1
|
Stamatopoulos K, Mistry N, Taskar K, Salem F, Butler JM, Tompson D. Physiologically Based Pharmacokinetic Modeling of Phosphate Prodrugs─Case Studies: Fostemsavir and Fostamatinib. Mol Pharm 2025; 22:2168-2181. [PMID: 40135517 DOI: 10.1021/acs.molpharmaceut.4c01362] [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] [Indexed: 03/27/2025]
Abstract
The aim of this work was to develop a physiologically based pharmacokinetic (PBPK) model for conversion of phosphate prodrugs to active drug via intestinal alkaline phosphatase (IAP) implementing a generalized modeling strategy. Fostemsavir and fostamatinib were chosen as model drugs since there is extensive clinical pharmacokinetic data following administration of oral formulations. LUA scripting was used to develop an "in vitro" to "in vivo" extrapolation of the conversion rate of prodrugs derived from Caco2 cell lines using an absolute IAP abundance approach. The Simcyp v23 platform was modified to generate a virtual population to reflect gastric emptying rates following administration of a moderate fat meal. The PBPK model predicted the results of three different extended-release (ER) tablets of fostemsavir under fasted and fed conditions as well as for powder in capsule and tablet immediate release (IR) formulations of fostamatinib. Retrospectively, the model was also able to assess the clinical relevance of the in vitro dissolution method to rate changes of different microcrystalline cellulose-based IR tablets of fostamatinib, observed in acidic media. All predictions were within 2-fold of the observed Cmax, AUC, and Tmax, with 81% being within 1.25-fold. The developed modeling strategy can be effectively adopted to increase the confidence of using PBPK modeling to prospectively assess the in vivo performance of phosphate prodrugs and support the development of optimal oral extended-release formulations for this class of drugs.
Collapse
Affiliation(s)
| | - Nena Mistry
- Biopharmaceutics, DPD, MDS, GSK, Ware SG12 0DP, U.K
| | - Kunal Taskar
- Drug Metabolism and Pharmacokinetics, GSK, Stevenage SG1 2NY, U.K
| | - Farzaneh Salem
- Drug Metabolism and Pharmacokinetics, GSK, Stevenage SG1 2NY, U.K
| | | | - Debra Tompson
- Clinical Pharmacology Modelling and Simulation, GSK, Medicines Research Centre, Stevenage SG1 2NY, U.K
| |
Collapse
|
2
|
Reppas C, Chorianopoulou C, Karkaletsi I, Dietrich S, Bakolia A, Vertzoni M. Simulation of Antral Conditions for Estimating Drug Apparent Equilibrium Solubility after a High-Calorie, High-Fat Meal. Mol Pharm 2025; 22:871-881. [PMID: 39811984 PMCID: PMC11795529 DOI: 10.1021/acs.molpharmaceut.4c01038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025]
Abstract
The simulation of antral conditions for estimating drug apparent equilibrium solubility after a high-calorie, high-fat meal is challenging. In this study, (1) we measured the apparent equilibrium solubility of two model lipophilic drugs, ketoconazole and danazol, in antral aspirates collected at various time points after a minced high-calorie, high-fat meal and a glass of water 30 min after initiation of meal administration, and we designated one point estimate for ketoconazole and one point estimate for danazol; (2) we evaluated the usefulness of FeSSGF-V2 and FEDGAS pH = 3 in reproducing the two point estimates; (3) we evaluated potential compositions of FeSSGF-V3 that simulate the pH, the buffer capacity toward both less acidic and more acidic values, and the antral lipid and protein contents with easily accessible, commercially available products, and (4) we identified the most useful composition of FeSSGF-V3 for reproducing the two point estimates. For both model drugs, apparent solubility in FeSSGF-V2 and in FEDGAS pH 3 deviated substantially from the corresponding point estimate. For FeSSGF-V3, hydrochloric acid, acetates, and FEDGASbuffer pH 3 were evaluated for regulating the pH and buffer capacity, FEDGASgel was used for simulating the lipid content, and Régilait skimmed milk powder was used for simulating the protein content. Level III FeSSGF-V3 prepared with hydrochloric acid, 6.1% (w/v) Régilait, and 2.83% (w/v) FEDGASgel, i.e., one-sixth of FEDGASgel concentration in FEDGAS pH 3, was comparatively the most useful medium for point estimating ketoconazole and danazol apparent solubility in antral contents after water administration in the fed state, induced as requested by regulatory authorities in oral drug bioavailability studies. Level III FeSSGF-V3 prepared by using hydrochloric acid as the principal pH controlling species could be useful in the evaluation of food effects on drug absorption with in silico physiologically based biopharmaceutics modeling approaches and, also, with biorelevant in vitro methodologies.
Collapse
Affiliation(s)
- Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou 15771, Greece
| | | | - Ioanna Karkaletsi
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou 15771, Greece
| | - Shirin Dietrich
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou 15771, Greece
| | - Andriani Bakolia
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou 15771, Greece
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou 15771, Greece
| |
Collapse
|
3
|
Taheri A, Almasri R, Wignall A, Schultz HB, Elz AS, Ariaee A, Bremmell KE, Joyce P, Prestidge CA. Enhancing the pharmacokinetics of abiraterone acetate through lipid-based formulations: addressing solubility and food effect challenges. Drug Deliv Transl Res 2024:10.1007/s13346-024-01755-y. [PMID: 39614037 DOI: 10.1007/s13346-024-01755-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
Abiraterone acetate, a prodrug of abiraterone, is an effective antiandrogen for treating metastatic prostate cancer. However, its poor aqueous solubility restricts oral bioavailability to under 10% in fasted conditions. Additionally, its pharmacokinetics are significantly influenced by food intake, leading to variable exposure that can impact treatment safety and efficacy. To overcome these challenges, we developed a series of lipid-based formulations aimed at reducing food effects and enhancing the fasted bioavailability of abiraterone acetate by incorporating the drug into colloidal delivery systems. Medium- and long-chain self-nanoemulsifying drug delivery systems (MC-SNEDDS and LC-SNEDDS) were formulated with abiraterone acetate loading at 80% of their respective preconcentrate equilibrium solubility. In-vitro gastrointestinal lipolysis experiments demonstrated that the SNEDDS formulations increased drug solubilisation by over 6-fold compared to pure abiraterone acetate and over 2-fold compared to the reference product after 60 min in the intestinal environment. In-vivo pharmacokinetic studies in rats revealed that both MC-SNEDDS and LC-SNEDDS formulations, along with their enteric-coated (EC) forms, exhibited enhanced bioavailability, with EC-LC-SNEDDS providing the highest performance, demonstrating a 7.32-fold increase in abiraterone exposure compared to the reference. Strong correlations were observed between in-vitro solubilisation and in-vivo AUC0 - 8 h (R2 = 0.980) and Cmax (R2 = 0.925). In-vivo pharmacokinetic studies in pigs demonstrated that EC-LC-SNEDDS improved drug systemic exposure in fasted conditions and mitigated positive food effects, showing a fed-to-fasted AUC0 - 8 h ratio of 108% compared to 334% with the reference. The developed lipid-based formulations hold promise in overcoming the pharmacokinetic challenges associated with abiraterone, potentially offering improved outcomes for patients.
Collapse
Affiliation(s)
- Ali Taheri
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Ruba Almasri
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Anthony Wignall
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Hayley B Schultz
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Aurelia S Elz
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Amin Ariaee
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Kristen E Bremmell
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Paul Joyce
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Clive A Prestidge
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia.
| |
Collapse
|
4
|
Heimbach T, Musuamba Tshinanu F, Raines K, Borges L, Kijima S, Malamatari M, Moody R, Veerasingham S, Seo P, Turner D, Fang L, Stillhart C, Bransford P, Ren X, Patel N, Sperry D, Chen H, Rostami-Hodjegan A, Lukacova V, Sun D, Nguefack JF, Carducci T, Grimstein M, Pepin X, Jamei M, Stamatopoulos K, Li M, Sanghavi M, Tannergren C, Mandula H, Zhao Z, Ju TR, Wagner C, Arora S, Wang M, Rullo G, Mitra A, Kollipara S, Chirumamilla SK, Polli JE, Mackie C. PBBM Considerations for Base Models, Model Validation, and Application Steps: Workshop Summary Report. Mol Pharm 2024; 21:5353-5372. [PMID: 39348508 PMCID: PMC11539057 DOI: 10.1021/acs.molpharmaceut.4c00758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 10/02/2024]
Abstract
The proceedings from the 30th August 2023 (Day 2) of the workshop "Physiologically Based Biopharmaceutics Models (PBBM) Best Practices for Drug Product Quality: Regulatory and Industry Perspectives" are provided herein. Day 2 covered PBBM case studies from six regulatory authorities which provided considerations for model verification, validation, and application based on the context of use (COU) of the model. PBBM case studies to define critical material attribute (CMA) specification settings, such as active pharmaceutical ingredient (API) particle size distributions (PSDs) were shared. PBBM case studies to define critical quality attributes (CQAs) such as the dissolution specification setting or to define the bioequivalence safe space were also discussed. Examples of PBBM using the credibility assessment framework, COU and model risk assessment, as well as scientific learnings from PBBM case studies are provided. Breakout session discussions highlighted current trends and barriers to application of PBBMs including: (a) PBBM credibility assessment framework and level of validation, (b) use of disposition parameters in PBBM and points to consider when iv data are not available, (c) conducting virtual bioequivalence trials and dealing with variability, (d) model acceptance criteria, and (e) application of PBBMs for establishing safe space and failure edges.
Collapse
Affiliation(s)
- Tycho Heimbach
- Pharmaceutical
Sciences and Clinical Supply, Merck &
Co., Inc., Rahway, New Jersey 07065, United States
| | - Flora Musuamba Tshinanu
- Belgian
Federal Agency for Medicines and Health Products, Galileelaan 5/03, Brussels 1210, Belgium
| | - Kimberly Raines
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Luiza Borges
- ANVISA, SIA Trecho 5 − Guará, Brasília, DF 71205-050, Brazil
| | - Shinichi Kijima
- Office of
New Drug V, Pharmaceuticals and Medical
Devices Agency (PMDA), Tokyo 100-0013, Japan
| | - Maria Malamatari
- Medicines
& Healthcare Products Regulatory Agency, 10 S Colonnade, London SW1W 9SZ, U.K.
| | - Rebecca Moody
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Shereeni Veerasingham
- Pharmaceutical
Drugs Directorate (PDD), Health Canada, 1600 Scott St, Ottawa, Ontario K1A 0K9, Canada
| | - Paul Seo
- Office of
Clinical Pharmacology (OCP), Office of Translational Sciences (OTS),
Center for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| | - David Turner
- Certara
Predictive
Technologies, Level 2-Acero, Simcyp Ltd, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Lanyan Fang
- Division
of Quantitative Methods and Modeling (DQMM), Office of Research and
Standards (ORS), Office of Generic Drugs (OGD), Center for Drug Evaluation
and Research (CDER), Food and Drug Administration
(FDA), Silver Spring, Maryland 20903-1058, United States
| | - Cordula Stillhart
- Pharmaceutical
R&D, F. Hoffmann-La Roche Ltd., Basel CH-4070, Switzerland
| | - Philip Bransford
- Data and
Computational Sciences, Vertex Pharmaceuticals,
Inc., Boston, Massachusetts 02210, United States
| | - Xiaojun Ren
- PK Sciences/Translational
Medicine, BioMedical Research, Novartis, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Nikunjkumar Patel
- Certara
Predictive
Technologies, Level 2-Acero, Simcyp Ltd, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - David Sperry
- Eli Lilly
and Company, Lilly Corporate
Center, Indianapolis, Indiana 46285, United States
| | - Hansong Chen
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Amin Rostami-Hodjegan
- Certara
Predictive
Technologies, Level 2-Acero, Simcyp Ltd, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
- Centre
for Applied Pharmacokinetic Research, University
of Manchester, Stopford Building, Oxford Road, Manchester M139PT, U.K.
| | - Viera Lukacova
- Simulations
Plus Inc., 42505 10th Street West, Lancaster, California 93534, United States
| | - Duxin Sun
- The University
of Michigan, North Campus Research Complex
(NCRC), 1600 Huron Parkway, Ann Arbor, Michigan 48109, United States
| | - Jean-Flaubert Nguefack
- Head of
Biopharmacy Team, Montpellier, Synthetics Platform, Global CMC, Sanofi, Paris 75008, France
| | - Tessa Carducci
- Analytical
Commercialization Technology, Merck &
Co., Inc., 126 E. Lincoln
Ave, Rahway, New Jersey 07065, United States
| | - Manuela Grimstein
- Office of
Clinical Pharmacology (OCP), Office of Translational Sciences (OTS),
Center for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| | - Xavier Pepin
- Simulations
Plus Inc., 42505 10th Street West, Lancaster, California 93534, United States
| | - Masoud Jamei
- Certara
Predictive
Technologies, Level 2-Acero, Simcyp Ltd, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | | | - Min Li
- Office of
Clinical Pharmacology (OCP), Office of Translational Sciences (OTS),
Center for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| | - Maitri Sanghavi
- Certara
Predictive
Technologies, Level 2-Acero, Simcyp Ltd, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - Christer Tannergren
- Biopharmaceutics
Science, New Modalities & Parenteral Product Development, Pharmaceutical
Technology & Development, Operations, AstraZeneca, Gothenburg 43183, Sweden
| | - Haritha Mandula
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Zhuojun Zhao
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Tzuchi Rob Ju
- Analytical
R&D, AbbVie Inc., 1 North Waukegan Road, North
Chicago, Illinois 60064, United States
| | - Christian Wagner
- Global
Drug Product Development, Global CMC Development, the Healthcare Business of Merck KGaA, Darmstadt 64293, Germany
| | - Sumit Arora
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Michael Wang
- Pharmaceutical
Sciences and Clinical Supply, Merck &
Co., Inc., Rahway, New Jersey 07065, United States
| | - Gregory Rullo
- Regulatory
CMC, AstraZeneca, 1 Medimmune Way, Gaithersburg, Maryland 20878, United States
| | - Amitava Mitra
- Clinical
Pharmacology, Kura Oncology Inc, Boston, Massachusetts 02210, United States
| | - Sivacharan Kollipara
- Biopharmaceutics
Group, Global Clinical Management, Integrated Product Development
Organization (IPDO), Dr. Reddy’s
Laboratories Ltd., Bachupally,
Medchal Malkajgiri District, Hyderabad, 500 090 Telangana, India
| | - Siri Kalyan Chirumamilla
- Certara
Predictive
Technologies, Level 2-Acero, Simcyp Ltd, 1 Concourse Way, Sheffield S1 2BJ, United Kingdom
| | - James E. Polli
- School
of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
| | - Claire Mackie
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| |
Collapse
|
5
|
Danielak D, Gajda M, Bołtromiuk T, Sulikowska K, Kubiak B, Romański M. Drug dissolution and transit in a heterogenous gastric chyme after fed administration: Semi-mechanistic modeling and simulations for an immediate-release and orodispersible tablets containing a poorly soluble drug. Eur J Pharm Biopharm 2024; 200:114341. [PMID: 38795785 DOI: 10.1016/j.ejpb.2024.114341] [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: 02/16/2024] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 05/28/2024]
Abstract
Mathematical models that treat the fed stomach content as a uniform entity emptied with a constant rate may not suffice to explain pharmacokinetic profiles recorded in clinical trials. In reality, phenomena such as the Magenstrasse or chyme areas of different pH and viscosity, play an important role in the intragastric drug dissolution and its transfer to the intestine. In this study, we investigated the data gathered in the bioequivalence trial between an immediate-release tablet (Reference) and an orally dispersible tablet (Test) with a poorly soluble weak base drug administered with or without water after a high-fat high-calorie breakfast. Maximum concentrations (Cmax) were significantly greater after administering the Reference product than the Test tablets, despite similar in vitro dissolution profiles. To explain this difference, we constructed a novel semi-mechanistic IVIVP model including a heterogeneous gastric chyme. The drug dissolution in vivo was modeled from the in vitro experiments in biorelevant media simulating gastric and intestinal fluids in the fed state (FEDGAS and FeSSIF). The key novelty of the model was separating the stomach contents into two compartments: isolated chyme (the viscous food content) that carries the drug slowly, and aq_chyme open for rapid Magenstrasse-like routes of drug transit. Drug distribution between these two compartments was both formulation- and administration-dependent, and recognized the respective drug fractions from the clinical pharmacokinetic data. The model's assumption about the nonuniform mixing of the API with the chyme, influencing differential drug dissolution and transit kinetics, led to simulating plasma concentration profiles that reflected well the variability observed in the clinical trial. The model indicated that, after administration, the Reference product mixes to a greater extent with aq_chyme, where the released drug dissolves better and transfers faster to the intestine. In conclusion, this novel approach underlines that diverse gastric emptying of different oral dosage forms may significantly impact pharmacokinetics and affect the outcomes of bioequivalence trials.
Collapse
Affiliation(s)
- Dorota Danielak
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Rokietnicka 3 St, 60-806, Poznań, Poland.
| | - Maciej Gajda
- Adamed Pharma S.A., Pieńków, Mariana Adamkiewicza 6A, 05-152 Czosnów, Poland.
| | - Tomasz Bołtromiuk
- Adamed Pharma S.A., Pieńków, Mariana Adamkiewicza 6A, 05-152 Czosnów, Poland.
| | | | - Bartłomiej Kubiak
- Adamed Pharma S.A., Pieńków, Mariana Adamkiewicza 6A, 05-152 Czosnów, Poland.
| | - Michał Romański
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, Rokietnicka 3 St, 60-806, Poznań, Poland.
| |
Collapse
|
6
|
Kollipara S, Martins FS, Sanghavi M, Santos GML, Saini A, Ahmed T. Role of Physiologically Based Biopharmaceutics Modeling (PBBM) in Fed Bioequivalence Study Waivers: Regulatory Outlook, Case Studies and Future Perspectives. J Pharm Sci 2024; 113:345-358. [PMID: 38043684 DOI: 10.1016/j.xphs.2023.11.030] [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: 09/14/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Over the past few decades, physiologically based biopharmaceutics modeling (PBBM) has demonstrated its utility in both new drug and generic product development. Applications of PBBM for fed bioequivalence study waivers is an upcoming area. Recently Innovation & Quality (IQ) consortium demonstrated utility of PBBM to avoid repeat food effect studies for new drugs. In the similar lines, the current manuscript aims to discuss role of PBBM in generic fed bioequivalence study waivers. Generic industry practices related to PBBM model development to predict fed bioequivalence was portrayed with special emphasis on fed bio-predictive media. Media that can simulate fed bioequivalence study outcome were discussed from practical perspective. In-depth analysis, collating the data from 36 products was performed to understand predictability of PBBM for fed bioequivalence. Cases where PBBM was successful to predict fed bioequivalence was correlated with BCS class, formulation category and type of food effect. Further, two case studies were presented wherein fed bioequivalence study waiver obtained with PBBM approach. Lastly, future direction in terms of fed bioequivalence study waivers, regulatory perspectives and best practices for PBBM were portrayed. Overall, this article paves a way to utilize PBBM for generic fed bioequivalence study waivers.
Collapse
Affiliation(s)
- Sivacharan Kollipara
- Biopharmaceutics Group, Global Clinical Management, Dr. Reddy's Laboratories Ltd., Integrated Product Development Organization (IPDO), Bachupally, Medchal Malkajgiri District, Hyderabad, Telangana 500 090, India
| | | | - Maitri Sanghavi
- Biopharmaceutics & Clinical Development, Pharmaceutical Technology Center (PTC), Zydus Lifesciences Ltd., NH-8A, Sarkhej-Bavla Highway, Moraiya, Ahmedabad-382210, Gujrat, India
| | | | - Anuj Saini
- Biopharmaceutics & Clinical Development, Pharmaceutical Technology Center (PTC), Zydus Lifesciences Ltd., NH-8A, Sarkhej-Bavla Highway, Moraiya, Ahmedabad-382210, Gujrat, India
| | - Tausif Ahmed
- Biopharmaceutics Group, Global Clinical Management, Dr. Reddy's Laboratories Ltd., Integrated Product Development Organization (IPDO), Bachupally, Medchal Malkajgiri District, Hyderabad, Telangana 500 090, India.
| |
Collapse
|
7
|
Stamatopoulos K, Mistry N, Fotaki N, Turner DB, Swift B. Physiologically Based Biopharmaceutics Model (PBBM) of Minimally Absorbed Locally Acting Drugs in the Gastrointestinal Tract-Case Study: Tenapanor. Pharmaceutics 2023; 15:2726. [PMID: 38140067 PMCID: PMC10747343 DOI: 10.3390/pharmaceutics15122726] [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: 10/11/2023] [Revised: 11/23/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
A physiologically based biopharmaceutics model (PBBM) was developed to predict stool and urine sodium content in response to tenapanor administration in healthy subjects. Tenapanor is a minimally absorbed small molecule that inhibits the sodium/hydrogen isoform 3 exchanger (NHE3). It is used to treat irritable bowel syndrome with constipation (IBS-C). Its mode of action in the gastrointestinal tract reduces the uptake of sodium, resulting in an increase in water secretion in the intestinal lumen and accelerating intestinal transit time. The strategy employed was to perform drug-drug interaction (DDI) modelling between sodium and tenapanor, with sodium as the "victim" administered as part of daily food intake and tenapanor as the "perpetrator" altering sodium absorption. Food effect was modelled, including meal-induced NHE3 activity using sodium as an inducer by normalising the induction kinetics of butyrate to sodium equivalents. The presented model successfully predicted both urine and stool sodium content in response to tenapanor dosed in healthy subjects (within 1.25-fold error) and provided insight into the clinical observations of tenapanor dosing time relative to meal ingestion. The PBBM model was applied retrospectively to assess the impact of different forms of tenapanor (free base vs. HCl salt) on its pharmacodynamic (PD) effect. The developed modelling strategy can be effectively adopted to increase confidence in using PBBM models for the prediction of the in vivo behaviour of minimally absorbed, locally acting drugs in the gastrointestinal tract, when other approaches (e.g., biomarkers or PD data) are not available.
Collapse
Affiliation(s)
| | - Nena Mistry
- Biopharmaceutics, DPD, MDS, GSK, Ware SG12 0DP, UK;
| | - Nikoletta Fotaki
- Centre for Therapeutic Innovation, Department of Life Sciences, University of Bath, Claverton Down, Bath BA2 7AY, UK;
| | | | | |
Collapse
|
8
|
Karnati P, Murthy A, Gundeti M, Ahmed T. Modelling Based Approaches to Support Generic Drug Regulatory Submissions-Practical Considerations and Case Studies. AAPS J 2023; 25:63. [PMID: 37353655 DOI: 10.1208/s12248-023-00831-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 06/03/2023] [Indexed: 06/25/2023] Open
Abstract
Model informed drug development (MiDD) is useful to predict in vivo exposure of drugs during various stages of the drug development process. This approach employs a variety of quantitative tools to assess the risks during the drug development process. One important tool in the MiDD tool kit is the Physiologically Based Pharmacokinetic Modelling (PBPK). This tool is extensively used to reduce the development cost and to accelerate the access of medicines to the patients. In this work, we provide an overview of PBPK modelling approaches in the generic drug development process, with a special emphasis on the bio-waiver applications. We describe herein approaches and common pitfalls while submitting model based justifications as a response to the regulatory deficiencies during the generic drug development process. With some in-house case studies, we have attempted to provide a clear path for PBPK model based justifications for bio-waivers. With this review, the gap between theoretical knowledge and practical application of modelling and simulation tools for generic drug product development could be potentially reduced.
Collapse
Affiliation(s)
- Prajwala Karnati
- Biopharmaceutics Department, Global Clinical Management, Dr. Reddy's Laboratories Ltd., Hyderabad, India
| | - Aditya Murthy
- Biopharmaceutics Department, Global Clinical Management, Dr. Reddy's Laboratories Ltd., Hyderabad, India
| | - Manoj Gundeti
- Biopharmaceutics Department, Global Clinical Management, Dr. Reddy's Laboratories Ltd., Hyderabad, India
| | - Tausif Ahmed
- Biopharmaceutics Department, Global Clinical Management, Dr. Reddy's Laboratories Ltd., Hyderabad, India.
| |
Collapse
|