51
|
Schlette EJ, Schlette EJ, Admirand J, Wierda W, Abruzzo L, Lin KI, O'Brien S, Lerner S, Keating MJ, Tam C. p53 expression by immunohistochemistry is an important determinant of survival in patients with chronic lymphocytic leukemia receiving frontline chemo-immunotherapy. Leuk Lymphoma 2009; 50:1597-605. [DOI: 10.1080/10428190903165241] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
52
|
Abstract
B-chronic lymphocytic leukemia (B-CLL) is characterized by a highly variable clinical course which has long remained a stumbling block for clinicians. This variability appears to arise from complex molecular alterations identified in malignant cells from patient subsets. Recent studies have focused in particular on identifying new molecular markers to help predict the most effective and adapted treatments. In addition to the mutation status of immunoglobulin variable heavy-chain region (IgVH) genes, which is a well-established predictive factor in B-CLL, these new markers include defects of cell factors involved in the maintenance of genome stability, such as telomere function, DNA repair, ATM and p53. Other predictive factors, such as tyrosine kinase Zap-70 and soluble factors found in patient sera, may be associated with B-cell receptor signal transduction. Interestingly, an alteration of these factors fits closely, though not strikingly, with the absence of somatic mutations in IgVH genes, suggesting that the latter may be due either to epigenetic events leading to an unstable genome or to an inherited defect in the immune response of malignant B-cells. Recent lessons from Zap-70 expression/phosphorylation suggest that some of these markers may reflect the defective pathways in B-CLL cells rather than being markers of cell malignancy per se. Furthermore, specific subsets of markers are found in patient cells resistant to treatment. Current studies on gene expression profiling and proteomic analyses should soon lead to a better understanding of how these pathways are affected, especially in multi-drug resistant B-CLL.
Collapse
Affiliation(s)
- Julien Bouley
- Laboratoire de Radiobiologie et Oncologie, CEA, DSV-DRR, Fontenay aux Roses, France
| | | | | | | |
Collapse
|
53
|
Zenz T, Mertens D, Döhner H, Stilgenbauer S. Molecular diagnostics in chronic lymphocytic leukemia – Pathogenetic and clinical implications. Leuk Lymphoma 2009; 49:864-73. [DOI: 10.1080/10428190701882955] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
54
|
Affiliation(s)
- Carla Kurkjian
- Advanced Developmental Therapeutics Training Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| | | | | |
Collapse
|
55
|
Van Bockstaele F, Verhasselt B, Philippé J. Prognostic markers in chronic lymphocytic leukemia: A comprehensive review. Blood Rev 2009; 23:25-47. [DOI: 10.1016/j.blre.2008.05.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
56
|
Butler T, Gribben JG. Biologic prognostic markers and their application in clinical trials and management of chronic lymphocytic leukaemia patients. ACTA ACUST UNITED AC 2008; 2:101-12. [DOI: 10.1517/17530059.2.1.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
57
|
Jacinto FV, Ballestar E, Ropero S, Esteller M. Discovery of epigenetically silenced genes by methylated DNA immunoprecipitation in colon cancer cells. Cancer Res 2007; 67:11481-6. [PMID: 18089774 DOI: 10.1158/0008-5472.can-07-2687] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CpG island promoter hypermethylation of tumor suppressor genes is a common hallmark of human cancer, and new large-scale epigenomic technologies might be useful in our attempts to define the complete DNA hypermethylome of tumor cells. Here, we report a functional search for hypermethylated CpG islands using the colorectal cancer cell line HCT-116, in which two major DNA methyltransferases, DNMT1 and DNMT3b, have been genetically disrupted (DKO cells). Using methylated DNA immunoprecipitation methodology in conjunction with promoter microarray analyses, we found that DKO cells experience a significant loss of hypermethylated CpG islands. Further characterization of these candidate sequences shows CpG island promoter hypermethylation and silencing of genes with potentially important roles in tumorigenesis, such as the Ras guanine nucleotide-releasing factor (RASGRF2), the apoptosis-associated basic helix-loop transcription factor (BHLHB9), and the homeobox gene (HOXD1). Hypermethylation of these genes occurs in premalignant lesions and accumulates during tumorigenesis. Thus, our results show the usefulness of DNMT genetic disruption strategies combined with methylated DNA immunoprecipitation in searching for unknown hypermethylated candidate genes in human cancer that might aid our understanding of the biology of the disease and be of potential translational use.
Collapse
Affiliation(s)
- Filipe V Jacinto
- Cancer Epigenetics Laboratory, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | | | | | | |
Collapse
|
58
|
Abstract
Genetic and epigenetic mechanisms contribute to the development of human tumors. However, the conventional analysis of neoplasias has preferentially focused on only one of these processes. This approach has led to a biased, primarily genetic view, of human tumorigenesis. Epigenetic alterations, such as aberrant DNA methylation, are sufficient to induce tumor formation, and can modify the incidence, and determine the type of tumor which will arise in genetic models of cancer. These observations raise important questions about the degree to which genetic and epigenetic mechanisms cooperate in human tumorigenesis, the identity of the specific cooperating genes and how these genes interact functionally to determine the diverse biological and clinical paths to tumor initiation and progression. These gaps in our knowledge are, in part, due to the lack of methods for full-scale integrated genetic and epigenetic analyses. The ultimate goal to fill these gaps would include sequencing relevant regions of the 3-billion nucleotide genome, and determining the methylation status of the 28-million CpG dinucleotide methylome at single nucleotide resolution in different types of neoplasias. Here, we review the emergence and advancement of technologies to map ever larger proportions of the cancer methylome, and the unique discovery potential of integrating these with cancer genomic data. We discuss the knowledge gained from these large-scale analyses in the context of gene discovery, therapeutic application and building a more widely applicable mechanism-based model of human tumorigenesis.
Collapse
Affiliation(s)
- Romulo M Brena
- Department of Molecular Genetics, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | | |
Collapse
|
59
|
Plass C, Byrd JC, Raval A, Tanner SM, de la Chapelle A. Molecular profiling of chronic lymphocytic leukaemia: genetics meets epigenetics to identify predisposing genes. Br J Haematol 2007; 139:744-52. [PMID: 17961188 DOI: 10.1111/j.1365-2141.2007.06875.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Molecular profiling may lead to a better understanding of a disease. This knowledge is especially important in malignancies, where multiple alterations are required during the progression from premalignant to malignant stages. Such information can be useful for the development of novel biomarkers that allow the prediction of a clinical course, response to treatment or early detection. Molecular data is also utilized to develop targeted therapies. Moreover, gene defects identified in profiling studies will help to understand the molecular pathways disrupted in the disease. This review provides an overview of molecular profiling approaches in chronic lymphocytic leukaemia (CLL). We will describe our current understanding of genetic alterations in CLL, the use of familial CLL for the identification of predisposing mutations, and the search for epigenetic alterations in CLL.
Collapse
Affiliation(s)
- Christoph Plass
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, the Comprehensive Cancer Center at the Ohio State University, Columbus, OH, USA.
| | | | | | | | | |
Collapse
|
60
|
Moskalyov EA, Eprintsev AT, Hoheisel JD. DNA methylation profiling in cancer: From single nucleotides towards the methylome. Mol Biol 2007. [DOI: 10.1134/s0026893307050068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
61
|
Blum W, Klisovic RB, Hackanson B, Liu Z, Liu S, Devine H, Vukosavljevic T, Huynh L, Lozanski G, Kefauver C, Plass C, Devine SM, Heerema NA, Murgo A, Chan KK, Grever MR, Byrd JC, Marcucci G. Phase I Study of Decitabine Alone or in Combination With Valproic Acid in Acute Myeloid Leukemia. J Clin Oncol 2007; 25:3884-91. [PMID: 17679729 DOI: 10.1200/jco.2006.09.4169] [Citation(s) in RCA: 244] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Purpose To determine an optimal biologic dose (OBD) of decitabine as a single agent and then the maximum-tolerated dose (MTD) of valproic acid (VA) combined with decitabine in acute myeloid leukemia (AML). Patients and Methods Twenty-five patients (median age, 70 years) were enrolled; 12 were untreated and 13 had relapsed AML. To determine an OBD (based on a gene re-expression end point), 14 patients received decitabine alone for 10 days. To determine the MTD, 11 patients received decitabine (at OBD, days 1 through 10) plus dose-escalating VA (days 5 through 21). Results The OBD of decitabine was 20 mg/m2/d intravenously, with limited nonhematologic toxicity. In patients treated with decitabine plus VA, dose-limiting encephalopathy occurred in two of two patients at VA 25 mg/kg/d and one of six patients at VA 20 mg/kg/d. Drug-induced re-expression of estrogen receptor (ER) was associated with clinical response (P ≤ .05). ER promoter demethylation, global DNA hypomethylation, depletion of DNA methyltransferase enzyme, and histone hyperacetylation were also observed. In an intent-to-treat analysis, the response rate was 44% (11 of 25). Of 21 assessable patients, 11 (52%) responded: four with morphologic and cytogenetic complete remission (CR; each had complex karyotype), four with incomplete CR, and three with partial remission. In untreated AML, four of nine assessable patients achieved CR. Clinical responses appeared similar for decitabine alone or with VA. Conclusion Low-dose decitabine was safe and showed encouraging clinical and biologic activity in AML, but the addition of VA led to encephalopathy at relatively low doses. On the basis of these results, additional studies of decitabine (20 mg/m2/d for 10 days) alone or with an alternative deacetylating agent are warranted.
Collapse
Affiliation(s)
- William Blum
- Department of Medicine, Division of Hematology and Oncology, The Ohio State University, Columbus, OH 43210, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Motiwala T, Majumder S, Kutay H, Smith DS, Neuberg DS, Lucas DM, Byrd JC, Grever M, Jacob ST. Methylation and silencing of protein tyrosine phosphatase receptor type O in chronic lymphocytic leukemia. Clin Cancer Res 2007; 13:3174-81. [PMID: 17545520 PMCID: PMC3074612 DOI: 10.1158/1078-0432.ccr-06-1720] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
PURPOSE Previous studies in our laboratory have shown the progressive methylation and suppression of the gene encoding protein tyrosine phosphatase, PTPRO, in the livers of rats fed a methyl-deficient diet that induces hepatocarcinogenesis. Subsequently, we observed the methylation of PTPRO in primary human lung tumors and also showed its potential tumor suppressor characteristics. The present study was undertaken to investigate whether the truncated form of PTPRO (PTPROt), specifically expressed in naïve B lymphocytes, was also methylated and suppressed in chronic lymphocytic leukemia (CLL), a disease generally affecting B lymphocytes. EXPERIMENTAL DESIGN AND RESULTS Initial screening showed that 60% of the 52 CLL samples analyzed using methylation-specific PCR assay were methylated compared with B lymphocytes from normal individuals, which were not methylated. The expression of PTPROt, as measured by semiquantitative reverse transcription-PCR, inversely correlated with methylation in the few samples tested. Analysis of additional samples (n = 50) by combined bisulfite restriction analysis showed that the PTPRO CpG island was methylated in 82% of patients with CLL compared with B lymphocytes from normal individuals. Furthermore, overall expression of PTPRO was reduced in CLL relative to normal lymphocytes. The PTPRO gene was also suppressed by methylation in the CLL cell line WaC3CD5, where it could be reactivated upon treatment with the DNA hypomethylating agent 5-AzaC. Ectopic expression of PTPROt in a nonexpressing cell line increased growth inhibition with fludarabine treatment, a therapy commonly used for CLL. CONCLUSION This study reveals the potential role of PTPRO methylation and silencing in CLL tumorigenesis and also provides a novel molecular target in the epigenetic therapy.
Collapse
MESH Headings
- B-Lymphocytes/metabolism
- DNA Methylation
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Gene Silencing
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Membrane Proteins/metabolism
- Membrane Proteins/physiology
- Models, Biological
- Polymerase Chain Reaction
- Protein Tyrosine Phosphatases/metabolism
- Protein Tyrosine Phosphatases/physiology
- Receptor-Like Protein Tyrosine Phosphatases, Class 3
- Reverse Transcriptase Polymerase Chain Reaction
- Sulfites/pharmacology
- Vidarabine/analogs & derivatives
- Vidarabine/pharmacology
Collapse
Affiliation(s)
- Tasneem Motiwala
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, Ohio
| | - Sarmila Majumder
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, Ohio
| | - Huban Kutay
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, Ohio
| | - David Spencer Smith
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, Ohio
| | - Donna S. Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David M. Lucas
- Division of Hematology-Oncology, Department of Internal Medicine, Ohio State University, Columbus, Ohio
| | - John C. Byrd
- Division of Hematology-Oncology, Department of Internal Medicine, Ohio State University, Columbus, Ohio
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio
| | - Michael Grever
- Division of Hematology-Oncology, Department of Internal Medicine, Ohio State University, Columbus, Ohio
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio
| | - Samson T. Jacob
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, Ohio
- Division of Hematology-Oncology, Department of Internal Medicine, Ohio State University, Columbus, Ohio
- Comprehensive Cancer Center, Ohio State University, Columbus, Ohio
| |
Collapse
|
63
|
Raval A, Tanner SM, Byrd JC, Angerman EB, Perko JD, Chen SS, Hackanson B, Grever MR, Lucas DM, Matkovic JJ, Lin TS, Kipps TJ, Murray F, Weisenburger D, Sanger W, Lynch J, Watson P, Jansen M, Yoshinaga Y, Rosenquist R, de Jong PJ, Coggill P, Beck S, Lynch H, de la Chapelle A, Plass C. Downregulation of death-associated protein kinase 1 (DAPK1) in chronic lymphocytic leukemia. Cell 2007; 129:879-90. [PMID: 17540169 PMCID: PMC4647864 DOI: 10.1016/j.cell.2007.03.043] [Citation(s) in RCA: 279] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2006] [Revised: 01/15/2007] [Accepted: 03/12/2007] [Indexed: 11/30/2022]
Abstract
The heritability of B cell chronic lymphocytic leukemia (CLL) is relatively high; however, no predisposing mutation has been convincingly identified. We show that loss or reduced expression of death-associated protein kinase 1 (DAPK1) underlies cases of heritable predisposition to CLL and the majority of sporadic CLL. Epigenetic silencing of DAPK1 by promoter methylation occurs in almost all sporadic CLL cases. Furthermore, we defined a disease haplotype, which segregates with the CLL phenotype in a large family. DAPK1 expression of the CLL allele is downregulated by 75% in germline cells due to increased HOXB7 binding. In the blood cells from affected family members, promoter methylation results in additional loss of DAPK1 expression. Thus, reduced expression of DAPK1 can result from germline predisposition, as well as epigenetic or somatic events causing or contributing to the CLL phenotype.
Collapse
Affiliation(s)
- Aparna Raval
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, The Comprehensive Cancer Center at The Ohio State University, Columbus, OH 43214, USA
| | - Stephan M. Tanner
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, The Comprehensive Cancer Center at The Ohio State University, Columbus, OH 43214, USA
| | - John C. Byrd
- Department of Internal Medicine, Division of Hematology and Oncology, The Ohio State University, Columbus, OH 43214, USA
| | - Elizabeth B. Angerman
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, The Comprehensive Cancer Center at The Ohio State University, Columbus, OH 43214, USA
| | - James D. Perko
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, The Comprehensive Cancer Center at The Ohio State University, Columbus, OH 43214, USA
| | - Shih-Shih Chen
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, The Comprehensive Cancer Center at The Ohio State University, Columbus, OH 43214, USA
| | - Björn Hackanson
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, The Comprehensive Cancer Center at The Ohio State University, Columbus, OH 43214, USA
- Department of Hematology/Oncology, University of Freiburg Medical Center, Freiburg, Germany
| | - Michael R. Grever
- Department of Internal Medicine, Division of Hematology and Oncology, The Ohio State University, Columbus, OH 43214, USA
| | - David M. Lucas
- Department of Internal Medicine, Division of Hematology and Oncology, The Ohio State University, Columbus, OH 43214, USA
| | - Jennifer J. Matkovic
- Department of Internal Medicine, Division of Hematology and Oncology, The Ohio State University, Columbus, OH 43214, USA
| | - Thomas S. Lin
- Department of Internal Medicine, Division of Hematology and Oncology, The Ohio State University, Columbus, OH 43214, USA
| | - Thomas J. Kipps
- Department of Internal Medicine, University of California at San Diego, San Diego, CA, 92093, USA
| | - Fiona Murray
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Dennis Weisenburger
- Department of Preventive Medicine and Public Health, Creighton University, Omaha, NB 68178, USA
| | - Warren Sanger
- Department of Preventive Medicine and Public Health, Creighton University, Omaha, NB 68178, USA
| | - Jane Lynch
- Department of Preventive Medicine and Public Health, Creighton University, Omaha, NB 68178, USA
| | - Patrice Watson
- Department of Preventive Medicine and Public Health, Creighton University, Omaha, NB 68178, USA
| | - Mary Jansen
- Department of Preventive Medicine and Public Health, Creighton University, Omaha, NB 68178, USA
| | - Yuko Yoshinaga
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Richard Rosenquist
- Department of Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Pieter J. de Jong
- Children's Hospital Oakland Research Institute, Oakland, CA 94609, USA
| | - Penny Coggill
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, UK
| | - Stephan Beck
- Wellcome Trust Sanger Institute, Genome Campus, Hinxton, Cambridge, UK
| | - Henry Lynch
- Department of Preventive Medicine and Public Health, Creighton University, Omaha, NB 68178, USA
| | - Albert de la Chapelle
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, The Comprehensive Cancer Center at The Ohio State University, Columbus, OH 43214, USA
- Corresponding author
| | - Christoph Plass
- Department of Molecular Virology, Immunology, and Medical Genetics, Human Cancer Genetics Program, The Comprehensive Cancer Center at The Ohio State University, Columbus, OH 43214, USA
- Corresponding author
| |
Collapse
|
64
|
Kainz B, Shehata M, Bilban M, Kienle D, Heintel D, Krömer-Holzinger E, Le T, Kröber A, Heller G, Schwarzinger I, Demirtas D, Chott A, Döhner H, Zöchbauer-Müller S, Fonatsch C, Zielinski C, Stilgenbauer S, Gaiger A, Wagner O, Jäger U. Overexpression of the paternally expressed gene10 (PEG10) from the imprinted locus on chromosome 7q21 in high-risk B-cell chronic lymphocytic leukemia. Int J Cancer 2007; 121:1984-1993. [PMID: 17621626 DOI: 10.1002/ijc.22929] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We report high expression of the maternally imprinted gene PEG10 in high-risk B-CLL defined by high LPL mRNA expression. Differential expression was initially identified by microarray analysis and confirmed by real time PCR in 42 B-CLL patients. mRNA expression ranged from 0.3- to 375.4-fold compared to normal peripheral blood mononuclear cells (PBMNC). Expression levels in CD19+ B-CLL cells were 100-fold higher than in B-cells from healthy donors. PEG10 expression levels in B-CLL patient samples remained stable over time even after chemotherapy. High PEG10 expression correlated with high LPL expression (p=0.001) and a positive Coombs' test (p=0.04). Interestingly, similar expression patterns were observed for the neighbouring imprinted gene sarcoglycan-epsilon (SGCE). Monoallelic expression and maintained imprinting of PEG10 were found by allele- or methylation-specific PCR. The intensity of intracellular staining of PEG10 protein corresponded to mRNA levels as confirmed by immunofluorescence staining. Short term knock-down of PEG10 in B-CLL cells and HepG2 cells was not associated with changes in cell survival but resulted in a significant change in the expression of 80 genes. However, long term inhibition of PEG10 led to induction of apoptosis in B-CLL cells. Our data indicate (i) a prognostic value of PEG10 in B-CLL patients; (ii) specific deregulation of the imprinted locus at 7q21 in high-risk B-CLL; (iii) a potential functional and biological role of PEG10 protein expression. Altogether, PEG10 represents a novel marker in B-CLL.
Collapse
MESH Headings
- Alleles
- Apoptosis Regulatory Proteins
- Biomarkers, Tumor
- Cell Line, Tumor
- Chromosomes, Human, Pair 7/genetics
- DNA Methylation
- DNA-Binding Proteins
- Down-Regulation
- Gene Expression Regulation, Neoplastic
- Genomic Imprinting/genetics
- Health
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Nuclear Proteins/genetics
- Polysaccharides/metabolism
- Proteins/genetics
- Proteins/metabolism
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- RNA-Binding Proteins
- Risk Factors
- Survival Rate
- Ubiquitin-Protein Ligases/genetics
Collapse
Affiliation(s)
- Birgit Kainz
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Medhat Shehata
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
- K. Landsteiner Institute for Cytokine and Tumor Microenvironment, Vienna, Austria
| | - Martin Bilban
- Department of Clinical Chemistry and Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Dirk Kienle
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Daniel Heintel
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | | | - Trang Le
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Alexander Kröber
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Gerwin Heller
- Division of Oncology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Ilse Schwarzinger
- Department of Clinical Chemistry and Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Dita Demirtas
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
- K. Landsteiner Institute for Cytokine and Tumor Microenvironment, Vienna, Austria
| | - Andreas Chott
- Department of Clinical Pathology, Medical University of Vienna, Vienna, Austria
| | - Hartmut Döhner
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
| | - Sabine Zöchbauer-Müller
- Division of Oncology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Christa Fonatsch
- Department of Human Genetics, Medical University of Vienna, Vienna, Austria
| | - Christoph Zielinski
- Division of Oncology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
- Center of Excellence in Clinical and Experimental Oncology (CLEXO), Vienna, Austria
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, University of Ulm, Ulm, Germany
- German CLL Study Group
| | - Alexander Gaiger
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Oswald Wagner
- Department of Clinical Chemistry and Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Center of Excellence in Clinical and Experimental Oncology (CLEXO), Vienna, Austria
| | - Ulrich Jäger
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
- Center of Excellence in Clinical and Experimental Oncology (CLEXO), Vienna, Austria
- German CLL Study Group
| |
Collapse
|
65
|
Abstract
The DNA methylation level in patients with chronic lymphocytic leukemia is generally lower than healthy individuals. Although DNA methylation is globally decreased, regional hypermethylation of gene promoters leads to gene silencing. Many of these genes have tumor suppressor phenotypes. Unlike mutations or deletions, hypermethylation is potentially reversible after inhibition with DNA methylation modulators. Myelodysplastic syndrome has been a model disease in which treatment of patients results in demethylation of specific genes. The story in patients with chronic lymphocytic leukemia is slowly unraveling as epigenetic modifications likely also play an important role. Ongoing clinical trials correlating clinical response to gene expression after treatment with DNA methylation inhibitors will ultimately allow us to better risk stratify and predict the subgroup of patients who will benefit from treatment with this class of drugs.
Collapse
Affiliation(s)
- Margaret K Yu
- Division of Hematology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
66
|
Gowda A, Byrd JC. Use of prognostic factors in risk stratification at diagnosis and time of treatment of patients with chronic lymphocytic leukemia. Curr Opin Hematol 2006; 13:266-72. [PMID: 16755224 DOI: 10.1097/01.moh.0000231425.46148.b0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW To review risk stratification strategies used in chronic lymphocytic leukemia at diagnosis to predict aggressiveness of disease and, at time of treatment, to predict duration of response. RECENT FINDINGS Several new prognostic factors can better assist clinicians in predicting the aggressiveness of chronic lymphocytic leukemia at diagnosis and the likelihood of maintaining a prolonged remission with treatment. This article reviews older prognostic factors such as beta2-microglobulin and thymidine kinase activity that have been partially validated by recently completed large studies. New prognostic factors such as interphase cytogenetics, immunoglobulin heavy-chain gene mutational analysis, and relevant secondary surrogate markers of immunoglobulin heavy-chain gene, including methylation of the zeta-associated protein gene, lipoprotein lipase overexpression, telomere length, and telomerase activity are reviewed. Some prognostic factors (interphase cytogenetics) but not others (immunoglobulin heavy-chain gene mutational status, zeta-associated protein expression) predict the duration of response to fludarabine-based combination strategies. SUMMARY Recent advances in risk stratification provide clinicians with tools to better predict outcome of chronic lymphocytic leukemia at the time of treatment and response to treatment at the time of developing symptomatic disease.
Collapse
MESH Headings
- Biomarkers, Tumor/analysis
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Prognosis
- Risk Factors
- Treatment Outcome
Collapse
Affiliation(s)
- Aruna Gowda
- Division of Hematology and Oncology, The Ohio State University, Columbus, Ohio 43210, USA
| | | |
Collapse
|
67
|
Meyer S, White DJ, Will AM, Eden T, Sim A, Brown R, Strathdee G. No evidence of significant silencing of Fanconi genes FANCF and FANCB or Nijmegen breakage syndrome gene NBS1 by DNA hyper-methylation in sporadic childhood leukaemia. Br J Haematol 2006; 134:61-3. [PMID: 16803569 DOI: 10.1111/j.1365-2141.2006.06107.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Fanconi anaemia (FA) and Nijmegen breakage syndrome (NBS) carry a high risk of haematological cancer. Affected cellular pathways may be modulated in sporadic malignancies and silencing of FANCF through methylation has been shown to cause somatic disruption of the FA pathway. Combined bisulphite restriction analysis for methylation of FANCF, FANCB and NBS1 was used to investigate 81 sporadic acute childhood leukaemias. No methylation was detected at any associated CpG sites analysed. This does not exclude very low levels of FANCF, FANCB or NBS1 methylation, but suggests other factors are responsible for chemo-sensitivity and chromosomal instability in sporadic childhood leukaemia.
Collapse
Affiliation(s)
- Stefan Meyer
- Department of Paediatric Haematology and Oncology, Central Manchester and Manchester Children's University Hospitals NHS Trust, Manchester, UK.
| | | | | | | | | | | | | |
Collapse
|
68
|
Buhl AM, Jurlander J, Geisler CH, Pedersen LB, Andersen MK, Josefsson P, Petersen JH, Leffers H. CLLU1 expression levels predict time to initiation of therapy and overall survival in chronic lymphocytic leukemia. Eur J Haematol 2006; 76:455-64. [PMID: 16529606 DOI: 10.1111/j.0902-4441.2005.t01-1-ejh2530.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Chronic lymphocytic leukemia (CLL) is an incurable disease with a highly variable clinical course. IgV(H) mutational status, chromosomal aberrations, CD38 expression and ZAP-70 expression are prognostic markers in CLL, however, they are not exclusively confined to this disease. We recently identified a novel CLL-specific gene (CLL upregulated gene1, CLLU1) that is exclusively upregulated in CLL cells. Here we describe our evaluation of the prognostic significance of CLLU1 in CLL. METHODS A cohort of 59 previously untreated CLL patients was studied. We determined the expression levels of two CLLU1 transcripts, cDNA1 and CDS, by quantitative RT-PCR. The relation between CLLU1 expression and time to therapy, overall survival and presence or absence of ZAP-70, CD38, chromosomal aberrations or IgV(H) mutations in the 59 patients was analyzed. RESULTS Analyzed as a continuous, quantitative parameter CLLU1 levels significantly predicted time from diagnosis to initiation of therapy (P < or = 0.0003) Analyzed as a categorical parameter, by segregation of the patients into groups with cDNA1 or CDS expression above or below the median, the CLLU1 levels significantly predicted time from diagnosis to initiation of therapy (P = 0.001) and predicted overall survival with borderline significance (P < or = 0.05). Patient stratification according to clinical stage, cytogenetics, IgV(H) mutational status, ZAP-70 and CD38, demonstrated significantly increased CLLU1 expression in all investigated CLL poor risk groups. CLLU1 expression levels contributed additional prognostic information to ZAP-70-positive patients. CONCLUSIONS CLLU1 is the first identified CLL specific gene. The CLLU1 mRNA expression level can predict time to initiation of treatment and survival in CLL patients.
Collapse
MESH Headings
- ADP-ribosyl Cyclase 1/biosynthesis
- ADP-ribosyl Cyclase 1/genetics
- Aged
- Aged, 80 and over
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/biosynthesis
- Biomarkers, Tumor/genetics
- Chromosome Aberrations
- Chromosomes, Human, Pair 12/genetics
- Cohort Studies
- DNA, Complementary/analysis
- Denmark/epidemiology
- Female
- Gene Expression Regulation, Neoplastic
- Genes, Immunoglobulin
- Humans
- Immunoglobulin Heavy Chains/biosynthesis
- Immunoglobulin Heavy Chains/genetics
- Immunoglobulin Variable Region/biosynthesis
- Immunoglobulin Variable Region/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Life Tables
- Male
- Middle Aged
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Organ Specificity
- Polymerase Chain Reaction
- Prognosis
- Proportional Hazards Models
- RNA, Long Noncoding
- RNA, Messenger/biosynthesis
- RNA, Neoplasm/biosynthesis
- Survival Analysis
- Time Factors
- Up-Regulation
- ZAP-70 Protein-Tyrosine Kinase/biosynthesis
- ZAP-70 Protein-Tyrosine Kinase/genetics
Collapse
Affiliation(s)
- Anne Mette Buhl
- Department of Hematology, The Leukemia Laboratory, Rigshospitalet, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
69
|
Abstract
A major obstacle to the expansion of abnormal cells with significant proliferative potential is the induction of programmed cell death. Consequently, oncogene-driven hyperproliferation must be associated with apoptosis inhibition to allow malignant outgrowth. The oncogenic cooperation of N-Myc and Twist-1 in the development of neuroblastoma, the most common and deadly solid tumour of childhood, perfectly illustrates such a process. N-Myc promotes cell proliferation, whereas Twist-1 counteracts its pro-apoptotic properties by knocking-down the ARF/p53 pathway. On the basis of numerous recent studies reporting its overexpression in a variety of human cancers, we discuss in this review the role of Twist-1 as a potent inhibitor of the cell safety programs engaged in response to an abnormal mitogenic activity.
Collapse
Affiliation(s)
- A Puisieux
- INSERM U590 Centre Léon Bérard, Université Claude Bernard Lyon 1, 69373 Lyon Cedex 08, France.
| | | | | |
Collapse
|
70
|
Brena RM, Auer H, Kornacker K, Hackanson B, Raval A, Byrd JC, Plass C. Accurate quantification of DNA methylation using combined bisulfite restriction analysis coupled with the Agilent 2100 Bioanalyzer platform. Nucleic Acids Res 2006; 34:e17. [PMID: 16464820 PMCID: PMC1361623 DOI: 10.1093/nar/gnj017] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
DNA methylation is the best-studied epigenetic modification and describes the conversion of cytosine to 5-methylcytosine. The importance of this phenomenon is that aberrant promoter hypermethylation is a common occurrence in cancer and is frequently associated with gene silencing. Various techniques are currently available for the analysis of DNA methylation. However, accurate and reproducible quantification of DNA methylation remains challenging. In this report, we describe Bio-COBRA (combined bisulfite restriction analysis coupled with the Agilent 2100 Bioanalyzer platform), as a novel approach to quantitative DNA methylation analysis. The combination of a well-established method, COBRA, which interrogates DNA methylation via the restriction enzyme analysis of PCR-amplified bisulfite treated DNAs, with the Bioanalyzer platform allows for the rapid and quantitative assessment of DNA methylation patterns in large sample sets. The sensitivity and reproducibility of Bio-COBRA make it a valuable tool for the analysis of DNA methylation in clinical samples, which could aid in the development of diagnostic and prognostic parameters with respect to disease detection and management.
Collapse
Affiliation(s)
- Romulo Martin Brena
- Department of Molecular Genetics, The Ohio State UniversityColumbus, OH, USA
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State UniversityColumbus, OH, USA
| | - Herbert Auer
- Columbus Children's Research InstituteColumbus, OH, USA
| | - Karl Kornacker
- Division of Sensory Biophysics, The Ohio State UniversityColumbus, OH, USA
| | - Björn Hackanson
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State UniversityColumbus, OH, USA
- Department of Hematology, University of Freiburg Medical CenterFreiburg, Germany
| | - Aparna Raval
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State UniversityColumbus, OH, USA
| | - John C. Byrd
- Department of Medicine and the Comprehensive Cancer Center, Divisions of Hematology-Oncology, The Ohio State UniversityColumbus, OH, USA
| | - Christoph Plass
- Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State UniversityColumbus, OH, USA
- To whom correspondence should be addressed at Division of Human Cancer Genetics, Medical Research Facility Room 464A, 420 West 12th Avenue, Columbus, OH 43210, USA. Tel: +1 614 292 6505; Fax: +1 614 688 4761;
| |
Collapse
|
71
|
Brena RM, Huang THM, Plass C. Quantitative assessment of DNA methylation: potential applications for disease diagnosis, classification, and prognosis in clinical settings. J Mol Med (Berl) 2006; 84:365-77. [PMID: 16416310 DOI: 10.1007/s00109-005-0034-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2005] [Accepted: 11/29/2005] [Indexed: 12/31/2022]
Abstract
Deregulation of the epigenome is now recognized as a major mechanism involved in the development and progression of human diseases such as cancer. As opposed to the irreversible nature of genetic events, which introduce changes in the primary DNA sequence, epigenetic modifications are reversible and leave the original DNA sequence intact. There is now evidence that the epigenetic landscape in humans undergoes modifications as the result of normal aging, with older individuals exhibiting higher levels of promoter hypermethylation compared to younger ones. Thus, it has been proposed that the higher incidence of certain disease in older individuals might be, in part, a consequence of an inherent change in the control and regulation of the epigenome. These observations are of remarkable clinical significance since the aberrant epigenetic changes characteristic of disease provide a unique platform for the development of new therapeutic approaches. In this review, we address the significance of DNA methylation changes that result or lead to disease, occur with aging, or may be the result of environmental exposure. We provide a detailed description of quantitative techniques currently available for the detection and analysis of DNA methylation and provide a comprehensive framework that may allow for the incorporation of protocols which include DNA methylation as a tool for disease diagnosis and classification, which could lead to the tailoring of therapeutic approaches designed to individual patient needs.
Collapse
Affiliation(s)
- Romulo Martin Brena
- Division of Human Cancer Genetics, Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | | | | |
Collapse
|
72
|
Abstract
5-Aza-2'-deoxycitidine (decitabine, Dacogen, Bloomington, MN) is a cytosine analogue that promotes hypomethylation of DNA and has documented efficacy in myeloid malignancies. Indeed, promising clinical results have been observed in acute myeloid leukemia (AML) and the myelodysplastic syndromes (MDS). Aberrant methylation has also been found in chronic leukemias, providing a rationale for investigating the use of decitabine in these diseases. There is clear evidence of molecular (hypomethylation) as well as hematologic and cytogenetic responses to decitabine in chronic myelogenous leukemia of all phases, including in patients resistant to imatinib mesylate. Clinical trials of decitabine in chronic lymphocytic leukemia are ongoing. There are many unanswered questions regarding optimizing this treatment for chronic leukemias, but successful proof-of-concept studies for hypomethylating agents move us closer to approaches that may have a significant impact on patient outcomes.
Collapse
Affiliation(s)
- Jean-Pierre J Issa
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | |
Collapse
|
73
|
Sausville EA, Carducci MA. Making Bad Cells Go Good: The Promise of Epigenetic Therapy. J Clin Oncol 2005; 23:3875-6. [PMID: 15946932 DOI: 10.1200/jco.2005.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|