Skip to main content
Top
Published in: Cancer and Metastasis Reviews 2/2015

01-06-2015

Epigenetics in lung cancer diagnosis and therapy

Authors: Aditi Mehta, Stephanie Dobersch, Addi J. Romero-Olmedo, Guillermo Barreto

Published in: Cancer and Metastasis Reviews | Issue 2/2015

Login to get access

Abstract

Lung cancer is the leading cause of cancer-related deaths worldwide. The initiation and progression of lung cancer is the result of the interaction between permanent genetic and dynamic epigenetic alterations. DNA methylation is the best studied epigenetic mark in human cancers. Altered DNA methylation in cancer was identified in 1983. Within 30 years of this discovery, DNA methylation inhibitors are used clinically to treat a variety of cancers, highlighting the importance of the epigenetic basis of cancer. In addition, histone modifications, nucleosome remodeling, and micro RNA (miRNA)-mediated gene regulation are also fundamental to tumor genesis. Distinct chromatin alterations occur in all stages of lung cancer, including initiation, growth, and metastasis. Therefore, stage-specific epigenetic changes can be used as powerful and reliable tools for early diagnosis of lung cancer and to monitor patient prognosis. Moreover, since epigenetic changes are dynamic and reversible, chromatin modifiers are promising targets for the development of more effective therapeutic strategies against cancer. This review summarizes the chromatin alterations in lung cancer, focusing on the diagnostic and therapeutic approaches targeting epigenetic modifications that could help to reduce the high case-fatality rate of this dreadful disease.
Literature
1.
go back to reference Balgkouranidou, I., Liloglou, T., & Lianidou, E. S. (2013). Lung cancer epigenetics: emerging biomarkers. Biomarkers in Medicine, 7(1), 49–58.PubMedCrossRef Balgkouranidou, I., Liloglou, T., & Lianidou, E. S. (2013). Lung cancer epigenetics: emerging biomarkers. Biomarkers in Medicine, 7(1), 49–58.PubMedCrossRef
2.
go back to reference Molina, J. R., et al. (2008). Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clinic Proceedings, 83(5), 584–594.PubMedCentralPubMedCrossRef Molina, J. R., et al. (2008). Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clinic Proceedings, 83(5), 584–594.PubMedCentralPubMedCrossRef
3.
go back to reference Herbst, R. S., Heymach, J. V., & Lippman, S. M. (2008). Lung cancer. New England Journal of Medicine, 359(13), 1367–1380.PubMedCrossRef Herbst, R. S., Heymach, J. V., & Lippman, S. M. (2008). Lung cancer. New England Journal of Medicine, 359(13), 1367–1380.PubMedCrossRef
4.
5.
go back to reference Nemeth, A., & Langst, G. (2004). Chromatin higher order structure: opening up chromatin for transcription. Briefings in Functional Genomics & Proteomics, 2(4), 334–343.CrossRef Nemeth, A., & Langst, G. (2004). Chromatin higher order structure: opening up chromatin for transcription. Briefings in Functional Genomics & Proteomics, 2(4), 334–343.CrossRef
7.
go back to reference Brzezianska, E., Dutkowska, A., & Antczak, A. (2013). The significance of epigenetic alterations in lung carcinogenesis. Molecular Biology Reports, 40(1), 309–325.PubMedCentralPubMedCrossRef Brzezianska, E., Dutkowska, A., & Antczak, A. (2013). The significance of epigenetic alterations in lung carcinogenesis. Molecular Biology Reports, 40(1), 309–325.PubMedCentralPubMedCrossRef
8.
go back to reference Ozturk, N., Singh, I., Mehta, A., Braun, T., & Barreto, G. HMGA proteins as modulators of chromatin structure during transcriptional activation. Frontiers in Cell and Developmental Biology. Ozturk, N., Singh, I., Mehta, A., Braun, T., & Barreto, G. HMGA proteins as modulators of chromatin structure during transcriptional activation. Frontiers in Cell and Developmental Biology.
9.
11.
go back to reference Cedar, H., & Bergman, Y. (2009). Linking DNA methylation and histone modification: patterns and paradigms. Nature Reviews Genetics, 10(5), 295–304.PubMedCrossRef Cedar, H., & Bergman, Y. (2009). Linking DNA methylation and histone modification: patterns and paradigms. Nature Reviews Genetics, 10(5), 295–304.PubMedCrossRef
12.
go back to reference Wang, Y., et al. (2004). Beyond the double helix: writing and reading the histone code. Novartis Foundation Symposium, 259, 3–17. discussion 17–21, 163–9.PubMedCrossRef Wang, Y., et al. (2004). Beyond the double helix: writing and reading the histone code. Novartis Foundation Symposium, 259, 3–17. discussion 17–21, 163–9.PubMedCrossRef
13.
go back to reference Knudson, A. G., Jr. (1971). Mutation and cancer: statistical study of retinoblastoma. Proceedings of the National Academy of Sciences of the United States of America, 68(4), 820–823.PubMedCentralPubMedCrossRef Knudson, A. G., Jr. (1971). Mutation and cancer: statistical study of retinoblastoma. Proceedings of the National Academy of Sciences of the United States of America, 68(4), 820–823.PubMedCentralPubMedCrossRef
14.
go back to reference Kim, H., et al. (2006). Elevated mRNA levels of DNA methyltransferase-1 as an independent prognostic factor in primary nonsmall cell lung cancer. Cancer, 107(5), 1042–1049.PubMedCrossRef Kim, H., et al. (2006). Elevated mRNA levels of DNA methyltransferase-1 as an independent prognostic factor in primary nonsmall cell lung cancer. Cancer, 107(5), 1042–1049.PubMedCrossRef
15.
go back to reference Lin, R. K., et al. (2007). Alteration of DNA methyltransferases contributes to 5′CpG methylation and poor prognosis in lung cancer. Lung Cancer, 55(2), 205–213.PubMedCrossRef Lin, R. K., et al. (2007). Alteration of DNA methyltransferases contributes to 5′CpG methylation and poor prognosis in lung cancer. Lung Cancer, 55(2), 205–213.PubMedCrossRef
16.
go back to reference Feinberg, A. P., & Vogelstein, B. (1983). Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature, 301(5895), 89–92.PubMedCrossRef Feinberg, A. P., & Vogelstein, B. (1983). Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature, 301(5895), 89–92.PubMedCrossRef
17.
go back to reference Weber, M., et al. (2005). Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells. Nature Genetics, 37(8), 853–862.PubMedCrossRef Weber, M., et al. (2005). Chromosome-wide and promoter-specific analyses identify sites of differential DNA methylation in normal and transformed human cells. Nature Genetics, 37(8), 853–862.PubMedCrossRef
18.
go back to reference Rauch, T. A., et al. (2008). High-resolution mapping of DNA hypermethylation and hypomethylation in lung cancer. Proceedings of the National Academy of Sciences of the United States of America, 105(1), 252–257.PubMedCentralPubMedCrossRef Rauch, T. A., et al. (2008). High-resolution mapping of DNA hypermethylation and hypomethylation in lung cancer. Proceedings of the National Academy of Sciences of the United States of America, 105(1), 252–257.PubMedCentralPubMedCrossRef
19.
go back to reference Lander, E. S., et al. (2001). Initial sequencing and analysis of the human genome. Nature, 409(6822), 860–921.PubMedCrossRef Lander, E. S., et al. (2001). Initial sequencing and analysis of the human genome. Nature, 409(6822), 860–921.PubMedCrossRef
20.
go back to reference Gaudet, F., et al. (2003). Induction of tumors in mice by genomic hypomethylation. Science, 300(5618), 489–492.PubMedCrossRef Gaudet, F., et al. (2003). Induction of tumors in mice by genomic hypomethylation. Science, 300(5618), 489–492.PubMedCrossRef
21.
go back to reference Gupta, P. K., et al. (1997). High frequency in vivo loss of heterozygosity is primarily a consequence of mitotic recombination. Cancer Research, 57(6), 1188–1193.PubMed Gupta, P. K., et al. (1997). High frequency in vivo loss of heterozygosity is primarily a consequence of mitotic recombination. Cancer Research, 57(6), 1188–1193.PubMed
23.
24.
go back to reference Kim, M., et al. (2004). Dnmt1 deficiency leads to enhanced microsatellite instability in mouse embryonic stem cells. Nucleic Acids Research, 32(19), 5742–5749.PubMedCentralPubMedCrossRef Kim, M., et al. (2004). Dnmt1 deficiency leads to enhanced microsatellite instability in mouse embryonic stem cells. Nucleic Acids Research, 32(19), 5742–5749.PubMedCentralPubMedCrossRef
25.
26.
28.
go back to reference Dupuy, A. J., et al. (2005). Mammalian mutagenesis using a highly mobile somatic Sleeping Beauty transposon system. Nature, 436(7048), 221–226.PubMedCrossRef Dupuy, A. J., et al. (2005). Mammalian mutagenesis using a highly mobile somatic Sleeping Beauty transposon system. Nature, 436(7048), 221–226.PubMedCrossRef
29.
go back to reference Collier, L. S., et al. (2005). Cancer gene discovery in solid tumours using transposon-based somatic mutagenesis in the mouse. Nature, 436(7048), 272–276.PubMedCrossRef Collier, L. S., et al. (2005). Cancer gene discovery in solid tumours using transposon-based somatic mutagenesis in the mouse. Nature, 436(7048), 272–276.PubMedCrossRef
30.
31.
go back to reference Lujambio, A., et al. (2010). CpG island hypermethylation-associated silencing of non-coding RNAs transcribed from ultraconserved regions in human cancer. Oncogene, 29(48), 6390–6401.PubMedCentralPubMedCrossRef Lujambio, A., et al. (2010). CpG island hypermethylation-associated silencing of non-coding RNAs transcribed from ultraconserved regions in human cancer. Oncogene, 29(48), 6390–6401.PubMedCentralPubMedCrossRef
32.
go back to reference Lin, S. H., et al. (2014). Genes suppressed by DNA methylation in non-small cell lung cancer reveal the epigenetics of epithelial-mesenchymal transition. BMC Genomics, 15, 1079.PubMedCentralPubMedCrossRef Lin, S. H., et al. (2014). Genes suppressed by DNA methylation in non-small cell lung cancer reveal the epigenetics of epithelial-mesenchymal transition. BMC Genomics, 15, 1079.PubMedCentralPubMedCrossRef
33.
go back to reference Thiery, J. P., et al. (2009). Epithelial-mesenchymal transitions in development and disease. Cell, 139(5), 871–890.PubMedCrossRef Thiery, J. P., et al. (2009). Epithelial-mesenchymal transitions in development and disease. Cell, 139(5), 871–890.PubMedCrossRef
34.
35.
go back to reference Le Bras, G. F., Taubenslag, K. J., & Andl, C. D. (2012). The regulation of cell-cell adhesion during epithelial-mesenchymal transition, motility and tumor progression. Cell Adhesion & Migration, 6(4), 365–373.CrossRef Le Bras, G. F., Taubenslag, K. J., & Andl, C. D. (2012). The regulation of cell-cell adhesion during epithelial-mesenchymal transition, motility and tumor progression. Cell Adhesion & Migration, 6(4), 365–373.CrossRef
36.
go back to reference Richardson, F., et al. (2012). The evaluation of E-Cadherin and vimentin as biomarkers of clinical outcomes among patients with non-small cell lung cancer treated with erlotinib as second- or third-line therapy. Anticancer Research, 32(2), 537–552.PubMed Richardson, F., et al. (2012). The evaluation of E-Cadherin and vimentin as biomarkers of clinical outcomes among patients with non-small cell lung cancer treated with erlotinib as second- or third-line therapy. Anticancer Research, 32(2), 537–552.PubMed
37.
go back to reference Xiao, D., & He, J. (2010). Epithelial mesenchymal transition and lung cancer. Journal of Thoracic Disease, 2(3), 154–159.PubMedCentralPubMed Xiao, D., & He, J. (2010). Epithelial mesenchymal transition and lung cancer. Journal of Thoracic Disease, 2(3), 154–159.PubMedCentralPubMed
38.
go back to reference Singh, A., & Settleman, J. (2010). EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene, 29(34), 4741–4751.PubMedCentralPubMedCrossRef Singh, A., & Settleman, J. (2010). EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene, 29(34), 4741–4751.PubMedCentralPubMedCrossRef
40.
go back to reference Yang, J., et al. (2004). Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell, 117(7), 927–939.PubMedCrossRef Yang, J., et al. (2004). Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell, 117(7), 927–939.PubMedCrossRef
41.
go back to reference Tran, P. T., et al. (2012). Twist1 suppresses senescence programs and thereby accelerates and maintains mutant Kras-induced lung tumorigenesis. PLoS Genetics, 8(5), e1002650.PubMedCentralPubMedCrossRef Tran, P. T., et al. (2012). Twist1 suppresses senescence programs and thereby accelerates and maintains mutant Kras-induced lung tumorigenesis. PLoS Genetics, 8(5), e1002650.PubMedCentralPubMedCrossRef
42.
go back to reference De Craene, B., & Berx, G. (2013). Regulatory networks defining EMT during cancer initiation and progression. Nature Reviews Cancer, 13(2), 97–110.PubMedCrossRef De Craene, B., & Berx, G. (2013). Regulatory networks defining EMT during cancer initiation and progression. Nature Reviews Cancer, 13(2), 97–110.PubMedCrossRef
43.
go back to reference Van’t Westeinde, S. C., & van Klaveren, R. J. (2011). Screening and early detection of lung cancer. Cancer Journal, 17(1), 3–10.CrossRef Van’t Westeinde, S. C., & van Klaveren, R. J. (2011). Screening and early detection of lung cancer. Cancer Journal, 17(1), 3–10.CrossRef
44.
go back to reference Mulshine, J. L. (2003). Screening for lung cancer: in pursuit of pre-metastatic disease. Nature Reviews Cancer, 3(1), 65–73.PubMedCrossRef Mulshine, J. L. (2003). Screening for lung cancer: in pursuit of pre-metastatic disease. Nature Reviews Cancer, 3(1), 65–73.PubMedCrossRef
45.
go back to reference Marshall, H. M., et al. (2013). Screening for lung cancer with low-dose computed tomography: a review of current status. Journal of Thoracic Disease, 5(Suppl 5), S524–S539.PubMedCentralPubMed Marshall, H. M., et al. (2013). Screening for lung cancer with low-dose computed tomography: a review of current status. Journal of Thoracic Disease, 5(Suppl 5), S524–S539.PubMedCentralPubMed
46.
go back to reference Siegel, R., Naishadham, D., & Jemal, A. (2013). Cancer statistics, 2013. CA: A Cancer Journal for Clinicians, 63(1), 11–30. Siegel, R., Naishadham, D., & Jemal, A. (2013). Cancer statistics, 2013. CA: A Cancer Journal for Clinicians, 63(1), 11–30.
47.
go back to reference Ahrendt, S. A., et al. (1999). Molecular detection of tumor cells in bronchoalveolar lavage fluid from patients with early stage lung cancer. Journal of the National Cancer Institute, 91(4), 332–339.PubMedCrossRef Ahrendt, S. A., et al. (1999). Molecular detection of tumor cells in bronchoalveolar lavage fluid from patients with early stage lung cancer. Journal of the National Cancer Institute, 91(4), 332–339.PubMedCrossRef
48.
go back to reference Selamat, S. A., et al. (2011). DNA methylation changes in atypical adenomatous hyperplasia, adenocarcinoma in situ, and lung adenocarcinoma. PLoS One, 6(6), e21443.PubMedCentralPubMedCrossRef Selamat, S. A., et al. (2011). DNA methylation changes in atypical adenomatous hyperplasia, adenocarcinoma in situ, and lung adenocarcinoma. PLoS One, 6(6), e21443.PubMedCentralPubMedCrossRef
49.
go back to reference Yanagawa, N., et al. (2003). Promoter hypermethylation of tumor suppressor and tumor-related genes in non-small cell lung cancers. Cancer Science, 94(7), 589–592.PubMedCrossRef Yanagawa, N., et al. (2003). Promoter hypermethylation of tumor suppressor and tumor-related genes in non-small cell lung cancers. Cancer Science, 94(7), 589–592.PubMedCrossRef
50.
go back to reference Pulling, L. C., et al. (2003). Promoter hypermethylation of the O6-methylguanine-DNA methyltransferase gene: more common in lung adenocarcinomas from never-smokers than smokers and associated with tumor progression. Cancer Research, 63(16), 4842–4848.PubMed Pulling, L. C., et al. (2003). Promoter hypermethylation of the O6-methylguanine-DNA methyltransferase gene: more common in lung adenocarcinomas from never-smokers than smokers and associated with tumor progression. Cancer Research, 63(16), 4842–4848.PubMed
51.
go back to reference Kim, J. S., et al. (2005). Aberrant methylation of H-cadherin (CDH13) promoter is associated with tumor progression in primary nonsmall cell lung carcinoma. Cancer, 104(9), 1825–1833.PubMedCrossRef Kim, J. S., et al. (2005). Aberrant methylation of H-cadherin (CDH13) promoter is associated with tumor progression in primary nonsmall cell lung carcinoma. Cancer, 104(9), 1825–1833.PubMedCrossRef
52.
go back to reference Lin, Q., et al. (2009). RASSF1A, APC, ESR1, ABCB1 and HOXC9, but not p16INK4A, DAPK1, PTEN and MT1G genes were frequently methylated in the stage I non-small cell lung cancer in China. Journal of Cancer Research and Clinical Oncology, 135(12), 1675–1684.PubMedCrossRef Lin, Q., et al. (2009). RASSF1A, APC, ESR1, ABCB1 and HOXC9, but not p16INK4A, DAPK1, PTEN and MT1G genes were frequently methylated in the stage I non-small cell lung cancer in China. Journal of Cancer Research and Clinical Oncology, 135(12), 1675–1684.PubMedCrossRef
53.
go back to reference Kneip, C., et al. (2011). SHOX2 DNA methylation is a biomarker for the diagnosis of lung cancer in plasma. Journal of Thoracic Oncology, 6(10), 1632–1638.PubMedCrossRef Kneip, C., et al. (2011). SHOX2 DNA methylation is a biomarker for the diagnosis of lung cancer in plasma. Journal of Thoracic Oncology, 6(10), 1632–1638.PubMedCrossRef
54.
go back to reference Tsou, J. A., et al. (2007). Identification of a panel of sensitive and specific DNA methylation markers for lung adenocarcinoma. Molecular Cancer, 6, 70.PubMedCentralPubMedCrossRef Tsou, J. A., et al. (2007). Identification of a panel of sensitive and specific DNA methylation markers for lung adenocarcinoma. Molecular Cancer, 6, 70.PubMedCentralPubMedCrossRef
55.
go back to reference Niklinska, W., et al. (2009). Prognostic significance of DAPK and RASSF1A promoter hypermethylation in non-small cell lung cancer (NSCLC). Folia Histochemica et Cytobiologica, 47(2), 275–280.PubMedCrossRef Niklinska, W., et al. (2009). Prognostic significance of DAPK and RASSF1A promoter hypermethylation in non-small cell lung cancer (NSCLC). Folia Histochemica et Cytobiologica, 47(2), 275–280.PubMedCrossRef
56.
go back to reference Risse, E. K., et al. (1985). Sputum cytology by the Saccomanno method in diagnosing lung malignancy. Diagnostic Cytopathology, 1(4), 286–291.PubMedCrossRef Risse, E. K., et al. (1985). Sputum cytology by the Saccomanno method in diagnosing lung malignancy. Diagnostic Cytopathology, 1(4), 286–291.PubMedCrossRef
57.
go back to reference Hubers, A. J., et al. (2014). Combined sputum hypermethylation and eNose analysis for lung cancer diagnosis. Journal of Clinical Pathology, 67(8), 707–711.PubMedCrossRef Hubers, A. J., et al. (2014). Combined sputum hypermethylation and eNose analysis for lung cancer diagnosis. Journal of Clinical Pathology, 67(8), 707–711.PubMedCrossRef
58.
go back to reference Leng, S., et al. (2012). Defining a gene promoter methylation signature in sputum for lung cancer risk assessment. Clinical Cancer Research, 18(12), 3387–3395.PubMedCentralPubMedCrossRef Leng, S., et al. (2012). Defining a gene promoter methylation signature in sputum for lung cancer risk assessment. Clinical Cancer Research, 18(12), 3387–3395.PubMedCentralPubMedCrossRef
59.
go back to reference Wang, Y. C., et al. (2003). Inactivation of hMLH1 and hMSH2 by promoter methylation in primary non-small cell lung tumors and matched sputum samples. Journal of Clinical Investigation, 111(6), 887–895.PubMedCentralPubMedCrossRef Wang, Y. C., et al. (2003). Inactivation of hMLH1 and hMSH2 by promoter methylation in primary non-small cell lung tumors and matched sputum samples. Journal of Clinical Investigation, 111(6), 887–895.PubMedCentralPubMedCrossRef
60.
go back to reference Liu, Y., et al. (2003). Hypermethylation of p16INK4a in Chinese lung cancer patients: biological and clinical implications. Carcinogenesis, 24(12), 1897–1901.PubMedCrossRef Liu, Y., et al. (2003). Hypermethylation of p16INK4a in Chinese lung cancer patients: biological and clinical implications. Carcinogenesis, 24(12), 1897–1901.PubMedCrossRef
61.
go back to reference Destro, A., et al. (2004). K-ras and p16(INK4A)alterations in sputum of NSCLC patients and in heavy asymptomatic chronic smokers. Lung Cancer, 44(1), 23–32.PubMedCrossRef Destro, A., et al. (2004). K-ras and p16(INK4A)alterations in sputum of NSCLC patients and in heavy asymptomatic chronic smokers. Lung Cancer, 44(1), 23–32.PubMedCrossRef
62.
go back to reference Belinsky, S. A., et al. (2007). Predicting gene promoter methylation in non-small-cell lung cancer by evaluating sputum and serum. British Journal of Cancer, 96(8), 1278–1283.PubMedCentralPubMedCrossRef Belinsky, S. A., et al. (2007). Predicting gene promoter methylation in non-small-cell lung cancer by evaluating sputum and serum. British Journal of Cancer, 96(8), 1278–1283.PubMedCentralPubMedCrossRef
63.
go back to reference Cirincione, R., et al. (2006). Methylation profile in tumor and sputum samples of lung cancer patients detected by spiral computed tomography: a nested case–control study. International Journal of Cancer, 118(5), 1248–1253.CrossRef Cirincione, R., et al. (2006). Methylation profile in tumor and sputum samples of lung cancer patients detected by spiral computed tomography: a nested case–control study. International Journal of Cancer, 118(5), 1248–1253.CrossRef
64.
go back to reference Ammanagi, A. S., et al. (2012). Sputum cytology in suspected cases of carcinoma of lung (Sputum cytology a poor man’s bronchoscopy!). Lung India, 29(1), 19–23.PubMedCentralPubMedCrossRef Ammanagi, A. S., et al. (2012). Sputum cytology in suspected cases of carcinoma of lung (Sputum cytology a poor man’s bronchoscopy!). Lung India, 29(1), 19–23.PubMedCentralPubMedCrossRef
65.
go back to reference Hsu, H. S., et al. (2007). Characterization of a multiple epigenetic marker panel for lung cancer detection and risk assessment in plasma. Cancer, 110(9), 2019–2026.PubMedCrossRef Hsu, H. S., et al. (2007). Characterization of a multiple epigenetic marker panel for lung cancer detection and risk assessment in plasma. Cancer, 110(9), 2019–2026.PubMedCrossRef
66.
go back to reference Suga, Y., et al. (2008). Quantitative p16 and ESR1 methylation in the peripheral blood of patients with non-small cell lung cancer. Oncology Reports, 20(5), 1137–1142.PubMed Suga, Y., et al. (2008). Quantitative p16 and ESR1 methylation in the peripheral blood of patients with non-small cell lung cancer. Oncology Reports, 20(5), 1137–1142.PubMed
67.
go back to reference Bearzatto, A., et al. (2002). p16(INK4A) Hypermethylation detected by fluorescent methylation-specific PCR in plasmas from non-small cell lung cancer. Clinical Cancer Research, 8(12), 3782–3787.PubMed Bearzatto, A., et al. (2002). p16(INK4A) Hypermethylation detected by fluorescent methylation-specific PCR in plasmas from non-small cell lung cancer. Clinical Cancer Research, 8(12), 3782–3787.PubMed
68.
go back to reference Russo, A. L., et al. (2005). Differential DNA hypermethylation of critical genes mediates the stage-specific tobacco smoke-induced neoplastic progression of lung cancer. Clinical Cancer Research, 11(7), 2466–2470.PubMedCrossRef Russo, A. L., et al. (2005). Differential DNA hypermethylation of critical genes mediates the stage-specific tobacco smoke-induced neoplastic progression of lung cancer. Clinical Cancer Research, 11(7), 2466–2470.PubMedCrossRef
69.
go back to reference Hanahan, D., & Weinberg, R. A. (2011). Hallmarks of cancer: the next generation. Cell, 144(5), 646–674.PubMedCrossRef Hanahan, D., & Weinberg, R. A. (2011). Hallmarks of cancer: the next generation. Cell, 144(5), 646–674.PubMedCrossRef
70.
go back to reference Boedeker, E., Friedel, G., & Walles, T. (2012). Sniffer dogs as part of a bimodal bionic research approach to develop a lung cancer screening. Interactive Cardiovascular and Thoracic Surgery, 14(5), 511–515.PubMedCentralPubMedCrossRef Boedeker, E., Friedel, G., & Walles, T. (2012). Sniffer dogs as part of a bimodal bionic research approach to develop a lung cancer screening. Interactive Cardiovascular and Thoracic Surgery, 14(5), 511–515.PubMedCentralPubMedCrossRef
71.
go back to reference Rattray, N. J., et al. (2014). Taking your breath away: metabolomics breathes life in to personalized medicine. Trends in Biotechnology, 32(10), 538–548.PubMedCrossRef Rattray, N. J., et al. (2014). Taking your breath away: metabolomics breathes life in to personalized medicine. Trends in Biotechnology, 32(10), 538–548.PubMedCrossRef
72.
go back to reference Dent, A. G., Sutedja, T. G., & Zimmerman, P. V. (2013). Exhaled breath analysis for lung cancer. Journal of Thoracic Disease, 5(Suppl 5), S540–S550.PubMedCentralPubMed Dent, A. G., Sutedja, T. G., & Zimmerman, P. V. (2013). Exhaled breath analysis for lung cancer. Journal of Thoracic Disease, 5(Suppl 5), S540–S550.PubMedCentralPubMed
73.
go back to reference Xiao, P., et al. (2014). Methylation of P16 in exhaled breath condensate for diagnosis of non-small cell lung cancer. Lung Cancer, 83(1), 56–60.PubMedCrossRef Xiao, P., et al. (2014). Methylation of P16 in exhaled breath condensate for diagnosis of non-small cell lung cancer. Lung Cancer, 83(1), 56–60.PubMedCrossRef
74.
go back to reference Han, W., et al. (2009). Gene promoter methylation assayed in exhaled breath, with differences in smokers and lung cancer patients. Respiratory Research, 10, 86.PubMedCentralPubMedCrossRef Han, W., et al. (2009). Gene promoter methylation assayed in exhaled breath, with differences in smokers and lung cancer patients. Respiratory Research, 10, 86.PubMedCentralPubMedCrossRef
75.
go back to reference Filipowicz, W., Bhattacharyya, S. N., & Sonenberg, N. (2008). Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nature Reviews Genetics, 9(2), 102–114.PubMedCrossRef Filipowicz, W., Bhattacharyya, S. N., & Sonenberg, N. (2008). Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nature Reviews Genetics, 9(2), 102–114.PubMedCrossRef
77.
go back to reference Calin, G. A., et al. (2004). MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proceedings of the National Academy of Sciences of the United States of America, 101(32), 11755–11760.PubMedCentralPubMedCrossRef Calin, G. A., et al. (2004). MicroRNA profiling reveals distinct signatures in B cell chronic lymphocytic leukemias. Proceedings of the National Academy of Sciences of the United States of America, 101(32), 11755–11760.PubMedCentralPubMedCrossRef
78.
go back to reference Lu, J., et al. (2005). MicroRNA expression profiles classify human cancers. Nature, 435(7043), 834–838.PubMedCrossRef Lu, J., et al. (2005). MicroRNA expression profiles classify human cancers. Nature, 435(7043), 834–838.PubMedCrossRef
80.
go back to reference Johnson, S. M., et al. (2005). RAS is regulated by the let-7 microRNA family. Cell, 120(5), 635–647.PubMedCrossRef Johnson, S. M., et al. (2005). RAS is regulated by the let-7 microRNA family. Cell, 120(5), 635–647.PubMedCrossRef
81.
go back to reference Takamizawa, J., et al. (2004). Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Research, 64(11), 3753–3756.PubMedCrossRef Takamizawa, J., et al. (2004). Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Research, 64(11), 3753–3756.PubMedCrossRef
82.
go back to reference Hayashita, Y., et al. (2005). A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Research, 65(21), 9628–9632.PubMedCrossRef Hayashita, Y., et al. (2005). A polycistronic microRNA cluster, miR-17-92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Research, 65(21), 9628–9632.PubMedCrossRef
83.
84.
go back to reference Boeri, M., et al. (2011). MicroRNA signatures in tissues and plasma predict development and prognosis of computed tomography detected lung cancer. Proceedings of the National Academy of Sciences of the United States of America, 108(9), 3713–3718.PubMedCentralPubMedCrossRef Boeri, M., et al. (2011). MicroRNA signatures in tissues and plasma predict development and prognosis of computed tomography detected lung cancer. Proceedings of the National Academy of Sciences of the United States of America, 108(9), 3713–3718.PubMedCentralPubMedCrossRef
85.
86.
go back to reference Nair, V. S., Maeda, L. S., & Ioannidis, J. P. (2012). Clinical outcome prediction by microRNAs in human cancer: a systematic review. Journal of the National Cancer Institute, 104(7), 528–540.PubMedCentralPubMedCrossRef Nair, V. S., Maeda, L. S., & Ioannidis, J. P. (2012). Clinical outcome prediction by microRNAs in human cancer: a systematic review. Journal of the National Cancer Institute, 104(7), 528–540.PubMedCentralPubMedCrossRef
87.
go back to reference Cagle, P. T., & Chirieac, L. R. (2012). Advances in treatment of lung cancer with targeted therapy. Archives of Pathology and Laboratory Medicine, 136(5), 504–509.PubMedCrossRef Cagle, P. T., & Chirieac, L. R. (2012). Advances in treatment of lung cancer with targeted therapy. Archives of Pathology and Laboratory Medicine, 136(5), 504–509.PubMedCrossRef
88.
go back to reference (2011) In The diagnosis and treatment of lung cancer (Update). Cardiff (UK). (2011) In The diagnosis and treatment of lung cancer (Update). Cardiff (UK).
89.
go back to reference Schuchert, M. J., et al. (2010). Sublobar resection for early-stage lung cancer. Seminars in Thoracic and Cardiovascular Surgery, 22(1), 22–31.PubMedCrossRef Schuchert, M. J., et al. (2010). Sublobar resection for early-stage lung cancer. Seminars in Thoracic and Cardiovascular Surgery, 22(1), 22–31.PubMedCrossRef
90.
go back to reference Sonke, J. J., & Belderbos, J. (2010). Adaptive radiotherapy for lung cancer. Seminars in Radiation Oncology, 20(2), 94–106.PubMedCrossRef Sonke, J. J., & Belderbos, J. (2010). Adaptive radiotherapy for lung cancer. Seminars in Radiation Oncology, 20(2), 94–106.PubMedCrossRef
91.
go back to reference Pfister, D. G., et al. (2004). American Society of Clinical Oncology treatment of unresectable non-small-cell lung cancer guideline: update 2003. Journal of Clinical Oncology, 22(2), 330–353.PubMedCrossRef Pfister, D. G., et al. (2004). American Society of Clinical Oncology treatment of unresectable non-small-cell lung cancer guideline: update 2003. Journal of Clinical Oncology, 22(2), 330–353.PubMedCrossRef
92.
go back to reference Simone, C. B., 2nd, et al. (2013). Stereotactic body radiation therapy for lung cancer. Chest, 143(6), 1784–1790.PubMedCrossRef Simone, C. B., 2nd, et al. (2013). Stereotactic body radiation therapy for lung cancer. Chest, 143(6), 1784–1790.PubMedCrossRef
94.
go back to reference Ma, P.C. (2012). Personalized targeted therapy in advanced non-small cell lung cancer. Cleveland Clinic Journal of Medicine, (79 Electronic Suppl 1), eS56–60. Ma, P.C. (2012). Personalized targeted therapy in advanced non-small cell lung cancer. Cleveland Clinic Journal of Medicine, (79 Electronic Suppl 1), eS56–60.
95.
go back to reference Sandler, A., et al. (2006). Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. New England Journal of Medicine, 355(24), 2542–2550.PubMedCrossRef Sandler, A., et al. (2006). Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. New England Journal of Medicine, 355(24), 2542–2550.PubMedCrossRef
99.
go back to reference Chuang, J. C., & Jones, P. A. (2007). Epigenetics and microRNAs. Pediatric Research, 61(5 Pt 2), 24R–29R.PubMedCrossRef Chuang, J. C., & Jones, P. A. (2007). Epigenetics and microRNAs. Pediatric Research, 61(5 Pt 2), 24R–29R.PubMedCrossRef
100.
go back to reference Dawson, M. A., & Kouzarides, T. (2012). Cancer epigenetics: from mechanism to therapy. Cell, 150(1), 12–27.PubMedCrossRef Dawson, M. A., & Kouzarides, T. (2012). Cancer epigenetics: from mechanism to therapy. Cell, 150(1), 12–27.PubMedCrossRef
101.
go back to reference Keohane, C., Mesa, R., & Harrison, C. (2013). The role of JAK1/2 inhibitors in the treatment of chronic myeloproliferative neoplasms. American Society of Clinical Oncology Educational Book, 301–305. Keohane, C., Mesa, R., & Harrison, C. (2013). The role of JAK1/2 inhibitors in the treatment of chronic myeloproliferative neoplasms. American Society of Clinical Oncology Educational Book, 301–305.
103.
go back to reference Lyko, F., & Brown, R. (2005). DNA methyltransferase inhibitors and the development of epigenetic cancer therapies. Journal of the National Cancer Institute, 97(20), 1498–1506.PubMedCrossRef Lyko, F., & Brown, R. (2005). DNA methyltransferase inhibitors and the development of epigenetic cancer therapies. Journal of the National Cancer Institute, 97(20), 1498–1506.PubMedCrossRef
105.
go back to reference Christman, J. K. (2002). 5-Azacytidine and 5-aza-2′-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene, 21(35), 5483–5495.PubMedCrossRef Christman, J. K. (2002). 5-Azacytidine and 5-aza-2′-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene, 21(35), 5483–5495.PubMedCrossRef
106.
go back to reference Komashko, V. M., & Farnham, P. J. (2010). 5-azacytidine treatment reorganizes genomic histone modification patterns. Epigenetics, 5(3), 229–240.PubMedCrossRef Komashko, V. M., & Farnham, P. J. (2010). 5-azacytidine treatment reorganizes genomic histone modification patterns. Epigenetics, 5(3), 229–240.PubMedCrossRef
107.
go back to reference Kim, H. J., & Bae, S. C. (2011). Histone deacetylase inhibitors: molecular mechanisms of action and clinical trials as anti-cancer drugs. American Journal of Translational Research, 3(2), 166–179.PubMedCentralPubMed Kim, H. J., & Bae, S. C. (2011). Histone deacetylase inhibitors: molecular mechanisms of action and clinical trials as anti-cancer drugs. American Journal of Translational Research, 3(2), 166–179.PubMedCentralPubMed
108.
go back to reference Brazelle, W., et al. (2010). Histone deacetylase inhibitors downregulate checkpoint kinase 1 expression to induce cell death in non-small cell lung cancer cells. PLoS One, 5(12), e14335.PubMedCentralPubMedCrossRef Brazelle, W., et al. (2010). Histone deacetylase inhibitors downregulate checkpoint kinase 1 expression to induce cell death in non-small cell lung cancer cells. PLoS One, 5(12), e14335.PubMedCentralPubMedCrossRef
109.
go back to reference Tang, Y. A., et al. (2010). A novel histone deacetylase inhibitor exhibits antitumor activity via apoptosis induction, F-actin disruption and gene acetylation in lung cancer. PLoS One, 5(9), e12417.PubMedCentralPubMedCrossRef Tang, Y. A., et al. (2010). A novel histone deacetylase inhibitor exhibits antitumor activity via apoptosis induction, F-actin disruption and gene acetylation in lung cancer. PLoS One, 5(9), e12417.PubMedCentralPubMedCrossRef
110.
go back to reference Perrino, E., et al. (2008). New sulfurated derivatives of valproic acid with enhanced histone deacetylase inhibitory activity. Bioorganic and Medicinal Chemistry Letters, 18(6), 1893–1897.PubMedCrossRef Perrino, E., et al. (2008). New sulfurated derivatives of valproic acid with enhanced histone deacetylase inhibitory activity. Bioorganic and Medicinal Chemistry Letters, 18(6), 1893–1897.PubMedCrossRef
111.
go back to reference Tesei, A., et al. (2012). Organosulfur derivatives of the HDAC inhibitor valproic acid sensitize human lung cancer cell lines to apoptosis and to cisplatin cytotoxicity. Journal of Cellular Physiology, 227(10), 3389–3396.PubMedCrossRef Tesei, A., et al. (2012). Organosulfur derivatives of the HDAC inhibitor valproic acid sensitize human lung cancer cell lines to apoptosis and to cisplatin cytotoxicity. Journal of Cellular Physiology, 227(10), 3389–3396.PubMedCrossRef
112.
go back to reference Zhang, W., et al. (2009). Histone deacetylase inhibitor romidepsin enhances anti-tumor effect of erlotinib in non-small cell lung cancer (NSCLC) cell lines. Journal of Thoracic Oncology, 4(2), 161–166.PubMedCentralPubMedCrossRef Zhang, W., et al. (2009). Histone deacetylase inhibitor romidepsin enhances anti-tumor effect of erlotinib in non-small cell lung cancer (NSCLC) cell lines. Journal of Thoracic Oncology, 4(2), 161–166.PubMedCentralPubMedCrossRef
113.
go back to reference Karthik, S., et al. (2014). Romidepsin induces cell cycle arrest, apoptosis, histone hyperacetylation and reduces matrix metalloproteinases 2 and 9 expression in bortezomib sensitized non-small cell lung cancer cells. Biomedicine and Pharmacotherapy, 68(3), 327–334.PubMedCrossRef Karthik, S., et al. (2014). Romidepsin induces cell cycle arrest, apoptosis, histone hyperacetylation and reduces matrix metalloproteinases 2 and 9 expression in bortezomib sensitized non-small cell lung cancer cells. Biomedicine and Pharmacotherapy, 68(3), 327–334.PubMedCrossRef
114.
go back to reference Papeleu, P., et al. (2003). Trichostatin A induces differential cell cycle arrests but does not induce apoptosis in primary cultures of mitogen-stimulated rat hepatocytes. Journal of Hepatology, 39(3), 374–382.PubMedCrossRef Papeleu, P., et al. (2003). Trichostatin A induces differential cell cycle arrests but does not induce apoptosis in primary cultures of mitogen-stimulated rat hepatocytes. Journal of Hepatology, 39(3), 374–382.PubMedCrossRef
115.
go back to reference Mukhopadhyay, N. K., et al. (2006). Effectiveness of trichostatin A as a potential candidate for anticancer therapy in non-small-cell lung cancer. Annals of Thoracic Surgery, 81(3), 1034–1042.PubMedCrossRef Mukhopadhyay, N. K., et al. (2006). Effectiveness of trichostatin A as a potential candidate for anticancer therapy in non-small-cell lung cancer. Annals of Thoracic Surgery, 81(3), 1034–1042.PubMedCrossRef
116.
go back to reference Platta, C. S., et al. (2007). The HDAC inhibitor trichostatin A inhibits growth of small cell lung cancer cells. Journal of Surgical Research, 142(2), 219–226.PubMedCrossRef Platta, C. S., et al. (2007). The HDAC inhibitor trichostatin A inhibits growth of small cell lung cancer cells. Journal of Surgical Research, 142(2), 219–226.PubMedCrossRef
117.
go back to reference Juergens, R.A., & Rudin, C.M. (2013). Aberrant epigenetic regulation: a central contributor to lung carcinogenesis and a new therapeutic target. American Society of Clinical Oncology Educational Book. Juergens, R.A., & Rudin, C.M. (2013). Aberrant epigenetic regulation: a central contributor to lung carcinogenesis and a new therapeutic target. American Society of Clinical Oncology Educational Book.
118.
go back to reference Petta, V., et al. (2013). Histones and lung cancer: are the histone deacetylases a promising therapeutic target? Cancer Chemotherapy and Pharmacology, 72(5), 935–952.PubMedCrossRef Petta, V., et al. (2013). Histones and lung cancer: are the histone deacetylases a promising therapeutic target? Cancer Chemotherapy and Pharmacology, 72(5), 935–952.PubMedCrossRef
119.
go back to reference Li, C. T., et al. (2011). Vorinostat, SAHA, represses telomerase activity via epigenetic regulation of telomerase reverse transcriptase in non-small cell lung cancer cells. Journal of Cellular Biochemistry, 112(10), 3044–3053.PubMedCrossRef Li, C. T., et al. (2011). Vorinostat, SAHA, represses telomerase activity via epigenetic regulation of telomerase reverse transcriptase in non-small cell lung cancer cells. Journal of Cellular Biochemistry, 112(10), 3044–3053.PubMedCrossRef
120.
go back to reference Forde, P. M., Brahmer, J. R., & Kelly, R. J. (2014). New strategies in lung cancer: epigenetic therapy for non-small cell lung cancer. Clinical Cancer Research, 20(9), 2244–2248.PubMedCentralPubMedCrossRef Forde, P. M., Brahmer, J. R., & Kelly, R. J. (2014). New strategies in lung cancer: epigenetic therapy for non-small cell lung cancer. Clinical Cancer Research, 20(9), 2244–2248.PubMedCentralPubMedCrossRef
121.
go back to reference Belinsky, S. A., et al. (2011). Combination therapy with Vidaza and entinostat suppresses tumor growth and reprograms the epigenome in an orthotopic lung cancer model. Cancer Research, 71(2), 454–462.PubMedCentralPubMedCrossRef Belinsky, S. A., et al. (2011). Combination therapy with Vidaza and entinostat suppresses tumor growth and reprograms the epigenome in an orthotopic lung cancer model. Cancer Research, 71(2), 454–462.PubMedCentralPubMedCrossRef
122.
go back to reference Noro, R., et al. (2007). PTEN inactivation in lung cancer cells and the effect of its recovery on treatment with epidermal growth factor receptor tyrosine kinase inhibitors. International Journal of Oncology, 31(5), 1157–1163.PubMed Noro, R., et al. (2007). PTEN inactivation in lung cancer cells and the effect of its recovery on treatment with epidermal growth factor receptor tyrosine kinase inhibitors. International Journal of Oncology, 31(5), 1157–1163.PubMed
123.
go back to reference Peipp, M., et al. (2002). A recombinant CD7-specific single-chain immunotoxin is a potent inducer of apoptosis in acute leukemic T cells. Cancer Research, 62(10), 2848–2855.PubMed Peipp, M., et al. (2002). A recombinant CD7-specific single-chain immunotoxin is a potent inducer of apoptosis in acute leukemic T cells. Cancer Research, 62(10), 2848–2855.PubMed
124.
go back to reference Deng, X. K., Nesbit, L. A., & Morrow, K. J., Jr. (2003). Recombinant single-chain variable fragment antibodies directed against Clostridium difficile toxin B produced by use of an optimized phage display system. Clinical and Diagnostic Laboratory Immunology, 10(4), 587–595.PubMedCentralPubMed Deng, X. K., Nesbit, L. A., & Morrow, K. J., Jr. (2003). Recombinant single-chain variable fragment antibodies directed against Clostridium difficile toxin B produced by use of an optimized phage display system. Clinical and Diagnostic Laboratory Immunology, 10(4), 587–595.PubMedCentralPubMed
126.
go back to reference Wang, R., et al. (2013). Engineering production of functional scFv antibody in E. coli by co-expressing the molecule chaperone Skp. Frontiers in Cellular and Infection Microbiology, 3, 72.PubMedCentralPubMed Wang, R., et al. (2013). Engineering production of functional scFv antibody in E. coli by co-expressing the molecule chaperone Skp. Frontiers in Cellular and Infection Microbiology, 3, 72.PubMedCentralPubMed
128.
go back to reference Xu, X., et al. (2012). Evidence for type II cells as cells of origin of K-Ras-induced distal lung adenocarcinoma. Proceedings of the National Academy of Sciences of the United States of America, 109(13), 4910–4915.PubMedCentralPubMedCrossRef Xu, X., et al. (2012). Evidence for type II cells as cells of origin of K-Ras-induced distal lung adenocarcinoma. Proceedings of the National Academy of Sciences of the United States of America, 109(13), 4910–4915.PubMedCentralPubMedCrossRef
129.
go back to reference Oh, Y. K., & Park, T. G. (2009). siRNA delivery systems for cancer treatment. Advanced Drug Delivery Reviews, 61(10), 850–862.PubMedCrossRef Oh, Y. K., & Park, T. G. (2009). siRNA delivery systems for cancer treatment. Advanced Drug Delivery Reviews, 61(10), 850–862.PubMedCrossRef
Metadata
Title
Epigenetics in lung cancer diagnosis and therapy
Authors
Aditi Mehta
Stephanie Dobersch
Addi J. Romero-Olmedo
Guillermo Barreto
Publication date
01-06-2015
Publisher
Springer US
Published in
Cancer and Metastasis Reviews / Issue 2/2015
Print ISSN: 0167-7659
Electronic ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-015-9563-3

Other articles of this Issue 2/2015

Cancer and Metastasis Reviews 2/2015 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