to treatment can develop due to ABL kinase mutations such as T315I or activation of compensatory signaling pathways such as PI3K/AKT and SRC, which allow the leukemic cells to survive despite the presence of TKIs (48). These mechanisms of resistance contribute to treatment failure and disease progression. In these cases, next-generation TKIs such as ponatinib and asciminib, which target resistant mutations, can be effective, although they may be associated with more severe side effects (49–51). Combining TKIs with other therapeutic modalities, including chemotherapy or stem cell transplantation, may be necessary for patients with resistant disease to achieve long-term disease control (52). Evidence on e13a3 variant outcomes is summarized in Table II. Further multi-center studies are needed to validate these prognostic outcomes in CML.
In this fusion transcript, the ABL1 breakpoint resides within intron 2, generating a chimeric mRNA linking BCR e14 to ABL1 e3. This rearrangement induces structural and functional alterations in the fusion protein, dysregulating intracellular signaling pathways to promote leukemic cell proliferation, survival and immune evasion. A previous study employed customized RT-PCR coupled with Sanger sequencing to confirm this fusion mRNA (53), concurrently identifying non-synonymous mutations in TP53, FMS-like tyrosine kinase 3, KIT Proto-Oncogene, Receptor Tyrosine Kinase(KIT) and paired box 5, underscoring the molecular heterogeneity. Another case report documented methylenetetrahydrofolate reductase mutation in a patient with CML harboring this BCR-ABL1 fusion, providing insights for future investigations (54). A study identified a BCR-ABL1 fusion in a rare CML case, where the breakpoints occurred at BCR intron 14 and ABL1 intron 2, using NGS (53). This unique fusion led to a compromised SH3 domain, which was associated with altered drug response and distinct clinical manifestations (53). These findings emphasize the critical role of SH3 domain loss in modulating therapeutic outcomes and the molecular heterogeneity underlying CML.
TKI monotherapy with imatinib or nilotinib has been used in e14a3-positive cases. A Chinese study (11) reported a 67-year-old Ph-positive female with coexisting e13a3 and e14a3 variants, the first documented instance of dual rare BCR-ABL1 fusions in China, who achieved therapeutic response with imatinib. Vaniawala et al (55) reported e14a3 BCR-ABL1 fusion in a 30-year-old male managed solely with imatinib. Nilotinib was similarly employed to treat a 52-year-old male by Massimino et al (56), with both cases achieving a treatment response.
Personalized combination regimens have also been explored. In a study by Lyu et al (53), hydroxyurea was initially administered for rapid leukocytosis control prior to TKI initiation, with subsequent imatinib dose reduction (from 400 to 300 mg/day) due to intolerance, emphasizing individualized dosing. A previous study (57) reported sequential intolerance to imatinib and dasatinib, ultimately transitioning to hydroxyurea monotherapy.
Most patients with e14a3 variant CML exhibit favorable prognoses, achieving sustained hematological, cytogenetic and molecular remissions. In a Chinese cohort (11), imatinib monotherapy induced rapid MR, with CCyR attainment within 3 months and notable BCR-ABL1-e14a3 transcript reduction. Nilotinib-treated cases similarly demonstrated favorable outcomes (56). Combination regimens have shown variable efficacy: One study reported TKI monotherapy achieving CHR at 2 months, MMR at 3 months and sustained transcript negativity for 9 years without kinase domain mutations (21). Adjuvant agents such as interferon, hydroxyurea, and aspirin were incorporated, though immunomodulatory effects of interferon yielded inconsistent results compared with prior reports (57,58). Conversely, a patient requiring hydroxyurea-nilotinib combination therapy achieved hematological and molecular remission after dose adjustment (54). Key e14a3 variant case reports and outcomes are summarized in Table III.
The e1a3 transcript arises from direct fusion of e1 of the BCR gene to e3 (a3) of the ABL1 gene, skipping ABL1 e2 (a2). This structural alteration results in a truncated fusion protein lacking approximately two-thirds of the sequence encoding the SH3 domain within the ABL1 moiety (8). Distinct from common variants, its unique fusion junction may perturb subcellular localization, substrate specificity and signaling pathways, thereby disrupting cellular homeostasis. This transcript is relatively rare, with a single case (4.8%) identified among patients with CML in a Syrian study (59). Additionally, its occurrence has been documented in Ph+ ALL and AML (31). Some researchers have posited that a subset of e1a3 BCR-ABL1-positive ALL cases may represent undiagnosed CML in lymphoid blast crisis, requiring exclusion through comprehensive clinical history review (60). A previous study (61) identified multiple atypical BCR-ABL1 transcripts in CML, challenging prior assumptions of singular fusion dominance. Conventional RT-PCR frequently fails to detect these transcripts, often yielding false-negative results (62), while RNA sequencing (RNA-seq) uncovers their presence, highlighting the necessity for advanced molecular diagnostics in clinical practice. The e1a3 and e6a2 BCR-ABL1 transcripts are characterized by unique fusion breakpoints within the ABL1 gene (63,64). These isoforms are less common and associated with more aggressive disease progression, including early blast crisis and resistance to standard TKIs (65,66). These isoforms demonstrate TKI resistance and often require multimodal therapy, including the use of third-generation TKIs or stem cell transplantation (67,68).
Unlike e13a3 and e14a3 variants, dasatinib serves as the primary TKI for e1a3-positive CML. The majority of reported cases demonstrate an indolent clinical course (62,69). A previous study (64) reported a patient achieving CCyR following immediate dasatinib initiation (140 mg/day). An 80-year-old Ph-positive male treated with 400 mg/day imatinib attained rapid CCyR and hematological normalization but subsequently developed lymphoblastic crisis at 5 months, suggesting a risk of ALL transformation (8). Combination therapies, including dasatinib with nilotinib or ponatinib, have shown variable efficacy (68,70); the T315I mutation frequently serves as the primary resistance mechanism, necessitating the switch to third-generation TKIs (71).
Innovative approaches, such as third-generation TKIs combined with ASCT, have been employed in a previous study (72). A 56-year-old female patient maintained disease-free status post-ASCT with continued olverembatinib therapy, underscoring the potential of next-generation TKIs and ASCT in managing this rare subtype (72).
Prognoses for e1a3 variant CML patients exhibit marked heterogeneity. A Japanese male (64) achieved CCyR by 6 months with dasatinib, despite presenting with extramedullary leukemia lacking leukocytosis, which is rare in CML. A previous case (72) demonstrated sustained remission post-ASCT and olverembatinib maintenance, yet developed isolated central nervous system (CNS) infiltration without hematological/cytogenetic relapse, implicating the CNS as a potential sanctuary site. Due to the blood-brain barrier and relatively immune-privileged status, conventional systemically administered chemotherapeutic and targeted therapeutic agents often fail to achieve effective concentrations within the CNS. This allows cancer cells to evade treatment, survive, and cause a relapse in this sanctuary site, while the rest of the body may still be in a state of remission. A patient harboring the e1a3 fusion, typically associated with aggressive disease, maintained stable, untreated CML, challenging the association between BCR-ABL1 variants and clinical severity (64). Key e1a3 variant case reports and outcomes are summarized in Table IV.
The e1a2 variant arises from fusion between e1 of the BCR gene and e2 of the ABL1 gene, generating a chimeric protein with distinct structural and functional properties. By contrast with the canonical p210 isoform, the p190 variant lacks central e13 and 14 of the BCR gene. Despite this truncation, the p190 fusion protein retains notably enhanced TK activity, which remains sufficient to drive leukemogenesis (73,74). The e1a2 transcript is rare in CML, accounting for ~1.8% of cases in a cohort of 2,322 patients treated with TKIs, including 1,326 male and 996 female patients, with a median age of 48 years (range 18–88) (14,75), In the aforementioned study, 41 patients (1.8%) exhibited the e1a2 fusion, confirmed by RT-PCR. This variant is associated with a distinct phenotype marked by monocytosis, absence of basophilia and blast crisis presentation at initial diagnosis in 61% of cases, significantly higher than in patients with canonical transcripts (76). In a study by Gong et al (52), 16 of 41 patients with the e1a2 transcript presented with blast crisis at initial diagnosis, two had accelerated phase and 23 were in chronic phase (76) The frequency of monocytosis at initial diagnosis was confirmed in 10 patients with available blood counts, showing a median of 11.5% (range, 5–36%), with seven patients exhibiting monocytosis >10% (76). In Ph-positive adult ALL, the e1a2 variant accounts for 61.2% of cases, as reported in a national cohort of 67 patients with Ph+ ALL, and is typically associated with elevated leukocyte count and lymphoid lineage differentiation (77). RT-PCR for BCR-ABL1 detection, and Sanger sequencing are employed to confirm atypical fusion transcripts.
Imatinib remains the primary therapeutic agent for e1a2-positive leukemia. A standard initial dose of 400 mg/day is administered in patients with ALL and CML, with dose escalation or TKI switching considered for suboptimal responders. However, it is important to consider the impact of side effects on daily life (78). Common adverse reactions such as fatigue, gastrointestinal disturbance and skin rashes can significantly disrupt daily activities and affect the overall quality of life (79). These side effects should be weighed when selecting the most appropriate treatment regimen, and supportive care may be necessary to improve patient comfort during therapy (80). A previous study (81) documented a patient with Ph-negative CML achieving molecular remission (undetectable e1a2 BCR-ABL1 by RT-PCR) after 2 months of imatinib monotherapy, alongside rare cyclical leukocyte fluctuations and spontaneous normalization without intervention. However, resistance to TKIs is a major challenge, particularly in patients with mutations in the ABL kinase domain, such as the T315I mutation, which notably impairs the binding of TKIs to the BCR-ABL1 fusion protein (82,83). These mutations lead to decreased efficacy of first- and second-generation TKIs. Additionally, compensatory signaling pathways, including the PI3K/AKT and SRC kinase pathways, may be activated, allowing leukemic cells to bypass the inhibition of BCR-ABL1, contributing to treatment resistance (84). To overcome these mechanisms of resistance, third-generation TKIs such as ponatinib and asciminib, which are designed to target BCR-ABL1 with T315I mutations and other resistant forms, show promising results (50,51,85). However, these agents also cause more severe side effects, including cardiovascular complications, which should be managed. Combining TKIs with other therapeutic strategies, such as chemotherapy or immunotherapy, may provide an alternative approach to overcome resistance, but this requires consideration of the risk-to-benefit ratio for each patient (86–89).
Combination approaches are frequently employed, often incorporating consolidation chemotherapy post-remission (68,90). Japanese protocols (91) combine hydroxyurea with TKIs. For relapsed/refractory cases, TKI substitution or chemotherapy intensification may be pursued. A previous study (92) reported initial imatinib-induced symptom resolution and 2.5-log BCR-ABL1 reduction, followed by leukemic transformation at 6 months necessitating high-dose chemotherapy and nilotinib. Transcript isoform switching may be a potential molecular mechanism underlying disease recurrence (93).
Prognostic heterogeneity characterizes e1a2 variant CML. While some patients achieve durable remission with imatinib-based regimens (81,92,94,95), clonal dynamics complicate outcomes. Multivariable analyses have confirmed that e1a2 BCR-ABL1 serves as an independent adverse prognostic factor, with a median OS of 69.5 months. Given its clinical behavior resembling Ph-positive ALL, certain researchers advocate classifying e1a2 as a distinct high-risk subtype of CML (76). One case (96) harbored dual e13a3 and e1a2 clones, developing imatinib resistance linked to e1a2 persistence despite achieving CHR, ultimately progressing to blast crisis and death. Notably, resistance occurred without ABL1 kinase domain mutations, suggesting alternative mechanisms. A separate study (97) described extramedullary blast crisis at TKI initiation, mirroring e1a3 cases, yet subsequent multimodal therapy (TKIs, ASCT) achieved sustained complete molecular remission (CMR) for >48 months. These findings underscore the necessity for comprehensive molecular monitoring and adaptive therapeutic strategies. Key e1a2 variant case reports and outcomes are summarized in Table V.
e6a2 variant results from fusion between e6 of the BCR gene and e2 of the ABL1 gene. This rearrangement alters the fusion protein structure, conferring distinct TK activity and protein interaction profiles compared with common variants, thereby dysregulating multiple intracellular signaling pathways to drive leukemogenesis (98). This transcript accounts for 0.02–2.30% of all BCR-ABL1-positive CML cases. Although most patients present in chronic phase, up to 40% of cases are diagnosed in accelerated phase or blast crisis, with this variant frequently demonstrating an aggressive clinical course (9,65). This transcript was also detected in a case of ABL (99). Notably, conventional RT-qPCR may fail to detect the e6a2 BCR-ABL1 transcript, necessitating specialized RT-PCR strategies for rare fusion detection (66). Zagaria et al (100) employed ddPCR for e6a2 transcript quantification, leveraging its high sensitivity, absolute quantification without standard curves and multiplexing capabilities. Concurrent additional sex combs-like 1(ASXL1) mutations, identified via NGS in e6a2-positive cases, may synergize with the fusion to promote acute transformation (65). Domains retained or missing in each subtype fusion protein are presented in Table VI.
TKIs including imatinib (101), nilotinib (63) and dasatinib (102) are used for e6a2 variant management, with imatinib remaining the cornerstone. However, certain scholars advocate upfront use of second-generation TKIs or ASCT to circumvent suboptimal responses to imatinib (103). In one CML case (102), initial hydroxyurea therapy for thrombocytosis was discontinued due to neutropenia, followed by successful imatinib 400 mg/day administration. In vitro sensitivity assay measuring Crk-like protein and phosphorylated-Src family kinase (Tyr416) inhibition confirmed imatinib responsiveness, guiding therapeutic decisions (103).
Combination regimens integrating chemotherapy and TKIs are employed in refractory cases. Crampe et al (65) reported a patient achieving hematological and morphological remission (BCR-ABL1/ABL1: 0.06%) with imatinib dose escalation (from 400 to 600 mg/day), though subsequent sepsis necessitated allogeneic ASCT. Furthermore, targeted therapies may confer prognostic benefits in patients harboring co-occurring ASXL1 mutations.
Prognoses for e6a2 variant CML exhibit marked variability, with frequent fatal outcomes. While some achieve sustained remission post-TKI monotherapy (63,102) or ASCT (65), others experience rapid progression. Prognostic indices indicate that despite a subset of patients exhibiting low-risk Sokal scores (63), OS rates remain inferior to those observed in patients with common transcript subtypes. A patient with Ph-positive CML maintained CCyR for 6 months on dasatinib despite notable eosinophilic hyperplasia with atypical precursors (a morphology potentially linked to the e6a2 transcript) (100). Conversely, Beel et al (66) documented rapid blast crisis within 3 months of TKI initiation, culminating in fatal multidrug-resistant bacteremia post-ASCT. Rohon et al (103) advocated early ASCT or clinical trial enrollment for e6a2-positive cases following short-term TKI or dual Src/ABL inhibitor therapy.
Aggressive presentations include iliac sarcoma at diagnosis (104) and novel BCR-ABL1 kinase domain mutations (K245E, L284S) emerging during imatinib therapy, culminating in blast crisis CML and death (105). These findings underscore the need for personalized strategies addressing variant-specific biology. Key e6a2 variant case reports and outcomes are summarized in Table VII.
The e8a2 variant is rare in CML cases. It is characterized by fusion between e8 of the BCR gene and exon a2 of ABL1, with insertion of a 127-bp sequence from e8 of Ral GEF with PH domain and SH3 binding motif 1 (106). Studies demonstrate that the generation of this transcript requires at least three chromosomal breaks (106,107): The first occurs within ABL1 intron 1b, causing inversion and insertion of this region downstream of BCR e8; the second occurs at BCR intron 8, facilitating fusion of BCR e8 with ABL1 a2; the third may involve additional chromosomes, forming a four-way translocation (107). While the BCR-ABL1 e8a2 transcript is predominantly observed in CML, its occurrence in ALL remains rare (106,108). A Uruguayan study (109) documented a rare four-way translocation t(1;17;9;22)(p35;q24;q44;q11) in a 51-year-old female patient with e8a2-positive CML, highlighting the complexity of chromosomal rearrangements in leukemogenesis. Researchers (110) have identified somatic mutations in tumor protein p53 binding protein 2 and cadherin-10 via whole-exome sequencing, absent in typical CML or healthy controls, suggesting potential BCR-ABL1-driven mutagenesis. Burmeister et al (107) proposed a mechanistic model for cryptic exon activation, generating transcripts containing 55-bp ABL1 intron 1b sequences. While the 55-bp insertion has been suggested as a potential prerequisite for sustaining kinase activity (111), documented cases demonstrate that insertion-free e8a2 retains oncoprotein-coding capacity, suggesting molecular heterogeneity (112,113). The e8a2 and e19a2 variants are rare and associated with complex chromosomal rearrangements. These variants can be considered distinct from typical BCR-ABL transcripts. due to their distinct structural features, including the involvement of additional chromosomal breaks that contribute to unique functional properties of the fusion proteins. These isoforms are less commonly associated with early TKI resistance but are often found in patients with advanced disease or when conventional diagnostic techniques fail.
TKI regimens (imatinib, dasatinib, nilotinib) constitute the primary therapeutic approach for e8a2-positive CML, often supplemented by individualized protocols. Dasatinib is prioritized as frontline therapy (110), with adjunctive measures such as thromboprophylaxis and allopurinol administration tailored to patient-specific factors, including age, comorbidity and disease phase. Close clinical monitoring ensures timely regimen optimization.
Most patients with e8a2 variant CML show a tendency towards favorable outcomes under TKI therapy (114); while initial studies associated the e8a2 transcript with thrombocytosis and suggested a poorer prognosis, more recent findings indicate that prognosis may vary, and the evidence remains inconclusive (107,115). Imatinib-treated cases typically demonstrate robust responses, while interferon-intolerant patients achieve CHR and CMR (BCR-ABL1 <0.001%) within 6 weeks, sustained beyond 6 months (110). Tchirkov et al (116) validated real-time RT-PCR for precise molecular monitoring, enabling therapeutic efficacy assessment and relapse prediction. Key e8a2 variant case reports and outcomes are summarized in Table VIII.
The e19a2 (µ-BCR-ABL1) transcript arises from aberrant fusion of BCR intron 19 to ABL1 exon a2, encoding a 230-kDa fusion protein (117). Its formation involves submicroscopic insertion events, resulting in FISH-negative/RT-PCR-positive detection. While p230 retains BCR oligomerization domains and ABL1 TK activity, structural divergence from p210 may compromise kinase-dependent signaling efficiency. Sequencing of cDNA microproducts (118) has identified mutations in ABL1 e4-9, while WGS uncovered a 122-kb ABL1 insertion into the BCR locus (117).
Management of e19a2-positive leukemia involves sequential or combinatorial TKI regimens. Patients frequently achieve MMR through sequential use of nilotinib, dasatinib or ponatinib. Imatinib, though initially employed, is often substituted due to resistance, thrombocytopenia, fluid retention or drug interactions (119,120). Dose escalation may partially restore hematological/cytogenetic responses in resistant cases. For imatinib-resistant patients harboring the E355G mutation, second-generation TKIs such as nilotinib induce major cytogenetic responses, offering alternative therapeutic avenues (121). Allogeneic ASCT is utilized in select cases (122), primarily because it remains the only potentially curative treatment for CML. It becomes a critical salvage treatment option when patients develop resistance to or intolerable severe side effects from multiple TKIs, or when the disease progresses from the chronic phase to the prognostically unfavorable accelerated or blast phase.
The prognosis of patients with e19a2 variant CML is influenced by genetic architecture, therapeutic regimen and individual comorbidities. Evidence indicates a trend towards favorable outcomes, but individual responses differ (119,122). While studies have linked the e19a2 transcript to an indolent phenotype (115,123), accumulating cases demonstrate clinical courses indistinguishable from classic CML (117), with potential heightened aggressiveness (124). Second-generation TKIs such as nilotinib and dasatinib demonstrate robust efficacy, exemplified by a 72-year-old patient with chronic-phase CML who achieved CCyR at 6 months and MMR at 12 months with dasatinib combined with hydroxyurea and interferon adjuncts, underscoring its utility as frontline therapy (120). Disease progression may occur in certain cases, manifesting as leukocytosis or marrow dysplasia. Notably, a patient managed with nilotinib required dose interruptions due to grade 2 hepatotoxicity yet maintained sustained CCyR and deep MR during long-term follow-up, aligning with findings by Crampe et al (121) and Ernst et al (122), which confirmed nilotinib durable efficacy following treatment interruptions (125). Hydroxyurea monotherapy has also stabilized leukocyte counts without complications in select cases (126–128).
Resistance mechanisms pose challenges. An Italian study (118) reported a dasatinib-resistant T315I mutation, typically associated with TKI refractoriness, where dose escalation partially restored hematological and cytogenetic responses, suggesting salvage potential in mutation-positive patients. Clonal evolution, including double Ph chromosomes and tetraploidy detected via FISH and cytogenetics in an imatinib-treated patient (129), culminated in fatal blast crisis within 2 years, emphasizing the need for personalized strategies in e19a2 BCR-ABL1-positive CML. e19a2 variant case reports and outcomes are summarized in Table IX.
The e12a2 variant, a rare subtype, arises from fusion between e12 of the BCR gene and exon a2 of ABL1. Investigators employed primer sets (BCR-10 and ABL1-4) in RT-PCR assays to detect uncommon e12a2 BCR-ABL1 fusion transcripts, identifying an 18-bp insertion derived from ABL1 intron 1b at the junctional site (130). Notably, this isoform may co-occur with common transcripts, suggesting clonal heterogeneity or molecular evolution during disease progression (130).
Therapeutic approaches involve sequential TKIs (imatinib, dasatinib, nilotinib, bosutinib, ponatinib) (130). A 59-year-old male patient with CML who developed resistance to imatinib (130) achieved CCyR and MR3 within 6 months of nilotinib escalation (800 mg/day). Due to cardiovascular adverse events, therapy was subsequently transitioned to ponatinib (15 mg/day), maintaining a MR4 for 6 years. However, management complexity arises from frequent requirement for multiple drugs, dose-limiting toxicity and treatment-associated burdens, necessitating rigorous monitoring. The paucity of reported e12a2 BCR-ABL1 cases underscores the need for expanded cohort studies to elucidate its impact on disease progression and prognosis.
The e18a2 transcript is a rare BCR-ABL1 fusion variant arising from t(9;22)(q34;q11) chromosomal translocation, which juxtaposes e18 of the BCR gene with exon 2 (a2) of ABL1, encoding a 225 kDa fusion protein (p225) (131,132). The breakpoint within the µ-BCR region retains nearly complete BCR sequences, including calcium-binding and GTPase-Activating Protein domains specific for the Rac GTPase and coiled-coil oligomerization motifs, while preserving the intact TK domain of ABL1 (133). The e18a2 transcript is rare in CML, with an estimated incidence <1%. A recent study documented a 49-year-old patient with CML initially misclassified as e19a2-positive; relapse evaluation failed due to negative conventional RT-qPCR targeting common isoforms, highlighting diagnostic challenges (134). Coexistence of e18a2 with e19a2 transcripts further complicates detection (135).
Current therapeutic evidence for e18a2 remains sparse (4,134), though insights may be extrapolated from other rare variants. In a 16-year-old female patient with CML harboring e18a2 (136), initial RQ-PCR failed to detect the transcript, yielding false-negative results. Treatment commenced with hydroxyurea and imatinib 600 mg/day, later decreased to 400 mg/day due to thrombocytopenia. CHR was achieved by day 56, followed by major cytogenetic response by day 106. Customized RQ-PCR monitoring revealed a decline in tumor burden to 1×10−3 by month 15. Imatinib was safely re-escalated to 600 mg/day without relapse, demonstrating favorable tolerability. The prognostic value of e18a2 remains contentious due to limited sample sizes and undefined molecular kinetics.
The e13a1 transcript is characterized by the replacement of the terminal 38 bp of BCR e13 with a 37-bp sequence derived from ABL1 intron 1–2/e1, resulting in bidirectional disruption of exon junction architecture. Notably, a G>A point mutation within the inserted sequence substitutes glutamine with lysine at position 27 (137), potentially altering local charge distribution and impacting drug-binding efficiency, though direct experimental evidence remains lacking. A previous study documented a 69-year-old patient with CML initially yielding negative results with TaqMan RT-q and multiplex PCR assays; subsequent Sanger sequencing of single-step PCR products confirmed the e13a1 transcript. The patient achieved sustained MR (BCR-ABL1/ABL1 levels ranging from MR4.5 to MMR) following imatinib therapy, underscoring therapeutic efficacy while necessitating long-term surveillance.
Certain BCR-ABL1 transcripts reported in the literature are rare (4,61), with their clinical significance poorly defined. For example, e1a4 and e1a5 variants have been described exclusively in Ph+ ALL (61), although large-scale epidemiological data validating their prevalence or clinical relevance are lacking. The e8a4 variant was detected in a patient with Sézary syndrome (138); to the best of our knowledge, however, there have been no subsequent studies investigating this variant, and its direct association with BCR-ABL1-driven oncogenesis requires further exploration. The existence of these rare transcripts suggests certain variants may emerge selectively within specific disease subtypes or individuals. Nevertheless, due to limitations in detection technologies and the paucity of reported cases, numerous potential variants may remain undetected or systematically uncharacterized.
The growing recognition of atypical BCR-ABL1 fusion transcripts in CML underscores the need for nuanced diagnostic and therapeutic strategies (4,26). While canonical isoforms dominate clinical practice, atypical variants such as e13a3, e14a3, e1a3, e1a2, e6a2 and e8a2 exhibit distinct molecular architectures that notably influence disease biology, therapeutic responsiveness and clinical outcomes (139).
Advances in understanding atypical fusions have revealed distinct structural configurations that alter fusion protein function, dysregulating intracellular signaling, proliferation, differentiation and apoptosis. Therapeutic strategies combining TKIs, chemotherapy and ASCT demonstrate variable efficacy across subtypes. While certain patients achieve durable remission, others experience refractory disease or rapid progression, highlighting pronounced inter-variant prognostic heterogeneity. For example, e13a3 and e8a2 variants are frequently associated with indolent disease course and favorable responses to TKIs, as evidenced by sustained cytogenetic and molecular remissions in multiple case series (7,44,110,116). Conversely, patients with e1a2 and e6a2 isoforms experience more rapid disease progression, including accelerated phase or blast crisis. In refractory cases, 5-year OS rates are 40–70%. Notably, patients with e1a2-positive CML often present with lymphoid blast crisis-like features, including monocytosis and absence of basophilia, mirroring Ph-positive ALL. Similarly, e6a2 cases show elevated rates of clonal evolution and resistance mutations, necessitating early escalation to second-generation TKIs or ASCT. These findings emphasize that atypical transcripts are not uniformly benign and require vigilant monitoring. TKI responsiveness varies significantly across atypical variants. While imatinib remains effective for e13a3 and e8a2, e1a2 and e6a2 subtypes often require early transition to second- or third-generation TKIs due to intrinsic resistance. Dose adjustments or combination regimens may salvage responses in resistant cases, as demonstrated in patients with e6a2 achieving molecular remission post-ASCT. However, therapeutic decisions must balance efficacy against toxicity, particularly in elderly or comorbid populations. For example, dose reduction mitigates hepatotoxicity while maintaining remission in e14a3 cases.
Conventional diagnostic methods, such as standard RT-PCR or FISH, may fail to detect rare fusion isoforms due to primer mismatches or cryptic chromosomal rearrangements. For example, e1a3 and e6a2 transcripts are frequently missed by routine assays, leading to delayed diagnosis and inappropriate therapeutic choices. Complementary techniques, including multiplex RT-PCR, nested PCR and RNA-seq, are essential for identifying atypical breakpoints and coexisting mutations that may drive disease progression. ddPCR further enhances sensitivity for minimal residual disease monitoring, particularly for low-abundance transcripts such as e6a2.
Data on atypical transcripts derive predominantly from case reports and small cohorts, limiting statistical power and generalizability. To address these challenges, multicenter collaborative studies are required to expand case accrual and establish robust genomic databases. Mechanistic investigations should delineate molecular pathways and crosstalk between atypical fusions and ancillary signaling networks, informing precision therapeutics. Diagnostic innovation should prioritize high-sensitivity/specificity assays for rare fusion detection, enabling early intervention. Therapeutic development requires variant-tailored approaches integrating genomic profiling, disease stage and patient comorbidities, alongside intensified research into resistance mechanisms and salvage strategies. Longitudinal studies assessing long-term outcomes and survivorship are key to optimize holistic care, refine prognostic stratification and ultimately improve survival for patients with atypical BCR-ABL1-positive leukemia.
Not applicable.
The present study was supported by the Taishan Youth Scholar Foundation of Shandong Province (grant no. tsqn201812140).
Not applicable.
XZ wrote the manuscript and constructed figures and tables. AL, DK and PZ revised the manuscript. YS and NS designed the methodology. Data authentication is not applicable. All authors have read and approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Foroni L, Wilson G, Gerrard G, Mason J, Grimwade D, White HE, de Castro DG, Austin S, Awan A, Burt E, et al: Guidelines for the measurement of BCR-ABL1 transcripts in chronic myeloid leukaemia. Br J Haematol. 153:179–1790. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Stella S, Gottardi EM, Favout V, Barragan Gonzalez E, Errichiello S, Vitale SR, Fava C, Luciano L, Stagno F, Grimaldi F, et al: The Q-LAMP method represents a valid and rapid alternative for the detection of the BCR-ABL1 rearrangement in Philadelphia-positive leukemias. Int J Mol Sci. 20:61062019. View Article : Google Scholar : PubMed/NCBI | |
|
Jajosky AN and Lichtman MA: Uncommon phenotypes of BCR::ABL1-positive chronic myelogenous leukemia. Haematologica. 110:1912–1920. 2025.PubMed/NCBI | |
|
Breccia M: Atypical CML: Diagnosis and treatment. Hematology Am Soc Hematol Educ Program. 2023:476–482. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Yan Z, Shi L, Li W, Liu W, Galderisi C, Spittle C, Spittle C and Li J: A novel Next-generation sequencing assay for the identification of BCR::ABL1 Transcript type and accurate and sensitive detection of TKI-resistant mutations. J Appl Lab Med. 9:886–900. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Cross NCP, Ernst T, Branford S, Cayuela JM, Deininger M, Fabarius A, Kim DDH, Machova Polakova K, Radich JP, Hehlmann R, et al: European LeukemiaNet laboratory recommendations for the diagnosis and management of chronic myeloid leukemia. Leukemia. 37:2150–2167. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Leske IB and Hantschel O: The e13a3 (b2a3) and e14a3 (b3a3) BCR::ABL1 isoforms are resistant to asciminib. Leukemia. 38:2041–2045. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Z: The e1a3 BCR-ABL1 fusion transcript in Philadelphia chromosome-positive acute lymphoblastic leukaemia: A case report. Hematology. 28:21860402023. View Article : Google Scholar : PubMed/NCBI | |
|
Baccarani M, Castagnetti F, Gugliotta G, Rosti G, Soverini S, Albeer A and Pfirrmann M; International BCR-ABL Study Group, : The proportion of different BCR-ABL1 transcript types in chronic myeloid leukemia. An international overview. Leukemia. 33:1173–1183. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Ding L, Chen Q, Chen K, Jiang Y, Li G, Chen Q, Bai D, Gao D, Deng M, Zhang H and Xu B: Simvastatin potentiates the cell-killing activity of imatinib in imatinib-resistant chronic myeloid leukemia cells mainly through PI3K/AKT pathway attenuation and Myc downregulation. Eur J Pharmacol. 913:1746332021. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Zhang Y, Meng X, Chen S, Wang T, Zhang L and Ma X: Chronic myeloid leukemia with two rare fusion gene transcripts of atypical BCR::ABL: A case report and literature review. Medicine (Baltimore). 103:e367282024. View Article : Google Scholar : PubMed/NCBI | |
|
Schäfer V, White HE, Gerrard G, Möbius S, Saussele S, Franke GN, Mahon FX, Talmaci R, Colomer D, Soverini S, et al: Assessment of individual molecular response in chronic myeloid leukemia patients with atypical BCR-ABL1 fusion transcripts: Recommendations by the EUTOS cooperative network. J Cancer Res Clin Oncol. 147:3081–3089. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hoffmann VS, Baccarani M, Hasford J, Castagnetti F, Di Raimondo F, Casado LF, Turkina A, Zackova D, Ossenkoppele G, Zaritskey A, et al: Treatment and outcome of 2904 CML patients from the EUTOS population-based registry. Leukemia. 31:593–60. 20171 View Article : Google Scholar : PubMed/NCBI | |
|
Hughes T, Deininger M, Hochhaus A, Branford S, Radich J, Kaeda J, Baccarani M, Cortes J, Cross NC, Druker BJ, et al: Monitoring CML patients responding to treatment with tyrosine kinase inhibitors: Review and recommendations for harmonizing current methodology for detecting BCR-ABL transcripts and kinase domain mutations and for expressing results. Blood. 108:28–37. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Lin X, Huang H and Chen P: Retrospective analysis of the clinical features of 172 patients with BCR-ABL1-negative chronic myeloproliferative neoplasms. Mol Cytogenet. 13:82020. View Article : Google Scholar : PubMed/NCBI | |
|
Tang Z, Wang W, Toruner GA, Hu S, Fang H, Xu J, You MJ, Medeiros LJ, Khoury JD and Tang G: Optical genome mapping for detection of BCR::ABL1-another tool in our toolbox. Genes (Basel). 15:13572024. View Article : Google Scholar : PubMed/NCBI | |
|
Liu B, Zhang W and Ma H: Complete cytogenetic response to Nilotinib in a chronic myeloid leukemia case with a rare e13a3(b2a3) BCR-ABL fusion transcript: A case report. Mol Med Rep. 13:2635–2638. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Avila M and Meric-Bernstam F: Next-generation sequencing for the general cancer patient. Clin Adv Hematol Oncol. 17:447–454. 2019.PubMed/NCBI | |
|
Sorokin M, Rabushko E, Rozenberg JM, Mohammad T, Seryakov A, Sekacheva M and Buzdin A: Clinically relevant fusion oncogenes: Detection and practical implications. Ther Adv Med Oncol. 14:175883592211441082022. View Article : Google Scholar : PubMed/NCBI | |
|
Lee H, Seo J, Shin S, Lee ST and Choi JR: Development and validation of sensitive BCR::ABL1 fusion gene quantitation using next-generation sequencing. Cancer Cell Int. 23:1062023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang X, Sun H, Su Y and Yi H: Long-term molecular remission after treatment with imatinib in a chronic myeloid leukemia patient with extreme thrombocytosis harboring rare e14a3 (b3a3) BCR::ABL1 transcript: A case report. Curr Oncol. 29:8171–8179. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Blachly JS, Walter RB and Hourigan CS: The present and future of measurable residual disease testing in acute myeloid leukemia. Haematologica. 107:2810–2822. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Denk D, Bradtke J, König M and Strehl S: PAX5 fusion genes in t(7;9)(q11.2;p13) leukemia: A case report and review of the literature. Mol Cytogenet. 7:132014. View Article : Google Scholar : PubMed/NCBI | |
|
Kosik P, Skorvaga M and Belyaev I: Incidence of preleukemic fusion genes in healthy subjects. Neoplasma. 63:659–672. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Elgehama A, Chen W, Pang J, Mi S, Li J, Guo W, Wang X, Gao J, Yu B, Shen Y and Xu Q: Blockade of the interaction between Bcr-Abl and PTB1B by small molecule SBF-1 to overcome Imatinib-resistance of chronic myeloid leukemia cells. Cancer Lett. 372:82–88. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Szuber N, Orazi A and Tefferi A: Chronic neutrophilic leukemia and atypical chronic myeloid leukemia: 2024 update on diagnosis, genetics, risk stratification, and management. Am J Hematol. 99:1360–1387. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Ha J, Cheong JW, Shin S, Lee ST and Choi JR: Chronic myeloid leukemia with rare variant b2a3 (e13a3) BCR-ABL1 fusion. Ann Lab Med. 36:287–289. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Hai A, Kizilbash NA, Zaidi SHH, Alruwaili J and Shahzad K: Differences in structural elements of Bcr-Abl oncoprotein isoforms in Chronic Myelogenous Leukemia. Bioinformation. 10:108–114. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Gross AW, Zhang X and Ren R: Bcr-Abl with an SH3 deletion retains the ability to induce a myeloproliferative disease in mice, yet c-Abl activated by an SH3 deletion induces only lymphoid malignancy. Mol Cell Biol. 19:6918–6928. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Liu LG, Tanaka H, Ito K, Kyo T, Ito T and Kimura A: Chronic myelogenous leukemia with e13a3 (b2a3) type of BCR-ABL transcript having a DNA breakpoint between ABL exons a2 and a3. Am J Hematol. 74:268–272. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Burmeister T, Schwartz S, Taubald A, Jost E, Lipp T, Schneller F, Diedrich H, Thomssen H, Mey UJ, Eucker J, et al: Atypical BCR-ABL mRNA transcripts in adult acute lymphoblastic leukemia. Haematologica. 92:1699–1702. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Crampe M, Kearney L, O'Brien D, Bacon CL, O'Shea D and Langabeer SE: Molecular monitoring in adult philadelphia Chromosome-positive acute lymphoblastic leukemia with the variant e13a3 BCR-ABL1 fusion. Case Rep Hematol. 2019:96350702019.PubMed/NCBI | |
|
Phan CL, Tan SN, Tan SM, Kadir SSSA, Ramli NLM, Lim TO and Ng CC: A variant e13a3 BCR-ABL1 fusion transcript in refractory adult B-cell acute lymphoblastic leukemia achieving complete remission with CAR-Tcell therapy. Cancer Genet. 250-251:20–24. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Duan MH, Li H and Cai H: A rare e13a3 (b2a3) BCR-ABL1 fusion transcript with normal karyotype in chronic myeloid leukemia: The challenges in diagnosis and monitoring minimal residual disease (MRD). Leuk Res. 59:8–11. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Waclaw J, Sacha T and Stoklosa T: Imatinib in the treatment of chronic myeloid leukemia: Current perspectives on optimal dose. Blood and Lymphatic Cancer: Targets Ther. 5:101–108. 2015. | |
|
Fava C, Rege-Cambrin G and Saglio G: Imatinib: The First-Line CML Therapy. Chronic Myeloid Leukemia [Internet]. Hehlmann R: Cham: Springer International Publishing; pp. 49–59. 2021, https://doi.org/10.1007/978-3-030-71913-5_4 View Article : Google Scholar | |
|
McCarron SL, Langabeer SE, Bolger K, Haslam K, Crampe M, Kelly J and Morrell R: Molecular response to imatinib in chronic myeloid leukaemia with a variant e13a3 BCR-ABL1 fusion. Med Oncol. 32:4522015. View Article : Google Scholar : PubMed/NCBI | |
|
Marzocchi G, Castagnetti F, Luatti S, Baldazzi C, Stacchini M, Gugliotta G, Amabile M, Specchia G, Sessarego M, Giussani U, et al: Variant Philadelphia translocations: Molecular-cytogenetic characterization and prognostic influence on frontline imatinib therapy, a GIMEMA Working Party on CML analysis. Blood. 117:6793–6800. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Lim TH, Tien SL, Lim P and Lim AST: The incidence and patterns of BCR/ABL rearrangements in chronic myeloid leukaemia (CML) using fluorescence in situ hybridisation (FISH). Ann Acad Med Singap. 34:533–538. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Masuko M, Furukawa T, Abe T, Wada R, Maruyama S, Kitajima T, Shibasaki Y, Toba K, Okada M and Aizawa Y: A chronic myeloid leukemia patient with atypical karyotype and BCR-ABL e13a3 transcript caused by complex chromosome rearrangement. Int J Hematol. 90:230–234. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou X, Li MR and Shan NN: Chronic myeloid leukemia with the e13a3 atypical fusion gene: A case report. Oncol Lett. 29:3192025. View Article : Google Scholar : PubMed/NCBI | |
|
Soverini S: Resistance mutations in CML and how we approach them. Hematology Am Soc Hematol Educ Program. 2023:469–475. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Xu D, Claudiani S, Naresh K, Mucklow S, Neelakantan P, Yebra E, Apperley JF, Khorashad J and Milojkovic D: Blast crisis of chronic myeloid leukemia with plasmacytoid dendritic cell phenotype associated with a rare fusion transcript, e13a3 BCR-ABL1. Leuk Lymphoma. 60:3090–3091. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Massimino M, Stella S, Tirrò E, Consoli ML, Pennisi MS, Puma A, Vitale SR, Romano C, Zammit V, Stagno F, et al: Efficacy of dasatinib in a very elderly CML patient expressing a rare E13a3 Bcr-Abl1 fusion transcript: A case report. Anticancer Res. 39:3949–3954. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wang YL, Bagg A, Pear W, Nowell PC and Hess JL: Chronic myelogenous leukemia: Laboratory diagnosis and monitoring. Genes Chromosomes Cancer. 32:97–11. 20011 View Article : Google Scholar : PubMed/NCBI | |
|
Caldemeyer L, Dugan M, Edwards J and Akard L: Long-term side effects of tyrosine kinase inhibitors in chronic myeloid leukemia. Curr Hematol Malig Rep. 11:71–79. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Narra RK, Flynn KE and Atallah E: Chronic myeloid leukemia-the promise of tyrosine kinase inhibitor discontinuation. Curr Hematol Malig Rep. 12:415–4123. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Santos FPS and Ravandi F: Advances in treatment of chronic myelogenous Leukemia-new treatment options with tyrosine kinase inhibitors. Leuk Lymphoma. 50 (Suppl 2):S16–S26. 2009. View Article : Google Scholar | |
|
Hughes TP, Mauro MJ, Cortes JE, Minami H, Rea D, DeAngelo DJ, Breccia M, Goh YT, Talpaz M, Hochhaus A, et al: Asciminib in chronic myeloid leukemia after ABL kinase inhibitor failure. N Engl J Med. 381:2315–2326. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Haddad FG, Issa GC, Jabbour E and Yilmaz M: Ponatinib for the treatment of adult patients with resistant or intolerant Chronic-phase chronic myeloid leukemia. Expert Opin Pharmacother. 23:751–758. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cortes JE, Sasaki K, Kim DW, Hughes TP, Etienne G, Mauro MJ, Hochhaus A, Lang F, Heinrich MC, Breccia M, et al: Asciminib monotherapy in patients with chronic-phase chronic myeloid leukemia with the T315I mutation after ≥1 prior tyrosine kinase inhibitor: 2-year follow-up results. Leukemia. 38:1522–1533. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Singh VK and Coumar MS: Chronic myeloid leukemia: Existing therapeutic options and strategies to overcome drug resistance. Mini Rev Med Chem. 19:333–345. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Lyu X, Yang J, Wang X, Hu J, Liu B, Zhao Y, Guo Z, Liu B, Fan R and Song Y: A novel BCR-ABL1 fusion gene identified by next-generation sequencing in chronic myeloid leukemia. Mol Cytogenet. 9:472016. View Article : Google Scholar : PubMed/NCBI | |
|
Chisti MM and Sanders DS: Chronic myeloid leukemia with b3a3 (e14a3) fusion: A rare BCR/ABL rearrangement presenting with thrombocytosis-Does MTHFR polymorphism matter. Case Rep Oncol. 11:485–492. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Vaniawala S, Acharya A, Parekh H and Mukhopadhyaya PN: Rare e14a3 (b3a3) BCR-ABL fusion in chronic myeloid leukemia in India: The threats and challenges in monitoring minimal residual disease (MRD). Anal Cell Pathol (Amst). 36:85–92. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Massimino M, Stella S, Tirrò E, Consoli ML, Pennisi MS, Puma A, Vitale SR, Romano C, Zammit V, Stagno F, et al: Rapid decline of Philadelphia-positive metaphases after nilotinib treatment in a CML patient expressing a rare e14a3 BCR-ABL1 fusion transcript: A case report. Oncol Lett. 18:2648–2653. 2019.PubMed/NCBI | |
|
Jinawath N, Norris-Kirby A, Smith BD, Gocke CD, Batista DA, Griffin CA and Murphy KM: A rare e14a3 (b3a3) BCR-ABL fusion transcript in chronic myeloid leukemia: Diagnostic challenges in clinical laboratory practice. J Mol Diagn. 11:359–363. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
O'Brien SG, Guilhot F, Larson RA, Gathmann I, Baccarani M, Cervantes F, Cornelissen JJ, Fischer T, Hochhaus A, Hughes T, et al: Imatinib Compared with Interferon and Low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 348:994–1004. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Farhat-Maghribi S, Habbal W and Monem F: Frequency of BCR-ABL transcript types in Syrian CML patients. J Oncol. 2016:84208532016. View Article : Google Scholar : PubMed/NCBI | |
|
Martinez-Serra J, Del Campo R, Gutierrez A, Antich JL, Ginard M, Durán MA, Bento L, Ros T, Amat JC, Vidal C, et al: Chronic myeloid leukemia with an e1a3 BCR-ABL fusion protein: Transformation to lymphoid blast crisis. Biomark Res. 2:142014. View Article : Google Scholar : PubMed/NCBI | |
|
Sun H, Yan Z and Zhang S: Three atypical BCR/ABL transcripts detected simultaneously in a Philadelphia-positive acute lymphoblastic leukemia patient showing resistance to tyrosine kinase inhibitors. Int J Hematol. 117:134–136. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Shin SY, Cho JH, Kim HJ, Jang JH, Lee ST and Kim SH: Two cases of acute lymphoblastic leukemia with an e1a3 BCR-ABL1 fusion transcript. Ann Lab Med. 35:159–161. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Manzella L, Tirrò E, Vitale SR, Puma A, Consoli ML, Tambè L, Pennisi MS, DI Gregorio S, Romano C, Tomarchio C, et al: Optimal response in a patient with CML expressing BCR-ABL1 E6A2 fusion transcript with nilotinib therapy: A case report. In Vivo. 34:1481–1486. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Miyashita N, Onozawa M, Suto K, Fujisawa S, Okazaki N, Hidaka D, Ohigashi H, Yasumoto A, Sugita J, Hashimoto D, et al: Aleukemic extramedullary blast crisis as an initial presentation of chronic myeloid leukemia with E1A3 BCR-ABL1 fusion transcript. Intern Med. 61:1049–1054. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Crampe M, Haslam K, Groarke E, Kelleher E, O'Shea D, Conneally E and Langabeer SE: Chronic myeloid leukemia with an e6a2 BCR-ABL1 fusion transcript: Cooperating mutations at blast crisis and molecular monitoring. Case Rep Hematol. 2017:90717022017.PubMed/NCBI | |
|
Beel KA, Lemmens J, Vranckx H, Maertens J and Vandenberghe P: CML with e6a2 BCR-ABL1 transcript: An aggressive entity? Ann Hematol. 90:1241–1243. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Hochhaus A, Breccia M, Saglio G, García-Gutiérrez V, Réa D, Janssen J and Apperley J: Expert Opinion-management of chronic myeloid leukemia after resistance to second-generation tyrosine kinase inhibitors. Leukemia. 34:1495–1502. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Hochhaus A, Ernst T, Eigendorff E and La Rosée P: Causes of resistance and treatment choices of second- and third-line treatment in chronic myelogenous leukemia patients. Ann Hematol. 94 (Suppl 2):S133–S140. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Roman J, Jimenez A, Barrios M, Castillejo JA, Maldonado J and Torres A: E1A3 as a unique, naturally occurring BCR-ABL transcript in an indolent case of chronic myeloid leukaemia. Br J Haematol. 114:635–637. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Chen R and Chen B: The role of dasatinib in the management of chronic myeloid leukemia. Drug Des Devel Ther. 9:773–779. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Sheets JW, Eulitt P, He R, Olteanu H, Coombs CC, Foster MC, Montgomery ND and Zeidner JF: Philadelphia chromosome-positive acute myeloid leukemia with e1a3 BCR-ABL1 fusion transcript. Hemasphere. 4:e4842020. View Article : Google Scholar : PubMed/NCBI | |
|
Qiang X, Wen Q, Li J, Chen S, Tao T, Zhang H, Wang P, Peng X, Feng Y and Zhang X: Isolated central nervous system infiltrated and progressed to acute myeloid leukemia from chronic myeloid leukemia with e1a3 BCR-ABL1 transcript: A rare case report and literature review. Cancer Manag Res. 17:35–43. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
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. 9:2960–2970. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Kearney L, Crampe M and Langabeer SE: Frequency and spectrum of atypical BCR-ABL1 transcripts in chronic myeloid leukemia. Exp Oncol. 42:78–79. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Arana-Trejo RM, Ruíz Sánchez E, Ignacio-Ibarra G, Báez de la Fuente E, Garces O, Gómez Morales E, Castro Granados M, Ovilla Martínez R, Rubio-Borja ME, Solís Anaya L, et al: BCR/ABL p210, p190 and p230 fusion genes in 250 Mexican patients with chronic myeloid leukaemia (CML). Clin Lab Haematol. 24:145–150. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Gong Z, Medeiros LJ, Cortes JE, Zheng L, Khoury JD, Wang W, Tang G, Loghavi S, Luthra R, Yang W, et al: Clinical and prognostic significance of e1a2 BCR-ABL1 transcript subtype in chronic myeloid leukemia. Blood Cancer J. 7:e5832017. View Article : Google Scholar : PubMed/NCBI | |
|
Bhreathnach Ú, Kearney L and Langabeer SE: Prevalence of atypical BCR-ABL1 transcript types in adult Philadelphia chromosome-positive acute lymphoblastic leukemia: Implications for measurable residual disease. Hematol Transfus Cell Ther. 44:130–131. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wasif K, Wasif N and Saif MW: Imatinib-induced ototoxicity in a patient with gastrointestinal stromal tumor (GIST). Cureus. 8:e8482016.PubMed/NCBI | |
|
Shin H, Choi SY, Kee KM, Kim SH, Yang SY, Jung SY, Noh H, Zang DY, Kim DW and Lee JI: Comprehensive analyses of safety and efficacy toward individualizing imatinib dosage in patients with chronic myeloid leukemia. Int J Hematol. 111:417–426. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Joensuu H, Trent JC and Reichardt P: Practical management of tyrosine kinase inhibitor-associated side effects in GIST. Cancer Treat Rev. 37:75–88. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Radojkovic M, Ristic S, Pavlovic S and Colovic M: Molecular response to imatinib in patient with Ph negative p190 BCR-ABL transcript positive chronic myeloid leukemia with cyclic leukocytosis. Leuk Res. 33:e10–e12. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Sun J, Hu R, Han M, Tan Y, Xie M, Gao S and Hu JF: Mechanisms underlying therapeutic resistance of tyrosine kinase inhibitors in chronic myeloid leukemia. Int J Biol Sci. 20:175–181. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Moore FR, Yang F and Press RD: Detection of BCR-ABL1 kinase domain mutations causing imatinib resistance in chronic myelogenous leukemia. Methods Mol Biol. 999:25–39. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Ben-Batalla I, Erdmann R, Jørgensen H, Mitchell R, Ernst T, von Amsberg G, Schafhausen P, Velthaus JL, Rankin S and Clark RE: Axl blockade by BGB324 inhibits BCR-ABL tyrosine kinase Inhibitor-sensitive and -resistant chronic myeloid leukemia. Clin Cancer Res. 23:2289–2300. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yeung DT, Shanmuganathan N and Hughes TP: Asciminib: A new therapeutic option in chronic-phase CML with treatment failure. Blood. 139:3474–3479. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cioccio J and Claxton D: Therapy of acute myeloid leukemia: Therapeutic targeting of tyrosine kinases. Expert Opin Investig Drugs. 28:337–349. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Pan Q, Lu Y, Xie L, Wu D, Liu R, Gao W, Luo K, He B and Pu Y: Recent advances in boosting EGFR tyrosine kinase inhibitors-based cancer therapy. Mol Pharm. 20:829–852. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Illmer T and Ehninger G: FLT3 kinase inhibitors in the management of acute myeloid leukemia. Clin Lymphoma Myeloma. 8 (Suppl 1):S24–S34. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Cui Q, Liang P, Dai H, Cui W, Cai M, Ding Z, Ma Q, Yin J, Li Z, Liu S, et al: Case report: CD38-directed CAR-T cell therapy: A novel immunotherapy targeting CD38-positive blasts overcomes TKI and chemotherapy resistance of myeloid chronic myeloid leukemia in blastic phase. Front Immunol. 13:10129812022. View Article : Google Scholar : PubMed/NCBI | |
|
Khan AM, Munir A, Asrani R and Najjar S: Acute myeloid leukemia with Philadelphia chromosome, near-tetraploidy, and 5q deletion. Cureus. 11:e56062019.PubMed/NCBI | |
|
Ohsaka A, Shiina S, Kobayashi M, Kudo H and Kawaguchi R: Philadelphia Chromosome-positive chronic myeloid leukemia expressing p190(BCR-ABL). Intern Med. 41:1183–1187. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Tucker D, Hamilton MS, Kerr JP, Wickham C and Hunter H: Lytic bone disease as the presenting feature of Philadelphia-positive monosomy 7 myelodysplasia progressing to acute myeloid leukaemia. Gene. 501:219–221. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Stella S, Massimino M, Tirrò E, Vitale SR, Scalise L, Leotta S, Pennisi MS, Puma A, Romano C, Stagno F, et al: B-ALL relapses after autologous stem cell transplantation associated with a shift from e1a2 to e14a2 BCR-ABL transcripts: A case report. Anticancer Res. 39:431–435. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Balatzenko G, Guenova M, Kalinova I, Belcheva M, Hristozova H and Kaleva V: Simultaneous occurrence of ETV6-RUNX1 and BCR-ABL1 (e1a2) transcripts in a child with B-cell acute lymphoblastic leukemia. Cancer Genet Mar. 206:97–101. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Langabeer SE, Crampe M, Haslam K, Kelly J and Cahill MR: Sustained clinical remission despite suboptimal molecular response to imatinib in e1a2 BCR-ABL chronic myeloid leukemia. Leuk Res. 34:e176–e177. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Agirre X, Román-Gómez J, Vázquez I, Jiménez-Velasco A, Larráyoz MJ, Lahortiga I, Andreu EJ, Márquez J, Beltrán de Heredia JM, Odero MD, et al: Coexistence of different clonal populations harboring the b3a2 (p210) and e1a2 (p190) BCR-ABL1 fusion transcripts in chronic myelogenous leukemia resistant to imatinib. Cancer Genet Cytogenet. 60:22–26. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Ai DI, Liu W, Lu G, Patel KP and Chen ZI: Extramedullary blast crisis as initial presentation in chronic myeloid leukemia with the e1a2 BCR-ABL1 transcript: A case report. Mol Clin Oncol. 3:1319–1322. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Dupont M, Jourdan E and Chiesa J: Identification of E6A2 BCR-ABL fusion in a Philadelphia-positive CML. Leukemia. 14:2011–2012. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Staal-Viliare A, Latger-Cannard V, Rault JP, Didion J, Grégoire MJ, Bologna S, Witz B, Jonveaux P, Lecompte T and Rio Y: A case of de novo acute basophilic leukaemia: Diagnostic criteria and review of the literature. Ann Biol Clin (Paris). 64:361–365. 2006.(In French). PubMed/NCBI | |
|
Zagaria A, Anelli L, Coccaro N, Tota G, Casieri P, Cellamare A, Impera L, Brunetti C, Minervini A, Minervini CF, et al: BCR-ABL1 e6a2 transcript in chronic myeloid leukemia: Biological features and molecular monitoring by droplet digital PCR. Virchows Arch. 467:357–363. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Popovici C, Cailleres S, David M, Lafage-Pochitaloff M, Sainty D and Mozziconacci MJ: E6a2 BCR-ABL fusion with BCR exon 5-deleted transcript in a Philadelphia positive CML responsive to Imatinib. Leuk Lymphoma. 46:1375–1377. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Breccia M, Cannella L, Diverio D, Streponi P, Nanni M, Stefanizzi C, Natalino F, Mecarocci S and Alimena G: Isolated thrombocytosis as first sign of chronic myeloid leukemia with e6a2 BCR/ABL fusion transcript, JAK2 negativity and complete response to imatinib. Leuk Res. 32:177–180. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Rohon P, Divoka M, Calabkova L, Mojzikova R, Katrincsakova B, Rusinakova Z, Lapcikova A, Raida L, Faber E, Jarosova M, et al: Identification of e6a2 BCR-ABL fusion in a Philadelphia-positive CML with marked basophilia: Implications for treatment strategy. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 155:187–190. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Torres F, Ivanova-Dragoeva A, Pereira M, Veiga J, Rodrigues AS, Sousa AB, Tavares P and Fernandes AR: An e6a2 BCR-ABL fusion transcript in a CML patient having an iliac chloroma at initial presentation. Leuk Lymphoma. 48:1034–1037. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Schnittger S, Bacher U, Kern W, Haferlach T, Hertenstein B and Haferlach C: A new case with rare e6a2 BCR-ABL fusion transcript developing two new resistance mutations during imatinib mesylate, which were replaced by T315I after subsequent dasatinib treatment. Leukemia. 22:856–858. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
McCarron SL, Kelly J, Coen N, McCabe S, Fay M, O'Dwyer M, Hayden PJ and Langabeer SE: A novel e8a2 BCR-ABL1 fusion with insertion of RALGPS1 exon 8 in a patient with relapsed Philadelphia chromosome-positive acute lymphoblastic leukemia. Leuk Lymphoma. 52:919–921. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Burmeister T, Bullinger L and le Coutre P: The recurrent atypical e8a2 BCR::ABL1 transcript with insertion of an inverted 55 base pair ABL1 Intron 1b sequence: A detailed molecular analysis. Acta Haematol. 146:413–418. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Mroczkowska A, Jaźwiec B, Urbańska-Rakus J, Szymanowska S, Tessmann A, Pająk S, Machnik K, Haus O and Wróbel T: A case report of pediatric acute lymphoblastic leukemia with e8a2 BCR/ABL1 fusion transcript. BMC Med Genomics. 15:202022. View Article : Google Scholar : PubMed/NCBI | |
|
Riva E, Manrique Arechavaleta G, De Almeida C, Costa V, Fernandez Del Campo M, Ifran González S and Uriarte R: A novel e8a2 BCR-ABL1 fusion with insertion of MAST2 exon 2 in a four-way translocation t (1;17;9;22) (p35;q24;q44;q11) in a patient with chronic myeloid leukemia. Leuk Lymphoma. 57:203–205. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Cheng Z, Yan WZ, Liu SF, Hu CH and Zhang GS: Molecular characterization and therapeutic reaction to dasatinib in a CML patient harboring a novel e8a2 BCR-ABL1 transcript with a somatic mutation in TP53BP2 and cadherin-10 genes. Leuk Lymphoma. 59:233–236. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Branford S and Apperley JF: Measurable residual disease in chronic myeloid leukemia. Haematologica. 107:2794–809. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Cundell MJ, Hutter LH, Nunes Bastos R, Poser E, Holder J, Mohammed S, Novak B and Barr FA: A PP2A-B55 recognition signal controls substrate dephosphorylation kinetics during mitotic exit. J Cell Biol. 214:539–554. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Gelens L, Qian J, Bollen M and Saurin AT: The importance of Kinase-phosphatase integration: Lessons from mitosis. Trends Cell Biol. 28:6–21. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Jin C, Zhu X, Xiao M, Liu S, Liu X, Liu J, Xu X, Yi S and Meng L: A Novel e8a2BCR-ABL1 fusion transcript without insertion sequence in a patient with chronic myeloid leukemia. Ann Lab Med. 38:169–171. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Langabeer SE, McCarron SL, Kelly J, Krawczyk J, McPherson S, Perera K and Murphy PT: Chronic myeloid leukemia with e19a2 BCR-ABL1 transcripts and marked thrombocytosis: The role of molecular monitoring. Case Rep Hematol. 2012:4587162012.PubMed/NCBI | |
|
Tchirkov A, Couderc JL, Périssel B, Goumy C, Regnier A, Uhrhammer N, Verrelle P and Berger M: Major molecular response to imatinib in a patient with chronic myeloid leukemia expressing a novel form of e8a2 BCR-ABL transcript. Leukemia. 20:167–168. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
May PC, Reid AG, Robinson ME, Khorashad JS, Milojkovic D, Claudiani S; Genomics England Research Consortium, ; Willis F, Apperley JF and Innes AJ: FISH-negative BCR::ABL1-positive e19a2 chronic myeloid leukaemia: The most cryptic of insertions. BMC Med Genomics. 16:1722023. View Article : Google Scholar : PubMed/NCBI | |
|
Cea M, Cirmena G, Garuti A, Rocco I, Palermo C, Cagnetta A, Moran E, Colombo N, Grasso R, Fugazza G, et al: A T315I mutation in e19a2 BCR/ABL1 chronic myeloid leukemia responding to dasatinib. Leuk Res. 34:e240–e242. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Popovici C, Charbonnier A, Gisserot O, Aguilon P, Rémy V, Olschwang S and Mozziconacci MJ: Y253H mutation appearing in a micro-BCR-ABL (e19a2) CML. Leuk Res. 32:361–362. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Ikeda K, Harada-Shirado K, Matsumoto H, Noji H, Ogawa K and Takeishi Y: Molecular response of e19a2 BCR-ABL1 chronic myeloid leukemia with double Philadelphia chromosome to dasatinib. J Clin Oncol. 34:e130–e133. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Crampe M, Garry J, Langabeer SE and Murphy PT: Sustained molecular response with nilotinib in Imatinib-intolerant chronic myeloid leukaemia with an e19a2 BCR-ABL1 fusion. Hematol Oncol Stem Cell Ther. 9:168–169. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Ernst P, Rinke J, Franke GN, Dicker F, Haferlach T, Ernst T and Hochhaus A: Treatment-free remission after third-line therapy with asciminib in chronic myeloid leukemia with an atypical e19a2 BCR::ABL1 transcript and T315I mutation. Leukemia. 38:2037–2040. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Mondal BC, Majumdar S, Dasgupta UB, Chaudhuri U, Chakrabarti P and Bhattacharyya S: e19a2 BCR-ABL fusion transcript in typical chronic myeloid leukaemia: A report of two cases. J Clin Pathol. 59:1102–1103. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
McCarron SL, Maher K, Kelly J, Ryan MF and Langabeer SE: Rapid evolution to blast crisis associated with a Q252H ABL1 kinase domain mutation in e19a2 BCR-ABL1 chronic myeloid leukaemia. Case Rep Hematol. 2013:4907402013.PubMed/NCBI | |
|
Kajiguchi T, Okuno S, Ohno T and Abe A: Molecular response to nilotinib in a patient with imatinib-intolerant e19a2-positive chronic myeloid leukemia. Intern Med. 53:2801–2804. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Santos FK de S and Maia CN: Patients with sickle cell disease taking hydroxyurea in the hemocentro regional de Montes Claros. Rev Bras Hematol Hemoter. 33:105–109. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Krakoff IH: Clinical and Pharmacologic Effects of Hydroxyurea. Antineoplastic and Immunosuppressive Agents: Part II [Internet]. Sartorelli AC and Johns DG: Springer; Berlin, Heidelberg: pp. 789–792. 1975, https://doi.org/10.1007/978-3-642-65806-8_43 View Article : Google Scholar | |
|
Kamidani R, Chiba N, Kuroda A, Uchida A and Okada H: Successful therapeutic leukapheresis for chronic myeloid leukemia identified by persistent erection: A case report. Cureus. 16:e613512024.PubMed/NCBI | |
|
Oshikawa G, Kurosu T, Arai A, Murakami N and Miura O: Clonal evolution with double Ph followed by tetraploidy in imatinib-treated chronic myeloid leukemia with e19a2 transcript in transformation. Cancer Genet Cytogenet. 199:56–61. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Stella S, Massimino M, Tirrò E, Vitale SR, Accurso V, Puma A, Pennisi MS, DI Gregorio S, Romano C, DI Raimondo F, et al: Detection and clinical implications of a novel BCR-ABL1 E12A2 Insertion/deletion in a CML patient expressing the E13A2 isoform. Anticancer Res. 39:6965–6971. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Alikian M, Gale RP, Apperley JF and Foroni L: Molecular techniques for the personalised management of patients with chronic myeloid leukaemia. Biomol Detect Quantif. 11:4–20. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Suttorp M, Millot F, Sembill S, Deutsch H and Metzler M: Definition, epidemiology, pathophysiology, and essential criteria for diagnosis of pediatric chronic myeloid leukemia. Cancers (Basel). 13:7982021. View Article : Google Scholar : PubMed/NCBI | |
|
Wu CC, Beird HC, Zhang J and Futreal PA: FusionPathway: Prediction of pathways and therapeutic targets associated with gene fusions in cancer. PLoS Comput Biol. 14:e10062662018. View Article : Google Scholar : PubMed/NCBI | |
|
Pretzsch T, Progscha S and Burmeister T: Diagnostic ambiguity caused by an atypical e18a2 BCR::ABL1 transcript in a chronic myeloid leukemia patient. Case Rep Hematol. 2024:94391342024.PubMed/NCBI | |
|
Sheng HX, Zhou LN, Chen JL, Hu GL, Wang QH, Gao DG, Liao L, Yang Y, Sun T, Chen H and Zhang B: Concurrence of e18a2 and e19a2 in Ph (+) chronic myelogenous leukemia: A case report and literature review. Zhonghua Xue Ye Xue Za Zhi. 38:799–802. 2017.(In Chinese). PubMed/NCBI | |
|
van der Velden VHJ, Beverloo HB, Hoogeveen PG and Zwaan CM: A novel BCR-ABL fusion transcript (e18a2) in a child with chronic myeloid leukemia. Leukemia. 21:833–835. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Naumann N, Bross-Bach U, Seifarth W, Fabarius A, Hofmann WK, Saußele S and Spiess B: A new aberrantly spliced BCR-ABL1 transcript variant (e13a1) identified in routine monitoring using different quantitative reverse transcription polymerase chain reaction techniques in a patient with chronic myeloid leukemia. EJHaem. 3:1339–1342. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Callet-Bauchu E, Salles G, Gazzo S, Dalle S, Berger F and Hayette S: Identification of a novel e8/a4 BCR/ABL fusion transcript in a case of a transformed Sézary syndrome. Haematologica. 92:1277–1278. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Wang H, Han C, Gong B, Liu Y, Liu K, Gu R, Wang Y, Wei H, Mi Y, Liu B and Wang J: Clinical features and treatment response to TKIs in chronic myeloid leukemia patients with atypical BCR::ABL1 transcripts. Leuk Res. 155:1077332025. View Article : Google Scholar : PubMed/NCBI | |
|
Zangrando A, Intini F, te Kronnie G and Basso G: Validation of NG2 antigen in identifying BP-ALL patients with MLL rearrangements using qualitative and quantitative flow cytometry: A prospective study. Leukemia. 22:858–861. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Park IJ, Lim YA, Lee WG, Park JS, Kim HC, Lee HJ and Cho SR: A case of chronic myelogenous leukemia with e8a2 fusion transcript. Cancer Genet Cytogenet. 185:106–108. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Lee J, Kim DS, Lee HS, Choi SI and Cho YG: Concurrence of e1a2 and e19a2 BCR-ABL1 fusion transcripts in a typical case of chronic myeloid leukemia. Ann Lab Med. 37:74–76. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Bennour A, Beaufils N, Sennana H, Meddeb B, Saad A and Gabert J: E355G mutation appearing in a patient with e19a2 chronic myeloid leukaemia resistant to imatinib. J Clin Pathol. 63:737–740. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Langabeer SE, McCarron SL, Carroll P, Kelly J, O'Dwyer M and Conneally E: Molecular response to first line nilotinib in a patient with e19a2 BCR-ABL1 chronic myeloid leukemia. Leuk Res. 35:e169–e170. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Jeon EK, Lim J, Kim M, Yahng SA, Lee SE, Cho BS, Kim YJ, Kim HJ, Min CK, Cho SG, et al: The first case of acute lymphoblastic leukemia with the e19a2 BCR-ABL1 transcript: Imatinib therapy followed by unrelated donor transplantation induces a durable molecular response. Leukemia. 25:366–367. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Qin YZ, Jiang Q, Jiang H, Lai YY, Shi HX, Chen WM, Yu L and Huang XJ: Prevalence and outcomes of uncommon BCR-ABL1 fusion transcripts in patients with chronic myeloid leukaemia: Data from a single centre. Br J Haematol. 182:693–700. 2018. View Article : Google Scholar : PubMed/NCBI |