Skip to main content
Current Genomics logoLink to Current Genomics
. 2010 Nov;11(7):513–518. doi: 10.2174/138920210793175903

Lung Cancer: Are we up to the Challenge?

Luca Esposito 1, Daniele Conti 2,3, Ramyasri Ailavajhala 4, Nansie Khalil 4, Antonio Giordano 2,4,*
PMCID: PMC3048313  PMID: 21532835

Abstract

Lung cancer is the leading cause of cancer deaths worldwide among both men and women, with more than 1 million deaths annually. Non–small cell lung cancer (NSCLC) accounts for about 80% of all lung cancers.

Although recent advances have been made in diagnosis and treatment strategies, the prognosis of NSCLC patients is poor and it is basically due to a lack of early diagnostic tools.

However, in the last years genetic and biochemical studies have provided more information about the protein and gene’s mutations involved in lung tumors. Additionally, recent proteomic and microRNA’s approaches have been introduced to help biomarker discovery.

Here we would like to discuss the most recent discoveries in lung cancer pathways, focusing on the genetic and epigenetic factors that play a crucial role in malignant cell proliferation, and how they could be helpful in diagnosis and targeted therapy.

Keywords: Lung cancer, oncosuppressors, oncogenes, epigenetics of lung cancer, diagnostic tools for lung cancer.

INTRODUCTION

Lung cancer is the leading cause of cancer-related deaths over the world, among both men and women, with an incidence of over 200000 new cases per year and a very high mortality rate. It is responsible for more deaths than breast, colon and prostate tumors combined [1].

Lung tumors can be divided into two histological groups: non-small cell lung cancer (NSCLC) (80.4%) and small cell lung cancer (SCLC) (16.8%) [2]. NSCLC, consisting mainly of adenocarcinoma, squamous cell and large cell carcinoma, accounts for almost 80% of lung cancer cases, whereas SCLC is slightly more common and all known cases are due to cigarette smoking.

Many factors potentially contribute to lung cancer formation, e.g. tobacco smoke, ionizing radiation and viral infection, although the mechanisms involved in lung carcinogenesis remain largely unknown.

Lung cancer is often suspected on the basis of abnormal chest imaging and/or non-specific symptoms. Bronchoscopy is generally used as an initial diagnostic tool, consisting of cytopathologic examination of bronchoalveolar lavage, endobronchial brushings and biopsies from the suspect area. Despite this procedure being specific for lung cancer, its sensitivity is low [3]. Therefore, more invasive and expensive diagnostic tests are often required, delaying the diagnosis and the subsequent treatment initiation. Indeed, the main problem of lung cancer disease is a lack of early-diagnosis tools, resulting in more than 60% of patients diagnosed with advanced or metastatic disease [4, 5] and therefore not eligible for a curative surgical resection.

The overall five-year survival rate for patients with NSCLC is less than 15% and has remained largely unchanged for the last three decades. Despite new drugs and therapeutic regimens, surgical resection remains the most promising chance for the ~25% of patients who present early-stage disease (stage I-IIIA), although 65% will relapse within two years [6].

These data suggest that strategies for the early detection of lung cancer, aimed at identifying the tumor at a stage in which it is small and locally defined, are urgently needed to increase significantly the chances of a cure [4]. Recent evidence shows that low dose spiral computed tomography (CT) detects lung cancer at smaller sizes and earlier than chest X-ray (CXR) that failed to identify 79% of lung cancers that were smaller than 2 cm [7].

Another major problem linked to lung cancer disease, as for other cancers, is the need to find new and more tailored chemotherapeutical.

The variable response to chemotherapy with platinum-based regimens in NSCLC is well recognized clinically. Patients crossing from one regimen to another in clinical trials commonly show responses [8], suggesting that it might be possible to optimize the therapy for individual patients once has been determined which regimen would be most effective.

In this short review we summarize what are the main genetic alterations involved in lung cancer disease and what is the prospective for the future development of new biomarker and diagnostic tools to improve the early detection of lung cancer.

GENETIC AND EPIGENETIC ALTERATIONS IN LUNG CANCER

Different factors contribute to lung cancer formation: tobacco smoke, ionizing radiation and viral infection are among the most well established. However, the mechanisms involved in lung carcinogenesis remain largely unknown to date [9].

As most other cancers, several genes are involved in lung cancer disease [9], which is initiated by the activation of oncogenes or inactivation of tumor suppressor genes [10].

The proto-oncogene KRAS is often mutated and is responsible for 10–30% of lung adenocarcinomas [11, 12].

MYC and Cyclin D1 [13] are amplified and over-expressed in 2.5–10% and 5% of NSCLC, respectively.

ERBB2 (also known as HER-2/neu) or BCL2 over-expression are involved in 25% of cases [14]. Farther, many studies have found ERBB2 mutations (exons 19-20) in a small subset of patients. These mutations often represent early events in the carcinogenesis of lung adenocarcinoma in never smokers [15, 16]. Novel mutations in BRAF were identified through systematic resequencing of oncogenes and are present in about 2% of adenocarcinoma patients and restricted to tumors that did not show KRAS mutations [17, 18].

The epidermal growth factor receptor (EGFR) that regulates cell proliferation, apoptosis, angiogenesis and tumor invasion [11] is over-expressed or in certain cases affected by oncogenic mutations in NSCLC and is one of the major target for lung cancer therapy. Furthermore, some activating mutations in EGFR, mainly deletion mutations in exon 19 and the single L858R point mutation in exon 21, are associated with increased response and survival after tyrosine kinase inhibitor therapy, whereas the T790M point mutation or insertion mutations in exon 20 of EGFR, are associated with failure to respond [19-22].

Basically, mutations involving EGFR, ERBB2 and KRAS are mutually exclusive and are thought to represent early events in the carcinogenesis of lung adenocarcinoma in never (EGFR and ERBB2) and current smokers (KRAS) [23].

Other oncogenes whose expression has been found altered in lung cancer, include MET, NKX2-1, and PIK3CA [11].

The tyrosine kinase protein, SRC is also overexpressed and activated in epithelial tumors, and the levels of expression or activation generally correlate with disease progression [24], although activating mutations are rare [25]. Studies have shown that SRC is activated in NSCLC tumor tissues [26, 27], and its inhibition leads to decreased anchorage-dependent cell growth and to cell cycle arrest and apoptosis [28, 29].

Recently, the fusion of the anaplastic lymphoma kinase (ALK) with the echinoderm microtubule-associated protein-like 4 (EML4) has been identified in a subset of NSCLC. Approximately 5% of all NSCLC cases contain an EML4-ALK translocation. It occurs in mutual exclusion to EGFR and KRAS mutations and is associated with nonsmokers. Since ALK tyrosine kinase activity is oncogenic both in vitro and in vivo, new ALK kinase inhibitors are being evaluated in pre-clinical trials for lung cancer [30, 31].

Also, inactivation of tumor suppressor genes plays an important role in lung carcinogenesis, as for example the tumor suppressor gene TP53 that is mutated in 60–75% of lung cancer including both NSCLC and SCLC [32].

Another important tumor suppressor gene is LKB1, whose loss-of-function mutation/deletion is observed in 30% lung adenocarcinomas and 20% of squamous cell carcinomas [33, 34].

Recently it has been shown a clearly link between SCR and LKB1 in NSCLC disease. In particular, SRC and FAK signaling pathways are activated in lung cancers when the tumor suppressor LKB1 is deleted. These findings suggest the use of unique combinatorial therapies for treatment of lung cancers [35].

The role of RB family genes in lung cancer malignancy has been long examined but remains unclear to date.

What is known is that the tumor suppressor RB1 is inactivated in a broad range of human tumors [36, 37], including pediatric retinoblastomas and about 90% of human SCLC. When RB1 is not itself mutated, other alterations in members of the RB pathway are found in human tumors [37-39]. For instance, RB1 is rarely found mutated in lung adenocarcinomas whereas p16INK4a, an upstream activator of the RB1 protein and the two related proteins p107 and p130, is frequently inactivated in this tumor type [40].

Recent studies provided genetic evidence that RB1 and p130 have the capacity to act as suppressors of lung adenocarcinoma development, confirming the broad tumor-suppressor potential of the RB family genes and raising the possibility that re-activation or induction of RB family function in lung cancers may be used to slow tumor growth in patients [41].

Genetic polymorphisms are also indicated to be involved in lung carcinogenesis, e.g. interleukin-1 [42], cytochrome P450 [43], apoptosis promoters such as caspase-8 [44] and DNA repair proteins such as XRCC1 [45].

Epigenetic modifications are now well recognized to significantly contribute to lung cancer tumorigenesis. For example, a great number of aberrantly methylated genes have been identified in lung cancer. A well-studied example is the aberrant promoter methylation of the tumor suppressor gene p16 which leads to gene silencing, an early event in tumorigenesis [46-48]. Additional examples include H-cadherin [49], death-associated protein kinase 1 (DAPK1) [50], 14-3-3 sigma [51] and the candidate tumor suppressor gene RASSF1A [52].

Although research based on known genes, proteins and epigenetic alteration has already yielded new information, during the past decade the microRNA (miRNA) research-field has been extensively studied and it may also lend insights into the biology of lung cancer, as well as for cancer in general.

MiRNAs are small non-coding RNAs about 22 nt long that play key roles in gene expression regulation by modulating the translation and degradation of target mRNAs through base pairing to partially complementary sites [53-55].

MmiRNA microarray analysis for lung cancer were recently examined to investigate miRNA involvement in lung carcinogenesis and the results obtained show that miRNAs could discriminate lung cancers from healthy lung tissues, suggesting that miRNA expression profiles could be diagnostic and prognostic markers of lung cancer [56] and also allow for the differential diagnosis between lung adenocarcinoma and mesothelioma [57].

Recent studies have reported some examples of miRNAs implication in lung cancer. For example miR-29 family members directly target both DNA methyltransferases DNMT 3A and -3B. In particular, it has been shown that expression of miR-29 family members is inversely correlated with DNMT3A and -3B expression in lung cancers and that these miRNAs down-modulate expression levels of both enzymes. Furthermore, enforced expressionof these miRNAs in lung cancer cells leads to reduced global DNA methylation, restores expression of TSGs, and inhibits tumorigenicity both in vitro and in vivo [58].

Another important report shows that miR-107 and miR-185 are down regulated in lung cancer compared with normal lung and their reintroduction in NSCLC cell lines is able to suppress cell growth [59]. Similarly, miR-15a/miR-16, which induce RB1-mediated cell cycle arrest by the down-regulation of G1 cyclins are downregulated in NSCLC [60].

Farthermore, we must remember the role of telomere and telomerases that recently have been demonstrated to be involved in lung cancer.

Telomeres are nucleoprotein complexes located at the end of eukaryotic chromosomes, which role is to prevent them from degradation, end to-end fusion and rearrangement.

Recently, telomere length has been proposed as prognostic factor in NSCLC, reflecting indirectly chromosomal instability.

Telomerase and telomeric complex play a key role in lung tumor progression. As telomere maintenance is essential to tumor cell proliferation, several approaches have been developed to target either telomerase or telomeric complex. Anti telomerase strategies can either target hTERT or hTERC, in addition to modulation of telomerase regulators at the transcriptional and post-transcriptional levels. More recently, the use of specific ligands leading to G quadruplex telomeric structure stabilization and therefore to limitation of telomerase accessibility to its target appears as a promising area of development [61].

DIAGNOSTIC TOOLS IN LUNG CANCER

Lung cancer is the most prominent cause of cancer-related mortality worldwide. About 60% of those diagnosed with lung cancer die within one year after diagnosis and the five-year survival for all patients with lung cancer is only 16%, a percentage that has not improved significantly in the past 10 years [1]. Although many insights into the molecular pathology of lung tumors have been achieved, additional information is critical for the development of targeted treatments and of early diagnostic methods. Diagnosis and accurate staging of lung cancer is essential for selection of appropriate curative or palliative therapy and affects patient prognosis. Both invasive and non-invasive procedures are used for this purpose.

Thorax computerized tomography (CT) and positron emission tomography (PET) are non-invasive techniques used to detect lymph node involvement and histological sampling of lymph nodes, but often these techniques are not sufficient and thus invasive techniques such as transbronchial needle aspiration (TBNA), needle aspiration by endobronchial ultrasound (EBUS) and mediastinoscopy are frequently used to recover histological samples from lymph nodes [62]. Since patients often present poor general conditions or severe hypoxemia due to coexisting diseases (COPD, heart failure, etc.), it may not be possible to use invasive procedures for diagnosis and staging in some of the patients with lung cancer.

Recent studies provided first evidence for the potential usefulness of a blood-based test for lung cancer diagnosis [63].

For example the mutational status of EGFR can be readily detected in primary tumors and the correlation between EGFR mutations and EGFR TKI (Tyrosine Kinase Inhibitors) sensitivity has been validated in several clinical trials, but it may be difficult to obtain tumor tissues for such analysis. Therefore, since plasma samples of patients with NSCLC often contain circulating DNA derived from tumor tissues, plasma samples have been used for detecting genetic alterations, in particular for EGFR [64].

The research of new biomarkers is important not only for improving diagnosis but also it may offer promise in optimizing treatment. Understanding the genetic mechanisms affecting drug activity and response to treatment is a major challenge for establishing an individualized chemotherapy.

To date, cisplatin and platinum-containing drugs are routinely used for treatment of NSCLC and are known to have vital role.

DNA adducts, due to cisplatin treatment, are mainly repaired by nucleotide excision repair pathway and proteins such as excision repair cross complementing 1 (ERCC1), ribonucleotide reductase subunit M1 (RRM1) and breast cancer susceptibility gene 1 (BRCA1) have an important role in this process.

Recently, ERCC1, RRM1 and BRCA1 and TS (Thymidylate synthase) [65] have been confirmed as predictive markers of treatment response and survival benefit in patients with lung cancer and prospective studies investigating the efficacy of their determination in larger set of patients are currently ongoing [66].

But to date more than two thirds of lung cancer tumors are diagnosed at late stages when the survival rate is low [4] and thus the main purpose and also the major obstacle of lung cancer research therapy is to detect lung cancer at an early stage and when it is still locally defined.

CONCLUSIONS

Lung cancer is the leading cause of death over the world and the only chance of cure for patients affected from this kind of cancer is surgical resection.

This is mainly due to the fact that several factors are involved in lung cancer develoment and progression and to date the diagnostic methods available for an early and efficient detection are not sufficient.

Although lung cancer research data have accumulated dramatically during the past several years, there is no database specifically focusing on lung cancer molecular biology available yet.

In this short review we have summarized some of the factors contributing to lung cancer, but it is just an overview of the most important proteins involved in lung cancer disease, which are often mutated or present an unusual pattern of expression compared to the healthy tissue.

All together these data are important for understanding the nature behind this type of cancer and also to understand that basic research on proteins, miRNAs and all the other epigenetic modifications could be used to develop more powerful diagnostic tools, as well as prognostic or predictive markers or even for the development of new targeting therapies for lung cancer.

REFERENCES

  • 1.Jemal A, Siegel R, Ward E, Murray T, Xu J, Thun MJ. Cancer statistics, 2007. CA Cancer J. Clin. 2007;57(1):43–66. doi: 10.3322/canjclin.57.1.43. [DOI] [PubMed] [Google Scholar]
  • 2.Travis WD, Travis LB, Devesa SS. Lung cancer. Cancer. 1995;75(1 Suppl):191–202. doi: 10.1002/1097-0142(19950101)75:1+<191::aid-cncr2820751307>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
  • 3.Schreiber G, McCrory DC. Performance characteristics of different modalities for diagnosis of suspected lung cancer: summary of published evidence. Chest. 2003;123(1 Suppl):115S–128S. doi: 10.1378/chest.123.1_suppl.115s. [DOI] [PubMed] [Google Scholar]
  • 4.Henschke C I, Yankelevitz D F. CT screening for lung cancer: update 2007. Oncologist. 2008;13(1):65–78. doi: 10.1634/theoncologist.2007-0153. [DOI] [PubMed] [Google Scholar]
  • 5.Spira A, Beane J, Shah V, Liu G, Schembri F, Yang X, Palma J, Brody JS. Effects of cigarette smoke on the human airway epithelial cell transcriptome. Proc. Natl. Acad. Sci. USA. 2004;101(27):10143–10148. doi: 10.1073/pnas.0401422101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sandler A, Gray R, Perry MC, Brahmer J, Schiller JH, Dowlati A, Lilenbaum R, Johnson DH. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N. Engl. J. Med. 2006;355(24):2542–2550. doi: 10.1056/NEJMoa061884. [DOI] [PubMed] [Google Scholar]
  • 7.Sone S, Li F, Yang ZG, Honda T, Maruyama Y, Takashima S, Hasegawa M, Kawakami S, Kubo K, Haniuda M, Yamanda T. Results of three-year mass screening programme for lung cancer using mobile low-dose spiral computed tomography scanner. Br. J. Cancer. 2001;84(1):25–32. doi: 10.1054/bjoc.2000.1531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Fossella FV, DeVore R, Kerr RN, Crawford J, Natale RR, Dunphy F, Kalman L, Miller V, Lee JS, Moore M, Gandara D, Karp D, Vokes E, Kris M, Kim Y, Gamza F, Hammershaimb L. Randomized phase III trial of docetaxel versus vinorelbine or ifosfamide in patients with advanced non-small-cell lung cancer previously treated with platinum-containing chemotherapy regimens. The TAX 320 Non-Small Cell Lung Cancer Study Group. J. Clin. Oncol. 2000;18(12):2354–2362. doi: 10.1200/JCO.2000.18.12.2354. [DOI] [PubMed] [Google Scholar]
  • 9.Wang L, Xiong Y, Sun Y, Fang Z, Li L, Ji H, Shi T. HLungDB: an integrated database of human lung cancer research. Nucleic Acids Res. 2010;38(Database issue):D665–9. doi: 10.1093/nar/gkp945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Fong KM, Sekido Y, Gazdar AF, Minna JD. Lung cancer. 9: Molecular biology of lung cancer: clinical implications. Thorax. 2003;58(10):892–900. doi: 10.1136/thorax.58.10.892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Herbst RS, Heymach JV, Lippman SM. Lung cancer. N. Engl. J. Med. 2008;359(13):1367–1380. doi: 10.1056/NEJMra0802714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Aviel-Ronen S, Blackhall FH, Shepherd FA, Tsao MS. Kras mutations in non-small-cell lung carcinoma: a review. Clin. Lung Cancer. 2006;8(1):30–38. doi: 10.3816/CLC.2006.n.030. [DOI] [PubMed] [Google Scholar]
  • 13.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. J. Cancer Res. Clin. Oncol. 1999;125(2):61–70. doi: 10.1007/s004320050243. [DOI] [PubMed] [Google Scholar]
  • 14.Salgia R, Skarin AT. Molecular abnormalities in lung cancer. J. Clin. Oncol. 1998;16(3):1207–1217. doi: 10.1200/JCO.1998.16.3.1207. [DOI] [PubMed] [Google Scholar]
  • 15.Shigematsu H, Takahashi T, Nomura M, Majmudar K, Suzuki M, Lee H, Wistuba II, Fong KM, Toyooka S, Shimizu N, Fujisawa T, Minna JD, Gazdar AF. Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer Res. 2005;65(5):1642–1646. doi: 10.1158/0008-5472.CAN-04-4235. [DOI] [PubMed] [Google Scholar]
  • 16.Buttitta F, Barassi F, Fresu G, Felicioni L, Chella A, Paolizzi D, Lattanzio G, Salvatore S, Camplese PP, Rosini S, Iarussi T, Mucilli F, Sacco R, Mezzetti A, Marchetti A. Mutational analysis of the HER2 gene in lung tumors from Caucasian patients: mutations are mainly present in adenocarcinomas with bronchioloalveolar features. Int. J. Cancer. 2006;119:2586–2591. doi: 10.1002/ijc.22143. [DOI] [PubMed] [Google Scholar]
  • 17.Naoki K, Chen TH, Richards WG, Sugarbaker DJ, Meyerson M. Missense mutations of the BRAF gene in human lung adenocarcinoma. Cancer Res. 2002;62(23):7001–7003. [PubMed] [Google Scholar]
  • 18.Brose MS, Volpe P, Feldman M, Kumar M, Rishi I, Gerrero R, Einhorn E, Herlyn M, Minna J, Nicholson A, Roth JA, Albelda SM, Davies H, Cox C, Brignell G, Stephens P, Futreal PA, Wooster R, Stratton MR, Weber BL. BRAF and RAS mutations in human lung cancer and melanoma. Cancer Res. 2002;62(23):6997–7000. [PubMed] [Google Scholar]
  • 19.Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan BW, Harris PL, Haserlat SM, Supko JG, Haluska FG, Louis DN, Christiani DC, Settleman J, Haber DA. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N. Engl. J. Med. 2004;350:2129–2139. doi: 10.1056/NEJMoa040938. [DOI] [PubMed] [Google Scholar]
  • 20.Paez JG, Jänne PA, Lee JC, Tracy S, Greulich H, Gabriel S, Herman P, Kaye FJ, Lindeman N, Boggon TJ, Naoki K, Sasaki H, Fujii Y, Eck MJ, Sellers WR, Johnson BE, Meyerson M. EGFRmutations in lung cancer: correlation with clinical response to gefitinib therapy. Science. 2004;304:1497–1500. doi: 10.1126/science.1099314. [DOI] [PubMed] [Google Scholar]
  • 21.Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I, Singh B, Heelan R, Rusch V, Fulton L, Mardis E, Kupfer D, Wilson R, Kris M, Varmus H. EGF receptor gene mutations are common in lung cancers from “never smokers” and are associated with sensitivity of tumors to gefitinib and erlotinib. Proc. Natl. Acad. Sci.USA. 2004;101:13306–13311. doi: 10.1073/pnas.0405220101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sequist LV, Engelman JA, Lynch TJ. Toward noninvasive genomic screening of lung cancer patients. J. Clin. Oncol. 2009;27(16):2589–2591. doi: 10.1200/JCO.2008.20.4875. [DOI] [PubMed] [Google Scholar]
  • 23.Sartori G, Cavazza A, Bertolini F, Longo L, Marchioni A, Costantini M, Barbieri F, Migaldi M, Rossi G. A subset of lung adenocarcinomas and atypical adenomatous hyperplasia-associated foci are genotypically related: an EGFR, HER2, and Kras mutational analysis. Am. J. Clin. Pathol. 2008;129(2):202–210. doi: 10.1309/THU13F3JRJVWLM30. [DOI] [PubMed] [Google Scholar]
  • 24.Irby RB, Yeatman TJ. Role of Src expression and activation in human cancer. Oncogene. 2000;19(49):5636–5642. doi: 10.1038/sj.onc.1203912. [DOI] [PubMed] [Google Scholar]
  • 25.Irby RB, Mao W, Coppola D, Kang J, Loubeau JM, Tru-deau W, Karl R, Fujita DJ, Jove R, Yeatman TJ. Activating SRC mutation in a subset of advanced human colon cancers. Nat. Genet. 1999;21(2):187–190. doi: 10.1038/5971. [DOI] [PubMed] [Google Scholar]
  • 26.Masaki T, Igarashi K, Tokuda M, Yukimasa S, Han F, Jin YJ, Li JQ, Yoneyama H, Uchida N, Fujita J, Yoshiji H, Wa-tanabe S, Kurokohchi K, Kuriyama S. pp60c-src activation in lung adenocarcinoma. Eur. J. Cancer. 2003;39(10):1447–1455. doi: 10.1016/s0959-8049(03)00276-4. [DOI] [PubMed] [Google Scholar]
  • 27.Mazurenko NN, Kogan EA, Zborovskaya IB, Kisseljov FL. Expression of pp60c-src in human small cell and non-small cell lung carcinomas. Eur. J. Cancer. 1992;28(2-3):372–377. doi: 10.1016/s0959-8049(05)80056-5. [DOI] [PubMed] [Google Scholar]
  • 28.Laird AD, Li G, Moss KG, Blake RA, Broome MA, Cherrington JM, Mendel DB. Src family kinase activity is required for signal tranducer and activator of transcription 3 and focal adhesion kinase phosphorylation and vascular endothelial growth factor signaling in vivo and for anchorage-dependent and -independent growth of human tumor cells. Mol. Cancer Ther. 2003;2(5):461–469. [PubMed] [Google Scholar]
  • 29.Song L, Turkson J, Karras JG, Jove R, Haura EB. Activation of Stat3 by receptor tyrosine kinases and cytokines regulates survival in human non-small cell carcinoma cells. Oncogene. 2003;22(27):4150–4165. doi: 10.1038/sj.onc.1206479. [DOI] [PubMed] [Google Scholar]
  • 30.Sasaki T, Rodig SJ, Chirieac LR, Jänne PA. The biology and treatment of EML4-ALK non-small cell lung cancer. Eur. J. Cancer. 2010;46(10):1773–80. doi: 10.1016/j.ejca.2010.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wong DW, Leung EL, So KK, Tam IY, Sihoe AD, Cheng LC, Ho KK, Au JS, Chung LP, Pik Wong M. 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(8):1723–1733. doi: 10.1002/cncr.24181. [DOI] [PubMed] [Google Scholar]
  • 32.Devereux TR, Taylor JA, Barrett JC. Molecular mechanisms of lung cancer. Interaction of environmental and genetic factors. Giles F. Filley Lecture. Chest. 1996;109(3 Suppl):14S–19S. doi: 10.1378/chest.109.3_supplement.14s. [DOI] [PubMed] [Google Scholar]
  • 33.Ji H, Ramsey MR, Hayes DN, Fan C, McNamara K, Kozlowski P, Torrice C, Wu MC, Shimamura T, Perera SA, Liang MC, Cai D, Naumov GN, Bao L, Contreras CM, Li D, Chen L, Krishnamurthy J, Koivunen J, Chirieac LR, Padera RF, Bronson RT, Lindeman NI, Christiani DC, Lin X, Shapiro GI, Janne PA, Johnson BE, Meyerson M, Kwiatkowski DJ, Castrillon DH, Bardeesy N, Sharpless NE, Wong KK. LKB1 modulates lung cancer differentiation and metastasis. Nature. 2007;448(7155):807–810. doi: 10.1038/nature06030. [DOI] [PubMed] [Google Scholar]
  • 34.Ding L, Getz G, Wheeler DA, Mardis ER, McLellan MD, Cibulskis K, Sougnez C, Greulich H, Muzny DM, Morgan MB, Fulton L, Fulton RS, Zhang Q, Wendl MC, Lawrence MS, Larson DE, Chen K, Dooling DJ, Sabo A, Hawes AC, Shen H, Jhangiani SN, Lewis LR, Hall O, Zhu Y, Mathew T, Ren Y, Yao J, Scherer SE, Clerc K, Metcalf GA, Ng B, Milosavljevic A, Gonzalez-Garay ML, Osborne JR, Meyer R, Shi X, Tang Y, Koboldt DC, Lin L, Abbott R, Miner TL, Pohl C, Fewell G, Haipek C, Schmidt H, Dunford-Shore BH, Kraja A, Crosby SD, Sawyer CS, Vickery T, Sander S, Robinson J, Winckler W, Baldwin J, Chirieac LR, Dutt A, Fennell T, Hanna M, Johnson BE, Onofrio RC, Thomas RK, Tonon G, Weir BA, Zhao X, Ziaugra L, Zody MC, Giordano T, Orringer MB, Roth JA, Spitz MR, Wistuba II, Ozenberger B, Good PJ, Chang AC, Beer DG, Watson MA, Ladanyi M, Broderick S, Yoshizawa A, Travis WD, Pao W, Province MA, Weinstock GM, Varmus HE, Gabriel SB, Lander ES, Gibbs RA, Meyerson M, Wilson RK. Somatic mutations affect key pathways in lung adenocarcinoma. Nature. 2008;455(7216):1069–1075. doi: 10.1038/nature07423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Slack-Davis J, Dasilva JO, Parsons SJ. LKB1 and Src: Antagonistic Regulators of Tumor Growth and Metastasis. Cancer Cell. 2010;17(6):547–559. doi: 10.1016/j.ccr.2010.05.016. [DOI] [PubMed] [Google Scholar]
  • 36.Classon M, Harlow E. The retinoblastoma tumour suppressor in development and cancer. Nat. Rev. Cancer. 2002;2(12):910–917. doi: 10.1038/nrc950. [DOI] [PubMed] [Google Scholar]
  • 37.Sherr CJ, McCormick F. The RB and p53 pathways in cancer. Cancer Cell. 2002;2(2):103–112. doi: 10.1016/s1535-6108(02)00102-2. [DOI] [PubMed] [Google Scholar]
  • 38.Grasemann C, Gratias S, Stephan H, Schuler A, Schramm A, Klein-Hitpass L, Rieder H, Schneider S, Kappes F, Eggert A, Lohmann DR. Gains and overexpression identify DEK and E2F3 as targets of chromosome 6p gains in retinoblastoma. Oncogene. 2005;24(42):6441–6449. doi: 10.1038/sj.onc.1208792. [DOI] [PubMed] [Google Scholar]
  • 39.Olsson AY, Feber A, Edwards S, Te Poele R, Giddings I, Merson S, Cooper CS. Role of E2F3 expression in modulating cellular proliferation rate in human bladder and prostate cancer cells. Oncogene. 2007;26(7):1028–1037. doi: 10.1038/sj.onc.1209854. [DOI] [PubMed] [Google Scholar]
  • 40.Wistuba II, Gazdar AF, Minna JD. Molecular genetics of small cell lung carcinoma. Semin. Oncol. 2001;28(2 Suppl 4):3–13. [PubMed] [Google Scholar]
  • 41.Ho VM, Schaffer BE, Karnezis AN, Park KS, Sage J. The retinoblastoma gene Rb and its family member p130 suppress lung adenocarcinoma induced by oncogenic K-Ras. Oncogene. 2009;28(10):1393–1399. doi: 10.1038/onc.2008.491. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Engels EA, Wu X, Gu J, Dong Q, Liu J, Spitz MR. Systematic evaluation of genetic variants in the inflammation pathway and risk of lung cancer. Cancer Res. 2007;67(13):6520–6527. doi: 10.1158/0008-5472.CAN-07-0370. [DOI] [PubMed] [Google Scholar]
  • 43.Wenzlaff AS, Cote ML, Bock CH, Land SJ, Santer SK, Schwartz DR, Schwartz AG. CYP1A1 and CYP1B1 polymorphisms and risk of lung cancer among never smokers: a population-based study. Carcinogenesis. 2005;26(12):2207–2212. doi: 10.1093/carcin/bgi191. [DOI] [PubMed] [Google Scholar]
  • 44.Son JW, Kang HK, Chae MH, Choi JE, Park JM, Lee WK, Kim CH, Kim DS, Kam S, Kang YM, Park JY. Polymorphisms in the caspase-8 gene and the risk of lung cancer. Cancer, Genet. Cytogenet. 2006;169(2):121–127. doi: 10.1016/j.cancergencyto.2006.04.001. [DOI] [PubMed] [Google Scholar]
  • 45.Yin J, Vogel U, Ma Y, Qi R, Sun Z, Wang H. The DNA repair gene XRCC1 and genetic susceptibility of lung cancer in a northeastern Chinese population. Lung Cancer. 2007;56(2):153–160. doi: 10.1016/j.lungcan.2006.12.012. [DOI] [PubMed] [Google Scholar]
  • 46.Zochbauer-Muller S, Fong KM, Virmani AK, Geradts J, Gazdar AF, Minna JD. Aberrant promoter methylation of multiple genes in non-small cell lung cancers. Cancer Res. 2001;61(1):249–255. [PubMed] [Google Scholar]
  • 47.Seike M, Gemma A, Hosoya Y, Hemmi S, Taniguchi Y, Fukuda Y, Yamanaka N, Kudoh S. Increase in the frequency of p16INK4 gene inactivation by hypermethylation in lung cancer during the process of metastasis and its relation to the status of p53. Clin. Cancer Res. 2000;6(11):4307–4313. [PubMed] [Google Scholar]
  • 48.Kersting M, Friedl C, Kraus A, Behn M, Pankow W, Schuermann M. Differential frequencies of p16(INK4a) promoter hypermethylation, p53 mutation, and K-ras mutation in exfoliative material mark the development of lung cancer in symptomatic chronic smokers. J. Clin. Oncol. 2000;18(18):3221–3229. doi: 10.1200/JCO.2000.18.18.3221. [DOI] [PubMed] [Google Scholar]
  • 49.Sato M, Mori Y, Sakurada A, Fujimura S, Horii A. The H-cadherin(CDH13) gene is inactivated in human lung cancer. Hum. Genet. 1998;103(1):96–101. doi: 10.1007/s004390050790. [DOI] [PubMed] [Google Scholar]
  • 50.Tang X, Khuri FR, Lee JJ, Kemp BL, Liu D, Hong WK, Mao L. Hypermethylation of the death-associated protein(DAP) kinase promoter and aggressiveness in stage I non-small-cell lung cancer. J. Natl. Cancer Inst. 2000;92(18):1511–1516. doi: 10.1093/jnci/92.18.1511. [DOI] [PubMed] [Google Scholar]
  • 51.Osada H, Tatematsu Y, Yatabe Y, Nakagawa T, Konishi H, Harano T, Tezel E, Takada M, Takahashi T. Frequent and histological type-specific inactivation of 14-3-3sigma in human lung cancers. Oncogene. 2002;21(15):2418–2424. doi: 10.1038/sj.onc.1205303. [DOI] [PubMed] [Google Scholar]
  • 52.Dammann R, Li C, Yoon JH, Chin PL, Bates S, Pfeifer GP. Epigenetic inactivation of a RAS association domain family protein from the lung tumour suppressor locus 3p21.3. Nat. Genet. 2000;25(3):315–319. doi: 10.1038/77083. [DOI] [PubMed] [Google Scholar]
  • 53.Lagos-Quintana M, Rauhut R, Lendeckel W, Tuschl T. Identification of novel genes coding for small expressed RNAs. Science. 2001;294(5543):853–858. doi: 10.1126/science.1064921. [DOI] [PubMed] [Google Scholar]
  • 54.Lau NC, Lim LP, Weinstein EG, Bartel DP. An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science. 2001;294(5543):858–862. doi: 10.1126/science.1065062. [DOI] [PubMed] [Google Scholar]
  • 55.Lee RC, Ambros V. An extensive class of small RNAs in Caenorhabditis elegans. Science. 2001;294(5543):862–864. doi: 10.1126/science.1065329. [DOI] [PubMed] [Google Scholar]
  • 56.Yanaihara N, Caplen N, Bowman E, Seike M, Kumamoto K, Yi M, Stephens RM, Okamoto A, Yokota J, Tanaka T, Calin GA, Liu CG, Croce CM, Harris CC. Unique microRNA molecular profiles in lung cancer diagnosis and prognosis. Cancer Cell. 2006;9(3):189–198. doi: 10.1016/j.ccr.2006.01.025. [DOI] [PubMed] [Google Scholar]
  • 57.Gee GV, Koestler DC, Christensen BC, Sugarbaker DJ, Ugolini D, Ivaldi GP, Resnick MB, Houseman EA, Kelsey KT, Marsit CJ. Downregulated MicroRNAs in the differential diagnosis of malignant pleural mesothelioma. Int. J. Cancer. 2010 doi: 10.1002/ijc.25285. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Fabbri M, Garzon R, Cimmino A, Liu Z, Zanesi N, Callegari E, Liu S, Alder H, Costinean S, Fernandez-Cymering C, Volinia S, Guler G, Morrison CD, Chan KK, Marcucci G, Calin GA, Huebner K, Croce CM. MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc. Natl. Acad. Sci. USA. 2007;104(40):15805–15810. doi: 10.1073/pnas.0707628104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Takahashi Y, Forrest AR, Maeno E, Hashimoto T, Daub CO, Yasuda J. MiR-107 and MiR-185 can induce cell cycle arrest in human non small cell lung cancer cell lines. PLoS One. 2009;4(8):e6677. doi: 10.1371/journal.pone.0006677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Bandi N, Zbinden S, Gugger M, Arnold M, Kocher V, Hasan L, Kappeler A, Brunner T, Vassella E. miR-15a and miR-16 are implicated in cell cycle regulation in a Rb-dependent manner and are frequently deleted or down-regulated in non-small cell lung cancer. Cancer Res. 2009;69(13):5553–5559. doi: 10.1158/0008-5472.CAN-08-4277. [DOI] [PubMed] [Google Scholar]
  • 61.Lantuéjoul S, Salon C, Soria JC, Brambilla E. Telomerase expression in lung preneoplasia and neoplasia. Int. J. Cancer. 2007;120:1835–1841. doi: 10.1002/ijc.22473. [DOI] [PubMed] [Google Scholar]
  • 62.Leidinger P, Keller A, Heisel S, Ludwig N, Rheinheimer S, Klein V, Andres C, Staratschek-Jox A, Wolf J, Stoelben E, Stephan B, Stehle I, Hamacher J, Huwer H, Lenhof HP, Meese E. Identification of lung cancer with high sensitivity and specificity by blood testing. Respir. Res. 2010;11:18. doi: 10.1186/1465-9921-11-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Goksel T, Akkoclu A. Pattern of lung cancer in Turkey, 1994-1998. Respiration. 2002;69(3):207–210. doi: 10.1159/000063621. [DOI] [PubMed] [Google Scholar]
  • 64.Bai H, Mao L, Wang HS, Zhao J, Yang L, An TT, Wang X, Duan CJ, Wu NM, Guo ZQ, Liu YX, Liu HN, Wang YY, Wang J. Epidermal Growth Factor Receptor Mutations in Plasma DNA Samples Predict Tumor Response in Chinese Patients With Stages IIIB to IV Non-Small-Cell Lung Cancer. J. Clin. Oncol. 2009;27:2653–2659. doi: 10.1200/JCO.2008.17.3930. [DOI] [PubMed] [Google Scholar]
  • 65.Ceppi P, Papotti M, Scagliotti G. New strategies for targeting the therapy of NSCLC: the role of ERCC1 and TS. Adv. Med. Sci. 2010;55(1):22–5. doi: 10.2478/v10039-010-0017-4. [DOI] [PubMed] [Google Scholar]
  • 66.Su C, Zhou S, Zhang L, Ren S, Xu J, Zhang J, Lv M, Zhang J, Zhou C. ERCC1, RRM1 and BRCA1 mRNA expression levels and clinical outcome of advanced non-small cell lung cancer. Med. Oncol. 2010 doi: 10.1007/s12032-010-9553-9. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]

Articles from Current Genomics are provided here courtesy of Bentham Science Publishers

RESOURCES