return to agenda

Biomarkers of Toxicity and Surrogate Endpoints for Safety

 

Objectives

 

·        Review the current status of toxicology biomarkers in drug development and clinical safety assessments

-          Do we need more information on biomarkers, and are we using our current information wisely?

·        Demonstrate that biomarkers can serve as early predictors of insidious adverse effects related to chronic drug exposures

-          What is needed to establish the utility of a biomarker as an indicator of clinically significant toxicology?

     Discuss the development of genetically modified rodent models as better predictors of drug effects in humans

-          Can or should transgenic humanized models be developed in other species besides the mouse?

     Discuss the clinical application of pharmacogenetics to predict and prevent adverse drug effects

-          Do the benefits outweigh the costs/risks associated with clinical pharmacogenetic profiling?

     Identify technological approaches that will provide opportunities for developing more and better toxicology biomarkers

-          What are the roles of industry, academia, and the government in the development of toxicology biomarkers?

-          What are the current barriers to the development of toxicology biomarkers?

 

Agenda

Moderators:   Frank Sistare, Ph.D., Center for Drug Evaluation and Research, U.S. Food and Drug Administration Jason D. Morrow, M.D., Vanderbilt University School of Medicine

 

Introduction

Kenneth Olden, Ph.D., Director, National Institute of Environmental Health Sciences

Presentation I

The Status of Toxicology Biomarkers and Safety Evaluation Approaches

James T. MacGregor, Ph.D., Center for Drug Evaluation and Research, U.S. Food and Drug Administration

Presentation II

Cardiac Troponin T as a Biomarker for Monitoring Chronic Doxorubicin Cardiomyopathy

Eugene H. Herman, Ph.D., Center for Drug Evaluation and Research, U.S. Food and Drug Administration

Steven Lipshultz, M.D., University of Rochester Medical Center

 

Presentation III

Preclinical Toxicology Model:  Comparing Different Human Variant Alleles in a Mouse Model

Daniel W. Nebert, M.D., University of Cincinnati Medical Center

Pharmacogenetics as Applied to Human Drug Safety Testing

Richard M. Weinshilboum, M.D., Mayo Clinic and Foundation

The Role of Mass Spectrometry in the Development of Biomarkers

Ian A. Blair, Ph.D., University of Pennsylvania

Toxicogenomics in Safety Assessment

Ronald D. Tyler, D.V.M., Ph.D., GlaxoWellcome Company, United Kingdom

Presentation IV

Gene Expression Analysis for Toxicology:  Moving Beyond Phenomenology

Spencer B. Farr, Ph.D., Phase I Toxicology

Open Discussion

Summary of Session Recommendations

Jason D. Morrow, M.D.

Frank Sistare, Ph.D.

 

ABSTRACTS

 

The Status of Toxicity Biomarkers and Safety Evaluation Approaches

James T. MacGregor, Ph.D.

Safety evaluation has traditionally relied on the measurement of clinical chemistry and hematology parameters that serve as biomarkers of cellular integrity (e.g., serum transaminases) and homeostasis of the organism (e.g., blood urea nitrogen, creatinine, electrolytes) or as indicator cell populations (e.g., white blood cell, host‑defense cell counts).  Recently, advances in biological understanding have created the opportunity for new surrogate markers of toxicity, including (1) inducible damage‑specific responses; (2) transgenic animals with humanized characteristics, accelerated tumor development, and other favorable traits; (3) in vitro human cell models that retain in vivo characteristics; (4) chemokine and/or cytokine signals as surrogates for tissue damage; (5) more specific and sensitive molecular markers of organ damage; and (6) the opportunity to use hybridization array, proteomic, and imaging technologies to improve conventional biomarker assays.  These new technologies can be expected to improve the clinical and nonclinical interface and improve the approaches to both nonclinical and clinical safety studies.

 

Key References

MacGregor JT. Transgenic animal models for mutagenesis studies: Role in mutagenesis research and regulatory testing. Environ Mol Mutagen 1998;38:106‑109.

MacGregor JT, Farr S, Tucker JD, Heddle JA, Tice RR, Turteltaub KW. New molecular endpoints and methods for routine toxicity testing. Fund Appl Toxicol 1995;26:156‑173.

MacGregor JT, Tucker JD, Eastmond DA, Wyrobek AJ. Integration of cytogenetic assays with toxicology studies. Environ Mol Mutagen 1995;25:328‑337.

 

Cardiac Troponin T as a Biomarker for Monitoring Chronic

Doxorubicin Cardiomyopathy

Eugene H. Herman, Ph.D.; Jun Ahang, M.D.; Steven Lipshultz, M.D.; Nader Rifai, Ph.D.; Kazuyo Takeda, M.D.; Zu‑Xi Yu, M.D.; Douglas P. Chadwick, B.S.; and Victor Ferrans, M.D., Ph.D.

The present study sought to determine whether cardiac troponin T (cTnT), a cardiac protein used to diagnose ischemic myocardial injury, also may be useful in detecting doxorubicin (DXR) cardiotoxicity.  Spontaneously hypertensive rats (SHRs) were given 1 mg/kg DXR weekly.  Myocardial tissues and serum samples were collected and analyzed after 2,4,6,8,10, or 12 weeks of treatment.  DXR lesion scores were assessed semiquantitatively by light microscopy, and serum levels of cTnT were quantified by an ELISA method (Enzyum).  Increases of cTnT (0.03‑0.05 ng/mL) in serum and lesion scores of 1 or 1.5 were noted in 1/5 and 2/5 SHRs given 2 or 4 mg/kg DXR, respectively.  All SHRs given 6 mg/kg or more DXR had elevations of cTnT in serum and myocardial lesions.  The average cTnT concentrations and the average lesion scores increased with the cumulative DXR dose (0.13 versus 0.40 ng/ml and 1.4 versus 3.0) in SHRs given 6 and 12 mg/kg DXR, respectively.  It is concluded that cTnT is released from DXR‑damaged myocytes, and measurements of serum levels of this protein can provide a sensitive means for assessing the early and continuing cardiotoxic effects of DXR.

 

Key References

Herman E, Lipshultz S, Rifai N, et al. Use of cardiac troponin T levels as an indicator of doxorubicin‑induced cardiotoxicity. Cancer Res 1998;58:195‑197.

Lipshultz SE, Rifai N, Sallan SE, Lipsitz SR, Dalton V, Sacks DB, Ottlinger ME. Predictive value of serum cardiac troponin T (cTnT) in pediatric patients at risk for myocardial injury. J Am Coll Cardiol 1997;96:2641-2648.

 

Preclinical Toxicology Model:  Comparing Different Human Variant Alleles  in a Mouse Model

Daniel W. Nebert, M.D.; Michael J. Carvan III, Ph.D.; Gary W. Stuart, Ph.D.; and

Tim Dalton, Ph.D.

Striking interindividual differences in environmental toxicity and cancer susceptibility among human populations often reflect polymorphisms in drug‑metabolizing enzymes (DMEs) and receptors that control DME levels.  Compared with interspecies (human‑mouse) differences of twofold to perhaps twentyfold, human interindividual variability in DME activities or DME receptor affinity has often been shown to vary fourfold to more than 10,000‑fold.  How can these different human alleles be studied most effectively and efficiently in an experimental animal model?  Transgenic knock‑in technology can be used to insert unique human alleles in place of the orthologous mouse genes.  The knock‑in of each gene is a separate targeting event, however, requiring (1) construction of the targeting vector and transfection into embryonic stem cells, (2) generation of a targeted mouse, and (3) backcross breeding of the knock‑in mouse (at least six times) to produce a suitable genetically homogeneous (>99 percent) background (for decreasing interindividual variability).  These experiments require years to complete—making this very powerful technology inefficient for routine applications.  If, on the other hand, the initial knock‑in targeting vector includes sequences that would allow the knocked‑in gene to be exchanged (possibly, even repeatedly) for yet another new allele, then testing a battery of human polymorphic alleles in transgenic mice could be accomplished in several months instead of several years.  This gene swapping can be done by zygotic injection of the human allele cassette into the fertilized ovum of the parental knock‑in mouse strain or by cloning mice from fibroblasts containing the nucleus wherein each human allele has already been swapped.  In mouse cells, we have succeeded in gene swapping by exchanging one gene (including its regulatory regions) flanked by heterotypic lox sites with a second gene (including its regulatory regions) flanked by heterotypic lox sites.  This research is supported in part by National Institutes of Health grants R01 ES07058, R01 ES08147, R01 AG09235, R01 ES06321, and P30 ES06096.

 

Pharmacogenetics as Applied to Human Drug Safety Testing

Richard M. Weinshilboum, M.D.

A large number of functionally significant, common, genetic polymorphisms for enzymes that participate in drug and xenobiotic biotransformation have been described over the past three decades.  We now appreciate that genetic polymorphisms for both phase I and phase II drug‑ and xenobiotic‑metabolizing enzymes can be important factors for variation in toxicity after exposure to these agents.  Prototypic examples of genetically polymorphic drug‑metabolizing enzymes will be described in the context of the rapid advances that are occurring in human genomics as well as molecular pharmacology and toxicology.

The Role of Mass Spectrometry in the Development of Biomarkers

Ian A. Blair, Ph.D.

The quantitation and identification of DNA adducts in biological samples as molecular dosimeters of both exogenous and endogenous carcinogens require sophisticated analytical methodology because adducts are normally present in such low concentrations.  Mass spectrometric techniques have high sensitivity and are highly specific because only ions derived from the analyte of interest are monitored.  The development of liquid chromatography/mass spectrometry methodology based on the use of atmospheric pressure ionization‑based techniques of electrospray ionization, ionspray, and atmospheric pressure chemical ionization has had a profound effect on our ability to identify analytes of interest when only trace amounts of material are available.  These techniques are extremely robust and are amenable to the quantitation of DNA adducts in biological samples.  High sensitivity can be attained by the use of selected reaction monitoring of a specific product ion after collision‑induced dissociation has been performed on the protonated molecular ion derived from the analyte of interest.  The ability to use this methodology as a dosimeter for environmental chemicals was exemplified in recent studies we have conducted on butadiene exposure. Liquid chromatography/mass spectrometry, in combination with stable isotope methodology, provided a robust, sensitive, and accurate means to quantify the butadiene derived N7‑guanine adducts.  Using this methodology, it was possible to demonstrate the presence of two diastereomeric forms of trihydroxybutylguanine in liver DNA and to assess the excretion of these DNA adducts in the urine.  Studies on endogenous carcinogens have relied more heavily on the use of gas chromatography/electron capture negative ion chemical ionization mass spectrometry because of the high sensitivity of this technique.  However, liquid chromatography/mass spectrometry has been very useful for identifying novel targets for molecular dosimeters.  Using this methodology, we have recently identified a new reactive electrophilic aldehyde from the breakdown of lipid peroxides that form covalent adducts with DNA bases.  The characterization of these DNA adducts has provided two novel biomarkers that can be employed to assess exposure to oxidative stress.  This research is supported by National Institutes of Health grant CA63878.

 

Key References

Chaudhary AK, Nokubo M, Marnett LJ, Blair IA. Analysis of the malondialdehyde‑2'‑ deoxyguanosine adduct in rat liver DNA by gas chromatography/electron capture negative chemical ionization mass spectrometry. Biol Mass Spectrom 1994;23:457‑464.

Chaudhary AK, Nokubo M, Oglesby TD, Marnett LJ, Blair IA. Characterization of endogenous DNA adducts by liquid chromatography/electrospray ionization tandem mass spectrometry. J Mass Spectrom 1995;30:1157‑1166.

Chaudhary AK, Nokubo M, Reddy GR, Yeola SN, Morrow J, Blair IA, Marnett LJ. Detection of endogenous malondialdehyde‑deoxyguanosine adducts in human liver. Science 1994;265:1580‑1582.

Chaudhary AK, Reddy GR, Blair IA, Marnett LJ. Characterization of an N6‑oxopropenyl‑2'‑deoxyadenosine adduct in malondialdehyde‑modified DNA using liquid chromatography/electrospray ionization tandem mass spectrometry. Carcinogenesis 1996;17:1167‑1170.

Kambouris SJ, Chaudhary AK, Blair IA. Liquid chromatography/electrospray ionization tandem mass spectroscopy (LC/ESI MS/MS) analysis of 1,2‑epoxybutene adducts of purine deoxynucleosides. Toxicology 1996;113:331‑335.

McCoull KD, Rindgen D, Blair IA, Penning TM. Synthesis and characterization of polycyclic aromatic hydrocarbon ortho‑quinone depurinating N7‑guanine adducts. Chem Res Toxicol 1999;In press.

Muller M, Belas FJ, Blair IA, Guengerich FP. Analysis of 1,N2‑ethenoguanine and 5,6,7,9‑tetrahydro‑7‑hydroxy‑9‑oxoimidazo[1,2‑a]purine in DNA treated with 2‑chlorooxirane by high performance liquid chromotography/electrospray mass spectrometry and comparison of amounts to other DNA adducts. Chem Res Toxicol 1997;10:242‑247.

Oe T, Kambouris SJ, Walker VE, Meng Q, Recio L, Wherli S, Chaudhary AK, Blair IA. Persistence of N7‑(1,2:3,4‑trihydroxy)guanine adducts in the livers of mice and rats exposed to 1,3‑butadiene. Chem Res Toxicol 1999;12:247‑257.

Rouzer CA, Chaudhary AK, Nokubo M, Ferguson DM, Reddy GR, Blair IA, Marnett LJ. Analysis of the malondialdehyde‑2'‑deoxyguanosine adduct pyrimidopurinone in human leukocyte DNA by gas chromatography/electron capture/negative chemical ionization/mass spectrometry. Chem Res Toxicol 1997;10:181‑188.

 

Toxicogenomics in Safety Assessment

Ronald D. Tyler, D.V.M., Ph.D.

Most toxicities that are not lethal within minutes are likely to have some impact on gene expression, be a result of gene expression, or both.  Some of the gene expression changes (GECs) that occur as toxicity develops are expected to be unique to the mechanism of toxicity (e.g., free radical production, inhibition of cellular respiration).  Some GECs are expected to be unique to the type of toxicity (e.g., apoptosis, nongenotoxic carcinogenicity) but common among mechanisms that cause the same type of toxicity.  Other GECs are expected to be adaptive in response to changes in such things as blood pressure, nutrition, and so forth.  Determination of GECs that reliably indicate the above would allow development of gene expression‑based biomarkers of toxicity.  The most valuable GECs will probably be critical core genes that have been conserved during evolution.  These GECs will be shared among species, greatly improving interspecies comparisons and extrapolations.  If gene expression analysis in discovery and toxicology studies identify GECs indicative of toxicity and in vitro studies with human and animal cell lines establish that the human gene homologs behave as the animal genes do, GECs can be used as biomarkers of toxicity in clinical trials and patients.  These biomarkers will be especially valuable when they occur in easily assessable tissues (e.g., lymphocytes) and for early detection of chronic toxicity.  Although functional or morphologic evidence of chronic toxicities may take weeks to years to develop, characteristic GECs likely occur in hours or days.  Identification of these GECs would enable early detection of the chronic toxicity, markedly shortening necessary animal studies and protecting clinical trial subjects and patients.  To fully harvest the benefit of gene expression analysis, reliable broad gene coverage technologies, computer systems capable of manipulating large data sets, and scientists that understand pathophysiology at the molecular and transcriptional level are necessary.  Development of these technologies, systems, and scientists should be a major focus of future resource commitment.

Gene Expression Analysis for Toxicology:  Moving Beyond Phenomenology

Spencer B. Farr, Ph.D.

The science of toxicology is changing radically.  There are two fundamental forces compelling this change:  (1) high‑throughput chemical library synthesis and screening and (2) the human genome project and attendant technology for rapid polymorphism and expression analysis.  Toxicologists are faced with the challenge of developing high‑throughput toxicity screens to keep pace with the increased number of “hits” derived from upstream screening activities.  In addition, toxicologists will need to better understand the interaction between a potential drug and the individual genetic makeup of the patient exposed to that compound.  To make meaningful progress in either area, toxicologists will have to effectively assimilate and wisely use an avalanche of information.  Without appropriate bioinformatics developed specifically for these challenges, the effort will be stunted.  The purpose of this presentation is to present an overview of the interplay among screening, molecular toxicologic analysis, and associated informatics.

 

Additional Thoughts on Toxicity Biomarkers:  The Isoprostanes, Indices of Oxidative Stress In Vivo

Jason D. Morrow, M.D.

Over the past decade, the role that oxidant stress plays in human pathophysiology has received considerable attention.  A number of biomarkers have been developed to measure oxidative injury although most have serious shortcomings when applied to the assessment of oxidant stress in vivo.  We have previously described a series of prostaglandin F2‑like compounds, termed F2‑isoprostanes (F2‑IsoPs), produced independently of the cyclooxygenase enzyme by the free‑radical‑catalyzed peroxidation of arachidonic acid in vivo in humans.  A large body of evidence suggests that quantification of these compounds is an accurate index of lipid peroxidation in vitro and in vivo.  F2‑IsoPs can be quantified by mass spectrometry or immunologically.  A particular interest of mine is the role of oxidative injury in atherosclerosis and neurodegenerative disorders such as Alzheimer's disease.  We have found that risk factors for atherosclerosis are associated with markedly increased circulating levels of F2‑IsoPs and that antioxidants such as vitamin E, which decrease the incidence of atherosclerosis, suppress plasma F2‑IsoP levels.  Furthermore, F2‑IsoPs are selectively increased in cerebrospinal fluid from patients with Alzheimer's disease, and levels correlate with disease severity.  Therefore, these studies suggest that quantification of F2‑IsoPs may provide a useful biomarker of oxidant stress in humans.

 

Key References

Montine TJ, Morrow JD. Increased CSF F2‑isoprostane levels in probable Alzheimer's disease. Neurology 1999;52:562‑565.

Morrow JD, et al. The isoprostanes‑unique bioactive markers of lipid peroxidation. Prog Lipid Res 1997;36:1‑21.

Roberts LJ, Morrow JD. Isoprostanes as markers of lipid peroxidation in atherosclerosis. In: Molecular and Cellular Basis of Inflammation. Serhan CN (ed.) Totowa NJ: Humana Press, 1998;141‑163