Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-ttngx Total loading time: 0 Render date: 2024-06-08T10:26:09.630Z Has data issue: false hasContentIssue false

43 - Lung cancer

from Part 3.1 - Molecular pathology: carcinomas

Published online by Cambridge University Press:  05 February 2015

Jill E. Larsen
Affiliation:
Hamon Center for herapeutic Oncology Research, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
John D. Minna
Affiliation:
Hamon Center for herapeutic Oncology Research, Simmons Cancer Center University of Texas Southwestern Medical Center, Dallas, TX, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

DNA damage leads to lung cancer

Approximately 85% of lung cancers are caused by tobacco-smoke-acquired carcinogenesis, while worldwide, ~15–25% of lung cancer cases occur in lifetime “never smokers” (less than 100 cigarettes in a lifetime). These etiologic differences are associated with distinct differences in tumor-acquired molecular changes, such as EGFR mutations in never-smoking lung cancers (1,2). Of importance, with cessation of cigarette smoking from public health initiatives, ~50% of all newly diagnosed cases of lung cancer occur in former smokers who ceased smoking >5 years previously. Never-smoking lung cancers represent a distinct disease that occurs more frequently in women and East Asians, targets the distal airways, is usually adenocarcinoma, and frequently has acquired EGFR mutations making it very responsive to EGFR-targeted therapies (1). A multi-step process involving genetic and epigenetic alterations resulting from DNA damage (usually from cigarette smoking) transforms normal lung epithelium into lung cancer and results in “field defects” in histologically normal lung epithelium, as well as a variety of histologic pre-neoplastic/pre-malignant lesions (3,4). The culmination of these changes lead to lung cancers exhibiting all six of the “hallmarks of cancer” (self-sufficiency of growth signals, insensitivity to growth-inhibitory (anti-growth) signals, evasion of programmed cell death (apoptosis), limitless replicative potential, sustained angiogenesis, and tissue invasion and metastasis; 5). Current information on the molecular steps and their timing in pre-neoplasia, primary cancer, and metastatic disease is the subject of this chapter (6). The identification and characterization of key molecular changes – often involving oncogenes and tumor suppressor genes (TSGs), and importantly, the associated “tumor cell vulnerabilities” that accompany these oncogenotype changes – in the development and progression of lung cancer are of fundamental importance for improving the prevention, early detection, and treatment of this disease. Ultimately these findings need to be translated to the clinic by using molecular alterations, such as biomarkers for early detection, targets for prevention, signatures for personalizing prognosis and therapy selection for each patient, and therapeutic targets to selectively kill or inhibit the growth of lung cancer.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 506 - 525
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Sun, S, Schiller, JH, Gazdar, AF. Lung cancer in never smokers – a different disease. Nature Reviews Cancer 2007;7:778–90.CrossRefGoogle ScholarPubMed
Scagliotti, GV, Longo, M, Novello, S. Non-small cell lung cancer in never smokers. Current Opinion in Oncology 2009;21:99–104.CrossRefGoogle ScholarPubMed
Wistuba, II, Gazdar, AF. Lung cancer preneoplasia. Annual Review of Pathology 2006;1:331–48.CrossRefGoogle ScholarPubMed
Sekido, Y, Fong, KM, Minna, JD. Progress in understanding the molecular pathogenesis of human lung cancer. Biochimica et Biophysica Acta 1998;1378:F21–59.Google ScholarPubMed
Hanahan, D, Weinberg, RA. The hallmarks of cancer. Cell 2000;100:57–70.CrossRefGoogle ScholarPubMed
Nowell, PC. The clonal evolution of tumor cell populations. Science 1976;194:23–8.CrossRefGoogle ScholarPubMed
Risch, A, Plass, C. Lung cancer epigenetics and genetics. International Journal of Cancer 2008;123:1–7.CrossRefGoogle ScholarPubMed
Herbst, RS, Heymach, JV, Lippman, SM. Lung cancer. New England Journal of Medicine 2008;359:1367–80.CrossRefGoogle ScholarPubMed
Amos, CI, Wu, X, Broderick, P, et al. Genome-wide association scan of tag SNPs identifies a susceptibility locus for lung cancer at 15q25.1. Nature Genetics 2008;40:616–22.CrossRefGoogle ScholarPubMed
Hung, RJ, McKay, JD, Gaborieau, V, et al. A susceptibility locus for lung cancer maps to nicotinic acetylcholine receptor subunit genes on 15q25. Nature 2008;452:633–7.CrossRefGoogle ScholarPubMed
Thorgeirsson, TE, Geller, F, Sulem, P, et al. A variant associated with nicotine dependence, lung cancer and peripheral arterial disease. Nature 2008;452:638–42.CrossRefGoogle ScholarPubMed
Le Marchand, L, Derby, KS, Murphy, SE, et al. Smokers with the CHRNA lung cancer-associated variants are exposed to higher levels of nicotine equivalents and a carcinogenic tobacco-specific nitrosamine. Cancer Research 2008;68:9137–40.CrossRefGoogle Scholar
Spitz, MR, Amos, CI, Dong, Q, Lin, J, Wu, X. The CHRNA5-A3 region on chromosome 15q24–25.1 is a risk factor both for nicotine dependence and for lung cancer. Journal of the National Cancer Institute 2008;100:1552–6.CrossRefGoogle ScholarPubMed
You, M, Wang, D, Liu, P, et al. Fine mapping of chromosome 6q23–25 region in familial lung cancer families reveals RGS17 as a likely candidate gene. Clinical Cancer Research 2009;15:2666–74.CrossRefGoogle ScholarPubMed
Albanell, J, Lonardo, F, Rusch, V, et al. High telomerase activity in primary lung cancers:association with increased cell proliferation rates and advanced pathologic stage. Journal of the National Cancer Institute 1997;89:1609–15.CrossRefGoogle ScholarPubMed
Hiyama, K, Hiyama, E, Ishioka, S, et al. Telomerase activity in small-cell and non-small-cell lung cancers. Journal of the National Cancer Institute 1995;87:895–902.CrossRefGoogle ScholarPubMed
Frias, C, Garcia-Aranda, C, De Juan, C, et al. Telomere shortening is associated with poor prognosis and telomerase activity correlates with DNA repair impairment in non-small cell lung cancer. Lung Cancer 2008;60:416–25.CrossRefGoogle ScholarPubMed
Kwei, KA, Kim, YH, Girard, L, et al. Genomic profiling identifies TITF1 as a lineage-specific oncogene amplified in lung cancer. Oncogene 2008;27:3635–40.CrossRefGoogle ScholarPubMed
Weir, BA, Woo, MS, Getz, G, et al. Characterizing the cancer genome in lung adenocarcinoma. Nature 2007;450:893–8.CrossRefGoogle ScholarPubMed
Merlino, GT, Xu, YH, Ishii, S, et al. Amplification and enhanced expression of the epidermal growth factor receptor gene in A431 human carcinoma cells. Science 1984;224:417–19.CrossRefGoogle ScholarPubMed
Lynch, TJ, Bell, DW, Sordella, R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. New England Journal of Medicine 2004;350:2129–39.CrossRefGoogle ScholarPubMed
Pao, W, Miller, V, Zakowski, M, et al. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proceedings of the National Academy of Sciences USA 2004;101:13 306–11.CrossRefGoogle ScholarPubMed
Paez, JG, Janne, PA, Lee, JC, et al. EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. Science 2004;304:1497–500.CrossRefGoogle ScholarPubMed
Massion, PP, Kuo, WL, Stokoe, D, et al. Genomic copy number analysis of non-small cell lung cancer using array comparative genomic hybridization: implications of the phosphatidylinositol 3-kinase pathway. Cancer Research 2002;62:3636–40.Google ScholarPubMed
Friend, SH, Bernards, R, Rogelj, S, et al. A human DNA segment with properties of the gene that predisposes to retinoblastoma and osteosarcoma. Nature 1986;323:643–6.CrossRefGoogle ScholarPubMed
Nobori, T, Miura, K, Wu, DJ, et al. Deletions of the cyclin-dependent kinase-4 inhibitor gene in multiple human cancers. Nature 1994;368:753–6.CrossRefGoogle ScholarPubMed
Steck, PA, Pershouse, MA, Jasser, SA, et al. Identification of a candidate tumour suppressor gene, MMAC1, at chromosome 10q23.3 that is mutated in multiple advanced cancers. Nature Genetics 1997;15:356–62.CrossRefGoogle ScholarPubMed
Weinstein, IB. Cancer. Addiction to oncogenes: the Achilles heal of cancer. Science 2002;297:63–4.CrossRefGoogle Scholar
Normanno, N, Bianco, C, Strizzi, L, et al. The ERBB receptors and their ligands in cancer:an overview. Current Drug Targets 2005;6:243–57.CrossRefGoogle ScholarPubMed
Rusch, V, Baselga, J, Cordon-Cardo, C, et al. Differential expression of the epidermal growth factor receptor and its ligands in primary non-small cell lung cancers and adjacent benign lung. Cancer Research 1993;53:2379–85.Google ScholarPubMed
Franklin, WA, Veve, R, Hirsch, FR, Helfrich, BA, Bunn, PA, Jr. Epidermal growth factor receptor family in lung cancer and premalignancy. Seminars on Oncology 2002;29:3–14.CrossRefGoogle ScholarPubMed
Herbst, RS. Review of epidermal growth factor receptor biology. International Journal of Radiation Oncology, Biology, Physics 2004;59:21–6.CrossRefGoogle ScholarPubMed
Fujino, S, Enokibori, T, Tezuka, N, et al. A comparison of epidermal growth factor receptor levels and other prognostic parameters in non-small cell lung cancer. European Journal of Cancer 1996;32A:2070–4.CrossRefGoogle ScholarPubMed
Eberhard, DA, Johnson, BE, Amler, LC, et al. Mutations in the epidermal growth factor receptor and in KRAS are predictive and prognostic indicators in patients with non-small-cell lung cancer treated with chemotherapy alone and in combination with erlotinib. Journal of Clinical Oncology 2005;23:5900–9.CrossRefGoogle ScholarPubMed
Gazdar, AF, Shigematsu, H, Herz, J, Minna, JD. Mutations and addiction to EGFR:the Achilles ‘heal’ of lung cancers? Trends Molecular Medicine 2004;10:481–6.CrossRefGoogle ScholarPubMed
Pao, W, Miller, VA, Politi, KA, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Medicine 2005;2:e73.Google ScholarPubMed
Pao, W, Wang, TY, Riely, GJ, et al. KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib. PLoS Medicine 2005;2:e17.Google ScholarPubMed
Shigematsu, H, Lin, L, Takahashi, T, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. Journal of the National Cancer Institute 2005;97:339–46.CrossRefGoogle ScholarPubMed
Sordella, R, Bell, DW, Haber, DA, Settleman, J. Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science 2004;305:1163–7.CrossRefGoogle ScholarPubMed
Bean, J, Brennan, C, Shih, JY, et al. MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib. Proceedings of the National Academy of Sciences USA 2007;104:20 932–7.CrossRefGoogle ScholarPubMed
Engelman, JA, Zejnullahu, K, Mitsudomi, T, et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007;316:1039–43.CrossRefGoogle ScholarPubMed
Gao, SP, Mark, KG, Leslie, K, et al. Mutations in the EGFR kinase domain mediate STAT3 activation via IL-6 production in human lung adenocarcinomas. Journal of Clinical Investigation 2007;117:3846–56.CrossRefGoogle ScholarPubMed
Ishihara, K, Hirano, T. Molecular basis of the cell specificity of cytokine action. Biochimica et Biophysica Acta 2002;1592:281–96.CrossRefGoogle ScholarPubMed
Hong, DS, Angelo, LS, Kurzrock, R. Interleukin-6 and its receptor in cancer:implications for Translational Therapeutics. Cancer 2007;110:1911–28.CrossRefGoogle ScholarPubMed
Riely, GJ, Kris, MG, Rosenbaum, D, et al. Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clinical Cancer Research 2008;14:5731–4.CrossRefGoogle ScholarPubMed
Singh, A, Greninger, P, Rhodes, D, et al. A gene expression signature associated with “K-Ras addiction” reveals regulators of EMT and tumor cell survival. Cancer Cell 2009;15:489–500.CrossRefGoogle ScholarPubMed
Meyerson, M. Cancer:broken genes in solid tumours. Nature 2007;448:545–6.CrossRefGoogle ScholarPubMed
Inamura, K, Takeuchi, K, Togashi, Y, et al. EML4-ALK fusion is linked to histological characteristics in a subset of lung cancers. Journal of Thoracic Oncology 2008;3:13–17.CrossRefGoogle Scholar
Soda, M, Choi, YL, Enomoto, M, et al. Identification of the transforming EML4-ALK fusion gene in non-small-cell lung cancer. Nature 2007;448:561–6.CrossRefGoogle ScholarPubMed
Mano, H. Non-solid oncogenes in solid tumors:EML4-ALK fusion genes in lung cancer. Cancer Science 2008;99:2349–55.CrossRef
Koivunen, JP, Mermel, C, Zejnullahu, K, et al. EML4-ALK fusion gene and efficacy of an ALK kinase inhibitor in lung cancer. Clinical Cancer Research 2008;14:4275–83.CrossRefGoogle ScholarPubMed
Shaw, AT, Yeap, BY, Mino-Kenudson, M, et al. Clinical features and outcome of patients with non-small-cell lung cancer who harbor EML4-ALK. Journal of Clinical Oncology 2009;27:4247–53.CrossRefGoogle ScholarPubMed
Horn, L, Pao, W. EML4-ALK:honing in on a new target in non-small-cell lung cancer. Journal of Clinical Oncology 2009;27:4232–5.CrossRefGoogle ScholarPubMed
Inamura, K, Takeuchi, K, Togashi, Y, et al. EML4-ALK lung cancers are characterized by rare other mutations, a TTF-1 cell lineage, an acinar histology, and young onset. Modern Pathology 2009;22:508–15.CrossRefGoogle ScholarPubMed
Wong, DW, Leung, EL, So, KK, et al. The EML4-ALK fusion gene is involved in various histologic types of lung cancers from nonsmokers with wild-type EGFR and KRAS. Cancer 2009;115:1723–33.CrossRefGoogle ScholarPubMed
Martelli, MP, Sozzi, G, Hernandez, L, et al. EML4-ALK rearrangement in non-small cell lung cancer and non-tumor lung tissues. American Journal of Pathology 2009;174:661–70.CrossRefGoogle ScholarPubMed
West, KA, Linnoila, IR, Belinsky, SA, Harris, CC, Dennis, PA. Tobacco carcinogen-induced cellular transformation increases activation of the phosphatidylinositol 3ʹ-kinase/Akt pathway in vitro and in vivo. Cancer Research 2004;64:446–51.CrossRefGoogle ScholarPubMed
Kendall, J, Liu, Q, Bakleh, A, et al. Oncogenic cooperation and coamplification of developmental transcription factor genes in lung cancer. Proceedings of the National Academy of Sciences USA 2007;104:16 663–8.CrossRefGoogle ScholarPubMed
Bass, AJ, Watanabe, H, Mermel, CH, et al. SOX2 is an amplified lineage-survival oncogene in lung and esophageal squamous cell carcinomas. Nature Genetics 2009;41:1238–42.CrossRefGoogle ScholarPubMed
Bingle, CD. Thyroid transcription factor-1. International Journal of Biochemistry and Cell Biology 1997;29:1471–3.CrossRefGoogle ScholarPubMed
Ikeda, K, Clark, JC, Shaw-White, JR, et al. Gene structure and expression of human thyroid transcription factor-1 in respiratory epithelial cells. Journal of Biological Chemistry 1995;270:8108–14.CrossRefGoogle ScholarPubMed
Visakorpi, T, Hyytinen, E, Koivisto, P, et al. In vivo amplification of the androgen receptor gene and progression of human prostate cancer. Nature Genetics 1995;9:401–6.CrossRefGoogle ScholarPubMed
Garraway, LA, Widlund, HR, Rubin, MA, et al. Integrative genomic analyses identify MITF as a lineage survival oncogene amplified in malignant melanoma. Nature 2005;436:117–22.CrossRefGoogle ScholarPubMed
Holst, F, Stahl, PR, Ruiz, C, et al. Estrogen receptor alpha (ESR1) gene amplification is frequent in breast cancer. Nature Genetics 2007;39:655–60.CrossRefGoogle ScholarPubMed
Garraway, LA, Sellers, WR. Lineage dependency and lineage-survival oncogenes in human cancer. Nature Reviews Cancer 2006;6:593–602.CrossRefGoogle ScholarPubMed
Knudson, AG, Jr. The ninth Gordon Hamilton-Fairley memorial lecture. Hereditary cancers: clues to mechanisms of carcinogenesis. British Journal of Cancer 1989;59:661–6.CrossRefGoogle ScholarPubMed
Breuer, RH, Postmus, PE, Smit, EF. Molecular pathology of non-small-cell lung cancer. Respiration 2005;72:313–30.CrossRefGoogle ScholarPubMed
Takahashi, T, Nau, MM, Chiba, I, et al. p53:a frequent target for genetic abnormalities in lung cancer. Science 1989;246:491–4.CrossRefGoogle ScholarPubMed
Hollstein, M, Sidransky, D, Vogelstein, B, Harris, CC. p53 mutations in human cancers. Science 1991;253:49–53.CrossRefGoogle ScholarPubMed
Greenblatt, MS, Bennett, WP, Hollstein, M, Harris, CC. Mutations in the p53 tumor suppressor gene:clues to cancer etiology and molecular pathogenesis. Cancer Research 1994;54:4855–78.Google ScholarPubMed
van Oijen, MG, Slootweg, PJ. Gain-of-function mutations in the tumor suppressor gene p53. Clinical Cancer Research 2000;6:2138–45.Google ScholarPubMed
Ventura, A, Kirsch, DG, McLaughlin, ME, et al. Restoration of p53 function leads to tumour regression in vivo. Nature 2007;445:661–5.CrossRefGoogle ScholarPubMed
Harbour, JW, Lai, SL, Whang-Peng, J, et al. Abnormalities in structure and expression of the human retinoblastoma gene in SCLC. Science 1988;241:353–7.CrossRefGoogle ScholarPubMed
Yokota, J, Mori, N, Akiyama, T, et al. Multiple genetic alterations in small-cell lung carcinoma. Princess Takamatsu Symposia 1989;20:43–8.Google ScholarPubMed
Otterson, GA, Kratzke, RA, Coxon, A, Kim, YW, Kaye, FJ. Absence of p16INK4 protein is restricted to the subset of lung cancer lines that retains wildtype RB. Oncogene 1994;9:3375–8.Google ScholarPubMed
Wistuba, II, Behrens, C, Virmani, AK, et al. High resolution chromosome 3p allelotyping of human lung cancer and preneoplastic/preinvasive bronchial epithelium reveals multiple, discontinuous sites of 3p allele loss and three regions of frequent breakpoints. Cancer Research 2000;60:1949–60.Google ScholarPubMed
Ito, M, Ito, G, Kondo, M, et al. Frequent inactivation of RASSF1A, BLU, and SEMA3B on 3p21.3 by promoter hypermethylation and allele loss in non-small cell lung cancer. Cancer Letters 2005;225:131–9.CrossRefGoogle ScholarPubMed
Kuroki, T, Trapasso, F, Yendamuri, S, et al. Allelic loss on chromosome 3p21.3 and promoter hypermethylation of semaphorin 3B in non-small cell lung cancer. Cancer Research 2003;63:3352–5.Google ScholarPubMed
Feng, Q, Hawes, SE, Stern, JE, et al. DNA methylation in tumor and matched normal tissues from non-small cell lung cancer patients. Cancer Epidemiology, Biomarkers and Prevention 2008;17:645–54.CrossRefGoogle ScholarPubMed
Wistuba, II, Gazdar, AF, Minna, JD. Molecular genetics of small cell lung carcinoma. Seminars on Oncology 2001;28:3–13.CrossRefGoogle ScholarPubMed
Zochbauer-Muller, S, Minna, JD, Gazdar, AF. Aberrant DNA methylation in lung cancer:biological and clinical implications. Oncologist 2002;7:451–7.CrossRefGoogle ScholarPubMed
Siprashvili, Z, Sozzi, G, Barnes, LD, et al. Replacement of Fhit in cancer cells suppresses tumorigenicity. Proceedings of the National Academy of Sciences USA 1997;94:13 771–6.CrossRefGoogle ScholarPubMed
Agathanggelou, A, Cooper, WN, Latif, F. Role of the Ras-association domain family 1 tumor suppressor gene in human cancers. Cancer Research 2005;65:3497–508.CrossRefGoogle ScholarPubMed
Ji, L, Roth, JA. Tumor suppressor FUS1 signaling pathway. Journal of Thoracic Oncology 2008;3:327–30.CrossRefGoogle ScholarPubMed
Deng, WG, Kawashima, H, Wu, G, et al. Synergistic tumor suppression by coexpression of FUS1 and p53 is associated with down-regulation of murine double minute-2 and activation of the apoptotic protease-activating factor 1-dependent apoptotic pathway in human non-small cell lung cancer cells. Cancer Research 2007;67:709–17.CrossRefGoogle ScholarPubMed
Tomizawa, Y, Sekido, Y, Kondo, M, et al. Inhibition of lung cancer cell growth and induction of apoptosis after reexpression of 3p21.3 candidate tumor suppressor gene SEMA3B. Proceedings of the National Academy of Sciences USA 2001;98:13 954–9.CrossRefGoogle ScholarPubMed
Ochi, K, Mori, T, Toyama, Y, Nakamura, Y, Arakawa, H. Identification of semaphorin3B as a direct target of p53. Neoplasia 2002;4:82–7.CrossRefGoogle ScholarPubMed
Castro-Rivera, E, Ran, S, Brekken, RA, Minna, JD. Semaphorin 3B inhibits the phosphatidylinositol 3-kinase/Akt pathway through neuropilin-1 in lung and breast cancer cells. Cancer Research 2008;68:8295–303.CrossRefGoogle ScholarPubMed
Brambilla, E, Constantin, B, Drabkin, H, Roche, J. Semaphorin SEMA3F localization in malignant human lung and cell lines: a suggested role in cell adhesion and cell migration. American Journal of Pathology 2000;156:939–50.CrossRefGoogle ScholarPubMed
Kessler, O, Shraga-Heled, N, Lange, T, et al. Semaphorin-3F is an inhibitor of tumor angiogenesis. Cancer Research 2004;64:1008–15.CrossRefGoogle ScholarPubMed
Alessi, DR, Sakamoto, K, Bayascas, JR. LKB1-dependent signaling pathways. Annual Review of Biochemistry 2006;75:137–63.CrossRefGoogle ScholarPubMed
Hemminki, A, Markie, D, Tomlinson, I, et al. A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature 1998;391:184–7.CrossRefGoogle ScholarPubMed
Matsumoto, S, Iwakawa, R, Takahashi, K, et al. Prevalence and specificity of LKB1 genetic alterations in lung cancers. Oncogene 2007;26:5911–18.CrossRefGoogle ScholarPubMed
Shaw, RJ, Bardeesy, N, Manning, BD, et al. The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell 2004;6:91–9.CrossRefGoogle ScholarPubMed
Onozato, R, Kosaka, T, Achiwa, H, et al. LKB1 gene mutations in Japanese lung cancer patients. Cancer Science 2007;98:1747–51.CrossRefGoogle ScholarPubMed
Sanchez-Cespedes, M, Parrella, P, Esteller, M, et al. Inactivation of LKB1/STK11 is a common event in adenocarcinomas of the lung. Cancer Research 2002;62:3659–62.Google ScholarPubMed
Carretero, J, Medina, PP, Pio, R, Montuenga, LM, Sanchez-Cespedes, M. Novel and natural knockout lung cancer cell lines for the LKB1/STK11 tumor suppressor gene. Oncogene 2004;23:4037–40.CrossRefGoogle ScholarPubMed
Mahoney, CL, Choudhury, B, Davies, H, et al. LKB1/KRAS mutant lung cancers constitute a genetic subset of NSCLC with increased sensitivity to MAPK and mTOR signalling inhibition. British Journal of Cancer 2009;100:370–5.CrossRefGoogle ScholarPubMed
Bird, A.DNA methylation patterns and epigenetic memory. Genes and Development 2002;16:6–21.CrossRefGoogle ScholarPubMed
Baylin, SB, Esteller, M, Rountree, MR, et al. Aberrant patterns of DNA methylation, chromatin formation and gene expression in cancer. Human Molecular Genetics 2001;10:687–92.CrossRefGoogle Scholar
Rauch, TA, Zhong, X, Wu, X, et al. High-resolution mapping of DNA hypermethylation and hypomethylation in lung cancer. Proceedings of the National Academy of Sciences USA 2008;105:252–7.CrossRefGoogle ScholarPubMed
Shames, DS, Girard, L, Gao, B, et al. A genome-wide screen for promoter methylation in lung cancer identifies novel methylation markers for multiple malignancies. PLoS Medicine 2006;3:e486.CrossRefGoogle ScholarPubMed
Zhong, S, Fields, CR, Su, N, Pan, YX, Robertson, KD. Pharmacologic inhibition of epigenetic modifications, coupled with gene expression profiling, reveals novel targets of aberrant DNA methylation and histone deacetylation in lung cancer. Oncogene 2007;26:2621–34.CrossRefGoogle ScholarPubMed
Brena, RM, Morrison, C, Liyanarachchi, S, et al. Aberrant DNA methylation of OLIG1, a novel prognostic factor in non-small cell lung cancer. PLoS Medicine 2007;4:e108.CrossRefGoogle ScholarPubMed
Pfeifer, GP, Rauch, TA. DNA methylation patterns in lung carcinomas. Seminars in Cancer Biology 2009;19:181–7.CrossRefGoogle ScholarPubMed
Metzler, M, Wilda, M, Busch, K, Viehmann, S, Borkhardt, A. High expression of precursor microRNA-155/BIC RNA in children with Burkitt lymphoma. Genes, Chromosomes and Cancer 2004;39:167–9.CrossRefGoogle ScholarPubMed
Michael, MZ, van Holst Pellekaan, NG, Young, GP, James, RJ. Reduced accumulation of specific microRNAs in colorectal neoplasia. Molecular Cancer Research 2003;1:882–91.Google ScholarPubMed
Calin, GA, Dumitru, CD, Shimizu, M, et al. Frequent deletions and down-regulation of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. Proceedings of the National Academy of Sciences USA 2002;99:15 524–9.CrossRefGoogle ScholarPubMed
Eis, PS, Tam, W, Sun, L, et al. Accumulation of miR-155 and BIC RNA in human B cell lymphomas. Proceedings of the National Academy of Sciences USA 2005;102:3627–32.CrossRefGoogle ScholarPubMed
He, L, Thomson, JM, Hemann, MT, et al. A microRNA polycistron as a potential human oncogene. Nature 2005;435:828–33.CrossRefGoogle ScholarPubMed
Ota, A, Tagawa, H, Karnan, S, et al. Identification and characterization of a novel gene, C13orf25, as a target for 13q31-q32 amplification in malignant lymphoma. Cancer Research 2004;64:3087–95.CrossRefGoogle ScholarPubMed
Voorhoeve, PM, le Sage, C, Schrier, M, et al. A genetic screen implicates miRNA-372 and miRNA-373 as oncogenes in testicular germ cell tumors. Cell 2006;124:1169–81.CrossRefGoogle ScholarPubMed
Takamizawa, J, Konishi, H, Yanagisawa, K, et al. Reduced expression of the let-7 microRNAs in human lung cancers in association with shortened postoperative survival. Cancer Research 2004;64:3753–6.CrossRefGoogle ScholarPubMed
Hayashita, Y, Osada, H, Tatematsu, Y, et al. A polycistronic microRNA cluster, miR-17–92, is overexpressed in human lung cancers and enhances cell proliferation. Cancer Research 2005;65:9628–32.CrossRefGoogle ScholarPubMed
Yu, SL, Chen, HY, Chang, GC, et al. MicroRNA signature predicts survival and relapse in lung cancer. Cancer Cell 2008;13:48–57.CrossRefGoogle ScholarPubMed
Yanaihara, N, Caplen, N, Bowman, E, et al. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell 2006;9:189–98.CrossRefGoogle ScholarPubMed
Fabbri, M, Garzon, R, Cimmino, A, et al. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proceedings of the National Academy of Sciences USA 2007;104:15 805–10.CrossRefGoogle ScholarPubMed
Crawford, M, Brawner, E, Batte, K, et al. MicroRNA-126 inhibits invasion in non-small cell lung carcinoma cell lines. Biochemical and Biophysical Research Communications 2008;373:607–12.CrossRefGoogle ScholarPubMed
Nasser, MW, Datta, J, Nuovo, G, et al. Downregulation of microRNA-1 (miR-1) in lung cancer: Suppression of tumorigenic property of lung cancer cells and their sensitization to doxorubicin induced apoptosis bymiR-1. Journal of Biological Chemistry 2008;283:33 394–405.CrossRefGoogle Scholar
Weiss, GJ, Bemis, LT, Nakajima, E, et al. EGFR regulation by microRNA in lung cancer: correlation with clinical response and survival to gefitinib and EGFR expression in cell lines. Annals of Oncology 2008;19:1053–9.CrossRefGoogle ScholarPubMed
Markou, A, Tsaroucha, EG, Kaklamanis, L, et al. Prognostic value of mature microRNA-21 and microRNA-205 overexpression in non-small cell lung cancer by quantitative real-time RT-PCR. Clinical Chemistry 2008;54:1696–704.CrossRefGoogle ScholarPubMed
Garofalo, M, Quintavalle, C, Di Leva, G, et al. MicroRNA signatures of TRAIL resistance in human non-small cell lung cancer. Oncogene 2008;27:3845–55.CrossRefGoogle ScholarPubMed
Volinia, S, Calin, GA, Liu, CG, et al. A microRNA expression signature of human solid tumors defines cancer gene targets. Proceedings of the National Academy of Sciences USA 2006;103:2257–61.CrossRefGoogle ScholarPubMed
Raponi, M, Dossey, L, Jatkoe, T, et al. MicroRNA classifiers for predicting prognosis of squamous cell lung cancer. Cancer Research 2009;69:5776–83.CrossRefGoogle ScholarPubMed
Seike, M, Goto, A, Okano, T, et al. MiR-21 is an EGFR-regulated anti-apoptotic factor in lung cancer in never-smokers. Proceedings of the National Academy of Sciences USA 2009;106:12 085–90.CrossRefGoogle ScholarPubMed
Du, L, Schageman, JJ, Subauste, MC, et al. miR-93, miR-98, and miR-197 regulate expression of tumor suppressor gene FUS1. Molecular Cancer Research 2009;7:1234–43.CrossRefGoogle ScholarPubMed
Johnson, SM, Grosshans, H, Shingara, J, et al. RAS is regulated by the let-7 microRNA family. Cell 2005;120:635–47.CrossRefGoogle ScholarPubMed
Lee, YS, Dutta, A. The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. Genes and Development 2007;21:1025–30.CrossRefGoogle ScholarPubMed
Mayr, C, Hemann, MT, Bartel, DP. Disrupting the pairing between let-7 and Hmga2 enhances oncogenic transformation. Science 2007;315:1576–9.CrossRefGoogle ScholarPubMed
Johnson, CD, Esquela-Kerscher, A, Stefani, G, et al. The let-7 microRNA represses cell proliferation pathways in human cells. Cancer Research 2007;67:7713–22.CrossRefGoogle ScholarPubMed
Esquela-Kerscher, A, Trang, P, Wiggins, JF, et al. The let-7 microRNA reduces tumor growth in mouse models of lung cancer. Cell Cycle 2008;7:759–64.CrossRefGoogle ScholarPubMed
Kumar, MS, Erkeland, SJ, Pester, RE, et al. Suppression of non-small cell lung tumor development by the let-7 microRNA family. Proceedings of the National Academy of Sciences USA 2008;105:3903–8.CrossRefGoogle ScholarPubMed
Bertolini, G, Roz, L, Perego, P, et al. Highly tumorigenic lung cancer CD133+ cells display stem-like features and are spared by cisplatin treatment. Proceedings of the National Academy of Sciences USA 2009;106:16 281–6.CrossRefGoogle ScholarPubMed
Tirino, V, Camerlingo, R, Franco, R, et al. The role of CD133 in the identification and characterisation of tumour-initiating cells in non-small-cell lung cancer. European Journal of Cardiothoracic Surgery 2009;36:446–53.CrossRefGoogle ScholarPubMed
Chen, YC, Hsu, HS, Chen, YW, et al. Oct-4 expression maintained cancer stem-like properties in lung cancer-derived CD133-positive cells. PLoS One 2008;3:e2637.CrossRefGoogle ScholarPubMed
Jiang, F, Qiu, Q, Khanna, A, et al. Aldehyde dehydrogenase 1 is a tumor stem cell-associated marker in lung cancer. Molecular Cancer Research 2009;7:330–8.CrossRefGoogle ScholarPubMed
Kitamura, H, Okudela, K, Yazawa, T, Sato, H, Shimoyamada, H. Cancer stem cell:Implications in cancer biology and therapy with special reference to lung cancer. Lung Cancer 2009;66:275–81.CrossRefGoogle ScholarPubMed
Ho, MM, Ng, AV, Lam, S, Hung, JY. Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer Research 2007;67:4827–33.CrossRefGoogle ScholarPubMed
Eramo, A, Lotti, F, Sette, G, et al. Identification and expansion of the tumorigenic lung cancer stem cell population. Cell Death and Differentiation 2008;15:504–14.CrossRefGoogle ScholarPubMed
Watkins, DN, Berman, DM, Burkholder, SG, et al. Hedgehog signalling within airway epithelial progenitors and in small-cell lung cancer. Nature 2003;422:313–17.CrossRefGoogle ScholarPubMed
Watkins, DN, Berman, DM, Baylin, SB. Hedgehog signaling:progenitor phenotype in small-cell lung cancer. Cell Cycle 2003;2:196–8.CrossRefGoogle ScholarPubMed
Daniel, VC, Peacock, CD, Watkins, DN. Developmental signalling pathways in lung cancer. Respirology 2006;11:234–40.CrossRefGoogle ScholarPubMed
Olsen, CL, Hsu, PP, Glienke, J, Rubanyi, GM, Brooks, AR. Hedgehog-interacting protein is highly expressed in endothelial cells but down-regulated during angiogenesis and in several human tumors. BMC Cancer 2004;4:43.CrossRefGoogle ScholarPubMed
Nickoloff, BJ, Osborne, BA, Miele, L. Notch signaling as a therapeutic target in cancer:a new approach to the development of cell fate modifying agents. Oncogene 2003;22:6598–608.CrossRefGoogle Scholar
Klein, F, Kotb, WF, Petersen, I. Incidence of human papilloma virus in lung cancer. Lung Cancer 2008;65:13–18.CrossRefGoogle ScholarPubMed
Werness, BA, Levine, AJ, Howley, PM. Association of human papillomavirus types 16 and 18 E6 proteins with p53. Science 1990;248:76–9.CrossRefGoogle ScholarPubMed
Dyson, N, Howley, PM, Munger, K, Harlow, E. The human papilloma virus-16 E7 oncoprotein is able to bind to the retinoblastoma gene product. Science 1989;243:934–7.CrossRefGoogle ScholarPubMed
Beer, DG, Kardia, SL, Huang, CC, et al. Gene-expression profiles predict survival of patients with lung adenocarcinoma. Nature Medicine 2002;8:816–24.CrossRefGoogle ScholarPubMed
Bild, AH, Yao, G, Chang, JT, et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 2006;439:353–7.CrossRefGoogle ScholarPubMed
Potti, A, Dressman, HK, Bild, A, et al. Genomic signatures to guide the use of chemotherapeutics. Nature Medicine 2006;12:1294–300.CrossRefGoogle ScholarPubMed
Shedden, K, Taylor, JM, Enkemann, SA, et al. Gene expression-based survival prediction in lung adenocarcinoma: a multi-site, blinded validation study. Nature Medicine 2008;14:822–7.CrossRefGoogle ScholarPubMed
Anguiano, A, Nevins, JR, Potti, A. Toward the individualization of lung cancer therapy. Cancer 2008;113:1760–7.CrossRefGoogle ScholarPubMed
Xie, Y, Minna, JD. Predicting the future for people with lung cancer. Nature Medicine 2008;14:812–13.CrossRefGoogle ScholarPubMed
Tonon, G, Wong, KK, Maulik, G, et al. High-resolution genomic profiles of human lung cancer. Proceedings of the National Academy of Sciences USA 2005;102:9625–30.CrossRefGoogle ScholarPubMed
Zhao, X, Weir, BA, LaFramboise, T, et al. Homozygous deletions and chromosome amplifications in human lung carcinomas revealed by single nucleotide polymorphism array analysis. Cancer Research 2005;65:5561–70.CrossRefGoogle ScholarPubMed
Shibata, T, Uryu, S, Kokubu, A, et al. Genetic classification of lung adenocarcinoma based on array-based comparative genomic hybridization analysis:its association with clinicopathologic features. Clinical Cancer Research 2005;11:6177–85.CrossRefGoogle ScholarPubMed
Ding, L, Getz, G, Wheeler, DA, et al. Somatic mutations affect key pathways in lung adenocarcinoma. Nature 2008;455:1069–75.CrossRefGoogle ScholarPubMed
Whitehurst, AW, Bodemann, BO, Cardenas, J, et al. Synthetic lethal screen identification of chemosensitizer loci in cancer cells. Nature 2007;446:815–19.CrossRefGoogle ScholarPubMed
Luo, J, Emanuele, MJ, Li, D, et al. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell 2009;137:835–48.CrossRefGoogle ScholarPubMed
Scholl, C, Frohling, S, Dunn, IF, et al. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell 2009;137:821–34.CrossRefGoogle ScholarPubMed
Guo, W, Wu, S, Liu, J, Fang, B. Identification of a small molecule with synthetic lethality for K-ras and protein kinase C iota. Cancer Research 2008;68:7403–8.CrossRefGoogle ScholarPubMed
Duex, JE, Sorkin, A. RNA interference screen identifies Usp18 as a regulator of epidermal growth factor receptor synthesis. Molecular Biology of the Cell 2009;20:1833–44.CrossRefGoogle ScholarPubMed
Yamanaka, S, Gu, Z, Sato, M, et al. siRNA targeting against EGFR, a promising candidate for a novel therapeutic application to lung adenocarcinoma. Pathobiology 2008;75:2–8.CrossRefGoogle ScholarPubMed
Arteaga, CL. Overview of epidermal growth factor receptor biology and its role as a therapeutic target in human neoplasia. Seminars on Oncology 2002;29:3–9.CrossRefGoogle ScholarPubMed
Sekido, Y, Obata, Y, Ueda, R, et al. Preferential expression of c-kit protooncogene transcripts in small cell lung cancer. Cancer Research 1991;51:2416–19.Google ScholarPubMed
Boldrini, L, Ursino, S, Gisfredi, S, et al. Expression and mutational status of c-kit in small-cell lung cancer:prognostic relevance. Clinical Cancer Research 2004;10:4101–8.CrossRefGoogle ScholarPubMed
Kobayashi, S, Boggon, TJ, Dayaram, T, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. New England Journal of Medicine 2005;352:786–92.CrossRefGoogle ScholarPubMed
Balak, MN, Gong, Y, Riely, GJ, et al. Novel D761Y and common secondary T790M mutations in epidermal growth factor receptor-mutant lung adenocarcinomas with acquired resistance to kinase inhibitors. Clinical Cancer Research 2006;12:6494–501.CrossRefGoogle ScholarPubMed
Kosaka, T, Yatabe, Y, Endoh, H, et al. Analysis of epidermal growth factor receptor gene mutation in patients with non-small cell lung cancer and acquired resistance to gefitinib. Clinical Cancer Research 2006;12:5764–9.CrossRefGoogle ScholarPubMed
Kwak, EL, Sordella, R, Bell, DW, et al. Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib. Proceedings of the National Academy of Sciences USA 2005;102:7665–70.CrossRefGoogle ScholarPubMed
Kosaka, T, Yatabe, Y, Endoh, H, et al. Mutations of the epidermal growth factor receptor gene in lung cancer:biological and clinical implications. Cancer Research 2004;64:8919–23.CrossRefGoogle ScholarPubMed
Mitsudomi, T, Kosaka, T, Yatabe, Y. Biological and clinical implications of EGFR mutations in lung cancer. International Journal of Clinical Oncology 2006;11:190–8.CrossRefGoogle ScholarPubMed
Gazdar, AF.Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. Oncogene 2009;28:S24–31.CrossRefGoogle ScholarPubMed
Mascaux, C, Iannino, N, Martin, B, et al. The role of RAS oncogene in survival of patients with lung cancer:a systematic review of the literature with meta-analysis. British Journal of Cancer 2005;92:131–9.CrossRefGoogle ScholarPubMed
Shields, JM, Pruitt, K, McFall, A, Shaub, A, Der, CJ. Understanding Ras :’it ain't over ‘til it's over’. Trends in Cell Biology 2000;10:147–54.CrossRefGoogle ScholarPubMed
Rodenhuis, S, Slebos, RJ. Clinical significance of ras oncogene activation in human lung cancer. Cancer Research 1992;52:2665s–9s.Google ScholarPubMed
Ji, H, Wang, Z, Perera, SA, et al. Mutations in BRAF and KRAS converge on activation of the mitogen-activated protein kinase pathway in lung cancer mouse models. Cancer Research 2007;67:4933–9.CrossRefGoogle ScholarPubMed
Adhikary, S, Eilers, M. Transcriptional regulation and transformation by Myc proteins. Nature Reviews Molecular Cell Biology 2005;6:635–45.CrossRefGoogle ScholarPubMed
Meyer, N, Penn, LZ. Reflecting on 25 years with MYC. Nature Reviews Cancer 2008;8:976–90.CrossRefGoogle ScholarPubMed
Nau, MM, Brooks, BJ, Jr., Carney, DN, et al. Human small-cell lung cancers show amplification and expression of the N-myc gene. Proceedings of the National Academy of Sciences USA 1986;83:1092–6.CrossRefGoogle ScholarPubMed
Broers, JL, Viallet, J, Jensen, SM, et al. Expression of c-myc in progenitor cells of the bronchopulmonary epithelium and in a large number of non-small cell lung cancers. American Journal of Respiratory Cell and Molecular Biology 1993;9:33–43.CrossRefGoogle Scholar
Fong, KM, Sekido, Y, Minna, JD. Molecular pathogenesis of lung cancer. Journal of Thoracic and Cardiovascular Surgery 1999;118:1136–52.CrossRefGoogle ScholarPubMed
Vivanco, I, Sawyers, CL. The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nature Reviews Cancer 2002;2:489–501.CrossRefGoogle ScholarPubMed
Kawano, O, Sasaki, H, Endo, K, et al. PIK3CA mutation status in Japanese lung cancer patients. Lung Cancer 2006;54:209–15.CrossRefGoogle ScholarPubMed
Garnis, C, Lockwood, WW, Vucic, E, et al. High resolution analysis of non-small cell lung cancer cell lines by whole genome tiling path array CGH. International Journal of Cancer 2006;118:1556–64.CrossRefGoogle ScholarPubMed
Yamamoto, H, Shigematsu, H, Nomura, M, et al. PIK3CA mutations and copy number gains in human lung cancers. Cancer Research 2008;68:6913–21.CrossRefGoogle ScholarPubMed
Guertin, DA, Sabatini, DM. Defining the role of mTOR in cancer. Cancer Cell 2007;12:9–22.CrossRefGoogle Scholar
Maehama, T, Dixon, JE. The tumor suppressor, PTEN/MMAC1, dephosphorylates the lipid second messenger, phosphatidylinositol 3,4,5-trisphosphate. Journal of Biological Chemistry 1998;273:13 375–8.CrossRefGoogle ScholarPubMed
Soria, JC, Lee, HY, Lee, JI, et al. Lack of PTEN expression in non-small cell lung cancer could be related to promoter methylation. Clinical Cancer Research 2002;8:1178–84.Google ScholarPubMed
Marsit, CJ, Zheng, S, Aldape, K, et al. PTEN expression in non-small-cell lung cancer:evaluating its relation to tumor characteristics, allelic loss, and epigenetic alteration. Human Pathology 2005;36:768–76.CrossRefGoogle ScholarPubMed
Higashiyama, M, Doi, O, Kodama, K, et al. MDM2 gene amplification and expression in non-small-cell lung cancer:immunohistochemical expression of its protein is a favourable prognostic marker in patients without p53 protein accumulation. British Journal of Cancer 1997;75:1302–8.CrossRefGoogle ScholarPubMed
Gazzeri, S, Della Valle, V, Chaussade, L, et al. The human p19ARF protein encoded by the beta transcript of the p16INK4a gene is frequently lost in small cell lung cancer. Cancer Research 1998;58:3926–31.Google ScholarPubMed
Vonlanthen, S, Heighway, J, Tschan, MP, et al. Expression of p16INK4a/p16alpha and p19ARF/p16beta is frequently altered in non-small cell lung cancer and correlates with p53 overexpression. Oncogene 1998;17:2779–85.CrossRefGoogle ScholarPubMed
Ohtani, N, Yamakoshi, K, Takahashi, A, Hara, E. The p16INK4a-RB pathway:molecular link between cellular senescence and tumor suppression. Journal of Medical Investigation 2004;51:146–53.CrossRefGoogle ScholarPubMed
Reissmann, PT, Koga, H, Takahashi, R, et al. Inactivation of the retinoblastoma susceptibility gene in non-small-cell lung cancer. The Lung Cancer Study Group. Oncogene 1993;8:1913–19.Google ScholarPubMed
Merlo, A, Gabrielson, E, Askin, F, Sidransky, D. Frequent loss of chromosome 9 in human primary non-small cell lung cancer. Cancer Research 1994;54:640–2.Google ScholarPubMed
Brambilla, E, Moro, D, Gazzeri, S, Brambilla, C. Alterations of expression of Rb, p16(INK4A) and cyclin D1 in non-small cell lung carcinoma and their clinical significance. Journal of Pathology 1999;188:351–60.3.0.CO;2-W>CrossRefGoogle ScholarPubMed
Sato, M, Takahashi, K, Nagayama, K, et al. Identification of chromosome arm 9p as the most frequent target of homozygous deletions in lung cancer. Genes, Chromosomes and Cancer 2005;44:405–14.CrossRefGoogle ScholarPubMed
Esteller, M.Cancer epigenetics: DNA methylation and chromatin alterations in human cancer. Advances in Experimental Medicine and Biology 2003;532:39–49.CrossRefGoogle ScholarPubMed
Kotake, Y, Cao, R, Viatour, P, et al. pRB family proteins are required for H3K27 trimethylation and Polycomb repression complexes binding to and silencing p16INK4alpha tumor suppressor gene. Genes and Development 2007;21:49–54.CrossRefGoogle ScholarPubMed
Rubin, LL, de Sauvage, FJ. Targeting the Hedgehog pathway in cancer. Nature Reviews Drug Discovery 2006;5:1026–33.CrossRefGoogle Scholar
Riobo, NA, Lu, K, Emerson, CP, Jr. Hedgehog signal transduction: signal integration and cross talk in development and cancer. Cell Cycle 2006;5:1612–15.CrossRefGoogle ScholarPubMed
Lauth, M, Toftgard, R. Non-canonical activation of GLI transcription factors: implications for targeted anti-cancer therapy. Cell Cycle 2007;6:2458–63.CrossRefGoogle ScholarPubMed
Yuan, Z, Goetz, JA, Singh, S, et al. Frequent requirement of hedgehog signaling in non-small cell lung carcinoma. Oncogene 2007;26:1046–55.CrossRefGoogle ScholarPubMed
He, B, You, L, Uematsu, K, et al. A monoclonal antibody against Wnt-1 induces apoptosis in human cancer cells. Neoplasia 2004;6:7–14.CrossRefGoogle ScholarPubMed
You, L, He, B, Xu, Z, et al. Inhibition of Wnt-2-mediated signaling induces programmed cell death in non-small-cell lung cancer cells. Oncogene 2004;23:6170–4.CrossRefGoogle ScholarPubMed
Winn, RA, Van Scoyk, M, Hammond, M, et al. Antitumorigenic effect of Wnt 7a and Fzd 9 in non-small cell lung cancer cells is mediated through ERK-5-dependent activation of peroxisome proliferator-activated receptor gamma. Journal of Biological Chemistry 2006;281:26 943–50.CrossRefGoogle ScholarPubMed
Fukui, T, Kondo, M, Ito, G, et al. Transcriptional silencing of secreted frizzled related protein 1 (SFRP 1) by promoter hypermethylation in non-small-cell lung cancer. Oncogene 2005;24:6323–7.CrossRefGoogle ScholarPubMed
Mazieres, J, He, B, You, L, et al. Wnt inhibitory factor-1 is silenced by promoter hypermethylation in human lung cancer. Cancer Research 2004;64:4717–20.CrossRefGoogle ScholarPubMed
Uematsu, K, He, B, You, L, et al. Activation of the Wnt pathway in non small cell lung cancer: evidence of dishevelled overexpression. Oncogene 2003;22:7218–21.CrossRefGoogle ScholarPubMed
Wissmann, C, Wild, PJ, Kaiser, S, et al. WIF1, a component of the Wnt pathway, is down-regulated in prostate, breast, lung, and bladder cancer. Journal of Pathology 2003;201:204–12.CrossRefGoogle Scholar
Dang, TP, Eichenberger, S, Gonzalez, A, Olson, S, Carbone, DP. Constitutive activation of Notch3 inhibits terminal epithelial differentiation in lungs of transgenic mice. Oncogene 2003;22:1988–97.CrossRefGoogle ScholarPubMed
Politi, K, Feirt, N, Kitajewski, J. Notch in mammary gland development and breast cancer. Seminars on Cancer Biology 2004;14:341–7.CrossRefGoogle ScholarPubMed
Parr, C, Watkins, G, Jiang, WG. The possible correlation of Notch-1 and Notch-2 with clinical outcome and tumour clinicopathological parameters in human breast cancer. International Journal of Molecular Medicine 2004;14:779–86.Google ScholarPubMed
Hainaud, P, Contreres, JO, Villemain, A, et al. The role of the vascular endothelial growth factor-Delta-like 4 ligand/Notch4-ephrin B2 cascade in tumor vessel remodeling and endothelial cell functions. Cancer Research 2006;66:8501–10.CrossRefGoogle ScholarPubMed
Sansone, P, Storci, G, Tavolari, S, et al. IL-6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. Journal of Clinical Investigation 2007;117:3988–4002.CrossRefGoogle ScholarPubMed
Haruki, N, Kawaguchi, KS, Eichenberger, S, et al. Dominant-negative Notch3 receptor inhibits mitogen-activated protein kinase pathway and the growth of human lung cancers. Cancer Research 2005;65:3555–61.CrossRefGoogle ScholarPubMed
Sun, S, Schiller, JH, Spinola, M, Minna, JD. New molecularly targeted therapies for lung cancer. Journal of Clinical Investigation 2007;117:2740–50.CrossRefGoogle ScholarPubMed
Shigematsu, H, Gazdar, AF. Somatic mutations of epidermal growth factor receptor signaling pathway in lung cancers. International Journal of Cancer 2006;118:257–62.CrossRefGoogle ScholarPubMed
Yousem, SA, Nikiforova, M, Nikiforov, Y. The histopathology of BRAF-V600E-mutated lung adenocarcinoma. American Journal of Surgical Pathology 2008;32:1317–21.CrossRefGoogle ScholarPubMed
Hirsch, FR, Varella-Garcia, M, Bunn, PA, Jr., et al. Epidermal growth factor receptor in non-small-cell lung carcinomas:correlation between gene copy number and protein expression and impact on prognosis. Journal of Clinical Oncology 2003;21:3798–807.CrossRefGoogle ScholarPubMed
Nakamura, H, Saji, H, Ogata, A, et al. Correlation between encoded protein overexpression and copy number of the HER2 gene with survival in non-small cell lung cancer. International Journal of Cancer 2003;103:61–6.CrossRefGoogle ScholarPubMed
Hirashima, N, Takahashi, W, Yoshii, S, Yamane, T, Ooi, A. Protein overexpression and gene amplification of c-erb B-2 in pulmonary carcinomas:a comparative immunohistochemical and fluorescence in situ hybridization study. Modern Pathology 2001;14:556–62.CrossRefGoogle ScholarPubMed
Tatematsu, A, Shimizu, J, Murakami, Y, et al. Epidermal growth factor receptor mutations in small cell lung cancer. Clinical Cancer Research 2008;14:6092–6.CrossRefGoogle ScholarPubMed
Swanton, C, Futreal, A, Eisen, T. Her2-targeted therapies in non-small cell lung cancer. Clinical Cancer Research 2006;12:4377s–83s.CrossRefGoogle ScholarPubMed
Rodenhuis, S, van de Wetering, ML, Mooi, WJ, et al. Mutational activation of the K-ras oncogene: a possible pathogenetic factor in adenocarcinoma of the lung. New England Journal of Medicine 1987;317:929–35.CrossRefGoogle ScholarPubMed
De Biasi, F, Del Sal, G, Hand, PH. Evidence of enhancement of the ras oncogene protein product (p21) in a spectrum of human tumors. International Journal of Cancer 1989;43:431–5.CrossRefGoogle Scholar
Potiron, VA, Roche, J, Drabkin, HA. Semaphorins and their receptors in lung cancer. Cancer Letters 2008;273:1–14.CrossRefGoogle ScholarPubMed
Samuels, Y, Wang, Z, Bardelli, A, et al. High frequency of mutations of the PIK3CA gene in human cancers. Science 2004;304:554.CrossRefGoogle ScholarPubMed
Lee, JW, Soung, YH, Kim, SY, et al. PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas. Oncogene 2005;24:1477–80.CrossRefGoogle ScholarPubMed
Davies, H, Hunter, C, Smith, R, et al. Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Research 2005;65:7591–5.CrossRefGoogle ScholarPubMed
Micke, P, Hengstler, JG, Ros, R, et al. c-erbB-2 expression in small-cell lung cancer is associated with poor prognosis. International Journal of Cancer 2001;92:474–9.CrossRefGoogle ScholarPubMed
Potti, A, Willardson, J, Forseen, C, et al. Predictive role of HER-2/neu overexpression and clinical features at initial presentation in patients with extensive stage small cell lung carcinoma. Lung Cancer 2002;36:257–61.CrossRefGoogle ScholarPubMed
Dworakowska, D, Jassem, E, Jassem, J, et al. MDM2 gene amplification: a new independent factor of adverse prognosis in non-small cell lung cancer (NSCLC). Lung Cancer 2004;43:285–95.CrossRefGoogle Scholar
Cappuzzo, F, Janne, PA, Skokan, M, et al. MET increased gene copy number and primary resistance to gefitinib therapy in non-small-cell lung cancer patients. Annals of Oncology 2008;20:298–304.CrossRefGoogle ScholarPubMed
Beau-Faller, M, Ruppert, AM, Voegeli, AC, et al. MET gene copy number in non-small cell lung cancer: molecular analysis in a targeted tyrosine kinase inhibitor naive cohort. Journal of Thoracic Oncology 2008;3:331–9.CrossRefGoogle Scholar
Richardson, GE, Johnson, BE. The biology of lung cancer. Seminars on Oncology 1993;20:105–27.Google ScholarPubMed
Johnson, BE, Russell, E, Simmons, AM, et al. MYC family DNA amplification in 126 tumor cell lines from patients with small cell lung cancer. Journal of Cell Biochemistry 1996;24:210–17.CrossRefGoogle ScholarPubMed
Ibson, JM, Waters, JJ, Twentyman, PR, Bleehen, NM, Rabbitts, PH. Oncogene amplification and chromosomal abnormalities in small cell lung cancer. Journal of Cell Biochemistry 1987;33:267–88.CrossRefGoogle ScholarPubMed
Shiraishi, M, Noguchi, M, Shimosato, Y, Sekiya, T. Amplification of protooncogenes in surgical specimens of human lung carcinomas. Cancer Research 1989;49:6474–9.Google ScholarPubMed
Miller, CT, Chen, G, Gharib, TG, et al. Increased C-CRK proto-oncogene expression is associated with an aggressive phenotype in lung adenocarcinomas. Oncogene 2003;22:7950–7.CrossRefGoogle ScholarPubMed
Pezzella, F, Turley, H, Kuzu, I, et al. bcl-2 protein in non-small-cell lung carcinoma. New England Journal of Medicine 1993;329:690–4.CrossRefGoogle ScholarPubMed
Kaiser, U, Schilli, M, Haag, U, et al. Expression of bcl-2–protein in small cell lung cancer. Lung Cancer 1996;15:31–40.CrossRefGoogle ScholarPubMed
Reissmann, PT, Koga, H, Figlin, RA, Holmes, EC, Slamon, DJ. Amplification and overexpression of the cyclin D1 and epidermal growth factor receptor genes in non-small-cell lung cancer. Lung Cancer Study Group. Journal of Cancer Research and Clinical Oncology 1999;125:61–70.CrossRefGoogle ScholarPubMed
Eren, B, Sar, M, Oz, B, Dincbas, FH. MMP-2, TIMP-2 and CD44v6 expression in non-small-cell lung carcinomas. Annals of the Academy of Medicine Singapore 2008;37:32–9.Google ScholarPubMed
Junker, K, Wiethege, T, Muller, KM. Pathology of small-cell lung cancer. Journal of Cancer Research and Clinical Oncology 2000;126:361–8.CrossRefGoogle ScholarPubMed
Micke, P, Basrai, M, Faldum, A, et al. Characterization of c-kit expression in small cell lung cancer:prognostic and therapeutic implications. Clinical Cancer Research 2003;9:188–94.Google ScholarPubMed
Cook, RM, Miller, YE, Bunn, PA, Jr. Small cell lung cancer:etiology, biology, clinical features, staging, and treatment. Current Problems in Cancer 1993;17:69–141.CrossRefGoogle Scholar
Araki, K, Ishii, G, Yokose, T, et al. Frequent overexpression of the c-kit protein in large cell neuroendocrine carcinoma of the lung. Lung Cancer 2003;40:173–80.CrossRefGoogle ScholarPubMed
Rygaard, K, Nakamura, T, Spang-Thomsen, M. Expression of the proto-oncogenes c-met and c-kit and their ligands, hepatocyte growth factor/scatter factor and stem cell factor, in SCLC cell lines and xenografts. British Journal of Cancer 1993;67:37–46.CrossRefGoogle ScholarPubMed
Plummer, H, 3rd, Catlett, J, Leftwich, J, et al. c-myc expression correlates with suppression of c-kit protooncogene expression in small cell lung cancer cell lines. Cancer Research 1993;53:4337–42.Google ScholarPubMed
Hibi, K, Takahashi, T, Sekido, Y, et al. Coexpression of the stem cell factor and the c-kit genes in small-cell lung cancer. Oncogene 1991;6:2291–6.Google ScholarPubMed
Weiner, DB, Nordberg, J, Robinson, R, et al. Expression of the neu gene-encoded protein (P185neu) in human non-small cell carcinomas of the lung. Cancer Research 1990;50:421–5.Google ScholarPubMed
Schneider, PM, Hung, MC, Chiocca, SM, et al. Differential expression of the c-erbB-2 gene in human small cell and non-small cell lung cancer. Cancer Research 1989;49:4968–71.Google ScholarPubMed
Fernandes, A, Hamburger, AW, Gerwin, BI. ERBB-2 kinase is required for constitutive stat 3 activation in malignant human lung epithelial cells. International Journal of Cancer 1999;83:564–70.3.0.CO;2-Q>CrossRefGoogle ScholarPubMed
Rygaard, K, Vindelov, LL, Spang-Thomsen, M. Expression of myc family oncoproteins in small-cell lung-cancer cell lines and xenografts. International Journal of Cancer 1993;54:144–52.CrossRefGoogle ScholarPubMed
Takahashi, T, Obata, Y, Sekido, Y, et al. Expression and amplification of myc gene family in small cell lung cancer and its relation to biological characteristics. Cancer Research 1989;49:2683–8.Google ScholarPubMed
Spencer, CA, Groudine, M. Control of c-myc regulation in normal and neoplastic cells. Advances in Cancer Research 1991;56:1–48.CrossRefGoogle ScholarPubMed
Zhang, P, Gao, WY, Turner, S, Ducatman, BS. Gleevec (STI-571) inhibits lung cancer cell growth (A549) and potentiates the cisplatin effect in vitro. Molecular Cancer 2003;2:1.CrossRefGoogle ScholarPubMed
Rikova, K, Guo, A, Zeng, Q, et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 2007;131:1190–203.CrossRefGoogle ScholarPubMed
Johnson, FM, Krug, LM, Tran, HT, et al. Phase I studies of imatinib mesylate combined with cisplatin and irinotecan in patients with small cell lung carcinoma. Cancer 2006;106:366–74.CrossRefGoogle ScholarPubMed
Rossi, G, Cavazza, A, Marchioni, A, et al. Role of chemotherapy and the receptor tyrosine kinases KIT, PDGFRalpha, PDGFRbeta, and Met in large-cell neuroendocrine carcinoma of the lung. Journal of Clinical Oncology 2005;23:8774–85.CrossRefGoogle ScholarPubMed
Ji, H, Ramsey, MR, Hayes, DN, et al. LKB1 modulates lung cancer differentiation and metastasis. Nature 2007;448:807–10.CrossRefGoogle ScholarPubMed
Koivunen, JP, Kim, J, Lee, J, et al. Mutations in the LKB1 tumour suppressor are frequently detected in tumours from Caucasian but not Asian lung cancer patients. British Journal of Cancer 2008;99:245–52.CrossRefGoogle Scholar
Carbone, DP, Mitsudomi, T, Chiba, I, et al. p53 immunostaining positivity is associated with reduced survival and is imperfectly correlated with gene mutations in resected non-small cell lung cancer. A preliminary report of LCSG 871. Chest 1994;106:377S–81S.Google ScholarPubMed
Wistuba, II, Berry, J, Behrens, C, et al. Molecular changes in the bronchial epithelium of patients with small cell lung cancer. Clinical Cancer Research 2000;6:2604–10.Google ScholarPubMed
Chiba, I, Takahashi, T, Nau, MM, et al. Mutations in the p53 gene are frequent in primary, resected non-small cell lung cancer. Lung Cancer Study Group. Oncogene 1990;5:1603–10.Google ScholarPubMed
Shimizu, E, Zhao, M, Shinohara, A, et al. Differential expressions of cyclin A and the retinoblastoma gene product in histological subtypes of lung cancer cell lines. Journal of Cancer Research and Clinical Oncology 1997;123:533–8.CrossRefGoogle ScholarPubMed
Salgia, R, Skarin, AT. Molecular abnormalities in lung cancer. Journal of Clinical Oncology 1998;16:1207–17.CrossRefGoogle ScholarPubMed
Hensel, CH, Hsieh, CL, Gazdar, AF, et al. Altered structure and expression of the human retinoblastoma susceptibility gene in small cell lung cancer. Cancer Research 1990;50:3067–72.Google ScholarPubMed
Girard, L, Zochbauer-Muller, S, Virmani, AK, Gazdar, AF, Minna, JD. Genome-wide allelotyping of lung cancer identifies new regions of allelic loss, differences between small cell lung cancer and non-small cell lung cancer, and loci clustering. Cancer Research 2000;60:4894–906.Google ScholarPubMed
Virmani, AK, Fong, KM, Kodagoda, D, et al. Allelotyping demonstrates common and distinct patterns of chromosomal loss in human lung cancer types. Genes, Chromosomes and Cancer 1998;21:308–19.3.0.CO;2-2>CrossRefGoogle ScholarPubMed
Thiberville, L, Payne, P, Vielkinds, J, et al. Evidence of cumulative gene losses with progression of premalignant epithelial lesions to carcinoma of the bronchus. Cancer Research 1995;55:5133–9.Google ScholarPubMed
Sunaga, N, Miyajima, K, Suzuki, M, et al. Different roles for caveolin-1 in the development of non-small cell lung cancer versus small cell lung cancer. Cancer Research 2004;64:4277–85.CrossRefGoogle ScholarPubMed
Mori, S, Ito, G, Usami, N, et al. p53 apoptotic pathway molecules are frequently and simultaneously altered in nonsmall cell lung carcinoma. Cancer 2004;100:1673–82.CrossRefGoogle ScholarPubMed
Prudkin, L, Behrens, C, Liu, DD, et al. Loss and reduction of FUS1 protein expression is a frequent phenomenon in the pathogenesis of lung cancer. Clinical Cancer Research 2008;14:41–7.CrossRefGoogle ScholarPubMed
Safar, AM, Spencer, H, 3rd, Su, X, et al. Methylation profiling of archived non-small cell lung cancer: a promising prognostic system. Clinical Cancer Research 2005;11:4400–5.CrossRefGoogle ScholarPubMed
Toyooka, S, Toyooka, KO, Maruyama, R, et al. DNA methylation profiles of lung tumors. Molecular Cancer Therapeutics 2001;1:61–7.Google ScholarPubMed
Shimamoto, T, Ohyashiki, JH, Hirano, T, Kato, H, Ohyashiki, K. Hypermethylation of E-cadherin gene is frequent and independent of p16INK4A methylation in non-small cell lung cancer: potential prognostic implication. Oncology Reports 2004;12:389–95.Google ScholarPubMed
Jarmalaite, S, Kannio, A, Anttila, S, Lazutka, JR, Husgafvel-Pursiainen, K. Aberrant p16 promoter methylation in smokers and former smokers with nonsmall cell lung cancer. International Journal of Cancer 2003;106:913–18.CrossRefGoogle ScholarPubMed
Esteller, M, Sanchez-Cespedes, M, Rosell, R, et al. Detection of aberrant promoter hypermethylation of tumor suppressor genes in serum DNA from non-small cell lung cancer patients. Cancer Research 1999;59:67–70.Google ScholarPubMed
Kim, DH, Nelson, HH, Wiencke, JK, et al. p16(INK4a) and histology-specific methylation of CpG islands by exposure to tobacco smoke in non-small cell lung cancer. Cancer Research 2001;61:3419–24.Google ScholarPubMed
Tang, X, Khuri, FR, Lee, JJ, et al. Hypermethylation of the death-associated protein (DAP) kinase promoter and aggressiveness in stage I non-small-cell lung cancer. Journal of the National Cancer Institute 2000;92:1511–16.CrossRefGoogle ScholarPubMed
Kim, DS, Cha, SI, Lee, JH, et al. Aberrant DNA methylation profiles of non-small cell lung cancers in a Korean population. Lung Cancer 2007;58:1–6.CrossRefGoogle Scholar
Suh, YA, Lee, HY, Virmani, A, et al. Loss of retinoic acid receptor beta gene expression is linked to aberrant histone H3 acetylation in lung cancer cell lines. Cancer Research 2002;62:3945–9.Google ScholarPubMed
Burbee, DG, Forgacs, E, Zochbauer-Muller, S, et al. Epigenetic inactivation of RASSF1A in lung and breast cancers and malignant phenotype suppression. Journal of the National Cancer Institute 2001;93:691–9.CrossRefGoogle ScholarPubMed
Dammann, R, Li, C, Yoon, JH, et al. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nature Genetics 2000;25:315–19.CrossRefGoogle ScholarPubMed
Speicher, MR, Gwyn Ballard, S, Ward, DC. Karyotyping human chromosomes by combinatorial multi-fluor FISH. Nature Genetics 1996;12:368–75.CrossRefGoogle ScholarPubMed
Balsara, BR, Testa, JR. Chromosomal imbalances in human lung cancer. Oncogene 2002;21:6877–83.CrossRefGoogle ScholarPubMed
Fong, KM, Kida, Y, Zimmerman, PV, Ikenaga, M, Smith, PJ. Loss of heterozygosity frequently affects chromosome 17q in non-small cell lung cancer. Cancer Research 1995;55:4268–72.Google ScholarPubMed
Luk, C, Tsao, MS, Bayani, J, Shepherd, F, Squire, JA. Molecular cytogenetic analysis of non-small cell lung carcinoma by spectral karyotyping and comparative genomic hybridization. Cancer Genetics and Cytogenetics 2001;125:87–99.CrossRefGoogle ScholarPubMed
Petersen, I, Bujard, M, Petersen, S, et al. Patterns of chromosomal imbalances in adenocarcinoma and squamous cell carcinoma of the lung. Cancer Research 1997;57:2331–5.Google ScholarPubMed
Petersen, I, Langreck, H, Wolf, G, et al. Small-cell lung cancer is characterized by a high incidence of deletions on chromosomes 3p, 4q, 5q, 10q, 13q and 17p. British Journal of Cancer 1997;75:79–86.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • Lung cancer
    • By Jill E. Larsen, Hamon Center for herapeutic Oncology Research, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA, John D. Minna, Hamon Center for herapeutic Oncology Research, Simmons Cancer Center University of Texas Southwestern Medical Center, Dallas, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.044
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • Lung cancer
    • By Jill E. Larsen, Hamon Center for herapeutic Oncology Research, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA, John D. Minna, Hamon Center for herapeutic Oncology Research, Simmons Cancer Center University of Texas Southwestern Medical Center, Dallas, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.044
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • Lung cancer
    • By Jill E. Larsen, Hamon Center for herapeutic Oncology Research, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA, John D. Minna, Hamon Center for herapeutic Oncology Research, Simmons Cancer Center University of Texas Southwestern Medical Center, Dallas, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.044
Available formats
×