Skip to main content
Top
Published in: Tumor Biology 10/2016

01-10-2016 | Review

Biomarkers of genome instability and cancer epigenetics

Authors: Adriana H. O. Reis, Fernando R. Vargas, Bernardo Lemos

Published in: Tumor Biology | Issue 10/2016

Login to get access

Abstract

Tumorigenesis is a multistep process involving genetic and epigenetic alterations that drive somatic evolution from normal human cells to malignant derivatives. Collectively, genetic and epigenetic alterations might be combined into biomarkers for the assessment of risk, the detection of early stage tumors, and accurate tumor characterization before and after treatment. Recent efforts have provided systematic approaches to cancer genomics through the application of massive sequencing of specific tumor types. Here, we review biomarkers of genome instability and epigenetics. Cancer evolvability and adaptation emerge through genetic and epigenetic lesions of a variety of sizes and qualities—from point mutations and small insertions/deletions to large-scale chromosomal rearrangements, alterations in whole chromosome copy number, preferential allelic expression of cancer risk alleles, and mechanisms that increase tumor mutation rates. We also review specific epigenetic mechanisms that facilitate or hinder tumor adaptation, including DNA methylation, histone modification, nucleosome remodeling, transcription factor activity, and small non-coding RNAs. Given the complexity of the carcinogenic process, the challenge ahead will be to interpret disparate signals across hundreds of genes and summarize these signals into a single actionable diagnosis that translates into specific treatments. Another challenge is to refine preventive efforts through the identification of epigenetic processes that mediate increased cancer rates in individuals exposed to sources of toxic environmental stress and pollution, specially through development and early childhood.
Literature
2.
go back to reference Chappell G et al. Genetic and epigenetic changes in fibrosis-associated hepatocarcinogenesis in mice. Int J Cancer. 2014;134(12):2778–88. Chappell G et al. Genetic and epigenetic changes in fibrosis-associated hepatocarcinogenesis in mice. Int J Cancer. 2014;134(12):2778–88.
4.
5.
go back to reference Almendro V, Marusyk A, Polyak K. Cellular heterogeneity and molecular evolution in cancer. Annu Rev Pathol. 2013;8:277–302.PubMedCrossRef Almendro V, Marusyk A, Polyak K. Cellular heterogeneity and molecular evolution in cancer. Annu Rev Pathol. 2013;8:277–302.PubMedCrossRef
6.
go back to reference Almendro V et al. Inference of tumor evolution during chemotherapy by computational modeling and in situ analysis of genetic and phenotypic cellular diversity. Cell Rep. 2014;6(3):514–27.PubMedPubMedCentralCrossRef Almendro V et al. Inference of tumor evolution during chemotherapy by computational modeling and in situ analysis of genetic and phenotypic cellular diversity. Cell Rep. 2014;6(3):514–27.PubMedPubMedCentralCrossRef
7.
go back to reference Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338–45.PubMedCrossRef Burrell RA, McGranahan N, Bartek J, Swanton C. The causes and consequences of genetic heterogeneity in cancer evolution. Nature. 2013;501(7467):338–45.PubMedCrossRef
9.
go back to reference Strand M, Prolla TA, Liskay RM, Petes TD. Destabilization of tracts of simple repetitive DNA in yeast by mutations affecting DNA mismatch repair. Nature. 1993;365(6443):274–6.PubMedCrossRef Strand M, Prolla TA, Liskay RM, Petes TD. Destabilization of tracts of simple repetitive DNA in yeast by mutations affecting DNA mismatch repair. Nature. 1993;365(6443):274–6.PubMedCrossRef
10.
go back to reference Parsons R et al. Hypermutability and mismatch repair deficiency in RER+ tumor cells. Cell. 1993;75(6):1227–36.PubMedCrossRef Parsons R et al. Hypermutability and mismatch repair deficiency in RER+ tumor cells. Cell. 1993;75(6):1227–36.PubMedCrossRef
11.
12.
go back to reference Aplan PD. Causes of oncogenic chromosomal translocation. Trends in Genetics: TIG. 2006;22(1):46–55.PubMedCrossRef Aplan PD. Causes of oncogenic chromosomal translocation. Trends in Genetics: TIG. 2006;22(1):46–55.PubMedCrossRef
13.
go back to reference Nowell PC, Hungerford DA. Chromosome studies on normal and leukemic human leukocytes. J Natl Cancer Inst. 1960;25:85–109.PubMed Nowell PC, Hungerford DA. Chromosome studies on normal and leukemic human leukocytes. J Natl Cancer Inst. 1960;25:85–109.PubMed
14.
go back to reference Hermans A et al. Unique fusion of bcr and c-abl genes in Philadelphia chromosome positive acute lymphoblastic leukemia. Cell. 1987;51(1):33–40.PubMedCrossRef Hermans A et al. Unique fusion of bcr and c-abl genes in Philadelphia chromosome positive acute lymphoblastic leukemia. Cell. 1987;51(1):33–40.PubMedCrossRef
15.
go back to reference Manolov G, Manolova Y. Marker band in one chromosome 14 from Burkitt lymphomas. Nature. 1972;237(5349):33–4.PubMedCrossRef Manolov G, Manolova Y. Marker band in one chromosome 14 from Burkitt lymphomas. Nature. 1972;237(5349):33–4.PubMedCrossRef
16.
go back to reference Zech L, Haglund U, Nilsson K, Klein G. Characteristic chromosomal abnormalities in biopsies and lymphoid-cell lines from patients with Burkitt and non-Burkitt lymphomas. Int J Cancer. 1976;17(1):47–56.PubMedCrossRef Zech L, Haglund U, Nilsson K, Klein G. Characteristic chromosomal abnormalities in biopsies and lymphoid-cell lines from patients with Burkitt and non-Burkitt lymphomas. Int J Cancer. 1976;17(1):47–56.PubMedCrossRef
17.
go back to reference Unniraman S, Zhou S, Schatz DG. Identification of an AID-independent pathway for chromosomal translocations between the Igh switch region and Myc. Nat Immunol. 2004;5(11):1117–23.PubMedCrossRef Unniraman S, Zhou S, Schatz DG. Identification of an AID-independent pathway for chromosomal translocations between the Igh switch region and Myc. Nat Immunol. 2004;5(11):1117–23.PubMedCrossRef
19.
go back to reference McGranahan N, Burrell RA, Endesfelder D, Novelli MR, Swanton C. Cancer chromosomal instability: therapeutic and diagnostic challenges. EMBO Rep. 2012;13(6):528–38.PubMedPubMedCentralCrossRef McGranahan N, Burrell RA, Endesfelder D, Novelli MR, Swanton C. Cancer chromosomal instability: therapeutic and diagnostic challenges. EMBO Rep. 2012;13(6):528–38.PubMedPubMedCentralCrossRef
20.
go back to reference Bettington M et al. The serrated pathway to colorectal carcinoma: current concepts and challenges. Histopathology. 2013;62(3):367–86.PubMedCrossRef Bettington M et al. The serrated pathway to colorectal carcinoma: current concepts and challenges. Histopathology. 2013;62(3):367–86.PubMedCrossRef
22.
go back to reference Issa JP. Colon Cancer: it’s CIN or CIMP. Clinical Cancer Research: an official journal of the American Association for Cancer Research. 2008;14(19):5939–40.CrossRef Issa JP. Colon Cancer: it’s CIN or CIMP. Clinical Cancer Research: an official journal of the American Association for Cancer Research. 2008;14(19):5939–40.CrossRef
23.
go back to reference Karageorgos I et al. Identification of cancer predisposition variants in apparently healthy individuals using a next-generation sequencing-based family genomics approach. Human Genomics. 2015;9:12.PubMedPubMedCentralCrossRef Karageorgos I et al. Identification of cancer predisposition variants in apparently healthy individuals using a next-generation sequencing-based family genomics approach. Human Genomics. 2015;9:12.PubMedPubMedCentralCrossRef
24.
go back to reference Loo LW et al. Cis-expression QTL analysis of established colorectal cancer risk variants in colon tumors and adjacent normal tissue. PloS one. 2012;7(2):e30477.PubMedPubMedCentralCrossRef Loo LW et al. Cis-expression QTL analysis of established colorectal cancer risk variants in colon tumors and adjacent normal tissue. PloS one. 2012;7(2):e30477.PubMedPubMedCentralCrossRef
25.
go back to reference Sille FC, Thomas R, Smith MT, Conde L, Skibola CF. Post-GWAS functional characterization of susceptibility variants for chronic lymphocytic leukemia. PLoS One. 2012;7(1):e29632.PubMedPubMedCentralCrossRef Sille FC, Thomas R, Smith MT, Conde L, Skibola CF. Post-GWAS functional characterization of susceptibility variants for chronic lymphocytic leukemia. PLoS One. 2012;7(1):e29632.PubMedPubMedCentralCrossRef
27.
go back to reference Kim HS, Minna JD, White MA. GWAS meets TCGA to illuminate mechanisms of cancer predisposition. Cell. 2013;152(3):387–9.PubMedCrossRef Kim HS, Minna JD, White MA. GWAS meets TCGA to illuminate mechanisms of cancer predisposition. Cell. 2013;152(3):387–9.PubMedCrossRef
28.
go back to reference Hindorff LA, Gillanders EM, Manolio TA. Genetic architecture of cancer and other complex diseases: lessons learned and future directions. Carcinogenesis. 2011;32(7):945–54.PubMedPubMedCentralCrossRef Hindorff LA, Gillanders EM, Manolio TA. Genetic architecture of cancer and other complex diseases: lessons learned and future directions. Carcinogenesis. 2011;32(7):945–54.PubMedPubMedCentralCrossRef
29.
go back to reference Gray PN, Dunlop CL, Elliott AM. Not all next generation sequencing diagnostics are created equal: understanding the nuances of solid tumor assay design for somatic mutation detection. Cancers. 2015;7(3):1313–32.PubMedPubMedCentralCrossRef Gray PN, Dunlop CL, Elliott AM. Not all next generation sequencing diagnostics are created equal: understanding the nuances of solid tumor assay design for somatic mutation detection. Cancers. 2015;7(3):1313–32.PubMedPubMedCentralCrossRef
31.
32.
go back to reference Verma M et al. Epigenetic research in cancer epidemiology: trends, opportunities, and challenges. Cancer Epidemiol Biomark Prev. 2013;23(2):223–33.CrossRef Verma M et al. Epigenetic research in cancer epidemiology: trends, opportunities, and challenges. Cancer Epidemiol Biomark Prev. 2013;23(2):223–33.CrossRef
33.
go back to reference Kalari S, Pfeifer GP. Identification of driver and passenger DNA methylation in cancer by epigenomic analysis. Adv Genet. 2010;70:277–308.PubMedPubMedCentral Kalari S, Pfeifer GP. Identification of driver and passenger DNA methylation in cancer by epigenomic analysis. Adv Genet. 2010;70:277–308.PubMedPubMedCentral
34.
go back to reference Uemura M et al. Overexpression of ribosomal RNA in prostate cancer is common but not linked to rDNA promoter hypomethylation. Oncogene. 2011;31(10):1254–63.PubMedPubMedCentralCrossRef Uemura M et al. Overexpression of ribosomal RNA in prostate cancer is common but not linked to rDNA promoter hypomethylation. Oncogene. 2011;31(10):1254–63.PubMedPubMedCentralCrossRef
36.
go back to reference Belinsky SA et al. Aberrant methylation of p16(INK4a) is an early event in lung cancer and a potential biomarker for early diagnosis. Proc Natl Acad Sci U S A. 1998;95(20):11891–6.PubMedPubMedCentralCrossRef Belinsky SA et al. Aberrant methylation of p16(INK4a) is an early event in lung cancer and a potential biomarker for early diagnosis. Proc Natl Acad Sci U S A. 1998;95(20):11891–6.PubMedPubMedCentralCrossRef
37.
go back to reference Wong IH et al. Detection of aberrant p16 methylation in the plasma and serum of liver cancer patients. Cancer Res. 1999;59(1):71–3.PubMed Wong IH et al. Detection of aberrant p16 methylation in the plasma and serum of liver cancer patients. Cancer Res. 1999;59(1):71–3.PubMed
38.
go back to reference Zou HZ et al. Detection of aberrant p16 methylation in the serum of colorectal cancer patients. Clinical Cancer Research: an official journal of the American Association for Cancer Research. 2002;8(1):188–91. Zou HZ et al. Detection of aberrant p16 methylation in the serum of colorectal cancer patients. Clinical Cancer Research: an official journal of the American Association for Cancer Research. 2002;8(1):188–91.
39.
go back to reference Esteller M et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000a;343(19):1350–4.PubMedCrossRef Esteller M et al. Inactivation of the DNA-repair gene MGMT and the clinical response of gliomas to alkylating agents. N Engl J Med. 2000a;343(19):1350–4.PubMedCrossRef
40.
go back to reference Shen L et al. MGMT promoter methylation and field defect in sporadic colorectal cancer. J Natl Cancer Inst. 2005;97(18):1330–8.PubMedCrossRef Shen L et al. MGMT promoter methylation and field defect in sporadic colorectal cancer. J Natl Cancer Inst. 2005;97(18):1330–8.PubMedCrossRef
41.
go back to reference Harden SV, Guo Z, Epstein JI, Sidransky D. Quantitative GSTP1 methylation clearly distinguishes benign prostatic tissue and limited prostate adenocarcinoma. J Urol. 2003;169(3):1138–42.PubMedCrossRef Harden SV, Guo Z, Epstein JI, Sidransky D. Quantitative GSTP1 methylation clearly distinguishes benign prostatic tissue and limited prostate adenocarcinoma. J Urol. 2003;169(3):1138–42.PubMedCrossRef
42.
go back to reference Nakamichi I et al. Correlation between promoter hypermethylation of GSTP1 and response to chemotherapy in diffuse large B cell lymphoma. Ann Hematol. 2007;86(8):557–64.PubMedCrossRef Nakamichi I et al. Correlation between promoter hypermethylation of GSTP1 and response to chemotherapy in diffuse large B cell lymphoma. Ann Hematol. 2007;86(8):557–64.PubMedCrossRef
43.
go back to reference Hashad DI, Hashad MM, Talaat IM, Ibrahim MA. Role of glutathione-S-transferase P1 hypermethylation in molecular detection of prostate cancer. Genet Test Mol Biomarkers. 2011;15(10):667–70.PubMedCrossRef Hashad DI, Hashad MM, Talaat IM, Ibrahim MA. Role of glutathione-S-transferase P1 hypermethylation in molecular detection of prostate cancer. Genet Test Mol Biomarkers. 2011;15(10):667–70.PubMedCrossRef
44.
go back to reference Saxena A et al. GSTP1 methylation and polymorphism increase the risk of breast cancer and the effects of diet and lifestyle in breast cancer patients. Exp Ther Med. 2012;4(6):1097–103.PubMedPubMedCentral Saxena A et al. GSTP1 methylation and polymorphism increase the risk of breast cancer and the effects of diet and lifestyle in breast cancer patients. Exp Ther Med. 2012;4(6):1097–103.PubMedPubMedCentral
45.
go back to reference Esteller M, Corn PG, Baylin SB, Herman JG. A gene hypermethylation profile of human cancer. Cancer Res. 2001;61(8):3225–9.PubMed Esteller M, Corn PG, Baylin SB, Herman JG. A gene hypermethylation profile of human cancer. Cancer Res. 2001;61(8):3225–9.PubMed
46.
go back to reference Menigatti M et al. Methylation pattern of different regions of the MLH1 promoter and silencing of gene expression in hereditary and sporadic colorectal cancer. Genes Chromosomes Cancer. 2001;31(4):357–61.PubMedCrossRef Menigatti M et al. Methylation pattern of different regions of the MLH1 promoter and silencing of gene expression in hereditary and sporadic colorectal cancer. Genes Chromosomes Cancer. 2001;31(4):357–61.PubMedCrossRef
47.
go back to reference Bischoff J et al. hMLH1 promoter hypermethylation and MSI status in human endometrial carcinomas with and without metastases. Clin Exp Metastasis. 2012;29(8):889–900.PubMedCrossRef Bischoff J et al. hMLH1 promoter hypermethylation and MSI status in human endometrial carcinomas with and without metastases. Clin Exp Metastasis. 2012;29(8):889–900.PubMedCrossRef
48.
go back to reference Ozdemir F, Altinisik J, Karateke A, Coksuer H, Buyru N. Methylation of tumor suppressor genes in ovarian cancer. Exp Ther Med. 2012;4(6):1092–6.PubMedPubMedCentral Ozdemir F, Altinisik J, Karateke A, Coksuer H, Buyru N. Methylation of tumor suppressor genes in ovarian cancer. Exp Ther Med. 2012;4(6):1092–6.PubMedPubMedCentral
49.
go back to reference Dobrovic A, Simpfendorfer D. Methylation of the BRCA1 gene in sporadic breast cancer. Cancer Res. 1997;57(16):3347–50.PubMed Dobrovic A, Simpfendorfer D. Methylation of the BRCA1 gene in sporadic breast cancer. Cancer Res. 1997;57(16):3347–50.PubMed
50.
go back to reference Esteller M et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst. 2000a;92(7):564–9.PubMedCrossRef Esteller M et al. Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors. J Natl Cancer Inst. 2000a;92(7):564–9.PubMedCrossRef
51.
go back to reference Bennett KL et al. Frequently methylated tumor suppressor genes in head and neck squamous cell carcinoma. Cancer Res. 2008;68(12):4494–9.PubMedCrossRef Bennett KL et al. Frequently methylated tumor suppressor genes in head and neck squamous cell carcinoma. Cancer Res. 2008;68(12):4494–9.PubMedCrossRef
52.
53.
go back to reference Lofton-Day C et al. DNA methylation biomarkers for blood-based colorectal cancer screening. Clin Chem. 2008;54(2):414–23.PubMedCrossRef Lofton-Day C et al. DNA methylation biomarkers for blood-based colorectal cancer screening. Clin Chem. 2008;54(2):414–23.PubMedCrossRef
54.
go back to reference Connolly D et al. Septin 9 isoform expression, localization, and epigenetic changes during human and mouse breast cancer progression. Breast Cancer Res. 2011;13(4):R76.PubMedPubMedCentralCrossRef Connolly D et al. Septin 9 isoform expression, localization, and epigenetic changes during human and mouse breast cancer progression. Breast Cancer Res. 2011;13(4):R76.PubMedPubMedCentralCrossRef
55.
go back to reference Ogino S et al. LINE-1 hypomethylation is inversely associated with microsatellite instability and CpG island methylator phenotype in colorectal cancer. Int J Cancer. 2008;122(12):2767–73.PubMedPubMedCentralCrossRef Ogino S et al. LINE-1 hypomethylation is inversely associated with microsatellite instability and CpG island methylator phenotype in colorectal cancer. Int J Cancer. 2008;122(12):2767–73.PubMedPubMedCentralCrossRef
56.
go back to reference Igarashi S et al. A novel correlation between LINE-1 hypomethylation and the malignancy of gastrointestinal stromal tumors. Clinical Cancer Research: an official journal of the American Association for Cancer Research. 2010;16(21):5114–23.CrossRef Igarashi S et al. A novel correlation between LINE-1 hypomethylation and the malignancy of gastrointestinal stromal tumors. Clinical Cancer Research: an official journal of the American Association for Cancer Research. 2010;16(21):5114–23.CrossRef
57.
go back to reference Martinez JG et al. Hypomethylation of LINE-1, and not centromeric SAT-alpha, is associated with centromeric instability in head and neck squamous cell carcinoma. Cell Oncol (Dordr). 2012;35(4):259–67.CrossRef Martinez JG et al. Hypomethylation of LINE-1, and not centromeric SAT-alpha, is associated with centromeric instability in head and neck squamous cell carcinoma. Cell Oncol (Dordr). 2012;35(4):259–67.CrossRef
58.
go back to reference Nishida N et al. Unique association between global DNA hypomethylation and chromosomal alterations in human hepatocellular carcinoma. PLoS One. 2013;8(9):e72312.PubMedPubMedCentralCrossRef Nishida N et al. Unique association between global DNA hypomethylation and chromosomal alterations in human hepatocellular carcinoma. PLoS One. 2013;8(9):e72312.PubMedPubMedCentralCrossRef
59.
go back to reference Raval A et al. Reduced rRNA expression and increased rDNA promoter methylation in CD34+ cells of patients with myelodysplastic syndromes. Blood. 2012;120(24):4812–8.PubMedCrossRef Raval A et al. Reduced rRNA expression and increased rDNA promoter methylation in CD34+ cells of patients with myelodysplastic syndromes. Blood. 2012;120(24):4812–8.PubMedCrossRef
63.
go back to reference Jaju RJ et al. A novel gene, NSD1, is fused to NUP98 in the t(5;11)(q35;p15.5) in de novo childhood acute myeloid leukemia. Blood. 2001;98(4):1264–7.PubMedCrossRef Jaju RJ et al. A novel gene, NSD1, is fused to NUP98 in the t(5;11)(q35;p15.5) in de novo childhood acute myeloid leukemia. Blood. 2001;98(4):1264–7.PubMedCrossRef
64.
go back to reference Rosati R et al. NUP98 is fused to the NSD3 gene in acute myeloid leukemia associated with t(8;11)(p11.2;p15). Blood. 2002;99(10):3857–60.PubMedCrossRef Rosati R et al. NUP98 is fused to the NSD3 gene in acute myeloid leukemia associated with t(8;11)(p11.2;p15). Blood. 2002;99(10):3857–60.PubMedCrossRef
66.
68.
go back to reference Hammerman PS et al. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489(7417):519–25.CrossRef Hammerman PS et al. Comprehensive genomic characterization of squamous cell lung cancers. Nature. 2012;489(7417):519–25.CrossRef
69.
70.
go back to reference Zang ZJ et al. Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat Genet. 2012;44(5):570–4.PubMedCrossRef Zang ZJ et al. Exome sequencing of gastric adenocarcinoma identifies recurrent somatic mutations in cell adhesion and chromatin remodeling genes. Nat Genet. 2012;44(5):570–4.PubMedCrossRef
73.
74.
go back to reference Choi JH et al. Expression profile of histone deacetylase 1 in gastric cancer tissues. Jpn J Cancer Res. 2001;92(12):1300–4.PubMedCrossRef Choi JH et al. Expression profile of histone deacetylase 1 in gastric cancer tissues. Jpn J Cancer Res. 2001;92(12):1300–4.PubMedCrossRef
75.
go back to reference Halkidou K et al. Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer. Prostate. 2004;59(2):177–89.PubMedCrossRef Halkidou K et al. Upregulation and nuclear recruitment of HDAC1 in hormone refractory prostate cancer. Prostate. 2004;59(2):177–89.PubMedCrossRef
76.
go back to reference Zhang Z et al. Quantitation of HDAC1 mRNA expression in invasive carcinoma of the breast*. Breast Cancer Res Treat. 2005;94(1):11–6.PubMedCrossRef Zhang Z et al. Quantitation of HDAC1 mRNA expression in invasive carcinoma of the breast*. Breast Cancer Res Treat. 2005;94(1):11–6.PubMedCrossRef
77.
go back to reference Burdelski C et al. HDAC1 overexpression independently predicts biochemical recurrence and is associated with rapid tumor cell proliferation and genomic instability in prostate cancer. Exp Mol Pathol. 2015;98(3):419–26.PubMedCrossRef Burdelski C et al. HDAC1 overexpression independently predicts biochemical recurrence and is associated with rapid tumor cell proliferation and genomic instability in prostate cancer. Exp Mol Pathol. 2015;98(3):419–26.PubMedCrossRef
78.
go back to reference Zhu P et al. Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis. Cancer Cell. 2004;5(5):455–63.PubMedCrossRef Zhu P et al. Induction of HDAC2 expression upon loss of APC in colorectal tumorigenesis. Cancer Cell. 2004;5(5):455–63.PubMedCrossRef
79.
go back to reference Song J et al. Increased expression of histone deacetylase 2 is found in human gastric cancer. APMIS. 2005;113(4):264–8.PubMedCrossRef Song J et al. Increased expression of histone deacetylase 2 is found in human gastric cancer. APMIS. 2005;113(4):264–8.PubMedCrossRef
80.
go back to reference Huang R et al. The role of HDAC2 in chromatin remodelling and response to chemotherapy in ovarian cancer. Oncotarget. 2016;7(4):4695–711.PubMed Huang R et al. The role of HDAC2 in chromatin remodelling and response to chemotherapy in ovarian cancer. Oncotarget. 2016;7(4):4695–711.PubMed
81.
go back to reference Wilson AJ et al. Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer. J Biol Chem. 2006;281(19):13548–58.PubMedCrossRef Wilson AJ et al. Histone deacetylase 3 (HDAC3) and other class I HDACs regulate colon cell maturation and p21 expression and are deregulated in human colon cancer. J Biol Chem. 2006;281(19):13548–58.PubMedCrossRef
82.
go back to reference Jiao F et al. Aberrant expression of nuclear HDAC3 and cytoplasmic CDH1 predict a poor prognosis for patients with pancreatic cancer. Oncotarget. 2016:16505–16. Jiao F et al. Aberrant expression of nuclear HDAC3 and cytoplasmic CDH1 predict a poor prognosis for patients with pancreatic cancer. Oncotarget. 2016:16505–16.
83.
go back to reference Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 2006;5(9):769–84.PubMedCrossRef Bolden JE, Peart MJ, Johnstone RW. Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov. 2006;5(9):769–84.PubMedCrossRef
84.
go back to reference Nakagawa M et al. Expression profile of class I histone deacetylases in human cancer tissues. Oncol Rep. 2007;18(4):769–74.PubMed Nakagawa M et al. Expression profile of class I histone deacetylases in human cancer tissues. Oncol Rep. 2007;18(4):769–74.PubMed
85.
go back to reference Oehme I et al. Histone deacetylase 8 in neuroblastoma tumorigenesis. Clinical Cancer Research: an official journal of the American Association for Cancer Research. 2009;15(1):91–9.CrossRef Oehme I et al. Histone deacetylase 8 in neuroblastoma tumorigenesis. Clinical Cancer Research: an official journal of the American Association for Cancer Research. 2009;15(1):91–9.CrossRef
86.
go back to reference Li L et al. Deacetylation of tumor-suppressor MST1 in Hippo pathway induces its degradation through HBXIP-elevated HDAC6 in promotion of breast cancer growth. . Oncogene. 2015. doi:10.1038/onc.2015.476. Li L et al. Deacetylation of tumor-suppressor MST1 in Hippo pathway induces its degradation through HBXIP-elevated HDAC6 in promotion of breast cancer growth. . Oncogene. 2015. doi:10.​1038/​onc.​2015.​476.
87.
go back to reference Hsieh CL, et al. Alterations in histone deacetylase 8 lead to cell migration and poor prognosis in breast cancer. Life Sciences. 2016. Hsieh CL, et al. Alterations in histone deacetylase 8 lead to cell migration and poor prognosis in breast cancer. Life Sciences. 2016.
88.
go back to reference Marquard L et al. Histone deacetylase 1, 2, 6 and acetylated histone H4 in B- and T-cell lymphomas. Histopathology. 2009;54(6):688–98.PubMedCrossRef Marquard L et al. Histone deacetylase 1, 2, 6 and acetylated histone H4 in B- and T-cell lymphomas. Histopathology. 2009;54(6):688–98.PubMedCrossRef
89.
go back to reference Adams H, Fritzsche FR, Dirnhofer S, Kristiansen G, Tzankov A. Class I histone deacetylases 1, 2 and 3 are highly expressed in classical Hodgkin’s lymphoma. Expert Opin Ther Targets. 2010;14(6):577–84.PubMedCrossRef Adams H, Fritzsche FR, Dirnhofer S, Kristiansen G, Tzankov A. Class I histone deacetylases 1, 2 and 3 are highly expressed in classical Hodgkin’s lymphoma. Expert Opin Ther Targets. 2010;14(6):577–84.PubMedCrossRef
90.
go back to reference Hrabeta J, Stiborova M, Adam V, Kizek R, & Eckschlager, T. Histone deacetylase inhibitors in cancer therapy. A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2013. Hrabeta J, Stiborova M, Adam V, Kizek R, & Eckschlager, T. Histone deacetylase inhibitors in cancer therapy. A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 2013.
91.
go back to reference Zhang J et al. microRNA-22 Downregulated in hepatocellular carcinoma and correlated with prognosis, suppresses cell proliferation and tumourigenicity. Br J Cancer. 2010;103(8):1215–20.PubMedPubMedCentralCrossRef Zhang J et al. microRNA-22 Downregulated in hepatocellular carcinoma and correlated with prognosis, suppresses cell proliferation and tumourigenicity. Br J Cancer. 2010;103(8):1215–20.PubMedPubMedCentralCrossRef
92.
97.
go back to reference Fujimoto A et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 2012;44(7):760–4.PubMedCrossRef Fujimoto A et al. Whole-genome sequencing of liver cancers identifies etiological influences on mutation patterns and recurrent mutations in chromatin regulators. Nat Genet. 2012;44(7):760–4.PubMedCrossRef
99.
go back to reference Huang J et al. Exome sequencing of hepatitis B virus-associated hepatocellular carcinoma. Nat Genet. 2012;44(10):1117–21.PubMedCrossRef Huang J et al. Exome sequencing of hepatitis B virus-associated hepatocellular carcinoma. Nat Genet. 2012;44(10):1117–21.PubMedCrossRef
103.
107.
108.
go back to reference Kunej T et al. Epigenetic regulation of microRNAs in cancer: an integrated review of literature. Mutat Res. 2011;717(1–2):77–84.PubMedCrossRef Kunej T et al. Epigenetic regulation of microRNAs in cancer: an integrated review of literature. Mutat Res. 2011;717(1–2):77–84.PubMedCrossRef
109.
go back to reference Dohner H et al. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med. 2000;343(26):1910–6.PubMedCrossRef Dohner H et al. Genomic aberrations and survival in chronic lymphocytic leukemia. N Engl J Med. 2000;343(26):1910–6.PubMedCrossRef
110.
go back to reference Calin GA et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S AProc Natl Acad Sci U S A. 2002;99(24):15524–9.CrossRef Calin GA et al. Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proc Natl Acad Sci U S AProc Natl Acad Sci U S A. 2002;99(24):15524–9.CrossRef
111.
112.
go back to reference Reis AH, Vargas FR, Lemos B. More epigenetic hits than meets the eye: microRNAs and genes associated with the tumorigenesis of retinoblastoma. Front Genet. 2012;3:284.PubMedPubMedCentral Reis AH, Vargas FR, Lemos B. More epigenetic hits than meets the eye: microRNAs and genes associated with the tumorigenesis of retinoblastoma. Front Genet. 2012;3:284.PubMedPubMedCentral
114.
115.
go back to reference Tian X, Xu G. Clinical value of lncRNA MALAT1 as a prognostic marker in human cancer: systematic review and meta-analysis. BMJ open. 2015;5(9):e008653.PubMedPubMedCentralCrossRef Tian X, Xu G. Clinical value of lncRNA MALAT1 as a prognostic marker in human cancer: systematic review and meta-analysis. BMJ open. 2015;5(9):e008653.PubMedPubMedCentralCrossRef
116.
go back to reference Zhu J et al. Long noncoding RNA MEG3 interacts with p53 protein and regulates partial p53 target genes in hepatoma cells. PLoS One. 2015;10(10):e0139790.PubMedPubMedCentralCrossRef Zhu J et al. Long noncoding RNA MEG3 interacts with p53 protein and regulates partial p53 target genes in hepatoma cells. PLoS One. 2015;10(10):e0139790.PubMedPubMedCentralCrossRef
117.
go back to reference Tani H, Torimura M, Akimitsu N. The RNA degradation pathway regulates the function of GAS5 a non-coding RNA in mammalian cells. PLoS One. 2013;8(1):e55684.PubMedPubMedCentralCrossRef Tani H, Torimura M, Akimitsu N. The RNA degradation pathway regulates the function of GAS5 a non-coding RNA in mammalian cells. PLoS One. 2013;8(1):e55684.PubMedPubMedCentralCrossRef
118.
go back to reference Wang Y, Liu XJ, Yao XD. Function of PCA3 in prostate tissue and clinical research progress on developing a PCA3 score. Chin J Cancer Res. 2014;26(4):493–500.PubMedPubMedCentral Wang Y, Liu XJ, Yao XD. Function of PCA3 in prostate tissue and clinical research progress on developing a PCA3 score. Chin J Cancer Res. 2014;26(4):493–500.PubMedPubMedCentral
Metadata
Title
Biomarkers of genome instability and cancer epigenetics
Authors
Adriana H. O. Reis
Fernando R. Vargas
Bernardo Lemos
Publication date
01-10-2016
Publisher
Springer Netherlands
Published in
Tumor Biology / Issue 10/2016
Print ISSN: 1010-4283
Electronic ISSN: 1423-0380
DOI
https://doi.org/10.1007/s13277-016-5278-5

Other articles of this Issue 10/2016

Tumor Biology 10/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine