scholarly journals Lymphoid blast transformation in an MPN with BCR-JAK2 treated with ruxolitinib: putative mechanisms of resistance

2021 ◽  
Vol 5 (17) ◽  
pp. 3492-3496
Author(s):  
Justin Anthony Chen ◽  
Yanli Hou ◽  
Krishna M. Roskin ◽  
Daniel A. Arber ◽  
Charles D. Bangs ◽  
...  

Abstract The basis for acquired resistance to JAK inhibition in patients with JAK2-driven hematologic malignancies is not well understood. We report a patient with a myeloproliferative neoplasm (MPN) with a BCR activator of RhoGEF and GTPase (BCR)–JAK2 fusion with initial hematologic response to ruxolitinib who rapidly developed B-lymphoid blast transformation. We analyzed pre-ruxolitinib and blast transformation samples using genome sequencing, DNA mate-pair sequencing (MPseq), RNA sequencing (RNA-seq), and chromosomal microarray to characterize possible mechanisms of resistance. No resistance mutations in the BCR-JAK2 fusion gene or transcript were identified, and fusion transcript expression levels remained stable. However, at the time of blast transformation, MPseq detected a new IKZF1 copy-number loss, which is predicted to result in loss of normal IKZF1 protein translation. RNA-seq revealed significant upregulation of genes negatively regulated by IKZF1, including IL7R and CRLF2. Disease progression was also characterized by adaptation to an activated B-cell receptor (BCR)–like signaling phenotype, with marked upregulation of genes such as CD79A, CD79B, IGLL1, VPREB1, BLNK, ZAP70, RAG1, and RAG2. In summary, IKZF1 deletion and a switch from cytokine dependence to activated BCR-like signaling phenotype represent putative mechanisms of ruxolitinib resistance in this case, recapitulating preclinical data on resistance to JAK inhibition in CRLF2-rearranged Philadelphia chromosome-like acute lymphoblastic leukemia.

2020 ◽  
Vol 21 (16) ◽  
pp. 5776 ◽  
Author(s):  
Lukasz Komorowski ◽  
Klaudyna Fidyt ◽  
Elżbieta Patkowska ◽  
Malgorzata Firczuk

Philadelphia chromosome (Ph) results from a translocation between the breakpoint cluster region (BCR) gene on chromosome 9 and ABL proto-oncogene 1 (ABL1) gene on chromosome 22. The fusion gene, BCR-ABL1, is a constitutively active tyrosine kinase which promotes development of leukemia. Depending on the breakpoint site within the BCR gene, different isoforms of BCR-ABL1 exist, with p210 and p190 being the most prevalent. P210 isoform is the hallmark of chronic myeloid leukemia (CML), while p190 isoform is expressed in majority of Ph-positive B cell acute lymphoblastic leukemia (Ph+ B-ALL) cases. The crucial component of treatment protocols of CML and Ph+ B-ALL patients are tyrosine kinase inhibitors (TKIs), drugs which target both BCR-ABL1 isoforms. While TKIs therapy is successful in great majority of CML patients, Ph+ B-ALL often relapses as a drug-resistant disease. Recently, the high-throughput genomic and proteomic analyses revealed significant differences between CML and Ph+ B-ALL. In this review we summarize recent discoveries related to differential signaling pathways mediated by different BCR-ABL1 isoforms, lineage-specific genetic lesions, and metabolic reprogramming. In particular, we emphasize the features distinguishing Ph+ B-ALL from CML and focus on potential therapeutic approaches exploiting those characteristics, which could improve the treatment of Ph+ B-ALL.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 4362-4362
Author(s):  
Kazuhiro Nishii ◽  
Fumihiko Monma ◽  
Felipe Lorenzo ◽  
Naoyuki Katayama ◽  
Hiroshi Shiku

Abstract The occurrence of acute bilineage leukemia is thought to be the malignant transformation of a myeloid or lymphoid leukemic progenitor with the potential to differentiate into the other lineages; however, the mechanisms of this lineage switch are not well understood. Here, we show the extremely rare case of adult Philadelphia chromosome positive acute bilineage leukemia, characterized by T-cell acute lymphoblastic leukemia, CD7+CD5+CD14−, and acute myelomonocytic leukemia, CD7−CD5−CD14+. Chromosome analysis showed 46,XY,del(7)(p11.2),t(9;22)(q34;q11.2) in all metaphase and leukemic cells expressed a minor BCR/ABL chimeric gene. When the CD5+CD14− and CD5−CD14+ cells were sorted, a fusion gene of BCR/ABL and a same clonal rearranged band of a T-cell receptor (TCR) gene were detected in both populations. Nucleotide sequencing of the TCRg gene revealed the clonal rearrangement of the V8-JGT2 complex in both populations. Over-expression of PU.1, which plays a fundamental role in myelomonocyte development was found in the sorted CD34+CD7+ and CD5−CD14+, but not CD5+CD14− cells. These results suggest that leukemic progenitor cells in the T-lineage with del(7),t(9;22) chromosome have the potential to differentiate into myeloid lineage and enforced PU.1 expression may contribute in part of this phenomenon. Studies of bilineage leukemia will be important for the understanding of lineage commitment and switch in hematopoietic cells.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2574-2574 ◽  
Author(s):  
Hiroaki Shimizu ◽  
Akihiko Yokohama ◽  
Hiroko Koya ◽  
Rumi Shimizu ◽  
Nahoko Hatsumi ◽  
...  

Abstract Abstract 2574 Background: Mixed phenotype acute leukemia (MPAL) has historically been known as biphenotypic acute leukemia (BAL), and novel diagnostic criteria for this disease entity are described in the World Health Organization (WHO) classification 4th edition. As the most common recurrent genetic abnormality observed in MPAL is the bcr-abl fusion gene, Philadelphia chromosome-positive MPAL (Ph+MPAL) has been recognized as one distinctive disease entity. The prognosis of Ph+B-cell acute lymphoblastic leukemia (Ph+B-ALL) has been dramatically improved with the introduction of imatinib, and the goal of this study was to determine whether imatinib results in a survival benefit in the context of Ph+MPAL. Patients and Methods: We retrospectively analyzed 42 consecutive adult patients who were diagnosed with Ph+AL between January 2001 and March 2012 at Gunma University Hospital and Saiseikai Maebashi Hospital in Gunma, Japan. Ph+AL was diagnosed based on detection of the bcr-abl fusion gene with the polymerase chain reaction method and the presence of more than 20% of blasts in the peripheral blood and/or bone marrow. Patients with a previous history of chronic myelogenous leukemia were excluded. The lineage of leukemia cells was defined according to the WHO classification 4th edition. All patients received intensive chemotherapy and concurrent administration of imatinib. The c2-test was used for comparison of binary variables. The Mann-Whitney U test was used for comparison of continuous variables. Overall survival (OS) rate was estimated by the Kaplan-Meier method and were compared using the log-rank test. P < 0.05 was considered as statistically significant. Results: According to the WHO classification 4th edition, 13 (31%) patients were categorized as Ph+MPAL (positive for both myeloid and B-cell lineage), 27 (64%) patients were categorized as B-cell lineage acute lymphoblastic leukemia (Ph+B-ALL), and two (5%) patients were categorized as acute myeloid leukemia (Ph+AML). Patients with Ph+AML were excluded from this study, as the number of patients was relatively small. Of the 40 Ph+AL patients, 23 patients were men, and 17 were women, and the median age was 53 years (range, 16–75 years). Age, sex, white blood cell counts, lactate dehydrogenase levels, and the prevalence of additional cytogenetic abnormalities at diagnosis were not significantly different when comparing the groups, although patients with Ph+MPAL showed significantly higher frequency of major bcr-abl gene than those with Ph+B-ALL (69% and 19%, respectively; p < 0.01). Immunophenotypic analysis revealed that Ph+MPAL patients expressed CD10 and CD34 with significantly lower frequency than Ph+B-ALL patients. Notably, positivity of myeloid antigens (CD13 and 33) was similar between both groups. The complete response (CR) rates after the initial induction therapy were not significantly different when comparing Ph+MPAL and Ph+B-ALL (100% vs. 85%, respectively, p = 0.14). Likewise, the 5-year-OS rate was similar when comparing patients with Ph+MPAL and Ph+B-ALL (55% vs. 53%, respectively, p = 0.87). Of the 13 patients with Ph+MPAL, six patients received AML-type chemotherapy, and seven patients received ALL-type chemotherapy as the initial induction therapy. All patients achieved CR after the initial induction therapy, and there was no significant difference in 5-year OS according to the therapeutic strategy (AML-type vs. ALL-type), (50% vs. 63%, respectively, p = 0.71). Among 12 patients younger than 65 years old who were alive at >3 months after the diagnosis, eight patients underwent allogeneic hematopoietic stem cell transplantation (allo SCT). The 5-year OS was significantly better for patients who underwent allo-SCT than for those who received chemotherapy alone (58% vs. 20%, respectively, p = 0.05). Conclusion: Among adult Ph+AL patients, mixed phenotype was more frequently observed than expected, and Ph+MPAL patients showed unique clinical features, including immunophenotype and the type of bcr-abl fusion gene. Although BAL has been considered as a negative prognostic factor, Ph+MPAL patients showed comparable prognosis to those with Ph+B-ALL who received imatinib-containing intensive chemotherapy. Therefore, the established theory that mixed phenotype is associated with poor outcomes should be revisited among these patients. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
1999 ◽  
Vol 94 (6) ◽  
pp. 2048-2055
Author(s):  
A. Cignetti ◽  
E. Bryant ◽  
B. Allione ◽  
A. Vitale ◽  
R. Foa ◽  
...  

CD34+ hematopoietic stem cells from normal individuals and from patients with chronic myelogenous leukemia can be induced to differentiate into dendritic cells (DC). The aim of the current study was to determine whether acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL) cells could be induced to differentiate into DC. CD34+ AML-M2 cells with chromosome 7 monosomy were cultured in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor  (TNF), and interleukin-4 (IL-4). After 3 weeks of culture, 35% of the AML-M2 cells showed DC morphology and phenotype. The DC phenotype was defined as upmodulation of the costimulatory molecules CD80 and CD86 and the expression of CD1a or CD83. The leukemic nature of the DC was validated by detection of chromosome 7 monosomy in sorted DC populations by fluorescence in situ hybridization (FISH). CD34+ leukemic cells from 2 B-ALL patients with the Philadelphia chromosome were similarly cultured, but in the presence of CD40-ligand and IL-4. After 4 days of culture, more than 58% of the ALL cells showed DC morphology and phenotype. The leukemic nature of the DC was validated by detection of the bcr-abl fusion gene in sorted DC populations by FISH. In functional studies, the leukemic DC were highly superior to the parental leukemic blasts for inducing allogeneic T-cell responses. Thus, CD34+ AML and ALL cells can be induced to differentiate into leukemic DC with morphologic, phenotypic, and functional similarities to normal DC.


2021 ◽  
Author(s):  
Enrico Bracco ◽  
M. Shahzad Ali ◽  
Stefano Magnati ◽  
Giuseppe Saglio

The aberrant tyrosine phosphorylation, either due to constitutive tyrosine kinases (TKs) or to inactivation of protein tyrosine phosphatases (PTPs), is a widespread feature of many cancerous cells. The BCR-ABL fusion protein, which arises from the Philadelphia chromosome, is a molecular distinct and peculiar trait of some kind of leukemia, namely Chronic Myeloid and Acute Lymphoblastic Leukemia, and displays constitutive tyrosine kinase activity. In the chapter, we will highlight the milestones that had led to the identification of the BCR-ABL fusion gene and its role as the only molecular pathogenic event sufficient to elicit and sustain chronic myeloid leukemia. We will also discuss the effort made to unveil the molecular mechanisms of action of the chimeric tyrosine kinase that eventually lead to aberrant cell proliferation and impaired cell-death. Furthermore, we will also review the lesson learned from the selective inhibition of BCR-ABL which currently represent a breakthrough in the treatment of several tumors characterized by defective tyrosine kinase activity.


2021 ◽  
Vol 10 (16) ◽  
pp. 1182-1184
Author(s):  
Shakib Hasan Sheikh ◽  
Vaishali Tembhare ◽  
Seema Singh ◽  
Savita Pohekar ◽  
Samruddhi Gujar

Excessive growth of mature granulocytes in the bone marrow induces chronic myeloid leukemia. The excess neoplastic granulocytes travel massively into the peripheral blood and in the end invade the liver and spleen. The protein encoded on the Philadelphia chromosome by the newly created BCR-ABL gene interferes with normal cell cycle activities, including regulating cell proliferation. Philadelphia chromosome is present in 90 - 95 percent of chronic myeloid leukemia (CML) patients. Their involvement is often a vital indicator of persistent disease or posttreatment relapse. However, for the diagnosis of CML, the presence of the Philadelphia chromosome is not specific since it is also present in acute lymphocytic leukemia (ALL) and rarely in acute myeloid leukemia (AML). 1 Chronic myeloid leukemia is a myeloproliferative neoplasm (MPN) characterised by involvement of the fusion gene BCRABL1 located in the Philadelphia chromosome. In reactive neutrophilia or chronic neutrophilic leukemia, the Ph chromosome is pathognomonic to CML and is never registered.2,3


2021 ◽  
Vol 28 (3) ◽  
pp. 1790-1794
Author(s):  
Hans G. Drexler ◽  
Stefan Nagel ◽  
Hilmar Quentmeier

Chronic neutrophilic leukemia (CNL) is a rare myeloproliferative neoplasm that is genetically characterized by the absence of both the Philadelphia chromosome and BCR-ABL1 fusion gene and the high prevalence of mutations in the colony-stimulating factor 3 receptor (CSF3R). Additional disease-modifying mutations have been recognized in CNL samples, portraying a distinct mutational landscape. Despite the growing knowledge base on genomic aberrations, further progress could be gained from the availability of representative models of CNL. To address this gap, we screened a large panel of available leukemia cell lines, followed by a detailed mutational investigation with focus on the CNL-associated candidate driver genes. The sister cell lines CNLBC-1 and MOLM-20 were derived from a patient with CNL and carry CNL-typical molecular hallmarks, namely mutations in several genes, such as CSF3R, ASXL1, EZH2, NRAS, and SETBP1. The use of these validated and comprehensively characterized models will benefit the understanding of the pathobiology of CNL and help inform therapeutic strategies.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 4390-4390
Author(s):  
Hilmar Quentmeier ◽  
Jan Cools ◽  
Roderick A.F. MacLeod ◽  
Peter Marynen ◽  
Cord C. Uphoff ◽  
...  

Abstract The Philadelphia chromosome (Ph) is the result of the chromosomal translocation t(9;22)(q34;q11), leading to the BCR-ABL fusion gene. The Ph chromosome is the hallmark of chronic myeloid leukemia, but is also detected in acute lymphoblastic leukemia (ALL), particularly in adults. The majority of Ph-positive ALL cases belong to the category of B-cell precursor ALL, whereas Ph-positive T-ALL cases are rather rare. The MHH-TALL1 cell line was established from the peripheral blood of an 11-year-old boy with T-ALL in 1993. PCR analysis of primary tumor cells failed to reveal the existence of a BCR-ABL fusion. Interestingly however, conventional cytogenetics and fluorescence in situ hybridization performed on the cell line showed a 3-way t(1;9;22)(q32;q34;q11) rearrangement effecting Ph formation. As with the patient, standard RT-PCR of the various known BCR-ABL fusion transcripts was negative in the cell line. However, a weak band, about 600 bp larger than the usual e1-a2 BCR-ABL transcript was detected, and subsequently confirmed on reanalysis after optimizing PCR conditions. Sequencing of the RT-PCR product showed that MHH-TALL1 expressed an e6-a2 BCR-ABL fusion transcript. Northern and Western blot analyses demonstrated that the BCR-ABL gene products were expressed at very low levels only. It may be speculated that the presence of this novel BCR-ABL variant has been overlooked in previous analyses, because (i) PCR conditions used to screen for BCR-ABL fusion transcripts were not optimal to detect this variant and (ii) a weak signal running at the “wrong” size might have been neglected. In summary, we report a novel BCR-ABL fusion variant expressed in a T-ALL cell line. Our data raise the intriguing possibility that some BCR-ABL negative cases may express the novel e6-a2 transcript described herein. This might have an impact on treatment of the respective patients with ABL kinase inhibitors.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 5349-5349
Author(s):  
Jennifer McNeer ◽  
Gordana Raca ◽  
Sandeep Gurbuxani ◽  
Jodi Mayfield ◽  
Wendy Stock

Abstract Background: The success in treatment of B acute lymphoblastic leukemia (B-ALL) is partially attributable to careful risk stratification of patients, based largely on biology of disease and disease response to therapy. Recently, gene expression profiling and subsequent high-throughput sequencing of B-ALL has resulted in the identification of a novel high-risk subset of patients with an activated kinase signature similar to patients with Philadelphia chromosome (Ph)-positive ALL, which is currently known as Ph-like ALL (Mullighan et al Nature 2007, Den Boer et al Lancet Oncol 2009). Identification of patients with Ph-like ALL using standard diagnostic techniques will be of great clinical importance since the activated kinases present in these leukemias may be therapeutically targeted. Here we report two patients with B-ALL who had residual disease after induction, neither of whom had a classic high-risk karyotype. Using chromosomal microarray analysis (CMA) we identified targetable kinase mutations in each patient that resulted in a change in therapy and eradication of minimal residual disease (MRD). Patient 1: An 11 year old female presented with fever, malaise, and decreased appetite. The initial WBC count was 28.2 K/µL with 65% blasts, hemoglobin was 5.9 gm/dL, and platelets were 46 K/µL. Flow cytometry confirmed B-ALL. Cytogenetic analysis revealed the following karyotype: 46,XX,add(5)(q13.3),add(6)(q13)[13]/46,XX[5]. She was started on four-drug induction chemotherapy, but at the end of induction, there were 90% blasts in the marrow, consistent with induction failure. Using CMA, multiple regions of copy number loss were detected, one of which was a microdeletion at 5q32-q33.3 with breakpoints located in the EBF1 and PDGFRB genes. Fluorescence in situ hybridization (FISH) analysis confirmed the presence of a PDGFRB rearrangement. The deletion likely resulted in the EBF1-PDGFRB fusion, previously described in rare cases of high-risk B-ALL. Based on previous reports that hematologic malignancies with PDGFRB fusions respond to the addition of a tyrosine kinase inhibitor (TKI) to therapy, imatinib was added to the chemotherapy backbone, and then dasatinib for broader tyrosine kinase inhibition. At the end of 8 weeks of this consolidation treatment there was no morphologic or immunophenotypic evidence of residual leukemia. Patient 2: A 28 year old male presented with abdominal pain, fatigue, and weight loss. The initial WBC was 210.7 K/µL with 94% blasts, hemoglobin was 8.8 gm/dL, and platelets were 43 K/µL. Flow cytometry confirmed B-ALL, and cytogenetic analysis showed one abnormal clone: 45,X,-Y,del(9)(p13p24)[11]. The patient received a four-drug induction, and at the end of induction therapy, MRD was present at a level of 4%. After 8 weeks of consolidation chemotherapy, MRD remained at a level of 1.3%. Several aberrations were detected by CMA, including a 360 kb copy number gain within the 9q34.12-34.13 segment of the long arm of chromosome 9. It involved the chromosome segment between the NUP214 and ABL1 genes, likely resulting in formation of a NUP214-ABL1 fusion gene and upregulation of the ABL1 kinase activity. Dasatinib was added to the chemotherapy regimen, and eight weeks later the marrow had no evidence of MRD. Discussion: We successfully employed CMA to identify 2 patients with B-ALL and a targetable fusion gene. Until now, CMA has been used to identify submicroscopic genetic lesions in ALL, often in genes that drive leukemogenesis. It has not yet been used for identification of patients with Ph-like ALL. Based on these two index cases, we have now begun prospectively to analyze leukemic blasts of patients with B-ALL who do not have classically prognostic cytogenetics using CMA and a panel of selected FISH probes. Using this approach, we have been able to identify Ph-like genetic lesions, and we plan to utilize this information to tailor therapy with early introduction of an appropriate TKI in order to achieve MRD-negative remissions. Figure 1 Figure Legend: CMA results for patients 1 and 2. Top panel: Array plot showing the 8.6Mb 5q32-q33.3 deletion (red bar) in patient 1, that fuses exon 11 of the EBF1 gene with exon 15 of PDGFRB. Bottom panel: The 360kb duplication at 9q34 (blue bar) in patient 2, with the breakpoints within the ABL1 gene and the NUP214 gene, resulting in the NUP214-ABL1 fusion. Figure 1. Figure Legend: CMA results for patients 1 and 2. Top panel: Array plot showing the 8.6Mb 5q32-q33.3 deletion (red bar) in patient 1, that fuses exon 11 of the EBF1 gene with exon 15 of PDGFRB. Bottom panel: The 360kb duplication at 9q34 (blue bar) in patient 2, with the breakpoints within the ABL1 gene and the NUP214 gene, resulting in the NUP214-ABL1 fusion. Disclosures Off Label Use: Use of tyrosine kinase inhibitors as adjunct therapy in Ph-like B-ALL.


Sign in / Sign up

Export Citation Format

Share Document