Metabolomics approaches for discovering biomarkers of drug-induced hepatotoxicity and nephrotoxicity

https://doi.org/10.1016/j.taap.2009.11.019Get rights and content

Abstract

Hepatotoxicity and nephrotoxicity are two major reasons that drugs are withdrawn post-market, and hence it is of major concern to both the FDA and pharmaceutical companies. The number of cases of serious adverse effects (SAEs) in marketed drugs has climbed faster than the number of total drug prescriptions issued. In some cases, preclinical animal studies fail to identify the potential toxicity of a new chemical entity (NCE) under development. The current clinical chemistry biomarkers of liver and kidney injury are inadequate in terms of sensitivity and/or specificity, prompting the need to discover new translational specific biomarkers of organ injury. Metabolomics along with genomics and proteomics technologies have the capability of providing translational diagnostic and prognostic biomarkers specific for early stages of liver and kidney injury. Metabolomics has several advantages over the other omics platforms such as ease of sample preparation, data acquisition and use of biofluids collected through minimally invasive procedures in preclinical and clinical studies. The metabolomics platform is reviewed with particular emphasis on applications involving drug-induced hepatotoxicity and nephrotoxicity. Analytical platforms for metabolomics, chemometrics for mining metabolomics data and the applications of the metabolomics technologies are covered in detail with emphasis on recent work in the field.

Introduction

New drugs can be approved by the Food and Drug Administration (FDA) after passing a series of clinical trials that demonstrate therapeutic efficacy and lack of serious adverse effects. During clinical trials, many of the serious side effects occur as a result of liver or kidney injury. One reason for the late detection of serous adverse effects induced by drugs is the lack of efficient and accurate injury biomarkers that can translate from the preclinical studies to clinical studies. Novel and specific translational biomarkers of injury in the liver and kidney induced by drug toxicity are therefore demanded that will reduce the time and cost associated with drug discovery and approval.

Clinical trials typically assess serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), and bilirubin levels to diagnose liver injuries and blood urea nitrogen (BUN) and creatinine to monitor acute kidney injury. In the case of BUN (Tilson and Moser, 1992, Bleecker, 2000, Adamcova et al., 2005, Vaidya and Bonventre, 2006) and creatinine, these clinical biomarkers do not deviate from normal levels until significant renal injury has already occurred (Espandiari et al., 2007, Zhou et al., 2008). Other biomarkers are either non-specific or inaccurate. ALT is a biomarker for hepatocyte injury, but its blood level can increase for a variety of reasons (Lewis, 1984, Sherman, 1991, Zeisel et al., 1991, Vaidya and Bonventre, 2006); its value alone therefore is not a reliable biomarker for liver injury (Amacher, 1998, Dufour et al., 2000, Kaplowitz, 2001, Kaplowitz, 2005, Dhami et al., 1979, Yamada et al., 1984a, Yamada et al., 1984b, Waner and Nyska, 1991). Therefore, the pharmaceutical industry and regulatory agencies have invested much effort toward discovering novel biomarkers that are organ specific and benefit both preclinical and clinical studies.

Preclinical studies can determine some adverse effects before investigational new drug (IND) submissions but many drug candidates enter clinical trials and subsequently cause adverse effects. The major reason for ending a drug in phase I trials is toxicity (Suter et al., 2004), but toxicity is still a reason for stopping drugs in phase II and phase III clinical trials. After a drug hits the market, toxicity is the major reason a drug is removed from the shelf or given a black box warning label. The fact that the number of serious adverse effects is growing faster than the number of prescriptions (Moore et al., 2007) clearly indicates there is a failure of animal safety studies to translate to human safety. The prevalence and associated costs of the failure of a drug candidate during the clinical trials and post-market significantly increase the cost of the drug development expenditure (Lazarou et al., 1998, Pirmohamed et al., 2004). Therefore, there is need for new efficient biomarkers that can translate effectively from preclinical trials to human studies. Additionally, personalized medicine biomarkers can be used to predict whether that individual will respond favorably or adversely to a drug or medical therapy. Metabolomics can play a role in providing translational biomarkers for organ injury and for predicting a susceptible subset of the population that should avoid specific drugs.

Global metabolic profiling is an emerging technology offering promise for identifying early toxicity biomarkers that are specific indicators of damage to a particular organ. The term global metabolic profiling encompasses both metabolomics and metabonomics studies, which have slightly different definitions. Metabolomics refers to the measurement of the metabolite pool that exists within a cell under a particular set of conditions (Fiehn, 2002) while metabonomics describes “the quantitative measurement of the dynamic multiparametric metabolic response of living systems to pathophysiological stimuli or genetic modification” (Nicholson et al., 1999). Metabolomics and metabonomics are metabolic profiling methods that offer the opportunity to identify biomarkers or patterns of biomarker changes related to drug toxicity in biofluid samples, such as urine and blood, which can be collected with relative ease. The overall pattern of metabolic biomarkers is expected to differ based upon the origin of the toxicity, specifically the organ that is targeted by a particular drug compound (Robertson et al., 2000, Lindon et al., 2005a, Lindon et al., 2005b, Ebbels et al., 2007, Nicholson et al., 2007). The terms metabolic profiling, metabolomics and metabonomics have been increasingly used interchangeably throughout the literature and this review uses all these terms at specific points because the titles of the manuscripts we are referencing used only one of these terms. We use metabolic profiling in giving general statements about metabonomics and metabolomics.

Section snippets

Metabolomics procedures

A general flow chart showing the logistical steps necessary for conducting metabolomics studies is displayed in Fig. 1. The first step is sample generation from a toxicity study. A well-planned toxicity study with multiple doses, multiple time points for sample collection and preparation are very important; if performed improperly the data generated could be invalid. Protocols for sample generation and collection need to be carefully designed before the study begins, so effects from factors

Metabolomics in drug toxicity

One of the major research areas that metabolic profiling will continue to impact is the investigation of drug toxicity (Nicholson et al., 2002, Ebbels et al., 2003, Heijne et al., 2005, Robertson, 2005, Portilla et al., 2006, Schnackenberg et al., 2006). There is a strong need to develop new biomarkers that can accurately predict toxicity in the preclinical development of new chemical entities (NCEs) early in the drug development process and are translatable to the clinic. Metabolic profiling

Importance of understanding phenotypic factors in drug toxicity studies

A better understanding of gene–environment interactions is needed for personalized medicine to be successful. Fig. 6 shows how genes and the environment interact with transcriptomics, proteomics, and metabolomics. The end product of transcriptomics is metabolites. Genetics relates the inherited genetic DNA of a patient with their health status and responses to medical treatments. Transcriptomics evaluates the level of mRNA expression, proteomics investigates alterations in proteins, and

Conclusions

Metabolomics has shown the potential to add physiological and metabolic pathway information to drug toxicity studies. Initially, NMR was the primary analytical technique used in metabolomics studies because of its ease of use, inherent quantitative ability and high reproducibility, but lately many groups have used LC/MS or GC/MS because of their greater sensitivity and selectivity capabilities. In the future, we expect that both NMR and MS analyses will be used in metabolomics studies as this

Acknowledgments and disclaimer

The views presented in this article do not necessarily reflect those of the U.S. Food and Drug Administration.

References (153)

  • GriffinJ.L.

    Metabonomics: NMR spectroscopy and pattern recognition analysis of body fluids and tissues for characterisation of xenobiotic toxicity and disease diagnosis

    Curr. Opin. Chem. Biol.

    (2003)
  • GriffinJ.L. et al.

    Cellular environment of metabolites and a metabonomic study of tamoxifen in endometrial cells using gradient high resolution magic angle spinning 1H NMR spectroscopy

    Biochim. Biophys. Acta

    (2003)
  • HolmesE. et al.

    The application of NMR-based metabonomics in neurological disorders

    NeuroRx

    (2006)
  • HolmesE. et al.

    Metabolomic phenotyping in health and disease

    Cell

    (2008)
  • KeunH.

    Metabonomic modeling of drug toxicity

    Pharmacol. Ther.

    (2006)
  • LenzE.M. et al.

    Cyclosporin A-induced changes in endogenous metabolites in rat urine: a metabonomic investigation using high field 1H NMR spectroscopy, HPLC-TOF/MS and chemometrics

    J. Pharm. Biomed. Anal.

    (2004)
  • LindonJ.C. et al.

    Contemporary issues in toxicology the role of metabonomics in toxicology and its evaluation by the COMET project

    Toxicol. Appl. Pharmacol.

    (2003)
  • AdamcovaM. et al.

    Troponin as a marker of myocardiac damage in drug-induced cardiotoxicity

    Expert Opin. Drug Saf.

    (2005)
  • AmarP.J. et al.

    Acetaminophen safety and hepatotoxicity—where do we go from here?

    Expert Opin. Drug Saf.

    (2007)
  • AssfalgM. et al.

    Evidence of different metabolic phenotypes in humans

    PNAS

    (2008)
  • BaileyW.J. et al.

    Molecular profiling approaches for identifying novel biomarkers

    Expert Opin. Drug Saf.

    (2004)
  • Beckwith-HallB.M. et al.

    Nuclear magnetic resonance spectroscopic and principal components analysis investigations into biochemical effects of three model hepatotoxins

    Chem. Res. Toxicol.

    (1998)
  • BegerR.D. et al.

    Metabonomic models of human pancreatic cancer using 1D proton NMR spectra of lipids in plasma

    Metabolomics

    (2006)
  • BegerR.D. et al.

    Single valproic acid treatment inhibits glycogen and RNA ribose turnover while disrupting glucose-derived cholesterol synthesis in liver as revealed by the (U-13C6)-d-glucose tracer in mice

    Metabolomics

    (2009)
  • BerniniP. et al.

    Individual human phenotypes in metabolic space and time

    J. Prot. Res.

    (2009)
  • BertramH.C. et al.

    Anal. Chem.

    (2007)
  • BessemsJ.G.M. et al.

    Paracetamol (Acetaminophen)-induced toxicity: molecular and biochemical mechanisms, analogues, and protective approaches

    Crit. Rev. Toxicol.

    (2001)
  • BollardM.E. et al.

    NMR-based metabonomic approaches for evaluating physiological influences on biofluid composition

    NMR Biomed.

    (2005)
  • BoudonckK.J. et al.

    Discovery of metabolomics biomarkers for early detection of nephrotoxicity

    Toxicol. Pathol.

    (2009)
  • BrazmaA. et al.

    Minimum information about a microarray experiment (MIAME)-toward standards for microarray data

    Nat. Genet.

    (2001)
  • CarrieriD. et al.

    Identification and quantification of water-soluble metabolites by cryoprobe-assisted nuclear magnetic resonance spectroscopy applied to microbial fermentation

    Magn. Reson. Chem.

    (2009)
  • ChenP. et al.

    Metabonomics and diabetes mellitus

    Adv. Ther.

    (2007)
  • ClaytonT.A. et al.

    Pharmaco-metabonomic phenotyping and personalized drug treatment

    Nature

    (2006)
  • ClaytonT.A. et al.

    Pharmacometabonomic identification of a significant host-microbiome metabolic interaction affecting human drug metabolism

    Proc. Natl. Acad. Sci. U. S. A.

    (2009)
  • CoenM. et al.

    An integrated metabonomic investigation of acetaminophen toxicity in the mouse using NMR spectroscopy

    Chem. Res. Toxicol.

    (2003)
  • CraigA. et al.

    Systems toxicology: integrated genomic, proteomic and metabonomic analysis of methapyrilene induced hepatotoxicity in the rat

    J. Proteome Res.

    (2006)
  • CrockfordD.J. et al.

    Curve-fitting method for direct quatitation of compounds in complex biological mixtures using 1H NMR: application in metabonomic toxicology studies

    Anal. Chem.

    (2005)
  • CrockfordD.J. et al.

    Statistical heterospectroscopy, an approach to the integrated analysis of NMR and UPLC-MS data sets: application in metabonomic toxicology studies

    Anal. Chem.

    (2006)
  • DieterleF. et al.

    Probabilistic quotient normalization as robust method to account for dilution of complex biological mixtures. Application in 1H NMR metabonomics

    Anal. Chem.

    (2006)
  • DemingS.N.

    Chemometrics: an overview

    Clin. Chem.

    (1986)
  • DhamiM.S. et al.

    Decreased aminotransferase activity of serum and various tissues in the rat after cefazolin treatment

    Clin. Chem.

    (1979)
  • DufourD.R. et al.

    Diagnosis and monitoring of hepatic injury: II. Recommendations for use of laboratory tests in screening, diagnosis, and monitoring

    Clin. Chem.

    (2000)
  • DumasM.E. et al.

    Assessment of analytical reproducibility of 1H NMR spectroscopy based metabonomics for large-scale epidemiological research: the INTERMAP study

    Anal. Chem.

    (2006)
  • DumasM.E. et al.

    Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice

    Proc. Natl. Acad. Sci. U. S. A.

    (2006)
  • EbbelsT.M. et al.

    Prediction and classification of drug toxicity using probabilistic modeling of temporal metabolic data: the consortium on metabonomic toxicology screening approach

    J. Proteome Res.

    (2007)
  • EspandiariP. et al.

    The utility of a rodent model in detecting pediatric drug-induced nephrotoxicity

    Tox. Sci.

    (2007)
  • EspandiariP. et al.

    Age-related differences in susceptibility to toxic effects of valproic acid in rats

    J. Appl. Toxicol.

    (2008)
  • FerraraC.T. et al.

    Genetic networks of liver metabolism revealed by integration of metabolomic and transcriptional profiling

    PLoS Genet

    (2008)
  • FiehnO.

    Metabolomics–The link between genotypes and phenotypes

    Plant Mol. Biol.

    (2002)
  • GarrodS. et al.

    Integrated metabonomic analysis of the multiorgan effects of hydrazine toxicity in the rat

    Chem. Res. Toxicol.

    (2005)
  • Cited by (0)

    View full text