1
|
Carfagna MA, Ahmed CS, Butler S, Fukushima T, Houser W, Jensen N, Paisley B, Leuenroth-Quinn S, Snyder K, Vispute S, Wang W, Ali MY. Cross study analyses of SEND data: toxicity profile classification. Toxicol Sci 2024; 200:277-286. [PMID: 38851876 DOI: 10.1093/toxsci/kfae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2024] Open
Abstract
A SEND toxicology data transformation, harmonization, and analysis platform were created to improve the identification of unique findings related to the intended target, species, and duration of dosing using data from multiple studies. The lack of a standardized digital format for data analysis had impeded large-scale analysis of in vivo toxicology studies. The CDISC SEND standard enables the analysis of data from multiple studies performed by different laboratories. This work describes methods to analyze data and automate cross-study analysis of toxicology studies. Cross-study analysis can be used to understand a single compound's toxicity profile across all studies performed and/or to evaluate on-target versus off-target toxicity for multiple compounds intended for the same pharmacological target. This work involved development of data harmonization/transformation strategies to enable cross-study analysis of both numerical and categorical SEND data. Four de-identified SEND datasets from the BioCelerate database were used for the analyses. Toxicity profiles for key organ systems were developed for liver, kidney, male reproductive tract, endocrine system, and hematopoietic system using SEND domains. A cross-study analysis dashboard with a built-in user-defined scoring system was created for custom analyses, including visualizations to evaluate data at the organ system level and drill down into individual animal data. This data analysis provides the tools for scientists to compare toxicity profiles across multiple studies using SEND. A cross-study analysis of 2 different compounds intended for the same pharmacological target is described and the analyses indicate potential on-target effects to liver, kidney, and hematopoietic systems.
Collapse
Affiliation(s)
| | - Cm Sabbir Ahmed
- US Food & Drug Administration, Silver Spring, MD 20901, United States
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, United States
| | - Susan Butler
- US Food & Drug Administration, Silver Spring, MD 20901, United States
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, United States
| | | | - William Houser
- Bristol Myers Squibb, New Brunswick, NJ 08901, United States
| | | | | | | | - Kevin Snyder
- US Food & Drug Administration, Silver Spring, MD 20901, United States
| | | | - Wenxian Wang
- Bristol Myers Squibb, New Brunswick, NJ 08901, United States
| | - Md Yousuf Ali
- US Food & Drug Administration, Silver Spring, MD 20901, United States
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, United States
| |
Collapse
|
2
|
Palazzi X, Anger LT, Boulineau T, Grevot A, Guffroy M, Henson K, Hoepp N, Jacobsen M, Kale VP, Kreeger J, Lane JH, Li D, Muster W, Paisley B, Ramaiah L, Robertson N, Shultz V, Steger Hartmann T, Westhouse R. Points to consider regarding the use and implementation of virtual controls in nonclinical general toxicology studies. Regul Toxicol Pharmacol 2024; 150:105632. [PMID: 38679316 DOI: 10.1016/j.yrtph.2024.105632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
The replacement of a proportion of concurrent controls by virtual controls in nonclinical safety studies has gained traction over the last few years. This is supported by foundational work, encouraged by regulators, and aligned with societal expectations regarding the use of animals in research. This paper provides an overview of the points to consider for any institution on the verge of implementing this concept, with emphasis given on database creation, risks, and discipline-specific perspectives.
Collapse
Affiliation(s)
- Xavier Palazzi
- Drug Safety Research and Development, Pfizer Inc, 445, Eastern Point Road, Groton CT, USA.
| | - Lennart T Anger
- Safety Assessment, Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Theresa Boulineau
- Nonclinical Drug Safety, Boehringer Ingelheim, 900 Ridgebury Road, Ridgefield, CT, 06877, USA
| | - Armelle Grevot
- Preclinical Safety, Novartis AG, Fabrikstrasse, Basel, Switzerland
| | - Magali Guffroy
- Preclinical Safety, AbbVie, 1 North Waukegan Road, R46G/AP13A-3, North Chicago, IL, 60064, USA
| | - Kristin Henson
- Preclinical Safety, Novartis Pharmaceuticals Corporation, One Health Plaza, East Hanover, NJ, 07936, USA
| | - Natalie Hoepp
- Nonclinical Drug Safety, Merck and Co., Inc., Rahway, NJ, USA
| | - Matt Jacobsen
- Clinical Pharmacology and Safety Sciences, AstraZeneca, Biomedical Campus, 1 Francis Crick Ave, Cambridge, UK
| | - Vijay P Kale
- Nonclinical Safety, Bristol Myers Squibb, 1 Squibb Dr, New Brunswick, NJ, 08901, USA
| | - John Kreeger
- Non-Clinical Safety, GSK, 1250 S. Collegeville Rd, Collegeville, PA, USA
| | - Joan H Lane
- Translational Safety & Bioanalytical Sciences, Amgen, Inc., 1 Amgen Center Dr, Thousand Oaks, CA, 91320, USA
| | - Dingzhou Li
- Global Biometrics & Data Management, Pfizer Inc, 445, Eastern Point Road, Groton CT, USA
| | - Wolfgang Muster
- Pharmaceutical Sciences, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070, Basel, Switzerland
| | - Brianna Paisley
- iBAR ADMET, Eli Lilly and Company, 893 Delaware St, Indianapolis, IN, USA
| | - Lila Ramaiah
- Preclinical Sciences and Translational Safety, Johnson & Johnson, 1400 McKean Road, PO Box 776, Spring House, PA, 19477, USA
| | - Nicola Robertson
- Non-Clinical Safety, GSK, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Valerie Shultz
- Nonclinical Development, Organon, 4000 Chemical Rd, Suite 500, Plymouth Meeting, PA, 19462, USA
| | - Thomas Steger Hartmann
- Investigational Toxicology, BAYER AG, Pharmaceuticals, Muellerstrasse 178, 13342, Berlin, Germany
| | - Richard Westhouse
- Toxicology and Pathology, Agios Pharmaceuticals, 88 Sidney Street, Cambridge, MA, USA
| |
Collapse
|
3
|
Yue QX, Ding RF, Chen WH, Wu LY, Liu K, Ji ZL. Mining Real-World Big Data to Characterize Adverse Drug Reaction Quantitatively: Mixed Methods Study. J Med Internet Res 2024; 26:e48572. [PMID: 38700923 PMCID: PMC11102038 DOI: 10.2196/48572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 03/18/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Adverse drug reactions (ADRs), which are the phenotypic manifestations of clinical drug toxicity in humans, are a major concern in precision clinical medicine. A comprehensive evaluation of ADRs is helpful for unbiased supervision of marketed drugs and for discovering new drugs with high success rates. OBJECTIVE In current practice, drug safety evaluation is often oversimplified to the occurrence or nonoccurrence of ADRs. Given the limitations of current qualitative methods, there is an urgent need for a quantitative evaluation model to improve pharmacovigilance and the accurate assessment of drug safety. METHODS In this study, we developed a mathematical model, namely the Adverse Drug Reaction Classification System (ADReCS) severity-grading model, for the quantitative characterization of ADR severity, a crucial feature for evaluating the impact of ADRs on human health. The model was constructed by mining millions of real-world historical adverse drug event reports. A new parameter called Severity_score was introduced to measure the severity of ADRs, and upper and lower score boundaries were determined for 5 severity grades. RESULTS The ADReCS severity-grading model exhibited excellent consistency (99.22%) with the expert-grading system, the Common Terminology Criteria for Adverse Events. Hence, we graded the severity of 6277 standard ADRs for 129,407 drug-ADR pairs. Moreover, we calculated the occurrence rates of 6272 distinct ADRs for 127,763 drug-ADR pairs in large patient populations by mining real-world medication prescriptions. With the quantitative features, we demonstrated example applications in systematically elucidating ADR mechanisms and thereby discovered a list of drugs with improper dosages. CONCLUSIONS In summary, this study represents the first comprehensive determination of both ADR severity grades and ADR frequencies. This endeavor establishes a strong foundation for future artificial intelligence applications in discovering new drugs with high efficacy and low toxicity. It also heralds a paradigm shift in clinical toxicity research, moving from qualitative description to quantitative evaluation.
Collapse
Affiliation(s)
- Qi-Xuan Yue
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Ruo-Fan Ding
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Wei-Hao Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Lv-Ying Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Ke Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Zhi-Liang Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Chemical Biology, Xiamen University, Xiamen, China
| |
Collapse
|
4
|
Anzai T, Myatt GJ, Hall F, Finney B, Nakagawa K, Iwata H, Anzai R, Dickinson A, Freer M, Nakae D, Onodera H, Matsuyama T. Drug review process advancement and required manufacturer and contract research oraganization responses. J Toxicol Pathol 2024; 37:45-53. [PMID: 38584971 PMCID: PMC10995435 DOI: 10.1293/tox.2023-0106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/09/2023] [Indexed: 04/09/2024] Open
Abstract
The United States Senate passed the "FDA Modernization Act 2.0." on September 29, 2022. Although the effectiveness of this Bill, which aims to eliminate the mandatory use of laboratory animals in new drug development, is limited, it represents a significant trend that will change the shape of drug applications in the United States and other countries. However, pharmaceutical companies have not taken major steps towards the complete elimination of animal testing from the standpoint of product safety, where they prioritize patient safety. Nonetheless, society is becoming increasingly opposed to animal testing, and efforts will be made to use fewer animals and conduct fewer animal tests as a natural and reasonable response. These changes eventually alter the shape of new drug applications. Based on the assumption that fewer animal tests will be conducted or fewer animals will be used in testing, this study explored bioinformatics and new technologies as alternatives to compensate for reduced information and provide a picture of how future new drug applications may look. The authors also discuss the directions that pharmaceutical companies and nonclinical contract research organizations should adopt to promote the replacement, reduction, and refinement of animals used in research, teaching, testing, and exhibitions.
Collapse
Affiliation(s)
- Takayuki Anzai
- Showa University School of Medicine, 1-5-8 Hatanodai,
Shinagawa, Tokyo 142-0064, Japan
| | - Glenn J. Myatt
- Instem, Diamond Way, Stone Business Park, Stone,
Staffordshire, ST150SD, United Kingdom
| | - Frances Hall
- Instem, Diamond Way, Stone Business Park, Stone,
Staffordshire, ST150SD, United Kingdom
| | - Brenda Finney
- Instem, Diamond Way, Stone Business Park, Stone,
Staffordshire, ST150SD, United Kingdom
| | - Kenshi Nakagawa
- Showa University School of Medicine, 1-5-8 Hatanodai,
Shinagawa, Tokyo 142-0064, Japan
| | - Hijiri Iwata
- LunaPath Laboratory of Toxicologic Pathology, 3-5-1
Aoihigashi, Naka-ku Hamamatsu-shi, Shizuoka 433-8114, Japan
| | - Reo Anzai
- Tokyo University of the Arts, 5000 Omonma, Toride-shi,
Ibaraki 302-0001, Japan
| | - Anne Dickinson
- Newcastle University, Newcastle upon Tyne, NE1 7RU, United
Kingdom
- Alcyomics, The Biosphere, Draymans Way, Newcastle upon Tyne
NE4 5BX, United Kingdom
| | - Matthew Freer
- Alcyomics, The Biosphere, Draymans Way, Newcastle upon Tyne
NE4 5BX, United Kingdom
| | - Dai Nakae
- Teikyo Heisei University, 4-1 Uruido-Minami, Ichihara-shi,
Chiba 290-0193, Japan
| | - Hiroshi Onodera
- National Institute of Health Sciences, 3-25-26 Tono-machi,
Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Takaaki Matsuyama
- Showa University School of Medicine, 1-5-8 Hatanodai,
Shinagawa, Tokyo 142-0064, Japan
| |
Collapse
|
5
|
Martens M, Stierum R, Schymanski EL, Evelo CT, Aalizadeh R, Aladjov H, Arturi K, Audouze K, Babica P, Berka K, Bessems J, Blaha L, Bolton EE, Cases M, Damalas DΕ, Dave K, Dilger M, Exner T, Geerke DP, Grafström R, Gray A, Hancock JM, Hollert H, Jeliazkova N, Jennen D, Jourdan F, Kahlem P, Klanova J, Kleinjans J, Kondic T, Kone B, Lynch I, Maran U, Martinez Cuesta S, Ménager H, Neumann S, Nymark P, Oberacher H, Ramirez N, Remy S, Rocca-Serra P, Salek RM, Sallach B, Sansone SA, Sanz F, Sarimveis H, Sarntivijai S, Schulze T, Slobodnik J, Spjuth O, Tedds J, Thomaidis N, Weber RJ, van Westen GJ, Wheelock CE, Williams AJ, Witters H, Zdrazil B, Županič A, Willighagen EL. ELIXIR and Toxicology: a community in development. F1000Res 2023; 10:ELIXIR-1129. [PMID: 37842337 PMCID: PMC10568213 DOI: 10.12688/f1000research.74502.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2023] [Indexed: 10/17/2023] Open
Abstract
Toxicology has been an active research field for many decades, with academic, industrial and government involvement. Modern omics and computational approaches are changing the field, from merely disease-specific observational models into target-specific predictive models. Traditionally, toxicology has strong links with other fields such as biology, chemistry, pharmacology and medicine. With the rise of synthetic and new engineered materials, alongside ongoing prioritisation needs in chemical risk assessment for existing chemicals, early predictive evaluations are becoming of utmost importance to both scientific and regulatory purposes. ELIXIR is an intergovernmental organisation that brings together life science resources from across Europe. To coordinate the linkage of various life science efforts around modern predictive toxicology, the establishment of a new ELIXIR Community is seen as instrumental. In the past few years, joint efforts, building on incidental overlap, have been piloted in the context of ELIXIR. For example, the EU-ToxRisk, diXa, HeCaToS, transQST, and the nanotoxicology community have worked with the ELIXIR TeSS, Bioschemas, and Compute Platforms and activities. In 2018, a core group of interested parties wrote a proposal, outlining a sketch of what this new ELIXIR Toxicology Community would look like. A recent workshop (held September 30th to October 1st, 2020) extended this into an ELIXIR Toxicology roadmap and a shortlist of limited investment-high gain collaborations to give body to this new community. This Whitepaper outlines the results of these efforts and defines our vision of the ELIXIR Toxicology Community and how it complements other ELIXIR activities.
Collapse
Affiliation(s)
- Marvin Martens
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, 6229 ER, The Netherlands
| | - Rob Stierum
- Risk Analysis for Products In Development (RAPID), Netherlands Organisation for applied scientific research TNO, Utrecht, 3584 CB, The Netherlands
| | - Emma L. Schymanski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, 4367, Luxembourg
| | - Chris T. Evelo
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, 6229 ER, The Netherlands
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, Maastricht, 6229 EN, The Netherlands
| | - Reza Aalizadeh
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, 15771, Greece
| | - Hristo Aladjov
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, 1113, Bulgaria
| | - Kasia Arturi
- Department Environmental Chemistry, Swiss Federal Institute of Aquatic Science and Technology, Dübendorf, 8600, Switzerland
| | | | - Pavel Babica
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Karel Berka
- Department of Physical Chemistry, Palacky University Olomouc, Olomouc, 77146, Czech Republic
| | | | - Ludek Blaha
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Evan E. Bolton
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | | | - Dimitrios Ε. Damalas
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, 15771, Greece
| | - Kirtan Dave
- School of Science, GSFC University, Gujarat, 391750, India
| | - Marco Dilger
- Forschungs- und Beratungsinstitut Gefahrstoffe (FoBiG) GmbH, Freiburg im Breisgau, 79106, Germany
| | | | - Daan P. Geerke
- AIMMS Division of Molecular Toxicology, Vrije Universiteit, Amsterdam, 1081 HZ, The Netherlands
| | - Roland Grafström
- Department of Toxicology, Misvik Biology, Turku, 20520, Finland
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, 17177, Sweden
| | - Alasdair Gray
- Department of Computer Science, Heriot-Watt University, Edinburgh, UK
| | | | - Henner Hollert
- Department Evolutionary Ecology & Environmental Toxicology (E3T), Goethe-University, Frankfurt, D-60438, Germany
| | | | - Danyel Jennen
- Department of Toxicogenomics, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Fabien Jourdan
- MetaboHUB, French metabolomics infrastructure in Metabolomics and Fluxomics, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, Toulouse, France
| | - Pascal Kahlem
- Scientific Network Management SL, Barcelona, 08015, Spain
| | - Jana Klanova
- RECETOX, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jos Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, 6200 MD, The Netherlands
| | - Todor Kondic
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, 4367, Luxembourg
| | - Boï Kone
- Faculty of Pharmacy, Malaria Research and Training Center, Bamako, BP:1805, Mali
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, UK, Birmingham, B15 2TT, UK
| | - Uko Maran
- Institute of Chemistry, University of Tartu, Tartu, 50411, Estonia
| | | | - Hervé Ménager
- Institut Français de Bioinformatique, Evry, F-91000, France
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Paris, F-75015, France
| | - Steffen Neumann
- Research group Bioinformatics and Scientific Data, Leibniz Institute of Plant Biochemistry, Halle, 06120, Germany
| | - Penny Nymark
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, 17177, Sweden
| | - Herbert Oberacher
- Institute of Legal Medicine and Core Facility Metabolomics, Medical University of Innsbruck, Innsbruck, A-6020, Austria
| | - Noelia Ramirez
- Institut d'Investigacio Sanitaria Pere Virgili-Universitat Rovira i Virgili, Tarragona, 43007, Spain
| | | | - Philippe Rocca-Serra
- Data Readiness Group, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Reza M. Salek
- International Agency for Research on Cancer, World Health Organisation, Lyon, 69372, France
| | - Brett Sallach
- Department of Environment and Geography, University of York, UK, York, YO10 5NG, UK
| | | | - Ferran Sanz
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, 08003, Spain
| | | | | | - Tobias Schulze
- Helmholtz Centre for Environmental Research - UFZ, Leipzig, 04318, Germany
| | | | - Ola Spjuth
- Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Uppsala, SE-75124, Sweden
| | - Jonathan Tedds
- ELIXIR Hub, Wellcome Genome Campus, Cambridge, CB10 1SD, UK
| | - Nikolaos Thomaidis
- Laboratory of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Athens, 15771, Greece
| | - Ralf J.M. Weber
- School of Biosciences, University of Birmingham, UK, Birmingham, B15 2TT, UK
| | - Gerard J.P. van Westen
- Division of Drug Discovery and Safety, Leiden Academic Center for Drug Research, Leiden, 2333 CC, The Netherlands
| | - Craig E. Wheelock
- Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm SE-141-86, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177, Sweden
| | - Antony J. Williams
- Center for Computational Toxicology and Exposure, United States Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | | | - Barbara Zdrazil
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, 1090, Austria
| | - Anže Županič
- Department Biotechnology and Systems Biology, National Institute of Biology, Ljubljana, 1000, Slovenia
| | - Egon L. Willighagen
- Department of Bioinformatics - BiGCaT, Maastricht University, Maastricht, 6229 ER, The Netherlands
| |
Collapse
|
6
|
Naga D, Dimitrakopoulou S, Roberts S, Husar E, Mohr S, Booler H, Musvasva E. CSL-Tox: an open-source analytical framework for the comparison of short-term and long-term toxicity end points and assessing the need of chronic studies in drug development. Sci Rep 2023; 13:14865. [PMID: 37684321 PMCID: PMC10491674 DOI: 10.1038/s41598-023-41899-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
In-vivo toxicity assessment is an important step prior to clinical development and is still the main source of data for overall risk assessment of a new molecular entity (NCE). All in-vivo studies are performed according to regulatory requirements and many efforts have been exerted to minimize these studies in accordance with the (Replacement, Reduction and Refinement) 3Rs principle. Many aspects of in-vivo toxicology packages can be optimized to reduce animal use, including the number of studies performed as well as study durations, which is the main focus of this analysis. We performed a statistical comparison of adverse findings observed in 116 short-term versus 78 long-term in-house or in-house sponsored Contract Research Organizations (CRO) studies, in order to explore the possibility of using only short-term studies as a prediction tool for the longer-term effects. All the data analyzed in this study was manually extracted from the toxicology reports (in PDF formats) to construct the dataset. Annotation of treatment related findings was one of the challenges faced during this work. A specific focus was therefore put on the summary and conclusion sections of the reports since they contain expert assessments on whether the findings were considered adverse or were attributed to other reasons. Our analysis showed a general good concordance between short-term and long-term toxicity findings for large molecules and the majority of small molecules. Less concordance was seen for certain body organs, which can be named as "target organ systems' findings". While this work supports the minimization of long-term studies, a larger-scale effort would be needed to provide more evidence. We therefore present the steps performed in this study as an open-source R workflow for the Comparison of Short-term and Long-term Toxicity studies (CSL-Tox). The dataset used in the work is provided to allow researchers to reproduce such analysis, re-evaluate the statistical tools used and promote large-scale application of this study. Important aspects of animal research reproducibility are highlighted in this work, specifically, the necessity of a reproducible adverse effects reporting system and utilization of the controlled terminologies in-vivo toxicology reports and finally the importance of open-source analytical workflows that can be assessed by other scientists in the field of preclinical toxicology.
Collapse
Affiliation(s)
- Doha Naga
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
- Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria.
| | - Smaragda Dimitrakopoulou
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Sonia Roberts
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Elisabeth Husar
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Susanne Mohr
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Helen Booler
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Eunice Musvasva
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland.
| |
Collapse
|
7
|
Smajić A, Rami I, Sosnin S, Ecker GF. Identifying Differences in the Performance of Machine Learning Models for Off-Targets Trained on Publicly Available and Proprietary Data Sets. Chem Res Toxicol 2023; 36:1300-1312. [PMID: 37439496 PMCID: PMC10445286 DOI: 10.1021/acs.chemrestox.3c00042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Indexed: 07/14/2023]
Abstract
Each year, publicly available databases are updated with new compounds from different research institutions. Positive experimental outcomes are more likely to be reported; therefore, they account for a considerable fraction of these entries. Established publicly available databases such as ChEMBL allow researchers to use information without constrictions and create predictive tools for a broad spectrum of applications in the field of toxicology. Therefore, we investigated the distribution of positive and nonpositive entries within ChEMBL for a set of off-targets and its impact on the performance of classification models when applied to pharmaceutical industry data sets. Results indicate that models trained on publicly available data tend to overpredict positives, and models based on industry data sets predict negatives more often than those built using publicly available data sets. This is strengthened even further by the visualization of the prediction space for a set of 10,000 compounds, which makes it possible to identify regions in the chemical space where predictions converge. Finally, we highlight the utilization of these models for consensus modeling for potential adverse events prediction.
Collapse
Affiliation(s)
- Aljoša Smajić
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Iris Rami
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Sergey Sosnin
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Gerhard F. Ecker
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| |
Collapse
|
8
|
Sanz F, Pognan F, Steger-Hartmann T, Díaz C, Asakura S, Amberg A, Bécourt-Lhote N, Blomberg N, Bosc N, Briggs K, Bringezu F, Brulle-Wohlhueter C, Brunak S, Bueters R, Callegaro G, Capella-Gutierrez S, Centeno E, Corvi J, Cronin MTD, Drew P, Duchateau-Nguyen G, Ecker GF, Escher S, Felix E, Ferreiro M, Frericks M, Furlong LI, Geiger R, George C, Grandits M, Ivanov-Draganov D, Kilgour-Christie J, Kiziloren T, Kors JA, Koyama N, Kreuchwig A, Leach AR, Mayer MA, Monecke P, Muster W, Nakazawa CM, Nicholson G, Parry R, Pastor M, Piñero J, Oberhauser N, Ramírez-Anguita JM, Rodrigo A, Smajic A, Schaefer M, Schieferdecker S, Soininen I, Terricabras E, Trairatphisan P, Turner SC, Valencia A, van de Water B, van der Lei JL, van Mulligen EM, Vock E, Wilkinson D. eTRANSAFE: data science to empower translational safety assessment. Nat Rev Drug Discov 2023; 22:605-606. [PMID: 37316648 DOI: 10.1038/d41573-023-00099-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
9
|
Steger-Hartmann T, Kreuchwig A, Wang K, Birzele F, Draganov D, Gaudio S, Rothfuss A. Perspectives of data science in preclinical safety assessment. Drug Discov Today 2023:103642. [PMID: 37244565 DOI: 10.1016/j.drudis.2023.103642] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 05/29/2023]
Abstract
The data landscape in preclinical safety assessment is fundamentally changing because of not only emerging new data types, such as human systems biology, or real-world data (RWD) from clinical trials, but also technological advancements in data-processing software and analytical tools based on deep learning approaches. The recent developments of data science are illustrated with use cases for the three factors: predictive safety (new in silico tools), insight generation (new data for outstanding questions); and reverse translation (extrapolating from clinical experience to resolve preclinical questions). Further advances in this field can be expected if companies focus on overcoming identified challenges related to a lack of platforms and data silos and assuring appropriate training of data scientists within the preclinical safety teams.
Collapse
Affiliation(s)
| | - Annika Kreuchwig
- Investigational Toxicology, Bayer AG, Pharmaceuticals, 13353 Berlin, Germany
| | - Ken Wang
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences F. Hoffmann-La-Roche AG, Basel, Switzerland
| | - Fabian Birzele
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences F. Hoffmann-La-Roche AG, Basel, Switzerland
| | - Dragomir Draganov
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences F. Hoffmann-La-Roche AG, Basel, Switzerland
| | - Stefano Gaudio
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences F. Hoffmann-La-Roche AG, Basel, Switzerland
| | - Andreas Rothfuss
- Roche Pharmaceutical Research and Early Development, Pharmaceutical Sciences F. Hoffmann-La-Roche AG, Basel, Switzerland
| |
Collapse
|
10
|
Cronin MTD, Belfield SJ, Briggs KA, Enoch SJ, Firman JW, Frericks M, Garrard C, Maccallum PH, Madden JC, Pastor M, Sanz F, Soininen I, Sousoni D. Making in silico predictive models for toxicology FAIR. Regul Toxicol Pharmacol 2023; 140:105385. [PMID: 37037390 DOI: 10.1016/j.yrtph.2023.105385] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/18/2023] [Accepted: 04/07/2023] [Indexed: 04/12/2023]
Abstract
In silico predictive models for toxicology include quantitative structure-activity relationship (QSAR) and physiologically based kinetic (PBK) approaches to predict physico-chemical and ADME properties, toxicological effects and internal exposure. Such models are used to fill data gaps as part of chemical risk assessment. There is a growing need to ensure in silico predictive models for toxicology are available for use and reproducible. This paper describes how the FAIR (Findable, Accessible, Interoperable, Reusable) principles, developed for data sharing, have been applied to in silico predictive models. In particular, this investigation has focussed on how the FAIR principles could be applied to improved regulatory acceptance of predictions from such models. Eighteen principles have been developed that cover all aspects of FAIR. It is intended that FAIRification of in silico predictive models for toxicology will increase their use and acceptance.
Collapse
Affiliation(s)
- Mark T D Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
| | - Samuel J Belfield
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Katharine A Briggs
- Lhasa Limited, Granary Wharf House, 2 Canal Wharf, Holbeck, Leeds, LS11 5PS, UK
| | - Steven J Enoch
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - James W Firman
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Markus Frericks
- BASF SE, APD/ET - Li 444, Speyerer St 2, 67117, Limburgerhof, Germany
| | - Clare Garrard
- ELIXIR, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK
| | - Peter H Maccallum
- ELIXIR, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK
| | - Judith C Madden
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Manuel Pastor
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Dept. of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, Carrer Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Ferran Sanz
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Dept. of Medicine and Life Sciences (MELIS), Universitat Pompeu Fabra, Carrer Dr. Aiguader 88, 08003, Barcelona, Spain
| | - Inari Soininen
- Synapse Research Management Partners SL, Calle Velazquez 94, planta 1, 28006, Madrid, Spain
| | - Despoina Sousoni
- ELIXIR, Wellcome Genome Campus, Hinxton, Cambridgeshire, CB10 1SD, UK
| |
Collapse
|
11
|
Pognan F, Beilmann M, Boonen HCM, Czich A, Dear G, Hewitt P, Mow T, Oinonen T, Roth A, Steger-Hartmann T, Valentin JP, Van Goethem F, Weaver RJ, Newham P. The evolving role of investigative toxicology in the pharmaceutical industry. Nat Rev Drug Discov 2023; 22:317-335. [PMID: 36781957 PMCID: PMC9924869 DOI: 10.1038/s41573-022-00633-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 02/15/2023]
Abstract
For decades, preclinical toxicology was essentially a descriptive discipline in which treatment-related effects were carefully reported and used as a basis to calculate safety margins for drug candidates. In recent years, however, technological advances have increasingly enabled researchers to gain insights into toxicity mechanisms, supporting greater understanding of species relevance and translatability to humans, prediction of safety events, mitigation of side effects and development of safety biomarkers. Consequently, investigative (or mechanistic) toxicology has been gaining momentum and is now a key capability in the pharmaceutical industry. Here, we provide an overview of the current status of the field using case studies and discuss the potential impact of ongoing technological developments, based on a survey of investigative toxicologists from 14 European-based medium-sized to large pharmaceutical companies.
Collapse
Affiliation(s)
- Francois Pognan
- Discovery and Investigative Safety, Novartis Pharma AG, Basel, Switzerland.
| | - Mario Beilmann
- Nonclinical Drug Safety Germany, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Harrie C M Boonen
- Drug Safety, Dept of Exploratory Toxicology, Lundbeck A/S, Valby, Denmark
| | | | - Gordon Dear
- In Vitro In Vivo Translation, GlaxoSmithKline David Jack Centre for Research, Ware, UK
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Tomas Mow
- Safety Pharmacology and Early Toxicology, Novo Nordisk A/S, Maaloev, Denmark
| | - Teija Oinonen
- Preclinical Safety, Orion Corporation, Espoo, Finland
| | - Adrian Roth
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | | | | | - Freddy Van Goethem
- Predictive, Investigative & Translational Toxicology, Nonclinical Safety, Janssen Research & Development, Beerse, Belgium
| | - Richard J Weaver
- Innovation Life Cycle Management, Institut de Recherches Internationales Servier, Suresnes, France
| | - Peter Newham
- Clinical Pharmacology and Safety Sciences, AstraZeneca R&D, Cambridge, UK.
| |
Collapse
|
12
|
Wright PSR, Smith GF, Briggs KA, Thomas R, Maglennon G, Mikulskis P, Chapman M, Greene N, Phillips BU, Bender A. Retrospective analysis of the potential use of virtual control groups in preclinical toxicity assessment using the eTOX database. Regul Toxicol Pharmacol 2023; 138:105309. [PMID: 36481280 DOI: 10.1016/j.yrtph.2022.105309] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/17/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022]
Abstract
Virtual Control Groups (VCGs) based on Historical Control Data (HCD) in preclinical toxicity testing have the potential to reduce animal usage. As a case study we retrospectively analyzed the impact of replacing Concurrent Control Groups (CCGs) with VCGs on the treatment-relatedness of 28 selected histopathological findings reported in either rat or dog in the eTOX database. We developed a novel methodology whereby statistical predictions of treatment-relatedness using either CCGs or VCGs of varying covariate similarity to CCGs were compared to designations from original toxicologist reports; and changes in agreement were used to quantify changes in study outcomes. Generally, the best agreement was achieved when CCGs were replaced with VCGs with the highest level of similarity; the same species, strain, sex, administration route, and vehicle. For example, balanced accuracies for rat findings were 0.704 (predictions based on CCGs) vs. 0.702 (predictions based on VCGs). Moreover, we identified covariates which resulted in poorer identification of treatment-relatedness. This was related to an increasing incidence rate divergence in HCD relative to CCGs. Future databases which collect data at the individual animal level including study details such as animal age and testing facility are required to build adequate VCGs to accurately identify treatment-related effects.
Collapse
Affiliation(s)
| | - Graham F Smith
- AstraZeneca, Data Science and AI, Clinical Pharmacology and Safety Sciences, R&D, Cambridge, United Kingdom
| | | | | | - Gareth Maglennon
- AstraZeneca, Oncology Pathology, Clinical Pharmacology and Safety Sciences, R&D, Melbourn, United Kingdom
| | - Paulius Mikulskis
- AstraZeneca, Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, Gothenburg, Sweden
| | - Melissa Chapman
- AstraZeneca, Toxicology, Clinical Pharmacology and Safety Sciences, R&D, Melbourn, United Kingdom
| | - Nigel Greene
- AstraZeneca, Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, Waltham, MA, USA
| | - Benjamin U Phillips
- AstraZeneca, Data Sciences and Quantitative Biology, Discovery Sciences, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Andreas Bender
- University of Cambridge, Chemistry, Cambridge, United Kingdom.
| |
Collapse
|
13
|
Gurjanov A, Kreuchwig A, Steger-Hartmann T, Vaas LAI. Hurdles and signposts on the road to virtual control groups-A case study illustrating the influence of anesthesia protocols on electrolyte levels in rats. Front Pharmacol 2023; 14:1142534. [PMID: 37153793 PMCID: PMC10159271 DOI: 10.3389/fphar.2023.1142534] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/31/2023] [Indexed: 05/10/2023] Open
Abstract
Introduction: Virtual Control Groups (VCGs) represent the concept of using historical control data from legacy animal studies to replace concurrent control group (CCG) animals. Based on the data curation and sharing activities of the Innovative Medicine Initiatives project eTRANSAFE (enhancing TRANSlational SAFEty Assessment through Integrative Knowledge Management) the ViCoG working group was established with the objectives of i) collecting suitable historical control data sets from preclinical toxicity studies, ii) evaluating statistical methodologies for building adequate and regulatory acceptable VCGs from historical control data, and iii) sharing those control-group data across multiple pharmaceutical companies. During the qualification process of VCGs a particular focus was put on the identification of hidden confounders in the data sets, which might impair the adequate matching of VCGs with the CCG. Methods: During our analyses we identified such a hidden confounder, namely, the choice of the anesthetic procedure used in animal experiments before blood withdrawal. Anesthesia using CO2 may elevate the levels of some electrolytes such as calcium in blood, while the use of isoflurane is known to lower these values. Identification of such hidden confounders is particularly important if the underlying experimental information (e.g., on the anesthetic procedure) is not routinely recorded in the standard raw data files, such as SEND (Standard for Exchange of Non-clinical Data). We therefore analyzed how the replacement of CCGs with VCGs would affect the reproducibility of treatment-related findings regarding electrolyte values (potassium, calcium, sodium, and phosphate). The analyses were performed using a legacy rat systemic toxicity study consisting of a control and three treatment groups conducted according to pertinent OECD guidelines. In the report of this study treatment-related hypercalcemia was reported. The rats in this study were anesthetized with isoflurane. Results: Replacing the CCGs with VCGs derived from studies comprising both anesthetics resulted in a shift of control electrolyte parameters. Instead of the originally reported hypercalcemia the use of VCG led to fallacious conclusions of no observed effect or hypocalcemia. Discussion: Our study highlights the importance of a rigorous statistical analysis including the detection and elimination of hidden confounders prior to the implementation of the VCG concept.
Collapse
Affiliation(s)
- A. Gurjanov
- Bayer AG, Pharmaceuticals, Investigational Toxicology, Berlin, Germany
- *Correspondence: A. Gurjanov,
| | - A. Kreuchwig
- Bayer AG, Pharmaceuticals, Investigational Toxicology, Berlin, Germany
| | | | - L. A. I. Vaas
- Bayer AG, Pharmaceuticals, Research and Pre-Clinical Statistics Group, Berlin, Germany
| |
Collapse
|
14
|
P21-04 “Please, sir, I want some more…” data: Guidelines for effective sharing of preclinical data. Toxicol Lett 2022. [DOI: 10.1016/j.toxlet.2022.07.684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
Madden JC, Thompson CV. Pharmacokinetic Tools and Applications. Methods Mol Biol 2022; 2425:57-83. [PMID: 35188628 DOI: 10.1007/978-1-0716-1960-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Drug toxicity, as well as therapeutic activity, is contingent upon the parent drug, or a derivative thereof, reaching the relevant site of action in the body, at sufficient concentration, over a given period of time. Thus, the potential to truly elicit an effect is governed by both the intrinsic activity/toxicity of the drug (or its transformation products) and its pharmacokinetic profile. As the pharmaceutical industry has become increasingly aware of the role of pharmacokinetics in determining drug activity and toxicity, the range of software, both freely available and commercial, to predict relevant properties has proliferated. Such tools can be considered on three different levels, applicable at different stages within the drug development process and providing increasing detail and relevance of information. Level (i) is the prediction of fundamental physicochemical properties that can be used to screen vast virtual libraries of potential candidates. Level (ii), predicting the individual absorption, distribution, metabolism, and excretion (ADME) characteristics of potential drugs, can also be applied to many compounds simultaneously. Level (iii), predicting the concentration-time profile of a drug in blood or specific tissues/organs for individuals or a population, is the most sophisticated level of prediction, applied to fewer candidates. In this chapter, in silico tools for predicting ADME-relevant properties, across these three levels, and the applications of this information, are described using exemplar, freely available resources. Further resources are signposted but not all are considered in detail as the purpose here is more to provide an introduction to the capabilities and practicalities of the tools, rather than to provide an exhaustive review of all the tools available.
Collapse
Affiliation(s)
- Judith C Madden
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK.
| | - Courtney V Thompson
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| |
Collapse
|
16
|
Brigo A, Naga D, Muster W. Increasing the Value of Data Within a Large Pharmaceutical Company Through In Silico Models. Methods Mol Biol 2022; 2425:637-674. [PMID: 35188649 DOI: 10.1007/978-1-0716-1960-5_24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The present contribution describes how in silico models and methods are applied at different stages of the drug discovery process in the pharmaceutical industry. A description of the most relevant computational methods and tools is given along with an evaluation of their performance in the assessment of potential genotoxic impurities and the prediction of off-target in vitro pharmacology. The challenges of predicting the outcome of highly complex in vivo studies are discussed followed by considerations on how novel ways to manage, store, exchange, and analyze data may advance knowledge and facilitate modeling efforts. In this context, the current status of broad data sharing initiatives, namely, eTOX and eTransafe, will be described along with related projects that could significantly reduce the use of animals in drug discovery in the future.
Collapse
Affiliation(s)
- Alessandro Brigo
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Basel, Switzerland.
| | - Doha Naga
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Basel, Switzerland
- Department of Pharmaceutical Chemistry, Group of Pharmacoinformatics, University of Vienna, Wien, Austria
| | - Wolfgang Muster
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Centre Basel, Basel, Switzerland
| |
Collapse
|
17
|
Pastor M, Sanz F, Bringezu F. Development of In Silico Methods for Toxicity Prediction in Collaboration Between Academia and the Pharmaceutical Industry. Methods Mol Biol 2022; 2425:119-131. [PMID: 35188630 DOI: 10.1007/978-1-0716-1960-5_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The pharmaceutical industry would benefit from the collaboration with academic groups in the development of predictive safety models using the newest computational technologies. However, this collaboration is sometimes hampered by the handling of confidential proprietary information and different working practices in both environments. In this manuscript, we propose a strategy for facilitating this collaboration, based on the use of modeling frameworks developed for facilitating the use of sensitive data, as well as the development, interchange, hosting, and use of predictive models in production. The strategy is illustrated with a real example in which we used Flame, an open-source modeling framework developed in our group, for the development of an in silico eye irritation model. The model was based on bibliographic data, refined during the company-academic group collaboration, and enriched with the incorporation of confidential data, yielding a useful model that was validated experimentally.
Collapse
Affiliation(s)
- Manuel Pastor
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
| | - Ferran Sanz
- Research Programme on Biomedical Informatics (GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Frank Bringezu
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|