1
|
Navarro R, Frago S, Hangiu O, Erce-Llamazares A, Lázaro-Gorines R, Morcillo MA, Rodriguez-Peralto JL, Sanz L, Compte M, Alvarez-Vallina L. Pharmacokinetics and safety of LEAD-452, an EGFR-specific 4-1BB-agonistic trimerbody in non-human primates. Toxicol Appl Pharmacol 2024; 487:116961. [PMID: 38740095 DOI: 10.1016/j.taap.2024.116961] [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: 12/20/2023] [Revised: 04/29/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
LEAD-452 is a humanized bispecific EGFR-targeted 4-1BB-agonistic trimerbody with a unique trimeric configuration compared to other 4-1BB-specific antibodies that are currently in development. Indeed, enhanced tumor-specific costimulation and very remarkable safety and efficacy profiles have been observed in mouse models. Here, we conducted for the first time a preclinical pharmacokinetic and toxicity study in non-human primates (NHP) (Macaca fascicularis). LEAD-452 exhibits comparable binding affinity for human and macaque targets, indicating its pharmacological significance for safety testing across species. The NHP were administered LEAD-452 in a series of ascending doses, ranging from 0.1 mg/kg to 10 mg/kg, and repeated doses up to 20 mg/kg. The administration of LEAD-452 was found to be clinically well tolerated, with no major related adverse effects observed. Furthermore, there have been no reported cases of liver toxicity, thrombocytopenia, and neutropenia, which are commonly associated with treatments using conventional anti-4-1BB IgG-based antibodies. In addition, neither IgM nor IgG-based anti-drug antibodies were detected in serum samples from NHP during the study, regardless of the dose of LEAD-452 administered. These results support the clinical development of LEAD-452 for the treatment of solid tumors.
Collapse
Affiliation(s)
- Rocío Navarro
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain
| | - Susana Frago
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain
| | - Oana Hangiu
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain; Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Ainhoa Erce-Llamazares
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain; Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain
| | - Rodrigo Lázaro-Gorines
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain; H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
| | - Miguel A Morcillo
- Medical Applications of Ionizing Radiations Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - José L Rodriguez-Peralto
- Department of Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain; Department of Pathology, Universidad Complutense, Madrid, Spain; Cutaneous Oncology Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain; Centro de Investigación Biomédica en Red en Oncología (CIBERONC), Madrid, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Fundación para la Investigación Biomédica Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Marta Compte
- Department of Antibody Engineering, Leadartis SL, Tres Cantos, Madrid, Spain.
| | - Luis Alvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain; Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain; H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain.
| |
Collapse
|
2
|
Mecklenburg L, Lenz S, Hempel G. How important are concurrent vehicle control groups in (sub)chronic non-human primate toxicity studies conducted in pharmaceutical development? An opportunity to reduce animal numbers. PLoS One 2023; 18:e0282404. [PMID: 37535533 PMCID: PMC10399820 DOI: 10.1371/journal.pone.0282404] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023] Open
Abstract
Safety assessment of human pharmaceuticals demands extensive animal experiments before a compound can be tested in patients or released on the market. Such experiments typically include concurrent vehicle control groups. Reconsidering the need for concurrent controls could support the strive to reduce the use of animals for scientific purposes. We reviewed reports from 20 (sub)chronic toxicity studies that were conducted in non-human primates (NHP) to characterize hazards of novel human pharmaceuticals. Firstly, we determined the toxicological endpoints that were identified to characterize the hazard. Secondly, we evaluated if the hazard could have been identified without reference to the concurrent controls. Thirdly, we employed an alternative statistical method to test for any significant change related to dose level or time. We found that toxicologically relevant hazards were identifiable without reference to concurrent controls, because individual measurements could be compared with pre-dosing values or because individual measurements could be compared to historical reference data. Effects that could not be evaluated without reference to concurrent controls were clinical observations and organ weights for which appropriate historical reference data was not available, or immune responses that could not be compared to pre-dosing measurements because their magnitude would change over time. Our investigation indicates that concurrent control groups in (sub)chronic NHP toxicity studies are of limited relevance for reaching the study objective. Under certain conditions, regulatory (sub)chronic NHP toxicity studies represent a good starting point to implement virtual control groups rather than concurrent control groups in nonclinical safety testing.
Collapse
Affiliation(s)
| | - Sarah Lenz
- Labcorp Early Development Services GmbH, Muenster, Germany
- Institute for Pharmaceutical and Medicinal Chemistry, Westphalian Wilhelms University, Muenster, Germany
| | - Georg Hempel
- Institute for Pharmaceutical and Medicinal Chemistry, Westphalian Wilhelms University, Muenster, Germany
| |
Collapse
|
3
|
Tyszkiewicz C, Hwang SK, Manickam B, Jakubczak B, Walters KM, Bolt MW, Santos R, Liu CN. Sex-related differences in retinal function in Wistar rats: implications for toxicity and safety studies. FRONTIERS IN TOXICOLOGY 2023; 5:1176665. [PMID: 37313214 PMCID: PMC10259507 DOI: 10.3389/ftox.2023.1176665] [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: 02/28/2023] [Accepted: 05/10/2023] [Indexed: 06/15/2023] Open
Abstract
Introduction: Wistar Han rats are a preferred strain of rodents for general toxicology and safety pharmacology studies in drug development. In some of these studies, visual functional tests that assess for retinal toxicity are included as an additional endpoint. Although the influence of gender on human retinal function has been documented for more than 6 decades, preclinically it is still uncertain if there are differences in retinal function between naïve male and female Wistar Han rats. Methods: In this study, sex-related differences in the retinal function were quantified by analyzing electroretinography (ERG) in 7-9-week-old (n = 52 males and 51 females) and 21-23-week-old Wistar Han rats (n = 48 males and 51 females). Optokinetic tracking response, brainstem auditory evoked potential, ultrasonic vocalization and histology were tested and evaluated in a subset of animals to investigate the potential compensation mechanisms of spontaneous blindness. Results/Discussion: Absence of scotopic and photopic ERG responses was found in 13% of 7-9-week-old (7/52) and 19% of 21-23-week-old males (9/48), but none of female rats (0/51). The averaged amplitudes of rod- and cone-mediated ERG b-wave responses obtained from males were significantly smaller than the amplitudes of the same responses from age-matched females (-43% and -26%, respectively) at 7-9 weeks of age. There was no difference in the retinal and brain morphology, brainstem auditory responses, or ultrasonic vocalizations between the animals with normal and abnormal ERGs at 21-23 weeks of age. In summary, male Wistar Han rats had altered retinal responses, including a complete lack of responses to test flash stimuli (i.e., blindness), when compared with female rats at 7-9 and 21-23 weeks of age. Therefore, sex differences should be considered when using Wistar Han rats in toxicity and safety pharmacology studies with regards to data interpretation of retinal functional assessments.
Collapse
Affiliation(s)
| | | | | | - Ben Jakubczak
- Comparative Medicine, Pfizer, Groton, CT, United States
| | - Karen M. Walters
- Drug Safety Research and Development, Pfizer, Groton, CT, United States
| | - Michael W. Bolt
- Drug Safety Research and Development, Pfizer, Cambridge, Massachusetts, United States
| | | | | |
Collapse
|
4
|
Gurjanov A, Kreuchwig A, Steger-Hartmann T, Vaas LAI. Hurdles and signposts on the road to virtual control groups-A case study illustrating the influence of anesthesia protocols on electrolyte levels in rats. Front Pharmacol 2023; 14:1142534. [PMID: 37153793 PMCID: PMC10159271 DOI: 10.3389/fphar.2023.1142534] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/31/2023] [Indexed: 05/10/2023] Open
Abstract
Introduction: Virtual Control Groups (VCGs) represent the concept of using historical control data from legacy animal studies to replace concurrent control group (CCG) animals. Based on the data curation and sharing activities of the Innovative Medicine Initiatives project eTRANSAFE (enhancing TRANSlational SAFEty Assessment through Integrative Knowledge Management) the ViCoG working group was established with the objectives of i) collecting suitable historical control data sets from preclinical toxicity studies, ii) evaluating statistical methodologies for building adequate and regulatory acceptable VCGs from historical control data, and iii) sharing those control-group data across multiple pharmaceutical companies. During the qualification process of VCGs a particular focus was put on the identification of hidden confounders in the data sets, which might impair the adequate matching of VCGs with the CCG. Methods: During our analyses we identified such a hidden confounder, namely, the choice of the anesthetic procedure used in animal experiments before blood withdrawal. Anesthesia using CO2 may elevate the levels of some electrolytes such as calcium in blood, while the use of isoflurane is known to lower these values. Identification of such hidden confounders is particularly important if the underlying experimental information (e.g., on the anesthetic procedure) is not routinely recorded in the standard raw data files, such as SEND (Standard for Exchange of Non-clinical Data). We therefore analyzed how the replacement of CCGs with VCGs would affect the reproducibility of treatment-related findings regarding electrolyte values (potassium, calcium, sodium, and phosphate). The analyses were performed using a legacy rat systemic toxicity study consisting of a control and three treatment groups conducted according to pertinent OECD guidelines. In the report of this study treatment-related hypercalcemia was reported. The rats in this study were anesthetized with isoflurane. Results: Replacing the CCGs with VCGs derived from studies comprising both anesthetics resulted in a shift of control electrolyte parameters. Instead of the originally reported hypercalcemia the use of VCG led to fallacious conclusions of no observed effect or hypocalcemia. Discussion: Our study highlights the importance of a rigorous statistical analysis including the detection and elimination of hidden confounders prior to the implementation of the VCG concept.
Collapse
Affiliation(s)
- A. Gurjanov
- Bayer AG, Pharmaceuticals, Investigational Toxicology, Berlin, Germany
- *Correspondence: A. Gurjanov,
| | - A. Kreuchwig
- Bayer AG, Pharmaceuticals, Investigational Toxicology, Berlin, Germany
| | | | - L. A. I. Vaas
- Bayer AG, Pharmaceuticals, Research and Pre-Clinical Statistics Group, Berlin, Germany
| |
Collapse
|
5
|
Baldrick P. Genotoxicity test battery - An assessment of its utility in early drug development. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2021; 868-869:503388. [PMID: 34454694 DOI: 10.1016/j.mrgentox.2021.503388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 11/27/2022]
Abstract
Formal requirements for genotoxicity testing of drug candidates to support clinical entry have been in place since the issue of initial regulatory guidance over 25 years ago and subsequent update a decade ago. An evaluation of such testing, supporting first clinical entry of 108 small molecule drug candidates over the last decade, showed that the most common approach (75 % of tested compounds) was for a Good Laboratory Practice test battery in the form of 2 in vitro (a bacterial reverse mutation and a mammalian cell) assays and one in vivo assay. The majority of other tested compounds involved in vitro testing only in bacterial reverse mutation and mammalian cell assays. Testing using a bacterial reverse mutation assay and an in vivo assessment of genotoxicity with 2 different tissues was limited to 2 occasions. For in vitro mammalian cell testing, the chromosome aberration test was most commonly used (70 % occasions), followed by a micronucleus test (16 % occasions) or a mouse lymphoma assay (14 % occasions). For in vivo evaluation, the most common test was a rodent bone marrow micronucleus test (87 % occasions). A positive in vitro mammalian cell assay result was seen on 13 % occasions but was not confirmed with further in vivo testing and the drug candidates were taken into the clinic. In conclusion, the present evaluation showed that the current test battery paradigm for genotoxicity testing has an integral part in supporting clinical entry to confirm candidate drugs taken into the clinic are unlikely to have genotoxic activity.
Collapse
Affiliation(s)
- Paul Baldrick
- Product Development and Market Access Consulting, Clinical Development & Commercialisation Services, Labcorp Drug Development Inc. (formerly Covance), Harrogate, North Yorkshire, HG3 1PY, United Kingdom.
| |
Collapse
|
6
|
Baldrick P, Reichl A. Nonclinical & clinical interface - extrapolation of nonclinical data to support Phase I clinical studies. Regul Toxicol Pharmacol 2021; 121:104869. [DOI: 10.1016/j.yrtph.2021.104869] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/09/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023]
|
7
|
Prior H, Haworth R, Labram B, Roberts R, Wolfreys A, Sewell F. Justification for species selection for pharmaceutical toxicity studies. Toxicol Res (Camb) 2020; 9:758-770. [PMID: 33442468 PMCID: PMC7786171 DOI: 10.1093/toxres/tfaa081] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Toxicity studies using mammalian species are generally required to provide safety data to support clinical development and licencing registration for potential new pharmaceuticals. International regulatory guidelines outline recommendations for the order (rodent and/or non-rodent) and number of species, retaining flexibility for development of a diverse range of drug modalities in a manner relevant for each specific new medicine. Selection of the appropriate toxicology species involves consideration of scientific, ethical and practical factors, with individual companies likely having different perspectives and preferences regarding weighting of various aspects dependent upon molecule characteristics and previous experience of specific targets or molecule classes. This article summarizes presentations from a symposium at the 2019 Annual Congress of the British Toxicology Society on the topic of species selection for pharmaceutical toxicity studies. This symposium included an overview of results from a National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs) and Association of British Pharmaceutical Industry (ABPI) international collaboration that reviewed the use of one or two species in regulatory toxicology studies and justification for the species selected within each programme. Perspectives from two pharmaceutical companies described their processes for species selection for evaluation of biologics, and justification for selection of the minipig as a toxicological species for small molecules. This article summarizes discussions on the scientific justification and other considerations taken into account to ensure the most appropriate animal species are used for toxicity studies to meet regulatory requirements and to provide the most value for informing project decisions.
Collapse
Affiliation(s)
- Helen Prior
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), 215 Euston Rd, London, NW1 2BE, UK
| | | | - Briony Labram
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), 215 Euston Rd, London, NW1 2BE, UK
| | - Ruth Roberts
- ApconiX, Alderley Park, Alderley Edge, SK10 4TG, UK
| | | | - Fiona Sewell
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), 215 Euston Rd, London, NW1 2BE, UK
| |
Collapse
|
8
|
Rossi O, Citiulo F, Mancini F. Outer membrane vesicles: moving within the intricate labyrinth of assays that can predict risks of reactogenicity in humans. Hum Vaccin Immunother 2020; 17:601-613. [PMID: 32687736 PMCID: PMC7899674 DOI: 10.1080/21645515.2020.1780092] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Outer membrane vesicles (OMV) are exosomes naturally released from the surface of Gram-negative bacteria. Since the ’80s, OMVs have been proposed as powerful vaccine platforms due to their intrinsic self-adjuvanticity and ability to present multiple antigens in natural conformation. However, the presence of several pathogen-associated molecular patterns (PAMPs), especially lipid A, has raised concerns about potential systemic reactogenicity in humans. Recently, chemical and genetic approaches allowed to efficiently modulate the balance between reactogenicity and immunogenicity for the use of OMV in humans. Several assays (monocyte activation test, rabbit pyrogenicity test, limulus amebocyte lysate, human transfectant cells, and toxicology studies) were developed to test, with highly predictive potential, the risk of reactogenicity in humans before moving to clinical use. In this review, we provide a historical perspective on how different assays were and can be used to successfully evaluate systemic reactogenicity during clinical development and after licensure.
Collapse
Affiliation(s)
- Omar Rossi
- GSK Vaccines Institute for Global Health S.r.l (GVGH) , Siena, Italy
| | - Francesco Citiulo
- GSK Vaccines Institute for Global Health S.r.l (GVGH) , Siena, Italy
| | - Francesca Mancini
- GSK Vaccines Institute for Global Health S.r.l (GVGH) , Siena, Italy
| |
Collapse
|
9
|
Prior H, Baldrick P, Beken S, Booler H, Bower N, Brooker P, Brown P, Burlinson B, Burns-Naas LA, Casey W, Chapman M, Clarke D, de Haan L, Doehr O, Downes N, Flaherty M, Gellatly N, Moesgaard SG, Harris J, Holbrook M, Hui J, Jones D, Jones K, Kedar H, Mahl A, Manninen A, McGuire A, Mortimer-Cassen E, Peraza M, Pugsley MK, Richard J, Roberts R, Roosen W, Rothfuss A, Schoenmakers A, Sewell F, Weaver R, Weir L, Wolfreys A, Kimber I. Opportunities for use of one species for longer-term toxicology testing during drug development: A cross-industry evaluation. Regul Toxicol Pharmacol 2020; 113:104624. [PMID: 32126256 DOI: 10.1016/j.yrtph.2020.104624] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 12/21/2022]
Abstract
An international expert working group representing 37 organisations (pharmaceutical/biotechnology companies, contract research organisations, academic institutions and regulatory bodies) collaborated in a data sharing exercise to evaluate the utility of two species within regulatory general toxicology studies. Anonymised data on 172 drug candidates (92 small molecules, 46 monoclonal antibodies, 15 recombinant proteins, 13 synthetic peptides and 6 antibody-drug conjugates) were submitted by 18 organisations. The use of one or two species across molecule types, the frequency for reduction to a single species within the package of general toxicology studies, and a comparison of target organ toxicities identified in each species in both short and longer-term studies were determined. Reduction to a single species for longer-term toxicity studies, as used for the development of biologicals (ICHS6(R1) guideline) was only applied for 8/133 drug candidates, but might have been possible for more, regardless of drug modality, as similar target organ toxicity profiles were identified in the short-term studies. However, definition and harmonisation around the criteria for similarity of toxicity profiles is needed to enable wider consideration of these principles. Analysis of a more robust dataset would be required to provide clear, evidence-based recommendations for expansion of these principles to small molecules or other modalities where two species toxicity testing is currently recommended.
Collapse
Affiliation(s)
- Helen Prior
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), Gibbs Building, 215 Euston Road, London, NW1 2BE, UK.
| | - Paul Baldrick
- Covance Laboratories Ltd, Otley Road, Harrogate, HG3 1PY, UK
| | - Sonja Beken
- Federal Agency for Medicines and Health Products (FAMHP), Victor Hortaplace 40/40, Brussels, 1060, Belgium
| | - Helen Booler
- Genentech, Inc, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Nancy Bower
- Eisai Inc, 155 Tice Blvd, Woodcliff Lake, NJ, 07677, USA
| | - Paul Brooker
- Board member, NC3Rs, Gibbs Building, 215 Euston Road, London, NW1 2BE, UK
| | - Paul Brown
- Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD, 20993, USA
| | | | | | - Warren Casey
- National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods (NICEATM), National Institute of Environmental Health Sciences, P.O. Box 12233, Research Triangle Park, NC, 27709, USA
| | - Melissa Chapman
- Oncology Safety, Clinical Pharmacology and Safety Sciences,R&D, AstraZeneca, Cambridge, UK
| | - David Clarke
- Lilly Research Laboratories, Indianapolis, IN, 46285, USA
| | - Lolke de Haan
- Biologics and Advanced Therapeutics Safety, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Olaf Doehr
- Bayer Pharma AG, Müllerstrasse 170, 13353, Berlin, Germany
| | - Noel Downes
- Sequani Limited, Bromyard Rd, Ledbury, Herefordshire, HR8 1LH, UK
| | - Meghan Flaherty
- Takeda Pharmaceuticals, 300 Massachusetts Ave, Cambridge, MA, 02139, USA
| | - Nichola Gellatly
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), Gibbs Building, 215 Euston Road, London, NW1 2BE, UK
| | | | - Jennifer Harris
- Association of British Pharmaceutical Industry (ABPI), 105 Victoria Street, London, SW1E 6QT, UK
| | | | - Julia Hui
- Celgene, 86 Morris Avenue, Summit, NJ, 07901, USA
| | - David Jones
- Medicines Healthcare products Regulatory Agency (MHRA) 10 South Colonnade, Canary Wharf, London, E14 4PU, UK
| | | | | | - Andreas Mahl
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | | | - Aidan McGuire
- Charles River Laboratories, Preclinical Services, Tranent, Edinburgh, EH33 2NE, UK
| | - Elisabeth Mortimer-Cassen
- Regulatory Safety Centre of Excellence, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Marjorie Peraza
- Pfizer Drug Safety Research and Development, 300 Technology Square, Cambridge, MA, 02139, USA
| | | | - Jacques Richard
- Sanofi, 371 Rue du Professeur Blayac, Montpellier, 34000, France
| | - Ruth Roberts
- ApconiX, Alderley Park, Alderley Edge, Cheshire, SK10 4TG, UK
| | - Wendy Roosen
- Janssen Research & Development, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Andreas Rothfuss
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, CH - 4070, Basel, Switzerland
| | | | - Fiona Sewell
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), Gibbs Building, 215 Euston Road, London, NW1 2BE, UK
| | - Richard Weaver
- Institut de Recherches Internationales Servier, Biopharmacy, 92284, Suresnes, Cedex, France
| | - Lucinda Weir
- GlaxoSmithKline, Park Road, Ware, Hertfordshire, SG12 0DP, UK
| | | | - Ian Kimber
- University of Manchester, Faculty of Biology, Medicine and Health, Oxford Rd, Manchester, M13 9PL, UK
| |
Collapse
|
10
|
Baldrick P. Nonclinical Immunotoxicity Testing in the Pharmaceutical World: The Past, Present, and Future. Ther Innov Regul Sci 2019:2168479019864555. [PMID: 31409131 DOI: 10.1177/2168479019864555] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
An examination for potential direct or indirect adverse effects on the immune system (immunotoxicity) is an established component of nonclinical testing to support safe use of new drugs. Testing recommendations occur in various regulatory guidance documents, especially ICH S8, and these will be presented. Key evaluation usually occurs in toxicology studies with further investigative work a consideration if a positive signal is seen. Expectations around whether findings may occur are related to the type of compound being developed, including a chemically synthesized small molecule, a small molecule oncology drug, a biopharmaceutical, an oligonucleotide, a gene therapy/stem cell product, a vaccine, or reformulation of drugs in liposomes or depots. Examples of immunotoxicity/immunogenicity findings will be discussed for all of these types of compound. Overall, it can be concluded that our main tool for evaluation of potential immunotoxicity/immunogenicity for a new drug still remains standard toxicology study testing with key assessment for effects on clinical pathology and lymphoid organs/tissues (weights and cellularity). Additional evaluation from studies using a T cell-dependent antibody response (TDAR) and lymphocyte phenotyping is also valuable, if needed. Thus, using the tools from the past, it is the role of toxicologists to work with clinical teams now and in the future, to interpret findings from nonclinical testing to possible adverse findings in humans.
Collapse
Affiliation(s)
- Paul Baldrick
- 1 Covance Laboratories Ltd, England, United Kingdom
- 2 Lincoln School of Pharmacy, University of Lincoln, United Kingdom
| |
Collapse
|
11
|
Troth SP, Simutis F, Friedman GS, Todd S, Sistare FD. Kidney Safety Assessment: Current Practices in Drug Development. Semin Nephrol 2019; 39:120-131. [DOI: 10.1016/j.semnephrol.2018.12.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
12
|
Prior H, Baldrick P, de Haan L, Downes N, Jones K, Mortimer-Cassen E, Kimber I. Reviewing the Utility of Two Species in General Toxicology Related to Drug Development. Int J Toxicol 2018. [PMCID: PMC5881785 DOI: 10.1177/1091581818760564] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
As part of the safety assessment of new drugs, the use of two species (a rodent and a nonrodent) for regulatory toxicology studies is the typical approach taken for small molecules. For biologics, species selection is dictated by pharmacological relevance, and single species toxicology packages (typically using the nonhuman primate) are common. The UK National Centre for the Replacement, Refinement, and Reduction of Animals in Research and the Association of the British Pharmaceutical Industry are collaborating on a project to review the utility of two species in regulatory toxicology studies, with the aim to explore whether there are wider circumstances when data from a single species could be sufficient to enable safe progression in humans. An international working group consisting of 37 representatives from pharmaceutical and biotechnology companies, contract research organizations, academia, and regulatory bodies is coordinating a large-scale data sharing exercise to examine the potential for changes in current practice to reduce the number of species used for nonclinical safety testing at different stages of development. The challenge will be to determine whether two species toxicology adds significant value or whether in some instances data from a single species are sufficient (across a broader range of molecules than is currently the case) without compromising human safety.
Collapse
Affiliation(s)
- Helen Prior
- National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), London, United Kingdom
| | - Paul Baldrick
- Nonclinical and Biological Discovery Expert Network (NaBDEN), The Association of the British Pharmaceutical Industry (ABPI), London, United Kingdom
| | - Lolke de Haan
- Nonclinical and Biological Discovery Expert Network (NaBDEN), The Association of the British Pharmaceutical Industry (ABPI), London, United Kingdom
| | - Noel Downes
- Nonclinical and Biological Discovery Expert Network (NaBDEN), The Association of the British Pharmaceutical Industry (ABPI), London, United Kingdom
| | - Keith Jones
- Nonclinical and Biological Discovery Expert Network (NaBDEN), The Association of the British Pharmaceutical Industry (ABPI), London, United Kingdom
| | - Elisabeth Mortimer-Cassen
- Nonclinical and Biological Discovery Expert Network (NaBDEN), The Association of the British Pharmaceutical Industry (ABPI), London, United Kingdom
| | - Ian Kimber
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
13
|
Baldrick P. Getting a molecule into the clinic: Nonclinical testing and starting dose considerations. Regul Toxicol Pharmacol 2017; 89:95-100. [DOI: 10.1016/j.yrtph.2017.07.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/21/2017] [Accepted: 07/23/2017] [Indexed: 10/19/2022]
|
14
|
Redfern WS, Tse K, Grant C, Keerie A, Simpson DJ, Pedersen JC, Rimmer V, Leslie L, Klein SK, Karp NA, Sillito R, Chartsias A, Lukins T, Heward J, Vickers C, Chapman K, Armstrong JD. Automated recording of home cage activity and temperature of individual rats housed in social groups: The Rodent Big Brother project. PLoS One 2017; 12:e0181068. [PMID: 28877172 PMCID: PMC5587114 DOI: 10.1371/journal.pone.0181068] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/26/2017] [Indexed: 12/04/2022] Open
Abstract
Measuring the activity and temperature of rats is commonly required in biomedical research. Conventional approaches necessitate single housing, which affects their behavior and wellbeing. We have used a subcutaneous radiofrequency identification (RFID) transponder to measure ambulatory activity and temperature of individual rats when group-housed in conventional, rack-mounted home cages. The transponder location and temperature is detected by a matrix of antennae in a baseplate under the cage. An infrared high-definition camera acquires side-view video of the cage and also enables automated detection of vertical activity. Validation studies showed that baseplate-derived ambulatory activity correlated well with manual tracking and with side-view whole-cage video pixel movement. This technology enables individual behavioral and temperature data to be acquired continuously from group-housed rats in their familiar, home cage environment. We demonstrate its ability to reliably detect naturally occurring behavioral effects, extending beyond the capabilities of routine observational tests and conventional monitoring equipment. It has numerous potential applications including safety pharmacology, toxicology, circadian biology, disease models and drug discovery.
Collapse
Affiliation(s)
- William S. Redfern
- Drug Safety and Metabolism, AstraZeneca R&D, Babraham Research Campus, Cambridge, United Kingdom
| | - Karen Tse
- Drug Safety and Metabolism, AstraZeneca R&D, Babraham Research Campus, Cambridge, United Kingdom
| | - Claire Grant
- Drug Safety and Metabolism, AstraZeneca R&D, Alderley Park, Cheshire, United Kingdom
| | - Amy Keerie
- Drug Safety and Metabolism, AstraZeneca R&D, Babraham Research Campus, Cambridge, United Kingdom
| | - David J. Simpson
- Drug Safety and Metabolism, AstraZeneca R&D, Alderley Park, Cheshire, United Kingdom
| | - John C. Pedersen
- Drug Safety and Metabolism, AstraZeneca R&D, Babraham Research Campus, Cambridge, United Kingdom
| | - Victoria Rimmer
- Drug Safety and Metabolism, AstraZeneca R&D, Alderley Park, Cheshire, United Kingdom
| | - Lauren Leslie
- Drug Safety and Metabolism, AstraZeneca R&D, Alderley Park, Cheshire, United Kingdom
| | - Stephanie K. Klein
- Drug Safety and Metabolism, AstraZeneca R&D, Babraham Research Campus, Cambridge, United Kingdom
| | - Natasha A. Karp
- Quantitative Biology, IMED, AstraZeneca, Darwin Building (Unit 310), Cambridge Science Park, Cambridge, United Kingdom
| | | | | | - Tim Lukins
- Actual Analytics Ltd, Edinburgh, United Kingdom
| | | | | | | | - J. Douglas Armstrong
- Actual Analytics Ltd, Edinburgh, United Kingdom
- School of Informatics, University of Edinburgh, Appleton Tower, Edinburgh, United Kingdom
| |
Collapse
|
15
|
Sistare FD, Mattes WB, LeCluyse EL. The Promise of New Technologies to Reduce, Refine, or Replace Animal Use while Reducing Risks of Drug Induced Liver Injury in Pharmaceutical Development. ILAR J 2017; 57:186-211. [DOI: 10.1093/ilar/ilw025] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 07/25/2016] [Accepted: 09/13/2016] [Indexed: 12/19/2022] Open
|
16
|
Prior H, Sewell F, Stewart J. Overview of 3Rs opportunities in drug discovery and development using non-human primates. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.ddmod.2017.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
17
|
Sewell F, Edwards J, Prior H, Robinson S. Opportunities to Apply the 3Rs in Safety Assessment Programs. ILAR J 2016; 57:234-245. [PMID: 28053076 PMCID: PMC5886346 DOI: 10.1093/ilar/ilw024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 08/01/2016] [Accepted: 09/01/2016] [Indexed: 12/16/2022] Open
Abstract
Before a potential new medicine can be administered to humans it is essential that its safety is adequately assessed. Safety assessment in animals forms an integral part of this process, from early drug discovery and initial candidate selection to the program of recommended regulatory tests in animals. The 3Rs (replacement, reduction, and refinement of animals in research) are integrated in the current regulatory requirements and expectations and, in the EU, provide a legal and ethical framework for in vivo research to ensure the scientific objectives are met whilst minimizing animal use and maintaining high animal welfare standards. Though the regulations are designed to uncover potential risks, they are intended to be flexible, so that the most appropriate approach can be taken for an individual product. This article outlines current and future opportunities to apply the 3Rs in safety assessment programs for pharmaceuticals, and the potential (scientific, financial, and ethical) benefits to the industry, across the drug discovery and development process. For example, improvements to, or the development of, novel, early screens (e.g., in vitro, in silico, or nonmammalian screens) designed to identify compounds with undesirable characteristics earlier in development have the potential to reduce late-stage attrition by improving the selection of compounds that require regulatory testing in animals. Opportunities also exist within the current regulatory framework to simultaneously reduce and/or refine animal use and improve scientific outcomes through improvements to technical procedures and/or adjustments to study designs. It is important that approaches to safety assessment are continuously reviewed and challenged to ensure they are science-driven and predictive of relevant effects in humans.
Collapse
Affiliation(s)
- Fiona Sewell
- Fiona Sewell, PhD, is a Programme Manager in Toxicology and Regulatory Sciences at the UK National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), London, UK.Joanna Edwards, PhD, is a Programme Manager in Technology Development at the NC3Rs, London, UK.Helen Prior, PhD, is a Programme Manager in Drug Development at the NC3Rs, London, UK.Sally Robinson, PhD, is Head of Laboratory Animal Sciences at AstraZeneca, Alderley Park, UK
| | - Joanna Edwards
- Fiona Sewell, PhD, is a Programme Manager in Toxicology and Regulatory Sciences at the UK National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), London, UK.Joanna Edwards, PhD, is a Programme Manager in Technology Development at the NC3Rs, London, UK.Helen Prior, PhD, is a Programme Manager in Drug Development at the NC3Rs, London, UK.Sally Robinson, PhD, is Head of Laboratory Animal Sciences at AstraZeneca, Alderley Park, UK
| | - Helen Prior
- Fiona Sewell, PhD, is a Programme Manager in Toxicology and Regulatory Sciences at the UK National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), London, UK.Joanna Edwards, PhD, is a Programme Manager in Technology Development at the NC3Rs, London, UK.Helen Prior, PhD, is a Programme Manager in Drug Development at the NC3Rs, London, UK.Sally Robinson, PhD, is Head of Laboratory Animal Sciences at AstraZeneca, Alderley Park, UK
| | - Sally Robinson
- Fiona Sewell, PhD, is a Programme Manager in Toxicology and Regulatory Sciences at the UK National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs), London, UK.Joanna Edwards, PhD, is a Programme Manager in Technology Development at the NC3Rs, London, UK.Helen Prior, PhD, is a Programme Manager in Drug Development at the NC3Rs, London, UK.Sally Robinson, PhD, is Head of Laboratory Animal Sciences at AstraZeneca, Alderley Park, UK
| |
Collapse
|
18
|
Abstract
This article summarizes the relevant definitions related to biomarkers; reviews the general processes related to biomarker discovery and ultimate acceptance and use; and finally summarizes and reviews, to the extent possible, examples of the types of biomarkers used in animal species within veterinary clinical practice and human and veterinary drug development. We highlight opportunities for collaboration and coordination of research within the veterinary community and leveraging of resources from human medicine to support biomarker discovery and validation efforts for veterinary medicine.
Collapse
Affiliation(s)
- Michael J Myers
- Center for Veterinary Medicine, Food and Drug Administration, Rockville, Maryland 20855;
| | - Emily R Smith
- Center for Veterinary Medicine, Food and Drug Administration, Rockville, Maryland 20855;
| | - Phillip G Turfle
- Center for Veterinary Medicine, Food and Drug Administration, Rockville, Maryland 20855;
| |
Collapse
|
19
|
Roberts R, Callander R, Duffy P, Jacobsen M, Knight R, Boobis A. Target organ profiles in toxicity studies supporting human dosing: Does severity progress with longer duration of exposure? Regul Toxicol Pharmacol 2015; 73:737-46. [PMID: 26517939 DOI: 10.1016/j.yrtph.2015.10.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Revised: 10/19/2015] [Accepted: 10/22/2015] [Indexed: 10/22/2022]
Abstract
We have previously reported the profile of target organs (defined as organs showing histopathological changes) in rodent and non-rodent toxicity studies conducted prior to first-time-in-man (FTiM) for 77 AstraZeneca candidate drugs (CDs). Here, we test the assumption that toxicity is exacerbated by dosing duration by comparing the incidence and severity of target organ toxicities in these ≤ 6 week FTiM studies with those observed in subsequent subchronic/chronic (≥ 3 month) studies. Looking at the effect of dosing duration on severity (pathological score) and incidence (percentage of animals within the group) for the 39 CDs that met the criteria for inclusion (comparable doses between FTiM and subchronic/chronic studies), new toxicities appeared for 31 target organs but existing ones resolved for 29 target organs. Increased severity was more frequent for rodent (16 target organs) than for non-rodent (4 target organs). Most notable changes were a large increase in severity/incidence in liver and in non-rodent lung in contrast to a large decrease in severity and incidence for kidneys/ureter and for the non-rodent thymus. Overall this analysis shows that, even with continued exposure, target organ toxicities of CDs are as likely to show partial or complete recovery as they are to progress in severity.
Collapse
Affiliation(s)
| | - Richard Callander
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Paul Duffy
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Matt Jacobsen
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Richard Knight
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, UK
| | | |
Collapse
|
20
|
Abstract
Drug-induced liver injury (DILI) is the most common organ toxicity encountered in regulatory animal toxicology studies required prior to the clinical development of new drug candidates. Very few reports have evaluated the value of these studies for predicting DILI in humans. Indeed, compounds inducing liver toxicity in regulatory toxicology studies are not always correlated with a risk of DILI in humans. Conversely, compounds associated with the occurrence of DILI in phase 3 studies or after market release are often tested negative in regulatory toxicology studies. Idiosyncratic DILI is a rare event that is precipitated in an individual by the simultaneous occurrence of several critical factors. These factors may relate to the host (e.g. human leukocyte antigen polymorphism, inflammation), the drug (e.g. reactive metabolites) or the environment (e.g. diet/microbiota). This type of toxicity therefore cannot be detected in conventional animal toxicology studies. Several animal models have recently been proposed for the identification of drugs with the potential to cause idiosyncratic DILI: rats treated with lipopolysaccharide, Sod2(+/-) mice, panels of inbred mouse strains or chimeric mice with humanized livers. These models are not suitable for use in the prospective screening of new drug candidates. Humans therefore constitute the best model for predicting and assessing idiopathic DILI.
Collapse
|
21
|
Levine DJ, Stöhr J, Falese LE, Ollesch J, Wille H, Prusiner SB, Long JR. Mechanism of scrapie prion precipitation with phosphotungstate anions. ACS Chem Biol 2015; 10:1269-77. [PMID: 25695325 PMCID: PMC4437617 DOI: 10.1021/cb5006239] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
![]()
The phosphotungstate anion (PTA)
is widely used to facilitate the
precipitation of disease-causing prion protein (PrPSc)
from infected tissue for applications in structural studies and diagnostic
approaches. However, the mechanism of this precipitation is not understood.
In order to elucidate the nature of the PTA interaction with PrPSc under physiological conditions, solutions of PTA were characterized
by NMR spectroscopy at varying pH. At neutral pH, the parent [PW12O40]3– ion decomposes to give
a lacunary [PW11O39]7– (PW11) complex and a single orthotungstate anion [WO4]2– (WO4). To measure the efficacy of
each component of PTA, increasing concentrations of PW11, WO4, and mixtures thereof were used to precipitate PrPSc from brain homogenates of scrapie prion-infected mice. The
amount of PrPSc isolated, quantified by ELISA and immunoblotting,
revealed that both PW11 and WO4 contribute to
PrPSc precipitation. Incubation with sarkosyl, PTA, or
individual components of PTA resulted in separation of higher-density
PrP aggregates from the neuronal lipid monosialotetrahexosylganglioside
(GM1), as observed by sucrose gradient centrifugation. These experiments
revealed that yield and purity of PrPSc were greater with
polyoxometalates (POMs), which substantially supported the separation
of lipids from PrPSc in the samples. Interaction of POMs
and sarkosyl with brain homogenates promoted the formation of fibrillar
PrPSc aggregates prior to centrifugation, likely through
the separation of lipids like GM1 from PrPSc. We propose
that this separation of lipids from PrP is a major factor governing
the facile precipitation of PrPSc by PTA from tissue and
might be optimized further for the detection of prions.
Collapse
Affiliation(s)
- Dana J. Levine
- Department
of Chemistry, University of California, Berkeley, 211 Lewis Hall, Berkeley, California 94720, United States
- Institute
for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, California 94143, United States
| | - Jan Stöhr
- Institute
for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, California 94143, United States
- Department
of Neurology, University of California, San Francisco, 675 Nelson
Rising Lane, San Francisco, California 94143, United States
| | - Lillian E. Falese
- Institute
for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, California 94143, United States
| | - Julian Ollesch
- Institute
for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, California 94143, United States
| | - Holger Wille
- Institute
for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, California 94143, United States
- Department
of Neurology, University of California, San Francisco, 675 Nelson
Rising Lane, San Francisco, California 94143, United States
| | - Stanley B. Prusiner
- Institute
for Neurodegenerative Diseases, University of California, San Francisco, 675 Nelson Rising Lane, San Francisco, California 94143, United States
- Department
of Neurology, University of California, San Francisco, 675 Nelson
Rising Lane, San Francisco, California 94143, United States
| | - Jeffrey R. Long
- Department
of Chemistry, University of California, Berkeley, 211 Lewis Hall, Berkeley, California 94720, United States
| |
Collapse
|
22
|
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
|
23
|
Bollag G. Setting up a kinase discovery and development project. Curr Top Microbiol Immunol 2014; 355:3-18. [PMID: 21809194 DOI: 10.1007/82_2011_159] [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: 02/11/2023]
Abstract
Discovery of novel kinase inhibitors has matured rapidly over the last decade. Paramount to the successful development of kinase inhibitors is appropriate selectivity for validated targets. Many different approaches have been applied over the years, with varied results. There are currently thirteen different small molecule protein kinase inhibitors on the marketplace. Interestingly, a majority of these compounds lack precise selectivity for specific targets. This will change in the coming years, as technology for achieving improved selectivity becomes more widely applied. This chapter will focus on some of the critical considerations in setting up a kinase discovery and development project, citing examples particularly targeting the Raf kinases.
Collapse
|
24
|
Bailey J. A response to the ABPI's letter to the use of dogs in predicting drug Toxicity in humans. Altern Lab Anim 2014; 42:149-53. [PMID: 24984299 DOI: 10.1177/026119291404200208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
25
|
|
26
|
Detection of hepatotoxicity potential with metabolite profiling (metabolomics) of rat plasma. Toxicol Lett 2014; 230:467-78. [PMID: 25086301 DOI: 10.1016/j.toxlet.2014.07.021] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 12/14/2022]
Abstract
While conventional parameters used to detect hepatotoxicity in drug safety assessment studies are generally informative, the need remains for parameters that can detect the potential for hepatotoxicity at lower doses and/or at earlier time points. Previous work has shown that metabolite profiling (metabonomics/metabolomics) can detect signals of potential hepatotoxicity in rats treated with doxorubicin at doses that do not elicit hepatotoxicity as monitored with conventional parameters. The current study extended this observation to the question of whether such signals could be detected in rats treated with compounds that can elicit hepatotoxicity in humans (i.e., drug-induced liver injury, DILI) but have not been reported to do so in rats. Nine compounds were selected on the basis of their known DILI potential, with six other compounds chosen as negative for DILI potential. A database of rat plasma metabolite profiles, MetaMap(®)Tox (developed by metanomics GmbH and BASF SE) was used for both metabolite profiles and mode of action (MoA) metabolite signatures for a number of known toxicities. Eight of the nine compounds with DILI potential elicited metabolite profiles that matched with MoA patterns of various rat liver toxicities, including cholestasis, oxidative stress, acetaminophen-type toxicity and peroxisome proliferation. By contrast, only one of the six non-DILI compounds showed a weak match with rat liver toxicity. These results suggest that metabolite profiling may indeed have promise to detect signals of hepatotoxicity in rats treated with compounds having DILI potential.
Collapse
|
27
|
Sewell F, Chapman K, Baldrick P, Brewster D, Broadmeadow A, Brown P, Burns-Naas LA, Clarke J, Constan A, Couch J, Czupalla O, Danks A, DeGeorge J, de Haan L, Hettinger K, Hill M, Festag M, Jacobs A, Jacobson-Kram D, Kopytek S, Lorenz H, Moesgaard SG, Moore E, Pasanen M, Perry R, Ragan I, Robinson S, Schmitt PM, Short B, Lima BS, Smith D, Sparrow S, van Bekkum Y, Jones D. Recommendations from a global cross-company data sharing initiative on the incorporation of recovery phase animals in safety assessment studies to support first-in-human clinical trials. Regul Toxicol Pharmacol 2014; 70:413-29. [PMID: 25078890 DOI: 10.1016/j.yrtph.2014.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 07/17/2014] [Accepted: 07/18/2014] [Indexed: 11/29/2022]
Abstract
An international expert group which includes 30 organisations (pharmaceutical companies, contract research organisations, academic institutions and regulatory bodies) has shared data on the use of recovery animals in the assessment of pharmaceutical safety for early development. These data have been used as an evidence-base to make recommendations on the inclusion of recovery animals in toxicology studies to achieve scientific objectives, while reducing animal use. Recovery animals are used in pharmaceutical development to provide information on the potential for a toxic effect to translate into long-term human risk. They are included on toxicology studies to assess whether effects observed during dosing persist or reverse once treatment ends. The group devised a questionnaire to collect information on the use of recovery animals in general regulatory toxicology studies to support first-in-human studies. Questions focused on study design, the rationale behind inclusion or exclusion and the impact this had on internal and regulatory decisions. Data on 137 compounds (including 53 biologicals and 78 small molecules) from 259 studies showed wide variation in where, when and why recovery animals were included. An analysis of individual study and programme design shows that there are opportunities to reduce the use of recovery animals without impacting drug development.
Collapse
Affiliation(s)
- Fiona Sewell
- UK National Centre for the Replacement, Refinement & Reduction of Animals in Research (NC3Rs), Gibbs Building, 215 Euston Road, London NW1 2BE, UK.
| | - Kathryn Chapman
- UK National Centre for the Replacement, Refinement & Reduction of Animals in Research (NC3Rs), Gibbs Building, 215 Euston Road, London NW1 2BE, UK
| | - Paul Baldrick
- Covance Laboratories Ltd, Otley Road, Harrogate HG3 1PY, UK
| | - David Brewster
- Vertex Pharmaceuticals Inc., 50 Northern Avenue, Boston, MA 02139, USA
| | | | - Paul Brown
- Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | | | | | - Alex Constan
- Infinity Pharmaceuticals, 780 Memorial Drive, Cambridge, MA 02139, USA
| | - Jessica Couch
- Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | - Andy Danks
- Charles River Laboratories, Preclinical Services, Tranent, Edinburgh EH33 2NE, UK
| | - Joseph DeGeorge
- Merck, 770 Sumneytown Pike, Mailstop WP45-201, West Point, PA 19486, USA
| | | | - Klaudia Hettinger
- Austrian Agency for Health and Food Safety, Traisengasse 5, 1200 Vienna, Austria
| | - Marilyn Hill
- Novartis Institutes for BioMedical Research (NIBR), Basel, Switzerland
| | - Matthias Festag
- Roche Pharmaceutical Research and Early Development, Roche Innovation Center Basel, CH - 4070 Basel, Switzerland
| | - Abby Jacobs
- Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - David Jacobson-Kram
- Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | | | - Helga Lorenz
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | | | - Emma Moore
- Huntingdon Life Sciences Ltd, Alconbury, Huntingdon, Cambridgeshire PE28 4HS, UK
| | - Markku Pasanen
- University of Eastern Finland, Faculty of Health Sciences, School of Pharmacy, Kuopio, Finland
| | - Rick Perry
- Pfizer Drug Safety Research and Development, 455 Eastern Point Rd., Groton, CT 06340, USA
| | - Ian Ragan
- Board member, NC3Rs, Gibbs Building, 215 Euston Road, London NW1 2BE, UK
| | - Sally Robinson
- AstraZeneca, Alderley Park, Macclesfield, Cheshire SK10 4TG, UK
| | - Petra M Schmitt
- Paul-Ehrlich-Institute, Federal Agency for Vaccines and Biomedicines, Langen, Germany
| | - Brian Short
- Allergan, Drug Safety Evaluation, 2525 Dupont Dr, RD-2A, Irvine, CA 92612-1599, USA
| | | | - Diane Smith
- Millenium: The Takeda Oncology Company, 40 Landsdowne St., Cambridge, MA, USA
| | - Sue Sparrow
- GlaxoSmithKline, Park Road, Ware, Hertfordshire SG12 0DP, UK
| | - Yvette van Bekkum
- Janssen Research & Development, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - David Jones
- Medicines Healthcare Products Regulatory Agency (MHRA), UK
| |
Collapse
|
28
|
Horner S, Robinson S, Lees D, Callander R, Roberts R. Target organ profiles in toxicity studies supporting human dosing: an assessment of recovery and chronic dosing. Regul Toxicol Pharmacol 2014; 70:270-85. [PMID: 25020275 DOI: 10.1016/j.yrtph.2014.07.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/20/2014] [Accepted: 07/07/2014] [Indexed: 11/17/2022]
Abstract
We have previously reported the profile of toxic effects with respect to target organs (defined as organs showing histopathological changes) observed in rodent and non-rodent toxicity studies conducted prior to first time in man (FTIM) for 77 AstraZeneca candidate drugs (CDs) across a range of therapy areas. The main objectives of the current study were twofold; to determine which target organs observed in the FTIM studies recovered after a dose free recovery period and to determine which additional target organs were observed in subsequent chronic (⩾3month) studies required to support longer term clinical dosing. The analysis showed that ⩾86% of findings in studies supporting FTIM either fully or partially resolved at the end of the recovery period, with profiles of recovery that were similar whether the CD progressed into man or not and across different therapy areas. Compared to observations in FTIM studies, chronic studies identified toxicities in an additional 39% of target organs. Overall these data demonstrate that chronic studies in both rodents and non-rodents provide valuable information for the risk assessment for longer term dosing in humans. In addition, the high levels of recovery demonstrated in this analysis suggest that inclusion of recovery assessments on FTIM studies should be on a case-by-case basis driven by a positive indication of need. This is in line with ICH non-clinical guidance that states that reversibility of severe nonclinical toxicities of potential clinic relevance should be assessed 'when appropriate', but that the evaluation can be based on a study of reversibility or on a scientific assessment.
Collapse
Affiliation(s)
- Steve Horner
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Sally Robinson
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, UK
| | - David Lees
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Richard Callander
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, UK
| | - Ruth Roberts
- Drug Safety and Metabolism, AstraZeneca, Alderley Park, Macclesfield, UK.
| |
Collapse
|
29
|
Ahuja V, Sharma S. Drug safety testing paradigm, current progress and future challenges: an overview. J Appl Toxicol 2013; 34:576-94. [PMID: 24777877 DOI: 10.1002/jat.2935] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 08/08/2013] [Accepted: 08/22/2013] [Indexed: 12/29/2022]
Abstract
Early assessment of the toxicity potential of new molecules in pharmaceutical industry is a multi-dimensional task involving predictive systems and screening approaches to aid in the optimization of lead compounds prior to their entry into development phase. Due to the high attrition rate in the pharma industry in last few years, it has become imperative for the nonclinical toxicologist to focus on novel approaches which could be helpful for early screening of drug candidates. The need is that the toxicologists should change their classical approach to a more investigative approach. This review discusses the developments that allow toxicologists to anticipate safety problems and plan ways to address them earlier than ever before. This includes progress in the field of in vitro models, surrogate models, molecular toxicology, 'omics' technologies, translational safety biomarkers, stem-cell based assays and preclinical imaging. The traditional boundaries between teams focusing on efficacy/ safety and preclinical/ clinical aspects in the pharma industry are disappearing, and translational research-centric organizations with a focused vision of bringing drugs forward safely and rapidly are emerging. Today's toxicologist should collaborate with medicinal chemists, pharmacologists, and clinicians and these value-adding contributions will change traditional toxicologists from side-effect identifiers to drug development enablers.
Collapse
Affiliation(s)
- Varun Ahuja
- Drug Safety Assessment, Novel Drug Discovery and Development, Lupin Limited (Research Park), 46A/47A, Nande Village, MulshiTaluka, Pune, 412 115, India
| | | |
Collapse
|
30
|
Horner S, Ryan D, Robinson S, Callander R, Stamp K, Roberts RA. Target organ toxicities in studies conducted to support first time in man dosing: An analysis across species and therapy areas. Regul Toxicol Pharmacol 2013; 65:334-43. [DOI: 10.1016/j.yrtph.2013.02.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/28/2013] [Accepted: 02/11/2013] [Indexed: 01/08/2023]
|
31
|
Hernández-Abreu O, Torres-Piedra M, García-Jiménez S, Ibarra-Barajas M, Villalobos-Molina R, Montes S, Rembao D, Estrada-Soto S. Dose-dependent antihypertensive determination and toxicological studies of tilianin isolated from Agastache mexicana. JOURNAL OF ETHNOPHARMACOLOGY 2013; 146:187-91. [PMID: 23276782 DOI: 10.1016/j.jep.2012.12.029] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 12/03/2012] [Accepted: 12/20/2012] [Indexed: 05/04/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Agastache mexicana is used in Mexican traditional medicine for the treatment of hypertension, anxiety and related diseases. AIM OF THE STUDY Current work was developed to establish pharmacological/toxicological parameters of tilianin, a flavone extracted from Agastache mexicana in order to propose it for clinical trials. MATERIALS AND METHODS Acute and sub-acute toxicology studies in Imprinting Control Region (ICR) mice and median effective dose (ED50) determination in conscious spontaneously hypertensive rats (SHR) were done. RESULTS A median lethal dose (LD50) of 6624 mg/kg (6201, 7076) in mice and significant antihypertensive effect (ED50=53.51 mg/kg) in SHR were determined. Moreover, sub-acute oral administration of tilianin did not alter body weight, clinical chemistry parameters (alanine amino-transferase, aspartate amino-transferase, total cholesterol, high density lipoprotein, low density lipoprotein, triglycerides, glucose and insulin), and also did not induce any toxic or adverse effects on kidney, heart, liver, and lung functions. CONCLUSIONS We have shown that tilianin, isolated from Agastache mexicana, was not toxic for rodents. Also, its antihypertensive effect was dose-dependent and ED50 (53.51 mg/kg) calculated was lesser than LD50 determined (6624 mg/kg), which suggest a wide range of pharmacology-toxicology patterns. Results support the hypothesis that tilianin must be investigated and developed for clinical trials as antihypertensive drug.
Collapse
Affiliation(s)
- Oswaldo Hernández-Abreu
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Av. Universidad 1001 Col. Chamilpa, 62209 Cuernavaca, Morelos, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
32
|
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
|
33
|
Baldrick P. Safety evaluation of biological drugs: What are toxicology studies in primates telling us? Regul Toxicol Pharmacol 2011; 59:227-36. [DOI: 10.1016/j.yrtph.2010.10.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 09/23/2010] [Accepted: 10/05/2010] [Indexed: 10/19/2022]
|
34
|
Roth A, Boess F, Landes C, Steiner G, Freichel C, Plancher JM, Raab S, de Vera Mudry C, Weiser T, Suter L. Gene expression-based in vivo and in vitro prediction of liver toxicity allows compound selection at an early stage of drug development. J Biochem Mol Toxicol 2010; 25:183-94. [DOI: 10.1002/jbt.20375] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
35
|
Milton MN, Horvath CJ. The EMEA Guideline on First-in-Human Clinical Trials and Its Impact on Pharmaceutical Development. Toxicol Pathol 2009; 37:363-71. [DOI: 10.1177/0192623309332997] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Mark N. Milton
- Tempo Pharmaceuticals, Cambridge, Massachusetts 02142, USA
| | | |
Collapse
|
36
|
Mattes WB, Walker EG. Translational Toxicology and the Work of the Predictive Safety Testing Consortium. Clin Pharmacol Ther 2009; 85:327-30. [DOI: 10.1038/clpt.2008.270] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
37
|
Safety evaluation to support First-In-Man investigations I: Kinetic and safety pharmacology studies. Regul Toxicol Pharmacol 2008; 51:230-6. [DOI: 10.1016/j.yrtph.2008.04.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2008] [Accepted: 04/11/2008] [Indexed: 11/18/2022]
|