Clonal Evolution of Gene Mutations Involving DNA Methylation in the Progression of CMML to Secondary AML

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 4120-4120
Author(s):  
Hsiao-Wen Kao ◽  
Ming-Chung Kuo ◽  
Po-Nan Wang ◽  
Jin-Hou Wu ◽  
Tung-Huei Lin ◽  
...  

Abstract Background and Aim: The molecular pathogenesis of progression of chronic myelomonocytic leukemia (CMML) to secondary acute myeloid leukemia (sAML) remains incompletely understood. Genemutations involving DNA methylation in the transformation of CMML to sAML were investigated in matched paired CMML/sAML bone marrow samples to determine the roles of TET2, DNMT3A, IDH1 and IDH2 mutations in the evolution of CMML to sAML. Material and Methods: 106 CMML (63 CMML-1 and 43 CMML-2) patients were analyzed for TET2, DNMT3A, IDH1, and IDH2 mutations at the initial diagnosis. 33 patients had paired CMML/sAML bone marrow samples for comparative analyses. Mutational analysis of TET2 was performed by PCR followed by direct sequencing for PCR products amplified with primer pairs covering the whole coding sequences (exons 3-11). DNMT3A mutations (exons 2-23) were screened by denaturing high-performance liquid chromatography (DHPLC) with adding GC-clamps to the primers to facilitate mutation detection.Samples with abnormal DHPLC profile were then directly sequenced. The hot spots of IDH1 and IDH2 genes on exon 4 were PCR-amplified from gDNA and subjected for direct sequencing. Additional gene mutations were analyzed by PCR-based assays with direct sequencing. The allele burden of gene mutations was measured at both CMML and sAML phases by pyrosequencing with a detection sensitivity of 5%. Results: The frequencies of TET2, DNMT3A, IDH1 and IDH2 in 106 CMML patients were 39.8% (41/103), 8.5% (9/106), 0% (0/106), and 7.5% (8/106), respectively. These epigenetic gene mutations were mostly mutually exclusive. Of the 31 paired CMML/sAML samples examined for DNMT3A, 4 had DNMT3A mutations at diagnosis; the mutation status, patterns and allele burden remained unchanged at sAML phases. None of 33 patients acquired IDH1 mutation and one acquired IDH2 mutation during sAML progression. Of the 30 patients with paired samples analyzed for TET2 mutation, 12 patients had TET2 mutations at both CMML and sAML phases; 10 patients retained the same TET2 mutations with stable allele burden, one patient had clonal expansion of TET2 mutation, and the other patient acquired 3rdTET2 mutation at sAML progression along with expansion of a preexisted TET2 mutant clone and one stable TET2 mutant subclone. Another patient harboring TET2 mutation at CMML diagnosis lost themutation at sAML progression. Acquisition of additional gene mutations during sAML evolution was detected in 5 TET2-mutated patients, including RUNX1, CEBPA, FLT3- ITD, JAK2 V617F, NPM1, SRSF2, and CSF3R, either alone or in combination. Conclusions: Our results showed that TET2 and DNMT3A mutational status and allele burden remained unchanged during the progression of CMML to sAML except that rare patients might have expansion or emergence of TET2 subclone at sAML phase. Acquisition of additional gene mutations occurred in half of TET2-mutated patients during the progression of CMML to sAML. Grant support: NHRI- EX103-10003NI and MOHW103-TD-B-111-09 Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1969-1969
Author(s):  
Ming-Chung Kuo ◽  
Po-Nan Wang ◽  
Po Dunn ◽  
Tung-Liang Lin ◽  
Jin-Hou Wu ◽  
...  

Abstract Background and Purpose: The molecular pathogenesis of progression of myelodysplastic syndromes (MDS) to secondary acute myeloid leukemia (sAML) remains incompletely understood. We studied genemutations involving spliceosome in the transformation of MDS to sAML by comparing matched paired MDS/sAML bone marrow samples to determine the roles of SRSF2, U2AF1 SF3B1, and ZRSR2 mutations in the evolution of MDS to sAML. Patients and Methods: One hundred and forty-nine de novo MDS patients (2 RCUD, 5 RARS, 27 RCMD, 52 RAEB-1, and 63 RAEB-2) were examined forspliceosome mutations at initial diagnosis and 93 patients progressed to sAML with a median follow-up of 16.4 months. Seventy one of them had paired MDS/sAML bone marrow samples available for comparative analyses. Mutational analyses of spliceosome were performed by pyrosequencing with a detection sensitivity of 5% for U2AF1 (exons 2 and 6) and SRSF2 (P95 of exon 1) mutations and by direct sequencing for SF3B1 (exons13-16) and ZRSR2 (whole coding exons 1-11) mutations. Additional 29 gene mutations known to involve in myeloid neoplasms were also analyzed by direct sequencing or next-generation sequencing (NGS, Ion Torrent PGM) followed by Sanger sequencing validation; NGS was mainly used for mutation detection of epigenetic regulators and cohesin complex. The allele burden of targeted genes was determined by pyrosequencing and/or NGS. Results: The frequencies of U2AF1, SRSF2, SF3B1, and ZRSR2 mutations in the 149 MDS patients were 14.8% (22/149), 12.8% (19/149), 12.1% (17/141), and 7.5% (10/133), respectively. Together, spliceosome mutations occurred in 51.5% of MDS patients at initial diagnosis and were mutually exclusive. All the 5 RARS patients had SF3B1 mutations. Co-existed mutations with epigenetic regulators were detected in 36 patients (52.9%), with RUNX1 in 13 (19.1%), with cohesin complex including STAG2, SMC3, RAD21,or SMC1A, in 13 (19.1%), with signaling pathways including RAS, PTPN11, JAK2, or FLT3-TKD in 4 (5.9%), BCOR in 2, and one each with CEBPa and SETBP1. There were no differences in blood counts, percentage of blasts in bone marrow and blood, WHO subtype, cytogenetic risk group, and IPSS-R between spliceosome-mutated and -unmutated patients. Of the patients carrying spliceosome mutations, only SRSF2 mutation had prognostic impact on predicting a higher risk of sAML transformation (P = 0.025) and sAML-free survival (median 10.8 months, 95% CI 5.1-16.5 months) compared to SRSF2-unmutated patients (median 17 months, 95% CI 8.5-25.5 months, P = 0.050). Of the 71 paired MDS/sAML samples, 37 (52.1%) had spliceosome mutations at diagnosis; the mutational status and patterns remained unchanged in all the 37 matched sAML samples but allele burden was apparently increased in the sAML samples with SRSF2 (P = 0.017) or SF3B1 (P = 0.015) mutations. In addition, one ZRSR2 mutant clone and two SRSF2 mutant clones evolved during sAML progression. Acquisition of other mutated genes was found in 37 spliceosome-mutated patients at sAML phases, including RUNX1 in 5, N-RAS in 5, CEBPa in 4, K-RAS in 3, FLT3-ITD in 3, and one each with ASXL1, TET2, STAG2, WT1, PTPN11, CBL, FLT3-TKD, and C-FMS. Notably, of the 3 patients acquiring spliceosome mutations, none gained other mutated genes during sAML transformation.Clonal expansion of other mutated genes were observed in 4 cases with RUNX1, in 2 with TET2, and one each with N-RAS, CEBPa, ASXL1, SMC1A, and STAG2. Conclusions: Our results showed that spliceosome mutations occurred in more than half of de novo MDS patients at initial diagnosis. Clonal expansion, evolution, or unchanged allele burden of spliceosome mutations might occur during sAML transformation with frequent acquisition of additional mutated genes. SRSF2 mutation predicted a higher risk and more rapid sAML transformation. (Grants support: NHRI-EX103-10003NI, MOHW103-TD-B-111-09 and CMRPG3D1532) Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 1455-1455
Author(s):  
Jung Sook Ha ◽  
Jae Hee Lee ◽  
Sung Gyun Park ◽  
Nam Hee Ryoo ◽  
Dong Suk Jeon ◽  
...  

Abstract Abstract 1455 Background: Since the acquired somatic mutation, JAK2 V617F, was discovered as a first molecular marker of myeloproliferative neoplasms (MPN), and it has been detected variably in each MPN subtypes. However, JAK2 V617F does not found in all of MPN cases and not necessarily specific to a particular clinicpathologic entity. Recently, mutation of the putative tumor suppressor gene, Ten-Eleven-Translocation-2(TET2), has been identified in MPN patients. However, the frequency of TET2 mutation or its relationship with JAK2 V617F mutation or pathologic function in MPN has not been concluded, yet. The aim of our study was to evaluate the frequency of TET2 in MPN patients, and whether there is any correlation of TET2 mutation with JAK2V617F mutation or the clinicohematologic parameters. Materials and Methods: Total 99 adult MPN patients (18 PV, 62 ET, 11 PMF and 8 MPN unclassified) whose bone marrow cells had been stored from 2007 to 2010 at point of first diagnosis were included in this study. Hematological diagnoses and subtyping were reconfirmed according to the 2008 WHO classification and clinicohematologic datas were collected from patient records. Direct sequencing for TET2(exon3–11) and JAK2 (exons 12 and 14) were performed using an ABI 3730XL DNA analyzer. The JAK2V617F allele burdens were determined by pyrosequencing for samples available and MPL was analyzed by allele-specific PCR. Results: The overall TET2 mutational frequency was 12.1%, and disease-specific mutational frequencies were 22.2% in PV, 9.7% in ET and 18.2% in PMF. The found mutations included 11 mutations, 7 frame-shift (p.Lys95AsnfsX18, p.Gln967AsnfsX40, p.Lys1022GlufsX4, p.Asp1314MetfsX49, p.Gln1534AlafsX43, p.Tyr1618LeufsX4, p.Leu1609GlufsX45), 1 nonsense (p.Gly1735X), 1 missense (Q599R) and 2 splicing mutations (c.3409+1G>T, c.4044+2insT). Those mutations most frequently involved exon 3(four mutations) and exon 11(four mutaions), and rarely intron 3, intron 8 and exon 7. None of the mutations were associated with a karyotypically apparent 4q24 rearrangement. All patients were also screened for JAK2 V617F, and the overall JAK2 V617F positive rate was 68%(94.4% in PV, 69.4% in ET, 45.5% in PMF and 37.5% in MPN, unclassified). All TET2 mutations occurred in JAK2 V617F positive cases. JAK2 exon12 mutation was not found in all patients. MPL W515L was found in one ET patient who also carried JAK2V617F, but not TET2 mutation. Information on JAK2 V617F allele burden was available in 78 patients. Considering all 99 patients, the patient age, hematologic indexes (leukocyte count, neutrophil fraction, lymphocyte fraction, monocyte fraction, Hb, Hct and platelet count), the frequency of organomegaly, marrow fibrosis or thrombotic/hemorrhagic complications were not different according to carrying TET2 mutation. However, TET2 mutation was more frequently found in JAK2 V617F carriers than non-carriers (P=0.008), but JAK2 V617F allele burden did not correlated with the presence of mutant TET2. When analysis was performed for each PV, ET, and PMF (no TET2 mutation in MPN-unclassifiable patients), correlation between TET2 and JAK2 V617F mutational status was not found in each subtypes (P=0.078 in PV, P=0.099 in ET and P=0.182 in PMF). However, the JAK2 V617F allele burden was significantly higher in PMF harboring TET2 mutation than PMF patients did not (88.0 ± 4.3% vs 19.1 ± 28.7%, P=0.034). In statistical analysis for the correlations of clinicohematologic parameters with TET2 mutation in each PV, ET and PMF patients, only a few statistically significant results were identified. The presence of TET2 mutation was correlated with high Hct in PMF (47.4 ± 5.4 vs 25.5 ± 6.2, P=0.037), and TET2 positive ET patients showed relatively higher frequency of organomegaly compared to ET patients without TET2 mutation (50% vs 19.6%, P=0.018). Conclusions: The overall and disease-specific frequencies of TET2 mutation in our study are similar with previous studies, and frame-shift mutation is the most frequent mutation type. There is no specific relationship between JAK2 V617F and TET2 mutation occurrence, but TET2 mutant PMF has higher JAK2 V617F allele burden than non-mutant. TET2 mutation is also associated with a higher Hct in PMF and higher frequency of organomegaly in ET. Larger scale studies involving more MPN patients are needed. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 1407-1407
Author(s):  
Hsiao-Wen Kao ◽  
Lee-Yung Shih ◽  
Ming-Chung Kuo ◽  
Tung-Liang Lin ◽  
Sung-Tzu Liang ◽  
...  

Abstract Background and purpose Abnormalities of genes regulating DNA methylation have been described in acute myeloid leukemia (AML). MLL protein is a transcriptional regulator and governs proper hematopoiesis through its histone methyltransferase activity. AML with partial tandem duplication of MLL (MLL-PTD) was associated with an unfavorable prognosis. The cooperating roles of MLL-PTD with other mutated genes regulating DNA methylation have not been comprehensively studied in AML. We aimed to determine the prevalence and clinical impact of mutations of DNA methylation regulators in AML with MLL-PTD. Materials and methods Bone marrow samples from 98 AML patients with MLL-PTD were analyzed for gene mutations of TET2, DNMT3A, IDH1 and IDH2. MLL-PTD was screened by RT-PCR and confirmed by real-time quantitative PCR assays. The mutational analysis was performed with PCR assays followed by direct sequencing for TET2 (whole coding exons 3–11) and IDH1/2 (hotspots exon 4). For the detection of DNMT3A mutations, the PCR products amplified for entire coding exons 2 to 23 were first screened with denaturing high-performance liquid chromatography followed by direct sequencing for the abnormal profiles. Results The frequency of TET2, IDH1, IDH2 and DNMT3A mutations in AML patients with MLL-PTD was 17.0% (16/94), 10.2% (10/98), 18.4% (18/98), and 31.6% (31/98), respectively. Taken together, 61.1% of patients with MLL-PTD had at least one mutated gene of DNA methylation regulators. TET2, IDH1 and IDH2 mutations were mutually exclusive with each other whereas DNMT3A mutations frequently co-existed with other DNA methylation modifiers:TET2 (n=8), IDH1 (n=5) and IDH2 (n=4). No differences were observed between the mutation status of the DNA methylation modifiers and clinico-hematologic features of patients with MLL-PTD except that TET2 (P=0.012) and DNMT3A (P=0.024) mutations were associated with older age. Of the 55 MLL-PTD patients who received standard chemotherapy, IDH2 mutation was associated with a lower complete remission rate (25.0% vs 67.8%, P=0.018), while DNMT3A mutations conferred an inferior event-free survival (0.0 vs 6.8 months, P=0.027) and overall survival (6.0 vs 11.5 months, P=0.032). In multivariate analysis, older age (P=0.008) and DNMT3A mutations (P=0.049) were independent adverse factors for overall survival. The crosstalk between MLL-PTD and genes involving DNA methylation in the leukemogenesis of AML warrants further investigation. Conclusions Gene mutations involving DNA methylation frequently co-existed in AML patients with MLL-PTD, especially DNMT3A mutations which conferred a poor outcome. Our study demonstrated the importance of genetic alterations involving DNA methylation in the pathogenesis of MLL-PTD AML and provided potential epigenetic-targeted therapy. Grant support The work was supported by NHRI-EX93-9011SL, NSC95-2314-B-195-001, NSC96-2314-B-195-006-MY3, NSC97-2314-B-182-011-MY3 and MMH-E-101-09. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4983-4983 ◽  
Author(s):  
Hongxing Liu ◽  
Fang Wang ◽  
Wen Teng ◽  
Yuehui Lin ◽  
Junfang Yang ◽  
...  

Abstract Background Acute myeloid leukemia (AML) is a heterogeneous disease with respect to presentation and clinical outcome. In recent years, more and more somatic mutations and their clinical significance were identified in AML. Most AML patients carry multiple gene mutations and the repertoire of mutations changing during the disease process, which determine the patient's unique clinical manifestations. Herein we recommend using the novel word ”mutaome¡± for representing the repertoire of somatic gene mutations in a specific tumor tissue, and aimed to establish a panel of mutation profiling protocol and retrospective profiling using archived bone marrow smear for clinical use in AML. Methods and Cases Mutation profiling protocol for CEBPA, DNMT3A, FLT3-ITD/TKD, IDH1, IDH2, KIT, NPM1, PHF6 and TET2 by PCR and Sanger sequencing was established, with the detection sensitivity about 15% to 20%. Bone Marrow (BM) or peripheral blood (PB) was collected form patients with newly diagnosed or relapsed. Archived BM smear on glass slides at the time of newly diagnosed were used for retrospective mutation analysis. Results 1) Totally 61 archived smear, 106 fresh BM or PB and 2 paraffin-embedded pathological specimens from 157 patients were analyzed. Age ranged from 1 to 77 years old, with the median age of 27 years old. 2) 60.4% (102/169) samples carrying at least one mutation, 51.0% (53/102) of them carrying 2 or more mutations, 30.4% (31/102) carrying mutations within 2 or more different genes. The number of mutated sample for each gene is 33 for CEBPA¡¢9 DNMT3A¡¢35 FLT3¡¢6 IDH1¡¢7 IDH2¡¢7 KIT¡¢19 NPM1¡¢8 PHF6¡¢and 21 TET2. 3) Paired archived smear and relapsed samples were analyzed for 11 cases, 9 of them showing difference, detailed results shown in Table 1. 4) Totally 53 patients come to our hospital after certain treatment, with the tumor cells below 15% and without AML gene mutation results. For these patients, archived BM smear samples were used for retrospective mutation profiling and then to guide targeted therapy and stratified evaluation. Conclusions Panel testing for gene mutations is effective method for detection of AML molecular markers. For the patients came with partial remission and without gene mutation result, archived bone marrow smear sample can be used for retrospective mutation analysis and then help guiding targeted therapy and stratified evaluation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1993-1993
Author(s):  
Xiaohui Zhang ◽  
Alan F List ◽  
Jeffrey E. Lancet ◽  
Song Jinming ◽  
Lynn C. Moscinski ◽  
...  

Abstract Background: Pancytopenia and bone marrow aplasia/hypoplasia are caused by a heterogeneous group of disorders, most commonly aplastic anemia (AA), hypoplastic myelodysplastic syndrome (MDS), paroxysmal nocturnal hemoglobinuria (PNH), and T-cell large granular lymphocytosis (T-LGL). Clinical and morphological distinction among these entities is often challenging, particularly between AA and hypoplastic MDS. This study is to examine the clinicopathological and genetic features of a group of AA and hypoplastic MDS patients, with or without concurrent T-LGL and/or PNH, in order to better understand and differentiate the two entities. Methods and Materials: We retrieved 45 cases with cytopenias and hypoplastic bone marrow at Moffitt Cancer Center. Peripheral blood complete blood counts, bone marrow morphological findings, flow cytometric analyses for LGL and PNH, and cytogenetics data were extracted from electronic medical records. Targeted next-generation sequencing (54 myeloid neoplasm related genes) was performed on the bone marrow. Results: The 45 patients showed peripheral blood cytopenias and bone marrow aplasia or marked hypocellularity. There were 26 cases diagnosed with AA with no morphologic evidence of dysplasia or increased blasts in the bone marrow, and 19 cases diagnosed with hypoplastic MDS based on morphological and cytogenetic criteria. In the meantime, distinct T-LGL population was identified in 2 of 11 cases with AA (18.2%) and 3 of 11 cases with hypoplastic MDS (27.3%); PNH clones were identified in 8 of 17 cases with AA (47%) and 3 of 11 cases with hypoplastic MDS (27.3%). Clonal cytogenetic abnormalities were found in 2 of 25 cases with AA (8%) and 11 of 18 cases with hypoplastic MDS (61.1%). Twelve of 26 cases of AA (46%) showed one or more gene mutations with allele burden ranging from 7% to 52%, and most of these cases (9 of 12; 75%) involved only one gene. In contrast, 15 of 19 cases of hypoplastic MDS (78.9%) had one or more gene mutations with allele burden ranging from 19% to 53%. Seven of the 15 cases (46.7%) had two or more gene mutations. The most common mutated genes in the two groups in this study were ASXL1 and TET2. Conclusion: Although there are overlapping clinical and morphological features between AA and hypoplastic MDS, differences are present between the two entities including presence of PNH clones, cytogenetic changes, and gene mutation frequencies. These features may help to make differential diagnosis and identify the cases with more progression potential. Clinical outcomes with different treatment and larger scale studies are needed to better characterize and define the two different entities. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2786-2786
Author(s):  
Hsuan-Jen Shih ◽  
Lee-Yung Shih ◽  
Hung Chang ◽  
Ming-Chung Kuo ◽  
Tung-Huei Lin ◽  
...  

Abstract Background and purpose ASXL1 and EZH2 are histone modifiers. Mutations of ASXL1 and EZH2 genes have been described in both myelodyspolastic syndromes (MDS) and acute myeloid leukemia (AML). The role of ASXL1 or EZH2 mutations in the progression from MDS to secondary AML (sAML) is unclear. We aimed to determine the clinical relevance of ASXL1 and EZH2 mutations in patients with MDS and to investigate the role of ASXL1 or EZH2 mutation with its cooperating mutated genes in sAML progression. Methods ASXL1 and EZH2 mutations were analyzed on bone marrow samples from 126 patients with de novo MDS (45 RAEB1, 53 RAEB2, 24 RCMD and 4 RARS). Paired matched MDS/sAML samples were available for ASXL1 mutational analysis in 58 patients and for EZH2 in 54 patients. Mutational analysis of ASXL1 exon 12 was performed by PCR assays followed by direct sequencing, EZH2 mutations were first screened with denaturing high-performance liquid chromatography on amplified PCR fragments covering the whole coding sequencing from exons 2 to 20 of EZH2 gene followed by sequencing of the abnormal profile. Additional 20 known gene mutations in myeloid neoplasms were also examined in patients carrying ASXL1 or EZH2 mutations. Relative allele frequency was determined by pyrosequencing. Results Among the 126 patients, ASXL1 mutations were detected in 18 (14.3%) patients and EZH2 in 11 patients (8.7%), 3 of them had both mutations. Taken together, 20.6% of patients carried mutations of ASXL1 and/or EZH2. ASXL1-mutated patients had male predominance (17 out of 18 patients, P=0.012) and fewer circulating blasts (P=0.007). ASXL1-mutated and -unmutated patients had no difference in hemoglobin levels, white blood cell counts, platelet counts, cytogenetics, WHO subtypes, bone marrow blasts, IPSS-R or risk to sAML. ASXL1 mutations had no impact on overall survival (P=0.765) or time to sAML transformation (P=0.605). No significant difference was observed between EZH2 mutation status and clinicohematologic features or outcomes. Of the 58 paired samples, ASXL1 mutations were detected in 8 cases at diagnosis of MDS, all were also present at the time of sAML progression with no difference in the mutant allele burden (P=0.614), 2 patients acquired ASXL1 mutations. Five had EZH2 mutations at both phases of disease which exhibited a similar allele frequency (P= 0.434), none lost and one acquired EZH2 mutation at sAML phase. Progression to sAML was accompanied by additional gene mutations including RUNX1 (n=4), TET2 (n=3), CEBPα (n=2), PTPN11 (n=2), and one each for FLT3-ITD, CBL, MLL-PTD, DNMT3A, IDH2 and EZH2 in ASXL1-mutated patients; TET2 (n=4), RUNX1 (n=3), N-RAS (n=2) and single cases for DNMT3A, IDH1, and ASXL1 in EZH2-mutated patients. Cooperating mutations were either detected in both MDS and sAML samples or newly appeared in sAML samples except one who lost JAK2V617F mutation while acquired EZH2 and N-RAS mutations during sAML progression. Conclusions Our study on a large cohort of paired MDS and sAML samples demonstrated that ASXL1 or EZH2 mutations remained stable at both phases of disease in most patients. Clonal evolution can occur and cooperation of additional gene mutations is frequently detected in patients harboring ASXL1 or EZH2 mutations in the progression of MDS to sAML. Grant support This work was supported by NHRI-EX102-10003NI and DOH102-TD-C-111-006, Taiwan. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 40 (4) ◽  
pp. 321-329
Author(s):  
Talal AlMaghamsi ◽  
Naeem Iqbal ◽  
Nabil Abdullrahman Al-Esaei ◽  
Muhsina Mohammed ◽  
Kamel Zein Eddin ◽  
...  

ABSTRACT BACKGROUND: Some mutations of the cystic fibrosis transmembrane regulator ( CFTR ) gene may impair spermatogenesis or cause a congenital absence of the vas deferens that manifests as isolated male infertility. OBJECTIVE: Assess the frequency and analyze the spectrum of CFTR gene variations in Saudi men with primary infertility. DESIGN: Prospective, cross-sectional. SETTING: Tertiary care specialist hospital in Jeddah. PATIENTS AND METHODS: Genomic DNA was extracted from peripheral blood samples of Saudi men who presented with primary infertility to the outpatient andrology clinic with either azoospermia or oligoasthenoteratozoospermia. Polymerase chain reaction and direct sequencing were used to identify all variants of the CFTR gene. MAIN OUTCOME MEASURES: Proportion of the patients with a mutant CFTR gene and the spectrum of CFTR gene variations. SAMPLE SIZE: 50 infertile Saudi men. RESULTS: This study identified 10 CFTR gene variants in 7 (14%) subjects (100 chromosomes). The detected variants and polymorphisms were: c.1408G>A, c.4389G>A, c.2562T>G, c.869+11C>T, c.2909-92G>A, c.3469-65C>A, c.1210-6delT, c.1210-6T>A, c.2988+1G>A, and c.1210-13GT>TG. CONCLUSION: We demonstrated that 14% of the study subjects had one or more CFTR mutations and these were compounded in most of the affected patients. The spectrum of CFTR gene mutations in these subjects was similar to the mutations reported in other studies throughout the world. LIMITATIONS: Small sample size and the lack of a control group. CONFLICTS OF INTEREST: None.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4225-4225
Author(s):  
Rie Ohba ◽  
Kazumichi Furuyama ◽  
Shigeru Tsuchiya ◽  
Atsushi Manabe ◽  
Etsuro Ito ◽  
...  

Abstract Abstract 4225 Introduction: Sideroblastic anemia is characterized by anemia with the emergence of ring sideroblasts in the bone marrow. There are two forms of sideroblastic anemia. One is an inherited sideroblastic anemia, and another is an acquired sideroblastic anemia. Because sideroblastic anemia is a rare disease, there are few comprehensive studies of sideroblastic anemia, including clinical and genetic information. In the present study, we have performed national-wide survey of sideroblastic anemia in Japan to investigate the epidemiology and pathogenesis of the disease. Method: This study consists of three-step surveys. First, the patients of sideroblastic anemia were searched by simple questionnaire to hematological department of hospitals in all areas in Japan (first investigation). Next, detailed clinical information of sideroblastic anemia patients were collected (second investigation). Survey items were age of onset, gender, family history, hematology, bone marrow and biochemical findings. Then, genetic analyses of patients who were suspected inherited sideroblastic anemia were performed (third investigation). For the genetic analysis, mutations of ALAS2, ABC7, GLRX5, SLC25A38, which are known to be responsible genes for inherited sideroblastic anemia, were examined. Result: At the first investigation, sideroblastic anemia patients were surveyed in the 1086 institutions of Japan. There were 14 cases of confirmed or suspected cases of inherited sideroblastic anemia and 285 cases of suspected or confirmed cases of acquired sideroblastic anemia. These patients were subjects of second investigation. As of August 9, data of 99 patients have been collected. In these cases there are 7 cases of confirmed inherited sideroblastic anemia, 7 cases of suspected inherited sideroblastic anemia, 28 cases of refractory anemia with ring sideroblasts (MDS-RARS) and 57 cases of refractory cytopenia with multilineage dysplasia with ring sideroblasts (MDS-RCMD-RS). Median age of onset was 70.5 years old in MDS-RCMD-RS cases, whereas that of cases in inherited sideroblastic anemia was 14 years old. Hemoglobin level in inherited sideroblastic anemia and RCMD-RS was 6.7g/dl and 8.3g/dl. MCV was 64.7 fl, 105.1 fl in inherited sideroblastic anemia and RCMD-RS. There was thus significant different between MCV level in inherited sideroblastic anemia and RCMD-RS. Chromosomal abnormality of +8 and idic (X) (q13), associated with ABC7 gene, were detected in 7 and 2 of 25 RCMD-RS patients, respectively. At the moment, 4 confirmed and 4 suspected patients of inherited sideroblastic anemia proceeded to third investigation. As a result, mutations of ALAS2, which is the first enzyme of heme biosynthesis in erythroid cells, have been identified in 6 out of 8 patients. The amino acid mutations were detected in exon 5 (R170C, R170L), exon 9 (R411C, R452C), and exon 11 (V562A). For patients without mutations in ALAS2 gene, mutations of other genes related to inherited sideroblastic anemia have been analyzed; however, no mutations are identified so far. Conclusion: The results showed that RCMD-RS is most common in sideroblastic anemia, and XLSA is most frequent type of inherited sideroblastic anemia. However, there are significant number of suspected cases of inherited sideroblastic anemia. The genetic analysis of these cases, including RARS without chromosomal anomaly, is currently in progress. This research has been supported by Grant-in-Aid for Scientific Research from Ministry of Health, Labour and Welfare of Japan. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4992-4992 ◽  
Author(s):  
Christian Langer ◽  
Mariam Ibañez ◽  
Peter Liebisch ◽  
Thorsten Zenz ◽  
Stefan Knop ◽  
...  

Abstract Abstract 4992 Multiple myeloma (MM) is characterized by frequent and complex genomic abnormalities. However, most genetic abnormalities are already present in the precursor state of MM, the monoclonal gammopathy of undetermined significance (MGUS). Therefore, it is likely that secondary genetic events might contribute to the development from MGUS to symptomatic MM. Mutations in isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2) genes have recently been described as relatively frequent molecular lesions in gliomas and in acute myeloid leukemia (AML). However no larger study has so far examined the frequency of IDH1 and IDH2 gene mutations in patients (pts) with MM. Methods: In 188 pts with MM genomic DNA from CD138 sorted plasma cells was used for analyses. Exon 4 of both IDH1 and IDH2 were amplified by PCR and the amplicons were analyzed using a combination of denaturing high-performance liquid chromatography and DNA sequencing. All patients were also characterized by a comprehensive set of FISH probes for the presence of recurring cytogenetic abnormalities. Results: 185 out of 188 samples were evaluable for analyses. One missense mutation in the IDH2 gene (c.G419A) was identified in the cohort of 185 MM pts (0.5%). This mutation was described as the most frequent IDH2 mutation in AML and is predicted to cause an amino acid change from arginine to glutamin at position 140 (p.R140Q). On cytogenetic analysis this patient harbored a translocation t(11;14) resulting in aberrant expression of CCND1. Additionally, in 15 pts (8%) the recently described single nucleotide polymorphism (SNP) in the IDH1 gene (rs11554137) was detected that has been reported as an adverse prognostic factor in cytogenetically normal AML. Summary: Mutations in the IDH1/2 genes are a rare event in MM (0.5%). Further studies are warranted to address the issue if IDH1/2 mutations are restricted to distinct genetic subgroups as for example the group of MM pts with translocation t(11;14). Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 5259-5259
Author(s):  
Ali Tabarroki ◽  
Valeria Visconte ◽  
Heesun J. Rogers ◽  
Juraj Bodo ◽  
Li Zhang ◽  
...  

Abstract Pharmacologic therapies that target the JAK-STAT pathway are clinically used to alleviate splenomegaly and disease-related constitutional symptoms in MF. However, it is clear that some patients develop intolerance or resistant to this therapy. Furthermore, there are MF related complications especially cytopenias that are not alleviated by these therapies. Therefore, alternative and complementarytherapies are warranted in the management of MF. We hypothesized that other pathways downstream of the JAK-STAT signaling pathway can play a role in the pathophysiology of MF. We used whole exome (WES) and RNA sequencing technologies to interrogate new molecular markers and pathways which can serve as novel targets for this disease. In 4 MF patients [JAK2 mutant (MUT) =2, and wild type (WT) =2], WES was performed using the Illumina platform. All of the variants were filtered based on PHRED score (>=30) with coverage was set at 30X. Analysis of data in JAK2/MPL WT patients demonstrated the presence of 263 candidate genes. After clarifying the status of tumor nucleotide variants in each gene compared to germline (CD3+) fraction, 7 genes (RBL1, ADSS, ZNF717,MUC4, TUBB4Q and CDC25A) were selected for further somatic confirmation by direct sequencing. Among these genes, only alteration in CDC25A, a regulator of cyclinE/cdk2 (cyclin-dependent kinase-2) and cyclinA/cdk2 kinase, was confirmed to be somatic. This genetic change was previously reported as somatic by WES in lung cancer although not confirmed by direct sequencing (Bartkova et al, Nature, 2005, Apr 14; 434 (7035):864-70). Based on these observations and since CDC25A acts as a downstream effector of JAK-STAT signaling, we hypothesized that, CDC25A phosphatase, may be a driver in MF pathogenesis. The transcriptome of two patients, one MUT and one WT for JAK2 was then analyzed. RNA was isolated from bone marrow (BM) cells of healthy individuals (HI) (N=3). cDNA was made from 1.5-3 ug of RNA and fragmented for library preparation. RNA-sequencing was performed on 20 million sequence reads. Paired-end 90 base pair reads were generated on an Illumina HiSeq2000 sequencer and aligned to the human genome 19. RNA-splicing patterns were analyzed by a bioinformatics algorithm and gene expression analysis was carried out using GSEA (Visconte V; Blood. 2012). By using FDR<0.2, 11,460 genes were expressed. Further analysis demonstrated, CDC25A was over-expressed in both cases compared to HI but interestingly more highly expressed in JAK2 WT cases [fold change (FC): 0.39].This finding was validated by performing Western blotting and immunohistochemistry (IHC). To evaluate the protein expression of CDC25A in 10 MF (JAK2 MUT=5, JAK WT=5) patients and 5 HI, western blotting was performed; and higher expression in WT and MUT compared to HI were observed. Furthermore, its expression was also higher in WT compared to MUT cases. This is in contrast to a previous report by Gautier EF et al (Blood, 2012 Feb 2;119 (5):1190-9) where CDC25A expression was less in JAK2 WT cases compared to MUT cases. IHC was performed to confirm the difference of expression level of CDC25A in JAK2 MUT and WT bone marrow samples (N=8). IHC showed that JAK2 WT samples had many positive megakaryocytes stained with CDC25A antibody (>80%) while JAK2 MUT samples had only a few positive megakaryocytes (<20%). To test the feasibility of targeting this pathway in patients with MF and to assess for differential response between JAK2 MUT and WT cases, a potent cell permeable 7-substituted quinolinedione derivedCDC25 phosphatase inhibitor (NSC663284) was tested in JAK2 MUT (N=2) vs WT (N=1). Cell proliferation was determined by Trypan Blue and MTT assay after cell exposure to different concentrations of the inhibitor [3, 5, 7, 10 and 30uM] in 24 hours observation. NSC663284 induced higher dose-dependent cell growth inhibition in JAK2 WT compared to MUT cases (% of viable cells in WT vs MUT using previously mentioned concentrations, 3 uM= 98% vs 86%, 5 uM= 93% VS 77%, 7 uM= 88% vs 65%, 10 uM= 71% vs 43% and 30 uM= 25% vs 61%; p=0.01).In conclusion, CDC25A is more highly expressed in patients with wild type JAK2 compared to the mutant counterpart and primary cells from WT JAK2 patients demonstrate higher sensitivity to CDC25A inhibition, warranting further clinical testing of this therapeutic strategy. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document