B. Allen, C. Hack, and H. Clewell, Use of Markov Chain Monte Carlo Analysis with a Physiologically-Based Pharmacokinetic Model of Methylmercury to Estimate Exposures in U.S. Women of Childbearing Age, Risk Analysis, vol.25, issue.2, pp.947-959, 2007.
DOI : 10.1111/0272-4332.00061

S. Beal and L. Sheiner, Estimating population kinetics, Crit Rev Biomed Eng, vol.8, issue.3, pp.195-222, 1982.

P. Bernillon and F. Bois, Statistical Issues in Toxicokinetic Modeling: A Bayesian Perspective, Environmental Health Perspectives, vol.108, issue.s5, pp.883-893, 2000.
DOI : 10.1289/ehp.00108s5883

URL : https://hal.archives-ouvertes.fr/ineris-00961852

F. Bois, Physiologically Based Modelling and Prediction of Drug Interactions, Basic & Clinical Pharmacology & Toxicology, vol.31, issue.9, pp.154-161, 2010.
DOI : 10.1111/j.1742-7843.2009.00488.x

F. Bois and P. Compton-quintana, Sensitivity analysis of a new model of carcinogenesis, Journal of Theoretical Biology, vol.159, issue.3, pp.361-375, 1992.
DOI : 10.1016/S0022-5193(05)80730-1

F. Bois, A. Gelman, J. Jiang, D. Maszle, L. Zeise et al., Population toxicokinetics of tetrachloroethylene, Archives of Toxicology, vol.70, issue.6, pp.347-355, 1996.
DOI : 10.1007/s002040050284

URL : https://hal.archives-ouvertes.fr/ineris-00969532

F. Bois, M. Jamei, and H. Clewell, PBPK modelling of inter-individual variability in the pharmacokinetics of environmental chemicals, Toxicology, vol.278, issue.3, pp.256-267, 2010.
DOI : 10.1016/j.tox.2010.06.007

URL : https://hal.archives-ouvertes.fr/ineris-00961755

F. Bois, G. Krowech, and L. Zeise, Modeling Human Interindividual Variability in Metabolism and Risk: The Example of 4-Aminobiphenyl, Risk Analysis, vol.266, issue.Supplement 10, pp.205-213, 1995.
DOI : 10.1111/j.1539-6924.1995.tb00314.x

F. Bois, L. Zeise, and T. Tozer, Precision and sensitivity of pharmacokinetic models for cancer risk assessment: Tetrachloroethylene in mice, rats, and humans, Toxicology and Applied Pharmacology, vol.102, issue.2, pp.300-315, 1990.
DOI : 10.1016/0041-008X(90)90029-T

B. Bradford, E. Lock, O. Kosyk, S. Kim, T. Uehara et al., Interstrain Differences in the Liver Effects of Trichloroethylene in a Multistrain Panel of Inbred Mice, Toxicological Sciences, vol.120, issue.1, pp.206-217, 2011.
DOI : 10.1093/toxsci/kfq362

J. Brown, T. Bateson, and W. Mcdonnell, Effects of Exposure to 0.06 ppm Ozone on FEV1 in Humans: A Secondary Analysis of Existing Data, Environmental Health Perspectives, vol.116, issue.8, pp.1023-1026, 2008.
DOI : 10.1289/ehp.11396

D. Burmaster and D. Murray, A Trivariate Distribution for the Height, Weight, and Fat of Adult Men*, Risk Analysis, vol.60, issue.4, pp.385-389, 1998.
DOI : 10.1111/j.1539-6924.1998.tb00352.x

Y. Chen, J. Zhu, P. Lum, X. Yang, S. Pinto et al., Variations in DNA elucidate molecular networks that cause disease, Nature, vol.140, issue.7186, pp.429-435, 2008.
DOI : 10.1038/nature06757

URL : https://hal.archives-ouvertes.fr/hal-00283069

W. Chiu, S. Euling, C. Scott, and R. Subramaniam, Approaches to advancing quantitative human health risk assessment of environ mental chemi cals in the postgenomic era, Toxicol Appl Pharmacol, p.19, 2010.

W. Chiu, M. Okino, and M. Evans, Characterizing uncertainty and population variability in the toxicokinetics of trichloroethylene and metabolites in mice, rats, and humans using an updated database, physiologically based pharmacokinetic (PBPK) model, and Bayesian approach, Toxicology and Applied Pharmacology, vol.241, issue.1, pp.36-60, 2009.
DOI : 10.1016/j.taap.2009.07.032

F. Collins, G. Gray, and J. Bucher, TOXICOLOGY: Transforming Environmental Health Protection, Science, vol.319, issue.5865, pp.906-907, 2008.
DOI : 10.1126/science.1154619

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2679521

K. Crump, C. Chen, W. Chiu, T. Louis, C. Portier et al., What Role for Biologically Based Dose???Response Models in Estimating Low-Dose Risk?, Environmental Health Perspectives, vol.118, issue.5, pp.585-588, 2010.
DOI : 10.1289/ehp.0901249

C. Csajka and D. Verotta, Pharmacokinetic???Pharmacodynamic Modelling: History and Perspectives, Journal of Pharmacokinetics and Pharmacodynamics, vol.21, issue.3, pp.227-279, 2006.
DOI : 10.1007/s10928-005-9002-0

J. Dorne, Metabolism, variability and risk assessment, Toxicology, vol.268, issue.3, pp.156-164, 2010.
DOI : 10.1016/j.tox.2009.11.004

M. Dumas, S. Wilder, M. Bihoreau, R. Barton, J. Fearnside et al., Direct quantitative trait locus mapping of mammalian metabolic phenotypes in diabetic and normoglycemic rat models, Nature Genetics, vol.75, issue.5, pp.666-672, 2007.
DOI : 10.1038/ng2026

R. Durbin, G. Abecasis, D. Altshuler, A. Auton, L. Brooks et al., A map of human genome variation from population-scale sequencing, Nature, vol.10, issue.7319, pp.1061-1073, 2010.
DOI : 10.1038/nature09534

URL : https://hal.archives-ouvertes.fr/cea-00904997

V. Emilsson, G. Tholeifsson, B. Zhang, A. Leonardson, F. Zink et al., Genetics of gene expression and its effect on disease, Nature, vol.138, issue.7186, pp.423-428, 2008.
DOI : 10.1038/nature06758

C. Gieger, L. Geistlinger, E. Altmaier, M. De-angelis, F. Kronenberg et al., Genetics Meets Metabolomics: A Genome-Wide Association Study of Metabolite Profiles in Human Serum, PLoS Genetics, vol.6, issue.11, 2008.
DOI : 10.1371/journal.pgen.1000282.s009

G. Ginsberg, A. K. Guyton, K. Sonawane, and B. , Polymorphism in the DNA repair enzyme XRCC1: Utility of current database and implications for human health risk assessment, Mutation Research/Reviews in Mutation Research, vol.727, issue.1-2, pp.1-15, 2011.
DOI : 10.1016/j.mrrev.2011.02.001

G. Ginsberg, K. Guyton, D. Johns, J. Schimek, K. Angle et al., Genetic polymorphism in metabolism and host defense enzymes: Implications for human health risk assessment, Critical Reviews in Toxicology, vol.10, issue.6, pp.575-619, 2010.
DOI : 10.1080/15287390801988772

G. Ginsberg, D. Hattis, and B. Sonawane, Incorporating pharmacokinetic differences between children and adults in assessing children's risks to environmental toxicants, Toxicology and Applied Pharmacology, vol.198, issue.2, pp.164-183, 2004.
DOI : 10.1016/j.taap.2003.10.010

G. Ginsberg, D. Hattis, B. Sonawane, A. Russ, P. Banati et al., Evaluation of Child/Adult Pharmacokinetic Differences from a Database Derived from the Therapeutic Drug Literature, Toxicological Sciences, vol.66, issue.2, pp.185-200, 2002.
DOI : 10.1093/toxsci/66.2.185

G. Ginsberg, P. Neafsey, D. Hattis, K. Guyton, D. Johns et al., Genetic Polymorphism in Paraoxonase 1 (PON1): Population Distribution of PON1 Activity, Journal of Toxicology and Environmental Health, Part B, vol.48, issue.5-6, pp.473-507, 2009.
DOI : 10.1007/s10038-004-0176-x

G. Ginsberg, S. Smolenski, D. Hattis, K. Guyton, D. Johns et al., Genetic Polymorphism in Glutathione Transferases (GST): Population Distribution of GSTM1, T1, and P1 Conjugating Activity, Journal of Toxicology and Environmental Health, Part B, vol.32, issue.2, pp.389-439, 2009.
DOI : 10.1515/CCLM.2004.059

G. Ginsberg, S. Smolenski, P. Neafsey, D. Hattis, K. Walker et al., The Influence of Genetic Polymorphisms on Population Variability in Six Xenobiotic-Metabolizing Enzymes, Journal of Toxicology and Environmental Health, Part B, vol.23, issue.5-6, pp.307-333, 2009.
DOI : 10.1080/15287390801988772

T. Giorgino, D. Abramo, M. Gervasio, F. , D. Fabritiis et al., Exploring the kinetics of drug binding to the hERG channel ? Environmental Health Perspectives through large-scale simulations [Abstract], Proceedings of the 2010 Virtual Physiological Human Conference, 30 September?1, pp.248-250, 2010.

Y. Guo, P. Lu, E. Farrell, X. Zhang, P. Weller et al., In silico and in vitro pharmacogenetic analysis in mice, Proceedings of the National Academy of Sciences, vol.104, issue.45, pp.17735-17740, 2007.
DOI : 10.1073/pnas.0700724104

Y. Guo, P. Weller, E. Farrell, P. Cheung, B. Fitch et al., In silico pharmacogenetics of warfarin metabolism, Nature Biotechnology, vol.23, issue.5, pp.531-536, 2006.
DOI : 10.1038/nbt1195

K. Guyton, A. Kyle, J. Aubrecht, V. Cogliano, D. Eastmond et al., Improving prediction of chemi cal carcinogenicity by considering multiple mechanisms and applying toxicogenomic approaches, Mutat Res Rev Mutat Res, vol.681, pp.2-3230, 2009.

C. Hack, Bayesian analysis of physiologically based toxico kinetic and toxicodynamic models, Toxicology, vol.2212, issue.3, pp.241-248, 2006.

A. Hamade, V. Misra, R. Rabold, and C. Tankersley, Age-related changes in cardiac and respiratory adaptation to acute ozone and carbon black exposures: Interstrain variation in mice, Inhalation Toxicology, vol.7, issue.sup2, pp.84-94, 2010.
DOI : 10.1080/08958370290084674

A. Harrill, P. Ross, D. Gatti, D. Threadgill, and I. Rusyn, Population-Based Discovery of Toxicogenomics Biomarkers for Hepatotoxicity Using a Laboratory Strain Diversity Panel, Toxicological Sciences, vol.110, issue.1, pp.235-243, 2009.
DOI : 10.1093/toxsci/kfp096

A. Harrill, P. Watkins, S. Su, P. Ross, D. Harbourt et al., Mouse population-guided resequencing reveals that variants in CD44 contribute to acetaminophen-induced liver injury in humans, Genome Research, vol.19, issue.9, pp.1507-1515, 2009.
DOI : 10.1101/gr.090241.108

D. Hattis, Human variability in susceptibility, Environmental Toxicology and Pharmacology, vol.4, issue.3-4, pp.3-4195, 1997.
DOI : 10.1016/S1382-6689(97)10012-6

D. Hattis, S. Baird, and R. Goble, A STRAW MAN PROPOSAL FOR A QUANTITATIVE DEFINITION OF THE RfD, Drug and Chemical Toxicology, vol.19, issue.4, pp.403-436, 2002.
DOI : 10.1093/toxsci/66.2.185

D. Hattis, G. Ginsberg, B. Sonawane, S. Smolenski, A. Russ et al., Differences in Pharmacokinetics Between Children and Adults-II. Children's Variability in Drug Elimination Half-Lives and in Some Parameters Needed for Physiologically-Based Pharmacokinetic Modeling, Risk Analysis, vol.24, issue.1, pp.117-142, 2003.
DOI : 10.1111/1539-6924.00295

D. Hattis, A. Russ, R. Goble, P. Banati, and M. Chu, Human Interindividual Variability in Susceptibility to Airborne Particles, Risk Analysis, vol.21, issue.4, pp.585-599, 2001.
DOI : 10.1111/0272-4332.214137

M. Hernandez, J. Lay, B. Harris, C. Esther, . Jr et al., Atopic asthmatic subjects but not atopic subjects without asthma have enhanced inflammatory response to ozone, Journal of Allergy and Clinical Immunology, vol.126, issue.3, 2010.
DOI : 10.1016/j.jaci.2010.06.043

J. Holloway, S. Francis, S. Fong, K. Yang, and I. , Genomics and the respiratory effects of air pollution exposure, Respirology, vol.183, issue.159, pp.590-600, 2012.
DOI : 10.1111/j.1440-1843.2012.02164.x

R. Huang, S. Johnatty, E. Gamazon, H. Im, D. Ziliak et al., Platinum Sensitivity???Related Germline Polymorphism Discovered via a Cell-Based Approach and Analysis of Its Association with Outcome in Ovarian Cancer Patients, Clinical Cancer Research, vol.17, issue.16, pp.5490-5500, 2011.
DOI : 10.1158/1078-0432.CCR-11-0724

M. Hucka, A. Finney, H. Sauro, H. Bolouri, J. Doyle et al., The systems biology markup language (SBML): a medium for representation and exchange of biochemical network models, Bioinformatics, vol.19, issue.4, pp.524-531, 2003.
DOI : 10.1093/bioinformatics/btg015

C. Hutter, J. Chang-claude, M. Slattery, B. Pflugeisen, Y. Lin et al., Characterization of Gene-Environment Interactions for Colorectal Cancer Susceptibility Loci, Cancer Research, vol.72, issue.8, pp.2036-2044, 2012.
DOI : 10.1158/0008-5472.CAN-11-4067

T. Illig, C. Gieger, G. Zhai, W. Römisch-margl, R. Wang-sattler et al., A genome-wide perspective of genetic variation in human metabolism, Nature Genetics, vol.78, issue.2, pp.137-141, 2010.
DOI : 10.1038/ng.507

M. Jamei, S. Marciniak, K. Feng, A. Barnett, G. Tucker et al., Population-based ADME Simulator, Expert Opinion on Drug Metabolism & Toxicology, vol.45, issue.2, pp.211-223, 2009.
DOI : 10.1080/00498250701620726

X. Jia, X. Song, M. Shima, K. Tamura, F. Deng et al., Acute effect of ambient ozone on heart rate variability in healthy elderly subjects, Journal of Exposure Science and Environmental Epidemiology, vol.94, issue.5, pp.541-547, 2011.
DOI : 10.1289/ehp.8337

G. Johanson, F. Jonsson, and F. Bois, Development of new technique for risk assessment using physiologically based toxico kinetic models, Am J Ind Med, vol.1, pp.101-103, 1999.

F. Jonsson, E. Jonsson, F. Bois, and S. Marshall, The Application of a Bayesian Approach to the Analysis of a Complex, Mechanistically Based Model, Journal of Biopharmaceutical Statistics, vol.1, issue.1, pp.65-92, 2007.
DOI : 10.1093/clinids/2.2.282

URL : https://hal.archives-ouvertes.fr/ineris-00963050

R. Judson, R. Kavlock, R. Setzer, E. Hubal, M. Martin et al., Estimating Toxicity-Related Biological Pathway Altering Doses for High-Throughput Chemical Risk Assessment, Chemical Research in Toxicology, vol.24, issue.4, pp.451-462, 2011.
DOI : 10.1021/tx100428e

G. Kedderis, Extrapolation of in vitro enzyme induction data to humans in vivo, Chemico-Biological Interactions, vol.107, issue.1-2, pp.109-121, 1997.
DOI : 10.1016/S0009-2797(97)00076-8

M. Khoury, M. Bowen, W. Burke, R. Coates, N. Dowling et al., Current Priorities for Public Health Practice in Addressing the Role of Human Genomics in Improving Population Health, American Journal of Preventive Medicine, vol.40, issue.4, pp.486-493, 2011.
DOI : 10.1016/j.amepre.2010.12.009

S. Kleeberger, R. Levitt, L. Zhang, M. Longphre, J. Harkema et al., Linkage analysis of susceptibility to ozone-induced lung inflammation in inbred mice, Nature Genetics, vol.269, issue.83, pp.475-478, 1997.
DOI : 10.1038/ng1297-475

I. Koturbash, A. Scherhag, J. Sorrentino, K. Sexton, W. Bodnar et al., Epigenetic Mechanisms of Mouse Interstrain Variability in Genotoxicity of the Environmental Toxicant 1,3-Butadiene, Toxicological Sciences, vol.122, issue.2, pp.448-456, 2011.
DOI : 10.1093/toxsci/kfr133

L. Thangue, N. Kerr, and D. , Predictive biomarkers: a paradigm shift towards personalized cancer medicine, Nature Reviews Clinical Oncology, vol.363, issue.10, pp.587-596, 2011.
DOI : 10.1038/nrclinonc.2011.121

E. Lock, N. Abdo, R. Huang, M. Xia, O. Kosyk et al., Quantitative High-Throughput Screening for Chemical Toxicity in a Population-Based In Vitro Model, Toxicological Sciences, vol.126, issue.2, pp.578-588, 2012.
DOI : 10.1093/toxsci/kfs023

N. Luke, M. Devito, I. Shah, and H. El-masri, Development of a Quantitative Model of Pregnane X Receptor (PXR) Mediated Xenobiotic Metabolizing Enzyme Induction, Bulletin of Mathematical Biology, vol.46, issue.3, pp.1799-1819, 2010.
DOI : 10.1007/s11538-010-9508-5

N. Luke, R. Sams, . Ii, M. Devito, R. Conolly et al., Development of a Quantitative Model Incorporating Key Events in a Hepatotoxic Mode of Action to Predict Tumor Incidence, Toxicological Sciences, vol.115, issue.1, pp.253-266, 2010.
DOI : 10.1093/toxsci/kfq021

T. Manolio, Genomewide association studies and assessment of the risk of disease, N Engl J Med, vol.363, issue.2, pp.166-176, 2010.

T. Manolio, F. Collins, N. Cox, D. Goldstein, L. Hindorff et al., Finding the missing heritability of complex diseases, Nature, vol.41, issue.7265, pp.747-753, 2009.
DOI : 10.1038/nature08494

M. Martin, D. Dix, R. Judson, R. Kavlock, D. Reif et al., Impact of Environmental Chemicals on Key Transcription Regulators and Correlation to Toxicity End Points within EPA???s ToxCast Program, Chemical Research in Toxicology, vol.23, issue.3, pp.578-590, 2010.
DOI : 10.1021/tx900325g

E. Mclanahan, M. Andersen, and J. Fisher, A Biologically Based Dose-Response Model for Dietary Iodide and the Hypothalamic-Pituitary-Thyroid Axis in the Adult Rat: Evaluation of Iodide Deficiency, Toxicological Sciences, vol.102, issue.2, pp.241-253, 2008.
DOI : 10.1093/toxsci/kfm312

H. Mortensen and S. Euling, Integrating mechanistic and polymorphism data to characterize human genetic susceptibility for environ mental chemi cal risk assessment in the 21st century, Toxicol Appl Pharmacol, p.15, 2011.

P. Neafsey, G. Ginsberg, D. Hattis, D. Johns, K. Guyton et al., Genetic Polymorphism in CYP2E1: Population Distribution of CYP2E1 Activity, Journal of Toxicology and Environmental Health, Part B, vol.41, issue.5-6, pp.362-388, 2009.
DOI : 10.1038/clpt.1993.125

P. Neafsey, G. Ginsberg, D. Hattis, and B. Sonawane, Genetic Polymorphism in Cytochrome P450 2D6 (CYP2D6): Population Distribution of CYP2D6 Activity, Journal of Toxicology and Environmental Health, Part B, vol.387, issue.5-6, pp.334-361, 2009.
DOI : 10.1007/s00210-003-0832-2

A. Nong, Y. Tan, M. Krolski, J. Wang, C. Lunchick et al., Bayesian calibration of a physiologically based pharmaco kinetic/pharmaco dynamic model of carbaryl cholinesterase inhibition, J Toxicol Environ Health A, issue.20, pp.711363-1381, 2008.

O. Shea, S. Schwarz, J. Kosyk, O. Ross, P. Ha et al., In Vitro Screening for Population Variability in Chemical Toxicity, Toxicological Sciences, vol.119, issue.2, pp.398-407, 2011.
DOI : 10.1093/toxsci/kfq322

C. Patel, J. Bhattacharya, and A. Butte, An Environment-Wide Association Study (EWAS) on Type 2 Diabetes Mellitus, PLoS ONE, vol.2, issue.5, 2010.
DOI : 10.1371/journal.pone.0010746.s010

E. Peters, A. Motsinger-reif, T. Havener, L. Everitt, N. Hardison et al., Pharmacogenomic characterization of US FDA-approved cytotoxic drugs, Pharmacogenomics, vol.12, issue.10, pp.1407-1415, 2011.
DOI : 10.2217/pgs.11.92

E. Phillips and S. Mallal, Pharmacogenetics of drug hypersensitivity, Pharmacogenomics, vol.11, issue.7, pp.973-987, 2010.
DOI : 10.2217/pgs.10.77

C. Portier and N. Kaplan, Variability of safe dose estimates when using complicated models of the carcinogenic process A case study: Methylene chloride, Fundamental and Applied Toxicology, vol.13, issue.3, pp.533-544, 1989.
DOI : 10.1016/0272-0590(89)90290-X

D. Prows, H. Shertzer, M. Daly, C. Sidman, and G. Leikauf, Genetic analysis of ozone-induced acute lung injury in sensitive and resistant strains of mice, Nature Genetics, vol.102, issue.4, pp.471-474, 1997.
DOI : 10.1038/ng1297-471

S. Rappaport and M. Smith, Environment and Disease Risks, Science, vol.330, issue.6003, pp.460-461, 2010.
DOI : 10.1126/science.1192603

D. Reif, M. Martin, S. Tan, K. Houck, R. Judson et al., Endocrine Profiling and Prioritization of Environmental Chemicals Using ToxCast Data, Environmental Health Perspectives, vol.118, issue.12, pp.1714-1720, 2010.
DOI : 10.1289/ehp.1002180

N. Rosenberg, L. Huang, E. Jewett, Z. Szpiech, I. Jankovic et al., Genome-wide association studies in diverse populations, Nature Reviews Genetics, vol.58, issue.5, pp.356-366, 2010.
DOI : 10.1038/nrg2760

D. Rotroff, B. Wetmore, D. Dix, S. Ferguson, H. Clewell et al., Incorporating Human Dosimetry and Exposure into High-Throughput In Vitro Toxicity Screening, Toxicological Sciences, vol.117, issue.2, pp.348-358, 2010.
DOI : 10.1093/toxsci/kfq220

A. Roy, D. Loke, N. Saha, O. Viegas, J. Tay et al., Interrelationships of Serum Paraoxonase, Serum Lipids and Apolipoproteins in Normal Pregnancy, Gynecologic and Obstetric Investigation, vol.38, issue.1, pp.10-13, 1994.
DOI : 10.1159/000292435

I. Rusyn, D. Gatti, T. Wiltshire, T. Wiltshire, S. Kleeberger et al., Toxicogenetics: population-based testing of drug and chemical safety in mouse models, Pharmacogenomics, vol.11, issue.8, pp.1127-1136, 2010.
DOI : 10.2217/pgs.10.100

S. Sadiq, D. Wright, O. Kenway, and P. Coveney, Accurate Ensemble Molecular Dynamics Binding Free Energy Ranking of Multidrug-Resistant HIV-1 Proteases, Journal of Chemical Information and Modeling, vol.50, issue.5, pp.890-905, 2010.
DOI : 10.1021/ci100007w

E. Schadt, Molecular networks as sensors and drivers of common human diseases, Nature, vol.324, issue.7261, pp.218-223, 2009.
DOI : 10.1038/nature08454

E. Schadt and J. Björkegren, NEW: Network-Enabled Wisdom in Biology, Medicine, and Health Care, Science Translational Medicine, vol.4, issue.115, p.3002132, 2012.
DOI : 10.1126/scitranslmed.3002132

P. Schuster, Modeling in biological chemistry. From biochemical kinetics to systems biology, Monatshefte fur Chemie, pp.427-446, 2008.
DOI : 10.1007/s00706-008-0892-6

R. Spear and F. Bois, Parameter variability and the interpretation of physiologically based pharmaco kinetic modeling results, Environ Health Perspect, vol.11, pp.61-66, 1994.

K. Suhre, S. Shin, A. Petersen, R. Mohney, D. Meredith et al., Human metabolic individuality in biomedical and pharmaceutical research, Nature, vol.279, issue.7362, pp.54-60, 2011.
DOI : 10.1038/nature10354

C. Thompson, D. Johns, B. Sonawane, H. Barton, D. Hattis et al., Database for Physiologically Based Pharmacokinetic (PBPK) Modeling: Physiological Data for Healthy and Health-Impaired Elderly, Journal of Toxicology and Environmental Health, Part B, vol.38, issue.1, pp.1-24, 2009.
DOI : 10.1093/ageing/28.1.29

J. Wakefield, Bayesian individualization via sampling-based methods, Journal of Pharmacokinetics and Biopharmaceutics, vol.91, issue.1, pp.103-131, 1996.
DOI : 10.1007/BF02353512

K. Walker, G. Ginsberg, D. Hattis, D. Johns, K. Guyton et al., -Acetyltransferase (NAT): Population Distribution of NAT1 and NAT2 Activity, Journal of Toxicology and Environmental Health, Part B, vol.37, issue.1, pp.5-6440, 2009.
DOI : 10.1007/s002280000203

URL : https://hal.archives-ouvertes.fr/hal-00857435

V. Watson, A. Motsinger-reif, N. Hardison, E. Peters, T. Havener et al., Identification and Replication of Loci Involved in Camptothecin-Induced Cytotoxicity Using CEPH Pedigrees, PLoS ONE, vol.11, issue.5, p.17561, 2011.
DOI : 10.1371/journal.pone.0017561.s011

M. Welsh, L. Mangravite, M. Medina, K. Tantisira, W. Zhang et al., Pharmacogenomic Discovery Using Cell-Based Models, Pharmacological Reviews, vol.61, issue.4, pp.413-429, 2009.
DOI : 10.1124/pr.109.001461

C. Wild, Complementing the Genome with an "Exposome": The Outstanding Challenge of Environmental Exposure Measurement in Molecular Epidemiology, Cancer Epidemiology Biomarkers & Prevention, vol.14, issue.8, pp.1847-1850, 2005.
DOI : 10.1158/1055-9965.EPI-05-0456

S. Willmann, K. Höhn, A. Edginton, M. Sevestre, J. Solodenko et al., Development of a physiology-based whole-body population model for assessing the influence of individual variability on the pharmaco kinetics of drugs, 2007.

A. Wood, R. Harrison, S. Semple, J. Ayres, and R. Stockley, Outdoor air pollution is associated with rapid decline of lung function in ??-1-antitrypsin deficiency, Occupational and Environmental Medicine, vol.67, issue.8, pp.556-561, 2010.
DOI : 10.1136/oem.2009.047589

T. Woodruff, L. Zeise, D. Axelrad, K. Guyton, S. Janssen et al., Meeting Report: Moving Upstream???Evaluating Adverse Upstream End Points for Improved Risk Assessment and Decision-Making, Environmental Health Perspectives, vol.116, issue.11, pp.1568-1575, 2008.
DOI : 10.1289/ehp.11516

M. Xia, R. Huang, K. Witt, N. Southall, J. Fostel et al., Compound Cytotoxicity Profiling Using Quantitative High-Throughput Screening, Environmental Health Perspectives, vol.116, issue.3, pp.284-291, 2008.
DOI : 10.1289/ehp.10727

M. Xia, R. Huang, V. Guo, N. Southall, M. Cho et al., Identification of compounds that potentiate CREB signaling as possible enhancers of long-term memory, Proceedings of the National Academy of Sciences, vol.106, issue.7, pp.2412-2417, 2009.
DOI : 10.1073/pnas.0813020106

H. Zhu, I. Rusyn, A. Richard, and A. Tropsha, The Use of Cell Viability Assay Data Improves the Prediction Accuracy of Conventional Quantitative Structure Activity Relationship Models of Animal Carcinogenicity, Environmental Health Perspectives, vol.116, pp.506-513, 2008.
DOI : 10.1289/ehp.10573