Laura Custer
Bristol-Myers Squibb
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Laura Custer.
Mutation Research-genetic Toxicology and Environmental Mutagenesis | 2010
Andreas Rothfuss; Mike O’Donovan; Marlies De Boeck; Dominique Brault; Andreas Czich; Laura Custer; Shuichi Hamada; Ulla Plappert-Helbig; Makoto Hayashi; Jonathan Howe; Andrew R. Kraynak; Bas-jan van der Leede; Madoka Nakajima; Catherine C. Priestley; Véronique Thybaud; Kazuhiko Saigo; Satin Sawant; Jing Shi; Richard D. Storer; Melanie Struwe; Esther Vock; Sheila M. Galloway
A collaborative trial was conducted to evaluate the possibility of integrating the rat-liver Comet assay into repeat-dose toxicity studies. Fourteen laboratories from Europe, Japan and the USA tested fifteen chemicals. Two chemicals had been previously shown to induce micronuclei in an acute protocol, but were found negative in a 4-week Micronucleus (MN) Assay (benzo[a]pyrene and 1,2-dimethylhydrazine; Hamada et al., 2001); four genotoxic rat-liver carcinogens that were negative in the MN assay in bone marrow or blood (2,6-dinitrotoluene, dimethylnitrosamine, 1,2-dibromomethane, and 2-amino-3-methylimidazo[4,5-f]quinoline); three compounds used in the ongoing JaCVAM (Japanese Center for the Validation of Alternative Methods) validation study of the acute liver Comet assay (2,4-diaminotoluene, 2,6-diaminotoluene and acrylamide); three pharmaceutical-like compounds (chlordiazepoxide, pyrimethamine and gemifloxacin), and three non-genotoxic rodent liver carcinogens (methapyrilene, clofibrate and phenobarbital). Male rats received oral administrations of the test compounds, daily for two or four weeks. The top dose was meant to be the highest dose producing clinical signs or histopathological effects without causing mortality, i.e. the 28-day maximum tolerated dose. The liver Comet assay was performed according to published recommendations and following the protocol for the ongoing JaCVAM validation trial. Laboratories provided liver Comet assay data obtained at the end of the long-term (2- or 4-week) studies together with an evaluation of liver histology. Most of the test compounds were also investigated in the liver Comet assay after short-term (1-3 daily) administration to compare the sensitivity of the two study designs. MN analyses were conducted in bone marrow or peripheral blood for most of the compounds to determine whether the liver Comet assay could complement the MN assay for the detection of genotoxins after long-term treatment. Most of the liver genotoxins were positive and the three non-genotoxic carcinogens gave negative result in the liver Comet assay after long-term administration. There was a high concordance between short- and long-term Comet assay results. Most compounds when tested up to the maximum tolerated dose were correctly detected in both short- and long-term studies. Discrepant results were obtained with 2,6 diaminotoluene (negative in the short-term, but positive in the long-term study), phenobarbital (positive in the short-term, but negative in the long-term study) and gemifloxacin (positive in the short-term, but negative in the long-term study). The overall results indicate that the liver Comet assay can be integrated within repeat-dose toxicity studies and efficiently complements the MN assay in detecting genotoxins. Practical aspects of integrating genotoxicity endpoints into repeat-dose studies were evaluated, e.g. by investigating the effect of blood sampling, as typically performed during toxicity studies, on the Comet and MN assays. The bleeding protocols used here did not affect the conclusions of the Comet assay or of the MN assays in blood and bone marrow. Although bleeding generally increased reticulocyte frequencies, the sensitivity of the response in the MN assay was not altered. These findings indicate that all animals in a toxicity study (main-study animals as well as toxicokinetic (TK) satellite animals) could be used for evaluating genotoxicity. However, possible logistical issues with scheduling of the necropsies and the need to conduct electrophoresis promptly after tissue sampling suggest that the use of TK animals could be simpler. The data so far do not indicate that liver proliferation or toxicity confound the results of the liver Comet assay. As was also true for other genotoxicity assays, criteria for evaluation of Comet assay results and statistical analyses differed among laboratories. Whereas comprehensive advice on statistical analysis is available in the literature, agreement is needed on applying consistent criteria.
Regulatory Toxicology and Pharmacology | 2013
Andreas Sutter; Alexander Amberg; Scott Boyer; Alessandro Brigo; Joseph F. Contrera; Laura Custer; Krista L. Dobo; Véronique Gervais; Susanne Glowienke; Jacky Van Gompel; Nigel Greene; Wolfgang Muster; John Nicolette; M. Vijayaraj Reddy; Véronique Thybaud; Esther Vock; Angela White; Lutz Müller
Genotoxicity hazard identification is part of the impurity qualification process for drug substances and products, the first step of which being the prediction of their potential DNA reactivity using in silico (quantitative) structure-activity relationship (Q)SAR models/systems. This white paper provides information relevant to the development of the draft harmonized tripartite guideline ICH M7 on potentially DNA-reactive/mutagenic impurities in pharmaceuticals and their application in practice. It explains relevant (Q)SAR methodologies as well as the added value of expert knowledge. Moreover, the predictive value of the different methodologies analyzed in two surveys conveyed in the US and European pharmaceutical industry is compared: most pharmaceutical companies used a rule-based expert system as their primary methodology, yielding negative predictivity values of ⩾78% in all participating companies. A further increase (>90%) was often achieved by an additional expert review and/or a second QSAR methodology. Also in the latter case, an expert review was mandatory, especially when conflicting results were obtained. Based on the available data, we concluded that a rule-based expert system complemented by either expert knowledge or a second (Q)SAR model is appropriate. A maximal transparency of the assessment process (e.g. methods, results, arguments of weight-of-evidence approach) achieved by e.g. data sharing initiatives and the use of standards for reporting will enable regulators to fully understand the results of the analysis. Overall, the procedures presented here for structure-based assessment are considered appropriate for regulatory submissions in the scope of ICH M7.
Environmental and Molecular Mutagenesis | 2011
Stephen D. Dertinger; Souk Phonethepswath; Pamela Weller; John Nicolette; Joel Murray; Paul Sonders; Hans-Werner Vohr; Jing Shi; Ljubica Krsmanovic; Carol Gleason; Laura Custer; Andrew Henwood; Kevin Sweder; Leon F. Stankowski; Daniel J. Roberts; Amanda Giddings; Julia Kenny; Anthony M. Lynch; Céline Defrain; Fabrice Nesslany; Bas‐jan M. van der Leede; Terry Van Doninck; Ann Schuermans; Kentaro Tanaka; Yoshie Hiwata; Osamu Tajima; Eleanor Wilde; Azeddine Elhajouji; William C. Gunther; Catherine Thiffeault
A collaborative international trial was conducted to evaluate the reproducibility and transferability of an in vivo mutation assay based on the enumeration of CD59‐negative rat erythrocytes, a phenotype that is indicative of Pig‐a gene mutation. Fourteen laboratories participated in this study, where anti‐CD59‐PE, SYTO 13 dye, and flow cytometry were used to determine the frequency of CD59‐negative erythrocytes (RBCCD59−) and CD59‐negative reticulocytes (RETCD59−). To provide samples with a range of mutant phenotype cell frequencies, male rats were exposed to N‐ethyl‐N‐nitrosourea (ENU) via oral gavage for three consecutive days (Days 1–3). Each laboratory studied 0, 20, and 40 mg ENU/kg/day (n = 5 per group). Three sites also evaluated 4 mg/kg/day. At a minimum, blood samples were collected three times: predosing and on Days 15 and 30. Blood samples were processed according to a standardized sample processing and data acquisition protocol, and three endpoints were measured: %reticulocytes, frequency of RETCD59−, and frequency of RBCCD59−. The methodology was found to be reproducible, as the analysis of technical replicates resulted in experimental coefficients of variation that approached theoretical values. Good transferability was evident from the similar kinetics and magnitude of the dose‐related responses that were observed among different laboratories. Concordance correlation coefficients showed a high level of agreement between the reference site and the test sites (range: 0.87–0.99). Collectively, these data demonstrate that with adequate training of personnel, flow cytometric analysis is capable of reliably enumerating mutant phenotype erythrocytes, thereby providing a robust in vivo mutation assay that is readily transferable across laboratories. Environ. Mol. Mutagen. 2011.
Environmental and Molecular Mutagenesis | 2011
Kerry L. Dearfield; Véronique Thybaud; Michael C. Cimino; Laura Custer; Andreas Czich; James Harvey; Susan D. Hester; James H. Kim; David Kirkland; Dan D. Levy; Elisabeth Lorge; Martha M. Moore; Gladys Ouédraogo-Arras; Maik Schuler; Willi Suter; Kevin Sweder; Kirk Tarlo; Jan van Benthem; Freddy Van Goethem; Kristine L. Witt
Appropriate follow‐up actions and decisions are needed when evaluating and interpreting clear positive results obtained in the in vitro assays used in the initial genotoxicity screening battery (i.e., the battery of tests generally required by regulatory authorities) to assist in overall risk‐based decision making concerning the potential effects of human exposure to the agent under test. Over the past few years, the International Life Sciences Institute (ILSI) Health and Environmental Sciences Institute (HESI) Project Committee on the Relevance and Follow‐up of Positive Results in In Vitro Genetic Toxicity (IVGT) Testing developed a decision process flow chart to be applied in case of clear positive results in vitro. It provides for a variety of different possibilities and allows flexibility in choosing follow‐up action(s), depending on the results obtained in the initial battery of assays and available information. The intent of the Review Subgroup was not to provide a prescriptive testing strategy, but rather to reinforce the concept of weighing the totality of the evidence. The Review Subgroup of the IVGT committee highlighted the importance of properly analyzing the existing data, and considering potential confounding factors (e.g., possible interactions with the test systems, presence of impurities, irrelevant metabolism), and chemical modes of action when analyzing and interpreting positive results in the in vitro genotoxicity assays and determining appropriate follow‐up testing. The Review Subgroup also examined the characteristics, strengths, and limitations of each of the existing in vitro and in vivo genotoxicity assays to determine their usefulness in any follow‐up testing. Environ. Mol. Mutagen., 2011.
Environmental and Molecular Mutagenesis | 2011
Anthony M. Lynch; Amanda Giddings; Laura Custer; Carol Gleason; Andrew Henwood; Mike Aylott; Julia Kenny
N‐methyl‐N‐nitrosourea (MNU) was evaluated in the in vivo Pig‐a mutation assay as part of an International Collaborative Trial to investigate laboratory reproducibility, 28‐day study integration, and comparative analysis with micronucleus (MN), comet, and clinical pathology endpoints. Male Sprague Dawley rats were treated for 28 days with doses of 0, 2.5, 5, and 10 mg MNU/kg/day in two independent laboratories, GlaxoSmithKline (GSK) and Bristol Myers Squibb (BMS). Additional studies investigated the low‐dose region (<2.5 mg/kg/day). Reticulocytes were evaluated for Pig‐a phenotypic mutation, CD59‐negative reticulocytes/erythrocytes (RETsCD59−/ RBCsCD59−) on Days 1, 4, 15, 29, 43, and 57, and for micronucleated reticulocytes (MN‐RETs) on Days 4 and 29. Comet analysis was conducted for liver and whole blood, and hematology and clinical chemistry was investigated. Dose‐dependent increases in the frequency of RETsCD59− and RBCsCD59− were observed by Day 15 or 29, respectively. Dose‐dependent increases were observed in %MN‐RET on Days 4 and 29, and in mean %tail intensity in liver and in blood. Hematology/clinical chemistry data demonstrated bone marrow toxicity. Data comparison between GSK and BMS indicated a high degree of concordance with the Pig‐a mutation assay results, consistent with previous observations with MNU and N‐ethyl‐N‐nitrosourea. These data confirm that complementary genotoxicity endpoints can be effectively incorporated into routine toxicology studies, a strategy that can provide information on gene mutation, chromosome damage, and DNA strand breaks in a single repeat dose rodent study. Collectively, this would reduce animal usage while providing valuable genetic toxicity information within the context of other toxicological endpoints. Environ. Mol. Mutagen., 2011.
Regulatory Toxicology and Pharmacology | 2015
Chris Barber; Alexander Amberg; Laura Custer; Krista L. Dobo; Susanne Glowienke; Jacky Van Gompel; Steve Gutsell; Jim Harvey; Masamitsu Honma; Michelle O. Kenyon; Naomi L. Kruhlak; Wolfgang Muster; Lidiya Stavitskaya; Andrew Teasdale; Jonathan D. Vessey; Joerg Wichard
The ICH M7 guidelines for the assessment and control of DNA reactive (mutagenic) impurities in pharmaceuticals allows for the consideration of in silico predictions in place of in vitro studies. This represents a significant advance in the acceptance of (Q)SAR models and has resulted from positive interactions between modellers, regulatory agencies and industry with a shared purpose of developing effective processes to minimise risk. This paper discusses key scientific principles that should be applied when evaluating in silico predictions with a focus on accuracy and scientific rigour that will support a consistent and practical route to regulatory submission.
Regulatory Toxicology and Pharmacology | 2016
Alexander Amberg; Lisa Beilke; Joel P. Bercu; Dave Bower; Alessandro Brigo; Kevin P. Cross; Laura Custer; Krista L. Dobo; Eric Dowdy; Kevin A. Ford; Susanne Glowienke; Jacky Van Gompel; James Harvey; Catrin Hasselgren; Masamitsu Honma; Robert A. Jolly; Raymond Kemper; Michelle O. Kenyon; Naomi L. Kruhlak; Penny Leavitt; Scott Miller; Wolfgang Muster; John Nicolette; Andreja Plaper; Mark W. Powley; Donald P. Quigley; M. Vijayaraj Reddy; Hans-Peter Spirkl; Lidiya Stavitskaya; Andrew Teasdale
The ICH M7 guideline describes a consistent approach to identify, categorize, and control DNA reactive, mutagenic, impurities in pharmaceutical products to limit the potential carcinogenic risk related to such impurities. This paper outlines a series of principles and procedures to consider when generating (Q)SAR assessments aligned with the ICH M7 guideline to be included in a regulatory submission. In the absence of adequate experimental data, the results from two complementary (Q)SAR methodologies may be combined to support an initial hazard classification. This may be followed by an assessment of additional information that serves as the basis for an expert review to support or refute the predictions. This paper elucidates scenarios where additional expert knowledge may be beneficial, what such an expert review may contain, and how the results and accompanying considerations may be documented. Furthermore, the use of these principles and procedures to yield a consistent and robust (Q)SAR-based argument to support impurity qualification for regulatory purposes is described in this manuscript.
Regulatory Toxicology and Pharmacology | 2016
Ernst Ahlberg; Alexander Amberg; Lisa Beilke; David Bower; Kevin P. Cross; Laura Custer; Kevin A. Ford; Jacky Van Gompel; James Harvey; Masamitsu Honma; Robert A. Jolly; Elisabeth Joossens; Raymond Kemper; Michelle O. Kenyon; Naomi L. Kruhlak; Lara Kuhnke; Penny Leavitt; Russell T. Naven; Claire L. Neilan; Donald P. Quigley; Dana Shuey; Hans-Peter Spirkl; Lidiya Stavitskaya; Andrew Teasdale; Angela White; Joerg Wichard; Craig Zwickl; Glenn J. Myatt
Statistical-based and expert rule-based models built using public domain mutagenicity knowledge and data are routinely used for computational (Q)SAR assessments of pharmaceutical impurities in line with the approach recommended in the ICH M7 guideline. Knowledge from proprietary corporate mutagenicity databases could be used to increase the predictive performance for selected chemical classes as well as expand the applicability domain of these (Q)SAR models. This paper outlines a mechanism for sharing knowledge without the release of proprietary data. Primary aromatic amine mutagenicity was selected as a case study because this chemical class is often encountered in pharmaceutical impurity analysis and mutagenicity of aromatic amines is currently difficult to predict. As part of this analysis, a series of aromatic amine substructures were defined and the number of mutagenic and non-mutagenic examples for each chemical substructure calculated across a series of public and proprietary mutagenicity databases. This information was pooled across all sources to identify structural classes that activate or deactivate aromatic amine mutagenicity. This structure activity knowledge, in combination with newly released primary aromatic amine data, was incorporated into Leadscopes expert rule-based and statistical-based (Q)SAR models where increased predictive performance was demonstrated.
Environmental and Molecular Mutagenesis | 2012
Dorothea K. Torous; Souk Phonethepswath; Svetlana L. Avlasevich; Jared Mereness; Steven M. Bryce; Jeffrey C. Bemis; Pamela Weller; Sara Bell; Carol Gleason; Laura Custer; James T. MacGregor; Stephen D. Dertinger
Combining multiple genetic toxicology endpoints into a single in vivo study, and/or integrating one or more genotoxicity assays into general toxicology studies, is attractive because it reduces animal use and enables comprehensive comparative analysis using toxicity, metabolism, and pharmacokinetic information from the same animal. This laboratory has developed flow cytometric scoring techniques for monitoring two blood‐based genotoxicity endpoints—micronucleated reticulocyte frequency and gene mutation at the Pig‐a locus—thereby making combination and integration studies practical. The ability to effectively monitor these endpoints in short‐term and repeated dosing schedules was investigated with the carcinogen/noncarcinogen pair benzo(a)pyrene (BP) and pyrene (Pyr). Male Sprague‐Dawley rats were treated via oral gavage for 3 or 28 consecutive days with several dose levels of Pyr, including maximum tolerated doses. BP exposure was administered by the same route but at one dose level, 250 or 125 mg/kg/day for 3‐day and 28‐day studies, respectively. Serial blood samples were collected up to Day 45, and were analyzed for Pig‐a mutation with a dual labeling method (SYTO 13 in combination with anti‐CD59‐PE) that facilitated mutant cell frequency measurements in both total erythrocytes and the reticulocyte subpopulation. A mutant cell enrichment step based on immunomagnetic column separation was used to increase the statistical power of the assay. BP induced robust mutant reticulocyte responses by Day 15, and elevated frequencies persisted until study termination. Mutant erythrocyte responses lagged mutant reticulocyte responses, with peak incidences observed on Day 30 of the 3‐day study (43‐fold increase) and on Day 42 of the 28‐day study (171‐fold increase). No mutagenic effects were apparent for Pyr. Blood samples collected on Day 4, and Day 29 for the 28‐day study, were evaluated for micronucleated reticulocyte frequency. Significant increases in micronucleus frequencies were observed with BP, whereas Pyr had no effect. These results demonstrate that Pig‐a and micronucleus endpoints discriminate between these structurally related carcinogenic and noncarcinogenic agents. Furthermore, the high sensitivity demonstrated with the enrichment protocol indicates that the Pig‐a endpoint is suitable for both repeated‐dose and acute studies, allowing integration of mutagenic and clastogenic endpoints into on‐going toxicology studies, and use as a short‐term assay that provides efficient screening and mechanistic information in vivo. Environ. Mol. Mutagen. 2012.
Regulatory Toxicology and Pharmacology | 2018
Glenn J. Myatt; Ernst Ahlberg; Yumi Akahori; David Allen; Alexander Amberg; Lennart T. Anger; Aynur O. Aptula; Scott S. Auerbach; Lisa Beilke; Phillip Bellion; Romualdo Benigni; Joel P. Bercu; Ewan D. Booth; Dave Bower; Alessandro Brigo; Natalie Burden; Zoryana Cammerer; Mark T. D. Cronin; Kevin P. Cross; Laura Custer; Magdalena Dettwiler; Krista L. Dobo; Kevin A. Ford; Marie C. Fortin; Samantha E. Gad-McDonald; Nichola Gellatly; Véronique Gervais; Kyle P. Glover; Susanne Glowienke; Jacky Van Gompel
The present publication surveys several applications of in silico (i.e., computational) toxicology approaches across different industries and institutions. It highlights the need to develop standardized protocols when conducting toxicity-related predictions. This contribution articulates the information needed for protocols to support in silico predictions for major toxicological endpoints of concern (e.g., genetic toxicity, carcinogenicity, acute toxicity, reproductive toxicity, developmental toxicity) across several industries and regulatory bodies. Such novel in silico toxicology (IST) protocols, when fully developed and implemented, will ensure in silico toxicological assessments are performed and evaluated in a consistent, reproducible, and well-documented manner across industries and regulatory bodies to support wider uptake and acceptance of the approaches. The development of IST protocols is an initiative developed through a collaboration among an international consortium to reflect the state-of-the-art in in silico toxicology for hazard identification and characterization. A general outline for describing the development of such protocols is included and it is based on in silico predictions and/or available experimental data for a defined series of relevant toxicological effects or mechanisms. The publication presents a novel approach for determining the reliability of in silico predictions alongside experimental data. In addition, we discuss how to determine the level of confidence in the assessment based on the relevance and reliability of the information.