Genes associated to radiation response in NSCLC from genomics data
1. Introduction
Lung cancer remains as one of the most aggressive cancer types with nearly 1.6 million new cases worldwide each year. There are an estimated 222,520 new cases and 157,300 deaths from lung cancer in the United States in 2010 [1]. Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer, comprising three major histological subtypes: adenocarcinoma, squamous cell carcinoma, and large cell carcinoma. Chronic exposure to carcinogens drives genetic and epigenetic damage that can result in lung epithelial cells progressively acquiring growth and/or survival advantages, giving as a result the generation of tumor cells. Studies have shown that some specific molecules contribute to sporadic tumors of lung cancer; even now, they are useful as predictive biomarkers. Mutations in at least one of the established lung cancer driver genes including
2. Lung cancer
Lung cancer remains as one of the most aggressive cancer types with nearly 1.6 million new cases worldwide each year. In 2010, in the United States were estimated 222,520 new cases and 157,300 deaths from lung cancer [1]. Non-small cell lung cancer (NSCLC) subtype represents 85% of all cases of lung cancer, while small cell lung cancer (SCLC) subtype comprises 15%. Histologically, NSCLC is classified as adenocarcinoma, squamous cell carcinoma, and large cell carcinoma. This classification has important implications for the clinical management and prognosis of the disease [3]. Yet early detection methods are not extensively used in the wider population, malignancy is most commonly diagnosed at a late stage resulting in poor patient survival. Overall 5-year survival rates for lung cancer vary globally but are consistently low (7.5-16%) [1]. Approximately 40% of patients with advanced unresectable disease at the time of diagnosis have a poor prognosis. At present, no single chemo-radiation therapy regimen can be considered standard; despite the treatment choice for unresectable stage III NSCLC, a platinum-based chemotherapy regimen and thoracic radiation are concurrently administered. Chemotherapy concurrently with chest radiation therapy significantly improves the survival of patients with unresectable stage IIIA and IIIB disease. Decades of research have increased understanding the lung cancer as a multistep process involving genetic and epigenetic alterations, through which, resulting DNA damage transforms normal epithelial cells that progressively acquire growth and/or survival advantages until cancer arises [2,4-7]. Malignant transformation of lung epithelial cells is characterized by genetic instability, which can exist at the chromosomal level (with large-scale loss or gain of genomic material, translocations, and microsatellite instability) or at the nucleotide level (with single or several nucleotide base changes). Moreover, lung cancer is also related to genomic and epigenomic changes at the transcriptome (with altered gene and microRNA expression) and proteome [8-11] level. As many kinds of tumors, molecular abnormalities in lung cancer cells are typically targeted to proto-oncogenes, tumor suppressor genes, DNA repair genes, and other genes that can promote outgrowth and immortality of affected cells [12,13]. It is accepted that the successful discovery, validation and implementation of specific molecular markers for early diagnosis, clinical surveillance and determination of tumor response to therapeutic intervention could improve survival rates for patients, but only few biomarkers turned out to be useful in the clinic.
3. Genome biomarkers: The opening to personalized medicine in lung cancer
Nowadays, molecular and genetic studies have shown that some specific molecules contribute to sporadic tumors of lung cancer; they are useful as therapeutic targets and predictive biomarkers [16]. Recently, the National Cancer Institute’s lung cancer mutation consortium (NCI´s LCMC) performed such a study on more than 800 lung adenocarcinoma tumor specimens, examining mutations in established lung cancer driver genes (

Figure 1.
EGFR and KRAS mutations in NSCLC. Mutations in extracellular domain of EGFR have been implicated in resistance to treatment with mAb against EGFR. Mutations in TK domain are most common in NSCLC, including L858R and E746-A750 deletion in exon 19. These mutations are target for small molecules inhibitors of tyrosine kinases domain (TKI). T790M is a mutation related to resistance to TKI treatment. Mutations in codon 12 or 13 of
Mutated EGFR are present in 10-15% of NSCLC tumors [2,17]. Mutant EGFRs (either by exon 19 deletion or punctual mutation in exon 21 known as L858R) show an increased amount and duration of EGFR activation compared with wild-type receptors [18]. Mutated EGFR can activate RAS/RAF/MEK/MAPK and phosphoinositide 3-kinase (PI3K)/AKT and STAT3/STAT5 pathways [19-21]. Beside the importance of EGFR on lung carcinogenesis, some other molecules have been described as molecular markers for prognosis and therapeutic targets. Gene amplification and mutations in the kinase domain of C-erbB2 (HER-2/neu), a member of EGFR family, have been identified in patients with lung adenocarcinomas with a frequency of less than 5% and 5 to 10% respectively, and its overexpression are involved in ~25% of NSCLC cases [22]. EGFR and HER-2 kinase domain mutations have similar associations with female sex, non-smoking status and Asian background in patients with adenocarcinoma [15,22]. RAS/RAF/MEK/MAPK pathway is involved in signaling downstream from EGFR leading the growth and tumor progression in NSCLC. Activating
Some other molecules have been identified based on expression and genomic data such as MYC and Cyclin D1 which are amplified and over-expressed in 2.5–10% and 5% of NSCLC respectively, while BCL-2 over-expression is involved in ~25% of cases of NSCLC [8,16]. Recent data have shown that methylation of the promoter regions of genes is a common event in NSCLC, which contributes to oncogenes over-expression or tumor genes suppressors silenced. These epigenetic changes may be an early event in NSCLC, since that promoter region of p16 gene is frequently methylated in smokers and premalignant lesion of lung cancer [27]. PI3K-AKT-mTOR pathway is altered in NSCLC. AKT overexpression has been described in a subgroup of NSCLC tumors jointly with mutations or amplification of PIK3CA gene. These genomic modifications are related with enhanced activity of PI3-K pathway mainly in squamous cell carcinoma tumors [28]. On the other hand, tissues of smoker patients show higher levels of angiogenic factors such as VEGF. VEGF expression increases in relationship with tumoral grade, which in turn, correlates with increased microvessel density, development and poor prognosis of lung cancer. Tumoral angiogenesis and angiogenic factors are regulated by hypoxic inductor factor (HIF) 1α and 2α or through oncogenes as

Figure 2.
EGFR pathway in NSCLC. Mutations, amplification or overexpression of growth factors receptors such as EGFR, HER-2 and C-MET are most frequent in NSCLC tumors from non-smokers patients. All these genetic alterations have been observed commonly in adenocarcinomas, women and Asiatic ethnicity. EML4/ALK fusion gene is associated to NSCLC from young and non-smokers patients. KRAS mutations and signaling pathway depending to KRAS are most frequent in smoker patients. PI3K signaling pathway modifications are most frequently observed in squamous cell carcinomas.
4. Molecular and radiology therapies in lung cancer
4.1. Molecular therapy: Response and biological resistance
EGFR exhibits overexpression or aberrant activation in 50 to 90% of NSCLC. Mutations in EGFR allow sustained activation of EGF signaling for tumor cell survival, therefore, has been development targeted inhibitors for this molecule [16]. mAbs target the extracellular domain of EGFR and small molecules that inhibit intracellular EGFR tyrosine kinase domain function. In 2004, a significant advancement in the treatment of NSCLC was made following the observation that somatic mutations in the kinase domain of EGFR strongly correlated with sensitivity to EGFR TKIs [31, 32]. EGFR mutations are particularly prevalent in a patient subgroups with specific characteristics as adenocarcinoma histology, women, never smokers, and East Asian ethnicity [36]. This subgroup shows an exquisite sensitivity and marked tumor response to TKIs treatment. Despites the results obtained with biological therapies, there is a group of patients who do not respond to molecular therapy. Moreover, there is another group of patients with EGFR mutant lung cancer who initially respond to TKI treatment, but subsequently develop disease progression after a median of 10 to 14 months on treatment with biological therapy [37,38]. Hence, no optimal therapy thereafter has yet been established. Presumably, tumors do not respond because their molecular lesions are downstream of the therapeutic target [39]. Resistance to biologic therapy in NSCLC has been associated with EGFR exon-20 insertions [40] or a secondary T790M mutation [41], KRAS mutation [42], or amplification of the MET proto-oncogene [43,44], where MET is a transmembrane receptor with a tyrosine kinase domain, which activates signaling survival depending to PI3K and MAPK pathways. Of importance, Some reports showed that inhibition of MET signaling can restore sensitivity to TKIs [45]. HER-2 kinase domain mutations are associated with resistance to EGFR TKIs, but also with sensitivity to HER-2-targeted therapy [46].
Genomics data have provided information for developing targeted therapies in lung cancer patients based upon identification of cancer-specific vulnerabilities and set the stage for molecular biomarkers that provide information on clinical outcome and response to treatment. It has become clear that molecular-targeted cancer therapies can only reach their full potential through appropriate patient selection. In addition, there are now large clinical studies of lung cancer showing distinct chemotherapy and radiation responses. The majority of patients with lung cancer display advanced disease, these patients have obtained modest improvements in overall survival and quality of life through the use of systemic chemotherapy; however, the survival is still low, getting a median survival of 8 to 10 months [1]. Once recurred or metastasized, the disease is essentially incurable with survival rates at 5 years of less than 5%, and this has improved only marginally during the past 25 years [1]. The substantial genetic heterogeneity inherent to human cancers as an indicator of distinct phenotypes makes the identification of patients most likely to benefit from a given anticancer agent challenging. The description of molecules associated with resistance or sensitivity to cytotoxic treatments will improve personalized therapy for lung cancer. Radiotherapy, alone or in combination with surgery or chemotherapy, plays a critical role in the management of lung cancer. More than 60% of lung cancer patients receive radiotherapy at least once during the course of their disease [47].
4.2. Role of EGFR pathways in resistance and sensibility to radiotherapy
NSCLC tumors exhibit a wide response spectrum to radiation therapy but the molecular basis for this responsiveness is unknown. Some patients with NSCLC have a good response to radiation therapy with long-term local control while others relapse even with high dose treatment [48]. Many factors are involved in biological process of lung damage induced by radiation. At the molecular level, it is established that ionizing radiation causes various types of cellular damage; the creation of DNA breaks represents the principal damage induced by direct action of ionizing radiation or indirect action provoked by reactive species oxygen (ROS). Inadequately repaired DNA breaks leads to loss of cell clonogenicity via the generation of lethal chromosomal aberrations or the direct induction of apoptosis [49]. In addition to DNA breaks, ROS rapidly triggers the production of cytokines, growth factors, and more ROS, ultimately leading to chronic oxidative stress, hypoxia and the nonhealing tissue response in the lung [50,51]. Tumor radioresistance, including intrinsic resistance before treatments and acquired resistance during radiotherapy, is one of the main obstacles for radiotherapy efficiency for NSCLC. Some of the most important mechanisms associated with radioresistance in cancer including checkpoint pathway, mismatch repair process, and DNA damage repair [52-54]. Accumulating evidence suggests that radioresistance is often correlated with some genes, such as p53 [55] and EGFR [56]. In this regard, targeting EGFR pathway activation radiosensitizes human cancer cells [57-59], suggesting that the presence of overexpressed or activated oncogenes such as EGFR or RAS may be a mechanism for increased cellular resistance to radiation. In some models, it has been demonstrated that EGFR/Ras/Raf/MEK/ERK signaling may be activated in response to radiation, promoting cancer cell survival and proliferation [52-54,60] (Figure 3).
Variations in NSCLC responses to radiotherapy alone or in combination with chemotherapy or biological therapy are most likely due in the majority of cases to the genetic and epigenetic constitution of tumors [61,62]. In NSCLC, EGFR and KRAS oncogenes play an important role as prognostic factors; therefore, their role in radioresistance has been documented [63]. NSCLC cell lines harboring EGFR with mutations in tyrosine kinase domain were many folds more sensitive to radiation compared to cell lines with wild type EGFR. Radiosensitivity of NSCLC cell lines with mutant EGFR and human bronchial epithelial cells stably expressing mutant forms of EGFR was attributed to delayed DNA repair kinetics, defective radiation-induced arrest during DNA synthesis or mitosis, and pronounced increases in apoptosis or the occurrence of micronuclei [63]. Apparently, mutant EGFR is unable to translocate into the nucleus, which hinders its interaction with DNA-dependent protein kinase (DNA-PK), which is a fundamental enzyme for repair radiation-induced double strand breaks [63]. Besides of the promising role of mutant EGFR in radiosensitivity, the effort by blocking EGFR pathway to induce better response to radiotherapy has been limited. Inhibition of the EGFR by TKI or mAb, has been shown to

Figure 3.
Role of EGFR pathway in radioresistance and radiosensibility in NSCLC. Aberrantly activation of EGFR pathways, including receptor mutations, KRAS activation, PI3K/AKT/mTOR pathway activation allows expression of specific genes for to regulate apoptosis, DNA repair, cell cycle and cell proliferation in order to get resistance to radiation.
radiosensitize a limited number of NSCLC cell lines
5. Lung cancer radiogenomics
Radiotherapy has played a key role in the control of tumor growth in many cancer patients, including lung cancer. Studies that originated more than 40 years ago [72,73] have indicated that tumors respond to radiotherapy by initiating a process called accelerated repopulation. In this process, the few surviving cells that escaped death after exposure to radiotherapy or chemotherapy can rapidly repopulate the badly damaged tumor by proliferating at a markedly faster pace. This phenomenon suggested that tumoral heterogeneity permits a cell population in the tumor to have advantages to avoid cell death induced by radiation. Cellular senescence, DNA repair and cell cycle checkpoint are cellular mechanisms that influence the resistance to radiotherapy. However, the molecular mechanisms that regulate the radioresistance phenotype have not been clear in cancer. For this reason, some research groups have focused in the study of biological models to obtain genomic and proteomic signatures in order to find genes and proteins that could predict radiosensitivity or radioresistance in lung tumors (Table 1). Although such researches have contributed to a partial understanding of the mechanisms underlying cellular radioresistance, the comprehensive functional mechanisms remain largely elusive. This may be quite reasonable since the mechanisms of radioresistance are a complex multigene interaction. In this sense, Torres-Roca
c-Jun* | [75] |
HDAC-1 | |
RELA (p65 subunit NFkB | |
PKC-beta | |
Sumo1 | |
c-Ab1 | |
STAT1 | |
AR | |
CDK1 | |
IRF1 | |
[76] | |
XRCC5 | |
ERCC5 | |
ERCC1 | |
RAD9A | |
ERCC4 | |
[76] | |
MDM2* | |
BCL-2 | |
PKC-2 | |
PIM2 | |
[77] | |
DDB2 | |
LOX | |
CDH2 | |
CR4AB | |
Livin α* | [79] |
[77] | |
GBP-1 | |
CD83 | |
TNNC1 | |
TP53I3* | [78] |
Table 1.
The authors developed a radiation classifier to calculate the radiosensitivity of tumor cell lines based on basal gene expression profiles obtained from the literature. They predicted the survival fraction to 2 Gy (SF2) value in 22 of 35 cell lines from the National Cancer Institute, a result significantly different from chance (P = 0.0002). In their approach, radiation sensitivity as a continuous variable, significance analysis of microarrays is used for gene selection, and a multivariate linear regression model is used for radiosensitivity prediction. In gene selection, they identified three novel genes: RbAp48, RGS19, and R5PIA, whose expression values correlated with radiation sensitivity. Exogenous overexpression of RbAp48 into three cancer cell lines (HS-578T, MALME-3M, and MDA-MB-231) induced radiosensitization (1.5- to 2-fold), moreover, higher proportion of transfected cells with RbAp48 were in G2-M phase of the cell cycle (27% versus 5%). Finally, RbAp48 overexpression is correlated with dephosphorylation of Akt, suggesting that RbAp48 might be exerting its effect radiosensitized by antagonizing the Ras pathway, but it could also do so through PI3K. The authors establish that radiation sensitivity can be predicted based on gene expression profiles and they introduce a genomic approach to the identification of novel molecular markers of radiation sensitivity. Despite of results in different tumor cell lines, this work included only four NSCLC cell lines and they were able to predict correct SF2 values for only two of them [74]. So, the study should be performed on a broader panel of NSCLC cell lines. In lung cancer, multiple studies have identified a wide array of genetic and epigenetic alterations, including mutations in DNA sequence, DNA copy number changes, aberrant DNA promoter methylation, changes in mRNA, microRNAs and protein expression [8], revealing many potential determinants and signaling pathways governing lung tumorigenesis and progression. Gene expression profiling analysis allows for an increase in the understanding of the molecular mechanisms and pathways that involve radioresistance. Thus, the strategy followed by Torres-Roca and collaborators can be applied to gene expression data reported in lung cancer, in order to identify new molecular targets for radiotherapy response. In this sense, we know that the response of tumor cells to radiation is accompanied by complex changes in the gene expression pattern. Based on mRNA expression profiles and systems-biology approach, Eschrich
A problem in radiogenomics research is the difficulty to determine what fraction of the tumor cell population is radioresistant after a course of radiotherapy. For understand the radiation-mediated changes in gene expression that might result in different responses to radiation, Guo W
researches have been focused in describing specific molecules that revert the radioresistant phenotype. It is well known that there is a large amount of cell death during cytotoxic cancer therapy such as radiotherapy; therefore, radioresistance is associated with deregulation of apoptosis proteins. Sun
6. Proteomics of radiation response in lung cancer
Despite proteomics being useful to find molecular markers associated to lung cancer cells [82], in radiation resistance research there are very few studies focused on applying proteomics to find new markers associated to radiotherapy response in lung cancer. Recently, Wei R
7. Conclusion
One of the most important problems in lung oncology is lack of suitable biomarkers as therapeutic targets or the absence of predictors of therapy response. The genetic heterogeneity of the lung tumors influences the initial molecular resistance to therapies, but also in the development of resistance during treatment. The molecular mechanisms that influence the resistance to biological or radiological treatments, referring to the resistance mechanisms occurring naturally because of the carcinogenic process, or those developing as a result of evolutionary pressure that tumor cells undergoing during the treatment administration, is a barrier that has not been fully elucidated. With current genomics and proteomics studies in lung cancer focused on solving the mystery of therapeutic resistance, it has been possible to identify molecules that may serve as prognostic markers of response to radiological and molecular therapy resistance. Genes and proteins that regulate cell proliferation and survival, including signaling molecules and transcription factors such as KRAS, BRAF, PI3K, MAPK, mTOR, JAK2, STAT, survivin and others have demonstrated to be part of the molecular machinery that regulates therapeutic resistance. Moreover, gene and protein expression profiling of lung cancer has focused specifically on searching predictive markers to radiotherapy. Some studies have generated data on molecules involved in radioresistance or radiosensitivity either natural or acquired. Using therapeutic doses of radiation in
Acknowledgement
Authors gratefully acknowledge the financial support from the National Council of Science and Technology (CONACyT), Mexico (grants 115552 and 115591), and The Institute of Science and Technology (ICyT-DF), Mexico (grant PIUTE147).
References
- 1.
Global cancer statistics. CA Cancer Journal for ClinicianJemal A Bray F Center M. M Ferlay J Ward E Forman D 2011 61 2 69 90 - 2.
Identification of driver mutations in tumor specimens from 1000 patients with lung adenocarcinoma: the NCI’s lung cancer mutation consortium (LCMC) [abstract CRA7506]. Journal of Clinical OncologyKris M. G Johnson B. E Kwiatkowski D. J Iafrate A. J Wistuba I. I Aronson S. L Engelman J. A Shyr Y Khuri F. R Rudin C. M Garon E. B Pao W Schiller J. H Haura E. B Shirai K Giaccone G Berry L. D Kugler K Minna J. D Bunn P. A 2011 Suppl). - 3.
Lung cancer preneoplasia. Annual Review of PathologyWistuba I. I Gazdar A. F 2006 1 331 - 4.
Progress in understanding the molecular pathogenesis of human lung cancer. Biochemical and Biophysical ActaSekido Y Fong K. M Minna J. D 1998 F21 59 - 5.
The changing epidemiology of lung cancer with a focus on screening. British Medical JournalSilvestri G. A Alberg A. J Ravenel J 2009 339 451 - 6.
Clonal genetic alterations in the lungs of current and former smokers. Journal of National Cancer InstituteMao L Lee J. S Kurie J. M Fan Y. H Lippman S. M Lee J. J Ro J. Y Broxson A Yu R Morice R. C Kemp B. L Khuri F. R Walsh G. L Hittelman W. N Hong W. K 1997 89 12 857 862 - 7.
Lung cancer in never smokers-a different disease. Nature Reviews CancerSun S Schiller J. H Gazdar A. F 2007 7 10 778 790 - 8.
Prognostic molecular markers in non-small cell lung cancer. Lung CancerNikliński J Niklińska W Laudanski J Chyczewska E Chyczewski L 2001 4 Suppl 2 S53-58. - 9.
Mechanistic roles of noncoding RNAs in lung cancer biology and their clinical implications. Genetic Research InternationalEnfield K. S Pikor L. A Martinez V. D Lam W. L 2012 ID:737416. - 10.
Gene-Promoter Hypermethylation as a biomarker in lung cancer. Nature Reviews CancerBelinsky S. A 2004 4 9 707 717 - 11.
Proteomic approaches in lung cancer biomarker development. Expert Review of ProteomicsCho J. Y Sung H. J 2009 6 1 27 42 - 12.
A translational view of the molecular pathogenesis of lung cancer. Journal of Thoracic OncologySato M Shames D. S Gazdar A. F Minna J. D 2007 2 4 327 343 - 13.
The mutation spectrum revealed by paired genome sequences from a lung cancer patient. NatureLee W Jiang Z Liu J Haverty P. M Guan Y Stinson J Yue P Zhang Y Pant K. P Bhatt D Ha C Johnson S Kennemer M. I Mohan S Nazarenko I Watanabe C Sparks A. B Shames D. S Gentleman R De Sauvage F. J Stern H Pandita A Ballinger D. G Drmanac R Modrusan Z Seshagiri S Zhang Z 2010 465 7297 473 477 - 14.
Tsao MS. K-ras mutations in non-small-cell lung carcinoma: a review. Clinical Lung CancerAviel-ronen S Blackhall F. H Shepherd F. A 2006 8 1 30 38 - 15.
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 Science USAPao 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 2004 101 36 13306 13311 - 16.
Key signaling pathways and targets in lung cancer therapy. Clinical Lung CancerChoong N. W Salgia R Vokes E. E 2007 Suppl 2 S52 S60. - 17.
EGFR tyrosine kinase domain mutations are detected in histologically normal respiratory epithelium in lung cancer patients. Cancer ResearchTang X Shigematsu H Bekele B. N Roth J. A Minna J. D Hong W. K Gazdar A. F Wistuba I. I 2005 65 17 7568 7572 - 18.
Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. New England Journal of MedicineLynch T. J Bell D. W Sordella R Gurubhagavatula S Okimoto R. A Brannigan B. W Harris P. L Haserlat S. M Supko J. G Haluska F. G Louis D. N Christiani D. C Settleman J Haber D. A 2004 350 21 2129 2139 - 19.
Gefitinib sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. ScienceSordella R Bell D. W Haber D. A Settleman J 2004 305 5687 1163 1167 - 20.
Epidermal growth factor receptor and K-Ras in non-small cell lung cancer-molecular pathways involved and targeted therapies. World Journal of Clinical OncologyDe Mello R. A Marques D. S Medeiros R Araujo A. M 2011 2 11 367 376 - 21.
Blocking the PI3K/AKT and MEK/ERK signaling pathways can overcome gefitinib-resistance in non-small cell lung cancer cell lines. Advances in Medical ScienceLi H Schmid-bindert G Wang D Zhao Y Yang X Su B Zhou C 2011 56 2 275 284 - 22.
Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer ResearchShigematsu H Takahashi T Nomura M Majmudar K Suzuki M Lee H Wistuba I. I Fong K. M Toyooka S Shimizu N Fujisawa T Minna J. D Gazdar A. F 2005 65 5 1642 1646 - 23.
Frequency and distinctive spectrum of KRAS mutations in never smokers with lung adenocarcinoma. Clinical Cancer ResearchRiely G. J Kris M. G Rosenbaum D Marks J Li A Chitale D. A Nafa K Riedel E. R Hsu M Pao W Miller V. A Ladanyi M 2008 14 18 5731 5734 - 24.
Clinical significance of BRAF gene mutations in patients with non-small cell lung cancer. Anticancer ResearchKobayashi M Sonobe M Takahashi T Yoshizawa A Ishikawa M Kikuchi R Okubo K Huang C. L Date H 2011 31 12 4619 4623 - 25.
Identification of the transforming EML4-ALK fusion gene in non-small cell lung cancer. NatureSoda M Choi Y. L Enomoto M Takada S Yamashita Y Ishikawa S Fujiwara S Watanabe H Kurashina K Hatanaka H Bando M Ohno S Ishikawa Y Aburatani H Niki T Sohara Y Sugiyama Y Mano H 2007 448 7153 561 566 - 26.
BRAF and RAS mutations in human lung cancer and melanoma. Cancer ResearchBrose M. S Volpe P Feldman M Kumar M Rishi I Gerrero R Einhorn E Herlyn M Minna J Nicholson A Roth J. A Albelda S. M Davies H Cox C Brignell G Stephens P Futreal P. A Wooster R Stratton M. R Weber B. L 2002 62 23 6997 7000 - 27.
Promoter hypermethylation of multiple genes in sputum precedes lung cancer incidence in a high-risk cohort. Cancer ResearchBelinsky S. A Liechty K. C Gentry F. D Wolf H. J Rogers J Vu K Haney J Kennedy T. C Hirsch F. R Miller Y Franklin W. A Herman J. G Baylin S. B Bunn P. A Byers T 2006 66 6 3338 3344 - 28.
Clarifying the spectrum of driver oncogene mutations in biomarker-verified squamous carcinoma of lung: lack of EGFR/KRAS and presence of PIK3CA/AKT1 mutations. Clinical Cancer ResearchRekhtman N Paik P. K Arcila M. E Tafe L. J Oxnard G. R Moreira A. L Travis W. D Zakowski M. F Kris M. G Ladanyi M 2012 18 4 1167 1176 - 29.
Prognostic role of angiogenesis in non-small cell lung cancer. Anticancer ResearchGiatromanolaki A 2001 B4373 4382 - 30.
Molecular biomarkers of lung carcinoma. Front Bioscience. (Elite Ed).Vrabec-branica B Gajovic S 2012 4 865 - 31.
Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. New England of Journal MedicineLynch T. J Bell D. W Sordella R Gurubhagavatula S Okimoto R. A Brannigan B. W Harris P. L Haserlat S. M Supko J. G Haluska F. G Louis D. N Christiani D. C Settleman J Haber D. A 2004 350 21 2129 2139 - 32.
EGFR mutations in lung cancer: correlation with clinical response to gefitinib therapy. SciencePaez J. G Jänne P. A Lee J. C Tracy S Greulich H Gabriel S Herman P Kaye F. J Lindeman N Boggon T. J Naoki K Sasaki H Fujii Y Eck M. J Sellers W. R Johnson B. E Meyerson M 2004 304 5676 1497 500 - 33.
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 OncologyEberhard D. A Johnson B. E Amler L. C Goddard A. D Heldens S. L Herbst R. S Ince W. L Jänne P. A Januario T Johnson D. H Klein P Miller V. A Ostland M. A Ramies D. A Sebisanovic D Stinson J. A Zhang Y. R Seshagiri S Hillan K. J 2005 23 25 5900 5909 - 34.
Non-small-cell lung cancers with kinase domain mutations in the epidermal growth factor receptor are sensitive to ionizing radiation. Cancer ResearchDas A. K Sato M Story M. D 2006 66 19 9601 9608 - 35.
Bunn PA Jr. Targeted therapies for non-small-cell lung cancer: biology, rationale, and preclinical results from a radiation oncology perspective. International Journal of Radiation Oncology Biology PhysicRaben D Helfrich B 2004 Suppl)27 38 - 36.
A comparison of epidermal growth factor receptor levels and other prognostic parameters in non-small cell lung cancer. European Journal of CancerFujino S Enokibori T Tezuka N Asada Y Inoue S Kato H Mori A 1996 A(12) 2070-2074. - 37.
Garcia Campelo R, Moreno MA, Catot S, Rolfo C, Reguart N, Palmero R, Sánchez JM, Bastus R, Mayo C, Bertran-Alamillo J, Molina MA, Sanchez JJ, Taron M; Spanish Lung Cancer Group. Screening for epidermal growth factor receptor mutations in lung cancer. New England of Journal MedicineRosell R Moran T Queralt C Porta R Cardenal F Camps C Majem M Lopez-vivanco G Isla D Provencio M Insa A Massuti B Gonzalez-larriba J. L Paz-ares L Bover I 2009 361 10 958 967 - 38.
Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. New England Journal of MedicineMok T. S Wu Y. L Thongprasert S Yang C. H Chu D. T Saijo N Sunpaweravong P Han B Margono B Ichinose Y Nishiwaki Y Ohe Y Yang J. J Chewaskulyong B Jiang H Duffield E. L Watkins C. L Armour A. A Fukuoka M 2009 361 10 947 957 - 39.
Activating and resistance mutations of EGFR in non-small-cell lung cancer: role in clinical response to EGFR tyrosine kinase inhibitors. OncogeneGazdar A. F 2009 Suppl 1) S24 31 - 40.
EGFR exon 20 insertion mutations in non-small-cell lung cancer: preclinical data and clinical implications. Lancet OncologyYasuda H Kobayashi S Costa D. B 2012 e23 31 - 41.
Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS MedicinePao W Miller V. A Politi K. A Riely G. J Somwar R Zakowski M. F Kris M. G Varmus H 2005 e73. - 42.
KRAS mutations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib. PLoS MedicinePao W Wang T. Y Riely G. J Miller V. A Pan Q Ladanyi M Zakowski M. F Heelan R. T Kris M. G Varmus H. E 2005 e17. - 43.
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 Science USABean J Brennan C Shih J. Y Riely G Viale A Wang L Chitale D Motoi N Szoke J Broderick S Balak M Chang W. C Yu C. J Gazdar A Pass H Rusch V Gerald W Huang S. F Yang P. C Miller V Ladanyi M Yang C. H Pao W 2007 104 52 20932 20937 - 44.
MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. ScienceEngelman J. A Zejnullahu K Mitsudomi T Song Y Hyland C Park J. O Lindeman N Gale C. M Zhao X Christensen J Kosaka T Holmes A. J Rogers A. M Cappuzzo F Mok T Lee C Johnson B. E Cantley L. C Jänne P. A 2007 316 5827 1039 1043 - 45.
Combined EGFR/MET or EGFR/HSP90 inhibition is effective in the treatment of lung cancers codriven by mutant EGFR containing T790M and MET. Cancer ResearchXu L Kikuchi E Xu C Ebi H Ercan D Cheng K. A Padera R Engelman J. A Jänne P. A Shapiro G. I Shimamura T Wong K. K 2012 72 13 3302 3311 - 46.
HER2 kinase domain mutation results in constitutive phosphorylation and activation of HER2 and EGFR and resistance to EGFR tyrosine kinase inhibitors. Cancer CellWang S. E Narasanna A Perez-torres M Xiang B Wu F. Y Yang S Carpenter G Gazdar A. F Muthuswamy S. K Arteaga C. L 2006 10 25 - 47.
Ten Haken RK, Schipper MJ, Sullivan MA, Chen M, Lopez C, Kalemkerian GP, Hayman JA. High-dose radiation improved local tumor control and overall survival in patients with inoperable/unresectable non-small-cell lung cancer: long-term results of a radiation dose escalation study. International Journal of Radiation Oncology Biology PhysicsKong F. M 2005 63 2 324 333 - 48.
Curran WJ; Radiation Therapy Oncology Group. Phase II trial of postoperative adjuvant paclitaxel/carboplatin and thoracic radiotherapy in resected stage II and IIIA non-small-cell lung cancer: promising long-term results of the Radiation Therapy Oncology Group-RTOG 9705. Journal of Clinical OncologyBradley J. D Paulus R Graham M. V Ettinger D. S Johnstone D. W Pilepich M. V Machtay M Komaki R Atkins J 2005 23 15 3480 3487 - 49.
Repair of radiation damage to DNA. British Journal of CancerWillers H Dahm-daphi J Powell S. N 2004 90 1297 - 50.
Small W Jr, Folz R, Anscher MS. Radiation-induced lung injury. Seminars in Radiation OncologyMarks L. B Yu X Vujaskovic Z 2003 13 3 333 345 - 51.
Using biological markers to predict risk of radiation injury. Seminars Radiation OncologyFleckenstein K Gauter-fleckenstein B Jackson I. L Rabbani Z Anscher M Vujaskovic Z 2007 17 2 89 98 - 52.
Ionizing radiation activates Erb-B receptor dependent Akt andContessa J. N Hampton J Lammering G Mikkelsen R. B Dent P Valerie K Schmidt-ullrich R. K 70 S6 kinase signaling in carcinoma cells. Oncogene2002 - 53.
Direct evidence for the contribution of activated N-ras and K-ras oncogenes to increased intrinsic radiation resistance in human tumor cell lines. Cancer ResearchBernhard E. J Stanbridge E. J Gupta S Gupta A. K Soto D Bakanauskas V. J Cerniglia G. J Muschel R. J Mckenna W. G 2000 60 23 6597 6600 - 54.
Radiation-induced EGFR signaling and control of DNA-damage repair. International Journal of Radiation BiologyRodemann H. P Dittmann K Toulany M 2007 83 781 - 55.
gene mutation and increased radiosensitivity in rat lung embryo epithelial cells during neoplastic development. Cancer ResearchBiard D. S Martin M Rhun Y. L Duthu A Lefaix J. L May E May P Concomitant p 1994 54 13 3361 3364 - 56.
Further characterization of the epidermal growth factor receptor ligand 11C-PD153035. Chinese Medical Journal (Engl)Wang H Yu J. M Yang G. R Song X. R Sun X. R Zhao S. Q Wang X. W Zhao W 2007 120 11 960 964 - 57.
Molecular mechanisms of resistance to therapies targeting the epidermal growth factor receptor. Clinical Cancer ResearchCamp E. R Summy J Bauer T. W Liu W Gallick G. E Ellis L. M 2005 11 397 - 58.
Mechanisms of enhanced radiation response following epidermal growth factor receptor signaling inhibition by erlotinib (Tarceva). Cancer Research.Chinnaiyan P Huang S Vallabhaneni G Armstrong E Varambally S Tomlins S. A Chinnaiyan A. M Harari P. M 2005 65 8 3328 3335 - 59.
Radiation-induced proliferation of the human A431 squamous carcinoma cells is dependent on EGFR tyrosine phosphorylation. OncogeneSchmidt-ullrich R. K Mikkelsen R. B Dent P Todd D. G Valerie K Kavanagh B. D Contessa J. N Rorrer W. K Chen P. B 1997 15 10 1191 1197 - 60.
Epidermal growth factor and ionizing radiation up-regulate the DNA repair genes XRCC1 and ERCC1 in DU145 and LNCaP prostate carcinoma through MAPK signaling. Radiation ResearchYacoub A Mckinstry R Hinman D Chung T Dent P Hagan M. P 2003 159 439 - 61.
Integration of molecular profiling into the lung cancer clinic. Clinical Cancer ResearchPao W Kris M. G Iafrate A. J 2009 15 17 5317 5322 - 62.
Tumor mRNA expression profiles predict responses to chemotherapy. Journal of Clinical OncologyMinna J. D Girard L Xie Y 2007 25 28 4329 4336 - 63.
Somatic mutations in the tyrosine kinase domain of epidermal growth factor receptor (EGFR) abrogate EGFR-mediated radioprotection in non-small cell lung carcinoma. Cancer ResearchDas A. K Chen B. P Story M. D 2007 67 11 5267 5274 - 64.
Mechanisms of enhanced radiation response following epidermal growth factor receptor signaling inhibition by erlotinib (Tarceva). Cancer ResearchChinnaiyan P Huang S Vallabhaneni G 2005 65 8 3328 3335 - 65.
Targeted therapy against VEGFR and EGFR with ZD6474 enhances the therapeutic efficacy of irradiation in an orthotopic model of human non-small-cell lung cancer. International Journal of Radiation Oncology Biology PhysicsShibuya K Komaki R Shintani T 2007 69 5 1534 1543 - 66.
EGF receptor inhibition radiosensitizes NSCLC cells by inducing senescence in cells sustaining DNA double-strand breaks. Cancer ResearchWang M Morsbach F Sander D Gheorghiu L Nanda A Benes C Kriegs M Krause M Dikomey E Baumann M Dahm-daphi J Settleman J Willers H 2011 71 19 6261 6269 - 67.
The epidermal growth factor receptor modulates DNA double-strand break repair by regulating non-homologous end-joining. DNA Repair (Amst).Kriegs M Kasten-pisula U Rieckmann T Holst K Saker J Dahm-daphi J Dikomey E 2010 9 8 889 897 - 68.
Requirement of Tyr-992 and Tyr-1173 in phosphorylation of the epidermal growth factor receptor by ionizing radiation and modulation by SHP2. Journal of Biological ChemistrySturla L. M Amorino G Alexander M. S Mikkelsen R. B Valerie K Schmidt-ullrichr R. K 2005 280 14597 - 69.
Radioresistance of K-Ras mutated human tumor cells is mediated through EGFR-dependent activation of PI3K-AKT pathway. Radiotherapy OncologyToulany M Dittmann K Kruger M Baumann M Rodemann H. P 2005 76 143 - 70.
Inhibition of JAK2 signaling by TG101209 enhances radiotherapy in lung cancer models. Journal of Thoracic OncologySun Y Moretti L Giacalone N. J Schleicher S Speirs C. K Carbone D. P Lu B 2011 6 4 699 706 - 71.
Survivin as a therapeutic target for radiation sensitization in lung cancer. Cancer Research.Lu B Mu Y Cao C Zeng F Schneider S Tan J Price J Chen J Freeman M Hallahan D. E 2004 64 2840 - 72.
Changes of cell proliferation characteristics in a rat rhabdomyosarcoma before and after x-irradiation. European Journal of CancerHermens A. F Barendsen G. W 1969 5 173 - 73.
Repopulation of gamma-irradiated Lewis lung carcinoma by malignant cells and host macrophage progenitors. British Journal of CancerStephens T. C Currie G. A Peacock J. H 1978 38 5 573 582 - 74.
Prediction of radiation sensitivity using a gene expression classifier. Cancer ResearchTorres-roca J. F Eschrich S Zhao H Bloom G Sung J Mccarthy S Cantor A. B Scuto A Li C Zhang S Jove R Yeatman T 2005 65 16 7169 7176 - 75.
Systems biology modeling of the radiation sensitivity network: a biomarker discovery platform. International Journal of Radiation Oncology Biology PhysicsEschrich S Zhang H Zhao H 2009 75 2 497 505 - 76.
Identification of differentially expressed genes contributing to radioresistance in lung cancer cells using microarray analysis. Radiation ResearchGuo W. F Lin R. X Huang J Zhou Z Yang J Guo G. Z Wang S. Q 2005 164 27 - 77.
Identification of differential gene expression profiles of radioresistant lung cancer cell line established by fractionated ionizing radiation in vitro. Chinese Medicine JournalQing-yong X Yuan G Yan L Wei-zhi Y Xiang-ying X 2008 121 18 1830 1837 - 78.
Differential gene expression profiles of radioresistant non-small-cell lung cancer cell lines established by fractionated irradiation: tumor proteinLee Y. S Oh J. H Yoon S Kwon M. S Song C. W Kim K. H Cho M. J Mollah M. L Je Y. J Kim Y. D Kim C. D Lee J. H 53 protein 3 confers sensitivity to ionizing radiation. International Journal of Radiation Oncology Biology Physics2010 - 79.
Role of inhibitor of apoptosis protein Livin in radiation resistance in non small cell lung cancer. Cancer Biotherapy and RadiopharmacySun J. G Liao R. X Zhang S. X Duan Y. Z Zhuo W. L Wang X. X Wang Z. X Li D. Z Chen Z. T 2011 26 5 585 592 - 80.
PTEN/pAkt/Jung I. L Kang H. J Kim K. C Kim I. G 53 signaling pathway correlates with the radioresponse of non-small cell lung cancer. International Journal of Molecular Medicine2010 - 81.
Overexpression of STAT3 potentiates growth, survival, and radioresistance of non-small-cell lung cancer (NSCLC) cells. Journal of Surgical ResearchYin Z. J Jin F. G Liu T. G Fu E. Q Xie Y. H Sun R. L 2011 171 2 675 683 - 82.
Mass spectrometry-based proteomics: The road to lung cancer biomarker discovery. Mass Spectrometry Reviews.Indovina P Marcelli E Pentimalli F Tanganelli P Tarro G Giordano A 2012 DOI: mas.21355. - 83.
Comparative proteomic and radiobiological analyses in human lung adenocarcinoma cells. Molecular and Cellular BiochemistryWei R Zhang Y Shen L Jiang W Li C Zhong M Xie Y Yang D He L Zhou Q 2012