scholarly journals Outgrowth of a CSF3R-mutant clone drives a second myeloproliferative neoplasm in a chronic myeloid leukemia patient: a case report

2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Sarah A. Carratt ◽  
Diana Brewer ◽  
Julia E. Maxson ◽  
Brian J. Druker ◽  
Theodore P. Braun

Abstract Background Chronic myeloid leukemia (CML) and chronic neutrophilic leukemia (CNL) are two myeloproliferative neoplasms with mutually exclusive diagnostic criteria. A hallmark of CML is the Philadelphia chromosome (Ph), which results in a BCR-ABL1 fusion gene and constitutive tyrosine kinase activity. CNL is a Ph-negative neoplasm and is defined in part by the presence of CSF3R mutations, which drive constative JAK/STAT signaling. Case presentation Here, we report the exceedingly rare co-occurrence of two granulocytic myeloproliferative neoplasms in a 69-year old male patient. After an initial diagnosis of chronic myeloid leukemia, the patient’s clinical course was shaped by hematologic toxicity, the emergence of treatment-resistant BCR-ABL1 clones, and the expansion of a CSF3R-mutant clone without ABL1 mutations under selective pressure from tyrosine kinase inhibitors. The emergence of the CSF3R-mutant, neutrophilic clone led to the diagnosis of CNL as a second myeloproliferative neoplasm in the same patient. Conclusions This is the first reported case of CNL arising subsequent to CML, which occurred under selective pressure from targeted therapy in a patient with complex clonal architecture. Patients with such molecularly complex disease may ultimately benefit from combination therapy that targets multiple oncogenic pathways.

1999 ◽  
Vol 189 (9) ◽  
pp. 1399-1412 ◽  
Author(s):  
Shaoguang Li ◽  
Robert L. Ilaria ◽  
Ryan P. Million ◽  
George Q. Daley ◽  
Richard A. Van Etten

The product of the Philadelphia chromosome (Ph) translocation, the BCR/ABL oncogene, exists in three principal forms (P190, P210, and P230 BCR/ABL) that are found in distinct forms of Ph-positive leukemia, suggesting the three proteins have different leukemogenic activity. We have directly compared the tyrosine kinase activity, in vitro transformation properties, and in vivo leukemogenic activity of the P190, P210, and P230 forms of BCR/ABL. P230 exhibited lower intrinsic tyrosine kinase activity than P210 and P190. Although all three oncogenes transformed both myeloid (32D cl3) and lymphoid (Ba/F3) interleukin (IL)-3–dependent cell lines to become independent of IL-3 for survival and growth, their ability to stimulate proliferation of Ba/F3 lymphoid cells differed and correlated directly with tyrosine kinase activity. In a murine bone marrow transduction/transplantation model, the three forms of BCR/ABL were equally potent in the induction of a chronic myeloid leukemia (CML)–like myeloproliferative syndrome in recipient mice when 5-fluorouracil (5-FU)–treated donors were used. Analysis of proviral integration showed the CML-like disease to be polyclonal and to involve multiple myeloid and B lymphoid lineages, implicating a primitive multipotential target cell. Secondary transplantation revealed that only certain minor clones gave rise to day 12 spleen colonies and induced disease in secondary recipients, suggesting heterogeneity among the target cell population. In contrast, when marrow from non– 5-FU–treated donors was used, a mixture of CML-like disease, B lymphoid acute leukemia, and macrophage tumors was observed in recipients. P190 BCR/ABL induced lymphoid leukemia with shorter latency than P210 or P230. The lymphoid leukemias and macrophage tumors had provirus integration patterns that were oligo- or monoclonal and limited to the tumor cells, suggesting a lineage-restricted target cell with a requirement for additional events in addition to BCR/ABL transduction for full malignant transformation. These results do not support the hypothesis that P230 BCR/ABL induces a distinct and less aggressive form of CML in humans, and suggest that the rarity of P190 BCR/ABL in human CML may reflect infrequent BCR intron 1 breakpoints during the genesis of the Ph chromosome in stem cells, rather than intrinsic differences in myeloid leukemogenicity between P190 and P210.


2019 ◽  
Vol 18 ◽  
pp. 153303381987990 ◽  
Author(s):  
Mohammed Hussein Kamareddine ◽  
Youssef Ghosn ◽  
Antonios Tawk ◽  
Carlos Elia ◽  
Walid Alam ◽  
...  

Chronic myeloid leukemia is a myeloproliferative neoplasm that occurs more prominently in the older population, with a peak incidence at ages 45 to 85 years and a median age at diagnosis of 65 years. This disease comprises roughly 15% of all leukemias in adults. It is a clonal stem cell disorder of myeloid cells characterized by the presence of t(9;22) chromosomal translocation, also known as the Philadelphia chromosome, or its byproducts BCR-ABL fusion protein/messenger RNA, leading to the expression of a protein with enhanced tyrosine kinase activity. This fusion protein has become the main therapeutic target in chronic myeloid leukemia therapy, with imatinib displaying superior antileukemic effects, placing it at the forefront of current treatment protocols and displaying great efficacy. Alternatively, nanomedicine and employing nanoparticles as drug delivery systems may represent new approaches in future anticancer therapy. This review focuses primarily on the use of organic nanoparticles aimed at chronic myeloid leukemia therapy in both in vitro and in vivo settings, by going through a thorough survey of published literature. After a brief introduction on the pathogenesis of chronic myeloid leukemia, a description of conventional, first- and second-line, treatment modalities of chronic myeloid leukemia is presented. Finally, some of the general applications of nanostrategies in medicine are presented, with a detailed focus on organic nanocarriers and their constituents used in chronic myeloid leukemia treatment from the literature.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 5455-5455
Author(s):  
Chandralekha Ashangari ◽  
Praveen K. Tumula

Abstract Introduction: Atypical chronic myeloid leukemia (aCML), BCR-ABL1 negative is a rare myelodysplastic syndromes (MDS)/myeloproliferative neoplasm (MPN) for which no current standard of care exists. We present one of the rare presentations of aCML in an elderly patient. Case: A 76 year old male presented to the Hematology clinic for consultation after discharge from local hospital for elevated WBC count. Past medical history was significant for COPD, acid reflux, peripheral arterial disease and hypertension. Physical exam was unremarkable. Initial labs were significant for leukocytosis of 30 k/cu mm, anemia with Hb 10 gm/dl, thrombocytosis 695,000 with neutrophilia of ANC 25,200. Peripheral blood was negative for JAK2 V617F and BCR-ABL. Peripheral blood flow cytometry showed granulocytic left shift with 1.5% myeloblasts. Bone marrow biopsy suggestive of hypercellular marrow (100%) with myeloid predominance, atypical megakaryocytes, increased ring sideroblasts (49% of NRBC), increased blasts (5%) and dysgranulopoeisis over all suggestive of Myelodyplastic Syndrome/Chronic Myeloproliferative Disorder (MDS/MPD). Cytogenetics were positive for U2AF1 positive, CSF3R T6181, CSF3R Q776 pathognomonicof atypical CML and negative for BCR-ABL, FLT3. He was considered transplant ineligible. He was started on Azacitadine and is currently receiving 2nd cycle therapy. He is also receiving darbepoeitin periodically to avoid frequent transfusions. He is currently transfusion independent. Discussion: Increased WBC count (e.g., cutoffs of >40×109/L or 50×109/L), increased percentage of peripheral blood myeloid precursors, female sex, and older age are adverse prognostic factors for overall survival or leukemia-free survival in aCML. aCML cases lack in Philadelphia chromosome. Overall 50-65% of patients show cytogenetic abnormalities. The most frequent is +8 (25%). Other changes such as -7 and del(12p) have also been recurrently observed. Patients with aCML have an estimated median survival between 14 and 30 months. aCML tends to exhibit a more aggressive clinical course than other MDS/MPN subtypes. Figure. Figure. Disclosures No relevant conflicts of interest to declare.


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


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 5134-5134
Author(s):  
Natalia Weich ◽  
Cristian Ferri ◽  
Elena Beatriz Moiraghi ◽  
Raquel Maria Nelida Bengio ◽  
Isabel Amanda Giere ◽  
...  

Abstract Chronic myeloid leukemia (CML) is specifically associated with the t(9;22)(q34;q11) reciprocal translocation giving rise to the Philadelphia chromosome and the subsequent formation of the BCR/ABL1 fusion gene, encoding a constitutively active tyrosine kinase. Advances in targeted therapies in chronic phase CML, notably the use of tyrosine kinase inhibitors (TKIs) such as imatinib mesylate (IM), have achieved successful treatment outcomes. However, some patients fail to achieve optimal response, and a substantial proportion of patients develop resistance to IM, which is frequently associated with mutations in the ABL kinase domain. Although BCR/ABL1 fusion oncogene is a key molecular marker involved in the pathogenesis and the clinical course of CML, molecular or cellular events that initiate leukemogenesis or drive translocation of the BCR/ABL1 genes are incompletely understood and little is known about individual susceptibility to this disease. Moreover, it is still unclear whether BCR/ABL1 oncoprotein alone is sufficient to explain the full range of clinical responses to ITKs. There is mounting evidence that genetic factors may play an important role in susceptibility to CML and variability in drug responsiveness. Polymorphic variants of several genes are linked to variations in expression, function, drug disposition and drug response and are potential factors accounting for susceptibility to complex diseases or drug resistance as they can uniquely influence the quality and quantity of gene product. Association studies have been performed to identify genetic variants associated with CML susceptibility and progression, but data are lacking for Argentina. In the present study, we determined the distribution of polymorphisms on TP53 tumor suppressor gene, drug transporter (ABCB1) and drug-metabolizing (glutathione-S-transferases, GSTs) genes to identify markers of susceptibility and pharmacogenetic response in Argentinian patients with CML. Genomic DNA samples from peripheral blood of 141 patients (69 females/72 males, median age 50.8 ± 1.3 years,) treated with IM and 141 age and sex matched healthy controls were evaluated. IM therapy failure was defined by cytogenetics and qRT-PCR in 2 consecutive studies, finding 76 cases that fail treatment. All individuals provided their informed consent according to institutional guidelines and the study was approved by the Ethics Committee of our Institution. GSTM1 and GSTT1 gene deletion polymorphisms and single nucleotide polymorphisms (SNPs) in GSTP1 (313A> G), TP53 (215C> G) and ABCB1 (3435C> T, 1236 C> T and 2677G>T/A) were determined using PCR-based methods. BCR/ABL1 transcript level was analyzed using RT-PCR and ABL1 mutations were identified by RT-PCR and sequencing. Comparison of genotypes between patients and controls as well as regarding clinical parameters was performed by logistic regression. The Kaplan-Meier curves were analyzed using the log-rank test. The level of significance was p <0.05. The case/control association analysis highlighted that carriers of TP53 CC+CG genotypes influence CML susceptibility (p=0.007; OR: 4.5; CI: 1.5-13.3), but no other markers were significantly involved. Next, the pharmacogenetic analysis was performed associating different genotypes with molecular response and treatment failure. We determined that patients carrying GSTM1 -present genotype did not reached major molecular response (p=0.038; OR: 2.9; CI: 1.16-7.4). In addition, average time to treatment change was significantly lower for carriers of the following genotypes: ABCB1 1236TT (23 months), ABCB1 3435TT (13.6 months) and GSTM1 -present (53 months), respect to patients with other variants (p=0.017; p=0.0046 and p=0.04, respectively). Finally, patients with TP53 CC+CG polymorphisms had an increased risk of progression (p=0.039) and lower overall survival (p=0.017). These results indicate that TP53 genotype may represent a genetic cofactor that influences susceptibility to CML and genotypic variations of TP53, ABCB1 and GSTM1 may modulate the response to imatinib. Disclosures Moiraghi: Bristol Myers Squibb: Speakers Bureau; Novartis: Speakers Bureau. Bengio:Novartis: Speakers Bureau; Bristol Myers Squibb: Speakers Bureau.


Sign in / Sign up

Export Citation Format

Share Document