Polycythemia Vera: Redefinition in the Genomic Era

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1754-1754
Author(s):  
Alison R. Moliterno ◽  
Donna M. Williams ◽  
Michael A McDevitt ◽  
Brady L Stein ◽  
Jonathan M. Gerber ◽  
...  

Abstract Abstract 1754 For more than 100 years since the initial descriptions, polycythemia vera (PV) was defined by an aggregation of clinical and laboratory features, reported to be more common in males than females, and diagnosed on average at age 60 (Modan Blood 1965, Berlin Sem Hematol 1975). The 2005 discovery of somatic mutations in the JAK2 gene introduced the molecular era of PV and required redefinition of this disease entity. We established a prospective, observational cohort of 556 patients evaluated in our center between 2005 and 2011, of which 273 had a PV phase at some point during their myeloproliferative neoplasm (MPN). Serial samples were obtained from each patient for genomic analyses, including neutrophil JAK2V617F allele burdens, clinical karyotypes, SNP-array karyotypes, JAK2 and ASXL1 sequencing and copy number variation, allele burden analysis in sorted hematopoietic stem cell (HSC) fractions, and whole exome sequencing. These data were used to define the relationship of genotype to clinical phenotype with regard to PV epidemiology, natural history and disease transformation. Thirty three percent of the cohort was evaluated within 1 year from PV diagnosis and the median MPN disease duration at the last update of the cohort was 9 years (range 1–52 years). As of 7/2012, of the 273 PV patient cohort, 47 had antecedent essential thrombocytosis (ET/PV), 176 had PV, 43 had developed post-PV myelofibrosis (PPVMF) and 7 had developed acute leukemia (AML) (PPVAML). 270 of the 273 PV patients had JAK2 mutations, either V617F (264, 97%) or exon 12 (6, 2%); the remaining 3 (1%) are molecularly undefined. Women outnumbered men (169/104; ratio 1.6), even when stratified by ET/PV (2.1), PV (1.6), PPVMF (1.4) and PPVAML (1.3). Age at PV diagnosis was significantly younger in women, 54 (range 8–88), compared to men, 56.5 (range 15–77) (p=0.022), and the proportion diagnosed before age 40 was 26% in women compared to 10.5% in men. PPVMF occurred on average after 9 years (range 2–53 years) of PV at a median age of 62.5 years. PPVAML occurred on average after 10 years (range 3–28 years) of PV, at a median age of 71 years, significantly higher than the age at PPVMF (p=0.038). Aside from JAK2V617F, acquired 9pUPD was the most common genomic lesion in PV, occurring in 57% within the first year after PV diagnosis, in 84% of PPVMF and 100% of PPVAML patients. Studied prospectively, the prevalence of 9pUPD increased from 0 to 40% in 11 patients transitioning from ET to PV, and increased from 59% to 75% in 30 PV patients from year 1 to year 6 after diagnosis, but stayed at 90% in 11 patients pre and post transformation to PPVMF. Chromosomal loss/gain was not highly prevalent during PV (2%) in contrast to PPVMF (64%) and PPVAML (100%). The most frequent chromosomal abnormalities in PPVMF were trisomy 9 (27%), 13q deletion (12%), 1q gain(12%), 20qdeletion (8%) and 11qdel (8%), whereas the most common chromosomal abnormalities in PPVAML were 5qdel or −5 (75%), and 7qdeletion (50%), both of which were often found in the setting of complex changes (75%). Genomic lesions identified in PV and PPVMF, including JAK2V617F, 9pUPD, 11qdel, and ASXL1 mutations, were detected at high allele burdens by quantitative allele assays in flow-sorted, pluripotent HSCs. We conclude that acquisition of a JAK2 mutation is implicated in the vast majority (99%) of PV patients, that PV occurs more often in women, and that younger women (<40) particularly are at higher risk than younger men. Genomic lesions in PV and PPVMF arise and accumulate in a primitive HSC population. 9pUPD is a common occurrence during transition from JAK2V617F+ ET to PV, and while highly prevalent, age and time dependent in PV, 9pUPD is not sufficient to generate PPVMF or PPVAML. In PPVMF, JAK2 mutations associate with specific recurrent chromosomal changes that are also found in normal individuals with advancing age (9pUPD, 13qdel, 20qdel, 11qdel; Nature Genetics 44, 2012). JAK2 mutations with 9pUPD enhance the acquisition of age-associated and therapy- associated genomic instability lesions, promoting the development of PPVMF and PPVAML. Given the molecular epidemiology of PV, it will be crucial recognize and reduce the risk factors that lead to the excess acquisition of PV in young women, to identify the risk factors that lead to 9pUPD, to study whether targeted therapy can prevent the development of 9pUPD, and to avoid genotoxic therapy that accelerates genomic instability in PV. Disclosures: Streiff: sanofi-aventis: Consultancy, Honoraria; BristolMyersSquibb: Research Funding; Eisai: Consultancy; Janssen Healthcare: Consultancy; Daiichi-Sankyo: Consultancy.

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3205-3205
Author(s):  
Donna M. Williams ◽  
Michael A McDevitt ◽  
Brady L Stein ◽  
Ophelia Rogers ◽  
Michael B. Streiff ◽  
...  

Abstract Introduction: Essential thrombocytosis (ET) is a hematopoietic stem cell disease defined by acquired mutations in JAK2, CALR or MPL. Variability in natural history, phenotypic transformation to PV or MF, and thrombosis risk exist within ET with age, gender, disease duration, mutant allele burden and other factors operating as modifiers. Methods: We established a prospective, observational cohort of 605 ET, polycythemia vera (PV) and myelofibrosis (MF) patients enrolled between 2005 and 2013 with which to study the genomic modifiers of the MPN. We obtained neutrophil JAK2V617F allele burdens for all patients, and MPL and CALR mutation status for the JAK2V617F-negative subset. SNP-array karyotypes and ASXL1mutation analyses were obtained on a subset of the cohort. Results: Twenty-five percent of the cohort was newly diagnosed and the median MPN disease duration of the cohort was 8 years (range 1-52 years). Females comprised the majority of the entire MPN cohort, whether considering newly diagnosed (80/155;51%) or prevalent cases (370/605;62%). Females comprised the majority of ET patients, whether considering newly diagnosed (34/55;61%), or prevalent cases (71/101;70%). JAK2V617F was present in 60% of ET. While CALR mutations were present in 67% of JAK2V617F-negative ET, they comprised a smaller relative proportion in females (55%) compared to males (88%; p=0.0047), due to an excess of JAK2-CALR-MPL mutation-negative (triple negative (TN)) ET in females (34%) compared to males (8%; p=0.0142). At last follow up, females comprised 70% of JAK2V617F-positive ET, 70% of JAK2V617F-negative ET, 57% of CALR mutation-positive ET, 86% of MPL-mutation positive ET and 90% of TN ET. Similar to reported findings, JAK2V617F-positive ET patients were older at diagnosis (48.7 vs 42.5 yrs,p=0.005), had lower platelet counts and a higher prevalence of chemotherapy use compared to JAK2V617F-negative ET. Strikingly, abdominal venous thrombosis events were restricted to the JAK2V617F-positive ET cohort, occurring in 4%, and predominantly in females. A family history of an MPN was similar between groups, described in 13% of CALR and TN mutation ET compared to 11% of JAK2V617F-positive ET. ET patients had a low prevalence of chromosomal loss, gain or acquired uniparental disomy (UPD) compared to PV or MF patients. UPD occurred in 14% of JAK2V617F-negative ET, and encompassed the CALR gene in one patient with a CALR mutation, the MPL gene in a patient with an MPL mutation, and the TET2 gene in two others. UPD was present in 23% of JAK2V617F-positive ET, and did not encompass regions present in the JAK2V617F-negative ET cases. ASXL1 mutations were absent in all ET cases. While JAK2V617F-positive ET patients may transform to PV, transformation to PV in CALR, MPL or TN mutation patients was not observed. Transformation to post-ET MF occurred in all ET genotypes, and despite differences in time to transformation (mean 10.2 years in JAK2V617F-positive ET compared to 18.3 years in CALR, MPL and TN mutation ET (p=0.005)), average age at MF transformation was similar (62 years). Both UPD and chromosomal loss/gain were prevalent in ET patients who transformed to MF, regardless of genotype. However, 9pUPD was restricted to JAK2V617F-positive patients. Chromosomal loss regions in 20q and 17q were present in both JAK2V617F and CALR positive post-ETMF, as were ASXL1 mutations (40% in JAK2V617F -positive post-ETMF and 75% in JAK2V617F-negative post-ETMF). Conclusions: The genetic drivers of phenotype within ET may differ (JAK2, CALR, MPL, TN), yet all ET subtypes share female predominance, similar rates of familial clustering, and similar age at transformation and chromosomal instability lesions that accompany post-ET MF. TN ET is not yet defined by a genetic lesion, yet shares similar epidemiologic, natural history and genetic features as mutation defined ET. We conclude that TN ET is likely a clonal myeloid disease, and that either there are other ET driver lesions, or that allele burdens of JAK2, CALR or MPL lesions are below the limits of detection of standard screening assays in TN ET. Given the molecular epidemiology of MPN, it will be crucial recognize and reduce the risk factors that lead to the excess acquisition of ET in women, to identify the risk factors that lead to disease transformation, and to develop targeted therapy that can extinguish ET clones while avoiding therapy that accelerates genomic instability in ET. Disclosures Moliterno: Incyte: Consultancy.


Hematology ◽  
2005 ◽  
Vol 2005 (1) ◽  
pp. 195-200 ◽  
Author(s):  
William Vainchenker ◽  
Stefan N. Constantinescu

Abstract Myeloproliferative disorders (MPDs) are heterogeneous diseases that occur at the level of a multipotent hematopoietic stem cell. They are characterized by increased blood cell production related to cytokine hypersensitivity and virtually normal cell maturation. The molecular pathogenesis of the MPDs has been poorly understood, except for chronic myeloid leukemia (CML), where the Bcr-Abl fusion protein exhibits constitutive kinase activity. Since some rare MPDs are also related to a dysregulated kinase activity, a similar mechanism was thought to be likely responsible for the more frequent MPDs. We investigated the mechanisms of endogenous erythroid colony formation (EEC) by polycythemia vera (PV) erythroid progenitor cells and found that EEC formation was abolished by a pharmacological inhibitor of JAK2 as well as an siRNA against JAK2. JAK2 sequencing revealed a unique mutation in the JH2 domain leading to a V617F substitution in more than 80% of the PV samples. This mutation in the pseudokinase autoinhibitory domain results in constitutive kinase activity and induces cytokine hypersensitivity or independence of factor-dependent cell lines. Retroviral transduction of the mutant JAK2 into murine HSC leads to the development of an MPD with polycythemia. The same mutation was found in about 50% of patients with idiopathic myelofibrosis (IMF) and 30% of patients with essential thrombocythemia (ET). Using different approaches, four other teams have obtained similar results. The identification of the JAK2 mutation represents a major advance in our understanding of the molecular pathogenesis of MPDs that will likely permit a new classification of these diseases and the development of novel therapeutic approaches.


2021 ◽  
Vol 28 ◽  
pp. 107327482110468
Author(s):  
Niloofar Allahverdi ◽  
Mohamed Yassin ◽  
Mohamed Ibrahim

Myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell disorders characterized by the overproduction of mature myeloid cells and are often associated with an acquired genetic mutation of Janus Kinase2V617F. Various epidemiological studies have indicated associations between environmental factors, lifestyle factors, and host characteristics with developing MPNs. This review aims to collect and summarize the existing information on these risk factors and establish their association with pathogenesis MPNs. Medline, Embase, PubMed, and grey literature were systematically searched using key terms for MPNs, and epidemiological study designs, that is, cross-sectional studies, case-control, and cohort, that investigated the risk factors for MPNs published were identified. Out of the 4621 articles identified, 20 met the selection criteria and were included in this review. Heterogeneity, study reliability, and bias were assessed. A significant association was found between smoking and the development of MPNs. This relationship has been explained by the substantial increase in several proinflammatory mediators and systematic oxidative stress causing hyperstimulation of myeloid compartments leading to the development of MPNs. Obesity was modestly linked with an increased risk of MPNs. The underlying mechanisms have been linked to changes in endocrine, metabolic, and inflammatory systems. No strong association was found between exposure to hazardous substances, that is, benzene and MPNs, but further investigation on the effects of increased levels and duration of exposure on hematopoietic stem cells will be beneficial. Unique individual and host variations have been determined as a modifier of disease pathogenesis and phenotype variations. There is a higher incidence rate of females developing MPNs, specifically ET, than males with higher PV incidence. Therefore, gender contributes to the heterogeneity in myeloproliferative neoplasm. Studies identified as part of this review are very diverse. Thus, further in-depth assessment to explore the role of these etiological factors associated with MPNs is warranted.


Cancers ◽  
2021 ◽  
Vol 13 (19) ◽  
pp. 4968
Author(s):  
Martina Barone ◽  
Monica Barone ◽  
Francesca Ricci ◽  
Giuseppe Auteri ◽  
Francesco Fabbri ◽  
...  

Polycythemia vera is a myeloproliferative neoplasm with increased risk of thrombosis and progression to myelofibrosis. However, no disease-specific risk factors have been identified so far. Circulating extracellular vesicles (EVs) are mostly of megakaryocyte (MK-EVs) and platelet (PLT-EVs) origin and, along with phosphatidylethanolamine (PE)-EVs, play a role in cancer and thrombosis. Interestingly, circulating microbial components/microbes have been recently indicated as potential modifiers of inflammation and coagulation. Here, we investigated phenotype and microbial DNA cargo of EVs after isolation from the plasma of 38 patients with polycythemia vera. Increased proportion of MK-EVs and reduced proportion of PLT-EVs identify patients with thrombosis history. Interestingly, EVs from patients with thrombosis history were depleted in Staphylococcus DNA but enriched in DNA from Actinobacteria members as well as Anaerococcus. In addition, patients with thrombosis history had also lower levels of lipopolysaccharide-associated EVs. In regard to fibrosis, along with increased proportion of PE-EVs, the EVs of patients with marrow fibrosis were enriched in DNA from Collinsella and Flavobacterium. Here, we identified a polycythemia-vera-specific host/microbial EV-based signature associated to thrombosis history and marrow fibrosis. These data may contribute to refining PV prognosis and to identifying novel druggable targets.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2805-2805
Author(s):  
Mathias Vilaine ◽  
Damla Olcaydu ◽  
Ashot Harutyunyan ◽  
Jonathan Bergeman ◽  
Tiab Mourad ◽  
...  

Abstract Abstract 2805 Background: Adequate expression and function of Jak2 in hematopoietic progenitors is critical for normal myelopoiesis. The JAK2 46/1 (GGCC) haplotype, a congenital particularity, predisposes to myeloproliferative neoplasm (MPN) both independently and through mutation of the JAK2 gene. The JAK2 V617F mutation and acquired homozygous status for JAK2 V617F are frequent in MPN. JAK2 V617F homozygosity is currently explained acquisition of the JAK2 V617F mutation followed by mitotic homologous recombination (HR) of JAK2 occurred between wild-type and mutant alleles, leading to uniparental disomy (UPD) of chromosome 9p (9pUPD). Here we report the cases of 2 PV patients (Na1061 and Na1253) with acquired homozygous status for the JAK2 46/1 haplotype yet their granulocytes carried <20% JAK2 V617F. Aim: To determine whether HR of JAK2 can precede the V617F mutation in MPN. Methods: Granulocyte DNA and CD3+ lymphocyte DNA were examined in parallel with qPCR assays specific for the wild type and 46/1 haplotypes using rs12343867, a JAK2 intron 14 marker, as well as 4 other single nucleotide polymorphisms (SNP) on chromosome 9p. 9pUPD clonality and length were determined using SNP array studies. Results: For both patients, lymphocytes were heterozygous for the 46/1 haplotype, confirming that granulocyte 46/1 homozygosity was acquired. Direct sequencing of the JAK2 and GNE genes and SNP array analyses revealed homologous recombination of part of the JAK2 gene (exons 6–19, patient Na1061) and of the complete 46/1 JAK2 haplotype (patient Na1253). Furthermore, for both patients, full length sequencing of JAK2 cDNA revealed no additional mutation. In both cases, HR of wild-type JAK2 was associated with growth advantage and high expression of recombined JAK2. For both patients, further SNP array analyses revealed partial 9pUPD concerning <30% cells, which correlated with %JAK2 V617F and was consistent with 9pUPD having occurred after JAK2 V617F (Figure 1). The distortion of SNP allelic differences was higher at the telomeric end than in the centromeric region of chr. 9p. This indicated 2 distinct partial 9pUPDs for Na1061 and 1 partial 9pUPD for Na1253. Conclusion: Homologous recombination involving wild type JAK2 can precede JAK2 mutation and 9pUPD in MPN. Thus multiple paths and diverse alterations of the JAK2 gene can lead to MPN in individuals carrying the JAK2 GGCC haplotype. We propose a new model with JAK2 HR as early event, followed or not by JAK2 mutation, or/and JAK2 mutation(s) facilitating subsequent recombination resulting in 9pUPD and JAK2 V617F homozygosity. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4961-4961
Author(s):  
Charikleia Kelaidi ◽  
Dimitrios Kokkinidis ◽  
Maria Protopappa ◽  
Georgios Papaioannou ◽  
Ioannis Batsis ◽  
...  

Abstract Abstract 4961 Background: Platelet increase under azacitidine in patients with myelodysplastic syndrome (MDS) has been acknowledged as an early predictive factor of response to treatment. However, extreme thrombocytosis under azacitidine has not been reported. Methods: We studied consecutive patients with MDS or MDS/myeloproliferative neoplasm (MDS/MPN) who had platelet counts near or over 1, 000 G/L under azacitidine. Results: Four patients, sex ratio 1:1, with median age of 65 years, had extreme thrombocytosis under azacitidine. Baseline characteristics were: WHO classification RAEB-2/CMML-1/CMML-2 in 2/1/1 patients, median platelet count 248 G/L (<400 G/L in all), normal karyotype/+8, −9/−7 in 2/1/1 patients, IPSS low/int-2/high in 1/2/1 patients. None had reticulinic fibrosis or ring sideroblasts>15% at baseline. A median number of 8 cycles of azacitidine was administered. Individual platelet counts reached 2, 960 G/L, 800 G/L, 1, 188 G/L and 2, 740 G/L. Thrombocytosis occured early after treatment onset or resumption (Figure 1). Histologic findings under treatment were: Increased cellularity (N=4), micromegakaryocytes and other signs of megakaryocytic dysplasia (N=4), reticulinic fibrosis grade I and II in 1 and 2 patients, respectively. JAK2 V617F mutation was detected in 1 patient (with maximum platelet count of 2, 900 G/L) and was undetectable in the remaining patients. None had a thrombotic or hemorrhagic event. Two patients had a concomitant increase of WBC count. Response to azacitidine was CR, PR and stable disease in 1/1/2 patients. Three patients received hydroxyurea (HU) in addition to azacitidine and one patient underwent hematopoietic stem cell transplantation (HCT). AML transformation occurred in 1 patient 25 months after azacitidine onset. Median overall survival after azacitidine onset was 25 months. Conclusion: Extreme thrombocytosis of the range of essential thrombocytosis, with megakaryocytic dysplasia and hyperplasia, was noted under azacitidine in 4 patients with MDS-MDS/MPN and normal baseline platelet count. Hypothetically, azacitidine may induce the expression of critical genes of megakaryopoiesis or platelet release in patients with rare mutations. Notably, JAK2 mutation was detected in only one patient. Alternatively, demethylation could unmask an underlying unclassified MDS/MPN similar to RARS-T. Disclosures: No relevant conflicts of interest to declare.


Antioxidants ◽  
2022 ◽  
Vol 11 (1) ◽  
pp. 113
Author(s):  
Elena Genovese ◽  
Margherita Mirabile ◽  
Sebastiano Rontauroli ◽  
Stefano Sartini ◽  
Sebastian Fantini ◽  
...  

Myelofibrosis (MF) is the Philadelphia-negative myeloproliferative neoplasm characterized by the worst prognosis and no response to conventional therapy. Driver mutations in JAK2 and CALR impact on JAK-STAT pathway activation but also on the production of reactive oxygen species (ROS). ROS play a pivotal role in inflammation-induced oxidative damage to cellular components including DNA, therefore leading to greater genomic instability and promoting cell transformation. In order to unveil the role of driver mutations in oxidative stress, we assessed ROS levels in CD34+ hematopoietic stem/progenitor cells of MF patients. Our results demonstrated that ROS production in CD34+ cells from CALR-mutated MF patients is far greater compared with patients harboring JAK2 mutation, and this leads to increased oxidative DNA damage. Moreover, CALR-mutant cells show less superoxide dismutase (SOD) antioxidant activity than JAK2-mutated ones. Here, we show that high plasma levels of total antioxidant capacity (TAC) correlate with detrimental clinical features, such as high levels of lactate dehydrogenase (LDH) and circulating CD34+ cells. Moreover, in JAK2-mutated patients, high plasma level of TAC is also associated with a poor overall survival (OS), and multivariate analysis demonstrated that high TAC classification is an independent prognostic factor allowing the identification of patients with inferior OS in both DIPSS lowest and highest categories. Altogether, our data suggest that a different capability to respond to oxidative stress can be one of the mechanisms underlying disease progression of myelofibrosis.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 101-101
Author(s):  
Xiaoli Wang ◽  
Cing Siang Hu ◽  
Joseph Tripodi ◽  
Vesna Najfeld ◽  
Bruce Petersen ◽  
...  

Abstract Myeloproliferative neoplasm-blast phase (MPN-BP) and de novo acute myeloid leukemia (AML) each have distinct mutational patterns and clinical courses. MPN-BP patients have a particularly dismal prognosis with a median survival of less than 6 months with currently available therapies. So far, the cellular hierarchy that characterizes MPN-BP and the evolution of various leukemia-initiating clones (LIC) in MPN-BP have not been well delineated. We therefore established an in vivo MPN-BP xenograft model to address these questions. Among the 22 patients with MPN-BP studied 11 were cytogenetically normal while the remainder had multiple chromosomal abnormalities including del(5), del(20q), del(14), +1q, del(17p). 86% of the patients had at least 2 myeloid malignancy gene mutations including JAK2, ASXL1,TET2, MPL, SF3B1, RUNX1, U2AF1, PTPN11, IDH1/2, SRSF2 and TP53. These findings indicate that MPN-BP is characterized by multiple mutational events and cytogenetic abnormalities. T cell-depleted mononuclear cells from 8 of 14 patients engrafted in NSG mice {>0.5% hCD45+ cells in bone marrow (BM)}. Among them, samples from 6 patients resulted in a high degree of hCD45+ cell chimerism (34.6±6.4% in BM) and recapitulated numerous aspects of MPN-BP within 4 months, including the presence of at least 20% hCD45dimCD33+ cells or hCD34+ cells, or at least 20% blasts as detected by morphological examination of the marrow and leukemia cell dissemination to the spleen and PB. These mice had a 2.8±0.6- fold increase in splenic weight as compared to mice receiving PBS alone. The leukemic mice were characterized by reduced blood counts, suggesting that MPN-BP cells suppressed normal murine hematopoiesis, or led to cytopenias due to hyper-splenism. Moreover, the greater degrees of blast cell chimerism and the higher frequency of leukemia initiating cells as determined by limiting dilution analyses correlated with a shorter time to leukemia initiation and an inferior clinical outcome of the transplanted NSG mice. Grafts from each of these 6 MPN-BP patients produced a large number of donor-derived myeloid cells and a smaller number of lymphoid cells (mostly CD3+ and few CD19+). Cells belonging to each of these lineages and leukemic cells in primary recipients produced from Pts 4, 5, 6 and 11 had an identical proportion of chromosomally abnormal and mutated cells as primary cells [Pt 4: JAK2V617F, TET2 and PHF6; Pt 5 and 11: Del (20q), +8; Pt 6: +1q, del(17p)], except that a small proportion of T cells from Pts 5 and 11 lacked chromosomal abnormalities. Furthermore, the degree of MPN-BP engraftment and leukemic cell burden increased with the subsequent 3 serial transplantations even when the recipients received progressively smaller numbers of MPN-BP cells from the prior recipient. Primary Pt 6 originally had a JAK2V617F+ PV but lost JAK2V617F at the time the MPN-BP occurred at which time there were two clonal cell populations, one with +1q (12%) and the other del(17p) (80%), the site of the TP53 gene, as well as normal cells (8%). In the primary recipient NSG the donor derived cells were JAK2V617F- but contained +1q (1%) and del(17p) (98.5%) and cytogenetically normal (0.5%). +1q and JAK2V617F were not observed, while cells containing the TP53 deletion alone were detected in donor derived leukemic cells, mature myeloid and T cells in the secondary and subsequent serial recipients. Furthermore, del(17p) was found in phenotypically isolated HSCs, MPPs, MLPs, CMPs, GMPs, MEPs, and mature T cells within the CD33- cell fraction as well as CD45dimCD33+ AML blasts selected from primary MPN-BP cells from Pt6. However, +1q was found exclusively in purified MLPs and MEP. These observations establish that cytogenetic and mutational events that lead to MPN-BP occur at different stages along the developmental HSC hierarchy and that a small population of normal HSCs persist. Furthermore, in JAK2V617F+ MPNs that develop MPN-BP and lose JAK2V617F, additional cytogenetic events occur at different stages along the JAK2V617F- MPN-BP-stem cell hierarchy. Our ability to serially transplant the LIC from these patients has allowed us to create the first MPN-BP PDX model that will not only extend our understanding of MPN-BP stem cell biology but might also prove useful for screening drugs to treat MPN-BP. Disclosures Rampal: Jazz: Consultancy, Honoraria; Incyte: Honoraria, Research Funding; Stemline: Research Funding; Constellation: Research Funding; Celgene: Honoraria. Mascarenhas:Incyte: Membership on an entity's Board of Directors or advisory committees, Research Funding; CTI Biopharma: Membership on an entity's Board of Directors or advisory committees, Research Funding; Merck: Research Funding; Roche: Research Funding; Novartis: Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees; Promedior: Research Funding; Janssen: Research Funding. Hoffman:Formation Biologics: Research Funding; Summer Road: Research Funding; Merus: Research Funding; Incyte: Research Funding; Janssen: Research Funding.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 2608-2608
Author(s):  
Frank Stegelmann ◽  
Martin Griesshammer ◽  
Karlheinz Holzmann ◽  
Frank G Rücker ◽  
Carmen Blersch ◽  
...  

Abstract Abstract 2608 Poster Board II-584 Background: Myeloproliferative neoplasms (MPN) represent a heterogeneous group of acquired hematopoietic stem cell disorders. Clonality leads to exceeding production of myeloid cells resulting in an inherent tendency for thrombotic and hemorrhagic complications as well as transformation into acute myeloid leukemia (sAML). While vascular complications predominantly account for the morbidity in essential thrombocythemia (ET) and polycythemia vera (PV), the mortality of MPN is significantly related to leukemic transformation. Secondary AML occurs more frequently in primary and secondary myelofibrosis (PMF and SMF) than in ET and PV, and the risk for leukemic transformation increases with the duration of the disease. The molecular basis underlying the progression of MPN is poorly understood. Clonal evolution due to genomic instability is considered to play an important role. Aim: To identify genomic lesions associated with leukemic transformation, we applied 250K single-nucleotide polymorphism (SNP) arrays that allow for genome-wide screening of both copy-number alterations (CNAs) and copy-neutral runs of homozygosity (ROH) at high resolution. Method: An unpaired SNP-array analysis of 23 sAML samples was performed [former diagnosis: ET, n=5; PV, n=7; PMF, n=9; SMF, n=2;]. An own set of 30 reference samples was used for normalization. CNAs and ROH were analyzed by CNAG 3.0 software. Aberrations were compared with the 250K SNP-array dataset of 151 MPN patients [ET, n=45; PV, n=45; PMF, n=47, SMF, n=14]. In one sAML patient corresponding SNP-array data from the time of ET diagnosis were available. Results: CNAs were present in 15 of 23 (65%) sAML patients. Thirty-five percent of cases (n=8) exhibited complex genomic aberrations with up to 20 CNAs in one patient (range 5–20). The most frequent larger (>5 Mb) CNAs were trisomy 8 (n=7), gain of 1q, loss of 5q, and deletion in 6p25-pter (16.2–26.7 Mb) and 20q11-q13 (13.6–16.9 Mb) in three cases each, followed by gain in 3q24-qter (51.7 and 54.1 Mb) in two patients. Of note, one case with deletion in 17p12-pter (64.4 Mb) encompassing TP53 and a second with gain in 21q22.12-qter (11.8 Mb) were identified; in the latter one the proximal breakpoint of the gain was located at RUNX1. Smaller CNAs (<5 Mb) were restricted to single cases with four cases exhibiting micro-deletions ranging from 0.7 to 2.7 Mb in size. Interestingly, three chromosomal regions harbour single genes: 11p11.2 (FOLH1), 18q21.2 (TCF4), and 21q22.12 (RUNX1). ROH comprising the terminal end of the chromosome were detectable in 13 of 23 (57%) sAML cases. The most frequent ROH included the JAK2 locus in 9p24 (n=6; 15.6–38.7 Mb), followed by ROH in 17p13-pter (16.3 and 17.7 Mb) covering TP53 and an overlapping segment in 1p32-pter (53 Mb) affecting MPL in two cases each. All cases with 9p ROH were JAK2 V617F mutated, whereas the MPL W515L mutation was found in one of the two 1p ROH cases. Moreover, sequencing analyses in both patients with ROH in 17p revealed TP53 missense mutations in exon 7 and exon 8, respectively. In addition, non-recurrent ROH covering the long arm of chromosome 7, 11, and 21 as well as ROH in the chromosomal segments 14q32-qter (12.6 Mb) and 17q31-qter (31 Mb) were identified. In one sAML patient SNP-array data performed at the time of ET were available for comparative analysis. While 20q deletion was present as sole aberration in ET, complex genomic aberrations (7 CNAs) were identified after development of sAML. Merging the results from our recent 250K SNP-array analysis (Stegelmann et al., Blood 2008; 112: Abstract #2794) on 61 PMF and SMF cases with data from this study, we were able to identify a second case with micro-deletion in 12q24. The commonly deleted region of both cases is 1.3 Mb in size and encompasses TCF1 as a novel recurrent aberration in MPN. Conclusion: In summary, our data on a large series of well-defined sAML cases that evolved from MPN demonstrate that 250K SNP-array profiling is an excellent tool to identify genomic aberrations. In contrast to MPN, genomic alterations in sAML are characterized by a marked complexity reflecting both genomic instability and genetic heterogeneity of sAML. However, in our study several regions of interest including recurrently affected candidate genes such as TCF1, RUNX1, and TP53 were identified that need to be further investigated on a single gene level. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (1) ◽  
pp. 177-181 ◽  
Author(s):  
Shubha Anand ◽  
Frances Stedham ◽  
Philip Beer ◽  
Emma Gudgin ◽  
Christina A. Ortmann ◽  
...  

Abstract The JAK2 V617F mutation is present in the majority of patients with a myeloproliferative neoplasm (MPN) and is sufficient to recapitulate an MPN in murine models. However, the consequences of JAK2 mutations for myeloid differentiation are poorly understood. After systematic analyses of a large cohort of JAK2-mutated MPN patients, we demonstrate in vivo that JAK2 mutations do not alter hematopoietic stem and progenitor cell com-partment size or in vitro behavior but generate expansion of later myeloid differentiation compartments, where homozygous expression of the mutation confers an added proliferative advantage at the single-cell level. In addition, we demonstrate that these findings may be partially explained by the expression pattern of JAK2, which markedly increases on myeloid differentiation. Our findings have potential clinical relevance, as they predict that JAK2 inhibitors may control myeloproliferation, but may have limited efficacy in eradicating the leukemic stem cells that sustain the human MPN.


Sign in / Sign up

Export Citation Format

Share Document