Toxicogenomics

Last updated

Toxicogenomics is a subdiscipline of pharmacology that deals with the collection, interpretation, and storage of information about gene and protein activity within a particular cell or tissue of an organism in response to exposure to toxic substances. Toxicogenomics combines toxicology with genomics or other high-throughput molecular profiling technologies such as transcriptomics, proteomics and metabolomics. [1] [2] Toxicogenomics endeavors to elucidate the molecular mechanisms evolved in the expression of toxicity, and to derive molecular expression patterns (i.e., molecular biomarkers) that predict toxicity or the genetic susceptibility to it.

Contents

Pharmaceutical research

In pharmaceutical research, toxicogenomics is defined as the study of the structure and function of the genome as it responds to adverse xenobiotic exposure. It is the toxicological subdiscipline of pharmacogenomics, which is broadly defined as the study of inter-individual variations in whole-genome or candidate gene single-nucleotide polymorphism maps, haplotype markers, and alterations in gene expression that might correlate with drug responses. [3] [4] Though the term toxicogenomics first appeared in the literature in 1999, [5] it was by that time already in common use within the pharmaceutical industry as its origin was driven by marketing strategies from vendor companies. The term is still not universally accepted, and others have offered alternative terms such as chemogenomics to describe essentially the same field of study. [6]

Bioinformatics

The nature and complexity of the data (in volume and variability) demands highly developed processes of automated handling and storage. The analysis usually involves a wide array of bioinformatics and statistics, [7] often including statistical classification approaches. [8]

Drug discovery

In pharmaceutical drug discovery and development, toxicogenomics is used to study possible adverse (i.e. toxic) effects of pharmaceutical drugs in defined model systems in order to draw conclusions on the toxic risk to patients or the environment. Both the United States Environmental Protection Agency (EPA) and the Food and Drug Administration (FDA) currently preclude basing regulatory decision-making on genomics data alone. However, they do encourage the voluntary submission of well-documented, quality genomics data. Both agencies are considering the use of submitted data on a case-by-case basis for assessment purposes (e.g., to help elucidate mechanism of action or contribute to a weight-of-evidence approach) or for populating relevant comparative databases by encouraging parallel submissions of genomics data and traditional toxicological test results. [9]

Public projects

Chemical Effects in Biological Systems is a project hosted by the National Institute of Environmental Health Sciences building a knowledge base of toxicology studies including study design, clinical pathology, and histopathology and toxicogenomics data. [10] [11]

InnoMed PredTox assesses the value of combining results from various omics technologies together with the results from more conventional toxicology methods in more informed decision-making in preclinical safety evaluation. [12]

Open TG-GATEs (Toxicogenomics Project-Genomics Assisted Toxicity Evaluation System) is a Japanese public-private effort which has published gene expression and pathology information for more than 170 compounds (mostly drugs). [13]

The Predictive Safety Testing Consortium aims to identify and clinically qualify safety biomarkers for regulatory use as part of the FDA's "Critical Path Initiative". [12]

ToxCast is a program for Predicting Hazard, Characterizing Toxicity Pathways, and Prioritizing the Toxicity Testing of Environmental Chemicals at the United States Environmental Protection Agency. [14]

Tox21 is a federal collaboration involving the National Institutes of Health (NIH), Environmental Protection Agency (EPA), and Food and Drug Administration (FDA), is aimed at developing better toxicity assessment methods. [15] Within this project the toxic effects of chemical compounds on cell lines derived from the 1000 Genomes Project individuals were assessed and associations with genetic markers were determined. [16] Parts of this data were used in the NIEHS-NCATS-UNC DREAM Toxicogenetics Challenge in order to determine methods for cytotoxicity predictions for individuals. [17] [18]

See also

Related Research Articles

Pharmacology Branch of biology concerning drugs

Pharmacology is a branch of medicine, biology and pharmaceutical sciences concerned with drug or medication action, where a drug may be defined as any artificial, natural, or endogenous molecule which exerts a biochemical or physiological effect on the cell, tissue, organ, or organism. More specifically, it is the study of the interactions that occur between a living organism and chemicals that affect normal or abnormal biochemical function. If substances have medicinal properties, they are considered pharmaceuticals.

Toxicology Study of substances harmful to living organisms

Toxicology is a scientific discipline, overlapping with biology, chemistry, pharmacology, and medicine, that involves the study of the adverse effects of chemical substances on living organisms and the practice of diagnosing and treating exposures to toxins and toxicants. The relationship between dose and its effects on the exposed organism is of high significance in toxicology. Factors that influence chemical toxicity include the dosage, duration of exposure, route of exposure, species, age, sex, and environment. Toxicologists are experts on poisons and poisoning. There is a movement for evidence-based toxicology as part of the larger movement towards evidence-based practices. Toxicology is currently contributing to the field of Cancer research, since some toxins can be used as drugs for killing tumor cells. One prime example of this is Ribosome Inactivating Proteins, tested in the treatment of Leukemia.

Perfluorooctanesulfonic acid Fluorosurfactant and persistent organic pollutant

Perfluorooctanesulfonic acid (PFOS) is an anthropogenic (human-made) fluorosurfactant, now regarded as a global pollutant. PFOS was the key ingredient in Scotchgard, a fabric protector made by 3M, and numerous stain repellents. It was added to Annex B of the Stockholm Convention on Persistent Organic Pollutants in May 2009. PFOS can be synthesized in industrial production or result from the degradation of precursors. PFOS levels that have been detected in wildlife are considered high enough to affect health parameters, and recently higher serum levels of PFOS were found to be associated with increased risk of chronic kidney disease in the general US population. "This association was independent of confounders such as age, sex, race/ethnicity, body mass index, diabetes, hypertension, and serum cholesterol level."

Endocrine disruptor Chemicals that can interfere with endocrine or hormonal systems

Endocrine disruptors, sometimes also referred to as hormonally active agents, endocrine disrupting chemicals, or endocrine disrupting compounds are chemicals that can interfere with endocrine systems. These disruptions can cause cancerous tumors, birth defects, and other developmental disorders. Found in many household and industrial products, endocrine disruptors "interfere with the synthesis, secretion, transport, binding, action, or elimination of natural hormones in the body that are responsible for development, behavior, fertility, and maintenance of homeostasis ."

Pharmacogenomics Study of the role of the genome in drug response

Pharmacogenomics is the study of the role of the genome in drug response. Its name reflects its combining of pharmacology and genomics. Pharmacogenomics analyzes how the genetic makeup of an individual affects their response to drugs. It deals with the influence of acquired and inherited genetic variation on drug response in patients by correlating gene expression or single-nucleotide polymorphisms with pharmacokinetics and pharmacodynamics. The term pharmacogenomics is often used interchangeably with pharmacogenetics. Although both terms relate to drug response based on genetic influences, pharmacogenetics focuses on single drug-gene interactions, while pharmacogenomics encompasses a more genome-wide association approach, incorporating genomics and epigenetics while dealing with the effects of multiple genes on drug response.

Personalized medicine Medical model that tailors medical practices to the individual patient

Personalized medicine, also referred to as precision medicine, is a medical model that separates people into different groups—with medical decisions, practices, interventions and/or products being tailored to the individual patient based on their predicted response or risk of disease. The terms personalized medicine, precision medicine, stratified medicine and P4 medicine are used interchangeably to describe this concept though some authors and organisations use these expressions separately to indicate particular nuances.

Thiopurine methyltransferase

Thiopurine methyltransferase or thiopurine S-methyltransferase (TPMT) is an enzyme that in humans is encoded by the TPMT gene. A pseudogene for this locus is located on chromosome 18q.

KEGG Collection of bioinformatics databases

KEGG is a collection of databases dealing with genomes, biological pathways, diseases, drugs, and chemical substances. KEGG is utilized for bioinformatics research and education, including data analysis in genomics, metagenomics, metabolomics and other omics studies, modeling and simulation in systems biology, and translational research in drug development.

AS3MT

Arsenite methyltransferase is an enzyme that in humans is encoded by the AS3MT gene.

The Comparative Toxicogenomics Database (CTD) is a public website and research tool launched in November 2004 that curates scientific data describing relationships between chemicals/drugs, genes/proteins, diseases, taxa, phenotypes, GO annotations, pathways, and interaction modules. The database is maintained by the Department of Biological Sciences at North Carolina State University.

The Hazardous Substances Data Bank (HSDB) is a toxicology database on the U.S. National Library of Medicine's (NLM) Toxicology Data Network (TOXNET). It focuses on the toxicology of potentially hazardous chemicals, and includes information on human exposure, industrial hygiene, emergency handling procedures, environmental fate, regulatory requirements, and related areas. All data are referenced and derived from a core set of books, government documents, technical reports, and selected primary journal literature. Prior to 2020, all entries were peer-reviewed by a Scientific Review Panel (SRP), members of which represented a spectrum of professions and interests. Last Chairs of the SRP are Dr. Marcel J. Cassavant, MD, Toxicology Group, and Dr. Roland Everett Langford, PhD, Environmental Fate Group. The SRP was terminated due to budget cuts and realignment of the NLM.

Critical Path Institute (C-Path) is a non-profit organization created to improve the drug development process; its consortia include more than 1,600 scientists from government regulatory and research agencies, academia, patient organizations, and bio-pharmaceutical companies.

The Environmental Mutagenesis and Genomics Society (EMGS) is a scientific society "for the promotion of critical scientific knowledge and research into the causes and consequences of damage to the genome and epigenome in order to inform and support national and international efforts to ensure a healthy, sustainable environment for future generations."

ArrayTrack

ArrayTrack is a multi-purpose bioinformatics tool primarily used for microarray data management, analysis, and interpretation. ArrayTrack was developed to support in-house filter array research for the U.S. Food and Drug Administration in 2001 and was made freely available to the public as an integrated research tool for microarrays in 2003. Since then, ArrayTrack has averaged about 5,000 users per year. It is regularly updated by the National Center for Toxicological Research.

The Klimisch score is a method of assessing the reliability of toxicological studies, mainly for regulatory purposes, that was proposed by H.J. Klimisch, M. Andreae and U. Tillmann of the chemical company BASF in 1997 in a paper entitled A Systematic Approach for Evaluating the Quality of Experimental Toxicological and Ecotoxicological Data which was published in Regulatory Toxicology and Pharmacology. It assigns studies to one of four categories as follows:

The non-profit Evidence-based Toxicology Collaboration (EBTC) comprises a group of scientists and experts with ties to governmental and non-governmental agencies, chemical and pharmaceutical companies, and academia that have banded together to promote the use of what are known as "evidence-based approaches" in toxicology. The discipline of evidence-based toxicology (EBT) is a process for transparently, consistently, and objectively assessing available scientific evidence in order to answer questions in toxicology. EBT has the potential to address concerns in the toxicological community about the limitations of current approaches. These include concerns related to transparency in decision making, synthesis of different types of evidence, and the assessment of bias and credibility. The evidence-based methods and approaches now being proposed for toxicology are widely used in medicine, which is the basis for their nomenclature. The need to improve how the performance of toxicological test methods is assessed was the main impetus for translating these tools to toxicology.

The Pharmacogenomics Knowledge Base (PharmGKB) is a publicly available, online knowledge base responsible for the aggregation, curation, integration and dissemination of knowledge regarding the impact of human genetic variation on drug response. It is funded by the National Institutes of Health (NIH) National Institute of General Medical Sciences (NIGMS), and is a partner of the NIH Pharmacogenomics Research Network (PGRN). It has been managed at Stanford University since its inception in 2000.

Bisphenol F is an organic compound with the chemical formula (HOC
6
H
4
)
2
CH
2
. It is structurally related to bisphenol A, a popular and controversial plasticizer, as both belong to the category of molecules known as bisphenols, which feature two phenol groups connected via a linking group. In BPF, the two aromatic rings are linked by a methylene connecting group. In response to the BPA controversy, BPF is finding increased use.

CompTox Chemicals Dashboard Chemical database

The CompTox Chemicals Dashboard is a freely accessible online database created and maintained by the U.S. Environmental Protection Agency (EPA). The database provides access to multiple types of data including physicochemical properties, environmental fate and transport, exposure, usage, in vivo toxicity, and in vitro bioassay. EPA and other scientists use the data and models contained within the dashboard to help identify chemicals that require further testing and reduce the use of animals in chemical testing. The Dashboard is also used to provide public access to information from EPA Action Plans, e.g. around perfluorinated alkylated substances.

Cancer pharmacogenomics

Cancer pharmacogenomics is the study of how variances in the genome influences an individual’s response to different cancer drug treatments. It is a subset of the broader field of pharmacogenomics, which is the area of study aimed at understanding how genetic variants influence drug efficacy and toxicity.

References

  1. National Research Council (US) Committee on Communicating Toxicogenomics Information to Nonexperts (2005). Communicating Toxicogenomics Information to Nonexperts: A Workshop Summary. The National Academies Press. doi:10.17226/11179. ISBN   978-0-309-09538-9. PMID   20669449.
  2. Hamadeh HK, Afshari CA, eds. (2004). Toxicogenomics: Principles and Applications. Hoboken, NJ: Wiley-Liss. ISBN   0-471-43417-5.
    Omenn GS (November 2004). "Book Review: Toxicogenomics: Principles and Applications". Environ. Health Perspect. 112 (16): A962. doi:10.1289/ehp.112-1247673. PMC   1247673 .
  3. Lesko LJ, Woodcock J (2004). "Translation of pharmacogenomics and pharmacogenetics: a regulatory perspective". Nature Reviews. Drug Discovery. 3 (9): 763–9. doi:10.1038/nrd1499. PMID   15340386. S2CID   33145913.
  4. Lesko LJ, Salerno RA, Spear BB, Anderson DC, Anderson T, Brazell C, Collins J, Dorner A, Essayan D, Gomez-Mancilla B, Hackett J, Huang SM, Ide S, Killinger J, Leighton J, Mansfield E, Meyer R, Ryan SG, Schmith V, Shaw P, Sistare F, Watson M, Worobec A (2003). "Pharmacogenetics and pharmacogenomics in drug development and regulatory decision making: report of the first FDA-PWG-PhRMA-DruSafe Workshop". Journal of Clinical Pharmacology. 43 (4): 342–58. doi:10.1177/0091270003252244. PMID   12723455. S2CID   5902873.
  5. Nuwaysir EF, Bittner M, Trent J, Barrett JC, Afshari CA (1999). "Microarrays and toxicology: the advent of toxicogenomics". Molecular Carcinogenesis. 24 (3): 153–9. doi:10.1002/(SICI)1098-2744(199903)24:3<153::AID-MC1>3.0.CO;2-P. PMID   10204799.
  6. Fielden MR, Pearson C, Brennan R, Kolaja KL (2005). "Preclinical drug safety analysis by chemogenomic profiling in the liver". American Journal of Pharmacogenomics. 5 (3): 161–71. doi:10.2165/00129785-200505030-00003. PMID   15952870. S2CID   24834075.
  7. Mattes WB, Pettit SD, Sansone SA, Bushel PR, Waters MD (March 2004). "Database development in toxicogenomics: issues and efforts". Environmental Health Perspectives. 112 (4): 495–505. doi:10.1289/ehp.6697. PMC   1241904 . PMID   15033600. Archived from the original on 2008-07-04.
  8. Ellinger-Ziegelbauer H, Gmuender H, Bandenburg A, Ahr HJ (January 2008). "Prediction of a carcinogenic potential of rat hepatocarcinogens using toxicogenomics analysis of short-term in vivo studies". Mutation Research. 637 (1–2): 23–39. doi:10.1016/j.mrfmmm.2007.06.010. PMID   17689568.
  9. Corvi R, Ahr HJ, Albertini S, Blakey DH, Clerici L, Coecke S, Douglas GR, Gribaldo L, Groten JP, Haase B, Hamernik K, Hartung T, Inoue T, Indans I, Maurici D, Orphanides G, Rembges D, Sansone SA, Snape JR, Toda E, Tong W, van Delft JH, Weis B, Schechtman LM (March 2006). "Meeting report: Validation of toxicogenomics-based test systems: ECVAM-ICCVAM/NICEATM considerations for regulatory use". Environmental Health Perspectives. 114 (3): 420–9. doi:10.1289/ehp.8247. PMC   1392237 . PMID   16507466. Archived from the original on 2008-10-17.
  10. Waters MD, Fostel JM (2004). "Toxicogenomics and systems toxicology: aims and prospects". Nature Reviews Genetics. 5 (12): 938–948. doi:10.1038/nrg1493. PMID   15573125. S2CID   1368324.
  11. Collins BC, Clarke A, Kitteringham NR, Gallagher WM, Pennington SR (October 2007). "Use of proteomics for the discovery of early markers of drug toxicity". Expert Opinion on Drug Metabolism & Toxicology. 3 (5): 689–704. doi:10.1517/17425255.3.5.689. PMID   17916055. S2CID   27489795.
  12. 1 2 Mattes WB (2008). "Public Consortium Efforts in Toxicogenomics". In Mendrick DL, Mattes WB (eds.). Essential Concepts in Toxicogenomics. Methods in Molecular Biology. 460. pp. 221–238. doi:10.1007/978-1-60327-048-9_11. ISBN   978-1-58829-638-2. PMID   18449490.
  13. Igarashi Y, Nakatsu N, Yamashita T, Ono A, Ohno Y, Urushidani T, Yamada H (January 2015). "Open TG-GATEs: a large-scale toxicogenomics database". Nucleic Acids Research. 43 (Database issue): D921–7. doi:10.1093/nar/gku955. PMC   4384023 . PMID   25313160.
  14. Dix DJ, Houck KA, Martin MT, Richard AM, Setzer RW, Kavlock RJ (January 2007). "The ToxCast program for prioritizing toxicity testing of environmental chemicals". Toxicological Sciences. 95 (1): 5–12. doi: 10.1093/toxsci/kfl103 . PMID   16963515.
  15. "Toxicology in the 21st century project". www.ncats.nih.gov.
  16. Abdo N, Xia M, Brown CC, Kosyk O, Huang R, Sakamuru S, Zhou YH, Jack JR, Gallins P, Xia K, Li Y, Chiu WA, Motsinger-Reif AA, Austin CP, Tice RR, Rusyn I, Wright FA (May 2015). "Population-based in vitro hazard and concentration-response assessment of chemicals: the 1000 genomes high-throughput screening study". Environmental Health Perspectives. 123 (5): 458–66. doi:10.1289/ehp.1408775. PMC   4421772 . PMID   25622337.
  17. "NIEHS-NCATS-UNC-DREAM Toxicogenetics Challenge". Sage Bionetworks.
  18. "DeepTox: Deep Learning for Toxicity Prediction". Institute of Bioinformatics, Johannes Kepler University Linz.