Targeted Cancer Exome Sequencing Discovers Novel Recurrent Mutations In MPN

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 ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3913-3913
Author(s):  
Sabina Swierczek ◽  
Christine Bellanne-Chantelot ◽  
Donghoon Yoon ◽  
Cecile Saint-Martin ◽  
Soo Jin Kim ◽  
...  

Abstract Abstract 3913 Poster Board III-849 The Ten-Eleven Translocation (TET) 2 gene is a tumor suppressor gene. Its mutations of which are frequently found in PV and other myeloid malignancies. The published evidence suggests that a TET2 mutational burden is present in a higher proportion than JAK2V617F in PV stem cells; its mutations typically precede the JAK2V617F mutation, and are preferentially expressed in myeloid cells. We studied 40 PV patients. Two had known TET2 mutation, one with an intronic mutation (3954+2T>A) predicted to cause aberrant splicing, and the other with a deletion of a single nucleotide in exon 3 (3138delT) leading to the predicted truncation of the TET2 peptide. We quantitated the mutational burden of JAK2V617F and TET2 and the clonality of blood lineages using X-chromosome allelic usage ratios in the blood cells and BFU-E colonies. We also followed the mutational burden of the JAK2V617F and TET2 somatic mutations and expression of TET2 mRNA and monitored the proportion of polyclonal cells, in in vitro expanded erythroid progenitors. These data were compared to the PV patients without known TET2 mutations. Using an X-chromosome-based transcriptional clonality assay, the PV patients had predominantly clonal reticulocytes, granulocytes, platelets and, in those available, CD34-positive cells. Studies of individual BFU-E found that in two PV patients, the TET2 mutations followed rather than preceded the JAK2V617F mutation. We report that only a fraction of clonal CD34+ cells carry the TET2 mutation, and demonstrate that a small proportion of largely polyclonal T cells also carry the TET2 mutation. We report that the presence of both JAK2V617 and TET2 mutations favors accumulation of the mutated erythroid progenitors, while in similar conditions the PV JAK2V617-positive and TET2-negative cells are at a proliferative disadvantage compared to normal erythroid progenitors. We also examined the clonality of these in vitro-expanded erythroid progenitors to determine if the dormant minor populations of nonclonal hematopoietic cells are preferentially expanded along with those belonging to the PV clone. We found that some PV erythroid progenitors with JAK2V617F but no known TET2 mutations became polyclonal after in vitro erythroid expansion, while two PV patients with JAK2V617F and TET2 mutations remained clonal, suggesting that the TET2 mutated clonal progenitors retained their proliferative advantage. Lastly, compared to normal erythroid progenitors wherein TET2 mRNA increases with erythroid maturation, it decreases in PV erythroid progenitors regardless of the presence of a TET2 mutation. As predicted, the intronic TET2 mutation causing aberrant splicing had decreased TET2 expression compared to controls and other PV samples in all cells examined. However, the TET2 mRNA transcript in peripheral blood granulocytes and platelets in JAK2V617F positive PV, regardless of TET2 mutations, was significantly increased compared with normal controls. We conclude that loss-of-function TET2 mutations in the two studied PV subjects are not the PV initiating events. Our data suggest that these TET2 mutations in PV preferentially affect the erythroid lineage, contribute to increased erythroid proliferation, and cause relative inhibition of PV granulopoiesis and megakaryopoiesis. However, in aggregate, these in vitro data also suggest that the acquisition of the TET2 somatic mutations increases the aggressivity of the PV clone. Disclosures: No relevant conflicts of interest to declare.


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.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 808-808 ◽  
Author(s):  
Raajit K. Rampal ◽  
Suveg Pandey ◽  
Omar Abdel-Wahab ◽  
Jennifer J Tsai ◽  
Taghi Manshouri ◽  
...  

Abstract Abstract 808 A subset of patients with Philadelphia-chromosome negative myeloproliferative neoplasms (MPNs) (Polycythemia Vera (PV), Essential Thrombocytosis (ET), and Primary Myelofibrosis (PMF)) subsequently transform to acute myeloid leukemia (AML). Leukemic transformation (LT) after MPN occurs in as many as 23% of PMF patients within 10 years of diagnosis, and in 4–8% of PV and ET patients in the first 18 years after diagnosis. The development of AML after an antecedent MPN is associated with a dismal clinical outcome, and is associated with a poor response to conventional anti-leukemic therapies. Although somatic mutations in the JAK-STAT signaling pathway, including in JAK2 and MPL, occur in the majority of MPN patients, the somatic mutations that drive LT from a pre-existing MPN have not been fully delineated. Recent candidate mutational studies have identified recurrent somatic mutations in a subset of known leukemogenic disease alleles at the time of transformation from MPN to AML, including mutations in TP53, IDH1/2, TET2 and SRSF2 as well as deletions in IKZF1. However, the functional contribution of these specific genetic events to LT has not been delineated, and genetically accurate models of transformation of Philadelphia-chromosome negative MPN to AML have not been reported to date. In order to develop a genetically accurate murine model of LT, we have modeled expression of JAK2V617F mutation in combination with TP53 loss in vivo to further our understanding of progression from MPN to AML and to use this preclinical model of LT to test novel therapies. Bone marrow (BM) cells from C57/Bl6 Tp53−/− and littermate control mice were infected with JAK2V617F-IRES-GFP retrovirus, followed by transplantation of transduced cells into lethally irradiated congenic recipients. Of note, transplantation of JAK2V617F/Tp53−/− cells, but not JAK2V617F positive cells was associated with impaired survival; 50% of mice injected with JAK2V617F/Tp53−/− cells died by day 100, whereas all mice injected with JAK2V617F positive cells survived 100 days or longer (p=0.011) (figure 1). Mice injected with JAK2V617F/Tp53−/− cells presented with significant leukocytosis, with a mean WBC of 38.4 in mice engrafted with JAK2V617F/Tp53−/− cells compared with 11.4 in JAK2V617F/Tp53 wildtype mice. At the time of sacrifice, all mice engrafted with JAK2V617F/Tp53−/− cells had increased numbers of blasts in the peripheral blood and bone marrow, as assessed by morphologic evaluation and flow cytometric analysis which noted CD117 expression on leukemic blasts. BM cells from mice engrafted with JAK2V617F/Tp53−/− cells were characterized by increased serial replating (>10 platings), which was not observed in plating studies with JAK2V617F positive cells. In addition, we noted that the disease from JAK2V617F/Tp53−/− cells, but not JAK2V617F positive cells, was transplantable into secondary recipients consistent with increased self-renewal in vivo. We have begun testing the efficacy of novel therapies in this murine model, using both in vitro assays and in vivo studies in secondary transplantation studies. Treatment with the JAK kinase inhibitors INCB18424 and CYT 387 resulted in dose-dependent inhibition of colony formation in vitro. The combination of INCB18424 and Decitabine (which has demonstrated clinical efficacy in post-MPN-AML) is associated with synergistic inhibitory effects in vitro. Based on these results, we are performing in vivo studies with INCB18424, Decitabine, and INCB18424 + Decitabine, and results from these preclinical therapeutic studies will be presented in detail. Taken together, our data demonstrate that expression of JAK2V617F plus Tp53 loss, a genoptype commonly seen in patients who transform to AML after MPN, efficiently models LT in vivo. This model can now be utilized to examine the mechanisms of leukemic transformation, including assessment of the leukemic cell of origin in transformed disease. In addition this model can be utilized to test novel therapeutic strategies in a preclinical setting, which can be used to inform clinical trials in this poor-risk hematologic malignancy. Figure 1: Survival curve of mice transplanted with JAK2V617F in presence and absence of Tp53 Figure 1:. Survival curve of mice transplanted with JAK2V617F in presence and absence of Tp53 Disclosures: Verstovsek: Incyte Corporation: Research Funding; Novartis: Research Funding; AstraZeneca: Research Funding; Celgene: Research Funding; SBIO: Research Funding; Lilly Oncology: Research Funding; Bristol-Myers: Research Funding; Geron Corp.: Research Funding; Gilead: Research Funding; YM Biosciences: Research Funding; Roche: Research Funding; NS Pharma: Research Funding; Infinity Pharmaceuticals: Research Funding.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 798-798
Author(s):  
Baobing Zhao ◽  
Yang Mei ◽  
Ronen Sumagin ◽  
Jing Yang ◽  
Chelsea Thorsheim ◽  
...  

Abstract V617F driver mutation of JAK2 is the leading cause of the Philadelphia-chromosome-negative myeloproliferative neoplasms (MPNs). Studies on the pathogenesis of JAK2V617F positive MPNs have primarily focused on deregulation of JAK2 and STAT activities. It remains unclear exactly how JAK2-STAT downstream targets are involved in the development of the MPNs. We previously reported that pleckstrin-2 (Plek2) plays an important role in terminal erythropoiesis in vitro. Here we show that Plek2 is a downstream target of the JAK2-STAT5 pathway in erythroid, megakaryocytic, and granulocytic cells. Plek2 is significantly upregulated by JAK2V617F in both transduced primary mouse hematopoietic cells and in patients with JAK2V617F positive MPNs. Using a JAK2V617F knockin mouse model, we demonstrated that loss of Plek2 ameliorated JAK2V617F-induced myeloproliferative phenotypes including reticulocytosis, thrombocytosis, neutrophilia, and splenomegaly, thereby reverting the widespread vascular occlusions and lethality of JAK2V617F knockin mice (Figure 1A). These phenotypes were also transplantable indicating the role of Plek2 in mediating the pathogenesis of JAK2V617F-induced MPNs is cell-intrinsic (Figure 1B). Our study identifies Plek2 as a novel effector of JAK2-STAT5 pathway and a key factor in the pathogenesis of JAK2V617F-induced MPNs. Figure 1. Loss of Plek2 Rescues The Lethality of JAK2V617F Knockin Mice. (A) Kaplan-Meier survival analysis of indicated mice. Both males and females were included in each group. JAK2+/+Plek2+/+mice, n=34; JAK2+/+Plek2-/-mice, n=34;JAK2VF/+Plek2+/+ mice, n=36; JAK2VF/+Plek2-/- mice, n=36. (B) Kaplan-Meier survival analysis of the transplanted mice. JAK2+/+Plek2+/+mice, n=10; JAK2+/+Plek2-/-mice, n=10;JAK2VF/+Plek2+/+ mice, n=17; JAK2VF/+Plek2-/- mice, n=17. Figure 1. Loss of Plek2 Rescues The Lethality of JAK2V617F Knockin Mice. (A) Kaplan-Meier survival analysis of indicated mice. Both males and females were included in each group. JAK2+/+Plek2+/+mice, n=34; JAK2+/+Plek2-/-mice, n=34;JAK2VF/+Plek2+/+ mice, n=36; JAK2VF/+Plek2-/- mice, n=36. (B) Kaplan-Meier survival analysis of the transplanted mice. JAK2+/+Plek2+/+mice, n=10; JAK2+/+Plek2-/-mice, n=10;JAK2VF/+Plek2+/+ mice, n=17; JAK2VF/+Plek2-/- mice, n=17. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3214-3214
Author(s):  
Sucha Nand ◽  
Manuel O. Diaz ◽  
David E. Marinier ◽  
Mark A. Walshauser ◽  
Amy B Rosenfeld ◽  
...  

Abstract Background: Familial MPNs are uncommon disorders that, like sporadic cases, are characterized by clonal hematopoiesis and presence of somatic mutations, e.g. JAK2, CALR, MPL and occasionally TET2. There is little information, however, about germ-line mutations in these families that may explain the low penetrance hereditary predisposition. Methods: We studied five families with MPNs, each with at least 2 affected members. After obtaining an informed consent, clinical data was obtained from the patients’ electronic medical records. Blood and buccal samples were collected from patients and unaffected relatives. Exome sequencing was performed on the blood DNA samples using Agilent SureSelect Human All Exon V5+UTRs exome capture kit followed by massively parallel sequencing with Illumina HiSeq 2000. Sanger sequencing was then done on both the blood and buccal swab DNA samples to validate selected gene variants and to differentiate the nature of those variants (germ line or somatic). Results: The 5 families participating in this study had the following diagnoses: 1. Mother: polycythemia vera (PV); son: essential thrombocythemia (ET), 2. Mother: primary myelofibrosis (MF); daughter: unclassifiable MPN (UMPN), 3. Father: PV; son: PV, 4. Sister: MF; sister: MF, 5. Two aunts: MF; niece; UMPN. Six patients were positive for JAK2, V617F mutation. Blood and buccal samples were collected from 5 patients and 4 relatives. In all 5 families, the pro-band was younger at the time of diagnosis than his/her affected relatives. The clinical course of the MPNs appeared to be similar to the sporadic form. Exome sequencing revealed TET2 mutations in 2 probands. In addition, novel non-synonymous mutations in several candidate genes, KMT2D, KMT2C, NBEAL1, NBEAL2, AHNAK2, RNF213, were identified in the blood samples from the patients but not their unaffected relatives. These include two novel KMT2D mutations in two unrelated families. These 2 mutations were also found in the matching buccal swab samples, indicating that they are germ line mutations. Discussion: KMT2D and KMT2C mutations have been previously identified as somatic mutations in lymphoid malignancies, including non-Hodgkin’s lymphomas (Morin 2011), and as germ line compound heterozygote mutations in infant MLL and ALL (Valentine 2014). About 32% of diffuse large cell lymphoma and 89% of follicular lymphoma have somatic mutations of KMT2D. NBEAL2 germ line mutations are associated with familial gray platelet syndrome, where some patients have myelofibrosis (Gunay-Aygun 2011). To our knowledge, this is the first report describing germ line mutations in familial MPNs. The possible role of these mutations in predisposition to MPN will be discussed. Studies on additional families with MPNs are planned. 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.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3827-3827
Author(s):  
Francesca Ferraro ◽  
Christopher A Miller ◽  
Amy Abdalla ◽  
Nichole Helton ◽  
Nathan Salomonis ◽  
...  

Currently, it is not clear why some patients with acute myeloid leukemia (AML) can be "cured" with chemotherapy alone; are they living with small amounts of disease that is held in check by immunologic (or other) mechanisms, or is their disease really eradicated? The percentage of cytogenetically normal AML patients who have long (>5 years) first remissions (LFRs) after chemotherapy alone is low (about 9.1% in patients <60 years and 1.6% in >60 years1). For this reason, most intermediate risk patients are offered allogeneic transplantation to decrease their risk for relapse. To better understand mechanisms of chemotherapy sensitivity in AML, we performed an analysis of the mutation landscape and persistence, using samples from 8 normal karyotype LFR patients (without CEBPA mutations) who received standard "7+3" induction and high dose cytarabine consolidation as their only therapy. The mean age at diagnosis was 43.5 years, and the mean follow up in first remission is 7.6 years; none of these patients has relapsed to date. For each case, we performed enhanced exome sequencing at diagnosis (235x coverage of the entire exome, and ~1008x coverage of recurrently mutated AML genes). Each case had at least one documented AML driver mutation, with a median of 29 somatic mutations in the exome space. We created probes for 225 mutations (mean 28 per case), and performed error-corrected sequencing (Haloplex) for all available remission samples. The mean depth of Haloplex coverage was 1607x, and each sample had at least one AML-specific mutation assayed, with a sensitivity of 1 cell in 1,750 (0.06%). 7/8 patients demonstrated complete clearance of all mutations in all remission samples tested, which was confirmed with digital droplet PCR for 5 cases, with a sensitivity of detection of 1 cell in 100,000. In one case, we detected a persistent ancestral clone harboring DNMT3AR882H, which can be associated with long first remissions for some patients2. Strikingly, the founding clone in all 8 cases had one or more somatic mutations in genes known to drive cell proliferation (e.g. MYC, FLT3, NRAS, PTPN11, Figure 1 top panel). These are usually subclonal mutations that occur late during leukemic progression, suggesting that the presence of a "proliferative hit" in the founding clone might be important for chemotherapy clearance of all the AML cells in a given patient. To support this hypothesis, we analyzed the mutational clearance of 82 AML cases with paired diagnosis and day 30 post-chemotherapy bone marrow samples. We observed that, whether present in the founding clone or in subclones, mutations in MYC, CEBPA, FLT3, NRAS, and PTPN11 cleared after induction chemotherapy in all samples, while other mutations were often persistent at day 30 (e.g. DNMT3A, IDH1, IDH2, NPM1, TET2; Figure 1 bottom panel). Compared to other published sequencing studies of AML, MYC and NRAS mutations were significantly enriched in this small cohort (MYC p= 0.002, and NRAS p= 0.034), with MYC enrichment being particularly striking (37.5% versus 1.8%). All MYC mutations were canonical single base substitutions occurring in the highly conserved MYC Box 2 domain at the N-terminus of MYC (p.P74Q or p.T73N). Overexpression of MYCP74Q in murine hematopoietic progenitors prolonged MYC half life (89 min vs. 44 min for wild type), and enhanced cytarabine sensitivity at all concentrations tested (range 10-1000 nM, p=0.0003), both in vitro and in a MYC-driven leukemia model in vivo. MYC expression measured with flow cytometry in the blasts of the LFR samples was significantly higher (p=0.045) compared to unfavorable risk (complex karyotype) or other intermediate risk categories, but similar to good risk AML (biallelic CEBPA mutations, core binding factor fusion-associated AML, and AML with isolated NPMc), suggesting that activation of the MYC pathway may represent a shared feature of chemosensitive patients. Taken together, these data suggest that some intermediate patients who are effectively "cured" with chemotherapy alone may not have persistent subclinical disease, nor retained ancestral clones that could potentially contribute to relapse. Importantly, these patients often have mutations driving cell proliferation in the founding clone, indicating that the presence of specific mutations in all malignant cells may be critical for complete AML cell clearance with chemotherapy. 1. Blood Adv. 2018 Jul 10; 2(13): 1645-1650 2. N Engl J Med 2018; 378:1189-1199 Disclosures No relevant conflicts of interest to declare.


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 ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4979-4979
Author(s):  
Edmond A. Bendaly ◽  
Saud S. Rahman ◽  
Samiah Zafar ◽  
Karen Haglof ◽  
Sherif Ibrahim ◽  
...  

Abstract Abstract 4979 Introduction The JAK2V617F mutation accounts for most cases of myeloproliferative neoplasms (MPN). Only a few case reports of MPN following cytotoxic chemotherapy have been reported, and all of them were published prior to the discovery of the JAK2V617F mutation. We report a series of 6 patients who developed a JAK2V617F positive MPN following cytotoxic chemotherapy. Patients From 2006 to 2009, 6 patients with a history of a hematologic or an oncologic malignancy who developed an MPN were identified and their medical records retrospectively reviewed. One patient had acute lymphoblastic leukemia, 1 had Hodgkin lymphoma, 1 had squamous cell carcinoma of the head and neck, 1 had cervical cancer, and 2 had breast cancer. All patients were in remission from their primary malignancies at the time the MPN was diagnosed. Five were females. The median age at diagnosis was 72 years. Median time to development of the myeloproliferative neoplasm was 14 years. Type of chemotherapy exposure, MPN diagnosis and time to MPN in each case is shown in the table below. The JAK2V617F mutation was detected either in the peripheral blood or the bone marrow of all patients. There was no predominance of any specific MPN diagnosis. Patients who received platinum-based chemotherapy developed the MPN sooner than those who received alkylators (6 vs 17.5 years respectively). Treatment consisted of phlebotomy, hydroxyurea, anagrelide, aspirin or a combination as deemed appropriate by the treating hematologist. Conclusion These findings lead us to hypothesize whether the development of JAK2V617F positive MPN may be related to prior exposure to cytotoxic chemotherapy. Exposure to platinum-based chemotherapy may cause the disorder to appear sooner compared with exposure to alkylators. Recently, JAK2V617F positive MPN was found to be strongly associated with a specific constitutional haplotype, 46/11 suggesting increased susceptibility to this mutation. Chromosomal analyses are planned to show whether any of the reported patients exhibit this haplotype. Ref: 1.Jones et al, Nat Genet. 2009 Apr;41(4):446-9. 2009 Mar 15. The authors have no relevant disclosure. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document