51
|
Kundu MG. Methods and Applications of Sample Size Calculation and Recalculation in Clinical Trials. J Biopharm Stat 2021. [DOI: 10.1080/10543406.2021.1992247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
52
|
Toussaint B, Hillaireau H, Jaccoulet E, Cailleau C, Legrand P, Ambroise Y, Fattal E. Interspecies comparison of plasma metabolism and sample stabilization for quantitative bioanalyses: Application to (R)-CE3F4 in preclinical development, including metabolite identification by high-resolution mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1183:122943. [PMID: 34666890 DOI: 10.1016/j.jchromb.2021.122943] [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: 04/15/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 01/01/2023]
Abstract
The CE3F4 is an inhibitor of the type 1 exchange protein directly activated by cAMP (EPAC1), which is involved in numerous signaling pathways. The inhibition of EPAC1 shows promising results in vitro and in vivo in different cardiac pathological situations like hypertrophic signaling, contributing to heart failure, or arrhythmia. An HPLC-UV method with a simple and fast sample treatment allowed the quantification of (R)-CE3F4. Sample treatment consisted of simple protein precipitation with 50 µL of ethanol and 150 µL of acetonitrile for a 50 µL biological sample. Two wavelengths were used according to the origin of plasma (220 or 250 nm for human samples and 250 nm for murine samples). Accuracy profile was evaluated for both wavelengths, and the method was in agreement with the criteria given by the EMA in the guideline for bioanalytical method validation for human and mouse plasma samples. The run time was 12 min allowing the detection of the (R)-CE3F4 and a metabolite. This study further permitted understanding the behavior of CE3F4 in plasma by highlighting an important difference between humans and rodents on plasma metabolism and may impact future in vivo studies related to this molecule and translation of results between animal models and humans. Using paraoxon as a metabolism inhibitor was crucial for the stabilization of (R)-CE3F4 in murine samples. HPLC-UV and HPLC-MS/MS studies were conducted to confirm metabolite structure and consequently, the main metabolic pathway in murine plasma.
Collapse
Affiliation(s)
- Balthazar Toussaint
- Université Paris-Saclay, CNRS, Institut Galien Paris Sud, 92296 Châtenay-Malabry, France; Département de Recherche et Développement Pharmaceutique, Agence Générale des Équipements et Produits de Santé (AGEPS), Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
| | - Hervé Hillaireau
- Université Paris-Saclay, CNRS, Institut Galien Paris Sud, 92296 Châtenay-Malabry, France
| | - Emmanuel Jaccoulet
- Université Paris-Saclay, CNRS, Institut Galien Paris Sud, 92296 Châtenay-Malabry, France; Hôpital européen Georges Pompidou (HEGP), Service Pharmacie (AP-HP), Paris, France
| | - Catherine Cailleau
- Université Paris-Saclay, CNRS, Institut Galien Paris Sud, 92296 Châtenay-Malabry, France
| | - Pauline Legrand
- Département de Recherche et Développement Pharmaceutique, Agence Générale des Équipements et Produits de Santé (AGEPS), Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France; Université de Paris, Faculté de sciences pharmaceutiques et biologiques, Unité de Technologies Chimiques et Biologiques pour la Santé (UTCBS), CNRS UMR8258, Inserm U1022, Paris, France
| | - Yves Ambroise
- Université Paris-Saclay, CEA, Institut des Sciences du Vivant Frederic Joliot, 91191 Gif-sur-Yvette, France
| | - Elias Fattal
- Université Paris-Saclay, CNRS, Institut Galien Paris Sud, 92296 Châtenay-Malabry, France.
| |
Collapse
|
53
|
Bauer RJ, Hooker AC, Mentre F. Tutorial for $DESIGN in NONMEM: Clinical trial evaluation and optimization. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1452-1465. [PMID: 34559958 PMCID: PMC8674001 DOI: 10.1002/psp4.12713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 12/02/2022]
Abstract
This NONMEM tutorial shows how to evaluate and optimize clinical trial designs, using algorithms developed in design software, such as PopED and PFIM 4.0. Parameter precision and model parameter estimability is obtained by assessing the Fisher Information Matrix (FIM), providing expected model parameter uncertainty. Model parameter identifiability may be uncovered by very large standard errors or inability to invert an FIM. Because evaluation of FIM is more efficient than clinical trial simulation, more designs can be investigated, and the design of a clinical trial can be optimized. This tutorial provides simple and complex pharmacokinetic/pharmacodynamic examples on obtaining optimal sample times, doses, or best division of subjects among design groups. Robust design techniques accounting for likely variability among subjects are also shown. A design evaluator and optimizer within NONMEM allows any control stream first developed for trial design exploration to be subsequently used for estimation of parameters of simulated or clinical data, without transferring the model to another software. Conversely, a model developed in NONMEM could be used for design optimization. In addition, the $DESIGN feature can be used on any model file and dataset combination to retrospectively evaluate the model parameter uncertainty one would expect given that the model generated the data, particularly if outliers of the actual data prevent a reasonable assessment of the variance‐covariance. The NONMEM trial design feature is suitable for standard continuous data, whereas more elaborate trial designs or with noncontinuous data‐types can still be accomplished in optimal design dedicated software like PopED and PFIM.
Collapse
Affiliation(s)
- Robert J Bauer
- Pharmacometrics, R&D, ICON Clinical Research, LLC, Gaithersburg, Maryland, USA
| | | | | |
Collapse
|
54
|
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
|
55
|
Collignon O, Burman C, Posch M, Schiel A. Collaborative Platform Trials to Fight COVID-19: Methodological and Regulatory Considerations for a Better Societal Outcome. Clin Pharmacol Ther 2021; 110:311-320. [PMID: 33506495 PMCID: PMC8014457 DOI: 10.1002/cpt.2183] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/19/2021] [Indexed: 12/19/2022]
Abstract
For the development of coronavirus disease 2019 (COVID-19) drugs during the ongoing pandemic, speed is of essence whereas quality of evidence is of paramount importance. Although thousands of COVID-19 trials were rapidly started, many are unlikely to provide robust statistical evidence and meet regulatory standards (e.g., because of lack of randomization or insufficient power). This has led to an inefficient use of time and resources. With more coordination, the sheer number of patients in these trials might have generated convincing data for several investigational treatments. Collaborative platform trials, comparing several drugs to a shared control arm, are an attractive solution. Those trials can utilize a variety of adaptive design features in order to accelerate the finding of life-saving treatments. In this paper, we discuss several possible designs, illustrate them via simulations, and also discuss challenges, such as the heterogeneity of the target population, time-varying standard of care, and the potentially high number of false hypothesis rejections in phase II and phase III trials. We provide corresponding regulatory perspectives on approval and reimbursement, and note that the optimal design of a platform trial will differ with our societal objective and by stakeholder. Hasty approvals may delay the development of better alternatives, whereas searching relentlessly for the single most efficacious treatment may indirectly diminish the number of lives saved as time is lost. We point out the need for incentivizing developers to participate in collaborative evidence-generation initiatives when a positive return on investment is not met.
Collapse
Affiliation(s)
| | - Carl‐Fredrik Burman
- Statistical Innovation, Data Science, and Artificial IntelligenceAstraZeneca R&DGothenburgSweden
| | - Martin Posch
- Section for Medical StatisticsCenter for Medical Statistics, Informatics, and Intelligent SystemsMedical University of ViennaViennaAustria
| | | |
Collapse
|
56
|
Ryeznik Y, Sverdlov O, Svensson EM, Montepiedra G, Hooker AC, Wong WK. Pharmacometrics meets statistics-A synergy for modern drug development. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:1134-1149. [PMID: 34318621 PMCID: PMC8520751 DOI: 10.1002/psp4.12696] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 05/17/2021] [Accepted: 07/02/2021] [Indexed: 01/20/2023]
Abstract
Modern drug development problems are very complex and require integration of various scientific fields. Traditionally, statistical methods have been the primary tool for design and analysis of clinical trials. Increasingly, pharmacometric approaches using physiology-based drug and disease models are applied in this context. In this paper, we show that statistics and pharmacometrics have more in common than what keeps them apart, and collectively, the synergy from these two quantitative disciplines can provide greater advances in clinical research and development, resulting in novel and more effective medicines to patients with medical need.
Collapse
Affiliation(s)
- Yevgen Ryeznik
- BioPharma Early Biometrics and Statistical Innovation, Data Science & AI, R&D Biopharmaceuticals, AstraZeneca, Gothenburg, Sweden
| | - Oleksandr Sverdlov
- Early Development Analytics, Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Elin M Svensson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden.,Department of Pharmacy, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Grace Montepiedra
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | | | - Weng Kee Wong
- Department of Biostatistics, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
57
|
Qu Y, Park SY, Wu Q, Shen W. A unified approach for evaluating the prediction of treatment effect across different types of endpoints. Pharm Stat 2021; 21:4-16. [PMID: 34268857 DOI: 10.1002/pst.2149] [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: 10/28/2020] [Revised: 03/18/2021] [Accepted: 06/01/2021] [Indexed: 11/06/2022]
Abstract
Phase 2 and 3 development failure is one of the key factors for high drug development cost. Robust prediction of a candidate drug's efficacy and safety profile could potentially improve the success rate of the drug development. Therefore, systematic evaluation of the prediction is important for learning and continuous improvement of the prediction. In this article, we proposed a set of unified criteria that allow to evaluate the predictions across different endpoints, indications and development stages: standardized bias (SB), standardized mean squared errors (SMSE), and credibility of prediction. We applied the SB and SMSE to the predicted treatment effects for 54 comparisons in 5 compounds in immunology and diabetes.
Collapse
Affiliation(s)
- Yongming Qu
- Global Statistical Sciences, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - So Young Park
- Global Statistical Sciences, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Qiwei Wu
- Global Statistical Sciences, Eli Lilly and Company, Indianapolis, Indiana, USA
| | - Wei Shen
- Global Statistical Sciences, Eli Lilly and Company, Indianapolis, Indiana, USA
| |
Collapse
|
58
|
Vinks AA, Barrett JS. Model-Informed Pediatric Drug Development: Application of Pharmacometrics to Define the Right Dose for Children. J Clin Pharmacol 2021; 61 Suppl 1:S52-S59. [PMID: 34185897 DOI: 10.1002/jcph.1841] [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: 12/29/2020] [Accepted: 02/16/2021] [Indexed: 12/26/2022]
Abstract
One of the biggest challenges in pediatric drug development is defining a safe and effective dose in pediatric populations, which span across a wide age and development range from neonates to adolescents. Model-informed drug development approaches are particularly suited to address knowledge gaps including data leveraging to increase the success of pediatric studies. Considering the often limited number of patients available for study and logistic difficulties to collect the necessary data in pediatric populations, the application of pharmacometrics and modeling and simulation techniques can improve clinical trial efficiency, increase the probability of regulatory success, and optimize therapeutic individualization in support of dedicated trials. This review describes the state of pediatric model-informed drug development to define the right dose for children and provides suggestions for future development.
Collapse
Affiliation(s)
- Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Jeffrey S Barrett
- Quantitative Medicine, Critical Path Institute, Tucson, Arizona, USA
| |
Collapse
|
59
|
Comets E, Rodrigues C, Jullien V, Ursino M. Conditional Non-parametric Bootstrap for Non-linear Mixed Effect Models. Pharm Res 2021; 38:1057-1066. [PMID: 34075519 DOI: 10.1007/s11095-021-03052-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/03/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE Non-linear mixed effect models are widely used and increasingly integrated into decision-making processes. Propagating uncertainty is an important element of this process, and while standard errors (SE) on pa- rameters are most often computed using asymptotic approaches, alternative methods such as the bootstrap are also available. In this article, we propose a modified residual parametric bootstrap taking into account the different levels of variability involved in these models. METHODS The proposed approach uses samples from the individual conditional distribution, and was implemented in R using the saemix algorithm. We performed a simulation study to assess its performance in different scenarios, comparing it to the asymptotic approximation and to standard bootstraps in terms of coverage, also looking at bias in the parameters and their SE. RESULTS Simulations with an Emax model with different designs and sigmoidicity factors showed a similar coverage rate to the parametric bootstrap, while requiring less hypotheses. Bootstrap improved coverage in several scenarios compared to the asymptotic method especially for the variance param-eters. However, all bootstraps were sensitive to estimation bias in the original datasets. CONCLUSIONS The conditional bootstrap provided better coverage rate than the traditional residual bootstrap, while preserving the structure of the data generating process.
Collapse
Affiliation(s)
- Emmanuelle Comets
- Universit́e de Paris, INSERM IAME; INSERM, CIC 1414; Rennes-1 University, France 16 rue Henri Huchard, 75018, Paris, France.
| | | | - Vincent Jullien
- UF Pharmacologie, GH Paris Seine Saint-Denis, Universit́e Paris, 13, Paris, France
| | - Moreno Ursino
- Unit of Clinical Epidemiology, Assistance Publique-Hôpitaux de Paris, CHU Robert Debré, Université de Paris, Sorbonne Paris-Cité, Inserm U1123 and CIC-EC 1426, Paris, F-75019, France
- Inserm, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, Paris, F-75006, France
- Inria, HeKA, F-75006, Paris, France
| |
Collapse
|
60
|
A hybrid modeling approach for assessing mechanistic models of small molecule partitioning in vivo using a machine learning-integrated modeling platform. Sci Rep 2021; 11:11143. [PMID: 34045592 PMCID: PMC8160209 DOI: 10.1038/s41598-021-90637-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
Prediction of the first-in-human dosing regimens is a critical step in drug development and requires accurate quantitation of drug distribution. Traditional in vivo studies used to characterize clinical candidate’s volume of distribution are error-prone, time- and cost-intensive and lack reproducibility in clinical settings. The paper demonstrates how a computational platform integrating machine learning optimization with mechanistic modeling can be used to simulate compound plasma concentration profile and predict tissue-plasma partition coefficients with high accuracy by varying the lipophilicity descriptor logP. The approach applied to chemically diverse small molecules resulted in comparable geometric mean fold-errors of 1.50 and 1.63 in pharmacokinetic outputs for direct tissue:plasma partition and hybrid logP optimization, with the latter enabling prediction of tissue permeation that can be used to guide toxicity and efficacy dosing in human subjects. The optimization simulations required to achieve these results were parallelized on the AWS cloud and generated outputs in under 5 h. Accuracy, speed, and scalability of the framework indicate that it can be used to assess the relevance of other mechanistic relationships implicated in pharmacokinetic-pharmacodynamic phenomena with a lower risk of overfitting datasets and generate large database of physiologically-relevant drug disposition for further integration with machine learning models.
Collapse
|
61
|
Wiklund SJ, Burman CF. Selection bias, investment decisions and treatment effect distributions. Pharm Stat 2021; 20:1168-1182. [PMID: 34002467 PMCID: PMC9290610 DOI: 10.1002/pst.2132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 04/09/2021] [Accepted: 05/03/2021] [Indexed: 11/08/2022]
Abstract
When making decisions regarding the investment and design for a Phase 3 programme in the development of a new drug, the results from preceding Phase 2 trials are an important source of information. However, only projects in which the Phase 2 results show promising treatment effects will typically be considered for a Phase 3 investment decision. This implies that, for those projects where Phase 3 is pursued, the underlying Phase 2 estimates are subject to selection bias. We will in this article investigate the nature of this selection bias based on a selection of distributions for the treatment effect. We illustrate some properties of Bayesian estimates, providing shrinkage of the Phase 2 estimate to counteract the selection bias. We further give some empirical guidance regarding the choice of prior distribution and comment on the consequences for decision-making in investment and planning for Phase 3 programmes.
Collapse
|
62
|
Fediuk DJ, Nucci G, Dawra VK, Callegari E, Zhou S, Musante CJ, Liang Y, Sweeney K, Sahasrabudhe V. End-to-end application of model-informed drug development for ertugliflozin, a novel sodium-glucose cotransporter 2 inhibitor. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:529-542. [PMID: 33932126 PMCID: PMC8213419 DOI: 10.1002/psp4.12633] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/02/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022]
Abstract
Model-informed drug development (MIDD) is critical in all stages of the drug-development process and almost all regulatory submissions for new agents incorporate some form of modeling and simulation. This review describes the MIDD approaches used in the end-to-end development of ertugliflozin, a sodium-glucose cotransporter 2 inhibitor approved for the treatment of adults with type 2 diabetes mellitus. Approaches included (1) quantitative systems pharmacology modeling to predict dose-response relationships, (2) dose-response modeling and model-based meta-analysis for dose selection and efficacy comparisons, (3) population pharmacokinetics (PKs) modeling to characterize PKs and quantify population variability in PK parameters, (4) regression modeling to evaluate ertugliflozin dose-proportionality and the impact of uridine 5'-diphospho-glucuronosyltransferase (UGT) 1A9 genotype on ertugliflozin PKs, and (5) physiologically-based PK modeling to assess the risk of UGT-mediated drug-drug interactions. These end-to-end MIDD approaches for ertugliflozin facilitated decision making, resulted in time/cost savings, and supported registration and labeling.
Collapse
Affiliation(s)
| | | | | | | | - Susan Zhou
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | | | | | | |
Collapse
|
63
|
McIntosh A, Sverdlov O, Yu L, Kaufmann P. Clinical Design and Analysis Strategies for the Development of Gene Therapies: Considerations for Quantitative Drug Development in the Age of Genetic Medicine. Clin Pharmacol Ther 2021; 110:1207-1215. [PMID: 33666225 DOI: 10.1002/cpt.2224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/01/2021] [Indexed: 12/19/2022]
Abstract
Cell and gene therapies have shown enormous promise across a range of diseases in recent years. Numerous adoptive cell therapy modalities as well as systemic and direct-to-target tissue gene transfer administrations are currently in clinical development. The clinical trial design, development, reporting, and analysis of novel cell and gene therapies can differ significantly from established practices for small molecule drugs and biologics. Here, we discuss important quantitative considerations and key competencies for drug developers in preclinical requirements, trial design, and lifecycle planning for gene therapies. We argue that the unique development path of gene therapies requires practicing quantitative drug developers-statisticians, pharmacometricians, pharmacokineticists, epidemiologists, and medical and translational science leads-to exercise active collaboration and cross-functional learning across development stages.
Collapse
Affiliation(s)
| | | | - Li Yu
- Novartis Gene Therapies, Bannockburn, Illinois, USA
| | | |
Collapse
|
64
|
Quan H, Kang T, Fan C, Lu X, Chen X, Luo X, Wei L. Trial monitoring via a futility criterion for interim results on a count data endpoint and a continuous endpoint. Contemp Clin Trials 2021; 103:106316. [PMID: 33571688 DOI: 10.1016/j.cct.2021.106316] [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: 09/23/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 10/22/2022]
Abstract
Assumptions made at design stage regarding the true treatment effect, background event rate and other factors may not always hold. Thus, long-term and large-scale studies may be designed with an interim analysis in order that the trials may be stopped early due to futility to save resource. There are many considerations of trial conducts for this type of trials. In this paper, we use a mock study to illustrate systematically the thinking and procedures for trial monitoring with a futility criterion for the interim results on a count data endpoint and a continuous endpoint. We focus on the discussions of blinded trial monitoring, the probability of meeting the futility criterion, conditional power/probability of success, Bayesian inference, potential delayed treatment effect and subgroup analysis. The experience should be applicable to future studies with similar features.
Collapse
Affiliation(s)
- Hui Quan
- Biostatistics and Programming, Sanofi, 55 Corporate Drive, Bridgewater, NJ 08807, United States of America.
| | - Tong Kang
- Biostatistics and Programming, Sanofi, 55 Corporate Drive, Bridgewater, NJ 08807, United States of America
| | - Chunpeng Fan
- Biostatistics and Programming, Sanofi, 55 Corporate Drive, Bridgewater, NJ 08807, United States of America
| | - Xin Lu
- Biostatistics and Programming, Sanofi, 55 Corporate Drive, Bridgewater, NJ 08807, United States of America
| | - Xun Chen
- Biostatistics and Programming, Sanofi, 55 Corporate Drive, Bridgewater, NJ 08807, United States of America
| | - Xiaodong Luo
- Biostatistics and Programming, Sanofi, 55 Corporate Drive, Bridgewater, NJ 08807, United States of America
| | - Lynn Wei
- Biostatistics and Programming, Sanofi, 55 Corporate Drive, Bridgewater, NJ 08807, United States of America
| |
Collapse
|
65
|
Vlachakis D, Vlamos P. Mathematical Multidimensional Modelling and Structural Artificial Intelligence Pipelines Provide Insights for the Designing of Highly Specific AntiSARS-CoV2 Agents. MATHEMATICS IN COMPUTER SCIENCE 2021; 15. [PMCID: PMC8205651 DOI: 10.1007/s11786-021-00517-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
COVID19 is the most impactful pandemic of recent times worldwide. It is a highly infectious disease that is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 virus), To date there is specific drug nor vaccination against COVID19. Therefor the need for novel and pioneering anti-COVID19 is of paramount importance. In this direction, computer-aided drug design constitutes a very promising antiviral approach for the discovery and analysis of drugs and molecules with biological activity against SARS-CoV2. In silico modelling takes advantage of the massive amounts of biological and chemical data available on the nature of the interactions between the targeted systems and molecules, as well as the rapid progress of computational tools and software. Herein, we describe the potential of the merging of mathematical modelling, artificial intelligence and learning techniques into seamless computational pipelines for the rapid and efficient discovery and design of potent anti- SARS-CoV-2 modulators.
Collapse
Affiliation(s)
- Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, Genetics and Computational Biology Group, School of Applied Biology and Biotechnology, Agricultural University of Athens, Iera Odos 75 Str. GR11855, Athens, Greece
- Laboratory of Molecular Endocrinology, Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Soranou Ephessiou Str. GR11527, Athens, Greece
- University Research Institute of Maternal and Child Health and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Thivon 1 & Papadiamantopoulou Str. GR11527, Athens, Greece
| | - Panayiotis Vlamos
- Department of Informatics, Ionian University, Plateia Tsirigoti 7, 49100 Corfu, Greece
| |
Collapse
|
66
|
Egnell AC, Johansson S, Chen C, Berges A. Clinical Pharmacology Modeling and Simulation in Drug Development. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11546-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
67
|
Barrett JS. Time to Expect More From Pharmacometrics. Clin Pharmacol Ther 2020; 108:1129-1131. [PMID: 32562273 PMCID: PMC7687126 DOI: 10.1002/cpt.1914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/15/2020] [Indexed: 11/12/2022]
Affiliation(s)
- Jeffrey S. Barrett
- Bill & Melinda Gates Medical Research InstituteCambridgeMassachusettsUSA
- Present address:
Critical Path InstituteTusconArizonaUSA
| |
Collapse
|
68
|
Anziano RJ, Milligan PA. Model Informed Drug Development: Collaboration Through A Common Framework. Clin Pharmacol Ther 2020; 110:1165-1167. [PMID: 33111309 PMCID: PMC8596610 DOI: 10.1002/cpt.2066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/23/2020] [Indexed: 11/07/2022]
|
69
|
Darwich AS, Polasek TM, Aronson JK, Ogungbenro K, Wright DFB, Achour B, Reny JL, Daali Y, Eiermann B, Cook J, Lesko L, McLachlan AJ, Rostami-Hodjegan A. Model-Informed Precision Dosing: Background, Requirements, Validation, Implementation, and Forward Trajectory of Individualizing Drug Therapy. Annu Rev Pharmacol Toxicol 2020; 61:225-245. [PMID: 33035445 DOI: 10.1146/annurev-pharmtox-033020-113257] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Model-informed precision dosing (MIPD) has become synonymous with modern approaches for individualizing drug therapy, in which the characteristics of each patient are considered as opposed to applying a one-size-fits-all alternative. This review provides a brief account of the current knowledge, practices, and opinions on MIPD while defining an achievable vision for MIPD in clinical care based on available evidence. We begin with a historical perspective on variability in dose requirements and then discuss technical aspects of MIPD, including the need for clinical decision support tools, practical validation, and implementation of MIPD in health care. We also discuss novel ways to characterize patient variability beyond the common perceptions of genetic control. Finally, we address current debates on MIPD from the perspectives of the new drug development, health economics, and drug regulations.
Collapse
Affiliation(s)
- Adam S Darwich
- Logistics and Informatics in Health Care, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), KTH Royal Institute of Technology, SE-141 57 Huddinge, Sweden
| | - Thomas M Polasek
- Department of Clinical Pharmacology, Royal Adelaide Hospital, Adelaide, South Australia 5000, Australia.,Centre for Medicine Use and Safety, Monash University, Melbourne, Victoria 3052, Australia.,Certara, Princeton, New Jersey 08540, USA
| | - Jeffrey K Aronson
- Centre for Evidence Based Medicine, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford OX2 6GG, United Kingdom
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester M13 9PT, United Kingdom;
| | | | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Jean-Luc Reny
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland.,Division of General Internal Medicine, Geneva University Hospitals, CH-1211 Geneva, Switzerland
| | - Youssef Daali
- Geneva Platelet Group, Faculty of Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Birgit Eiermann
- Inera AB, Swedish Association of Local Authorities and Regions, SE-118 93 Stockholm, Sweden
| | - Jack Cook
- Drug Safety Research & Development, Pfizer Inc., Groton, Connecticut 06340, USA
| | - Lawrence Lesko
- Center for Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, Florida 32827, USA
| | - Andrew J McLachlan
- School of Pharmacy, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Amin Rostami-Hodjegan
- Certara, Princeton, New Jersey 08540, USA.,Centre for Applied Pharmacokinetic Research, The University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
70
|
Srinivasan M, White A, Chaturvedula A, Vozmediano V, Schmidt S, Plouffe L, Wingate LT. Incorporating Pharmacometrics into Pharmacoeconomic Models: Applications from Drug Development. PHARMACOECONOMICS 2020; 38:1031-1042. [PMID: 32734572 PMCID: PMC7578131 DOI: 10.1007/s40273-020-00944-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Pharmacometrics is the science of quantifying the relationship between the pharmacokinetics and pharmacodynamics of drugs in combination with disease models and trial information to aid in drug development and dosing optimization for clinical practice. Considering the variability in the dose-concentration-effect relationship of drugs, an opportunity exists in linking pharmacokinetic and pharmacodynamic model-based estimates with pharmacoeconomic models. This link may provide early estimates of the cost effectiveness of drug therapies, thus informing late-stage drug development, pricing, and reimbursement decisions. Published case studies have demonstrated how integrated pharmacokinetic-pharmacodynamic-pharmacoeconomic models can complement traditional pharmacoeconomic analyses by identifying the impact of specific patient sub-groups, dose, dosing schedules, and adherence on the cost effectiveness of drugs, thus providing a mechanistic basis to predict the economic value of new drugs. Greater collaboration between the pharmacoeconomics and pharmacometrics community can enable methodological improvements in pharmacokinetic-pharmacodynamic-pharmacoeconomic models to support drug development.
Collapse
Affiliation(s)
- Meenakshi Srinivasan
- University of North Texas System College of Pharmacy, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Annesha White
- University of North Texas System College of Pharmacy, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA.
| | - Ayyappa Chaturvedula
- University of North Texas System College of Pharmacy, 3500 Camp Bowie Blvd, Fort Worth, TX, 76107, USA
| | - Valvanera Vozmediano
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Orlando, FL, USA
| | | | | |
Collapse
|
71
|
Chen X, Wang DD, Li ZP. Analysis of time course and dose effect of tacrolimus on proteinuria in lupus nephritis patients. J Clin Pharm Ther 2020; 46:106-113. [PMID: 32974902 DOI: 10.1111/jcpt.13260] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/11/2020] [Accepted: 08/17/2020] [Indexed: 12/21/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVES Tacrolimus is used to treat patients with lupus nephritis; however, its time course and dose effect on proteinuria in lupus nephritis patients remain unknown. The purpose of this study was to determine the time course and dose effect of tacrolimus on proteinuria in lupus nephritis patients via model-based meta-analysis (MBMA). METHODS PubMed, Web of Science, Cochrane Library and ClinicalTrials.gov databases were systematically searched for information on the efficacy of tacrolimus against proteinuria in lupus nephritis patients. Useful data were extracted to build a model for the population studied using a non-linear mixed-effect model (NONMEM). This model was applied to simulate time course of tacrolimus on proteinuria using Monte Carlo simulations. RESULTS Ten clinical studies that recruited 222 patients with lupus nephritis were included. Based on various diagnostic plots, we found that the established model described the observed data reasonably well. In addition, the typical Emax and ET50 of tacrolimus for 24-hour proteinuria in lupus nephritis patients were -5.88 g and 0.37 months, respectively. The baseline value of 24-hour proteinuria affected Emax . No significant dose-response relationship was observed in the range of tacrolimus concentration used in the present study (3-10 ng/mL), indicating that the effect of tacrolimus on proteinuria depends on effective concentration range and not the dose. However, the time course relationship was obvious; the efficacy of tacrolimus increased over time, reaching a plateau (80% Emax ) at approximately 1.48 months from the beginning of treatment. WHAT IS NEW AND CONCLUSION When the concentration range of tacrolimus is maintained at 3-10 ng/mL, at least 1.48 months of treatment is required to achieve a better outcome with regard to proteinuria in lupus nephritis patients.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Pharmacy, Children's Hospital of Fudan University, Shanghai, China
| | - Dong-Dong Wang
- Department of Pharmacy, Children's Hospital of Fudan University, Shanghai, China
| | - Zhi-Ping Li
- Department of Pharmacy, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
72
|
Bi Y, Liu J, Li L, Yu J, Bhattaram A, Bewernitz M, Li RJ, Liu C, Earp J, Ma L, Zhuang L, Yang Y, Zhang X, Zhu H, Wang Y. Role of Model-Informed Drug Development in Pediatric Drug Development, Regulatory Evaluation, and Labeling. J Clin Pharmacol 2020; 59 Suppl 1:S104-S111. [PMID: 31502691 DOI: 10.1002/jcph.1478] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 11/12/2022]
Abstract
The unique challenges in pediatric drug development require efficient and innovative tools. Model-informed drug development (MIDD) offers many powerful tools that have been frequently applied in pediatric drug development. MIDD refers to the application of quantitative models to integrate and leverage existing knowledge to bridge knowledge gaps and facilitate development and decision-making processes. This article discusses the current practices and visions of applying MIDD in pediatric drug development, regulatory evaluation, and labeling, with detailed examples. The application of MIDD in pediatric drug development can be broadly classified into 3 categories: leveraging knowledge for bridging the gap, dose selection and optimization, and informing clinical trial design. In particular, MIDD can provide evidence for the assumption of exposure-response similarity in bridging existing knowledge from reference to target population, support the dose selection and optimization based on the "exposure-matching" principle in the pediatric population, and increase the efficiency and success rate of pediatric trials. In addition, the role of physiologically based pharmacokinetics in drug-drug interaction in children and adolescents and in utilizing ontogeny data to predict pharmacokinetics in neonates and infants has also been illustrated. Moving forward, MIDD should be incorporated into all pediatric drug development programs at every stage to inform clinical trial design and dose selection, with both its strengths and limitations clearly laid out. The accumulated experience and knowledge of MIDD has and will continue to drive regulatory policy development and refinement, which will ultimately improve the consistency and efficiency of pediatric drug development.
Collapse
Affiliation(s)
- Youwei Bi
- Food and Drug Administration, Silver Spring, MD, USA
| | - Jiang Liu
- Food and Drug Administration, Silver Spring, MD, USA
| | - Lingjue Li
- Food and Drug Administration, Silver Spring, MD, USA
| | - Jingyu Yu
- Food and Drug Administration, Silver Spring, MD, USA
| | | | | | - Ruo-Jing Li
- Food and Drug Administration, Silver Spring, MD, USA
| | - Chao Liu
- Food and Drug Administration, Silver Spring, MD, USA
| | - Justin Earp
- Food and Drug Administration, Silver Spring, MD, USA
| | - Lian Ma
- Food and Drug Administration, Silver Spring, MD, USA
| | - Luning Zhuang
- Food and Drug Administration, Silver Spring, MD, USA
| | - Yuching Yang
- Food and Drug Administration, Silver Spring, MD, USA
| | - Xinyuan Zhang
- Food and Drug Administration, Silver Spring, MD, USA
| | - Hao Zhu
- Food and Drug Administration, Silver Spring, MD, USA
| | - Yaning Wang
- Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
73
|
Hartmann S, Biliouris K, Lesko LJ, Nowak-Göttl U, Trame MN. Quantitative Systems Pharmacology Model-Based Predictions of Clinical Endpoints to Optimize Warfarin and Rivaroxaban Anti-Thrombosis Therapy. Front Pharmacol 2020; 11:1041. [PMID: 32765265 PMCID: PMC7381140 DOI: 10.3389/fphar.2020.01041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/26/2020] [Indexed: 11/25/2022] Open
Abstract
Background Tight monitoring of efficacy and safety of anticoagulants such as warfarin is imperative to optimize the benefit-risk ratio of anticoagulants in patients. The standard tests used are measurements of prothrombin time (PT), usually expressed as international normalized ratio (INR), and activated partial thromboplastin time (aPTT). Objective To leverage a previously developed quantitative systems pharmacology (QSP) model of the human coagulation network to predict INR and aPTT for warfarin and rivaroxaban, respectively. Methods A modeling and simulation approach was used to predict INR and aPTT measurements of patients receiving steady-state anticoagulation therapy. A previously developed QSP model was leveraged for the present analysis. The effect of genetic polymorphisms known to influence dose response of warfarin (CYP2C9, VKORC1) were implemented into the model by modifying warfarin clearance (CYP2C9 *1: 0.2 L/h; *2: 0.14 L/h, *3: 0.04 L/h) and the concentration of available vitamin K (VKORC1 GA: −22% from normal vitamin K concentration; AA: −44% from normal vitamin K concentration). Virtual patient populations were used to assess the ability of the model to accurately predict routine INR and aPTT measurements from patients under long-term anticoagulant therapy. Results The introduced model accurately described the observed INR of patients receiving long-term warfarin treatment. The model was able to demonstrate the influence of genetic polymorphisms of CYP2C9 and VKORC1 on the INR measurements. Additionally, the model was successfully used to predict aPTT measurements for patients receiving long-term rivaroxaban therapy. Conclusion The QSP model accurately predicted INR and aPTT measurements observed during routine therapeutic drug monitoring. This is an exemplar of how a QSP model can be adapted and used as a model-based precision dosing tool during clinical practice and drug development to predict efficacy and safety of anticoagulants to ultimately help optimize anti-thrombotic therapy.
Collapse
Affiliation(s)
- Sonja Hartmann
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, FL, United States
| | - Konstantinos Biliouris
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, FL, United States
| | - Lawrence J Lesko
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, FL, United States
| | - Ulrike Nowak-Göttl
- Thrombosis & Hemostasis Treatment Center, Institute of Clinical Chemistry, University of Schleswig-Holstein, Germany
| | - Mirjam N Trame
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics, University of Florida, Orlando, FL, United States
| |
Collapse
|
74
|
Assessing parameter uncertainty in small-n pharmacometric analyses: value of the log-likelihood profiling-based sampling importance resampling (LLP-SIR) technique. J Pharmacokinet Pharmacodyn 2020; 47:219-228. [PMID: 32248328 PMCID: PMC7289778 DOI: 10.1007/s10928-020-09682-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 03/26/2020] [Indexed: 01/23/2023]
Abstract
Assessing parameter uncertainty is a crucial step in pharmacometric workflows. Small datasets with ten or fewer subjects appear regularly in drug development and therapeutic use, but it is unclear which method to assess parameter uncertainty is preferable in such situations. The aim of this study was to (i) systematically evaluate the performance of standard error (SE), bootstrap (BS), log-likelihood profiling (LLP), Bayesian approaches (BAY) and sampling importance resampling (SIR) to assess parameter uncertainty in small datasets and (ii) to evaluate methods to provide proposal distributions for the SIR. A simulation study was conducted and the 0-95% confidence interval (CI) and coverage for each parameter was evaluated and compared to reference CIs derived by stochastic simulation and estimation (SSE). A newly proposed LLP-SIR, combining the proposal distribution provided by LLP with SIR, was included in addition to conventional SE-SIR and BS-SIR. Additionally, the methods were applied to a clinical dataset. The determined CIs differed substantially across the methods. The CIs of SE, BS, LLP and BAY were not in line with the reference in datasets with ≤ 10 subjects. The best alignment was found for the LLP-SIR, which also provided the best coverage results among the SIR methods. The best overall results regarding the coverage were provided by LLP and BAY across all parameters and dataset sizes. To conclude, the popular SE and BS methods are not suitable to derive parameter uncertainty in small datasets containing ≤ 10 subjects, while best performances were observed with LLP, BAY and LLP-SIR.
Collapse
|
75
|
Sassen SDT, Zwaan CM, van der Sluis IM, Mathôt RAA. Pharmacokinetics and population pharmacokinetics in pediatric oncology. Pediatr Blood Cancer 2020; 67:e28132. [PMID: 31876123 DOI: 10.1002/pbc.28132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 11/19/2019] [Accepted: 11/24/2019] [Indexed: 12/28/2022]
Abstract
Pharmacokinetic research has become increasingly important in pediatric oncology as it can have direct clinical implications and is a crucial component in individualized medicine. Population pharmacokinetics has become a popular method especially in children, due to the potential for sparse sampling, flexible sampling times, computing of heterogeneous data, and identification of variability sources. However, population pharmacokinetic reports can be complex and difficult to interpret. The aim of this article is to provide a basic explanation of population pharmacokinetics, using clinical examples from the field of pediatric oncology, to facilitate the translation of pharmacokinetic research into the daily clinic.
Collapse
Affiliation(s)
- Sebastiaan D T Sassen
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - C Michel Zwaan
- Department of Pediatric Oncology, Erasmus MC-Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | - Ron A A Mathôt
- Department of Hospital Pharmacy, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
76
|
Ayyar VS, Jusko WJ. Transitioning from Basic toward Systems Pharmacodynamic Models: Lessons from Corticosteroids. Pharmacol Rev 2020; 72:414-438. [PMID: 32123034 PMCID: PMC7058984 DOI: 10.1124/pr.119.018101] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Technology in bioanalysis, -omics, and computation have evolved over the past half century to allow for comprehensive assessments of the molecular to whole body pharmacology of diverse corticosteroids. Such studies have advanced pharmacokinetic and pharmacodynamic (PK/PD) concepts and models that often generalize across various classes of drugs. These models encompass the "pillars" of pharmacology, namely PK and target drug exposure, the mass-law interactions of drugs with receptors/targets, and the consequent turnover and homeostatic control of genes, biomarkers, physiologic responses, and disease symptoms. Pharmacokinetic methodology utilizes noncompartmental, compartmental, reversible, physiologic [full physiologically based pharmacokinetic (PBPK) and minimal PBPK], and target-mediated drug disposition models using a growing array of pharmacometric considerations and software. Basic PK/PD models have emerged (simple direct, biophase, slow receptor binding, indirect response, irreversible, turnover with inactivation, and transduction models) that place emphasis on parsimony, are mechanistic in nature, and serve as highly useful "top-down" methods of quantitating the actions of diverse drugs. These are often components of more complex quantitative systems pharmacology (QSP) models that explain the array of responses to various drugs, including corticosteroids. Progressively deeper mechanistic appreciation of PBPK, drug-target interactions, and systems physiology from the molecular (genomic, proteomic, metabolomic) to cellular to whole body levels provides the foundation for enhanced PK/PD to comprehensive QSP models. Our research based on cell, animal, clinical, and theoretical studies with corticosteroids have provided ideas and quantitative methods that have broadly advanced the fields of PK/PD and QSP modeling and illustrates the transition toward a global, systems understanding of actions of diverse drugs. SIGNIFICANCE STATEMENT: Over the past half century, pharmacokinetics (PK) and pharmacokinetics/pharmacodynamics (PK/PD) have evolved to provide an array of mechanism-based models that help quantitate the disposition and actions of most drugs. We describe how many basic PK and PK/PD model components were identified and often applied to the diverse properties of corticosteroids (CS). The CS have complications in disposition and a wide array of simple receptor-to complex gene-mediated actions in multiple organs. Continued assessments of such complexities have offered opportunities to develop models ranging from simple PK to enhanced PK/PD to quantitative systems pharmacology (QSP) that help explain therapeutic and adverse CS effects. Concurrent development of state-of-the-art PK, PK/PD, and QSP models are described alongside experimental studies that revealed diverse CS actions.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Department of Pharmaceutical Sciences University at Buffalo, School of Pharmacy and Pharmaceutical Sciences, Buffalo, New York
| | - William J Jusko
- Department of Pharmaceutical Sciences University at Buffalo, School of Pharmacy and Pharmaceutical Sciences, Buffalo, New York
| |
Collapse
|
77
|
Krishnaswami S, Austin D, Della Pasqua O, Gastonguay MR, Gobburu J, van der Graaf PH, Ouellet D, Tannenbaum S, Visser SAG. MID3: Mission Impossible or Model-Informed Drug Discovery and Development? Point-Counterpoint Discussions on Key Challenges. Clin Pharmacol Ther 2020; 107:762-772. [PMID: 31955417 PMCID: PMC7158219 DOI: 10.1002/cpt.1788] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/03/2020] [Indexed: 11/12/2022]
|
78
|
Cheng Q, Huang J, Xu L, Li Y, Li H, Shen Y, Zheng Q, Li L. Analysis of Time-Course, Dose-Effect, and Influencing Factors of Antidepressants in the Treatment of Acute Adult Patients With Major Depression. Int J Neuropsychopharmacol 2020; 23:76-87. [PMID: 31774497 PMCID: PMC7094001 DOI: 10.1093/ijnp/pyz062] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 11/12/2019] [Accepted: 11/26/2019] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Model-based meta-analysis was used to describe the time-course and dose-effect relationships of antidepressants and also simultaneously investigate the impact of various factors on drug efficacy. METHODS This study is a reanalysis of a published network meta-analysis. Only placebo-controlled trials were included in this study. The change rate in depression rating scale scores from baseline was used as an efficacy indicator because a continuous variable is more likely to reflect subtle differences in efficacy between drugs. RESULTS A total 230 studies containing 64 346 patients were included in the analysis. The results showed that the number of study sites (single or multi-center) and the type of setting (inpatient or noninpatient) are important factors affecting the efficacy of antidepressants. After deducting the placebo effect, the maximum pure drug efficacy value of inpatients was 18.4% higher than that of noninpatients, and maximum pure drug efficacy value of single-center trials was 10.2% higher than that of multi-central trials. Amitriptyline showed the highest drug efficacy. The remaining 18 antidepressants were comparable or had little difference. Within the approved dose range, no significant dose-response relationship was observed. However, the time-course relationship is obvious for all antidepressants. In terms of safety, with the exception of amitriptyline, the dropout rate due to adverse events of other drugs was not more than 10% higher than that of the placebo group. CONCLUSION The number of study sites and the type of setting are significant impact factors for the efficacy of antidepressants. Except for amitriptyline, the other 18 antidepressants have little difference in efficacy and safety.
Collapse
Affiliation(s)
- Qingqing Cheng
- Center for Drug of Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jihan Huang
- Center for Drug of Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Xu
- Center for Drug of Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunfei Li
- Center for Drug of Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huafang Li
- Shanghai Mental Health Center, Shanghai, China
| | - Yifeng Shen
- Shanghai Mental Health Center, Shanghai, China
| | - Qingshan Zheng
- Center for Drug of Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lujin Li
- Center for Drug of Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
79
|
Peters SA, Petersson C, Blaukat A, Halle JP, Dolgos H. Prediction of active human dose: learnings from 20 years of Merck KGaA experience, illustrated by case studies. Drug Discov Today 2020; 25:909-919. [PMID: 31981792 DOI: 10.1016/j.drudis.2020.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/24/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022]
Abstract
High-quality dose predictions based on a good understanding of target engagement is one of the main translational goals in drug development. Here, we systematically evaluate active human dose predictions for 15 Merck KGaA/EMD Serono assets spanning several modalities and therapeutic areas. Using case studies, we illustrate the value of adhering to the translational best practices of having an exposure-response relationship in an appropriate animal model; having validated, translatable pharmacodynamic (PD) biomarkers measurable in Phase I populations in the right tissue; having a deeper understanding of biology; and capturing uncertainties in predictions. Given the gap in publications on the subject, we believe that the learnings from this unique diverse data set, which are generic to the industry, will trigger actions to improve future predictions.
Collapse
Affiliation(s)
- Sheila Annie Peters
- Translational Quantitative Pharmacology, Translational Medicine, Biopharma, Global R&D, Merck Healthcare, Frankfurter Str. 250, 64293 Darmstadt, Germany.
| | - Carl Petersson
- Drug Metabolism and Disposition, Discovery Technology, Biopharma, Global R&D, Merck Healthcare, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Andree Blaukat
- Translational Innovation Platform Oncology, Biopharma, Global R&D, Merck Healthcare, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Joern-Peter Halle
- Translational Innovation Platform Immuno Oncology, Biopharma, Global R&D, Merck Healthcare, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Hugues Dolgos
- Biopharmacy Center of Excellence, Servier RD, Suresnes, 92150, France
| |
Collapse
|
80
|
Quan H, Chen X, Lan Y, Luo X, Kubiak R, Bonnet N, Paux G. Applications of Bayesian analysis to proof‐of‐concept trial planning and decision making. Pharm Stat 2020; 19:468-481. [DOI: 10.1002/pst.1985] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 07/23/2019] [Accepted: 10/15/2019] [Indexed: 11/10/2022]
Affiliation(s)
- Hui Quan
- Biostatistics and ProgrammingSanofi Bridgewater New Jersey
| | - Xun Chen
- Biostatistics and ProgrammingSanofi Bridgewater New Jersey
| | - Yu Lan
- Biostatistics and ProgrammingSanofi Bridgewater New Jersey
| | - Xiaodong Luo
- Biostatistics and ProgrammingSanofi Bridgewater New Jersey
| | - Rene Kubiak
- Biostatistics and ProgrammingSanofi Bridgewater New Jersey
| | - Nicolas Bonnet
- Biostatistics and ProgrammingSanofi Bridgewater New Jersey
| | - Gautier Paux
- Biostatistics and ProgrammingSanofi Bridgewater New Jersey
| |
Collapse
|
81
|
Xu Y, Constantine F, Yuan Y, Pritchett YL. ASIED: a Bayesian adaptive subgroup-identification enrichment design. J Biopharm Stat 2019; 30:623-638. [PMID: 31782938 DOI: 10.1080/10543406.2019.1696356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Developing targeted therapies based on patients' baseline characteristics and genomic profiles such as biomarkers has gained growing interests in recent years. Depending on patients' clinical characteristics, the expression of specific biomarkers or their combinations, different patient subgroups could respond differently to the same treatment. An ideal design, especially at the proof of concept stage, should search for such subgroups and make dynamic adaptation as the trial goes on. When no prior knowledge is available on whether the treatment works on the all-comer population or only works on the subgroup defined by one biomarker or several biomarkers, it is necessary to incorporate the adaptive estimation of the heterogeneous treatment effect to the decision-making at interim analyses. To address this problem, we propose an Adaptive Subgroup-Identification Enrichment Design, ASIED, to simultaneously search for predictive biomarkers, identify the subgroups with differential treatment effects, and modify study entry criteria at interim analyses when justified. More importantly, we construct robust quantitative decision-making rules for population enrichment when the interim outcomes are heterogeneous in the context of a multilevel target product profile, which defines the minimal and targeted levels of treatment effect. Through extensive simulations, the ASIED is demonstrated to achieve desirable operating characteristics and compare favorably against alternatives.
Collapse
Affiliation(s)
- Yanxun Xu
- Department of Applied Mathematics and Statistics, Johns Hopkins University , Baltimore, Maryland, USA
| | - Florica Constantine
- Department of Applied Mathematics and Statistics, Johns Hopkins University , Baltimore, Maryland, USA
| | - Yuan Yuan
- Late Respiratory Inflammatory and Autoimmunity Biometrics, AstraZeneca , Gaithersburg, Maryland, USA
| | - Yili L Pritchett
- Biometrics, G1 Therapeutics, Inc., Research Triangle Park , NC, USA
| |
Collapse
|
82
|
Madabushi R, Wang Y, Zineh I. A Holistic and Integrative Approach for Advancing Model-Informed Drug Development. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2019; 8:9-11. [PMID: 30582301 PMCID: PMC6363064 DOI: 10.1002/psp4.12379] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Rajanikanth Madabushi
- Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Yaning Wang
- Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Issam Zineh
- Office of Clinical Pharmacology, Office of Translational Sciences, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
83
|
Fidler M, Wilkins JJ, Hooijmaijers R, Post TM, Schoemaker R, Trame MN, Xiong Y, Wang W. Nonlinear Mixed-Effects Model Development and Simulation Using nlmixr and Related R Open-Source Packages. CPT Pharmacometrics Syst Pharmacol 2019; 8:621-633. [PMID: 31207186 PMCID: PMC6765694 DOI: 10.1002/psp4.12445] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 04/29/2019] [Indexed: 12/15/2022] Open
Abstract
nlmixr is a free and open-source R package for fitting nonlinear pharmacokinetic (PK), pharmacodynamic (PD), joint PK-PD, and quantitative systems pharmacology mixed-effects models. Currently, nlmixr is capable of fitting both traditional compartmental PK models as well as more complex models implemented using ordinary differential equations. We believe that, over time, it will become a capable, credible alternative to commercial software tools, such as NONMEM, Monolix, and Phoenix NLME.
Collapse
Affiliation(s)
| | | | | | | | | | - Mirjam N. Trame
- Novartis Institutes for BioMedical ResearchCambridgeMassachusettsUSA
| | | | - Wenping Wang
- Novartis Pharmaceuticals CorporationEast HanoverNew JerseyUSA
| |
Collapse
|
84
|
Olson EJ, Mahar KM, Haws TF, Fossler MJ, Gao F, de Gouville AC, Sprecher DL, Lepore JJ. A Randomized, Placebo-Controlled Trial to Assess the Effects of 8 Weeks of Administration of GSK256073, a Selective GPR109A Agonist, on High-Density Lipoprotein Cholesterol in Subjects With Dyslipidemia. Clin Pharmacol Drug Dev 2019; 8:871-883. [PMID: 31268250 DOI: 10.1002/cpdd.704] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/07/2019] [Indexed: 11/11/2022]
Abstract
GPR109A (HM74A), a G-protein-coupled receptor, is hypothesized to mediate lipid and lipoprotein changes and dermal flushing associated with niacin administration. GSK256073 (8-chloro-3-pentyl-1H-purine-2,6[3H,7H]-dione) is a selective GPR109A agonist shown to suppress fatty acid levels and produce mild flushing in short-term clinical studies. This study evaluated the effects of GSK256073 on lipids in subjects with low high-density lipoprotein cholesterol (HDLc). Subjects (n = 80) were randomized (1:1:1:1) to receive GSK256073 5, 50, or 150 mg/day or matching placebo for 8 weeks. The primary end point was determining the GSK256073 exposure-response relationship for change from baseline in HDLc. No significant exposure response was observed between GSK256073 and HDLc levels. GSK256073 did not significantly alter HDLc levels versus placebo, but rather revealed a trend at the 150-mg dose for a nonsignificant decrease in HDLc (-6.31%; P = .12) and an increase in triglycerides (median, 24.4%; 95% confidence interval, 7.3%-41.6%). Flushing was reported in 21%, 25%, and 60% of subjects (5, 50, and 150 mg, respectively) versus 24% for placebo. Results indicated that selective activation of the GPR109A receptor with GSK256073 did not produce niacin-like lipid effects. These findings add to the increasing evidence that niacin-mediated lipoprotein changes occur predominantly via GPR109A-independent pathways.
Collapse
Affiliation(s)
- Eric J Olson
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline, Collegeville, PA, USA
| | - Kelly M Mahar
- Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, Collegeville, PA, USA
| | - Thomas F Haws
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline, Collegeville, PA, USA
| | - Michael J Fossler
- Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, Collegeville, PA, USA
| | - Feng Gao
- Clinical Statistics, Metabolic Pathways and Cardiovascular Unit, GlaxoSmithKline, Collegeville, PA, USA
| | | | - Dennis L Sprecher
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline, Collegeville, PA, USA
| | - John J Lepore
- Clinical Pharmacology and Experimental Medicine, GlaxoSmithKline, Collegeville, PA, USA
| |
Collapse
|
85
|
Seurat J, Nguyen TT, Mentré F. Robust designs accounting for model uncertainty in longitudinal studies with binary outcomes. Stat Methods Med Res 2019; 29:934-952. [DOI: 10.1177/0962280219850588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
To optimize designs for longitudinal studies analyzed by mixed-effect models with binary outcomes, the Fisher information matrix can be used. Optimal design approaches, however, require a priori knowledge of the model. We aim to propose, for the first time, a robust design approach accounting for model uncertainty in longitudinal trials with two treatment groups, assuming mixed-effect logistic models. To optimize designs given one model, we compute several optimality criteria based on Fisher information matrix evaluated by the new approach based on Monte-Carlo/Hamiltonian Monte-Carlo. We propose to use the DDS-optimality criterion, as it ensures a compromise between the precision of estimation of the parameters, and hence the Wald test power, and the overall precision of parameter estimation. To account for model uncertainty, we assume candidate models with their respective weights. We compute robust design across these models using compound DDS-optimality. Using the Fisher information matrix, we propose to predict the average power over these models. Evaluating this approach by clinical trial simulations, we show that the robust design is efficient across all models, allowing one to achieve good power of test. The proposed design strategy is a new and relevant approach to design longitudinal studies with binary outcomes, accounting for model uncertainty.
Collapse
Affiliation(s)
- Jérémy Seurat
- IAME, UMR 1137, INSERM, Université Paris Diderot, Paris, France
| | - Thu Thuy Nguyen
- IAME, UMR 1137, INSERM, Université Paris Diderot, Paris, France
| | - France Mentré
- IAME, UMR 1137, INSERM, Université Paris Diderot, Paris, France
| |
Collapse
|
86
|
Kowalski KG. Integration of Pharmacometric and Statistical Analyses Using Clinical Trial Simulations to Enhance Quantitative Decision Making in Clinical Drug Development. Stat Biopharm Res 2019. [DOI: 10.1080/19466315.2018.1560361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
87
|
Wang Y, Zhu H, Madabushi R, Liu Q, Huang S, Zineh I. Model‐Informed Drug Development: Current US Regulatory Practice and Future Considerations. Clin Pharmacol Ther 2019; 105:899-911. [DOI: 10.1002/cpt.1363] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 12/26/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Yaning Wang
- Office of Clinical PharmacologyOffice of Translational SciencesUS Food and Drug Administration Silver Spring Maryland USA
| | - Hao Zhu
- Office of Clinical PharmacologyOffice of Translational SciencesUS Food and Drug Administration Silver Spring Maryland USA
| | - Rajanikanth Madabushi
- Office of Clinical PharmacologyOffice of Translational SciencesUS Food and Drug Administration Silver Spring Maryland USA
| | - Qi Liu
- Office of Clinical PharmacologyOffice of Translational SciencesUS Food and Drug Administration Silver Spring Maryland USA
| | - Shiew‐Mei Huang
- Office of Clinical PharmacologyOffice of Translational SciencesUS Food and Drug Administration Silver Spring Maryland USA
| | - Issam Zineh
- Office of Clinical PharmacologyOffice of Translational SciencesUS Food and Drug Administration Silver Spring Maryland USA
| |
Collapse
|
88
|
Riggs MM, Cremers S. Pharmacometrics and systems pharmacology for metabolic bone diseases. Br J Clin Pharmacol 2019; 85:1136-1146. [PMID: 30690761 DOI: 10.1111/bcp.13881] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/30/2018] [Accepted: 01/19/2019] [Indexed: 12/20/2022] Open
Abstract
Mathematical modelling and simulation (M&S) of drug concentrations, pharmacologic effects and the (patho)physiologic systems within which they interact can be powerful tools for the preclinical, translational and clinical development of drugs. Indeed, the Prescription Drug User Fee Act (PDUFA VI), incorporated as part of the FDA Reauthorization Act of 2017 (FDARA), highlights the goal of advancing model-informed drug development (MIDD). MIDD can benefit development across many drug classes, including for metabolic bone diseases such as osteoporosis, cancer-related and numerous rare metabolic bone diseases; conditions characterized by significant morbidity and mortality. A drought looms in terms of the availability of new drugs to better treat these devastating diseases. This review provides an overview of several M&S approaches ranging from simple pharmacokinetic to integrated pharmacometric and systems pharmacology modelling. Examples are included to illustrate the use of these approaches during the development of several drugs for metabolic bone diseases such as bisphosphonates, denosumab, teriparatide and sclerostin inhibitors (romosozumab and blosozumab).
Collapse
Affiliation(s)
| | - Serge Cremers
- Departments of Pathology & Cell Biology and Medicine, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
89
|
Ibrahim MMA, Ueckert S, Freiberga S, Kjellsson MC, Karlsson MO. Model-Based Conditional Weighted Residuals Analysis for Structural Model Assessment. AAPS JOURNAL 2019; 21:34. [PMID: 30815754 PMCID: PMC6394649 DOI: 10.1208/s12248-019-0305-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 01/30/2019] [Indexed: 11/30/2022]
Abstract
Nonlinear mixed effects models are widely used to describe longitudinal data to improve the efficiency of drug development process or increase the understanding of the studied disease. In such settings, the appropriateness of the modeling assumptions is critical in order to draw correct conclusions and must be carefully assessed for any substantial violations. Here, we propose a new method for structure model assessment, based on assessment of bias in conditional weighted residuals (CWRES). We illustrate this method by assessing prediction bias in two integrated models for glucose homeostasis, the integrated glucose-insulin (IGI) model, and the integrated minimal model (IMM). One dataset was simulated from each model then analyzed with the two models. CWRES outputted from each model fitting were modeled to capture systematic trends in CWRES as well as the magnitude of structural model misspecifications in terms of difference in objective function values (ΔOFVBias). The estimates of CWRES bias were used to calculate the corresponding bias in conditional predictions by the inversion of first-order conditional estimation method’s covariance equation. Time, glucose, and insulin concentration predictions were the investigated independent variables. The new method identified correctly the bias in glucose sub-model of the integrated minimal model (IMM), when this bias occurred, and calculated the absolute and proportional magnitude of the resulting bias. CWRES bias versus the independent variables agreed well with the true trends of misspecification. This method is fast easily automated diagnostic tool for model development/evaluation process, and it is already implemented as part of the Perl-speaks-NONMEM software.
Collapse
Affiliation(s)
- Moustafa M A Ibrahim
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.,Department of Pharmacy Practice, Helwan University, Cairo, Egypt
| | - Sebastian Ueckert
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Svetlana Freiberga
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Maria C Kjellsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - Mats O Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
90
|
Adiraj Iyer M, Eddington DT. Storing and releasing rhodamine as a model hydrophobic compound in polydimethylsiloxane microfluidic devices. LAB ON A CHIP 2019; 19:574-579. [PMID: 30681692 DOI: 10.1039/c9lc00039a] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Polydimethylsiloxane (PDMS) is a ubiquitous material used in soft lithography and microfluidics. Due to its hydrophobic nature, PDMS tends to absorb small hydrophobic molecules, and is seen as a major disadvantage of the material in pharmaceutical and cell culture studies. While there have been extensive reports of attempts to treat PDMS to limit or block this absorption, little attention has been given to using this property as a feature in microfluidic devices. In this work, we leverage the ability of PDMS to store hydrophobic molecules inside the PDMS matrix and release them over time in a sustained manner.
Collapse
Affiliation(s)
- M Adiraj Iyer
- Dept of Bioengineering, University of Illinois at Chicago, Chicago, USA.
| | | |
Collapse
|
91
|
Glassman PM, Balthasar JP. Physiologically-based modeling of monoclonal antibody pharmacokinetics in drug discovery and development. Drug Metab Pharmacokinet 2019; 34:3-13. [PMID: 30522890 PMCID: PMC6378116 DOI: 10.1016/j.dmpk.2018.11.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/11/2018] [Accepted: 11/19/2018] [Indexed: 12/20/2022]
Abstract
Over the past few decades, monoclonal antibodies (mAbs) have become one of the most important and fastest growing classes of therapeutic molecules, with applications in a wide variety of disease areas. As such, understanding of the determinants of mAb pharmacokinetic (PK) processes (absorption, distribution, metabolism, and elimination) is crucial in developing safe and efficacious therapeutics. In the present review, we discuss the use of physiologically-based pharmacokinetic (PBPK) models as an approach to characterize the in vivo behavior of mAbs, in the context of the key PK processes that should be considered in these models. Additionally, we discuss current and potential future applications of PBPK in the drug discovery and development timeline for mAbs, spanning from identification of potential target molecules to prediction of potential drug-drug interactions. Finally, we conclude with a discussion of currently available PBPK models for mAbs that could be implemented in the drug development process.
Collapse
Affiliation(s)
- Patrick M Glassman
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214 United States; Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104 United States
| | - Joseph P Balthasar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214 United States.
| |
Collapse
|
92
|
Ermakov S, Schmidt BJ, Musante CJ, Thalhauser CJ. A Survey of Software Tool Utilization and Capabilities for Quantitative Systems Pharmacology: What We Have and What We Need. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2018; 8:62-76. [PMID: 30417600 PMCID: PMC6389347 DOI: 10.1002/psp4.12373] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/29/2018] [Indexed: 12/14/2022]
Abstract
Quantitative systems pharmacology (QSP) is a rapidly emerging discipline with application across a spectrum of challenges facing the pharmaceutical industry, including mechanistically informed prioritization of target pathways and combinations in discovery, target population, and dose expansion decisions early in clinical development, and analyses for regulatory authorities late in clinical development. QSP's development has influences from physiologic modeling, systems biology, physiologically‐based pharmacokinetic modeling, and pharmacometrics. Given a varied scientific heritage, a variety of tools to accomplish the demands of model development, application, and model‐based analysis of available data have been developed. We report the outcome from a community survey and resulting analysis of how modelers view the impact and growth of QSP, how they utilize existing tools, and capabilities they need improved to further accelerate their impact on drug development. These results serve as a benchmark and roadmap for advancements to the QSP tool set.
Collapse
|
93
|
Swift B, Jain L, White C, Chandrasekaran V, Bhandari A, Hughes DA, Jadhav PR. Innovation at the Intersection of Clinical Trials and Real-World Data Science to Advance Patient Care. Clin Transl Sci 2018; 11:450-460. [PMID: 29768712 PMCID: PMC6132367 DOI: 10.1111/cts.12559] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/29/2018] [Indexed: 02/01/2023] Open
Abstract
While efficacy and safety data collected from randomized clinical trials are the evidentiary standard for determining market authorization, this alone may no longer be sufficient to address the needs of key stakeholders (regulators, providers, and payers) and guarantee long-term success of pharmaceutical products. There is a heightened interest from stakeholders on understanding the use of real-world evidence (RWE) to substantiate benefit-risk assessment and support the value of a new drug. This review provides an overview of real-world data (RWD) and related advances in the regulatory framework, and discusses their impact on clinical research and development. A framework for linking drug development decisions with the value proposition of the drug, utilizing pharmacokinetic-pharmacodynamic-pharmacoeconomic models, is introduced. The summary presented here is based on the presentations and discussion at the symposium entitled Innovation at the Intersection of Clinical Trials and Real-World Data to Advance Patient Care at the American Society for Clinical Pharmacology and Therapeutics (ASCPT) 2017 Annual Meeting.
Collapse
Affiliation(s)
| | - Lokesh Jain
- Quantitative Pharmacology and PharmacometricsMerck & Co., Inc.RahwayNew JerseyUSA
| | - Craig White
- Harvard PhD program in Health PolicyCambridgeMassachusettsUSA
| | - Vasu Chandrasekaran
- Center for Observational and Real World EvidenceMerck & Co., Inc.BostonMassachusettsUSA
| | - Aman Bhandari
- Center for Observational and Real World EvidenceMerck & Co., Inc.BostonMassachusettsUSA
| | - Dyfrig A. Hughes
- Centre for Health Economics and Medicines EvaluationBangor UniversityBangorGwyneddUK
| | - Pravin R. Jadhav
- Corporate ProjectsResearch & Development (R&D) InnovationOtsuka Pharmaceutical Development and Commercialization (OPDC)PrincetonNew JerseyUSA
| |
Collapse
|
94
|
Deng J, Jhandey A, Zhu X, Yang Z, Yik KFP, Zuo Z, Lam TN. In silico drug absorption tract: An agent-based biomimetic model for human oral drug absorption. PLoS One 2018; 13:e0203361. [PMID: 30169515 PMCID: PMC6118387 DOI: 10.1371/journal.pone.0203361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 08/20/2018] [Indexed: 11/26/2022] Open
Abstract
Background An agent-based modeling approach has been suggested as an alternative to traditional, equation-based modeling methods for describing oral drug absorption. It enables researchers to gain a better understanding of the pharmacokinetic (PK) mechanisms of a drug. This project demonstrates that a biomimetic agent-based model can adequately describe the absorption and disposition kinetics both of midazolam and clonazepam. Methods An agent-based biomimetic model, in silico drug absorption tract (ISDAT), was built to mimic oral drug absorption in humans. The model consisted of distinct spaces, membranes, and metabolic enzymes, and it was altogether representative of human physiology relating to oral drug absorption. Simulated experiments were run with the model, and the results were compared to the referent data from clinical equivalence trials. Acceptable similarity was verified by pre-specified criteria, which included 1) qualitative visual matching between the clinical and simulated concentration-time profiles, 2) quantitative similarity indices, namely, weighted root mean squared error (RMSE), and weighted mean absolute percentage error (MAPE) and 3) descriptive similarity which requires less than 25% difference between key PK parameters calculated by the clinical and the simulated concentration-time profiles. The model and its parameters were iteratively refined until all similarity criteria were met. Furthermore, simulated PK experiments were conducted to predict bioavailability (F). For better visualization, a graphical user interface for the model was developed and a video is available in Supporting Information. Results Simulation results satisfied all three levels of similarity criteria for both drugs. The weighted RMSE was 0.51 and 0.92, and the weighted MAPE was 5.99% and 8.43% for midazolam and clonazepam, respectively. Calculated PK parameter values, including area under the curve (AUC), peak plasma drug concentration (Cmax), time to reach Cmax (Tmax), terminal elimination rate constant (Kel), terminal elimination half life (T1/2), apparent oral clearance (CL/F), and apparent volume of distribution (V/F), were reasonable compared to the referent values. The predicted absolute oral bioavailability (F) was 44% for midazolam (literature reported value, 31–72%) and 93% (literature reported value, ≥ 90%) for clonazepam. Conclusion The ISDAT met all the pre-specified similarity criteria for both midazolam and clonazepam, and demonstrated its ability to describe absorption kinetics of both drugs. Therefore, the validated ISDAT can be a promising platform for further research into the use of similar in silico models for drug absorption kinetics.
Collapse
Affiliation(s)
- Jianyuan Deng
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Anika Jhandey
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Xiao Zhu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Zhibo Yang
- Department of Computer Science, Stony Brook University, Stony Brook, NY, United States of America
| | - Kin Fu Patrick Yik
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Zhong Zuo
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tai Ning Lam
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- * E-mail:
| |
Collapse
|
95
|
Opportunities and pitfalls in clinical proof-of-concept: principles and examples. Drug Discov Today 2018; 23:776-787. [PMID: 29406264 DOI: 10.1016/j.drudis.2018.01.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/11/2017] [Accepted: 01/19/2018] [Indexed: 12/13/2022]
Abstract
Clinical proof-of-concept trials crucially inform major resource deployment decisions. This paper discusses several mechanisms for enhancing their rigour and efficiency. The importance of careful consideration when using a surrogate endpoint is illustrated; situational effectiveness of run-in patient enrichment is explored; a versatile tool is introduced to ensure a strong pharmacological underpinning; the benefits of dose-titration are revealed by simulation; and the importance of adequately scheduled observations is shown. The general process of model-based trial design and analysis is described and several examples demonstrate the value in historical data, simulation-guided design, model-based analysis and trial adaptation informed by interim analysis.
Collapse
|
96
|
Gomeni R, Bressolle-Gomeni F, Fava M. Is It Time for Going Beyond the P-Value Paradigm With the Estimation of the Probability of Clinical Benefit as a Criterion for Assessing the Outcomes of a Clinical Trial? A Case Study in Patients With Major Depressive Disorder. J Clin Pharmacol 2018; 58:740-749. [PMID: 29372561 DOI: 10.1002/jcph.1074] [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: 11/02/2017] [Accepted: 12/05/2017] [Indexed: 11/07/2022]
Abstract
The conventional statistical methodologies for evaluating treatment effect are based on hypothesis testing (P-value). Alternative measurements of treatment effect have been proposed for anti-infective treatments using the probability of target attainment. A general framework is proposed to extend this methodology to other therapeutic areas. A disease trial model is used for estimating the probability of reaching a treatment effect associated with relevant clinical benefits, in complement to the evaluation of the probability of rejecting the null hypothesis. A case study is presented in depression, where disease status is evaluated using bounded clinical scores (Hamilton Depression Rating Scale), and detectable treatment effect is inversely proportional to placebo response. The β-regression approach is used to model Hamilton scale scores, and a placebo-related criterion is proposed for determining the clinical benefit. The probability of reaching a clinical benefit represents a reliable criterion for replacing the P-value paradigm in the assessment of the outcomes of clinical trials.
Collapse
Affiliation(s)
| | | | - Maurizio Fava
- Psychiatry Department, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
97
|
Morgan P, Brown DG, Lennard S, Anderton MJ, Barrett JC, Eriksson U, Fidock M, Hamrén B, Johnson A, March RE, Matcham J, Mettetal J, Nicholls DJ, Platz S, Rees S, Snowden MA, Pangalos MN. Impact of a five-dimensional framework on R&D productivity at AstraZeneca. Nat Rev Drug Discov 2018; 17:167-181. [DOI: 10.1038/nrd.2017.244] [Citation(s) in RCA: 269] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
98
|
Dunyak J, Mitchell P, Hamrén B, Helmlinger G, Matcham J, Stanski D, Al-Huniti N. Integrating dose estimation into a decision-making framework for model-based drug development. Pharm Stat 2018; 17:155-168. [PMID: 29322659 DOI: 10.1002/pst.1841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 09/11/2017] [Accepted: 10/10/2017] [Indexed: 12/12/2022]
Abstract
Model-informed drug discovery and development offers the promise of more efficient clinical development, with increased productivity and reduced cost through scientific decision making and risk management. Go/no-go development decisions in the pharmaceutical industry are often driven by effect size estimates, with the goal of meeting commercially generated target profiles. Sufficient efficacy is critical for eventual success, but the decision to advance development phase is also dependent on adequate knowledge of appropriate dose and dose-response. Doses which are too high or low pose risk of clinical or commercial failure. This paper addresses this issue and continues the evolution of formal decision frameworks in drug development. Here, we consider the integration of both efficacy and dose-response estimation accuracy into the go/no-go decision process, using a model-based approach. Using prespecified target and lower reference values associated with both efficacy and dose accuracy, we build a decision framework to more completely characterize development risk. Given the limited knowledge of dose response in early development, our approach incorporates a set of dose-response models and uses model averaging. The approach and its operating characteristics are illustrated through simulation. Finally, we demonstrate the decision approach on a post hoc analysis of the phase 2 data for naloxegol (a drug approved for opioid-induced constipation).
Collapse
|
99
|
Rouse R, Zineh I, Strauss DG. Regulatory Science - An Underappreciated Component of Translational Research. Trends Pharmacol Sci 2018; 39:225-229. [PMID: 29329784 DOI: 10.1016/j.tips.2017.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022]
Abstract
Translational science refers to translating basic scientific findings to practical application (i.e., 'bench-to-bedside'). An underappreciated aspect of translational science is regulatory science. Herein, we focus on the importance of regulatory science to facilitate development of innovative new drugs and optimize use of approved drugs, with a call for community participation.
Collapse
Affiliation(s)
- Rodney Rouse
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Issam Zineh
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - David G Strauss
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA.
| |
Collapse
|
100
|
Abbiati RA, Savoca A, Manca D. An engineering oriented approach to physiologically based pharmacokinetic and pharmacodynamic modeling. COMPUTER AIDED CHEMICAL ENGINEERING 2018. [DOI: 10.1016/b978-0-444-63964-6.00002-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|