Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Expert Opin Drug Metab Toxicol ; 12(12): 1463-1477, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27548410

ABSTRACT

INTRODUCTION: If a drug is found to be an inducer of hepatic drug metabolizing enzymes via activation of nuclear receptors such as pregnane X receptor (PXR) or constitutive androstane receptor (CAR), it is likely that drug transporters regulated through these same receptors will be induced as well. This review highlights what is currently known about the molecular mechanisms that regulate transporter expression and where the research is directed. Areas covered: This review is focused on publications that describe the role of activated hepatic nuclear receptors in the subsequent regulation of drug uptake and/or efflux transporters following exposure to xenobiotics. Expert opinion: Many of the published studies on the role of nuclear receptors in the regulation of drug transporters involve non-human test animals. But due to species response differences, these associations are not always applicable to humans. For this reason, some relevant human in vitro models have been developed, such as primary or cryopreserved human hepatocytes, human liver slices, or HepG2 or HuH7 cell lines transiently or stably transfected with PXR expression and reporter constructs as well as in vivo models such as PXR-humanized mice. These human-relevant test systems will continue to be developed and applied for the testing of investigational drugs.


Subject(s)
Liver/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Xenobiotics/metabolism , Animals , Biological Transport/physiology , Cell Line , Constitutive Androstane Receptor , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Humans , Mice , Pregnane X Receptor , Receptors, Steroid/metabolism , Species Specificity
2.
Biomarkers ; 21(5): 387-403, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26983778

ABSTRACT

Context The silencing or activation of cancer-associated genes by epigenetic mechanisms can ultimately lead to the clonal expansion of cancer cells. Objective The aim of this review is to summarize all relevant epigenetic biomarkers that have been proposed to date for the diagnosis of some prevalent human cancers. Methods A Medline search for the terms epigenetic biomarkers, human cancers, DNA methylation, histone modifications and microRNAs was performed. Results One hundred fifty-seven relevant publications were found and reviewed. Conclusion To date, a significant number of potential epigenetic cancer biomarkers of human cancer have been investigated, and some have advanced to clinical implementation.


Subject(s)
Biomarkers, Tumor/genetics , Epigenesis, Genetic , Genes, Neoplasm/genetics , Neoplasms/diagnosis , DNA Methylation , Histone Code , Humans , MEDLINE , MicroRNAs , Neoplasms/genetics
3.
Biomarkers ; 19(7): 541-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25189636

ABSTRACT

CONTEXT: The definitive standard for the diagnosis of nonalcoholic fatty liver disease (NAFLD) is clinico-pathological correlation, but frequently the only laboratory abnormality is an elevation of serum aminotransferases. OBJECTIVE: This has resulted in the search for more specific laboratory biomarkers. METHODS: The literature was searched for novel plasma/serum markers of NAFLD. RESULTS: Studies reviewed here included histologically-confirmed patients presenting some stage of NAFLD and monitored one or more novel serum/plasma biomarkers. CONCLUSION: The most promising application of some of these novel biomarkers for the detection and quantification of NAFLD and particularly NASH appears to be in the combination of several into diagnostic panels.


Subject(s)
Liver/metabolism , Non-alcoholic Fatty Liver Disease/blood , Biomarkers/blood , Disease Progression , Humans , Liver/pathology , MicroRNAs/blood , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Predictive Value of Tests , Prognosis , Risk Factors , Severity of Illness Index
4.
Semin Liver Dis ; 34(2): 205-14, 2014 May.
Article in English | MEDLINE | ID: mdl-24879984

ABSTRACT

Several drugs have been associated with the potential for drug-induced hepatic steatosis (DIHS) and/or phospholipidosis (DIPL), a lysosomal storage disorder. Drug-induced hepatic steatosis is generally a chronic but reversible affliction and may involve drug accumulation in the liver. Fat accumulation may be either macrovesicular or microvesicular in nature. Commonly used medications associated with DIHS include amiodarone, valproate, tamoxifen, methotrexate, and some chemotherapeutic and antiretroviral agents. Two recently approved medications for the treatment of hereditary homozygous hypercholesterolemia have also been noted to cause hepatic steatosis. For some compounds such as methotrexate and tamoxifen, the underlying metabolic risk factors such as obesity and metabolic syndrome may exacerbate their potential to cause DIHS and its progression. In this article, the authors discuss the preclinical screening and mechanisms of DIHS and DIPL, and review specific examples of drugs commonly used in clinical practice that are known to cause DIHS.


Subject(s)
Fatty Liver/chemically induced , Fatty Liver/metabolism , Lysosomal Storage Diseases/chemically induced , Mitochondria, Liver/metabolism , Phospholipids/metabolism , Amiodarone/adverse effects , Animals , Anti-Arrhythmia Agents/adverse effects , Anti-Retroviral Agents/adverse effects , Anticholesteremic Agents/adverse effects , Anticonvulsants/adverse effects , Antineoplastic Agents, Hormonal/adverse effects , Drug Evaluation, Preclinical , Fatty Liver/pathology , Humans , Immunosuppressive Agents/adverse effects , Methotrexate/adverse effects , Mitochondria, Liver/drug effects , Tamoxifen/adverse effects , Valproic Acid/adverse effects
5.
Regul Toxicol Pharmacol ; 68(1): 76-84, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24275050

ABSTRACT

Diethanolamine (DEA) has been listed on the State of California's Proposition 65 List. This listing is based in part on tumors reported in a National Toxicology Program (NTP) 2-year dermal carcinogenicity study in mice which found clear evidence of carcinogenic activity in B6C3F1 mice based on increased incidences of liver neoplasms in both sexes, and increased incidences of renal tubule neoplasms in males. Although considerable controversy exists on the relevance of the NTP study to humans, industries are obligated to comply with the Proposition 65 labeling requirement and drinking water discharge prohibition, unless they are able to demonstrate that DEA levels in their products are below a specific No Significant Risk Level (NSRL). The State of California has not published an NSRL for DEA. In this article, a NSRL of 5.6 µg/day and a life-stage-adjusted NSRL(adj) of 1.4 µg/day are derived from the NTP carcinogenicity study using a benchmark dose modeling method based on the incidence of hepatocellular carcinomas in female mice, in accordance with the guidelines of California EPA.


Subject(s)
Carcinogens/standards , Ethanolamines/standards , Animals , California , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Ethanolamines/pharmacokinetics , Ethanolamines/toxicity , Female , Government Regulation , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/prevention & control , Male , Mice , Mutagenicity Tests , Rats , Rats, Inbred F344 , Risk Assessment/standards , Skin/metabolism , Skin Absorption , State Government
6.
Hum Exp Toxicol ; 33(9): 928-39, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24299907

ABSTRACT

Adverse drug reactions (ADRs) can involve all tissues and organs, but liver injuries are considered among the most serious. A number of prospective, multicenter studies have confirmed a higher risk of ADRs in general among female subjects compared to a male cohort. Although drug-induced liver injury (DILI) is infrequently encountered, the preponderance of evidence suggests that women appear to be more susceptible than men to fulminate hepatic/acute liver failure especially in response to some anti-infective drugs and to autoimmune-like hepatitis following exposure to certain other therapeutic drugs. A number of hypotheses have been proposed to explain this sex difference in susceptibility to DILI. Collectively, these hypotheses suggest three basic sex-dependent mechanisms that include differences in various aspects of drug pharmacokinetics (PK) or pharmacodynamics following the administration of certain drugs; specific hormonal effects or interactions with immunomodulating agents or signaling molecules; and differences in the adverse response of the immune system to some drugs, reactive drug metabolites, or drug-protein adducts. At the preclinical drug safety stage, there is a need for more research on hormonal effects on drug PK and for additional research on gender differences in aberrant immune responses that may lead to idiosyncratic DILI in some female patients. Because the detection of rare but serious hepatic ADRs requires the exposure of very large patient populations, pharmacovigilance networks will continue to play a key role in the postmarketing surveillance for their detection and reporting.


Subject(s)
Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Female , Humans , Liver/drug effects , Male , Risk Factors , Sex Factors
7.
Genome Med ; 5(9): 85, 2013.
Article in English | MEDLINE | ID: mdl-24073687

ABSTRACT

Biomarkers of drug-induced liver injury (DILI) are essential for the diagnosis of severe cases of DILI in clinical trials and clinical practice, but the currently used biomarker paradigm detects damage after it has occurred and has limited prognostic value. The development of new biomarker strategies that improve the diagnosis of DILI by providing increased specificity and/or by identifying individual patients who are at risk for DILI is therefore crucial. See related Research, http://genomemedicine.com/content/5/9/86.

8.
Mol Diagn Ther ; 17(6): 343-54, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23868512

ABSTRACT

Drug-induced liver injury (DILI) remains a rare but serious complication in drug therapy that is a primary cause of drug failure during clinical trials. Conventional biomarkers, particularly the serum transaminases and bilirubin, serve as useful indicators of hepatocellular or cholestatic liver injury, respectively, but only after substantial and sometimes irreversible tissue damage. Ideally, more sensitive biomarkers that respond very early before irreversible injury has occurred would offer improved outcomes. Novel biomarkers are initially being developed in animal models exposed to intrinsically hepatotoxic stimuli. However, the eventual translation to human populations, even those with known risk factors that predispose the liver to drug toxicity, would be the fundamental goal. Ultimately, some might even be applicable for the early identification of individuals predisposed to idiosyncratic hepatotoxicity potential. This article reviews recent progress in the discovery and qualification of novel biomarkers for DILI and delineates the path to eventual utilization for risk assessment. Some major categories of plasma or serum biomarkers surveyed include proteins, cytokines, circulating mRNAs, and microRNAs.


Subject(s)
Biomarkers/blood , Chemical and Drug Induced Liver Injury/diagnosis , Chemical and Drug Induced Liver Injury/metabolism , Liver/metabolism , Animals , Chemical and Drug Induced Liver Injury/genetics , Cytokines/blood , Databases, Bibliographic , Disease Models, Animal , Humans , Metabolomics , MicroRNAs/blood , MicroRNAs/metabolism , RNA, Messenger/blood , RNA, Messenger/metabolism , Risk Assessment , Risk Factors
9.
Toxicol Pathol ; 40(5): 810-8, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22504321

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) represent therapeutic targets for the management of type 2 diabetes mellitus and dyslipidemia. Rodent carcinogenicity studies have revealed a link between γ and dual γ/α PPAR agonist treatment and the increased incidence of subcutaneous (SC) liposarcomas/fibrosarcomas or hemangiosarcomas, but very little has been reported for potent and selective PPARα agonists. We present a mode of action framework for the development of SC mesenchymal tumors in rodents given PPAR agonists. (1) Tumor promotion results from pharmacologically mediated recruitment (proliferation and differentiation), thermogenesis and adipogenesis of stromovascular cells, and subsequent generation of oxidative free radicals. (2) Tumor initiation consists of chemotype-driven mitochondrial dysfunction causing uncontrolled oxidative stress and permanent DNA damage. Promotion is characterized by enhanced adipogenesis in the SC adipose tissue, where the baseline PPARγ expression and responsiveness to PPARγ ligands is the highest, and by thermogenesis through expression of the uncoupling protein 1 (UCP-1) and the PPARγ co-activator 1 α (PGC-1α), two factors more highly expressed in brown versus white adipose tissue. Initiation is supported by the demonstration of mitochondrial uncoupling and OXPHOS Complexes dysfunction (Complexes III, IV and V) by compounds associated with increased incidences of sarcomas (muraglitazar and troglitazone), but not others lacking malignant tumor effects (pioglitazone, rosiglitazone).


Subject(s)
Hypoglycemic Agents/toxicity , PPAR alpha/agonists , PPAR gamma/agonists , Sarcoma/chemically induced , Adipogenesis/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Cell Differentiation , Chromans/toxicity , DNA Damage/drug effects , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/therapy , Glycine/analogs & derivatives , Glycine/toxicity , Ion Channels/genetics , Ion Channels/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxazoles/toxicity , Oxidative Stress/drug effects , PPAR alpha/genetics , PPAR alpha/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Pioglitazone , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Rats , Rodentia/metabolism , Rosiglitazone , Sarcoma/pathology , Thermogenesis/drug effects , Thiazolidinediones/toxicity , Transcription Factors/genetics , Transcription Factors/metabolism , Troglitazone , Uncoupling Protein 1
10.
Expert Opin Drug Metab Toxicol ; 8(3): 335-47, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22288564

ABSTRACT

INTRODUCTION: Idiosyncratic drug-induced liver injury (DILI) is a rare, serious and sometimes fatal condition that lacks an effective clinical countermeasure. Polymorphisms of bioactivation/toxification pathways via the Phase I drug-metabolizing enzymes, Phase II detoxification reactions and Phase III excretion/transport, together with immunological factors, are predisposing factors for some DILI. AREAS COVERED: This review focuses on the seminal role of hepatic drug metabolism in the idiosyncratic toxicity response and the potential role of genetic polymorphisms in specific drug-metabolizing enzymes or transporters. Furthermore, the article looks at recent developments for the assessment of those mechanisms, in order to avoid them in novel drug candidates that are still under development. Examples of proposed or confirmed metabolic mechanisms from the current literature for marketed or previously marketed drugs are summarized in detail. The contributions of Phase I, II and III and other enzymatic pathways in the idiosyncratic response are reviewed with particular emphasis on gene polymorphisms that might explain why some individuals respond in an aberrant manner. EXPERT OPINION: Toxicologists continue to focus their efforts to define the idiosyncratic response at the biochemical and molecular levels. This has resulted in the development of some early drug screening tools based on such characteristics as the daily dose, metabolite covalent binding and polymorphisms in drug-metabolizing enzymes. While it will take years to fully assess the impact of these recently developed assessment tools, research on genetically based differences in hepatic metabolic pathways will continue, aided by the establishment of DILI registries for improved patient access.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Liver/metabolism , Animals , Biotransformation/genetics , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Evidence-Based Medicine , Genetic Predisposition to Disease , Humans , Liver/enzymology , Mitochondria, Liver/metabolism , Oxidative Stress/genetics , Pharmacogenetics , Phenotype , Polymorphism, Genetic , Risk Assessment , Risk Factors , Toxicity Tests
11.
Toxicol Pathol ; 40(3): 435-47, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22301950

ABSTRACT

Weak peroxisome proliferator-activated receptor (PPAR) α agonists (fibrates) are used to treat dyslipidemia. This study compared the effects of the potent and selective PPARα agonist CP-778875 on peroxisomal ß-oxidation and cardiac and/or skeletal muscle injury with those of the weak PPARα agonist fenofibrate. We hypothesized that these muscle effects are mediated through the PPARα receptor, leading to increased ß-oxidation and consequent oxidative stress. CP-778875 (5 or 500 mg/kg) and fenofibrate (600 or 2,000→1,200 mg/kg, dose lowered because of intolerance) were administered to rats for six weeks. Standard end points, serum troponin I, heart and skeletal muscle ß-oxidation of palmitoyl-CoA, and acyl co-oxidase (AOX) mRNA were assessed. Both compounds dose-dependently increased the incidence and/or severity of cardiomyocyte degeneration and necrosis, heart weight, troponin I, and skeletal muscle degeneration. Mean heart ß-oxidation (3.4- to 5.1-fold control) and AOX mRNA (2.4- to 3.2-fold control) were increased with CP-778875 500 mg/kg and both doses of fenofibrate. ß-Oxidation of skeletal muscle was not affected by either compound; however, a significant increase in AOX mRNA (1.6- to 2.1-fold control) was observed with CP-778875 500 mg/kg and both doses of fenofibrate. Taken together, these findings were consistent with PPARα agonism and support the link between increased cardiac and skeletal muscle ß-oxidation and resultant muscle injury in the rat.


Subject(s)
Fenofibrate/toxicity , Heart/drug effects , Muscle, Skeletal/drug effects , Oxidative Stress/drug effects , PPAR alpha/agonists , Animals , Blood Chemical Analysis , Body Weight , Dose-Response Relationship, Drug , Female , Fenofibrate/pharmacokinetics , Liver/chemistry , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , Muscle Proteins/metabolism , Muscle, Skeletal/chemistry , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/chemically induced , Muscular Diseases/metabolism , Myocardium/chemistry , Myocardium/metabolism , Myocardium/pathology , NAD/metabolism , Peroxisomes/metabolism , Rats , Rats, Sprague-Dawley , Toxicity Tests , Troponin I/blood , Troponin I/metabolism
12.
Expert Opin Drug Metab Toxicol ; 7(8): 949-65, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21510823

ABSTRACT

INTRODUCTION: Hepatic steatosis is the histological observation of numerous lipid inclusions due to an excess accumulation of triacylglycerols. They are a concern with new therapeutic candidates because they signify altered lipid metabolism that can progress to more serious liver toxicity. AREAS COVERED: This article is based on an article search using the PubMed database from 1987 to 2011 and confirms associations for several previously marketed drugs with four basic hepatocellular mechanisms. The article also describes how these mechanisms are controlled by master regulators of lipid metabolism, which include gene transcription factors, nuclear receptors, hormonal signaling, energy sensing proteins, endoplasmic reticulum stress signaling and certain key metabolic intermediates. EXPERT OPINION: Drug-induced hepatic steatosis is typically not detectable by conventional means other than invasive histological examinations. By understanding the basic mechanisms, key regulators and energy signaling systems of the liver, the investigator is better equipped to avoid xenobiotics with steatogenic potential in the drug discovery or early development process. There are now a number of methods for detecting this potential, specifically gene expression or metabolomic profiling and pathway analysis or mechanism-based in vitro systems.


Subject(s)
Fatty Liver/chemically induced , Fatty Liver/pathology , Liver/pathology , Xenobiotics/adverse effects , Adipose Tissue/metabolism , Animals , Endoplasmic Reticulum/metabolism , Gene Expression Regulation/drug effects , Humans , Lipid Metabolism/drug effects , Receptors, Cytoplasmic and Nuclear/drug effects , Signal Transduction/drug effects , Stress, Physiological , Transcription Factors/genetics , Transcription Factors/metabolism , Triglycerides/biosynthesis , Triglycerides/blood , Xenobiotics/metabolism
13.
Toxicology ; 279(1-3): 10-8, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-20974209

ABSTRACT

Hepatic steatosis is characterized by the accumulation of lipid droplets in the liver. Although relatively benign, simple steatosis can eventually lead to the development of steatohepatitis, a more serious condition characterized by fibrosis, cirrhosis, and eventual liver failure if the underlying cause is not eliminated. According to the "two hit" theory of steatohepatitis, the initial hit involves fat accumulation in the liver, and a second hit leads to inflammation and subsequent tissue injury. Because some xenobiotics target liver fatty acid metabolism, especially mitochondrial ß-oxidation, it is important to avoid potential drug candidates that can contribute to either the initiation of liver steatosis or progression to the more injurious steatohepatitis. The gold standard for the detection of these types of hepatic effects is histopathological examination of liver tissue. In animal studies, these examinations are slow, restricted to a single sampling time, and limited tissue sections. Recent literature suggests that rapid in vitro screening methods can be used early in the drug R&D process to identify compounds with steatotic potential. Further, progress in the identification of potential serum or plasma protein biomarkers for these liver changes may provide additional in vivo tools to the preclinical study toxicologist. This review summarizes recent developments for in vitro screening and in vivo biomarker detection for steatotic drug candidates.


Subject(s)
Chemical and Drug Induced Liver Injury/diagnosis , Drug-Related Side Effects and Adverse Reactions , Fatty Liver/diagnosis , Animals , Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Drug Design , Drug Evaluation, Preclinical/methods , Fatty Liver/chemically induced , Fatty Liver/pathology , Humans , Mitochondria/metabolism , Toxicity Tests/methods
14.
Toxicol Mech Methods ; 20(4): 159-66, 2010 May.
Article in English | MEDLINE | ID: mdl-20218941

ABSTRACT

The induction of hepatic cytochrome P450 (CYP) enzymes, conjugating enzymes, and drug transporters involved in the phase I-III metabolism of xenobiotics is frequently encountered in pre-clinical drug safety studies. As xenobiotics, new drug entities can serve as ligands to three major nuclear receptors; the aryl hydrocarbon receptor (AhR), the constitutive androstane receptor (CAR), and the pregnane X receptor (PXR). These act as xenosensors that often coordinate gene expression with several other nuclear receptors normally involved in endobiotic metabolism. A subsequent gene activation cascade can result in altered liver weights and histopathology and, in some cases, reduced therapeutic efficacy if the drug under test is also a substrate for the induced metabolic enzymes. In humans, CYP induction can result in therapeutic failure for autoinducers or drug-drug interactions if the pharmacokinetic and pharmacodynamic properties of co-administered drugs are altered because they are substrates for the induced enzymes. In addition to CYP gene expression, nuclear receptor proteins regulate the expression of complex gene networks, and therefore mediate the metabolism and modify the effects of steroid hormones, fat-soluble vitamins, and free fatty acids on the metabolic, reproductive, and developmental processes of mammals. CAR and PXR also regulate hepatic energy metabolism through cross-talk with insulin- or glucagon-responsive transcription factors. This review examines the perturbation of these endogenous regulatory systems by xenobiotic CYP inducers, which have potential pathophysiological consequences ranging from alterations in the biological clock to adverse effects on the cardiovascular system of pre-clinical species.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Xenobiotics/pharmacology , Enzyme Induction
15.
Toxicol Appl Pharmacol ; 245(1): 134-42, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20219512

ABSTRACT

Biomarkers are biometric measurements that provide critical quantitative information about the biological condition of the animal or individual being tested. In drug safety studies, established toxicity biomarkers are used along with other conventional study data to determine dose-limiting organ toxicity, and to define species sensitivity for new chemical entities intended for possible use as human medicines. A continuing goal of drug safety scientists in the pharmaceutical industry is to discover and develop better trans-species biomarkers that can be used to determine target organ toxicities for preclinical species in short-term studies at dose levels that are some multiple of the intended human dose and again later in full development for monitoring clinical trials at lower therapeutic doses. Of particular value are early, predictive, noninvasive biomarkers that have in vitro, in vivo, and clinical transferability. Such translational biomarkers bridge animal testing used in preclinical science and human studies that are part of subsequent clinical testing. Although suitable for in vivo preclinical regulatory studies, conventional hepatic safety biomarkers are basically confirmatory markers because they signal organ toxicity after some pathological damage has occurred, and are therefore not well-suited for short-term, predictive screening assays early in the discovery-to-development progression of new chemical entities (NCEs) available in limited quantities. Efforts between regulatory agencies and the pharmaceutical industry are underway for the coordinated discovery, qualification, verification and validation of early predictive toxicity biomarkers. Early predictive safety biomarkers are those that are detectable and quantifiable prior to the onset of irreversible tissue injury and which are associated with a mechanism of action relevant to a specific type of potential hepatic injury. Potential drug toxicity biomarkers are typically endogenous macromolecules in biological fluids with varying immunoreactivity which can present bioanalytical challenges when first discovered. The potential success of these efforts is greatly enhanced by recent advances in two closely linked technologies, toxicoproteomics and targeted, quantitative mass spectrometry. This review focuses on the examination of the current status of these technologies as they relate to the discovery and development of novel preclinical biomarkers of hepatotoxicity. A critical assessment of the current literature reveals two distinct lines of safety biomarker investigation, (1) peripheral fluid biomarkers of organ toxicity and (2) tissue or cell-based toxicity signatures. Improved peripheral fluid biomarkers should allow the sensitive detection of potential organ toxicity prior to the onset of overt organ pathology. Advancements in tissue or cell-based toxicity biomarkers will provide sensitive in vitro or ex vivo screening systems based on toxicity pathway markers. An examination of the current practices in clinical pathology and the critical evaluation of some recently proposed biomarker candidates in comparison to the desired characteristics of an ideal toxicity biomarker lead this author to conclude that a combination of selected biomarkers will be more informative if not predictive of potential animal organ toxicity than any single biomarker, new or old. For the practical assessment of combinations of conventional and/or novel toxicity biomarkers in rodent and large animal preclinical species, mass spectrometry has emerged as the premier analytical tool compared to specific immunoassays or functional assays. Selected and multiple reaction monitoring mass spectrometry applications make it possible for this same basic technology to be used in the progressive stages of biomarker discovery, development, and more importantly, routine study applications without the use of specific antibody reagents. This technology combined with other "omics" technologies can provide added selectivity and sensitivity in preclinical drug safety testing.


Subject(s)
Biomarkers/metabolism , Chemical and Drug Induced Liver Injury/metabolism , Proteome/metabolism , Toxicity Tests/methods , Animals , Biomarkers/chemistry , Mass Spectrometry , Toxicity Tests/instrumentation
16.
Drug Chem Toxicol ; 31(4): 427-45, 2008.
Article in English | MEDLINE | ID: mdl-18850354

ABSTRACT

Dexamethasone (DEXA) administration has been associated with serum alanine aminotransferase (ALT) elevations that may result from enhanced ALT expression. The aim of our current study was to compare liver vs. serum ALT activity and to examine the onset of any hepatocellular changes. Groups of 4 male Sprague-Dawley rats were administered a single dose of DEXA or corn oil at 12, 16, and 24 h prior to euthanasia or once-daily for 2, 3, or 4 days. All (nonfasted) rats were necropsied together on Day 5. While DEXA incrementally increased liver ALT activity in the 1-, 2-, 3-, and 4-day treatment groups (maximal, 3.7-fold), liver aspartate aminotransferase (AST) never exceeded 1.4-fold over control. Significant hepatic glycogen elevations were detected after DEXA treatment, which correlated with microscopic observations. Serum ALT, AST, sorbitol dehydrogenase, and glutamate dehydrogenase (GLDH) increased after 2, 3, and 4 days of DEXA dosing (1.3-10.3-fold). DEXA-related necropsy findings included pale livers consistent with glycogen deposition. The relative percent liver to body weight was elevated in all DEXA-treated rats. Hepatocellular necrosis was observed in 1/4 rats at 12 h, 2/4 rats at 2 days, 4/4 rats at 3 days, and 3/4 rats at 4 days. DEXA treatment <2 days failed to produce consistent evidence of hepatic injury, as detected by serum biomarkers and pathology assessment. However, early DEXA treatment did correlate with apparent ALT induction. Ultimately, this may explain some early asymptomatic serum ALT elevations seen clinically.


Subject(s)
Alanine Transaminase/metabolism , Dexamethasone/administration & dosage , Glucocorticoids/administration & dosage , Liver/drug effects , Alanine Transaminase/blood , Animals , Biomarkers/metabolism , Glycogen/metabolism , Liver/enzymology , Liver/pathology , Male , Necrosis , Rats , Rats, Sprague-Dawley , Time Factors , Up-Regulation
17.
Toxicol In Vitro ; 20(8): 1488-99, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16782301

ABSTRACT

Xenobiotic-inflammation interactions lead to hepatotoxicity in vivo. Selected xenobiotic agents (acetaminophen, APAP; chlorpromazine, CPZ; allyl alcohol, AlOH; monocrotaline, MCT) for which this occurs were evaluated for ability to elicit the release of Kupffer cell (KC)-derived inflammatory mediators and to modulate lipopolysaccharide (LPS)-stimulated release of these mediators. Using KCs and hepatocytes (HPCs) isolated from rat, KC/HPC cocultures were treated with either LPS, xenobiotic, vehicle or a combination. Six hours later, the release of inflammatory mediators was assessed. LPS alone caused a concentration-dependent increase in TNF-alpha release but had no significant effect on the release of PGE(2). APAP by itself did not alter release of TNF-alpha, PGE(2), IL-10, Gro/KC or IFN-gamma; however, in the presence of LPS, APAP enhanced LPS-induced TNF-alpha and Gro/KC release. APAP also attenuated LPS-induced increases in IL-10 and MCP-1. CPZ alone caused a concentration-dependent increase in TNF-alpha release, which was approximately additive in the presence of LPS. AlOH alone did not affect TNF-alpha release, but decreased TNF-alpha production in the presence of LPS. AlOH increased PGE(2) production, and this effect was potentiated in the presence of LPS. MCT by itself did not affect release of TNF-alpha but increased the response to LPS. Neither MCT, LPS, nor the combination affected production of PGE(2). These results demonstrate that KC/HPC cocultures can be used to evaluate interactions of xenobiotics with LPS. Furthermore, data from these studies qualitatively mirror reported data from whole animal studies, suggesting that this model could be useful for predicting aspects of xenobiotic-inflammation interactions in vivo.


Subject(s)
Hepatocytes/drug effects , Inflammation/pathology , Kupffer Cells/drug effects , Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Animals , Antipsychotic Agents/toxicity , Carcinogens/toxicity , Cell Separation , Chlorpromazine/toxicity , Coculture Techniques , Drug Interactions , Enzyme-Linked Immunosorbent Assay , Humans , Immunoassay , Immunohistochemistry , Lipopolysaccharides/pharmacology , Male , Monocrotaline/toxicity , Potassium Chloride/pharmacology , Propanols/toxicity , Rats , Rats, Sprague-Dawley
18.
Curr Drug Metab ; 7(3): 219-29, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16611018

ABSTRACT

Severe adverse drug responses are infrequent but occasionally serious events that are not readily predictable at the preclinical development level using only non-human or in vitro models. A common characteristic of the more serious toxicities is generation of short-lived and highly reactive electrophilic species in some individuals. The objective here is to review the literature for toxicological mechanisms that underlie known adverse drug reactions and then categorize the biological consequences of reactive chemical intermediates and radicals in terms of human risk factors and known metabolic variables. Xenobiotics described as being associated with rare but potentially serious adverse events affecting liver, skin, or causing blood dyscrasias tend to have three of four essential characteristics, (1) they are capable of forming short-lived reactive intermediates (RI) or free radicals in target tissues under ideal conditions that are distinct from primary metabolic products, (2) these RI escape/overwhelm the detoxification mechanisms associated with the site of origin or form toxic conjugates, (3) the unconjugated RI must either selectively damage critical proteins or other key macromolecules or (4) the RI acts as a hapten and stimulates an immunological (hypersensitivity) response or overcomes tolerance. Some risk factors may increase the probability of susceptibility, but this remains an active area of research. Because of the complexity of the pathogenesis of some injuries and the role of individual factors, no highly predictive in vitro screening methods are available; however, several methods are evolving that may be used to reveal mechanisms of action when a serious adverse effect is encountered.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Animals , Antioxidants/metabolism , Biotransformation , Free Radicals/adverse effects , Free Radicals/chemistry , Humans , Pharmaceutical Preparations/chemistry , Reactive Oxygen Species/metabolism , Risk Factors , Toxicogenetics
19.
Food Chem Toxicol ; 44(4): 528-37, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16274908

ABSTRACT

The purpose of this investigation was to examine the relationship among hepatic microsomal enzyme induction, liver weight, histological evidence of hepatic injury, and serum clinical chemistry markers of hepatic origin in the cynomolgus monkey. We report here the results from independent toxicology studies for 10 investigative drug candidates representing four therapeutic classes. Study conditions were selected to elicit target organ toxicity. We found that six of the 10 compounds altered cytochrome P450-associated activities in both male and female monkeys, two in females only, and one altered similar activities in males only. Frequently, significant treatment-related elevations in NADPH cytochrome c reductase and ethylmorphine N-demethylase were noted. When the results from all 10 studies were pooled, 14 cytochrome P450-associated activities were significantly increased and five were decreased in males compared to 15 significantly increased and three decreased in the females. Treatment-associated liver weight increases were noted in four studies. Except for hepatocellular hypertrophy in one study, no significant treatment-related microscopic changes in liver and no elevations of serum biomarkers commonly associated with liver toxicity were observed in any of the studies that demonstrated significant hepatic enzyme induction. Compared to parallel rat studies, one compound was an inducer only in monkeys and one was an inducer only in rats. Significant elevations of microsomal drug-metabolizing enzymes in the cynomolgus monkey liver are not accompanied by substantial hepatic changes except for hepatomegaly. These alterations in the hepatic drug-metabolizing enzyme system were benign based the absence of histopathological lesions and serum biomarkers of hepatobiliary toxicity.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Animals , Cytochrome P-450 Enzyme System/drug effects , Enzyme Induction , Female , Haplorhini , Liver/drug effects , Liver/pathology , Male , No-Observed-Adverse-Effect Level , Rats , Species Specificity , Toxicity Tests
20.
Toxicol Mech Methods ; 16(7): 385-94, 2006.
Article in English | MEDLINE | ID: mdl-20021012

ABSTRACT

The assessment of hepatic microsomal enzyme induction at the completion of preclinical toxicology studies in rodents and large mammals provides a wealth of information to the toxicologist and pharmacokineticist regarding how the drug-metabolizing system of the hepatocyte endoplasmic reticulum responded to high-dose levels of a xenobiotic designed for a specific pharmacological target in any of several target organs. The interpretation of these data can be greatly enhanced by a clear understanding of how this system functions and what the immediate and long-term ramifications are to organs and organ systems. This review focuses on how drugs modify the hepatic cytochrome P450 system, how those modifications are detected, the various consequences of these modifications, and some differences in the induction response among species.

SELECTION OF CITATIONS
SEARCH DETAIL
...