1
|
Chaturvedi S, Sonawane A. Recapitulating the potential contribution of protein S-palmitoylation in cancer. Cancer Metastasis Rev 2024; 44:20. [PMID: 39725785 DOI: 10.1007/s10555-024-10217-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/14/2024] [Indexed: 12/28/2024]
Abstract
Protein S-palmitoylation is a reversible form of protein lipidation in which the formation of a thioester bond occurs between a cysteine (Cys) residue of a protein and a 16-carbon fatty acid chain. This modification is catalyzed by a family of palmitoyl acyl transferases, the DHHC enzymes, so called because of their Asp-His-His-Cys (DHHC) catalytic motif. Deregulation of DHHC enzymes has been linked to various diseases, including cancer and infections. Cancer, a major cause of global mortality, is characterized by features like uncontrolled cell growth, resistance to cell death, angiogenesis, invasion, and metastasis. Several of these processes are controlled by DHHC-mediated S-palmitoylation of oncogenes or tumor suppressors, including growth factor receptors (e.g., EGFR), kinases (e.g., AKT), and transcription factors (e.g., β-catenin). Dynamic regulation of S-palmitoylation is also governed by protein depalmitoylases. These enzymes balance the cycling of palmitoylation and regulate cellular signaling, cell growth, and its organization. Given the significance of S-palmitoylation in cancer, the DHHCs and protein depalmitoylases are promising targets for cancer therapy. Here we summarize the catalytic mechanisms of DHHC enzymes and depalmitoylases, their role in cancer progression and prevention, as well as the crosstalk of palmitoylation with other post-translational modifications. Additionally, we discuss the methods to detect S-palmitoylation, the limitations of available DHHC-targeting inhibitors, and ongoing research efforts to address these obstacles.
Collapse
Affiliation(s)
- Suchi Chaturvedi
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, 453552, Simrol, Madhya Pradesh, India
| | - Avinash Sonawane
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, 453552, Simrol, Madhya Pradesh, India.
| |
Collapse
|
2
|
Khan MA, Mutahir S, Jabar G, Wenwei Z, Tariq MA, Almehizia AA, Mustafa M. DFT, Molecular Docking, ADME, and Cardiotoxicity Studies of Persuasive Thiazoles as Potential Inhibitors of the Main Protease of SARS-CoV-2. Chem Biodivers 2024; 21:e202401775. [PMID: 39161231 DOI: 10.1002/cbdv.202401775] [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] [Received: 07/22/2024] [Accepted: 08/19/2024] [Indexed: 08/21/2024]
Abstract
This study explores the capability of thiazoles as potent inhibitors of SARS-CoV-2 Mpro. Seventeen thiazoles (1-17) were screened for their linking affinity with the active site of SARS-CoV-2 Mpro and compared with the FDA-recommended antiviral drugs, Remdesivir and Baricitinib. Density Functional Theory (DFT) calculations provided electronic and energetic properties of these ligands, shedding light on their stability and reactivity. Molecular docking analysis revealed that thiazole derivatives exhibited favorable linking affinities with various functional sites of SARS-CoV-2 proteins, including spike receptor-linking zone, nucleocapsid protein N-terminal RNA linking zone, and Mpro. Notably, compounds 3, 10, and 12 displayed the best interaction with 6LZG as compared to FDA-approved antiviral drugs Remdesivir and Baricitinib, while compounds 1, 10, and 8 exhibited strong linking with 6 M3 M and also better than Remdesivir and Baricitinib. Additionally, compounds 3, 1, and 6 showed promising interactions with 6LU7 but only compound 3 performed better than Baricitinib. An ADME (Absorption, Distribution, Metabolism, and Excretion) study provided insights into the pharmacokinetics and drug-likeness of these compounds, with all ligands demonstrating good physicochemical characteristics, lipophilicity, water solubility, pharmacokinetics, drug-likeness, and medicinal chemistry attributes. The results suggest that these selected thiazole derivatives hold promise as potential candidates for further drug development.
Collapse
Affiliation(s)
- Muhammad Asim Khan
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, China
| | - Sadaf Mutahir
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, China
| | - Gauhar Jabar
- Department of Chemistry, University of Sialkot, Sialkot, 51300, Pakistan
| | - Zhao Wenwei
- School of Chemistry and Chemical Engineering, Linyi University, Linyi, 276005, China
| | | | - Abdulrahman A Almehizia
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Saudi Arabia
| | - Muhammad Mustafa
- Department of Chemistry, University of Sialkot, Sialkot, 51300, Pakistan
| |
Collapse
|
3
|
Hewitt P, Hartmann A, Tornesi B, Ferry-Martin S, Valentin JP, Desert P, Gresham S, Demarta-Gatsi C, Venishetty VK, Kolly C. Importance of tailored non-clinical safety testing of novel antimalarial drugs: Industry best-practice. Regul Toxicol Pharmacol 2024; 154:105736. [PMID: 39515409 DOI: 10.1016/j.yrtph.2024.105736] [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: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
Malaria is an acute, debilitating parasitic illness. There were 249 million cases of malaria in 2022, resulting in 608,000 deaths globally, 76% of which were children ≤5 years. The unique nature of this disease (recurrences leading to re-treatments and numerous organ systems affected), specific clinical treatment regimens, poor compliance, and diversity of affected populations (predominantly pediatrics, women of childbearing potential, pregnant and lactating women), often makes standard testing approaches inadequate, and tailor-made safety assessments are more appropriate. We provide best practice recommendations based on company experience for the non-clinical safety assessment of antimalarial drugs, with a focus on small molecules since they represent the majority of drug candidates for this illness. We focus on specific testing considerations for repeat dose toxicity studies, including combination toxicity assessments, since new drug treatment regimens typically foresee short treatment durations to improve compliance (i.e., 1 day) with combinations of compounds to improve efficacy and limit potential resistance. Due to the target population, the timing of reproductive, developmental, and juvenile toxicity studies may be earlier than general testing roadmaps for other small molecule drugs. In conclusion, key recommendations presented should enable a more effective and efficient development path whilst protecting clinical trial participants and patients.
Collapse
Affiliation(s)
- Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany.
| | | | - Belen Tornesi
- Non-Clinical Pharmacology & Toxicology, Medicines for Malaria Venture, Geneva, Switzerland
| | - Sandrine Ferry-Martin
- Nonclinical Drug Safety, Merck Research Laboratories, Merck Sharp & Dohme, Clermont-Ferrand, France
| | - Jean-Pierre Valentin
- Early Clinical Development & Translational Science, Non-Clinical Safety Evaluation, UCB Pharma, Braine L'Alleud, Belgium
| | - Paul Desert
- Nonclinical Safety, Sanofi, Marcy l'Etoile, France
| | | | - Claudia Demarta-Gatsi
- Global Health R&D of Merck Healthcare, Ares Trading S.A., (a subsidiary of Merck KGaA, Darmstadt, Germany), Switzerland
| | | | - Carine Kolly
- Preclinical Safety, Novartis Pharma AG, Basel, Switzerland
| |
Collapse
|
4
|
Redfern WS, Pollard CE, Holbrook M, Islam B, Abbasi M, Mahmud J, Lambert K, Haslam A, Jo H, Khalidi H, Bielecka Z, Starkey J, Ellinger T, Bryan S, Savas A, Andrews S, Aspbury R, Rosenbrier Ribeiro L, Henderson Park KA, Vargas HM, Gilmer CR. Predicting clinical outcomes from off-target receptor interactions using Secondary Intelligence™. J Pharmacol Toxicol Methods 2024; 131:107570. [PMID: 39577752 DOI: 10.1016/j.vascn.2024.107570] [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: 09/23/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024]
Abstract
Adverse effects due to off-target activity can be predicted by careful comparison of the relationship between expected plasma concentration and off-target activity of the test compound with that of reference drugs targeting that receptor for their therapeutic efficacy. The ratio between plasma concentration (unbound) and the Ki at the receptor is a surrogate measure reflecting receptor occupancy. Where data are available for reference drugs, we have curated and evaluated this at 100 receptors, 72 of which can involve both negative and positive modulations by drugs: a total of 172 'receptor modulations'. This provides a quantitative framework upon which to achieve consistent risk assessment of off-target interactions across receptors, across compounds and between assessors. It therefore represents a significant departure from an opinion-based to an evidence-based approach to secondary pharmacology. Demonstration of proof-of-principle was achieved for one of the receptor interactions (α1A-adrenoceptor antagonism leading to postural hypotension in clinical use) due to the availability of high-quality off-target Ki data for >30 drugs at this receptor.
Collapse
Affiliation(s)
- Will S Redfern
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom.
| | - Chris E Pollard
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Mark Holbrook
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Barira Islam
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Mitra Abbasi
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Joanne Mahmud
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Katie Lambert
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Augustus Haslam
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Heeseung Jo
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Hiba Khalidi
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Zofia Bielecka
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom; Pharmacoepidemiology and Pharmacoeconomics Unit, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Kraków 30-688, Poland
| | - Josh Starkey
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Thomas Ellinger
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Simon Bryan
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Angeli Savas
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Steve Andrews
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Rob Aspbury
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | | | | | - Hugo M Vargas
- Translational Safety Research, TS&BA, Amgen Inc., Thousand Oaks, CA 91320, USA
| | - Clare R Gilmer
- Certara Predictive Technologies, Certara UK Limited, Level 2-Acero, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| |
Collapse
|
5
|
Burbank M, Kukic P, Ouedraogo G, Kenna JG, Hewitt NJ, Armstrong D, Otto-Bruc A, Ebmeyer J, Boettcher M, Willox I, Mahony C. In vitro pharmacologic profiling aids systemic toxicity assessment of chemicals. Toxicol Appl Pharmacol 2024; 492:117131. [PMID: 39437896 DOI: 10.1016/j.taap.2024.117131] [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: 07/18/2024] [Revised: 10/02/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
An adapted in vitro pharmacology profiling panel (APPP) was developed that included targets used in the pharmaceutical industry alongside additional targets whose cellular functions have been linked to systemic toxicities. This panel of 83 target assays was used to profile the activities of 129 cosmetic relevant chemicals with diverse chemical structures, physiochemical properties and cosmetic use types. Internal data consistency was proved robust, as evidenced by the reproducibility between single concentration and concentration-response data and showed good concordance with data reported in the ToxCast and drug excipient datasets. We discuss how the data can be analyzed and multiple potential contexts of use illustrated by case studies, alongside other new approach methodologies, to support cosmetic chemical risk assessments that do not require data from new animal studies.
Collapse
Affiliation(s)
| | - Predrag Kukic
- Unilever Safety and Environmental Assurance Centre, Bedfordshire, MK 44 1LQ, UK
| | | | - J Gerry Kenna
- Cosmetics Europe, 40 Avenue Hermann-Debroux, 1160 Brussels, Belgium
| | - Nicola J Hewitt
- Cosmetics Europe, 40 Avenue Hermann-Debroux, 1160 Brussels, Belgium
| | | | | | | | | | - Ian Willox
- Eurofins Cerep, Celle-Lévescault, France
| | | |
Collapse
|
6
|
Gerets HHJ, Delaunois A, Cardenas A, Class R, Fleurance R, de Haro T, Laleu B, Lowe MA, Rosseels ML, Valentin JP. Assessing the interplay between off-target promiscuity, cytotoxicity, and tolerability in rodents to improve the safety profile of novel anti-malarial plasmepsin X inhibitors. Toxicol Sci 2024; 201:311-320. [PMID: 38976649 DOI: 10.1093/toxsci/kfae086] [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: 07/10/2024] Open
Abstract
Within drug development, high off-target promiscuity as well as potent cytotoxicity, are associated with a high attrition rate. We investigated the safety profile of novel plasmepsin X (PMX) inhibitors for the treatment of malaria. In our screening cascade, a total of 249 PMX compounds were profiled in a panel of in vitro secondary pharmacology assays containing 44 targets (SafetyScreen44 panel) and in a cytotoxicity assay in HepG2 cells using ATP as an endpoint. Six of the lead compounds were subsequently tested in a 7-d rat toxicology study, and/or in a cardiovascular study in guinea pigs. Overall, compounds with high cytotoxicity in HepG2 cells correlated with high promiscuity (off-target hit rate >20%) in the SafetyScreen44 panel and were associated with poor tolerability in vivo (decedents, morbidity, adverse clinical signs, or severe cardiovascular effects). Some side effects observed in rats or guinea pigs could putatively be linked with hits in the secondary pharmacological profiling, such as the M1 or M2 muscarinic acetylcholine receptor, opioid µ and/or κ receptors or hERG/CaV1.2/Na+ channels, which were common to >50% the compounds tested in vivo. In summary, compounds showing high cytotoxicity and high promiscuity are likely to be poorly tolerated in vivo. However, such associations do not necessarily imply a causal relationship. Identifying the targets that cause these undesirable effects is key for early safety risk assessment. A tiered approach, based on a set of in vitro assays, helps selecting the compounds with highest likelihood of success to proceed to in vivo toxicology studies.
Collapse
Affiliation(s)
| | | | | | - Reiner Class
- UCB Biopharma SRL, 1420 Braine-l'Alleud, Belgium
| | | | | | - Benoît Laleu
- MMV Medicines for Malaria Venture, ICC, 1215 Geneva, Switzerland
| | - Martin A Lowe
- UK Branch of UCB Pharma, SL1 3WE Slough, United Kingdom
| | | | | |
Collapse
|
7
|
Brennan RJ, Jenkinson S, Brown A, Delaunois A, Dumotier B, Pannirselvam M, Rao M, Ribeiro LR, Schmidt F, Sibony A, Timsit Y, Sales VT, Armstrong D, Lagrutta A, Mittlestadt SW, Naven R, Peri R, Roberts S, Vergis JM, Valentin JP. The state of the art in secondary pharmacology and its impact on the safety of new medicines. Nat Rev Drug Discov 2024; 23:525-545. [PMID: 38773351 DOI: 10.1038/s41573-024-00942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/05/2024] [Indexed: 05/23/2024]
Abstract
Secondary pharmacology screening of investigational small-molecule drugs for potentially adverse off-target activities has become standard practice in pharmaceutical research and development, and regulatory agencies are increasingly requesting data on activity against targets with recognized adverse effect relationships. However, the screening strategies and target panels used by pharmaceutical companies may vary substantially. To help identify commonalities and differences, as well as to highlight opportunities for further optimization of secondary pharmacology assessment, we conducted a broad-ranging survey across 18 companies under the auspices of the DruSafe leadership group of the International Consortium for Innovation and Quality in Pharmaceutical Development. Based on our analysis of this survey and discussions and additional research within the group, we present here an overview of the current state of the art in secondary pharmacology screening. We discuss best practices, including additional safety-associated targets not covered by most current screening panels, and present approaches for interpreting and reporting off-target activities. We also provide an assessment of the safety impact of secondary pharmacology screening, and a perspective on opportunities and challenges in this rapidly developing field.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Mohan Rao
- Janssen Research & Development, San Diego, CA, USA
- Neurocrine Biosciences, San Diego, CA, USA
| | - Lyn Rosenbrier Ribeiro
- UCB Biopharma, Braine-l'Alleud, Belgium
- AstraZeneca, Cambridge, UK
- Grunenthal, Berkshire, UK
| | | | | | - Yoav Timsit
- Novartis Biomedical Research, Cambridge, MA, USA
- Blueprint Medicines, Cambridge, MA, USA
| | | | - Duncan Armstrong
- Novartis Biomedical Research, Cambridge, MA, USA
- Armstrong Pharmacology, Macclesfield, UK
| | | | | | - Russell Naven
- Takeda Pharmaceuticals, Cambridge, MA, USA
- Novartis Biomedical Research, Cambridge, MA, USA
| | - Ravikumar Peri
- Takeda Pharmaceuticals, Cambridge, MA, USA
- Alexion Pharmaceuticals, Wilmington, DE, USA
| | - Sonia Roberts
- Roche Pharma Research and Early Development, Roche Innovation Center, Basel, Switzerland
| | - James M Vergis
- Faegre Drinker Biddle and Reath, LLP, Washington, DC, USA
| | | |
Collapse
|
8
|
Dumotier BM, Urban L. Preclinical mitigation of 5-HT2B agonism-related cardiac valvulopathy revisited. J Pharmacol Toxicol Methods 2024; 128:107542. [PMID: 39032441 DOI: 10.1016/j.vascn.2024.107542] [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: 05/16/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
Cardiac valvulopathy (Cardiac Valve Disease; CVD) associated with off-target activation of the 5-hydroxytryptamine (5-HT) 2B receptor has been well recognized, but is still poorly predicted during drug development. The regulatory guidance proposes the use of 5-HT2B binding data (i.e., Ki values) and free maximum therapeutic exposure (Cmax) to calculate safety margins as a threshold of detection (>10) for eliminating the risk of drug-induced cardiac valvulopathy. In this paper, we provide additional recommendations for preclinical prediction of CVD risk based on clinical pharmacodynamic and pharmacokinetic data obtained from drugs with or without 5-HT2B receptor activation. Our investigations showed that 5-HT2B agonist affinity of molecules tested in an in vitro 5-HT2B cell-based functional assay, placed in perspective to their sustained plasma exposure (AUCs) and not to their peak plasma exposure, Cmax (i.e., maximum therapeutic exposure) provide a solid basis for interpreting 5-HT2B data, for calculating safety margins and then, accurately differentiate drugs associated with a clinical risk of CVD from those which are not (despite having some agonist 5-HT2B activity). In addition, we discuss the risk of multi-organ fibrosis linked to 5-HT2B receptor activation, often underestimated, however well reported in FAERS for 5-HT2B agonists. We believe that our recommendations have the potential to mitigate the risk for the clinical development of CVD and fibrosis.
Collapse
Affiliation(s)
- Bérengère M Dumotier
- Novartis Biomedical Research, Translational Medicine, Preclinical Safety, Basel CH-4056, Switzerland.
| | - Laszlo Urban
- Novartis Biomedical Research, Translational Medicine, Preclinical Safety, Cambridge, MA 02139, United States
| |
Collapse
|
9
|
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
|
10
|
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
|
11
|
Cheng Y, Ji C, Xu J, Chen R, Guo Y, Bian Q, Shen Z, Zhang B. LCK-SafeScreen-Model: An Advanced Ensemble Machine Learning Approach for Estimating the Binding Affinity between Compounds and LCK Target. Molecules 2023; 28:7382. [PMID: 37959801 PMCID: PMC10650606 DOI: 10.3390/molecules28217382] [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: 08/28/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
The lymphocyte-specific protein tyrosine kinase (LCK) is a critical target in leukemia treatment. However, potential off-target interactions involving LCK can lead to unintended consequences. This underscores the importance of accurately predicting the inhibitory reactions of drug molecules with LCK during the research and development stage. To address this, we introduce an advanced ensemble machine learning technique designed to estimate the binding affinity between molecules and LCK. This comprehensive method includes the generation and selection of molecular fingerprints, the design of the machine learning model, hyperparameter tuning, and a model ensemble. Through rigorous optimization, the predictive capabilities of our model have been significantly enhanced, raising test R2 values from 0.644 to 0.730 and reducing test RMSE values from 0.841 to 0.732. Utilizing these advancements, our refined ensemble model was employed to screen an MCE -like drug library. Through screening, we selected the top ten scoring compounds, and tested them using the ADP-Glo bioactivity assay. Subsequently, we employed molecular docking techniques to further validate the binding mode analysis of these compounds with LCK. The exceptional predictive accuracy of our model in identifying LCK inhibitors not only emphasizes its effectiveness in projecting LCK-related safety panel predictions but also in discovering new LCK inhibitors. For added user convenience, we have also established a webserver, and a GitHub repository to share the project.
Collapse
Affiliation(s)
- Ying Cheng
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.C.); (C.J.); (J.X.)
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (R.C.); (Y.G.); (Q.B.)
| | - Cong Ji
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.C.); (C.J.); (J.X.)
| | - Jun Xu
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.C.); (C.J.); (J.X.)
- Department of Pharmacy, Huzhou Central Hospital, Huzhou 313000, China
| | - Roufen Chen
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (R.C.); (Y.G.); (Q.B.)
| | - Yu Guo
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (R.C.); (Y.G.); (Q.B.)
| | - Qingyu Bian
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (R.C.); (Y.G.); (Q.B.)
| | - Zheyuan Shen
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (R.C.); (Y.G.); (Q.B.)
| | - Bo Zhang
- College of Pharmaceutical Sciences, Hangzhou First People’s Hospital, Zhejiang Chinese Medical University, Hangzhou 311402, China; (Y.C.); (C.J.); (J.X.)
| |
Collapse
|
12
|
Santillo MF, Sprando RL. Predicting binding between 55 cannabinoids and 4,799 biological targets by in silico methods. J Appl Toxicol 2023; 43:1476-1487. [PMID: 37101313 DOI: 10.1002/jat.4478] [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] [Received: 03/20/2023] [Revised: 04/11/2023] [Accepted: 04/22/2023] [Indexed: 04/28/2023]
Abstract
Recently, there has been an increase in cannabis-derived products being marketed as foods, dietary supplements, and other consumer products. Cannabis contains over a hundred cannabinoids, many of which have unknown physiological effects. Since there are large numbers of cannabinoids, and many are not commercially available for in vitro testing, an in silico tool (Chemotargets Clarity software) was used to predict binding between 55 cannabinoids and 4,799 biological targets (enzymes, ion channels, receptors, and transporters). This tool relied on quantitative structure activity relationships (QSAR), structural similarity, and other approaches to predict binding. From this screening, 827 cannabinoid-target binding pairs were predicted, which included 143 unique targets. Many cannabinoids sharing core structures (cannabinoid "types") had similar binding profiles, whereas most cannabinoids containing carboxylic acid groups were similar without regards to their core structure. For some of the binding predictions (43), in vitro binding data were available, and they agreed well with in silico binding data (median fourfold difference in binding concentrations). Finally, clinical adverse effects associated with 22 predicted targets were identified from an online database (Clarivate Off-X), providing important insights on potential human health hazards. Overall, in silico biological target predictions are a rapid means to identify potential hazards due to cannabinoid-target interactions, and the data can be used to prioritize subsequent in vitro and in vivo testing.
Collapse
Affiliation(s)
- Michael F Santillo
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, Maryland, USA
| | - Robert L Sprando
- Division of Toxicology, Office of Applied Research and Safety Assessment, Center for Food Safety and Applied Nutrition, US Food and Drug Administration, Laurel, Maryland, USA
| |
Collapse
|
13
|
Valentin JP, Sibony A, Rosseels ML, Delaunois A. "Appraisal of state-of-the-art" The 2021 Distinguished Service Award of the Safety Pharmacology Society: Reflecting on the past to tackle challenges ahead. J Pharmacol Toxicol Methods 2023; 123:107269. [PMID: 37149063 DOI: 10.1016/j.vascn.2023.107269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 05/03/2023] [Indexed: 05/08/2023]
Abstract
This appraisal of state-of-the-art manuscript highlights and expands upon the thoughts conveyed in the lecture of Dr. Jean-Pierre Valentin, recipient of the 2021 Distinguished Service Award of the Safety Pharmacology Society, given on the 2nd December 2021. The article reflects on the strengths, weaknesses, opportunities, and threats that surrounded the evolution of safety and secondary pharmacology over the last 3 decades with a particular emphasis on pharmaceutical drug development delivery, scientific and technological innovation, complexities of regulatory framework and people leadership and development. The article further built on learnings from past experiences to tackle constantly emerging issues and evolving landscape whilst being cognizant of the challenges facing these disciplines in the broader drug development and societal context.
Collapse
Affiliation(s)
- Jean-Pierre Valentin
- UCB-Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine L'Alleud, Belgium.
| | - Alicia Sibony
- UCB-Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine L'Alleud, Belgium
| | - Marie-Luce Rosseels
- UCB-Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine L'Alleud, Belgium
| | - Annie Delaunois
- UCB-Biopharma SRL, Early Solutions, Development Science, Non-Clinical Safety Evaluation, Braine L'Alleud, Belgium
| |
Collapse
|
14
|
Wang X, Wang T, Fan X, Zhang Z, Wang Y, Li Z. A Molecular Toolbox of Positron Emission Tomography Tracers for General Anesthesia Mechanism Research. J Med Chem 2023; 66:6463-6497. [PMID: 37145921 DOI: 10.1021/acs.jmedchem.2c01965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
With appropriate radiotracers, positron emission tomography (PET) allows direct or indirect monitoring of the spatial and temporal distribution of anesthetics, neurotransmitters, and biomarkers, making it an indispensable tool for studying the general anesthesia mechanism. In this Perspective, PET tracers that have been recruited in general anesthesia research are introduced in the following order: 1) 11C/18F-labeled anesthetics, i.e., PET tracers made from inhaled and intravenous anesthetics; 2) PET tracers targeting anesthesia-related receptors, e.g., neurotransmitters and voltage-gated ion channels; and 3) PET tracers for studying anesthesia-related neurophysiological effects and neurotoxicity. The radiosynthesis, pharmacodynamics, and pharmacokinetics of the above PET tracers are mainly discussed to provide a practical molecular toolbox for radiochemists, anesthesiologists, and those who are interested in general anesthesia.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Tao Wang
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaowei Fan
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhao Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zijing Li
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
15
|
Pognan F, Beilmann M, Boonen HCM, Czich A, Dear G, Hewitt P, Mow T, Oinonen T, Roth A, Steger-Hartmann T, Valentin JP, Van Goethem F, Weaver RJ, Newham P. The evolving role of investigative toxicology in the pharmaceutical industry. Nat Rev Drug Discov 2023; 22:317-335. [PMID: 36781957 PMCID: PMC9924869 DOI: 10.1038/s41573-022-00633-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 02/15/2023]
Abstract
For decades, preclinical toxicology was essentially a descriptive discipline in which treatment-related effects were carefully reported and used as a basis to calculate safety margins for drug candidates. In recent years, however, technological advances have increasingly enabled researchers to gain insights into toxicity mechanisms, supporting greater understanding of species relevance and translatability to humans, prediction of safety events, mitigation of side effects and development of safety biomarkers. Consequently, investigative (or mechanistic) toxicology has been gaining momentum and is now a key capability in the pharmaceutical industry. Here, we provide an overview of the current status of the field using case studies and discuss the potential impact of ongoing technological developments, based on a survey of investigative toxicologists from 14 European-based medium-sized to large pharmaceutical companies.
Collapse
Affiliation(s)
- Francois Pognan
- Discovery and Investigative Safety, Novartis Pharma AG, Basel, Switzerland.
| | - Mario Beilmann
- Nonclinical Drug Safety Germany, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Harrie C M Boonen
- Drug Safety, Dept of Exploratory Toxicology, Lundbeck A/S, Valby, Denmark
| | | | - Gordon Dear
- In Vitro In Vivo Translation, GlaxoSmithKline David Jack Centre for Research, Ware, UK
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Tomas Mow
- Safety Pharmacology and Early Toxicology, Novo Nordisk A/S, Maaloev, Denmark
| | - Teija Oinonen
- Preclinical Safety, Orion Corporation, Espoo, Finland
| | - Adrian Roth
- Pharmaceutical Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | | | | | - Freddy Van Goethem
- Predictive, Investigative & Translational Toxicology, Nonclinical Safety, Janssen Research & Development, Beerse, Belgium
| | - Richard J Weaver
- Innovation Life Cycle Management, Institut de Recherches Internationales Servier, Suresnes, France
| | - Peter Newham
- Clinical Pharmacology and Safety Sciences, AstraZeneca R&D, Cambridge, UK.
| |
Collapse
|
16
|
De Vita S, Chini MG, Bifulco G, Lauro G. Target identification by structure-based computational approaches: Recent advances and perspectives. Bioorg Med Chem Lett 2023; 83:129171. [PMID: 36739998 DOI: 10.1016/j.bmcl.2023.129171] [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: 08/05/2022] [Revised: 12/15/2022] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
The use of computational techniques in the early stages of drug discovery has recently experienced a boost, especially in the target identification step. Finding the biological partner(s) for new or existing synthetic and/or natural compounds by "wet" approaches may be challenging; therefore, preliminary in silico screening is even more recommended. After a brief overview of some of the most known target identification techniques, recent advances in structure-based computational approaches for target identification are reported in this digest, focusing on Inverse Virtual Screening and its recent applications. Moreover, future perspectives concerning the use of such methodologies, coupled or not with other approaches, are analyzed.
Collapse
Affiliation(s)
- Simona De Vita
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy
| | - Maria Giovanna Chini
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche (IS), Italy
| | - Giuseppe Bifulco
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy.
| | - Gianluigi Lauro
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano (SA), Italy.
| |
Collapse
|
17
|
Middleton AM, Reynolds J, Cable S, Baltazar MT, Li H, Bevan S, Carmichael PL, Dent MP, Hatherell S, Houghton J, Kukic P, Liddell M, Malcomber S, Nicol B, Park B, Patel H, Scott S, Sparham C, Walker P, White A. Are Non-animal Systemic Safety Assessments Protective? A Toolbox and Workflow. Toxicol Sci 2022; 189:124-147. [PMID: 35822611 PMCID: PMC9412174 DOI: 10.1093/toxsci/kfac068] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
An important question in toxicological risk assessment is whether non-animal new approach methodologies (NAMs) can be used to make safety decisions that are protective of human health, without being overly conservative. In this work, we propose a core NAM toolbox and workflow for conducting systemic safety assessments for adult consumers. We also present an approach for evaluating how protective and useful the toolbox and workflow are by benchmarking against historical safety decisions. The toolbox includes physiologically based kinetic (PBK) models to estimate systemic Cmax levels in humans, and 3 bioactivity platforms, comprising high-throughput transcriptomics, a cell stress panel, and in vitro pharmacological profiling, from which points of departure are estimated. A Bayesian model was developed to quantify the uncertainty in the Cmax estimates depending on how the PBK models were parameterized. The feasibility of the evaluation approach was tested using 24 exposure scenarios from 10 chemicals, some of which would be considered high risk from a consumer goods perspective (eg, drugs that are systemically bioactive) and some low risk (eg, existing food or cosmetic ingredients). Using novel protectiveness and utility metrics, it was shown that up to 69% (9/13) of the low risk scenarios could be identified as such using the toolbox, whilst being protective against all (5/5) the high-risk ones. The results demonstrated how robust safety decisions could be made without using animal data. This work will enable a full evaluation to assess how protective and useful the toolbox and workflow are across a broader range of chemical-exposure scenarios.
Collapse
Affiliation(s)
| | - Joe Reynolds
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Sophie Cable
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | | | - Hequn Li
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | | | - Paul L Carmichael
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Matthew Philip Dent
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Sarah Hatherell
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Jade Houghton
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Predrag Kukic
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Mark Liddell
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Sophie Malcomber
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Beate Nicol
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | | | - Hiral Patel
- Charles River Laboratories, Cambridgeshire, CB10 1XL, UK
| | - Sharon Scott
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Chris Sparham
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| | - Paul Walker
- Cyprotex Discovery Ltd, Cheshire SK10 4TG, UK
| | - Andrew White
- Unilever Safety and Environmental Assurance Centre, Bedfordshire MK44 1LQ, UK
| |
Collapse
|
18
|
Ruan C, Wang Y, Zhang X, Lyu J, Zhang N, Ma Y, Shi C, Qu G, Ye M. Matrix Thermal Shift Assay for Fast Construction of Multidimensional Ligand-Target Space. Anal Chem 2022; 94:6482-6490. [PMID: 35442643 DOI: 10.1021/acs.analchem.1c04627] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Existing thermal shift-based mass spectrometry approaches are able to identify target proteins without chemical modification of the ligand, but they are suffering from complicated workflows with limited throughput. Herein, we present a new thermal shift-based method, termed matrix thermal shift assay (mTSA), for fast deconvolution of ligand-binding targets and binding affinities at the proteome level. In mTSA, a sample matrix, treated horizontally with five different compound concentrations and vertically with five technical replicates of each condition, was denatured at a single temperature to induce protein precipitation, and then, data-independent acquisition was employed for quick protein quantification. Compared with previous thermal shift assays, the analysis throughput of mTSA was significantly improved, but the costs as well as efforts were reduced. More importantly, the matrix experiment design allowed simultaneous computation of the statistical significance and fitting of the dose-response profiles, which can be combined to enable a more accurate identification of target proteins, as well as reporting binding affinities between the ligand and individual targets. Using a pan-specific kinase inhibitor, staurosporine, we demonstrated a 36% improvement in screening sensitivity over the traditional thermal proteome profiling (TPP) and a comparable sensitivity with a latest two-dimensional TPP. Finally, mTSA was successfully applied to delineate the target landscape of perfluorooctanesulfonic acid (PFOS), a persistent organic pollutant that is hard to perform modification on, and revealed several potential targets that might account for the toxicities of PFOS.
Collapse
Affiliation(s)
- Chengfei Ruan
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
| | - Xiaolei Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
| | - Jiawen Lyu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Na Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanni Ma
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunzhen Shi
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Guangbo Qu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Mingliang Ye
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian 116023, China
| |
Collapse
|
19
|
Accurate in silico simulation of the rabbit Purkinje fiber electrophysiological assay to facilitate early pharmaceutical cardiosafety assessment: Dream or reality? J Pharmacol Toxicol Methods 2022; 115:107172. [DOI: 10.1016/j.vascn.2022.107172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 03/31/2022] [Accepted: 04/08/2022] [Indexed: 11/24/2022]
|
20
|
Ponting DJ, Burns MJ, Foster RS, Hemingway R, Kocks G, MacMillan DS, Shannon-Little AL, Tennant RE, Tidmarsh JR, Yeo DJ. Use of Lhasa Limited Products for the In Silico Prediction of Drug Toxicity. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2425:435-478. [PMID: 35188642 DOI: 10.1007/978-1-0716-1960-5_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lhasa Limited have had a role in the in silico prediction of drug and other chemical toxicity for over 30 years. This role has always been multifaceted, both as a provider of predictive software such as Derek Nexus, and as an honest broker for the sharing of proprietary chemical and toxicity data. A changing regulatory environment and the drive for the Replacement, Reduction and Refinement (the 3Rs) of animal testing have led both to increased acceptance of in silico predictions and a desire for the sharing of data to reduce duplicate testing. The combination of these factors has led to Lhasa Limited providing a suite of products and coordinating numerous data-sharing consortia that do indeed facilitate a significant reduction in the testing burden that companies would otherwise be laboring under. Many of these products and consortia can be organized into workflows for specific regulatory use cases, and it is these that will be used to frame the narrative in this chapter.
Collapse
|
21
|
Ball K, Bruin G, Escandon E, Funk C, Pereira JN, Yang TY, Yu H. Characterizing the pharmacokinetics and biodistribution of therapeutic proteins: an industry white paper. Drug Metab Dispos 2022; 50:858-866. [PMID: 35149542 DOI: 10.1124/dmd.121.000463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022] Open
Abstract
Characterization of the pharmacokinetics (PK) and biodistribution of therapeutic proteins (TPs) is a hot topic within the pharmaceutical industry, particularly with an ever-increasing catalog of novel modality TPs. Here, we review the current practices, and provide a summary of extensive cross-company discussions as well as a survey completed by International Consortium for Innovation and Quality (IQ consortium) members on this theme. A wide variety of in vitro, in vivo and in silico techniques are currently used to assess PK and biodistribution of TPs, and we discuss the relevance of these from an industry perspective, focusing on PK/PD understanding at the preclinical stage of development, and translation to human. We consider that the 'traditional in vivo biodistribution study' is becoming insufficient as a standalone tool, and thorough characterization of the interaction of the TP with its target(s), target biology, and off-target interactions at a microscopic scale are key to understand the overall biodistribution at a full-body scale. Our summary of the current challenges and our recommendations to address these issues could provide insight into the implementation of best practices in this area of drug development, and continued cross-company collaboration will be of tremendous value. Significance Statement The Innovation & Quality Consortium (IQ) Translational and ADME Sciences Leadership Group (TALG) working group for the ADME of therapeutic proteins evaluates the current practices, recent advances, and challenges in characterizing the PK and biodistribution of therapeutic proteins during drug development, and proposes recommendations to address these issues. Incorporating the in vitro, in vivo and in silico approaches discussed herein may provide a pragmatic framework to increase early understanding of PK/PD relationships, and aid translational modelling for first-in-human dose predictions.
Collapse
Affiliation(s)
| | - Gerard Bruin
- Novartis Institutes for Biomedical Research, Switzerland
| | | | - Christoph Funk
- Dept. of Drug Metabolism and Pharmacokinetics, F. Hoffmann-La Roche Ltd., Switzerland
| | | | | | - Hongbin Yu
- Boehringer Ingelheim Pharmaceuticals, Inc, United States
| |
Collapse
|
22
|
High-throughput screening for identifying acetylcholinesterase inhibitors: Insights on novel inhibitors and the use of liver microsomes. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:65-67. [PMID: 35058177 PMCID: PMC9884470 DOI: 10.1016/j.slasd.2021.10.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Rapid, higher throughput, and predictive toxicological methods are needed to assess vast numbers of chemicals with unknown safety profiles. A current effort towards this goal is Toxicology in the 21st Century (Tox21), a United States government consortium using a battery of in vitro assays to screen a library of 10,000 compounds relevant to food, drug, and environmental safety. Recently, we implemented in vitro assays for measuring acetylcholinesterase (AChE) inhibition, a mechanism of toxicity, into Tox21's high-throughput screening campaign (Li S., et al. Environ Health Persp 2021;129:047008, doi:10.1289/EHP6993). In this Commentary, we provide detailed insights on two topics related to our article: (1) prioritizing recently discovered AChE inhibitors from our screening based upon physiological relevance and (2) incorporating human liver microsomes into the AChE inhibition assay to identify metabolically active AChE inhibitors.
Collapse
|
23
|
Halland N, Schmidt F, Weiss T, Li Z, Czech J, Saas J, Ding-Pfennigdorff D, Dreyer MK, Strübing C, Nazare M. Rational Design of Highly Potent, Selective, and Bioavailable SGK1 Protein Kinase Inhibitors for the Treatment of Osteoarthritis. J Med Chem 2021; 65:1567-1584. [PMID: 34931844 DOI: 10.1021/acs.jmedchem.1c01601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The serine/threonine kinase SGK1 is an activator of the β-catenin pathway and a powerful stimulator of cartilage degradation that is found to be upregulated under genomic control in diseased osteoarthritic cartilage. Today, no oral disease-modifying treatments are available and chronic treatment in this indication sets high requirements for the drug selectivity, pharmacokinetic, and safety profile. We describe the identification of a highly selective druglike 1H-pyrazolo[3,4-d]pyrimidine SGK1 inhibitor 17a that matches both safety and pharmacokinetic requirements for oral dosing. Rational compound design was facilitated by a novel hSGK1 co-crystal structure, and multiple ligand-based computer models were applied to guide the chemical optimization of the compound ADMET and selectivity profiles. Compounds were selected for subchronic proof of mechanism studies in the mouse femoral head cartilage explant model, and compound 17a emerged as a druglike SGK1 inhibitor, with a highly optimized profile suitable for oral dosing as a novel, potentially disease-modifying agent for osteoarthritis.
Collapse
Affiliation(s)
- Nis Halland
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Friedemann Schmidt
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Tilo Weiss
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Ziyu Li
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Jörg Czech
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Joachim Saas
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | | | - Matthias K Dreyer
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Carsten Strübing
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Marc Nazare
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
| |
Collapse
|
24
|
Löscher W, Kaila K. CNS pharmacology of NKCC1 inhibitors. Neuropharmacology 2021; 205:108910. [PMID: 34883135 DOI: 10.1016/j.neuropharm.2021.108910] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 12/21/2022]
Abstract
The Na-K-2Cl cotransporter NKCC1 and the neuron-specific K-Cl cotransporter KCC2 are considered attractive CNS drug targets because altered neuronal chloride regulation and consequent effects on GABAergic signaling have been implicated in numerous CNS disorders. While KCC2 modulators are not yet clinically available, the loop diuretic bumetanide has been used off-label in attempts to treat brain disorders and as a tool for NKCC1 inhibition in preclinical models. Bumetanide is known to have anticonvulsant and neuroprotective effects under some pathophysiological conditions. However, as shown in several species from neonates to adults (mice, rats, dogs, and by extrapolation in humans), at the low clinical doses of bumetanide approved for diuresis, this drug has negligible access into the CNS, reaching levels that are much lower than what is needed to inhibit NKCC1 in cells within the brain parenchyma. Several drug discovery strategies have been initiated over the last ∼15 years to develop brain-permeant compounds that, ideally, should be selective for NKCC1 to eliminate the diuresis mediated by inhibition of renal NKCC2. The strategies employed to improve the pharmacokinetic and pharmacodynamic properties of NKCC1 blockers include evaluation of other clinically approved loop diuretics; development of lipophilic prodrugs of bumetanide; development of side-chain derivatives of bumetanide; and unbiased high-throughput screening approaches of drug discovery based on large chemical compound libraries. The main outcomes are that (1), non-acidic loop diuretics such as azosemide and torasemide may have advantages as NKCC1 inhibitors vs. bumetanide; (2), bumetanide prodrugs lead to significantly higher brain levels than the parent drug and have lower diuretic activity; (3), the novel bumetanide side-chain derivatives do not exhibit any functionally relevant improvement of CNS accessibility or NKCC1 selectivity vs. bumetanide; (4) novel compounds discovered by high-throughput screening may resolve some of the inherent problems of bumetanide, but as yet this has not been achieved. Thus, further research is needed to optimize the design of brain-permeant NKCC1 inhibitors. In parallel, a major challenge is to identify the mechanisms whereby various NKCC1-expressing cellular targets of these drugs within (e.g., neurons, oligodendrocytes or astrocytes) and outside the brain parenchyma (e.g., the blood-brain barrier, the choroid plexus, and the endocrine system), as well as molecular off-target effects, might contribute to their reported therapeutic and adverse effects.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Dept. of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, Germany; Center for Systems Neuroscience Hannover, Germany.
| | - Kai Kaila
- Molecular and Integrative Biosciences and Neuroscience Center (HiLIFE), University of Helsinki, Finland
| |
Collapse
|
25
|
Eguida M, Rognan D. Unexpected similarity between HIV-1 reverse transcriptase and tumor necrosis factor binding sites revealed by computer vision. J Cheminform 2021; 13:90. [PMID: 34814950 PMCID: PMC8609734 DOI: 10.1186/s13321-021-00567-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/06/2021] [Indexed: 11/10/2022] Open
Abstract
Rationalizing the identification of hidden similarities across the repertoire of druggable protein cavities remains a major hurdle to a true proteome-wide structure-based discovery of novel drug candidates. We recently described a new computational approach (ProCare), inspired by numerical image processing, to identify local similarities in fragment-based subpockets. During the validation of the method, we unexpectedly identified a possible similarity in the binding pockets of two unrelated targets, human tumor necrosis factor alpha (TNF-α) and HIV-1 reverse transcriptase (HIV-1 RT). Microscale thermophoresis experiments confirmed the ProCare prediction as two of the three tested and FDA-approved HIV-1 RT inhibitors indeed bind to soluble human TNF-α trimer. Interestingly, the herein disclosed similarity could be revealed neither by state-of-the-art binding sites comparison methods nor by ligand-based pairwise similarity searches, suggesting that the point cloud registration approach implemented in ProCare, is uniquely suited to identify local and unobvious similarities among totally unrelated targets.
Collapse
Affiliation(s)
- Merveille Eguida
- Laboratoire d'Innovation Thérapeutique, UMR 7200 CNRS, Université de Strasbourg, 67400, Illkirch, France
| | - Didier Rognan
- Laboratoire d'Innovation Thérapeutique, UMR 7200 CNRS, Université de Strasbourg, 67400, Illkirch, France.
| |
Collapse
|
26
|
Bassan A, Alves VM, Amberg A, Anger LT, Auerbach S, Beilke L, Bender A, Cronin MT, Cross KP, Hsieh JH, Greene N, Kemper R, Kim MT, Mumtaz M, Noeske T, Pavan M, Pletz J, Russo DP, Sabnis Y, Schaefer M, Szabo DT, Valentin JP, Wichard J, Williams D, Woolley D, Zwickl C, Myatt GJ. In silico approaches in organ toxicity hazard assessment: current status and future needs in predicting liver toxicity. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 20:100187. [PMID: 35340402 PMCID: PMC8955833 DOI: 10.1016/j.comtox.2021.100187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Hepatotoxicity is one of the most frequently observed adverse effects resulting from exposure to a xenobiotic. For example, in pharmaceutical research and development it is one of the major reasons for drug withdrawals, clinical failures, and discontinuation of drug candidates. The development of faster and cheaper methods to assess hepatotoxicity that are both more sustainable and more informative is critically needed. The biological mechanisms and processes underpinning hepatotoxicity are summarized and experimental approaches to support the prediction of hepatotoxicity are described, including toxicokinetic considerations. The paper describes the increasingly important role of in silico approaches and highlights challenges to the adoption of these methods including the lack of a commonly agreed upon protocol for performing such an assessment and the need for in silico solutions that take dose into consideration. A proposed framework for the integration of in silico and experimental information is provided along with a case study describing how computational methods have been used to successfully respond to a regulatory question concerning non-genotoxic impurities in chemically synthesized pharmaceuticals.
Collapse
Affiliation(s)
- Arianna Bassan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova, Italy
| | - Vinicius M. Alves
- The National Institute of Environmental Health Sciences, Division of the National Toxicology, Program, Research Triangle Park, NC 27709, USA
| | - Alexander Amberg
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926 Frankfurt am Main, Germany
| | | | - Scott Auerbach
- The National Institute of Environmental Health Sciences, Division of the National Toxicology, Program, Research Triangle Park, NC 27709, USA
| | - Lisa Beilke
- Toxicology Solutions Inc., San Diego, CA, USA
| | - Andreas Bender
- AI and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW
| | - Mark T.D. Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | | | - Jui-Hua Hsieh
- The National Institute of Environmental Health Sciences, Division of the National Toxicology, Program, Research Triangle Park, NC 27709, USA
| | - Nigel Greene
- Data Science and AI, DSM, IMED Biotech Unit, AstraZeneca, Boston, USA
| | - Raymond Kemper
- Nuvalent, One Broadway, 14th floor, Cambridge, MA, 02142, USA
| | - Marlene T. Kim
- US Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD, 20993, USA
| | - Moiz Mumtaz
- Office of the Associate Director for Science (OADS), Agency for Toxic Substances and Disease, Registry, US Department of Health and Human Services, Atlanta, GA, USA
| | - Tobias Noeske
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Manuela Pavan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova, Italy
| | - Julia Pletz
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Daniel P. Russo
- Department of Chemistry, Rutgers University, Camden, NJ 08102, USA
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ 08102, USA
| | - Yogesh Sabnis
- UCB Biopharma SRL, Chemin du Foriest – B-1420 Braine-l’Alleud, Belgium
| | - Markus Schaefer
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926 Frankfurt am Main, Germany
| | | | | | - Joerg Wichard
- Bayer AG, Genetic Toxicology, Müllerstr. 178, 13353 Berlin, Germany
| | - Dominic Williams
- Functional & Mechanistic Safety, Clinical Pharmacology & Safety Sciences, AstraZeneca, Darwin Building 310, Cambridge Science Park, Milton Rd, Cambridge CB4 0FZ, UK
| | - David Woolley
- ForthTox Limited, PO Box 13550, Linlithgow, EH49 7YU, UK
| | - Craig Zwickl
- Transendix LLC, 1407 Moores Manor, Indianapolis, IN 46229, USA
| | | |
Collapse
|
27
|
Bassan A, Alves VM, Amberg A, Anger LT, Beilke L, Bender A, Bernal A, Cronin MT, Hsieh JH, Johnson C, Kemper R, Mumtaz M, Neilson L, Pavan M, Pointon A, Pletz J, Ruiz P, Russo DP, Sabnis Y, Sandhu R, Schaefer M, Stavitskaya L, Szabo DT, Valentin JP, Woolley D, Zwickl C, Myatt GJ. In silico approaches in organ toxicity hazard assessment: Current status and future needs for predicting heart, kidney and lung toxicities. COMPUTATIONAL TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 20:100188. [PMID: 35721273 PMCID: PMC9205464 DOI: 10.1016/j.comtox.2021.100188] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The kidneys, heart and lungs are vital organ systems evaluated as part of acute or chronic toxicity assessments. New methodologies are being developed to predict these adverse effects based on in vitro and in silico approaches. This paper reviews the current state of the art in predicting these organ toxicities. It outlines the biological basis, processes and endpoints for kidney toxicity, pulmonary toxicity, respiratory irritation and sensitization as well as functional and structural cardiac toxicities. The review also covers current experimental approaches, including off-target panels from secondary pharmacology batteries. Current in silico approaches for prediction of these effects and mechanisms are described as well as obstacles to the use of in silico methods. Ultimately, a commonly accepted protocol for performing such assessment would be a valuable resource to expand the use of such approaches across different regulatory and industrial applications. However, a number of factors impede their widespread deployment including a lack of a comprehensive mechanistic understanding, limited in vitro testing approaches and limited in vivo databases suitable for modeling, a limited understanding of how to incorporate absorption, distribution, metabolism, and excretion (ADME) considerations into the overall process, a lack of in silico models designed to predict a safe dose and an accepted framework for organizing the key characteristics of these organ toxicants.
Collapse
Affiliation(s)
- Arianna Bassan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova, Italy
| | - Vinicius M. Alves
- The National Institute of Environmental Health Sciences, Division of the National Toxicology Program, Research Triangle Park, NC 27709, United States
| | - Alexander Amberg
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926 Frankfurt am Main, Germany
| | - Lennart T. Anger
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, United States
| | - Lisa Beilke
- Toxicology Solutions Inc., San Diego, CA, United States
| | - Andreas Bender
- AI and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United States
| | | | - Mark T.D. Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Jui-Hua Hsieh
- The National Institute of Environmental Health Sciences, Division of the National Toxicology Program, Research Triangle Park, NC 27709, United States
| | | | - Raymond Kemper
- Nuvalent, One Broadway, 14th floor, Cambridge, MA 02142, United States
| | - Moiz Mumtaz
- Agency for Toxic Substances and Disease Registry, US Department of Health and Human Services, Atlanta, GA, United States
| | - Louise Neilson
- Broughton Nicotine Services, Oak Tree House, West Craven Drive, Earby, Lancashire BB18 6JZ UK
| | - Manuela Pavan
- Innovatune srl, Via Giulio Zanon 130/D, 35129 Padova, Italy
| | - Amy Pointon
- Functional and Mechanistic Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Julia Pletz
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Patricia Ruiz
- Agency for Toxic Substances and Disease Registry, US Department of Health and Human Services, Atlanta, GA, United States
| | - Daniel P. Russo
- The Rutgers Center for Computational and Integrative Biology, Camden, NJ 08102, United States
- Department of Chemistry, Rutgers University, Camden, NJ 08102, United States
| | - Yogesh Sabnis
- UCB Biopharma SRL, Chemin du Foriest, B-1420 Braine-l’Alleud, Belgium
| | - Reena Sandhu
- SafeDose Ltd., 20 Dundas Street West, Suite 921, Toronto, Ontario M5G2H1, Canada
| | - Markus Schaefer
- Sanofi, R&D Preclinical Safety Frankfurt, Industriepark Hoechst, D-65926 Frankfurt am Main, Germany
| | - Lidiya Stavitskaya
- US Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| | | | | | - David Woolley
- ForthTox Limited, PO Box 13550, Linlithgow, EH49 7YU, UK
| | - Craig Zwickl
- Transendix LLC, 1407 Moores Manor, Indianapolis, IN 46229, United States
| | - Glenn J. Myatt
- Instem, 1393 Dublin Road, Columbus, OH 43215, United States
| |
Collapse
|
28
|
Dodson A, Mi K, Russo DP, Scott C, Saulnier M, Snyder K, Racz R. Aggregation and analysis of secondary pharmacology data from investigational new drug submissions at the US Food and Drug Administration. J Pharmacol Toxicol Methods 2021; 111:107098. [PMID: 34229067 DOI: 10.1016/j.vascn.2021.107098] [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: 03/29/2021] [Revised: 06/14/2021] [Accepted: 06/29/2021] [Indexed: 01/23/2023]
Abstract
Secondary pharmacology studies are utilized by the pharmaceutical industry as a cost-efficient tool to identify potential safety liabilities of drugs before entering Phase 1 clinical trials. These studies are recommended by the Food and Drug Administration (FDA) as a part of the Investigational New Drug (IND) application. However, despite the utility of these assays, there is little guidance on which targets should be screened and which format should be used. Here, we evaluated 226 secondary pharmacology profiles obtained from close to 90 unique sponsors. The results indicated that the most tested target in our set was the GABA benzodiazepine receptor (tested 168 times), the most hit target was adenosine 3 (hit 24 times), and the target with the highest hit percentage was the quinone reductase 2 (NQO2) receptor (hit 29% of the time). The overall results were largely consistent with those observed in previous publications. However, this study also identified the need for improvement in the submission process of secondary pharmacology studies by industry, which could enhance their utility for regulatory purpose. FDA-industry collaborative working groups will utilize this data to determine the best methods for regulatory submission of these studies and evaluate the need for a standard target panel.
Collapse
Affiliation(s)
- Andrew Dodson
- Division of Applied Regulatory Science, Food and Drug Administration, Silver Spring, MD 20993, United States of America; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, United States of America
| | - Kevin Mi
- Office of New Drugs, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| | - Daniel P Russo
- Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, United States of America; Office of New Drugs, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| | - Christina Scott
- Division of Applied Regulatory Science, Food and Drug Administration, Silver Spring, MD 20993, United States of America; Oak Ridge Institute for Science and Education, Oak Ridge, TN 37830, United States of America
| | - Muriel Saulnier
- Office of New Drugs, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| | - Kevin Snyder
- Office of New Drugs, Food and Drug Administration, Silver Spring, MD 20993, United States of America
| | - Rebecca Racz
- Division of Applied Regulatory Science, Food and Drug Administration, Silver Spring, MD 20993, United States of America.
| |
Collapse
|
29
|
Dixit VA, Singh P. A property-response perspective on modern toxicity assessment and drug toxicity index (DTI). In Silico Pharmacol 2021; 9:37. [PMID: 34017677 PMCID: PMC8124026 DOI: 10.1007/s40203-021-00096-9] [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: 02/04/2021] [Accepted: 05/05/2021] [Indexed: 11/26/2022] Open
Abstract
Toxicity related failures in drug discovery and clinical development have motivated scientists and regulators to develop a wide range of in-vitro, in-silico tools coupled with data science methods. Older drug discovery rules are being constantly modified to churn out any hidden predictive value. Nonetheless, the dose-response concepts remain central to all these methods. Over the last 2 decades medicinal chemists, and pharmacologists have observed that different physicochemical, and pharmacological properties capture trends in toxic responses. We propose that these observations should be viewed in a comprehensive property-response framework where dose is only a factor that modifies the inherent toxicity potential. We then introduce the recently proposed "Drug Toxicity Index (DTI)" and briefly summarize its applications. A webserver is available to calculate DTI values (https://all-tool-kit.github.io/Web-Tool.html).
Collapse
Affiliation(s)
- Vaibhav A. Dixit
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani (BITS Pilani), Vidya Vihar Campus, Street number 41, Pilani, Rajasthan 333031 India
| | - Pragati Singh
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani (BITS Pilani), Vidya Vihar Campus, Street number 41, Pilani, Rajasthan 333031 India
| |
Collapse
|
30
|
Smit IA, Afzal AM, Allen CHG, Svensson F, Hanser T, Bender A. Systematic Analysis of Protein Targets Associated with Adverse Events of Drugs from Clinical Trials and Postmarketing Reports. Chem Res Toxicol 2020; 34:365-384. [PMID: 33351593 DOI: 10.1021/acs.chemrestox.0c00294] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Adverse drug reactions (ADRs) are undesired effects of medicines that can harm patients and are a significant source of attrition in drug development. ADRs are anticipated by routinely screening drugs against secondary pharmacology protein panels. However, there is still a lack of quantitative information on the links between these off-target proteins and the reporting of ADRs in humans. Here, we present a systematic analysis of associations between measured and predicted in vitro bioactivities of drugs and adverse events (AEs) in humans from two sources of data: the Side Effect Resource, derived from clinical trials, and the Food and Drug Administration Adverse Event Reporting System, derived from postmarketing surveillance. The ratio of a drug's therapeutic unbound plasma concentration over the drug's in vitro potency against a given protein was used to select proteins most likely to be relevant to in vivo effects. In examining individual target bioactivities as predictors of AEs, we found a trade-off between the positive predictive value and the fraction of drugs with AEs that can be detected. However, considering sets of multiple targets for the same AE can help identify a greater fraction of AE-associated drugs. Of the 45 targets with statistically significant associations to AEs, 30 are included on existing safety target panels. The remaining 15 targets include 9 carbonic anhydrases, of which CA5B is significantly associated with cholestatic jaundice. We include the full quantitative data on associations between measured and predicted in vitro bioactivities and AEs in humans in this work, which can be used to make a more informed selection of safety profiling targets.
Collapse
Affiliation(s)
- Ines A Smit
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Avid M Afzal
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Chad H G Allen
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Fredrik Svensson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| | - Thierry Hanser
- Lhasa Limited, Granary Wharf House, 2 Canal Wharf, Leeds LS11 5PS, United Kingdom
| | - Andreas Bender
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, United Kingdom
| |
Collapse
|