1
|
Lewandowski M, Riedel TP, Krug JD, Kleindienst TE, Meier MJ, Long AS, Warren SH, DeMarini DM. Molecular Compositions, Mutagenicity, and Mutation Spectra of Atmospheric Oxidation Products of Alkenes and Dienes Initiated by NOx + UV or Ozone: A Structure-Activity Analysis. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:18846-18855. [PMID: 39374177 PMCID: PMC11588028 DOI: 10.1021/acs.est.4c04603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Photooxidation products resulting from volatile organic compounds (VOCs) reacting with sunlight are important contributors to gas-phase air pollution. We characterized the product-weighted mutagenic potencies (rev m3 mgC-1 h-1) in Salmonella TA100 of atmospheres resulting from the hydroxyl radical (OH)-initiated photochemical oxidation of 11 C4 or C5 alkenes or dienes in the presence of nitric oxide (NO) and from the ozonolysis of four VOCs without NO (isoprene; 1,3-pentadiene; 1,4-pentadiene; and 1,3-butadiene). Irradiated atmospheres from precursors with a single C═C bond (3-methyl-1-butene, 2-methyl-1-butene, cis/trans-2-pentene, 2-methyl-2-butene, 1-butene, and 1-pentene) had low potencies (<5), whereas linear dienes with terminal C═C bonds had high potencies (50-65). Dienes with a branched structure (isoprene) or internal C═C bonds (1,3-pentadiene) had intermediate potencies (15-20). No VOCs were mutagenic without photochemical oxidation. VOCs+O3 in the dark produced less mutagenic atmospheres than photochemistry in the presence of NO. Atmospheres induced primarily C to T and C to A mutations, the main base substitutions in nonsmoker lung cancer. Atmospheres from the photooxidation of isoprene and 1,3-pentadiene also induced GG to TT, the signature mutation of peroxyacetyl nitrate. Five molecular compositions identified by Chemical Ionization Mass Spectrometry (CIMS), most containing nitrogen, correlated (r = 0.76-0.85) with the mutagenic potencies of irradiated atmospheres; most had a likely nitrate functional group. Assessment of the mutagenicity of emitted VOCs should consider VOC photooxidation products, especially dienes with terminal C═C bonds, as these products likely contribute to overall health effects from ambient air pollution.
Collapse
Affiliation(s)
- Michael Lewandowski
- Center for Environmental Measurement and Modeling, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Theran P. Riedel
- Center for Environmental Measurement and Modeling, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Jonathan D. Krug
- Center for Environmental Measurement and Modeling, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Tadeusz E. Kleindienst
- Center for Environmental Measurement and Modeling, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Matthew J. Meier
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Alexandra S. Long
- Existing Substances Risk Assessment Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, ON K1A 0K9, Canada
| | - Sarah H. Warren
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - David M. DeMarini
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| |
Collapse
|
2
|
Pan S, Li Y, Zhang J. 6-Shogaol prevents benzo (A) pyrene-exposed lung carcinogenesis via modulating PRDX1-associated oxidative stress, inflammation, and proliferation in mouse models. ENVIRONMENTAL TOXICOLOGY 2024; 39:75-84. [PMID: 37638803 DOI: 10.1002/tox.23946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/13/2023] [Indexed: 08/29/2023]
Abstract
In this study, we have investigated the chemopreventive role of 6-shogaol (6-SGL) on benzopyrene (BaP) exposed lung carcinogenesis by modulating PRDX1-associated oxidative stress, inflammation, and proliferation in Swiss albino mouse models. Mice were exposed to BaP (50 mg/kg b.wt) orally twice a week for four consecutive weeks and maintained for 16 weeks, respectively. 6-SGL (30 mg/kg b.wt) were orally administered to mouse 1 h before BaP exposure for 16 weeks. After the experiment's termination, 6-SGL (30 mg/kg b.wt) prevented the loss in body weight, increased lung weight, and the total number of tumors in the mice. Moreover, we observed that 6-SGL treatment reverted the activity of BaP-induced lipid peroxidation and antioxidants in mice. Also, 6-SGL impeded the phosphorylation of MAPK family proteins such as Erk1, p38, and Jnk1 in BaP-exposed mice. PRDX1 is an essential antioxidant protein that scavenges toxic radicals and enhances several antioxidant proteins. Overexpression of PRDX1 substantially inhibits MAPKs, proliferation, and inflammation signaling axis. Hence, PRDX1 is thought to be a novel targeting protein for preventing BaP-induced lung cancer. In this study, we have obtained the 6-SGL treatment in a mouse model that reverted BaP-induced depletion of PRDX1 expression. Moreover, pretreatment of 6-SGL (30 mg/kg b.wt) significantly inhibited enhanced proinflammatory cytokines (TNF-α, IL-6, IL-β1, IL-10) and proliferative markers (Cyclin-D1, Cyclin-D2, and PCNA) in BaP-exposed mice. The histopathological studies also confirmed that 6-SGL effectively protected the cells with less damage. Thus, the study demonstrated that 6-SGL could be a potential phytochemical and act as a chemopreventive agent in BaP-induced lung cancer by enhancing PRDX1 expression.
Collapse
Affiliation(s)
- Shuang Pan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Yaming Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jinzhao Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| |
Collapse
|
3
|
Karlow JA, Pehrsson EC, Xing X, Watson M, Devarakonda S, Govindan R, Wang T. Non-small Cell Lung Cancer Epigenomes Exhibit Altered DNA Methylation in Smokers and Never-smokers. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:991-1013. [PMID: 37742993 PMCID: PMC10928376 DOI: 10.1016/j.gpb.2023.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 02/11/2023] [Accepted: 03/14/2023] [Indexed: 09/26/2023]
Abstract
Epigenetic alterations are widespread in cancer and can complement genetic alterations to influence cancer progression and treatment outcome. To determine the potential contribution of DNAmethylation alterations to tumor phenotype in non-small cell lung cancer (NSCLC) in both smoker and never-smoker patients, we performed genome-wide profiling of DNA methylation in 17 primary NSCLC tumors and 10 matched normal lung samples using the complementary assays, methylated DNA immunoprecipitation sequencing (MeDIP-seq) and methylation sensitive restriction enzyme sequencing (MRE-seq). We reported recurrent methylation changes in the promoters of several genes, many previously implicated in cancer, including FAM83A and SEPT9 (hypomethylation), as well as PCDH7, NKX2-1, and SOX17 (hypermethylation). Although many methylation changes between tumors and their paired normal samples were shared across patients, several were specific to a particular smoking status. For example, never-smokers displayed a greater proportion of hypomethylated differentially methylated regions (hypoDMRs) and a greater number of recurrently hypomethylated promoters, including those of ASPSCR1, TOP2A, DPP9, and USP39, all previously linked to cancer. Changes outside of promoters were also widespread and often recurrent, particularly methylation loss over repetitive elements, highly enriched for ERV1 subfamilies. Recurrent hypoDMRs were enriched for several transcription factor binding motifs, often for genes involved in signaling and cell proliferation. For example, 71% of recurrent promoter hypoDMRs contained a motif for NKX2-1. Finally, the majority of DMRs were located within an active chromatin state in tissues profiled using the Roadmap Epigenomics data, suggesting that methylation changes may contribute to altered regulatory programs through the adaptation of cell type-specific expression programs.
Collapse
Affiliation(s)
- Jennifer A Karlow
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erica C Pehrsson
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark Watson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Siddhartha Devarakonda
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ramaswamy Govindan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA.
| |
Collapse
|
4
|
Wang P, Sun S, Lam S, Lockwood WW. New insights into the biology and development of lung cancer in never smokers-implications for early detection and treatment. J Transl Med 2023; 21:585. [PMID: 37653450 PMCID: PMC10472682 DOI: 10.1186/s12967-023-04430-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/10/2023] [Indexed: 09/02/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide. Despite never smokers comprising between 10 and 25% of all cases, lung cancer in never smokers (LCNS) is relatively under characterized from an etiological and biological perspective. The application of multi-omics techniques on large patient cohorts has significantly advanced the current understanding of LCNS tumor biology. By synthesizing the findings of multi-omics studies on LCNS from a clinical perspective, we can directly translate knowledge regarding tumor biology into implications for patient care. Primarily focused on never smokers with lung adenocarcinoma, this review details the predominance of driver mutations, particularly in East Asian patients, as well as the frequency and importance of germline variants in LCNS. The mutational patterns present in LCNS tumors are thoroughly explored, highlighting the high abundance of the APOBEC signature. Moreover, this review recognizes the spectrum of immune profiles present in LCNS tumors and posits how it can be translated to treatment selection. The recurring and novel insights from multi-omics studies on LCNS tumor biology have a wide range of clinical implications. Risk factors such as exposure to outdoor air pollution, second hand smoke, and potentially diet have a genomic imprint in LCNS at varying degrees, and although they do not encompass all LCNS cases, they can be leveraged to stratify risk. Germline variants similarly contribute to a notable proportion of LCNS, which warrants detailed documentation of family history of lung cancer among never smokers and demonstrates value in developing testing for pathogenic variants in never smokers for early detection in the future. Molecular driver subtypes and specific co-mutations and mutational signatures have prognostic value in LCNS and can guide treatment selection. LCNS tumors with no known driver alterations tend to be stem-like and genes contributing to this state may serve as potential therapeutic targets. Overall, the comprehensive findings of multi-omics studies exert a wide influence on clinical management and future research directions in the realm of LCNS.
Collapse
Affiliation(s)
- Peiyao Wang
- Department of Integrative Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Sophie Sun
- Department of Medical Oncology, British Columbia Cancer Agency Vancouver, Vancouver, BC, Canada
| | - Stephen Lam
- Department of Integrative Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada
| | - William W Lockwood
- Department of Integrative Oncology, British Columbia Cancer Research Institute, Vancouver, BC, Canada.
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Andrzejczak A, Partyka A, Wiśniewski A, Porębska I, Pawełczyk K, Ptaszkowski K, Kuśnierczyk P, Jasek M, Karabon L. The association of BTLA gene polymorphisms with non-small lung cancer risk in smokers and never-smokers. Front Immunol 2023; 13:1006639. [PMID: 36741370 PMCID: PMC9893504 DOI: 10.3389/fimmu.2022.1006639] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Introduction Lung cancer is the predominant cause of death among cancer patients and non-small cell lung cancer (NSCLC) is the most common type. Cigarette smoking is the prevailing risk factor for NSCLC, nevertheless, this cancer is also diagnosed in never-smokers. B and T lymphocyte attenuator (BTLA) belongs to immunological checkpoints which are key regulatory molecules of the immune response. A growing body of evidence highlights the important role of BTLA in cancer. In our previous studies, we showed a significant association between BTLA gene variants and susceptibility to chronic lymphoblastic leukemia and renal cell carcinoma in the Polish population. The present study aimed to analyze the impact of BTLA polymorphic variants on the susceptibility to NSCLC and NSCLC patients' overall survival (OS). Methods Using TaqMan probes we genotyped seven BTLA single-nucleotide polymorphisms (SNPs): rs2705511, rs1982809, rs9288952, rs9288953, rs1844089, rs11921669 and rs2633582 with the use of ViiA 7 Real-Time PCR System. Results We found that rs1982809 within BTLA is associated with NSCLC risk, where carriers of rs1982809G allele (AG+GG genotypes) were more frequent in patients compared to controls. In subgroup analyses, we also noticed that rs1982809G carriers are significantly overrepresented in never-smokers, but not in smokers compared to controls. Additionally, the global distribution of the haplotypes differed between the never-smokers and smokers, where haplotypes A G G C A, C G A C G, and C G A T G were more frequent in never-smoking patients. Furthermore, the presence rs1982809G (AG+GG genotypes) allele as well as the presence of rs9288953T allele (CT+TT genotypes) increased NSCLC risk in females' patients. After stratification by histological type, we noticed that rs1982809G and rs2705511C carriers were more frequent among adenocarcinoma patients. Moreover, rs1982809G and rs2705511C correlated with the more advanced stages of NSCLC (stage II and III), but not with stage IV. Furthermore, we showed that rs2705511 and rs1982809 significantly modified OS, while rs9288952 tend to be associated with patients' survival. Conclusion Our results indicate that BTLA polymorphic variants may be considered low penetrating risk factors for NSCLC especially in never-smokers, and in females, and are associated with OS of NSCLC patients.
Collapse
Affiliation(s)
- Anna Andrzejczak
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland,*Correspondence: Anna Andrzejczak, ; Lidia Karabon,
| | - Anna Partyka
- Laboratory of Immunopathology, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Andrzej Wiśniewski
- Laboratory of Immunogenetics and Tissue Immunology, Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Irena Porębska
- Department of Pulmonology and Lung Oncology, Wrocław Medical University, Wrocław, Poland
| | - Konrad Pawełczyk
- Departament of Thoracic Surgery, Lower Silesian Centre of Oncology, Pulmonology and Haematology, Wrocław, Poland
| | - Kuba Ptaszkowski
- Department of Clinical Biomechanics and Physiotherapy in Motor System Disorders, Wrocław Medical University, Wrocław, Poland
| | - Piotr Kuśnierczyk
- Laboratory of Immunogenetics and Tissue Immunology, Department of Clinical Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Monika Jasek
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Lidia Karabon
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland,*Correspondence: Anna Andrzejczak, ; Lidia Karabon,
| |
Collapse
|
6
|
Kuśnierczyk P. Genetic differences between smokers and never-smokers with lung cancer. Front Immunol 2023; 14:1063716. [PMID: 36817482 PMCID: PMC9932279 DOI: 10.3389/fimmu.2023.1063716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023] Open
Abstract
Smoking is a major risk factor for lung cancer, therefore lung cancer epidemiological trends reflect the past trends of cigarette smoking to a great extent. The geographic patterns in mortality closely follow those in incidence. Although lung cancer is strongly associated with cigarette smoking, only about 15% of smokers get lung cancer, and also some never-smokers develop this malignancy. Although less frequent, lung cancer in never smokers is the seventh leading cause of cancer deaths in both sexes worldwide. Lung cancer in smokers and never-smokers differs in many aspects: in histological types, environmental factors representing a risk, and in genes associated with this disease. In this review, we will focus on the genetic differences between lung cancer in smokers versus never-smokers: gene expression, germ-line polymorphisms, gene mutations, as well as ethnic and gender differences. Finally, treatment options for smokers and never-smokers will be briefly reviewed.
Collapse
Affiliation(s)
- Piotr Kuśnierczyk
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
7
|
Gray ER, Mordaka JM, Christoforou ER, von Bargen K, Potts ND, Xyrafaki C, Silva AL, Stolarek-Januszkiewicz M, Anton K, Powalowska PK, Andreazza S, Tomassini A, Palmer RN, Cooke A, Osborne RJ, Balmforth BW. Ultra-sensitive molecular detection of gene fusions from RNA using ASPYRE. BMC Med Genomics 2022; 15:215. [PMID: 36224552 PMCID: PMC9555097 DOI: 10.1186/s12920-022-01363-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/23/2022] [Indexed: 12/04/2022] Open
Abstract
Background RNA is a critical analyte for unambiguous detection of actionable mutations used to guide treatment decisions in oncology. Currently available methods for gene fusion detection include molecular or antibody-based assays, which suffer from either being limited to single-gene targeting, lack of sensitivity, or long turnaround time. The sensitivity and predictive value of next generation sequencing DNA-based assays to detect fusions by sequencing intronic regions is variable, due to the extensive size of introns. The required depth of sequencing and input nucleic acid required can be prohibitive; in addition it is not certain that predicted gene fusions are actually expressed. Results Herein we describe a method based on pyrophosphorolysis to include detection of gene fusions from RNA, with identical assay steps and conditions to detect somatic mutations in DNA [1], permitting concurrent assessment of DNA and RNA in a single instrument run. Conclusion The limit of detection was under 6 molecules/ 6 µL target volume. The workflow and instrumentation required are akin to PCR assays, and the entire assay from extracted nucleic acid to sample analysis can be completed within a single day. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01363-0.
Collapse
Affiliation(s)
- Eleanor R Gray
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Justyna M Mordaka
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | | | - Kristine von Bargen
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Nicola D Potts
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Christina Xyrafaki
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Ana-Luisa Silva
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | | | - Katarzyna Anton
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Paulina K Powalowska
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Simonetta Andreazza
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Alessandro Tomassini
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Rebecca N Palmer
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Aishling Cooke
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Robert J Osborne
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England
| | - Barnaby W Balmforth
- Biofidelity Ltd, 330 Cambridge Science Park, Milton road, CB4 0WN, Cambridge, England.
| |
Collapse
|
8
|
Oróstica KY, Saez-Hidalgo J, de Santiago PR, Rivas S, Contreras S, Navarro G, Asenjo JA, Olivera-Nappa Á, Armisén R. Total mutational load and clinical features as predictors of the metastatic status in lung adenocarcinoma and squamous cell carcinoma patients. Lab Invest 2022; 20:373. [PMID: 35982500 PMCID: PMC9389677 DOI: 10.1186/s12967-022-03572-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 08/04/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Recently, extensive cancer genomic studies have revealed mutational and clinical data of large cohorts of cancer patients. For example, the Pan-Lung Cancer 2016 dataset (part of The Cancer Genome Atlas project), summarises the mutational and clinical profiles of different subtypes of Lung Cancer (LC). Mutational and clinical signatures have been used independently for tumour typification and prediction of metastasis in LC patients. Is it then possible to achieve better typifications and predictions when combining both data streams? METHODS In a cohort of 1144 Lung Adenocarcinoma (LUAD) and Lung Squamous Cell Carcinoma (LSCC) patients, we studied the number of missense mutations (hereafter, the Total Mutational Load TML) and distribution of clinical variables, for different classes of patients. Using the TML and different sets of clinical variables (tumour stage, age, sex, smoking status, and packs of cigarettes smoked per year), we built Random Forest classification models that calculate the likelihood of developing metastasis. RESULTS We found that LC patients different in age, smoking status, and tumour type had significantly different mean TMLs. Although TML was an informative feature, its effect was secondary to the "tumour stage" feature. However, its contribution to the classification is not redundant with the latter; models trained using both TML and tumour stage performed better than models trained using only one of these variables. We found that models trained in the entire dataset (i.e., without using dimensionality reduction techniques) and without resampling achieved the highest performance, with an F1 score of 0.64 (95%CrI [0.62, 0.66]). CONCLUSIONS Clinical variables and TML should be considered together when assessing the likelihood of LC patients progressing to metastatic states, as the information these encode is not redundant. Altogether, we provide new evidence of the need for comprehensive diagnostic tools for metastasis.
Collapse
Affiliation(s)
- Karen Y Oróstica
- Instituto de Investigación Interdisciplinaria, Vicerrectoría Académica, Universidad de Talca, 3460000, Talca, Chile
| | - Juan Saez-Hidalgo
- Centre for Biotechnology and Bioengineering (CeBiB), Department of Chemical Engineering, Biotechnology and Materials, University of Chile, 8370456, Santiago, Chile.,Department of Computer Science, University of Chile, 8370459, Santiago, Chile
| | - Pamela R de Santiago
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Solange Rivas
- Department of Basic Clinical Oncology, Faculty of Medicine, University of Chile, Santiago, Chile.,Centro de Genética Y Genómica, Instituto de Ciencias E Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, 7590943, Santiago, Chile
| | - Sebastian Contreras
- Centre for Biotechnology and Bioengineering (CeBiB), Department of Chemical Engineering, Biotechnology and Materials, University of Chile, 8370456, Santiago, Chile.,Max Planck Institute for Dynamics and Self-Organization, Göttingen, Germany
| | - Gonzalo Navarro
- Centre for Biotechnology and Bioengineering (CeBiB), Department of Chemical Engineering, Biotechnology and Materials, University of Chile, 8370456, Santiago, Chile.,Department of Computer Science, University of Chile, 8370459, Santiago, Chile
| | - Juan A Asenjo
- Centre for Biotechnology and Bioengineering (CeBiB), Department of Chemical Engineering, Biotechnology and Materials, University of Chile, 8370456, Santiago, Chile
| | - Álvaro Olivera-Nappa
- Centre for Biotechnology and Bioengineering (CeBiB), Department of Chemical Engineering, Biotechnology and Materials, University of Chile, 8370456, Santiago, Chile.
| | - Ricardo Armisén
- Centro de Genética Y Genómica, Instituto de Ciencias E Innovación en Medicina, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, 7590943, Santiago, Chile.
| |
Collapse
|
9
|
Cheng ES, Weber M, Steinberg J, Yu XQ. Lung cancer risk in never-smokers: An overview of environmental and genetic factors. Chin J Cancer Res 2021; 33:548-562. [PMID: 34815629 PMCID: PMC8580800 DOI: 10.21147/j.issn.1000-9604.2021.05.02] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/17/2021] [Indexed: 01/22/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality globally, accounting for 1.8 million deaths in 2020. While the vast majority are caused by tobacco smoking, 15%-25% of all lung cancer cases occur in lifelong never-smokers. The International Agency for Research on Cancer (IARC) has classified multiple agents with sufficient evidence for lung carcinogenesis in humans, which include tobacco smoking, as well as several environmental exposures such as radon, second-hand tobacco smoke, outdoor air pollution, household combustion of coal and several occupational hazards. However, the IARC evaluation had not been stratified based on smoking status, and notably lung cancer in never-smokers (LCINS) has different epidemiological, clinicopathologic and molecular characteristics from lung cancer in ever-smokers. Among several risk factors proposed for the development of LCINS, environmental factors have the most available evidence for their association with LCINS and their roles cannot be overemphasized. Additionally, while initial genetic studies largely focused on lung cancer as a whole, recent studies have also identified genetic risk factors for LCINS. This article presents an overview of several environmental factors associated with LCINS, and some of the emerging evidence for genetic factors associated with LCINS. An increased understanding of the risk factors associated with LCINS not only helps to evaluate a never-smoker's personal risk for lung cancer, but also has important public health implications for the prevention and early detection of the disease. Conclusive evidence on causal associations could inform longer-term policy reform in a range of areas including occupational health and safety, urban design, energy use and particle emissions, and the importance of considering the impacts of second-hand smoke in tobacco control policy.
Collapse
Affiliation(s)
- Elvin S Cheng
- The Daffodil Centre, the University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW 2011, Australia
| | - Marianne Weber
- The Daffodil Centre, the University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW 2011, Australia
| | - Julia Steinberg
- The Daffodil Centre, the University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW 2011, Australia
| | - Xue Qin Yu
- The Daffodil Centre, the University of Sydney, a joint venture with Cancer Council NSW, Sydney, NSW 2011, Australia
| |
Collapse
|
10
|
Wiśniewski A, Sobczyński M, Pawełczyk K, Porębska I, Jasek M, Wagner M, Niepiekło-Miniewska W, Kowal A, Dubis J, Jędruchniewicz N, Kuśnierczyk P. Polymorphisms of Antigen-Presenting Machinery Genes in Non-Small Cell Lung Cancer: Different Impact on Disease Risk and Clinical Parameters in Smokers and Never-Smokers. Front Immunol 2021; 12:664474. [PMID: 34149699 PMCID: PMC8212834 DOI: 10.3389/fimmu.2021.664474] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 05/05/2021] [Indexed: 12/25/2022] Open
Abstract
Lung cancer is strongly associated with cigarette smoking; nevertheless some never-smokers develop cancer. Immune eradication of cancer cells is dependent on polymorphisms of HLA class I molecules and antigen-processing machinery (APM) components. We have already published highly significant associations of single nucleotide polymorphisms (SNPs) of the ERAP1 gene with non-small cell lung cancer (NSCLC) in Chinese, but not in Polish populations. However, the smoking status of participants was not known in the previous study. Here, we compared the distribution of APM polymorphic variants in larger cohorts of Polish patients with NSCLC and controls, stratified according to their smoking status. We found significant but opposite associations in never-smokers and in smokers of all tested SNPs (rs26653, rs2287987, rs30187, and rs27044) but one (rs26618) in ERAP1. No significant associations were seen in other genes. Haplotype analysis indicated that the distribution of many ERAP1/2 haplotypes is opposite, depending on smoking status. Additionally, haplotypic combination of low activity ERAP1 and the lack of an active form of ERAP2 seems to favor the disease in never-smokers. We also revealed interesting associations of some APM polymorphisms with: age at diagnosis (ERAP1 rs26653), disease stage (ERAP1 rs27044, PSMB9 rs17587), overall survival (ERAP1 rs30187), and response to chemotherapy (ERAP1 rs27044). The results presented here may suggest the important role for ERAP1 in the anti-cancer response, which is different in smokers versus never-smokers, depending to some extent on the presence of ERAP2, and affecting NSCLC clinical course.
Collapse
Affiliation(s)
- Andrzej Wiśniewski
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Maciej Sobczyński
- Department of Bioinformatics and Genomics, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Konrad Pawełczyk
- Department and Clinic of Thoracic Surgery, Wrocław Medical University, Wrocław, Poland
| | - Irena Porębska
- Department of Pulmonology and Lung Oncology, Wrocław Medical University, Wrocław, Poland
| | - Monika Jasek
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Marta Wagner
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Wanda Niepiekło-Miniewska
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Aneta Kowal
- Department of Pulmonology and Lung Oncology, Wrocław Medical University, Wrocław, Poland
| | - Joanna Dubis
- Research and Development Centre, Regional Specialist Hospital in Wrocław, Wrocław, Poland
| | - Natalia Jędruchniewicz
- Research and Development Centre, Regional Specialist Hospital in Wrocław, Wrocław, Poland
| | - Piotr Kuśnierczyk
- Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
11
|
Associations of Variability in Metabolic Parameters with Lung Cancer: A Nationwide Population-Based Study. Cancers (Basel) 2021; 13:cancers13081982. [PMID: 33924149 PMCID: PMC8074362 DOI: 10.3390/cancers13081982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/27/2021] [Accepted: 04/14/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Lung cancer is currently the most common cancer worldwide. This study investigates whether visit-to-visit variability in metabolic parameters is associated with lung cancer risk. We found that a high variability in fasting blood glucose, systolic blood pressure, total cholesterol, and body weight were each associated with increased risk of lung cancer. A higher number of high-variability parameters were also associated with increased lung cancer risk. Further research is needed to examine whether reducing variability can lead to decreased lung cancer risk. Abstract We investigated whether visit-to-visit variability in metabolic parameters is associated with lung cancer risk. We used nationally representative data from the Korean National Health Insurance System, and 8,011,209 lung-cancer-free subjects who underwent over three health examinations from 2005 to 2010 were followed until 2017. Variability of fasting blood glucose, total cholesterol, systolic blood pressure, and body weight were measured by the variability independent of the mean, assessed by quartiles. There were 44,982 lung cancer events. The hazard ratio (HR) and 95% confidence interval (CI) for lung cancer risk was 1.07 (1.04, 1.10) for fasting blood glucose in the highest quartile, 1.08 (1.05, 1.10) for systolic blood pressure, 1.04 (1.01, 1.07) for weight, and 1.11 (1.08, 1.14) for total cholesterol. When comparing ≥3 vs. 0 high-variability metabolic parameters, the HR for lung cancer was 1.18 (95% CI, 1.14, 1.22). However, while ≥3 high-variability parameters showed an increased lung cancer risk in men (HR 1.26, 95% CI 1.21, 1.31), women did not show increased risk (HR 0.99, 95% CI 0.92, 1.06). High variability in each metabolic parameter, and a higher number of high-variability parameters, were associated with increased lung cancer risk.
Collapse
|
12
|
Non-small cell lung cancer in never- and ever-smokers: Is it the same disease? J Thorac Cardiovasc Surg 2020; 161:1903-1917.e9. [PMID: 32893009 DOI: 10.1016/j.jtcvs.2020.03.175] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 02/28/2020] [Accepted: 03/12/2020] [Indexed: 11/20/2022]
Abstract
OBJECTIVES To investigate differences in presentation, pathology, and outcomes after resection of non-small cell lung cancer (NSCLC) in never-smokers versus ever-smokers. METHODS From January 2006 to July 2016, 172 never-smokers and 1376 ever-smokers with NSCLC underwent pulmonary resection. The 2 cohorts were matched on patient characteristics, histopathological cancer cell type, and pathological stage group using a weighted balancing score, and overall survival and cancer recurrence were compared by pathological stage. Random forests for survival was used to identify granular cancer characteristics with different survival and cancer recurrence importance between groups. RESULTS In never-smokers, the prevalence of NSCLC was more frequent in women than in men (63% [n = 109] vs 45% [n = 63]). Compared with ever-smokers, never-smokers had less upper-lobe disease (53% [n = 91] vs 62% [n = 855]) and more adenocarcinoma (88% [n = 151] vs 62% [n = 845]). Postoperative complications were similar. Never-smokers had a lower prevalence of non-lung cancer deaths than ever-smokers (13% vs 23% at 5 years; P = .006). Among matched pairs, never-smokers had better overall survival at 5 years in pathological stage I (96% vs 78%), but worse survival in stage II (54% vs 78%). Tumor size, N category, and histopathological cell type were more important drivers of mortality and cancer recurrence in never-smokers than in ever-smokers. CONCLUSIONS NSCLC in never-smokers affects women more than men and presents with different anatomic and histopathological distributions. Matched never-smokers have better or equivalent outcomes than ever-smokers in pathological stage I cancer, but are less likely to survive and to be cured of cancer as tumor burden increases. These findings suggest that there might be unique tumor or host behaviors differentially impacting survival of never- and ever-smoking patients with NSCLC.
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW With the development of targeted therapies, the treatment strategy of patients with advanced or metastatic non-small cell lung cancer (NSCLC) has changed tremendously. In this review, we focus on the different aspects of the treatment of oncogene-driven NSCLC. RECENT FINDINGS Patients with an EGFR or ALK alteration show a better clinical outcome with tyrosine kinase inhibitor (TKI) treatment compared to chemotherapy.Patients with a ROS1 rearrangement or a BRAF V600E mutation show favorable clinical outcome with TKI treatment compared to chemotherapy, although randomized trials are not available.Patients on TKIs will eventually develop disease progression because of acquired resistance.The treatment with immunotherapy in EGFR and ALK-positive NSCLC patients did not improve overall survival over that of chemotherapy.Blood-based genetic analysis provides the opportunity to noninvasively screen patients for the presence of oncogenic drivers and to monitor resistance during TKI treatment. SUMMARY Targeted molecular therapies are now standard of care for patients with oncogene-driven NSCLC with a good clinical benefit and minimal toxicity. The role of immunotherapy in patients with molecular alterations is still unclear. Blood-based genotyping has gained interest in the diagnostic and resistance monitoring setting for patients with NSCLC.
Collapse
|
14
|
Evaluation of models for predicting the probability of malignancy in patients with pulmonary nodules. Biosci Rep 2020; 40:222159. [PMID: 32068231 PMCID: PMC7048676 DOI: 10.1042/bsr20193875] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/12/2020] [Accepted: 02/12/2020] [Indexed: 01/06/2023] Open
Abstract
Objectives: The post-imaging, mathematical predictive model was established by combining demographic and imaging characteristics with a pulmonary nodule risk score. The prediction model provides directions for the treatment of pulmonary nodules. Many studies have established predictive models for pulmonary nodules in different populations. However, the predictive factors contained in each model were significantly different. We hypothesized that applying different models to local research groups will make a difference in predicting the benign and malignant lung nodules, distinguishing between early and late lung cancers, and between adenocarcinoma and squamous cell carcinoma. In the present study, we compared four widely used and well-known mathematical prediction models. Materials and methods: We performed a retrospective study of 496 patients from January 2017 to October 2019, they were diagnosed with nodules by pathological. We evaluate models’ performance by viewing 425 malignant and 71 benign patients’ computed tomography results. At the same time, we use the calibration curve and the area under the receiver operating characteristic curve whose abbreviation is AUC to assess one model’s predictive performance. Results: We find that in distinguishing the Benign and the Malignancy, Peking University People’s Hospital model possessed excellent performance (AUC = 0.63), as well as differentiating between early and late lung cancers (AUC = 0.67) and identifying lung adenocarcinoma (AUC = 0.61). While in the identification of lung squamous cell carcinoma, the Veterans Affairs model performed the best (AUC = 0.69). Conclusions: Geographic disparities are an extremely important influence factors, and which clinical features contained in the mathematical prediction model are the key to affect the precision and accuracy.
Collapse
|
15
|
Löfling L, Karimi A, Sandin F, Bahmanyar S, Kieler H, Lambe M, Lamberg K, Wagenius G. Clinical characteristics and survival in non-small cell lung cancer patients by smoking history: a population-based cohort study. Acta Oncol 2019; 58:1618-1627. [PMID: 31373239 DOI: 10.1080/0284186x.2019.1638521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Introduction: Approximately, 10-15% of lung cancer patients have never smoked. Previous epidemiological studies on non-tobacco associated lung cancer have been hampered by selected data from a small number of hospitals or limited numbers of patients. By use of data from large population-based registers with national coverage, this study aims to compare characteristics and survival of patients with non-small cell lung cancer (NSCLC) with different smoking histories.Methods: Swedish national population-based registers were used to retrieve data on patients diagnosed with primary NSCLC between 2002 and 2016. The Kaplan-Meier method and Cox proportional hazard models were used to estimate overall survival and lung cancer-specific survival by smoking history.Results: In total, 41,262 patients with NSCLC were included. Of those, 4624 (11%) had never smoked. Never-smokers were more often women and older compared to ever smokers (current and former). Adenocarcinoma was proportionally more common in never-smokers (77%) compared to current (52%) and former smokers (57%). Stage IV disease was more common in never-smokers (57%) than in current (48%) and former smokers (48%). Epidermal growth factor receptor mutation was observed more in never-smokers (37%) compared to current (5%) and former smokers (9%). Both lung cancer-specific and overall survival were higher for never-smokers compared to current smokers.Conclusions: The observed differences in characteristics between never-smokers and smokers, and the higher survival in never-smokers compared to smokers from this large population-based study provide further evidence that lung cancer in never-smokers is clinically different to tobacco-associated lung cancer. The findings from this study emphasise the need for an improved understanding of genetics, pathogenesis, mechanisms and progression of non-tobacco associated lung cancer that may help prevent lung cancer or identify individually targeted treatments.
Collapse
Affiliation(s)
- Lukas Löfling
- Department of Medicine – Solna, Centre for Pharmacoepidemiology, Karolinska Institutet, T2 Karolinska University Hospital, Solna, Sweden
| | - Annette Karimi
- Department of Medical Science, Uppsala University Hospital, Uppsala, Sweden
| | | | - Shahram Bahmanyar
- Department of Medicine – Solna, Centre for Pharmacoepidemiology, Karolinska Institutet, T2 Karolinska University Hospital, Solna, Sweden
| | - Helle Kieler
- Department of Medicine – Solna, Centre for Pharmacoepidemiology, Karolinska Institutet, T2 Karolinska University Hospital, Solna, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Mats Lambe
- Regional Cancer Centre, Uppsala-Örebro, Uppsala, Sweden
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Solna, Sweden
| | - Kristina Lamberg
- Department of Medical Science, Uppsala University Hospital, Uppsala, Sweden
| | - Gunnar Wagenius
- Department of Oncology, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
16
|
Zhong J, Ren X, Chen Z, Zhang H, Zhou L, Yuan J, Li P, Chen X, Liu W, Wu D, Yang X, Liu J. miR-21-5p promotes lung adenocarcinoma progression partially through targeting SET/TAF-Iα. Life Sci 2019; 231:116539. [PMID: 31176779 DOI: 10.1016/j.lfs.2019.06.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/29/2019] [Accepted: 06/05/2019] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Although SET(I2PP2A) and miRNAs are reported to play a pivotal role in lung cancer, the underlying mechanisms have remained obscure. To address this issue, we investigated how miRNAs and SET participate in the progression of lung cancer. METHODS miRNAs that target SET were predicted from multiple miRNA databases. Three human NSCLC cell lines and two normal lung cell lines were used to evaluate aberrant miRNA and SET expressions. A dual luciferase reporter assay system was employed to verify the interaction between miRNA and SET. Stable miRNA knockdown and SET overexpression in A549 cells were achieved through lentivirus transfection; the corresponding influences on lung cancer progression were also examined. RESULTS In this study, A549 was the sole cell line to lack SET/TAF-Iα expression, which was inversely correlated with the up-regulation of miR-21-5p. SET was subsequently revealed as the direct target site of miR-21-5p in A549 cells. The stable miR-21-5p knockdown and SET/TAF-Iα overexpression were shown to markedly enhance the expression of SET/TAF-Iα and to inhibit the migration, invasion, proliferation as well as the in vivo tumorigenicity of A549 cells. CONCLUSION We suggest that SET/TAF-Iα might be a tumor suppressing factor regulated by miR-21-5p in lung adenocarcinoma. This might provide a target for lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Jiacheng Zhong
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xiaohu Ren
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Zhihong Chen
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Hang Zhang
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Li Zhou
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Jianhui Yuan
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Ping Li
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xiao Chen
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Wei Liu
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Desheng Wu
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Xifei Yang
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Jianjun Liu
- Institute of Toxicology, Shenzhen Center for Disease Control and Prevention, No 8 Longyuan Road, Nanshan District, Shenzhen 518055, Guangdong, China.
| |
Collapse
|
17
|
Shi K, Li N, Yang M, Li W. Identification of Key Genes and Pathways in Female Lung Cancer Patients Who Never Smoked by a Bioinformatics Analysis. J Cancer 2019; 10:51-60. [PMID: 30662525 PMCID: PMC6329865 DOI: 10.7150/jca.26908] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 10/17/2018] [Indexed: 01/10/2023] Open
Abstract
Smoking is considered the major risk factor for lung cancer, but only a small portion of female lung adenocarcinoma patients are associated with smoking. Thus, identifying crucial genes and pathways related to nonsmoking female lung cancer patients is of great importance. Gene expression profiles were downloaded from the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) databases. The R software packages were applied to screen the differentially expressed genes (DEGs). GO term enrichment and KEGG pathway analyses were carried out using DAVID tools. The protein-protein interaction (PPI) network was constructed by Cytoscape software. In total, 487 downregulated and 199 upregulated DEGs were identified. The down-regulated DEGs were mainly enriched for behavior and response to wounding, and the upregulated DEGs were significantly enriched for multicellular organismal metabolic process and cell division. The KEGG pathway analysis revealed that the downregulated DEGs were significantly enriched for cell adhesion molecules and neuroactive ligand-receptor interaction, while the upregulated DEGs were mainly enriched for cell cycle and the p53 signaling pathway. The top 10 hub genes and top 3 gene interaction modules were selected from the PPI network. Of the ten hub genes, a high expression of five genes was related to a poor OS in female lung cancer patients who never smoked, including IL6, CXCR2, FPR2, PPBP and HBA1. However, a low expression of GNG11, LRRK2, CDH5, CAV1 and SELE was associated with a worse OS for the female lung cancer patients who never smoked. In conclusion, our study provides novel insight for a better understanding of the pathogenesis of nonsmoking female lung cancer, and these identified DEGs may serve as biomarkers for diagnostics and treatment.
Collapse
Affiliation(s)
- Ke Shi
- Department of Geriatrics, Clinical Laboratory, Xiangya Hospital of Central South University, Changsha, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, People's Republic of China
| | - Na Li
- Department of Pathology, the First Affiliated Hospital of Hunan University of Medicine, Huaihua, People's Republic of China
| | - Meilan Yang
- Department of Pathology, the First Affiliated Hospital of Hunan University of Medicine, Huaihua, People's Republic of China
| | - Wei Li
- Department of Geriatrics, Clinical Laboratory, Xiangya Hospital of Central South University, Changsha, People's Republic of China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, People's Republic of China
| |
Collapse
|
18
|
Gridelli C, de Castro Carpeno J, Dingemans AMC, Griesinger F, Grossi F, Langer C, Ohe Y, Syrigos K, Thatcher N, Das-Gupta A, Truman M, Donica M, Smoljanovic V, Bennouna J. Safety and Efficacy of Bevacizumab Plus Standard-of-Care Treatment Beyond Disease Progression in Patients With Advanced Non-Small Cell Lung Cancer: The AvaALL Randomized Clinical Trial. JAMA Oncol 2018; 4:e183486. [PMID: 30177994 DOI: 10.1001/jamaoncol.2018.3486] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Bevacizumab treatment beyond progression has been investigated in breast and metastatic colorectal cancers. Avastin in All Lines Lung (AvaALL) is the first randomized phase 3 study of bevacizumab across multiple lines of treatment beyond progression in non-small cell lung cancer (NSCLC). Objective To assess the efficacy and safety of continuous bevacizumab treatment beyond first progression in NSCLC. Design, Setting, and Participants AvaALL was a randomized, open-label, phase 3b trial, conducted from 2011 to 2015 in 123 centers worldwide. Patients with nonsquamous NSCLC previously treated with first-line bevacizumab plus platinum-doublet chemotherapy and at least 2 cycles of bevacizumab maintenance were randomized (1:1) at first progression to receive bevacizumab plus standard of care (SOC) or SOC alone. Interventions Patients received bevacizumab (7.5 or 15 mg/kg intravenously every 21 days) and/or investigator's choice of SOC. For subsequent lines, patients treated with bevacizumab received SOC with or without bevacizumab; the SOC arm received SOC only. Main Outcomes and Measures The primary outcome was overall survival (OS). Secondary outcomes included progression-free survival from first to second (PFS2) and third progression (PFS3), time to second (TTP2) and third progression (TTP3), and safety. Results Between June 2011 and January 2015, 485 patients (median age, 63.0 years [range, 26-84 years]; 293 [60.4%] male) were randomized. Median OS was not significantly longer with bevacizumab plus SOC vs SOC alone: 11.9 (90% CI, 10.2-13.7) vs 10.2 (90% CI, 8.6-11.9) months (hazard ratio [HR], 0.84; 90% CI, 0.71-1.00; P = .104). Median PFS2 was numerically longer with bevacizumab plus SOC vs SOC alone: 5.5 (90% CI, 4.2-5.7) vs 4.0 (90% CI, 3.4-4.3) months (HR, 0.83; 90% CI, 0.70-0.98; P = .06). Median PFS3 appeared longer with bevacizumab plus SOC vs SOC alone: 4.0 (90% CI, 2.9-4.5) vs 2.6 (90% CI, 2.3-2.9) months (HR, 0.63; 90% CI, 0.49-0.83), as did TTP2 and TTP3. Grade 3/4 adverse events were more frequent with bevacizumab plus SOC (186 [76.5%]) vs SOC alone (140 [60.3%]). No new safety signals were observed. Conclusions and Relevance The primary end point was not met; however, OS was underpowered according to initial statistical assumptions. Continued therapy beyond first progression led to improved PFS3 (but not PFS2), TTP2, and TTP3. Although a result with P = .06 for PFS2 would conventionally be considered significant at a specified 2-sided α of .10, in the absence of adjustments for multiplicity, this result could be a chance finding. No new safety signals were identified with bevacizumab treatment beyond progression. Trial Registration clinicaltrialsregister.eu Identifier: 2010-022645-14; ClinicalTrials.gov identifier: NCT01351415.
Collapse
Affiliation(s)
- Cesare Gridelli
- Division of Medical Oncology, S.G. Moscati Hospital, Avellino, Italy
| | | | - Anne-Marie C Dingemans
- Department of Pulmonology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Frank Griesinger
- Department of Hematology and Oncology, University Department of Internal Medicine-Oncology, Pius-Hospital, University of Oldenburg, Oldenburg, Germany
| | - Francesco Grossi
- Lung Cancer Unit, Ospedale Policlinico San Martino, Genova, Italy
| | - Corey Langer
- Thoracic Oncology Unit, Abramson Cancer Center, University of Pennsylvania, Philadelphia
| | - Yuichiro Ohe
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | | | - Nick Thatcher
- Christie Hospital NHS Trust, Manchester, United Kingdom
| | | | - Matt Truman
- Roche Products Pty Ltd, Sydney, New South Wales, Australia.,Now with OzBiostat Pty Ltd, Manly, New South Wales, Australia
| | | | | | | |
Collapse
|
19
|
Calabrese EJ. The additive to background assumption in cancer risk assessment: A reappraisal. ENVIRONMENTAL RESEARCH 2018; 166:175-204. [PMID: 29890424 DOI: 10.1016/j.envres.2018.05.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/08/2018] [Accepted: 05/11/2018] [Indexed: 05/21/2023]
Abstract
The assumption that chemical and radiation induced cancers act in a manner that is additive to background was proposed in the mid-1970s. It was adopted by the U.S. Environmental Protection Agency (EPA) in 1986 and then subsequently by other regulatory agencies worldwide for cancer risk assessment. It ensured that cancer risks at low doses act in a linear fashion. The additive to background process assumes that the mechanism(s) resulting in induced (i.e., treatment related) and spontaneous (i.e., control group) cancers are identical. This assumption could not be properly evaluated due to inadequate mechanistic data when it was proposed in the 1970s. Using the findings of modern molecular toxicology, including oncogene activation/mutation, gene regulation, and molecular pathway analyses, the additive to background assumption was evaluated in the present paper. Based on published studies with 45 carcinogens over 13 diverse mammalian models and for a broad range of tumor types compelling evidence indicates that carcinogen-induced tumors are mediated in general via mechanisms that are not identical to those affecting the occurrence of the same type of spontaneous tumors in appropriate control groups. These findings, which challenge a fundamental assumption of the additive to background concept, have significant implications for cancer risk assessment policy, regulatory agency practices, as well as fundamental concepts of cancer biology.
Collapse
Affiliation(s)
- Edward J Calabrese
- Department of Environmental Health Sciences, University of Massachusetts, Morrill I, N344, Amherst, MA 01003, United States.
| |
Collapse
|
20
|
Zavala J, Krug JD, Warren SH, Krantz QT, King C, McKee J, Gavett SH, Lewandowski M, Lonneman WA, Kleindienst TE, Meier MJ, Higuchi M, Gilmour MI, DeMarini DM. Evaluation of an Air Quality Health Index for Predicting the Mutagenicity of Simulated Atmospheres. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2018; 52:3045-3053. [PMID: 29406743 PMCID: PMC5858694 DOI: 10.1021/acs.est.8b00613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
No study has evaluated the mutagenicity of atmospheres with a calculated air quality health index (AQHI). Thus, we generated in a UV-light-containing reaction chamber two simulated atmospheres (SAs) with similar AQHIs but different proportions of criteria pollutants and evaluated them for mutagenicity in three Salmonella strains at the air-agar interface. We continuously injected into the chamber gasoline, nitric oxide, and ammonium sulfate, as well as either α-pinene to produce SA-PM, which had a high concentration of particulate matter (PM): 119 ppb ozone (O3), 321 ppb NO2, and 1007 μg/m3 PM2.5; or isoprene to produce SA-O3, which had a high ozone (O3) concentration: 415 ppb O3, 633 ppb NO2, and 55 μg/m3 PM2.5. Neither PM2.5 extracts, NO2, or O3 alone, nor nonphoto-oxidized mixtures were mutagenic or cytotoxic. Both photo-oxidized atmospheres were largely direct-acting base-substitution mutagens with similar mutagenic potencies in TA100 and TA104. The mutagenic potencies [(revertants/h)/(mgC/m3)] of SA-PM (4.3 ± 0.4) and SA-O3 (9.5 ± 1.3) in TA100 were significantly different ( P < 0.0001), but the mutation spectra were not ( P = 0.16), being ∼54% C → T and ∼46% C → A. Thus, the AQHI may have some predictive value for the mutagenicity of the gas phase of air.
Collapse
Affiliation(s)
- Jose Zavala
- ORISE Research Fellow, National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Jonathan D. Krug
- National Environmental Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Sarah H. Warren
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Q. Todd Krantz
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Charly King
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - John McKee
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Stephen H. Gavett
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Michael Lewandowski
- National Environmental Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - William A. Lonneman
- National Environmental Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Tadeusz E. Kleindienst
- National Environmental Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - Matthew J. Meier
- Biology Department, Carleton University, Ottawa, Ontario K1S 5B6, Canada
| | - Mark Higuchi
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - M. Ian Gilmour
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| | - David M. DeMarini
- National Health and Environmental Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711, United States
| |
Collapse
|
21
|
Imperial R, Toor OM, Hussain A, Subramanian J, Masood A. Comprehensive pancancer genomic analysis reveals (RTK)-RAS-RAF-MEK as a key dysregulated pathway in cancer: Its clinical implications. Semin Cancer Biol 2017; 54:14-28. [PMID: 29175106 DOI: 10.1016/j.semcancer.2017.11.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 11/19/2017] [Indexed: 02/07/2023]
Abstract
Recent advances in Next Generation Sequencing (NGS) have provided remarkable insights into the genomic characteristics of human cancers that have spurred a revolution in the field of oncology. The mitogen-activated protein kinase pathway (MAPK) and its activating cell receptor, the receptor tyrosine kinases (RTKs), which together encompass the (RTK)-RAS-RAF-MEK-ERK axis, are central to oncogenesis. A pan-cancer genomics analysis presented in this review is made possible by large collaborative projects, including The Cancer Genome Atlas (TCGA), the International Cancer Genome Consortium (ICGC), and others. Landmark studies contributing to these projects have revealed alterations in cell signaling cascades that vary between cancer types and within tumors themselves. We review several of these studies in major tumor types to highlight recent advances in our understanding of the role of (RTK)-RAS-RAF alterations in cancer. Further studies are needed to increase the statistical power to detect clinically relevant low-frequency mutations, in addition to the known (RTK)-RAS-RAF pathway alterations, and to refine the resolution of the genomic landscape that defines these cancer mutations. The (RTK)-RAS-RAF-MEK-ERK mutation status, and their prognostic value, are also examined and correlated with clinical phenotypes. Treatments targeting various components of this pathway are ongoing, and are often effective initially in defined subgroups of patients. However, resistance to these agents can develop through adaptive mechanisms. With our steady increase in understanding the molecular biology of cancer, ongoing evaluation and monitoring through genomic analysis will continue to provide important information to the clinician in the context of treatment selection, response, resistance and outcomes.
Collapse
Affiliation(s)
- Robin Imperial
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA
| | - Omer M Toor
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA; Division of Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA; Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA
| | - Arif Hussain
- Division of Oncology, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA; The Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Janakiraman Subramanian
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA; Division of Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA; Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA
| | - Ashiq Masood
- Department of Medicine, University of Missouri Kansas City School of Medicine, Kansas City, MO 64108, USA; Division of Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA; Center for Precision Oncology, Saint Luke's Cancer Institute, Kansas City, MO 64111, USA.
| |
Collapse
|
22
|
Environmental tobacco smoke exposure and EGFR and ALK alterations in never smokers' lung cancer. Results from the LCRINS study. Cancer Lett 2017; 411:130-135. [PMID: 28987389 DOI: 10.1016/j.canlet.2017.09.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/22/2017] [Accepted: 09/25/2017] [Indexed: 01/02/2023]
Abstract
Environmental tobacco smoke (ETS) exposure is a main risk factor of lung cancer in never smokers. Epidermal Growth Factor Receptor (EGFR) mutations and ALK translocations are more frequent in never smokers' lung cancer than in ever-smokers. We performed a multicenter case-control study to assess if ETS exposure is associated with the presence of EGFR mutations and its types and if ALK translocations were related with ETS exposure. All patients were never smokers and had confirmed lung cancer diagnosis. ETS exposure during childhood showed a negative association on the probability of EGRF mutation though not significant. Exposure during adulthood, at home or at workplace, did not show any association with EGFR mutation. The mutation type L858R seemed the most associated with a lower probability of EGFR alterations for ETS exposure at home in adult life. There is no apparent association between ETS exposure and ALK translocation. These results might suggest that ETS exposure during childhood or at home in adult life could influence the EGFR mutations profile in lung cancer in never smokers, reducing the probability of presenting EFGR mutation.
Collapse
|
23
|
Akhtar N, Bansal JG. Risk factors of Lung Cancer in nonsmoker. Curr Probl Cancer 2017; 41:328-339. [PMID: 28823540 DOI: 10.1016/j.currproblcancer.2017.07.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/12/2017] [Accepted: 07/17/2017] [Indexed: 12/18/2022]
Abstract
Generally, the cause of lung cancer is attributed to tobacco smoking. But many of the new lung cancer cases have been reported in nonsmokers. Apart from smoking; air pollution, environmental exposure, mutations, and single-nucleotide polymorphisms are known to be associated with lung cancer. Improper diet, alcohol consumption, marijuana smoking, estrogen, infections with human papillomavirus (HPV), HIV, and Epstein-Barr virus are suggested to be linked with lung cancer but clear evidences to ascertain their relation is not available. This article provides a comprehensive review of various risk factors and the underlying molecular mechanisms responsible for increasing the incidence of lung cancer. The pathologic, histologic, and genetic differences exist with lung cancer among smokers and nonsmokers. A better understanding of the risk factors, differences in pathology and molecular features of lung cancer in smokers and nonsmokers and the mode of action of various carcinogens will facilitate the prevention and management of lung cancer.
Collapse
Affiliation(s)
- Nahid Akhtar
- Department of Bioengineering and Biosciences, Lovely Professional University (LPU), Phagwara, Punjab, India
| | - Jeena Gupta Bansal
- Department of Bioengineering and Biosciences, Lovely Professional University (LPU), Phagwara, Punjab, India.
| |
Collapse
|
24
|
Cha YK, Lee HY, Ahn MJ, Park K, Ahn JS, Sun JM, Choi YL, Lee KS. The impact of smoking status on radiologic tumor progression patterns and response to epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors in lung adenocarcinoma with activating EGFR mutations. J Thorac Dis 2017; 8:3175-3186. [PMID: 28066597 DOI: 10.21037/jtd.2016.11.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the impact of smoking on the treatment outcome of epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) in patients with EGFR-mutant lung adenocarcinoma, with consideration of other factors including radiologic tumor progression pattern according to patient smoking status. METHODS A total of 224 patients with EGFR mutant lung adenocarcinomas that were treated with EGFR-TKIs were retrospectively reviewed. Radiologic tumor progression pattern and treatment outcomes were evaluated according to smoking history. RESULTS There were no significant differences in radiologic tumor progression pattern based on smoking status. There were no significant differences in survival between never-smokers and smokers or among never-, former-, and current-smokers, but there was a trend of shorter progression free survival (PFS) and poorer overall survival (OS) in smokers compared with never-smokers. In multivariate analysis, long-term smokers had shorter PFS and poorer OS than those who had never smoked. CONCLUSIONS A history of smoking had no significant effect on radiologic tumor progression pattern; however, smoking history is a negative predictive factor of survival in patients with EGFR-mutant lung adenocarcinoma undergoing EGFR-TKI therapy.
Collapse
Affiliation(s)
- Yoon Ki Cha
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea;; Department of Radiology, Dongguk University Ilsan Hospital, Dongguk University, Goyang, South Korea
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoon-La Choi
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung Soo Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Ruano-Ravina A, Torres-Durán M, Kelsey KT, Parente-Lamelas I, Leiro-Fernández V, Abdulkader I, Abal-Arca J, Montero-Martínez C, Vidal-García I, Amenedo M, Castro-Añón O, Golpe-Gómez A, González-Barcala J, Martínez C, Guzmán-Taveras R, Provencio M, Mejuto-Martí MJ, Fernández-Villar A, Barros-Dios JM. Residential radon, EGFR mutations and ALK alterations in never-smoking lung cancer cases. Eur Respir J 2016; 48:1462-1470. [PMID: 27799390 DOI: 10.1183/13993003.00407-2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/26/2016] [Indexed: 11/05/2022]
Abstract
The aim of this study was to assess if residential radon exposure might cause EGFR mutations or ALK rearrangements in never-smokers.We designed a multicentre case-control study in a radon-prone area (Galicia, Spain); only lung cancer cases were included in the study. We obtained residential radon measurements and clinical information for all the participants. We compared the median values of residential radon between patients with EGFR mutations or ALK rearrangements versus those without them.323 patients were included. Median age was 70 years and 19.5% were males. 42 and 15% of patients were EGFR- and ALK-positive, respectively. The most frequent EGFR alterations were exon 19 deletions and exon 21 (L858R) single-point substitution mutations. ALK-positive patients were 10 years younger than ALK-negative patients. Residential radon levels were two-fold higher in patients with exon 19 deletions compared with patients with exon 21 (L858R) single-point substitution mutations (216 versus 118 Bq·m-3; p=0.057). There were no differences in residential radon levels by EGFR mutation status. ALK-positive patients (n=12) essentially had two-fold residential radon levels compared with ALK-negative patients (290 versus 164 Bq·m-3, respectively).Residential radon may have a role in the molecular signature of lung cancer in never-smokers, although more studies with larger sample sizes are needed to support this hypothesis.
Collapse
Affiliation(s)
- Alberto Ruano-Ravina
- Dept of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain .,CIBER de Epidemiología y Salud Pública, CIBERESP, Madrid, Spain.,Dept of Epidemiology, Brown School of Public Health, Brown University, Providence, RI, USA
| | | | - Karl T Kelsey
- Dept of Epidemiology, Brown School of Public Health, Brown University, Providence, RI, USA
| | | | | | - Ihab Abdulkader
- Service of Pathology, Santiago de Compostela University Clinic Hospital, Santiago de Compostela, Spain
| | - José Abal-Arca
- Service of Pneumology, Ourense Hospital Complex, Ourense, Spain
| | | | - Iria Vidal-García
- Service of Pneumology, University Hospital Complex of A Coruña, A Coruña, Spain
| | | | | | - Antonio Golpe-Gómez
- Service of Pneumology, Santiago de Compostela University Clinic Hospital, Santiago de Compostela, Spain
| | - Javier González-Barcala
- Service of Pneumology, Santiago de Compostela University Clinic Hospital, Santiago de Compostela, Spain
| | - Cristina Martínez
- National Institute of Silicosis, University Hospital of Asturias, Oviedo, Spain
| | | | - Mariano Provencio
- Service of Oncology, Puerta de Hierro University Hospital, Madrid, Spain
| | | | | | - Juan Miguel Barros-Dios
- Dept of Preventive Medicine and Public Health, University of Santiago de Compostela, Santiago de Compostela, Spain.,CIBER de Epidemiología y Salud Pública, CIBERESP, Madrid, Spain.,Service of Preventive Medicine, University Hospital Complex of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
26
|
Asić K. Dominant mechanisms of primary resistance differ from dominant mechanisms of secondary resistance to targeted therapies. Crit Rev Oncol Hematol 2016; 97:178-96. [DOI: 10.1016/j.critrevonc.2015.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 06/18/2015] [Accepted: 08/04/2015] [Indexed: 02/07/2023] Open
|