Poster: MPN-207: Is the Absence of JAK2V617F Mutation a Risk Factor for Bleeding in Philadelphia Chromosome-Negative Myeloproliferative Neoplasms?

2021 ◽  
Vol 21 ◽  
pp. S234
Author(s):  
Shima A. Ahmed ◽  
Ghada E.M Abdallah ◽  
Mai M Aly
2014 ◽  
Vol 60 (2) ◽  
pp. 44-48
Author(s):  
Annamária Szántó ◽  
Zsuzsanna Pap ◽  
Z Pávai ◽  
I Benedek ◽  
Judit Beáta Köpeczi ◽  
...  

Abstract Background: The elucidation of the genetic background of the myeloproliferative neoplasms completely changed the management of these disorders: the presence of the Philadelphia chromosome and/or the BCR-ABL oncogene is pathognomonic for chronic myeloid leukemia and identification of JAK2 gene mutations are useful in polycytemia vera (PV), essential thrombocytemia (ET) and myelofibrosis (PMF). The aim of this study was to investigate the role of molecular biology tests in the management of myeloproliferative neoplasms. Materials and methods: We tested the blood samples of 117 patients between April 2008 and February 2013 at the Molecular Biology of UMF Târgu Mureș using RQ-PCR (for M-BCR-ABL oncogene) and/or allele-specific PCR (for JAK2V617F mutation). Results: Thirty-two patients presented the M-BCR-ABL oncogene, 16 of them were regularly tested as a follow-up of the administered therapy: the majority of chronic phase patients presented decreasing or stable values, while in case of accelerated phase and blast phase the M-BCR-ABL values increased or remained at the same level. Twenty patients were identified with the JAK2V617F mutation: 8 patients with PV, 4 with ET, 3 with PMF, 4 with unclassifiable chronic myeloproliferative disease and 1 patient with chronic myelomonocytic leukemia. There was no case of concomitant occurance of both molecular markers. Conclusions: Molecular biology testing plays an important role in the management of myeloproliferative neoplasms: identification of the molecular markers confirms the final diagnosis, excluding secondary causes of abnormal blood count parameters. Regular monitoring of MBCR- ABL expression level is useful in the follow-up of therapeutic efficiency.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 809-809
Author(s):  
Hajime Akada ◽  
Saeko Akada ◽  
Dongqing Yan ◽  
Robert Hutchison ◽  
Golam Mohi

Abstract Abstract 809 The activating JAK2V617F mutation is the most common mutation found in Philadelphia chromosome (Ph)-negative myeloproliferative neoplasms (MPNs), which include polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). Although a majority of MPN patients carry heterozygous JAK2V617F mutation, loss of heterozygosity (LOH) on chromosome 9p involving JAK2 has been observed in ∼30% of patients with MPNs particularly in PV and PMF. JAK2V617F homozygosity through 9pLOH has been linked to more severe MPN phenotype. However, the contribution of 9pLOH in the pathogenesis of MPNs remains unclear. To investigate the role of wild-type JAK2 in MPNs induced by JAK2V617F, we have utilized conditional Jak2 knock-out and Jak2V617F knock-in alleles and generated heterozygous, hemizygous and homozygous Jak2V617F mice. Whereas heterozygous Jak2V617F expression results in a polycythemia vera-like disease in mice, loss of wild-type Jak2 allele in hemizygous or homozygous Jak2V617F mice results in a significantly greater increase in reticulocytes, white blood cells, neutrophils and platelets in the peripheral blood and larger spleen size. We also have found that hemizygous or homozygous Jak2V617F expression significantly increased megakaryocyte-erythroid progenitors in the bone marrow and spleens and marked infiltration of neutrophils in the liver compared with heterozygous Jak2V617F. More importantly, hemizygous or homozygous Jak2V617F mice show accelerated myelofibrosis compared with heterozygous Jak2V617F-expressing mice. Thus, loss of wild type Jak2 allele increases myeloid cell expansion and enhances the severity of the MPN. Together, these results suggest that wild-type Jak2 serves as a negative regulator of MPN induced by Jak2V617F. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 28 (2) ◽  
pp. 1544-1557
Author(s):  
Chris Varghese ◽  
Tracey Immanuel ◽  
Anna Ruskova ◽  
Edward Theakston ◽  
Maggie L. Kalev-Zylinska

Background: There is a paucity of data on ethnic disparities in patients with the classical Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs): polycythaemia vera (PV), essential thrombocythaemia (ET) and primary myelofibrosis (PMF). Methods: This study analysed the demographic data for PV, ET and PMF collected by the New Zealand Cancer Registry (NZCR) between 2010 and 2017. Results: We found that the NZCR capture rates were lower than average international incidence rates for PV and ET, but higher for PMF (0.76, 0.99 and 0.82 per 100,000, respectively). PV patients were older and had worse outcomes than expected, which suggests these patients were reported to the registry at an advanced stage of their disease. Polynesian patients with all MPN subtypes, PV, ET and PMF, were younger than their European counterparts both at the time of diagnosis and death (p < 0.001). Male gender was an independent risk factor for mortality from PV and PMF (hazard ratios (HR) of 1.43 and 1.81, respectively; p < 0.05), and Māori ethnicity was an independent risk factor for mortality from PMF (HR: 2.94; p = 0.006). Conclusions: New Zealand Polynesian patients may have increased genetic predisposition to MPN, thus we advocate for modern genetic testing in this ethnic group to identify the cause. Further work is also required to identify modifiable risk factors for mortality in MPN, in particular those associated with male gender and Māori ethnicity; the results may benefit all patients with MPN.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2962-2962
Author(s):  
Stephan Wilmes ◽  
Maximillian Hafer ◽  
Tess A Stanly ◽  
Ignacio Moraga ◽  
Joni Vuorio ◽  
...  

Janus kinase (JAK2)V617F is the most common mutation found in patients with Philadelphia chromosome negative myeloproliferative neoplasms (Ph- MPNs). The discovery of this mutation over 15 years ago revolutionised MPN diagnosis and inspired the development of JAK inhibitors as new therapeutic interventions. However, despite extensive structural and biophysical studies using JAK2 domains in isolation, the exact molecular mechanisms of JAK2V617F activation remains elusive. We have previously demonstrated that expression of the thrombopoietin (TPO) receptor, MPL, which interacts directly with JAK2, is essential for disease development in a mouse model of a JAK2V617F-positiveMPN (Blood 2014 124:3956-3963). Using total internal reflection fluorescence (TIRF) microscopy, we visualized MPL interaction dynamics in live cells on single molecule level. Effective cell surface MPL fluorescence labelling and dual-color imaging allowed us to determine the level of MPL dimerization under various experimental conditions. Using this assay, we clearly established that MPL is monomeric at physiologically relevant receptor densities. However, TPO stimulation results in significant dimerization of MPL (>50%) and an equilibrium between monomers and dimers. This counters the current dogma that MPL exists at the membrane as a pre-formed dimer. Strikingly, we found that JAK2V617F shifts this monomer-dimer equilibrium leading to significant TPO-independent MPL dimerization providing a novel mechanistic model of oncogenic JAK2 activation. To highlight the role of ligand-independent receptor dimerization in JAK2 activation, we compared three groups of autoactivating mutations in the PK domain covering the FERM-SH2 (FS2)-PK linker region (Group I), residues in the proximity of the αC helix (Group II) and at the autoinhibitory PK-TK interface (Group III). Consistent MPL dimerization was only observed for mutations in groups I and II. Mutations in these groups both localize to a potential homomeric PK/PK interface that has been implicated as a switch of JAK activation. Using MD simulations, we also found that the FERM domain of JAK2 strongly interacts with the inner leaflet of the lipid bilayer of the plasma membrane via a single hydrophobic residue (L224) surrounded by several positively charged residues that allows the region to act as a membrane anchor. This tight coupling to the membrane enforces an appropriate orientation between the JAKs within the receptor dimers required for optimal intermolecular PK/PK interaction that is critical for receptor dimerization. To interfere with membrane anchoring, we introduced a negative charge in this position (L224E). Strikingly, ligand-independent MPL dimerization and activation by JAK2V617F was dramatically reduced upon introducing L224E, supporting the vital importance of L224 for orienting JAK2 at the membrane to allow productive PK-PK interactions. Here, we demonstrate that JAK2V617F mutation acts by altering and strengthening the intermolecular interactions involving the PK/PK dimerization interface. In essence, these mutations drive cytoplasmic stabilization of receptor-JAK dimers, bypassing extracellular stabilization of dimers via cytokine binding. These results provide critical and entirely novel mechanistic insights into signal initiation in MPNs and readdress the roles of receptor-associated proteins. Disclosures Hubbard: Ajax Therapeutics, Inc.: Membership on an entity's Board of Directors or advisory committees, Other: Co-Founder.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4099-4099
Author(s):  
Elena Tenedini ◽  
Isabella Bernardis ◽  
Valentina Artusi ◽  
Lucia Artuso ◽  
Enrica Roncaglia ◽  
...  

Abstract The discovery of the JAK2V617F mutation in 2005 [Kralovics R, N Engl J Med 2005] represented a major breakthrough in the understanding of the molecular pathogenesis of Philadelphia chromosome negative chronic myeloproliferative neoplasms (MPN). Nevertheless several observations suggest that the JAK2V617F mutation may not be the disease funding mutation, at least in most instances. Therefore, a great deal of effort is ongoing with the aim to identifying novel genetic lesions contributing to the disease pathogenesis. The two major theoretical and technical drawbacks to the identification of new somatic mutations are represented, respectively, by the huge number of genes potentially involved in tumorigenesis of MPN and by the availability of a “pure” germline control DNA. Buccal swabs and saliva have been generally considered as readily available sources of DNA of non-hematopoietic origin, but detection of the JAK2V617F mutation in at least some of these samples indeed suggested the presence of myeloid cell contamination [Levine RL, Cancer Cell 2005]. So, in order to discover novel mutations in MPN using upfront technologies based on next-generation sequencing (NGS) we designed a “cancer exome” capture panel of 2000 unique genes and microRNAs. This panel was used to capture libraries generated from genomic DNA extracted from granulocytes and in vitro expanded CD3+ T-lymphocytes as germline control, in a cohort of 20 MPN patients. These captured libraries were than massively sequenced using the Roche 454 FLX platform. DNA samples had been collected at the diagnosis of PV in 9 subjects and PMF in 6 subjects, while the remaining 5 DNA samples were from 5 of the 9 PV patients at the time they evolved to post-PV myelofibrosis. After extensive bioinformatics analysis and multiple control adjustments, we finally produced a list of 171 novel “true” somatic mutations occurring in genes and microRNAs coding regions of those MPN samples; some of these mutations have been already described in MPN, whereas novel variants represent the vast majority. Despite patients harbored different numbers of somatic mutations, spanning from four to twenty-one variants, only 22 genes appear recurrently mutated. It is worth of notice the acquisition of additional mutations and/or the occurrence of loss of some mutations at the time of disease evolution from PV to a post-PV Myelofibrosis in the five patients for whom samples were available at both disease phases. Some of them, either acquired (NTRK1, PRDM2, BRCA2 and BARD1) or lost (APC, CARS, MLL3 and FAT2) had been found also in another PV or PMF sample. To test the recurrence of these mutations, we screened a different cohort of 189 patients composed by PMF (91 samples), PV (50 patients) and post-PV Myelofibrosis (48 samples) by Ion AmpliSeq technology on an Ion Torrent PGM platform. Deep amplicon sequencing of granulocytes DNA achieved a sample median of 1000-fold coverage. Excluding JAK2, MPL, IDH2, ASXL1 known variants, for 7 genes (SCRIB, MIR662, BARD1, TCF12, FAT4, DAP3, NRAS) we demonstrated in MPN a global mutation frequency greater than the 3%. Whereas some new variants need functional validation to prove causal mechanisms, some other mutations have a well-known pathogenic role in solid cancers but here are described for the first time in MPN. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2368-2368 ◽  
Author(s):  
Yue Han ◽  
Shixiang Zhao ◽  
Wenjuan Zhang ◽  
Jiannong Cen ◽  
Wei Zhang ◽  
...  

Abstract Background Microparticles (MPs) are small membrane vesicles that are classified as red blood cell MPs (RMPs), platelet-derived MPs (PMPs), tissue factor MPs (TF+MPs) and endothelial MPs (EMPs) based on their origins. Philadelphia chromosome-negative myeloproliferative neoplasms (Ph-MPN) are disorders characterized by abnormal hematopoiesis, thrombosis, JAK2V617F mutation. Although MPs are considered as biomarkers reflecting procoagulant state in cancer patients, their involvement in the patients with Ph-MPN remains unclear. Our objective in this study was to measure the alterations of the four MPs types in the patients with MPN and to evaluate their correlations with JAK2V617F mutation and some clinical complications, especially for thrombosis and splenomegaly. Methods Sixty-seven patients with MPN were enrolled in this study, including 12 polycythaemia vera (PV), 49 essential thrombocythemia (ET) and 6 primary myelofibrosis (PMF). 30 healthy donors were selected as normal controls. Venous blood was anticoagulated with sodium citrate (1:9). Using flow cytometry, plasma samples were measured for RMPs, PMPs, TF+MPs and EMPs with phycoerythrin (PE)-conjugated monoclonal antibodies CD235a, CD61, CD142, and CD62E, respectively. Forward scatter was set in scale using fluorescent microspheres of 0.8μm and standard fluorescent microbeads (0-0.8μm) in diameter were used to set the microparticle gate. Data were expressed as median (M) and interquartile range (IQR). Meanwhile, genomic DNA was extracted from mononuclear cells and amplified by allele specific polymerase chain reaction (PCR). Results (1) Patients with MPN showed significantly higher plasma levels for all four MPs compared with healthy donors (P<0.05), namely 49.0/μl (15.8-109.5/μl) vs 21.0/μl (13.8-32.6/μl) for RMPs, 181.2/μl(75.8-1111.6/μl) vs 74.9/μl (55.5-115.4/μl) for PMPs, 48.1/μl (13.1-72.4/μl) vs 31.0/μl (14.9-47.6/μl) for TF+MPs and 310.2/μl (128.6-1130.5/μl) vs 155.9/μl (100.3-227.6/μl) for EMPs. (2) Among different subtypes of MPN, PMPs were higher in patients with PMF than patients with PV and ET (P<0.05), but there was no significant difference between PV and ET group. No obvious difference was found in RMPs, TF+MPs and EMPs among different subtypes of MPN patients. (3) MPN patients with JAK2V617F mutation (n=34) were found to have higher plasma levels of TF+MPs and RMPs than those without mutation (P<0.05) and this difference was not found for PMPs and EMPs. (4) MPN patients with various thrombotic complications (n=10) showed higher levels of all four types of MPs than those without thrombotic complications (n=31) (P<0.05). Elevated MP levels were also found in patients with splenomegaly (n=19) compared to those without splenomegaly (n=14) (P<0.05). Conclusion Higher levels of MPs were observed in MPN patients compared with healthy controls, especially in patients complicated with thrombosis and splenomegaly, which reflects a prothrombotic state. Moreover, significantly increased TF+MPs and RMPs were found in MPN patients with JAK2V617F mutatioin. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3068-3068
Author(s):  
Hiraku Takei ◽  
Yoko Edahiro ◽  
Lihua Li ◽  
Yoshihisa Mizukami ◽  
Misa Imai ◽  
...  

Abstract Philadelphia-chromosome negative myeloproliferative neoplasms (MPNs) consists of different clinical entities that include polycythemia vera (PV) and essential thrombocythemia (ET). Despite of differential clinical features, JAK2V617F mutation is found in both PV and ET, leaving the cause of differential biological phenotypes by an oncogene obscure. Previously, studies have shown that higher allele frequencies or expression of JAK2V617F are associated with PV symptom/features in patients or model animals, respectively, suggesting that the copy number of JAK2V617F modulates hematopoietic cell differentiation and thus exhibits differential clinical features. However, this remained elusive in human hematopoiesis. To examine the impact of the zygosity of JAK2V617F allele on hematopoietic differentiation in human cells, we established induced pluripotent stem cells (iPSCs) harboring heterozygous- and homozygous-JAK2V617F mutation using genome-editing techniques from normal iPSCs. The introduction of JAK2V617F mutation with one or two copies did not alter the pluripotency of iPSCs and their capacity to differentiate into hematopoietic stem/progenitor cells (HSPCs) in vitro. When we induced hematopoietic cell differentiation from HSPCs, factor-independent erythropoiesis and megakaryopoiesis were induced in both heterozygous and homozygous JAK2V617F-HSPCs. Furthermore, homozygous JAK2V617F-HSPCs showed higher potential for erythropoiesis compared to the ones with heterozygous JAK2V617F, while the zygosity of JAK2V617F showed less effect on megakaryopoiesis. To further understand the molecular mechanism of hematopoietic cell differentiation modulated by differential copy number of JAK2V617F, we analyzed the activation of JAK-STAT signal by immunoblot analysis. The activation of JAK-STAT signals was more prominent in HSPCs harboring homozygous JAK2V617F, than those with heterozygous JAK2V617F. This suggested that the level of JAK2 phosphorylation was positively correlated with the copy number of JAK2V617F. These observations implied followings: 1) heterozygous JAK2V617F was sufficient to promote the development of MPN by inducing the factor-independent erythropoiesis and megakaryopoiesis; 2) the zygosity of JAK2V617F determined the disease phenotypes of MPNs by modulating erythropoiesis but not megakaryopoiesis; and 3) the homozygous JAK2V617F increased JAK-STAT signaling in HSPCs, promoting an increased erythropoiesis. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Shirong Wen ◽  
Wenxiao Zhang ◽  
Yiping Fei ◽  
Ke Guan ◽  
Hui Zhao ◽  
...  

Abstract Background: The ischemic cerebrovascular disease (ICVD) is major thrombotic complication of Philadelphia chromosome (Ph)-negative myeloproliferative neoplasms (Ph-negative MPNs) which included essential thrombocythemia (ET), polycythemia vera (PV) and primary myelofibrosis (PMF) leading to high disability and mortality rates. However, risk factors attributable to ICVD in Ph-negative MPNs patients are still not understood. This study aimed to identify risk factors for ICVD in Ph-negative MPNs. Methods: Patients with Ph-negative MPNs were divided into ICVD and non-ICVD groups. The demographic, biochemical, genetic parameters (JAK2V617F and CALR mutations) were assessed. The association between these factors and ICVD was assessed using logistic regression analysis.Results: One hundred eighty five Ph-negative MPNs patients (82 ET, 78 PV, and 25 PMF) were recruited,and 57 (30.8%) had ICVD which was higher than recently published data. The higher prevalence of hypertension(59.6% vs 32.0%), smoking(22.8% vs 6.3%), drinking(22.8% vs 6.3%), JAK2V617F mutation(78.9% vs 63.3%), the percentage of neutrophils( 78.76% vs 71.73%), the lower prevalence of CALR mutation(3.2% vs 22.2%) and the percentage of basophils(0.82% vs 1.40%) were found in the ICVD group comparing with the non-ICVD group. The frequency of ICVD events was significantly higher in patients with JAK2V617F mutation than in those without (35.7% and 20.3%, P=0.034). Multivariate logistic regression analysis showed that hypertension (OR=2.464, 95%CI 1.218-4.983, p=0.012) and smoking (OR=5.426, 95%CI 1.919-15.340, p=0.001) were significantly positively associated with ICVD events. For ET patients, both smoking (OR=4.414, 95%CI 1.079-15.685, p=0.038) and increased percentage of neutrophils (OR=1.080, 95%CI 1.019-1.144, p=0.009) were independently associated with ICVD incidence. Hypertension (OR=4.647, 95%CI 1.215-17.781, p=0.025), smoking (OR=6.065, 95%CI 1.083-33.951, p=0.040), and increased percentage of lymphocytes (OR=1.039, 95%CI 1.002-1.078, p=0.039) were all positively correlated with ICVD risk in PV patients. Conclusions: Our data suggest that hypertension, smoking, higher percentage of neutrophils and lymphocytes rather than JAK2V617F and CALR mutations may be associated with an elevated risk of ICVD in Ph-negative MPNs patients, although the relative role of each factor may vary in the individual subgroup. Additional studies of large patient cohorts will be essential to validate these findings.


Leukemia ◽  
2009 ◽  
Vol 23 (9) ◽  
pp. 1577-1586 ◽  
Author(s):  
J Wang ◽  
T Ishii ◽  
W Zhang ◽  
S Sozer ◽  
Y Dai ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document