1
|
Weddell J, Gulati A, Yamada A. Recommendations for a standardized publication protocol for a QSP model. J Pharmacokinet Pharmacodyn 2024:10.1007/s10928-024-09943-6. [PMID: 39390205 DOI: 10.1007/s10928-024-09943-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024]
Abstract
Development of a Quantitative Systems Pharmacology (QSP) model is a long process with many iterative steps. Lack of standard practices for publishing QSP models has resulted in limited model reproducibility within the field. Multiple studies have identified that model reproducibility is a large challenge, especially for QSP models. This work aimed to investigate the causes of QSP model reproducibility issues and suggest standard practices as a potential solution to ensure QSP models are reproducible. In addition, a protocol is suggested as a guidance towards better publication strategy across journals, hoping to enable QSP knowledge preservation.
Collapse
Affiliation(s)
- Jared Weddell
- Early Development, New Technology, Astellas Pharma Inc, Northbrook, IL, USA
| | - Abhishek Gulati
- Quantitative Pharmacology & Pharmacometrics, Merck & Co. Inc, West Point, PA, USA.
| | - Akihiro Yamada
- Early Development, New Technology, Astellas Pharma Inc, Tokyo, Japan
| |
Collapse
|
2
|
Meid AD, Scherkl C, Metzner M, Czock D, Seidling HM. Real-World Application of a Quantitative Systems Pharmacology (QSP) Model to Predict Potassium Concentrations from Electronic Health Records: A Pilot Case towards Prescribing Monitoring of Spironolactone. Pharmaceuticals (Basel) 2024; 17:1041. [PMID: 39204148 PMCID: PMC11357243 DOI: 10.3390/ph17081041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Quantitative systems pharmacology (QSP) models are rarely applied prospectively for decision-making in clinical practice. We therefore aimed to operationalize a QSP model for potas-sium homeostasis to predict potassium trajectories based on spironolactone administrations. For this purpose, we proposed a general workflow that was applied to electronic health records (EHR) from patients treated in a German tertiary care hospital. The workflow steps included model exploration, local and global sensitivity analyses (SA), identifiability analysis (IA) of model parameters, and specification of their inter-individual variability (IIV). Patient covariates, selected parameters, and IIV then defined prior information for the Bayesian a posteriori prediction of individual potassium trajectories of the following day. Following these steps, the successfully operationalized QSP model was interactively explored via a Shiny app. SA and IA yielded five influential and estimable parameters (extracellular fluid volume, hyperaldosteronism, mineral corticoid receptor abundance, potassium intake, sodium intake) for Bayesian prediction. The operationalized model was validated in nine pilot patients and showed satisfactory performance based on the (absolute) average fold error. This provides proof-of-principle for a Prescribing Monitoring of potassium concentrations in a hospital system, which could suggest preemptive clinical measures and therefore potentially avoid dangerous hyperkalemia or hypokalemia.
Collapse
Affiliation(s)
- Andreas D. Meid
- Internal Medicine IX: Department of Clinical Pharmacology and Pharmacoepidemiology, Medical Faculty Heidelberg/Heidelberg University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Camilo Scherkl
- Internal Medicine IX: Department of Clinical Pharmacology and Pharmacoepidemiology, Medical Faculty Heidelberg/Heidelberg University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Michael Metzner
- Internal Medicine IX: Department of Clinical Pharmacology and Pharmacoepidemiology, Medical Faculty Heidelberg/Heidelberg University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - David Czock
- Internal Medicine IX: Department of Clinical Pharmacology and Pharmacoepidemiology, Medical Faculty Heidelberg/Heidelberg University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Hanna M. Seidling
- Internal Medicine IX: Department of Clinical Pharmacology and Pharmacoepidemiology, Medical Faculty Heidelberg/Heidelberg University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
- Internal Medicine IX: Department of Clinical Pharmacology and Pharmacoepidemiology—Cooperation Unit Clinical Pharmacy, Medical Faculty Heidelberg/Heidelberg University Hospital, Heidelberg University, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| |
Collapse
|
3
|
van Noort M, Ruppert M. Two new user-friendly methods to assess pharmacometric parameter identifiability on categorical and continuous scales. CPT Pharmacometrics Syst Pharmacol 2024; 13:1160-1169. [PMID: 38715388 PMCID: PMC11247123 DOI: 10.1002/psp4.13147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 03/07/2024] [Accepted: 04/10/2024] [Indexed: 07/16/2024] Open
Abstract
Parameter identifiability methods assess whether the parameters of a model are uniquely determined by the observations. While the success of a model fit can provide some information on this, it can be valuable to determine identifiability before any fit has been attempted, or to separate identifiability from other issues. Two concepts that lean themselves well for identifiability analysis and have been underutilized are the sensitivity matrix (SM) and the Fisher information matrix (FIM). This paper presents two newly developed methods, one based on the SM and one based on the FIM. Both methods can assess local identifiability for a wide set of models, can be used with limited effort, and are freely available. The methods require the proposed model in the form of a set of differential equations, the parameter values, and the study design as input. They can be used a priori, as they do not need observed values or a successful model fit. Traditional methods provide a single categorical (yes/no) answer to the question of identifiability. In many cases, this is not very informative, and identifiability depends on study design (e.g., dose levels or observation times) and parameter values. Indicators on a continuous scale characterizing the level of identifiability would provide more detailed and relevant information, for example, to guide model development. Our two methods provide both categorical and continuous indicators. Both methods indicate which parameter combinations are difficult to identify by calculating the directions in parameter space that are least identifiable. The methods were validated with an example problem.
Collapse
|
4
|
van Noort M, Ruppert M, DeJongh J, Marostica E, Bosch R, Mešić E, Snelder N. Navigating the landscape of parameter identifiability methods: A workflow recommendation for model development. CPT Pharmacometrics Syst Pharmacol 2024; 13:1170-1179. [PMID: 38715385 PMCID: PMC11247124 DOI: 10.1002/psp4.13148] [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: 10/02/2023] [Revised: 03/07/2024] [Accepted: 04/10/2024] [Indexed: 07/16/2024] Open
Abstract
In pharmacometric modeling, it is often important to know whether the data is sufficiently rich to identify the parameters of a proposed model. While it may be possible to assess this based on the results of a model fit, it is often difficult to disentangle identifiability issues from other model fitting and numerical problems. Furthermore, it can be of value to ascertain identifiability beforehand. This paper compares four methods for parameter identifiability, namely Differential Algebra for Identifiability of SYstems (DAISY), the sensitivity matrix method (SMM), Aliasing, and the Fisher information matrix method (FIMM). We discuss the characteristics of the methods and apply them to a set of applications, consisting of frequently used PK model structures, with suitable dosing regimens and sampling times. These applications were selected to validate the methods and demonstrate their usefulness. While traditional identifiability analysis provides a categorical result [PLoS One, 6, 2011, e27755; CPT Pharmacometrics Syst Pharmacol, 8, 2019, 259; Bioinformatics, 30, 2014, 1440], we argue that in practice a continuous scale better reflects the limitations on the data and is more informative. The methods were generally consistent in their evaluation of the applications. The Fisher information matrix method seemed to provide the most consistent answers. All methods provided information on the parameters that were unidentifiable. Some of the results were unexpected, indicating identifiability issues where none were foreseen, but could be explained upon further analysis. This illustrated the usefulness of identifiability assessment.
Collapse
Affiliation(s)
| | | | | | | | | | - Emir Mešić
- LAP&P Consultants BV, Leiden, The Netherlands
| | | |
Collapse
|
5
|
Cucurull-Sanchez L. An industry perspective on current QSP trends in drug development. J Pharmacokinet Pharmacodyn 2024:10.1007/s10928-024-09905-y. [PMID: 38443663 DOI: 10.1007/s10928-024-09905-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 02/07/2024] [Indexed: 03/07/2024]
Abstract
2023 marks the 10th anniversary of Natpara's submission to the US FDA, which led to the first recorded regulatory interaction where a decision was supported by Quantitative and Systems Pharmacology (QSP) simulations. It had taken about 5 years for the timid QSP discipline to emerge as an effective Model-Informed Drug Development (MIDD) tool with visible impact in the pharmaceutical industry. Since then, the presence of QSP in the regulatory environment has continued to increase, to the point that the Agency reported 60 QSP submissions in 2020 alone, representing ~ 4% of their annual IND submissions [1]. What sort of industry mindset has enabled QSP to reach this level of success? How does QSP fit within the MIDD paradigm? Does QSP mean the same to Discovery and to Clinical Development projects? How do 'platforms' compare to 'fit-for-purpose' QSP models in an industrial setting? Can QSP and empirical Pharmacokinetic-Pharmacodynamic (PKPD) modelling be complementary? What level of validation is required to inform drug development decisions? This article reflects on all these questions, in particular addressing those audiences with limited line-of-sight into the drug industry decision-making machinery.
Collapse
|
6
|
Lemaire V, Hu C, van der Graaf PH, Chang S, Wang W. No Recipe for Quantitative Systems Pharmacology Model Validation, but a Balancing Act Between Risk and Cost. Clin Pharmacol Ther 2024; 115:25-28. [PMID: 37943003 DOI: 10.1002/cpt.3082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/16/2023] [Indexed: 11/10/2023]
Affiliation(s)
- Vincent Lemaire
- Clinical Pharmacology, Genentech, South San Francisco, California, USA
| | - Chuanpu Hu
- Clinical Pharmacology, Pharmacometrics, Disposition and Bioanalysis, Bristol Myers Squibb, Princeton, New Jersey, USA
| | | | - Steve Chang
- Immunetrics, Inc., Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
7
|
van Wijk RC, Mockeliunas L, van den Hoogen G, Upton CM, Diacon AH, Simonsson USH. Reproducibility in pharmacometrics applied in a phase III trial of BCG-vaccination for COVID-19. Sci Rep 2023; 13:16292. [PMID: 37770596 PMCID: PMC10539503 DOI: 10.1038/s41598-023-43412-3] [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: 06/15/2022] [Accepted: 09/23/2023] [Indexed: 09/30/2023] Open
Abstract
Large clinical trials often generate complex and large datasets which need to be presented frequently throughout the trial for interim analysis or to inform a data safety monitory board (DSMB). In addition, reliable and traceability are required to ensure reproducibility in pharmacometric data analysis. A reproducible pharmacometric analysis workflow was developed during a large clinical trial involving 1000 participants over one year testing Bacillus Calmette-Guérin (BCG) (re)vaccination in coronavirus disease 2019 (COVID-19) morbidity and mortality in frontline health care workers. The workflow was designed to review data iteratively during the trial, compile frequent reports to the DSMB, and prepare for rapid pharmacometric analysis. Clinical trial datasets (n = 41) were transferred iteratively throughout the trial for review. An RMarkdown based pharmacometric processing script was written to automatically generate reports for evaluation by the DSMB. Reports were compiled, reviewed, and sent to the DSMB on average three days after the data cut-off, reflecting the trial progress in real-time. The script was also utilized to prepare for the trial pharmacometric analyses. The same source data was used to create analysis datasets in NONMEM format and to support model script development. The primary endpoint analysis was completed three days after data lock and unblinding, and the secondary endpoint analyses two weeks later. The constructive collaboration between clinical, data management, and pharmacometric teams enabled this efficient, timely, and reproducible pharmacometrics workflow.
Collapse
Affiliation(s)
- Rob C van Wijk
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 75124, Uppsala, Sweden
| | - Laurynas Mockeliunas
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 75124, Uppsala, Sweden
| | | | | | | | - Ulrika S H Simonsson
- Department of Pharmaceutical Biosciences, Uppsala University, Box 591, 75124, Uppsala, Sweden.
| |
Collapse
|
8
|
Aldibani HKA, Rajput AJ, Rostami-Hodjegan A. In-depth analysis of patterns in selection of different physiologically-based pharmacokinetic modeling tools: Part II - Assessment of model reusability and comparison between open and non-open source-code software. Biopharm Drug Dispos 2023; 44:292-300. [PMID: 37083940 DOI: 10.1002/bdd.2360] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/17/2022] [Accepted: 04/04/2023] [Indexed: 04/22/2023]
Abstract
Whilst the reproducibility of models in the area of systems biology and quantitative systems pharmacology has been the focus of attention lately, the concept of 'reusability' is not addressed. With the advent of the 'Model Master File' dominating some regulatory discussions on pharmaceutical applications of physiologically-based pharmacokinetic (PBPK) models, reusability becomes a vital aspect of confidence in their use. Herein, we define 'reusability' specifically in the context of PBPK models and investigate the influence of open versus non-open source-code (NOSC) nature of the software on the extent of 'reusability'. Original articles (n = 145) that were associated with the development of novel PBPK models were identified as source models and citations to these reports, which involved further PBPK model development, were explored (n > 1800) for reuse cases of the source PBPK model whether in full or partial form. The nature of source-code was a major determinant of external reusability for PBPK models (>50% of the NOSC models as opposed <25% of open source-code [OSC]). Full reusability of the models was not common and mostly involved internal reuse of the OSC model (by the group who had previously developed the original model). The results were stratified by the software utilised (various), organisations involved (academia, industry, regulatory), and type of reusability (full vs. partial). The clear link between external reuse of models and NOSC PBPK software might stem from many elements related to quality and trust that require further investigation, and challenges the unfounded notion that OSC models are associated with higher uptake for reuse.
Collapse
Affiliation(s)
| | - Arham Jamaal Rajput
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, UK
- Certara UK Limited, Sheffield, UK
| |
Collapse
|
9
|
Jin YB, Liang XC, Cai JH, Wang K, Wang CY, Wang WH, Chen XL, Bao S. Mechanism of action of icaritin on uterine corpus endometrial carcinoma based on network pharmacology and experimental evaluation. Front Oncol 2023; 13:1205604. [PMID: 37538114 PMCID: PMC10394632 DOI: 10.3389/fonc.2023.1205604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/28/2023] [Indexed: 08/05/2023] Open
Abstract
Background Uterine corpus endometrial carcinoma (UCEC) belongs to a group of epithelial malignant tumors. Icaritin is the main active compound of Epimedii Folium. Icaritin has been utilized to induce UCEC cells to death. Methods We wished to identify potential targets for icaritin in the treatment of UCEC, as well as to provide a groundwork for future studies into its pharmacologic mechanism of action. Network pharmacology was employed to conduct investigations on icaritin. Target proteins were chosen from the components of icaritin for UCEC treatment. A protein-protein interaction (PPI) network was established using overlapping genes. Analyses of enrichment of function and signaling pathways were undertaken using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases, respectively, to select "hub genes". Finally, experiments were carried out to ascertain the effect of icaritin on endometrial cancer (HEC-1-A) cells. Results We demonstrated that icaritin has bioactive components and putative targets that are therapeutically important. Icaritin treatment induced sustained activation of the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt pathway) and inhibited growth of HEC-1-A cells. Conclusion Our data provide a rationale for preclinical and clinical evaluations of icaritin for UCEC therapy.
Collapse
Affiliation(s)
- Yan-Bin Jin
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, Hainan, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
| | - Xiao-Chen Liang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, Hainan, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
| | - Jun-Hong Cai
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
| | - Kang Wang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
| | - Chen-Yang Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wen-Hua Wang
- Department of Obstetrics and Gynecology, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiu-Li Chen
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, Hainan, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
| | - Shan Bao
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, Hainan, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, Hainan, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, Hainan, China
| |
Collapse
|
10
|
A Review of Quantitative Systems Pharmacology Models of the Coagulation Cascade: Opportunities for Improved Usability. Pharmaceutics 2023; 15:pharmaceutics15030918. [PMID: 36986779 PMCID: PMC10054658 DOI: 10.3390/pharmaceutics15030918] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
Despite the numerous therapeutic options to treat bleeding or thrombosis, a comprehensive quantitative mechanistic understanding of the effects of these and potential novel therapies is lacking. Recently, the quality of quantitative systems pharmacology (QSP) models of the coagulation cascade has improved, simulating the interactions between proteases, cofactors, regulators, fibrin, and therapeutic responses under different clinical scenarios. We aim to review the literature on QSP models to assess the unique capabilities and reusability of these models. We systematically searched the literature and BioModels database reviewing systems biology (SB) and QSP models. The purpose and scope of most of these models are redundant with only two SB models serving as the basis for QSP models. Primarily three QSP models have a comprehensive scope and are systematically linked between SB and more recent QSP models. The biological scope of recent QSP models has expanded to enable simulations of previously unexplainable clotting events and the drug effects for treating bleeding or thrombosis. Overall, the field of coagulation appears to suffer from unclear connections between models and irreproducible code as previously reported. The reusability of future QSP models can improve by adopting model equations from validated QSP models, clearly documenting the purpose and modifications, and sharing reproducible code. The capabilities of future QSP models can improve from more rigorous validation by capturing a broader range of responses to therapies from individual patient measurements and integrating blood flow and platelet dynamics to closely represent in vivo bleeding or thrombosis risk.
Collapse
|
11
|
Desikan R, Jayachandran P. CYTOCON DB: A versatile database of human cell and molecule concentrations for accelerating model development. CPT Pharmacometrics Syst Pharmacol 2023; 12:5-7. [PMID: 36633255 PMCID: PMC9835113 DOI: 10.1002/psp4.12897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 11/17/2022] [Indexed: 01/13/2023] Open
Affiliation(s)
- Rajat Desikan
- Clinical Pharmacology Modeling & Simulation (CPMS)GSKStevenageUK
| | | |
Collapse
|
12
|
Parker C, Nelson E, Zhang T. VeVaPy, a Python Platform for Efficient Verification and Validation of Systems Biology Models with Demonstrations Using Hypothalamic-Pituitary-Adrenal Axis Models. ENTROPY (BASEL, SWITZERLAND) 2022; 24:1747. [PMID: 36554152 PMCID: PMC9777964 DOI: 10.3390/e24121747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 06/17/2023]
Abstract
In order for mathematical models to make credible contributions, it is essential for them to be verified and validated. Currently, verification and validation (V&V) of these models does not meet the expectations of the system biology and systems pharmacology communities. Partially as a result of this shortfall, systemic V&V of existing models currently requires a lot of time and effort. In order to facilitate systemic V&V of chosen hypothalamic-pituitary-adrenal (HPA) axis models, we have developed a computational framework named VeVaPy-taking care to follow the recommended best practices regarding the development of mathematical models. VeVaPy includes four functional modules coded in Python, and the source code is publicly available. We demonstrate that VeVaPy can help us efficiently verify and validate the five HPA axis models we have chosen. Supplied with new and independent data, VeVaPy outputs objective V&V benchmarks for each model. We believe that VeVaPy will help future researchers with basic modeling and programming experience to efficiently verify and validate mathematical models from the fields of systems biology and systems pharmacology.
Collapse
Affiliation(s)
- Christopher Parker
- Department of Pharmacology & Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Erik Nelson
- Department of Psychiatry & Behavioral Neuroscience, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Tongli Zhang
- Department of Pharmacology & Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
13
|
Androulakis IP. Towards a comprehensive assessment of QSP models: what would it take? J Pharmacokinet Pharmacodyn 2022:10.1007/s10928-022-09820-0. [PMID: 35962928 PMCID: PMC9922790 DOI: 10.1007/s10928-022-09820-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 07/15/2022] [Indexed: 10/15/2022]
Abstract
Quantitative Systems Pharmacology (QSP) has emerged as a powerful ensemble of approaches aiming at developing integrated mathematical and computational models elucidating the complex interactions between pharmacology, physiology, and disease. As the field grows and matures its applications expand beyond the boundaries of research and development and slowly enter the decision making and regulatory arenas. However, widespread acceptance and eventual adoption of a new modeling approach requires assessment criteria and quantifiable metrics that establish credibility and increase confidence in model predictions. QSP aims to provide an integrated understanding of pathology in the context of therapeutic interventions. Because of its ambitious nature and the fact that QSP emerged in an uncoordinated manner as a result of activities distributed across organizations and academic institutions, high entropy characterizes the tools, methods, and computational methodologies and approaches used. The eventual acceptance of QSP model predictions as supporting material for an application to a regulatory agency will require that two key aspects are considered: (1) increase confidence in the QSP framework, which drives standardization and assessment; and (2) careful articulation of the expectations. Both rely heavily on our ability to rigorously and consistently assess QSP models. In this manuscript, we wish to discuss the meaning and purpose of such an assessment in the context of QSP model development and elaborate on the differentiating features of QSP that render such an endeavor challenging. We argue that QSP establishes a conceptual, integrative framework rather than a specific and well-defined computational methodology. QSP elicits the use of a wide variety of modeling and computational methodologies optimized with respect to specific applications and available data modalities, which exceed the data structures employed by chemometrics and PK/PD models. While the range of options fosters creativity and promises to substantially advance our ability to design pharmaceutical interventions rationally and optimally, our expectations of QSP models need to be clearly articulated and agreed on, with assessment emphasizing the scope of QSP studies rather than the methods used. Nevertheless, QSP should not be considered an independent approach, rather one of many in the broader continuum of computational models.
Collapse
Affiliation(s)
- Ioannis P Androulakis
- Biomedical Engineering Department and Chemical & Biochemical Engineering Department, Rutgers, The State University of New Jersey, New Brunswick, USA.
| |
Collapse
|
14
|
Chan JR, Allen R, Boras B, Cabal A, Damian V, Gibbons FD, Gulati A, Hosseini I, Kearns JD, Saito R, Cucurull-Sanchez L, Selimkhanov J, Stein AM, Umehara K, Wang G, Wang W, Neves-Zaph S. Current practices for QSP model assessment: an IQ consortium survey. J Pharmacokinet Pharmacodyn 2022:10.1007/s10928-022-09811-1. [PMID: 35953664 PMCID: PMC9371373 DOI: 10.1007/s10928-022-09811-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/03/2022] [Indexed: 12/02/2022]
Abstract
Quantitative Systems Pharmacology (QSP) modeling is increasingly applied in the pharmaceutical industry to influence decision making across a wide range of stages from early discovery to clinical development to post-marketing activities. Development of standards for how these models are constructed, assessed, and communicated is of active interest to the modeling community and regulators but is complicated by the wide variability in the structures and intended uses of the underlying models and the diverse expertise of QSP modelers. With this in mind, the IQ Consortium conducted a survey across the pharmaceutical/biotech industry to understand current practices for QSP modeling. This article presents the survey results and provides insights into current practices and methods used by QSP practitioners based on model type and the intended use at various stages of drug development. The survey also highlights key areas for future development including better integration with statistical methods, standardization of approaches towards virtual populations, and increased use of QSP models for late-stage clinical development and regulatory submissions.
Collapse
Affiliation(s)
- Jason R Chan
- Global PKPD and Pharmacometrics, Eli Lilly and Company, Indianapolis, IN, 46285, USA.
| | - Richard Allen
- Worldwide Research, Development and Medical, Pfizer Inc. Kendall Square, Cambridge, MA, 02139, USA
| | - Britton Boras
- Worldwide Research, Development and Medical, Pfizer Inc.,, La Jolla, CA, 92121, USA
| | | | | | | | | | | | - Jeffrey D Kearns
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Ryuta Saito
- Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, Yokohama, Japan
| | | | | | - Andrew M Stein
- Pharmacometrics, Novartis Institutes of BioMedical Research, Cambridge, MA, USA
| | - Kenichi Umehara
- Roche Pharmaceutical Research and Early Development, Basel, Switzerland
| | - Guanyu Wang
- Drug Metabolism and Pharmacokinetics, Vertex Pharmaceuticals, Abingdon, Oxfordshire, UK
| | - Weirong Wang
- Clinical Pharmacology and Pharmacometrics, Janssen Research and Development, LLC, Spring House, PA, 19477, USA
| | | |
Collapse
|
15
|
Brown LV, Coles MC, McConnell M, Ratushny AV, Gaffney EA. Analysis of cellular kinetic models suggest that physiologically based model parameters may be inherently, practically unidentifiable. J Pharmacokinet Pharmacodyn 2022; 49:539-556. [PMID: 35933452 PMCID: PMC9508223 DOI: 10.1007/s10928-022-09819-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 07/13/2022] [Indexed: 11/30/2022]
Abstract
Physiologically-based pharmacokinetic and cellular kinetic models are used extensively to predict concentration profiles of drugs or adoptively transferred cells in patients and laboratory animals. Models are fit to data by the numerical optimisation of appropriate parameter values. When quantities such as the area under the curve are all that is desired, only a close qualitative fit to data is required. When the biological interpretation of the model that produced the fit is important, an assessment of uncertainties is often also warranted. Often, a goal of fitting PBPK models to data is to estimate parameter values, which can then be used to assess characteristics of the fit system or applied to inform new modelling efforts and extrapolation, to inform a prediction under new conditions. However, the parameters that yield a particular model output may not necessarily be unique, in which case the parameters are said to be unidentifiable. We show that the parameters in three published physiologically-based pharmacokinetic models are practically (deterministically) unidentifiable and that it is challenging to assess the associated parameter uncertainty with simple curve fitting techniques. This result could affect many physiologically-based pharmacokinetic models, and we advocate more widespread use of thorough techniques and analyses to address these issues, such as established Markov Chain Monte Carlo and Bayesian methodologies. Greater handling and reporting of uncertainty and identifiability of fit parameters would directly and positively impact interpretation and translation for physiologically-based model applications, enhancing their capacity to inform new model development efforts and extrapolation in support of future clinical decision-making.
Collapse
Affiliation(s)
- Liam V Brown
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK.
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | - Mark C Coles
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Mark McConnell
- Bristol Myers Squibb, Seattle, WA, USA
- Currently Chinook Therapeutics, Seattle, WA, USA
| | | | - Eamonn A Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Desikan R, Padmanabhan P, Kierzek AM, van der Graaf PH. Mechanistic Models of COVID-19: Insights into Disease Progression, Vaccines, and Therapeutics. Int J Antimicrob Agents 2022; 60:106606. [PMID: 35588969 PMCID: PMC9110059 DOI: 10.1016/j.ijantimicag.2022.106606] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 04/27/2022] [Accepted: 05/08/2022] [Indexed: 12/02/2022]
Abstract
The COVID-19 pandemic has severely impacted health systems and economies worldwide. Significant global efforts are therefore ongoing to improve vaccine efficacies, optimize vaccine deployment, and develop new antiviral therapies to combat the pandemic. Mechanistic viral dynamics and quantitative systems pharmacology models of SARS-CoV-2 infection, vaccines, immunomodulatory agents, and antiviral therapeutics have played a key role in advancing our understanding of SARS-CoV-2 pathogenesis and transmission, the interplay between innate and adaptive immunity to influence the outcomes of infection, effectiveness of treatments, mechanisms and performance of COVID-19 vaccines, and the impact of emerging SARS-CoV-2 variants. Here, we review some of the critical insights provided by these models and discuss the challenges ahead.
Collapse
Affiliation(s)
- Rajat Desikan
- Quantitative Systems Pharmacology (QSP) group, Certara, Sheffield and Canterbury, United Kingdom.
| | - Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Andrzej M Kierzek
- Quantitative Systems Pharmacology (QSP) group, Certara, Sheffield and Canterbury, United Kingdom; School of Biosciences and Medicine, University of Surrey, Guildford, United Kingdom
| | - Piet H van der Graaf
- Quantitative Systems Pharmacology (QSP) group, Certara, Sheffield and Canterbury, United Kingdom; Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
17
|
Miyano T, Irvine AD, Tanaka RJ. Model-based meta-analysis to optimise S. aureus-targeted therapies for atopic dermatitis. JID INNOVATIONS 2022; 2:100110. [PMID: 35757782 PMCID: PMC9214323 DOI: 10.1016/j.xjidi.2022.100110] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/29/2022] Open
Abstract
Several clinical trials of Staphylococcus aureus (S. aureus)‒targeted therapies for atopic dermatitis (AD) have shown conflicting results about whether they improve AD severity scores. This study performs a model-based meta-analysis to investigate the possible causes of these conflicting results and suggests how to improve the efficacies of S. aureus‒targeted therapies. We developed a mathematical model that describes systems-level AD pathogenesis involving dynamic interactions between S. aureus and coagulase-negative Staphylococcus (CoNS). Our model simulation reproduced the clinically observed detrimental effects of the application of S. hominis A9 and flucloxacillin on AD severity and showed that these effects disappeared if the bactericidal activity against CoNS was removed. A hypothetical (modeled) eradication of S. aureus by 3.0 log10 colony-forming unit per cm2 without killing CoNS achieved Eczema Area and Severity Index 75 comparable with that of dupilumab. This efficacy was potentiated if dupilumab was administered in conjunction with S. aureus eradication (Eczema Area and Severity Index 75 at week 16) (S. aureus eradication: 66.7%, dupilumab 61.6% and combination 87.8%). The improved efficacy was also seen for virtual dupilumab poor responders. Our model simulation suggests that killing CoNS worsens AD severity and that S. aureus‒specific eradication without killing CoNS could be effective for patients with AD, including dupilumab poor responders. This study will contribute to designing promising S. aureus‒targeted therapy.
Collapse
Affiliation(s)
- Takuya Miyano
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Alan D. Irvine
- Pediatric Dermatology, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, College of Medicine, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | - Reiko J. Tanaka
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Correspondence: Reiko J. Tanaka, Department of Bioengineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom.
| |
Collapse
|
18
|
Fitzpatrick R, Stefan MI. Validation Through Collaboration: Encouraging Team Efforts to Ensure Internal and External Validity of Computational Models of Biochemical Pathways. Neuroinformatics 2022; 20:277-284. [PMID: 35543917 PMCID: PMC9537119 DOI: 10.1007/s12021-022-09584-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2022] [Indexed: 01/09/2023]
Abstract
Computational modelling of biochemical reaction pathways is an increasingly important part of neuroscience research. In order to be useful, computational models need to be valid in two senses: First, they need to be consistent with experimental data and able to make testable predictions (external validity). Second, they need to be internally consistent and independently reproducible (internal validity). Here, we discuss both types of validity and provide a brief overview of tools and technologies used to ensure they are met. We also suggest the introduction of new collaborative technologies to ensure model validity: an incentivised experimental database for external validity and reproducibility audits for internal validity. Both rely on FAIR principles and on collaborative science practices.
Collapse
Affiliation(s)
- Richard Fitzpatrick
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK ,School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Melanie I. Stefan
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK ,ZJU-UoE Institute, Zhejiang University, Haining, China
| |
Collapse
|
19
|
Braakman S, Pathmanathan P, Moore H. Evaluation framework for systems models. CPT Pharmacometrics Syst Pharmacol 2021; 11:264-289. [PMID: 34921743 PMCID: PMC8923730 DOI: 10.1002/psp4.12755] [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: 11/04/2019] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 12/16/2022] Open
Abstract
As decisions in drug development increasingly rely on predictions from mechanistic systems models, assessing the predictive capability of such models is becoming more important. Several frameworks for the development of quantitative systems pharmacology (QSP) models have been proposed. In this paper, we add to this body of work with a framework that focuses on the appropriate use of qualitative and quantitative model evaluation methods. We provide details and references for those wishing to apply these methods, which include sensitivity and identifiability analyses, as well as concepts such as validation and uncertainty quantification. Many of these methods have been used successfully in other fields, but are not as common in QSP modeling. We illustrate how to apply these methods to evaluate QSP models, and propose methods to use in two case studies. We also share examples of misleading results when inappropriate analyses are used.
Collapse
Affiliation(s)
- Sietse Braakman
- Application Engineering, MathWorks Inc, Natick, Massachusetts, USA
| | - Pras Pathmanathan
- Office of Science and Engineering Laboratories (OSEL), Center for Devices and Radiological Health (CDRH), US Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | - Helen Moore
- Laboratory for Systems Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
20
|
Aghamiri SS, Amin R, Helikar T. Recent applications of quantitative systems pharmacology and machine learning models across diseases. J Pharmacokinet Pharmacodyn 2021; 49:19-37. [PMID: 34671863 PMCID: PMC8528185 DOI: 10.1007/s10928-021-09790-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/07/2021] [Indexed: 12/29/2022]
Abstract
Quantitative systems pharmacology (QSP) is a quantitative and mechanistic platform describing the phenotypic interaction between drugs, biological networks, and disease conditions to predict optimal therapeutic response. In this meta-analysis study, we review the utility of the QSP platform in drug development and therapeutic strategies based on recent publications (2019-2021). We gathered recent original QSP models and described the diversity of their applications based on therapeutic areas, methodologies, software platforms, and functionalities. The collection and investigation of these publications can assist in providing a repository of recent QSP studies to facilitate the discovery and further reusability of QSP models. Our review shows that the largest number of QSP efforts in recent years is in Immuno-Oncology. We also addressed the benefits of integrative approaches in this field by presenting the applications of Machine Learning methods for drug discovery and QSP models. Based on this meta-analysis, we discuss the advantages and limitations of QSP models and propose fields where the QSP approach constitutes a valuable interface for more investigations to tackle complex diseases and improve drug development.
Collapse
Affiliation(s)
- Sara Sadat Aghamiri
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Rada Amin
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA.
| | - Tomáš Helikar
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA.
| |
Collapse
|
21
|
Kapitanov GI, Chabot JR, Narula J, Roy M, Neubert H, Palandra J, Farrokhi V, Johnson JS, Webster R, Jones HM. A Mechanistic Site-Of-Action Model: A Tool for Informing Right Target, Right Compound, And Right Dose for Therapeutic Antagonistic Antibody Programs. FRONTIERS IN BIOINFORMATICS 2021; 1:731340. [DOI: 10.3389/fbinf.2021.731340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Quantitative modeling is increasingly utilized in the drug discovery and development process, from the initial stages of target selection, through clinical studies. The modeling can provide guidance on three major questions–is this the right target, what are the right compound properties, and what is the right dose for moving the best possible candidate forward. In this manuscript, we present a site-of-action modeling framework which we apply to monoclonal antibodies against soluble targets. We give a comprehensive overview of how we construct the model and how we parametrize it and include several examples of how to apply this framework for answering the questions postulated above. The utilities and limitations of this approach are discussed.
Collapse
|
22
|
Rowland Yeo K, Hennig S, Krishnaswami S, Strydom N, Ayyar VS, French J, Sinha V, Sobie E, Zhao P, Friberg LE, Mentré F. CPT: Pharmacometrics & Systems Pharmacology - Inception, Maturation, and Future Vision. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:649-657. [PMID: 34298582 PMCID: PMC8302238 DOI: 10.1002/psp4.12680] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022]
Affiliation(s)
| | | | | | - Natasha Strydom
- Department of Bioengineering and Therapeutic Sciences, School of Pharmacy, University of California, San Francisco, CA, USA
| | | | | | | | - Eric Sobie
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ping Zhao
- Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Lena E Friberg
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
23
|
Bai JPF, Schmidt BJ, Gadkar KG, Damian V, Earp JC, Friedrich C, van der Graaf PH, Madabushi R, Musante CJ, Naik K, Rogge M, Zhu H. FDA-Industry Scientific Exchange on assessing quantitative systems pharmacology models in clinical drug development: a meeting report, summary of challenges/gaps, and future perspective. AAPS JOURNAL 2021; 23:60. [PMID: 33931790 DOI: 10.1208/s12248-021-00585-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023]
Abstract
The pharmaceutical industry is actively applying quantitative systems pharmacology (QSP) to make internal decisions and guide drug development. To facilitate the eventual development of a common framework for assessing the credibility of QSP models for clinical drug development, scientists from US Food and Drug Administration and the pharmaceutical industry organized a full-day virtual Scientific Exchange on July 1, 2020. An assessment form was used to ensure consistency in the evaluation process. Among the cases presented, QSP was applied to various therapeutic areas. Applications mostly focused on phase 2 dose selection. Model transparency, including details on expert knowledge and data used for model development, was identified as a major factor for robust model assessment. The case studies demonstrated some commonalities in the workflow of QSP model development, calibration, and validation but differ in the size, scope, and complexity of QSP models, in the acceptance criteria for model calibration and validation, and in the algorithms/approaches used for creating virtual patient populations. Though efforts are being made to build the credibility of QSP models and the confidence is increasing in applying QSP for internal decisions at the clinical stages of drug development, there are still many challenges facing QSP application to late stage drug development. The QSP community needs a strategic plan that includes the ability and flexibility to Adapt, to establish Common expectations for model Credibility needed to inform drug Labeling and patient care, and to AIM to achieve the goal (ACCLAIM).
Collapse
Affiliation(s)
- Jane P F Bai
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20903, USA.
| | - Brian J Schmidt
- Quantitative Clinical Pharmacology, Bristol Myers Squibb, Princeton, New Jersey, USA.
| | - Kapil G Gadkar
- Development Sciences, Genentech Inc., South San Francisco, California, 94080, USA. .,Denali Therapeutics, San Francisco, California, USA.
| | - Valeriu Damian
- GSK R&D - Upper Providence, 1250 S Collegeville Rd, Collegeville, Pennsylvania, 19426, USA
| | - Justin C Earp
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | | | - Piet H van der Graaf
- Certara, Canterbury, CT2 7FG, UK.,Leiden Academic Centre for Drug Research, Leiden, 2333, CC, the Netherlands
| | - Rajanikanth Madabushi
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Cynthia J Musante
- Early Clinical Development, Pfizer Worldwide Research, Development, & Medical, 1 Portland Street, Cambridge, Massachusetts, 02139, USA
| | - Kunal Naik
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Mark Rogge
- Quantitative Translational Science, Takeda Pharmaceuticals International Co, 40 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
| | - Hao Zhu
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| |
Collapse
|
24
|
|
25
|
Bai JPF, Earp JC, Strauss DG, Zhu H. A Perspective on Quantitative Systems Pharmacology Applications to Clinical Drug Development. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 9:675-677. [PMID: 33159491 PMCID: PMC7762807 DOI: 10.1002/psp4.12567] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 10/14/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Jane P F Bai
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Justin C Earp
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - David G Strauss
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hao Zhu
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
26
|
Betts A, van der Graaf PH. Mechanistic Quantitative Pharmacology Strategies for the Early Clinical Development of Bispecific Antibodies in Oncology. Clin Pharmacol Ther 2020; 108:528-541. [PMID: 32579234 PMCID: PMC7484986 DOI: 10.1002/cpt.1961] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023]
Abstract
Bispecific antibodies (bsAbs) have become an integral component of the therapeutic research strategy to treat cancer. In addition to clinically validated immune cell re‐targeting, bsAbs are being designed for tumor targeting and as dual immune modulators. Explorative preclinical and emerging clinical data indicate potential for enhanced efficacy and reduced systemic toxicity. However, bsAbs are a complex modality with challenges to overcome in early clinical trials, including selection of relevant starting doses using a minimal anticipated biological effect level approach, and predicting efficacious dose despite nonintuitive dose response relationships. Multiple factors can contribute to variability in the clinic, including differences in functional affinity due to avidity, receptor expression, effector to target cell ratio, and presence of soluble target. Mechanistic modeling approaches are a powerful integrative tool to understand the complexities and aid in clinical translation, trial design, and prediction of regimens and strategies to reduce dose limiting toxicities of bsAbs. In this tutorial, the use of mechanistic modeling to impact decision making for bsAbs is presented and illustrated using case study examples.
Collapse
Affiliation(s)
- Alison Betts
- Applied Biomath, Concord, Massachusetts, USA.,Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands
| | - Piet H van der Graaf
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden, The Netherlands.,Certara, Canterbury, UK
| |
Collapse
|
27
|
Carter SJ, Chouhan B, Sharma P, Chappell MJ. Prediction of Clinical Transporter-Mediated Drug-Drug Interactions via Comeasurement of Pitavastatin and Eltrombopag in Human Hepatocyte Models. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 9:211-221. [PMID: 32142598 PMCID: PMC7179958 DOI: 10.1002/psp4.12505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 01/27/2020] [Indexed: 11/21/2022]
Abstract
A structurally identifiable micro‐rate constant mechanistic model was used to describe the interaction between pitavastatin and eltrombopag, with improved goodness‐of‐fit values through comeasurement of pitavastatin and eltrombopag. Transporter association and dissociation rate constants and passive rates out of the cell were similar between pitavastatin and eltrombopag. Translocation into the cell through transporter‐mediated uptake was six times greater for pitavastatin, leading to pronounced inhibition of pitavastatin uptake by eltrombopag. The passive rate into the cell was 91 times smaller for pitavastatin compared with eltrombopag. A semimechanistic physiologically‐based pharmacokinetic (PBPK) model was developed to evaluate the potential for clinical drug–drug interactions (DDIs). The PBPK model predicted a twofold increase in the pitavastatin peak blood concentration and area under the concentration‐time curve in the presence of eltrombopag in simulated healthy volunteers. The use of structural identifiability supporting experimental design combined with robust micro‐rate constant parameter estimates and a semimechanistic PBPK model gave more informed predictions of transporter‐mediated DDIs.
Collapse
Affiliation(s)
- Simon J Carter
- Biomedical and Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK
| | - Bhavik Chouhan
- Functional & Mechanistic Safety, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca R&D, Gothenburg, Sweden
| | - Pradeep Sharma
- Clinical Pharmacology & Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca R&D, Cambridge, UK
| | - Michael J Chappell
- Biomedical and Biological Systems Laboratory, School of Engineering, University of Warwick, Coventry, UK
| |
Collapse
|
28
|
Hosseini I, Feigelman J, Gajjala A, Susilo M, Ramakrishnan V, Ramanujan S, Gadkar K. gQSPSim: A SimBiology-Based GUI for Standardized QSP Model Development and Application. CPT Pharmacometrics Syst Pharmacol 2020; 9:165-176. [PMID: 31957304 PMCID: PMC7080534 DOI: 10.1002/psp4.12494] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/25/2019] [Indexed: 01/07/2023] Open
Abstract
Quantitative systems pharmacology (QSP) models are often implemented using a wide variety of technical workflows and methodologies. To facilitate reproducibility, transparency, portability, and reuse for QSP models, we have developed gQSPSim, a graphical user interface–based MATLAB application that performs key steps in QSP model development and analyses. The capabilities of gQSPSim include (i) model calibration using global and local optimization methods, (ii) development of virtual subjects to explore variability and uncertainty in the represented biology, and (iii) simulations of virtual populations for different interventions. gQSPSim works with SimBiology‐built models using components such as species, doses, variants, and rules. All functionalities are equipped with an interactive visualization interface and the ability to generate presentation‐ready figures. In addition, standardized gQSPSim sessions can be shared and saved for future extension and reuse. In this work, we demonstrate gQSPSim’s capabilities with a standard target‐mediated drug disposition model and a published model of anti‐proprotein convertase subtilisin/kexin type 9 (PCSK9) treatment of hypercholesterolemia.
Collapse
Affiliation(s)
- Iraj Hosseini
- Genentech Inc., South San Francisco, California, USA
| | | | - Anita Gajjala
- Consulting Services, MathWorks, Natick, Massachusetts, USA
| | - Monica Susilo
- Genentech Inc., South San Francisco, California, USA
| | | | | | - Kapil Gadkar
- Genentech Inc., South San Francisco, California, USA
| |
Collapse
|
29
|
Barrett JS, Bucci-Rechtweg C, Amy Cheung SY, Gamalo-Siebers M, Haertter S, Karres J, Marquard J, Mulugeta Y, Ollivier C, Strougo A, Yanoff L, Yao L, Zeitler P. Pediatric Extrapolation in Type 2 Diabetes: Future Implications of a Workshop. Clin Pharmacol Ther 2020; 108:29-39. [PMID: 32017043 PMCID: PMC7383960 DOI: 10.1002/cpt.1805] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 01/27/2020] [Indexed: 12/30/2022]
Abstract
Extrapolation from adults to youth with type 2 diabetes (T2D) is challenged by differences in disease progression and manifestation. This manuscript presents the results of a mock-team workshop focused on examining the typical team-based decision process used to propose a pediatric development plan for T2D addressing the viability of extrapolation. The workshop was held at the American Society for Clinical Pharmacology and Therapeutics (ASCPT) in Orlando, Florida on March 21, 2018.
Collapse
Affiliation(s)
- Jeffrey S Barrett
- Quantitative Sciences, Bill & Melinda Gates Medical Research Institute, Cambridge, Massachusetts, USA
| | - Christina Bucci-Rechtweg
- Pediatric & Maternal Health Policy, Regulatory Affairs, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | | | - Sebastian Haertter
- Translational Med & Clinical Pharmacology, Boehringer Ingelheim, Biberach, Germany
| | - Janina Karres
- Paediatric Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Jan Marquard
- Global Clinical Development CardioMetabolism, Boehringer Ingelheim, Ingelheim, Germany
| | - Yeruk Mulugeta
- Division of Pediatric and Maternal Health, Office of New Drugs, Center for Drug Evaluation and Research, Washington, DC, USA
| | | | - Ashley Strougo
- Translational Medicine, Pharmacokinetics, Dynamics and Metabolism, Sanofi, Frankfurt, Germany
| | - Lisa Yanoff
- Division of Metabolism and Endocrinology Products, Office of New Drugs, Center for Drug Evaluation and Research, Washington, DC, USA
| | - Lynne Yao
- Division of Pediatric and Maternal Health, Office of New Drugs, Center for Drug Evaluation and Research, Washington, DC, USA
| | - Philip Zeitler
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
30
|
Kirouac DC. Open Models for Clinical Pharmacology. Clin Pharmacol Ther 2020; 107:700-702. [PMID: 31983060 DOI: 10.1002/cpt.1738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/22/2019] [Indexed: 12/16/2022]
|
31
|
Bradshaw EL, Spilker ME, Zang R, Bansal L, He H, Jones RD, Le K, Penney M, Schuck E, Topp B, Tsai A, Xu C, Nijsen MJ, Chan JR. Applications of Quantitative Systems Pharmacology in Model-Informed Drug Discovery: Perspective on Impact and Opportunities. CPT Pharmacometrics Syst Pharmacol 2019; 8:777-791. [PMID: 31535440 PMCID: PMC6875708 DOI: 10.1002/psp4.12463] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
Quantitative systems pharmacology (QSP) approaches have been increasingly applied in the pharmaceutical since the landmark white paper published in 2011 by a National Institutes of Health working group brought attention to the discipline. In this perspective, we discuss QSP in the context of other modeling approaches and highlight the impact of QSP across various stages of drug development and therapeutic areas. We discuss challenges to the field as well as future opportunities.
Collapse
Affiliation(s)
| | - Mary E. Spilker
- Pfizer Worldwide Research and DevelopmentSan DiegoCaliforniaUSA
| | | | | | - Handan He
- Novartis Institutes for Biomedical ResearchEast HanoverNew JerseyUSA
| | | | - Kha Le
- AgiosCambridgeMassachusettsUSA
| | | | | | | | - Alice Tsai
- Vertex Pharmaceuticals IncorporatedBostonMassachusettsUSA
| | | | | | | |
Collapse
|
32
|
Bai JPF, Earp JC, Pillai VC. Translational Quantitative Systems Pharmacology in Drug Development: from Current Landscape to Good Practices. AAPS JOURNAL 2019; 21:72. [PMID: 31161268 DOI: 10.1208/s12248-019-0339-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/07/2019] [Indexed: 12/12/2022]
Abstract
Systems pharmacology approaches have the capability of quantitatively linking the key biological molecules relevant to a drug candidate's mechanism of action (drug-induced signaling pathways) to the clinical biomarkers associated with the proposed target disease, thereby quantitatively facilitating its development and life cycle management. In this review, the model attributes of published quantitative systems pharmacology (QSP) modeling for lowering cholesterol, treating salt-sensitive hypertension, and treating rare diseases as well as describing bone homeostasis and related pharmacological effects are critically reviewed with respect to model quality, calibration, validation, and performance. We further reviewed the common practices in optimizing QSP modeling and prediction. Notably, leveraging genetics and genomic studies for model calibration and validation is common. Statistical and quantitative assessment of QSP prediction and handling of model uncertainty are, however, mostly lacking as are the quantitative and statistical criteria for assessing QSP predictions and the covariance matrix of coefficients between the parameters in a validated virtual population. To accelerate advances and application of QSP with consistent quality, a list of key questions is proposed to be addressed when assessing the quality of a QSP model in hopes of stimulating the scientific community to set common expectations. The common expectations as to what constitutes the best QSP modeling practices, which the scientific community supports, will advance QSP modeling in the realm of informed drug development. In the long run, good practices will extend the life cycles of QSP models beyond the life cycles of individual drugs.
Collapse
Affiliation(s)
- Jane P F Bai
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20903, USA.
| | - Justin C Earp
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| | - Venkateswaran C Pillai
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20903, USA
| |
Collapse
|
33
|
Ramanujan S, Chan JR, Friedrich CM, Thalhauser CJ. A Flexible Approach for Context-Dependent Assessment of Quantitative Systems Pharmacology Models. CPT Pharmacometrics Syst Pharmacol 2019; 8:340-343. [PMID: 30983158 PMCID: PMC6617835 DOI: 10.1002/psp4.12409] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 03/26/2019] [Indexed: 12/12/2022] Open
Affiliation(s)
| | - Jason R. Chan
- Eli Lilly and CoLilly Corporate CenterIndianapolisIndianaUSA
| | | | | |
Collapse
|
34
|
Weis M, Baillie R, Friedrich C. Considerations for Adapting Pre-existing Mechanistic Quantitative Systems Pharmacology Models for New Research Contexts. Front Pharmacol 2019; 10:416. [PMID: 31057411 PMCID: PMC6482345 DOI: 10.3389/fphar.2019.00416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
|
35
|
Zineh I. Quantitative Systems Pharmacology: A Regulatory Perspective on Translation. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:336-339. [PMID: 30924594 PMCID: PMC6618141 DOI: 10.1002/psp4.12403] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/07/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Issam Zineh
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|