1
|
Malik P, Mian P. Physiologically Based Pharmacokinetic Modeling to Refine Dosing of Posaconazole in Young Children. Clin Ther 2025; 47:261-270. [PMID: 39827022 DOI: 10.1016/j.clinthera.2024.12.018] [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: 08/16/2024] [Revised: 12/07/2024] [Accepted: 12/30/2024] [Indexed: 01/22/2025]
Abstract
PURPOSE Posaconazole is a broad-spectrum antifungal for treating and preventing invasive fungal infections (IFIs) in immunocompromised individuals, including children as young as 2 years. Available in delayed-release (DR) oral suspension, intravenous formulation, and older immediate-release (IR) formulation (off-label in younger children), dosing harmonization across age groups and formulations remains inconsistent. This inconsistency arises from the unique physiology of young children and posaconazole's pH-dependent absorption. Limited pharmacokinetic (PK) data for children under 2 years complicates dosing, as absorption, distribution, metabolism, and excretion processes are underdeveloped and age-dependent. This work aims to harmonize pediatric dosing for children aged 2 to 7 years and extend dosing guidance for those aged 6 months to 2 years using physiologically-based PK (PBPK) modeling. METHODS An adult PBPK model was created using posaconazole's physicochemical properties and ADME characteristics with virtual populations from PK-Sim. Calibrated with single-dose data from healthy subjects, the model was verified by predicting PK following multiple doses in adults at risk for IFIs. The model was then scaled to children, accounting for developmental anatomy and physiology, including UGT1A4 ontogeny. The pediatric model was evaluated against observed data from children aged 2 to 7 years. Simulations were conducted to harmonize dosing across formulations and extend dosing to children as young as 6 months, acknowledging standard plasma concentration targets for treatment of IFIs (1000 ng/mL) as well as prophylaxis (700 ng/mL). FINDINGS The pediatric model adequately captured observed PK data from literature following all three formulations. The IR oral suspension is impractical and likely subtherapeutic for most children under 7 years due to solubility limits. Intravenous doses of 11-13 mg/kg once daily (QD) may be optimal for treatment, and 8 to 9 mg/kg QD for prophylaxis, varying by age. Oral DR suspension doses of 12 to 14 mg/kg QD for treatment and 8.5 to 10 mg/kg QD for prophylaxis may be optimal, also age-dependent. Dividing the total daily dose by a factor of 0.7 and administering twice daily can achieve similar trough levels. IMPLICATIONS PBPK modeling for posaconazole bridges the gap between PK principles and clinical practice, potentially improving therapeutic outcomes and minimizing risks associated with inadequate dosing in pediatric patients.
Collapse
Affiliation(s)
- Paul Malik
- Ionis Pharmaceuticals Inc, Carlsbad, California.
| | - Paola Mian
- University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| |
Collapse
|
2
|
Ng T, Kou D. Evaluation of the impact of mucin on supersaturation and permeation of BCS class 2 basic drugs. J Pharm Sci 2024; 113:3272-3278. [PMID: 39179030 DOI: 10.1016/j.xphs.2024.08.016] [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: 07/18/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
This study evaluated the impact of mucin on supersaturation and permeation of BCS Class 2 basic drugs in a pH-shift, 2-stage model using three model compounds, dipyridamole, ricobendazole, and Compound A. The three compounds showed various degrees of supersaturation (DoS) in Stage 2 and modest to no increases in flux with the presence of mucin in the dissolution media. Mucin's impact on DoS and flux, if any, appeared to be compound specific and possibly related to its pKa and ionization state. Overall, the increases in supersaturation and permeation due to mucin ranged from modest to minimal for the three model compounds under the conditions tested. The pH-shift model using MacroFLUX was able to monitor gastric and intestinal dissolution and simultaneously assess the effect of intestinal mucin on supersaturation and flux.
Collapse
Affiliation(s)
- Tania Ng
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Dawen Kou
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
3
|
Pepin X, Arora S, Borges L, Cano-Vega M, Carducci T, Chatterjee P, Chen G, Cristofoletti R, Dallmann A, Delvadia P, Dressman J, Fotaki N, Gray E, Heimbach T, Holte Ø, Kijima S, Kotzagiorgis E, Lennernäs H, Lindahl A, Loebenberg R, Mackie C, Malamatari M, McAllister M, Mitra A, Moody R, Mudie D, Musuamba Tshinanu F, Polli JE, Rege B, Ren X, Rullo G, Scherholz M, Song I, Stillhart C, Suarez-Sharp S, Tannergren C, Tsakalozou E, Veerasingham S, Wagner C, Seo P. Parameterization of Physiologically Based Biopharmaceutics Models: Workshop Summary Report. Mol Pharm 2024; 21:3697-3731. [PMID: 38946085 PMCID: PMC11304397 DOI: 10.1021/acs.molpharmaceut.4c00526] [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: 05/13/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
This Article shares the proceedings from the August 29th, 2023 (day 1) workshop "Physiologically Based Biopharmaceutics Modeling (PBBM) Best Practices for Drug Product Quality: Regulatory and Industry Perspectives". The focus of the day was on model parametrization; regulatory authorities from Canada, the USA, Sweden, Belgium, and Norway presented their views on PBBM case studies submitted by industry members of the IQ consortium. The presentations shared key questions raised by regulators during the mock exercise, regarding the PBBM input parameters and their justification. These presentations also shed light on the regulatory assessment processes, content, and format requirements for future PBBM regulatory submissions. In addition, the day 1 breakout presentations and discussions gave the opportunity to share best practices around key questions faced by scientists when parametrizing PBBMs. Key questions included measurement and integration of drug substance solubility for crystalline vs amorphous drugs; impact of excipients on apparent drug solubility/supersaturation; modeling of acid-base reactions at the surface of the dissolving drug; choice of dissolution methods according to the formulation and drug properties with a view to predict the in vivo performance; mechanistic modeling of in vitro product dissolution data to predict in vivo dissolution for various patient populations/species; best practices for characterization of drug precipitation from simple or complex formulations and integration of the data in PBBM; incorporation of drug permeability into PBBM for various routes of uptake and prediction of permeability along the GI tract.
Collapse
Affiliation(s)
- Xavier Pepin
- Regulatory
Affairs, Simulations Plus Inc., 42505 10th Street West, Lancaster, California 93534-7059, United States
| | - Sumit Arora
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Luiza Borges
- ANVISA, SIA Trecho 5́, Guara, Brasília, Federal District 71205-050, Brazil
| | - Mario Cano-Vega
- Drug
Product Technologies, Amgen Inc., Thousand Oaks, California 91320-1799, United
States
| | - Tessa Carducci
- Analytical
Commercialization Technology, Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, New Jersey 07065, United States
| | - Parnali Chatterjee
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Grace Chen
- Takeda
Development Center Americas Inc., 300 Shire Way, Lexington, Massachusetts 02421, United States
| | - Rodrigo Cristofoletti
- College
of Pharmacy, University of Florida, 6550 Sanger Rd., Orlando, Florida 32827, United States
| | - André Dallmann
- Bayer
HealthCare SAS, 59000 Lille, France, on behalf of Bayer
AG, Pharmacometrics/Modeling and Simulation, Systems Pharmacology
& Medicine, PBPK, Leverkusen, Germany
| | - Poonam Delvadia
- Office
of Translational Science, Office of Clinical Pharmacology (OCP), Center
for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, Frankfurt am Main 60596, Germany
| | - Nikoletta Fotaki
- University of Bath, Claverton Down, Bath BA2
7AY, United Kingdom
| | - Elizabeth Gray
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Tycho Heimbach
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Øyvind Holte
- Norwegian Medical Products Agency, Oslo 0213, Norway
| | - Shinichi Kijima
- Office
of New Drug V, Pharmaceuticals and Medical
Devices Agency (PMDA), Tokyo 100-0013, Japan
| | - Evangelos Kotzagiorgis
- European Medicines Agency (EMA), Domenico Scarlattilaan 6, Amsterdam 1083 HS, The Netherlands
| | - Hans Lennernäs
- Translational
Drug Discovery and Development, Department of Pharmaceutical Bioscience, Uppsala University, Uppsala 751 05, Sweden
| | | | - Raimar Loebenberg
- Faculty
of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmontonton T6G 2E1, Canada
| | - Claire Mackie
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Maria Malamatari
- Medicines & Healthcare Products Regulatory Agency, 10 S Colonnade, London SW1W 9SZ, United Kingdom
| | - Mark McAllister
- Global
Biopharmaceutics, Drug Product Design, Pfizer, Sandwich CT13 9NJ, United Kingdom
| | - Amitava Mitra
- Clinical
Pharmacology, Kura Oncology Inc., Boston, Massachusetts 02210, United States
| | - 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
| | - Deanna Mudie
- Global
Research and Development, Small Molecules, Lonza, 63045 NE Corporate
Pl., Bend, Oregon 97701, United States
| | - Flora Musuamba Tshinanu
- Belgian Federal Agency for Medicines and Health Products, Galileelaan 5/03, Brussel 1210, Belgium
| | - James E. Polli
- School
of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
| | - Bhagwant Rege
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Xiaojun Ren
- PK
Sciences/Translational Medicine, BioMedical Research, Novartis, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Gregory Rullo
- Regulatory
CMC, AstraZeneca, 1 Medimmune Way, Gaithersburg, Maryland 20878, United States
| | - Megerle Scherholz
- Pharmaceutical
Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Ivy Song
- Takeda
Development Center Americas Inc., 300 Shire Way, Lexington, Massachusetts 02421, United States
| | - Cordula Stillhart
- Pharmaceutical
R&D, F. Hoffmann-La Roche Ltd., Basel 4070, Switzerland
| | - Sandra Suarez-Sharp
- Regulatory
Affairs, Simulations Plus Inc., 42505 10th Street West, Lancaster, California 93534-7059, United States
| | - Christer Tannergren
- Biopharmaceutics
Science, New Modalities & Parenteral Product Development, Pharmaceutical
Technology & Development, Operations, AstraZeneca, Gothenburg 431 50, Sweden
| | - Eleftheria Tsakalozou
- Division
of Quantitative Methods and Modeling, Office of Research and Standards,
Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20903-1058, United
States
| | - Shereeni Veerasingham
- Pharmaceutical
Drugs Directorate (PDD), Health Canada, 1600 Scott St., Ottawa K1A 0K9, Canada
| | - Christian Wagner
- Global
Drug Product Development, Global CMC Development, the Healthcare Business of Merck KGaA, Darmstadt D-64293, Germany
| | - Paul Seo
- Office
of Translational Science, Office of Clinical Pharmacology (OCP), Center
for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| |
Collapse
|
4
|
Dietrich S, Ceulemans J, Hermans E, Argyropoulos T, Goumas K, Vertzoni M, Reppas C. Understanding the Conditions Under Which Drugs are Transferred from the Stomach Through the Upper Small Intestine After a High-Calorie, High-Fat Meal. J Pharm Sci 2024; 113:1546-1554. [PMID: 38218315 DOI: 10.1016/j.xphs.2024.01.001] [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: 12/07/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 01/15/2024]
Abstract
Information on the conditions under which drugs are transferred from the stomach through the upper small intestine after a high-calorie, high-fat meal is very limited. To simulate the drug presence after disintegration and arrival in the antral region, paracetamol solution and Sporanox® amorphous solid dispersion pellets at two dose levels were administered to the antrum of 8 healthy adults 30 min after administration of a high-calorie, high-fat meal on a crossover basis. The overall median buffer capacity of antral contents was estimated to be 18.0 and 24.0 mmol/ml/ΔpH when titrating with NaOH and HCl, respectively. The corresponding values for the contents of upper the small intestine were 14.0 and 16.8 mmol/ml/ΔpH, respectively. The drug transfer process from the antrum through the upper small intestine occurred with apparent first-order kinetics. The best estimate for the antral emptying half-life was 39min and 45min for paracetamol and itraconazole, respectively, the apparent volume of contents of the upper small intestine was more than double compared with previously reported values in the fasted state, the half-life of drug elimination from the upper small intestine was similar to recent estimates for highly permeable drugs in the fasted state, and the apparent volume of antral contents during the first couple of hours post drug administration was 303mL. Information collected in this study could increase the reliability of in silico and/or in vitro modelling approaches applied in clinical drug development.
Collapse
Affiliation(s)
- Shirin Dietrich
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece; Pharmaceutical and Material Sciences, Pharmaceutical Product Development and Supply, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Jens Ceulemans
- Pharmaceutical and Material Sciences, Pharmaceutical Product Development and Supply, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Eline Hermans
- Pharmaceutical and Material Sciences, Pharmaceutical Product Development and Supply, Janssen Pharmaceutica NV, Beerse, Belgium
| | | | - Konstantinos Goumas
- Department of Gastroenterology, Red Cross Hospital of Athens, Athens, Greece
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece
| | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece.
| |
Collapse
|
5
|
van Aken GA. Computer modeling of digestive processes in the alimentary tract and their physiological regulation mechanisms: closing the gap between digestion models and in vivo behavior. Front Nutr 2024; 11:1339711. [PMID: 38606020 PMCID: PMC11007706 DOI: 10.3389/fnut.2024.1339711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/15/2024] [Indexed: 04/13/2024] Open
Abstract
Introduction A model has been developed for in silico simulation of digestion and its physiological feedback mechanisms. Methods The model is based on known physiology described in the literature and is able to describe the complexity of many simultaneous processes related to food digestion. Results Despite the early stage of development of the model, it already encompasses a large number of processes that occur simultaneously, enabling the prediction of a large number of post-prandial physiological markers, which can be highly functional in combination with in vitro, organ-on-a-chip and digital twin models purposed to measure the physiological properties of organs and to predict the effect of adjusted food composition in normal and diseased states. Discussion Input from and collaboration between science fileds is needed to further develop and refine the model and to connect with in vitro, in vivo, and ex vivo (organ-on-a-chip) models.
Collapse
|
6
|
Holzem FL, Petrig Schaffland J, Brandl M, Bauer-Brandl A, Stillhart C. Using molecularly dissolved drug concentrations in PBBMs improves the prediction of oral absorption from supersaturating formulations. Eur J Pharm Sci 2024; 194:106703. [PMID: 38224722 DOI: 10.1016/j.ejps.2024.106703] [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: 10/23/2023] [Revised: 12/21/2023] [Accepted: 01/12/2024] [Indexed: 01/17/2024]
Abstract
Predicting the absorption of drugs from enabling formulations is still challenging due to the limited capabilities of standard physiologically based biopharmaceutics models (PBBMs) to capture complex absorption processes. Amongst others, it is often assumed that both, molecularly and apparently dissolved drug in the gastrointestinal lumen are prone to absorption. A recently introduced method for measuring concentrations of molecularly dissolved drug in a dynamic in vitro dissolution setup using microdialysis has opened new opportunities to test this hypothesis and refine mechanistic PBBM approaches. In the present study, we compared results of PBBMs that used either molecularly or apparently dissolved concentrations in the simulated gastrointestinal lumen as input parameters. The in vitro dissolution data from three supersaturating formulations of Posaconazole (PCZ) were used as model input. The modeling outcome was verified using PCZ concentration vs. time profiles measured in human intestinal aspirates and in the blood plasma. When using apparently dissolved drug concentrations (i.e., the sum of colloid-associated and molecularly dissolved drug) the simulated systemic plasma exposures were overpredicted, most pronouncedly with the ASD-based tablet. However, if the concentrations of molecularly dissolved drug were used as input values, the PBBM resulted in accurate prediction of systemic exposures for all three PCZ formulations. The present study impressively demonstrated the value of considering molecularly dissolved drug concentrations as input value for PBBMs of supersaturating drug formulations.
Collapse
Affiliation(s)
- Florentin Lukas Holzem
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark; Pharmaceutical R&D, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Jeannine Petrig Schaffland
- Roche Pharmaceutical Research & Early Development, Pre-Clinical CMC, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| | - Martin Brandl
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Annette Bauer-Brandl
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Cordula Stillhart
- Pharmaceutical R&D, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| |
Collapse
|
7
|
Djuris J, Cvijic S, Djekic L. Model-Informed Drug Development: In Silico Assessment of Drug Bioperformance following Oral and Percutaneous Administration. Pharmaceuticals (Basel) 2024; 17:177. [PMID: 38399392 PMCID: PMC10892858 DOI: 10.3390/ph17020177] [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] [Received: 11/03/2023] [Revised: 12/23/2023] [Accepted: 12/29/2023] [Indexed: 02/25/2024] Open
Abstract
The pharmaceutical industry has faced significant changes in recent years, primarily influenced by regulatory standards, market competition, and the need to accelerate drug development. Model-informed drug development (MIDD) leverages quantitative computational models to facilitate decision-making processes. This approach sheds light on the complex interplay between the influence of a drug's performance and the resulting clinical outcomes. This comprehensive review aims to explain the mechanisms that control the dissolution and/or release of drugs and their subsequent permeation through biological membranes. Furthermore, the importance of simulating these processes through a variety of in silico models is emphasized. Advanced compartmental absorption models provide an analytical framework to understand the kinetics of transit, dissolution, and absorption associated with orally administered drugs. In contrast, for topical and transdermal drug delivery systems, the prediction of drug permeation is predominantly based on quantitative structure-permeation relationships and molecular dynamics simulations. This review describes a variety of modeling strategies, ranging from mechanistic to empirical equations, and highlights the growing importance of state-of-the-art tools such as artificial intelligence, as well as advanced imaging and spectroscopic techniques.
Collapse
Affiliation(s)
- Jelena Djuris
- Department of Pharmaceutical Technology and Cosmetology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (S.C.); (L.D.)
| | | | | |
Collapse
|
8
|
Cheong EJY, Chin SY, Ng ZW, Yap TJ, Cheong EZB, Wang Z, Chan ECY. Unraveling Complexities in the Absorption and Disposition Kinetics of Abiraterone via Iterative PBPK Model Development and Refinement. Clin Pharmacokinet 2023; 62:1243-1261. [PMID: 37405634 DOI: 10.1007/s40262-023-01266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 07/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Abiraterone is a first-in-class inhibitor of cytochrome P450 17A1 (CYP17A1), and its pharmacokinetic (PK) profile is susceptible to intrinsic and extrinsic variabilities. Potential associations between abiraterone concentrations and pharmacodynamic consequences in prostate cancer may demand further dosage optimization to balance therapeutic outcomes. Consequently, we aim to develop a physiologically based pharmacokinetic (PBPK) model for abiraterone via a middle-out approach to prospectively interrogate the untested, albeit clinically relevant, scenarios. METHODS To characterize in vivo hydrolysis of prodrug abiraterone acetate (AA) and supersaturation of abiraterone, in vitro aqueous solubility data, biorelevant measurements, and supersaturation and precipitation parameters were utilized for mechanistic absorption simulation. CYP3A4-mediated N-oxidation and sulfotransferase 2A1-catalyzed sulfation of abiraterone were subsequently quantified in human liver subcellular systems. Iterative PBPK model refinement involved evaluation of potential organic anion transporting polypeptide (OATP)-mediated abiraterone uptake in transfected cells in the absence and presence of albumin. RESULTS The developed PBPK model recapitulated the duodenal concentration-time profile of both AA and abiraterone after simulated AA administration. Our findings established abiraterone as a substrate of hepatic OATP1B3 to recapitulate its unbound metabolic intrinsic clearance. Further consideration of a transporter-induced protein-binding shift established accurate translational scaling factors and extrapolated the sinusoidal uptake process. Subsequent simulations effectively predicted the PK of abiraterone upon single and multiple dosing. CONCLUSION Our systematic development of the abiraterone PBPK model has demonstrated its application for the prospective interrogation of the individual or combined influences of potential interindividual variabilities influencing the systemic exposure of abiraterone.
Collapse
Affiliation(s)
- Eleanor Jing Yi Cheong
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Sheng Yuan Chin
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Zheng Wei Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Ting Jian Yap
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Ervin Zhi Bin Cheong
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Ziteng Wang
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore.
| |
Collapse
|
9
|
Denninger A, Becker T, Westedt U, Wagner KG. Advanced In Vivo Prediction by Introducing Biphasic Dissolution Data into PBPK Models. Pharmaceutics 2023; 15:1978. [PMID: 37514164 PMCID: PMC10386266 DOI: 10.3390/pharmaceutics15071978] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Coupling biorelevant in vitro dissolution with in silico physiological-based pharmacokinetic (PBPK) tools represents a promising method to describe and predict the in vivo performance of drug candidates in formulation development including non-passive transport, prodrug activation, and first-pass metabolism. The objective of the present study was to assess the predictability of human pharmacokinetics by using biphasic dissolution results obtained with the previously established BiPHa+ assay and PBPK tools. For six commercial drug products, formulated by different enabling technologies, the respective organic partitioning profiles were processed with two PBPK in silico modeling tools, namely PK-Sim and GastroPlus®, similar to extended-release dissolution profiles. Thus, a mechanistic dissolution/precipitation model of the assessed drug products was not required. The developed elimination/distribution models were used to simulate the pharmacokinetics of the evaluated drug products and compared with available human data. In essence, an in vitro to in vivo extrapolation (IVIVE) was successfully developed. Organic partitioning profiles obtained from the BiPHa+ dissolution analysis enabled highly accurate predictions of the pharmacokinetic behavior of the investigated drug products. In addition, PBPK models of (pro-)drugs with pronounced first-pass metabolism enabled adjustment of the solely passive diffusion predicting organic partitioning profiles, and increased prediction accuracy further.
Collapse
Affiliation(s)
- Alexander Denninger
- Department of Pharmaceutical Technology, University of Bonn, Gerhard-Domagk-Strasse 3, 53121 Bonn, Germany
- Corden Pharma GmbH, Otto-Hahn-Strasse, 68723 Plankstadt, Germany
| | - Tim Becker
- Department of Pharmaceutical Technology, University of Bonn, Gerhard-Domagk-Strasse 3, 53121 Bonn, Germany
| | - Ulrich Westedt
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Karl G Wagner
- Department of Pharmaceutical Technology, University of Bonn, Gerhard-Domagk-Strasse 3, 53121 Bonn, Germany
| |
Collapse
|
10
|
Kane Z, Cheng I, McGarrity O, Chiesa R, Klein N, Cortina-Borja M, Standing JF, Gastine S. Model Based Estimation of Posaconazole Tablet and Suspension Bioavailability in Hospitalized Children Using Real-World Therapeutic Drug Monitoring Data in Patients Receiving Intravenous and Oral Dosing. Antimicrob Agents Chemother 2023; 67:e0007723. [PMID: 37260401 PMCID: PMC10353366 DOI: 10.1128/aac.00077-23] [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: 01/16/2023] [Accepted: 04/18/2023] [Indexed: 06/02/2023] Open
Abstract
Invasive fungal infections are a major cause of morbidity and mortality for immunocompromised patients. Posaconazole is approved for treatment and prophylaxis of invasive fungal infection in adult patients, with intravenous, oral suspension, and gastroresistant/delayed-released tablet formulations available. In Europe, until very recently, posaconazole was used off-label in children, although a new delayed-release suspension approved for pediatric use is expected to become available soon. A population pharmacokinetic model was developed which uses posaconazole therapeutic drug monitoring data following intravenous and oral dosing in hospitalized children, thus enabling estimation of pediatric suspension and tablet oral bioavailability. In total, 297 therapeutic drug monitoring plasma levels from 104 children were included in this analysis. The final model was a one-compartment model with first-order absorption and nonlinear elimination. Allometric scaling on clearance and volume of distribution was included a priori. Tablet bioavailability was estimated to be 66%. Suspension bioavailability was estimated to decrease with increasing doses, ranging from 3.8% to 32.2% in this study population. Additionally, concomitant use of proton pump-inhibitors was detected as a significant covariate, reducing suspension bioavailability by 41.0%. This is the first population pharmacokinetic study to model posaconazole data from hospitalized children following intravenous, tablet, and suspension dosing simultaneously. The incorporation of saturable posaconazole clearance into the model has been key to the credible joint estimation of tablet and suspension bioavailability. To aid rational posaconazole dosing in children, this model was used alongside published pharmacodynamic targets to predict the probability of target attainment using typical pediatric dosing regimen.
Collapse
Affiliation(s)
- Zoe Kane
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Iek Cheng
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Pharmacy, Great Ormond Street Hospital, London, United Kingdom
| | - Orlagh McGarrity
- Department of Pharmacy, Great Ormond Street Hospital, London, United Kingdom
| | - Robert Chiesa
- Department of Bone Marrow Transplantation, Great Ormond Street Hospital, London, United Kingdom
| | - Nigel Klein
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Mario Cortina-Borja
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Joseph F. Standing
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- Department of Pharmacy, Great Ormond Street Hospital, London, United Kingdom
| | - Silke Gastine
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
11
|
Yang E, Yu K, Lee S. Prediction of gastric pH-mediated drug exposure using physiologically-based pharmacokinetic modeling: A case study of itraconazole. CPT Pharmacometrics Syst Pharmacol 2023; 12:865-877. [PMID: 36967484 PMCID: PMC10272297 DOI: 10.1002/psp4.12959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 05/24/2024] Open
Abstract
Abnormal gastric acidity, including achlorhydria, can act as a significant source of variability in orally administered drugs especially with pH-sensitive solubility profiles, such as weak bases, potentially resulting in an undesirable therapeutic response. This study aimed to evaluate the utility of physiologically-based pharmacokinetic (PBPK) modeling in the prediction of gastric pH-mediated drug exposure by using itraconazole, a weak base, as a case. An itraconazole PBPK model was developed on the mechanistic basis of its absorption kinetics in a middle-out manner from a stepwise in vitro-in vivo extrapolation to in vivo refinement. Afterward, an independent prospective clinical study evaluating gastric pH and itraconazole pharmacokinetics (PKs) under normal gastric acidity and esomeprazole-induced gastric hypoacidity was conducted for model validation. Validation was performed by comparing the predicted data with the clinical observations, and the valid model was subsequently applied to predict PK changes under achlorhydria. The developed itraconazole PBPK model showed reasonable reproducibility for gastric pH-mediated exposure observed in the clinical investigation. Based on the model-based simulations, itraconazole exposure was expected to be decreased up to 65% under achlorhydria, and furthermore, gastric pH-mediated exposure could be mechanistically interpreted according to sequential variation in total solubility, dissolution, and absorption. This study suggested the utility of PBPK modeling in the prediction of gastric pH-mediated exposure, especially for drugs whose absorption is susceptible to gastric pH. Our findings will serve as a leading model for further mechanistic assessment of exposure depending on gastric pH for various drugs, ultimately contributing to personalized pharmacotherapy.
Collapse
Affiliation(s)
- Eunsol Yang
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and Hospital101 Daehak‐ro, Jongno‐guSeoul03080Republic of Korea
- Kidney Research InstituteSeoul National University Medical Research Center103 Daehak‐ro, Jongno‐guSeoul03080Republic of Korea
| | - Kyung‐Sang Yu
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and Hospital101 Daehak‐ro, Jongno‐guSeoul03080Republic of Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and TherapeuticsSeoul National University College of Medicine and Hospital101 Daehak‐ro, Jongno‐guSeoul03080Republic of Korea
| |
Collapse
|
12
|
Yamamoto H, Shanker R, Sugano K. Application of Population Balance Model to Simulate Precipitation of Weak Base and Zwitterionic Drugs in Gastrointestinal pH Environment. Mol Pharm 2023; 20:2266-2275. [PMID: 36929729 DOI: 10.1021/acs.molpharmaceut.3c00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
The purpose of the present study was to evaluate whether the population balance model (PBM) could be a suitable model for the precipitation of weak base and zwitterionic drugs in the gastrointestinal pH environment. Five poorly soluble drugs were used as model drugs (dipyridamole, haloperidol, papaverine, phenazopyridine, and tosufloxacin). PBM consists of the equations for primary nucleation, secondary nucleation, and particle growth. Each equation has two empirical parameters. The pH shift (pH-dumping) precipitation test (pH 3.0 to 6.5) was used to determine the model parameters for each drug. It was difficult to determine all six parameters by simultaneously fitting them to the precipitation profiles. Therefore, the number of model parameters was reduced from six to three by neglecting the secondary nucleation process and applying a common exponent number for the particle growth equation. Despite reducing the parameter number, PBM appropriately described the precipitation profiles in the pH shift tests. The constructed PBM model was then used to predict the precipitation profiles in an artificial stomach-intestine transfer (ASIT) test. PBM appropriately predicted the precipitation profiles in the ASIT test. These results suggested that PBM can be a suitable model to represent the precipitation of weak base and zwitterionic drugs in the gastrointestinal pH environment for biopharmaceutics modeling and simulation.
Collapse
Affiliation(s)
- Hibiki Yamamoto
- Molecular Pharmaceutics Lab., College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Ravi Shanker
- Pfizer Worldwide Research, Development, and Medical, 280 Shennecossett Road, Groton, Connecticut 06340, United States
| | - Kiyohiko Sugano
- Molecular Pharmaceutics Lab., College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1, Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| |
Collapse
|
13
|
Zhou D, Chen B, Sharma S, Tang W, Pepin X. Physiologically Based Absorption Modelling to Explore the Formulation and Gastric pH Changes on the Pharmacokinetics of Acalabrutinib. Pharm Res 2023; 40:375-386. [PMID: 35478298 DOI: 10.1007/s11095-022-03268-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 04/20/2022] [Indexed: 10/18/2022]
Abstract
Acalabrutinib, a selective Bruton's tyrosine kinase inhibitor, is a biopharmaceutics classification system class II drug. The aim of this study was to develop a physiologically based pharmacokinetic (PBPK) model to mechanistically describe absorption of immediate release capsule formulation of acalabrutinib in humans. Integration of in vitro biorelevant measurements, dissolution studies and in silico modelling provided clinically relevant inputs for the mechanistic absorption PBPK model. The batch specific dissolution data were integrated in two ways, by fitting a diffusion layer model scalar to the drug product dissolution with integration of drug substance laser diffraction particle size data, or by fitting a product particle size distribution to the dissolution data. The latter method proved more robust and biopredictive. In both cases, the drug surface solubility was well predicted by the Simcyp simulator. The model using the product particle size distribution (P-PSD) for each clinical batch adequately captured the PK profiles of acalabrutinib and its active metabolite. Average fold errors were 0.89 for both Cmax and AUC, suggesting good agreement between predicted and observed PK values. The model also accurately predicted pH-dependent drug-drug interactions between omeprazole and acalabrutinib, which was similar across all clinical formulations. The model predicted acalabrutinib geometric mean AUC ratios (with omeprazole vs acalabrutinib alone) were 0.51 and 0.68 for 2 batches of formulations, which are close to observed values of 0.43 and 0.51~0.63, respectively. The mechanistic absorption PBPK model could be potentially used for future applications such as optimizing formulations or predicting the PK for different batches of the drug product.
Collapse
Affiliation(s)
- Diansong Zhou
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, BioPharmaceuticals R&D, Boston, Massachusetts, USA. .,AstraZeneca, 35 Gatehouse Dr., Waltham, Massachusett, 02451, USA.
| | - Buyun Chen
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, BioPharmaceuticals R&D, South San Francisco, California, USA
| | - Shringi Sharma
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, BioPharmaceuticals R&D, South San Francisco, California, USA
| | - Weifeng Tang
- Clinical Pharmacology & Quantitative Pharmacology, AstraZeneca, BioPharmaceuticals R&D, Gaithersburg, Maryland, USA
| | - Xavier Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| |
Collapse
|
14
|
Wu D, Li M. Current State and Challenges of Physiologically Based Biopharmaceutics Modeling (PBBM) in Oral Drug Product Development. Pharm Res 2023; 40:321-336. [PMID: 36076007 DOI: 10.1007/s11095-022-03373-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/17/2022] [Indexed: 01/17/2023]
Abstract
Physiologically based biopharmaceutics modeling (PBBM) emphasizes the integration of physicochemical properties of drug substance and formulation characteristics with system physiological parameters to predict the absorption and pharmacokinetics (PK) of a drug product. PBBM has been successfully utilized in drug development from discovery to postapproval stages and covers a variety of applications. The use of PBBM facilitates drug development and can reduce the number of preclinical and clinical studies. In this review, we summarized the major applications of PBBM, which are classified into six categories: formulation selection and development, biopredictive dissolution method development, biopharmaceutics risk assessment, clinically relevant specification settings, food effect evaluation and pH-dependent drug-drug-interaction risk assessment. The current state of PBBM applications is illustrated with examples from published studies for each category of application. Despite the variety of PBBM applications, there are still many hurdles limiting the use of PBBM in drug development, that are associated with the complexity of gastrointestinal and human physiology, the knowledge gap between the in vitro and the in vivo behavior of drug products, the limitations of model interfaces, and the lack of agreed model validation criteria, among other issues. The challenges and essential considerations related to the use of PBBM are discussed in a question-based format along with the scientific thinking on future research directions. We hope this review can foster open discussions between the pharmaceutical industry and regulatory agencies and encourage collaborative research to fill the gaps, with the ultimate goal to maximize the applications of PBBM in oral drug product development.
Collapse
Affiliation(s)
- Di Wu
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ, 07065, USA
| | - Min Li
- 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
|
15
|
Pepin XJH, Hammarberg M, Mattinson A, Moir A. Physiologically Based Biopharmaceutics Model for Selumetinib Food Effect Investigation and Capsule Dissolution Safe Space - Part I: Adults. Pharm Res 2023; 40:387-403. [PMID: 36002614 DOI: 10.1007/s11095-022-03339-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/09/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE A physiologically based biopharmaceutics model (PBBM) was developed to mechanistically investigate the effect of formulation and food on selumetinib pharmacokinetics. METHODS Selumetinib is presented as a hydrogen sulfate salt, and in vitro and in vivo data were used to verify the precipitation rate to apply to simulations. Dissolution profiles observed for capsules and granules were used to derive product-particle size distributions for model input. The PBBM incorporated gut efflux and first-pass gut metabolism, based on intravenous and oral pharmacokinetic data, alongside in vitro data for the main enzyme isoform and P-glycoprotein efflux. The PBBM was validated across eight clinical scenarios. RESULTS The quality-control dissolution method for selumetinib capsules was found to be clinically relevant through PBBM validation. A safe space for capsule dissolution was established using a virtual batch. The effect of food (low fat vs high fat) on capsules and granules was elucidated by the PBBM. For capsules, a lower amount was dissolved in the fed state due to a pH increase in the stomach followed by higher precipitation in the small intestine. First-pass gut extraction is higher for capsules in the fed state due to drug dilution in the stomach chyme and reduced concentration in the lumen. The enteric-coated granules dissolve more slowly than capsules after stomach emptying, attenuating the difference in first-pass gut extraction between prandial states. CONCLUSIONS The PBBM was instrumental in understanding and explaining the different behaviors of the selumetinib formulations. The model can be used to predict the impact of food in humans.
Collapse
Affiliation(s)
- Xavier J H Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Maria Hammarberg
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden. .,AstraZeneca, Pepparedsleden, SE-431 83, Mölndal, Sweden.
| | - Alexandra Mattinson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Andrea Moir
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| |
Collapse
|
16
|
Franco YL, Da Silva L, Charbe N, Kinvig H, Kim S, Cristofoletti R. Integrating Forward and Reverse Translation in PBPK Modeling to Predict Food Effect on Oral Absorption of Weakly Basic Drugs. Pharm Res 2023; 40:405-418. [PMID: 36788156 DOI: 10.1007/s11095-023-03478-0] [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: 10/10/2022] [Accepted: 01/28/2023] [Indexed: 02/16/2023]
Abstract
INTRODUCTION Ketoconazole and posaconazole are two weakly basic broad-spectrum antifungals classified as Biopharmaceutics Classification System class II drugs, indicating that they are highly permeable, but exhibit poor solubility. As a result, oral bioavailability and clinical efficacy can be impacted by the formulation performance in the gastrointestinal system. In this work, we have leveraged in vitro biopharmaceutics and clinical data available in the literature to build physiologically based pharmacokinetic (PBPK) models for ketoconazole and posaconazole, to determine the suitability of forward in vitro-in vivo translation for characterization of in vivo drug precipitation, and to predict food effect. METHODS A stepwise modeling approach was utilized to derive key parameters related to absorption, such as drug solubility, dissolution, and precipitation kinetics from in vitro data. These parameters were then integrated into PBPK models for the simulation of ketoconazole and posaconazole plasma concentrations in the fasted and fed states. RESULTS Forward in vitro-in vivo translation of intestinal precipitation kinetics for both model drugs resulted in poor predictions of PK profiles. Therefore, a reverse translation approach was applied, based on limited fitting of precipitation-related parameters to clinical data. Subsequent simulations for ketoconazole and posaconazole demonstrated that fasted and fed state PK profiles for both drugs were adequately recapitulated. CONCLUSION The two examples presented in this paper show how middle-out modeling approaches can be used to predict the magnitude and direction of food effects provided the model is verified on fasted state PK data.
Collapse
Affiliation(s)
- Yesenia L Franco
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics (Lake Nona), University of Florida, Orlando, FL, USA
| | - Lais Da Silva
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics (Lake Nona), University of Florida, Orlando, FL, USA
| | - Nitin Charbe
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics (Lake Nona), University of Florida, Orlando, FL, USA
| | - Hannah Kinvig
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics (Lake Nona), University of Florida, Orlando, FL, USA
| | - Soyoung Kim
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics (Lake Nona), University of Florida, Orlando, FL, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics & Systems Pharmacology, Department of Pharmaceutics (Lake Nona), University of Florida, Orlando, FL, USA.
| |
Collapse
|
17
|
Moseson DE, Benson EG, Cao Z, Bhalla S, Wang F, Wang M, Zheng K, Narwankar PK, Simpson GJ, Taylor LS. Impact of Aluminum Oxide Nanocoating on Drug Release from Amorphous Solid Dispersion Particles. Mol Pharm 2023; 20:593-605. [PMID: 36346665 DOI: 10.1021/acs.molpharmaceut.2c00818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Atomic layer coating (ALC) is emerging as a particle engineering strategy to inhibit surface crystallization of amorphous solid dispersions (ASDs). In this study, we turn our attention to evaluating drug release behavior from ALC-coated ASDs, and begin to develop a mechanistic framework. Posaconazole/hydroxypropyl methylcellulose acetate succinate was used as a model system at both 25% and 50% drug loadings. ALC-coatings of aluminum oxide up to 40 nm were evaluated for water sorption kinetics and dissolution performance under a range of pH conditions. Scanning electron microscopy with energy dispersive X-ray analysis was used to investigate the microstructure of partially released ASD particles. Coating thickness and defect density (inferred from deposition rates) were found to impact water sorption kinetics. Despite reduced water sorption kinetics, the presence of a coating was not found to impact dissolution rates under conditions where rapid drug release was observed. Under slower releasing conditions, underlying matrix crystallization was reduced by the coating, enabling greater levels of drug release. These results demonstrate that water was able to penetrate through the ALC coating, hydrating the amorphous solid, which can initiate dissolution of drug and/or polymer (depending on pH conditions). Swelling of the ASD substrate subsequently occurs, disrupting and cracking the coating, which serves to facilitate rapid drug release. Water sorption kinetics are highlighted as a potential predictive tool to investigate the coating quality and its potential impact on dissolution performance. This study has implications for formulation design and evaluation of ALC-coated ASD particles.
Collapse
Affiliation(s)
- Dana E Moseson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Emily G Benson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ziyi Cao
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shradha Bhalla
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Fei Wang
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Miaojun Wang
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Kai Zheng
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Pravin K Narwankar
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Garth J Simpson
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
18
|
An Integrated Population Pharmacokinetic Analysis for Posaconazole Oral Suspension, Delayed-Release Tablet, and Intravenous Infusion in Healthy Volunteers. Drugs 2023; 83:75-86. [PMID: 36607589 DOI: 10.1007/s40265-022-01819-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Posaconazole is widely used for the prophylaxis and treatment of invasive fungal diseases. Because of the limited and variable absorption of the initially available oral suspension, a delayed-release tablet and intravenous formulation were developed. OBJECTIVE This study aimed to characterize the pharmacokinetics, including the absolute oral bioavailability, of all posaconazole formulations in healthy volunteers. METHODS Data from 182 healthy volunteers with 3898 densely sampled posaconazole concentrations were pooled from eight phase I clinical studies on the three formulations of various single and multiple dosage regimens between 50 and 400 mg. Analysis and simulations were performed using NONMEM 7.5.0. In the covariate analysis, the influence of food (fed vs fasted), nonlinearity, and for the delayed-release tablet, comedication (antacid, ranitidine, esomeprazole, and metoclopramide) were tested. RESULTS A two-compartment model with respectively, four and eight absorption transit compartments, best described the profiles of the oral suspension and delayed-release tablet. For the suspension, both a food effect and a dose-dependent nonlinear bioavailability were quantified, resulting in lower bioavailability when fasted or at a higher dose. The typical bioavailability of the suspension at 100 mg and 400 mg was derived to be respectively, 17.1% and 10.1% under fasted conditions and 59.1% and 49.2% under fed conditions. The absolute bioavailability of the delayed-release tablet was 58.8% (95% confidence interval 33.2-80.4) under fasted conditions and approached complete absorption under fed conditions for dosages up to 300 mg. Food intake reduced the absorption rate constant of the suspension by 52.2% (confidence interval 45.2-59.2). The impact of comedication on the absorption of the delayed-release tablet was not statistically significant. Model-based simulations indicate that under fed conditions, the licensed dosages of the three formulations yield a steady-state trough concentration ≥ 0.7 mg/L in over 90% of healthy volunteers. About 35% of healthy volunteers who receive the licensed 300-mg delayed-release tablet under fasted conditions do not achieve this target, while for the suspension this percentage varies between 55 and 85%, depending on the dose. CONCLUSIONS For both oral posaconazole formulations, we quantified bioavailability and absorption rate, including food effects, in healthy volunteers. The pharmacokinetic superiority of the delayed-release tablet was demonstrated under both fed and fasted conditions, compared with the oral suspension. The impact of food on the bioavailability of the delayed-release tablet was larger than anticipated, suggesting that administering the delayed-release tablet with food enhances absorption.
Collapse
|
19
|
A Physiologically-Based Pharmacokinetic Model of Ruxolitinib and Posaconazole to Predict CYP3A4-Mediated Drug-Drug Interaction Frequently Observed in Graft versus Host Disease Patients. Pharmaceutics 2022; 14:pharmaceutics14122556. [PMID: 36559050 PMCID: PMC9785192 DOI: 10.3390/pharmaceutics14122556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
Ruxolitinib (RUX) is approved for the treatment of steroid-refractory acute and chronic graft versus host disease (GvHD). It is predominantly metabolized via cytochrome P450 (CYP) 3A4. As patients with GvHD have an increased risk of invasive fungal infections, RUX is frequently combined with posaconazole (POS), a strong CYP3A4 inhibitor. Knowledge of RUX exposure under concomitant POS treatment is scarce and recommendations on dose modifications are inconsistent. A physiologically based pharmacokinetic (PBPK) model was developed to investigate the drug-drug interaction (DDI) between POS and RUX. The predicted RUX exposure was compared to observed concentrations in patients with GvHD in the clinical routine. PBPK models for RUX and POS were independently set up using PK-Sim® Version 11. Plasma concentration-time profiles were described successfully and all predicted area under the curve (AUC) values were within 2-fold of the observed values. The increase in RUX exposure was predicted with a DDI ratio of 1.21 (Cmax) and 1.59 (AUC). Standard dosing in patients with GvHD led to higher RUX exposure than expected, suggesting further dose reduction if combined with POS. The developed model can serve as a starting point for further simulations of the implemented DDI and can be extended to further perpetrators of CYP-mediated PK-DDIs or disease-specific physiological changes.
Collapse
|
20
|
Fisher MC, Alastruey-Izquierdo A, Berman J, Bicanic T, Bignell EM, Bowyer P, Bromley M, Brüggemann R, Garber G, Cornely OA, Gurr SJ, Harrison TS, Kuijper E, Rhodes J, Sheppard DC, Warris A, White PL, Xu J, Zwaan B, Verweij PE. Tackling the emerging threat of antifungal resistance to human health. Nat Rev Microbiol 2022; 20:557-571. [PMID: 35352028 PMCID: PMC8962932 DOI: 10.1038/s41579-022-00720-1] [Citation(s) in RCA: 512] [Impact Index Per Article: 170.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2022] [Indexed: 12/12/2022]
Abstract
Invasive fungal infections pose an important threat to public health and are an under-recognized component of antimicrobial resistance, an emerging crisis worldwide. Across a period of profound global environmental change and expanding at-risk populations, human-infecting pathogenic fungi are evolving resistance to all licensed systemic antifungal drugs. In this Review, we highlight the main mechanisms of antifungal resistance and explore the similarities and differences between bacterial and fungal resistance to antimicrobial control. We discuss the research and innovation topics that are needed for risk reduction strategies aimed at minimizing the emergence of resistance in pathogenic fungi. These topics include links between the environment and One Health, surveillance, diagnostics, routes of transmission, novel therapeutics and methods to mitigate hotspots for fungal adaptation. We emphasize the global efforts required to steward our existing antifungal armamentarium, and to direct the research and development of future therapies and interventions.
Collapse
Affiliation(s)
- Matthew C Fisher
- MRC Centre for Global Infectious Disease Outbreak Analysis, Imperial College London, London, UK.
| | - Ana Alastruey-Izquierdo
- Mycology Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | - Judith Berman
- Shmunis School of Biomedical and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Israel
| | - Tihana Bicanic
- Institute of Infection and Immunity, St George's University London, London, UK
| | - Elaine M Bignell
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Paul Bowyer
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Michael Bromley
- Manchester Fungal Infection Group, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Roger Brüggemann
- Department of Pharmacy, Radboudumc Institute for Health Sciences and Radboudumc - CWZ Centre of Expertise for Mycology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Gary Garber
- Department of Medicine and the School of Public Health and Epidemiology, University of Ottawa, Ottawa, Ontario, Canada
| | - Oliver A Cornely
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Cologne, Germany
| | | | - Thomas S Harrison
- Institute of Infection and Immunity, St George's University London, London, UK
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Ed Kuijper
- Centre for Infectious Diseases Research, Diagnostics and Laboratory Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Johanna Rhodes
- MRC Centre for Global Infectious Disease Outbreak Analysis, Imperial College London, London, UK
| | - Donald C Sheppard
- Infectious Disease in Global Health Program and McGill Interdisciplinary Initiative in Infection and Immunity, McGill University Health Centre, Montreal, Québec, Canada
| | - Adilia Warris
- MRC Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - P Lewis White
- Public Health Wales Mycology Reference Laboratory, University Hospital of Wales, Cardiff, UK
| | - Jianping Xu
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Bas Zwaan
- Department of Plant Science, Laboratory of Genetics, Wageningen University & Research, Wageningen, Netherlands
| | - Paul E Verweij
- Centre for Infectious Diseases Research, Diagnostics and Laboratory Surveillance, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands.
- Department of Medical Microbiology and Radboudumc - CWZ Centre of Expertise for Mycology, Radboud University Medical Centre, Nijmegen, Netherlands.
| |
Collapse
|
21
|
Developing Clinically Relevant Dissolution Specifications (CRDSs) for Oral Drug Products: Virtual Webinar Series. Pharmaceutics 2022; 14:pharmaceutics14051010. [PMID: 35631595 PMCID: PMC9148161 DOI: 10.3390/pharmaceutics14051010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/29/2022] [Accepted: 05/03/2022] [Indexed: 02/06/2023] Open
Abstract
A webinar series that was organised by the Academy of Pharmaceutical Sciences Biopharmaceutics focus group in 2021 focused on the challenges of developing clinically relevant dissolution specifications (CRDSs) for oral drug products. Industrial scientists, together with regulatory and academic scientists, came together through a series of six webinars, to discuss progress in the field, emerging trends, and areas for continued collaboration and harmonisation. Each webinar also hosted a Q&A session where participants could discuss the shared topic and information. Although it was clear from the presentations and Q&A sessions that we continue to make progress in the field of CRDSs and the utility/success of PBBM, there is also a need to continue the momentum and dialogue between the industry and regulators. Five key areas were identified which require further discussion and harmonisation.
Collapse
|
22
|
Ibrahim F. An enabling formulation of a weakly basic compound guided by Physiologically Based Biopharmaceutics Modeling (PBBM). J Pharm Sci 2022; 111:2490-2495. [DOI: 10.1016/j.xphs.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/04/2022] [Accepted: 04/04/2022] [Indexed: 11/30/2022]
|
23
|
Bego M, Patel N, Cristofoletti R, Rostami-Hodjegan A. Proof of Concept in Assignment of Within-Subject Variability During Virtual Bioequivalence Studies: Propagation of Intra-Subject Variation in Gastrointestinal Physiology Using Physiologically Based Pharmacokinetic Modeling. AAPS J 2022; 24:21. [PMID: 34988679 PMCID: PMC8817238 DOI: 10.1208/s12248-021-00672-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/04/2021] [Indexed: 12/11/2022] Open
Abstract
While the concept of ‘Virtual Bioequivalence’ (VBE) using a combination of modelling, in vitro tests and integration of pre-existing data on systems and drugs is growing from its infancy, building confidence on VBE outcomes requires demonstration of its ability not only in predicting formulation-dependent systemic exposure but also the expected degree of population variability. The concept of variation influencing the outcome of BE, despite being hidden with the cross-over nature of common BE studies, becomes evident when dealing with the acceptance criteria that consider the 90% confidence interval (CI) around the relative bioavailability. Hence, clinical studies comparing a reference product against itself may fail due to within-subject variations associated with the two occasions that the individual receives the same formulation. In this proof-of-concept study, we offer strategies to capture the most realistic predictions of CI around the pharmacokinetic parameters by propagating physiological variations through physiologically based pharmacokinetic modelling. The exercise indicates feasibility of the approach based on comparisons made between the simulated and observed WSV of pharmacokinetic parameters tested for a clinical bioequivalence case study. However, it also indicates that capturing WSV of a large array of physiological parameters using backward translation modelling from repeated BE studies of reference products would require a diverse set of drugs and formulations. The current case study of delayed-release formulation of posaconazole was able to declare certain combinations of WSV of physiological parameters as ‘not plausible’. The eliminated sets of WSV values would be applicable to PBPK models of other drugs and formulations. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Margareta Bego
- Agency for Medicinal Products and Medical Devices (HALMED), Zagreb, Croatia. .,Centre for Applied Pharmacokinetic Research (CAPKR), School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PL, UK.
| | - Nikunjkumar Patel
- Certara UK Limited, Simcyp Division, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PL, UK.,Certara UK Limited, Simcyp Division, 1 Concourse Way, Sheffield, S1 2BJ, UK
| |
Collapse
|
24
|
Kaur M, Yardley V, Wang K, Masania J, Arroo RRJ, Turner DB, Li M. Artemisinin Cocrystals for Bioavailability Enhancement. Part 2: In Vivo Bioavailability and Physiologically Based Pharmacokinetic Modeling. Mol Pharm 2021; 18:4272-4289. [PMID: 34748332 DOI: 10.1021/acs.molpharmaceut.1c00385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report the evaluation and prediction of the pharmacokinetic (PK) performance of artemisinin (ART) cocrystal formulations, that is, 1:1 artemisinin/orcinol (ART-ORC) and 2:1 artemisinin/resorcinol (ART2-RES), using in vivo murine animal and physiologically based pharmacokinetic (PBPK) models. The efficacy of the ART cocrystal formulations along with the parent drug ART was tested in mice infected with Plasmodium berghei. When given at the same dose, the ART cocrystal formulation showed a significant reduction in parasitaemia at day 4 after infection compared to ART alone. PK parameters including Cmax (maximum plasma concentration), Tmax (time to Cmax), and AUC (area under the curve) were obtained by determining drug concentrations in the plasma using liquid chromatography-high-resolution mass spectrometry (LC-HRMS), showing enhanced ART levels after dosage with the cocrystal formulations. The dose-response tests revealed that a significantly lower dose of the ART cocrystals in the formulation was required to achieve a similar therapeutic effect as ART alone. A PBPK model was developed using a PBPK mouse simulator to accurately predict the in vivo behavior of the cocrystal formulations by combining in vitro dissolution profiles with the properties of the parent drug ART. The study illustrated that information from classical in vitro and in vivo experimental investigations of the parent drug of ART formulations can be coupled with PBPK modeling to predict the PK parameters of an ART cocrystal formulation in an efficient manner. Therefore, the proposed modeling strategy could be used to establish in vitro and in vivo correlations for different cocrystals intended to improve dissolution properties and to support clinical candidate selection, contributing to the assessment of cocrystal developability and formulation development.
Collapse
Affiliation(s)
- Manreet Kaur
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - Vanessa Yardley
- Department of Infection & Immunity, Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, U.K
| | - Ke Wang
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - Jinit Masania
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - Randolph R J Arroo
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - David B Turner
- Certara UK Limited, Simcyp Division, Sheffield S1 2BJ, U.K
| | - Mingzhong Li
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| |
Collapse
|
25
|
Vinarov Z, Abrahamsson B, Artursson P, Batchelor H, Berben P, Bernkop-Schnürch A, Butler J, Ceulemans J, Davies N, Dupont D, Flaten GE, Fotaki N, Griffin BT, Jannin V, Keemink J, Kesisoglou F, Koziolek M, Kuentz M, Mackie A, Meléndez-Martínez AJ, McAllister M, Müllertz A, O'Driscoll CM, Parrott N, Paszkowska J, Pavek P, Porter CJH, Reppas C, Stillhart C, Sugano K, Toader E, Valentová K, Vertzoni M, De Wildt SN, Wilson CG, Augustijns P. Current challenges and future perspectives in oral absorption research: An opinion of the UNGAP network. Adv Drug Deliv Rev 2021; 171:289-331. [PMID: 33610694 DOI: 10.1016/j.addr.2021.02.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Although oral drug delivery is the preferred administration route and has been used for centuries, modern drug discovery and development pipelines challenge conventional formulation approaches and highlight the insufficient mechanistic understanding of processes critical to oral drug absorption. This review presents the opinion of UNGAP scientists on four key themes across the oral absorption landscape: (1) specific patient populations, (2) regional differences in the gastrointestinal tract, (3) advanced formulations and (4) food-drug interactions. The differences of oral absorption in pediatric and geriatric populations, the specific issues in colonic absorption, the formulation approaches for poorly water-soluble (small molecules) and poorly permeable (peptides, RNA etc.) drugs, as well as the vast realm of food effects, are some of the topics discussed in detail. The identified controversies and gaps in the current understanding of gastrointestinal absorption-related processes are used to create a roadmap for the future of oral drug absorption research.
Collapse
Affiliation(s)
- Zahari Vinarov
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Department of Chemical and Pharmaceutical Engineering, Sofia University, Sofia, Bulgaria
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Hannah Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Philippe Berben
- Pharmaceutical Development, UCB Pharma SA, Braine- l'Alleud, Belgium
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - James Butler
- GlaxoSmithKline Research and Development, Ware, United Kingdom
| | | | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Gøril Eide Flaten
- Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | | | | | | | | | - Martin Kuentz
- Institute for Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, Basel, Switzerland
| | - Alan Mackie
- School of Food Science & Nutrition, University of Leeds, Leeds, United Kingdom
| | | | | | - Anette Müllertz
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Petr Pavek
- Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
| | | | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Kiyohiko Sugano
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Elena Toader
- Faculty of Medicine, University of Medicine and Pharmacy of Iasi, Romania
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Saskia N De Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clive G Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
26
|
Hens B, Augustijns P, Lennernäs H, McAllister M, Abrahamsson B. Leveraging Oral Drug Development to a Next Level: Impact of the IMI-Funded OrBiTo Project on Patient Healthcare. Front Med (Lausanne) 2021; 8:480706. [PMID: 33748152 PMCID: PMC7973356 DOI: 10.3389/fmed.2021.480706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 02/12/2021] [Indexed: 11/13/2022] Open
Abstract
A thorough understanding of the behavior of drug formulations in the human gastrointestinal (GI) tract is essential when working in the field of oral drug development in a pharmaceutical company. For orally administered drug products, various GI processes, including disintegration of the drug formulation, drugrelease, dissolution, precipitation, degradation, dosage form transit and permeation, dictate absorption into the systemic circulation. These processes are not always fully captured in predictive in vitro and in silico tools, as commonly applied in the pre-clinical stage of formulation drug development. A collaborative initiative focused on the science of oral biopharmaceutics was established in 2012 between academic institutions and industrial companies to innovate, optimize and validate these in vitro and in silico biopharmaceutical tools. From that perspective, the predictive power of these models can be revised and, if necessary, optimized to improve the accuracy toward predictions of the in vivo performance of orally administered drug products in patients. The IMI/EFPIA-funded "Oral Bioavailability Tools (OrBiTo)" project aimed to improve our fundamental understanding of the GI absorption process. The gathered information was integrated into the development of new (or already existing) laboratory tests and computer-based methods in order to deliver more accurate predictions of drug product behavior in a real-life setting. These methods were validated with the use of industrial data. Crucially, the ultimate goal of the project was to set up a scientific framework (i.e., decision trees) to guide the use of these new tools in drug development. The project aimed to facilitate and accelerate the formulation development process and to significantly reduce the need for animal experiments in this area as well as for human clinical studies in the future. With respect to the positive outcome for patients, high-quality oral medicines will be developed where the required dose is well-calculated and consistently provides an optimal clinical effect. In a first step, this manuscript summarizes the setup of the project and how data were collected across the different work packages. In a second step, case studies of how this project contributed to improved knowledge of oral drug delivery which can be used to develop improved products for patients will be illustrated.
Collapse
Affiliation(s)
- Bart Hens
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Drug Product Design, Pfizer, Sandwich, United Kingdom
| | - Patrick Augustijns
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Hans Lennernäs
- Department of Pharmaceutical Biosciences and Technology, Uppsala University, Uppsala, Sweden
| | | | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology and Development, Operations, AstraZeneca Gothenburg, Mölndal, Sweden
| |
Collapse
|
27
|
O'Dwyer PJ, Box KJ, Dressman J, Griffin BT, Henze LJ, Litou C, Pentafragka C, Statelova M, Vertzoni M, Reppas C. Oral biopharmaceutics tools: recent progress from partnership through the Pharmaceutical Education and Research with Regulatory Links collaboration. J Pharm Pharmacol 2021; 73:437-446. [PMID: 33793836 DOI: 10.1093/jpp/rgaa055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To summarise key contributions of the Pharmaceutical Education and Research with Regulatory Links (PEARRL) project (2016-2020) to the optimisation of existing and the development of new biopharmaceutics tools for evaluating the in vivo performance of oral drug products during the development of new drugs and at the regulatory level. KEY FINDINGS Optimised biopharmaceutics tools: Based on new clinical data, the composition of biorelevant media for simulating the fed state conditions in the stomach was simplified. Strategies on how to incorporate biorelevant in vitro data of bio-enabling drug products into physiologically based pharmacokinetic (PBPK) modelling were proposed. Novel in vitro biopharmaceutics tools: Small-scale two-stage biphasic dissolution and dissolution-permeation setups were developed to facilitate understanding of the supersaturation effects and precipitation risks of orally administered drugs. A porcine fasted state simulated intestinal fluid was developed to improve predictions and interpretation of preclinical results using in vitro dissolution studies. Based on new clinical data, recommendations on the design of in vitro methodologies for evaluating the GI drug transfer process in the fed state were suggested. The optimized design of in vivo studies for investigating food effects: A food effect study protocol in the pig model was established which successfully predicted the food-dependent bioavailability of two model compounds. The effect of simulated infant fed state conditions in healthy adults on the oral absorption of model drugs was evaluated versus the fasted state and the fed state conditions, as defined by regulatory agencies for adults. Using PBPK modelling, the extrapolated fasted and infant fed conditions data appeared to be more useful to describe early drug exposure in infants, while extrapolation of data collected under fed state conditions, as defined by regulators for adults, failed to capture in vivo infant drug absorption. SUMMARY Substantial progress has been made in developing an advanced suite of biopharmaceutics tools for streamlining drug formulation screening and supporting regulatory applications. These advances in biopharmaceutics were achieved through networking opportunities and research collaborations provided under the H2020 funded PEARRL project.
Collapse
Affiliation(s)
- Patrick J O'Dwyer
- School of Pharmacy, University College Cork, Cork, Ireland.,Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK.,Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Karl J Box
- Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK
| | - Jennifer Dressman
- Institute of Translational Medicine and Pharmacology (ITMP), Fraunhofer Gesellschaft, Frankfurt am Main, Germany
| | | | - Laura J Henze
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Chara Litou
- Institute of Translational Medicine and Pharmacology (ITMP), Fraunhofer Gesellschaft, Frankfurt am Main, Germany
| | - Christina Pentafragka
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Marina Statelova
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Maria Vertzoni
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Christos Reppas
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| |
Collapse
|
28
|
Bhatnagar S, Mukherjee D, Salem AH, Miles D, Menon RM, Gibbs JP. Dose adjustment of venetoclax when co-administered with posaconazole: clinical drug-drug interaction predictions using a PBPK approach. Cancer Chemother Pharmacol 2021; 87:465-474. [PMID: 33398386 DOI: 10.1007/s00280-020-04179-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE Venetoclax, a targeted anticancer agent approved for the treatment of chronic lymphocytic leukemia and acute myeloid leukemia, is a substrate of cytochrome P450 (CYP) 3A enzyme (CYP3A4). Posaconazole, commonly used to prevent invasive fungal infections in neutropenic patients with hematological malignancies, potently inhibits CYP3A4. The purpose of this evaluation was to predict venetoclax exposures following co-administration of posaconazole at doses not previously studied clinically. METHODS Two physiologically based pharmacokinetic (PBPK) models were developed for posaconazole based on published parameters, one for an oral suspension and another for delayed released tablets. Parameter optimization, guided by sensitivity analyses, was conducted such that the models could replicate clinical exposures of posaconazole and drug-drug interactions with sensitive CYP3A substrates including venetoclax. The clinically verified posaconazole PBPK models were then utilized to predict DDI with a previously published venetoclax PBPK model at clinically relevant dosing scenarios. RESULTS The posaconazole PBPK models predicted posaconazole exposure and DDI related fold changes with acceptable prediction errors for both posaconazole formulations. The model predicted exposures of venetoclax, when co-administered with a 300 mg QD dose of delayed release tablets of posaconazole, were in concordance with observed data. Increasing the posaconazole dose to 500 mg QD increased venetoclax exposures by about 12% relative to 300 mg QD, which were still within the venetoclax safe exposure range. CONCLUSIONS The posaconazole PBPK models were developed and clinically verified. Predictions using the robust PBPK model confirmed the venetoclax label recommendation of 70 mg in the presence of posaconazole at doses up to 500 mg QD.
Collapse
Affiliation(s)
- Sumit Bhatnagar
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., 1 North Waukegan Road, Dept. R4PK, Bldg. AP31-3, North Chicago, IL, 60064, USA.
| | - Dwaipayan Mukherjee
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., 1 North Waukegan Road, Dept. R4PK, Bldg. AP31-3, North Chicago, IL, 60064, USA
| | - Ahmed Hamed Salem
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., 1 North Waukegan Road, Dept. R4PK, Bldg. AP31-3, North Chicago, IL, 60064, USA.,Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Dale Miles
- Clinical Pharmacology, Genentech Inc, South San Francisco, CA, USA
| | - Rajeev M Menon
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., 1 North Waukegan Road, Dept. R4PK, Bldg. AP31-3, North Chicago, IL, 60064, USA
| | - John P Gibbs
- Clinical Pharmacology and Pharmacometrics, AbbVie Inc., 1 North Waukegan Road, Dept. R4PK, Bldg. AP31-3, North Chicago, IL, 60064, USA
| |
Collapse
|
29
|
Abstract
Posaconazole is typically used for preventing invasive yeast and mold infections such as invasive aspergillosis in high-risk immunocompromised patients. The oral suspension was the first released formulation and many pharmacokinetic and pharmacodynamic studies of this formulation have been published. Erratic absorption profiles associated with this formulation were widely reported. Posaconazole exposure was found to be significantly influenced by food and many gastrointestinal conditions, including pH and motility. As a result, low posaconazole plasma concentrations were obtained in large groups of patients. These issues of erratic absorption urged the development of the subsequently marketed delayed-release tablet, which proved to be associated with higher and more stable exposure profiles. Shortly thereafter, an intravenous formulation was released for patients who are not able to take oral formulations. Both new formulations require a loading dose on day 1 to achieve high posaconazole concentrations more quickly, which was not possible with the oral suspension. So far, there appears to be no evidence of increased toxicity correlated to the higher posaconazole exposure achieved with the regimen for these formulations. The higher systemic availability of posaconazole for the delayed-release tablet and intravenous formulation have resulted in these two formulations being preferable for both prophylaxis and treatment of invasive fungal disease. This review aimed to integrate the current knowledge on posaconazole pharmacokinetics, pharmacodynamics, major toxicity, existing resistance, clinical experience in special populations, and new therapeutic strategies in order to get a clear understanding of the clinical use of this drug.
Collapse
|
30
|
Gao H, Wang W, Dong J, Ye Z, Ouyang D. An integrated computational methodology with data-driven machine learning, molecular modeling and PBPK modeling to accelerate solid dispersion formulation design. Eur J Pharm Biopharm 2020; 158:336-346. [PMID: 33301864 DOI: 10.1016/j.ejpb.2020.12.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/25/2020] [Accepted: 12/02/2020] [Indexed: 01/05/2023]
Abstract
Drugs in solid dispersion (SD) take advantage of fast and extended dissolution, thus attains a higher bioavailability than the crystal form. However, current development of SD relies on a random large-scale formulation screening method with low efficiency. Current research aims to integrate various computational tools, including machine learning (ML), molecular dynamic (MD) simulation and physiologically based pharmacokinetic (PBPK) modeling, to accelerate the development of SD formulations. Firstly, based on a dataset consisting of 674 dissolution profiles of SD, the random forest algorithm was used to construct a classification model to distinguish two types of dissolution profiles: "spring-and-parachute" and "maintain supersaturation", and a regression model to predict the time-dependent dissolution profiles. Both of the two prediction models showed good prediction performance. Moreover, feature importance was performed to help understand the key information that contributes to the model. After that, the vemurafenib (VEM) SD formulation in previous report was used as an example to validate the models. MD simulation was used to investigate the dissolution behavior of two SD formulations with two polymers (HPMCAS and Eudragit) at the molecular level. The results showed that the HPMCAS-based formulation resulted in faster dissolution than the Eudragit formulation, which agreed with the reported experimental results. Finally, a PBPK model was constructed to accurately predict the human pharmacokinetic profile of the VEM-HPMCAS SD formulation. In conclusion, combined computational tools have been developed to in silico predict formulation composition, in vitro release and in vivo absorption behavior of SD formulations. The integrated computational methodology will significantly facilitate pharmaceutical formulation development than the traditional trial-and-error approach in the laboratory.
Collapse
Affiliation(s)
- Hanlu Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China
| | - Wei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China
| | - Jie Dong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China
| | - Zhuyifan Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences (ICMS), University of Macau, Macau, China.
| |
Collapse
|
31
|
A combined in vitro in-silico approach to predict the oral bioavailability of borderline BCS Class II/IV weak base albendazole and its main metabolite albendazole sulfoxide. Eur J Pharm Sci 2020; 155:105552. [PMID: 32937212 DOI: 10.1016/j.ejps.2020.105552] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/28/2020] [Accepted: 09/11/2020] [Indexed: 12/17/2022]
Abstract
The aim of this study was to use a combined in vitro-in silico approach to develop a physiologically based pharmacokinetic model (PBPK) that predicts the bioavailability of albendazole (ABZ), a BCS class II/IV lipophilic weak base, and simulates its main metabolite albendazole sulphoxide (ABZSO) after oral administration of the current marketed dose of 400 mg in the fasted state. In vitro data was collected from solubility and dissolution tests performed with biorelevant media and transfer tests were carried out to evaluate the supersaturation and precipitation characteristics of ABZ upon gastric emptying. These in vitro results were used as biopharmaceutical inputs together with ABZ physicochemical properties including also permeability and in vitro metabolism data and information gathered from different clinical trials reported in the literature, were used to enable PBPK models to be developed using GastroPlus™ (version 9.7). As expected for this weak base with pKa = 3.6, ABZ exhibited a pronounced pH dependent solubility, with the solubility and extent of dissolution being greater at gastric pH and dropping significantly in the intestinal environment suggesting supersaturation and precipitation upon gastric emptying, which was confirmed by the transfer model experiments. PBPK models were set up for heathy volunteers using a full PBPK modeling approach and by implementing dynamic fluid volumes in the ACAT gut physiology in GastroPlus™. When coupling in vitro data (solubility values, dissolution rate and precipitation rate constant, etc.) for ABZ and with fitted values for the Vdss and liver systemic clearance of the sulfoxide metabolite to the PBPK model, the simulated profiles successfully predicated plasma concentrations of ABZ at 400 mg dose and simulated ABZSO at different ABZ dose levels and with different study populations, indicating the usefulness of combing in vitro biorelevant tools with PBPK modeling for the accurate prediction of ABZ bioavailability. The results obtained in this study also helped confirm that ABZ behaves as a BCS class IV compound.
Collapse
|
32
|
Komasaka T, Dressman J. Simulation of oral absorption from non-bioequivalent dosage forms of the salt of raltegravir, a poorly soluble acidic drug, using a physiologically based biopharmaceutical modeling (PBBM) approach. Eur J Pharm Sci 2020; 157:105630. [PMID: 33122010 DOI: 10.1016/j.ejps.2020.105630] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/13/2020] [Accepted: 10/23/2020] [Indexed: 11/24/2022]
Abstract
Non-bioequivalent plasma concentration profiles among different dosage forms of the salt of raltegravir, a poorly soluble acidic drug, were investigated using biorelevant in vitro testing combined with the commercial in silico software, Simcyp®. A suspension and a tablet dosage forms of raltegravir potassium were selected as the test formulations. While dissolution from the suspension was rapid, dissolution from the tablets was slow and delayed by pre-exposure to an acidic environment. Although the tablet was expected to have complex in vivo performance, plasma concentration profiles were successfully simulated when gastric emptying was taken into account as a key physiological factor in in vitro and in silico trials. The effect of pre-exposure to acid in the stomach on dissolution behavior in the intestine was estimated by two-stage in vitro dissolution testing. Based on these results, theoretical in vivo dissolution profiles for different gastric emptying times were inputted into the in silico model and plasma concentration profiles were simulated taking the distribution of individual gastric emptying times into account. The in vitro and in silico method presented in this report would be a practical approach to simulate oral absorption from various formulations of poorly soluble weak acids and their salts.
Collapse
Affiliation(s)
- Takao Komasaka
- Pharmaceutical Research Department, Mitsubishi Tanabe Pharma Corporation, 3-16-89, Kashima, Yodogawa-ku, Osaka 532-8505, Japan.
| | - Jennifer Dressman
- Fraunhofer Institute of Molecular Biology and Applied Ecology (IME), Division of Translational Pharmacology and Medicine (TMP), and Goethe University, Max-von-Laue Straße 9, D-60438 Frankfurt am Main, Germany
| |
Collapse
|
33
|
Parrott N, Suarez-Sharp S, Kesisoglou F, Pathak SM, Good D, Wagner C, Dallmann A, Mullin J, Patel N, Riedmaier AE, Mitra A, Raines K, Butler J, Kakhi M, Li M, Zhao Y, Tsakalozou E, Flanagan T, Dressman J, Pepin X. Best Practices in the Development and Validation of Physiologically Based Biopharmaceutics Modeling. A Workshop Summary Report. J Pharm Sci 2020; 110:584-593. [PMID: 33058891 DOI: 10.1016/j.xphs.2020.09.058] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/29/2020] [Accepted: 09/29/2020] [Indexed: 12/16/2022]
Abstract
This workshop report summarizes the proceedings of Day 2 of a three-day workshop on "Current State and Future Expectations of Translational Modeling Strategies toSupportDrug Product Development, Manufacturing Changes and Controls". From a drug product quality perspective, physiologically based biopharmaceutics modeling (PBBM) is a tool to link variations in the drug product quality attributes to in vivo outcomes enabling the establishment of clinically relevant drug product specifications (CRDPS). Day 2 of the workshop focused on best practices in developing, verifying and validating PBBM. This manuscript gives an overview of podium presentations and summarizes breakout (BO) session discussions related to (1) challenges and opportunities for using PBBM to assess the clinical impact of formulation and manufacturing changes on the in vivo performance of a drug product, (2) best practices to account for parameter uncertainty and variability during model development, (3) best practices in the development, verification and validation of PBBM and (4) opportunities and knowledge gaps related to leveraging PBBM for virtual bioequivalence simulations.
Collapse
Affiliation(s)
- Neil Parrott
- Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd. Grenzacherstrasse 124, CH-4070 Basel, Switzerland.
| | | | | | | | - David Good
- Biopharmaceutics, Bristol-Myers Squibb, New Brunswick, NJ, USA
| | - Christian Wagner
- Pharmaceutical Technologies, Chemical and Pharmaceutical Development, Merck KGaA, Darmstadt, Germany
| | - André Dallmann
- Clinical Pharmacometrics, Research & Development, Pharmaceuticals, Bayer AG, Leverkusen, Germany
| | - James Mullin
- Simulations Plus Inc., 42505 10th Street West, Lancaster, CA 93534, USA
| | | | | | - Amitava Mitra
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, Spring House, PA, USA
| | - Kimberly Raines
- Division of Biopharmaceutics, Office of New Drug Products, Office of Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - James Butler
- Biopharmaceutics, Drug Product Design & Dev, GlaxoSmithKline R&D, Ware, UK
| | - Maziar Kakhi
- Division of Product Quality Research, Office of Testing and Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Min Li
- Division of Biopharmaceutics, Office of New Drug Products, Office of Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Yang Zhao
- Division of Biopharmaceutics, Office of New Drug Products, Office of Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, MD, USA
| | - Eleftheria Tsakalozou
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Talia Flanagan
- Pharmaceutical Development, UCB Pharma SA, Braine l'Alleud, Belgium
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, Carl-von-Noorden-Platz 9, 60596 Frankfurt am Main, Germany
| | - Xavier Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| |
Collapse
|
34
|
Karatza E, Karalis V. Delay differential equations for the description of Irbesartan pharmacokinetics: A population approach to model absorption complexities leading to dual peaks. Eur J Pharm Sci 2020; 153:105498. [PMID: 32736091 DOI: 10.1016/j.ejps.2020.105498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/27/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Irbesartan is a poorly soluble BCS class II compound with weak acidic properties. After oral administration, dual peaks are noted in its concentration (C) - time (t) profile, a phenomenon that may be attributed to enterohepatic recirculation, gastric emptying and/or other absorption complexities related to its pH- and buffer capacity-dependent dissolution behavior. A population pharmacokinetic model, encompassing delay differential equations, was found the most appropriate approach to describe dual peaks in irbesartan's C-t profiles. Parameters estimated were: the absorption rate constant in the central compartment (ka = 0.304 h-1), the constant time delay between the administration and the absorption (T=1.68 h), the apparent volume of distribution of the central (V1/F = 13.8 L) and peripheral (V2/F = 85.8 L) compartment, the apparent clearance from the central compartment (CL/F = 13.5 L/h), and the inter-compartmental clearance (Q/F = 17.7 L/h). Using simulations, it was made evident that changing the time delay results in significant changes of peak plasma concentrations but not of its blood pressure-lowering effect. In conclusion, delay differential equations may be useful to model dual peaks arising from absorption complexities, while changes of the time delay that reflect physiological processes that take place before absorption may have significant implications in proving bioequivalence.
Collapse
Affiliation(s)
- Eleni Karatza
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece; Institute of Applied and Computational Mathematics (IACM) / Foundation of Research and Technology Hellas (FORTH).
| | - Vangelis Karalis
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15784 Athens, Greece; Institute of Applied and Computational Mathematics (IACM) / Foundation of Research and Technology Hellas (FORTH)
| |
Collapse
|
35
|
Jamei M, Abrahamsson B, Brown J, Bevernage J, Bolger MB, Heimbach T, Karlsson E, Kotzagiorgis E, Lindahl A, McAllister M, Mullin JM, Pepin X, Tistaert C, Turner DB, Kesisoglou F. Current status and future opportunities for incorporation of dissolution data in PBPK modeling for pharmaceutical development and regulatory applications: OrBiTo consortium commentary. Eur J Pharm Biopharm 2020; 155:55-68. [DOI: 10.1016/j.ejpb.2020.08.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 07/03/2020] [Accepted: 08/06/2020] [Indexed: 12/13/2022]
|
36
|
Mudie DM, Stewart AM, Biswas N, Brodeur TJ, Shepard KB, Smith A, Morgen MM, Baumann JM, Vodak DT. Novel High-Drug-Loaded Amorphous Dispersion Tablets of Posaconazole; In Vivo and In Vitro Assessment. Mol Pharm 2020; 17:4463-4472. [PMID: 32835489 DOI: 10.1021/acs.molpharmaceut.0c00471] [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] [Indexed: 01/22/2023]
Abstract
Amorphous solid dispersions (ASDs) can increase the bioavailability of drugs with poor aqueous solubility. However, concentration-sustaining dispersion polymers (CSPs) incorporated in ASDs can result in low drug loading and, therefore, a large dosage-form size or multiple units to meet dose requirements, potentially decreasing patient compliance. To address this challenge, a high-loaded dosage-form (HLDF) architecture for ASDs was developed, in which a drug is first spray-dried with a high glass-transition temperature (Tg) dispersion polymer to facilitate high drug loading while maintaining physical stability. The ASD is then granulated with a CSP designed to extend supersaturation in solution. The HLDF differs from traditional ASD architectures in which the dispersion polymer inside the ASD acts as the CSP. By strategically combining two different polymers, one "inside" and one "outside" the ASD, solubilization performance, physical stability, and overall drug loading are maximized. This study demonstrates in vivo performance of the HLDF architecture using posaconazole as a model drug. Two sizes of HLDF tablets were tested in beagle dogs, along with traditional ASD architecture (benchmark) tablets, ASD tablets without a CSP, and a commercial crystalline oral suspension (Noxafil OS). HLDF tablets performed equivalently to the benchmark tablets, the smaller HLDF tablet being 40% smaller (by mass) than the benchmark tablet. The HLDF tablets doubled the blood plasma AUC relative to Noxafil OS. In line with the in vivo outcome, in vitro results in a multicompartment dissolution apparatus demonstrated similar area under the curve (AUC) values in the intestinal compartment for ASD tablets. However, the in vitro data underpredicted the relative in vivo AUC of Noxafil OS compared to the ASD tablets. This study demonstrated that the HLDF approach can increase drug loadings while achieving good performance for ASD drug products.
Collapse
Affiliation(s)
- Deanna M Mudie
- Lonza Pharma and Biotech, Bend, Oregon 97703, United States
| | | | - Nishant Biswas
- Lonza Pharma and Biotech, Bend, Oregon 97703, United States
| | | | | | - Adam Smith
- Lonza Pharma and Biotech, Bend, Oregon 97703, United States
| | | | - John M Baumann
- Lonza Pharma and Biotech, Bend, Oregon 97703, United States
| | - David T Vodak
- Lonza Pharma and Biotech, Bend, Oregon 97703, United States
| |
Collapse
|
37
|
Matsumura N, Ono A, Akiyama Y, Fujita T, Sugano K. Bottom-Up Physiologically Based Oral Absorption Modeling of Free Weak Base Drugs. Pharmaceutics 2020; 12:E844. [PMID: 32899235 PMCID: PMC7558956 DOI: 10.3390/pharmaceutics12090844] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/24/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022] Open
Abstract
In this study, we systematically evaluated "bottom-up" physiologically based oral absorption modeling, focusing on free weak base drugs. The gastrointestinal unified theoretical framework (the GUT framework) was employed as a simple and transparent model. The oral absorption of poorly soluble free weak base drugs is affected by gastric pH. Alternation of bulk and solid surface pH by dissolving drug substances was considered in the model. Simple physicochemical properties such as pKa, the intrinsic solubility, and the bile micelle partition coefficient were used as input parameters. The fraction of a dose absorbed (Fa) in vivo was obtained by reanalyzing the pharmacokinetic data in the literature (15 drugs, a total of 85 Fa data). The AUC ratio with/without a gastric acid-reducing agent (AUCr) was collected from the literature (22 data). When gastric dissolution was neglected, Fa was underestimated (absolute average fold error (AAFE) = 1.85, average fold error (AFE) = 0.64). By considering gastric dissolution, predictability was improved (AAFE = 1.40, AFE = 1.04). AUCr was also appropriately predicted (AAFE = 1.54, AFE = 1.04). The Fa values of several drugs were slightly overestimated (less than 1.7-fold), probably due to neglecting particle growth in the small intestine. This modeling strategy will be of great importance for drug discovery and development.
Collapse
Affiliation(s)
- Naoya Matsumura
- Minase Research Institute, Ono Pharmaceutical Co., Ltd., 3-1-1 Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - Asami Ono
- Laboratory for Chemistry, Manufacturing, and Control, Pharmaceuticals Production & Technology Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan;
| | - Yoshiyuki Akiyama
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan;
| | - Takuya Fujita
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan;
| | - Kiyohiko Sugano
- Molecular Pharmaceutics Lab., College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan;
| |
Collapse
|
38
|
Biorelevant Two-Stage In Vitro Testing for rDCS Classification and in PBPK Modeling–Case Example Ritonavir. J Pharm Sci 2020; 109:2512-2526. [DOI: 10.1016/j.xphs.2020.04.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
|
39
|
PBPK modeling of CYP3A and P-gp substrates to predict drug-drug interactions in patients undergoing Roux-en-Y gastric bypass surgery. J Pharmacokinet Pharmacodyn 2020; 47:493-512. [PMID: 32710209 DOI: 10.1007/s10928-020-09701-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
Roux-en-Y gastric bypass surgery (RYGBS) is an effective surgical intervention to reduce mortality in morbidly obese patients. Following RYGBS, the disposition of drugs may be affected by anatomical alterations and changes in intestinal and hepatic drug metabolizing enzyme activity. The aim of this study was to better understand the drug-drug interaction (DDI) potential of CYP3A and P-gp inhibitors. The impacts of RYGBS on the absorption and metabolism of midazolam, acetaminophen, digoxin, and their major metabolites were simulated using physiologically-based pharmacokinetic (PBPK) modeling. PBPK models for verapamil and posaconazole were built to evaluate CYP3A- and P-gp-mediated DDIs pre- and post-RYGBS. The simulations suggest that for highly soluble drugs, such as verapamil, the predicted bioavailability was comparable pre- and post-RYGBS. For verapamil inhibition, RYGBS did not affect the fold-change of the predicted inhibited-to-control plasma AUC ratio or predicted inhibited-to-control peak plasma concentration ratio for either midazolam or digoxin. In contrast, the predicted bioavailability of posaconazole, a poorly soluble drug, decreased from 12% pre-RYGBS to 5% post-RYGBS. Compared to control, the predicted posaconazole-inhibited midazolam plasma AUC increased by 2.0-fold pre-RYGBS, but only increased by 1.6-fold post-RYGBS. A similar trend was predicted for pre- and post-RYGBS inhibited-to-control midazolam peak plasma concentration ratios (2.0- and 1.6-fold, respectively) following posaconazole inhibition. Absorption of highly soluble drugs was more rapid post-RYGBS, resulting in higher predicted midazolam peak plasma concentrations, which was further increased following inhibition by verapamil or posaconazole. To reduce the risk of a drug-drug interaction in patients post-RYGBS, the dose or frequency of object drugs may need to be decreased when administered with highly soluble inhibitor drugs, especially if toxicities are associated with plasma peak concentrations.
Collapse
|
40
|
Six years of progress in the oral biopharmaceutics area – A summary from the IMI OrBiTo project. Eur J Pharm Biopharm 2020; 152:236-247. [DOI: 10.1016/j.ejpb.2020.05.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/10/2020] [Indexed: 12/18/2022]
|
41
|
Arora S, Pansari A, Kilford P, Jamei M, Gardner I, Turner DB. Biopharmaceutic In Vitro In Vivo Extrapolation (IVIV_E) Informed Physiologically-Based Pharmacokinetic Model of Ritonavir Norvir Tablet Absorption in Humans Under Fasted and Fed State Conditions. Mol Pharm 2020; 17:2329-2344. [DOI: 10.1021/acs.molpharmaceut.0c00043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Sumit Arora
- Certara UK Limited, Simcyp Division, Level 2-Acero, Sheffield, S1 2BJ, U.K
| | - Amita Pansari
- Certara UK Limited, Simcyp Division, Level 2-Acero, Sheffield, S1 2BJ, U.K
| | - Peter Kilford
- Certara UK Limited, Simcyp Division, Level 2-Acero, Sheffield, S1 2BJ, U.K
| | - Masoud Jamei
- Certara UK Limited, Simcyp Division, Level 2-Acero, Sheffield, S1 2BJ, U.K
| | - Iain Gardner
- Certara UK Limited, Simcyp Division, Level 2-Acero, Sheffield, S1 2BJ, U.K
| | - David B. Turner
- Certara UK Limited, Simcyp Division, Level 2-Acero, Sheffield, S1 2BJ, U.K
| |
Collapse
|
42
|
Suarez-Sharp S, Lindahl A, Heimbach T, Rostami-Hodjegan A, Bolger MB, Ray Chaudhuri S, Hens B. Translational Modeling Strategies for Orally Administered Drug Products: Academic, Industrial and Regulatory Perspectives. Pharm Res 2020; 37:95. [DOI: 10.1007/s11095-020-02814-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022]
|
43
|
Pepin XJH, Parrott N, Dressman J, Delvadia P, Mitra A, Zhang X, Babiskin A, Kolhatkar V, Suarez-Sharp S. Current State and Future Expectations of Translational Modeling Strategies to Support Drug Product Development, Manufacturing Changes and Controls: A Workshop Summary Report. J Pharm Sci 2020; 110:555-566. [PMID: 32380182 DOI: 10.1016/j.xphs.2020.04.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022]
Abstract
The implementation of clinically relevant drug product specifications (CRDPS) depends on establishing a link between in vitro performance and in vivo exposure. The scientific community, including regulatory agencies, relies on biopharmaceutics tools on the in vitro performance side, while to enable the link to in vivo exposure, physiologically based pharmacokinetic (PBPK) modeling offers much promise. However, when it comes to PBPK applications in support of CRDPS, otherwise called physiologically based biopharmaceutics models (PBBM), the tools are not yet at the desired level. Currently, it is not possible to integrate detailed variations in chemistry, manufacturing and controls (CMC) attributes and parameters into these models in a way that can consistently predict their effect on local and systemic drug exposure. Specifically, to achieve the desired level, there is a need to advance the science and policy of PBBM. This manuscript summarizes the proceedings of a three-day workshop where the following themes were discussed: 1) Challenges in the development and implementation of in vitro biopredictive tools needed for successful mechanistic modeling; 2) Best practices in model development, verification and validation; and 3) Appropriate terminology (e.g., PBBM vs. PBPK models for biopharmaceutics applications) and applications of PBBM in support of drug product quality.
Collapse
Affiliation(s)
- Xavier J H Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | | | - Poonam Delvadia
- Division of Biopharmaceutics, Office of New Drug Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Amitava Mitra
- Clinical Development, Sandoz Inc (A Novartis Division), Princeton, NJ, USA
| | - Xinyuan Zhang
- Division of Pharmacometrics, Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Andrew Babiskin
- Division of Quantitative Methods and Modeling, Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Vidula Kolhatkar
- Division of Biopharmaceutics, Office of New Drug Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Sandra Suarez-Sharp
- Division of Biopharmaceutics, Office of New Drug Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
44
|
O’Dwyer PJ, Imanidis G, Box KJ, Reppas C. On the Usefulness of Two Small-Scale In Vitro Setups in the Evaluation of Luminal Precipitation of Lipophilic Weak Bases in Early Formulation Development. Pharmaceutics 2020; 12:pharmaceutics12030272. [PMID: 32188116 PMCID: PMC7151110 DOI: 10.3390/pharmaceutics12030272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/08/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023] Open
Abstract
A small-scale biphasic dissolution setup and a small-scale dissolution-permeation (D-P) setup were evaluated for their usefulness in simulating the luminal precipitation of three lipophilic weak bases—dipyridamole, ketoconazole and itraconazole. The transition from the gastric to intestinal environment was incorporated into both experimental procedures. Emulsification during the biphasic dissolution experiments had a minimal impact on the data, when appropriate risk mitigation steps were incorporated. Precipitation parameters estimated from the in vitro data were inputted into the Simcyp® physiologically based pharmacokinetic (PBPK) modelling software and simulated human plasma profiles were compared with previously published pharmacokinetic data. Average Cmax and AUC values estimated using experimentally derived precipitation parameters from the biphasic experiments deviated from corresponding published actual values less than values estimated using the default simulator parameters for precipitation. The slow rate of transport through the biomimetic membrane in the D-P setup limited its usefulness in forecasting the rates of in vivo precipitation used in the modelling of average plasma profiles.
Collapse
Affiliation(s)
- Patrick J. O’Dwyer
- Pion Inc. (UK) Ltd., Forest Row, East Sussex RH18 5DW, UK; (P.J.O.); (K.J.B.)
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, GR 157 84 Zografou, Greece
| | - Georgios Imanidis
- School of Life Sciences, Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Hofackerstrasse 30, 4132 Muttenz, Switzerland;
- Department of Pharmaceutical Sciences, University of Basel, CH 4056 Basel, Switzerland
| | - Karl J. Box
- Pion Inc. (UK) Ltd., Forest Row, East Sussex RH18 5DW, UK; (P.J.O.); (K.J.B.)
| | - Christos Reppas
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, GR 157 84 Zografou, Greece
- Correspondence: ; Tel.: +30-210-727-4678; Fax: +30-210-727-4027
| |
Collapse
|
45
|
Hens B, Masuy I, Deloose E, Mols R, Tack J, Augustijns P. Exploring the impact of real-life dosing conditions on intraluminal and systemic concentrations of atazanavir in parallel with gastric motility recording in healthy subjects. Eur J Pharm Biopharm 2020; 150:66-76. [PMID: 32113916 DOI: 10.1016/j.ejpb.2020.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 02/21/2020] [Accepted: 02/25/2020] [Indexed: 02/07/2023]
Abstract
This work strived to explore gastrointestinal (GI) dissolution, supersaturation and precipitation of the weakly basic drug atazanavir in humans under different 'real-life' intake conditions. The impact of GI pH and motility on these processes was thoroughly explored. In a cross-over study, atazanavir (Reyataz®) was orally administered to 5 healthy subjects with (i) a glass of water, (ii) a glass of Coca-Cola® and (iii) a glass of water under hypochlorhydric conditions (induced by concomitant intake of a proton-pump inhibitor (PPI)). After intake, GI fluids were aspirated from the stomach and the duodenum and, subsequently, analyzed for atazanavir. In parallel, blood samples were collected to assess systemic concentrations. In general, the results of this study revealed that the acidic gastric pH in combination with gastric residence time played a crucial role in the dissolution of atazanavir along the GI tract. After intake of atazanavir with a glass of water (i.e., reference condition), complete gastric dissolution was observed. After GI transfer, supersaturation was noticed for a limited amount of time (1.25 h). With respect to the Coca-Cola® condition, complete gastric dissolution was also observed. A delay in gastric emptying, highly likely caused by the caloric content (101 kcal), was responsible for delayed arrival of atazanavir into the upper small intestine, creating a longer time window of supersaturated concentrations in the duodenal segment (3.25 h) compared to the water condition. The longer period of supersaturated concentrations resulted in a slightly higher systemic exposure of atazanavir compared to the condition when atazanavir was taken with a glass of water. A remarkable observation was the creation (when the drug was given in the migrating motor complex (MMC) phase 2) or maintenance (when the drug was given in MMC phase 1) of a quiescent phase for up to 80 min. With respect to the PPI condition, negligible gastric and intestinal concentrations were observed, resulting in minimal systemic exposure for all subjects. It can be concluded that gastric pH and residence time play a pivotal role in the intestinal disposition of atazanavir in order to generate sufficiently high concentrations further down in the intestinal tract for a sufficient period of time, thus creating a beneficial driving force for intestinal absorption.
Collapse
Affiliation(s)
- Bart Hens
- Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Imke Masuy
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Eveline Deloose
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Raf Mols
- Drug Delivery and Disposition, KU Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | | |
Collapse
|
46
|
Comparison of induction methods for supersaturation: pH shift versus solvent shift. Int J Pharm 2020; 573:118862. [DOI: 10.1016/j.ijpharm.2019.118862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/06/2019] [Accepted: 11/07/2019] [Indexed: 12/27/2022]
|
47
|
Cristofoletti R, Hens B, Patel N, Esteban VV, Schmidt S, Dressman J. Integrating Drug- and Formulation-Related Properties With Gastrointestinal Tract Variability Using a Product-Specific Particle Size Approach: Case Example Ibuprofen. J Pharm Sci 2019; 108:3842-3847. [PMID: 31539541 DOI: 10.1016/j.xphs.2019.09.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/06/2019] [Accepted: 09/11/2019] [Indexed: 11/18/2022]
Abstract
In the present study, an in vitro-in vivo extrapolation of dissolution integrated to a physiologically based pharmacokinetics modeling approach, considering a product-specific particle size distribution and a self-buffering effect of the drug, is introduced and appears to be a promising translational modeling strategy to support drug product development, manufacturing changes and setting clinically relevant specifications for immediate release formulations containing ibuprofen and other weak acids with similar properties.
Collapse
Affiliation(s)
- Rodrigo Cristofoletti
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida 32827.
| | - Bart Hens
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Nikunjkumar Patel
- Simcyp Limited (A Certara Company), Blades Enterprise Centre, Sheffield, UK
| | - Valvanera Vozmediano Esteban
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida 32827
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, Florida 32827
| | - Jennifer Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
48
|
Insights on role of polymers in precipitation of celecoxib from supersaturated solutions as assessed by focused beam reflectance measurement (FBRM). Eur J Pharm Sci 2019; 137:104983. [PMID: 31271876 DOI: 10.1016/j.ejps.2019.104983] [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: 02/19/2019] [Revised: 06/06/2019] [Accepted: 06/30/2019] [Indexed: 01/23/2023]
Abstract
Supersaturating drug delivery systems (SDDS) have dominated the commercial and academic spheres owing to their potential in overcoming the solubility issue of poorly soluble drugs. Precipitation inhibitors are used as excipients in such formulations which has necessitated the development of supersaturation assays that evaluate their precipitation-inhibition efficacy. Such assays are able to give relative estimates of polymer efficacy ceteris paribus within a given set-up. However, the estimates of different laboratories cannot be compared with each other owing to high variability in procedure. Microarray plate method allows comprehensive replicates and decent statistics that make the method an edge over the other exploratory assays. In the current study, the precipitation-inhibition performance of three polymers on the precipitation of a model BCS class II drug was evaluated using the microarray plate method. Quantitative estimations were made through application of Poisson equation for nucleation rates and area under curve. Insights of the precipitation process at particle level were obtained through focused beam reflectance measurement (FBRM) technique coupled with end-process PVM imaging. Through real-time particle size analysis, FBRM technique demonstrated the potential for discerning the role of polymer as nucleation-inhibitor or crystal growth inhibitor. The events observed in the scaled-up FBRM analysis could be correlated with the events observed visually and spectrophotometrically. Powder X-ray diffraction and scanning electron microscopy were performed to capture the influence of polymers on the precipitates formed. This study was able to demonstrate the applicability of microarray plate method for quantitative estimations of precipitation kinetics that can be utilized for excipient screening for poorly soluble drugs having intra-luminal precipitation as a problem. FBRM analysis is highly valuable to gain mechanistic insights and put to rest the prevalent conjecture-based role attribution for polymers.
Collapse
|
49
|
Berben P, Ashworth L, Beato S, Bevernage J, Bruel JL, Butler J, Dressman J, Schäfer K, Hutchins P, Klumpp L, Mann J, Nicolai J, Ojala K, Patel S, Powell S, Rosenblatt K, Tomaszewska I, Williams J, Augustijns P. Biorelevant dissolution testing of a weak base: Interlaboratory reproducibility and investigation of parameters controlling in vitro precipitation. Eur J Pharm Biopharm 2019; 140:141-148. [DOI: 10.1016/j.ejpb.2019.04.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 02/27/2019] [Accepted: 04/29/2019] [Indexed: 12/19/2022]
|
50
|
Successful oral delivery of poorly water-soluble drugs both depends on the intraluminal behavior of drugs and of appropriate advanced drug delivery systems. Eur J Pharm Sci 2019; 137:104967. [PMID: 31252052 DOI: 10.1016/j.ejps.2019.104967] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/27/2019] [Accepted: 06/21/2019] [Indexed: 12/11/2022]
Abstract
Poorly water-soluble drugs continue to be a problematic, yet important class of pharmaceutical compounds for treatment of a wide range of diseases. Their prevalence in discovery is still high, and their development is usually limited by our lack of a complete understanding of how the complex chemical, physiological and biochemical processes that occur between administration and absorption individually and together impact on bioavailability. This review defines the challenge presented by these drugs, outlines contemporary strategies to solve this challenge, and consequent in silico and in vitro evaluation of the delivery technologies for poorly water-soluble drugs. The next steps and unmet needs are proposed to present a roadmap for future studies for the field to consider enabling progress in delivery of poorly water-soluble compounds.
Collapse
|