Non-Smoking-Associated Lung Cancer: A distinct Entity in Terms of Tumor Biology, Patient Characteristics and Impact of Hereditary Cancer Predisposition
Abstract
:1. Introduction
2. Squamous Cell Carcinoma in Non- or Never-Smokers
3. Adenocarcinoma in Non- or Never-Smokers: Patient Characteristics
4. Physical Inactivity
5. Asbestos and Radon
6. Immunological Changes: Tumor Microenvironment in Never-Smokers
7. Anaplastic Lymphoma Tyrosine Kinase -Rearrangement in Lung Adenocarcinoma in Non- and Never-Smokers
8. Conclusions
Funding
Conflicts of Interest
References
- Global Burden of Disease Cancer Collaboration. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived with Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol. 2017, 3, 524–548. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.; Naishadham, D.; Jemal, A. Cancer statistics, 2013. CA Cancer J. Clin. 2013, 63, 11–30. [Google Scholar] [CrossRef] [PubMed]
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
- Park, Y.R.; Bae, S.H.; Ji, W.; Seo, E.J.; Lee, J.C.; Kim, H.R.; Jang, S.J.; Chang-Min, C. GAB2 Amplification in Squamous Cell Lung Cancer of Non-Smokers. J. Korean Med. Sci. 2017, 32, 1784–1791. [Google Scholar] [CrossRef] [PubMed]
- Okazaki, I.; Ishikawa, S.; Ando, W.; Sohara, Y. Lung Adenocarcinoma in Never Smokers: Problems of Primary Prevention from Aspects of Susceptible Genes and Carcinogens. Anticancer Res. 2016, 36, 6207–6224. [Google Scholar] [CrossRef]
- Couraud, S.; Zalcman, G.; Milleron, B.; Morin, F.; Souquet, P.J. Lung cancer in never smokers–a review. Eur. J. Cancer 2012, 48, 1299–1311. [Google Scholar] [CrossRef] [PubMed]
- Sun, S.; Schiller, J.H.; Gazdar, A.F. Lung cancer in never smokers—a different disease. Nat. Rev. Cancer 2007, 7, 778–790. [Google Scholar] [CrossRef]
- Thun, M.J.; Hannan, L.M.; Adams-Campbell, L.L.; Boffetta, P.; Buring, J.E.; Feskanich, D.; Flanders, D.W.; Jee, S.H.; Katanoda, K.; Kolonel, L.N.; et al. Lung cancer occurrence in never-smokers: An analysis of 13 cohorts and 22 cancer registry studies. PLoS Med. 2008, 30, e185. [Google Scholar] [CrossRef]
- Kawaguchi, T.; Takada, M.; Kubo, A.; Matsumura, A.; Fukai, S.; Tamura, A.; Saito, R.; Kawahara, M.; Maruyama, Y. Gender, histology, and time of diagnosis are important factors for prognosis: Analysis of 1499 never-smokers with advanced non-small cell lung cancer in Japan. J. Thorac. Oncol. 2010, 5, 1011–1017. [Google Scholar] [CrossRef]
- Pao, W.; Miller, V.; Zakowski, M.; Doherty, J.; Politi, K.; Sarkaria, I.; Singh, B.; Heelan, R.; Rusch, V.; Fulton, L.; 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. Proc. Natl. Acad. Sci. USA 2004, 101, 13306–13311. [Google Scholar] [CrossRef]
- Samet, J.M. Lung Cancer, Smoking, and Obesity: It’s Complicated. J. Natl. Cancer Inst. 2018, 110, 795–796. [Google Scholar] [CrossRef] [PubMed]
- Samet, J.M. Tobacco smoking: The leading cause of preventable disease worldwide. Thorac. Surg. Clin. 2013, 23, 103–112. [Google Scholar] [CrossRef] [PubMed]
- Lynch, 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.; et al. 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. [Google Scholar] [CrossRef]
- Koo, L.C.; Ho, J.H. Worldwide epidemiological patterns of lung cancer in nonsmokers. Int. J. Epidemiol. 1990, 19, S14–S23. [Google Scholar] [CrossRef] [PubMed]
- Nordquist, L.T.; Simon, G.R.; Cantor, A.; Alberts, W.M.; Bepler, G. Improved survival in never-smokers vs current smokers with primary adenocarcinoma of the lung. Chest 2004, 126, 347–351. [Google Scholar] [CrossRef] [PubMed]
- Paez, J.G.; Janne, P.A.; Lee, J.C.; Tracy, S.; Greulich, H.; Gabriel, S.; Herman, P.; Kaye, F.J.; Lindeman, N.; Boggon, T.J.; et al. EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science 2004, 304, 1497–1500. [Google Scholar] [CrossRef] [PubMed]
- Malhotra, J.; Malvezzi, M.; Negri, E.; La Vecchia, C.; Boffetta, P. Risk factors for lung cancer worldwide. Eur. Respir. J. 2016, 48, 889–902. [Google Scholar] [CrossRef]
- Lee, D.H.; Lee, J.S.; Wang, J.; Hsia, T.C.; Wang, X.; Kim, J.; Orlando, M. Pemetrexed-Erlotinib, Pemetrexed Alone, or Erlotinib Alone as Second-Line Treatment for East Asian and Non-East Asian Never-Smokers with Locally Advanced or Metastatic Nonsquamous Non-small Cell Lung Cancer: Exploratory Subgroup Analysis of a Phase II Trial. Cancer Res. Treat. 2015, 47, 616–629. [Google Scholar]
- Miller, V.A.; Kris, M.G.; Shah, N.; Patel, J.; Azzoli, C.; Gomez, J.; Krug, L.M.; Pao, W.; Rizvi, N.; Pizzo, B.; et al. Bronchioloalveolar pathologic subtype and smoking history predict sensitivity to gefitinib in advanced non-small-cell lung cancer. J. Clin. Oncol. 2004, 22, 1103–1109. [Google Scholar] [CrossRef]
- Shigematsu, H.; Lin, L.; Takahashi, T.; Nomura, M.; Suzuki, M.; Wistuba, I.I.; Fong, K.M.; Lee, H.; Toyooka, S.; Shimizu, N.; et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J. Natl. Cancer Inst. 2005, 97, 339–346. [Google Scholar] [CrossRef]
- Islami, F.; Torre, L.A.; Jemal, A. Global trends of lung cancer mortality and smoking prevalence. Transl. Lung Cancer Res. 2015, 4, 327–338. [Google Scholar] [PubMed]
- Choi, J.R.; Park, S.Y.; Noh, O.K.; Koh, Y.W.; Kang, D.R. Gene mutation discovery research of non-smoking lung cancer patients due to indoor radon exposure. Ann. Occup. Environ. Med. 2016, 28, 13. [Google Scholar] [CrossRef] [PubMed]
- Secretan, B.; Straif, K.; Baan, R.; Grosse, Y.; El Ghissassi, F.; Bouvard, V.; Benbrahim-Tallaa, L.; Guha, N.; Freeman, C.; Galichet, L.; et al. A review of human carcinogens--Part E: Tobacco, areca nut, alcohol, coal smoke, and salted fish. Lancet Oncol. 2009, 10, 1033–1034. [Google Scholar] [CrossRef]
- Cheng, Y.W.; Chiou, H.L.; Sheu, G.T.; Hsieh, L.L.; Chen, J.T.; Chen, C.Y.; Su, J.M.; Lee, H. The association of human papillomavirus 16/18 infection with lung cancer among nonsmoking Taiwanese women. Cancer Res. 2001, 61, 2799–2803. [Google Scholar] [PubMed]
- Liang, H.Y.; Li, X.L.; Yu, X.S.; Guan, P.; Yin, Z.H.; He, Q.C.; Zhou, B.S. Facts and fiction of the relationship between preexisting tuberculosis and lung cancer risk: A systematic review. Int. J. Cancer 2009, 125, 2936–2944. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Li, J.; Wu, P.; Zhou, L.; Lu, B.; Ying, K.; Chen, E.; Lu, Y.; Liu, P. Smoker and non-smoker lung adenocarcinoma is characterized by distinct tumor immune microenvironments. Oncoimmunology 2018, 7, e1494677. [Google Scholar] [CrossRef] [PubMed]
- Brunelli, M.; Bria, E.; Nottegar, A.; Cingarlini, S.; Simionato, F.; Calio, A.; Eccher, A.; Parolini, C.; Iannucci, A.; Gilioli, E.; et al. True 3q chromosomal amplification in squamous cell lung carcinoma by FISH and aCGH molecular analysis: Impact on targeted drugs. PLoS ONE 2012, 7, e49689. [Google Scholar] [CrossRef]
- Cancer Genome Atlas Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012, 489, 519–525. [Google Scholar] [CrossRef]
- Forbes, S.A.; Beare, D.; Gunasekaran, P.; Leung, K.; Bindal, N.; Boutselakis, H.; Ding, M.; Bamford, S.; Cole, C.; Ward, S.; et al. COSMIC: Exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015, 43, D805–CD811. [Google Scholar] [CrossRef]
- Baldassarri, M.; Fallerini, C.; Cetta, F.; Ghisalberti, M.; Bellan, C.; Furini, S.; Spiga, O.; Crispino, S.; Gotti, G.; Ariani, F. Omic Approach in Non-smoker Female with Lung Squamous Cell Carcinoma Pinpoints to Germline Susceptibility and Personalized Medicine. Cancer Res. Treat. 2018, 50, 356–365. [Google Scholar] [CrossRef]
- Vanni, I.; Coco, S.; Bonfiglio, S.; Cittaro, D.; Genova, C.; Biello, F.; Mora, M.; Rossella, V.; Dal Bello, M.G.; Truini, A.; et al. Whole exome sequencing of independent lung adenocarcinoma, lung squamous cell carcinoma, and malignant peritoneal mesothelioma: A case report. Medicine (Baltimore) 2016, 95, e5447. [Google Scholar] [CrossRef] [PubMed]
- Tong, L. Acetyl-coenzyme A carboxylase: Crucial metabolic enzyme and attractive target for drug discovery. Cell Mol. Life Sci. 2005, 62, 1784–1803. [Google Scholar] [CrossRef] [PubMed]
- Garassino, M.C.; Crino, L.; Catino, A.; Ardizzoni, A.; Cortesi, E.; Cappuzzo, F.; Bordi, P.; Calabrò, L.; Barbieri, F.; Santo, A.; et al. Nivolumab in never-smokers with advanced squamous non-small cell lung cancer: Results from the Italian cohort of an expanded access program. Tumour Biol. 2018, 40, 1010428318815047. [Google Scholar] [CrossRef] [PubMed]
- Bonanno, L.; Calabrese, F.; Nardo, G.; Calistri, D.; Tebaldi, M.; Tedaldi, G.; Polo, V.; Vuljan, S.; Favaretto, A.; Conte, P.; et al. Morphological and genetic heterogeneity in multifocal lung adenocarcinoma: The case of a never-smoker woman. Lung Cancer 2016, 96, 52–55. [Google Scholar] [CrossRef] [PubMed]
- Okazaki, I.; Ishikawa, S.; Sohara, Y. Genes associated with succeptibility to lung adenocarcinoma among never smokers suggest the mechanism of disease. Anticancer Res. 2014, 34, 5229–5240. [Google Scholar]
- Yang, P.; Cerhan, J.R.; Vierkant, R.A.; Olson, J.E.; Vachon, C.M.; Limburg, P.J.; Parker, A.S.; Anderson, K.E.; Sellers, T.A. Adenocarcinoma of the lung is strongly associated with cigarette smoking: Further evidence from a prospective study of women. Am. J. Epidemiol. 2002, 156, 1114–1122. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Kent, J.W.; Olivier, M.; Ali, O.; Cerjak, D.; Broeckel, U.; Abdou, R.M.; Dyer, T.D.; Comuzzie, A.; Curran, J.E.; et al. A comprehensive analysis of adiponectin QTLs using SNP association, SNP cis-effects on peripheral blood gene expression and gene expression correlation identified novel metabolic syndrome, MetS) genes with potential role in carcinogenesis and systemic inflammation. BMC Med. Genomics 2013, 6, 14. [Google Scholar]
- Mazieres, J.; Rouquette, I.; Lepage, B.; Milia, J.; Brouchet, L.; Guibert, N.; Beau-Faller, M.; Validire, P.; Hofman, P.; Fouret, P. Specificities of lung adenocarcinoma in women who have never smoked. J. Thorac. Oncol. 2013, 8, 923–929. [Google Scholar] [CrossRef] [PubMed]
- Russo, A.; Autelitano, M.; Bisanti, L. Metabolic syndrome and cancer risk. Eur. J. Cancer 2008, 44, 293–297. [Google Scholar] [CrossRef]
- Flegal, K.M.; Graubard, B.I.; Williamson, D.F.; Gail, M.H. Cause-specific excess deaths associated with underweight, overweight, and obesity. JAMA 2007, 298, 2028–2037. [Google Scholar] [CrossRef]
- Lee, J.Y.; Jeon, I.; Lee, J.M.; Yoon, J.M.; Park, S.M. Diabetes mellitus as an independent risk factor for lung cancer: A meta-analysis of observational studies. Eur. J. Cancer 2013, 49, 2411–2423. [Google Scholar] [CrossRef] [PubMed]
- Cannioto, R.; Etter, J.L.; LaMonte, M.J.; Ray, A.D.; Joseph, J.M.; Al Qassim, E.; Eng, K.H.; Moysich, K.B. Lifetime Physical Inactivity is Associated with Lung Cancer Risk and Mortality. Cancer Treat. Res. Commun. 2018, 14, 37–45. [Google Scholar] [CrossRef] [PubMed]
- Patel, A.V.; Carter, B.D.; Stevens, V.L.; Gaudet, M.M.; Campbell, P.T.; Gapstur, S.M. The relationship between physical activity, obesity, and lung cancer risk by smoking status in a large prospective cohort of US adults. Cancer Causes Control. 2017, 28, 1357–1368. [Google Scholar] [CrossRef] [PubMed]
- Calle, E.E.; Rodriguez, C.; Jacobs, E.J.; Almon, M.L.; Chao, A.; McCullough, M.L.; McCullough, M.L.; Feigelson, H.S.; Thun, M.J. The American Cancer Society Cancer Prevention Study II Nutrition Cohort: Rationale, study design, and baseline characteristics. Cancer 2002, 94, 2490–2501. [Google Scholar] [CrossRef] [PubMed]
- Hassfjell, C.S.; Grimsrud, T.K.; Standring, W.J.F.; Tretli, S. Lung cancer incidence associated with radon exposure in Norwegian homes. Tidsskr. Nor. Laegeforen. 2017, 137, 14–15. [Google Scholar] [CrossRef]
- Kettunen, E.; Hernandez-Vargas, H.; Cros, M.P.; Durand, G.; Le Calvez-Kelm, F.; Stuopelyte, K.; Jarmalaite, S.; Salmenkivi, K.; Anttila, S.; Wolff, H.; et al. Asbestos-associated genome-wide DNA methylation changes in lung cancer. Int. J. Cancer 2017, 141, 2014–2029. [Google Scholar] [CrossRef] [PubMed]
- Santarelli, L.; Gaetani, S.; Monaco, F.; Bracci, M.; Valentino, M.; Amati, M.; Rubini, C.; Sabbatini, A.; Pasquini, E.; Zanotta, N.; et al. Four-miRNA Signature to Identify Asbestos-Related Lung Malignancies. Cancer Epidemiol. Biomarkers Prev. 2019, 28, 119–126. [Google Scholar] [CrossRef]
- Goncalves, R.B.; Coletta, R.D.; Silverio, K.G.; Benevides, L.; Casati, M.Z.; da Silva, J.S.; Nociti, F.H., Jr. Impact of smoking on inflammation: Overview of molecular mechanisms. Inflamm. Res. 2011, 60, 409–424. [Google Scholar] [CrossRef]
- Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef]
- Fridman, W.H.; Pages, F.; Sautes-Fridman, C.; Galon, J. The immune contexture in human tumours: Impact on clinical outcome. Nat. Rev. Cancer 2012, 12, 298–306. [Google Scholar] [CrossRef]
- Kawai, O.; Ishii, G.; Kubota, K.; Murata, Y.; Naito, Y.; Mizuno, T.; Aokage, K.; Saijo, N.; Nishiwaki, Y.; Gemma, A.; et al. Predominant infiltration of macrophages and CD8+) T Cells in cancer nests is a significant predictor of survival in stage IV nonsmall cell lung cancer. Cancer 2008, 113, 1387–1395. [Google Scholar] [CrossRef] [PubMed]
- Trojan, A.; Urosevic, M.; Dummer, R.; Giger, R.; Weder, W.; Stahel, R.A. Immune activation status of CD8+ T cells infiltrating non-small cell lung cancer. Lung Cancer 2004, 44, 143–147. [Google Scholar] [CrossRef] [PubMed]
- Hiraoka, K.; Miyamoto, M.; Cho, Y.; Suzuoki, M.; Oshikiri, T.; Nakakubo, Y.; Itoh, T.; Ohbuchi, T.; Kondo, S.; Katoh, H. Concurrent infiltration by CD8+ T cells and CD4+ T cells is a favourable prognostic factor in non-small-cell lung carcinoma. Br. J. Cancer 2006, 94, 275–280. [Google Scholar] [CrossRef]
- Tobin, N.P.; Foukakis, T.; De Petris, L.; Bergh, J. The importance of molecular markers for diagnosis and selection of targeted treatments in patients with cancer. J. Intern. Med. 2015, 278, 545–570. [Google Scholar] [CrossRef] [PubMed]
- Popper, H.H.; Ryska, A.; Timar, J.; Olszewski, W. Molecular testing in lung cancer in the era of precision medicine. Transl. Lung Cancer Res. 2014, 3, 291–300. [Google Scholar] [PubMed]
- Sampsonas, F.; Ryan, D.; McPhillips, D.; Breen, D.P. Molecular testing and personalized treatment of lung cancer. Curr. Mol. Pharmacol. 2014, 7, 22–32. [Google Scholar] [CrossRef]
- Williams, A.S.; Greer, W.; Bethune, D.; Craddock, K.J.; Flowerdew, G.; Xu, Z. ALK+ lung adenocarcinoma in never smokers and long-term ex-smokers: Prevalence and detection by immunohistochemistry and fluorescence in situ hybridization. Virchows Arch. 2016, 469, 533–540. [Google Scholar] [CrossRef]
- Rodig, S.J.; Mino-Kenudson, M.; Dacic, S.; Yeap, B.Y.; Shaw, A.; Barletta, J.A.; Stubbs, H.; Law, K.; Lindeman, N.; Mark, E.; et al. Unique clinicopathologic features characterize ALK-rearranged lung adenocarcinoma in the western population. Clin. Cancer Res. 2009, 15, 5216–5223. [Google Scholar] [CrossRef]
Tumor Type | Patient Characteristics | Aim Of The Study | Methods | Key Findings | Discussion/Conclusion | Reference |
---|---|---|---|---|---|---|
Adenocarcinoma (AC) | 1111 lung AC and 200 samples of adjacent normal tissue; comparison of smokers vs. non-smokers | Characterization of tumor microenvironment/pattern of tumor-associated immune cells | Online lung cancer data analysis via Gene Expression Omnibus (GEO); to determine the fraction of immune cells in tumors, a linear support vector regression-based method, CIBERSORT, was applied to estimate the relative ratios of 21 leukocyte subtypes | Distinct pathways were altered in lung carcinogenesis in ever-smokers and never-smokers. Never-smoker patients had a better outcome than ever-smoker patients. Mast cells and CD4+ memory T cells were associated with poor outcome when activated compared to the resting form; cigarette smoke induced activation of these immune cells | Tobacco smoke alters the composition of immune cells in lung adenocarcinoma; activation of CD4+ memory T cells and mast cells by smoking may be responsible for the worse outcome in smokers as compared to non-smokers | [26] |
AC | 1 never-smoker female patient with multifocal lung AC; after surgery the patient underwent treatment with EGFR-TKI | Assessment of morphological and genetic tumorheterogeneity | 30-gene next generation sequencing (NGS) panel, allowing for evaluation of intra- and inter-tumoral heterogeneity | The 3 lung tumors were confirmed independent according to NGS; identical EGFR, PIK3CA and TP53 mutations were found in one of the three primary tumors and in the metastasis that occurred later on | In this non-smoker female patient, some type of genetic cancer predispositon is likely, explaining the three genetically independent lung ACs and limited response to treatment | [34] |
Lung cancer (any histological type) | Prospectively studied population of women aged 55-69 years, who were followed up for 13 years | Evaluation of the role of metabolic syndrome in lung cancer | Prospective cohort study; focus on body mass index (BMI) and waist circumference | Patients with lung cancer had a significantly higher waist circumference | Abdominal obesity may increase the risk for lung cancer when stratifying for other common risk factors | [36] |
AC | 140 female AC patients, amongst them 63 never-smokers and 77 former or current smokers | Investigating the link of metabolic disorders and lung AC in never-smokers | Histologic analysis of tumor samples, smoking-associated vs. not smoking-associated tumors | In never-smokers, lipidic histologic differentiation was found significantly more often as compared to smokers | Non-smokers with a sedentary lifestyle, hyperlipidemia and other signs of metabolic disease might be at higher risk for lung cancer as compared to non-smokers without metabolic syndrome | [38] |
Lung cancer (any histological type) | 660 lung cancer patients and 1335 matched controls who did not suffer from any malignant disease | To determine whether physical inactivity increases lung cancer risk, and whether it increases mortality in case of lung cancer | Case-control study; assessment of the association of inactive lifestyle and risk of lung cancer via multivariate logistic regression analysis; Cox proportional hazard models were used for estimation of the connex of inactivity and mortality from lung cancer | Significant positive correlation of physical inactivity and risk of lung cancer; significant positive correlation between lifetime physical inactivity and lung cancer-related mortality; also significant for non-smoking lung cancer patients | Physical inactivity increases not only the risk of lung cancer but also lung cancer-related mortality | [42] |
Lung cancer (any histological type) | Data of 162679 men and women from the American Cancer Society Cancer Prevention Study-II Nutrition Cohort were analyzed, who were all free of cancer at enrollment in this study (1992-1993) | Assessment of the correlation of baseline physical activity and lung cancer incidence over the follow-up period | Baseline physical activity (MET-hours per week; none, 0.1 to <8.75; 8.75-17.4; >17.5 MET-hours/week), BMI and waist circumference were assessed in relation to lung cancer risk | Physical activity was not associated with lung cancer risk, except in former smokers who quit less than 10 years ago (for >17.5 MET hours/week); BMI was inversely associated with lung cancer risk | According to this study, physical activity is not a protective factor regarding the incidence of lung cancer | [43] |
AC | Never-smokers and long-term ex-smokers who quit >10 years prior to diagnosis with AC; 251 cases of resected lung AC (79 never-, and 172 ex-smokers) | To assess the prevalence of ALK-rearrangements in lung AC samples of lifetime non-smokers, as well as long-term ex-smokers (quit >10 years prior to diagnosis) | ALK-rearrangement was evaluated via fluorescence in situ hybridization FISH, and immunohistochemistry (IHC) | strong evidence of increased ALK gene rearrangement in the non-smoking population; no significant difference in ALK-rearrangement between the ex-smokers and the general population | ALK-rearrangement is more common in non-smoking patients | [57] |
Squamous cell carcinoma (SCC) | 19 patients suffering from SCC (8 non-smokers, 11 smokers) | Evaluation of genetic differences in SCC of smokers, as compared to non-smokers | Array comparative genomic hybridization (ArrayCGH); immunohistochemistry | 16 gene regions were significantly altered, according to ArrayCGH in non-smokers compared to smokers; the proto-oncogene GAB2 (11q14.1) was significantly amplified in non-smoking patients, and the GAB2 protein was upregulated as well | GAB2 amplification is likely to contribute to SCC development in non-smokers | [4] |
SCC | 1 non-smoker female patient with SCC and history of a SCC at the neck 2 years prior | To determine germline cancer predisposition in this special case | Assessment of somatic driver mutations via whole exome sequencing | Predominance of C>T transitions in tumoral lung tissue; usually not found in tobacco-smoke-associated lung cancer; no mutations of frequent driver genes of lung cancer were found | In this case a special oligogenic germline signature predisposed for SCC formation; personalized medicine is especially important in non-smoker patients | [30] |
SCC | 371 patients with SCC; among them were 31 never-smokers | Analyzing the response to nivolumab in SCC patients who are non-smokers | Nivolumab was administered at a dosage of 3 mg/kg every 2 weeks for a maximum of 24 months, and safety was monitored | The objective response rate, disease-control rate and median overall survival were comparable in the smoking vs. the never-smoking group | Safety and effecacy of nivolumab seems to be similar in never-smokers as compared to smokers with SCC | [33] |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Smolle, E.; Pichler, M. Non-Smoking-Associated Lung Cancer: A distinct Entity in Terms of Tumor Biology, Patient Characteristics and Impact of Hereditary Cancer Predisposition. Cancers 2019, 11, 204. https://doi.org/10.3390/cancers11020204
Smolle E, Pichler M. Non-Smoking-Associated Lung Cancer: A distinct Entity in Terms of Tumor Biology, Patient Characteristics and Impact of Hereditary Cancer Predisposition. Cancers. 2019; 11(2):204. https://doi.org/10.3390/cancers11020204
Chicago/Turabian StyleSmolle, Elisabeth, and Martin Pichler. 2019. "Non-Smoking-Associated Lung Cancer: A distinct Entity in Terms of Tumor Biology, Patient Characteristics and Impact of Hereditary Cancer Predisposition" Cancers 11, no. 2: 204. https://doi.org/10.3390/cancers11020204
APA StyleSmolle, E., & Pichler, M. (2019). Non-Smoking-Associated Lung Cancer: A distinct Entity in Terms of Tumor Biology, Patient Characteristics and Impact of Hereditary Cancer Predisposition. Cancers, 11(2), 204. https://doi.org/10.3390/cancers11020204