1
|
Moraga P, Prieto P, Conradie A, Benhayoun M, Rousell V, Davy M, Fuhr U, Antonijoan Arbos R, Abad‐Santos F, Portolés A, Van Duinen J, Carcas AJ, Borobia AM. Academia and industry agreement on a feasibility tool for first-time-in-human clinical trial units. Clin Transl Sci 2023; 16:2421-2428. [PMID: 37818923 PMCID: PMC10719454 DOI: 10.1111/cts.13655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/19/2023] [Accepted: 09/20/2023] [Indexed: 10/13/2023] Open
Abstract
First-time-in-human (FTIH) trials are designed to generate information on the safety, tolerability, as well as the pharmacokinetic and pharmacodynamics profile of new drugs. To ensure the safety of participants, these trials need to be conducted at specifically equipped phase I clinical trial units (CTUs). In accordance with the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) Guideline for Good Clinical Practice (GCP) and the European Union (EU) regulatory guidelines, one of the aims of the European Regime Accelerator for Tuberculosis (ERA4TB) project is to collaboratively create a feasibility tool, through a partnership between public and private entities, for the validation of CTUs selected to conduct FTIH trials. A feasibility form, encompassing nine sections, was created to gather information on the unit in relation to key attributes of FTIH trials. Collaboratively, industry and academic partners defined the minimal criteria to ensure the adherence of CTUs to the principles of ICH GCP and regulations outlined by the European Medicines Agency (EMA) for the execution of FTIH trials. Subsequently, all CTUs available for the project were assessed for FTIH trial eligibility. The introduction of the certification procedure through the feasibility tool within ERA4TB resulted in the accreditation of the five academic CTUs, which are now prepared to carry out FTIH trials as part of the Consortium. The developed feasibility tool aims to establish open and widely used minimum requirements for the validation of academic CTUs as FTIH units, marking it as the inaugural tool for CTU validation resulting from the collaboration between industry and academia within the ERA4TB project. The established partnership has enabled an innovative and novel way of working.
Collapse
Affiliation(s)
- Paloma Moraga
- Clinical Pharmacology DepartmentLa Paz University Hospital, IdiPAZMadridSpain
| | - Paula Prieto
- Clinical Pharmacology DepartmentLa Paz University Hospital, IdiPAZMadridSpain
| | - Almari Conradie
- Global Alliance for TB Drug Development (TB Alliance)New YorkNew YorkUSA
| | - Majda Benhayoun
- Global Alliance for TB Drug Development (TB Alliance)New YorkNew YorkUSA
| | | | | | - Uwe Fuhr
- Department of Pharmacology, Faculty of Medicine and University Hospital Cologne, Center for PharmacologyUniversity of CologneCologneGermany
| | - Rosa Antonijoan Arbos
- Clinical Pharmacology DepartmentHospital de la Santa Creu i Sant PauBarcelonaSpain
- Drug Research CenterInstitut d'Investigació Biomèdica Sant Pau, IIB Sant PauBarcelonaSpain
- Pharmacology and Therapeutics DepartmentUniversitat Autònoma de Barcelona (UAB)BellaterraSpain
| | - Francisco Abad‐Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto Teófilo HernandoUniversidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria La Princesa (IP)MadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd)Instituto de Salud Carlos IIIMadridSpain
| | - Antonio Portolés
- Clinical Pharmacology Department, Hospital Clínico San CarlosInstituto de Investigación Sanitaria del Hospital Clínico San Carlos, IdISSCMadridSpain
- Pharmacology and Toxicology Department, School of MedicineUniversidad Complutense de Madrid (UCM)MadridSpain
| | | | - Antonio J. Carcas
- Clinical Pharmacology DepartmentLa Paz University Hospital, IdiPAZMadridSpain
- Pharmacology Department, School of MedicineUniversidad Autónoma de MadridMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades InfecciosasInstituto de Salud Carlos IIIMadridSpain
| | - Alberto M. Borobia
- Clinical Pharmacology DepartmentLa Paz University Hospital, IdiPAZMadridSpain
- Pharmacology Department, School of MedicineUniversidad Autónoma de MadridMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades InfecciosasInstituto de Salud Carlos IIIMadridSpain
| | | |
Collapse
|
2
|
Li Q, He Y, Pan J. CrossFuse-XGBoost: accurate prediction of the maximum recommended daily dose through multi-feature fusion, cross-validation screening and extreme gradient boosting. Brief Bioinform 2023; 25:bbad511. [PMID: 38216539 PMCID: PMC10786712 DOI: 10.1093/bib/bbad511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 01/14/2024] Open
Abstract
In the drug development process, approximately 30% of failures are attributed to drug safety issues. In particular, the first-in-human (FIH) trial of a new drug represents one of the highest safety risks, and initial dose selection is crucial for ensuring safety in clinical trials. With traditional dose estimation methods, which extrapolate data from animals to humans, catastrophic events have occurred during Phase I clinical trials due to interspecies differences in compound sensitivity and unknown molecular mechanisms. To address this issue, this study proposes a CrossFuse-extreme gradient boosting (XGBoost) method that can directly predict the maximum recommended daily dose of a compound based on existing human research data, providing a reference for FIH dose selection. This method not only integrates multiple features, including molecular representations, physicochemical properties and compound-protein interactions, but also improves feature selection based on cross-validation. The results demonstrate that the CrossFuse-XGBoost method not only improves prediction accuracy compared to that of existing local weighted methods [k-nearest neighbor (k-NN) and variable k-NN (v-NN)] but also solves the low prediction coverage issue of v-NN, achieving full coverage of the external validation set and enabling more reliable predictions. Furthermore, this study offers a high level of interpretability by identifying the importance of different features in model construction. The 241 features with the most significant impact on the maximum recommended daily dose were selected, providing references for optimizing the structure of new compounds and guiding experimental research. The datasets and source code are freely available at https://github.com/cqmu-lq/CrossFuse-XGBoost.
Collapse
Affiliation(s)
- Qiang Li
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Yu He
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jianbo Pan
- Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
3
|
Solovyeva O, Dimairo M, Weir CJ, Hee SW, Espinasse A, Ursino M, Patel D, Kightley A, Hughes S, Jaki T, Mander A, Evans TRJ, Lee S, Hopewell S, Rantell KR, Chan AW, Bedding A, Stephens R, Richards D, Roberts L, Kirkpatrick J, de Bono J, Yap C. Development of consensus-driven SPIRIT and CONSORT extensions for early phase dose-finding trials: the DEFINE study. BMC Med 2023; 21:246. [PMID: 37408015 PMCID: PMC10324137 DOI: 10.1186/s12916-023-02937-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/12/2023] [Indexed: 07/07/2023] Open
Abstract
BACKGROUND Early phase dose-finding (EPDF) trials are crucial for the development of a new intervention and influence whether it should be investigated in further trials. Guidance exists for clinical trial protocols and completed trial reports in the SPIRIT and CONSORT guidelines, respectively. However, both guidelines and their extensions do not adequately address the characteristics of EPDF trials. Building on the SPIRIT and CONSORT checklists, the DEFINE study aims to develop international consensus-driven guidelines for EPDF trial protocols (SPIRIT-DEFINE) and reports (CONSORT-DEFINE). METHODS The initial generation of candidate items was informed by reviewing published EPDF trial reports. The early draft items were refined further through a review of the published and grey literature, analysis of real-world examples, citation and reference searches, and expert recommendations, followed by a two-round modified Delphi process. Patient and public involvement and engagement (PPIE) was pursued concurrently with the quantitative and thematic analysis of Delphi participants' feedback. RESULTS The Delphi survey included 79 new or modified SPIRIT-DEFINE (n = 36) and CONSORT-DEFINE (n = 43) extension candidate items. In Round One, 206 interdisciplinary stakeholders from 24 countries voted and 151 stakeholders voted in Round Two. Following Round One feedback, one item for CONSORT-DEFINE was added in Round Two. Of the 80 items, 60 met the threshold for inclusion (≥ 70% of respondents voted critical: 26 SPIRIT-DEFINE, 34 CONSORT-DEFINE), with the remaining 20 items to be further discussed at the consensus meeting. The parallel PPIE work resulted in the development of an EPDF lay summary toolkit consisting of a template with guidance notes and an exemplar. CONCLUSIONS By detailing the development journey of the DEFINE study and the decisions undertaken, we envision that this will enhance understanding and help researchers in the development of future guidelines. The SPIRIT-DEFINE and CONSORT-DEFINE guidelines will allow investigators to effectively address essential items that should be present in EPDF trial protocols and reports, thereby promoting transparency, comprehensiveness, and reproducibility. TRIAL REGISTRATION SPIRIT-DEFINE and CONSORT-DEFINE are registered with the EQUATOR Network ( https://www.equator-network.org/ ).
Collapse
Affiliation(s)
| | - Munyaradzi Dimairo
- Clinical Trials Research Unit, School of Health and Related Research, University of Sheffield, Sheffield, UK
| | - Christopher J Weir
- Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Siew Wan Hee
- University Hospitals Coventry & Warwickshire NHS Trust, Coventry, UK
- University of Warwick, Coventry, UK
| | | | - Moreno Ursino
- Inserm, Centre de Recherche Des Cordeliers, Sorbonne UniversitéUniversité Paris Cité, 75006, Paris, France
- HeKA, Inria Paris, 75015, Paris, France
- Unit of Clinical Epidemiology, AP-HP, CHU Robert Debré, CIC-EC 1426, Paris, France
- RECaP/F-CRIN, Inserm, 5400, Nancy, France
| | | | - Andrew Kightley
- Patient and Public Involvement and Engagement (PPIE) Lead, Lichfield, UK
| | | | - Thomas Jaki
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
- University of Regensburg, Regensburg, Germany
| | | | | | - Shing Lee
- Columbia University, Mailman School of Public Health, New York, USA
| | - Sally Hopewell
- Oxford Clinical Trials Research Unit, University of Oxford, Oxford, UK
| | | | - An-Wen Chan
- Department of Medicine, Women's College Research Institute, University of Toronto, Toronto, Canada
| | | | | | | | | | | | - Johann de Bono
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | | |
Collapse
|
4
|
Ferreira GS, Dijkstra FM, Veening-Griffioen DH, Boon WPC, Schellekens H, Moors EHM, van Meer PJK, Stuurman FE, van Gerven JMA. Translatability of preclinical to early clinical tolerable and pharmacologically active dose ranges for central nervous system active drugs. Transl Psychiatry 2023; 13:74. [PMID: 36859342 PMCID: PMC9977891 DOI: 10.1038/s41398-023-02353-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 01/25/2023] [Accepted: 02/01/2023] [Indexed: 03/03/2023] Open
Abstract
The primary purpose of this study was to assess the translatability of preclinical to early clinical tolerable and pharmacologically active dose ranges for central nervous system (CNS) active drugs. As a part of this, IBs were reviewed on reporting quality. Investigator's Brochures (IBs) of studies performed at the Centre for Human Drug Research (CHDR) reporting statistically significant results of CNS activity related to the drug's mechanism of action were included. The quality of IBs was assessed based on the presence of a rationale for the chosen animal model, completeness of pharmacokinetic (PK) results in reporting and internal validity information of the preclinical evidence. The IB-derisk tool was used to generate preclinical and early clinical data overviews data. For each compound, the overlap between pharmacologically active dose ranges and well-tolerated levels was calculated for three pharmacokinetic (PK) parameters: human equivalent dose (HED), maximum plasma concentration (Cmax) and area under the curve (AUC). Twenty-five IBs were included. In general, the quality of reporting in IBs was assessed as poor. About a third of studies did not explore the entire concentration-effect curve (pre)clinically. Single dose tolerability ranges were most accurately predicted by Cmax. Human equivalent dose and AUC were the best predictors of pharmacologically active ranges. Tolerable and pharmacologically active dose ranges in healthy volunteers can be reasonably well predicted from preclinical data with the IB-derisk tool. The translatability of preclinical studies can be improved by applying a higher reporting standard in IBs including comparable PK measurements across all preclinical and clinical studies.
Collapse
Affiliation(s)
- Guilherme S Ferreira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Francis M Dijkstra
- Centre for Human Drug Research, Leiden, The Netherlands.
- Leiden University Medical Centre, Leiden, The Netherlands.
| | - Désirée H Veening-Griffioen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Wouter P C Boon
- Copernicus Institute of Sustainable Development, Innovation Studies, Utrecht University, Utrecht, The Netherlands
| | - Huub Schellekens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ellen H M Moors
- Copernicus Institute of Sustainable Development, Innovation Studies, Utrecht University, Utrecht, The Netherlands
| | - Peter J K van Meer
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Medicines Evaluation Board (CBG), Utrecht, The Netherlands
| | - Frederik E Stuurman
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Medical Centre, Leiden, The Netherlands
| | - Joop M A van Gerven
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Medical Centre, Leiden, The Netherlands
- Central Committee on Research Involving Human Subjects (CCMO), Leiden, The Netherlands
| |
Collapse
|
5
|
Germovsek E, Cheng M, Giragossian C. Allometric scaling of therapeutic monoclonal antibodies in preclinical and clinical settings. MAbs 2021; 13:1964935. [PMID: 34530672 PMCID: PMC8463036 DOI: 10.1080/19420862.2021.1964935] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/19/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023] Open
Abstract
Constant technological advancement enabled the production of therapeutic monoclonal antibodies (mAbs) and will continue to contribute to their rapid expansion. Compared to small-molecule drugs, mAbs have favorable characteristics, but also more complex pharmacokinetics (PK), e.g., target-mediated nonlinear elimination and recycling by neonatal Fc-receptor. This review briefly discusses mAb biology, similarities and differences in PK processes across species and within human, and provides a detailed overview of allometric scaling approaches for translating mAb PK from preclinical species to human and extrapolating from adults to children. The approaches described here will remain vital in mAb drug development, although more data are needed, for example, from very young patients and mAbs with nonlinear PK, to allow for more confident conclusions and contribute to further growth of this field. Improving mAb PK predictions will facilitate better planning of (pediatric) clinical studies and enable progression toward the ultimate goal of expediting drug development.
Collapse
Affiliation(s)
- Eva Germovsek
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | - Ming Cheng
- Development Biologicals, Drug Metabolism And Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| | - Craig Giragossian
- Biotherapeutics Discovery, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, US
| |
Collapse
|