1
|
Friedrichs GS, Abernathy MM, Ackley D, Clark M, DaSilva JK, Foley CM, Greiter-Wilke A, Henderson KA, Kremer JJ, Morimoto BH, Paglialunga S, Pugsley MK, Regan CP, Rossman EI, Segretti JA, Traebert M, Vargas HM, Wisialowski TA. Reevaluating safety pharmacology respiratory studies within the ICH S7A core battery: A multi-company evaluation of preclinical utility and clinical translation. Regul Toxicol Pharmacol 2024; 153:105706. [PMID: 39293707 DOI: 10.1016/j.yrtph.2024.105706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/09/2024] [Accepted: 09/16/2024] [Indexed: 09/20/2024]
Abstract
Optimization of ICH safety guideline studies for inclusion into regulatory submissions is critical for resource conservation, animal use reduction, and efficient drug development. The ICH S7A guidance for Safety Pharmacology (SP) studies adopted in 2001 identified the core battery of studies to evaluate the acute safety of putative pharmaceutical molecules prior to First in Human (FIH) trials. To assess the utility of respiratory studies in predicting clinical AE's, seven pharmaceutical companies pooled preclinical and clinical respiratory findings. A large database of novel molecules included all relevant data from standard S7A respiratory (n = 459) and FIH studies (n = 309). The data were analyzed with respect to the progression of these molecules, clinical adverse event reporting of these same molecules, and achieved exposures. These S7A respiratory assay findings had no impact on compound progression, and only 12 of 309 drug candidates were 'positive' preclinically and reported a respiratory-related AE in clinical trials (i.e. cough, dyspnea, etc.), an overall incidence rate of 3.9%. Contingency tables/statistics support a lack of concordance of these preclinical assays. Overall, our extensive analysis clearly indicated that the preclinical respiratory assay fails to provide any prognostic value for detecting clinically relevant respiratory adverse events.
Collapse
Affiliation(s)
- G S Friedrichs
- Novartis Biomedical Research, 1 Health Plaza, East Hanover, NJ 07936-1080, USA.
| | - M M Abernathy
- Eli Lilly Corporate Center, 893 Delaware St, Indianapolis, IN 46225, USA
| | - D Ackley
- Eli Lilly Corporate Center, 893 Delaware St, Indianapolis, IN 46225, USA
| | - M Clark
- Ability Biologics, 3 Place Ville-Marie Suite 400, Montréal, Québec H3B 4W8, Canada
| | - J K DaSilva
- Pfizer Research and Development, 445 Eastern Point Rd Groton, CT 06340, USA
| | - C M Foley
- Abbvie Inc, North Chicago, IL, 60064, USA
| | - A Greiter-Wilke
- Roche Innovation Center Basel, Grenzacherstr. 124, CH 4070 Basel, Switzerland
| | - K A Henderson
- Amgen Inc., Translational Safety & Bioanalytical Sciences, Thousand Oaks, CA 91320, USA
| | - J J Kremer
- Novartis BioMedical Research, 220 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - B H Morimoto
- Alto Neuroscience, Inc. 369 S San Antonio Rd, Los Altos CA 94022, USA
| | | | - M K Pugsley
- Cytokinetics, 350 Oyster Point Blvd, South San Francisco, CA 94080, USA
| | - C P Regan
- Merck & Co., Inc., West Point, PA 19486, USA
| | - E I Rossman
- GSK, 1250 Collegeville Avenue, Collegeville, PA 19426, USA
| | | | - M Traebert
- Novartis BioMedical Research, Postfach, 4002 Basel, Switzerland
| | - H M Vargas
- Amgen Inc., Translational Safety & Bioanalytical Sciences, Thousand Oaks, CA 91320, USA
| | - T A Wisialowski
- Pfizer Research and Development, 445 Eastern Point Rd Groton, CT 06340, USA
| |
Collapse
|
2
|
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
|
3
|
Gould S, Templin MV. Off target toxicities and links with physicochemical properties of medicinal products, including antibiotics, oligonucleotides, lipid nanoparticles (with cationic and/or anionic charges). Data review suggests an emerging pattern. Toxicol Lett 2023; 384:14-29. [PMID: 37454775 DOI: 10.1016/j.toxlet.2023.07.011] [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: 01/10/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023]
Abstract
Toxicology is an essential part of any drug development plan. Circumnavigating the risk of failure because of a toxicity issue can be a challenge, and failure in late development is extremely costly. To identify potential risks, it requires more than just understanding the biological target. The toxicologist needs to consider a compound's structure, it's physicochemical properties (including the impact of the overall formulation), as well as the biological target (e.g., receptor interactions). Understanding the impact of the physicochemical properties can be used to predict potential toxicities in advance by incorporating key endpoints in early screening strategies and/or used to compare toxicity profiles across lead candidates. This review discussed the risks of off-target and/or non-specific toxicities that may be associated with the physicochemical properties of compounds, especially those carrying dominant positive or negative charges, including amphiphilic small molecules, peptides, oligonucleotides and lipids/liposomes/lipid nanoparticles. The latter of which are being seen more and more in drug development, including the recent Covid pandemic, where mRNA and lipid nanoparticle technology is playing more of a role in vaccine development. The translation between non-clinical and clinical data is also considered, questioning how a physicochemical driven toxicity may be more universal across species, which means that such toxicity may be reassuringly translatable between species and as such, this information may also be considered as a support to the 3 R's, particularly in the early screening stages of a drug development plan.
Collapse
|
4
|
Liu X, Zhang Y, Ward LD, Yan Q, Bohnuud T, Hernandez R, Lao S, Yuan J, Fan F. A proteomic platform to identify off-target proteins associated with therapeutic modalities that induce protein degradation or gene silencing. Sci Rep 2021; 11:15856. [PMID: 34349202 PMCID: PMC8338952 DOI: 10.1038/s41598-021-95354-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/12/2021] [Indexed: 12/31/2022] Open
Abstract
Novel modalities such as PROTAC and RNAi have the ability to inadvertently alter the abundance of endogenous proteins. Currently available in vitro secondary pharmacology assays, which evaluate off-target binding or activity of small molecules, do not fully assess the off-target effects of PROTAC and are not applicable to RNAi. To address this gap, we developed a proteomics-based platform to comprehensively evaluate the abundance of off-target proteins. First, we selected off-target proteins using genetics and pharmacology evidence. This process yielded 2813 proteins, which we refer to as the “selected off-target proteome” (SOTP). An iterative algorithm was then used to identify four human cell lines out of 932. The 4 cell lines collectively expressed ~ 80% of the SOTP based on transcriptome data. Second, we used mass spectrometry to quantify the intracellular and extracellular proteins from the selected cell lines. Among over 10,000 quantifiable proteins identified, 1828 were part of the predefined SOTP. The SOTP was designed to be easily modified or expanded, owing to the rational selection process developed and the label free LC–MS/MS approach chosen. This versatility inherent to our platform is essential to design fit-for-purpose studies that can address the dynamic questions faced in investigative toxicology.
Collapse
Affiliation(s)
- Xin Liu
- Amgen Inc., Translational Safety and Bioanalytical Sciences, 360 Binney St., Cambridge, MA, 02142, USA.,Novartis Institutes for Biomedical Research, 500 Technology Square, Cambridge, MA, 02139, USA
| | - Ye Zhang
- Amgen Inc., Translational Safety and Bioanalytical Sciences, 360 Binney St., Cambridge, MA, 02142, USA.,Novartis Institutes for Biomedical Research, 500 Technology Square, Cambridge, MA, 02139, USA
| | - Lucas D Ward
- Amgen Inc., Translational Safety and Bioanalytical Sciences, 360 Binney St., Cambridge, MA, 02142, USA.,Alnylam Pharmaceuticals, 300 Third St., Cambridge, MA, 02142, USA
| | - Qinghong Yan
- Amgen Inc., Translational Safety and Bioanalytical Sciences, 360 Binney St., Cambridge, MA, 02142, USA.,Fosun Pharma, 104 Carnegie Center Drive, Suite 204, Princeton, NJ, 08540, USA
| | - Tanggis Bohnuud
- Amgen Inc., Translational Safety and Bioanalytical Sciences, 360 Binney St., Cambridge, MA, 02142, USA.,Beam Pharmaceuticals, 26 Landsdowne St., Cambridge, MA, 02139, USA
| | - Rocio Hernandez
- Amgen Inc., Translational Safety and Bioanalytical Sciences, 360 Binney St., Cambridge, MA, 02142, USA.,Amgen Inc., Translational Safety and Bioanalytical Sciences, 1 Amgen Center Dr., Thousand Oaks, CA, 91320, USA
| | - Socheata Lao
- Amgen Inc., Translational Safety and Bioanalytical Sciences, 360 Binney St., Cambridge, MA, 02142, USA.,Amgen Inc., Translational Safety and Bioanalytical Sciences, 1120 Veteran Blvd, South San Francisco, CA, 94080, USA
| | - Jing Yuan
- Amgen Inc., Translational Safety and Bioanalytical Sciences, 360 Binney St., Cambridge, MA, 02142, USA.,Drug Safety Research and Development, Pfizer Inc., 1 Portland St., Cambridge, MA, 02139, USA
| | - Fan Fan
- Amgen Inc., Translational Safety and Bioanalytical Sciences, 360 Binney St., Cambridge, MA, 02142, USA. .,Amgen Inc., Translational Safety and Bioanalytical Sciences, 1120 Veteran Blvd, South San Francisco, CA, 94080, USA.
| |
Collapse
|
5
|
Iachetta G, Colistra N, Melle G, Deleye L, Tantussi F, De Angelis F, Dipalo M. Improving reliability and reducing costs of cardiotoxicity assessments using laser-induced cell poration on microelectrode arrays. Toxicol Appl Pharmacol 2021; 418:115480. [PMID: 33689843 DOI: 10.1016/j.taap.2021.115480] [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: 11/27/2020] [Revised: 02/07/2021] [Accepted: 03/02/2021] [Indexed: 10/22/2022]
Abstract
Drug-induced cardiotoxicity is a major barrier to drug development and a main cause of withdrawal of marketed drugs. Drugs can strongly alter the spontaneous functioning of the heart by interacting with the cardiac membrane ion channels. If these effects only surface during in vivo preclinical tests, clinical trials or worse after commercialization, the societal and economic burden will be significant and seriously hinder the efficient drug development process. Hence, cardiac safety pharmacology requires in vitro electrophysiological screening assays of all drug candidates to predict cardiotoxic effects before clinical trials. In the past 10 years, microelectrode array (MEA) technology began to be considered a valuable approach in pharmaceutical applications. However, an effective tool for high-throughput intracellular measurements, compatible with pharmaceutical standards, is not yet available. Here, we propose laser-induced optoacoustic poration combined with CMOS-MEA technology as a reliable and effective platform to detect cardiotoxicity. This approach enables the acquisition of high-quality action potential recordings from large numbers of cardiomyocytes within the same culture well, providing reliable data using single-well MEA devices and single cardiac syncytia per each drug. Thus, this technology could be applied in drug safety screening platforms reducing times and costs of cardiotoxicity assessments, while simultaneously improving the data reliability.
Collapse
Affiliation(s)
| | - Nicolò Colistra
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Giovanni Melle
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Lieselot Deleye
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | | | | | - Michele Dipalo
- Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy.
| |
Collapse
|
6
|
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: 2.8] [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
|
7
|
Jenkinson S, Schmidt F, Rosenbrier Ribeiro L, Delaunois A, Valentin JP. A practical guide to secondary pharmacology in drug discovery. J Pharmacol Toxicol Methods 2020; 105:106869. [PMID: 32302774 DOI: 10.1016/j.vascn.2020.106869] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/21/2020] [Accepted: 04/03/2020] [Indexed: 01/29/2023]
Abstract
Secondary pharmacological profiling is increasingly applied in pharmaceutical drug discovery to address unwanted pharmacological side effects of drug candidates before entering the clinic. Regulators, drug makers and patients share a demand for deep characterization of secondary pharmacology effects of novel drugs and their metabolites. The scope of such profiling has therefore expanded substantially in the past two decades, leading to the implementation of broad in silico profiling methods and focused in vitro off-target screening panels, to identify liabilities, but also opportunities, as early as possible. The pharmaceutical industry applies such panels at all stages of drug discovery routinely up to early development. Nevertheless, target composition, screening technologies, assay formats, interpretation and scheduling of panels can vary significantly between companies in the absence of dedicated guidelines. To contribute towards best practices in secondary pharmacology profiling, this review aims to summarize the state-of-the art in this field. Considerations are discussed with respect to panel design, screening strategy, implementation and interpretation of the data, including regulatory perspectives. The cascaded, or integrated, use of in silico and off-target profiling allows to exploit synergies for comprehensive safety assessment of drug candidates.
Collapse
Affiliation(s)
- Stephen Jenkinson
- Drug Safety Research and Development, Pfizer Inc., La Jolla, CA 92121, United States of America.
| | - Friedemann Schmidt
- Sanofi, R&D Preclinical Safety, Industriepark Höchst, 65926 Frankfurt/Main, Germany
| | - Lyn Rosenbrier Ribeiro
- Medicines Discovery Catapult, Block 35, Mereside, Alderley Park, Alderley Edge, SK10 4TG, United Kingdom
| | - Annie Delaunois
- UCB BioPharma SRL, Early Solutions, Development Science, Non-Clinical Safety, 1420 Braine L'Alleud, Walloon Region, Belgium
| | - Jean-Pierre Valentin
- UCB BioPharma SRL, Early Solutions, Development Science, Non-Clinical Safety, 1420 Braine L'Alleud, Walloon Region, Belgium
| |
Collapse
|
8
|
Sameshima T, Yukawa T, Hirozane Y, Yoshikawa M, Katoh T, Hara H, Yogo T, Miyahisa I, Okuda T, Miyamoto M, Naven R. Small-Scale Panel Comprising Diverse Gene Family Targets To Evaluate Compound Promiscuity. Chem Res Toxicol 2019; 33:154-161. [PMID: 31461269 DOI: 10.1021/acs.chemrestox.9b00128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the recent advances in the life sciences and the remarkable investment in drug discovery research, the success rate of small-molecule drug development remains low. Safety is the second most influential factor of drug attrition in clinical studies; thus, the selection of compounds with fewer toxicity concerns is crucial to increase the success rate of drug discovery. Compounds that promiscuously bind to multiple targets are likely to cause unexpected pharmacological activity that may lead to adverse effects. Therefore, avoiding such compounds during early research stages would contribute to identifying compounds with a higher chance of success in the clinic. To evaluate the interaction profile against a wide variety of targets, we constructed a small-scale promiscuity panel (PP) consisting of eight targets (ROCK1, PDE4D2, GR, PPARγ, 5-HT2B, adenosine A3, M1, and GABAA) that were selected from diverse gene families. The validity of this panel was confirmed by comparison with the promiscuity index evaluated from larger-scale panels. Analysis of data from the PP revealed that both lipophilicity and basicity are likely to increase promiscuity, while the molecular weight does not significantly contribute. Additionally, the promiscuity assessed using our PP correlated with the occurrence of both in vitro cytotoxicity and in vivo toxicity, suggesting that the PP is useful to identify compounds with fewer toxicity concerns. In summary, this small-scale and cost-effective PP can contribute to the identification of safer compounds that would lead to a reduction in drug attrition due to safety issues.
Collapse
Affiliation(s)
- Tomoya Sameshima
- Research , Takeda Pharmaceutical Company Limited , Fujisawa 251-8555 , Japan
| | - Tomoya Yukawa
- Research , Takeda Pharmaceuticals International, Inc. , Cambridge , Massachusetts 02139 , United States
| | - Yoshihiko Hirozane
- Research , Takeda Pharmaceutical Company Limited , Fujisawa 251-8555 , Japan
| | - Masato Yoshikawa
- Research , Takeda Pharmaceutical Company Limited , Fujisawa 251-8555 , Japan
| | - Taisuke Katoh
- Research , Takeda Pharmaceutical Company Limited , Fujisawa 251-8555 , Japan
| | - Hideto Hara
- Research , Takeda Pharmaceutical Company Limited , Fujisawa 251-8555 , Japan
| | - Takatoshi Yogo
- Research , Takeda Pharmaceutical Company Limited , Fujisawa 251-8555 , Japan
| | - Ikuo Miyahisa
- Research , Takeda Pharmaceutical Company Limited , Fujisawa 251-8555 , Japan
| | - Teruaki Okuda
- Research , Takeda Pharmaceutical Company Limited , Fujisawa 251-8555 , Japan
| | - Makoto Miyamoto
- Research , Takeda Pharmaceutical Company Limited , Fujisawa 251-8555 , Japan
| | - Russell Naven
- Research , Takeda Pharmaceuticals International, Inc. , Cambridge , Massachusetts 02139 , United States.,Research , Takeda California , San Diego , California 92121 , United States
| |
Collapse
|
9
|
McAleer CW, Pointon A, Long CJ, Brighton RL, Wilkin BD, Bridges LR, Narasimhan Sriram N, Fabre K, McDougall R, Muse VP, Mettetal JT, Srivastava A, Williams D, Schnepper MT, Roles JL, Shuler ML, Hickman JJ, Ewart L. On the potential of in vitro organ-chip models to define temporal pharmacokinetic-pharmacodynamic relationships. Sci Rep 2019; 9:9619. [PMID: 31270362 PMCID: PMC6610665 DOI: 10.1038/s41598-019-45656-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 06/07/2019] [Indexed: 12/20/2022] Open
Abstract
Functional human-on-a-chip systems hold great promise to enable quantitative translation to in vivo outcomes. Here, we explored this concept using a pumpless heart only and heart:liver system to evaluate the temporal pharmacokinetic/pharmacodynamic (PKPD) relationship for terfenadine. There was a time dependent drug-induced increase in field potential duration in the cardiac compartment in response to terfenadine and that response was modulated using a metabolically competent liver module that converted terfenadine to fexofenadine. Using this data, a mathematical model was developed to predict the effect of terfenadine in preclinical species. Developing confidence that microphysiological models could have a transformative effect on drug discovery, we also tested a previously discovered proprietary AstraZeneca small molecule and correctly determined the cardiotoxic response to its metabolite in the heart:liver system. Overall our findings serve as a guiding principle to future investigations of temporal concentration response relationships in these innovative in vitro models, especially, if validated across multiple time frames, with additional pharmacological mechanisms and molecules representing a broad chemical diversity.
Collapse
Affiliation(s)
| | - Amy Pointon
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Christopher J Long
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - Rocky L Brighton
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - Benjamin D Wilkin
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - L Richard Bridges
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | | | - Kristin Fabre
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | - Robin McDougall
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | - Victorine P Muse
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | - Jerome T Mettetal
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Waltham, USA
| | | | - Dominic Williams
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Mark T Schnepper
- NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA
| | - Jeff L Roles
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - Michael L Shuler
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA
| | - James J Hickman
- Hesperos, Inc., 3259 Progress Dr., Room 158, Orlando, FL, 32826-3230, USA.
- NanoScience Technology Center, 12424 Research Parkway, Suite 400, Orlando, FL, 32826, USA.
| | - Lorna Ewart
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
| |
Collapse
|
10
|
Hurrell T, Segeritz CP, Vallier L, Lilley KS, Cromarty AD. Proteomic Comparison of Various Hepatic Cell Cultures for Preclinical Safety Pharmacology. Toxicol Sci 2019; 164:229-239. [PMID: 29635369 DOI: 10.1093/toxsci/kfy084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Experimental drugs need to be screened for safety within time constraints. Hepatotoxicity is one concerning contributor to the failure of investigational new drugs and a major rationale for postmarketing withdrawal decisions. Ethical considerations in preclinical research force the requirement for highly predictive in vitro assays using human tissue which retains functionality reflective of primary tissue. Here, the proteome of cells commonly used to assess preclinical hepatotoxicity was compared. Primary human hepatocytes (PHHs), hepatocyte-like cells (HLCs) differentiated from human pluripotent stem cells, HepG2 cell monolayers and HepG2 cell 3D spheroids were cultured and collected as whole cell lysates. Over 6000 proteins were identified and quantified in terms of relative abundance in replicate proteomic experiments using isobaric tagging methods. Comparison of these quantitative data provides biological insight into the feasibility of using HLCs, HepG2 monolayers, and HepG2 3D spheroids for hepatotoxicity testing. Collectively these data reveal how HLCs differentiated for 35 days and HepG2 cells proteomes differ from one another and that of PHHs. HepG2 cells possess a strong cancer cell signature and do not adequately express key metabolic proteins which mark the hepatic phenotype, this was not substantially altered by culturing as 3D spheroids. These data suggest that while no single hepatic model reflects the diverse array of outcomes required to mimic the in vivo liver functions, that HLCs are the most suitable investigational avenue for replacing PHHs in vitro.
Collapse
Affiliation(s)
- Tracey Hurrell
- Department of Pharmacology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria 0007, South Africa
| | - Charis-Patricia Segeritz
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Ludovic Vallier
- Wellcome Trust-Medical Research Council Stem Cell Institute, Anne McLaren Laboratory for Regenerative Medicine, Department of Surgery, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Kathryn S Lilley
- Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge CB2 1QR, UK
| | - Allan Duncan Cromarty
- Department of Pharmacology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Pretoria 0007, South Africa
| |
Collapse
|
11
|
Fan F, Toledo Warshaviak D, Hamadeh HK, Dunn RT. The integration of pharmacophore-based 3D QSAR modeling and virtual screening in safety profiling: A case study to identify antagonistic activities against adenosine receptor, A2A, using 1,897 known drugs. PLoS One 2019; 14:e0204378. [PMID: 30605479 PMCID: PMC6317804 DOI: 10.1371/journal.pone.0204378] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 12/12/2018] [Indexed: 12/23/2022] Open
Abstract
Safety pharmacology screening against a wide range of unintended vital targets using in vitro assays is crucial to understand off-target interactions with drug candidates. With the increasing demand for in vitro assays, ligand- and structure-based virtual screening approaches have been evaluated for potential utilization in safety profiling. Although ligand based approaches have been actively applied in retrospective analysis or prospectively within well-defined chemical space during the early discovery stage (i.e., HTS screening and lead optimization), virtual screening is rarely implemented in later stage of drug discovery (i.e., safety). Here we present a case study to evaluate ligand-based 3D QSAR models built based on in vitro antagonistic activity data against adenosine receptor 2A (A2A). The resulting models, obtained from 268 chemically diverse compounds, were used to test a set of 1,897 chemically distinct drugs, simulating the real-world challenge of safety screening when presented with novel chemistry and a limited training set. Due to the unique requirements of safety screening versus discovery screening, the limitations of 3D QSAR methods (i.e., chemotypes, dependence on large training set, and prone to false positives) are less critical than early discovery screen. We demonstrated that 3D QSAR modeling can be effectively applied in safety assessment prior to in vitro assays, even with chemotypes that are drastically different from training compounds. It is also worth noting that our model is able to adequately make the mechanistic distinction between agonists and antagonists, which is important to inform subsequent in vivo studies. Overall, we present an in-depth analysis of the appropriate utilization and interpretation of pharmacophore-based 3D QSAR models for safety screening.
Collapse
Affiliation(s)
- Fan Fan
- Amgen Research, Department of Comparative Biology and Safety Sciences, Thousand Oaks, CA, United States of America
- * E-mail:
| | - Dora Toledo Warshaviak
- Schrodinger Inc., San Diego, CA, United States of America
- Department of Molecular Engineering, Amgen Inc., Thousand Oaks, CA, United States of America
| | - Hisham K. Hamadeh
- Amgen Research, Department of Comparative Biology and Safety Sciences, Thousand Oaks, CA, United States of America
| | - Robert T. Dunn
- Amgen Research, Department of Comparative Biology and Safety Sciences, Thousand Oaks, CA, United States of America
| |
Collapse
|
12
|
Hurrell T, Lilley KS, Cromarty AD. Proteomic responses of HepG2 cell monolayers and 3D spheroids to selected hepatotoxins. Toxicol Lett 2019; 300:40-50. [DOI: 10.1016/j.toxlet.2018.10.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 09/29/2018] [Accepted: 10/25/2018] [Indexed: 01/26/2023]
|
13
|
Uteng M, Urban L, Brees D, Muller PY, Kullak-Ublick GA, Bouchard P, Tougas G, Chibout SD. Safety differentiation: emerging competitive edge in drug development. Drug Discov Today 2018; 24:285-292. [PMID: 30244081 DOI: 10.1016/j.drudis.2018.09.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/24/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022]
Abstract
With increasing expectations to provide evidence of drug efficacy, safety, and cost-effectiveness, best-in-class drugs are a major value driver for the pharmaceutical industry. Superior safety is a key differentiation criterion that could be achieved through better risk:benefit profiles, safety margins, fewer contraindications, and improved patient compliance. To accomplish this, comparative safety assessments using innovative and adaptive nonclinical and clinical outcome-based approaches should be undertaken, and continuous strategic adjustments must be made as the risk:benefit profiles evolve. Key success criteria include scientific expertise and integration between all disciplines during the full extent of the drug development process.
Collapse
Affiliation(s)
- Marianne Uteng
- Novartis Institutes for Biomedical Research, Translational Medicine, Pre-Clinical Safety, Basel, Switzerland.
| | - Laszlo Urban
- Novartis Institutes for Biomedical Research, Translational Medicine, Pre-Clinical Safety, Cambridge, MA, USA
| | - Dominique Brees
- Novartis Institutes for Biomedical Research, Translational Medicine, Pre-Clinical Safety, Basel, Switzerland
| | | | - Gerd A Kullak-Ublick
- Novartis Pharma AG, Global Drug Development, Chief Medical Office and Patient Safety, Basel, Switzerland; Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Page Bouchard
- Novartis Institutes for Biomedical Research, Translational Medicine, Pre-Clinical Safety, Cambridge, MA, USA
| | - Gervais Tougas
- Novartis Pharma AG, Global Drug Development, Chief Medical Office and Patient Safety, Basel, Switzerland
| | - Salah-Dine Chibout
- Novartis Institutes for Biomedical Research, Translational Medicine, Pre-Clinical Safety, Basel, Switzerland
| |
Collapse
|
14
|
Mead AN, Amouzadeh HR, Chapman K, Ewart L, Giarola A, Jackson SJ, Jarvis P, Jordaan P, Redfern W, Traebert M, Valentin JP, Vargas HM. Assessing the predictive value of the rodent neurofunctional assessment for commonly reported adverse events in phase I clinical trials. Regul Toxicol Pharmacol 2016; 80:348-57. [DOI: 10.1016/j.yrtph.2016.05.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/20/2016] [Accepted: 05/02/2016] [Indexed: 11/28/2022]
|
15
|
Valentin JP, Guth B, Hamlin RL, Lainée P, Sarazan D, Skinner M. Functional Cardiac Safety Evaluation of Novel Therapeutics. METHODS AND PRINCIPLES IN MEDICINAL CHEMISTRY 2015. [DOI: 10.1002/9783527673643.ch10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
16
|
Secondary pharmacology data to assess potential off-target activity of new drugs: a regulatory perspective. Nat Rev Drug Discov 2015; 14:294. [PMID: 25792260 DOI: 10.1038/nrd3845-c1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
17
|
Abstract
Whereas pharmacological responses tend to be fairly rapid in onset and are therefore detectable after a single dose, some diminish on repeated dosing, and others increase in magnitude and therefore can be missed or underestimated in single-dose safety pharmacology studies. Safety pharmacology measurements can be incorporated into repeat-dose toxicity studies, either routinely or on an ad hoc basis. Drivers for this are both scientific (see above) and regulatory (e.g. ICH S6, S7, S9). There are inherent challenges in achieving this: the availability of suitable technical and scientific expertise in the test facility, unsuitable laboratory conditions, use of simultaneous (as opposed to staggered) dosing, requirement for toxicokinetic sampling, unsuitability of certain techniques (e.g. use of anaesthesia, surgical implantation, food restriction), equipment availability at close proximity and sensitivity of the methods to detect small, clinically relevant, changes. Nonetheless, 'fit-for-purpose' data can still be acquired without requiring additional animals. Examples include assessment of behaviour, sensorimotor, visual and autonomic functions, ambulatory ECG and blood pressure, echocardiography, respiratory, gastrointestinal, renal and hepatic function. This is entirely achievable if the safety pharmacology measurements are relatively unobtrusive, both with respect to the animals and to the toxicology study itself. Careful pharmacological validation of any methods used, and establishing their detection sensitivity, is vital to ensure the credibility of generated data.
Collapse
Affiliation(s)
- Will S Redfern
- Drug Safety and Metabolism, AstraZeneca R&D, Darwin Building, 310 Cambridge Science Park, Cambridge, CB4 0WG, UK,
| |
Collapse
|
18
|
Graham KL, Zhang JV, Lewén S, Burke TM, Dang T, Zoudilova M, Sobel RA, Butcher EC, Zabel BA. A novel CMKLR1 small molecule antagonist suppresses CNS autoimmune inflammatory disease. PLoS One 2014; 9:e112925. [PMID: 25437209 PMCID: PMC4249827 DOI: 10.1371/journal.pone.0112925] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 10/22/2014] [Indexed: 12/27/2022] Open
Abstract
Therapies that target leukocyte trafficking pathways can reduce disease activity and improve clinical outcomes in multiple sclerosis (MS). Experimental autoimmune encephalomyelitis (EAE) is a widely studied animal model that shares many clinical and histological features with MS. Chemokine-like receptor-1 (CMKLR1) is a chemoattractant receptor that is expressed by key effector cells in EAE and MS, including macrophages, subsets of dendritic cells, natural killer cells and microglia. We previously showed that CMKLR1-deficient (CMKLR1 KO) mice develop less severe clinical and histological EAE than wild-type mice. In this study, we sought to identify CMKLR1 inhibitors that would pharmaceutically recapitulate the CMKLR1 KO phenotype in EAE. We identified 2-(α-naphthoyl) ethyltrimethylammonium iodide (α-NETA) as a CMKLR1 small molecule antagonist that inhibits chemerin-stimulated β-arrestin2 association with CMKLR1, as well as chemerin-triggered CMKLR1+ cell migration. α-NETA significantly delayed the onset of EAE induced in C57BL/6 mice by both active immunization with myelin oligodendrocyte glycoprotein peptide 35-55 and by adoptive transfer of encephalitogenic T cells. In addition, α-NETA treatment significantly reduced mononuclear cell infiltrates within the CNS. This study provides additional proof-of-concept data that targeting CMKLR1:chemerin interactions may be beneficial in preventing or treating MS.
Collapse
MESH Headings
- Animals
- Arrestins/metabolism
- Brain/drug effects
- Brain/metabolism
- Cell Movement/drug effects
- Chemokines/metabolism
- Drug Evaluation, Preclinical
- Drug Stability
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Humans
- Intercellular Signaling Peptides and Proteins/metabolism
- Leukocytes/drug effects
- Mice
- Mice, Inbred C57BL
- Naphthalenes/adverse effects
- Naphthalenes/chemistry
- Naphthalenes/pharmacology
- Naphthalenes/therapeutic use
- Quaternary Ammonium Compounds/adverse effects
- Quaternary Ammonium Compounds/chemistry
- Quaternary Ammonium Compounds/pharmacology
- Quaternary Ammonium Compounds/therapeutic use
- Receptors, Chemokine
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Safety
- Spinal Cord/drug effects
- Spinal Cord/metabolism
- Structure-Activity Relationship
- beta-Arrestins
Collapse
Affiliation(s)
- Kareem L Graham
- Palo Alto Veterans Institute for Research and Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Jian V Zhang
- Palo Alto Veterans Institute for Research and Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Susanna Lewén
- Palo Alto Veterans Institute for Research and Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Thomas M Burke
- Palo Alto Veterans Institute for Research and Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| | - Ton Dang
- ChemoCentryx, Inc., Mountain View, California, United States of America
| | - Maria Zoudilova
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Raymond A Sobel
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Eugene C Butcher
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research and Veterans Affairs Palo Alto Health Care System, Palo Alto, California, United States of America
| |
Collapse
|
19
|
Keating C, Ewart L, Grundy L, Valentin JP, Grundy D. Translational potential of a mouse in vitro bioassay in predicting gastrointestinal adverse drug reactions in Phase I clinical trials. Neurogastroenterol Motil 2014; 26:980-9. [PMID: 24813024 PMCID: PMC4207192 DOI: 10.1111/nmo.12349] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 03/28/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND Motility-related gastrointestinal (GI) adverse drug reactions (GADRs) such as diarrhea and constipation are a common and deleterious feature associated with drug development. Novel biomarkers of GI function are therefore required to aid decision making on the GI liability of compounds in development. METHODS Fifteen compounds associated with or without clinical GADRs were used to assess the ability of an in vitro colonic motility bioassay to predict motility-related GADRs. Compounds were examined in a blinded fashion for their effects on mouse colonic peristaltic motor complexes in vitro. For each compound concentration-response relationships were determined and the results compared to clinical data. Compounds were also assessed using GI transit measurements obtained using an in vivo rat charcoal meal model. KEY RESULTS Within a clinically relevant dosing range, the in vitro assay identified five true and three false positives, four true and three false negatives, which gave a predictive capacity of 60%. The in vivo assay detected four true and four false positives, four false and three true negatives, giving rise to a predictive capacity for this model of 47%. CONCLUSIONS & INFERENCES Overall these results imply that both assays are poor predictors of GADRs. Further analysis would benefit from a larger compound set, but the data show a clear need for improved models for use in safety pharmacology assessment of GI motility.
Collapse
Affiliation(s)
- C Keating
- Department of Biomedical Sciences, University of SheffieldSheffield, UK
| | - L Ewart
- Department of Safety Pharmacology, Global Safety Assessment, AstraZeneca R&D Alderley ParkMacclesfield, UK
| | - L Grundy
- Department of Biomedical Sciences, University of SheffieldSheffield, UK
| | - JP Valentin
- Department of Safety Pharmacology, Global Safety Assessment, AstraZeneca R&D Alderley ParkMacclesfield, UK
| | - D Grundy
- Department of Biomedical Sciences, University of SheffieldSheffield, UK
| |
Collapse
|
20
|
Springer C, Sokolnicki KL. A fingerprint pair analysis of hERG inhibition data. Chem Cent J 2013; 7:167. [PMID: 24144230 PMCID: PMC3854090 DOI: 10.1186/1752-153x-7-167] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/10/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Drugs that bind to the human Ether-a-go-go Related Gene (hERG) potassium channel and block its ion conduction can lead to Torsade de Pointes (TdP), a fatal ventricular arrhythmia. Thus, compounds are screened for hERG inhibition in the drug development process; those found to be active face a difficult road to approval. Knowing which structural transformations reduce hERG binding would be helpful in the lead optimization phase of drug discovery. RESULTS To identify such transformations, we carried out a comprehensive analysis of all approximately 33,000 compound pairs in the Novartis internal database which have IC50 values in the dofetilide displacement assay. Most molecular transformations have only a single example in the data set; however, a few dozen transformations have sufficient numbers for statistical analysis. CONCLUSIONS We observe that transformations which increased polarity (for example adding an oxygen, or an sp2 nitrogen), decreased lipophilicity (removing carbons), or decreased positive charge consistently reduced hERG inhibition between 3- and 10-fold. The largest observed reduction in hERG was from a transformation from imidazole to methyl tetrazole. We also observe that some changes in aromatic ring substituents (for example hydrogen to methoxy) can also reduce hERG binding in vitro.
Collapse
Affiliation(s)
- Clayton Springer
- Novartis Institutes for BioMedical Research, 100 Technology Square, Cambridge, MA 02139, USA.
| | | |
Collapse
|
21
|
Harris K, Aylott M, Cui Y, Louttit JB, McMahon NC, Sridhar A. Comparison of Electrophysiological Data From Human-Induced Pluripotent Stem Cell–Derived Cardiomyocytes to Functional Preclinical Safety Assays. Toxicol Sci 2013; 134:412-26. [DOI: 10.1093/toxsci/kft113] [Citation(s) in RCA: 177] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
22
|
Redfern WS, Ewart LC, Lainée P, Pinches M, Robinson S, Valentin JP. Functional assessments in repeat-dose toxicity studies: the art of the possible. Toxicol Res (Camb) 2013. [DOI: 10.1039/c3tx20093k] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
23
|
Reducing safety-related drug attrition: the use of in vitro pharmacological profiling. Nat Rev Drug Discov 2012. [DOI: 10.1038/nrd3845] [Citation(s) in RCA: 470] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
An JC, Kwon KR. Effects of Sweet Bee Venom on the Central Nervous System in Rats -using the Functional Observational Battery-. J Pharmacopuncture 2011. [DOI: 10.3831/kpi.2011.14.3.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
25
|
Redfern WS, Valentin JP. Trends in safety pharmacology: Posters presented at the annual meetings of the Safety Pharmacology Society 2001–2010. J Pharmacol Toxicol Methods 2011; 64:102-10. [DOI: 10.1016/j.vascn.2011.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Accepted: 05/13/2011] [Indexed: 01/01/2023]
|
26
|
Valentin JP, Pollard C, Lainée P, Hammond T. Value of non-clinical cardiac repolarization assays in supporting the discovery and development of safer medicines. Br J Pharmacol 2010; 159:25-33. [PMID: 20141518 DOI: 10.1111/j.1476-5381.2009.00530.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Non-clinical QT-related assays aligned to the pharmaceutical drug discovery and development phases are used in several ways. During the early discovery phases, assays are used for hazard identification and wherever possible for hazard elimination. The data generated enable us to: (i) establish structure-activity relationships and thereby; (ii) influence the medicinal chemistry design and provide tools for effective decision making; and provide structure-activity data for in silico predictive databases; (iii) solve problems earlier; (iv) provide reassurance for compound or project to progress; and (v) refine strategies as scientific and technical knowledge grows. For compounds progressing into pre-clinical development, the 'core battery' QT-related data enable an integrated risk assessment to: (i) fulfil regulatory requirements; (ii) assess the safety and risk-benefit for compound progression to man; (iii) contribute to defining the starting dose during the phase I clinical trials; (iv) influence the design of the phase I clinical trials; (v) identify clinically relevant safety biomarkers; and (vi) contribute to the patient risk management plan. Once a compound progresses into clinical development, QT-related data can be applied in the context of risk management and risk mitigation. The data from 'follow-up' studies can be used to: (i) support regulatory approval; (ii) investigate discrepancies that may have emerged within and/or between non-clinical and clinical data; (iii) understand the mechanism of an undesirable pharmacodynamic effect; (iv) provide reassurance for progression into multiple dosing in humans and/or large-scale clinical trials; and (v) assess drug-drug interactions. Based on emerging data, the integrated risk assessment is then reviewed in this article, and the benefit-risk for compound progression was re-assessed. Project examples are provided to illustrate the impact of non-clinical data to support compound progression throughout the drug discovery and development phases, and regulatory approval.
Collapse
Affiliation(s)
- Jean-Pierre Valentin
- Safety Assessment UK, AstraZeneca, Mereside, Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK.
| | | | | | | |
Collapse
|
27
|
Hamon J, Whitebread S. In Vitro
Safety Pharmacology Profiling: An Important Tool to Decrease Attrition. ACTA ACUST UNITED AC 2010. [DOI: 10.1002/9783527627448.ch12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
28
|
Cavero I. Exploratory Safety Pharmacology: a new safety paradigm to de-risk drug candidates prior to selection for regulatory science investigations. Expert Opin Drug Saf 2009; 8:627-47. [DOI: 10.1517/14740330903362422] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
29
|
Valentin JP, Bialecki R, Ewart L, Hammond T, Leishmann D, Lindgren S, Martinez V, Pollard C, Redfern W, Wallis R. A framework to assess the translation of safety pharmacology data to humans. J Pharmacol Toxicol Methods 2009; 60:152-8. [DOI: 10.1016/j.vascn.2009.05.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 05/05/2009] [Indexed: 11/16/2022]
|
30
|
Report and recommendations of the workshop of the European Centre for the Validation of Alternative Methods for Drug-Induced Cardiotoxicity. Cardiovasc Toxicol 2009; 9:107-25. [PMID: 19572114 DOI: 10.1007/s12012-009-9045-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 04/17/2009] [Indexed: 10/20/2022]
Abstract
Cardiotoxicity is among the leading reasons for drug attrition and is therefore a core subject in non-clinical and clinical safety testing of new drugs. European Centre for the Validation of Alternative Methods held in March 2008 a workshop on "Alternative Methods for Drug-Induced Cardiotoxicity" in order to promote acceptance of alternative methods reducing, refining or replacing the use of laboratory animals in this field. This review reports the outcome of the workshop. The participants identified the major clinical manifestations, which are sensitive to conventional drugs, to be arrhythmias, contractility toxicity, ischaemia toxicity, secondary cardiotoxicity and valve toxicity. They gave an overview of the current use of alternative tests in cardiac safety assessments. Moreover, they elaborated on new cardiotoxicological endpoints for which alternative tests can have an impact and provided recommendations on how to cover them.
Collapse
|
31
|
Valentin JP, Hammond T. Safety and secondary pharmacology: Successes, threats, challenges and opportunities. J Pharmacol Toxicol Methods 2008; 58:77-87. [DOI: 10.1016/j.vascn.2008.05.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Accepted: 05/19/2008] [Indexed: 12/15/2022]
|
32
|
Harmer A, Abi-Gerges N, Easter A, Woods A, Lawrence C, Small B, Valentin JP, Pollard C. Optimisation and validation of a medium-throughput electrophysiology-based hNav1.5 assay using IonWorks™. J Pharmacol Toxicol Methods 2008; 57:30-41. [DOI: 10.1016/j.vascn.2007.09.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2007] [Accepted: 09/15/2007] [Indexed: 10/22/2022]
|
33
|
Moscardo E, Maurin A, Dorigatti R, Champeroux P, Richard S. An optimised methodology for the neurobehavioural assessment in rodents. J Pharmacol Toxicol Methods 2007; 56:239-55. [PMID: 17629504 DOI: 10.1016/j.vascn.2007.03.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2007] [Accepted: 03/30/2007] [Indexed: 10/23/2022]
Abstract
INTRODUCTION The most widely used test to identify undesired effects of drugs on the central and the peripheral nervous system is the neurobehavioural observation battery adapted from that first described by Irwin in mice. As a neurobehavioural assessment is based on observations; thus, all factors involved need to be controlled and standardised to make the data collected objective, reproducible, reliable and predictive of safety liabilities. METHODS An observation battery comprising 58 signs with assigned full details of numerical scores was defined, and a standard design with associated recording, presenting and analysing data system was established. Validation studies were conducted with chlorpromazine, amphetamine, diazepam or clonidine given orally to rats or mice, in order to assess if this methodology could clearly differentiate the profile of effects produced by these compounds. The analysis of data from 80 control rats allowed for the assessment of the normal behaviour in order to characterise the inter-individual, daytime-related variability and the habituation of animals to the procedure. RESULTS The reference compounds induced their typical and expected transient effects on neurobehaviour, observed both in the home cage and open-arena, and on body temperature. In particular, amphetamine induced a stimulation of the nervous system activities and marked hyperthermia. Chlorpromazine, diazepam and clonidine induced depressive, anxiolytic or sedative effects associated with hypothermia. The analysis of data collected in control animals allowed for the identification of 6 signs which scored differently from the assigned normality at the first handling occasion due to the characteristic fear reactions to the unknown, and 9 signs at 8 h post-dose due to the animal's habituation to experimental conditions and handling. DISCUSSION The neurobehavioural changes expected by reference compounds administration were detected. These results confirm that by using this methodology the normal behaviour of the rat and the mouse, the daytime-related variability and the habituation of animals can be characterised, allowing a refined, reliable and reproducible neurobehavioural assessment of test substances in rodents.
Collapse
Affiliation(s)
- Elena Moscardo
- Safety Pharmacology, Safety Assessment Department, GlaxoSmithKline R&D Centre, Via Fleming 4, VERONA (37135), Italy.
| | | | | | | | | |
Collapse
|
34
|
Easter A, Sharp TH, Valentin JP, Pollard CE. Pharmacological validation of a semi-automated in vitro hippocampal brain slice assay for assessment of seizure liability. J Pharmacol Toxicol Methods 2007; 56:223-33. [PMID: 17600733 DOI: 10.1016/j.vascn.2007.04.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2007] [Accepted: 04/16/2007] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Drug-induced seizures are a serious, life-threatening adverse drug reaction (ADR) that can result in the failure of drugs to be licensed for clinical use or withdrawn from the market. Seizure liability of potential drugs is traditionally assessed using animal models run during the later phases of the drug discovery process. Given the low throughput, high animal usage and high compound requirement associated with these assays, it would be advantageous to identify higher throughput, in vitro models that could be used to give an earlier assessment of seizure liability. The hippocampal brain slice is one possibility but conventionally allows recording from only one slice at a time. The aim of this study was to validate a semi-automated system (Slicemaster, Scientifica UK Ltd) which allows concurrent electrophysiological recording from multiple brain slices. METHODS Conventional electrophysiological recording techniques were used to record electrically evoked synaptic activity from rat hippocampal brain slices. Population spikes (PS) were evoked at 30 s intervals by electrical stimulation of the Schaffer collateral pathway and were recorded using extracellular electrodes positioned in the CA1 cell body layer. Responses were quantified as PS areas (the area above and below the 0 mV line). The effects of eight validation compounds known to cause seizures in vivo and/or in the clinic were assessed. RESULTS Seven out of eight compounds evoked a concentration-dependent increase in population spike (PS) area that was statistically significant at higher concentrations (P<0.05; ANOVA). At the highest test concentration the percentage effects (mean+/-s.e.m.), relative to vehicle, were: picrotoxin 212.9+/-28.8, pentylenetrazole (PTZ) 181.4+/-24.7, 4-AP 328.9+/-48.6, aminophylline 124.5+/-5.9, chlorpromazine 122.1+/-9.8, SNC-80 132.1+/-12.6 and penicillin 174.7+/-14.1. Physostigmine had no significant effect on PS area although a concentration-dependent change in the morphology of the response was evident. DISCUSSION All validation compounds evoked a statistically significant effect on synaptic activity in the rat hippocampal slice. Although similar effects have been described previously, this is the first time that the effects of a pharmacologically diverse set of compounds have been assessed using a standardised brain slice assay. Given the low compound usage and relatively high throughput associated with this assay, the hippocampal brain slice assay may facilitate earlier testing of convulsant liability than is currently possible using in vivo models.
Collapse
Affiliation(s)
- A Easter
- Safety Assessment UK, AstraZeneca R&D Alderley Park, Macclesfield, SK10 4TG, UK.
| | | | | | | |
Collapse
|
35
|
Faller B, Wang J, Zimmerlin A, Bell L, Hamon J, Whitebread S, Azzaoui K, Bojanic D, Urban L. High-throughputinvitroprofiling assays: lessons learnt from experiences at Novartis. Expert Opin Drug Metab Toxicol 2006; 2:823-33. [PMID: 17125403 DOI: 10.1517/17425255.2.6.823] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
This article reviews the use of a selection of in vitro assays implemented at Novartis and intends to address exposure and safety in early drug discovery. The authors' own experience, based on a large number of 'real' drug discovery compounds, is described to reflect on what has worked, where improvement is needed and how to best use the data for decision making. Possible strategies are discussed, and guidelines are provided on how to organise assays, extract value and contribute knowledge from the data.
Collapse
Affiliation(s)
- Bernard Faller
- Novartis Institutes for BioMedical Research, Basel, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Nolan ER, Feng MR, Koup JR, Liu J, Turluck D, Zhang Y, Paulissen JB, Olivier NB, Miller T, Bailie MB. A novel predictive pharmacokinetic/pharmacodynamic model of repolarization prolongation derived from the effects of terfenadine, cisapride and E-4031 in the conscious chronic av node—ablated, His bundle-paced dog. J Pharmacol Toxicol Methods 2006; 53:1-10. [PMID: 16399550 DOI: 10.1016/j.vascn.2005.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Accepted: 02/17/2005] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Terfenadine, cisapride, and E-4031, three drugs that prolong ventricular repolarization, were selected to evaluate the sensitivity of the conscious chronic atrioventricular node--ablated, His bundle-paced Dog for defining drug induced cardiac repolarization prolongation. A novel predictive pharmacokinetic/pharmacodynamic model of repolarization prolongation was generated from these data. METHODS Three male beagle dogs underwent radiofrequency AV nodal ablation, and placement of a His bundle-pacing lead and programmable pacemaker under anesthesia. Each dog was restrained in a sling for a series of increasing dose infusions of each drug while maintained at a constant heart rate of 80 beats/min. RT interval, a surrogate for QT interval in His bundle-paced dogs, was recorded throughout the experiment. RESULTS E-4031 induced a statistically significant RT prolongation at the highest three doses. Cisapride resulted in a dose-dependent increase in RT interval, which was statistically significant at the two highest doses. Terfenadine induced a dose-dependent RT interval prolongation with a statistically significant change occurring only at the highest dose. The relationship between drug concentration and RT interval change was described by a sigmoid E(max) model with an effect site. Maximum RT change (E(max)), free drug concentration at half of the maximum effect (EC(50)), and free drug concentration associated with a 10 ms RT prolongation (EC(10 ms)) were estimated. A linear correlation between EC(10 ms) and HERG IC(50) values was identified. DISCUSSION The conscious dog with His bundle-pacing detects delayed cardiac repolarization related to I(Kr) inhibition, and detects repolarization change induced by drugs with activity at multiple ion channels. A clinically relevant sensitivity and a linear correlation with in vitro HERG data make the conscious His bundle-paced dog a valuable tool for detecting repolarization effect of new chemical entities.
Collapse
Affiliation(s)
- Emily R Nolan
- Pfizer Global Research and Development, Ann Arbor, MI 48105, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gerloff T, Roots I. [Implications of pharmacogenetics in every-day practice]. Internist (Berl) 2005; 46:1270-7. [PMID: 16170512 DOI: 10.1007/s00108-005-1489-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Pharmacogenetics as one of the areas of clinical pharmacology addresses hereditary factors involved in individually different responses to drugs. Clinical trials combined with molecular genetics seek for underlying reasons influencing efficacy and toxicity of drugs. The declared goal of pharmacogenetics is to provide physicians with knowledge and tools to allow an individualized patient-directed pharmacotherapy. This concept is best evolved for clinical practice in the field of drug-metabolizing enzymes, especially for the cytochromes P450 (CYP) 2D6, CYP2C19 and thiopurine S-methyltransferase (TPMT). Patients with inherited enzyme deficiencies are at risk to accumulate excessive drug concentrations when treated with standard doses which may lead to adverse drug reactions or even to life-threatening conditions. Genetic factors are also involved in drug-target interactions (e. g. receptors). Prospective controlled clinical trials are needed to evaluate the benefit of pharmacogenetics for therapy outcome and to define its role in clinical practice.
Collapse
Affiliation(s)
- T Gerloff
- Institut für Klinische Pharmakologie, Charité Universitätsmedizin Berlin.
| | | |
Collapse
|
38
|
Redfern WS, Strang I, Storey S, Heys C, Barnard C, Lawton K, Hammond TG, Valentin JP. Spectrum of effects detected in the rat functional observational battery following oral administration of non-CNS targeted compounds. J Pharmacol Toxicol Methods 2005; 52:77-82. [PMID: 15936219 DOI: 10.1016/j.vascn.2005.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2005] [Accepted: 04/05/2005] [Indexed: 10/25/2022]
Abstract
INTRODUCTION The Functional Observational Battery (FOB) is a systematic evaluation of nervous system function in the rat, comprising more than 30 parameters across autonomic, neuromuscular, sensorimotor and behavioural domains. We have collated FOB outcomes from 50 compounds that were not targeted at CNS disorders, and would therefore be anticipated to have relatively few CNS side-effects, for evaluation of the FOB as part of the safety pharmacology 'core battery'. METHODS Male Han Wistar rats (200-300 g) were used, with n=6 per treatment group. Each compound was tested acutely at 3 dose levels (oral route), from the therapeutic dose up to either 100 times this dose or to the maximal tolerated dose (MTD). A vehicle control group was included in each study. RESULTS Effects were detected in the FOB for 94% of compounds tested. The commonest effects were weight loss/decreased body weight gain overnight post-dose (46% of compounds), and changes in core temperature (36%). Dose-related effects were observed with 62% of compounds; the commonest was decreased body weight gain (32%), followed by effects on tail flick latency (14%), landing foot splay (12%), decreased rectal temperature (10%), time to exit the centre circle in the open field (10%), diarrhoea/loose faeces (8%), respiratory effects (4%), grasping reflex (4%) and supported rears in the open field (4%). Remaining parameters were affected by < or =2% of compounds. DISCUSSION The value of doing the FOB as part of the safety pharmacology 'core battery' is emphasised by the fact that, even for non-CNS targeted compounds, the majority affected at least one of the parameters in the FOB. These data may also help to anticipate the most frequently required 'follow-up' studies.
Collapse
Affiliation(s)
- William S Redfern
- Safety Assessment UK, AstraZeneca R&D Alderley Park, Cheshire SK10 4TG, UK.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Ducroq J, Printemps R, Le Grand M. Additive effects of ziprasidone and d,l-sotalol on the action potential in rabbit Purkinje fibres and on the hERG potassium current. J Pharmacol Toxicol Methods 2005; 52:115-22. [PMID: 15922632 DOI: 10.1016/j.vascn.2005.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
INTRODUCTION Ziprasidone, an atypical antipsychotic has been shown to be devoid of cardiac adverse effects in spite of its propensity to prolong the QT-interval via a hERG current inhibition. However, the effects of ziprasidone on the action potential (AP) parameters have not been published yet. Moreover, very little information is available concerning pharmacodynamic interactions between ziprasidone and other hERG channel blockers. Thus, we investigated the putative interaction between ziprasidone and D,L-sotalol on the hERG channels at therapeutic concentrations and their consequences on the action potential prolongation. METHODS AP were recorded at 1 and 0.2 Hz. Increasing concentrations of ziprasidone (0.01-10 micromol/L) were successively superfused for 30 min alone or in D,L-sotalol 10 micromol/L pre-treated fibres. Moreover, the effects of ziprasidone, alone or in association with d,l-sotalol, were investigated on the hERG current. RESULTS Ziprasidone (1-10 microM) induced a concentration and reverse frequency-dependent increase in APD(90) (APD(90): +27% and +36%, respectively at 1 Hz and +50% and +70%, respectively at 0.2 Hz) due to a hERG current blockade (IC50: 0.24 micromol/L). A pre-treatment with D,L-sotalol 10 micromol/L led to an increase in APD(90) of +23% at 1 Hz, stable at 66+/-4 min. In these pre-treated fibres, ziprasidone (1 and 10 micromol/L) induced an additional AP prolongation (APD(90): +16% and +18%, respectively at 1 Hz) as compared to D,L-sotalol pre-treatment. Moreover, D,L-sotalol did not interact with the pharmacological profile of ziprasidone on the hERG channel. CONCLUSION The present study demonstrates that ziprasidone induces an AP prolongation due to its propensity to block the hERG channel. Moreover, ziprasidone and d,l-sotalol, superfused concomitantly exhibit additive effects on the AP duration since they do not interact as competitors for the hERG channel.
Collapse
Affiliation(s)
- Joffrey Ducroq
- Physiostim, Z.I. de Brénas, Lautrec, 81440 Lautrec, France.
| | | | | |
Collapse
|
40
|
Cavero I, Crumb W. ICH S7B draft guideline on the non-clinical strategy for testing delayed cardiac repolarisation risk of drugs: a critical analysis. Expert Opin Drug Saf 2005; 4:509-30. [PMID: 15934857 DOI: 10.1517/14740338.4.3.509] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The International Conference on Harmonization (ICH) stems from the initiative of three major world partners (Japan, USA, European Community) who composed a mutually accepted body of regulations concerning the safety, quality and efficacy requirements that new medicines have to meet in order to receive market approval. Documents on non-clinical safety pharmacology already composed by this organisation include two guidelines: the S7A adopted in 2000 and, its companion, the S7B guideline, in a draft form since 2001. The S7A guideline deals with general principles and recommendations on safety pharmacology studies designed to protect healthy volunteers and patients from potential drug-induced adverse reactions. The S7B recommends a general non-clinical testing strategy for determining the propensity of non-cardiovascular pharmaceuticals to delay ventricular repolarisation, an effect that at times progresses into life-threatening ventricular arrhythmia. In the most recent version of this document (June 2004), the strategy proposes experimental assays and a critical examination of other pertinent information for applying an 'evidence of risk' label to a compound. Regrettably, the guideline fails to deal satisfactorily with a number of crucial issues such as scoring the evidence of risk and the clinical consequences of such scoring. However, in the latter case, the S7B relies on the new ICH guideline E14 which is currently in preparation. E14 is the clinical counterpart of the S7B guideline which states that non-clinical data are a poor predictor of drug-induced repolarisation delay in humans. The present contribution summarises and assesses salient aspects of the S7A guideline as its founding principles are also applicable to the S7B guideline. The differences in strategies proposed by the various existing drafts of the latter document are critically examined together with some unresolved, crucial problems. The need for extending the objective of the S7B document to characterise the full electrophysiological profile of new pharmaceuticals is argued as this approach would more extensively assess the non-clinical cardiac safety of a drug. Finally, in order to overcome present difficulties in arriving at the definitive version of the S7B guideline, the Expert Working Group could reflect on the introduction of the S7B guideline recommendations in the S7A document, as originally intended, or on postponing the adoption of an harmonized text until the availability of novel scientific data allows solving presently contentious aspects of this and the E14 guidelines.
Collapse
|
41
|
Bass AS, Vargas HM, Kinter LB. Introduction to nonclinical safety pharmacology and the safety pharmacology society. J Pharmacol Toxicol Methods 2004; 49:141-4. [PMID: 15172009 DOI: 10.1016/j.vascn.2004.02.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
42
|
Ette EI, Garg V, Jayaraj A. A Rational Approach to Drug Development: The Exploratory Phase. ACTA ACUST UNITED AC 2004. [DOI: 10.1081/crp-200043648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
43
|
Preskorn SH. Relating clinical trials to psychiatric practice: part II: the gap between the usual patient in registration trials and in practice. J Psychiatr Pract 2003; 9:455-61. [PMID: 15985968 DOI: 10.1097/00131746-200311000-00008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Sheldon H Preskorn
- Department of Psychiatry, University of Kansas School of Medicine-Wichita, USA
| |
Collapse
|
44
|
Szafir D, Lelouët H, Imbs JL, Barouki R, Blayac J, Bons B, Caron J, Castot A, Childs M, Daurat V, Deligne J, Delmas C, Demarez J, Eschwege E, Garcia-Gibot A, Girard F, Guilhot J, Kreft-Jaïs C, Lagier G, Lamarque V, Lebrun-Vignes B, Lelouet A, Maillère P, Malvy J, Mazraani M, Moachon-Chauvelot L, Muzard D, Olivier P, Pappo M, Pastor E, Riché C, Sallière D, Thomas L. Prevention of Drug-Induced Risks. Therapie 2003. [DOI: 10.2515/therapie:2003036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Szafir D, Lelouët H, Imbs JL. Risque médicamenteux : prévention. Therapie 2003; 58:221-7. [PMID: 14655317 DOI: 10.2515/therapie:2003035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Effective prevention of drug-induced risks depends on an accurate understanding of their triggering or predisposing factors, and the quality of information on these available to prescribing practitioners and users. All preclinical and clinical data available on the proprietary medicinal product concerned should facilitate identification of a risk, and these data should be compared with existing data on drugs sharing the same mode of action or therapeutic strategy. This information should be based on a communication plan adapted to the context of the disease under treatment, the therapeutic alternatives available and the benefits expected.
Collapse
|
46
|
Pharmacoepidemiology and drug safety. Pharmacoepidemiol Drug Saf 2003; 12:161-76. [PMID: 12642981 DOI: 10.1002/pds.788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
47
|
Wakefield ID, Pollard C, Redfern WS, Hammond TG, Valentin JP. The application of in vitro methods to safety pharmacology. Fundam Clin Pharmacol 2002; 16:209-18. [PMID: 12165068 DOI: 10.1046/j.1472-8206.2002.00099.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The ICH S7A guideline defines safety pharmacology (SP) studies as those that investigate 'the potential undesirable pharmacodynamic effects of a substance on physiological functions in relation to exposure in the therapeutic range and above', and permits both in vivo and in vitro techniques, as appropriate. The implementation of these ICH guidelines by the pharmaceutical industry--whilst providing a welcome and long overdue clarity into the scientific rationale, timing and regulatory requirements for SP studies--has also generated new challenges, both logistical and scientific, which have a major impact on drug development. These factors have motivated us to consider the introduction of in vitro techniques at an early stage of SP evaluation. Amongst these factors are: the expanded range of study types and physiological parameters to be assessed, the increased 'front-loading' of SP at earlier stages of the drug discovery process; the greater number of new chemical entities (NCEs) to be tested, together with limited compound supply; the condensed time frames for drug development, the higher and quicker throughput of in vitro vs. in vivo tests; the increasing predictability of in vitro tests and application of the '3Rs' rule of animal welfare (reduction, replacement and refinement). Also, there is the failure of traditional in vivo safety evaluation to predict certain clinical side-effects. The use of molecular (e.g. fluorescence and cloned ion channel), cellular (e.g. patch clamp and isolated cardiac cells) and tissue-based (e.g. microelectrodes and Purkinje fibres) methods offers a wide portfolio of novel techniques for SP evaluation of NCEs at a pre-in vivo stage. Thus, innovative in vitro techniques will contribute significantly to the early SP evaluation of NCEs.
Collapse
Affiliation(s)
- Ian D Wakefield
- Safety Assessment UK, AstraZeneca R & D, Aldereley Park, Macclesfield, Cheshire, UK, SK10 4TG.
| | | | | | | | | |
Collapse
|