• This record comes from PubMed

Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: the challenge ahead

. 2015 Jun ; 36 Suppl 1 (Suppl 1) : S254-96.

Language English Country Great Britain, England Media print

Document type Journal Article, Research Support, N.I.H., Extramural, Research Support, Non-U.S. Gov't, Research Support, U.S. Gov't, Non-P.H.S., Review

Grant support
N01-ES35504 NIEHS NIH HHS - United States
T32 ES007126 NIEHS NIH HHS - United States
R03 DK089010 NIDDK NIH HHS - United States
KL2TR000063 NCATS NIH HHS - United States
R01-DK52825 NIDDK NIH HHS - United States
G12 RR003062 NCRR NIH HHS - United States
R01 CA184384 NCI NIH HHS - United States
G12 MD007581 NIMHD NIH HHS - United States
MR/L007215/1 Medical Research Council - United Kingdom
R01 CA113447 NCI NIH HHS - United States
CA163069 NCI NIH HHS - United States
P30 ES000210 NIEHS NIH HHS - United States
R13ES023276 NIEHS NIH HHS - United States
T32CA09151 NCI NIH HHS - United States
P20 GM103476 NIGMS NIH HHS - United States
R01-CA141704 NCI NIH HHS - United States
P50 CA114747 NCI NIH HHS - United States
UL1TR000039 NCATS NIH HHS - United States
HHSN27320140003C PHS HHS - United States
R01 CA192613 NCI NIH HHS - United States
R01 CA170378 PQ22 NCI NIH HHS - United States
R01-CA150214 NCI NIH HHS - United States
CA92306 NCI NIH HHS - United States
K01 DK075386 NIDDK NIH HHS - United States
T32 CA009151 NCI NIH HHS - United States
MOP-115019 CIHR - Canada
R01 CA92306-S1 NCI NIH HHS - United States
U54 CA151459 NCI NIH HHS - United States
R21 CA169964 NCI NIH HHS - United States
G12 MD007590 NIMHD NIH HHS - United States
ES016893 NIEHS NIH HHS - United States
CA116812 NCI NIH HHS - United States
T32 CA009686 NCI NIH HHS - United States
U54 MD007593 NIMHD NIH HHS - United States
MD007593 NIMHD NIH HHS - United States
P30 CA016058 NCI NIH HHS - United States
R01 DK105118 NIDDK NIH HHS - United States
R01-ES022968 NIEHS NIH HHS - United States
U54 CA149145 NCI NIH HHS - United States
MD004038 NIMHD NIH HHS - United States
G12MD007581 NIMHD NIH HHS - United States
R01 ES016893 NIEHS NIH HHS - United States
R01-CA61774 NCI NIH HHS - United States
R01 CA092306 NCI NIH HHS - United States
SC1CA153326 NCI NIH HHS - United States
Cancer Research UK - United Kingdom
R13 ES023276 NIEHS NIH HHS - United States
R01 CA118374 NCI NIH HHS - United States

Lifestyle factors are responsible for a considerable portion of cancer incidence worldwide, but credible estimates from the World Health Organization and the International Agency for Research on Cancer (IARC) suggest that the fraction of cancers attributable to toxic environmental exposures is between 7% and 19%. To explore the hypothesis that low-dose exposures to mixtures of chemicals in the environment may be combining to contribute to environmental carcinogenesis, we reviewed 11 hallmark phenotypes of cancer, multiple priority target sites for disruption in each area and prototypical chemical disruptors for all targets, this included dose-response characterizations, evidence of low-dose effects and cross-hallmark effects for all targets and chemicals. In total, 85 examples of chemicals were reviewed for actions on key pathways/mechanisms related to carcinogenesis. Only 15% (13/85) were found to have evidence of a dose-response threshold, whereas 59% (50/85) exerted low-dose effects. No dose-response information was found for the remaining 26% (22/85). Our analysis suggests that the cumulative effects of individual (non-carcinogenic) chemicals acting on different pathways, and a variety of related systems, organs, tissues and cells could plausibly conspire to produce carcinogenic synergies. Additional basic research on carcinogenesis and research focused on low-dose effects of chemical mixtures needs to be rigorously pursued before the merits of this hypothesis can be further advanced. However, the structure of the World Health Organization International Programme on Chemical Safety 'Mode of Action' framework should be revisited as it has inherent weaknesses that are not fully aligned with our current understanding of cancer biology.

Advanced Molecular Science Research Centre King George's Medical University Lucknow Uttar Pradesh 226 003 India

Analytische Milieu en Geochemie Vrije Universiteit Brussel Brussel B1050 Belgium

Baylor College of Medicine Houston TX 77030 USA

California Pacific Medical Center Research Institute 2100 Webster Street 401 San Francisco CA 94115 USA Getting to Know Cancer Room 229A 36 Arthur Street Truro Nova Scotia B2N 1X5 Canada Lancaster Environment Centre Lancaster University Bailrigg Lancaster LA1 4AP UK Institute for Health and the Environment University at Albany 5 University Pl Rensselaer NY 12144 USA Getting to Know Cancer Guelph N1G 1E4 Canada School of Biotechnology Faculty of Agriculture Biotechnology and Food Sciences Sultan Zainal Abidin University Tembila Campus 22200 Besut Terengganu Malaysia Department of Pharmacology and Toxicology Faculty of Pharmacy University of Navarra Pamplona 31008 Spain Department of Pharmacology and Toxicology Ernest Mario School of Pharmacy Rutgers State University of New Jersey Piscataway NJ 08854 USA Instituto de Biomedicina de Sevilla Consejo Superior de Investigaciones Cientificas Hospital Universitario Virgen del Rocio Univ de Sevilla Avda Manuel Siurot sn 41013 Sevilla Spain Department of Experimental and Clinical Medicine University of Firenze Florence 50134 Italy School of Biological Sciences University of Reading Hopkins Building Reading Berkshire RG6 6UB UK Department of Nutrition University of Oslo Oslo Norway Department of Biochemistry and Biology University of Bath Claverton Down Bath BA2 7AY UK Department of Public Health Sciences College of Medicine Pennsylvania State University Hershey PA 17033 USA Center for Environmental Carcinogenesis and Risk Assessment Environmental Protection and Health Prevention Agency 40126 Bologna Italy Department of Chemicals and Radiation Division of Environmental Medicine Norwegian Institute of Public Health Oslo N 0403 Norway Planet Biotechnologies Inc St Albert Alberta T8N 5K4 Canada Department of Molecular and Cellular Biochemistry University of Kentucky Lexington KY 40508 USA Spanish National Cancer Research Centre CNI

Cancer Research UK Cambridge Institute University of Cambridge Robinson Way Cambridge CB2 0RE UK

Center for Environment and Water Research Institute King Fahd University of Petroleum and Minerals Dhahran 3126 Saudi Arabia

Center for Environmental Carcinogenesis and Risk Assessment Environmental Protection and Health Prevention Agency 40126 Bologna Italy

Center for Experimental Molecular Medicine Academic Medical Center Meibergdreef 9 Amsterdam 1105 AZ The Netherlands

Center for Genomic Science of Fondazione Istituto Italiano di Tecnologia Via Adamello 16 20139 Milano Italy

Center for Radiological Research Columbia University Medical Center New York NY 10032 USA Instituto de Alta Investigacion Universidad de Tarapaca Arica Chile

Centre De Recherche En Cancerologie De Lyon Lyon U1052 UMR5286 France

Charles University Prague 3rd Faculty of Medicine CZ 100 00 Prague 10 Czech Republic

Computer Science Department Southern Illinois University Carbondale IL 62901 USA

Department of Anatomy and Cell Biology University of Western Ontario London Ontario N6A 3K7 Canada

Department of Anatomy College of Medicine Yeungnam University Daegu 705 717 South Korea

Department of Anatomy Physiology and Biochemistry The Swedish University of Agricultural Sciences PO Box 7011 VHC Almas Allé 4 SE 756 51 Uppsala Sweden

Department of Applied Medical Sciences University of Southern Maine 96 Falmouth St Portland ME 04104 USA

Department of Biochemistry and Biology University of Bath Claverton Down Bath BA2 7AY UK

Department of Biochemistry and Cancer Biology Meharry Medical College Nashville TN 37208 USA

Department of Biochemistry and Molecular Biology Dalhousie University Halifax Nova Scotia B3H 4R2 Canada

Department of Biochemistry Rush University Chicago IL 60612 USA

Department of Biological Chemical and Pharmaceutical Sciences and Technologies Polyclinic Plexus University of Palermo Palermo 90127 Italy

Department of Biological Chemical and Pharmaceutical Sciences and Technologies Polyclinic Plexus University of Palermo Palermo 90127 Italy Sbarro Institute for Cancer Research and Molecular Medicine Temple University Philadelphia PA 19122 USA

Department of Biological Chemistry Medical School University of Athens Institute of Molecular Medicine and Biomedical Research 10676 Athens Greece

Department of Biology Center for Cancer Research and Therapeutic Development Clark Atlanta University Atlanta GA 30314 USA

Department of Biology Geology and Environmental Sciences Alma Mater Studiorum Università di Bologna Via Francesco Selmi 3 40126 Bologna Italy Center for Applied Biomedical Research S Orsola Malpighi University Hospital Via Massarenti 9 40126 Bologna Italy National Institute of Biostructures and Biosystems Viale Medaglie d' Oro 305 00136 Roma Italy

Department of Biology Jackson State University Jackson MS 39217 USA

Department of Biomedical Engineering and Cancer Biology Vanderbilt University Nashville TN 37235 USA

Department of Biomedical Sciences Faculty of Medicine and Health Sciences 43400 Universiti Putra Malaysia Serdang Selangor Malaysia

Department of Biomedical Sciences University of Cagliari 09124 Cagliari Italy

Department of Bioscience and Biotechnology Sejong University Seoul 143 747 Korea

Department of Biosciences and Veterinary Public Health Faculty of Veterinary Medicine Swedish University of Agricultural Sciences PO Box 7028 75007 Uppsala Sweden

Department of Biotechnology and Biosciences University of Milano Bicocca 20126 Milan Italy SYSBIO Centre of Systems Biology Department of Biotechnology and Biosciences University of Milano Bicocca 20126 Milan Italy

Department of Cancer Biology Perelman School of Medicine at the University of Pennsylvania Philadelphia PA 19104 USA

Department of Cardiac Thoracic and Vascular Sciences Unit of Occupational Medicine University of Padova Padova 35128 Italy

Department of Cell Biology Pharmamar SAU Avda De los Reyes 1 28770 Colmenar Viejo Madrid Spain

Department of Cellular and Physiological Sciences Life Sciences Institute Faculty of Medicine The University of British Columbia Vancouver British Columbia V5Z 1M9 Canada

Department of Chemicals and Radiation Division of Environmental Medicine Norwegian Institute of Public Health Oslo N 0403 Norway

Department of Cytokinetics Institute of Biophysics Academy of Sciences of the Czech Republic Brno CZ 61265 Czech Republic

Department of Environmental and Occupational Health University of Arkansas for Medical Sciences Little Rock AR 72205 USA

Department of Environmental and Radiological Health Sciences Colorado State University Fort Collins CO 80523 1680 USA

Department of Environmental Health Indiana University School of Public Health Bloomington IN 47405 USA

Department of Experimental and Clinical Medicine University of Firenze Florence 50134 Italy

Department of Experimental Oncology Mediterranean Institute of Oncology Via Penninazzo 7 Viagrande 95029 Italy

Department of Genomic Medicine and Environmental Toxicology Institute for Biomedical Research National Autonomous University of Mexico Mexico City 04510 México

Department of Geriatric Medicine Kyoto University Hospital 54 Kawaharacho Shogoin Sakyo ku Kyoto 606 8507 Japan

Department of Head and Neck Surgery Head and Neck Cancer Research Johns Hopkins University School of Medicine Baltimore MD 21205 USA

Department of Hygiene Kawasaki Medical School Matsushima Kurashiki Okayama 701 0192 Japan

Department of Life Sciences College of Health and Life Sciences and the Health and Environment Theme Institute of Environment Health and Societies Brunel University Kingston Lane Uxbridge Middlesex UB8 3PH UK

Department of Life Sciences Tzu Chi University Hualien 970 Taiwan

Department of Medicine McGill University Montreal Quebec H4A 3J1 Canada

Department of Medicine Medical Oncology Rutgers Cancer Institute of New Jersey New Brunswick NJ 08903 USA

Department of Medicine Oncology and Pathology Stanford University Stanford CA 94305 USA

Department of Medicine University of Louisville Louisville KY 40202 USA Robley Rex VA Medical Center Louisville KY 40202 USA

Department of Microbiology and Immunology School of Medicine College of Medicine Taipei Medical University Taipei 11031 Taiwan

Department of Microbiology and Immunology Virginia Commonwealth University Massey Cancer Center Richmond VA 23298 USA

Department of Molecular and Cellular Biochemistry University of Kentucky Lexington KY 40508 USA

Department of Molecular and Environmental Toxicology University of Wisconsin Madison Madison WI 53706 USA

Department of Molecular and Experimental Medicine The Scripps Research Institute La Jolla CA 92037 USA

Department of Molecular Biology School of Osteopathic Medicine Rowan University Stratford NJ 08084 USA

Department of Molecular Medicine and Medical Biotechnology Federico 2 University of Naples 80131 Naples Italy

Department of Natural Science The City University of New York at Hostos Campus Bronx NY 10451 USA

Department of Nutrition University of Oslo Oslo Norway

Department of Obstetrics and Gynecology University of Melbourne Victoria 3052 Australia

Department of Otolaryngology Head and Neck Surgery University of Michigan Medical School Ann Arbor MI 48109 USA

Department of Pathology Kuwait University Safat 13110 Kuwait

Department of Pathology Stony Brook School of Medicine Stony Brook University The State University of New York Stony Brook NY 11794 8691 USA

Department of Pathology Wayne State University Detroit MI 48201 USA

Department of Pharmaceutical Chemistry Faculty of Pharmacy Kuwait University PO Box 24923 Safat 13110 Kuwait and

Department of Pharmaceutical Sciences West Virginia University Morgantown WV 26506 USA

Department of Pharmacological and Biomolecular Sciences Università degli Studi di Milano 20133 Milan Italy

Department of Pharmacology and Toxicology Ernest Mario School of Pharmacy Rutgers State University of New Jersey Piscataway NJ 08854 USA

Department of Pharmacology and Toxicology Faculty of Pharmacy University of Navarra Pamplona 31008 Spain

Department of Pharmacology and Toxicology University of Vienna Vienna A 1090 Austria Institute of Cancer Research Department of Medicine Medical University of Vienna Wien 1090 Austria

Department of Public Health Sciences College of Medicine Pennsylvania State University Hershey PA 17033 USA

Department of Surgery Pathology Immunology Duke University Medical Center Durham NC 27710 USA

Department of Surgery The Ohio State University College of Medicine The James Comprehensive Cancer Center Columbus OH 43210 USA

Department of Toxicology NUTRIM School for Nutrition Toxicology and Metabolism Maastricht University Maastricht 6200 The Netherlands

Department of Urology Kasr Al Ainy School of Medicine Cairo University El Manial Cairo 11559 Egypt

Department of Urology University of California Davis Sacramento CA 95817 USA

Departments of Neurosurgery and Biochemistry and Massey Cancer Center Virginia Commonwealth University Richmond VA 23298 USA

Departments of Obstetrics and Gynecology and Pathology Pennsylvania State University College of Medicine Hershey PA 17033 USA

Departments of Pharmacology and Hematology and Medical Oncology Emory University School of Medicine and Winship Cancer Institute Atlanta GA 30322 USA

Division of Environmental Health Sciences School of Public Health University of California Berkeley CA 94720 7360 USA

Division of Genetic and Molecular Toxicology National Center for Toxicological Research United States Food and Drug Administration Jefferson AR 72079 USA

Division of Hematology and Oncology Department of Pediatrics Children's Healthcare of Atlanta GA 30322 USA Department of Pediatrics Emory University School of Medicine Emory University Atlanta GA 30322 USA

Division of Preclinical Innovation National Center for Advancing Translational Sciences National Institutes of Health 9800 Medical Center Drive Bethesda MD 20892 3375 USA

Environmental and Molecular Toxicology Environmental Health Science Center Oregon State University Corvallis OR 97331 USA

Environmental Risk and Health Unit Flemish Institute for Technological Research 2400 Mol Belgium

Faculty of Medicine University of Otago Dunedin 9054 New Zealand

Geisel School of Medicine at Dartmouth Hanover NH 03755 USA Geisel School of Medicine at Dartmouth Hanover NH 03755 USA

Getting to Know Cancer Guelph N1G 1E4 Canada

Getting to Know Cancer Room 229A 36 Arthur Street Truro Nova Scotia B2N 1X5 Canada Lancaster Environment Centre Lancaster University Bailrigg Lancaster LA1 4AP UK

Hacettepe University Center for Stem Cell Research and Development Ankara 06640 Turkey

Harvard Medical School Brigham and Women's Hospital Boston MA 02115 USA

Human Genetics Unit Department of Pathology Faculty of Medicine Kuwait University Jabriya 13110 Kuwait

Human Safety and Environmental Research Department of Health Sciences College of North Atlantic Doha 24449 State of Qatar

Inflammation and Cancer Research National Cancer Institute National Institutes of Health Bethesda MD 20892 USA

INRS Institut Armand Frappier 531 Boulevard des Prairies Laval QC H7V 1B7 Canada

INSERM U1113 team 3 'Cell Signalling and Communication in Kidney and Prostate Cancer' University of Strasbourg Faculté de Médecine 67085 Strasbourg France

INSERM U1113 team 3 'Cell Signalling and Communication in Kidney and Prostate Cancer' University of Strasbourg Faculté de Médecine 67085 Strasbourg France Department of Cell and Developmental Biology University of California Irvine CA 92697 USA

Institut De Recerca Hospital Vall D'Hebron Passeig Vall d'Hebron 119 129 08035 Barcelona Spain

Institute for Health and the Environment University at Albany 5 University Pl Rensselaer NY 12144 USA

Instituto de Alta Investigacion Universidad de Tarapaca Arica Chile

Instituto de Biomedicina de Sevilla Consejo Superior de Investigaciones Cientificas Hospital Universitario Virgen del Rocio Univ de Sevilla Avda Manuel Siurot sn 41013 Sevilla Spain

Integrated Laboratory Systems Inc in support of the National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods RTP NC 27709 USA

Istituto di Genetica Molecolare CNR Via Abbiategrasso 207 27100 Pavia Italy

Laboratory for Cell Genetics Vrije Universiteit Brussel 1050 Brussels Belgium

Lancaster Environment Centre Lancaster University Bailrigg Lancaster LA1 4AP UK

Lineberger Comprehensive Cancer Center University of North Carolina at Chapel Hill NC 27599 USA

Lombardi Comprehensive Cancer Center Georgetown University Medical Center Washington DC 20057 USA

Mouse Models of Cancer Therapies Group Vall d'Hebron Institute of Oncology 08035 Barcelona Spain

Mouse Models of Cancer Therapies Group Vall d'Hebron Institute of Oncology Barcelona 08010 Spain

National Institute for Environmental Studies 16 2 Onogawa Tsukuba Ibraki 3058506 Japan

Planet Biotechnologies Inc St Albert Alberta T8N 5K4 Canada

Population Health and Prevention Research Prevention and Cancer Control Cancer Care Ontario Toronto Ontario M5G 2L7 Canada Departments of Epidemiology and Occupational and Environmental Health Dalla Lana School of Public Health University of Toronto Toronto Ontario M5T 3M7 Canada

Program in Genetics and Molecular Biology Graduate Division of Biological and Biomedical Sciences Emory University Atlanta GA 30322 USA

Regulatory Toxicology Research Division Bureau of Chemical Safety Food Directorate Health Canada Ottawa Ontario K1A 0K9 Canada

School of Biological Sciences The University of Reading Whiteknights Reading RG6 6UB England

School of Biological Sciences University of Reading Hopkins Building Reading Berkshire RG6 6UB UK

School of Biological Sciences University of Reading Reading RG6 6UB UK

School of Biomedical Sciences The Chinese University of Hong Kong Shatin NT Hong Kong SAR The People's Republic of China

School of Biotechnology Faculty of Agriculture Biotechnology and Food Sciences Sultan Zainal Abidin University Tembila Campus 22200 Besut Terengganu Malaysia

School of Biotechnology Yeungnam University Gyeongbuk 712 749 South Korea

School of Engineering and Materials Science Queen Mary University of London Mile End Road London E1 4NS UK

School of Public Health Curtin University Bentley WA 6102 Australia Department of Urology University of California Davis Sacramento CA 95817 USA

Siriraj Center of Excellence for Stem Cell Research Faculty of Medicine Siriraj Hospital Mahidol University Bangkok 10700 Thailand

Spanish National Cancer Research Centre CNIO Melchor Fernandez Almagro 3 28029 Madrid Spain

Stanford University Department of Medicine Division of Oncology Stanford CA 94305 USA

Superfund Research Program National Institute of Environmental Health Sciences Research Triangle Park NC 27560 USA

United States Army Institute of Public Health Toxicology Portfolio Health Effects Research Program Aberdeen Proving Ground Edgewood MD 21010 5403 USA

University of Puerto Rico Medical Sciences Campus School of Public Health Nutrition Program San Juan 00921 Puerto Rico

White River Junction Veterans Affairs Medical Center White River Junction VT 05009 USA Geisel School of Medicine at Dartmouth Hanover NH 03755 USA

Erratum In

PubMed

See more in PubMed

(2014) World cancer report 2014. In Wild C.P. and Stewart B.W (eds). World Health Organization.

Malhotra J. (2014) Molecular and genetic epidemiology of cancer in low- and medium-income countries. Ann. Glob. Health, 80, 418–425. PubMed

McGuinn L.A., et al. (2012) Cancer and environment: definitions and misconceptions. Environ. Res., 112, 230–234. PubMed PMC

Sankpal U.T., et al. (2012) Environmental factors in causing human cancers: emphasis on tumorigenesis. Tumour Biol., 33, 1265–1274. PubMed

Trosko J.E., et al. (2005) The emperor wears no clothes in the field of carcinogen risk assessment: ignored concepts in cancer risk assessment. Mutagenesis, 20, 81–92. PubMed

Christiani D.C. (2011) Combating environmental causes of cancer. N. Engl. J. Med., 364, 791–793. PubMed

Clapp R. (2011) Chemicals policy in the 2008-2009 President's Cancer Panel Report. New Solut., 21, 447–455. PubMed

Reuben S.H. (2008–2009) Reducing environmental cancer risk: what we can do now. In Panel T.P.s.C. (ed.), Bethesda, Maryland.

Parkin D.M., et al. (2011) The fraction of cancer attributable to lifestyle and environmental factors in the UK in 2010. Br. J. Cancer, 105(Suppl 2), S77–S81. PubMed PMC

(2009) Global Health Risks: Mortality and Burden of Disease Attributable to Selected Major Risks. World Health Organization, Geneva.

Straif K. (2008) The burden of occupational cancer. Occup. Environ. Med., 65, 787–788. PubMed

Vandenberg L.N., et al. (2012) Hormones and endocrine-disrupting chemicals: low-dose effects and nonmonotonic dose responses. Endocr. Rev., 33, 378–455. PubMed PMC

(2009) OECD Guidelines for the Testing of Chemicals, Section 4 Health Effects, Test No. 451: Carcinogenicity Studies. OECD.

Wignall J.A., et al. (2014) Standardizing benchmark dose calculations to improve science-based decisions in human health assessments. Environ. Health Perspect., 122, 499–505. PubMed PMC

Myers J.P., et al. (2009) A clash of old and new scientific concepts in toxicity, with important implications for public health. Environ. Health Perspect., 117, 1652–1655. PubMed PMC

Vandenberg L.N., et al. (2013) Regulatory decisions on endocrine disrupting chemicals should be based on the principles of endocrinology. Reprod. Toxicol., 38, 1–15. PubMed PMC

Bergman A., et al. (2013) Science and policy on endocrine disrupters must not be mixed: a reply to a “common sense” intervention by toxicology journal editors. Environ. Health, 12, 69. PubMed PMC

Ames B.N. (1979) Identifying environmental chemicals causing mutations and cancer. Science, 204, 587–593. PubMed

Armitage P., et al. (1954) The age distribution of cancer and a multi-stage theory of carcinogenesis. Br. J. Cancer, 8, 1–12. PubMed PMC

Truhaut R. (1990) [Recent progress in the evaluation of the dangers of chemical carcinogens]. J. Pharm. Belg., 45, 131–140. PubMed

Hanahan D., et al. (2000) The hallmarks of cancer. Cell, 100, 57–70. PubMed

Preston R.J. (2005) Extrapolations are the Achilles heel of risk assessment. Mutat. Res., 589, 153–157. PubMed

Hanahan D., et al. (2011) Hallmarks of cancer: the next generation. Cell, 144, 646–674. PubMed

Colotta F., et al. (2009) Cancer-related inflammation, the seventh hallmark of cancer: links to genetic instability. Carcinogenesis, 30, 1073–1081. PubMed

Warburg O. (ed.) (1930) The Metabolism of Tumours: Investigations from the Kaiser Wilhelm Institute for Biology, Berlin-Dahlen. Constable & Company Limited, London.

Aisenberg A.C. (1961) The Glycolysis and Respiration of Tumors. Academic Press, New York, NY.

Ankley G.T., et al. (2010) Adverse outcome pathways: a conceptual framework to support ecotoxicology research and risk assessment. Environ. Toxicol. Chem., 29, 730–741. PubMed

Groh K.J., et al. (2015) Development and application of the adverse outcome pathway framework for understanding and predicting chronic toxicity: I. Challenges and research needs in ecotoxicology. Chemosphere, 120, 764–777. PubMed

Kleinstreuer N.C., et al. (2013) In vitro perturbations of targets in cancer hallmark processes predict rodent chemical carcinogenesis. Toxicol. Sci., 131, 40–55. PubMed

Yates L.R., et al. (2012) Evolution of the cancer genome. Nat. Rev. Genet., 13, 795–806. PubMed PMC

(2001) National Toxicology Program’s report of the endocrine disruptors low dose peer review. National Institute of Environmental Health Sciences, National Toxicology Program, Research Triangle Park, NC. PubMed PMC

Melnick R., et al. (2002) Summary of the National Toxicology Program's report of the endocrine disruptors low-dose peer review. Environ. Health Perspect., 110, 427–431. PubMed PMC

Welshons W.V., et al. (2006) Large effects from small exposures. III. Endocrine mechanisms mediating effects of bisphenol A at levels of human exposure. Endocrinology, 147(Suppl 6), S56–S69. PubMed

Vandenberg L.N., et al. (2007) Human exposure to bisphenol A (BPA). Reprod. Toxicol., 24, 139–177. PubMed

Brucker-Davis F., et al. (2001) Significant effects of mild endogenous hormonal changes in humans: considerations for low-dose testing. Environ. Health Perspect., 109( Suppl 1 ), 21–26. PubMed PMC

EPA, U.S. The U.S. Environmental Protection Agency ToxCast Phase I/II data http://www.epa.gov/ncct/toxcast/data.html.

Taylor T.R., et al. (2011) Ziram activates mitogen-activated protein kinases and decreases cytolytic protein levels in human natural killer cells. Toxicol. Mech. Methods, 21, 577–584. PubMed PMC

McMahon T.A., et al. (2011) The fungicide chlorothalonil is nonlinearly associated with corticosterone levels, immunity, and mortality in amphibians. Environ. Health Perspect., 119, 1098–1103. PubMed PMC

Goldman J.M., et al. (2004) Methoxychlor-induced alterations in the histological expression of angiogenic factors in pituitary and uterus. J. Mol. Histol., 35, 363–375. PubMed

Chapin R.E., et al. (1997) The effects of perinatal/juvenile methoxychlor exposure on adult rat nervous, immune, and reproductive system function. Fundam. Appl. Toxicol., 40, 138–157. PubMed

Qian Y., et al. (2010) Perfluorooctane sulfonate (PFOS) induces reactive oxygen species (ROS) production in human microvascular endothelial cells: role in endothelial permeability. J. Toxicol. Environ. Health A, 73, 819–836. PubMed PMC

Hu J.X., et al. (2013) Toxic effects of cypermethrin on the male reproductive system: with emphasis on the androgen receptor. J. Appl. Toxicol., 33, 576–585. PubMed

Jin M., et al. (2010) Estrogenic activities of two synthetic pyrethroids and their metabolites. J. Environ. Sci. (China), 22, 290–296. PubMed

Kakko I., et al. (2004) Oestradiol potentiates the effects of certain pyrethroid compounds in the MCF7 human breast carcinoma cell line. Altern. Lab. Anim., 32, 383–390. PubMed

Feng Z., et al. (2006) Acrolein is a major cigarette-related lung cancer agent: preferential binding at p53 mutational hotspots and inhibition of DNA repair. Proc. Natl Acad. Sci. USA, 103, 15404–15409. PubMed PMC

Günther M., et al. (2008) Acrolein: unwanted side product or contribution to antiangiogenic properties of metronomic cyclophosphamide therapy? J. Cell. Mol. Med., 12(6B), 2704–2716. PubMed PMC

Luo C., et al. (2013) A cigarette component acrolein induces accelerated senescence in human diploid fibroblast IMR-90 cells. Biogerontology, 14, 503–511. PubMed

Roy J., et al. (2010) Acrolein induces apoptosis through the death receptor pathway in A549 lung cells: role of p53. Can. J. Physiol. Pharmacol., 88, 353–368. PubMed

Tanel A., et al. (2014) Acrolein activates cell survival and apoptotic death responses involving the endoplasmic reticulum in A549 lung cells. Biochim. Biophys. Acta, 1843, 827–835. PubMed

Tang M.S., et al. (2011) Acrolein induced DNA damage, mutagenicity and effect on DNA repair. Mol. Nutr. Food Res., 55, 1291–1300. PubMed PMC

Cabeza-Arvelaiz Y., et al. (2012) Transcriptome analysis of a rotenone model of parkinsonism reveals complex I-tied and -untied toxicity mechanisms common to neurodegenerative diseases. PLoS One, 7, e44700. PubMed PMC

Deng Y.T., et al. (2010) Rotenone induces apoptosis in MCF-7 human breast cancer cell-mediated ROS through JNK and p38 signaling. Mol. Carcinog., 49, 141–151. PubMed

Gonçalves A.P., et al. (2011) Involvement of p53 in cell death following cell cycle arrest and mitotic catastrophe induced by rotenone. Biochim. Biophys. Acta, 1813, 492–499. PubMed PMC

Li Y., et al. (2013) Copper induces cellular senescence in human glioblastoma multiforme cells through downregulation of Bmi-1. Oncol. Rep., 29, 1805–1810. PubMed

Ostrakhovitch E.A., et al. (2005) Role of p53 and reactive oxygen species in apoptotic response to copper and zinc in epithelial breast cancer cells. Apoptosis, 10, 111–121. PubMed

Parr-Sturgess C.A., et al. (2012) Copper modulates zinc metalloproteinase-dependent ectodomain shedding of key signaling and adhesion proteins and promotes the invasion of prostate cancer epithelial cells. Mol. Cancer Res., 10, 1282–1293. PubMed

Freitas M., et al. (2013) Nickel induces apoptosis in human neutrophils. Biometals, 26, 13–21. PubMed

Wu C.H., et al. (2012) Nickel-induced epithelial-mesenchymal transition by reactive oxygen species generation and E-cadherin promoter hypermethylation. J. Biol. Chem., 287, 25292–25302. PubMed PMC

Aimola P., et al. (2012) Cadmium induces p53-dependent apoptosis in human prostate epithelial cells. PLoS One, 7, e33647. PubMed PMC

Yuan D., et al. (2013) Long-term cadmium exposure leads to the enhancement of lymphocyte proliferation via down-regulating p16 by DNA hypermethylation. Mutat. Res., 757, 125–131. PubMed

Aluigi M.G., et al. (2010) Apoptosis as a specific biomarker of diazinon toxicity in NTera2-D1 cells. Chem. Biol. Interact., 187, 299–303. PubMed

Giordano G., et al. (2007) Organophosphorus insecticides chlorpyrifos and diazinon and oxidative stress in neuronal cells in a genetic model of glutathione deficiency. Toxicol. Appl. Pharmacol., 219, 181–189. PubMed

Gilsing A.M., et al. (2013) Dietary heme iron and the risk of colorectal cancer with specific mutations in KRAS and APC. Carcinogenesis, 34, 2757–2766. PubMed

Pluth J.M., et al. (1996) Increased frequency of specific genomic deletions resulting from in vitro malathion exposure. Cancer Res., 56, 2393–2399. PubMed

Chen Z.J., et al. (2014) Bisphenol A modulates colorectal cancer protein profile and promotes the metastasis via induction of epithelial to mesenchymal transitions. Arch Toxicol. PubMed

Zhu H., et al. (2010) Environmental endocrine disruptors promote invasion and metastasis of SK-N-SH human neuroblastoma cells. Oncol. Rep., 23, 129–139. PubMed

Pontillo C.A., et al. (2013) Action of hexachlorobenzene on tumor growth and metastasis in different experimental models. Toxicol. Appl. Pharmacol., 268, 331–342. PubMed

O'Brien D.W., et al. (2004) A mechanism of airway injury in an epithelial model of mucociliary clearance. Respir. Res., 5, 10. PubMed PMC

Ornstein D.L., et al. (2007) Iron stimulates urokinase plasminogen activator expression and activates NF-kappa B in human prostate cancer cells. Nutr. Cancer, 58, 115–126. PubMed

Mao L., et al. (2012) Circadian gating of epithelial-to-mesenchymal transition in breast cancer cells via melatonin-regulation of GSK3β. Mol. Endocrinol., 26, 1808–1820. PubMed PMC

Papagerakis S., et al. (2014) The circadian clock in oral health and diseases. J. Dent. Res., 93, 27–35. PubMed PMC

Bouskine A., et al. (2009) Low doses of bisphenol A promote human seminoma cell proliferation by activating PKA and PKG via a membrane G-protein-coupled estrogen receptor. Environ. Health Perspect., 117, 1053–1058. PubMed PMC

Hernández L.G., et al. (2013) A mode-of-action approach for the identification of genotoxic carcinogens. PLoS One, 8, e64532. PubMed PMC

Wetherill Y.B., et al. (2002) The xenoestrogen bisphenol A induces inappropriate androgen receptor activation and mitogenesis in prostatic adenocarcinoma cells. Mol. Cancer Ther., 1, 515–524. PubMed

Park S.H., et al. (2009) Cell growth of ovarian cancer cells is stimulated by xenoestrogens through an estrogen-dependent pathway, but their stimulation of cell growth appears not to be involved in the activation of the mitogen-activated protein kinases ERK-1 and p38. J. Reprod. Dev., 55, 23–29. PubMed

Wilkinson C.F., et al. (1996) A mechanistic interpretation of the oncogenicity of chlorothalonil in rodents and an assessment of human relevance. Regul. Toxicol. Pharmacol., 24(1 Pt 1), 69–84. PubMed

Vesselinovitch S.D., et al. (1983) Lindane bioassay studies and human cancer risk assessment. Toxicol. Pathol., 11, 12–22. PubMed

Wang Q.L., et al. (2013) Risk assessment of mouse gastric tissue cancer induced by dichlorvos and dimethoate. Oncol. Lett., 5, 1385–1389. PubMed PMC

Lee H.R., et al. (2012) Treatment with bisphenol A and methoxychlor results in the growth of human breast cancer cells and alteration of the expression of cell cycle-related genes, cyclin D1 and p21, via an estrogen receptor-dependent signaling pathway. Int. J. Mol. Med., 29, 883–890. PubMed

Stagg N.J., et al. (2012) Assessment of possible carcinogenicity of oxyfluorfen to humans using mode of action analysis of rodent liver effects. Toxicol. Sci., 128, 334–345. PubMed

Doull J., et al. (1999) A cancer risk assessment of di(2-ethylhexyl)phthalate: application of the new U.S. EPA Risk Assessment Guidelines. Regul. Toxicol. Pharmacol., 29, 327–357. PubMed

Mazzoleni G., et al. (1994) Influence of the herbicide Linuron on growth rate and gap-junctional intercellular communication of cultured endothelial cells. J. Environ. Pathol. Toxicol. Oncol., 13, 1–10. PubMed

Yasaei H., et al. (2013) Carcinogen-specific mutational and epigenetic alterations in INK4A, INK4B and p53 tumour-suppressor genes drive induced senescence bypass in normal diploid mammalian cells. Oncogene, 32, 171–179. PubMed

Singh K.P., et al. (2008) Allelic loss and mutations in a new ETRG-1 gene are early events in diethylstilbestrol-induced renal carcinogenesis in Syrian hamsters. Gene, 408, 18–26. PubMed

Tsutsui T., et al. (1994) Reserpine-induced cell transformation without detectable genetic effects in Syrian hamster embryo cells in culture. Carcinogenesis, 15, 11–14. PubMed

Martens U., et al. (1996) Low expression of the WAF1/CIP1 gene product, p21, in enzyme-altered foci induced in rat liver by diethylnitrosamine or phenobarbital. Cancer Lett., 104, 21–26. PubMed

Geter D.R., et al. (2014) Dose-response modeling of early molecular and cellular key events in the CAR-mediated hepatocarcinogenesis pathway. Toxicol. Sci., 138, 425–445. PubMed

Bader A., et al. (2011) Paracetamol treatment increases telomerase activity in rat embryonic liver cells. Pharmacol. Rep., 63, 1435–1441. PubMed

Tsuruga Y., et al. (2008) Establishment of immortalized human hepatocytes by introduction of HPV16 E6/E7 and hTERT as cell sources for liver cell-based therapy. Cell Transplant., 17, 1083–1094. PubMed

Nguyen T.H., et al. (2005) Treatment of acetaminophen-induced acute liver failure in the mouse with conditionally immortalized human hepatocytes. J. Hepatol., 43, 1031–1037. PubMed

Bode-Böger S.M., et al. (2005) Aspirin reduces endothelial cell senescence. Biochem. Biophys. Res. Commun., 334, 1226–1232. PubMed

Heinloth A.N., et al. (2004) Gene expression profiling of rat livers reveals indicators of potential adverse effects. Toxicol. Sci., 80, 193–202. PubMed

Jacob T., et al. (2009) The effect of cotinine on telomerase activity in human vascular smooth muscle cells. J. Cardiovasc. Surg. (Torino), 50, 345–349. PubMed

Brüne B., et al. (2001) Transcription factors p53 and HIF-1alpha as targets of nitric oxide. Cell. Signal., 13, 525–533. PubMed

Davis C.D., et al. (2000) Dietary selenium and arsenic affect DNA methylation in vitro in Caco-2 cells and in vivo in rat liver and colon. J. Nutr., 130, 2903–2909. PubMed

Arnér E.S., et al. (2006) The thioredoxin system in cancer. Semin. Cancer Biol., 16, 420–426. PubMed

vom Saal F.S., et al. (2007) Chapel Hill bisphenol A expert panel consensus statement: integration of mechanisms, effects in animals and potential to impact human health at current levels of exposure. Reprod. Toxicol., 24, 131–138. PubMed PMC

Qin X.Y., et al. (2012) Effects of bisphenol A exposure on the proliferation and senescence of normal human mammary epithelial cells. Cancer Biol. Ther., 13, 296–306. PubMed

Peluso M.E., et al. (2014) Bisphenol-A exposures and behavioural aberrations: median and linear spline and meta-regression analyses of 12 toxicity studies in rodents. Toxicology, 325, 200–208. PubMed

Fang C.C., et al. (2013) Cyprodinil as an activator of aryl hydrocarbon receptor. Toxicology, 304, 32–40. PubMed

Bharadwaj R., et al. (2004) The spindle checkpoint, aneuploidy, and cancer. Oncogene, 23, 2016–2027. PubMed

Orton F., et al. (2011) Widely used pesticides with previously unknown endocrine activity revealed as in vitro antiandrogens. Environ. Health Perspect., 119, 794–800. PubMed PMC

Tanaka T., et al. (2013) Effects of maternal exposure to imazalil on behavioral development in F₁-generation mice. Birth Defects Res. B Dev. Reprod. Toxicol., 98, 334–342. PubMed

Ahmad I., et al. (2008) The involvement of nitric oxide in maneb- and paraquat-induced oxidative stress in rat polymorphonuclear leukocytes. Free Radic. Res., 42, 849–862. PubMed

US Environmental Protection Agency (1988) US Integrated Risk Information System—Maneb (CASRN 12427-38-2). http://www.epa.gov/iris/subst/0249.htm .

Miller K.P., et al. (2006) Methoxychlor metabolites may cause ovarian toxicity through estrogen-regulated pathways. Toxicol. Sci., 93, 180–188. PubMed

Du X., et al. (2014) Perinatal exposure to low-dose methoxychlor impairs testicular development in C57BL/6 mice. PLoS One, 9, e103016. PubMed PMC

Palanza P., et al. (2001) Effects of prenatal exposure to low doses of diethylstilbestrol, o,p'DDT, and methoxychlor on postnatal growth and neurobehavioral development in male and female mice. Horm. Behav., 40, 252–265. PubMed

Du G., et al. (2013) Perfluorooctane sulfonate (PFOS) affects hormone receptor activity, steroidogenesis, and expression of endocrine-related genes in vitro and in vivo . Environ. Toxicol. Chem., 32, 353–360. PubMed

Kim H.S., et al. (2011) Induction of apoptosis and CYP4A1 expression in Sprague-Dawley rats exposed to low doses of perfluorooctane sulfonate. J. Toxicol. Sci., 36, 201–210. PubMed

Eveillard A., et al. (2009) Di-(2-ethylhexyl)-phthalate (DEHP) activates the constitutive androstane receptor (CAR): a novel signalling pathway sensitive to phthalates. Biochem. Pharmacol., 77, 1735–1746. PubMed

Nakai M., et al. (1999) Binding characteristics of dialkyl phthalates for the estrogen receptor. Biochem. Biophys. Res. Commun., 254, 311–314. PubMed

Grande S.W., et al. (2006) A dose-response study following in utero and lactational exposure to di(2-ethylhexyl)phthalate: effects on female rat reproductive development. Toxicol. Sci., 91, 247–254. PubMed

Kojima H., et al. (2011) Comparative study of human and mouse pregnane X receptor agonistic activity in 200 pesticides using in vitro reporter gene assays. Toxicology, 280, 77–87. PubMed

(2006) Phosalone Reregistration Eligibility Decision (RED). The United States Environmental Protection Agency Office of Pesticide Programs.

Li X., et al. (2013) Structure-dependent activities of hydroxylated polybrominated diphenyl ethers on human estrogen receptor. Toxicology, 309, 15–22. PubMed

Berger R.G., et al. (2014) Exposure to an environmentally relevant mixture of brominated flame retardants affects fetal development in Sprague-Dawley rats. Toxicology, 320, 56–66. PubMed

Hofmeister M.V., et al. (2004) Effects of the pesticides prochloraz and methiocarb on human estrogen receptor alpha and beta mRNA levels analyzed by on-line RT-PCR. Toxicol. In Vitro, 18, 427–433. PubMed

Jacobsen P.R., et al. (2012) Persistent developmental toxicity in rat offspring after low dose exposure to a mixture of endocrine disrupting pesticides. Reprod. Toxicol., 34, 237–250. PubMed

Kamanga-Sollo E., et al. (2008) Roles of IGF-I and the estrogen, androgen and IGF-I receptors in estradiol-17beta- and trenbolone acetate-stimulated proliferation of cultured bovine satellite cells. Domest. Anim. Endocrinol., 35, 88–97. PubMed

Yarrow J.F., et al. (2010) Tissue selectivity and potential clinical applications of trenbolone (17beta-hydroxyestra-4,9,11-trien-3-one): a potent anabolic steroid with reduced androgenic and estrogenic activity. Steroids, 75, 377–389. PubMed

Yan H., et al. (2008) Exposure to bisphenol A prenatally or in adulthood promotes T(H)2 cytokine production associated with reduction of CD4CD25 regulatory T cells. Environ. Health Perspect., 116, 514–519. PubMed PMC

Erden E.S., et al. (2014) Investigation of Bisphenol A as an endocrine disruptor, total thiol, malondialdehyde, and C-reactive protein levels in chronic obstructive pulmonary disease. Eur. Rev. Med. Pharmacol. Sci., 18, 3477–3483. PubMed

Kharrazian D. (2014) The potential roles of bisphenol A (BPA) pathogenesis in autoimmunity. Autoimmune Dis., 2014, 743616. PubMed PMC

Liu Y., et al. (2014) Modulation of cytokine expression in human macrophages by endocrine-disrupting chemical Bisphenol-A. Biochem. Biophys. Res. Commun., 451, 592–598. PubMed

Rogers J.A., et al. (2013) Review: endocrine disrupting chemicals and immune responses: a focus on bisphenol-A and its potential mechanisms. Mol. Immunol., 53, 421–430. PubMed

Deutschle T., et al. (2008) A controlled challenge study on di(2-ethylhexyl) phthalate (DEHP) in house dust and the immune response in human nasal mucosa of allergic subjects. Environ. Health Perspect., 116, 1487–1493. PubMed PMC

Peltier M.R., et al. (2012) Polybrominated diphenyl ethers enhance the production of proinflammatory cytokines by the placenta. Placenta, 33, 745–749. PubMed PMC

Park H.R., et al. (2014) Involvement of reactive oxygen species in brominated diphenyl ether-47-induced inflammatory cytokine release from human extravillous trophoblasts in vitro . Toxicol. Appl. Pharmacol., 274, 283–292. PubMed PMC

Park H.R., et al. (2014) Protective effect of nuclear factor E2-related factor 2 on inflammatory cytokine response to brominated diphenyl ether-47 in the HTR-8/SVneo human first trimester extravillous trophoblast cell line. Toxicol. Appl. Pharmacol., 281, 67–77. PubMed PMC

Koike E., et al. (2014) Penta- and octa-bromodiphenyl ethers promote proinflammatory protein expression in human bronchial epithelial cells in vitro . Toxicol. In Vitro, 28, 327–333. PubMed

Zhao S., et al. (2013) Sub-acute exposure to the herbicide atrazine suppresses cell immune functions in adolescent mice. Biosci. Trends, 7, 193–201. PubMed

Rowe A.M., et al. (2006) Immunomodulatory effects of maternal atrazine exposure on male Balb/c mice. Toxicol. Appl. Pharmacol., 214, 69–77. PubMed PMC

Skinner M.K., et al. (2007) Epigenetic transgenerational actions of vinclozolin on the development of disease and cancer. Crit. Rev. Oncog., 13, 75–82. PubMed PMC

Anway M.D., et al. (2008) Transgenerational effects of the endocrine disruptor vinclozolin on the prostate transcriptome and adult onset disease. Prostate, 68, 517–529. PubMed PMC

Cowin P.A., et al. (2008) Early-onset endocrine disruptor-induced prostatitis in the rat. Environ. Health Perspect., 116, 923–929. PubMed PMC

Zhou H.R., et al. (2003) Rapid, sequential activation of mitogen-activated protein kinases and transcription factors precedes proinflammatory cytokine mRNA expression in spleens of mice exposed to the trichothecene vomitoxin. Toxicol. Sci., 72, 130–142. PubMed

Shin S.G., et al. (2005) Suppression of inducible nitric oxide synthase and cyclooxygenase-2 expression in RAW 264.7 macrophages by sesquiterpene lactones. J. Toxicol. Environ. Health A, 68, 2119–2131. PubMed

Gollamudi S., et al. (2012) Concordant signaling pathways produced by pesticide exposure in mice correspond to pathways identified in human Parkinson's disease. PLoS One, 7, e36191. PubMed PMC

Morgan J.B., et al. (2010) The marine sponge metabolite mycothiazole: a novel prototype mitochondrial complex I inhibitor. Bioorg. Med. Chem., 18, 5988–5994. PubMed PMC

(1997) BASF Corporation Pyridaben (Sanmite) Pesticide Tolerance Petition 3/97, US EPA [PF-721; FRL-5592 -7], http//pmep.cce.cornell. edu/profiles/insect-mite/mevinphos-propargite/pyridaben/ pyridab_pet_0397.html (accessed 7 May 2015)

Barros S.P., et al. (2010) Triclosan inhibition of acute and chronic inflammatory gene pathways. J. Clin. Periodontol., 37, 412–418. PubMed

Wallet M.A., et al. (2013) Triclosan alters antimicrobial and inflammatory responses of epithelial cells. Oral Dis., 19, 296–302. PubMed PMC

Bhargava H.N., et al. (1996) Triclosan: applications and safety. Am. J. Infect. Control, 24, 209–218. PubMed

Winitthana T., et al. (2014) Triclosan potentiates epithelial-to-mesenchymal transition in anoikis-resistant human lung cancer cells. PLoS One, 9, e110851. PubMed PMC

Stoker T.E., et al. (2010) Triclosan exposure modulates estrogen-dependent responses in the female wistar rat. Toxicol. Sci., 117, 45–53. PubMed

Shioda T., et al. (2006) Importance of dosage standardization for interpreting transcriptomal signature profiles: evidence from studies of xenoestrogens. Proc. Natl Acad. Sci. USA, 103, 12033–12038. PubMed PMC

Welshons W.V., et al. (1999) Low-dose bioactivity of xenoestrogens in animals: fetal exposure to low doses of methoxychlor and other xenoestrogens increases adult prostate size in mice. Toxicol. Ind. Health, 15, 12–25. PubMed

Alyea R.A., et al. (2009) Differential regulation of dopamine transporter function and location by low concentrations of environmental estrogens and 17beta-estradiol. Environ. Health Perspect., 117, 778–783. PubMed PMC

Wozniak A.L., et al. (2005) Xenoestrogens at picomolar to nanomolar concentrations trigger membrane estrogen receptor-alpha-mediated Ca2+ fluxes and prolactin release in GH3/B6 pituitary tumor cells. Environ. Health Perspect., 113, 431–439. PubMed PMC

Jeng Y.J., et al. (2011) Combinations of physiologic estrogens with xenoestrogens alter ERK phosphorylation profiles in rat pituitary cells. Environ. Health Perspect., 119, 104–112. PubMed PMC

Cabaton N.J., et al. (2011) Perinatal exposure to environmentally relevant levels of bisphenol A decreases fertility and fecundity in CD-1 mice. Environ. Health Perspect., 119, 547–552. PubMed PMC

Jones B.A., et al. (2011) Pre- and postnatal bisphenol A treatment results in persistent deficits in the sexual behavior of male rats, but not female rats, in adulthood. Horm. Behav., 59, 246–251. PubMed

Lemos M.F., et al. (2010) Protein differential expression induced by endocrine disrupting compounds in a terrestrial isopod. Chemosphere, 79, 570–576. PubMed

Markey C.M., et al. (2001) The mouse uterotrophic assay: a reevaluation of its validity in assessing the estrogenicity of bisphenol A. Environ. Health Perspect., 109, 55–60. PubMed PMC

Filipov N.M., et al. (2005) Manganese potentiates in vitro production of proinflammatory cytokines and nitric oxide by microglia through a nuclear factor kappa B–dependent mechanism. Toxicol. Sci., 84, 139–148. PubMed

Knudsen T.B., et al. (2011) Disruption of embryonic vascular development in predictive toxicology. Birth Defects Res. Part C, 93, 312–323. PubMed

Qin R., et al. (2011) Protection by tetrahydroxystilbene glucoside against neurotoxicity induced by MPP+: the involvement of PI3K/Akt pathway activation. Toxicol. Lett., 202, 1–7. PubMed

Manfo F.P., et al. (2011) Effects of maneb on testosterone release in male rats. Drug Chem. Toxicol., 34, 120–128. PubMed

Matsushita T., et al. (1976) Experimental study on contact dermatitis caused by dithiocarbamates maneb, mancozeb, zineb, and their related compounds. Int. Arch. Occup. Environ. Health, 37, 169–178. PubMed

Barlow, B. et al. (2005) Modulation of antioxidant defense systems by the environmental pesticide Maneb in dopaminergic cells. Neurotoxicol., 26, 63–75 . PubMed

Kazantseva Y.A., et al. (2013) Dichlorodiphenyltrichloroethane technical mixture regulates cell cycle and apoptosis genes through the activation of CAR and ERα in mouse livers. Toxicol. Appl. Pharmacol., 271, 137–143. PubMed

Lin Z.X., et al. (1986) Inhibition of gap junctional intercellular communication in human teratocarcinoma cells by organochlorine pesticides. Toxicol. Appl. Pharmacol., 83, 10–19. PubMed

Ruch R.J., et al. (1987) Inhibition of intercellular communication between mouse hepatocytes by tumor promoters. Toxicol. Appl. Pharmacol., 87, 111–120. PubMed

Ventura C., et al. (2012) Differential mechanisms of action are involved in chlorpyrifos effects in estrogen-dependent or -independent breast cancer cells exposed to low or high concentrations of the pesticide. Toxicol. Lett., 213, 184–193. PubMed

Mense S.M., et al. (2006) The common insecticides cyfluthrin and chlorpyrifos alter the expression of a subset of genes with diverse functions in primary human astrocytes. Toxicol. Sci., 93, 125–135. PubMed

Santucci M.A., et al. (2003) Cell-cycle deregulation in BALB/c 3T3 cells transformed by 1,2-dibromoethane and folpet pesticides. Environ. Mol. Mutagen., 41, 315–321. PubMed

Albanito L., et al. (2008) G-protein-coupled receptor 30 and estrogen receptor-alpha are involved in the proliferative effects induced by atrazine in ovarian cancer cells. Environ. Health Perspect., 116, 1648–1655. PubMed PMC

Tsuda H., et al. (2005) High susceptibility of human c-Ha-ras proto-oncogene transgenic rats to carcinogenesis: a cancer-prone animal model. Cancer Sci., 96, 309–316. PubMed PMC

Wetzel L.T., et al. (1994) Chronic effects of atrazine on estrus and mammary tumor formation in female Sprague-Dawley and Fischer 344 rats. J. Toxicol. Environ. Health, 43, 169–182. PubMed

Andersson H., et al. (2012) Proangiogenic effects of environmentally relevant levels of bisphenol A in human primary endothelial cells. Arch. Toxicol., 86, 465–474. PubMed

Dairkee S.H., et al. (2013) Bisphenol-A-induced inactivation of the p53 axis underlying deregulation of proliferation kinetics, and cell death in non-malignant human breast epithelial cells. Carcinogenesis, 34, 703–712. PubMed PMC

Betancourt A.M., et al. (2012) Altered carcinogenesis and proteome in mammary glands of rats after prepubertal exposures to the hormonally active chemicals bisphenol A and genistein. J. Nutr, 142, 1382S–1388S. PubMed PMC

Andrysík Z., et al. (2013) Aryl hydrocarbon receptor-mediated disruption of contact inhibition is associated with connexin43 downregulation and inhibition of gap junctional intercellular communication. Arch. Toxicol., 87, 491–503. PubMed

Haber L.T., et al. (2000) Hazard identification and dose response of inhaled nickel-soluble salts. Regul. Toxicol. Pharmacol., 31, 210–230. PubMed

LN V., et al. (2013) Low dose effects of bisphenol A: an integrated review of in vitro, laboratory animal and human studies. Endocrine Disruptors, 1, e1.1–e1.20.

Tryphonas H., et al. (2004) Oral (gavage), in utero and post-natal exposure of Sprague-Dawley rats to low doses of tributyltin chloride. Part II: effects on the immune system. Food Chem. Toxicol., 42, 221–235. PubMed

Watanabe J., et al. (2013) Low dose of methylmercury (MeHg) exposure induces caspase mediated-apoptosis in cultured neural progenitor cells. J. Toxicol. Sci., 38, 931–935. PubMed

Petroni D., et al. (2012) Low-dose methylmercury-induced oxidative stress, cytotoxicity, and tau-hyperphosphorylation in human neuroblastoma (SH-SY5Y) cells. Environ. Toxicol., 27, 549–555. PubMed

McCormack A.L., et al. (2005) Role of oxidative stress in paraquat-induced dopaminergic cell degeneration. J. Neurochem., 93, 1030–1037. PubMed

Hartwig A., et al. (2002) Interference by toxic metal ions with DNA repair processes and cell cycle control: molecular mechanisms. Environ. Health Perspect., 110( Suppl 5 ), 797–799. PubMed PMC

Asmuss M., et al. (2000) Differential effects of toxic metal compounds on the activities of Fpg and XPA, two zinc finger proteins involved in DNA repair. Carcinogenesis, 21, 2097–2104. PubMed

McNeill D.R., et al. (2004) Inhibition of Ape1 nuclease activity by lead, iron, and cadmium. Environ. Health Perspect., 112, 799–804. PubMed PMC

Pottier G., et al. (2013) Lead exposure induces telomere instability in human cells. PLoS One, 8, e67501. PubMed PMC

Zhang X., et al. (2013) Environmental and occupational exposure to chemicals and telomere length in human studies. Occup. Environ. Med., 70, 743–749. PubMed

Exon J.H. (2006) A review of the toxicology of acrylamide. J. Toxicol. Environ. Health B. Crit. Rev., 9, 397–412. PubMed

Sickles D.W., et al. (2007) Acrylamide effects on kinesin-related proteins of the mitotic/meiotic spindle. Toxicol. Appl. Pharmacol., 222, 111–121. PubMed

Wang X., et al. (2013) Epigenotoxicity of environmental pollutants evaluated by a combination of DNA methylation inhibition and capillary electrophoresis-laser-induced fluorescence immunoassay. Anal. Bioanal. Chem., 405, 2435–2442. PubMed

Arita A., et al. (2012) Global levels of histone modifications in peripheral blood mononuclear cells of subjects with exposure to nickel. Environ. Health Perspect., 120, 198–203. PubMed PMC

Cantone L., et al. (2011) Inhalable metal-rich air particles and histone H3K4 dimethylation and H3K9 acetylation in a cross-sectional study of steel workers. Environ. Health Perspect., 119, 964–969. PubMed PMC

Chervona Y., et al. (2012) Carcinogenic metals and the epigenome: understanding the effect of nickel, arsenic, and chromium. Metallomics, 4, 619–627. PubMed PMC

Avissar-Whiting M., et al. (2010) Bisphenol A exposure leads to specific microRNA alterations in placental cells. Reprod. Toxicol., 29, 401–406. PubMed PMC

Roedel E.Q., et al. (2012) Pulmonary toxicity after exposure to military-relevant heavy metal tungsten alloy particles. Toxicol. Appl. Pharmacol., 259, 74–86. PubMed

Freyre-Fonseca V., et al. (2011) Titanium dioxide nanoparticles impair lung mitochondrial function. Toxicol. Lett., 202, 111–119. PubMed

Elhajouji A., et al. (2011) Potential thresholds for genotoxic effects by micronucleus scoring. Mutagenesis, 26, 199–204. PubMed

Ermler S., et al. (2013) Seven benzimidazole pesticides combined at sub-threshold levels induce micronuclei in vitro . Mutagenesis, 28, 417–426. PubMed PMC

Sargent L.M., et al. (2012) Single-walled carbon nanotube-induced mitotic disruption. Mutat. Res., 745, 28–37. PubMed PMC

Muller J., et al. (2008) Clastogenic and aneugenic effects of multi-wall carbon nanotubes in epithelial cells. Carcinogenesis, 29, 427–433. PubMed

Thomas D. (2010) Gene–environment-wide association studies: emerging approaches. Nat. Rev. Genet., 11, 259–272. PubMed PMC

Santella R.M., et al. (2005) DNA adducts, DNA repair genotype/phenotype and cancer risk. Mutat. Res., 592, 29–35. PubMed

(2011) Cytogenetic Dosimetry: Applications in Preparedness for and Response to Radiation Emergencies. International Atomic Energy Agency, Vienna.

De Lange T. (2005) Telomere-related genome instability in cancer. Cold Spring Harb. Symp. Quant. Biol., 70, 197–204. PubMed

Frias C., et al. (2012) Telomere dysfunction and genome instability. Front. Biosci. (Landmark Ed), 17, 2181–2196. PubMed

Hollstein M., et al. (1991) p53 mutations in human cancers. Science, 253, 49–53. PubMed

Jang J.W., et al. (2006) Isoform-specific ras activation and oncogene dependence during MYC- and Wnt-induced mammary tumorigenesis. Mol. Cell. Biol., 26, 8109–8121. PubMed PMC

Muñoz D.M., et al. (2013) Loss of p53 cooperates with K-ras activation to induce glioma formation in a region-independent manner. Glia, 61, 1862–1872. PubMed

Pierotti, M.A. et al. Mechanisms of oncogene activation. Kufe DW, Pollock RE, Weichselbaum RR, and et al. Holland-Frei Cancer Medicine. 6th. 2003. Hamilton (ON), BC Decker .

Mazzei F., et al. (2013) Role of MUTYH in human cancer. Mutat. Res., 743–744, 33–43. PubMed

Sancar A. (1995) Excision repair in mammalian cells. J. Biol. Chem., 270, 15915–15918. PubMed

Vineis P., et al. (2009) A field synopsis on low-penetrance variants in DNA repair genes and cancer susceptibility. J. Natl Cancer Inst., 101, 24–36. PubMed

Bohacek J., et al. (2013) Epigenetic inheritance of disease and disease risk. Neuropsychopharmacology, 38, 220–236. PubMed PMC

Esteller M. (2007) Cancer epigenomics: DNA methylomes and histone-modification maps. Nat. Rev. Genet., 8, 286–298. PubMed

Caffarelli E., et al. (2011) Epigenetic regulation in cancer development. Front. Biosci. (Landmark Ed), 16, 2682–2694. PubMed

Croce C.M. (2009) Causes and consequences of microRNA dysregulation in cancer. Nat. Rev. Genet., 10, 704–714. PubMed PMC

Wang Y., et al. (2013) MicroRNAs and DNA damage response: implications for cancer therapy. Cell Cycle, 12, 32–42. PubMed PMC

Devalle S., et al. (2012) Implications of aneuploidy for stem cell biology and brain therapeutics. Front. Cell. Neurosci., 6, 36. PubMed PMC

Linschooten J.O., et al. (2013) Paternal lifestyle as a potential source of germline mutations transmitted to offspring. FASEB J., 27, 2873–2879. PubMed PMC

Leyns L., et al. (2012) Genomic integrity of mouse embryonic stem cells. In Embryogenesis. Intech. pp. 333–358.

Blessing H., et al. (2004) Interaction of selenium compounds with zinc finger proteins involved in DNA repair. Eur. J. Biochem., 271, 3190–3199. PubMed

Zhang X., et al. (2013) Environmental and occupational exposure to chemicals and telomere length in human studies. Postgrad. Med. J., 89, 722–728. PubMed

Lombaert N., et al. (2013) Hard-metal (WC-Co) particles trigger a signaling cascade involving p38 MAPK, HIF-1α, HMOX1, and p53 activation in human PBMC. Arch. Toxicol., 87, 259–268. PubMed

Jugan M.L., et al. (2012) Titanium dioxide nanoparticles exhibit genotoxicity and impair DNA repair activity in A549 cells. Nanotoxicology, 6, 501–513. PubMed

Song M.F., et al. (2012) Metal nanoparticle-induced micronuclei and oxidative DNA damage in mice. J. Clin. Biochem. Nutr., 50, 211–216. PubMed PMC

Doshi T., et al. (2011) Hypermethylation of estrogen receptor promoter region in adult testis of rats exposed neonatally to bisphenol A. Toxicology, 289, 74–82. PubMed

Kundakovic M., et al. (2013) Sex-specific epigenetic disruption and behavioral changes following low-dose in utero bisphenol A exposure. Proc. Natl Acad. Sci. USA, 110, 9956–9961. PubMed PMC

Pupo M., et al. (2012) Bisphenol A induces gene expression changes and proliferative effects through GPER in breast cancer cells and cancer-associated fibroblasts. Environ. Health Perspect., 120, 1177–1182. PubMed PMC

Ribeiro-Varandas E., et al. (2013) Bisphenol A at concentrations found in human serum induces aneugenic effects in endothelial cells. Mutat. Res., 751, 27–33. PubMed

Marshall H. (2002) Fact sheet: carbendazim. Pesticides News, 57, 20–21.

Zhao Y., et al. (2010) Characterization and determination of chloro- and bromo-benzoquinones as new chlorination disinfection byproducts in drinking water. Anal. Chem., 82, 4599–4605. PubMed

Piao M.J., et al. (2011) Silver nanoparticles induce oxidative cell damage in human liver cells through inhibition of reduced glutathione and induction of mitochondria-involved apoptosis. Toxicol. Lett., 201, 92–100. PubMed

Gong C., et al. (2012) Methylation of PARP-1 promoter involved in the regulation of nano-SiO2-induced decrease of PARP-1 mRNA expression. Toxicol. Lett., 209, 264–269. PubMed

Choi A.O., et al. (2008) Quantum dot-induced epigenetic and genotoxic changes in human breast cancer cells. J. Mol. Med. (Berl), 86, 291–302. PubMed

Balansky R., et al. (2013) Transplacental clastogenic and epigenetic effects of gold nanoparticles in mice. Mutat. Res., 751–752, 42–48. PubMed

Liu Y., et al. (2013) Understanding the toxicity of carbon nanotubes. Acc. Chem. Res., 46, 702–713. PubMed

Chisholm H. (1910–1911) 11th Edition of Encyclopedia Britannica. Cambridge University Press, Cambridge, UK.

Stoker T.E., et al. (1999) Prepubertal exposure to compounds that increase prolactin secretion in the male rat: effects on the adult prostate. Biol. Reprod., 61, 1636–1643. PubMed

Ho S.M., et al. (2006) Developmental exposure to estradiol and bisphenol A increases susceptibility to prostate carcinogenesis and epigenetically regulates phosphodiesterase type 4 variant 4. Cancer Res., 66, 5624–5632. PubMed PMC

Riu A., et al. (2011) Characterization of novel ligands of ERalpha, Erbeta, and PPARgamma: the case of halogenated bisphenol A and their conjugated metabolites. Toxicol. Sci, 122, 372–82. PubMed

Riu A., et al. (2011) Peroxisome proliferator-activated receptor γ is a target for halogenated analogs of bisphenol A. Environ. Health Perspect., 119, 1227–1232. PubMed PMC

Thueson L.E., et al. (2015) In vitro exposure to the herbicide atrazine inhibits T cell activation, proliferation, and cytokine production and significantly increases the frequency of Foxp3+ regulatory T cells. Toxicol. Sci., 143, 418–429. PubMed PMC

Hooghe R.J., et al. (2000) Effects of selected herbicides on cytokine production in vitro . Life Sci., 66, 2519–2525. PubMed

Filipov N.M., et al. (2005) Immunotoxic effects of short-term atrazine exposure in young male C57BL/6 mice. Toxicol. Sci., 86, 324–332. PubMed

Karrow N.A., et al. (2005) Oral exposure to atrazine modulates cell-mediated immune function and decreases host resistance to the B16F10 tumor model in female B6C3F1 mice. Toxicology, 209, 15–28. PubMed

Basini G., et al. (2012) Atrazine disrupts steroidogenesis, VEGF and NO production in swine granulosa cells. Ecotoxicol. Environ. Saf., 85, 59–63. PubMed

Chen J.Y., et al. (2013) Immunotoxicity of atrazine in Balb/c mice. J. Environ. Sci. Health B., 48, 637–645. PubMed

Chen J., et al. (2015) Effects of atrazine on the proliferation and cytotoxicity of murine lymphocytes with the use of carboxyfluorescein succinimidyl ester-based flow cytometric approaches. Food Chem. Toxicol., 76, 61–69. PubMed

Danelli L., et al. (2015) Mast cells boost myeloid-derived suppressor cell activity and contribute to the development of tumor-favoring microenvironment. Cancer Immunol. Res., 3, 85–95. PubMed

Grimm E.A., et al. (2013) Molecular pathways: inflammation-associated nitric-oxide production as a cancer-supporting redox mechanism and a potential therapeutic target. Clin. Cancer Res., 19, 5557–5563. PubMed PMC

Costa A., et al. (2014) The role of reactive oxygen species and metabolism on cancer cells and their microenvironment. Semin. Cancer Biol., 25, 23–32. PubMed

Lei Y., et al. (2015) Redox regulation of inflammation: old elements, a new story. Med. Res. Rev., 35, 306–340. PubMed

Wu Y., et al. (2014) Molecular mechanisms underlying chronic inflammation-associated cancers. Cancer Lett., 345, 164–173. PubMed PMC

Zhang H.Y., et al. (2014) Perinatal exposure to 4-nonylphenol affects adipogenesis in first and second generation rats offspring. Toxicol. Lett., 225, 325–332. PubMed

Mitchison J. (1971) The Biology of the Cell Cycle. Cambridge University Press.

Keating M.T., et al. (1988) Autocrine stimulation of intracellular PDGF receptors in v-sis-transformed cells. Science, 239, 914–916. PubMed

Skobe M., et al. (1998) Tumorigenic conversion of immortal human keratinocytes through stromal cell activation. Proc. Natl Acad. Sci. USA, 95, 1050–1055. PubMed PMC

Lemmon M.A. (2009) Ligand-induced ErbB receptor dimerization. Exp. Cell Res., 315, 638–648. PubMed PMC

Kerkhoff E., et al. (1998) Cell cycle targets of Ras/Raf signalling. Oncogene, 17, 1457–1462. PubMed

Mezquita B., et al. (2014) Unlocking doors without keys: activation of Src by truncated C-terminal intracellular receptor tyrosine kinases lacking tyrosine kinase activity. Cells, 3, 92–111. PubMed PMC

Grünfeld H.T., et al. (2004) Effect of in vitro estrogenic pesticides on human oestrogen receptor alpha and beta mRNA levels. Toxicol. Lett., 151, 467–480. PubMed

Symonds D.A., et al. (2005) Methoxychlor induces proliferation of the mouse ovarian surface epithelium. Toxicol. Sci., 83, 355–362. PubMed

Murono E.P., et al. (2004) The effects of the reported active metabolite of methoxychlor, 2,2-bis(p-hydroxyphenyl)-1,1,1-trichloroethane, on testosterone formation by cultured Leydig cells from young adult rats. Reprod. Toxicol., 19, 135–146. PubMed

Gaido K.W., et al. (2000) Interaction of methoxychlor and related compounds with estrogen receptor alpha and beta, and androgen receptor: structure-activity studies. Mol. Pharmacol., 58, 852–858. PubMed

Paulose T., et al. (2011) Increased sensitivity of estrogen receptor alpha overexpressing antral follicles to methoxychlor and its metabolites. Toxicol. Sci., 120, 447–459. PubMed PMC

Wilard S, et al. (2009) Growth factors differentially augment the effects of HPTE on estrogen response element-mediated gene transcription in a dose- and time-dependent manner among human breast cancer cell lines. Res. J. Med. Med. Sci. 4, 171–180.

Kojima H., et al. (2010) Endocrine-disrupting potential of pesticides via nuclear receptors and aryl hydrocarbon receptor. J. Health Sci., 56, 374–386.

Noriega N.C., et al. (2005) Late gestational exposure to the fungicide prochloraz delays the onset of parturition and causes reproductive malformations in male but not female rat offspring. Biol. Reprod., 72, 1324–1335. PubMed

Cocco P. (2002) On the rumors about the silent spring. Review of the scientific evidence linking occupational and environmental pesticide exposure to endocrine disruption health effects. Cad. Saude. Publica., 18, 379–402. PubMed

Kleinstreuer N.C., et al. (2011) Identifying developmental toxicity pathways for a subset of ToxCast chemicals using human embryonic stem cells and metabolomics. Toxicol. Appl. Pharmacol., 257, 111–121. PubMed

Cummings A.M., et al. (1989) Antifertility effect of methoxychlor in female rats: dose- and time-dependent blockade of pregnancy. Toxicol. Appl. Pharmacol., 97, 454–462. PubMed

Gray L.E., Jr, et al. (1989) A dose-response analysis of methoxychlor-induced alterations of reproductive development and function in the rat. Fundam. Appl. Toxicol., 12, 92–108. PubMed

Metcalf J.L., et al. (1996) Methoxychlor mimics the action of 17 beta-estradiol on induction of uterine epidermal growth factor receptors in immature female rats. Reprod. Toxicol., 10, 393–399. PubMed

Kuiper G.G., et al. (1998) Interaction of estrogenic chemicals and phytoestrogens with estrogen receptor beta. Endocrinology, 139, 4252–4263. PubMed

Andersen H.R., et al. (2002) Effects of currently used pesticides in assays for estrogenicity, androgenicity, and aromatase activity in vitro . Toxicol. Appl. Pharmacol., 179, 1–12. PubMed

Vinggaard A.M., et al. (2006) Prochloraz: an imidazole fungicide with multiple mechanisms of action. Int. J. Androl., 29, 186–192. PubMed

Liu C., et al. (2011) Effects of prochloraz or propylthiouracil on the cross-talk between the HPG, HPA, and HPT axes in zebrafish. Environ. Sci. Technol., 45, 769–775. PubMed

Zhang W., et al. (2013) Known and emerging factors modulating estrogenic effects of estrogen-disrupting chemicals. Environ. Rev., 21, 1–12.

Medjakovic S., et al. (2014) Effect of nonpersistent pesticides on estrogen receptor, androgen receptor, and aryl hydrocarbon receptor. Environ. Toxicol., 29, 1201–1216. PubMed PMC

Jenkins S., et al. (2009) Oral exposure to bisphenol a increases dimethylbenzanthracene-induced mammary cancer in rats. Environ. Health Perspect., 117, 910–915. PubMed PMC

Goodson W.H., III, et al. (2011) Activation of the mTOR pathway by low levels of xenoestrogens in breast epithelial cells from high-risk women. Carcinogenesis, 32, 1724–1733. PubMed PMC

Meironyté D., et al. (1999) Analysis of polybrominated diphenyl ethers in Swedish human milk. A time-related trend study, 1972–1997. J. Toxicol. Environ. Health A, 58, 329–341. PubMed

Brown D.J., et al. (2004) Analysis of Ah receptor pathway activation by brominated flame retardants. Chemosphere, 55, 1509–1518. PubMed

Hsieh T.H., et al. (2012) Phthalates induce proliferation and invasiveness of estrogen receptor-negative breast cancer through the AhR/HDAC6/c-Myc signaling pathway. FASEB J., 26, 778–787. PubMed

Janjua N.R., et al. (2007) Systemic uptake of diethyl phthalate, dibutyl phthalate, and butyl paraben following whole-body topical application and reproductive and thyroid hormone levels in humans. Environ. Sci. Technol., 41, 5564–5570. PubMed

Liu W.L., et al. (2010) [Distribution characteristics of phthalic acid esters in soils and plants at e-waste recycling sites in Taizhou of Zhejiang, China]. Ying Yong Sheng Tai Xue Bao, 21, 489–494. PubMed

Wormuth M., et al. (2006) What are the sources of exposure to eight frequently used phthalic acid esters in Europeans? Risk Anal., 26, 803–824. PubMed

Galbraith H. (2002) Hormones in international meat production: biological, sociological and consumer issues. Nutr. Res. Rev., 15, 293–314. PubMed

Boettcher M., et al. (2011) Low-dose effects and biphasic effect profiles: is trenbolone a genotoxicant? Mutat. Res., 723, 152–157. PubMed

Kongsuwan K., et al. (2012) The effect of combination treatment with trenbolone acetate and estradiol-17β on skeletal muscle expression and plasma concentrations of oxytocin in sheep. Domest. Anim. Endocrinol., 43, 67–73. PubMed

Hotchkiss A.K., et al. (2007) An environmental androgen, 17beta-trenbolone, affects delayed-type hypersensitivity and reproductive tissues in male mice. J. Toxicol. Environ. Health A, 70, 138–140. PubMed

Zhao J.X., et al. (2011) Trenbolone enhances myogenic differentiation by enhancing β-catenin signaling in muscle-derived stem cells of cattle. Domest. Anim. Endocrinol., 40, 222–229. PubMed PMC

Ansari K.M., et al. (2010) Skin tumor promotion by argemone oil/alkaloid in mice: evidence for enhanced cell proliferation, ornithine decarboxylase, cyclooxygenase-2 and activation of MAPK/NF-kappaB pathway. Food Chem. Toxicol., 48, 132–138. PubMed

Mishra V., et al. (2012) Role of ErbB2 mediated AKT and MAPK pathway in gall bladder cell proliferation induced by argemone oil and butter yellow. Argemone oil and butter yellow induced gall bladder cell proliferation. Cell Biol. Toxicol., 28, 149–159. PubMed

Parker M. (1991) Nuclear Hormone Receptors: Molecular Mechanisms, Cellular Functions, Clinical Abnormalities. Academic Press, London.

Gulliver L.S.M. (2013) Estradiol synthesis and metabolism and risk of ovarian cancer in older women taking prescribed or plant-derived estrogen supplementation. J. Steroids Horm. Sci., S12:003.

Leroy B., et al. (2014) TP53 mutations in human cancer: database reassessment and prospects for the next decade. Hum. Mutat., 35, 672–688. PubMed

De Blasio A., et al. (2005) Differentiative pathway activated by 3-aminobenzamide, an inhibitor of PARP, in human osteosarcoma MG-63 cells. FEBS Lett., 579, 615–620. PubMed

Nielsen G.P., et al. (1998) CDKN2A gene deletions and loss of p16 expression occur in osteosarcomas that lack RB alterations. Am. J. Pathol., 153, 159–163. PubMed PMC

Pietruszewska W., et al. (2008) Loss of heterozygosity for Rb locus and pRb immunostaining in laryngeal cancer: a clinicopathologic, molecular and immunohistochemical study. Folia Histochem. Cytobiol., 46, 479–485. PubMed

Myong N.H. (2008) Cyclin D1 overexpression, p16 loss, and pRb inactivation play a key role in pulmonary carcinogenesis and have a prognostic implication for the long-term survival in non-small cell lung carcinoma patients. Cancer Res. Treat., 40, 45–52. PubMed PMC

Ikushima H., et al. (2010) TGFbeta signalling: a complex web in cancer progression. Nat. Rev. Cancer, 10, 415–424. PubMed

Su V., et al. (2014) Connexins: mechanisms regulating protein levels and intercellular communication. FEBS Lett., 588, 1212–1220. PubMed PMC

Li M.W., et al. (2010) Connexin 43 is critical to maintain the homeostasis of the blood-testis barrier via its effects on tight junction reassembly. Proc. Natl Acad. Sci. USA, 107, 17998–18003. PubMed PMC

Campos-Pereira F.D., et al. (2012) Early cytotoxic and genotoxic effects of atrazine on Wistar rat liver: a morphological, immunohistochemical, biochemical, and molecular study. Ecotoxicol. Environ. Saf., 78, 170–177. PubMed

Stenner-Liewen F., et al. (2003) Apoptosis and cancer: basic mechanisms and therapeutic opportunities in the postgenomic era. Cancer Res., 63, 263–268.

Thompson C.B. (1995) Apoptosis in the pathogenesis and treatment of disease. Science, 267, 1456–1462. PubMed

Shortt J., et al. (2012) Oncogenes in cell survival and cell death. Cold Spring Harb. Perspect. Biol., 4, a009829. PubMed PMC

Alberts B., et al. (2002) Extracellular control of cell division, cell growth, and apoptosis. In Molecular Biology of the Cell. Garland Science, New York, NY.

Roos W.P., et al. (2006) DNA damage-induced cell death by apoptosis. Trends Mol. Med., 12, 440–450. PubMed

Fridman J.S., et al. (2003) Control of apoptosis by p53. Oncogene, 22, 9030–9040. PubMed

Adams J.M. (2003) Ways of dying: multiple pathways to apoptosis. Genes Dev., 17, 2481–2495. PubMed

Deveraux Q.L., et al. (1999) IAP family proteins–suppressors of apoptosis. Genes Dev., 13, 239–252. PubMed

Yang Y.L., et al. (2000) The IAP family: endogenous caspase inhibitors with multiple biological activities. Cell Res., 10, 169–177. PubMed

Wu W., et al. (2013) Metabolic changes in cancer: beyond the Warburg effect. Acta Biochim. Biophys. Sin. (Shanghai), 45, 18–26. PubMed

Gonzalez M.J., et al. (2012) The bio-energetic theory of carcinogenesis. Med. Hypotheses, 79, 433–439. PubMed

Ferreira L.M., et al. (2012) Metabolic reprogramming of the tumor. Oncogene, 31, 3999–4011. PubMed

Yi C.H., et al. (2011) Metabolic regulation of protein N-alpha-acetylation by Bcl-xL promotes cell survival. Cell, 146, 607–620. PubMed PMC

Wu G.S. (2009) TRAIL as a target in anti-cancer therapy. Cancer Lett., 285, 1–5. PubMed

Klaunig J.E., et al. (1990) Gap-junctional intercellular communication and murine hepatic carcinogenesis. Prog. Clin. Biol. Res., 331, 277–291. PubMed

Carette D., et al. (2014) Connexin a check-point component of cell apoptosis in normal and physiopathological conditions. Biochimie, 101, 1–9. PubMed

Leung-Toung R., et al. (2006) Thiol proteases: inhibitors and potential therapeutic targets. Curr. Med. Chem., 13, 547–581. PubMed

Kim I.Y., et al. (2004) Phthalates inhibit tamoxifen-induced apoptosis in MCF-7 human breast cancer cells. J. Toxicol. Environ. Health A, 67, 2025–2035. PubMed

Corcelle E., et al. (2006) Disruption of autophagy at the maturation step by the carcinogen lindane is associated with the sustained mitogen-activated protein kinase/extracellular signal-regulated kinase activity. Cancer Res., 66, 6861–6870. PubMed

Kim J.Y., et al. (2014) Methoxychlor and triclosan stimulates ovarian cancer growth by regulating cell cycle- and apoptosis-related genes via an estrogen receptor-dependent pathway. Environ. Toxicol. Pharmacol., 37, 1264–1274. PubMed

Carnero A. (2013) Markers of cellular senescence. Methods Mol. Biol., 965, 63–81. PubMed

Serrano M., et al. (2001) Putting the stress on senescence. Curr. Opin. Cell Biol., 13, 748–753. PubMed

Shay J.W., et al. (2004) Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene, 23, 2919–2933. PubMed

Ohtani N., et al. (2004) The p16INK4a-RB pathway: molecular link between cellular senescence and tumor suppression. J. Med. Invest., 51, 146–153. PubMed

Sherr C.J., et al. (2002) The RB and p53 pathways in cancer. Cancer Cell, 2, 103–112. PubMed

Vergel M., et al. (2010) Bypassing cellular senescence by genetic screening tools. Clin. Transl. Oncol., 12, 410–417. PubMed

Zanella F., et al. (2010) Understanding FOXO, new views on old transcription factors. Curr. Cancer Drug Targets, 10, 135–146. PubMed

Ruiz L., et al. (2008) Characterization of the p53 response to oncogene-induced senescence. PLoS One, 3, e3230. PubMed PMC

Newbold R.F., et al. (1982) Induction of immortality is an early event in malignant transformation of mammalian cells by carcinogens. Nature, 299, 633–635. PubMed

Russo I., et al. (1998) A telomere-independent senescence mechanism is the sole barrier to Syrian hamster cell immortalization. Oncogene, 17, 3417–3426. PubMed

Newbold R.F., et al. (1980) Mutagenicity of carcinogenic methylating agents is associated with a specific DNA modification. Nature, 283, 596–599. PubMed

Lehman T.A., et al. (1993) p53 mutations in human immortalized epithelial cell lines. Carcinogenesis, 14, 833–839. PubMed

Lafarge-Frayssinet C., et al. (1989) Over expression of proto-oncogenes: ki-ras, fos and myc in rat liver cells treated in vitro by two liver tumor promoters: phenobarbital and biliverdin. Cancer Lett., 44, 191–198. PubMed

Arita A., et al. (2009) Epigenetics in metal carcinogenesis: nickel, arsenic, chromium and cadmium. Metallomics, 1, 222–228. PubMed PMC

Trott D.A., et al. (1995) Mechanisms involved in the immortalization of mammalian cells by ionizing radiation and chemical carcinogens. Carcinogenesis, 16, 193–204. PubMed

Rivedal E., et al. (2000) Morphological transformation and effect on gap junction intercellular communication in Syrian hamster embryo cells as screening tests for carcinogens devoid of mutagenic activity. Toxicol. In Vitro, 14, 185–192. PubMed

Zhou X., et al. (2009) Effects of nickel, chromate, and arsenite on histone 3 lysine methylation. Toxicol. Appl. Pharmacol., 236, 78–84. PubMed PMC

Creton S., et al. (2010) Acute toxicity testing of chemicals-Opportunities to avoid redundant testing and use alternative approaches. Crit. Rev. Toxicol., 40, 50–83. PubMed

Dickens F., et al. (1933) The metabolism of normal and tumour tissue: The effects of lactate, pyruvate and deprivation of substrate. Biochem. J., 27, 1134–1140. PubMed PMC

MEDES G., et al. (1953) Metabolism of neoplastic tissue. IV. A study of lipid synthesis in neoplastic tissue slices in vitro . Cancer Res., 13, 27–29. PubMed

Menendez J.A., et al. (2007) Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat. Rev. Cancer, 7, 763–777. PubMed

Deberardinis R.J., et al. (2008) Brick by brick: metabolism and tumor cell growth. Curr. Opin. Genet. Dev., 18, 54–61. PubMed PMC

Vander Heiden M.G., et al. (2011) Metabolic pathway alterations that support cell proliferation. Cold Spring Harb. Symp. Quant. Biol., 76, 325–334. PubMed

Currie E., et al. (2013) Cellular fatty acid metabolism and cancer. Cell Metab., 18, 153–161. PubMed PMC

Kamphorst J.J., et al. (2013) Hypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids. Proc. Natl Acad. Sci. USA, 110, 8882–8887. PubMed PMC

Lazebnik Y. (2010) What are the hallmarks of cancer? Nat. Rev. Cancer, 10, 232–233. PubMed

Berridge M.V., et al. (2010) Metabolic flexibility and cell hierarchy in metastatic cancer. Mitochondrion, 10, 584–588. PubMed

Floor S.L., et al. (2012) Hallmarks of cancer: of all cancer cells, all the time? Trends Mol. Med., 18, 509–515. PubMed

Newsholme E.A., et al. (1973) Regulation in Metabolism. Wiley, London, New York.

Shim H., et al. (1997) c-Myc transactivation of LDH-A: implications for tumor metabolism and growth. Proc. Natl Acad. Sci. USA, 94, 6658–6663. PubMed PMC

Fantin V.R., et al. (2006) Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell, 9, 425–434. PubMed

Stanton R.C. (2012) Glucose-6-phosphate dehydrogenase, NADPH, and cell survival. IUBMB Life, 64, 362–369. PubMed PMC

Raimundo N., et al. (2011) Revisiting the TCA cycle: signaling to tumor formation. Trends Mol. Med., 17, 641–649. PubMed PMC

Mullen A.R., et al. (2012) Genetically-defined metabolic reprogramming in cancer. Trends Endocrinol. Metab., 23, 552–559. PubMed PMC

Wallace D.C. (2005) A mitochondrial paradigm of metabolic and degenerative diseases, aging, and cancer: a dawn for evolutionary medicine. Annu. Rev. Genet., 39, 359–407. PubMed PMC

Santidrian A.F., et al. (2013) Mitochondrial complex I activity and NAD+/NADH balance regulate breast cancer progression. J. Clin. Invest., 123, 1068–1081. PubMed PMC

Lapuente-Brun E., et al. (2013) Supercomplex assembly determines electron flux in the mitochondrial electron transport chain. Science, 340, 1567–1570. PubMed

Zaidi N., et al. (2013) Lipogenesis and lipolysis: the pathways exploited by the cancer cells to acquire fatty acids. Prog. Lipid Res, 52, 585–589. PubMed PMC

Weidberg H., et al. (2009) Lipophagy: selective catabolism designed for lipids. Dev. Cell, 16, 628–630. PubMed

Liu K., et al. (2013) Regulation of lipid stores and metabolism by lipophagy. Cell Death Differ., 20, 3–11. PubMed PMC

Newsholme E.A., et al. (1991) Application of metabolic-control logic to fuel utilization and its significance in tumor cells. Adv. Enzyme Regul., 31, 225–246. PubMed

Kalhan S.C., et al. (2012) Resurgence of serine: an often neglected but indispensable amino Acid. J. Biol. Chem., 287, 19786–19791. PubMed PMC

Locasale J.W. (2013) Serine, glycine and one-carbon units: cancer metabolism in full circle. Nat. Rev. Cancer, 13, 572–583. PubMed PMC

Cho H.M., et al. (2000) Nucleotide sequence and differential expression of the human 3-phosphoglycerate dehydrogenase gene. Gene, 245, 193–201. PubMed

Owen O.E., et al. (2002) The key role of anaplerosis and cataplerosis for citric acid cycle function. J. Biol. Chem., 277, 30409–30412. PubMed

Meyerhof O. (1951) Mechanisms of glycolysis and fermentation. Can. J. Med. Sci., 29, 63–77. PubMed

Lowry O.H., et al. (1964) The relationships between substrates and enzymes of glycolysis in brain. J. Biol. Chem., 239, 31–42. PubMed

Warburg O., et al. (1958) [Partial anaerobiosis and radiation-sensitivity of cancer cells]. Arch. Biochem. Biophys., 78, 573–586. PubMed

Robey R.B. (2011) On dogma and the metabolic gestalt of tumor cells, Science, E-Letters. https://www.sciencemag.org/content/330/ 6009/1338/reply (7 May 2015, date last accessed).

Copley S.D. (2003) Enzymes with extra talents: moonlighting functions and catalytic promiscuity. Curr. Opin. Chem. Biol., 7, 265–272. PubMed

Luo W., et al. (2012) Emerging roles of PKM2 in cell metabolism and cancer progression. Trends Endocrinol. Metab., 23, 560–566. PubMed PMC

Gao X., et al. (2012) Pyruvate kinase M2 regulates gene transcription by acting as a protein kinase. Mol. Cell, 45, 598–609. PubMed PMC

Robey R.B., et al. (2006) Mitochondrial hexokinases, novel mediators of the antiapoptotic effects of growth factors and Akt. Oncogene, 25, 4683–4696. PubMed

Hu J., et al. (2013) Heterogeneity of tumor-induced gene expression changes in the human metabolic network. Nat. Biotechnol., 31, 522–529. PubMed PMC

Kacser H., et al. (1973) The control of flux. Symp. Soc. Exp. Biol., 27, 65–104. PubMed

Agarwal A.R., et al. (2013) Metabolic shift in lung alveolar cell mitochondria following acrolein exposure. Am. J. Physiol. Lung Cell. Mol. Physiol., 305, L764–L773. PubMed

Ishida S., et al. (2013) Bioavailable copper modulates oxidative phosphorylation and growth of tumors. Proc. Natl Acad. Sci. USA, 110, 19507–19512. PubMed PMC

George J., et al. (2011) Cypermethrin exposure leads to regulation of proteins expression involved in neoplastic transformation in mouse skin. Proteomics, 11, 4411–4421. PubMed

Tsitsimpikou C., et al. (2013) Histopathological lesions, oxidative stress and genotoxic effects in liver and kidneys following long term exposure of rabbits to diazinon and propoxur. Toxicology, 307, 109–114. PubMed

Abdollahi M., et al. (2004) Pesticides and oxidative stress: a review. Med. Sci. Monit., 10, RA141–RA147. PubMed

Folkman J. (1995) Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat. Med., 1, 27–31. PubMed

Carmeliet P. (2005) Angiogenesis in life, disease and medicine. Nature, 438, 932–936. PubMed

Folkman J. (1971) Tumor angiogenesis: therapeutic implications. N. Engl. J. Med., 285, 1182–1186. PubMed

Carmeliet P., et al. (2011) Molecular mechanisms and clinical applications of angiogenesis. Nature, 473, 298–307. PubMed PMC

Ferrara N., et al. (2005) Angiogenesis as a therapeutic target. Nature, 438, 967–974. PubMed

Folkman J. (2003) Fundamental concepts of the angiogenic process. Curr. Mol. Med., 3, 643–651. PubMed

Folkman J. (2003) Angiogenesis inhibitors: a new class of drugs. Cancer Biol. Ther., 2, S127–S133. PubMed

Siemann D.W., et al. (2005) Differentiation and definition of vascular-targeted therapies. Clin. Cancer Res., 11(2 Pt 1), 416–420. PubMed

Thorpe P.E. (2004) Vascular targeting agents as cancer therapeutics. Clin. Cancer Res., 10, 415–427. PubMed

Patterson D.M., et al. (2007) Vascular damaging agents. Clin. Oncol. (R. Coll. Radiol.), 19, 443–456. PubMed

Hu Z., et al. (1999) Targeting tumor vasculature endothelial cells and tumor cells for immunotherapy of human melanoma in a mouse xenograft model. Proc. Natl Acad. Sci. USA, 96, 8161–8166. PubMed PMC

Hu Z., et al. (2000) Intratumoral injection of adenoviral vectors encoding tumor-targeted immunoconjugates for cancer immunotherapy. Proc. Natl Acad. Sci. USA, 97, 9221–9225. PubMed PMC

Hu Z., et al. (2001) Targeting tissue factor on tumor vascular endothelial cells and tumor cells for immunotherapy in mouse models of prostatic cancer. Proc. Natl Acad. Sci. USA, 98, 12180–12185. PubMed PMC

Hu Z., et al. (2010) Natural killer cells are crucial for the efficacy of Icon (factor VII/human IgG1 Fc) immunotherapy in human tongue cancer. BMC Immunol., 11, 49. PubMed PMC

Konigsberg W.H., et al. (1988) Molecular cloning of the cDNA for human tissue factor. Cell, 52, 639–640. PubMed

Contrino J., et al. (1996) In situ detection of tissue factor in vascular endothelial cells: correlation with the malignant phenotype of human breast disease. Nat. Med., 2, 209–215. PubMed

Hu Z. (2011) Factor VII-Targeted Photodynamic Therapy for Breast Cancer and Its Therapeutic Potential for Other Solid Cancers and Leukemia, Breast Cancer—Current and Alternative Therapeutic Modalities. In EsraGunduz and MehmetGunduz (eds), InTech, Rijeka, Croatia. http://www.intechopen.com/articles/show/title/factor-vii-targeted-photodynamic-therapy-for-breast-cancer-and-its-therapeutic-potential-for-other-s ISBN: 978-953-307-776-5.

Hu Z., et al. (2010) Targeting tissue factor on tumour cells and angiogenic vascular endothelial cells by factor VII-targeted verteporfin photodynamic therapy for breast cancer in vitro and in vivo in mice. BMC Cancer, 10, 235. PubMed PMC

Duanmu J., et al. (2011) Effective treatment of chemoresistant breast cancer in vitro and in vivo by a factor VII-targeted photodynamic therapy. Br. J. Cancer, 104, 1401–1409. PubMed PMC

Hu Z., et al. (2011) Selective and effective killing of angiogenic vascular endothelial cells and cancer cells by targeting tissue factor using a factor VII-targeted photodynamic therapy for breast cancer. Breast Cancer Res. Treat., 126, 589–600. PubMed PMC

Jessen-Eller K.K.J., et al. (2002) A new invertebrate member of the p53 gene family is developmentally expressed and responds to polychlorinated biphenyls. Environ. Health Perspect., 110, 9. PubMed PMC

Taylor T.R., et al. (2011) Ziram activates mitogen-activated protein kinases and decreases cytolytic protein levels in human natural killer cells. Toxicol. Mech. Methods, 21, 8. PubMed PMC

Berx G., et al. (2009) Involvement of members of the cadherin superfamily in cancer. Cold Spring Harb. Perspect. Biol., 1, a003129. PubMed PMC

Klymkowsky M.W., et al. (2009) Epithelial-mesenchymal transition: a cancer researcher's conceptual friend and foe. Am. J. Pathol., 174, 1588–1593. PubMed PMC

Micalizzi D.S., et al. (2010) Epithelial-mesenchymal transition in cancer: parallels between normal development and tumor progression. J. Mammary Gland Biol. Neoplasia, 15, 117–134. PubMed PMC

Yang J., et al. (2008) Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev. Cell, 14, 818–829. PubMed

Kawata M., et al. (2012) TGF-β-induced epithelial-mesenchymal transition of A549 lung adenocarcinoma cells is enhanced by pro-inflammatory cytokines derived from RAW 264.7 macrophage cells. J. Biochem., 151, 205–216. PubMed

Nagase H., et al. (2006) Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc. Res., 69, 562–573. PubMed

Chen H.C., et al. (1998) Tyrosine phosphorylation of focal adhesion kinase stimulated by hepatocyte growth factor leads to mitogen-activated protein kinase activation. J. Biol. Chem., 273, 25777–25782. PubMed

Al-Mehdi A.B., et al. (2000) Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: a new model for metastasis. Nat. Med., 6, 100–102. PubMed

Chang C.C., et al. (2013) Connective tissue growth factor activates pluripotency genes and mesenchymal-epithelial transition in head and neck cancer cells. Cancer Res., 73, 4147–4157. PubMed

McCormick J.M., et al. (2010) Embryonic exposure to tetrabromobisphenol A and its metabolites, bisphenol A and tetrabromobisphenol A dimethyl ether disrupts normal zebrafish (Danio rerio) development and matrix metalloproteinase expression. Aquat. Toxicol., 100, 255–262. PubMed PMC

Ding S.Z., et al. (2013) Epithelial-mesenchymal transition during oncogenic transformation induced by hexavalent chromium involves reactive oxygen species-dependent mechanism in lung epithelial cells. Toxicol. Appl. Pharmacol., 269, 61–71. PubMed PMC

Hsiang C.Y., et al. (2007) Acetaldehyde induces matrix metalloproteinase-9 gene expression via nuclear factor-kappaB and activator protein 1 signaling pathways in human hepatocellular carcinoma cells: association with the invasive potential. Toxicol. Lett., 171, 78–86. PubMed

Seo J.H., et al. (2004) Helicobacter pylori in a Korean isolate activates mitogen-activated protein kinases, AP-1, and NF-kappaB and induces chemokine expression in gastric epithelial AGS cells. Lab. Invest., 84, 49–62. PubMed

Olumi A.F., et al. (1999) Carcinoma-associated fibroblasts direct tumor progression of initiated human prostatic epithelium. Cancer Res., 59, 5002–5011. PubMed PMC

Le Bitoux M.A., et al. (2008) Tumor-host interactions: the role of inflammation. Histochem. Cell Biol., 130, 1079–1090. PubMed

Maffini M.V., et al. (2005) Stromal regulation of neoplastic development: age-dependent normalization of neoplastic mammary cells by mammary stroma. Am. J. Pathol., 167, 1405–1410. PubMed PMC

Weaver V.M., et al. (2004) Watch thy neighbor: cancer is a communal affair. J. Cell Sci., 117, 1287–1290. PubMed

Maguer-Satta V. (2011) The stem cell niche: the black master of cancer. Cancer Stem Cells Theories and Practices, ISBN 978-953-307-225-8.

Laconi S., et al. (2001) A growth-constrained environment drives tumor progression invivo. Proc. Natl Acad. Sci. USA, 98, 7806–7811. PubMed PMC

Ding J., et al. (2009) TNF-alpha induction by nickel compounds is specific through ERKs/AP-1-dependent pathway in human bronchial epithelial cells. Curr. Cancer Drug Targets, 9, 81–90. PubMed

Li J., et al. (2004) Nickel compounds act through phosphatidylinositol-3-kinase/Akt-dependent, p70(S6k)-independent pathway to induce hypoxia inducible factor transactivation and Cap43 expression in mouse epidermal Cl41 cells. Cancer Res., 64, 94–101. PubMed

Ding J., et al. (2006) Nickel compounds render anti-apoptotic effect to human bronchial epithelial Beas-2B cells by induction of cyclooxygenase-2 through an IKKbeta/p65-dependent and IKKalpha- and p50-independent pathway. J. Biol. Chem., 281, 39022–39032. PubMed

Zhang J., et al. (2013) The alteration of miR-222 and its target genes in nickel-induced tumor. Biol. Trace Elem. Res., 152, 267–274. PubMed

Allard P., et al. (2010) Bisphenol A impairs the double-strand break repair machinery in the germline and causes chromosome abnormalities. Proc. Natl Acad. Sci. USA, 107, 20405–20410. PubMed PMC

Hassan Z.K., et al. (2012) Bisphenol A induces hepatotoxicity through oxidative stress in rat model. Oxid. Med. Cell. Longev., 2012, 194829. PubMed PMC

Takahashi A., et al. (2004) Bisphenol A from dental polycarbonate crown upregulates the expression of hTERT. J. Biomed. Mater. Res. B. Appl. Biomater., 71, 214–221. PubMed

Hurt K., et al. (2013) Tributyltin and dibutyltin alter secretion of tumor necrosis factor alpha from human natural killer cells and a mixture of T cells and natural killer cells. J. Appl. Toxicol., 33, 503–510. PubMed PMC

Patel E., et al. (2013) Methylmercury impairs motor function in early development and induces oxidative stress in cerebellar granule cells. Toxicol. Lett., 222, 265–272. PubMed

Sherwani S.I., et al. (2013) Eicosanoid signaling and vascular dysfunction: methylmercury-induced phospholipase D activation in vascular endothelial cells. Cell Biochem. Biophys., 67, 317–329. PubMed

Black A.T., et al. (2008) Increased oxidative stress and antioxidant expression in mouse keratinocytes following exposure to paraquat. Toxicol. Appl. Pharmacol., 231, 384–392. PubMed PMC

Chang X., et al. (2013) Paraquat inhibits cell viability via enhanced oxidative stress and apoptosis in human neural progenitor cells. Chem. Biol. Interact., 206, 248–255. PubMed

Khatami M. (2014) Chronic inflammation: synergistic interactions of recruiting macrophages (TAMs) and eosinophils (Eos) with host mast cells (MCs) and tumorigenesis in CALTs. M-CSF, suitable biomarker for cancer diagnosis! Cancers (Basel), 6, 297–322. PubMed PMC

Whiteside T.L. (2006) Immune suppression in cancer: effects on immune cells, mechanisms and future therapeutic intervention. Semin Cancer Biol., 16, 3–15. PubMed

Whiteside T.L. (2002) Tumor-induced death of immune cells: its mechanisms and consequences. Semin Cancer Biol., 12, 43–50. PubMed

Yang L., et al. (2010) TGF-β and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol., 31, 220–227. PubMed PMC

Mocellin S., et al. (2004) The multifaceted relationship between IL-10 and adaptive immunity: putting together the pieces of a puzzle. Cytokine Growth Factor Rev., 15, 61–76. PubMed

Zou W. (2005) Immunosuppressive networks in the tumour environment and their therapeutic relevance. Nat. Rev. Cancer, 5, 263–274. PubMed

IARC (2013) IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Agents classified by the IARC Monographs. International Agency for Research and Cancer, Lyon.

IARC (1982) Monograph on the Evaluation of Carcinogenic Risk to Humans. Some Industrial Chemicals and Dyestuffs. Di(2-ethylhexyl) Phthalate. International Agency for Research and Cancer, Lyon, pp. 257–280.

IARC (2000) Monograph on the Evaluation of Carcinogenic Risk to Humans. Some Industrial Chemicals. Di(2-ethylhexyl) Phthalate. International Agency for Research and Cancer, Lyon, pp. 41–148.

Odermatt A., et al. (2008) Disruption of glucocorticoid and mineralocorticoid receptor-mediated responses by environmental chemicals. CHIMIA Int. J. Chem., 62, 335–339.

Santos P.M., et al. (2009) Insights into yeast adaptive response to the agricultural fungicide mancozeb: a toxicoproteomics approach. Proteomics, 9, 657–670. PubMed

Kuster M., et al. (2009) Liquid chromatography–tandem mass spectrometric analysis and regulatory issues of polar pesticides in natural and treated waters. J. Chromatogr. A, 1216, 520–529. PubMed

Kim J.H., et al. (2013) Synergism of antifungal activity between mitochondrial respiration inhibitors and kojic acid. Molecules, 18, 1564–1581. PubMed PMC

Çayır A., et al. (2014) Micronuclei, nucleoplasmic bridges, and nuclear buds induced in human lymphocytes by the fungicide signum and its active ingredients (boscalid and pyraclostrobin). Environ. Toxicol, 29, 723–732. PubMed

Judson R.S., et al. (2010) In vitro screening of environmental chemicals for targeted testing prioritization: the ToxCast project. Environ. Health Persp. (Online), 118, 485. PubMed PMC

Pereboeva L., et al. (2013) DNA damage responses and oxidative stress in dyskeratosis congenita. PLoS One, 8, e76473. PubMed PMC

Pinchuk L.M., et al. (2007) In vitro atrazine exposure affects the phenotypic and functional maturation of dendritic cells. Toxicol. Appl. Pharmacol., 223, 206–217. PubMed PMC

Kavlock R., et al. (2012) Update on EPA’s ToxCast program: providing high throughput decision support tools for chemical risk management. Chem. Res. Toxicol., 25, 1287–1302. PubMed

Martin M.T., et al. (2011) Predictive model of rat reproductive toxicity from ToxCast high throughput screening. Biol. Reprod., 85, 327–339. PubMed

Metzler M., et al. (2001) Chemistry of natural and anthropogenic endocrine active compounds. In M.Metzler (ed) The Handbook of Environmental Chemistry Vol. 3, Part L. Endocrine Disruptors–Part I. Springer, Berlin Heidelberg, pp. 63–80.

Gatidou G., et al. (2007) Simultaneous determination of the endocrine disrupting compounds nonylphenol, nonylphenol ethoxylates, triclosan and bisphenol A in wastewater and sewage sludge by gas chromatography–mass spectrometry. J. Chromatogr. A, 1138, 32–41. PubMed

Foran C., et al. (2000) Developmental evaluation of a potential non-steroidal estrogen: triclosan. Mar. Environ. Res., 50, 153–156. PubMed

Ishibashi H., et al. (2004) Effects of triclosan on the early life stages and reproduction of medaka Oryzias latipes and induction of hepatic vitellogenin. Aquat. Toxicol., 67, 167–179. PubMed

Boyd G.R., et al. (2003) Pharmaceuticals and personal care products (PPCPs) in surface and treated waters of Louisiana, USA and Ontario, Canada. Sci. Total Environ., 311, 135–149. PubMed

Kolpin D.W., et al. (2002) Pharmaceuticals, hormones, and other organic wastewater contaminants in US streams, 1999–2000: a national reconnaissance. Environ. Sci. Technol., 36, 1202–1211. PubMed

Vandhana S., et al. (2013) Biochemical changes accompanying apoptotic cell death in retinoblastoma cancer cells treated with lipogenic enzyme inhibitors. Biochim. Biophys. Acta (BBA)-Molecular and Cell Biology of Lipids, 1831, 1458–1466. PubMed

Zuckerbraun H.L., et al. (1998) Triclosan, cytotoxicity, mode of action, and induction of apoptosis in human gingival cells in vitro . Eur. J. Oral Sci., 106, 628–636. PubMed

Terasaka H., et al. (2005) Cytotoxicity and apoptosis-inducing activity of bisphenol A and hydroquinone in HL-60 cells. Anticancer Res., 25, 2241–2247. PubMed

The Australian National Industrial Chemicals Notification and Assessment Scheme (2006) Diethylhexyl Phthalate (DEHP) Factsheet. CAS: 117-81-7. http://www.nicnas.gov.au/communications/publications/information-sheets/existing-chemical-info-sheets/diethylhexyl-phthalate-dehp-factsheet (7 May 2015, date last accessed).

Jobling S., et al. (1995) A variety of environmentally persistent chemicals, including some phthalate plasticizers, are weakly estrogenic. Environ. Health Persp., 103, 582. PubMed PMC

Hao C., et al. (2012) Perinatal exposure to diethyl-hexyl-phthalate induces obesity in mice. Front. Biosci. (Elite edition), 5, 725–733. PubMed

Moushumi Priya A., et al. (2012) Induction of Apoptosis and cell cycle arrest by Bis (2-ethylhexyl) phthalate produced by Marine Bacillus pumilus MB 40. Chem. Biol. Interact., 195, 133–143. PubMed

Park M.A., et al. (2012) Cell growth of BG-1 ovarian cancer cells is promoted by di-n-butyl phthalate and hexabromocyclododecane via upregulation of the cyclin D and cyclin-dependent kinase-4 genes. Mol. Med. Rep., 5, 761–766. PubMed

Kinoshita Y., et al. (2003) Induction of aromatase (CYP19) expression in breast cancer cells through a nongenomic action of estrogen receptor alpha. Cancer Res., 63, 3546–3555. PubMed

Prins G.S. (2008) Endocrine disruptors and prostate cancer risk. Endocr. Relat. Cancer, 15, 649–656. PubMed PMC

Laville N., et al. (2006) Modulation of aromatase activity and mRNA by various selected pesticides in the human choriocarcinoma JEG-3 cell line. Toxicology, 228, 98–108. PubMed

Bulun S.E., et al. (2007) Aromatase excess in cancers of breast, endometrium and ovary. J. Steroid Biochem. Mol. Biol., 106, 81–96. PubMed PMC

(2012) A Review of Human Carcinogens—Pharmaceuticals IARC Monographs on the Evaluation of Caricinogenic Risk to Humans. Vol. 100, World health Organization IARC, Geneva, Switzerland.

Paul A., et al. (2014) The breast cancer susceptibility genes (BRCA) in breast and ovarian cancers. Front. Biosci. (Landmark Ed), 19, 605–618. PubMed PMC

Supplementary Guidance for Conducting Health Risk Assessment of Chemical Mixtures (2000) U.S. Environmental Protection Agency Report No. EPA/630/R-00/002, Washington, DC.

Dellarco V., et al. (2012) Mode of action: moving toward a more relevant and efficient assessment paradigm. J. Nutr., 142, 2192S–2198S. PubMed

Meek M.E., et al. (2003) A framework for human relevance analysis of information on carcinogenic modes of action. Crit. Rev. Toxicol., 33, 591–653. PubMed

Boobis A.R., et al. (2006) IPCS framework for analyzing the relevance of a cancer mode of action for humans. Crit. Rev. Toxicol., 36, 781–792. PubMed

(2012) OECD Guidance Document 116 On The Conduct And Design Of Chronic Toxicity And Carcinogenicity Studies, Supporting Test Guidelines 451, 452 And 453, 2nd Edition, Series on Testing and Assessment, ENV/JM/MONO(2011)47. OECD Environment Directorate Joint Meeting of the Chemicals Committee and the Working Party on Chemicals, Pesticides and Biotechnology, Paris, France.

U.S.E.P.A. (2002) Guidance on Cumulative Risk Assessment of Pesticide Chemicals That Have a Common Mechanism of Toxicity. In Office of Pesticide Programs, Washington, D.C. 20460.

EFSA PPR Panel (EFSA Panel on Plant Protection Products and their Residues) (2013). Scientific opinion on the relevance of dissimilar mode of action and its appropriate application for cumulative risk assessment of pesticides residues in food. EFSA J., 11, 40.

Driedger A.A., et al. (1971) Demonstration of two types of DNA repair in X-irradiated Micrococcus radiodurans. Can. J. Microbiol., 17, 495–499. PubMed

Mason P.A., et al. (2003) Mismatch repair activity in mammalian mitochondria. Nucleic Acids Res., 31, 1052–1058. PubMed PMC

Heindorff K., et al. (1983) Genetic toxicology of ethylenediaminetetraacetic acid (EDTA). Mutat. Res., 115, 149–173. PubMed

Cory-Slechta, D. et al. (2008) Phthalates Cumulative Risk Assessment—The Tasks Ahead. In National Research Council, N.A.o.S., Board on Environmental Science and Technology, Committee on Phthalates Health Risks. National Academy Press, Washington, DC, p. 208.

Bucala R. (1996) MIF rediscovered: cytokine, pituitary hormone, and glucocorticoid-induced regulator of the immune response. FASEB J., 10, 1607–1613. PubMed

Bucala R., et al. (2007) Macrophage migration inhibitory factor: a probable link between inflammation and cancer. Immunity, 26, 281–285. PubMed

Grieb G., et al. (2010) Macrophage migration inhibitory factor (MIF): a promising biomarker. Drug News Perspect., 23, 257–264. PubMed PMC

Landesmann B., et al. (2013) Adverse outcome pathway-based screening strategies for an animal-free safety assessment of chemicals. Altern. Lab. Anim., 41, 461–471. PubMed

Malhotra J., et al. (2015) Effect of occupational exposures on lung cancer susceptibility: a study of gene-environment interaction analysis. Cancer Epidemiol. Biomarkers Prev., 24, 570–579. PubMed

Singh S., et al. (2012) Epigenetic effects of environmental chemicals bisphenol a and phthalates. Int. J. Mol. Sci., 13, 10143–10153. PubMed PMC

Vineis P., et al. (2010) Models of carcinogenesis: an overview. Carcinogenesis, 31, 1703–1709. PubMed PMC

Brash D., et al. (2009) The mysterious steps in carcinogenesis. Br. J. Cancer, 101, 379–380. PubMed PMC

Brash D., et al. (2009) The mysterious steps in carcinogenesis: addendum. Br. J. Cancer, 101, 1490. PubMed PMC

Rappaport S.M., et al. (2010) Epidemiology. Environment and disease risks. Science, 330, 460–461. PubMed PMC

Bisson W.H. (2012) Editorial: computational chemogenomics in drug design and discovery. Curr. Top. Med. Chem., 12, 1867–1868. PubMed

Tomasetti C., et al. (2015) Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science, 347, 78–81. PubMed PMC

Weinberg R.A. (2014) Coming full circle-from endless complexity to simplicity and back again. Cell, 157, 267–271. PubMed

Koutsogiannouli E., et al. (2013) Complexity in cancer biology: is systems biology the answer? Cancer Med., 2, 164–177. PubMed PMC

Alberghina L., et al. (2004) Systems biology and the molecular circuits of cancer. Chembiochem, 5, 1322–1333. PubMed

Carvalho R.N., et al. (2014) Mixtures of chemical pollutants at European legislation safety concentrations: how safe are they? Toxicol. Sci., 141, 218–233. PubMed PMC

Porter W.P., et al. (1999) Endocrine, immune, and behavioral effects of aldicarb (carbamate), atrazine (triazine) and nitrate (fertilizer) mixtures at groundwater concentrations. Toxicol. Ind. Health, 15, 133–150. PubMed

Tarone R.E., et al. (2011) Combating environmental causes of cancer. N. Engl. J. Med., 364, 2266–2267. PubMed

Willett W.C., et al. (2011) Combating environmental causes of cancer. N. Engl. J. Med., 364, 2266. PubMed

Richter E.D., et al. (2004) The precautionary principle, epidemiology and the ethics of delay. Int. J. Occup. Med. Environ. Health, 17, 9–16. PubMed

Pohl H.R., et al. (2010) Chemical risk assessment and uncertainty associated with extrapolation across exposure duration. Regul. Toxicol. Pharmacol., 57, 18–23. PubMed

Tice R.R., et al. (2013) Improving the human hazard characterization of chemicals: a Tox21 update. Environ. Health Perspect., 121, 756–765. PubMed PMC

Find record

Citation metrics

Loading data ...

Archiving options

Loading data ...