65 results on '"Francois Pognan"'
Search Results
2. Drug-induced phospholipidosis confounds drug repurposing for SARS-CoV-2
- Author
-
Veronica V. Rezelj, Audrey Fischer, Francesca Moretti, Romel Rosales, Olivier Schwartz, Jiankun Lyu, Francois Pognan, Andrew C. Kruse, Kris M. White, Marco Vignuzzi, Heiko Schadt, Ziyang Zhang, Henry R. O’Donnell, Michael Schotsaert, Kevan M. Shokat, Tia A. Tummino, Adolfo García-Sastre, Nevan J. Krogan, Jean-Rene Galarneau, Raveen Rathnasinghe, Blandine Monel, Benoit Fischer, Briana L. McGovern, Assaf Alon, Matthew J. O’Meara, Thomas Vallet, Brian K. Shoichet, Sonia Jangra, Laszlo Urban, University of California [San Francisco] (UC San Francisco), University of California (UC), Populations virales et Pathogenèse - Viral Populations and Pathogenesis, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Novartis Institutes for BioMedical Research (NIBR), University of Michigan [Ann Arbor], University of Michigan System, Virus et Immunité - Virus and immunity (CNRS-UMR3569), Icahn School of Medicine at Mount Sinai [New York] (MSSM), Harvard Medical School [Boston] (HMS), This work was supported by grants from the Defense Advanced Research Projects Agency (HR0011-19-2-0020 to B.K.S., N.J.K., A.G.-S., and K.M.S.), NIGMS R35GM122481 (to B.K.S.), National Institutes of Health (P50AI150476, U19AI135990, U19AI135972, R01AI143292, R01AI120694, P01AI063302, and R01AI122747 to N.J.K.), Excellence in Research Award (ERA) from the Laboratory for Genomics Research (LGR) (to N.J.K.), a collaboration between UCSF, UCB, and GSK (no. 133122P to N.J.K.), a Fast Grant for COVID-19 from the Emergent Ventures program at the Mercatus Center of George Mason University (to N.J.K.), funding from the Roddenberry Foundation (to N.J.K.), funding from F. Hoffmann–La Roche and Vir Biotechnology (to N.J.K.), gifts from QCRG philanthropic donors (to N.J.K.), funding from Institut Pasteur (to O.S. and M.V.), Urgence COVID-19 Fundraising Campaign of Institut Pasteur (to O.S. and M.V.), Labex IBEID (ANR-10-LABX-62-IBEID to O.S. and M.V.), ANR/FRM Flash Covid PROTEO-SARS-CoV-2 (to O.S.), IDISCOVR (to O.S.), National Institutes of Health (R01GM119185 to A.C.K.), partly supported by the Center for Research for Influenza Pathogenesis, a Center of Excellence for Influenza Research and Surveillance supported by the National Institute of Allergy and Infectious Diseases (contract no. HHSN272201400008C to A.G.-S.), a supplement to NIAID grant U19AI135972 and to DoD grant W81XWH-20-1-0270 (to A.G.-S.), a Fast Grant from the Mercatus Center (to A.G.-S.), the generous support of the JPB Foundation and the Open Philanthropy Project [research grant 2020-215611 (5384) to A.G.-S.], and anonymous donors (to A.G.-S.). Z.Z. is a Damon Runyon fellow supported by the Damon Runyon Cancer Research Foundation (DRG-2281-17)., We gratefully acknowledge the Région Ile-de-France (program DIM1Health) for the use of the Institut Pasteur imaging facility. We thank N. Aulner for assistance with image requisition and A. Danckaert for assistance with image analysis at Institut Pasteur, Paris. We thank R. Albrecht for support with the BSL3 facility and procedures at the Icahn School of Medicine at Mount Sinai, New York. We thank C. Hayden for the work with the transmission electron microscopy at Novartis. We thank T. R. O’Meara for reading the manuscript. We thank K. Obernier, M. Bouhaddou, and J. M. Fabius for contributions to the overall COVID-19 effort at QCRG., ANR-10-LABX-0062,IBEID,Integrative Biology of Emerging Infectious Diseases(2010), and ANR-20-COVI-0059,PROTEO-SARS-CoV-2,Protéomique du SARS-CoV-2(2020)
- Subjects
Drug ,General Science & Technology ,media_common.quotation_subject ,Microbial Sensitivity Tests ,Pharmacology ,Lipidoses ,Virus Replication ,Amiodarone ,Antiviral Agents ,Article ,Dose-Response Relationship ,Mice ,Surface-Active Agents ,Cations ,Chlorocebus aethiops ,Animals ,Humans ,Medicine ,Lung ,Vero Cells ,Phospholipids ,Repurposing ,media_common ,Phospholipidosis ,Multidisciplinary ,Dose-Response Relationship, Drug ,SARS-CoV-2 ,Drug discovery ,business.industry ,Drug Repositioning ,COVID-19 ,Hydroxychloroquine ,Molecular Pharmacology ,COVID-19 Drug Treatment ,Drug repositioning ,Infectious Diseases ,Good Health and Well Being ,5.1 Pharmaceuticals ,A549 Cells ,[SDV.MP.VIR]Life Sciences [q-bio]/Microbiology and Parasitology/Virology ,Female ,Development of treatments and therapeutic interventions ,business ,medicine.drug - Abstract
International audience; Repurposing drugs as treatments for COVID-19, the disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has drawn much attention. Beginning with sigma receptor ligands and expanding to other drugs from screening in the field, we became concerned that phospholipidosis was a shared mechanism underlying the antiviral activity of many repurposed drugs. For all of the 23 cationic amphiphilic drugs we tested, including hydroxychloroquine, azithromycin, amiodarone, and four others already in clinical trials, phospholipidosis was monotonically correlated with antiviral efficacy. Conversely, drugs active against the same targets that did not induce phospholipidosis were not antiviral. Phospholipidosis depends on the physicochemical properties of drugs and does not reflect specific target-based activities—rather, it may be considered a toxic confound in early drug discovery. Early detection of phospholipidosis could eliminate these artifacts, enabling a focus on molecules with therapeutic potential.
- Published
- 2021
- Full Text
- View/download PDF
3. OntoBrowser: a collaborative tool for curation of ontologies by subject matter experts.
- Author
-
Carlo Ravagli, Francois Pognan, and Philippe Marc
- Published
- 2017
- Full Text
- View/download PDF
4. Human Retinal Organoids Provide a Suitable Tool for Toxicological Investigations: A Comprehensive Validation Using Drugs and Compounds Affecting the Retina
- Author
-
Birthe Dorgau, Maria Georgiou, Alexander Chaudhary, Marina Moya-Molina, Joseph Collin, Rachel Queen, Gerrit Hilgen, Tracey Davey, Philip Hewitt, Michael Schmitt, Stefan Kustermann, Francois Pognan, David H Steel, Evelyne Sernagor, Lyle Armstrong, and Majlinda Lako
- Subjects
Organoids ,Digoxin ,Thioridazine ,Induced Pluripotent Stem Cells ,Humans ,B200 ,Cell Differentiation ,Cell Biology ,General Medicine ,B500 ,Retina ,Sildenafil Citrate ,Developmental Biology - Abstract
Retinal drug toxicity screening is essential for the development of safe treatment strategies for a large number of diseases. To this end, retinal organoids derived from human pluripotent stem cells (hPSCs) provide a suitable screening platform due to their similarity to the human retina and the ease of generation in large-scale formats. In this study, two hPSC cell lines were differentiated to retinal organoids, which comprised all key retinal cell types in multiple nuclear and synaptic layers. Single-cell RNA-Seq of retinal organoids indicated the maintenance of retinal ganglion cells and development of bipolar cells: both cell types segregated into several subtypes. Ketorolac, digoxin, thioridazine, sildenafil, ethanol, and methanol were selected as key compounds to screen on retinal organoids because of their well-known retinal toxicity profile described in the literature. Exposure of the hPSC-derived retinal organoids to digoxin, thioridazine, and sildenafil resulted in photoreceptor cell death, while digoxin and thioridazine additionally affected all other cell types, including Müller glia cells. All drug treatments caused activation of astrocytes, indicated by dendrites sprouting into neuroepithelium. The ability to respond to light was preserved in organoids although the number of responsive retinal ganglion cells decreased after drug exposure. These data indicate similar drug effects in organoids to those reported in in vivo models and/or in humans, thus providing the first robust experimental evidence of their suitability for toxicological studies.
- Published
- 2021
5. Guidelines for FAIR sharing of preclinical safety and off-target pharmacology data
- Author
-
Francois Pognan, Phil Drew, Nicolas Bosc, Tima Camara, Jan A. Kors, Katharine Briggs, Sirarat Sarntivijai, Manuel Pastor, William C Drewe, Thomas Steger-Hartmann, Erik M. van Mulligen, Carlos Díaz, Jordi Quintana, and Medical Informatics
- Subjects
Guiding Principles ,Drug Industry ,Data field ,Pharmacology ,Intellectual property ,Off-target pharmacology ,Pooling data ,03 medical and health sciences ,Animals ,Data Mining ,Computer Simulation ,FAIR ,Pharmaceutical industry ,0303 health sciences ,business.industry ,Information Dissemination ,030311 toxicology ,General Medicine ,Multiple-criteria decision analysis ,Preclinical ,3. Good health ,Data sharing ,Medical Laboratory Technology ,General partnership ,Business - Abstract
Pre-competitive data sharing can offer the pharmaceutical industry significant benefits in terms of reducing the time and costs involved in getting a new drug to market through more informed testing strategies and knowledge gained by pooling data. If sufficient data is shared and can be co-analyzed, then it can also offer the potential for reduced animal usage and improvements in the in silico prediction of toxicological effects. Data sharing benefits can be further enhanced by applying the FAIR Guiding Principles, reducing time spent curating, transforming and aggregating datasets and allowing more time for data mining and analysis. We hope to facilitate data sharing by other organizations and initiatives by describing lessons learned as part of the Enhancing TRANslational SAFEty Assessment through Integrative Knowledge Management (eTRANSAFE) project, an Innovative Medicines Initiative (IMI) partnership which aims to integrate publicly available data sources with proprietary preclinical and clinical data donated by pharmaceutical organizations. Methods to foster trust and overcome non-technical barriers to data sharing such as legal and IPR (intellectual property rights) are described, including the security requirements that pharmaceutical organizations generally expect to be met. We share the consensus achieved among pharmaceutical partners on decision criteria to be included in internal clearance procedures used to decide if data can be shared. We also report on the consensus achieved on specific data fields to be excluded from sharing for sensitive preclinical safety and pharmacology data that could otherwise not be shared. This project has received funding from the Innovative Medicines Initiative 2 Joint Undertaking under grant agreement No 777365 (“eTRANSAFE”). This Joint Undertaking receives support from the European Union’s Horizon 2020 research and innovation programme and EFPIA.
- Published
- 2021
6. Phospholipidosis is a shared mechanism underlying the in vitro antiviral activity of many repurposed drugs against SARS-CoV-2
- Author
-
Francois Pognan, Laszlo Urban, Heiko Schadt, Veronica V. Rezelj, Adolfo Garcia-Sastre, Olivier Schwartz, Blandine Monel, Nevan J. Krogan, Audrey Fischer, Ziyang Zhang, Brian K. Shoichet, Assaf Alon, Matthew J. O’Meara, Benoit Fischer, Kevan M. Shokat, Tia A. Tummino, Kris M. White, Marco Vignuzzi, Francesca Moretti, Jiankun Lyu, Thomas Vallet, Henry R. O’Donnell, and Andrew C. Kruse
- Subjects
Phospholipidosis ,Coronavirus disease 2019 (COVID-19) ,Drug discovery ,business.industry ,Mechanism (biology) ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Medicine ,Drug side effects ,Pharmacology ,business ,Article ,Repurposing ,In vitro - Abstract
Repurposing drugs as treatments for COVID-19 has drawn much attention. A common strategy has been to screen for established drugs, typically developed for other indications, that are antiviral in cells or organisms. Intriguingly, most of the drugs that have emerged from these campaigns, though diverse in structure, share a common physical property: cationic amphiphilicity. Provoked by the similarity of these repurposed drugs to those inducing phospholipidosis, a well-known drug side effect, we investigated phospholipidosis as a mechanism for antiviral activity. We tested 23 cationic amphiphilic drugs—including those from phenotypic screens and others that we ourselves had found—for induction of phospholipidosis in cell culture. We found that most of the repurposed drugs, which included hydroxychloroquine, azithromycin, amiodarone, and four others that have already progressed to clinical trials, induced phospholipidosis in the same concentration range as their antiviral activity; indeed, there was a strong monotonic correlation between antiviral efficacy and the magnitude of the phospholipidosis. Conversely, drugs active against the same targets that did not induce phospholipidosis were not antiviral. Phospholipidosis depends on the gross physical properties of drugs, and does not reflect specific target-based activities, rather it may be considered a confound in early drug discovery. Understanding its role in infection, and detecting its effects rapidly, will allow the community to better distinguish between drugs and lead compounds that more directly impact COVID-19 from the large proportion of molecules that manifest this confounding effect, saving much time, effort and cost., One Sentence Summary: Drug-induced phospholipidosis is a single mechanism that may explain the in vitro efficacy of a wide-variety of therapeutics repurposed for COVID-19.
- Published
- 2021
- Full Text
- View/download PDF
7. eTRANSAFE's Rosetta stone - a new approach to overcome safety translational hurdles
- Author
-
E van Mulligen, Francois Pognan, Jan A. Kors, J. van der Lei, and Thomas Steger-Hartmann
- Subjects
General Medicine ,Toxicology - Published
- 2021
- Full Text
- View/download PDF
8. Improving the Safety Assessment of Chemicals and Drug Candidates by the Integration of Bioinformatics and Chemoinformatics Data
- Author
-
Thomas Steger-Hartmann and Francois Pognan
- Subjects
0301 basic medicine ,Drug ,Databases, Factual ,Drug-Related Side Effects and Adverse Reactions ,Process (engineering) ,media_common.quotation_subject ,In silico ,Drug Evaluation, Preclinical ,Guidelines as Topic ,Context (language use) ,Toxicology ,Bioinformatics ,Risk Assessment ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Computer Simulation ,media_common ,Preclinical toxicity ,Pharmacology ,business.industry ,Computational Biology ,General Medicine ,Chemical industry ,3. Good health ,030104 developmental biology ,Cheminformatics ,030220 oncology & carcinogenesis ,ACROSS Project ,business ,Software - Abstract
The application of read-across and in silico tools for regulatory decision-making has been limited for pharmaceutical compounds to the assessment of genotoxic impurity. In contrast, the broad availability of toxicity data for industrial chemicals has triggered regulatory frameworks for read-across (e.g. ECHA Read-Across Assessment Framework), software tools and public databases for an automated process of gap filling in the context of safety assessment. This MiniReview provides an overview of the currently existing in silico and read-across approaches for chemicals together with recent developments for pharmaceutical compounds in these areas. It also highlights the differences and commonalities in the in silico safety assessment of industrial chemicals and drug candidates. Whereas toxicity data collection and sharing is now common practice for chemicals falling under the European REACH regulation (Registration, Evaluation, Authorisation and Restriction of Chemicals), the biggest hurdle for establishing preclinical safety databases for pharmaceutical compounds is the unwillingness to share proprietary data and lack of published data sets. In a recent consortium approach, thirteen pharmaceutical companies, eleven academic partners and six small to medium size enterprises (SMEs) of the bioinformatics sector joined forces over the last 7 years within the European Innovative Medicines Initiative project eTOX ('electronic toxicity') to design and implement a strategy for leveraging these preclinical data for small molecules and sharing them across project partners. The eTOX database has evolved as the largest preclinical toxicity database for drugs and drug candidates and currently contains more than 1900 different chemical structures and more than 8000 in vivo toxicity study data sets. It can be foreseen that the development and application of such databases for drugs or drug candidates will in the future also cross-fertilize the read-across and the in silico assessment of industrial or consumer chemicals particularly as soon as human safety data from clinical trials are integrated too.
- Published
- 2018
- Full Text
- View/download PDF
9. Mechanistic insights into selective killing of OXPHOS-dependent cancer cells by arctigenin
- Author
-
Franziska Paech, Francois Pognan, Virginie Riebel, Philippe Couttet, Stephan Krähenbühl, Marianne Uteng, Salah-Dine Chibout, Armin Wolf, and Karin Brecht
- Subjects
0301 basic medicine ,endocrine system diseases ,Antineoplastic Agents ,Apoptosis ,Oxidative phosphorylation ,Biology ,Mitochondrion ,Toxicology ,Lignans ,Oxidative Phosphorylation ,Necrosis ,03 medical and health sciences ,chemistry.chemical_compound ,Adenosine Triphosphate ,Oxygen Consumption ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Humans ,Furans ,Arctigenin ,Membrane Potential, Mitochondrial ,General Medicine ,Endoplasmic Reticulum Stress ,medicine.disease ,digestive system diseases ,Cell biology ,Mitochondrial toxicity ,030104 developmental biology ,Biochemistry ,chemistry ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Unfolded protein response ,Glycolysis - Abstract
Arctigenin has previously been identified as a potential anti-tumor treatment for advanced pancreatic cancer. However, the mechanism of how arctigenin kills cancer cells is not fully understood. In the present work we studied the mechanism of toxicity by arctigenin in the human pancreatic cell line, Panc-1, with special emphasis on the mitochondria. A comparison of Panc-1 cells cultured in glucose versus galactose medium was applied, allowing assessments of effects in glycolytic versus oxidative phosphorylation (OXPHOS)-dependent Panc-1 cells. For control purposes, the mitochondrial toxic response to treatment with arctigenin was compared to the anti-cancer drug, sorafenib, which is a tyrosine kinase inhibitor known for mitochondrial toxic off-target effects (Will et al., 2008). In both Panc-1 OXPHOS-dependent and glycolytic cells, arctigenin dissipated the mitochondrial membrane potential, which was demonstrated to be due to inhibition of the mitochondrial complexes II and IV. However, arctigenin selectively killed only the OXPHOS-dependent Panc-1 cells. This selective killing of OXPHOS-dependent Panc-1 cells was accompanied by generation of ER stress, mitochondrial membrane permeabilization and caspase activation leading to apoptosis and aponecrosis.
- Published
- 2017
- Full Text
- View/download PDF
10. Introducing the concept of virtual control groups into preclinical toxicology testing
- Author
-
Frank Bringezu, Alexander Amberg, Barber Christopher Gordon, Wolfgang Muster, Thomas Steger-Hartmann, Francois Pognan, Lea A. I. Vaas, Jörg Wichard, and Annika Kreuchwig
- Subjects
Pharmacology ,0303 health sciences ,Databases, Factual ,Computer science ,Information Dissemination ,Knowledge Bases ,Control (management) ,Drug Evaluation, Preclinical ,General Medicine ,Data science ,Outcome (game theory) ,3. Good health ,Data sharing ,Clinical trial ,03 medical and health sciences ,Medical Laboratory Technology ,0302 clinical medicine ,Proof of concept ,Research Design ,030220 oncology & carcinogenesis ,Toxicity Tests ,Duration (project management) ,Set (psychology) ,Construct (philosophy) ,030304 developmental biology - Abstract
Sharing legacy data from in vivo toxicity studies offers the opportunity to analyze the variability of control groups stratified for strain, age, duration of study, vehicle and other experimental conditions. Historical animal control group data may lead to a repository, which could be used to construct virtual control groups (VCGs) for toxicity studies. VCGs are an established concept in clinical trials, but the idea of replacing living beings with virtual data sets has so far not been introduced into the design of regulatory animal studies. The use of VCGs has the potential of a 25% reduction in animal use by replacing the control group animals with existing randomized data sets. Prerequisites for such an approach are the availability of large and well-structured control data sets as well as thorough statistical evaluations. the foundation of data sharing has been laid within the Innovative Medicines Initiatives projects eTOX and eTRANSAFE. For a proof of principle participating companies have started to collect control group data for subacute (4-week) GLP studies with Wistar rats (the strain preferentially used in Europe) and are characterizing these data for its variability. In a second step, the control group data will be shared among the companies and cross-company variability will be investigated. In a third step, a set of studies will be analyzed to assess whether the use of VCG data would have influenced the outcome of the study compared to the real control group.
- Published
- 2020
11. The eTRANSAFE Project on Translational Safety Assessment through Integrative Knowledge Management: Achievements and Perspectives
- Author
-
Francois Pognan, José M. Fernández, Ferran Sanz, Philip Drew, Alfonso Valencia, Johan van der Lei, Josep Saüch-Pitarch, Carlos Díaz, Juan Manuel Ramírez-Anguita, Giulia Callegaro, Manuel Pastor, Francesco Ronzano, Laura I. Furlong, Salvador Capella-Gutierrez, Emilio Centeno, Niklas Blomberg, Bob van de Water, Erik M. van Mulligen, Janet Piñero, Emre Guney, Philip Rowell, Javier Corvi, Thomas Steger-Hartmann, Jan A. Kors, Frank Bringezu, Katharine Briggs, William C Drewe, Miguel Angel Mayer, Barcelona Supercomputing Center, and Medical Informatics
- Subjects
read across ,drug safety ,Knowledge management ,Legacy data ,Computer science ,Informàtica::Sistemes d'informació [Àrees temàtiques de la UPC] ,data sharing ,lcsh:Medicine ,lcsh:RS1-441 ,Pharmaceutical Science ,Drug safety ,Integrative knowledge management ,Read across ,Toxicology ,Article ,lcsh:Pharmacy and materia medica ,03 medical and health sciences ,0302 clinical medicine ,Data query ,Drug Discovery ,Data mining ,030304 developmental biology ,0303 health sciences ,Computational model ,integrative knowledge management ,translational safety assessment ,Translational safety assessment ,business.industry ,lcsh:R ,Predictive modelling ,data mining ,3. Good health ,Visualization ,Data sharing ,Molecular Medicine ,Mineria de dades ,Modeling and simulation in science, engineering & technology ,predictive modelling ,business ,030217 neurology & neurosurgery ,toxicology - Abstract
eTRANSAFE is a research project funded within the Innovative Medicines Initiative (IMI), which aims at developing integrated databases and computational tools (the eTRANSAFE ToxHub) that support the translational safety assessment of new drugs by using legacy data provided by the pharmaceutical companies that participate in the project. The project objectives include the development of databases containing preclinical and clinical data, computational systems for translational analysis including tools for data query, analysis and visualization, as well as computational models to explain and predict drug safety events. This research received funding from the Innovative Medicines Initiative 2 Joint Undertaking under grant agreements eTRANSAFE (777365). This Joint Undertaking receives support from the European Union’s Horizon 2020 research and innovation programme and EFPIA companies in kind contribution. The Research Programme on Biomedical Informatics (GRIB) of IMIM and DCEXS-UPF is a member of the Spanish National Bioinformatics Institute (INB), PRB2-ISCIII and is supported by grant PT13/0001/0023, of the PE I + D + i 2013–2016, funded by ISCIII and FEDER. The GRIB also receive support from Agència de Gestió D’ajuts Universitaris i de Recerca Generalitat de Catalunya (AGAUR, ref.: 2017SGR01020). The DCEXS is a “Unidad de Excelencia María de Maeztu”, funded by the MINECO (ref: MDM-2014-0370).
- Published
- 2021
- Full Text
- View/download PDF
12. Colony-Stimulating Factor-1 Antibody Lacnotuzumab in a Phase 1 Healthy Volunteer Study and Mechanistic Investigation of Safety Outcomes
- Author
-
Yinuo Pang, Gaetane Woerly, Esther Sutter, Francois Pognan, Ursula Schramm, Yoav E Timsit, Marie Anne Valentin, Philippe Couttet, Armin Wolf, Beate Vogel, Ivan Demin, Birgit Jaitner, and Ronenn Roubenoff
- Subjects
0301 basic medicine ,Pharmacology ,biology ,business.industry ,Placebo ,Asymptomatic ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Pharmacokinetics ,Tolerability ,Pharmacodynamics ,biology.protein ,Molecular Medicine ,Medicine ,Creatine kinase ,medicine.symptom ,Receptor ,business ,Adverse effect ,030217 neurology & neurosurgery - Abstract
The colony-stimulating factor-1 (CSF-1) receptor pathway has been implicated in a variety of diseases, and CSF-1-dependent mechanisms are also involved in bloodborne protein clearance. Lacnotuzumab is a novel, high-affinity, humanized, anti-CSF-1 monoclonal antibody that prevents CSF-1-mediated receptor activation. This phase 1, two-part, double-blind study in healthy volunteers assessed the safety and tolerability of lacnotuzumab and its pharmacokinetics (PK) and pharmacodynamic properties. Part A (n = 36) was a single, ascending-dose assessment of eight lacnotuzumab doses (0.01-20 mg/kg); in part B (n = 16), lacnotuzumab was administered at either 5 or 10 mg/kg. In each study cohort, individuals were randomized 3:1 to lacnotuzumab or placebo. Lacnotuzumab was generally well tolerated. At higher doses (10 and 20 mg/kg), creatine kinase (CK) elevations (>5× the upper limit of normal, but asymptomatic and reversible) and mild transient periorbital swelling were reported. Most adverse events (AEs) were low-grade, no unexpected or novel AEs were observed, and there were no discontinuations for AEs. Free, unbound lacnotuzumab serum concentration-time profiles showed nonlinear PK across doses from 0.01 to 20 mg/kg, with faster apparent elimination at lower doses or concentrations; this finding was consistent with apparent target-mediated drug disposition. Lacnotuzumab also showed dose-dependent, on-target effects on multiple downstream biomarkers. Preclinical investigations of the CK elevation and periorbital swelling observed after lacnotuzumab administration suggest that these are reversible, nonpathological events linked to inhibition of the CSF-1 pathway. These data support further evaluation of lacnotuzumab in clinical studies.
- Published
- 2018
13. Bile Acid Sequestration by Cholestyramine Mitigates FGFR4 Inhibition-Induced ALT Elevation
- Author
-
Terrilyn A. Richardson, Chi-Hse Teng, Audrey Fischer, Marianne Schwald, William Kluwe, Joerg Andreas Mahl, Robin Alec Fairhurst, Luigi Manenti, Francois Pognan, Armin Wolf, Gerd A. Kullak-Ublick, Mathilde Lienard, Jacqueline Kinyamu-Akunda, Elizabeth Skuba, Salah-Dine Chibout, Andreas Weiss, Kuno Wuersch, Philippe Couttet, Heiko Schadt, Diana Graus Porta, Corinne Emotte, University of Zurich, and Kinyamu-Akunda, Jacqueline
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,medicine.drug_class ,Pyridines ,Cholestyramine Resin ,610 Medicine & health ,Toxicology ,Cholesterol 7 alpha-hydroxylase ,Piperazines ,Bile Acids and Salts ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Dogs ,Internal medicine ,Toxicity Tests ,medicine ,Animals ,Receptor, Fibroblast Growth Factor, Type 4 ,Protein Kinase Inhibitors ,Cholestyramine ,biology ,Bile acid ,Dose-Response Relationship, Drug ,3005 Toxicology ,FGF19 ,Alanine Transaminase ,Toxicokinetics ,Dose–response relationship ,030104 developmental biology ,Endocrinology ,Alanine transaminase ,chemistry ,Liver ,10199 Clinic for Clinical Pharmacology and Toxicology ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Taurolithocholic acid ,Taurodeoxycholic acid ,medicine.drug - Abstract
The FGF19- fibroblast growth factor receptor (FGFR4)-βKlotho (KLB) pathway plays an important role in the regulation of bile acid (BA) homeostasis. Aberrant activation of this pathway has been described in the development and progression of a subset of liver cancers including hepatocellular carcinoma, establishing FGFR4 as an attractive therapeutic target for such solid tumors. FGF401 is a highly selective FGFR4 kinase inhibitor being developed for hepatocellular carcinoma, currently in phase I/II clinical studies. In preclinical studies in mice and dogs, oral administration of FGF401 led to induction of Cyp7a1, elevation of its peripheral marker 7alpha-hydroxy-4-cholesten-3-one, increased BA pool size, decreased serum cholesterol and diarrhea in dogs. FGF401 was also associated with increases of serum aminotransferases, primarily alanine aminotransferase (ALT), in the absence of any observable adverse histopathological findings in the liver, or in any other organs. We hypothesized that the increase in ALT could be secondary to increased BAs and conducted an investigative study in dogs with FGF401 and coadministration of the BA sequestrant cholestyramine (CHO). CHO prevented and reversed FGF401-related increases in ALT in dogs in parallel to its ability to reduce BAs in the circulation. Correlation analysis showed that FGF401-mediated increases in ALT strongly correlated with increases in taurolithocholic acid and taurodeoxycholic acid, the major secondary BAs in dog plasma, indicating a mechanistic link between ALT elevation and changes in BA pool hydrophobicity. Thus, CHO may offer the potential to mitigate elevations in serum aminotransferases in human subjects that are caused by targeted FGFR4 inhibition and elevated intracellular BA levels.
- Published
- 2018
14. Determination of liver specific toxicities in rat hepatocytes by high content imaging during 2-week multiple treatment
- Author
-
Armin Wolf, Salah-Dine Chibout, Francois Pognan, Davide Germano, and Marianne Uteng
- Subjects
Male ,Chlorpromazine ,Amiodarone ,Pharmacology ,Toxicology ,Troglitazone ,03 medical and health sciences ,0302 clinical medicine ,In vitro model ,In vivo ,Long-term culture ,Culture Techniques ,Cyclosporin a ,Potassium Channel Blockers ,medicine ,Animals ,Hypoglycemic Agents ,Chromans ,Rats, Wistar ,Cells, Cultured ,030304 developmental biology ,Liver injury ,Phospholipidosis ,0303 health sciences ,business.industry ,Hepatotoxicity ,General Medicine ,medicine.disease ,Safety profiling ,Rats ,3. Good health ,medicine.anatomical_structure ,Gene Expression Regulation ,High content imaging ,030220 oncology & carcinogenesis ,Hepatocyte ,Cyclosporine ,Hepatocytes ,Dopamine Antagonists ,Thiazolidinediones ,Steatosis ,business ,Rat hepatocytes ,Immunosuppressive Agents ,medicine.drug - Abstract
DILI is a major safety issue during drug development and one of the leading causes for market withdrawal. Despite many efforts made in the past, the prediction of DILI using in vitro models remains very unreliable. In the present study, the well-established hepatocyte Collagen I-Matrigel™ sandwich culture was used, mimicking chronic drug treatment after multiple incubations for 14days. Ten drugs associated with different types of specific preclinical and clinical liver injury were evaluated at non-cytotoxic concentrations. Mrp2-mediated transport, intracellular accumulation of neutral lipids and phospholipids were selected as functional endpoints by using Cellomics™ Arrayscan® technology and assessed at five timepoints (day 1, 3, 7, 10, 14). Liver specific functional impairments after drug treatment were enhanced over time and could be monitored by HCI already after few days and before cytotoxicity. Phospholipidosis-inducing drugs Chlorpromazine and Amiodarone displayed the same response as in vivo. Cyclosporin A, Chlorpromazine, and Troglitazone inhibited Mrp2-mediated biliary transport, correlating with in vivo findings. Steatosis remained difficult to be reproduced under the current in vitro testing conditions, resulting into false negative and positive responses. The present results suggest that the repeated long-term treatment of rat hepatocytes in the Collagen I-Matrigel™ sandwich configuration might be a suitable tool for safety profiling of the potential to induce phospholipidosis and impair Mrp2-mediated transport processes, but not to predict steatosis.
- Published
- 2015
- Full Text
- View/download PDF
15. Ensuring confidence in predictions: A scheme to assess the scientific validity of in silico models
- Author
-
Mark Hewitt, Francois Pognan, Claire M. Ellison, Judith C. Madden, Jordi Munoz-Muriendas, Mark T. D. Cronin, Thomas Steger-Hartmann, and Manuel Pastor
- Subjects
Scheme (programming language) ,Computer science ,End user ,In silico ,Frame (networking) ,Reproducibility of Results ,Pharmaceutical Science ,Pilot Projects ,Legislation ,Models, Theoretical ,Bioinformatics ,RS ,3. Good health ,Drug development ,Risk analysis (engineering) ,Humans ,Computer Simulation ,Robustness (economics) ,computer ,Reliability (statistics) ,computer.programming_language - Abstract
The use of in silico tools within the drug development process to predict a wide range of properties including absorption, distribution, metabolism, elimination and toxicity has become increasingly important due to changes in legislation and both ethical and economic drivers to reduce animal testing. Whilst in silico tools have been used for decades there remains reluctance to accept predictions based on these methods particularly in regulatory settings. This apprehension arises in part due to lack of confidence in the reliability, robustness and applicability of the models. To address this issue we propose a scheme for the verification of in silico models that enables end users and modellers to assess the scientific validity of models in accordance with the principles of good computer modelling practice. We report here the implementation of the scheme within the Innovative Medicines Initiative project "eTOX" (electronic toxicity) and its application to the in silico models developed within the frame of this project.
- Published
- 2015
- Full Text
- View/download PDF
16. Detection, Elimination, Mitigation, and Prediction of Drug-Induced Liver Injury in Drug Discovery
- Author
-
Francois Pognan
- Subjects
Drug ,Liver injury ,business.industry ,Drug discovery ,media_common.quotation_subject ,Medicine ,Pharmacology ,business ,medicine.disease ,media_common - Published
- 2018
- Full Text
- View/download PDF
17. Reduced Activity of Sphingosine-1-Phosphate Lyase Induces Podocyte-related Glomerular Proteinuria, Skin Irritation, and Platelet Activation
- Author
-
David Ledieu, Anna Schubart, Francois Pognan, Esther Sutter, Jens Schümann, Pierre Moulin, Armelle Grevot, Serge Côté, Alessandro Piaia, Andreas Billich, Ursula Junker Walker, Christian Beerli, and Armin Wolf
- Subjects
Male ,medicine.medical_specialty ,Inflammation ,Biology ,Kidney ,Toxicology ,Pathology and Forensic Medicine ,Podocyte ,Mice ,In vivo ,Internal medicine ,Genetic model ,medicine ,Animals ,Platelet activation ,Molecular Biology ,Aldehyde-Lyases ,Skin ,Podocytes ,Cell Biology ,Platelet Activation ,Lyase ,Rats ,Proteinuria ,Tamoxifen ,medicine.anatomical_structure ,Endocrinology ,Toxicity ,Female ,lipids (amino acids, peptides, and proteins) ,medicine.symptom - Abstract
Sphingosine-1-phosphate (S1P) lyase is considered as a drug target in autoimmune diseases based on the protective effect of reducing activity of the enzyme in animal models of inflammation. Since S1P lyase deficiency in mice causes a severe, lethal phenotype, it was of interest to investigate any pathological alterations associated with only partially reduced activity of S1P lyase as may be encountered upon pharmacological inhibition. Both genetic reduction of S1P lyase activity in mice and inhibition of S1P lyase with a low-molecular-weight compound in rats consistently resulted in podocyte-based kidney toxicity, which is the most severe finding. In addition, skin irritation and platelet activation were observed in both instances. The similarity of the findings in both the genetic model and the pharmacological study supports the value of analyzing inducible partially target-deficient mice for safety assessment. If the findings described in rodents translate to humans, target-related toxicity, particularly podocyte dysfunction, may limit chronic systemic treatment of autoimmune diseases with S1P lyase inhibitors. Furthermore, partial deficiency or inhibition of S1P lyase appears to provide an in vivo rodent model to enable studies on the mechanism of podocyte dysfunction.
- Published
- 2015
- Full Text
- View/download PDF
18. Induction of hemangiosarcoma in mice after chronic treatment with S1P-modulator siponimod and its lack of relevance to rat and human
- Author
-
André Cordier, David Ledieu, Francois Pognan, Sarah B. Voytek, J. Andreas Mahl, Page Bouchard, Annabelle Heier, Marc Raccuglia, Salah-Dine Chibout, Maria Papoutsi, Diethilde Theil, Carine Kolly, Andreas Hartmann, Katie Kubek-Luck, Christian Trendelenburg, Natalie M. Claudio, and Patrick J. Devine
- Subjects
0301 basic medicine ,Male ,Health, Toxicology and Mutagenesis ,Hemangiosarcoma ,Administration, Oral ,Stimulation ,Mice, Inbred Strains ,Genotoxicity and Carcinogenicity ,Pharmacology ,Siponimod ,Toxicology ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,Species Specificity ,In vivo ,Benzyl Compounds ,Hemangiosarcoma (HSA) ,Animals ,Humans ,Rats, Wistar ,Receptor ,Toxicity Tests, Chronic ,Mitosis ,Cells, Cultured ,Placental growth factor (PLGF2) ,Placenta Growth Factor ,Chemistry ,Cell growth ,Endothelial Cells ,General Medicine ,In vitro ,Toxicokinetics ,Endothelial stem cell ,Receptors, Lysosphingolipid ,030104 developmental biology ,Azetidines ,Sphingosine-1-phosphate Receptor 1 (S1P1) ,Endothelium, Vascular ,Transcriptome ,Vascular endothelial cells - Abstract
A high incidence of hemangiosarcoma (HSA) was observed in mice treated for 2 years with siponimod, a sphingosine-1-phosphate receptor 1 (S1P1) functional antagonist, while no such tumors were observed in rats under the same treatment conditions. In 3-month rat (90 mg/kg/day) and 9-month mouse (25 and 75 mg/kg/day) in vivo mechanistic studies, vascular endothelial cell (VEC) activation was observed in both species, but VEC proliferation and persistent increases in circulating placental growth factor 2 (PLGF2) were only seen in the mouse. In mice, these effects were sustained over the 9-month study duration, while in rats increased mitotic gene expression was present at day 3 only and PLGF2 was induced only during the first week of treatment. In the mouse, the persistent VEC activation, mitosis induction, and PLGF2 stimulation likely led to sustained neo-angiogenesis which over life-long treatment may result in HSA formation. In rats, despite sustained VEC activation, the transient mitotic and PLGF2 stimuli did not result in the formation of HSA. In vitro, the mouse and rat primary endothelial cell cultures mirrored their respective in vivo findings for cell proliferation and PLGF2 release. Human VECs, like rat cells, were unresponsive to siponimod treatment with no proliferative response and no release of PLGF2 at all tested concentrations. Hence, it is suggested that the human cells also reproduce a lack of in vivo response to siponimod. In conclusion, the molecular mechanisms leading to siponimod-induced HSA in mice are considered species specific and likely irrelevant to humans. Electronic supplementary material The online version of this article (10.1007/s00204-018-2189-9) contains supplementary material, which is available to authorized users.
- Published
- 2017
19. Induction of hemangiosarcoma in mice after chronic treatment with S1P-modulator siponimod and its lack of relevance to rat and human
- Author
-
Francois Pognan
- Subjects
chemistry.chemical_compound ,Hemangiosarcoma ,Siponimod ,chemistry ,business.industry ,Cancer research ,Medicine ,General Medicine ,Toxicology ,business ,medicine.disease - Published
- 2018
- Full Text
- View/download PDF
20. Editor's Highlight: Comparative Renal Safety Assessment of the Hepatitis B Drugs, Adefovir, Tenofovir, Telbivudine and Entecavir in Rats
- Author
-
Elaine Tritto, Francois Pognan, Li Li, Alessandro Piaia, David Ledieu, Nicolau Beckmann, Armin Wolf, Valerie Dubost, Marianne Uteng, Pierre Moulin, Salah-Dine Chibout, and Andreas Mahl
- Subjects
Male ,Renal function ,Pharmacology ,Toxicology ,Kidney ,Antiviral Agents ,Rats, Sprague-Dawley ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Telbivudine ,medicine ,Adefovir ,Animals ,030212 general & internal medicine ,Creatinine ,business.industry ,Entecavir ,Hepatitis B ,medicine.disease ,Virology ,Magnetic Resonance Imaging ,Rats ,medicine.anatomical_structure ,chemistry ,Toxicity ,030211 gastroenterology & hepatology ,business ,medicine.drug - Abstract
The aim of this study was to determine the relative safety of four antiviral drugs (telbivudine, tenofovir, adefovir, entecavir) against hepatitis B virus with respect to kidney function and toxicity in male Spraque-Dawley rats. The antiviral drugs were administered once daily for four weeks by oral gavage at about 10 and 25-40 times the human equivalent dose. Main assessments included markers of renal toxicity in urine, magnetic resonance imaging (MRI) of kidney function, histopathology and electron microscopic examination. Administration of adefovir at 11 and 28 mg/kg for 4 weeks caused functional and morphological kidney alterations in a time- and dose-dependent manner, affecting mainly the proximal tubules and suggesting a mechanism of toxicity related to mitochondrial degeneration/depletion. Of note, the observed adefovir-induced reduction of kidney function was not detected by the standard method of glomerular filtration rate (GFR) measurements (clearance rate of the endogenous marker, creatinine), thereby emphasizing the superiority of MRI in terms of sensitive detection of GFR in rats. For the low dose of 300 mg/kg of tenofovir, minor kidney effects such as nuclear enlargement in the tubular epithelium, and hyaline droplets accumulation were detected, which was also observed for the low dose (11 mg/kg) of adefovir. No assessments could be done at the higher dose of 600/1000 mg/kg tenofovir due to gastrointestinal tract toxicity which prevented treatment of the animals for longer than 1 week. Entecavir at 1 and 3 mg/kg and telbivudine at 600 and 1600 mg/kg caused no toxicologically relevant effects on the kidney.
- Published
- 2016
21. Inroads to Predict in Vivo Toxicology—An Introduction to the eTOX Project
- Author
-
David K. Watson, Thomas Steger-Hartmann, Andreas Sutter, Montserrat Cases, Francois Pognan, David J. Heard, Ferran Sanz, Manuel Pastor, Christof H. Schwab, Katharine Briggs, and Jörg D. Wichard
- Subjects
Data Integration ,Decision support system ,Manual Curation ,Databases, Factual ,data sharing ,Knowledge Bases ,Drug Evaluation, Preclinical ,Review ,predictive toxicology ,computer.software_genre ,in vivo toxicity ,030226 pharmacology & pharmacy ,Bases de dades ,lcsh:Chemistry ,Toxicology ,0302 clinical medicine ,Predictive toxicology ,Computational models ,Data Mining ,Medicine ,ontology ,lcsh:QH301-705.5 ,Spectroscopy ,Pharmaceutical industry ,In vivo toxicity ,0303 health sciences ,Ontology ,QSAR ,Decision Support System ,General Medicine ,In vitro toxicity ,In silico toxicity ,3. Good health ,Computer Science Applications ,Knowledge Management ,Drug development ,Cheminformatics ,Medicaments -- Efectes secundaris ,histopathology ,Data integration ,in silico toxicity ,Drug-Related Side Effects and Adverse Reactions ,Information Dissemination ,Histopathology ,in vitro toxicity ,Expert Systems ,Risk Assessment ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,Deliverable ,Animals ,Humans ,Physical and Theoretical Chemistry ,Molecular Biology ,030304 developmental biology ,business.industry ,Organic Chemistry ,Data sharing ,lcsh:Biology (General) ,lcsh:QD1-999 ,computational models ,business ,computer - Abstract
There is a widespread awareness that the wealth of preclinical toxicity data that the pharmaceutical industry has generated in recent decades is not exploited as efficiently as it could be. Enhanced data availability for compound comparison ("read-across"), or for data mining to build predictive tools, should lead to a more efficient drug development process and contribute to the reduction of animal use (3Rs principle). In order to achieve these goals, a consortium approach, grouping numbers of relevant partners, is required. The eTOX ("electronic toxicity") consortium represents such a project and is a public-private partnership within the framework of the European Innovative Medicines Initiative (IMI). The project aims at the development of in silico prediction systems for organ and in vivo toxicity. The backbone of the project will be a database consisting of preclinical toxicity data for drug compounds or candidates extracted from previously unpublished, legacy reports from thirteen European and European operation-based pharmaceutical companies. The database will be enhanced by incorporation of publically available, high quality toxicology data. Seven academic institutes and five small-to-medium size enterprises (SMEs) contribute with their expertise in data gathering, database curation, data mining, chemoinformatics and predictive systems development. The outcome of the project will be a predictive system contributing to early potential hazard identification and risk assessment during the drug development process. The concept and strategy of the eTOX project is described here, together with current achievements and future deliverables.
- Published
- 2012
- Full Text
- View/download PDF
22. Investigation of Correlation Among Safety Biomarkers in Serum, Histopathological Examination, and Toxicogenomics
- Author
-
Paul Vancutsem, Marc DeCristofaro, Salah-Dine Chibout, Armin Wolf, Tao Wang, M. Craig Keselica, Francois Pognan, Marion Wiesmann, Elizabeth Skuba, Maria Papoutsi, Judit Markovits, Robert H. Spaet, James Sutton, Pierre Moulin, William Kluwe, and Lew Petryk
- Subjects
medicine.medical_specialty ,Pathology ,Necrosis ,Down-Regulation ,Aspartate transaminase ,Inflammation ,Biology ,Toxicology ,medicine.disease_cause ,Muscle hypertrophy ,Toxicity Tests ,medicine ,Animals ,Humans ,Aspartate Aminotransferases ,Benzothiazoles ,RNA, Messenger ,Rats, Wistar ,Picolinic Acids ,Liver injury ,Gene Expression Profiling ,Alanine Transaminase ,medicine.disease ,Rats ,Up-Regulation ,Macaca fascicularis ,Liver ,Pharmacogenetics ,biology.protein ,Histopathology ,medicine.symptom ,Toxicogenomics ,Biomarkers ,Oxidative stress - Abstract
This article addresses the issue of miscorrelation between hepatic injury biomarkers and histopathological findings in the drug development context. Our studies indicate that the use of toxicogenomics can aid in the drug development decision-making process associated with such miscorrelated data. BLZ945 was developed as a Colony-Stimulating Factor 1 Receptor (CSF-1R) inhibitor. Treatment of BLZ945 in rats and monkeys increased serum alanine aminotransferase (ALT) and aspartate transaminase (AST). However, liver hypertrophy was the only histopathological liver finding in rats, and there was no change in the livers of monkeys. Longer treatment of BLZ945 in rats for 6 weeks caused up to 6-fold elevation of ALT, yet hepatocyte necrosis was not detected microscopically. Toxicogenomic profiling of liver samples demonstrated that the genes associated with early response to liver injury, apoptosis/necrosis, inflammation, oxidative stress, and metabolic enzymes were upregulated. Studies are ongoing to evaluate the mechanisms underlying BL945-induced ALT and AST elevations.
- Published
- 2011
- Full Text
- View/download PDF
23. The Use of Rat Lens Explant Cultures to Study the Mechanism of Drug-Induced Cataractogenesis
- Author
-
Armin Wolf, Francois Pognan, Nandita Shangari, Lee Anne McLean, Elba Cruz, Parul Patel, Salah-Dine Chibout, Shruthi Sampath, Pierre Moulin, Steve Busch, Chiara Buono, Albert Reising, and Maya Gurnani
- Subjects
Male ,Agonist ,medicine.medical_specialty ,Cell Survival ,medicine.drug_class ,Acetates ,Toxicology ,Cataract ,PPAR agonist ,Tissue Culture Techniques ,Andrology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cataracts ,Ciglitazone ,Lens, Crystalline ,medicine ,Animals ,PPAR delta ,RNA, Messenger ,Rats, Wistar ,Eye Proteins ,Receptor ,Oligonucleotide Array Sequence Analysis ,030304 developmental biology ,0303 health sciences ,Chemistry ,Dimethyl sulfoxide ,Gene Expression Profiling ,medicine.disease ,Lens Fiber ,Rats ,Surgery ,Oxidative Stress ,Thiazoles ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Thiazolidinediones ,Explant culture - Abstract
Lens explant cultures were used to assess the mechanism of drug-induced cataractogenic potential of NVS001, a peroxisome proliferator-activated receptor delta (PPARδ) agonist, which resulted in cataract in all treated animals during a 13-week rat study. Ciglitazone, a PPARγ agonist and cataractogenic compound, was used as a positive control to validate this model. Rat lenses were extracted and cultured in medium supplemented with antibiotics for 24-h preincubation pretreatment. Lenses showing no signs of damage at the end of the preincubation pretreatment period were randomized into five experimental groups, (1) untreated control, (2) 0.1% dimethyl sulphoxide control, (3) 10μM NVS001, (4) 10μM ciglitazone, and (5) 10μM acetaminophen (negative control). Lenses were treated every 24 h after preincubation pretreatment for up to 48 h. Samples for viability, histology, and gene expression profiling were collected at 4, 24, and 48 h. There was a time-dependent increase in opacity, which correlated to a decrease in viability measured by adenosine triphosphate levels in NVS001 and ciglitazone-treated lenses compared with controls. NVS001 and ciglitazone had comparable cataractogenic effects after 48 h with histology showing rupture of the lens capsule, lens fiber degeneration, cortical lens vacuolation, and lens epithelial degeneration. Furthermore, no changes were seen when lenses were treated with acetaminophen. Gene expression analysis supported oxidative and osmotic stress, along with decreases in membrane and epithelial cell integrity as key factors in NVS001-induced cataracts. This study suggests that in vitro lens cultures can be used to assess cataractogenic potential of PPAR agonists and to study/understand the underlying molecular mechanism of cataractogenesis in rat.
- Published
- 2011
- Full Text
- View/download PDF
24. Imatinib does not induce cardiotoxicity at clinically relevant concentrations in preclinical studies
- Author
-
Francois Pognan, Min Dong, Armin Wolf, Ursula Junker, Jeffrey Tsao, David Ledieu, Ulrich W. Laengle, Danielle Roman, Anja Nussher, Olivier Grenet, Christian Trendelenburg, Gilles-Jacques Riviere, Philippe Couttet, Estelle Marrer, Elke Persohn, Marcia Heron, and Daniel Robert Roth
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Cmax ,Antineoplastic Agents ,Pharmacology ,Polymerase Chain Reaction ,Piperazines ,Mice ,hemic and lymphatic diseases ,Internal medicine ,medicine ,Animals ,Cytotoxic T cell ,Rats, Wistar ,neoplasms ,DNA Primers ,Cardiotoxicity ,Base Sequence ,business.industry ,Endoplasmic reticulum ,Heart ,Imatinib ,Hematology ,medicine.disease ,Rats ,Mice, Inbred C57BL ,Pyrimidines ,Endocrinology ,Imatinib mesylate ,Oncology ,Apoptosis ,Heart failure ,Benzamides ,Imatinib Mesylate ,business ,medicine.drug - Abstract
Cytotoxic concentrations of imatinib mesylate (10–50 μM) were required to trigger markers of apoptosis and endoplasmic reticulum stress response in neonatal rat ventricular myocytes and fibroblasts, with no significant differences observed between c-Abl silenced and nonsilenced cells. In mice, oral or intraperitoneal imatinib treatment did not induce cardiovascular pathology or heart failure. In rats, high doses of oral imatinib did result in some cardiac hypertrophy. Multi-organ toxicities may have increased the cardiac workload and contributed to the cardiac hypertrophy observed in rats only. These data suggest that imatinib is not cardiotoxic at clinically relevant concentrations (5 μM).
- Published
- 2010
- Full Text
- View/download PDF
25. Key Challenges and Opportunities Associated with the Use of In Vitro Models to Detect Human DILI: Integrated Risk Assessment and Mitigation Plans
- Author
-
Donna Dambach, Frederic Moulin, Okechukwu Ukairo, Richard J. Weaver, Franck A. Atienzar, Gilles Labbe, Philip Hewitt, J. Gerry Kenna, Eric A.G. Blomme, Minjun Chen, Yvonne Will, Laura Suter-Dick, Adrian Roth, and Francois Pognan
- Subjects
0301 basic medicine ,General Immunology and Microbiology ,business.industry ,Market Withdrawal ,lcsh:R ,Liver failure ,Data interpretation ,lcsh:Medicine ,Harmonization ,General Medicine ,Review Article ,General Biochemistry, Genetics and Molecular Biology ,3. Good health ,Toxicology ,03 medical and health sciences ,030104 developmental biology ,Risk analysis (engineering) ,Key (cryptography) ,Medicine ,Drug reaction ,business ,Risk assessment ,Pharmaceutical industry - Abstract
Drug-induced liver injury (DILI) is a major cause of late-stage clinical drug attrition, market withdrawal, black-box warnings, and acute liver failure. Consequently, it has been an area of focus for toxicologists and clinicians for several decades. In spite of considerable efforts, limited improvements in DILI prediction have been made and efforts to improve existing preclinical models or develop new test systems remain a high priority. While prediction of intrinsic DILI has improved, identifying compounds with a risk for idiosyncratic DILI (iDILI) remains extremely challenging because of the lack of a clear mechanistic understanding and the multifactorial pathogenesis of idiosyncratic drug reactions. Well-defined clinical diagnostic criteria and risk factors are also missing. This paper summarizes key data interpretation challenges, practical considerations, model limitations, and the need for an integrated risk assessment. As demonstrated through selected initiatives to address other types of toxicities, opportunities exist however for improvement, especially through better concerted efforts at harmonization of current, emerging and novel in vitro systems or through the establishment of strategies for implementation of preclinical DILI models across the pharmaceutical industry. Perspectives on the incorporation of newer technologies and the value of precompetitive consortia to identify useful practices are also discussed.
- Published
- 2016
26. Bile acids in drug induced liver injury: Key players and surrogate markers
- Author
-
Heiko Schadt, Salah-Dine Chibout, Michael Merz, Armin Wolf, Gerd A. Kullak-Ublick, Francois Pognan, University of Zurich, and Kullak-Ublick, Gerd A
- Subjects
0301 basic medicine ,medicine.drug_class ,Context (language use) ,610 Medicine & health ,Cholestasis, Intrahepatic ,Pharmacology ,digestive system ,Bile Acids and Salts ,03 medical and health sciences ,Cholestasis ,Enterohepatic Circulation ,medicine ,Homeostasis ,Humans ,2715 Gastroenterology ,Enterohepatic circulation ,Liver injury ,Hepatology ,Bile acid ,business.industry ,Gastroenterology ,Lipid metabolism ,Biological Transport ,medicine.disease ,3. Good health ,Diarrhea ,030104 developmental biology ,Biochemistry ,10199 Clinic for Clinical Pharmacology and Toxicology ,2721 Hepatology ,Bile Ducts ,medicine.symptom ,Chemical and Drug Induced Liver Injury ,business ,Biomarkers - Abstract
Bile acid research has gained great momentum since the role of bile acids as key signaling molecules in the enterohepatic circulation was discovered. Their physiological function in regulating their own homeostasis, as well as energy and lipid metabolism make them interesting targets for the pharmaceutical industry in the context of diseases such as bile acid induced diarrhea, bile acid induced cholestasis or nonalcoholic steatohepatitis. Changes in bile acid homeostasis are also linked to various types of drug-induced liver injury (DILI). However, the key question whether bile acids are surrogate markers for monitoring DILI or key pathogenic players in the onset and progression of DILI is under intense investigation. The purpose of this review is to summarize the different facets of bile acids in the context of normal physiology, hereditary defects of bile acid transport and DILI.
- Published
- 2016
27. Genomics, proteomics and metabonomics in toxicology: hopefully not ‘fashionomics’
- Author
-
Francois Pognan
- Subjects
Proteomics ,Pharmacology ,Principal Component Analysis ,Proteomics methods ,Metabolite ,Genomics ,Biology ,Toxicogenetics ,Toxicology ,chemistry.chemical_compound ,chemistry ,Genetics ,Animals ,Humans ,Molecular Medicine ,Toxicant - Abstract
Genomics, proteomics and metabonomics are applied to toxicology either as stand-alone technologies or in combination, with the intention of providing a more efficient assessment of the potential side effects of new chemical entities. Two different approaches are taken: a predictive/proactive strategy based on a statistical analogy of ‘signatures’ of drugs to many known toxicant gene or metabolite fingerprints; and a mechanistic/reactive strategy based on the in-depth biological analysis of the gene, protein or metabolite profiles induced by one or a few compounds of interest. This article focuses on the advantages and disadvantages of these technologies, as well as the many hurdles associated with both these approaches in toxicology that have to be considered before applying them to the assessment of future drugs.
- Published
- 2004
- Full Text
- View/download PDF
28. In utero transmission of Encephalitozoon cuniculi strain type I in rabbits
- Author
-
Francois Pognan and Philippe Baneux
- Subjects
Male ,Transplacental transmission ,Strain type ,Biology ,DNA, Ribosomal ,Polymerase Chain Reaction ,law.invention ,Pregnancy ,law ,parasitic diseases ,Animals ,Pregnancy Complications, Infectious ,Maternal-Fetal Exchange ,Encephalitozoon cuniculi ,reproductive and urinary physiology ,DNA Primers ,Fetus ,General Veterinary ,fungi ,virus diseases ,Sequence Analysis, DNA ,DNA, Protozoan ,biology.organism_classification ,Virology ,Infectious Disease Transmission, Vertical ,Transmission (mechanics) ,In utero ,Encephalitozoonosis ,Gestation ,Female ,Animal Science and Zoology ,Rabbits - Abstract
Pregnant rabbits were serologically diagnosed as having been infected with Encephalitozoon cuniculi. At necropsy at 28 days of gestation, does, placentas and fetuses were found to be infected with E. cuniculi strain type I as evidenced by using the nested-polymerase chain reaction (PCR) technique, thereby confirming vertical transplacental transmission.
- Published
- 2003
- Full Text
- View/download PDF
29. Genomics and Proteomics Analysis of Acetaminophen Toxicity in Mouse Liver
- Author
-
Francois Pognan, Joanne Shaw, Nicola Wallis, Stefan U. Ruepp, and Robert Tonge
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Time Factors ,Proteome ,NAPQI ,Kupffer Cells ,Blotting, Western ,Mitochondria, Liver ,Biology ,Mitochondrion ,Pharmacology ,Toxicology ,medicine.disease_cause ,DNA Adducts ,Mice ,chemistry.chemical_compound ,medicine ,Animals ,RNA, Messenger ,Acetaminophen ,Dose-Response Relationship, Drug ,Granulocyte-Macrophage Colony-Stimulating Factor ,Genomics ,Glutathione ,Microscopy, Electron ,Oxidative Stress ,medicine.anatomical_structure ,Liver ,chemistry ,Apoptosis ,Hepatocyte ,Toxicity ,Hepatocytes ,Electrophoresis, Polyacrylamide Gel ,Drug Overdose ,Oxidative stress ,medicine.drug - Abstract
Overdose of acetaminophen (APAP) causes severe centrilobular hepatic necrosis in humans and experimental animals. Here, to explore its mechanism, we administered APAP at subtoxic (150 mg/kg ip) and toxic (500 mg/kg ip) doses to overnight fasted mice. Animals were sacrificed at different time points from 15 min to 4 h postinjection. We assessed liver toxicity by plasma ALT activity and by electron microscopy. Using nylon filter arrays and RTQPCR, we performed genomics analysis in liver. We ran proteomics on liver mitochondrial subfractions using the newly developed quantitative fluorescent 2D-DIGE method (Amersham Pharmacia Biotech UK Limited). As soon as 15 min postinjection, centrilobular hepatocyte mitochondria were already slightly enlarged and GSH total content dropped by a third at top dose. GM-CSF mRNA, which is a granulocyte specific gene likely coming from resident Kupffer cells, was also induced to its maximum of 3-fold at both doses. Chaperone proteins Hsp10 and Hsp60 were readily decreased by half in mitochondria at both doses, most likely by leaking into cytoplasm. Although APAP is known as an apoptotic trigger, no apoptosis was observed at any time point. Most of the protein changes in mitochondria were present at 15 min postinjection, thus preceding most of the gene regulations. The decrease of ATP synthase subunits and beta-oxidation pathway proteins indicated a loss of energy production. As the morphology of mitochondria was also affected very early at top dose, we concluded that APAP toxicity was a direct action of its known reactive metabolite NAPQI, rather than a consequence of gene regulation. However, the latter will either worsen the toxicity or lead toward cell recovery depending on the cellular damage level.
- Published
- 2002
- Full Text
- View/download PDF
30. The IMI eTOX initiative – Data mining, read-across and predictive models for target evaluation and early drug candidate assessment
- Author
-
Ferran Sanz, Thomas Steger-Hartmann, and Francois Pognan
- Subjects
Computer science ,Drug candidate ,business.industry ,General Medicine ,Artificial intelligence ,Toxicology ,Machine learning ,computer.software_genre ,business ,computer - Published
- 2017
- Full Text
- View/download PDF
31. Learning from approved kinase inhibitors to better inform on the safety risks of specific kinases
- Author
-
Salah-Dine Chibout, Axel Vicart, Laszlo Urban, Francois Pognan, Berengere Dumotier, Jonathan G. Moggs, William Kluwe, Steven Whitebread, and Jacqueline Kinyamu-Akunda
- Subjects
business.industry ,Kinase ,Medicine ,General Medicine ,Pharmacology ,Toxicology ,business - Published
- 2017
- Full Text
- View/download PDF
32. Preclinical ontologies – A key feature for toxicity data exchange and mining
- Author
-
Francois Pognan, Carlo Ravagli, Philip Drew, and Philippe Marc
- Subjects
Toxicity data ,Feature (computer vision) ,Computer science ,business.industry ,Key (cryptography) ,General Medicine ,Artificial intelligence ,Toxicology ,business ,computer.software_genre ,computer ,Natural language processing - Published
- 2017
- Full Text
- View/download PDF
33. Phase II metabolism in human skin: skin explants show full coverage for glucuronidation, sulfation, N-acetylation, catechol methylation, and glutathione conjugation
- Author
-
Barbara Ling, Joanna Ashton-Chess, Barbara Bertschi, Hilmar Schiller, Olivier Kretz, Nenad Manevski, Dirk J. Schaefer, Reto Wettstein, Armin Wolf, Piet Swart, Markus Walles, Kamal Kumar Balavenkatraman, Francois Pognan, Karine Litherland, Gian Camenisch, and Peter Itin
- Subjects
Adult ,Male ,Diclofenac ,Glucuronidation ,Catechols ,Pharmaceutical Science ,Human skin ,Naphthols ,Methylation ,chemistry.chemical_compound ,Sulfation ,Glucuronides ,Humans ,Biotransformation ,Aged ,Skin ,Pharmacology ,Catechol ,integumentary system ,Sulfates ,Acetylation ,Glutathione ,Middle Aged ,Metabolic Detoxication, Phase II ,chemistry ,Biochemistry ,Liver ,Toxicity ,Female ,Drug metabolism - Abstract
Although skin is the largest organ of the human body, cutaneous drug metabolism is often overlooked, and existing experimental models are insufficiently validated. This proof-of-concept study investigated phase II biotransformation of 11 test substrates in fresh full-thickness human skin explants, a model containing all skin cell types. Results show that skin explants have significant capacity for glucuronidation, sulfation, N-acetylation, catechol methylation, and glutathione conjugation. Novel skin metabolites were identified, including acyl glucuronides of indomethacin and diclofenac, glucuronides of 17β-estradiol, N-acetylprocainamide, and methoxy derivatives of 4-nitrocatechol and 2,3-dihydroxynaphthalene. Measured activities for 10 μM substrate incubations spanned a 1000-fold: from the highest 4.758 pmol·mg skin(-1)·h(-1) for p-toluidine N-acetylation to the lowest 0.006 pmol·mg skin(-1)·h(-1) for 17β-estradiol 17-glucuronidation. Interindividual variability was 1.4- to 13.0-fold, the highest being 4-methylumbelliferone and diclofenac glucuronidation. Reaction rates were generally linear up to 4 hours, although 24-hour incubations enabled detection of metabolites in trace amounts. All reactions were unaffected by the inclusion of cosubstrates, and freezing of the fresh skin led to loss of glucuronidation activity. The predicted whole-skin intrinsic metabolic clearances were significantly lower compared with corresponding whole-liver intrinsic clearances, suggesting a relatively limited contribution of the skin to the body's total systemic phase II enzyme-mediated metabolic clearance. Nevertheless, the fresh full-thickness skin explants represent a suitable model to study cutaneous phase II metabolism not only in drug elimination but also in toxicity, as formation of acyl glucuronides and sulfate conjugates could play a role in skin adverse reactions.
- Published
- 2014
34. The eTOX data-sharing project to advance in silico drug-induced toxicity prediction
- Author
-
Montserrat Cases, Thomas Steger-Hartmann, Katharine Briggs, Ferran Sanz, Thomas Kleinöder, Francois Pognan, Christof H. Schwab, Jörg D. Wichard, Manuel Pastor, and Philippe Marc
- Subjects
Decision support system ,Medicaments -- Toxicologia ,in vitro toxicity ,decision support system ,in silico toxicity ,Databases, Pharmaceutical ,data sharing ,Intellectual property ,in vivo toxicity ,Bioinformatics ,computer.software_genre ,Predictive models ,lcsh:Chemistry ,Drug Discovery ,Medicine ,Data Mining ,ontologies ,lcsh:QH301-705.5 ,Spectroscopy ,Pharmaceutical industry ,In vivo toxicity ,QSAR ,General Medicine ,In vitro toxicity ,predictive models ,In silico toxicity ,3. Good health ,Computer Science Applications ,Drug development ,Pharmaceutical Preparations ,Vocabulary, Controlled ,Data integration ,Ontologies (Recuperació de la informació) ,read-across ,Drug-Related Side Effects and Adverse Reactions ,Public domain ,Models, Biological ,Catalysis ,Article ,Inorganic Chemistry ,Controlled vocabulary ,Ontologies ,Humans ,Computer Simulation ,Physical and Theoretical Chemistry ,Molecular Biology ,data integration ,business.industry ,Organic Chemistry ,Data science ,Data sharing ,lcsh:Biology (General) ,lcsh:QD1-999 ,Read-across ,business ,computer - Abstract
The high-quality in vivo preclinical safety data produced by the pharmaceutical industry during drug development, which follows numerous strict guidelines, are mostly not available in the public domain. These safety data are sometimes published as a condensed summary for the few compounds that reach the market, but the majority of studies are never made public and are often difficult to access in an automated way, even sometimes within the owning company itself. It is evident from many academic and industrial examples, that useful data mining and model development requires large and representative data sets and careful curation of the collected data. In 2010, under the auspices of the Innovative Medicines Initiative, the eTOX project started with the objective of extracting and sharing preclinical study data from paper or pdf archives of toxicology departments of the 13 participating pharmaceutical companies and using such data for establishing a detailed, well-curated database, which could then serve as source for read-across approaches (early assessment of the potential toxicity of a drug candidate by comparison of similar structure and/or effects) and training of predictive models. The paper describes the efforts undertaken to allow effective data sharing intellectual property (IP) protection and set up of adequate controlled vocabularies) and to establish the database (currently with over 4000 studies contributed by the pharma companies corresponding to more than 1400 compounds). In addition, the status of predictive models building and some specific features of the eTOX predictive system (eTOXsys) are presented as decision support knowledge-based tools for drug development process at an early stage. The research leading to these results has received support from the Innovative Medicines Initiative Joint Undertaking under grant agreement n° 115002 (eTOX), resources of which are composed of financial contribution from the European Union’s Seventh Framework Programme (FP7/2007-2013) and EFPIA companies’ in kind contributions. The authors would like to formally acknowledge the contribution to the eTOX project of all scientists and other staff involved
- Published
- 2014
35. Aldehyde oxidase activity in fresh human skin
- Author
-
Olivier Kretz, Reto Wettstein, Francois Pognan, Nenad Manevski, Armin Wolf, Kamal Kumar Balavenkatraman, Gian Camenisch, Barbara Ling, Piet Swart, Markus Walles, Dirk J. Schaefer, Barbara Bertschi, Hilmar Schiller, and Karine Litherland
- Subjects
Adult ,Male ,Toluidines ,Glucuronidation ,Pharmaceutical Science ,Human skin ,Hydroxylation ,Guanidines ,chemistry.chemical_compound ,Sulfation ,Humans ,Enzyme kinetics ,Aldehyde oxidase ,Aged ,Skin ,Pharmacology ,chemistry.chemical_classification ,Chemistry ,Middle Aged ,Hydralazine ,Metabolic Detoxication, Phase II ,Aldehyde Oxidase ,Kinetics ,Enzyme ,Biochemistry ,Pyrazoles ,Female ,Carbamates ,Drug metabolism - Abstract
Human aldehyde oxidase (AO) is a molybdoflavoenzyme that commonly oxidizes azaheterocycles in therapeutic drugs. Although high metabolic clearance by AO resulted in several drug failures, existing in vitro-in vivo correlations are often poor and the extrahepatic role of AO practically unknown. This study investigated enzymatic activity of AO in fresh human skin, the largest organ of the body, frequently exposed to therapeutic drugs and xenobiotics. Fresh, full-thickness human skin was obtained from 13 individual donors and assayed with two specific AO substrates: carbazeran and zoniporide. Human skin explants from all donors metabolized carbazeran to 4-hydroxycarbazeran and zoniporide to 2-oxo-zoniporide. Average rates of carbazeran and zoniporide hydroxylations were 1.301 and 0.164 pmol⋅mg skin(-1)⋅h(-1), resulting in 13 and 2% substrate turnover, respectively, after 24 hours of incubation with 10 μM substrate. Hydroxylation activities for the two substrates were significantly correlated (r(2) = 0.769), with interindividual variability ranging from 3-fold (zoniporide) to 6-fold (carbazeran). Inclusion of hydralazine, an irreversible inhibitor of AO, resulted in concentration-dependent decrease of hydroxylation activities, exceeding 90% inhibition of carbazeran 4-hydroxylation at 100 μM inhibitor. Reaction rates were linear up to 4 hours and well described by Michaelis-Menten enzyme kinetics. Comparison of carbazeran and zoniporide hydroxylation with rates of triclosan glucuronidation and sulfation and p-toluidine N-acetylation showed that cutaneous AO activity is comparable to tested phase II metabolic reactions, indicating a significant role of AO in cutaneous drug metabolism. To our best knowledge, this is the first report of AO enzymatic activity in human skin.
- Published
- 2014
36. High Content Imaging Approaches for In Vitro Toxicology
- Author
-
Francois Pognan, Marianne Uteng, Kamal Kumar Balavenkatraman, Armin Wolf, and Davide Germano
- Subjects
Chemistry ,In vitro toxicology ,High content imaging ,Biomedical engineering - Published
- 2014
- Full Text
- View/download PDF
37. Investigative safety science as a competitive advantage for Pharma
- Author
-
Michael Merz, André Cordier, Pierre Moulin, Jonathan G. Moggs, David J. Heard, Michele Leonard, Francois Pognan, Page Bouchard, Steve Busch, Salah-Dine Chibout, Michael Kammüller, and Dominique Brees
- Subjects
Pharmacology ,Noninvasive imaging ,Drug Industry ,Drug-Related Side Effects and Adverse Reactions ,business.industry ,Drug Evaluation, Preclinical ,Computational Biology ,General Medicine ,Toxicology ,Bioinformatics ,Competitive advantage ,Translational Research, Biomedical ,Expert opinion ,Models, Animal ,Toxicity Tests ,Medicine ,Clinical safety ,Animals ,Humans ,Engineering ethics ,business ,Pharmaceutical industry - Abstract
Following a US National Academy of Sciences report in 2007 entitled "Toxicity Testing of the 21st Century: a Vision and a Strategy," significant advances within translational drug safety sciences promise to revolutionize drug discovery and development. The purpose of this review is to outline why investigative safety science is a competitive advantage for the pharmaceutical industry.The article discusses the essential goals for modern investigative toxicologists including: cross-species target biology; molecular pathways of toxicity; and development of predictive tools, models and biomarkers that allow discovery researchers and clinicians to anticipate safety problems and plan ways to address them, earlier than ever before. Furthermore, the article emphasizes the importance of investigating unanticipated clinical safety signals through a combination of mechanistic preclinical studies and/or molecular characterization of clinical samples from affected organs.The traditional boundaries between pharma industry teams focusing on safety/efficacy and preclinical/clinical development are rapidly disappearing in favor of translational safety science-centric organizations with a vision of bringing more effective medicines forward safely and quickly. Comparative biology and mechanistic toxicology approaches facilitate: i) identifying translational safety biomarkers; ii) identifying new drug targets/indications; and iii) mitigating off-target toxicities. These value-adding safety science contributions will change traditional toxicologists from side-effect identifiers to drug development enablers.
- Published
- 2012
38. Perturbation of microRNAs in Rat Heart during Chronic Doxorubicin Treatment
- Author
-
Brian R. Berridge, Yasmina Bauer, Martin Letzkus, Philippe Couttet, Caterina Vacchi-Suzzi, Kevin Gerrish, Jonathan J. Lyon, Sandrine Bongiovanni, Francois Pognan, Olivier Grenet, Martin P. Vidgeon-Hart, Richard S. Paules, Frank Staedtler, Jonathan G. Moggs, and Hisham K. Hamadeh
- Subjects
Male ,Muscle Proteins ,Bioinformatics ,Toxicology ,Rats, Sprague-Dawley ,Molecular cell biology ,RNA interference ,Gene expression ,Etoposide ,Cellular Stress Responses ,Multidisciplinary ,Antibiotics, Antineoplastic ,GTPase-Activating Proteins ,Nuclear Proteins ,Genomics ,Animal Models ,Cardiovascular physiology ,Up-Regulation ,Nucleic acids ,Medicine ,Cardiomyopathies ,medicine.drug ,Research Article ,Transcriptional Activation ,Science ,Toxic Agents ,Predictive Toxicology ,Biology ,Model Organisms ,microRNA ,medicine ,Animals ,Humans ,Doxorubicin ,RNA, Messenger ,Adaptor Proteins, Signal Transducing ,Myocardium ,RNA stability ,medicine.disease ,Rats ,MicroRNAs ,HEK293 Cells ,RNA processing ,Heart failure ,Vacuoles ,Cancer research ,Rat ,RNA ,Topoisomerase-II Inhibitor ,Genome Expression Analysis ,Pharmacogenomics ,Transcriptome ,Biomarkers - Abstract
Anti-cancer therapy based on anthracyclines (DNA intercalating Topoisomerase II inhibitors) is limited by adverse effects of these compounds on the cardiovascular system, ultimately causing heart failure. Despite extensive investigations into the effects of doxorubicin on the cardiovascular system, the molecular mechanisms of toxicity remain largely unknown. MicroRNAs are endogenously transcribed non-coding 22 nucleotide long RNAs that regulate gene expression by decreasing mRNA stability and translation and play key roles in cardiac physiology and pathologies. Increasing doses of doxorubicin, but not etoposide (a Topoisomerase II inhibitor devoid of cardiovascular toxicity), specifically induced the up-regulation of miR-208b, miR-216b, miR-215, miR-34c and miR-367 in rat hearts. Furthermore, the lowest dosing regime (1 mg/kg/week for 2 weeks) led to a detectable increase of miR-216b in the absence of histopathological findings or alteration of classical cardiac stress biomarkers. In silico microRNA target predictions suggested that a number of doxorubicin-responsive microRNAs may regulate mRNAs involved in cardiac tissue remodeling. In particular miR-34c was able to mediate the DOX-induced changes of Sipa1 mRNA (a mitogen-induced Rap/Ran GTPase activating protein) at the post-transcriptional level and in a seed sequence dependent manner. Our results show that integrated heart tissue microRNA and mRNA profiling can provide valuable early genomic biomarkers of drug-induced cardiac injury as well as novel mechanistic insight into the underlying molecular pathways.
- Published
- 2012
39. Preclinical evaluation of potential nilotinib cardiotoxicity
- Author
-
Francois Pognan, Elke Persohn, Estelle Marrer, Wei Zhou, Philippe Couttet, Marcia Heron, Ursula Junker, Olivier Grenet, Armin Wolf, David Ledieu, Jeffrey Tsao, Danielle Roman, Andreas Mahl, and Min Dong
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Heart Ventricles ,Drug Evaluation, Preclinical ,Pharmacology ,Cardiotoxins ,Tyrosine-kinase inhibitor ,Rats, Sprague-Dawley ,Random Allocation ,In vivo ,Internal medicine ,medicine ,Animals ,Ventricular Function ,Myocytes, Cardiac ,Rats, Wistar ,Cells, Cultured ,Cardiotoxicity ,Dose-Response Relationship, Drug ,business.industry ,Heart ,Hematology ,medicine.disease ,Cardiovascular physiology ,Rats ,Endocrinology ,Pyrimidines ,Oncology ,Nilotinib ,Animals, Newborn ,Apoptosis ,Heart failure ,Toxicity ,business ,medicine.drug - Abstract
In vitro, concentrations ≥ 10 μM of nilotinib were needed to induce markers of cytotoxicity, apoptosis, and endoplasmic reticulum stress in both neonatal rat ventricular myocytes, a putative target tissue, and non-target heart fibroblasts, indicating a lack of cardiomyocyte-specific nilotinib toxicity in vitro. In rats, oral nilotinib treatment at 80 mg/kg for 4 weeks induced increased heart weight; however, this was not associated with relevant histopathological changes or effects on heart function. Thus, nilotinib at and above clinically relevant concentrations (4.27 μM) did not induce overt cardiovascular pathologies or heart failure in vitro or in vivo under study conditions.
- Published
- 2010
40. Mapping the epigenome — impact for toxicology
- Author
-
Francois Pognan, Salah-Dine Chibout, Soon-Siong Teo, Jonathan G. Moggs, and Jennifer Marlowe
- Subjects
Drug ,Immune Stimulation ,media_common.quotation_subject ,Human Epigenome Project ,Computational biology ,Epigenome ,Epigenetics ,Disease ,Biology ,Epigenomics ,media_common ,Epigenesis - Abstract
Recent advances in technological approaches for mapping and characterizing the epigenome are generating a wealth of new opportunities for exploring the relationship between epigenetic modifications, human disease and the therapeutic potential of pharmaceutical drugs. While the best examples for xenobiotic-induced epigenetic perturbations come from the field of non-genotoxic carcinogenesis, there is growing evidence for the relevance of epigenetic mechanisms associated with a wide range of disease areas and drug targets. The application of epigenomic profiling technologies to drug safety sciences has great potential for providing novel insights into the molecular basis of long-lasting cellular perturbations including increased susceptibility to disease and/or toxicity, memory of prior immune stimulation and/or drug exposure, and transgenerational effects.
- Published
- 2009
- Full Text
- View/download PDF
41. Mechanistic investigation of N,N-diethyl-4-(phenyl-piperidin-4-ylidenemethyl)-benzamide-induced insulin depletion in the rat and RINm5F cells
- Author
-
Calvert Louden, Francois Pognan, Monicah A. Otieno, Thomas J. Hudzik, Nicole Bavuso, Debra Wescott, Martin Dyroff, Joseph Milano, Yan Li, Khanh-Hui Bui, and Linda Foster-Brown
- Subjects
Agonist ,Blood Glucose ,medicine.medical_specialty ,Calcium Channels, L-Type ,medicine.drug_class ,medicine.medical_treatment ,Biology ,Toxicology ,Pertussis toxin ,chemistry.chemical_compound ,Piperidines ,In vivo ,Naltrindole ,Internal medicine ,Receptors, Opioid, delta ,medicine ,Animals ,Cyclizine ,Insulin ,RNA, Messenger ,Rats, Wistar ,Benzamide ,Pancreas ,Cells, Cultured ,Calcium metabolism ,Dose-Response Relationship, Drug ,Rats ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Benzamides ,Calcium ,medicine.drug - Abstract
These studies describe the effect of N,N-diethyl-4-(phenyl-piperidin-4-ylidenemethyl)-benzamide (AR-M100390), a delta-opioid agonist, on the pancreas and its mechanisms for pancreatic toxicity. Rats were treated with 5, 100, and 600 micromol/kg of AR-M100390 for 3 and/or 7 days; another group of rats treated with 600 micromol/kg of compound were allowed to recover for 14 days. AR-M100390 (600 micromol/kg) caused vacuolation in the beta-cell of the rat pancreas that was associated with depletion of insulin and hyperglycemia after 7 days of dosing. The loss of insulin by AR-M100390 was due to specific inhibition of rat insulin2 mRNA transcription in vivo. Insulin depletion and hyperglycemia were reversible. The effects of AR-M100390 in rats were reproduced in the rat pancreatic beta-cell line RINm5F, where it inhibited intracellular insulin content and secretion without affecting cell survival. Loss of insulin in vitro was also a result of specific inhibition of insulin2 mRNA transcription and was reversible. Pretreatment of cells with the delta-opioid antagonist naltrindole or pertussis toxin did not reverse loss of insulin in AR-M100390-treated cells suggesting that the effects were not mediated by the delta-opioid receptor. AR-M100390 inhibited KCl-mediated calcium mobilization in RINm5F cells, suggesting that L-type calcium channels found in these cells and in pancreatic beta-cells may partially play a role in the inhibition of insulin secretion by this compound. In summary, the in vitro and in vivo studies suggest that inhibition of insulin by AR-M100390 is due to a combination of inhibition of insulin synthesis and/or release.
- Published
- 2008
42. Toxicogenomics applied to predictive and exploratory toxicology for the safety assessment of new chemical entities: a long road with deep potholes
- Author
-
Francois Pognan
- Subjects
Toxicology ,Metabolomics ,Expression data ,Event based ,Protein database ,Identification (biology) ,Biology ,Toxicogenomics ,Predictive value ,Phenotype - Abstract
Toxicology is the perturbation of metabolism by external factors such as xenobiotics, environmental factors or drugs. As such, toxicology covers a broad range of fields from studies of the whole organism responses to minute biochemical events. Mechanistic toxicogenomics is an attempt to harness genomic tools to understand the physiological basis for a toxic event based on an analysis of transcriptional, translational or metabolomic profiles. These studies are complicated by non-toxic adaptive responses in transcript, protein or metabolite expression levels that have to be distinguished from those that are proximally related to the toxic event. Substantial progress has been made on the identification of biomarkers and the establishment of screens derived from such toxicogenomics studies. The ultimate goal, of course, is predictive toxicogenomics, which is an attempt to infer the likelihood of occurrence of a toxic event with exposure to a new agent based upon comparative responses with large databases of gene, protein or metabolite expression data. Gene expression databases are currently limited by the fact that measurable toxic phenotypes generally precede or at best coincide with the earliest observable changes in transcriptional profiles. Unfortunately, predictive protein databases have been limited by technical difficulties. Metabonomics-based databases, which would probably have the highest predictive value, are limited in turn by the inability to perform high dose studies in humans. This chapter will conclude by reviewing those elements of toxicogenomics that apply specifically to the development of anti-infectives and the potential for accuratelymodelling the toxicity of future drugs.
- Published
- 2007
- Full Text
- View/download PDF
43. Pharmacodynamic profiling of EGFR inhibitors in human keratinocytes
- Author
-
Francois Pognan, Kamal Kumar Balavenkatraman, Armin Wolf, B. Bertschi, Philippe Couttet, S-D. Chibout, U. Hopfer, N. Rathfelder, M. Marcellin, B. Greutmann, David Ledieu, A. Lambert, Axel Vicart, and Jonathan G. Moggs
- Subjects
business.industry ,Pharmacodynamics ,Medicine ,Profiling (information science) ,General Medicine ,Pharmacology ,Toxicology ,business ,EGFR inhibitors - Published
- 2013
- Full Text
- View/download PDF
44. Determination of liver specific toxicities in rat hepatocytes by High Content Imaging during 2-week multiple treatment
- Author
-
Salah-Dine Chibout, Olivier Grenet, Armin Wolf, Philippe Couttet, Francois Pognan, Davide Germano, and Marianne Uteng
- Subjects
business.industry ,Medicine ,General Medicine ,Pharmacology ,Toxicology ,business ,High content imaging - Published
- 2013
- Full Text
- View/download PDF
45. Modulation of notch processing by gamma-secretase inhibitors causes intestinal goblet cell metaplasia and induction of genes known to specify gut secretory lineage differentiation
- Author
-
Linda Foster-Brown, Joseph Milano, Anna Zacco, Claude Dagenais, Barry D. Greenberg, Robert Toms Jacobs, Paul J. Ciaccio, Jenny McKay, Francois Pognan, and Reto Gadient
- Subjects
Male ,medicine.medical_specialty ,Large Intestinal Glandular Cell ,Notch signaling pathway ,Apoptosis ,Receptors, Cell Surface ,Toxicology ,Benzodiazepines ,Dibenzazepines ,Internal medicine ,Metaplasia ,Cell Line, Tumor ,Endopeptidases ,medicine ,Animals ,Aspartic Acid Endopeptidases ,Humans ,RNA, Messenger ,HES1 ,Enzyme Inhibitors ,Rats, Wistar ,Receptor, Notch1 ,NeuroD ,Goblet cell ,Sulfonamides ,biology ,Dose-Response Relationship, Drug ,Cell Differentiation ,Molecular biology ,Rats ,medicine.anatomical_structure ,Endocrinology ,dBZ ,Gene Expression Regulation ,biology.protein ,Goblet Cells ,medicine.symptom ,Amyloid Precursor Protein Secretases ,Amyloid precursor protein secretase ,Biomarkers ,Signal Transduction ,Transcription Factors - Abstract
It is anticipated that gamma-secretase inhibitors (gamma-Sec-I) that modulate Notch processing will alter differentiation in tissues whose architecture is governed by Notch signaling. To explore this hypothesis, Han Wistar rats were dosed for up to 5 days with 10-100 micromol/kg b.i.d. gamma-Sec-I from three chemical series that inhibit Notch processing in vitro at various potencies (Notch IC(50)). These included an arylsulfonamide (AS) (142 nM), a dibenzazepine (DBZ) (1.7 nM), and a benzodiazepine (BZ) (2.2 nM). The DBZ and BZ caused dose-dependent intestinal goblet cell metaplasia. In contrast, the AS produced no detectable in vivo toxicity, despite higher exposure to free drug. In a time course using BZ, small intestinal crypt cell and large intestinal glandular cell epithelial apoptosis was observed on days 1-5, followed by goblet cell metaplasia on days 2-5 and crypt epithelial and glandular epithelial regenerative hyperplasia on days 4-5. Gene expression profiling of duodenal samples from BZ-dosed animals revealed significant time-dependent deregulation of mRNAs for various panendocrine, hormonal, and transcription factor genes. Somatostatin, secretin, mucin, CCK, and gastrin mRNAs were elevated twofold or more by day 2, and a number of candidate "early-predictive" genes were altered on days 1-2, remaining changed for 4-5 days; these included Delta1, NeuroD, Hes1-regulated adipsin, and the Hes-regulated transcriptional activator of gut secretory lineage differentiation, the rat homolog of Drosophila atonal, Rath1. Western blotting of fecal protein from BZ-and DBZ-dosed animals exhibited increased levels of both anti-Rath1 reactive protein and anti-adipsin reactive proteins, confirming their potential value as noninvasive biomarkers of intestinal goblet metaplasia.
- Published
- 2004
46. Integrated application of transcriptomics and metabonomics yields new insight into the toxicity due to paracetamol in the mouse
- Author
-
Francois Pognan, John C. Lindon, Stefan U. Ruepp, Muireann Coen, Ian D. Wilson, Jeremy K. Nicholson, and Eva M. Lenz
- Subjects
Male ,Very low-density lipoprotein ,Magnetic Resonance Spectroscopy ,Time Factors ,Clinical Biochemistry ,Pharmaceutical Science ,Gene Expression ,Mice, Inbred Strains ,Analytical Chemistry ,chemistry.chemical_compound ,Mice ,Drug Discovery ,Toxicity Tests ,medicine ,Animals ,Glycolysis ,Aspartate Aminotransferases ,RNA, Messenger ,Spectroscopy ,Acetaminophen ,Oligonucleotide Array Sequence Analysis ,Alanine ,Glycogen ,Dose-Response Relationship, Drug ,Reverse Transcriptase Polymerase Chain Reaction ,Lipid metabolism ,Alanine Transaminase ,Analgesics, Non-Narcotic ,Lipid Metabolism ,Metabolic pathway ,chemistry ,Biochemistry ,Liver ,Toxicity ,Proto-Oncogene Proteins c-fos ,Injections, Intraperitoneal ,medicine.drug - Abstract
Gene chip array (Affymetrix) data from liver tissue and high resolution 1H NMR spectra from intact liver tissue, tissue extracts and plasma have been analyzed to identify biochemical changes arising from hepatotoxicity in mice dosed with acetaminophen. These data sets have been co-interpreted in terms of common metabolic pathways. The principal metabolic changes comprised a decrease in hepatic glucose and glycogen in intact tissue, coupled with an increase in lipid content, with increases in the levels of glucose, pyruvate, acetate and lactate in plasma, and increases in alanine and lactate in the aqueous tissue extracts. Collectively these data provide evidence for an increased rate of hepatic glycolysis. The metabolic observations were consistent with the altered levels of gene expression relating to lipid and energy metabolism in liver which both preceded and were concurrent with the metabolic perturbations. The results show that these two technology platforms together offer a complementary view into cellular responses to toxic processes, providing new insight into the toxic consequences, even for well-studied therapeutic agents such as acetaminophen.
- Published
- 2003
47. An integrated metabonomic investigation of acetaminophen toxicity in the mouse using NMR spectroscopy
- Author
-
Muireann Coen, John C. Lindon, Francois Pognan, Ian D. Wilson, Jeremy K. Nicholson, and E. M. Lenz
- Subjects
Male ,Time Factors ,Mitochondria, Liver ,Carbohydrate metabolism ,Mitochondrion ,Toxicology ,chemistry.chemical_compound ,Mice ,Animals ,Glycolysis ,Nuclear Magnetic Resonance, Biomolecular ,Acetaminophen ,chemistry.chemical_classification ,Principal Component Analysis ,Glycogen ,Dose-Response Relationship, Drug ,Chemistry ,Fatty acid ,General Medicine ,Peroxisome ,Lipid Metabolism ,Lipids ,Citric acid cycle ,Glucose ,Biochemistry ,Liver ,Liver Extracts ,Polyunsaturated fatty acid - Abstract
An integrated metabonomics study using high-resolution 1H NMR spectroscopy has been applied to investigate the biochemical composition of intact liver tissue (using magic angle spinning), liver tissue extracts, and blood plasma samples obtained from control and acetaminophen-treated mice. Principal components analysis was used to visualize similarities and differences in biochemical profiles. The time- and dose-dependent biochemical effects of acetaminophen were related to the drug toxicity, as determined using histopathology. Metabolic effects in intact liver tissue and lipid soluble liver tissue extracts from animals treated with the high dose level of acetaminophen included an increase in lipid triglycerides and monounsaturated fatty acids together with a decrease in polyunsaturated fatty acids, indicating mitochondrial malfunction with concomitant compensatory increase of peroxisomal activity. In addition, a depletion of phospholipids was observed in treated liver tissue, which suggested an inhibition of enzymes involved in phospholipid synthesis. There was also a depletion in the levels of liver glucose and glycogen. In addition, the aqueous soluble liver tissue extracts from high dose animals also revealed an increase in lactate, alanine, and other amino acids, together with a decrease in glucose. Plasma spectra showed increases in glucose, acetate, pyruvate, and lactate. These observations all provide evidence for an increased rate of glycolysis. These findings could indicate a mitochondrial inability to use pyruvate in the citric acid cycle and also reveal the impairment of fatty acid beta-oxidation in liver mitochondria of such treated mice.
- Published
- 2003
48. Inter-laboratory validation of procedures for measuring 8-oxo-7,8-dihydroguanine/8-oxo-7,8-dihydro-2’-deoxyguanosine in DNA
- Author
-
C. Casalini, Nicole Phoa, Ruan M. Elliott, Jean-Luc Ravanat, Hiroshi Kasai, Rozenn Legall, Bénédicte Morin, Piero Dolara, Bente Jensen, Ana Lloret, Matthew J. Gordon, Christopher P. Wild, Bernd Epe, Rie Kido, Samantha Johnson, Karl E. Herbert, Eric Gremaud, Laura J. Hardie, Andrew Collins, Zdena Durackova, Rex M. Tyrrell, Siân Astley, Ryszard Olinski, Parul R. Patel, Henrik E. Poulsen, Laurent B. Fay, Pierre Duez, Andrea White, Pierre Cillard, Cristina Luceri, Annie Aarup Jensen, Csilla Mislanova, Michèle Tripier, Jean Cadet, Tim Hofer, Lucia Hlincíková, Mark D. Evans, Isabelle Morel, Henry Faure, Dina Chauhan, Angela Bailley, Chrisi Dunster, Ian D. Podmore, Karol Bialkowski, Odile Sergent, Jean-François Rees, Jose Viña, Francois Pognan, Peter Korytar, Allan Weimann, Anke Pelzer, Joseph Lunec, F. Guglielmi, Jacques Dubois, Thierry Douki, Frank J. Kelly, Catherine M. Gedik, Lennart Möller, Elizabeth C. Smith, Steffen Loft, Richard H. Stadler, Maria Dusinska, John O'Brien, Sharon G. Wood, Liliane Degand, Ann White, Julie Eakins, and Andrea Hartwig
- Subjects
Guanine ,Analytical chemistry ,Test sensitivity ,Thymus Gland ,Sensitivity and Specificity ,Biochemistry ,Gas Chromatography-Mass Spectrometry ,Mass Spectrometry ,Oxidative dna damage ,chemistry.chemical_compound ,8 oxo 7 8 dihydroguanine ,Animals ,Humans ,European commission ,Inter-laboratory ,Chromatography, High Pressure Liquid ,Chromatography ,Chemistry ,8 oxo 7 8 dihydro 2 deoxyguanosine ,DNA ,General Medicine ,Cattle ,Biomarkers ,Chromatography, Liquid ,DNA Damage - Abstract
The aim of ESCODD, a European Commission funded Concerted Action, is to improve the precision and accuracy of methods for measuring 8-oxo-7,8-dihydroguanine (8-oxoGua) or the nucleoside (8-oxodG). On two occasions, participating laboratories received samples of different concentrations of 8-oxodG for analysis. About half the results returned (for 8-oxodG) were within 20% of the median values. Coefficients of variation (for three identical samples) were commonly around 10%. A sample of calf thymus DNA was sent, dry, to all laboratories. Analysis of 8-oxoGua/8-oxodG in this sample was a test of hydrolysis methods. Almost half the reported results were within 20% of the median value, and half obtained a CVof less than 10%. In order to test sensitivity, as well as precision, DNA was treated with photosensitiser and light to introduce increasing amounts of 8-oxoGua and samples were sent to members. Median values calculated from all returned results were 45.6 (untreated), 53.9, 60.4 and 65.6 8-oxoGua/10(6) Gua; only seven laboratories detected the increase over the whole range, while all but one detected a dose response over two concentration intervals. Results in this trial reflect a continuing improvement in precision and accuracy. The next challenge will be the analysis of 8-oxodG in DNA isolated from cells or tissue, where the concentration is much lower than in calf thymus DNA.
- Published
- 2002
- Full Text
- View/download PDF
49. Bile acids profiling of rat plasma after treatment with Cyclosporin A
- Author
-
Heiko Schadt, Francois Pognan, Armin Wolf, Daniel P. Stiehl, Salah-Dine Chibout, Audrey Dietz, Carlos A. Penno, Catia Vicente Vieira, Pierre Moulin, and Marianne Schwald
- Subjects
Chemistry ,Cyclosporin a ,General Medicine ,Pharmacology ,Toxicology ,After treatment - Published
- 2014
- Full Text
- View/download PDF
50. Validation and development of fluorescence two-dimensional differential gel electrophoresis proteomics technology
- Author
-
Francois Pognan, Brian Middleton, Joanne Shaw, Steve Rayner, Janice Young, Robert Tonge, Matthew Davison, Edward Hawkins, Rachel Rowlinson, and Ian Currie
- Subjects
Gel electrophoresis ,Male ,Chromatography ,Proteome ,Chemistry ,Difference gel electrophoresis ,Proteins ,Proteomics ,Mass spectrometry ,Biochemistry ,Fluorescence ,Mass Spectrometry ,chemistry.chemical_compound ,Mice ,Liver ,Labelling ,Animals ,Electrophoresis, Gel, Two-Dimensional ,Cyanine ,Molecular Biology ,Two-Dimensional Difference Gel Electrophoresis ,Acetaminophen ,Biotechnology ,Fluorescent Dyes - Abstract
Fluorescence two-dimensional differential gel electrophoresis (2-D DIGE*) is a new development in protein detection for two-dimensional gels. Using mouse liver homogenates (control and paracetamol (N-acetyl-p-aminophenol, APAP)-treated), we have determined the quantitative variation in the 2-D DIGE process and established statistically valid thresholds for assigning quantitative changes between samples. Thresholds were dependent on normalised spot volume, ranged from approximately 1.2 fold for large volume spots to 3.5 fold for small volume spots and were not markedly affected by the particular cyanine dye combination or by multiple operators carrying out the dye labelling reaction. To minimise the thresholds, substantial user editing was required when using ImageMaster 2D-Elite software. The difference thresholds were applied to the test system and quantitative protein differences were determined using replicate gels of pool samples and single gels from multiple individual animals (control vs treated in each gel). Throughout, the differences revealed with a particular cyanine dye combination were mirrored almost without exception when the dye combination was reversed. Both pool and individual sample analyses provided unique data to the study. The inter-animal response variability in inbred mice was approximately nine times that contributed by the 2-D DIGE process. A number of the most frequently observed protein changes resulting from APAP-treatment were identified by mass spectrometry. Several of these can be rationalised based on available data on the mechanism of APAP hepatotoxicity but others cannot, indicating that proteomics can provide further insights into the biochemical basis of APAP toxicity.
- Published
- 2001
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.