The use of biomarkers and genetic epidemiology

Similar documents
Biomarkers of acrylamide

Genetic polymorphisms and head and neck cancer risk (Review)

dna oestrogen GENOTYPE REPORT Patient Name: Date of Birth: Sample Number: Referring Practitioner: Date Reported:

Genetics and Genomics: Influence on Individualization of Medication Regimes

Genomics and proteomics offers new hopes towards a personalized approach to lung cancer prevention and treatment

Department of Respiratory Medicine, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, China

PHASE I Detoxification: The First Line of Defense

Environmental exposures and the etiology of cancer. Paolo Vineis

Biomarkers in Public Health: Development and Applications

Environmental Management & Pollution Environmental and Chemical Carcinogenesis

Susceptibility towards formaldehyde-induced genotoxicity. Günter Speit

The importance of pharmacogenetics in the treatment of epilepsy

Toxicity profiles of heterocyclic aromatic amines Bettina Seeger and Pablo Steinberg

The Breast Cancer Family Registry: Description of Resource and some Applications

Toxicant Disposition and Metabolism. Jan Chambers Center for Environmental Health Sciences College of Veterinary Medicine

Pharmacogenetics in: Primary Care. Bradley T. Wajda D.O.

Caveat: Validation and Limitations of Phenotyping Methods for Drug Metabolizing Enzymes and Transporters

B. Incorrect! Compounds are made more polar, to increase their excretion.

Cancer Biomarkers and Susceptibility Factors. Thomas Kensler, PhD Bloomberg School of Public Health

Estrogen Report GENOTYPE REPORT OPTIMAL HEALTH FOR LIFE. John Doe NAME DATE OF BIRTH 7/31/1999 REFFERRING PRACTITIONER. Dr. Exercise/Diet specialist

optimal health for life genotype report Name: Date of Birth: Sample Number: Referring Practitioner: Date Reported:

Imagine that well before tumors

Welcome to your dna oestrogen report

Transformation of Breast Cancer in Taiwan

ENVIRONMENTAL FACTORS IN VIRUS-ASSOCIATED HUMAN CANCERS

Genetics and Pharmacogenetics in Human Complex Disorders (Example of Bipolar Disorder)

accesalabs.c

A DISSERTATION SUBMITTED TO THE FACULTY OF THE GRADUATE SCHOOL OF THE UNIVERSITY OF MINNESOTA BY. Yan Zhong

Falk Symposium 156: Genetics in Liver Disease. Pharmacogenetics. Gerd Kullak-Ublick

10/19/2017. How Nutritional Genomics Affects You in Nutrition Research and Practice Joyanna Hansen, PhD, RD & Kristin Guertin, PhD, MPH

CYP1A1, GSTM1 and NQO1 gene polymorphisms: Genetic risk factors for small cell lung cancer

Gene environment interaction in tobacco-related cancers

Interpretation of Epidemiologic Studies

International Agency for Research on Cancer (IARC) - Summaries & Evaluations

Welcome. to your dna osetrogen report. Welcome to your dna oestrogen report

Association between the -77T>C polymorphism in the DNA repair gene XRCC1 and lung cancer risk

Biomarkers of Industrial and Environmental Exposure to 1,3 Butadiene

5. Summary of Data Reported and Evaluation 5.1 Exposure data

Exposure Dose Health Effects

Chemical Carcinogenesis November

Evaluation of genome damage in subjects occupationally exposed to possible carcinogens. D. Želježi, M. Mladini, N. Kopjar, A. Hursidic Radulovic

Industrial Toxicology

MOLECULAR EPIDEMIOLOGY Afiono Agung Prasetyo Faculty of Medicine Sebelas Maret University Indonesia

PHARMACOGENETICS OF BREAST CANCER

Big data vs. the individual liver from a regulatory perspective

Early Life Development of Toxicokinetic Pathways: Framework Case Examples and Implications for Safety Assessment

Genetic polymorphisms in the drug detoxification genes and susceptibility to Urinary Bladder Cancer (UBC)

Etiology of Chronic Diseases. Complex Diseases Genes and Environment Initiative

A review of human carcinogens -Part F: Chemical agents and related occupations

Pharmacogenomics and Pharmacokinetics ^

Research Framework for Evaluating the Potential Mode(s)

TOXICOLOGY FOR ENGLISH SPEAKING STUDENTS

Gene-Environment Interactions

Objectives Making CYP450, Drug Interactions, & Pharmacogenetics Easy

ToxStrategies, Inc. and Summit Toxicology

Main Challenges Related to Measuring Biomarkers of Exposure of Bisphenol A and Triclosan

formaldehyde S-hydroxymethyl-glutatione S-formylglutatione Formic acid CO 2 + H 2 O (exhaled) Na + HCOO - (urine)

A sthma and allergies have become some of the most

B reast cancer is the most common cancer in

Association between the CYP1A1 polymorphisms and hepatocellular carcinoma: a meta-analysis

OraSure Test Cutoffs Urine Serum Oral Fluid Type of Exposure 500ng/mL 25ng/mL 10ng/mL. Second-Hand Smoke <100 ng/ml <15 ng/ml <3 ng/ml

2/28/2010. Pharmacogenomics and the Asian Population. Limited efficacy/response to drugs already on the market

2. BALcanOSH MEDNARODNA KONFERENCA ZA REGIONALNO SODELOVANJE, BLED, SLOVENIJA

Genetics of Nicotine Dependence & Impact of Smoking on Bladder Cancer Risk and Prognosis

Principles of Toxicology: The Study of Poisons

ENVIRONMENTAL TOXICOLOGY

Chemical Carcinogenesis November

Pharmacogenetics to tailor Drug Exposure and Outcomes in Kidney Transplantation

OVERALL HEALTH EFFECTS OF SWEDISH MATCH SNUS PRODUCTS

Cytokrom P450 (CYP) Hepatic Drug Metabolism. Medicines in plasma. Plasma concentration of a medicine. Eva Brittebo Dept Pharmaceutical Biosciences

Nicotine dependence treatment: A translational research approach

PHARMACOGENETICS. Marshalling the human genome to individualize drug therapy

Genomics (HMGP 7620) Pharmacogenomics.

Click to edit Master title style

Development of NJ Human Health-based Criteria and Standards

Record of the Consultation on Pharmacogenomics/Biomarkers

Chapter 2. Review of Literature

BIOMARKERS AND TOXICITY MECHANISMS 07 Mechanisms Metabolism & Detoxification. Luděk Bláha, PřF MU, RECETOX

Conditions. Name : dummy Age/sex : xx Y /x. Lab No : xxxxxxxxx. Rep Centre : xxxxxxxxxxx Ref by : Dr. xxxxxxxxxx

E Ltd. P4 Test Report. Test Report. Sex: Report Print: Comment: Summary of Test Results. cortisol value lower than 5.02 ng/ml. Stroke have deficiency.

Exposure (To Stressors) Autism Spectrum Disorders

application of biomarkers to disease

The potential impact of toxicogenomics on modern chemical risk assessment 3-MCPD and 3-MCPD fatty acid esters as examples

Original Policy Date

National Burden of Cancers Attributable to Secondhand Smoking in Indonesia

Incorporating Gene-Environment Information into Kinetic Models: Lessons Learned and Future Challenges

Read Across with Metabolomics for Phenoxy Herbicides a Case Study with MCPP BASF SE

Effect of variation of ABCB1 and GSTP1 on osteosarcoma survival after chemotherapy

Types of Electrophilic Carcinogenic Intermediates

CS2220 Introduction to Computational Biology

Deliverable 2.1 List of relevant genetic variants for pre-emptive PGx testing

Pharmacokinetics for Physicians. Assoc Prof. Noel E. Cranswick Clinical Pharmacologist Royal Children s Hospital Melbourne

Section 5.3: Preclinical safety data

Asingle inherited mutant gene may be enough to

Health Effects of Passive Smoking

NexGen Risk Assessment

Detox Genomics. Client Name:

A Regional Model of Lung Metabolism for Improving Species Dependent Descriptions of 1,3-Butadiene and its Metabolites

A Weight of Evidence Approach to Cancer Assessment. Alan R Boobis Imperial College London

Variability Due to Genetic Differences

Transcription:

The use of biomarkers and genetic epidemiology Alan R Boobis Imperial College London a.boobis@imperial.ac.uk Understanding Epidemiology for Chemical Risk Assessment 6-8 October, 2010, Cranfield

Biomarkers Biomarkers of Susceptibility Exposure Internal Dose Biologically Effective Dose Early Biological Response Altered Structure/ Function Clinical disease Prognosis Biomarkers of Exposure Biomarkers of Effect

Types of biomarker Biomarkers of exposure: Measurements that indicate that exposure to a chemical or chemical class has occurred, but do not provide knowledge of adverse effects at the level of the organism Biomarkers of effect: Measurements that indicate that both exposure and adverse effects have occurred Biomarkers of susceptibility: Measurements used to assess an organism s inherent or acquired limitation to cope with a chemical exposure

Some examples of biomarkers Biomarkers of exposure Body burden Exhaled breath Blood or urinary levels Internal dose Blood metabolite levels Urinary metabolite levels Protein adducts Plasma cholinesterase inhibition Biologically-effective dose DNA adducts Biomarkers of effect Sister chromatid exchanges Micronuclei Chromosomal damage Red cell cholinesterase inhibition Urinary beta-2-microglobulin Biomarkers of susceptibility Breathing rate Genotype or phenotype P450 Glutathione S-transferase Epoxide hydrolase DNA repair enzymes These divisions are not exact (e.g. DNA adducts could be considered a biomarker of effect rather than of exposure, micronuclei could be considered a biomarker of exposure rather than of effect)

Source of materials and methodologies for assessment of biomarkers MATERIAL METHODOLOGY Biofluids urine, serum, saliva bronchoalveolar lavage Cells peripheral blood cells cell culture Tissue necropsy biopsy Chemical and biochemical analysis Biofluid MRS RT-PCR/Taq Man In situ hybridization MRI/MRS of tissue Microarrays 2-D gel electrophoresis Flow cytometry

Biomarkers of exposure The concentration of the substance of interest The concentration of a product of its biotransformation A biological (non-critical or incidental) effect of exposure, for example through interaction with a non-target molecule or cell A biomarker of exposure indicates that exposure has occurred, but does not provide knowledge of an adverse effect at the level of the organism

RISK MANAGEMENT Exposure-effect continuum for environmental chemicals Source Fate and Transport Water, Air, Food, Soil, Dust, Sediment, Surfaces, Personal Care Products Metabolism Elimination Exposure Internal Dose Target Organ Dose Absorption following: Distribution Elimination Inhalation Ingestion Dermal Contact RISK ASSESSMENT Angerer et al. Tox Sci 93: 3-10 (2006) Biologically Effective Dose Effect Pharmacodynamic Processes

Cotinine as a biomarker of exposure to cigarette smoke Cotinine is a metabolite of nicotine that tracks exposure to tobacco smoke In nonsmokers, tracks exposure to secondhand smoke N N CH 3 O

Percentage of the population Exposure of the US population to tobacco smoke: Serum cotinine levels 5 4 ETS exposure (nonsmokers) Smokers 3 2 1 0 0.1 1.0 10 100 1000 Serum cotinine (ng/ml) NHANES III, 1988-1991

Biomarkers of exposure Compound Biomarker Source Styrene Benzene Mandelic acid, Phenylglyoxylic acid Benzene Benzene metabolites Urine Blood, urine, breath Urine Chloroform Chloroform Breath, blood Dioxins Dioxins Blood Fat PAHs 1-Hydroxypyrene Urine Heterocyclic aromatic amines Parent compound and metabolites Urine

Biomarkers of exposure Compound Biomarker Source Arsenic Arsenic Urine Lead Lead Blood, bone Nicotine Cotinine Urine DDT DDE Serum Nitrosating agents N-nitrosoamino acids urine

Post-exposure fate of a persistent chemical in blood and urine Blood Toxicant/Metabolite Hemoglobin Adduct DNA Adduct Albumin Adduct Urinary Adduct Urinary Metabolite 1 10 100 1000 Time (Days) Needham and Sexton. JEAEE 10: 611-629 (2000) Adapted from: Henderson et al. Crit Rev Toxicol 20: 65-82 (1989)

Post-exposure fate of a non-persistent chemical in blood and urine Blood Toxicant/Metabolite Urinary Metabolite Albumin Adduct DNA Adduct Hemoglobin Adduct If chemical forms an adduct: extends time window of exposure Urinary Adduct 1 10 100 1000 Time (Days) Needham and Sexton. JEAEE 10: 611-629 (2000) Adapted from: Henderson et al. Crit Rev Toxicol 20: 65-82 (1989)

Potential value of biomarkers in exposure assessment Confirmation of more conventional approaches to exposure assessment Corroborate estimates of exposure Reduce misclassification of exposure Combined exposures to similar compounds Estimate of internal or target dose Development and validation of PBPK models Bridge between studies in experimental animals and observations in humans Interindiviudal variation in exposure Susceptible sub-populations

Potential limitations of biomarkers in exposure assessment Often restricted to recent exposures Intraindividual variation over time Relationship between biomarker and dose often unknown (importance of PK) Reduced study size relative to other exposure assessments Need for individual sampling Invasiveness of measure Analytical issues Increased potential for confounding

Biomarkers of effect A measurable biochemical, physiological, behavioural or other alteration within an organism that, depending upon the magnitude, can be recognized as associated with an established or possible health impairment or disease Biomarkers of effect are often a necessary, though usually not sufficient step in the pathological process towards disease

Example of a biomarker of genotoxicity Micronuclei (± centromere) formation in lymphocytes isolated from peripheral blood can be used for assessment of genotoxicity potential (cells are cultured for 3 days with cytochalasin B block of cytokinesis)

Cardiac troponin Gold standard biomarker of myocardial injury in man Myofibrillar protein regulating contraction that leeks from injured cardiac cells 3 subunits: C binds Ca 2, I inhibits actin and myosin interaction at low [Ca 2 +], T binds tropomyosin

Use of ctnt in experimental animals O Brien et al, 1997

Proposed biomarkers of renal toxicity (HESI) Proximal Tubule Distal Tubule -GST -GST Collecting Duct No specific nephronal location RPA-1 (renal papillary antigen-1) Clusterin

Fold Change from Control Fold Change from concurrent control Fold Change from Control 25 20 15 10 5 Creatinine BUN U Prot GGT NAG -GST clusterin -GST RPA-1 Relative performance of biomarkers for renal toxicity Cisplatin 3 mg/kg (Wistar) 8 7 6 5 4 3 2 1 Gentamicin 50 mg/kg/day (SD ) BUN CREATININE U_VOL U_GGT U_NAG U_ALP U_PROTEIN GST GST RPA CLUSTERIN OSMOLALITY 0 HESI Day 2 Day 3 Day 5 25 20 15 10 5 0 NPAA (N-phenylanthranilic acid) 500 mg/kg/day (Wistar) Day 3 Day 7 Day 14 0 Day 7 Urine Glucose Urine Protein ALP LDH NAG GGT GST GST RPA1 Clusterin

Potential value of biomarkers of effect Understanding mode or mechanism of action and its human relevance Definition of nature and shape of doseresponse relationship Extrapolation from experimental doses to levels of human exposure and possible dose transitions Mechanistic basis of thresholds or points of departure in dose-response curve

Potential limitations of biomarkers of effect Uncertainty in qualitative and quantitative relationship between biomarker and toxic response Specificity of response Temporal relationship between exposure of relevance and measurement of biomarker of effect Possible confounding Inter-species extrapolation

Biomarkers of susceptibility Biomarkers of susceptibility should reflect individual susceptible to disease Such biomarkers are usually genetically determined but may also be acquired Biomarkers of susceptibility are independent of environmental exposure Biomarkers of susceptibility may affect either internal dose or toxicant response

Genetic epidemiology Gene-environment interactions Where environment represents a chemical exposure Interaction (effect modification) occurs when the estimate of effect of exposure (environment) depends on the level of another factor (gene) within the population Interaction is distinct from confounding (or selection or information bias), but rather a real difference in the effect of exposure in various subgroups that may be of considerable interest

Gene-environment interaction Environment/ lifestyle Genetics Pre-clinical disease Clinical disease Outcome Quality of life Mortality

Gene-environment interactions Some genetic factors and environmental exposures can cause disease independently Genetic defect (BRCA 1,2) causes breast cancer Exposure to oestradiol causes breast cancer Some responses occur only with combination of certain genetic factors and environmental exposures Asthma? Autoimmune diseases? Some genetic factors modulate extent of internal exposure GST polymorphisms regulate activation/inactivation of some solvents Some genetic factors modulate response to environmental exposures herg polymorphisms determine QTc prolongation by some drugs Environmental exposures can modulate genetic disease Lead exposure exacerbates severity of acute intermittent porphyria

Examples of polymorphisms affecting susceptibility Variants of several P450 genes associated with increased lung cancer risk in smokers e.g. CYP1A1-10% of Caucasians have one such variant Another variant present only in African-Americans Deletion of one of glutathione S-transferase genes (GSTM1) associated with increased risk of bladder and lung cancer from exposure to several toxic substances (e.g., PAHs, aflatoxin) 50% of Caucasians carry deletion

Example: Cancer (SMRs) Not U.S. U.S. Born U.S. Cauc Cancer Japan Born Stomach (M) 100 72 38 17 Intestine (F) 100 218 209 483 Breast (F) 100 166 136 591 (MacMahon B, Pugh TF. Epidemiology: Principles and Methods. Boston: Little, Brown and Co, 1970:178.)

Genetic polymorphisms: environmental susceptibility Many genetic polymorphisms identified affect response to xenobiotics Altered kinetics or dynamics e.g. genes affecting metabolism, detoxication, DNA repair, receptors, cell cycle control, etc. Many genes increase risk from exposure, but some have protective effect Environmental Genome Project has identified ~ 600 genes with alleles potentially exhibiting differential responses to environmental exposures

Genetic polymorphisms of some P450 enzymes Enzyme Major Effect % of PM mutation CYP1B1 Val 432 Leu Defective enzyme ~10% CYP2A6 Leu 160 His Defective enzyme <1% CYP2C9 Arg 144 Cys Altered specificity ~5% CYP2C19 Cryptic splice No enzyme 3% site in exon 5 CYP2D6 Mutation at intron 4/ No enzyme 8% exon 5 stop codon CYP2B6 Several Reduced enzyme ~75% CYP3A5 Mutation in intron 3 Reduced enzyme ~70% stop codon

Distribution of PMs for CYP2D6 3% 1% 0.5% 0% 6% 9% 6% 7% 10% 1% 2% 4% 7% 2%

Case Study: N-acetyltransferases (NAT) Two genes, NAT1 and NAT2 Catalytic transfer of acetyl group from donor (Acetyl-Co-A) to substrate (amine) Both are polymorphic, with multiple different allelic variants Phenotype associated with slow acetylation NAT1 / NAT2

Gene-environment interactions Polymorphism (NAT2) Slow acetylation Fast acetylation Bladder cancer relative risk Bladder cancer relative risk associated with smoking = 8 associated with smoking = 1.5 Interaction - effect of smoking is different in subjects with a specific polymorphic gene Better understanding of the effects of established toxicants Uncover low levels of risk previously masked by genetic heterogeneity NB: Data are for illustrative purposes only

Modulation of DNA adducts in smokers by genetic polymorphisms DNA adducts in relation to Genotype - / slow + / fast p GSTM1 1.30 1.03 0.05 GSTT1 1.28 1.12 0.41 NAT1 1.58 1.11 0.05 NAT2 1.29 1.03 0.06 Combinations of 3 genotypes GSTM1 -, NAT1 slow, NAT2 slow 2.03 GSTM1 +, NAT1 fast, NAT2 fast 0.91 Godschalk et al (2001)

Variability and toxic response: GST and tobacco smoke Glutathione S-transferase (GST) Metabolic enzyme Participates in metabolic detoxication of benzo(a)pyrene GST null donors are more sensitive to the induction of chromosomal aberrations due to tobacco smoke GST null also has been linked to increased risk of lung and bladder cancer in smokers Knowledge of genotype may impact behaviour Biomarker of susceptibility Norppa (2003). Mut Res

Interactions between smoking and GSTM1 6 5 4 (Adjusted odds ratios and 95% confidence intervals) 3.2 (1.6, 6.3) 6.5 (3.3, 13.0) 3 2 1 0 1.00 Never/ Positive 1.2 (0.7, 2.2) Never/ Null Ever/ Positive Ever/ Null Stucker et al. (1999), Int Epidmiol Assoc

Gene-environment interaction and lung cancer risk Adduct level GSTM1 OR Low present 1.0 Low null 2.3 High present 6.9 High null 16.2 Tang et al., Carcinogenesis 1998

Carcinogen-DNA adducts and cancer risk Study design: Type of cancer Risk Reference Cohort (smokers only) Lung 1.22 H. Bak, poster Porvoo Cohort (current smokers) Lung 2.98 Perera et al, 2002 Case-control Bladder 1.9 Benhamou et al, 2003 Case-control Breast (PAH) 1.97 Rundle et al, 2002 Case-control Breast (PhIP) 4.03 Zhu et al, 2003 Conclusion: Carcinogen-DNA adducts appear to be a risk indicator for cancer, especially in smokers

Functional and non-functional polymorphisms Functional polymorphism - a change in the DNA sequence results in a change in the expression or function of the protein, e.g. introduction of a premature termination codon Non-functional polymorphism - a change in the DNA sequence that has no effect on the expression or activity of the protein, e.g. change in DNA sequence within an intron

Importance of understanding functional consequence of a polymorphism (PON1) Cherry et al (2002). Lancet 359, 763-764. Cases were more likely than referents to have at least one R allele at position 192 (Gln to Arg substitution); odds ratio 1.93 (95% CI 1.24-3.01) More likely to to have diazoxonase activity below normal median (1.77, 1.18-2.67) Our results support the hypothesis that organophosphates contribute to the reported ill health of people who dip sheep Mackness et al (2003). Pharmacogenetics 13, 81-88. Farmers reporting chronic ill health due to organophosphate exposure have a higher proportion of the PON1-192R polymorphism This was associated with lower rates of diazoxon hydrolysis than the controls their ill health may be explained by a lower ability to detoxify diazoxon

Activity (U/ml) Activity (U/ml) Activity (U/ml) Interindividual variability in PON1 activity towards diazoxon 40 30 20 10 40 0 TT TC CC PON1-108 genotype 40 QQ QR RR 30 30 20 20 10 10 0 LueLeu LeuMet MetMet 0 GlnGln GlnArg ArgArg PON1 55 genotypes PON1 192 Genotype

SNP selection for association studies - Regulatory / Functional SNPs - Yuan (2006) FastSNP

Sample size requirement for geneenvironment interaction studies RR Hunter (2005)

Breast cancer case-only analysis of the effect of tobacco smoke exposure Genetic Never Passive Former Current Polymorphism (N) (N) (N) (N) CYP 1B1 Any Val 41 52 44 55 Leu/Leu 21 39 17 13 ORi (95%CI) 1 0.69 (0.35-1.37) 1.33 (0.59-2.96) 2.32 (1.00-5.38) SULT1A1 Arg/Arg 36 51 38 25 Any His 26 40 23 43 ORi (95%CI) 1 1.08 (0.55-2.11) 0.79 (0.37-1.68) 2.55 (1.21-5.36) COMT VaL/Val 22 26 19 18 Any Met 40 65 42 50 ORi (95%CI) 1 1.26 (0.62-2.57) 1.07 (0.49-2.35) 1.42 (0.65-3.13) (Nov, 2003)

Development of novel biomarkers Unique opportunities exist for the development of new biomarkers on the basis of genomics, proteomics, metabonomics, etc Ideally they should be mechanistically-related Initial development in animal models, and using a combination of in vitro and in vivo approaches Critical to the application of biomarkers is their validation

Expression platforms Transcriptomics Microarrays Proteomics Chromatography, 2D gel electrophoresis, mass spectrometry Protein arrays, yeast 2-hybrid screen Metabonomics Chromatography, nuclear magnetic resonance (NMR), mass spectrometry

-Omics in the search for novel biomarkers Top down, rather than bottom up, approach Possible identification of completely novel biomarkers Biomarkers comprising suites of analytes? Knowledge of the human genome informs choice and development of biomarkers Validation is a major issue Relevance, specificity, reproducibility,..

Genetic markers in the HLA-B region associated with abacavir hypersensitivity Roses (2002)

Biomarkers in epidemiology Genetic factors can contribute to disease risk from environmental chemicals Identified effects to date have generally been relatively modest Need for improved exposure assessment Statistical study design issues when exploring multiple polymorphisms Potential application of biomarkers of exposure and of effect New generation of biomarkers under development how should they be validated

Further information Smolders R et al (2009). J. Toxicol. Environ. Health, Part B,12:107-123 Wagner JA (2008). Annu. Rev. Pharmacol. Toxicol. 48:631-651 Altar CA et al (2008). Clin. Pharmac. Ther. 83:368-471