16 results on '"Gondek, Lukasz P."'
Search Results
2. Co-occurring mutations in ASXL1, SRSF2, and SETBP1 define a subset of myelodysplastic/ myeloproliferative neoplasm with neutrophilia.
- Author
-
Jain T, Ware AD, Dalton WB, Pasca S, Tsai HL, Gocke CD, Gondek LP, Xian RR, Borowitz MJ, and Levis MJ
- Subjects
- Male, Female, Humans, Leukocytosis, Hyperplasia, Mutation, Serine-Arginine Splicing Factors genetics, Repressor Proteins genetics, Carrier Proteins genetics, Nuclear Proteins genetics, Myelodysplastic-Myeloproliferative Diseases genetics, Myelodysplastic-Myeloproliferative Diseases pathology, Leukemia, Myeloid, Acute genetics
- Abstract
Identification of genomic signatures with consistent clinicopathological features in myelodysplastic/myeloproliferative neoplasm (MDS/MPN) is critical for improved diagnosis, elucidation of biology, inclusion in clinical trials, and development of therapies. We describe clinical and pathological features with co-existence of mutations in ASXL1 (missense or nonsense), SRSF2, and SKI homologous region of SETBP1, in 18 patients. Median age was 68 years with a male predominance (83%). Leukocytosis and neutrophilia were common at presentation. Marrow features included hypercellularity, granulocytic hyperplasia with megakaryocytic atypia, while the majority had myeloid hyperplasia and/or erythroid hypoplasia, myeloid dysplasia, and aberrant CD7 expression on blasts. Mutations in growth signaling pathways (RAS or JAK2) were noted at diagnosis or acquired during the disease course in 83% of patients. Two patients progressed upon acquisition of FLT3-TKD (acute myeloid leukemia) or KIT (aggressive systemic mastocytosis) mutations. The prognosis is poor with only two long-term survivors, thus far, who underwent blood or marrow transplantation. We propose that the presence of co-occurring ASXL1, SRSF2, and SETBP1 mutations can be diagnostic of a subtype of MDS/MPN with neutrophilia if clinical and morphological findings align. Our report underscores the association between genotype and phenotype within MDS/MPN and that genomic signatures should guide categorization of these entities., Competing Interests: Declaration of Competing Interest Jain: Institutional research support from CTI Biopharma, Kartos Therapeutics, Incyte; Advisory board participation with Care Dx, Bristol Myers Squibb, Incyte, Abbvie, CTI, Kite, Cogent Biosciences, Blueprint Medicine, Telios pharma, Protagonist therapeutics. Levis: Research funding from Astellas, FujiFilm; Consultancy for Abbvie, Amgen, Bristol-Myers Squibb, Daiichi-Sankyo, Jazz, Menarini, Syndax. The remaining authors report no potential relevant conflict of interest., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
3. CCRL2 affects the sensitivity of myelodysplastic syndrome and secondary acute myeloid leukemia cells to azacitidine.
- Author
-
Karantanos T, Teodorescu P, Arvanitis M, Perkins B, Jain T, DeZern AE, Dalton WB, Christodoulou I, Paun BC, Varadhan R, Esteb C, Rajkhowa T, Bonifant C, Gondek LP, Levis MJ, Yegnasubramanian S, Ghiaur G, and Jones RJ
- Subjects
- Humans, Azacitidine pharmacology, Azacitidine therapeutic use, Cell Line, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Better understanding of the biology of resistance to DNA methyltransferase (DNMT) inhibitors is required to identify therapies that can improve their efficacy for patients with high-risk myelodysplastic syndrome (MDS). CCRL2 is an atypical chemokine receptor that is upregulated in CD34+ cells from MDS patients and induces proliferation of MDS and secondary acute myeloid leukemia (sAML) cells. In this study, we evaluated any role that CCRL2 may have in the regulation of pathways associated with poor response or resistance to DNMT inhibitors. We found that CCRL2 knockdown in TF-1 cells downregulated DNA methylation and PRC2 activity pathways and increased DNMT suppression by azacitidine in MDS/sAML cell lines (MDS92, MDS-L and TF-1). Consistently, CCRL2 deletion increased the sensitivity of these cells to azacitidine in vitro and the efficacy of azacitidine in an MDS-L xenograft model. Furthermore, CCRL2 overexpression in MDS-L and TF-1 cells decreased their sensitivity to azacitidine. Finally, CCRL2 levels were higher in CD34+ cells from MDS and MDS/myeloproliferative neoplasm patients with poor response to DNMT inhibitors. In conclusion, we demonstrated that CCRL2 modulates epigenetic regulatory pathways, particularly DNMT levels, and affects the sensitivity of MDS/sAML cells to azacitidine. These results support CCRL2 targeting as having therapeutic potential in MDS/sAML.
- Published
- 2023
- Full Text
- View/download PDF
4. Venetoclax in combination with hypomethylating agent for the treatment of advanced myeloproliferative neoplasms and acute myeloid leukemia with extramedullary disease.
- Author
-
Sanber K, Ye K, Tsai HL, Newman M, Webster JA, Gojo I, Ghiaur G, Prince GT, Gondek LP, Smith BD, Levis MJ, DeZern AE, Ambinder AJ, Dalton WB, and Jain T
- Subjects
- Humans, Retrospective Studies, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Sulfonamides, Antineoplastic Combined Chemotherapy Protocols adverse effects, Azacitidine therapeutic use, Myeloproliferative Disorders diagnosis, Myeloproliferative Disorders drug therapy, Myeloproliferative Disorders genetics, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
The combination of venetoclax and hypomethylating agent (HMA/venetoclax) has emerged as a treatment option for patients with de novo acute myeloid leukemia (AML) who are unfit to receive intensive chemotherapy. In this single-center retrospective study, we evaluated clinical outcomes following treatment with HMA/venetoclax in 35 patients with advanced myeloproliferative neoplasms, myelodysplastic syndrome/myeloproliferative neoplasm overlap syndromes or AML with extramedullary disease. The composite complete remission (CR) rate (including confirmed/presumed complete cytogenetic response, acute leukemia response-complete, CR and CR with incomplete hematologic recovery) was 42.9% with median overall survival (OS) of 9.7 months. Complex karyotype was associated with inferior median OS (3.7 versus 12.2 months; p = 0.0002) and composite CR rate (22% versus 50.0%; p = 0.2444). Although SRSF2 mutations were associated with higher composite CR rate (80.0% versus 28.0%; p = 0.0082), this was not associated with longer median OS (10.9 versus 8.0 months; p = 0.2269). Future studies should include these patient subgroups.
- Published
- 2023
- Full Text
- View/download PDF
5. Allelic complexity of KMT2A partial tandem duplications in acute myeloid leukemia and myelodysplastic syndromes.
- Author
-
Tsai HK, Gibson CJ, Murdock HM, Davineni P, Harris MH, Wang ES, Gondek LP, Kim AS, Nardi V, and Lindsley RC
- Subjects
- Alleles, Disease Progression, Gene Duplication genetics, Humans, RNA, Histone-Lysine N-Methyltransferase genetics, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics, Myeloid-Lymphoid Leukemia Protein genetics
- Abstract
KMT2A partial tandem duplication (KMT2A-PTD) is an adverse risk factor in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS), a potential therapeutic target, and an attractive marker of measurable residual disease. High initial KMT2A-PTD RNA levels have been linked to poor prognosis, but mechanisms regulating KMT2A-PTD expression are not well understood. Although KMT2A-PTD has been reported to affect only a single allele, it has been theorized but not proven that genomic gains of a monoallelic KMT2A-PTD may occur, thereby potentially driving high expression and disease progression. In this study, we identified 94 patients with KMT2A-PTDs using targeted DNA next-generation sequencing (NGS) and found that 16% (15/94) had complex secondary events, including copy-neutral loss of heterozygosity and selective gain involving the KMT2A-PTD allele. High copy numbers indicating complexity were significantly enriched in AML vs MDS and correlated with higher RNA expression. Moreover, in serial samples, complexity was associated with relapse and secondary transformation. Taken together, we provide approaches to integrate quantitative and allelic assessment of KMT2A-PTDs into targeted DNA NGS and demonstrate that secondary genetic events occur in KMT2A-PTD by multiple mechanisms that may be linked to myeloid disease progression by driving increased expression from the affected allele., (© 2022 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
6. Impact of diagnostic genetics on remission MRD and transplantation outcomes in older patients with AML.
- Author
-
Murdock HM, Kim HT, Denlinger N, Vachhani P, Hambley B, Manning BS, Gier S, Cho C, Tsai HK, McCurdy S, Ho VT, Koreth J, Soiffer RJ, Ritz J, Carroll MP, Vasu S, Perales MA, Wang ES, Gondek LP, Devine S, Alyea EP, Lindsley RC, and Gibson CJ
- Subjects
- Aged, Humans, Neoplasm, Residual diagnosis, Neoplasm, Residual genetics, Recurrence, Retrospective Studies, Transplantation Conditioning, Transplantation, Homologous, Hematopoietic Stem Cell Transplantation, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy
- Abstract
Older patients with acute myeloid leukemia (AML) have high relapse risk and poor survival after allogeneic hematopoietic cell transplantation (HCT). Younger patients may receive myeloablative conditioning to mitigate relapse risk associated with high-risk genetics or measurable residual disease (MRD), but older adults typically receive reduced-intensity conditioning (RIC) to limit toxicity. To identify factors that drive HCT outcomes in older patients, we performed targeted mutational analysis (variant allele fraction ≥2%) on diagnostic samples from 295 patients with AML aged ≥60 years who underwent HCT in first complete remission, 91% of whom received RIC, and targeted duplex sequencing at remission in a subset comprising 192 patients. In a multivariable model for leukemia-free survival (LFS) including baseline genetic and clinical variables, we defined patients with low (3-year LFS, 85%), intermediate (55%), high (35%), and very high (7%) risk. Before HCT, 79.7% of patients had persistent baseline mutations, including 18.3% with only DNMT3A or TET2 (DT) mutations and 61.4% with other mutations (MRD positive). In univariable analysis, MRD positivity was associated with increased relapse and inferior LFS, compared with DT and MRD-negative mutations. However, in a multivariable model accounting for baseline risk, MRD positivity had no independent impact on LFS, most likely because of its significant association with diagnostic genetic characteristics, including MDS-associated gene mutations, TP53 mutations, and high-risk karyotype. In summary, molecular associations with MRD positivity and transplant outcomes in older patients with AML are driven primarily by baseline genetics, not by mutations present in remission. In this group of patients, where high-intensity conditioning carries substantial risk of toxicity, alternative approaches to mitigating MRD-associated relapse risk are needed., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
7. The role of the atypical chemokine receptor CCRL2 in myelodysplastic syndrome and secondary acute myeloid leukemia.
- Author
-
Karantanos T, Teodorescu P, Perkins B, Christodoulou I, Esteb C, Varadhan R, Helmenstine E, Rajkhowa T, Paun BC, Bonifant C, Dalton WB, Gondek LP, Moliterno AR, Levis MJ, Ghiaur G, and Jones RJ
- Subjects
- Cell Proliferation, Female, Hematopoiesis, Humans, Male, Signal Transduction, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes pathology
- Abstract
The identification of new pathways supporting the myelodysplastic syndrome (MDS) primitive cells growth is required to develop targeted therapies. Within myeloid malignancies, men have worse outcomes than women, suggesting male sex hormone-driven effects in malignant hematopoiesis. Androgen receptor promotes the expression of five granulocyte colony-stimulating factor receptor-regulated genes. Among them, CCRL2 encodes an atypical chemokine receptor regulating cytokine signaling in granulocytes, but its role in myeloid malignancies is unknown. Our study revealed that CCRL2 is up-regulated in primitive cells from patients with MDS and secondary acute myeloid leukemia (sAML). CCRL2 knockdown suppressed MDS92 and MDS-L cell growth and clonogenicity in vitro and in vivo and decreased JAK2/STAT3/STAT5 phosphorylation. CCRL2 coprecipitated with JAK2 and potentiated JAK2-STAT interaction. Erythroleukemia cells expressing JAK2V617F showed less effect of CCRL2 knockdown, whereas fedratinib potentiated the CCRL2 knockdown effect. Conclusively, our results implicate CCRL2 as an MDS/sAML cell growth mediator, partially through JAK2/STAT signaling.
- Published
- 2022
- Full Text
- View/download PDF
8. Gender-related differences in the outcomes and genomic landscape of patients with myelodysplastic syndrome/myeloproliferative neoplasm overlap syndromes.
- Author
-
Karantanos T, Gondek LP, Varadhan R, Moliterno AR, DeZern AE, Jones RJ, and Jain T
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Genomics, Hematologic Neoplasms mortality, Humans, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Myelodysplastic Syndromes mortality, Myeloproliferative Disorders mortality, Proto-Oncogene Mas, Retrospective Studies, Databases, Nucleic Acid, Hematologic Neoplasms genetics, Leukemia, Myeloid, Acute genetics, Mutation, Myelodysplastic Syndromes genetics, Myeloproliferative Disorders genetics, Neoplasm Proteins genetics, Sex Characteristics
- Abstract
Myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) overlap syndromes show a male predominance and men with MDS/MPN have worse outcomes, but it is unknown if the mutational burden differs between genders. We reviewed 167 patients with MDS/MPN and found that men had worse overall survival [hazard ratio (HR) 2·09, 95% confidence interval (CI) 1·16-3·75; P = 0·013] independent of subtype, Revised International Prognostic Scoring System score and age at diagnosis. We analysed the genomic data of a subset of 100 patients. Men had 0·88 more somatic mutations on average (95% CI 0·20-1·56, P = 0·011) independent of subtype, sample source and blast percentage. More somatic mutations was associated with a higher incidence of transformation to acute myeloid leukaemia (subdistribution HR 1·30, 95% CI 1·01-1·70; P = 0·046). Men had 0·70 more mutations in high-risk genes [additional sex combs like-1 (ASXL1), enhancer of zeste homolog 2 (EZH2), Runt-related transcription factor 1 (RUNX1), SET binding protein 1 (SETBP1), NRAS proto-oncogene, GTPase (NRAS), stromal antigen 2 (STAG2)] on average (95% CI 0·11-1·29, P = 0·021), and 13-times higher odds of harbouring an EZH2 mutation (95% CI 1·64-102·94, P = 0·015). The presence of an EZH2 mutation was associated with worse survival among men (HR 2·98, 95% CI 1·1-8·0; P = 0·031). Our present findings suggest that the worse outcomes in men with MDS/MPN are associated with a higher number of somatic mutations, especially in high-risk genes. These results warrant validation in larger cohorts and investigation of the underlying mechanisms., (© 2021 The Authors. British Journal of Haematology published by British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
9. Immunomodulation with pomalidomide at early lymphocyte recovery after induction chemotherapy in newly diagnosed AML and high-risk MDS.
- Author
-
Zeidner JF, Knaus HA, Zeidan AM, Blackford AL, Montiel-Esparza R, Hackl H, Prince GT, Gondek LP, Ghiaur G, Showel MM, DeZern AE, Pratz KW, Douglas Smith B, Levis MJ, Gore S, Coombs CC, Foster MC, Streicher H, Karp JE, Luznik L, and Gojo I
- Subjects
- Adult, Aged, Cytarabine administration & dosage, Disease-Free Survival, Etoposide administration & dosage, Female, Hexosamines administration & dosage, Humans, Immunomodulation drug effects, Induction Chemotherapy methods, Male, Maximum Tolerated Dose, Middle Aged, Remission Induction, Thalidomide administration & dosage, Treatment Outcome, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Immunologic Factors administration & dosage, Leukemia, Myeloid, Acute drug therapy, Myelodysplastic Syndromes drug therapy, Thalidomide analogs & derivatives
- Abstract
An immunosuppressive microenvironment promoting leukemia cell immune escape plays an important role in the pathogenesis of AML. Through its interaction with cereblon, a substrate receptor for the E3 ubiquitin ligase complex, pomalidomide leads to selective ubiquitination of transcription factors Aiolos and Ikaros thereby promoting immune modulation. In this phase I trial, 51 newly diagnosed non-favorable risk AML and high-risk MDS patients were enrolled and treated with AcDVP16 (cytarabine 667 mg/m
2 /day IV continuous infusion days 1-3, daunorubicin 45 mg/m2 IV days 1-3, etoposide 400 mg/m2 IV days 8-10) induction therapy followed by dose- and duration-escalation pomalidomide beginning at early lymphocyte recovery. Forty-three patients (AML: n = 39, MDS: n = 4) received pomalidomide. The maximum tolerated dose of pomalidomide was 4 mg for 21 consecutive days. The overall complete remission (CR + CRi) rate, median overall survival, and disease-free survival were 75%, 27.1 and 20.6 months, respectively. Subset analyses revealed 86% CR/CRi rate in AML patients with unfavorable-risk karyotype treated with pomalidomide. Pomalidomide significantly decreased Aiolos expression in both CD4+ and CD8+ peripheral blood and bone marrow T cells, promoted T cell differentiation, proliferation, and heightened their cytokine production. Finally, pomalidomide induced distinct gene expression changes in immune function-related ontologies in CD4+ and CD8+ T cells.- Published
- 2020
- Full Text
- View/download PDF
10. Hedgehog/GLI1 activation leads to leukemic transformation of myelodysplastic syndrome in vivo and GLI1 inhibition results in antitumor activity.
- Author
-
Lau BW, Huh K, Madero-Marroquin R, De Marchi F, Lim Y, Wang Q, Lobo F, Marchionni L, Smith DB, DeZern A, Levis MJ, Aplan PD, Matsui W, and Gondek LP
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic pathology, Granulocyte-Macrophage Progenitor Cells pathology, Hedgehog Proteins genetics, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes pathology, Pyridines pharmacology, Pyrimidines pharmacology, Zinc Finger Protein GLI1 genetics, Cell Transformation, Neoplastic metabolism, Granulocyte-Macrophage Progenitor Cells metabolism, Hedgehog Proteins metabolism, Leukemia, Myeloid, Acute metabolism, Myelodysplastic Syndromes metabolism, Zinc Finger Protein GLI1 metabolism
- Abstract
Myelodysplastic syndromes (MDSs) are stem cell disorders with risk of transformation to acute myeloid leukemia (AML). Gene expression profiling reveals transcriptional expression of GLI1, of Hedgehog (Hh) signaling, in poor-risk MDS/AML. Using a murine model of MDS we demonstrated that constitutive Hh/Gli1 activation accelerated leukemic transformation and decreased overall survival. Hh/Gli1 activation resulted in clonal expansion of phenotypically defined granulocyte macrophage progenitors (GMPs) and acquisition of self-renewal potential in a non-self-renewing progenitor compartment. Transcriptome analysis of GMPs revealed enrichment in gene signatures of self-renewal pathways, operating via direct Gli1 activation. Using human cell lines we demonstrated that in addition to canonical Hh signaling, GLI1 is activated in a Smoothened-independent manner. GLI1 knockdown or inhibition with GANT61 resulted in decreased proliferation and clonogenic potential. Our data suggest that GLI1 activation is frequent in MDS during disease progression and inhibition of GLI1 is an attractive therapeutic target for a subset of patients.
- Published
- 2019
- Full Text
- View/download PDF
11. Donor cell leukemia arising from clonal hematopoiesis after bone marrow transplantation.
- Author
-
Gondek LP, Zheng G, Ghiaur G, DeZern AE, Matsui W, Yegnasubramanian S, Lin MT, Levis M, Eshleman JR, Varadhan R, Tucker N, Jones R, and Gocke CD
- Subjects
- Aged, Female, Hematopoiesis physiology, Humans, Leukemia genetics, Male, Middle Aged, Tissue Donors, Bone Marrow Transplantation adverse effects, Leukemia etiology, Leukemia, Myeloid, Acute therapy, Lymphoma, B-Cell therapy, Transplantation, Homologous adverse effects
- Published
- 2016
- Full Text
- View/download PDF
12. Prognostic impact of SNP array karyotyping in myelodysplastic syndromes and related myeloid malignancies.
- Author
-
Tiu RV, Gondek LP, O'Keefe CL, Elson P, Huh J, Mohamedali A, Kulasekararaj A, Advani AS, Paquette R, List AF, Sekeres MA, McDevitt MA, Mufti GJ, and Maciejewski JP
- Subjects
- Adult, Aged, Aged, 80 and over, Chromosome Aberrations statistics & numerical data, Female, Genome-Wide Association Study, Humans, Karyotyping, Leukemia, Myeloid, Acute epidemiology, Male, Middle Aged, Myelodysplastic Syndromes epidemiology, Predictive Value of Tests, Prognosis, Risk Factors, Young Adult, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes genetics, Oligonucleotide Array Sequence Analysis, Polymorphism, Single Nucleotide
- Abstract
Single nucleotide polymorphism arrays (SNP-As) have emerged as an important tool in the identification of chromosomal defects undetected by metaphase cytogenetics (MC) in hematologic cancers, offering superior resolution of unbalanced chromosomal defects and acquired copy-neutral loss of heterozygosity. Myelodysplastic syndromes (MDSs) and related cancers share recurrent chromosomal defects and molecular lesions that predict outcomes. We hypothesized that combining SNP-A and MC could improve diagnosis/prognosis and further the molecular characterization of myeloid malignancies. We analyzed MC/SNP-A results from 430 patients (MDS = 250, MDS/myeloproliferative overlap neoplasm = 95, acute myeloid leukemia from MDS = 85). The frequency and clinical significance of genomic aberrations was compared between MC and MC plus SNP-A. Combined MC/SNP-A karyotyping lead to higher diagnostic yield of chromosomal defects (74% vs 44%, P < .0001), compared with MC alone, often through detection of novel lesions in patients with normal/noninformative (54%) and abnormal (62%) MC results. Newly detected SNP-A defects contributed to poorer prognosis for patients stratified by current morphologic and clinical risk schemes. The presence and number of new SNP-A detected lesions are independent predictors of overall and event-free survival. The significant diagnostic and prognostic contributions of SNP-A-detected defects in MDS and related diseases underscore the utility of SNP-A when combined with MC in hematologic malignancies.
- Published
- 2011
- Full Text
- View/download PDF
13. New lesions detected by single nucleotide polymorphism array-based chromosomal analysis have important clinical impact in acute myeloid leukemia.
- Author
-
Tiu RV, Gondek LP, O'Keefe CL, Huh J, Sekeres MA, Elson P, McDevitt MA, Wang XF, Levis MJ, Karp JE, Advani AS, and Maciejewski JP
- Subjects
- Chromosome Aberrations, Cytogenetic Analysis, Disease-Free Survival, Humans, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute mortality, Middle Aged, Nuclear Proteins genetics, Nucleophosmin, Prognosis, Sensitivity and Specificity, fms-Like Tyrosine Kinase 3 genetics, Karyotyping methods, Leukemia, Myeloid, Acute genetics, Polymorphism, Single Nucleotide
- Abstract
Purpose: Cytogenetics is the primary outcome predictor in acute myeloid leukemia (AML). Metaphase cytogenetics (MC) detects an abnormal karyotype in only half of patients with AML, however. Single nucleotide polymorphism arrays (SNP-A) can detect acquired somatic uniparental disomy (UPD) and other cryptic defects, even in samples deemed normal by MC. We hypothesized that SNP-A will improve detection of chromosomal defects in AML and that this would enhance the prognostic value of MC., Patients and Methods: We performed 250K and 6.0 SNP-A analyses on 140 patients with primary (p) and secondary (s) AML and correlated the results with clinical outcomes and Flt-3/nucleophosmin (NPM-1) status., Results: SNP-A is more sensitive than MC in detecting unbalanced lesions (pAML, 65% v 39%, P = .002; and sAML, 78% v 51%, P = .003). Acquired somatic UPD, not detectable by MC, was common in our AML cohort (29% in pAML and 35% in sAML). Patients with SNP-A lesions including acquired somatic UPD exhibited worse overall survival (OS) and event-free survival (EFS) in pAML with normal MC and in pAML/sAML with abnormal MC. SNP-A improved the predictive value of Flt-3 internal tandem duplication/NPM-1 status, with inferior survival seen in patients with additional SNP-A defects. Multivariate analyses confirmed the independent predictive value of SNP-A defects for OS (hazard ratio [HR] = 2.52; 95% CI, 1.29 to 5.22; P = .006) and EFS (HR = 1.72; 95% CI, 1.12 to 3.48; P = .04)., Conclusion: SNP-A analysis allows enhanced detection of chromosomal abnormalities and provides important prognostic impact in AML.
- Published
- 2009
- Full Text
- View/download PDF
14. Progressive chromatin repression and promoter methylation of CTNNA1 associated with advanced myeloid malignancies.
- Author
-
Ye Y, McDevitt MA, Guo M, Zhang W, Galm O, Gore SD, Karp JE, Maciejewski JP, Kowalski J, Tsai HL, Gondek LP, Tsai HC, Wang X, Hooker C, Smith BD, Carraway HE, and Herman JG
- Subjects
- Blotting, Western, Cells, Cultured, Chromatin Immunoprecipitation, DNA Methylation, Fluorescent Antibody Technique, Humans, Leukemia, Myeloid, Acute pathology, Luciferases metabolism, Lymphocytes metabolism, Myelodysplastic Syndromes pathology, RNA, Messenger genetics, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Chromatin physiology, Chromosomes, Human, Pair 5 genetics, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics, Promoter Regions, Genetic genetics, alpha Catenin genetics
- Abstract
Complete loss or deletion of the long arm of chromosome 5 is frequent in myelodysplastic syndrome (MDS) and acute myelogenous leukemia (AML). The putative gene(s) deleted and responsible for the pathogenesis of these poor prognosis hematologic disorders remain controversial. This study is a comprehensive analysis of previously implicated and novel genes for epigenetic inactivation in AML and MDS. In 146 AML cases, methylation of CTNNA1 was frequent, and more common in AML patients with 5q deletion (31%) than those without 5q deletion (14%), whereas no methylation of other 5q genes was observed. In 31 MDS cases, CTNNA1 methylation was only found in high-risk MDS (>or=RAEB2), but not in low-risk MDS (
- Published
- 2009
- Full Text
- View/download PDF
15. Aberrant DNA methylation is a dominant mechanism in MDS progression to AML.
- Author
-
Jiang Y, Dunbar A, Gondek LP, Mohan S, Rataul M, O'Keefe C, Sekeres M, Saunthararajah Y, and Maciejewski JP
- Subjects
- Adolescent, Adult, Aged, Anemia, Refractory genetics, Anemia, Refractory pathology, Anemia, Refractory, with Excess of Blasts genetics, Anemia, Refractory, with Excess of Blasts pathology, Bone Marrow metabolism, Bone Marrow pathology, Case-Control Studies, Chromosome Aberrations, CpG Islands, Disease Progression, Female, Humans, Karyotyping, Leukemia, Myeloid, Acute pathology, Male, Microarray Analysis, Middle Aged, Myelodysplastic Syndromes pathology, Polymorphism, Single Nucleotide genetics, Prognosis, Promoter Regions, Genetic genetics, RNA, Messenger genetics, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, Tumor Cells, Cultured, Young Adult, DNA Methylation, Frizzled Receptors genetics, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics, Receptors, G-Protein-Coupled genetics, Tumor Suppressor Proteins genetics
- Abstract
Myelodysplastic syndromes (MDSs) are clonal hematologic disorders that frequently represent an intermediate disease stage before progression to acute myeloid leukemia (AML). As such, study of MDS/AML can provide insight into the mechanisms of neoplastic evolution. In 184 patients with MDS and AML, DNA methylation microarray and high-density single nucleotide polymorphism array (SNP-A) karyotyping were used to assess the relative contributions of aberrant DNA methylation and chromosomal deletions to tumor-suppressor gene (TSG) silencing during disease progression. Aberrant methylation was seen in every sample, on average affecting 91 of 1505 CpG loci in early MDS and 179 of 1505 loci after blast transformation (refractory anemia with excess blasts [RAEB]/AML). In contrast, chromosome aberrations were seen in 79% of early MDS samples and 90% of RAEB/AML samples, and were not as widely distributed over the genome. Analysis of the most frequently aberrantly methylated genes identified FZD9 as a candidate TSG on chromosome 7. In patients with chromosome deletion at the FZD9 locus, aberrant methylation of the remaining allele was associated with the poorest clinical outcome. These results indicate that aberrant methylation can cooperate with chromosome deletions to silence TSG. However, the ubiquity, extent, and correlation with disease progression suggest that aberrant DNA methylation is the dominant mechanism for TSG silencing and clonal variation in MDS evolution to AML.
- Published
- 2009
- Full Text
- View/download PDF
16. Chromosomal lesions and uniparental disomy detected by SNP arrays in MDS, MDS/MPD, and MDS-derived AML.
- Author
-
Gondek LP, Tiu R, O'Keefe CL, Sekeres MA, Theil KS, and Maciejewski JP
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Cytogenetics, Humans, Karyotyping, Leukemia, Myeloid, Acute etiology, Leukemia, Myeloid, Acute pathology, Middle Aged, Myelodysplastic Syndromes complications, Myelodysplastic Syndromes pathology, Myeloproliferative Disorders pathology, Oligonucleotide Array Sequence Analysis, Prognosis, Chromosomes, Human genetics, Leukemia, Myeloid, Acute genetics, Myelodysplastic Syndromes genetics, Myeloproliferative Disorders genetics, Polymorphism, Single Nucleotide genetics, Uniparental Disomy genetics
- Abstract
Using metaphase cytogenetics (MC), chromosomal abnormalities are found in only a proportion of patients with myelodysplastic syndrome (MDS). We hypothesized that with new precise methods more cryptic karyotypic lesions can be uncovered that may show important clinical implications. We have applied 250K single nucleotide polymorphisms (SNP) arrays (SNP-A) to study chromosomal lesions in samples from 174 patients (94 MDS, 33 secondary acute myeloid leukemia [sAML], and 47 myelodysplastic/myeloproliferative disease [MDS/MPD]) and 76 controls. Using SNP-A, aberrations were found in around three-fourths of MDS, MDS/MPD, and sAML (vs 59%, 37%, 53% by MC; in 8% of patients MC was unsuccessful). Previously unrecognized lesions were detected in patients with normal MC and in those with known lesions. Moreover, segmental uniparental disomy (UPD) was found in 20% of MDS, 23% of sAML, and 35% of MDS/MPD patients, a lesion resulting in copy-neutral loss of heterozygosity undetectable by MC. The potential clinical significance of abnormalities detected by SNP-A, but not seen on MC, was demonstrated by their impact on overall survival. UPD involving chromosomes frequently affected by deletions may have prognostic implications similar to the deletions visible by MC. SNP-A-based karyotyping shows superior resolution for chromosomal defects, including UPD. This technique further complements MC to improve clinical prognosis and targeted therapies.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.