Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
2.
Drug Chem Toxicol ; 38(2): 212-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24975547

ABSTRACT

Fifty two steroids and 9 Vitamin D analogs were docked into ten crystallographically-defined DNA dinucleotide sites and two human topoisomerase II ATP binding sites using two computational programs, Autodock and Surflex. It is shown that both steroids and Vitamin D analogs exhibit a propensity for non-covalent intercalative binding to DNA. A higher predicted binding affinity was found, however, for steroids and the ATP binding site of topoisomerase; in fact these drugs exhibited among the highest topo II binding observed in over 1370 docked drugs. These findings along with genotoxicity data from 26 additional steroids not subjected to docking analysis, support a mechanism wherein the long known, but poorly understood, clastogenicity of steroids may be attributable to inhibition of topoisomerase. A "proof of principle" experiment with dexamethasone demonstrated this to be the likely mechanism of clastogenicity of, at least, this steroid. The generality of this proposed mechanism of genotoxicity across the steroids and vitamin-D analogs is discussed.


Subject(s)
DNA Topoisomerases, Type II/metabolism , DNA/metabolism , Steroids/toxicity , Vitamin D/toxicity , Binding Sites , Crystallography/methods , Humans , Molecular Docking Simulation , Mutagenicity Tests , Software , Steroids/metabolism , Vitamin D/analogs & derivatives , Vitamin D/metabolism
3.
Environ Mol Mutagen ; 55(9): 679-88, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25044351

ABSTRACT

Predictive toxicology plays an important role in the assessment of toxicity of chemicals and the drug development process. While there are several well-established in vitro and in vivo assays that are suitable for predictive toxicology, recent advances in high-throughput analytical technologies and model systems are expected to have a major impact on the field of predictive toxicology. This commentary provides an overview of the state of the current science and a brief discussion on future perspectives for the field of predictive toxicology for human toxicity. Computational models for predictive toxicology, needs for further refinement and obstacles to expand computational models to include additional classes of chemical compounds are highlighted. Functional and comparative genomics approaches in predictive toxicology are discussed with an emphasis on successful utilization of recently developed model systems for high-throughput analysis. The advantages of three-dimensional model systems and stem cells and their use in predictive toxicology testing are also described.


Subject(s)
Genomics/methods , Toxicology/methods , Animals , Chickens , Computer Simulation , DNA Repair/drug effects , High-Throughput Screening Assays/methods , Humans , Lymphocytes/drug effects , Models, Theoretical , Mutagenicity Tests , Stem Cells/drug effects , Yeasts/drug effects , Yeasts/genetics
4.
Environ Mol Mutagen ; 54(8): 668-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23893771

ABSTRACT

Noncovalent chemical/DNA interactions, for example, intercalation and groove-binding, may be more important to genomic integrity than previously appreciated, and there may very well be genotoxic consequences of that binding. It is of importance, then, to develop methods allowing a determination or prediction of such interactions. This would have particular utility in the pharmaceutical industry where genotoxicity is, for the most part, disallowed in new drug entities. We have previously used DNA docking simulations to assess if molecules had structure and charge characteristics which could accommodate noncovalent binding via, for example, electrostatic/hydrogen bonding. We here extend those earlier studies by examining a series of over 1,350 "launched" drugs for ability to noncovalently bind 10 different DNA sequences using two computational programs: Autodock and Surflex. These drugs were also evaluated for binding to the crystallographic ATP-binding site of human topoisomerase II. The results obtained clearly demonstrate multiple series of noncovalent DNA binding structure activity relationships which would not have been predicted based on cursory structural examination. Many drugs within these series are genotoxic although not via any commonly recognized structural covalent alerts. The present studies confirm previously implicated features such as N-dialkyl groups and specific N-aryl ketones as potential genotoxic chemical moieties acting through noncovalent mechanisms. These initial studies provide considerable evidence that DNA intercalation may be an important, largely overlooked, source of drug-induced genotoxicity and further suggest involvement of topoisomerase in that genotoxicity.


Subject(s)
Computer Simulation , DNA/chemistry , Drug Interactions , Intercalating Agents/chemistry , Models, Chemical , Binding Sites , Crystallography, X-Ray , DNA Topoisomerases, Type II/chemistry , Humans , Mutagenicity Tests
5.
Arthritis Rheum ; 63(12): 3988-97, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21953143

ABSTRACT

OBJECTIVE: To identify biomarkers that distinguish between active antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) and remission in a manner superior or complementary to established markers of systemic inflammation. METHODS: Markers of vascular injury and angiogenesis were measured before and after treatment in a large clinical trial in AAV: 163 subjects enrolled in the Rituximab in ANCA-Associated Vasculitis trial were screened for the present study. Serum levels of E-selectin, intercellular adhesion molecule 3 matrix metalloproteinase protein 1 (MMP-1), MMP-3, MMP-9, P-selectin, thrombomodulin, and vascular endothelial growth factor were measured at study screening (time of active disease) and at month 6. Erythrocyte sedimentation rate (ESR) and C-reactive protein (CRP) levels had been measured at the time of the clinical visit. The primary outcome measure was the difference in marker level between screening and month 6 among patients whose disease was in remission (Birmingham Vasculitis Activity Score for Wegener's granulomatosis [BVAS/WG] score of 0) at month 6. RESULTS: All patients had severe active vasculitis at screening (mean ± SD BVAS/WG score 8.6 ± 3.2). Among the 123 patients whose disease was clinically in remission at month 6, levels of all markers except E-selectin showed significant declines. MMP-3 levels were also higher among the 23 patients with active disease at month 6 than among the 123 patients whose disease was in remission. MMP-3 levels correlated weakly with ESR and CRP levels. CONCLUSION: Many markers of vascular injury and angiogenesis are elevated in severe active AAV and decline with treatment, but MMP-3 appears to distinguish active AAV from remission better than the other markers studied. Further study of MMP-3 is warranted to determine its clinical utility in combination with conventional markers of inflammation and ANCA titers.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/blood , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/physiopathology , E-Selectin/blood , Matrix Metalloproteinase 3/blood , Neovascularization, Pathologic/blood , Vascular System Injuries/blood , Adult , Aged , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/drug therapy , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antirheumatic Agents/therapeutic use , Biomarkers/blood , Blood Sedimentation , C-Reactive Protein/metabolism , Case-Control Studies , Diagnosis, Differential , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neovascularization, Pathologic/physiopathology , Remission Induction , Rituximab , Vascular System Injuries/physiopathology
6.
Environ Mol Mutagen ; 52(5): 339-54, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21538556

ABSTRACT

A symposium at the 40th anniversary of the Environmental Mutagen Society, held from October 24-28, 2009 in St. Louis, MO, surveyed the current status and future directions of genetic toxicology. This article summarizes the presentations and provides a perspective on the future. An abbreviated history is presented, highlighting the current standard battery of genotoxicity assays and persistent challenges. Application of computational toxicology to safety testing within a regulatory setting is discussed as a means for reducing the need for animal testing and human clinical trials, and current approaches and applications of in silico genotoxicity screening approaches across the pharmaceutical industry were surveyed and are reported here. The expanded use of toxicogenomics to illuminate mechanisms and bridge genotoxicity and carcinogenicity, and new public efforts to use high-throughput screening technologies to address lack of toxicity evaluation for the backlog of thousands of industrial chemicals in the environment are detailed. The Tox21 project involves coordinated efforts of four U.S. Government regulatory/research entities to use new and innovative assays to characterize key steps in toxicity pathways, including genotoxic and nongenotoxic mechanisms for carcinogenesis. Progress to date, highlighting preliminary test results from the National Toxicology Program is summarized. Finally, an overview is presented of ToxCast™, a related research program of the U.S. Environmental Protection Agency, using a broad array of high throughput and high content technologies for toxicity profiling of environmental chemicals, and computational toxicology modeling. Progress and challenges, including the pressing need to incorporate metabolic activation capability, are summarized.


Subject(s)
Environmental Monitoring/methods , Toxicogenetics/methods , Models, Theoretical , Toxicogenetics/trends , United States , United States Environmental Protection Agency
7.
Angle Orthod ; 81(3): 383-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21261488

ABSTRACT

OBJECTIVE: Utilize American Board of Orthodontics (ABO) cast/radiographic evaluation (CRE) to compare a series of 63 consecutive patients, finished with manual wire bending (conventional) treatment, vs a subsequent series of 69 consecutive patients, finished by the same orthodontist using the SureSmile™ (SS) method. MATERIALS AND METHODS: Records of 132 nonextraction patients were scored by a calibrated examiner blinded to treatment mode. Age and discrepancy index (DI) between groups were compared by t-tests. A chi-square test was used to compare for differences in sex and whether the patient was treated using braces only (no orthopedic correction). Analysis of covariance tested for differences in CRE outcomes and treatment times, with sex and DI included as covariates. A logarithmic transformation of CRE outcomes and treatment times was used because their distributions were skewed. Significance was defined as P < .05. RESULTS: Compared with conventional finishing, SS patients had significantly lower DI scores, less treatment time (∼7 months), and better CRE scores for first-order alignment-rotation and interproximal space closure; however, second-order root angulation (RA) was inferior. CONCLUSION: SS patients were treated in less time to better CRE scores for first-order rotation (AR) and interproximal space closure (IC) but on the average, malocclusions were less complex and second order root alignment was inferior, compared with patients finished with manual wire bending.


Subject(s)
Computer-Aided Design , Orthodontic Appliance Design , Orthodontic Appliances , Orthodontics, Corrective/methods , Therapy, Computer-Assisted , Analysis of Variance , Child , Female , Humans , Male , Malocclusion/therapy , Orthodontic Wires , Orthodontics, Corrective/instrumentation , Single-Blind Method , Time Factors , Treatment Outcome
8.
Birth Defects Res B Dev Reprod Toxicol ; 89(6): 474-84, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21058326

ABSTRACT

BACKGROUND: The effects of histamine H1 antagonist chlorcyclizine on rat palate development were characterized following in utero exposure. METHODS: To identify the optimum dose for inducing cleft palate, pregnant rats were administered 30, 60, or 90 mg/kg chlorcyclizine on Gestation Days 11 to 14. Fetal palate gene expression was also assessed after 90 mg/kg chlorcyclizine at 8, 15 and 30 hours post-dose on Gestation Day 14 using microarray and qRT-PCR. RESULTS: Rats in the 60- and 90-mg/kg groups exhibited adverse clinical signs and body weight loss. Rats in the 90-mg/kg group also demonstrated increases in late resorptions and decreases in fetal weight. Effects in the low-dose group were limited to decreases in body weight gain. Fetal assessment on Gestation Day 21 revealed that findings were limited to the 60- and 90-mg/kg groups, and included cleft palate (80% of litters for both groups), high arched palate, small nose, micrognathia, high domed head, digits shortened/absent and small limb. The fetal incidence of cleft palate was higher at 90 mg/kg, thus this dose was selected to assess palate gene expression. The altered genes associated with chlorcyclizine-induced cleft palate included Wnt5a, Bmp2, Bmp4, Fgf10, Fgfr2, Msx1, and Insig1 but the magnitude of the change was relatively small (1.5- to 2-fold). CONCLUSIONS: Expression of several genes involved in palate, limb and digit development was altered in the fetal palate following in utero exposure to chlorcyclizine. The subtle perturbation and interplay of these genes may have profound effects on the dynamics of fetal palate development.


Subject(s)
Cleft Palate/chemically induced , Embryo, Mammalian/drug effects , Histamine H1 Antagonists/toxicity , Palate/drug effects , Piperazines/toxicity , Animals , Biomarkers/metabolism , Cleft Palate/genetics , Cleft Palate/pathology , Embryo, Mammalian/abnormalities , Female , Fetal Resorption/chemically induced , Fetal Weight/drug effects , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Maternal Exposure , Microarray Analysis , Palate/abnormalities , Palate/metabolism , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Inbred Strains , Weight Gain
9.
Environ Mol Mutagen ; 51(8-9): 800-14, 2010.
Article in English | MEDLINE | ID: mdl-20872827

ABSTRACT

The battery of regulatory genotoxicity studies required in support of new drug registration has remained largely static since its inception some thirty years ago. The Ames mutagenicity assay still forms the foundation for this testing being highly reproducible and, for the most part, transparent. The same is not necessarily true of in vitro mammalian chromosome aberration assays since there is a fairly large and growing number of molecules without clear structural genotoxicity alerts (DEREK, MCASE), which are negative in Ames testing but positive in aberration studies, often only at high concentrations and/or cytotoxicity. Interpretation and risk assessment of these positive results can be problematic since there is no clear understanding of the process that generates them. The present paper builds on our previous observations suggesting that non covalent drug/DNA interactions, which are not adequately modeled in computational programs, may help explain some of these unexpected positive results. In particular, it is suggested that N-dimethyl groups and certain pyridine/piperidine aryl ketones may play a contributory role in genotoxicity, perhaps via DNA intercalation and topoisomerase inhibition. Clastogenicity arising from topoisomerase inhibition would be expected to be a threshold phenomenon and, as such, may carry a distinctly reduced risk relative to clastogenicity associated with covalent drug/DNA interactions.


Subject(s)
Mutagens/chemistry , Pharmaceutical Preparations/chemistry , Drug-Related Side Effects and Adverse Reactions , Humans , Ketones/chemistry , Molecular Structure , Mutagenicity Tests , Piperidines/chemistry , Pyridines/chemistry
10.
Toxicol Pathol ; 38(5): 738-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20585143

ABSTRACT

Treatment with drugs from multiple classes induces vascular injury with medial necrosis, hemorrhage, endothelial damage, and inflammation. Previous research has suggested early events might be occurring well in advance of the full lesions that appear forty-eight to seventy-two hours after dosing with SCH 351591, a PDE IV inhibitor. This study was performed to study early events in detail. Rats were dosed with 20 mg/kg of drug by gavage and sacrificed at times between fifteen and 240 minutes after dosing. Tissues were collected for histopathological analysis and gene expression studies. Serum was collected for biomarker analysis. The data from biomarker analysis showed a three-part response with an early phase that was maximal at fifteen to thirty minutes, a second phase from forty-five to 180 minutes, and the third phase that was starting to rise at four hours. The first phase included increases in lymphocytes, serum histamine, and serum nitrite. The second phase shows continued elevation of serum nitrite. The third phase was marked by an increase in serum GRO/CINC-1. At fifteen minutes, histopathology showed activation of mast cells, but not degranulation. Increases in endothelial activation and perivascular inflammatory cells were first apparent at thirty minutes and increased through 240 minutes.


Subject(s)
Cyclic N-Oxides/toxicity , Inflammation/chemically induced , Phosphodiesterase 4 Inhibitors/toxicity , Quinolines/toxicity , Vascular System Injuries/chemically induced , Animals , Biomarkers/analysis , Biomarkers/blood , Gene Expression/drug effects , Inflammation/pathology , Mesentery/blood supply , Mesentery/pathology , Rats , Rats, Sprague-Dawley , Time , Vascular System Injuries/blood , Vascular System Injuries/pathology
11.
Environ Health Perspect ; 118(7): 992-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20100679

ABSTRACT

BACKGROUND: In spite of the application of toxicogenomic (TGx) data to the field of toxicology for the past 10 years, the broad implementation and full impact of TGx for chemical and drug evaluation to improve decision making within organizations and by policy makers has not been achieved. OBJECTIVES: The goal of the Health and Environmental Sciences Institute (HESI) Committee on the Application of Genomics to Mechanism-based Risk Assessment was to construct and summarize a multisector survey, addressing key issues and perspectives on the current and future practical uses and challenges of implementing TGx data to facilitate discussions for decision making within organizations and by policy makers. METHODS: An online survey to probe the current status and future challenges facing the field of TGx for drug and chemical evaluation in experimental and nonclinical models was taken by scientists and scientific decision/policy makers actively engaged in the field of TGx within industrial, academic, and regulatory sectors of the United States, Europe, and Japan. For this survey, TGx refers specifically to the analysis of gene expression responses to evaluate xenobiotic exposure in experimental and preclinical models. RESULTS: The survey results are summarized from questions covering broad areas including technology used, organizational capacity and resource allocation, experimental approaches, data storage and exchange, perceptions of benefits and hurdles, and future expectations. CONCLUSIONS: The survey findings provide valuable information on the current state of the science of TGx applications and identify key areas in which TGx will have an impact as well as the key hurdles in applying TGx data to address issues. The findings serve as a public resource to facilitate discussions on the focus of future TGx efforts to ensure that a maximal benefit can be obtained from toxicogenomic studies.


Subject(s)
Risk Assessment/methods , Toxicogenetics/methods , Toxicogenetics/trends , Administrative Personnel , Data Collection , Europe , Gene Expression Regulation/drug effects , Humans , Internet , Japan , Medical Laboratory Personnel , United States , Xenobiotics/toxicity
12.
Cytometry A ; 75(11): 924-33, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19802874

ABSTRACT

Drug resistant tumor "side-populations," enriched in cancer stem cells and identified by reduced accumulation of Hoechst 33342 under ABCG2-mediated efflux, may compromise therapeutic outcome. Side-population cells have predicted resistance to minor groove ligands, including the DNA topoisomerase I poison topotecan. We have used a stable Hoechst 33342-resistant murine L cell system (HoeR415) to study resistance patterns, removing the need for SP isolation before microarray analysis of gene expression and the tracking of cell cycle dynamics and cytotoxicity. The majority of HoeR415 cells displayed a side-population phenotype comparable with that of the side-population resident in the ABCG2 over-expressing A549 lung cancer cell line. Photo-crosslinking showed direct protection against minor groove ligand residence on DNA, driven by ABCG2-mediated efflux and not arising from any binding competition with endogenous polyamines. The covalent minor-groove binding properties of the drug FCE24517 (tallimustine) prevented resistance suggesting a mechanism for overcoming SP-related drug resistance. Hoechst 33342-resistant murine cells showed lower but significant crossresistance to topotecan, again attributable to enhanced ABCG2 expression, enabling cells to evade S-phase arrest. Hoechst 33342/TPT-resistant cells showed limited ancillary gene expression changes that could modify cellular capacity to cope with chronic stress including over-expression of Aldh1a1 and Mgst1, but under-expression of Plk2 and Nnt. There was no evidence to link the putative stem cell marker ALDH1A1 with any augmentation of the TPT resistance phenotype. The study has implications for the patterns of drug resistance arising during tumor repopulation and the basal resistance to minor groove-binding drugs of tumor side-populations.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Benzimidazoles/pharmacology , Flow Cytometry/methods , Topotecan/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Cell Line, Tumor , Cell Separation , Cross-Linking Reagents/pharmacology , Distamycins/pharmacology , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/pharmacology , Humans , Ligands , Mice , Nitrogen Mustard Compounds/pharmacology , Polyamines/chemistry
13.
Drug Chem Toxicol ; 32(1): 17-20, 2009.
Article in English | MEDLINE | ID: mdl-19514934

ABSTRACT

The 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A reductase inhibitor, lovastatin (lova), has been reported to both sensitize to, and protect against, the toxic effects of the antitumor anthracycline doxorubicin (dox) in cellular and in vivo systems. The mechanism by which these effects occur has not yet been determined. In the present study, lova is shown to enhance the genotoxicity of dox in the V79 cell in vitro micronucleus assay and to do so, most likely, via noncovalent interaction with DNA adjacent to sites of dox binding. These studies confirm and extend the experimental evidence strongly suggesting the importance of noncovalent drug/DNA interactions in cellular responses to genotoxic stimuli.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Lovastatin/toxicity , Mutagens/toxicity , Animals , Cell Line , Cricetinae , Cricetulus , DNA/drug effects , DNA/metabolism , Drug Synergism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/toxicity , Micronucleus Tests
14.
J Pharmacol Toxicol Methods ; 60(1): 69-78, 2009.
Article in English | MEDLINE | ID: mdl-19422924

ABSTRACT

Identification of novel new molecules which hold the greatest promise of safe and effective therapies remains a continuous challenge to the pharmaceutical industry. This has led the industry to implement strategies for identification of the most promising candidates during the discovery phase and for their safe and expeditious advancement through development. Testing for potential liable properties in the discovery phase has included the evaluation of major areas of pharmaceutics that have led to failure such as its physical and pharmaceutical properties, drug metabolism and pharmacokinetic characteristics, various safety endpoints including pre-development safety pharmacology, general toxicology and genetic toxicology and interrogation of counter-screen data to identify off-target affinities (i.e., receptors, ion channels, transporters, kinases, etc.) that pose a concern. Amongst the many important areas of concern is the potential for toxicities of the major organ systems. To mitigate this concern, a strategy pursued is to identify the prominent toxicological properties of the candidate prior to its recommendation for development. The results of these studies in discovery allow exclusion of the candidate before the expenditure of resources and time typical of development. In addition, the discovery phase toxicology studies serve to address key questions that may have arisen from the study of another molecule, the phenotypic profile from pre-clinical models where the therapeutic target has been genetically modified or concerns that have been raised as a result of other investigations. Importantly, the results of the exploratory drug safety studies will be used by the sponsor to judge the potential risks associated with continued pursuit of a potential development candidate. In many ways, pre-clinical toxicological investigations in discovery serve the important objective of identifying the most promising candidates to progress into development and onto registration.


Subject(s)
Drug Discovery , Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions , Toxicology/methods , Animals , Drug Industry , Pharmaceutical Preparations/metabolism , Research Design
15.
Toxicol Sci ; 110(2): 341-52, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19465456

ABSTRACT

The genotoxicity testing battery is highly sensitive for detection of chemical carcinogens. However, it features a low specificity and provides only limited mechanistic information required for risk assessment of positive findings. This is especially important in case of positive findings in the in vitro chromosome damage assays, because chromosome damage may be also induced secondarily to cell death. An increasing body of evidence indicates that toxicogenomic analysis of cellular stress responses provides an insight into mechanisms of action of genotoxicants. To evaluate the utility of such a toxicogenomic analysis we evaluated gene expression profiles of TK6 cells treated with four model genotoxic agents using a targeted high density real-time PCR approach in a multilaboratory project coordinated by the Health and Environmental Sciences Institute Committee on the Application of Genomics in Mechanism-based Risk Assessment. We show that this gene profiling technology produced reproducible data across laboratories allowing us to conclude that expression analysis of a relevant gene set is capable of distinguishing compounds that cause DNA adducts or double strand breaks from those that interfere with mitotic spindle function or that cause chromosome damage as a consequence of cytotoxicity. Furthermore, our data suggest that the gene expression profiles at early time points are most likely to provide information relevant to mechanisms of genotoxic damage and that larger gene expression arrays will likely provide richer information for differentiating molecular mechanisms of action of genotoxicants. Although more compounds need to be tested to identify a robust molecular signature, this study confirms the potential of toxicogenomic analysis for investigation of genotoxic mechanisms.


Subject(s)
DNA Damage , Gene Expression Profiling , Gene Expression Regulation/drug effects , Laboratories , Mutagenicity Tests/methods , Mutagens/toxicity , Polymerase Chain Reaction , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromosome Aberrations/chemically induced , Cisplatin/toxicity , Cluster Analysis , DNA Adducts/metabolism , DNA Breaks, Double-Stranded , Dose-Response Relationship, Drug , Etoposide/toxicity , Gene Expression Profiling/standards , Humans , Laboratories/standards , Mutagenicity Tests/standards , Observer Variation , Paclitaxel/toxicity , Polymerase Chain Reaction/standards , Reproducibility of Results , Risk Assessment , Sodium Chloride/toxicity , Spindle Apparatus/drug effects , Time Factors
16.
Environ Mol Mutagen ; 50(6): 435-50, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19334052

ABSTRACT

Information from the 1999 through 2008 Physicians' Desk Reference (PDR) was used to evaluate the genotoxicity of marketed drugs. Where available, data regarding the rodent carcinogenicity results were included (PDR and Gold potency database). In addition, computational predictivity of genotoxicity (DEREK, MC4PC) is included and expanded upon from two previous reviews. The present paper contains genotoxicity data on 545 marketed drugs. Excluded from analysis were most cytotoxic anti-cancer and antiviral drugs, nucleosides (all with known mechanistic genotoxicity), steroids with class-specific genotoxicity and biologicals or peptide-based drugs. Per assay type, the percentage of positive drugs was: Bacterial mutagenesis assay: 38/525 (7.1%), in vitro chromosome aberrations: 88/380 (26.1%); mouse lymphoma assays (MLA): 32/163 (19.1%), in vivo cytogenetics: 49/438 (11.1%). The relationship among positive genetic toxicity findings, rodent carcinogenicity, and in silico prediction is discussed. Finally, supporting evidence is presented for the idea that the presence of an N-dialkyl group or piperidine aryl ketone may somehow be associated with genotoxicity, perhaps through DNA intercalation and consequent DNA topoisomerase II inhibition.


Subject(s)
Computational Biology , Drug-Related Side Effects and Adverse Reactions , Marketing , Animals , Carcinogenicity Tests , Chromosome Aberrations/drug effects , Mice , Mutagenicity Tests , Rats
17.
Chem Biol Interact ; 176(2-3): 252-60, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-19086139

ABSTRACT

Carcinogenicity of chemicals can currently only be evaluated in 2-year rodent bioassays. Therefore, the development of early biomarkers for carcinogenesis would result in substantial savings in time and expense. The current study investigates whether early changes in gene expression may be developed as markers for cancer. Animals were treated for 1 or 5 days with either non-genotoxic carcinogens or non-carcinogens and gene expression was analyzed by quantitative PCR (qPCR).We tested two gene signatures previously reported to detect non-genotoxic carcinogens. Using one gene signature it was confirmed that 3/3 nongenotoxic carcinogens and 2/2 non-carcinogens are correctly identified with data from 1 or 5 days of dosing. In contrast an alternative signature correctly identified 0/3 and 2/3 nongenotoxic carcinogens at 1 and 5 days of treatment, respectively and 2/2 non-carcinogens at both time-points. Additionally, we evaluated a novel panel of putative biomarker genes, from the literature, many of which have roles in cell growth and division, including myc, cdc2 and mcm6. These genes were significantly induced by non-genotoxic carcinogens and not by non-carcinogens. Using the average fold-induction across this panel, 2/3 non-genotoxic carcinogens were detected on both day 1 and day 5. These data support the idea that acute changes in gene expression may provide biomarkers for non-genotoxic carcinogenesis but also highlight interesting differences in the sensitivities of distinct gene signatures.


Subject(s)
Biological Assay/methods , Biomarkers/analysis , Carcinogens/toxicity , Gene Expression Regulation, Neoplastic/genetics , Predictive Value of Tests , Animals , Carcinogenicity Tests , Dose-Response Relationship, Drug , Male , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Risk Assessment , Time Factors
18.
Drug Chem Toxicol ; 31(4): 501-13, 2008.
Article in English | MEDLINE | ID: mdl-18850359

ABSTRACT

Assessment of cytochrome P450 (CYP) induction at the mRNA level in preclinical rodent studies has gained interest in recent years, but there are still concerns regarding correlations between the mRNA and the enzyme activity levels, especially in mice. The purpose of the present study was to systematically evaluate patterns of temporal changes of CYPs 1a1, 1a2, 2b10, 3a11, and 4a10 at mRNA, protein, and activity levels in order to determine to what extent mRNA levels could be used either qualitatively or quantitatively for the assessment of CYP enzyme induction. In this study, livers from male CD-1 mice treated daily with beta-naphthoflavone, phenobarbital, dexamethasone, clofibrate, and control vehicles were collected for RNA and microsomal analysis after 0.5, 1, 2, 4, and 8 days of daily dose. The results revealed a good correlation among mRNA, protein, and enzyme activity levels, with the best correlation at the time points between Days 2 and 8, suggesting that the appropriate time to monitor CYP mRNA may be beyond Day 2 of chemical treatments. Based on these results, we concluded that the mRNA approach is a useful tool to monitor CYP induction in mice, particularly when treatment duration is beyond 2 days.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Drug Evaluation, Preclinical/methods , Liver/drug effects , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Animals , Clofibrate/pharmacology , Cytochrome P-450 Enzyme System/genetics , Dexamethasone/pharmacology , Enzyme Induction/drug effects , Feasibility Studies , Isoenzymes , Liver/enzymology , Male , Mice , Phenobarbital/pharmacology , Reproducibility of Results , Time Factors , beta-Naphthoflavone/pharmacology
19.
Drug Chem Toxicol ; 31(4): 515-28, 2008.
Article in English | MEDLINE | ID: mdl-18850360

ABSTRACT

A large number of cationic amphiphilic drugs (CADs) are known to cause phospholipidosis (PLD) in vivo. In the present study, we have built upon our previous findings to further qualify the use of a fluorescently labeled phospholipid-based cell-culture assay to detect PLD-inducing drugs. In this paper, we demonstrate that 12 PLD-negative compounds and 11 drugs known to cause PLD in vivo are all correctly identified by using this assay. Interestingly, we found that in cells treated with certain CADs, the fluorescent phospholipid was sequestered in a very specific punctate pattern, which overlapped strongly with the staining pattern seen with a lysosomal marker protein. Our data also show that false positives can be generated with the fluorescence assay when compounds are used at concentrations that cause a >30% decrease in cell number in this assay. Confocal microscopy demonstrated that the staining pattern of fluorescent phospholipids in these cases may be differentiated from those of true positives by the fact that diffuse, rather than punctuate, fluorescence is observed. These studies confirm and expand our previous results showing that the fluorescent phospholipid assay is a highly sensitive, specific tool for detecting PLD-inducing drugs, if care is taken to rule out cytotoxicity-related artifact.


Subject(s)
Drug Evaluation, Preclinical/methods , Fluorescent Dyes/metabolism , Lipidoses/chemically induced , Molecular Probe Techniques , Phospholipids/metabolism , Artifacts , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Lipidoses/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Microscopy, Confocal , Reproducibility of Results , Time Factors
20.
Toxicol Pathol ; 36(6): 827-39, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18776163

ABSTRACT

Histopathological and immunohistochemical studies were conducted to characterize vascular injuries in rats treated with phosphodiesterase (PDE) IV inhibitors SCH 351591 or SCH 534385. Sprague-Dawley rats were administered PDE IV inhibitors by gavage at a range of doses and times. The two PDE IV inhibitors induced comparable levels of vascular injury, primarily in the mesentery and to a lesser extent in the pancreas, kidney, liver, small intestine, and stomach. Mesenteric vascular changes occurred as early as one hour, progressively developed over twenty-four to forty-eight hours, peaked at seventy-two hours, and gradually subsided from seven to nine days. The typical morphology of the vascular toxicity consisted of hemorrhage and necrosis of arterioles and arteries, microvascular injury, fibrin deposition, and perivascular inflammation of a variety of blood vessels. The incidence and severity of mesenteric vascular injury increased in a time- and dose-dependent manner in SCH 351591- or SCH 534385-treated rats. Mesenteric vascular injury was frequently associated with activation of mast cells (MC), endothelial cells (EC), and macrophages (MØ). Immunohistochemical studies showed increases in CD63 immunoreactivity of mesenteric MC and in nitrotyrosine immunoreactivity of mesenteric EC and MØ. The present study also provides a morphological and cellular basis for evaluating candidate biomarkers of drug-induced vascular injury.


Subject(s)
Blood Vessels/drug effects , Cyclic N-Oxides/toxicity , Phosphodiesterase 4 Inhibitors , Phosphodiesterase Inhibitors/toxicity , Quinolines/toxicity , Vascular Diseases/chemically induced , Vascular Diseases/pathology , Animals , Immunohistochemistry , Intestine, Small/blood supply , Intestine, Small/pathology , Kidney/blood supply , Kidney/pathology , Mesenteric Arteries/pathology , Pancreas/blood supply , Pancreas/pathology , Rats , Rats, Sprague-Dawley , Statistics, Nonparametric , Stomach/blood supply , Stomach/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...