1
|
Ruiz L, Jaramillo S, Calvo A, Torrente MA, Tassies D, Reverter JC, Blasi A, Troconiz I. Dynamics of thrombin generation: Filling the gap between the system pharmacology theory and clinical practice in clinical pharmacology and therapeutics. Pharmacol Res Perspect 2025; 13:e70014. [PMID: 39739766 DOI: 10.1002/prp2.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 08/27/2024] [Accepted: 09/01/2024] [Indexed: 01/02/2025] Open
Abstract
Mathematical models of thrombin generation (TG) that have been developed are based on a systems biology approach. Although this approach provides important information about the coagulation system, its clinical applicability is limited by its complexity and number of input variables required. The aim of this study was to develop a semimechanistic model able to describe TG in trauma and control patients. A dataset containing longitudinal data of TG assays and coagulation factors from 40 trauma patients and 20 control patients was used for model building. The model considered three fundamental processes: the degradation of tissue factor (TF) through a first-order process, the activation of factor II by the TF through a first-order process, and the degradation of thrombin through a first-order process. Model fitting was performed using a nonlinear mixed-effects approach. The condition of the patient (trauma and control) and coagulation factors were modelled as covariates. Model building demonstrated the presence of two additional processes that improved the predictive capacity of the model: the activation of factor II by TF governed by a second-order constant and, a mechanism of factor II activation by TF characterized by a 7-compartment transit chain governed by a second-order constant. In the covariate model only the inclusion of patient condition was significant. Model evaluation demonstrated excellent performance in describing the temporal pattern of TG in trauma and control patients. Thrombin generation can be adequately modelled using a semimechanistic approach. Its application in practice could help to better assess the risk of hemorrhage and/or thrombosis in different settings.
Collapse
|
2
|
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: 1.5] [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
|
3
|
Falkenhagen U, Knöchel J, Kloft C, Huisinga W. Deriving mechanism-based pharmacodynamic models by reducing quantitative systems pharmacology models: An application to warfarin. CPT Pharmacometrics Syst Pharmacol 2023; 12:432-443. [PMID: 36866520 PMCID: PMC10088086 DOI: 10.1002/psp4.12903] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/18/2022] [Accepted: 11/29/2022] [Indexed: 03/04/2023] Open
Abstract
Quantitative systems pharmacology (QSP) models integrate comprehensive qualitative and quantitative knowledge about pharmacologically relevant processes. We previously proposed a first approach to leverage the knowledge in QSP models to derive simpler, mechanism-based pharmacodynamic (PD) models. Their complexity, however, is typically still too large to be used in the population analysis of clinical data. Here, we extend the approach beyond state reduction to also include the simplification of reaction rates, elimination of reactions, and analytic solutions. We additionally ensure that the reduced model maintains a prespecified approximation quality not only for a reference individual but also for a diverse virtual population. We illustrate the extended approach for the warfarin effect on blood coagulation. Using the model-reduction approach, we derive a novel small-scale warfarin/international normalized ratio model and demonstrate its suitability for biomarker identification. Due to the systematic nature of the approach in comparison with empirical model building, the proposed model-reduction algorithm provides an improved rationale to build PD models also from QSP models in other applications.
Collapse
Affiliation(s)
- Undine Falkenhagen
- Institute of Mathematics, University of Potsdam, Potsdam, Germany.,Graduate Research Training Program PharMetrX: Pharmacometrics & Computational Disease Modelling, Freie Universität Berlin and University of Potsdam, Potsdam, Germany
| | - Jane Knöchel
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
| | - Charlotte Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Wilhelm Huisinga
- Institute of Mathematics, University of Potsdam, Potsdam, Germany
| |
Collapse
|
4
|
Sher A, Niederer SA, Mirams GR, Kirpichnikova A, Allen R, Pathmanathan P, Gavaghan DJ, van der Graaf PH, Noble D. A Quantitative Systems Pharmacology Perspective on the Importance of Parameter Identifiability. Bull Math Biol 2022; 84:39. [PMID: 35132487 PMCID: PMC8821410 DOI: 10.1007/s11538-021-00982-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 11/30/2021] [Indexed: 12/31/2022]
Abstract
There is an inherent tension in Quantitative Systems Pharmacology (QSP) between the need to incorporate mathematical descriptions of complex physiology and drug targets with the necessity of developing robust, predictive and well-constrained models. In addition to this, there is no “gold standard” for model development and assessment in QSP. Moreover, there can be confusion over terminology such as model and parameter identifiability; complex and simple models; virtual populations; and other concepts, which leads to potential miscommunication and misapplication of methodologies within modeling communities, both the QSP community and related disciplines. This perspective article highlights the pros and cons of using simple (often identifiable) vs. complex (more physiologically detailed but often non-identifiable) models, as well as aspects of parameter identifiability, sensitivity and inference methodologies for model development and analysis. The paper distills the central themes of the issue of identifiability and optimal model size and discusses open challenges.
Collapse
Affiliation(s)
- Anna Sher
- Pfizer Worldwide Research, Development and Medical, Massachusetts, USA.
| | | | - Gary R Mirams
- Centre for Mathematical Medicine and Biology, Mathematical Sciences, University of Nottingham, Nottingham, UK
| | | | - Richard Allen
- Pfizer Worldwide Research, Development and Medical, Massachusetts, USA
| | - Pras Pathmanathan
- Center for Devices and Radiological Health, U.S. Food and Drug Administration, Maryland, USA
| | - David J Gavaghan
- Department of Computer Science, University of Oxford, Oxford, UK
| | | | - Denis Noble
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
5
|
Derbalah A, Al-Sallami HS, Duffull SB. Reduction of quantitative systems pharmacology models using artificial neural networks. J Pharmacokinet Pharmacodyn 2021; 48:509-523. [PMID: 33651241 DOI: 10.1007/s10928-021-09742-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 02/11/2021] [Indexed: 12/16/2022]
Abstract
Quantitative systems pharmacology models are often highly complex and not amenable to further simulation and/or estimation analyses. Model-order reduction can be used to derive a mechanistically sound yet simpler model of the desired input-output relationship. In this study, we explore the use of artificial neural networks for approximating an input-output relationship within highly dimensional systems models. We illustrate this approach using a model of blood coagulation. The model consists of two components linked together through a highly dimensional discontinuous interface, which creates a difficulty for model reduction techniques. The proposed approach enables the development of an efficient approximation to complex models with the desired level of accuracy. The technique is applicable to a wide variety of models and provides substantial speed boost for use of such models in simulation and control purposes.
Collapse
Affiliation(s)
- Abdallah Derbalah
- School of Pharmacy, University of Otago, 18 Frederick St, North Dunedin, Dunedin, 9016, New Zealand.
| | - Hesham S Al-Sallami
- School of Pharmacy, University of Otago, 18 Frederick St, North Dunedin, Dunedin, 9016, New Zealand
| | - Stephen B Duffull
- School of Pharmacy, University of Otago, 18 Frederick St, North Dunedin, Dunedin, 9016, New Zealand
| |
Collapse
|
6
|
Derbalah A, Al‐Sallami H, Hasegawa C, Gulati A, Duffull SB. A framework for simplification of quantitative systems pharmacology models in clinical pharmacology. Br J Clin Pharmacol 2020; 88:1430-1440. [DOI: 10.1111/bcp.14451] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/13/2020] [Accepted: 06/22/2020] [Indexed: 12/12/2022] Open
Affiliation(s)
| | | | | | - Abhishek Gulati
- Clinical Pharmacology and Exploratory Development, Astellas Pharma Global Development Northbrook Illinois USA
| | | |
Collapse
|
7
|
Automated proper lumping for simplification of linear physiologically based pharmacokinetic systems. J Pharmacokinet Pharmacodyn 2019; 46:361-370. [PMID: 31227954 PMCID: PMC6656793 DOI: 10.1007/s10928-019-09644-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 06/12/2019] [Indexed: 01/24/2023]
Abstract
Physiologically based pharmacokinetic (PBPK) models are an important type of systems model used commonly in drug development before commencement of first-in-human studies. Due to structural complexity, these models are not easily utilised for future data-driven population pharmacokinetic (PK) analyses that require simpler models. In the current study we aimed to explore and automate methods of simplifying PBPK models using a proper lumping technique. A linear 17-state PBPK model for fentanyl was identified from the literature. Four methods were developed to search the optimal lumped model, including full enumeration (the reference method), non-adaptive random search (NARS), scree plot plus NARS, and simulated annealing (SA). For exploratory purposes, it was required that the total area under the fentanyl arterial concentration–time curve (AUC) between the lumped and original models differ by 0.002% at maximum. In full enumeration, a 4-state lumped model satisfying the exploratory criterion was found. In NARS, a lumped model with the same number of lumped states was found, requiring a large number of random samples. The scree plot provided a starting lumped model to NARS and the search completed within a short time. In SA, a 4-state lumped model was consistently delivered. In simplify an existing linear fentanyl PBPK model, SA was found to be robust and the most efficient and may be suitable for general application to other larger-scale linear systems. Ultimately, simplified PBPK systems with fundamental mechanisms may be readily used for data-driven PK analyses.
Collapse
|
8
|
Hasegawa C, Duffull SB. Automated Scale Reduction of Nonlinear QSP Models With an Illustrative Application to a Bone Biology System. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2018; 7:562-572. [PMID: 30043496 PMCID: PMC6157701 DOI: 10.1002/psp4.12324] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Integrating quantitative systems pharmacology (QSP) into pharmacokinetics/pharmacodynamics (PKPD) has resulted in models that are highly complex and often not amenable to further exploration via estimation or design. Because QSP models are usually depicted using nonlinear differential equations it is not straightforward to apply some model reduction techniques, such as proper lumping. In this study, we explore the combined use of linearization and proper lumping as a general method to simplification of a nonlinear QSP model. We illustrate this with a bone biology model and the reduced model was then applied to describe bone mineral density (BMD) changes due to denosumab dosing. The methodologies used in this study can be applied to other multiscale models for developing a mechanism-based structural model for future analyses.
Collapse
Affiliation(s)
- Chihiro Hasegawa
- School of Pharmacy, University of Otago, Dunedin, New Zealand.,Translational Medicine Center, Ono Pharmaceutical Co., Ltd., Osaka, Japan
| | | |
Collapse
|
9
|
Hecht M, Veigure R, Couchman L, S Barker CI, Standing JF, Takkis K, Evard H, Johnston A, Herodes K, Leito I, Kipper K. Utilization of data below the analytical limit of quantitation in pharmacokinetic analysis and modeling: promoting interdisciplinary debate. Bioanalysis 2018; 10:1229-1248. [PMID: 30033744 DOI: 10.4155/bio-2018-0078] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Traditionally, bioanalytical laboratories do not report actual concentrations for samples with results below the LOQ (BLQ) in pharmacokinetic studies. BLQ values are outside the method calibration range established during validation and no data are available to support the reliability of these values. However, ignoring BLQ data can contribute to bias and imprecision in model-based pharmacokinetic analyses. From this perspective, routine use of BLQ data would be advantageous. We would like to initiate an interdisciplinary debate on this important topic by summarizing the current concepts and use of BLQ data by regulators, pharmacometricians and bioanalysts. Through introducing the limit of detection and evaluating its variability, BLQ data could be released and utilized appropriately for pharmacokinetic research.
Collapse
Affiliation(s)
- Max Hecht
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Rūta Veigure
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
| | - Lewis Couchman
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Charlotte I S Barker
- Paediatric Infectious Diseases Research Group, Institute for Infection & Immunity, St George's University of London, London, SW17 0RE, UK
- Inflammation, Infection & Rheumatology Section, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
- Paediatric Infectious Diseases Unit, St George's University Hospitals NHS Foundation Trust, London, SW17 0RE, UK
| | - Joseph F Standing
- Paediatric Infectious Diseases Research Group, Institute for Infection & Immunity, St George's University of London, London, SW17 0RE, UK
- Inflammation, Infection & Rheumatology Section, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Kalev Takkis
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| | - Hanno Evard
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
| | - Atholl Johnston
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
- Clinical Pharmacology, Barts & The London School of Medicine & Dentistry, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Koit Herodes
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
| | - Ivo Leito
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
| | - Karin Kipper
- Chair of Analytical Chemistry, Institute of Chemistry, University of Tartu, 14a Ravila Street, 50411 Tartu, Estonia
- Analytical Services International, St George's University of London, Cranmer Terrace, London, SW17 0RE, UK
| |
Collapse
|
10
|
Snowden TJ, van der Graaf PH, Tindall MJ. Model reduction in mathematical pharmacology : Integration, reduction and linking of PBPK and systems biology models. J Pharmacokinet Pharmacodyn 2018; 45:537-555. [PMID: 29582349 PMCID: PMC6061126 DOI: 10.1007/s10928-018-9584-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 03/14/2018] [Indexed: 11/27/2022]
Abstract
In this paper we present a framework for the reduction and linking of physiologically based pharmacokinetic (PBPK) models with models of systems biology to describe the effects of drug administration across multiple scales. To address the issue of model complexity, we propose the reduction of each type of model separately prior to being linked. We highlight the use of balanced truncation in reducing the linear components of PBPK models, whilst proper lumping is shown to be efficient in reducing typically nonlinear systems biology type models. The overall methodology is demonstrated via two example systems; a model of bacterial chemotactic signalling in Escherichia coli and a model of extracellular regulatory kinase activation mediated via the extracellular growth factor and nerve growth factor receptor pathways. Each system is tested under the simulated administration of three hypothetical compounds; a strong base, a weak base, and an acid, mirroring the parameterisation of pindolol, midazolam, and thiopental, respectively. Our method can produce up to an 80% decrease in simulation time, allowing substantial speed-up for computationally intensive applications including parameter fitting or agent based modelling. The approach provides a straightforward means to construct simplified Quantitative Systems Pharmacology models that still provide significant insight into the mechanisms of drug action. Such a framework can potentially bridge pre-clinical and clinical modelling - providing an intermediate level of model granularity between classical, empirical approaches and mechanistic systems describing the molecular scale.
Collapse
Affiliation(s)
- Thomas J. Snowden
- Department of Mathematics and Statistics, University of Reading, Reading, RG6 6AX UK
- Certara QSP, University of Kent Innovation Centre, Canterbury, CT2 7FG UK
| | - Piet H. van der Graaf
- Certara QSP, University of Kent Innovation Centre, Canterbury, CT2 7FG UK
- Leiden Academic Centre for Drug Research, Universiteit Leiden, 2333 CC Leiden, The Netherlands
| | - Marcus J. Tindall
- Department of Mathematics and Statistics, University of Reading, Reading, RG6 6AX UK
- The Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6UR UK
| |
Collapse
|
11
|
Understanding and reducing complex systems pharmacology models based on a novel input-response index. J Pharmacokinet Pharmacodyn 2017; 45:139-157. [PMID: 29243176 DOI: 10.1007/s10928-017-9561-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 12/06/2017] [Indexed: 11/27/2022]
Abstract
A growing understanding of complex processes in biology has led to large-scale mechanistic models of pharmacologically relevant processes. These models are increasingly used to study the response of the system to a given input or stimulus, e.g., after drug administration. Understanding the input-response relationship, however, is often a challenging task due to the complexity of the interactions between its constituents as well as the size of the models. An approach that quantifies the importance of the different constituents for a given input-output relationship and allows to reduce the dynamics to its essential features is therefore highly desirable. In this article, we present a novel state- and time-dependent quantity called the input-response index that quantifies the importance of state variables for a given input-response relationship at a particular time. It is based on the concept of time-bounded controllability and observability, and defined with respect to a reference dynamics. In application to the brown snake venom-fibrinogen (Fg) network, the input-response indices give insight into the coordinated action of specific coagulation factors and about those factors that contribute only little to the response. We demonstrate how the indices can be used to reduce large-scale models in a two-step procedure: (i) elimination of states whose dynamics have only minor impact on the input-response relationship, and (ii) proper lumping of the remaining (lower order) model. In application to the brown snake venom-fibrinogen network, this resulted in a reduction from 62 to 8 state variables in the first step, and a further reduction to 5 state variables in the second step. We further illustrate that the sequence, in which a recursive algorithm eliminates and/or lumps state variables, has an impact on the final reduced model. The input-response indices are particularly suited to determine an informed sequence, since they are based on the dynamics of the original system. In summary, the novel measure of importance provides a powerful tool for analysing the complex dynamics of large-scale systems and a means for very efficient model order reduction of nonlinear systems.
Collapse
|
12
|
Hasegawa C, Duffull SB. Selection and Qualification of Simplified QSP Models When Using Model Order Reduction Techniques. AAPS JOURNAL 2017; 20:2. [PMID: 29181592 DOI: 10.1208/s12248-017-0170-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 11/05/2017] [Indexed: 01/04/2023]
Abstract
Quantitative systems pharmacology (QSP) models are increasingly used in drug development to provide a deep understanding of the mechanism of action of drugs and to identify appropriate disease targets. Such models are, however, not suitable for estimation purposes due to their high dimensionality. Based on any desired and specific input-output relationship, the system may be reduced to a model with fewer states and parameters. However, any simplification process will be a trade-off between model performance and complexity. In this study, we develop a weighted composite criterion which brings together the opposing indices of performance and dimensionality. The weighting factor can be determined by qualification of the simplified model based on a visual predictive check (VPC) using the precision of each parameter. The weighted criterion and model qualification techniques were illustrated with three examples: a simple compartmental pharmacokinetic model, a physiologically based pharmacokinetic (PBPK) example, and a semimechanistic model for bone mineral density. When considering the PBPK example, this automated search identified the same reduced model which had been detected in a previous report, as well as a simpler model which had not been previously identified. The simpler bone mineral density model provided an adequate description of the response even after 1 year from the initiation of treatment. The proposed criterion together with a VPC provides a natural way for model order reduction that can be fully automated and applied to multiscale models.
Collapse
Affiliation(s)
- Chihiro Hasegawa
- School of Pharmacy, University of Otago, Dunedin, New Zealand. .,Translational Medicine Center, Ono Pharmaceutical Co., Ltd., Osaka, Japan.
| | | |
Collapse
|
13
|
Ribba B, Grimm HP, Agoram B, Davies MR, Gadkar K, Niederer S, van Riel N, Timmis J, van der Graaf PH. Methodologies for Quantitative Systems Pharmacology (QSP) Models: Design and Estimation. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:496-498. [PMID: 28585415 PMCID: PMC5572127 DOI: 10.1002/psp4.12206] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 04/27/2017] [Accepted: 05/09/2017] [Indexed: 02/05/2023]
Abstract
With the increased interest in the application of quantitative systems pharmacology (QSP) models within medicine research and development, there is an increasing need to formalize model development and verification aspects. In February 2016, a workshop was held at Roche Pharma Research and Early Development to focus discussions on two critical methodological aspects of QSP model development: optimal structural granularity and parameter estimation. We here report in a perspective article a summary of presentations and discussions.
Collapse
Affiliation(s)
- B Ribba
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Basel, Switzerland
| | - H P Grimm
- Roche Pharmaceutical Research & Early Development, Roche Innovation Center Basel, Switzerland
| | - B Agoram
- MedImmune, Mountain View, California, USA
| | - M R Davies
- QT Informatics Limited, Macclesfield, UK
| | - K Gadkar
- Genentech, South San Francisco, California, USA
| | - S Niederer
- King's College London, Division of Imaging Sciences and Biomedical Engineering, London, UK
| | - N van Riel
- Eindhoven University of Technology, Department of Biomedical Engineering, Eindhoven, The Netherlands.,University of Amsterdam, Academic Medical Center, Amsterdam, The Netherlands
| | - J Timmis
- SimOmics Ltd, Department of Electronics, University of York, York, UK
| | - P H van der Graaf
- Leiden Academic Centre for Drug Research (LACDR), Leiden, The Netherlands.,Certara QSP, Canterbury, UK
| |
Collapse
|
14
|
Snowden TJ, van der Graaf PH, Tindall MJ. Methods of Model Reduction for Large-Scale Biological Systems: A Survey of Current Methods and Trends. Bull Math Biol 2017; 79:1449-1486. [PMID: 28656491 PMCID: PMC5498684 DOI: 10.1007/s11538-017-0277-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 03/30/2017] [Indexed: 01/31/2023]
Abstract
Complex models of biochemical reaction systems have become increasingly common in the systems biology literature. The complexity of such models can present a number of obstacles for their practical use, often making problems difficult to intuit or computationally intractable. Methods of model reduction can be employed to alleviate the issue of complexity by seeking to eliminate those portions of a reaction network that have little or no effect upon the outcomes of interest, hence yielding simplified systems that retain an accurate predictive capacity. This review paper seeks to provide a brief overview of a range of such methods and their application in the context of biochemical reaction network models. To achieve this, we provide a brief mathematical account of the main methods including timescale exploitation approaches, reduction via sensitivity analysis, optimisation methods, lumping, and singular value decomposition-based approaches. Methods are reviewed in the context of large-scale systems biology type models, and future areas of research are briefly discussed.
Collapse
Affiliation(s)
- Thomas J Snowden
- Department of Mathematics and Statistics, University of Reading, Reading, RG6 6AX, UK.,Certara QSP, University of Kent Innovation Centre, Canterbury, CT2 7FG, UK
| | - Piet H van der Graaf
- Certara QSP, University of Kent Innovation Centre, Canterbury, CT2 7FG, UK.,Leiden Academic Centre for Drug Research, Universiteit Leiden, Leiden, 2333 CC, Netherlands
| | - Marcus J Tindall
- Department of Mathematics and Statistics, University of Reading, Reading, RG6 6AX, UK. .,The Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6AX, UK.
| |
Collapse
|
15
|
Hasegawa C, Duffull SB. Exploring inductive linearization for pharmacokinetic–pharmacodynamic systems of nonlinear ordinary differential equations. J Pharmacokinet Pharmacodyn 2017; 45:35-47. [DOI: 10.1007/s10928-017-9527-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Accepted: 05/22/2017] [Indexed: 11/28/2022]
|
16
|
Snowden TJ, van der Graaf PH, Tindall MJ. A combined model reduction algorithm for controlled biochemical systems. BMC SYSTEMS BIOLOGY 2017; 11:17. [PMID: 28193218 PMCID: PMC5307760 DOI: 10.1186/s12918-017-0397-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2016] [Accepted: 01/18/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Systems Biology continues to produce increasingly large models of complex biochemical reaction networks. In applications requiring, for example, parameter estimation, the use of agent-based modelling approaches, or real-time simulation, this growing model complexity can present a significant hurdle. Often, however, not all portions of a model are of equal interest in a given setting. In such situations methods of model reduction offer one possible approach for addressing the issue of complexity by seeking to eliminate those portions of a pathway that can be shown to have the least effect upon the properties of interest. METHODS In this paper a model reduction algorithm bringing together the complementary aspects of proper lumping and empirical balanced truncation is presented. Additional contributions include the development of a criterion for the selection of state-variable elimination via conservation analysis and use of an 'averaged' lumping inverse. This combined algorithm is highly automatable and of particular applicability in the context of 'controlled' biochemical networks. RESULTS The algorithm is demonstrated here via application to two examples; an 11 dimensional model of bacterial chemotaxis in Escherichia coli and a 99 dimensional model of extracellular regulatory kinase activation (ERK) mediated via the epidermal growth factor (EGF) and nerve growth factor (NGF) receptor pathways. In the case of the chemotaxis model the algorithm was able to reduce the model to 2 state-variables producing a maximal relative error between the dynamics of the original and reduced models of only 2.8% whilst yielding a 26 fold speed up in simulation time. For the ERK activation model the algorithm was able to reduce the system to 7 state-variables, incurring a maximal relative error of 4.8%, and producing an approximately 10 fold speed up in the rate of simulation. Indices of controllability and observability are additionally developed and demonstrated throughout the paper. These provide insight into the relative importance of individual reactants in mediating a biochemical system's input-output response even for highly complex networks. CONCLUSIONS Through application, this paper demonstrates that combined model reduction methods can produce a significant simplification of complex Systems Biology models whilst retaining a high degree of predictive accuracy. In particular, it is shown that by combining the methods of proper lumping and empirical balanced truncation it is often possible to produce more accurate reductions than can be obtained by the use of either method in isolation.
Collapse
Affiliation(s)
- Thomas J Snowden
- Department of Mathematics and Statistics, University of Reading, Reading, RG6 6AX, UK.,Certara QSP, University of Kent Innovation Centre, Canterbury, CT2 7FG, UK
| | - Piet H van der Graaf
- Certara QSP, University of Kent Innovation Centre, Canterbury, CT2 7FG, UK.,Leiden Academic Centre for Drug Research, Universiteit Leiden, Leiden, NL-2333 CC, Netherlands
| | - Marcus J Tindall
- Department of Mathematics and Statistics, University of Reading, Reading, RG6 6AX, UK. .,The Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6AX, UK.
| |
Collapse
|
17
|
Duffull SB. A Philosophical Framework for Integrating Systems Pharmacology Models Into Pharmacometrics. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:649-655. [PMID: 27863137 PMCID: PMC5192992 DOI: 10.1002/psp4.12148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/07/2016] [Indexed: 12/14/2022]
Abstract
The framework for systems pharmacology style models does not naturally sit with the usual scientific dogma of parsimony and falsifiability based on deductive reasoning. This does not invalidate the importance or need for overarching models based on pharmacology to describe and understand complicated biological systems. However, it does require some consideration on how systems pharmacology fits into the overall scientific approach.
Collapse
Affiliation(s)
- S B Duffull
- Otago Pharmacometrics Group, School of Pharmacy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
Upton RN, Foster DJR, Abuhelwa AY. An introduction to physiologically-based pharmacokinetic models. Paediatr Anaesth 2016; 26:1036-1046. [PMID: 27550716 DOI: 10.1111/pan.12995] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/24/2016] [Indexed: 11/30/2022]
Abstract
Physiologically-based pharmacokinetic (PBPK) models represent drug kinetics in one or more 'real' organs (and hence require submodels of organs/tissues) and they describe 'whole-body' kinetics by joining together submodels with drug transport by blood flow as dictated by anatomy. They attempt to reproduce 'measureable' physiological and/or pharmacokinetic processes rather than more abstract rate constants and volumes. PBPK models may be built using a 'bottom-up' approach, where parameters are chosen from first principles, literature, or in vitro data as opposed to a 'top-down' approach, where all parameters are estimated from data. The basic principles of PBPK models are described, focusing on the equations for three individual organs: a single flow-limited compartment describing distribution only, a membrane-limited compartment describing distribution, and a single flow-limited compartment with elimination. These organ models are linked to make a basic three-compartment physiological model of the whole body. PBPK models are particularly suited to scaling kinetics across body size (e.g., adult to neonate) and species (e.g., animal to first-in-man) as physiology and pharmacology can be represented by independent parameters. Maturation models can be incorporated as for compartmental models. PBPK models are now available in commercial software packages, and are perhaps now more accessible than ever. Alternatively, even complex PBPK models can be represented in generic differential equation-solving software using the simple principles described here. The relative ease of constructing the code for PBPK models belies the most difficult aspect of their implementation-collecting, collating, and justifying the data used to parameterize the model.
Collapse
Affiliation(s)
- Richard N Upton
- Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia.
| | - David J R Foster
- Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Ahmad Y Abuhelwa
- Australian Centre for Pharmacometrics and Sansom Institute, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
19
|
Ruiz-Cerdá ML, Irurzun-Arana I, González-Garcia I, Hu C, Zhou H, Vermeulen A, Trocóniz IF, Gómez-Mantilla JD. Towards patient stratification and treatment in the autoimmune disease lupus erythematosus using a systems pharmacology approach. Eur J Pharm Sci 2016; 94:46-58. [DOI: 10.1016/j.ejps.2016.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 04/07/2016] [Accepted: 04/07/2016] [Indexed: 01/28/2023]
|
20
|
Wendling T, Tsamandouras N, Dumitras S, Pigeolet E, Ogungbenro K, Aarons L. Reduction of a Whole-Body Physiologically Based Pharmacokinetic Model to Stabilise the Bayesian Analysis of Clinical Data. AAPS JOURNAL 2015; 18:196-209. [PMID: 26538125 DOI: 10.1208/s12248-015-9840-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/15/2015] [Indexed: 12/27/2022]
Abstract
Whole-body physiologically based pharmacokinetic (PBPK) models are increasingly used in drug development for their ability to predict drug concentrations in clinically relevant tissues and to extrapolate across species, experimental conditions and sub-populations. A whole-body PBPK model can be fitted to clinical data using a Bayesian population approach. However, the analysis might be time consuming and numerically unstable if prior information on the model parameters is too vague given the complexity of the system. We suggest an approach where (i) a whole-body PBPK model is formally reduced using a Bayesian proper lumping method to retain the mechanistic interpretation of the system and account for parameter uncertainty, (ii) the simplified model is fitted to clinical data using Markov Chain Monte Carlo techniques and (iii) the optimised reduced PBPK model is used for extrapolation. A previously developed 16-compartment whole-body PBPK model for mavoglurant was reduced to 7 compartments while preserving plasma concentration-time profiles (median and variance) and giving emphasis to the brain (target site) and the liver (elimination site). The reduced model was numerically more stable than the whole-body model for the Bayesian analysis of mavoglurant pharmacokinetic data in healthy adult volunteers. Finally, the reduced yet mechanistic model could easily be scaled from adults to children and predict mavoglurant pharmacokinetics in children aged from 3 to 11 years with similar performance compared with the whole-body model. This study is a first example of the practicality of formal reduction of complex mechanistic models for Bayesian inference in drug development.
Collapse
Affiliation(s)
- Thierry Wendling
- Manchester Pharmacy School, The University of Manchester, Manchester, UK. .,Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, Basel, Switzerland.
| | | | - Swati Dumitras
- Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | | | - Kayode Ogungbenro
- Manchester Pharmacy School, The University of Manchester, Manchester, UK
| | - Leon Aarons
- Manchester Pharmacy School, The University of Manchester, Manchester, UK
| |
Collapse
|
21
|
Pan S, Korell J, Stamp LK, Duffull SB. Simplification of a pharmacokinetic model for red blood cell methotrexate disposition. Eur J Clin Pharmacol 2015; 71:1509-16. [DOI: 10.1007/s00228-015-1951-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 09/16/2015] [Indexed: 11/29/2022]
|
22
|
Biliouris K, Lavielle M, Trame MN. MatVPC: A User-Friendly MATLAB-Based Tool for the Simulation and Evaluation of Systems Pharmacology Models. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015; 4:547-57. [PMID: 26451334 PMCID: PMC4592534 DOI: 10.1002/psp4.12011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/10/2015] [Indexed: 12/13/2022]
Abstract
Quantitative systems pharmacology (QSP) models are progressively entering the arena of contemporary pharmacology. The efficient implementation and evaluation of complex QSP models necessitates the development of flexible computational tools that are built into QSP mainstream software. To this end, we present MatVPC, a versatile MATLAB-based tool that accommodates QSP models of any complexity level. MatVPC executes Monte Carlo simulations as well as automatic construction of visual predictive checks (VPCs) and quantified VPCs (QVPCs).
Collapse
Affiliation(s)
- K Biliouris
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida Orlando, Florida, USA
| | | | - M N Trame
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, University of Florida Orlando, Florida, USA
| |
Collapse
|