1
|
Vahle JL, Dybowski J, Graziano M, Hisada S, Lebron J, Nolte T, Steigerwalt R, Tsubota K, Sistare FD. ICH S1 prospective evaluation study and weight of evidence assessments: commentary from industry representatives. FRONTIERS IN TOXICOLOGY 2024; 6:1377990. [PMID: 38845817 PMCID: PMC11153695 DOI: 10.3389/ftox.2024.1377990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/03/2024] [Indexed: 06/09/2024] Open
Abstract
Industry representatives on the ICH S1B(R1) Expert Working Group (EWG) worked closely with colleagues from the Drug Regulatory Authorities to develop an addendum to the ICH S1B guideline on carcinogenicity studies that allows for a weight-of-evidence (WoE) carcinogenicity assessment in some cases, rather than conducting a 2-year rat carcinogenicity study. A subgroup of the EWG composed of regulators have published in this issue a detailed analysis of the Prospective Evaluation Study (PES) conducted under the auspices of the ICH S1B(R1) EWG. Based on the experience gained through the Prospective Evaluation Study (PES) process, industry members of the EWG have prepared the following commentary to aid sponsors in assessing the standard WoE factors, considering how novel investigative approaches may be used to support a WoE assessment, and preparing appropriate documentation of the WoE assessment for presentation to regulatory authorities. The commentary also reviews some of the implementation challenges sponsors must consider in developing a carcinogenicity assessment strategy. Finally, case examples drawn from previously marketed products are provided as a supplement to this commentary to provide additional examples of how WoE criteria may be applied. The information and opinions expressed in this commentary are aimed at increasing the quality of WoE assessments to ensure the successful implementation of this approach.
Collapse
Affiliation(s)
- John L. Vahle
- Lilly Research Laboratories, Indianapolis, IN, United States
| | - Joe Dybowski
- Alnylam Pharmaceuticals, Cambridge, MA, United States
| | | | - Shigeru Hisada
- Formerly ASKA Pharmaceutical Co., Ltd., Fujisawa-shi, Kanagawa, Japan
| | - Jose Lebron
- Merck & Co., Inc., Rahway, NJ, United States
| | - Thomas Nolte
- Development NCE, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | | | | | | |
Collapse
|
2
|
Bourcier T, McGovern T, Cavaliero T, Ebere G, Nishikawa A, Nishimura J, Ogawa K, Pasanen M, Vespa A, Van der Laan JW. ICH S1 prospective evaluation study: weight of evidence approach to predict outcome and value of 2-year rat carcinogenicity studies. A report from the regulatory authorities subgroup. FRONTIERS IN TOXICOLOGY 2024; 6:1353783. [PMID: 38665214 PMCID: PMC11043531 DOI: 10.3389/ftox.2024.1353783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/29/2024] [Indexed: 04/28/2024] Open
Abstract
Introduction: The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) initiated a process in 2012 to revise the S1B Guideline "Testing for Carcinogenicity of Pharmaceuticals". Previous retrospective analysis indicated the importance of histopathological risk factors in chronic toxicity studies, evidence of endocrine perturbation, and positive genetic toxicology results as potentially predictive indicators of carcinogenic risk. In addition, a relationship between pharmacodynamic activity and carcinogenicity outcome in long-term rodent studies has been reported. It was postulated that these factors could be evaluated in a Weight-of-Evidence (WoE) approach to predict the outcome of a 2-year rat study. Methods: The ICH S1B(R1) Expert Working Group (EWG) conducted a Prospective Evaluation Study (PES) to determine the regulatory feasibility of this WoE approach. Drug Regulatory Authorities (DRAs) evaluated 49 Carcinogenicity Assessment Documents (CADs), which describe the WoE for submitted pharmaceutical compounds. Each compound was categorized into a carcinogenic risk category including a statement of the value of the 2-year rat study. The outcome of the completed 2-year rat studies was evaluated in relation to the prospective CAD to determine the accuracy of predictions. Results: Based on the results of the PES, the EWG concluded that the evaluation process for assessing human carcinogenic risk of pharmaceuticals described in ICH S1B could be expanded to include a WoE approach. Approximately 27% of 2-year rat studies could be avoided in cases where DRAs and sponsors unanimously agreed that such a study would not add value. Discussion: Key factors supporting a WoE assessment were identified: data that inform carcinogenic potential based on drug target biology and the primary pharmacologic mechanism of the parent compound and major human metabolites; results from secondary pharmacology screens for this compound and major human metabolites that inform carcinogenic risk; histopathology data from repeated-dose toxicity studies; evidence for hormonal perturbation; genotoxicity data; and evidence of immune modulation. The outcome of the PES indicates that a WoE approach can be used in place of conducting a 2-year rat study for some pharmaceuticals. These data were used by the ICH S1B(R1) EWG to write the R1 Addendum to the S1B Guideline published in August 2022.
Collapse
Affiliation(s)
- Todd Bourcier
- Food and Drug Administration, Silver Spring, MD, United States
| | | | - Tania Cavaliero
- Swissmedic, Swiss Agency for Therapeutic Products, Bern, Switzerland
| | - Geoffrey Ebere
- Health Canada, Pharmaceutical Drugs Directorate, Ottawa, ON, Canada
| | | | | | - Kumiko Ogawa
- National Institute of Health Sciences, Kanagawa, Japan
| | - Markku Pasanen
- Faculty of Health Sciences, School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Alisa Vespa
- Health Canada, Pharmaceutical Drugs Directorate, Ottawa, ON, Canada
| | | |
Collapse
|
3
|
Bassan A, Steigerwalt R, Keller D, Beilke L, Bradley PM, Bringezu F, Brock WJ, Burns-Naas LA, Chambers J, Cross K, Dorato M, Elespuru R, Fuhrer D, Hall F, Hartke J, Jahnke GD, Kluxen FM, McDuffie E, Schmidt F, Valentin JP, Woolley D, Zane D, Myatt GJ. Developing a pragmatic consensus procedure supporting the ICH S1B(R1) weight of evidence carcinogenicity assessment. FRONTIERS IN TOXICOLOGY 2024; 6:1370045. [PMID: 38646442 PMCID: PMC11027748 DOI: 10.3389/ftox.2024.1370045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 03/04/2024] [Indexed: 04/23/2024] Open
Abstract
The ICH S1B carcinogenicity global testing guideline has been recently revised with a novel addendum that describes a comprehensive integrated Weight of Evidence (WoE) approach to determine the need for a 2-year rat carcinogenicity study. In the present work, experts from different organizations have joined efforts to standardize as much as possible a procedural framework for the integration of evidence associated with the different ICH S1B(R1) WoE criteria. The framework uses a pragmatic consensus procedure for carcinogenicity hazard assessment to facilitate transparent, consistent, and documented decision-making and it discusses best-practices both for the organization of studies and presentation of data in a format suitable for regulatory review. First, it is acknowledged that the six WoE factors described in the addendum form an integrated network of evidence within a holistic assessment framework that is used synergistically to analyze and explain safety signals. Second, the proposed standardized procedure builds upon different considerations related to the primary sources of evidence, mechanistic analysis, alternative methodologies and novel investigative approaches, metabolites, and reliability of the data and other acquired information. Each of the six WoE factors is described highlighting how they can contribute evidence for the overall WoE assessment. A suggested reporting format to summarize the cross-integration of evidence from the different WoE factors is also presented. This work also notes that even if a 2-year rat study is ultimately required, creating a WoE assessment is valuable in understanding the specific factors and levels of human carcinogenic risk better than have been identified previously with the 2-year rat bioassay alone.
Collapse
Affiliation(s)
| | | | - Douglas Keller
- Independent Consultant, Kennett Square, PA, United States
| | - Lisa Beilke
- Toxicology Solutions, Inc., Marana, AZ, United States
| | | | - Frank Bringezu
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - William J. Brock
- Brock Scientific Consulting, LLC, Hilton Head, SC, United States
| | | | | | | | | | | | - Douglas Fuhrer
- BioXcel Therapeutics, Inc., New Haven, CT, United States
| | | | - Jim Hartke
- Gilead Sciences, Inc., Foster City, CA, United States
| | | | | | - Eric McDuffie
- Neurocrine Bioscience, Inc., San Diego, CA, United States
| | | | | | | | - Doris Zane
- Gilead Sciences, Inc., Foster City, CA, United States
| | | |
Collapse
|
4
|
Tang W, Zhang X, Hong H, Chen J, Zhao Q, Wu F. Computational Nanotoxicology Models for Environmental Risk Assessment of Engineered Nanomaterials. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:155. [PMID: 38251120 PMCID: PMC10819018 DOI: 10.3390/nano14020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/08/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Although engineered nanomaterials (ENMs) have tremendous potential to generate technological benefits in numerous sectors, uncertainty on the risks of ENMs for human health and the environment may impede the advancement of novel materials. Traditionally, the risks of ENMs can be evaluated by experimental methods such as environmental field monitoring and animal-based toxicity testing. However, it is time-consuming, expensive, and impractical to evaluate the risk of the increasingly large number of ENMs with the experimental methods. On the contrary, with the advancement of artificial intelligence and machine learning, in silico methods have recently received more attention in the risk assessment of ENMs. This review discusses the key progress of computational nanotoxicology models for assessing the risks of ENMs, including material flow analysis models, multimedia environmental models, physiologically based toxicokinetics models, quantitative nanostructure-activity relationships, and meta-analysis. Several challenges are identified and a perspective is provided regarding how the challenges can be addressed.
Collapse
Affiliation(s)
- Weihao Tang
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-Environmental Pollution Control and Management, Institute of Eco-Environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Xuejiao Zhang
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-Environmental Pollution Control and Management, Institute of Eco-Environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China
| | - Huixiao Hong
- National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd., Jefferson, AR 72079, USA
| | - Jingwen Chen
- Key Laboratory of Industrial Ecology and Environmental Engineering (Ministry of Education), Dalian Key Laboratory on Chemicals Risk Control and Pollution Prevention Technology, School of Environmental Science and Technology, Dalian University of Technology, Dalian 116024, China
| | - Qing Zhao
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-Environmental Pollution Control and Management, Institute of Eco-Environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China
| | - Fengchang Wu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| |
Collapse
|
5
|
Rahmani K, Wu YC, Buck NR, Lau A, Hanlon PR. Retrospective analysis of carcinogenicity assessments within FDA-notified GRAS determinations. Hum Exp Toxicol 2024; 43:9603271241254338. [PMID: 39052968 DOI: 10.1177/09603271241254338] [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: 07/27/2024]
Abstract
Frameworks have been developed to standardize the assessment of carcinogenic potential in the pharmaceutical and agrochemical industries, building upon decades of research. Carcinogenicity is also evaluated during the safety evaluation of food substances, using a comprehensive approach unique to each substance. To better understand these approaches, a retrospective assessment was conducted on the publicly available database of substances notified to the United States Food and Drug Administration (US FDA) as being Generally Recognized As Safe (GRAS). The data contained within these GRAS notifications (GRNs) were reviewed for the methods used to evaluate carcinogenic potential (genotoxicity studies, 2-year bioassays, other pre-clinical animal studies) to identify patterns that could provide an understanding of how this assessment has been conducted for different categories of food substances. While different approaches to the safety evaluation were required to adapt to the unique food substances, the data in all notifications supported the conclusion of safety. The evaluation of food substances for carcinogenic potential must consider all available data, including identifying the need for when more data must be generated to support an evaluation. Due to the complexity of substances used in food, ranging from defined chemical entities to minimally processed agricultural commodities to live microorganisms, the approach to conducting the safety evaluation of food substances must be able to adapt to the most relevant scientifically supported approach. This paper illustrates the data commonly used to support the safety of different types of food substances and proposes an approach familiar to other product sectors.
Collapse
|
6
|
Pandiri AR, Auerbach SS, Stevens JL, Blomme EAG. Toxicogenomics Approaches to Address Toxicity and Carcinogenicity in the Liver. Toxicol Pathol 2023; 51:470-481. [PMID: 38288963 PMCID: PMC11014763 DOI: 10.1177/01926233241227942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/13/2024]
Abstract
Toxicogenomic technologies query the genome, transcriptome, proteome, and the epigenome in a variety of toxicological conditions. Due to practical considerations related to the dynamic range of the assays, sensitivity, cost, and technological limitations, transcriptomic approaches are predominantly used in toxicogenomics. Toxicogenomics is being used to understand the mechanisms of toxicity and carcinogenicity, evaluate the translational relevance of toxicological responses from in vivo and in vitro models, and identify predictive biomarkers of disease and exposure. In this session, a brief overview of various transcriptomic technologies and practical considerations related to experimental design was provided. The advantages of gene network analyses to define mechanisms were also discussed. An assessment of the utility of toxicogenomic technologies in the environmental and pharmaceutical space showed that these technologies are being increasingly used to gain mechanistic insights and determining the translational relevance of adverse findings. Within the environmental toxicology area, there is a broader regulatory consideration of benchmark doses derived from toxicogenomics data. In contrast, these approaches are mainly used for internal decision-making in pharmaceutical development. Finally, the development and application of toxicogenomic signatures for prediction of apical endpoints of regulatory concern continues to be area of intense research.
Collapse
Affiliation(s)
- Arun R Pandiri
- National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | - Scott S Auerbach
- National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | | | | |
Collapse
|
7
|
Audebert M, Assmann AS, Azqueta A, Babica P, Benfenati E, Bortoli S, Bouwman P, Braeuning A, Burgdorf T, Coumoul X, Debizet K, Dusinska M, Ertych N, Fahrer J, Fetz V, Le Hégarat L, López de Cerain A, Heusinkveld HJ, Hogeveen K, Jacobs MN, Luijten M, Raitano G, Recoules C, Rundén-Pran E, Saleh M, Sovadinová I, Stampar M, Thibol L, Tomkiewicz C, Vettorazzi A, Van de Water B, El Yamani N, Zegura B, Oelgeschläger M. New approach methodologies to facilitate and improve the hazard assessment of non-genotoxic carcinogens-a PARC project. FRONTIERS IN TOXICOLOGY 2023; 5:1220998. [PMID: 37492623 PMCID: PMC10364052 DOI: 10.3389/ftox.2023.1220998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 06/19/2023] [Indexed: 07/27/2023] Open
Abstract
Carcinogenic chemicals, or their metabolites, can be classified as genotoxic or non-genotoxic carcinogens (NGTxCs). Genotoxic compounds induce DNA damage, which can be detected by an established in vitro and in vivo battery of genotoxicity assays. For NGTxCs, DNA is not the primary target, and the possible modes of action (MoA) of NGTxCs are much more diverse than those of genotoxic compounds, and there is no specific in vitro assay for detecting NGTxCs. Therefore, the evaluation of the carcinogenic potential is still dependent on long-term studies in rodents. This 2-year bioassay, mainly applied for testing agrochemicals and pharmaceuticals, is time-consuming, costly and requires very high numbers of animals. More importantly, its relevance for human risk assessment is questionable due to the limited predictivity for human cancer risk, especially with regard to NGTxCs. Thus, there is an urgent need for a transition to new approach methodologies (NAMs), integrating human-relevant in vitro assays and in silico tools that better exploit the current knowledge of the multiple processes involved in carcinogenesis into a modern safety assessment toolbox. Here, we describe an integrative project that aims to use a variety of novel approaches to detect the carcinogenic potential of NGTxCs based on different mechanisms and pathways involved in carcinogenesis. The aim of this project is to contribute suitable assays for the safety assessment toolbox for an efficient and improved, internationally recognized hazard assessment of NGTxCs, and ultimately to contribute to reliable mechanism-based next-generation risk assessment for chemical carcinogens.
Collapse
Affiliation(s)
- Marc Audebert
- INRAE: Toxalim, INRAE, INP-ENVT, INP-EI-Purpan, Université de Toulouse 3 Paul Sabatier, Toulouse, France
| | - Ann-Sophie Assmann
- Department Experimental Toxicology and ZEBET, German Centre for the Protection of Laboratory Animals (Bf3R) and Department Food Safety, BfR: German Federal Institute for Risk Assessment, Berlin, Germany
| | - Amaya Azqueta
- Department of Pharmacology and Toxicology, School of Pharmacy and Nutrition, UNAV: University of Navarra, Pamplona, Spain
| | - Pavel Babica
- RECETOX: RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Emilio Benfenati
- IRFMN: Istituto di Ricerche Farmacologiche Mario Negri—IRCCS, Milan, Italy
| | - Sylvie Bortoli
- INSERM: INSERM UMR-S 1124 T3S—Université Paris Cité, Paris, France
| | - Peter Bouwman
- UL-LACDR: Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Albert Braeuning
- Department Experimental Toxicology and ZEBET, German Centre for the Protection of Laboratory Animals (Bf3R) and Department Food Safety, BfR: German Federal Institute for Risk Assessment, Berlin, Germany
| | - Tanja Burgdorf
- Department Experimental Toxicology and ZEBET, German Centre for the Protection of Laboratory Animals (Bf3R) and Department Food Safety, BfR: German Federal Institute for Risk Assessment, Berlin, Germany
| | - Xavier Coumoul
- INSERM: INSERM UMR-S 1124 T3S—Université Paris Cité, Paris, France
| | - Kloé Debizet
- INSERM: INSERM UMR-S 1124 T3S—Université Paris Cité, Paris, France
| | - Maria Dusinska
- Health Effects Laboratory, NILU: The Climate and Environmental Research Institute, Kjeller, Norway
| | - Norman Ertych
- Department Experimental Toxicology and ZEBET, German Centre for the Protection of Laboratory Animals (Bf3R) and Department Food Safety, BfR: German Federal Institute for Risk Assessment, Berlin, Germany
| | - Jörg Fahrer
- Department of Chemistry, RPTU: Division of Food Chemistry and Toxicology, Kaiserslautern, Germany
| | - Verena Fetz
- Department Experimental Toxicology and ZEBET, German Centre for the Protection of Laboratory Animals (Bf3R) and Department Food Safety, BfR: German Federal Institute for Risk Assessment, Berlin, Germany
| | - Ludovic Le Hégarat
- ANSES: French Agency for Food, Environmental and Occupational Health and Safety, Fougères Laboratory, Toxicology of Contaminants Unit, Fougères, France
| | - Adela López de Cerain
- Department of Pharmacology and Toxicology, School of Pharmacy and Nutrition, UNAV: University of Navarra, Pamplona, Spain
| | - Harm J. Heusinkveld
- RIVM: National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Kevin Hogeveen
- ANSES: French Agency for Food, Environmental and Occupational Health and Safety, Fougères Laboratory, Toxicology of Contaminants Unit, Fougères, France
| | - Miriam N. Jacobs
- Radiation, Chemical and Environmental Hazards, UKHSA: UK Health Security Agency, Chilton, Oxfordshire, United Kingdom
| | - Mirjam Luijten
- RIVM: National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Giuseppa Raitano
- IRFMN: Istituto di Ricerche Farmacologiche Mario Negri—IRCCS, Milan, Italy
| | - Cynthia Recoules
- INRAE: Toxalim, INRAE, INP-ENVT, INP-EI-Purpan, Université de Toulouse 3 Paul Sabatier, Toulouse, France
| | - Elise Rundén-Pran
- Health Effects Laboratory, NILU: The Climate and Environmental Research Institute, Kjeller, Norway
| | - Mariam Saleh
- ANSES: French Agency for Food, Environmental and Occupational Health and Safety, Fougères Laboratory, Toxicology of Contaminants Unit, Fougères, France
| | - Iva Sovadinová
- RECETOX: RECETOX, Faculty of Science, Masaryk University, Brno, Czechia
| | - Martina Stampar
- Department of Genetic Toxicology and Cancer Biology, NIB: National Institute of Biology, Ljubljana, Slovenia
| | - Lea Thibol
- Department of Chemistry, RPTU: Division of Food Chemistry and Toxicology, Kaiserslautern, Germany
| | | | - Ariane Vettorazzi
- Department of Pharmacology and Toxicology, School of Pharmacy and Nutrition, UNAV: University of Navarra, Pamplona, Spain
| | - Bob Van de Water
- UL-LACDR: Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Naouale El Yamani
- Health Effects Laboratory, NILU: The Climate and Environmental Research Institute, Kjeller, Norway
| | - Bojana Zegura
- Department of Genetic Toxicology and Cancer Biology, NIB: National Institute of Biology, Ljubljana, Slovenia
| | - Michael Oelgeschläger
- Department Experimental Toxicology and ZEBET, German Centre for the Protection of Laboratory Animals (Bf3R) and Department Food Safety, BfR: German Federal Institute for Risk Assessment, Berlin, Germany
| |
Collapse
|
8
|
Radi ZA, Khan N. Pathophysiology and human cancer risk assessment of pharmaceutical-induced thymoma in carcinogenicity studies. Toxicol Appl Pharmacol 2023; 466:116471. [PMID: 36934859 DOI: 10.1016/j.taap.2023.116471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 03/19/2023]
Abstract
Thymoma, a tumor of thymic lymphocytes or thymic epithelial cells (TECs), is a common spontaneous tumor in Wistar Han rats, especially in females with up to 18% incidence in controls. In addition to sex, there are rat strain differences in background incidence of thymomas such as Sprague Dawley versus Wistar Han rats. Human thymomas are very rare and without clear differences in incidence between males and females. Immunomodulatory and anti-inflammatory pharmaceutical drug classes, including Janus kinase inhibitors, increase the incidence of benign thymoma in two-year rat carcinogenicity studies. Potential non-genotoxic mechanisms that might contribute to the pathogenesis of thymoma development in one sex (female) Wistar Han rats include: (1) hormonal differences, (2) high proliferation rate of TECs, (3) delayed physiologic thymic involution, and/or (4) significant level of immunosuppression at high doses of a pharmaceutical drug. Factors to consider in the human cancer risk assessment of pharmaceutical-induced thymoma are: the genotoxicity of the test article, sex and strain of rats, exposure safety margins, and pathophysiologic differences and similarities of thymoma between rats and humans. Totality of weight of evidence approach and available data suggest thymomas observed in carcinogenicity studies of pharmaceutical drugs are not relevant for human risk at clinically relevant therapeutic doses.
Collapse
Affiliation(s)
- Zaher A Radi
- Pfizer Worldwide Research, Development and Medical, Drug Safety R&D, 1 Portland Street, Cambridge, MA 02140, USA.
| | - Nasir Khan
- Pfizer Worldwide Research, Development and Medical, Drug Safety R&D, Eastern Point Road, Groton, CT 06340, USA
| |
Collapse
|
9
|
Berry SCL, Cohen SM, Corton JC, de Camargo JLV, Eisenbrand G, Fukushima S, Greim H, Weber K, Rietjens IMCM, Strupp C. Letter to the Editors regarding "10% body weight (gain) change as criterion for the maximum tolerated dose: A critical analysis". Regul Toxicol Pharmacol 2023; 139:105362. [PMID: 36828242 DOI: 10.1016/j.yrtph.2023.105362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023]
Affiliation(s)
| | - Samuel M Cohen
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, USA
| | - J Christopher Corton
- Center for Computational Toxicology and Exposure, Office of Research and Development, Environmental Protection Agency, Durham, NC, 27711, USA
| | | | - Gerhard Eisenbrand
- Department of Chemistry, Div of Food Chemistry & Toxicology, University of Kaiserslautern, Heidelberg, Germany
| | | | | | | | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Wageningen, the Netherlands
| | | |
Collapse
|
10
|
Hilton GM, Corvi R, Luijten M, Mehta J, Wolf DC. Towards achieving a modern science-based paradigm for agrochemical carcinogenicity assessment. Regul Toxicol Pharmacol 2022; 137:105301. [PMID: 36436696 DOI: 10.1016/j.yrtph.2022.105301] [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: 08/30/2022] [Revised: 10/21/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
The rodent cancer bioassay has been the standard approach to fulfill regulatory requirements for assessing human carcinogenic potential of agrochemicals, food additives, industrial chemicals, and pharmaceuticals. Decades of research have described the limitations of the rodent cancer bioassay leading to international initiatives to seek alternatives and establish approaches that modernize carcinogenicity assessment. Biologically relevant approaches can provide mechanistic information and increased efficiency for evaluating hazard and risk of chemical carcinogenicity to humans. The application of human-relevant mechanistic understanding to support new approaches to carcinogenicity assessment will be invaluable for regulatory decision-making. The present work outlines the challenges and opportunities that authorities should consider as they come together to build a roadmap that leads to global acceptance and incorporation of fit-for-purpose, scientifically defensible new approaches for human-relevant carcinogenicity assessment of agrochemicals.
Collapse
Affiliation(s)
- Gina M Hilton
- PETA Science Consortium International e.V., Stuttgart, Germany.
| | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Mirjam Luijten
- National Institute for Public Health and the Environment, Netherlands
| | - Jyotigna Mehta
- ADAMA Agricultural Solutions Ltd, Reading, United Kingdom
| | | |
Collapse
|
11
|
Manuppello J, Slankster-Schmierer E, Baker E, Sullivan K. Animal use and opportunities for reduction in carcinogenicity studies supporting approved new drug applications in the U.S., 2015-2019. Regul Toxicol Pharmacol 2022; 137:105289. [PMID: 36379352 DOI: 10.1016/j.yrtph.2022.105289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/12/2022] [Accepted: 11/06/2022] [Indexed: 11/15/2022]
Abstract
A minimum of 65,341 rats and mice were used in 109 carcinogenicity studies conducted for new drug applications approved by the U.S. Food and Drug Administration from 2015 through 2019. By analyzing how these animals were used, we compared the potential for reducing animal use of implementing existing international guidelines and recommendations. The greatest reduction, 18.7%, would result from evaluating exposure by microsampling blood in main studies to replace toxicokinetics satellites, which used three-fold more mice than rats. A similar reduction, 17.3%, would result from replacing 33 long-term studies in mice with short-term studies in transgenic mice. Based on histopathology findings in chronic studies, 15 long-term studies in rats could have been waived, using 8410 fewer rats. Simply using single, rather than dual, negative control groups would result in a 7.8% reduction, and eliminating positive control groups would use 640 fewer transgenic mice. Combined, an estimated 46% reduction would be achieved, using approximately 29,876 fewer animals. The publication of an addendum to the main carcinogenicity testing guideline promises to decrease the number of long-term studies conducted in rats and mice and presents opportunity to promote full harmonization and implementation of related recommendations that would further dramatically reduce animal use.
Collapse
Affiliation(s)
- Joseph Manuppello
- Physicians Committee for Responsible Medicine, 5100 Wisconsin Ave., NW, Suite 400, Washington, DC, 20016-4131, USA.
| | - Eryn Slankster-Schmierer
- Physicians Committee for Responsible Medicine, 5100 Wisconsin Ave., NW, Suite 400, Washington, DC, 20016-4131, USA
| | - Elizabeth Baker
- Physicians Committee for Responsible Medicine, 5100 Wisconsin Ave., NW, Suite 400, Washington, DC, 20016-4131, USA
| | - Kristie Sullivan
- Physicians Committee for Responsible Medicine, 5100 Wisconsin Ave., NW, Suite 400, Washington, DC, 20016-4131, USA
| |
Collapse
|
12
|
Corton JC, Mitchell CA, Auerbach S, Bushel P, Ellinger-Ziegelbauer H, Escobar PA, Froetschl R, Harrill AH, Johnson K, Klaunig JE, Pandiri AR, Podtelezhnikov AA, Rager JE, Tanis KQ, van der Laan JW, Vespa A, Yauk CL, Pettit SD, Sistare FD. A Collaborative Initiative to Establish Genomic Biomarkers for Assessing Tumorigenic Potential to Reduce Reliance on Conventional Rodent Carcinogenicity Studies. Toxicol Sci 2022; 188:4-16. [PMID: 35404422 PMCID: PMC9238304 DOI: 10.1093/toxsci/kfac041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
There is growing recognition across broad sectors of the scientific community that use of genomic biomarkers has the potential to reduce the need for conventional rodent carcinogenicity studies of industrial chemicals, agrochemicals, and pharmaceuticals through a weight-of-evidence approach. These biomarkers fall into 2 major categories: (1) sets of gene transcripts that can identify distinct tumorigenic mechanisms of action; and (2) cancer driver gene mutations indicative of rapidly expanding growth-advantaged clonal cell populations. This call-to-action article describes a collaborative approach launched to develop and qualify biomarker gene expression panels that measure widely accepted molecular pathways linked to tumorigenesis and their activation levels to predict tumorigenic doses of chemicals from short-term exposures. Growing evidence suggests that application of such biomarker panels in short-term exposure rodent studies can identify both tumorigenic hazard and tumorigenic activation levels for chemical-induced carcinogenicity. In the future, this approach will be expanded to include methodologies examining mutations in key cancer driver gene mutation hotspots as biomarkers of both genotoxic and nongenotoxic chemical tumor risk. Analytical, technical, and biological validation studies of these complementary genomic tools are being undertaken by multisector and multidisciplinary collaborative teams within the Health and Environmental Sciences Institute. Success from these efforts will facilitate the transition from current heavy reliance on conventional 2-year rodent carcinogenicity studies to more rapid animal- and resource-sparing approaches for mechanism-based carcinogenicity evaluation supporting internal and regulatory decision-making.
Collapse
Affiliation(s)
- J Christopher Corton
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | - Constance A Mitchell
- Health and Environmental Sciences Institute, Washington, District of Columbia, USA
| | - Scott Auerbach
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Pierre Bushel
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, North Carolina, USA
| | | | - Patricia A Escobar
- Safety Assessment and Laboratory Animal Resources, Merck Sharp & Dohme Corp, West Point, Pennsylvania, USA
| | - Roland Froetschl
- BfArM-Bundesinstitut für Arzneimittel und Medizinprodukte, Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | - Alison H Harrill
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | - James E Klaunig
- Laboratory of Investigative Toxicology and Pathology, Department of Environmental and Occupational Health, Indiana School of Public Health, Indiana University, Bloomington, Indiana, USA
| | - Arun R Pandiri
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | | | - Julia E Rager
- The Institute for Environmental Health Solutions, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Keith Q Tanis
- Safety Assessment and Laboratory Animal Resources, Merck Sharp & Dohme Corp, West Point, Pennsylvania, USA
| | - Jan Willem van der Laan
- Section on Pharmacology, Toxicology and Kinetics, Medicines Evaluation Board, Utrecht, The Netherlands
| | - Alisa Vespa
- Therapeutic Products Directorate, Health Canada, Ottawa, Ontario, Canada
| | - Carole L Yauk
- Department of Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Syril D Pettit
- Health and Environmental Sciences Institute, Washington, District of Columbia, USA
| | - Frank D Sistare
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
13
|
Current Therapeutic Landscape and Safety Roadmap for Targeting the Aryl Hydrocarbon Receptor in Inflammatory Gastrointestinal Indications. Cells 2022; 11:cells11101708. [PMID: 35626744 PMCID: PMC9139855 DOI: 10.3390/cells11101708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/30/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023] Open
Abstract
Target modulation of the AhR for inflammatory gastrointestinal (GI) conditions holds great promise but also the potential for safety liabilities both within and beyond the GI tract. The ubiquitous expression of the AhR across mammalian tissues coupled with its role in diverse signaling pathways makes development of a “clean” AhR therapeutically challenging. Ligand promiscuity and diversity in context-specific AhR activation further complicates targeting the AhR for drug development due to limitations surrounding clinical translatability. Despite these concerns, several approaches to target the AhR have been explored such as small molecules, microbials, PROTACs, and oligonucleotide-based approaches. These various chemical modalities are not without safety liabilities and require unique de-risking strategies to parse out toxicities. Collectively, these programs can benefit from in silico and in vitro methodologies that investigate specific AhR pathway activation and have the potential to implement thresholding parameters to categorize AhR ligands as “high” or “low” risk for sustained AhR activation. Exploration into transcriptomic signatures for AhR safety assessment, incorporation of physiologically-relevant in vitro model systems, and investigation into chronic activation of the AhR by structurally diverse ligands will help address gaps in our understanding regarding AhR-dependent toxicities. Here, we review the role of the AhR within the GI tract, novel therapeutic modality approaches to target the AhR, key AhR-dependent safety liabilities, and relevant strategies that can be implemented to address drug safety concerns. Together, this review discusses the emerging therapeutic landscape of modalities targeting the AhR for inflammatory GI indications and offers a safety roadmap for AhR drug development.
Collapse
|
14
|
Hilton GM, Adcock C, Akerman G, Baldassari J, Battalora M, Casey W, Clippinger AJ, Cope R, Goetz A, Hayes AW, Papineni S, Peffer RC, Ramsingh D, Williamson Riffle B, Sanches da Rocha M, Ryan N, Scollon E, Visconti N, Wolf DC, Yan Z, Lowit A. Rethinking chronic toxicity and carcinogenicity assessment for agrochemicals project (ReCAAP): A reporting framework to support a weight of evidence safety assessment without long-term rodent bioassays. Regul Toxicol Pharmacol 2022; 131:105160. [PMID: 35311659 DOI: 10.1016/j.yrtph.2022.105160] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 11/17/2022]
Abstract
Rodent cancer bioassays have been long-required studies for regulatory assessment of human cancer hazard and risk. These studies use hundreds of animals, are resource intensive, and certain aspects of these studies have limited human relevance. The past 10 years have seen an exponential growth of new technologies with the potential to effectively evaluate human cancer hazard and risk while reducing, refining, or replacing animal use. To streamline and facilitate uptake of new technologies, a workgroup comprised of scientists from government, academia, non-governmental organizations, and industry stakeholders developed a framework for waiver rationales of rodent cancer bioassays for consideration in agrochemical safety assessment. The workgroup used an iterative approach, incorporating regulatory agency feedback, and identifying critical information to be considered in a risk assessment-based weight of evidence determination of the need for rodent cancer bioassays. The reporting framework described herein was developed to support a chronic toxicity and carcinogenicity study waiver rationale, which includes information on use pattern(s), exposure scenario(s), pesticidal mode-of-action, physicochemical properties, metabolism, toxicokinetics, toxicological data including mechanistic data, and chemical read-across from similar registered pesticides. The framework could also be applied to endpoints other than chronic toxicity and carcinogenicity, and for chemicals other than agrochemicals.
Collapse
Affiliation(s)
- Gina M Hilton
- PETA Science Consortium International e.V., Stuttgart, Germany.
| | - Catherine Adcock
- Health Canada, Pest Management Regulatory Agency, Ottawa, Ontario, Canada
| | - Gregory Akerman
- United States Environmental Protection Agency, Office of Pesticide Programs, Washington DC, USA
| | | | | | - Warren Casey
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | | | - Rhian Cope
- Australian Pesticides and Veterinary Medicines Authority, Armidale, New South Wales, Australia
| | - Amber Goetz
- Syngenta Crop Protection, LLC, Greensboro, NC, USA
| | - A Wallace Hayes
- University of South Florida College of Public Health, Tampa, FL, USA
| | | | | | - Deborah Ramsingh
- Health Canada, Pest Management Regulatory Agency, Ottawa, Ontario, Canada
| | | | | | - Natalia Ryan
- Syngenta Crop Protection, LLC, Greensboro, NC, USA
| | | | | | | | | | - Anna Lowit
- United States Environmental Protection Agency, Office of Pesticide Programs, Washington DC, USA
| |
Collapse
|
15
|
Hollingshead BD, Khan N, Schuler M, Radi Z. Development challenges for carcinogenicity risk assessments of topical drugs. J Toxicol Sci 2022; 47:1-11. [PMID: 34987136 DOI: 10.2131/jts.47.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The nonclinical safety package to support development and approval of drugs intended to be administered by topical application generally follows International Council for Harmonisation multidisciplinary 3 (ICH M3) and topic specific safety (ICH S) guidances. However, some aspects of topical drug development may require case-by-case determination of nonclinical safety strategies. The necessity to conduct a dermal rodent carcinogenicity study is one such example that is not considered an obligate component of a nonclinical safety data package for drug approval. While absence of systemic exposure, as stated in ICH M3, is a primary reason to forego a dermal carcinogenicity assessment, there may also be other factors for consideration in determining the need for a life-time carcinogencity study by dermal route to aid in the overall human cancer risk assessment. We therefore reviewed nonclinical carcinogencity data packages from drugs approved by the FDA or PMDA over a ~25 year time period to evaluate outcomes of oral versus topical carcinogencity studies and to understand their utility for informing the overall human risk assessment. We also discuss various other properties of topical small molecules that could impact the decisions to conduct a dermal life-time rodent carcinogenicity study. Collectively, the need to conduct 2-year dermal carcinogenicity studies in rodents should be determined case-by-case and consider scientific factors such existing systemic toxicity and carcinogenicity study data, anticipated drug exposures in skin, skin evaluation from the chronic minipig toxicity study, and genetic toxicity profile.
Collapse
Affiliation(s)
- Brett D Hollingshead
- Pfizer Drug Safety Research and Development, 1 Portland Street Cambridge, MA, USA
| | - Nasir Khan
- Pfizer Drug Safety Research and Development, 445 Eastern Point Road, Groton, CT, USA
| | - Maik Schuler
- Pfizer Drug Safety Research and Development, 445 Eastern Point Road, Groton, CT, USA
| | - Zaher Radi
- Pfizer Drug Safety Research and Development, 1 Portland Street Cambridge, MA, USA
| |
Collapse
|
16
|
Hisada S, Tsubota K, Inoue K, Yamada H, Ikeda T, Sistare FD. Survey of tumorigenic sensitivity in 6-month rasH2-Tg mice studies compared with 2-year rodent assays. J Toxicol Pathol 2022; 35:53-73. [PMID: 35221496 PMCID: PMC8828610 DOI: 10.1293/tox.2021-0031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/07/2021] [Indexed: 12/04/2022] Open
Abstract
The pharmacokinetic endpoint of a 25-fold increase in human exposure is one of the specified criteria for high-dose selection for 2-year carcinogenicity studies in rodents according to ICH S1C(R2). However, this criterion is not universally accepted for 6-month carcinogenicity tests in rasH2-Tg mice. To evaluate an appropriate multiple for rasH2-Tg mice, we evaluated data for 53 compounds across five categories of rasH2-Tg mouse-positive [(1) genotoxic and (2) non-genotoxic] carcinogens and rasH2-Tg mouse-negative [(3) non-genotoxic carcinogens with clear or uncertain human relevance; (4) non-genotoxic rodent-specific carcinogens; and (5) non-carcinogens], and surveyed their tumorigenic activities and high doses in rasH2-Tg mice and 2-year rodent models. Our survey indicated that area under the curve (AUC) margins (AMs) or body surface area-adjusted dose ratios (DRs) of tumorigenesis in rasH2-Tg mice to the maximum recommended human dose (MRHD) were 0.05- to 5.2-fold in 6 category (1) compounds with small differences between models and 0.2- to 47-fold in 7 category (2) including three 2-year rat study-negative compounds. Among all 53 compounds, including 40 compounds of the rasH2-Tg mouse-negative category (3), (4), and (5), no histopathologic risk factors for rodent neoplasia were induced only at doses above 50-fold AM or DR in rasH2-Tg mice except for two compounds, which induced hyperplasia and had no relationship with the tumors observed in the rasH2-Tg mouse or 2-year rodent studies. From the results of these surveys, we confirmed that exceeding a high dose level of 50-fold AM in rasH2-Tg mouse carcinogenicity studies does not appear to be of value.
Collapse
Affiliation(s)
- Shigeru Hisada
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- ASKA Pharmaceutical Co., Ltd., 26-1, Muraoka-Higashi
2-chome, Fujisawa-shi, Kanagawa 251-8555, Japan
| | - Kenjiro Tsubota
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- Astellas Pharma Inc, 21 Miyukigaoka, Tsukuba-shi, Ibaraki
305-8585, Japan
| | - Kenji Inoue
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- Maruho Co., Ltd., 93 Chudoji Awatacho, Shimogyo-ku, Kyoto
600-8815, Japan
| | - Hisaharu Yamada
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- Taisho Pharmaceutical Co., Ltd., 1-403 Yoshino-cho, Kita-ku,
Saitama 331-9530, Japan
| | - Takanori Ikeda
- Non-Clinical Evaluation Expert Committee, Drug Evaluation
Committee, The Japan Pharmaceutical Manufacturers Association, 2-3-11 Nihonbashi-Honcho,
Chuo-ku, Tokyo 103-0023, Japan
- MSD K.K., 1-13-12 Kudan-kita, Chiyoda-ku, Tokyo 102-8667,
Japan
| | - Frank D. Sistare
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA
19486-0004, USA
- Current Address: 315 Meadowmont Lane, Chapel Hill, NC 27517,
USA
| |
Collapse
|
17
|
Stalford SA, Cayley AN, de Oliveira AAF. Employing an adverse outcome pathway framework for weight-of-evidence assessment with application to the ICH S1B guidance addendum. Regul Toxicol Pharmacol 2021; 127:105071. [PMID: 34737134 DOI: 10.1016/j.yrtph.2021.105071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/14/2021] [Accepted: 10/25/2021] [Indexed: 10/20/2022]
Abstract
Across industry, there is a paradigm shift occurring for carcinogenicity testing, with the focus moving from long term animal studies to alternative approaches. Based on the explorative work done in recent years, the International Council for Harmonization (ICH) recently published a draft addendum to the S1B guidance, which allows for a weight-of-evidence (WoE) assessment to be conducted based on data gathered throughout the pharmaceutical development process and literature to mitigate some testing in rodents if the body of evidence clearly shows undertaking an animal lifetime study would not add value to the risk assessment. While several alternative approaches already exist, and other new approach methodologies (NAMs) are being explored, all of which can contribute to this WoE, it is important that all the evidence can be combined in a meaningful and consistent way to reach a conclusion. Adverse outcome pathways have been advocated as a framework for organising evidence in an integrated approach to testing and assessment, which gives context to data and can aid reaching a conclusion as to the adverse outcome (AO). This approach can be combined with a reasoning methodology to give a prediction for an AO and applied to the factors which need to be considered for the ICH S1B WoE to predict for carcinogenicity. Using this approach to the WoE assessment, consistent, scientifically robust, and transparent calls can be made as to whether conducting an animal carcinogenicity study would add value to a human risk assessment and mitigate the need to run animal studies unnecessarily.
Collapse
Affiliation(s)
- Susanne A Stalford
- Lhasa Limited, Granary Wharf House, 2 Canal Wharf, Leeds, LS11 5PS, United Kingdom.
| | - Alex N Cayley
- Lhasa Limited, Granary Wharf House, 2 Canal Wharf, Leeds, LS11 5PS, United Kingdom
| | | |
Collapse
|
18
|
Rethinking agrochemical safety assessment: A perspective. Regul Toxicol Pharmacol 2021; 127:105068. [PMID: 34678328 DOI: 10.1016/j.yrtph.2021.105068] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/09/2021] [Accepted: 10/18/2021] [Indexed: 11/20/2022]
Abstract
Agrochemical safety assessment has traditionally relied on the use of animals for toxicity testing, based on scientific understanding and test guidelines developed in the 1980s. However, since then, there have been significant advances in the toxicological sciences that have improved our understanding of mechanisms underpinning adverse human health effects. The time is ripe to 'rethink' approaches used for human safety assessments of agrochemicals to ensure they reflect current scientific understanding and increasingly embrace new opportunities to improve human relevance and predictivity, and to reduce the reliance on animals. Although the ultimate aim is to enable a paradigm shift and an overhaul of global regulatory data requirements, there is much that can be done now to ensure new opportunities and approaches are adopted and implemented within the current regulatory frameworks. This commentary reviews current initiatives and emerging opportunities to embrace new approaches to improve agrochemical safety assessment for humans, and considers various endpoints and initiatives (including acute toxicity, repeat dose toxicity studies, carcinogenicity, developmental and reproductive toxicity, exposure-driven approaches, inhalation toxicity, and data modelling). Realistic aspirations to improve safety assessment, incorporate new technologies and reduce reliance on animal testing without compromising protection goals are discussed.
Collapse
|
19
|
Desaulniers D, Vasseur P, Jacobs A, Aguila MC, Ertych N, Jacobs MN. Integration of Epigenetic Mechanisms into Non-Genotoxic Carcinogenicity Hazard Assessment: Focus on DNA Methylation and Histone Modifications. Int J Mol Sci 2021; 22:10969. [PMID: 34681626 PMCID: PMC8535778 DOI: 10.3390/ijms222010969] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
Epigenetics involves a series of mechanisms that entail histone and DNA covalent modifications and non-coding RNAs, and that collectively contribute to programing cell functions and differentiation. Epigenetic anomalies and DNA mutations are co-drivers of cellular dysfunctions, including carcinogenesis. Alterations of the epigenetic system occur in cancers whether the initial carcinogenic events are from genotoxic (GTxC) or non-genotoxic (NGTxC) carcinogens. NGTxC are not inherently DNA reactive, they do not have a unifying mode of action and as yet there are no regulatory test guidelines addressing mechanisms of NGTxC. To fil this gap, the Test Guideline Programme of the Organisation for Economic Cooperation and Development is developing a framework for an integrated approach for the testing and assessment (IATA) of NGTxC and is considering assays that address key events of cancer hallmarks. Here, with the intent of better understanding the applicability of epigenetic assays in chemical carcinogenicity assessment, we focus on DNA methylation and histone modifications and review: (1) epigenetic mechanisms contributing to carcinogenesis, (2) epigenetic mechanisms altered following exposure to arsenic, nickel, or phenobarbital in order to identify common carcinogen-specific mechanisms, (3) characteristics of a series of epigenetic assay types, and (4) epigenetic assay validation needs in the context of chemical hazard assessment. As a key component of numerous NGTxC mechanisms of action, epigenetic assays included in IATA assay combinations can contribute to improved chemical carcinogen identification for the better protection of public health.
Collapse
Affiliation(s)
- Daniel Desaulniers
- Environmental Health Sciences and Research Bureau, Hazard Identification Division, Health Canada, AL:2203B, Ottawa, ON K1A 0K9, Canada
| | - Paule Vasseur
- CNRS, LIEC, Université de Lorraine, 57070 Metz, France;
| | - Abigail Jacobs
- Independent at the Time of Publication, Previously US Food and Drug Administration, Rockville, MD 20852, USA;
| | - M. Cecilia Aguila
- Toxicology Team, Division of Human Food Safety, Center for Veterinary Medicine, US Food and Drug Administration, Department of Health and Human Services, Rockville, MD 20852, USA;
| | - Norman Ertych
- German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Diedersdorfer Weg 1, 12277 Berlin, Germany;
| | - Miriam N. Jacobs
- Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton OX11 0RQ, UK;
| |
Collapse
|
20
|
Podtelezhnikov AA, Monroe JJ, Aslamkhan AG, Pearson K, Qin C, Tamburino AM, Loboda AP, Glaab WE, Sistare FD, Tanis KQ. Quantitative Transcriptional Biomarkers of Xenobiotic Receptor Activation in Rat Liver for the Early Assessment of Drug Safety Liabilities. Toxicol Sci 2021; 175:98-112. [PMID: 32119089 DOI: 10.1093/toxsci/kfaa026] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The robust transcriptional plasticity of liver mediated through xenobiotic receptors underlies its ability to respond rapidly and effectively to diverse chemical stressors. Thus, drug-induced gene expression changes in liver serve not only as biomarkers of liver injury, but also as mechanistic sentinels of adaptation in metabolism, detoxification, and tissue protection from chemicals. Modern RNA sequencing methods offer an unmatched opportunity to quantitatively monitor these processes in parallel and to contextualize the spectrum of dose-dependent stress, adaptation, protection, and injury responses induced in liver by drug treatments. Using this approach, we profiled the transcriptional changes in rat liver following daily oral administration of 120 different compounds, many of which are known to be associated with clinical risk for drug-induced liver injury by diverse mechanisms. Clustering, correlation, and linear modeling analyses were used to identify and optimize coexpressed gene signatures modulated by drug treatment. Here, we specifically focused on prioritizing 9 key signatures for their pragmatic utility for routine monitoring in initial rat tolerability studies just prior to entering drug development. These signatures are associated with 5 canonical xenobiotic nuclear receptors (AHR, CAR, PXR, PPARα, ER), 3 mediators of reactive metabolite-mediated stress responses (NRF2, NRF1, P53), and 1 liver response following activation of the innate immune response. Comparing paradigm chemical inducers of each receptor to the other compounds surveyed enabled us to identify sets of optimized gene expression panels and associated scoring algorithms proposed as quantitative mechanistic biomarkers with high sensitivity, specificity, and quantitative accuracy. These findings were further qualified using public datasets, Open TG-GATEs and DrugMatrix, and internal development compounds. With broader collaboration and additional qualification, the quantitative toxicogenomic framework described here could inform candidate selection prior to committing to drug development, as well as complement and provide a deeper understanding of the conventional toxicology study endpoints used later in drug development.
Collapse
Affiliation(s)
| | - James J Monroe
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Amy G Aslamkhan
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Kara Pearson
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Chunhua Qin
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | | | | | - Warren E Glaab
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | - Frank D Sistare
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., West Point, Pennsylvania 19486-0004
| | | |
Collapse
|
21
|
Madia F, Pillo G, Worth A, Corvi R, Prieto P. Integration of data across toxicity endpoints for improved safety assessment of chemicals: the example of carcinogenicity assessment. Arch Toxicol 2021; 95:1971-1993. [PMID: 33830278 PMCID: PMC8166685 DOI: 10.1007/s00204-021-03035-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Abstract
In view of the need to enhance the assessment of consumer products called for in the EU Chemicals Strategy for Sustainability, we developed a methodology for evaluating hazard by combining information across different systemic toxicity endpoints and integrating the information with new approach methodologies. This integrates mechanistic information with a view to avoiding redundant in vivo studies, minimising reliance on apical endpoint tests and ultimately devising efficient testing strategies. Here, we present the application of our methodology to carcinogenicity assessment, mapping the available information from toxicity test methods across endpoints to the key characteristics of carcinogens. Test methods are deconstructed to allow the information they provide to be organised in a systematic way, enabling the description of the toxicity mechanisms leading to the adverse outcome. This integrated approach provides a flexible and resource-efficient means of fully exploiting test methods for which test guidelines are available to fulfil regulatory requirements for systemic toxicity assessment as well as identifying where new methods can be integrated.
Collapse
Affiliation(s)
- Federica Madia
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21027, Ispra, VA, Italy.
| | - Gelsomina Pillo
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21027, Ispra, VA, Italy
| | - Andrew Worth
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21027, Ispra, VA, Italy
| | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21027, Ispra, VA, Italy
| | - Pilar Prieto
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21027, Ispra, VA, Italy
| |
Collapse
|
22
|
Yamada T, Cohen SM, Lake BG. Critical evaluation of the human relevance of the mode of action for rodent liver tumor formation by activators of the constitutive androstane receptor (CAR). Crit Rev Toxicol 2021; 51:373-394. [PMID: 34264181 DOI: 10.1080/10408444.2021.1939654] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Many nongenotoxic chemicals have been shown to produce liver tumors in mice and/or rats by a mode of action (MOA) involving activation of the constitutive androstane receptor (CAR). Studies with phenobarbital (PB) and other compounds have identified the key events for this MOA: CAR activation; increased hepatocellular proliferation; altered foci formation; and ultimately the development of adenomas/carcinomas. In terms of human relevance, the pivotal species difference is that CAR activators are mitogenic agents in mouse and rat hepatocytes, but they do not stimulate increased hepatocellular proliferation in humans. This conclusion is supported by substantial in vitro studies with cultured rodent and human hepatocytes and also by in vivo studies with chimeric mice with human hepatocytes. Examination of the literature reveals many similarities in the hepatic effects and species differences between activators of rodent CAR and the peroxisome proliferator-activated receptor alpha (PPARα), with PPARα activators also not being mitogenic agents in human hepatocytes. Overall, a critical analysis of the available data demonstrates that the established MOA for rodent liver tumor formation by PB and other CAR activators is qualitatively not plausible for humans. This conclusion is supported by data from several human epidemiology studies.
Collapse
Affiliation(s)
- Tomoya Yamada
- Environmental Health Science Laboratory, Sumitomo Chemical Company, Ltd., Osaka, Japan
| | - Samuel M Cohen
- Department of Pathology and Microbiology, Havlik-Wall Professor of Oncology, University of Nebraska Medical Center, Nebraska Medical Center, Omaha, NE, USA
| | - Brian G Lake
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| |
Collapse
|
23
|
Halappanavar S, Nymark P, Krug HF, Clift MJD, Rothen-Rutishauser B, Vogel U. Non-Animal Strategies for Toxicity Assessment of Nanoscale Materials: Role of Adverse Outcome Pathways in the Selection of Endpoints. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007628. [PMID: 33559363 DOI: 10.1002/smll.202007628] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/08/2021] [Indexed: 06/12/2023]
Abstract
Faster, cheaper, sensitive, and mechanisms-based animal alternatives are needed to address the safety assessment needs of the growing number of nanomaterials (NM) and their sophisticated property variants. Specifically, strategies that help identify and prioritize alternative schemes involving individual test models, toxicity endpoints, and assays for the assessment of adverse outcomes, as well as strategies that enable validation and refinement of these schemes for the regulatory acceptance are needed. In this review, two strategies 1) the current nanotoxicology literature review and 2) the adverse outcome pathways (AOPs) framework, a systematic process that allows the assembly of available mechanistic information concerning a toxicological response in a simple modular format, are presented. The review highlights 1) the most frequently assessed and reported ad hoc in vivo and in vitro toxicity measurements in the literature, 2) various AOPs of relevance to inhalation toxicity of NM that are presently under development, and 3) their applicability in identifying key events of toxicity for targeted in vitro assay development. Finally, using an existing AOP for lung fibrosis, the specific combinations of cell types, exposure and test systems, and assays that are experimentally supported and thus, can be used for assessing NM-induced lung fibrosis, are proposed.
Collapse
Affiliation(s)
- Sabina Halappanavar
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, K1A0K9, Canada
- Department of Biology, University of Ottawa, Ottawa, K1N6N5, Canada
| | - Penny Nymark
- Institute of Environmental Medicine, Karolinska Institute, Nobels väg 13, Stockholm, 17177, Sweden
| | - Harald F Krug
- NanoCASE GmbH, St. Gallerstr. 58, Engelburg, 9032, Switzerland
| | - Martin J D Clift
- Institute of Life Science, Swansea University Medical School, Swansea University, Singleton Park, Swansea, Wales, SA2 8PP, UK
| | | | - Ulla Vogel
- National Research Centre for the Working Environment, Lersø Parkallé 105, Copenhagen, DK-2100, Denmark
- DTU Health Tech, Technical University of Denmark, Lyngby, DK-2800 Kgs., Denmark
| |
Collapse
|
24
|
Guzmán A, Encina G, Fernández de Henestrosa AR, Vila C, Tortajada A, Marín AP. Twenty six-week repeat dose oral rat toxicity study of cizolirtine, a substance-P and calcitonin gene-related peptide release modulator. Regul Toxicol Pharmacol 2021; 122:104916. [PMID: 33711392 DOI: 10.1016/j.yrtph.2021.104916] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 02/05/2023]
Abstract
Cizolirtine, a substance-P and calcitonin gene-related peptide release modulator developed for the treatment of pain and urinary incontinence, was orally administered for 26-weeks to rats at dosages of 20, 60 and 200 mg/kg/day. Clinical signs were limited to post-dosing salivation and brown staining on head and muzzle. There were slight decreases in bodyweight gain and slight increases in water consumption among cizolirtine-treated animals. Slight increases in plasma alkaline phosphatase activity, and cholesterol and phospholipid concentrations were observed in mid- and/or high-dose animals. Low urinary volume, pH and sodium and potassium outputs were observed after 12-weeks, and low urinary pH, low sodium and high potassium outputs at end of treatment. Increased relative (to bodyweight) liver weight was observed in high-dose animals. Treated males and high-dose females showed a dose-related increase in the incidence and severity of periacinar hepatocytic hypertrophy and midzonal/periacinar hepatocytic fat vacuolization. Increased incidences of hepatic clear cell foci were observed in all cizolirtine-treated male groups and, to a lesser extent, in treated females. Ovaries of treated females showed a dose-dependent increased incidence of absent corpora lutea and, occasionally, follicular cysts. The dosages of 20 and 60 mg/kg/day were considered as the No-Observed-Adverse-Effect Levels for males and females, respectively.
Collapse
Affiliation(s)
- Antonio Guzmán
- Toxicology Department, Welab Barcelona and Toxicology Department Esteve Pharmaceuticals, Spain.
| | - Gregorio Encina
- Development PK & Bioanalysis Department WeLab Barcelona and Development PK & Bioanalysis Department, Esteve Pharmaceuticals, Spain
| | | | - Cristina Vila
- Toxicology Department, Welab Barcelona and Toxicology Department Esteve Pharmaceuticals, Spain
| | - Araceli Tortajada
- Toxicology Department, Welab Barcelona and Toxicology Department Esteve Pharmaceuticals, Spain
| | - Ana-Paz Marín
- Toxicology Department, Welab Barcelona and Toxicology Department Esteve Pharmaceuticals, Spain
| |
Collapse
|
25
|
Heusinkveld H, Braakhuis H, Gommans R, Botham P, Corvaro M, van der Laan JW, Lewis D, Madia F, Manou I, Schorsch F, Wolterink G, Woutersen R, Corvi R, Mehta J, Luijten M. Towards a mechanism-based approach for the prediction of nongenotoxic carcinogenic potential of agrochemicals. Crit Rev Toxicol 2020; 50:725-739. [DOI: 10.1080/10408444.2020.1841732] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Harm Heusinkveld
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Hedwig Braakhuis
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Robin Gommans
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | | | | | | | | | - Federica Madia
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Irene Manou
- European Partnership for Alternative Approaches to Animal Testing (EPAA), Brussels, Belgium
| | | | - Gerrit Wolterink
- Centre for Nutrition, Prevention and Health Services, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Ruud Woutersen
- TNO Quality of Life, Zeist, and Wageningen University & Research, Wageningen, the Netherlands
| | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| |
Collapse
|
26
|
Corton JC, Korunes KL, Abedini J, El-Masri H, Brown J, Paul-Friedman K, Liu Y, Martini C, He S, Rooney J. Thresholds Derived From Common Measures in Rat Studies Are Predictive of Liver Tumorigenic Chemicals. Toxicol Pathol 2020; 48:857-874. [DOI: 10.1177/0192623320960412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We hypothesized that typical tissue and clinical chemistry (ClinChem) end points measured in rat toxicity studies exhibit chemical-independent biological thresholds beyond which cancer occurs. Using the rat in vivo TG-GATES study, 75 chemicals were examined across chemical-dose-time comparisons that could be linked to liver tumor outcomes. Thresholds for liver weight to body weight (LW/BW) and 21 serum ClinChem end points were defined as the maximum and minimum values for those exposures that did not lead to liver tumors in rats. Upper thresholds were identified for LW/BW (117%), aspartate aminotransferase (195%), alanine aminotransferase (141%), alkaline phosphatase (152%), and total bilirubin (115%), and lower thresholds were identified for phospholipids (82%), relative albumin (93%), total cholesterol (82%), and total protein (94%). Thresholds derived from the TG-GATES data set were consistent across other acute and subchronic rat studies. A training set of ClinChem and LW/BW thresholds derived from a 38 chemical training set from TG-GATES was predictive of liver tumor outcomes for a test set of 37 independent TG-GATES chemicals (91%). The thresholds were most predictive when applied to 7d treatments (98%). These findings provide support that biological thresholds for common end points in rodent studies can be used to predict chemical tumorigenic potential.
Collapse
Affiliation(s)
- J. Christopher Corton
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, USA
| | - Katharine L. Korunes
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, USA
- Department of Evolutionary Anthropology, Duke University, Durham, NC, USA
| | - Jaleh Abedini
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, USA
| | - Hisham El-Masri
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, USA
| | - Jason Brown
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, USA
| | - Katie Paul-Friedman
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, USA
| | - Ying Liu
- ASRC Federal, Research Triangle Park, NC, USA
| | | | - Shihan He
- ASRC Federal, Research Triangle Park, NC, USA
| | - John Rooney
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, USA
- Oak Ridge Institute for Science and Education (ORISE), National Health and Environmental Effects Research Laboratory (NHEERL), Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, NC, USA
| |
Collapse
|
27
|
Luijten M, Corvi R, Mehta J, Corvaro M, Delrue N, Felter S, Haas B, Hewitt NJ, Hilton G, Holmes T, Jacobs MN, Jacobs A, Lamplmair F, Lewis D, Madia F, Manou I, Melching-Kollmuss S, Schorsch F, Schütte K, Sewell F, Strupp C, van der Laan JW, Wolf DC, Wolterink G, Woutersen R, Zvonar Z, Heusinkveld H, Braakhuis H. A comprehensive view on mechanistic approaches for cancer risk assessment of non-genotoxic agrochemicals. Regul Toxicol Pharmacol 2020; 118:104789. [PMID: 33035627 DOI: 10.1016/j.yrtph.2020.104789] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/14/2020] [Accepted: 10/04/2020] [Indexed: 11/28/2022]
Abstract
Currently the only methods for non-genotoxic carcinogenic hazard assessment accepted by most regulatory authorities are lifetime carcinogenicity studies. However, these involve the use of large numbers of animals and the relevance of their predictive power and results has been scientifically challenged. With increased availability of innovative test methods and enhanced understanding of carcinogenic processes, it is believed that tumour formation can now be better predicted using mechanistic information. A workshop organised by the European Partnership on Alternative Approaches to Animal Testing brought together experts to discuss an alternative, mechanism-based approach for cancer risk assessment of agrochemicals. Data from a toolbox of test methods for detecting modes of action (MOAs) underlying non-genotoxic carcinogenicity are combined with information from subchronic toxicity studies in a weight-of-evidence approach to identify carcinogenic potential of a test substance. The workshop included interactive sessions to discuss the approach using case studies. These showed that fine-tuning is needed, to build confidence in the proposed approach, to ensure scientific correctness, and to address different regulatory needs. This novel approach was considered realistic, and its regulatory acceptance and implementation can be facilitated in the coming years through continued dialogue between all stakeholders and building confidence in alternative approaches.
Collapse
Affiliation(s)
- Mirjam Luijten
- National Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, the Netherlands.
| | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | | | | | - Nathalie Delrue
- Organisation for Economic Cooperation and Development (OECD), Paris, France
| | | | - Bodo Haas
- Federal Institute for Drugs and Medical Devices, Kurt-Georg-Kiesinger-Allee 3, 53175 Bonn, Germany
| | | | - Gina Hilton
- PETA International Science Consortium Ltd, London, UK
| | | | - Miriam N Jacobs
- Centre for Radiation, Chemical and Environmental Hazards (CRCE), Public Health England, UK
| | | | - Franz Lamplmair
- European Commission, DG Internal Market, Industry, Entrepreneurship and SMEs, Brussels, Belgium
| | | | - Federica Madia
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Irene Manou
- EPAA Industry Secretariat, Brussels, Belgium
| | | | | | | | - Fiona Sewell
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), London, UK
| | | | | | - Douglas C Wolf
- Syngenta Crop Protection, LLC, Greensboro, North Carolina, USA
| | - Gerrit Wolterink
- National Institute for Public Health and the Environment (RIVM), Centre for Nutrition, Prevention and Health Services, Bilthoven, the Netherlands
| | - Ruud Woutersen
- TNO Innovation for Life, Zeist; Wageningen University and Research, Wageningen, the Netherlands
| | | | - Harm Heusinkveld
- National Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, the Netherlands
| | - Hedwig Braakhuis
- National Institute for Public Health and the Environment (RIVM), Centre for Health Protection, Bilthoven, the Netherlands
| |
Collapse
|
28
|
Deshapriya US, Dinuka DLS, Ratnaweera PB, Ratnaweera CN. In silico study for prediction of novel bioactivities of the endophytic fungal alkaloid, mycoleptodiscin B for human targets. J Mol Graph Model 2020; 102:107767. [PMID: 33130394 DOI: 10.1016/j.jmgm.2020.107767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/23/2020] [Accepted: 09/28/2020] [Indexed: 11/28/2022]
Abstract
Mycoleptodiscin B is a natural product extracted from the endophytic fungus Mycoleptodiscus sp. found in Sri Lanka and Panama with experimentally unexplored activities for human targets. In this study, a computational methodology was applied to determine druggable targets of mycoleptodiscin B. According to the computational toxicity and pharmacokinetics assessment, mycoleptodiscin B was proven to be a suitable drug candidate. Druggable targets for this compound, aromatase, acidic plasma glycoprotein and androgen receptor, were predicted using reverse docking. A two-step validation of those targets was performed using conventional molecular docking and molecular dynamic (MD) simulations, resulting in aromatase being determined as the potential therapeutic target. Based on molecular mechanics/Generalized Born Surface Area (GBSA) free energies and ligand stability inside the active site cavity during its 120 ns MD run, it can be concluded that mycoleptodiscin B is a potent aromatase inhibitor and could be subjected to further in vitro and in vivo experiments in the drug development pipeline. Consequently, natural product chemists can quickly identify the hidden medicinal properties of their miracle compounds using the computational approach applied in this research.
Collapse
Affiliation(s)
- Uthpala S Deshapriya
- College of Chemical Sciences, Institute of Chemistry Ceylon, Rajagiriya, Sri Lanka; Faculty of Medicine, University of Colombo, Colombo, Sri Lanka
| | - D L Senal Dinuka
- College of Chemical Sciences, Institute of Chemistry Ceylon, Rajagiriya, Sri Lanka; Department of Chemistry, Mississippi State University, Mississippi State, USA
| | - Pamoda B Ratnaweera
- Department of Science and Technology, Faculty of Applied Sciences, Uva Wellassa University, Badulla, Sri Lanka
| | - Chinthaka N Ratnaweera
- College of Chemical Sciences, Institute of Chemistry Ceylon, Rajagiriya, Sri Lanka; Department of Chemistry, University of Ruhuna, Matara, Sri Lanka.
| |
Collapse
|
29
|
Lewis RW, Hill T, Corton JC. A set of six Gene expression biomarkers and their thresholds identify rat liver tumorigens in short-term assays. Toxicology 2020; 443:152547. [PMID: 32755643 PMCID: PMC10439517 DOI: 10.1016/j.tox.2020.152547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 02/01/2023]
Abstract
Traditional methods for cancer risk assessment are retrospective, resource-intensive, and not feasible for the vast majority of environmental chemicals. In earlier studies, we used a set of six biomarkers to accurately identify liver tumorigens in transcript profiles derived from chemically-treated rats using either a Toxicological Priority Index (ToxPi) approach or using derived biomarker thresholds for cancer. The biomarkers consisting of 7-113 genes are used to predict the most common liver cancer molecular initiating events: genotoxicity, cytotoxicity and activation of the xenobiotic receptors AhR, CAR, ER, and PPARα. In the present study, we apply and evaluate the performance of these methods for cancer prediction in an independent rat liver study of 44 chemicals (6 h-7d exposures) examined by Affymetrix arrays. In the first approach, ToxPi ranking of biomarker scores consistently gave the highest scores to tumorigenic chemical-dose pairs; balanced accuracies for identification of liver tumorigenic chemicals were up to 89 %. The second approach used tumorigenic thresholds derived in the present study or from our earlier study that were set at the maximum value for chemical-dose exposures without detectable liver tumor outcomes. Using these thresholds, balanced accuracies were up to 90 %. Both approaches identified all tumorigenic chemicals. Almost all of the tumorigenic chemicals activated more than one MIE. We also compared biomarker responses between two types of profiling platforms (Affymetrix full-genome array, TempO-Seq 1500+ array containing ∼2600 genes) and found that the lack of the full set of biomarker genes on the 1500+ array resulted in decreased ability to identify chemicals that activate the MIEs. Overall, these results demonstrate that predictive approaches based on the 6 biomarkers could be used in short-term assays to identify chemicals and their doses that induce liver tumors, the most common endpoint in rodent bioassays.
Collapse
Affiliation(s)
- Robert W Lewis
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, United States.
| | - Thomas Hill
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, United States; Oak Ridge Institute for Science and Education (ORISE) fellow Office of Research and Development, U.S. Environmental Protection Agency (EPA), Research Triangle Park, NC, United States.
| | - J Christopher Corton
- Center for Computational Toxicology and Exposure, U.S. EPA, Research Triangle Park, NC, United States.
| |
Collapse
|
30
|
Corton JC, Hill T, Sutherland JJ, Stevens JL, Rooney J. A Set of Six Gene Expression Biomarkers Identify Rat Liver Tumorigens in Short-term Assays. Toxicol Sci 2020; 177:11-26. [PMID: 32603430 PMCID: PMC8026143 DOI: 10.1093/toxsci/kfaa101] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Chemical-induced liver cancer occurs in rodents through well-characterized adverse outcome pathways. We hypothesized that measurement of the 6 most common molecular initiating events (MIEs) in liver cancer adverse outcome pathways in short-term assays using only gene expression will allow early identification of chemicals and their associated doses that are likely to be tumorigenic in the liver in 2-year bioassays. We tested this hypothesis using transcript data from a rat liver microarray compendium consisting of 2013 comparisons of 146 chemicals administered at doses with previously established effects on rat liver tumor induction. Five MIEs were measured using previously characterized gene expression biomarkers composed of gene sets predictive for genotoxicity and activation of 1 or more xenobiotic receptors (aryl hydrocarbon receptor, constitutive activated receptor, estrogen receptor, and peroxisome proliferator-activated receptor α). Because chronic injury can be important in tumorigenesis, we also developed a biomarker for cytotoxicity that had a 96% balanced accuracy. Characterization of the genes in each biomarker set using the unsupervised TXG-MAP network model demonstrated that the genes were associated with distinct functional coexpression modules. Using the Toxicological Priority Index to rank chemicals based on their ability to activate the MIEs showed that chemicals administered at tumorigenic doses clearly gave the highest ranked scores. Balanced accuracies using thresholds derived from either TG-GATES or DrugMatrix data sets to predict tumorigenicity in independent sets of chemicals were up to 93%. These results show that a MIE-directed approach using only gene expression biomarkers could be used in short-term assays to identify chemicals and their doses that cause tumors.
Collapse
Affiliation(s)
- J Christopher Corton
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
| | - Thomas Hill
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
- Oak Ridge Institute for Science and Education (ORISE)
| | | | - James L Stevens
- Indiana Biosciences Research Institute, Indianapolis, Indiana
- Paradox Found LLC, Apex, North Carolina
| | - John Rooney
- Center for Computational Toxicology and Exposure, U.S. Environmental Protection Agency (EPA), Research Triangle Park, North Carolina
- Oak Ridge Institute for Science and Education (ORISE)
- Integrated Lab Services, Research Triangle Park, NC 27560
| |
Collapse
|
31
|
Jacobs MN, Colacci A, Corvi R, Vaccari M, Aguila MC, Corvaro M, Delrue N, Desaulniers D, Ertych N, Jacobs A, Luijten M, Madia F, Nishikawa A, Ogawa K, Ohmori K, Paparella M, Sharma AK, Vasseur P. Chemical carcinogen safety testing: OECD expert group international consensus on the development of an integrated approach for the testing and assessment of chemical non-genotoxic carcinogens. Arch Toxicol 2020; 94:2899-2923. [PMID: 32594184 PMCID: PMC7395040 DOI: 10.1007/s00204-020-02784-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 05/07/2020] [Indexed: 12/26/2022]
Abstract
While regulatory requirements for carcinogenicity testing of chemicals vary according to product sector and regulatory jurisdiction, the standard approach starts with a battery of genotoxicity tests (which include mutagenicity assays). If any of the in vivo genotoxicity tests are positive, a lifetime rodent cancer bioassay may be requested, but under most chemical regulations (except plant protection, biocides, pharmaceuticals), this is rare. The decision to conduct further testing based on genotoxicity test outcomes creates a regulatory gap for the identification of non-genotoxic carcinogens (NGTxC). With the objective of addressing this gap, in 2016, the Organization of Economic Cooperation and Development (OECD) established an expert group to develop an integrated approach to the testing and assessment (IATA) of NGTxC. Through that work, a definition of NGTxC in a regulatory context was agreed. Using the adverse outcome pathway (AOP) concept, various cancer models were developed, and overarching mechanisms and modes of action were identified. After further refining and structuring with respect to the common hallmarks of cancer and knowing that NGTxC act through a large variety of specific mechanisms, with cell proliferation commonly being a unifying element, it became evident that a panel of tests covering multiple biological traits will be needed to populate the IATA. Consequently, in addition to literature and database investigation, the OECD opened a call for relevant assays in 2018 to receive suggestions. Here, we report on the definition of NGTxC, on the development of the overarching NGTxC IATA, and on the development of ranking parameters to evaluate the assays. Ultimately the intent is to select the best scoring assays for integration in an NGTxC IATA to better identify carcinogens and reduce public health hazards.
Collapse
Affiliation(s)
- Miriam N Jacobs
- Centre for Radiation, Chemical and Environmental Hazards (CRCE), Public Health England, Chilton, UK.
| | - Annamaria Colacci
- Center for Environment, Prevention and Health, Regional Agency for Prevention, Environment and Energy Emilia Romagna Region (Arpae), Bologna, Italy
| | - Raffaella Corvi
- European Commission Joint Research Centre (EC JRC), Ispra, Italy
| | - Monica Vaccari
- Center for Environment, Prevention and Health, Regional Agency for Prevention, Environment and Energy Emilia Romagna Region (Arpae), Bologna, Italy
| | | | | | - Nathalie Delrue
- Organisation for Economic Cooperation and Development (OECD), Paris, France
| | | | - Norman Ertych
- German Centre for the Protection of Laboratory Animals (Bf3R), German Federal Institute for Risk Assessment, Diedersdorfer Weg 1, 12277, Berlin, Germany
| | - Abigail Jacobs
- US Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Mirjam Luijten
- National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Federica Madia
- European Commission Joint Research Centre (EC JRC), Ispra, Italy
| | | | - Kumiko Ogawa
- National Institute of Health Sciences, Kawasaki, Japan
| | - Kiyomi Ohmori
- Kanagawa Prefectural Institute of Public Health, Chigasaki, Japan
| | - Martin Paparella
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | | | | |
Collapse
|
32
|
Suarez-Torres JD, Jimenez-Orozco FA, Ciangherotti CE. The 2-year rodent bioassay in drug and chemical carcinogenesis testing: Sensitivity, according to the framework of carcinogenic action. Toxicol Mech Methods 2020; 30:462-475. [PMID: 32338171 DOI: 10.1080/15376516.2020.1760986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The long-term rodent bioassay (RCB) has been the gold-standard for the pre-marketing prediction of chemical and drug carcinogenicity to humans. Nonetheless, the validity of this toxicity test has remained elusive for several decades. In the quest to uncover the performance of the RCB, its sensitivity (SEN) was charted as the first step. This appraisal was based on (a) chemicals with sufficient epidemiological evidence of carcinogenicity, and (b) other substances with limited epidemiological evidence, or remarkable classifications of carcinogenicity based on mechanistic or pharmacological data. In the present study, chemicals evaluated for their carcinogenicity to humans in IARC Monographs volumes 1-123, U.S. EPA IRIS Assessments, and U.S. NTP RoC were considered. This investigation gathered additional evidence supporting that, in hazard identification, the RCB is unwarranted for mutagenic or direct-acting genotoxicants. However, for purposes of risk assessment or management, the RCB might be justified whenever there is a lack of reliable and/or comprehensive epidemiological data. The RCB exhibited a significantly different SEN for threshold-based human carcinogens compared to non-threshold-based ones. With threshold-based chemicals, to increase the SEN of the testing from 80% (rat-RCB) to 90%, the 2-species RCB might be warranted. Nevertheless, the resolve would depend on the viewpoint, and on the future analysis of the overall performance of the RCB. In terms of SEN, and cancer hazard identification, the comparison between the RCB and alternative methods (e.g. rasH2 mouse, Tg.AC mouse) is now enabled.
Collapse
Affiliation(s)
- Jose D Suarez-Torres
- Department of Pharmacy, Universidad Nacional de Colombia, Bogotá, Colombia.,Department of Toxicology, Universidad Nacional de Colombia, Bogotá, Colombia.,Institute of Pharmaceutical Research, School of Pharmacy, Universidad Central de Venezuela, Caracas, Venezuela
| | - Fausto A Jimenez-Orozco
- Department of Pharmacology, Faculty of Medicine, Universidad Nacional Autónoma de México, Ciudad de Mexico, Mexico
| | - Carlos E Ciangherotti
- Institute of Pharmaceutical Research, School of Pharmacy, Universidad Central de Venezuela, Caracas, Venezuela.,Laboratory of Neuropeptides, School of Pharmacy, Universidad Central de Venezuela, Caracas, Venezuela
| |
Collapse
|
33
|
Bogdanffy MS, Lesniak J, Mangipudy R, Sistare FD, Colman K, Garcia-Tapia D, Monticello T, Blanset D. Tg.rasH2 Mouse Model for Assessing Carcinogenic Potential of Pharmaceuticals: Industry Survey of Current Practices. Int J Toxicol 2020; 39:198-206. [DOI: 10.1177/1091581820919896] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The Tg.rasH2 mouse was developed as an alternative model to the traditional 2-year mouse bioassay for pharmaceutical carcinogenicity testing. This model has found extensive use in support of pharmaceutical drug development over the last few decades. It has the potential to improve quality and timeliness, reduce animal usage, and in some instances allow expedient decision-making regarding the human carcinogenicity potential of a drug candidate. Despite the increased use of the Tg.rasH2 model, there has been no systematic survey of current practices in the design, interpretation of results from the bioassay, and global health authority perspectives. Therefore, the aim of this work was to poll the pharmaceutical industry on study design practices used in the dose range finding and definitive 6-month studies and on results relative to the ongoing negotiations to revise The International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use S1 Guidance. Twenty-two member companies of International Consortium for Innovation and Quality in Pharmaceutical Development DruSafe Leadership Group participated in the survey, sharing experiences from studies conducted with 55 test compounds between 2010 and 2018. The survey results provide very useful insights into study design and interpretation. Importantly, the results identified several key opportunities for reducing animal use and increasing the value of testing for potential human carcinogenicity using this model. Recommended changes to study designs that would reduce animal usage include eliminating the requirement to include positive control groups in every study, use of nontransgenic wild-type littermates in the dose range finding study, and use of microsampling to reduce or eliminate satellite groups for toxicokinetics.
Collapse
Affiliation(s)
- Matthew S. Bogdanffy
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | | | | | | | - Karyn Colman
- Novartis Institutes for BioMedical Research, One Health Plaza, East Hanover, NJ, USA
| | - David Garcia-Tapia
- Toxicology, Drug Disposition &PK/PD Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Thomas Monticello
- Translational Safety and Bioanalytical Sciences, Amgen Research, Thousand Oaks, CA, USA
| | - Diann Blanset
- Nonclinical Drug Safety, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| |
Collapse
|
34
|
Gooderham NJ, Cohen SM, Eisenbrand G, Fukushima S, Guengerich FP, Hecht SS, Rietjens IMCM, Rosol TJ, Bastaki M, Linman MJ, Taylor SV. The safety evaluation of food flavoring substances: the role of genotoxicity studies. Crit Rev Toxicol 2020; 50:1-27. [PMID: 32162576 DOI: 10.1080/10408444.2020.1712589] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Flavor and Extract Manufacturers Association (FEMA) Expert Panel relies on the weight of evidence from all available data in the safety evaluation of flavoring substances. This process includes data from genotoxicity studies designed to assess the potential of a chemical agent to react with DNA or otherwise cause changes to DNA, either in vitro or in vivo. The Panel has reviewed a large number of in vitro and in vivo genotoxicity studies during the course of its ongoing safety evaluations of flavorings. The adherence of genotoxicity studies to standardized protocols and guidelines, the biological relevance of the results from those studies, and the human relevance of these studies are all important considerations in assessing whether the results raise specific concerns for genotoxic potential. The Panel evaluates genotoxicity studies not only for evidence of genotoxicity hazard, but also for the probability of risk to the consumer in the context of exposure from their use as flavoring substances. The majority of flavoring substances have given no indication of genotoxic potential in studies evaluated by the FEMA Expert Panel. Examples illustrating the assessment of genotoxicity data for flavoring substances and the consideration of the factors noted above are provided. The weight of evidence approach adopted by the FEMA Expert Panel leads to a rational assessment of risk associated with consumer intake of flavoring substances under the conditions of use.
Collapse
Affiliation(s)
| | - Samuel M Cohen
- Havlik-Wall Professor of Oncology, Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Gerhard Eisenbrand
- Food Chemistry & Toxicology, University of Kaiserslautern (retired), Heidelberg, Germany
| | | | - F Peter Guengerich
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Stephen S Hecht
- Masonic Cancer Center and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | | | - Thomas J Rosol
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Maria Bastaki
- Flavor and Extract Manufacturers Association, Washington, DC, USA
| | - Matthew J Linman
- Flavor and Extract Manufacturers Association, Washington, DC, USA
| | - Sean V Taylor
- Flavor and Extract Manufacturers Association, Washington, DC, USA
| |
Collapse
|
35
|
Yauk CL, Harrill AH, Ellinger-Ziegelbauer H, van der Laan JW, Moggs J, Froetschl R, Sistare F, Pettit S. A cross-sector call to improve carcinogenicity risk assessment through use of genomic methodologies. Regul Toxicol Pharmacol 2020; 110:104526. [PMID: 31726190 PMCID: PMC7891877 DOI: 10.1016/j.yrtph.2019.104526] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/25/2019] [Accepted: 11/08/2019] [Indexed: 12/30/2022]
Abstract
Robust genomic approaches are now available to realize improvements in efficiencies and translational relevance of cancer risk assessments for drugs and chemicals. Mechanistic and pathway data generated via genomics provide opportunities to advance beyond historical reliance on apical endpoints of uncertain human relevance. Published research and regulatory evaluations include many examples for which genomic data have been applied to address cancer risk assessment as a health protection endpoint. The alignment of mature, robust, reproducible, and affordable technologies with increasing demands for reduced animal testing sets the stage for this important transition. We present our shared vision for change from leading scientists from academic, government, nonprofit, and industrial sectors and chemical and pharmaceutical safety applications. This call to action builds upon a 2017 workshop on "Advances and Roadblocks for Use of Genomics in Cancer Risk Assessment." The authors propose a path for implementation of innovative cancer risk assessment including incorporating genomic signatures to assess mechanistic relevance of carcinogenicity and enhanced use of genomics in benchmark dose and point of departure evaluations. Novel opportunities for the chemical and pharmaceutical sectors to combine expertise, resources, and objectives to achieve a common goal of improved human health protection are identified.
Collapse
Affiliation(s)
| | - Alison H Harrill
- National Institute of Environmental Health Sciences, Research Triangle, Park, NC, 27709, USA.
| | | | | | | | - Roland Froetschl
- BfArM Federal Institute for Drugs and Medical Devices, Bonn, Germany
| | | | - Syril Pettit
- Health and Environmental Sciences Institute, Washington, DC, USA
| |
Collapse
|
36
|
Harris KL, Myers MB, McKim KL, Elespuru RK, Parsons BL. Rationale and Roadmap for Developing Panels of Hotspot Cancer Driver Gene Mutations as Biomarkers of Cancer Risk. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2020; 61:152-175. [PMID: 31469467 PMCID: PMC6973253 DOI: 10.1002/em.22326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 05/24/2023]
Abstract
Cancer driver mutations (CDMs) are necessary and causal for carcinogenesis and have advantages as reporters of carcinogenic risk. However, little progress has been made toward developing measurements of CDMs as biomarkers for use in cancer risk assessment. Impediments for using a CDM-based metric to inform cancer risk include the complexity and stochastic nature of carcinogenesis, technical difficulty in quantifying low-frequency CDMs, and lack of established relationships between cancer driver mutant fractions and tumor incidence. Through literature review and database analyses, this review identifies the most promising targets to investigate as biomarkers of cancer risk. Mutational hotspots were discerned within the 20 most mutated genes across the 10 deadliest cancers. Forty genes were identified that encompass 108 mutational hotspot codons overrepresented in the COSMIC database; 424 different mutations within these hotspot codons account for approximately 63,000 tumors and their prevalence across tumor types is described. The review summarizes literature on the prevalence of CDMs in normal tissues and suggests such mutations are direct and indirect substrates for chemical carcinogenesis, which occurs in a spatially stochastic manner. Evidence that hotspot CDMs (hCDMs) frequently occur as tumor subpopulations is presented, indicating COSMIC data may underestimate mutation prevalence. Analyses of online databases show that genes containing hCDMs are enriched in functions related to intercellular communication. In its totality, the review provides a roadmap for the development of tissue-specific, CDM-based biomarkers of carcinogenic potential, comprised of batteries of hCDMs and can be measured by error-correct next-generation sequencing. Environ. Mol. Mutagen. 61:152-175, 2020. Published 2019. This article is a U.S. Government work and is in the public domain in the USA. Environmental and Molecular Mutagenesis published by Wiley Periodicals, Inc. on behalf of Environmental Mutagen Society.
Collapse
Affiliation(s)
- Kelly L. Harris
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| | - Meagan B. Myers
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| | - Karen L. McKim
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| | - Rosalie K. Elespuru
- Division of Biology, Chemistry and Materials ScienceCDRH/OSEL, US Food and Drug AdministrationSilver SpringMaryland
| | - Barbara L. Parsons
- Division of Genetic and Molecular ToxicologyNational Center for Toxicological Research, US Food and Drug AdministrationJeffersonArkansas
| |
Collapse
|
37
|
Sacaan A, Thibault S, Khan KN. Central nervous system tumors in 2-year rat carcinogenicity studies: perspectives on human risk assessment. J Toxicol Sci 2019; 44:643-655. [PMID: 31588056 DOI: 10.2131/jts.44.643] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Rodent in vivo carcinogenicity bioassays are required for human risk assessment and have been utilized in this capacity for decades. Accordingly, there is an abundance of data that could be accessed and analyzed to better understand the translatability of xenobiotic-induced rodent tumors to human risk assessment. In the past decade, various groups have published assessments of the value garnered by these life-time rodent studies. Results and recommendations from the International Council for Harmonization Expert Working Group (ICH-S1 EWG) on the predictability of the current testing paradigm and proposal for an integrated approach to human carcinogenicity risk assessment are pending. Central nervous system (CNS) tumors in rats are rare and translatability to human remains unknown. This review focuses on microglial cell tumors (MCT) of the CNS in rats including its classification, nomenclature, incidence and translatability to human risk assessment. Based on emerging immunohistochemistry (IHC) characterization, glial tumors previously thought of astrocytic origin are more likely MCTs. These may be considered rodent specific and glucose dysregulation may be one component contributing to their formation. Based on review of the literature, MCTs are rarely diagnosed in humans, thus this tumor type may be rat-specific. We propose to include MCTs as a tumor type in revised International Harmonization of Nomenclature and Diagnostic Criteria (INHAND) classification and all glial tumors to be classified as MCTs unless proven otherwise by IHC.
Collapse
Affiliation(s)
- Aida Sacaan
- Pfizer Inc. Drug Safety Research and Development
| | | | - K Nasir Khan
- Pfizer Inc. Drug Safety Research and Development
| |
Collapse
|
38
|
Laube B, Michaelsen S, Meischner V, Hartwig A, Epe B, Schwarz M. Classification or non-classification of substances with positive tumor findings in animal studies: Guidance by the German MAK commission. Regul Toxicol Pharmacol 2019; 108:104444. [PMID: 31433998 DOI: 10.1016/j.yrtph.2019.104444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/10/2019] [Accepted: 08/14/2019] [Indexed: 12/19/2022]
Abstract
One of the important tasks of the German Senate Commission for the Investigation of Health Hazards of Chemical Compounds in the Work Area (known as the MAK Commission) is in the evaluation of a potential for carcinogenicity of hazardous substances at the workplace. Often, this evaluation is critically based on data on carcinogenic responses seen in animal studies and, if positive tumor responses have been observed, this will mostly lead to a classification of the substance under investigation into one of the classes for carcinogens. However, there are cases where it can be demonstrated with a very high degree of confidence that the tumor findings in the experimental animals are not relevant for humans at the workplace and, therefore, the MAK Commission will not classify the respective substance into one of the classes for carcinogens. This paper will summarize the general criteria used by the MAK Commission for the categorization into "carcinogen" and "non-carcinogen" and compare this procedure with those used by other national and international organizations.
Collapse
Affiliation(s)
- Britta Laube
- Scientific Secretariat of the Senate Commission on the Investigation of Health Hazards of Chemical Compounds in the Work Area (MAK Commission), Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Sandra Michaelsen
- Scientific Secretariat of the Senate Commission on the Investigation of Health Hazards of Chemical Compounds in the Work Area (MAK Commission), Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Veronika Meischner
- Scientific Secretariat of the Senate Commission on the Investigation of Health Hazards of Chemical Compounds in the Work Area (MAK Commission), Institute for Applied Biosciences, Karlsruhe Institute of Technology (KIT), Department of Food Chemistry and Toxicology, Karlsruhe, Germany
| | - Andrea Hartwig
- Food Chemistry and Toxicology, Institute of Applied Bioscience, Karlsruhe Institute of Technology (KIT), Adenauerring 20a, 76131, Karlsruhe, Germany
| | - Bernd Epe
- Institute of Pharmacy and Biochemistry, University of Mainz, Staudingerweg 5, D-55099, Mainz, Germany
| | - Michael Schwarz
- Dept. of Experimental and Clinical Pharmacology and Toxicology, Dept. Toxicology, Eberhard Karls University, Wilhelmstr. 56, 72074, Tübingen, Germany.
| |
Collapse
|
39
|
Andrews PA, McNerney ME, DeGeorge JJ. Exposure assessments in reproductive and developmental toxicity testing: An IQ-DruSafe industry survey on current practices and experiences in support of exposure-based high dose selection. Regul Toxicol Pharmacol 2019; 107:104413. [PMID: 31229519 DOI: 10.1016/j.yrtph.2019.104413] [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: 05/08/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 11/17/2022]
Abstract
The draft ICH S5(R3) guideline includes an exposure-based endpoint as an option for selecting the high dose in developmental and reproductive toxicity (DART) studies. In 2016, IQ DruSafe conducted an anonymous survey to identify industry practices and experiences related to pharmacokinetic assessments in DART studies in order to facilitate a pragmatic data-driven approach to development of an acceptable multiple of the clinical exposure to be proposed for dose selection in the guideline. Questions in the survey were designed to explore pharmacokinetic differences in pregnant versus non-pregnant animals, and to assess exposure levels attained in the absence of maternal toxicity as well as DART outcomes in animal studies associated with those exposures. Small molecule and therapeutic proteins were analyzed separately. The key findings for small molecules were: a) differences in exposures between pregnant and non-pregnant animals were generally ≤3-fold, b) Cmax or AUC exposures ≥25-fold the clinical exposure were achieved in the absence of maternal toxicity for 31% and 23% of rat and rabbit developmental toxicity studies, respectively, and c) only 3.3% (5/153) and 1.6% (2/128) of the developmental toxicity studies were positive for malformations or embryofetal lethality in rats and rabbits, respectively, that were not observed until exposure margins were ≥25-fold.
Collapse
|
40
|
Qin C, Aslamkhan AG, Pearson K, Tanis KQ, Podtelezhnikov A, Frank E, Pacchione S, Pippert T, Glaab WE, Sistare FD. AhR Activation in Pharmaceutical Development: Applying Liver Gene Expression Biomarker Thresholds to Identify Doses Associated with Tumorigenic Risks in Rats. Toxicol Sci 2019; 171:46-55. [PMID: 31127949 DOI: 10.1093/toxsci/kfz125] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/16/2019] [Accepted: 05/18/2019] [Indexed: 12/13/2022] Open
Abstract
Aryl hydrocarbon receptor (AhR) activation is associated with carcinogenicity of non-genotoxic AhR-activating carcinogens such as 2, 3, 7, 8-tetrachlorodibenzodioxin (TCDD), and is often observed with drug candidate molecules in development and raises safety concerns. As downstream effectors of AhR signaling, the expression and activity of Cyp1a1 and Cyp1a2 genes are commonly monitored as evidence of AhR activation to inform carcinogenic risk of compounds in question. However, many marketed drugs and phytochemicals are reported to induce these Cyps modestly and are not associated with dioxin-like toxicity or carcinogenicity. We hypothesized that a threshold of AhR activation needs to be surpassed in a sustained manner in order for the dioxin-like toxicity to manifest, and a simple liver gene expression signature based on Cyp1a1 and Cyp1a2 from a short-term rat study could be used to assess AhR activation strength and differentiate tumorigenic dose levels from non-tumorigenic ones. To test this hypothesis, short term studies were conducted in Wistar Han rats with two AhR-activating carcinogens (TCDD and PCB126) at minimally carcinogenic and non-carcinogenic dose levels, and three AhR-activating non-carcinogens (omeprazole, mexiletine, and canagliflozin) at the top doses used in their reported 2-year rat carcinogenicity studies. A threshold of AhR activation was identified in rat liver that separated a meaningful "tumorigenic-strength AhR signal" from a statistically-significant AhR activation signal that was not associated with dioxin-like carcinogenicity. These studies also confirmed the importance of the sustainability of AhR activation for carcinogenic potential. A sustained activation of AhR above the threshold could thus be used in early pharmaceutical development to identify dose levels of drug candidates expected to exhibit dioxin-like carcinogenic potential.
Collapse
Affiliation(s)
- Chunhua Qin
- Department of Safety Assessment and Laboratory Animal Resources, Merck & Co. Inc., West Point, PA, USA
| | - Amy G Aslamkhan
- Department of Safety Assessment and Laboratory Animal Resources, Merck & Co. Inc., West Point, PA, USA
| | - Kara Pearson
- Department of Safety Assessment and Laboratory Animal Resources, Merck & Co. Inc., West Point, PA, USA
| | - Keith Q Tanis
- Department of Human Genetics and Pharmacogenomics, Merck & Co. Inc., West Point, PA, USA
| | - Alexei Podtelezhnikov
- Department of Human Genetics and Pharmacogenomics, Merck & Co. Inc., West Point, PA, USA
| | - Erika Frank
- Department of Safety Assessment and Laboratory Animal Resources, Merck & Co. Inc., West Point, PA, USA
| | - Stephen Pacchione
- Department of Safety Assessment and Laboratory Animal Resources, Merck & Co. Inc., West Point, PA, USA
| | - Todd Pippert
- Department of Safety Assessment and Laboratory Animal Resources, Merck & Co. Inc., West Point, PA, USA
| | - Warren E Glaab
- Department of Safety Assessment and Laboratory Animal Resources, Merck & Co. Inc., West Point, PA, USA
| | - Frank D Sistare
- Department of Safety Assessment and Laboratory Animal Resources, Merck & Co. Inc., West Point, PA, USA
| |
Collapse
|
41
|
Cohen SM, Boobis AR, Dellarco VL, Doe JE, Fenner-Crisp PA, Moretto A, Pastoor TP, Schoeny RS, Seed JG, Wolf DC. Chemical carcinogenicity revisited 3: Risk assessment of carcinogenic potential based on the current state of knowledge of carcinogenesis in humans. Regul Toxicol Pharmacol 2019; 103:100-105. [DOI: 10.1016/j.yrtph.2019.01.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 01/27/2023]
|
42
|
Kobets T, Iatropoulos MJ, Williams GM. Mechanisms of DNA-reactive and epigenetic chemical carcinogens: applications to carcinogenicity testing and risk assessment. Toxicol Res (Camb) 2019; 8:123-145. [PMID: 30997017 PMCID: PMC6417487 DOI: 10.1039/c8tx00250a] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 12/18/2018] [Indexed: 01/03/2023] Open
Abstract
Chemicals with carcinogenic activity in either animals or humans produce increases in neoplasia through diverse mechanisms. One mechanism is reaction with nuclear DNA. Other mechanisms consist of epigenetic effects involving either modifications of regulatory macromolecules or perturbation of cellular regulatory processes. The basis for distinguishing between carcinogens that have either DNA reactivity or an epigenetic activity as their primary mechanism of action is detailed in this review. In addition, important applications of information on these mechanisms of action to carcinogenicity testing and human risk assessment are discussed.
Collapse
Affiliation(s)
- Tetyana Kobets
- Department of Pathology , New York Medical College , Valhalla , NY 10595 , USA . ; ; Tel: +1 914-594-3105
| | - Michael J Iatropoulos
- Department of Pathology , New York Medical College , Valhalla , NY 10595 , USA . ; ; Tel: +1 914-594-3105
| | - Gary M Williams
- Department of Pathology , New York Medical College , Valhalla , NY 10595 , USA . ; ; Tel: +1 914-594-3105
| |
Collapse
|
43
|
Rooney J, Hill T, Qin C, Sistare FD, Corton JC. Adverse outcome pathway-driven identification of rat liver tumorigens in short-term assays. Toxicol Appl Pharmacol 2018; 356:99-113. [DOI: 10.1016/j.taap.2018.07.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/12/2018] [Accepted: 07/20/2018] [Indexed: 02/07/2023]
|
44
|
Dal Negro G, Eskes C, Belz S, Bertein C, Chlebus M, Corvaro M, Corvi R, Dhalluin S, Halder M, Harvey J, Hermann M, Hoffmann-Dörr S, Kilian K, Lambrigts D, Laroche C, Louhimies S, Mahony C, Manou I, McNamee P, Prieto P, Reid K, Roggen E, Schutte K, Stirling C, Uhlrich S, Weissenhorn R, Whelan M. One science-driven approach for the regulatory implementation of alternative methods: A multi-sector perspective. Regul Toxicol Pharmacol 2018; 99:33-49. [PMID: 30098372 DOI: 10.1016/j.yrtph.2018.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/05/2018] [Indexed: 12/28/2022]
Abstract
EU regulations call for the use of alternative methods to animal testing. During the last decade, an increasing number of alternative approaches have been formally adopted. In parallel, new 3Rs-relevant technologies and mechanistic approaches have increasingly contributed to hazard identification and risk assessment evolution. In this changing landscape, an EPAA meeting reviewed the challenges that different industry sectors face in the implementation of alternative methods following a science-driven approach. Although clear progress was acknowledged in animal testing reduction and refinement thanks to an integration of scientifically robust approaches, the following challenges were identified: i) further characterization of toxicity pathways; ii) development of assays covering current scientific gaps, iii) better characterization of links between in vitro readouts and outcome in the target species; iv) better definition of alternative method applicability domains, and v) appropriate implementation of the available approaches. For areas having regulatory adopted alternative methods (e.g., vaccine batch testing), harmonised acceptance across geographical regions was considered critical for broader application. Overall, the main constraints to the application of non-animal alternatives are the still existing gaps in scientific knowledge and technological limitations. The science-driven identification of most appropriate methods is key for furthering a multi-sectorial decrease in animal testing.
Collapse
Affiliation(s)
- Gianni Dal Negro
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, United Kingdom
| | - Chantra Eskes
- SeCAM Services and Consultation on Alternative Methods, Via Campagnora 1, 6983, Magliaso, Switzerland.
| | - Susanne Belz
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| | | | - Magda Chlebus
- European Federation of Pharmaceutical Industries and Associations (EFPIA), Rue du Trône 108, 1050, Brussels, Belgium
| | - Marco Corvaro
- ECPA - the European Crop Protection Association, 6 Avenue E. Van Nieuwenhuyse, 1160, Brussels, Belgium
| | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| | - Stephane Dhalluin
- L'Oréal Research & Innovation, 9 rue Pierre Dreyfus, 92110, Clichy, France
| | - Marlies Halder
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| | - Jim Harvey
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, United Kingdom
| | - Martina Hermann
- Henkel AG & Co. KGaA, Henkelstr. 67, 40589, Duesseldorf, Germany
| | | | - Karin Kilian
- European Commission, Directorate General for the Environment (DG ENV), Brussels, Belgium
| | - Denis Lambrigts
- GlaxoSmithKline Vaccines, 20 Avenue Fleming, 1300, Wavre, Belgium
| | - Charles Laroche
- European Partnership for Alternative Approaches to Animal Testing (EPAA), Av. Herrmann-Debroux 40, 1160, Brussels, Belgium
| | - Susanna Louhimies
- European Commission, Directorate General for the Environment (DG ENV), Brussels, Belgium
| | - Catherine Mahony
- The Procter & Gamble Company, Whitehall Lane, Egham, Surrey TW20 9NW, United Kingdom
| | - Irene Manou
- European Partnership for Alternative Approaches to Animal Testing (EPAA), Av. Herrmann-Debroux 40, 1160, Brussels, Belgium
| | - Pauline McNamee
- The Procter & Gamble Company, Whitehall Lane, Egham, Surrey TW20 9NW, United Kingdom
| | - Pilar Prieto
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| | - Kirsty Reid
- European Federation of Pharmaceutical Industries and Associations (EFPIA), Rue du Trône 108, 1050, Brussels, Belgium
| | - Erwin Roggen
- 3Rs Management and Consulting, Asavænget 14, 2800, Kongens Lyngby, Denmark
| | - Katrin Schutte
- European Commission, Directorate General for the Environment (DG ENV), Brussels, Belgium
| | | | - Sylvie Uhlrich
- Sanofi Pasteur, 1541 Av. Marcel Merieux, 69280, Marcy l'Etoile, France
| | - Renate Weissenhorn
- European Commission, Directorate General for Internal Market, Industry, Enterpreneurship and SME, Brussels, Belgium
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), Via E. Fermi 2749, 21017, Ispra, Italy
| |
Collapse
|
45
|
Goodman JI. Goodbye to the bioassay. Toxicol Res (Camb) 2018; 7:558-564. [PMID: 30090606 PMCID: PMC6062362 DOI: 10.1039/c8tx00004b] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 01/31/2018] [Indexed: 12/30/2022] Open
Abstract
It is time to say goodbye to the standard two-year rodent bioassay. While a few, primarily genotoxic, compounds which are clearly associated with human cancer test positive in the bioassay, there is no science-based, sound foundation for presuming it provides either a valid broad (across different chemicals) capability for discerning potential human carcinogens or a valid starting point for making human risk assessment decisions. The two basic assumptions underlying the bioassay are: (1) rodent carcinogens are human carcinogens; and (2) results obtained at high doses are indicative of results that will occur at lower, environmentally relevant, doses. Both of these assumptions are not correct. Furthermore, a reevaluation of National Toxicology Program bioassay data has revealed that if the dose group size were increased from 50 to 200 rodents per group the number of bioassays deemed to be positive would increase from approximately 50% to very close to 100%. Thus, under the extreme conditions of the bioassay (e.g., high doses, lifetime exposure and, at times, a non-physiological route of administration) virtually all chemicals tested could be made into rodent carcinogens. In recent years there have been a number of proposals to move away from the standard bioassay. In particular, a recently formulated decision tree (Cohen, 2017), which places an emphasis on dose-response relationships and invites the use of MOA information, provides a sound basis for moving on from the bioassay and towards a rational approach to both identify chemicals which appear to have the potential to cause cancer in humans and take dose-response relationships into consideration in order to place the extent, if any, of the risk they might pose into proper perspective.
Collapse
Affiliation(s)
- Jay I Goodman
- Department of Pharmacology and Toxicology , Michigan State University , East Lansing , MI 48824 , USA . ; Tel: +1-517-353-9346
| |
Collapse
|
46
|
McMullen PD, Andersen ME, Cholewa B, Clewell HJ, Dunnick KM, Hartman JK, Mansouri K, Minto MS, Nicolas CI, Phillips MB, Slattery S, Yoon M, Clewell RA. Evaluating opportunities for advancing the use of alternative methods in risk assessment through the development of fit-for-purpose in vitro assays. Toxicol In Vitro 2018; 48:310-317. [PMID: 29391263 DOI: 10.1016/j.tiv.2018.01.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/27/2017] [Accepted: 01/29/2018] [Indexed: 12/11/2022]
Abstract
An evolving regulatory, scientific, and legislative landscape is driving a fundamental change in how chemical safety decisions are made. As we move to implement changes, regulatory agencies and industry are beginning to adopt tiered approaches, which leverage high-throughput screening technologies for prioritization and read across, followed by interrogation of "hit chemicals" with more rigorous dose-response assessment either in fit-for-purpose human cell-based assays or with traditional in vivo tests. However, to date, suitable in vitro alternatives do not exist for the vast majority of the organ toxicities that form the basis of current regulatory decisions. To successfully support safety decisions, biologically relevant, quantitative, cell-based assays that evaluate dose-response and identify regions of safety for chemical exposure are required. This review evaluates the current state of the science in the development of such assays, identifies key gaps in the current tests, and recommends areas where research efforts may be focused to help move the risk assessment community towards more wide-spread use of in vitro methods. Our analysis suggests that a key shortcoming in the current efforts is the ability to test volatile compounds and to predict pulmonary toxicity. We present a mechanistically-based path forward for the development of a fit-for-purpose lung toxicity assay.
Collapse
Affiliation(s)
| | | | - Brian Cholewa
- ScitoVation, LLC., Research Triangle Park, NC 27709, United States
| | - Harvey J Clewell
- ScitoVation, LLC., Research Triangle Park, NC 27709, United States
| | | | | | - Kamel Mansouri
- ScitoVation, LLC., Research Triangle Park, NC 27709, United States
| | - Melyssa S Minto
- ScitoVation, LLC., Research Triangle Park, NC 27709, United States
| | | | | | - Scott Slattery
- ScitoVation, LLC., Research Triangle Park, NC 27709, United States
| | - Miyoung Yoon
- ScitoVation, LLC., Research Triangle Park, NC 27709, United States
| | | |
Collapse
|
47
|
Smirnova L, Kleinstreuer N, Corvi R, Levchenko A, Fitzpatrick SC, Hartung T. 3S - Systematic, systemic, and systems biology and toxicology. ALTEX 2018; 35:139-162. [PMID: 29677694 PMCID: PMC6696989 DOI: 10.14573/altex.1804051] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 04/06/2018] [Indexed: 12/11/2022]
Abstract
A biological system is more than the sum of its parts - it accomplishes many functions via synergy. Deconstructing the system down to the molecular mechanism level necessitates the complement of reconstructing functions on all levels, i.e., in our conceptualization of biology and its perturbations, our experimental models and computer modelling. Toxicology contains the somewhat arbitrary subclass "systemic toxicities"; however, there is no relevant toxic insult or general disease that is not systemic. At least inflammation and repair are involved that require coordinated signaling mechanisms across the organism. However, the more body components involved, the greater the challenge to reca-pitulate such toxicities using non-animal models. Here, the shortcomings of current systemic testing and the development of alternative approaches are summarized. We argue that we need a systematic approach to integrating existing knowledge as exemplified by systematic reviews and other evidence-based approaches. Such knowledge can guide us in modelling these systems using bioengineering and virtual computer models, i.e., via systems biology or systems toxicology approaches. Experimental multi-organ-on-chip and microphysiological systems (MPS) provide a more physiological view of the organism, facilitating more comprehensive coverage of systemic toxicities, i.e., the perturbation on organism level, without using substitute organisms (animals). The next challenge is to establish disease models, i.e., micropathophysiological systems (MPPS), to expand their utility to encompass biomedicine. Combining computational and experimental systems approaches and the chal-lenges of validating them are discussed. The suggested 3S approach promises to leverage 21st century technology and systematic thinking to achieve a paradigm change in studying systemic effects.
Collapse
Affiliation(s)
- Lena Smirnova
- Johns Hopkins University, Bloomberg School of Public Health, Center for Alternatives to Animal Testing (CAAT), Baltimore, MD, USA
| | | | - Raffaella Corvi
- European Commission, Joint Research Centre (JRC), EU Reference Laboratory for Alternatives to Animal Testing (EURL ECVAM), Ispra, (VA), Italy
| | - Andre Levchenko
- Yale Systems Biology Institute and Biomedical Engineering Department, Yale University, New Haven, CT, USA
| | - Suzanne C Fitzpatrick
- Food and Drug Administration (FDA), Center for Food Safety and Applied Nutrition, College Park, MD, USA
| | - Thomas Hartung
- Johns Hopkins University, Bloomberg School of Public Health, Center for Alternatives to Animal Testing (CAAT), Baltimore, MD, USA.
- CAAT-Europe, University of Konstanz, Konstanz, Germany
| |
Collapse
|
48
|
Bachtarzi H. Ex vivo and in vivo genome editing: a regulatory scientific framework from early development to clinical implementation. Regen Med 2017; 12:1015-1030. [PMID: 29243558 DOI: 10.2217/rme-2017-0095] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recent advances in human genome science have paved the way to a new class of human gene therapies based on gene editing, with the potential to provide a long-lasting curative strategy for many debilitating and complex disorders, for which there is an unmet medical need. Therapeutic genome editing encompasses both ex vivo and in vivo gene correction modalities, for which similar and also application-specific considerations apply, which dictate the overall strategy to be followed from a scientific, clinical and regulatory perspective. Here, the major regulatory barriers to successful clinical implementation are discussed, together with the key issues to be considered for generating safe (minimizing risks of tumorigenesis and off-target effects) and effective gene editing-based medicines for application in regenerative medicine.
Collapse
|
49
|
Modernizing Human Cancer Risk Assessment of Therapeutics. Trends Pharmacol Sci 2017; 39:232-247. [PMID: 29242029 DOI: 10.1016/j.tips.2017.11.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 12/14/2022]
Abstract
Cancer risk assessment of therapeutics is plagued by poor translatability of rodent models of carcinogenesis. In order to overcome this fundamental limitation, new approaches are needed that enable us to evaluate cancer risk directly in humans and human-based cellular models. Our enhanced understanding of the mechanisms of carcinogenesis and the influence of human genome sequence variation on cancer risk motivates us to re-evaluate how we assess the carcinogenic risk of therapeutics. This review will highlight new opportunities for applying this knowledge to the development of a battery of human-based in vitro models and biomarkers for assessing cancer risk of novel therapeutics.
Collapse
|
50
|
Corvi R, Madia F, Guyton KZ, Kasper P, Rudel R, Colacci A, Kleinjans J, Jennings P. Moving forward in carcinogenicity assessment: Report of an EURL ECVAM/ESTIV workshop. Toxicol In Vitro 2017; 45:278-286. [PMID: 28911985 PMCID: PMC5735222 DOI: 10.1016/j.tiv.2017.09.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/08/2017] [Accepted: 09/10/2017] [Indexed: 10/27/2022]
Abstract
There is an increased need to develop novel alternative approaches to the two-year rodent bioassay for the carcinogenicity assessment of substances where the rodent bioassay is still a basic requirement, as well as for those substances where animal use is banned or limited or where information gaps are identified within legislation. The current progress in this area was addressed in a EURL ECVAM- ESTIV workshop held in October 2016, in Juan les Pins. A number of initiatives were presented and discussed, including data-driven, technology-driven and pathway-driven approaches. Despite a seemingly diverse range of strategic developments, commonalities are emerging. For example, providing insight into carcinogenicity mechanisms is becoming an increasingly appreciated aspect of hazard assessment and is suggested to be the best strategy to drive new developments. Thus, now more than ever, there is a need to combine and focus efforts towards the integration of available information between sectors. Such cross-sectorial harmonisation will aid in building confidence in new approach methods leading to increased implementation and thus a decreased necessity for the two-year rodent bioassay.
Collapse
Affiliation(s)
- Raffaella Corvi
- European Commission, Joint Research Centre (JRC), EU Reference Laboratory for Alternatives to Animal Testing (EURL ECVAM), Ispra, (VA), Italy.
| | - Federica Madia
- European Commission, Joint Research Centre (JRC), EU Reference Laboratory for Alternatives to Animal Testing (EURL ECVAM), Ispra, (VA), Italy
| | - Kathryn Z Guyton
- Monographs Programme, International Agency for Research on Cancer, Lyon, France
| | - Peter Kasper
- Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | | | - Annamaria Colacci
- Centre for Environmental Toxicology and Risk Assessment, Environmental Protection and Health Prevention Agency, Emilia Romagna Region, Italy
| | - Jos Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, The Netherlands
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, HZ Amsterdam, The Netherlands
| |
Collapse
|