1
|
García MA, Paulos C, Ibarra Viñales M, Michelet R, Cabrera-Pérez MÁ, Aceituno A, Bone M, Ibacache M, Cortínez LI, Guzmán M. Modeling and Simulations in Latin-American Generic Markets: Perspectives from Chilean Local Industry, Regulatory Agency, and Academia. Mol Pharm 2024. [PMID: 39454202 DOI: 10.1021/acs.molpharmaceut.4c00764] [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: 10/27/2024]
Abstract
In the last 20 years, modeling and simulations (M&S) have gained increased attention in pharmaceutical sciences. International industry and world-reference agencies have used M&S to make cost-efficient decisions through the model-informed drug development (MIDD) framework. Modeling tools include biopredictive dissolution models, physiologically based pharmacokinetic models (PBPK), biopharmaceutic models (PBBM), and virtual bioequivalence, among many others. Regulatorily, health agencies are becoming more and more open to accept the use of M&S to support regulatory applications, including setting dissolution specifications, quality-by-design (QbD), postapproval changes (SUPAC), etc. Nonetheless, the potential of M&S has been only barely explored in Latin America (Latam) across different actors: industry, regulatory agencies, and even academia. In this manuscript, we discuss the challenges and opportunities for implementing M&S approaches in Latam. Perspectives of regional experts were shared in a workshop. Attendance (professionals from industry, regulator, academia, and clinicians) also shared their views via survey. The rational development of bioequivalent generics was considered the main opportunity for M&S in regional market, particularly the use of PBPK and PBBM. Nonetheless, a critical mass of modeling scientists is needed before Latin American industry and regulators can actually benefit from M&S. Collaborations (e.g., Academia-Industry and Academia-Regulatory) may be a path to develop applied research projects and train the future modelers. Reaching that critical mass, scientists from industry may apply modeling across generic drug development process and life cycle, while regulatory scientists may issue guidelines in local language to support regional industry. Only at that stage could the full potential of MIDD be reached in Latin American generic markets.
Collapse
Affiliation(s)
- Mauricio A García
- Departamento de Farmacia, Escuela de Química y Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Claudio Paulos
- Departamento de Farmacia, Escuela de Química y Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago 7820436, Chile
| | - Manuel Ibarra Viñales
- Department of Pharmaceutical Sciences, Faculty of Chemistry, Universidad de la República, Montevideo 11800, Uruguay
| | - Robin Michelet
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universität Berlin, Kelchstraße 31, Berlin 14195, Germany
- qPharmetra LLC, Berlin 14195, Germany
| | - Miguel Ángel Cabrera-Pérez
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias, Universidad Católica del Norte, Antofagasta 1240000, Chile
| | - Alexis Aceituno
- National Drug Agency Department, Institute of Public Health (ISP), Santiago 7780050, Chile
- University of Valparaíso, Faculty of Pharmacy, Valparaíso 2381850, Chile
| | - Michelle Bone
- National Drug Agency Department, Institute of Public Health (ISP), Santiago 7780050, Chile
| | - Mauricio Ibacache
- División Anestesiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
| | - Luis Ignacio Cortínez
- División Anestesiología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago de Chile 7820436, Chile
| | - Marcelo Guzmán
- Validations and Bioequivalence, Laboratorio Milab, Grupo FEMSA, Santiago 8380000, Chile
| |
Collapse
|
2
|
Kowthavarapu VK, Charbe NB, Gupta C, Iakovleva T, Stillhart C, Parrott NJ, Schmidt S, Cristofoletti R. Mechanistic Modeling of In Vitro Biopharmaceutic Data for a Weak Acid Drug: A Pathway Towards Deriving Fundamental Parameters for Physiologically Based Biopharmaceutic Modeling. AAPS J 2024; 26:44. [PMID: 38575716 DOI: 10.1208/s12248-024-00912-y] [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: 12/14/2023] [Accepted: 03/17/2024] [Indexed: 04/06/2024] Open
Abstract
Mechanistic modeling of in vitro experiments using metabolic enzyme systems enables the extrapolation of metabolic clearance for in vitro-in vivo predictions. This is particularly important for successful clearance predictions using physiologically based pharmacokinetic (PBPK) modeling. The concept of mechanistic modeling can also be extended to biopharmaceutics, where in vitro data is used to predict the in vivo pharmacokinetic profile of the drug. This approach further allows for the identification of parameters that are critical for oral drug absorption in vivo. However, the routine use of this analysis approach has been hindered by the lack of an integrated analysis workflow. The objective of this tutorial is to (1) review processes and parameters contributing to oral drug absorption in increasing levels of complexity, (2) outline a general physiologically based biopharmaceutic modeling workflow for weak acids, and (3) illustrate the outlined concepts via an ibuprofen (i.e., a weak, poorly soluble acid) case example in order to provide practical guidance on how to integrate biopharmaceutic and physiological data to better understand oral drug absorption. In the future, we plan to explore the usefulness of this tutorial/roadmap to inform the development of PBPK models for BCS 2 weak bases, by expanding the stepwise modeling approach to accommodate more intricate scenarios, including the presence of diprotic basic compounds and acidifying agents within the formulation.
Collapse
Affiliation(s)
- Venkata Krishna Kowthavarapu
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Nitin Bharat Charbe
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Churni Gupta
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Tatiana Iakovleva
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Cordula Stillhart
- Pharmaceutical Research & Development, Formulation & Process Development, F. Hoffmann-La Roche Ltd., 4070, Basel, Switzerland
| | - Neil John Parrott
- Pharmaceutical Research & Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., 4070, Basel, Switzerland
| | - Stephan Schmidt
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA
| | - Rodrigo Cristofoletti
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics Lake Nona (Orlando), College of Pharmacy, University of Florida, 6550 Sanger Road, Office 467, Orlando, Florida, 32827, USA.
| |
Collapse
|
3
|
Yoshida H, Morita T, Abe Y, Inagaki A, Tomita N, Izutsu KI, Sato Y. Effects of Apex Size on Dissolution Profiles in the USP II Paddle Apparatus. AAPS PharmSciTech 2023; 25:9. [PMID: 38158516 DOI: 10.1208/s12249-023-02722-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/05/2023] [Indexed: 01/03/2024] Open
Abstract
The use of apex vessels may solve coning problems associated with dissolution testing. However, excessive dissolution acceleration can reduce the discriminatory power. This study aimed to clarify how different apex vessel sizes affect the dissolution behavior of cone-forming formulations. Five apex vessels with different heights, centralities, and compendial vessels were used. The paddle rotation speed at which the coning phenomenon resolved was measured using standard particles of different densities. Three model formulations-USP prednisone tablets, atorvastatin calcium hydrate tablets, and levofloxacin fine granules-were selected, and dissolution tests were conducted at 30-100 revolutions per minute (rpm). Compared to the compendial vessels, the disappearance of standard particles at the apex base at lower paddle speeds in apex vessels was observed. Standard particles tended to remain in the center of the apex vessels and disappear at rotational speeds comparable to those of the compendial vessels. Dissolution increased in an apex height-dependent manner in the model formulations, except for the atorvastatin calcium hydrate tablets at 50 rpm. For levofloxacin fine granules, dissolution was also improved by reducing the paddle agitation speed to 30 rpm in the compendial vessels. Differences in apex centrality by 3 mm did not affect the dissolution rate. Our results indicate that apex vessels with low apex heights have a mount-resolving effect, but the degree of dissolution improvement by avoiding the coning phenomenon depends on the formulation characteristics used in the dissolution tests.
Collapse
Affiliation(s)
- Hiroyuki Yoshida
- Division of Drugs, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan.
| | - Tokio Morita
- Division of Drugs, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Yasuhiro Abe
- Division of Drugs, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Aoi Inagaki
- Division of Drugs, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Naomi Tomita
- Division of Drugs, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| | - Ken-Ichi Izutsu
- Department of Pharmaceutical Sciences, International University of Health and Welfare, Tochigi, Japan
| | - Yoji Sato
- Division of Drugs, National Institute of Health Sciences, 3-25-26, Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan
| |
Collapse
|
4
|
Mansuroglu Y, Dressman J. Factors That Influence Sustained Release from Hot-Melt Extrudates. Pharmaceutics 2023; 15:1996. [PMID: 37514182 PMCID: PMC10386192 DOI: 10.3390/pharmaceutics15071996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Hot-melt extrusion is a well-established tool in the pharmaceutical industry, mostly implemented to increase the solubility of poorly soluble drugs. A less frequent application of this technique is to obtain formulations with extended release. This study investigated the influence of polymer choice, drug loading, milling and hydrodynamics on the release of a model drug, flurbiprofen, from sustained-release hot-melt extrudates with Eudragit polymers. The choice of polymer and degree of particle size reduction of the extrudate by milling were the two key influences on the release profile: the percentage release after 12 h varied from 6% (2 mm threads) to 84% (particle size <125 µm) for Eudragit RL extrudates vs. 4.5 to 62% for the corresponding Eudragit RS extrudates. By contrast, the release profile was largely independent of drug loading and robust to hydrodynamics in the dissolution vessel. Thus, hot-melt extrusion offers the ability to tailor the release of the API to the therapeutic indication through a combination of particle size and polymer choice while providing robustness over a wide range of hydrodynamic conditions.
Collapse
Affiliation(s)
- Yaser Mansuroglu
- Fraunhofer Institute of Translational Medicine and Pharmacology, Theodor-Stern-Kai.7, 60596 Frankfurt am Main, Germany
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, Theodor-Stern-Kai.7, 60596 Frankfurt am Main, Germany
| |
Collapse
|
5
|
Pepin X, McAlpine V, Moir A, Mann J. Acalabrutinib Maleate Tablets: The Physiologically Based Biopharmaceutics Model behind the Drug Product Dissolution Specification. Mol Pharm 2023; 20:2181-2193. [PMID: 36859819 DOI: 10.1021/acs.molpharmaceut.3c00005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Acalabrutinib maleate tablets correspond to an improved formulation compared to acalabrutinib capsules as they can be dosed with and without acid reducing agents and therefore benefit more cancer patients. The dissolution specification for the drug product was determined using all the information available on the drug safety, efficacy, and in vitro performance. In addition, a physiologically based biopharmaceutics model was developed for acalabrutinib maleate tablets on the back of a previously published model for acalabrutinib capsules to establish that the proposed drug product dissolution specification would ensure safe and effective products for all patients including those under acid reducing agent treatment. The model was built, validated, and used to predict the exposure of virtual batches where the dissolution was slower than that of the clinical target. A combination of exposure prediction and the use of a PK-PD model allowed it to be demonstrated that the proposed drug product dissolution specification was acceptable. This combination of models enabled a larger safe space than would have been granted by consideration of bioequivalence only.
Collapse
Affiliation(s)
- Xavier Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Charter Way, SK10 2NA Macclesfield, United Kingdom
| | - Vivien McAlpine
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, SK10 2NA Macclesfield, United Kingdom
| | - Andrea Moir
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, SK10 2NA Macclesfield, United Kingdom
| | - James Mann
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, SK10 2NA Macclesfield, United Kingdom
| |
Collapse
|
6
|
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: 6] [Impact Index Per Article: 6.0] [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
|
7
|
Zhang F, Wu X, Wu K, Yu M, Liu B, Wang H. Predicting the Pharmacokinetics of Orally Administered Drugs across BCS Classes 1-4 by Virtual Bioequivalence Model. Mol Pharm 2023; 20:395-408. [PMID: 36469444 DOI: 10.1021/acs.molpharmaceut.2c00688] [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: 12/12/2022]
Abstract
To evaluate the influence of solubility and permeability on the pharmacokinetic prediction performance of orally administered drugs using avirtual bioequivalence (VBE) model, a total of 23 orally administered drugs covering Biopharmaceutics Classification System (BCS) classes 1-4 were selected. A VBE model (i.e., a physiologically based pharmacokinetic model integrated with dissolution data) based on a B2O simulator was applied for pharmacokinetic (PK) prediction in a virtual population. Parameter sensitivity analysis was used for input parameter selection. The predictive performances of PK parameters (i.e., AUC0-t, Cmax, and Tmax), PK profiles, and bioequivalence (BE) results were evaluated using the twofold error, average fold error (AFE), absolute average fold error (AAFE), and BE reassessment metrics. All models successfully simulated the mean PK profiles, with AAFE < 2 and AFE ranging from 0.58 to 1.66. As for the PK parameters, except for the time of peak concentration, Tmax, of isosorbide mononitrate, other simulated PK parameters were all within a twofold error. The simulated PK behaviors were comparable to the observed ones, both for test (T) and reference (R) products, and the simulated T/R arithmetic mean ratios were all within 0.88-1.16 of the observed values. These four evaluation metrics were distributed equally among BCS class 1-4 drugs. The VBE model showed powerful performance to predict the PK behavior of orally administered drugs with various combinations of solubility and permeability, irrespective of the BCS category.
Collapse
Affiliation(s)
- Fan Zhang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing100730, China
| | - Xiaofei Wu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing100730, China
| | - Keheng Wu
- Yinghan Pharmaceutical Technology (Shanghai) Co., Ltd, Shanghai201100, China
| | - Mengyang Yu
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing100730, China
| | - Bo Liu
- Wuhan Institute of Technology, Wuhan, Hubei430205, China
| | - Hongyun Wang
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing100730, China
| |
Collapse
|
8
|
Anand O, Pepin XJH, Kolhatkar V, Seo P. The Use of Physiologically Based Pharmacokinetic Analyses-in Biopharmaceutics Applications -Regulatory and Industry Perspectives. Pharm Res 2022; 39:1681-1700. [PMID: 35585448 DOI: 10.1007/s11095-022-03280-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/27/2022] [Indexed: 12/18/2022]
Abstract
The use of physiologically based pharmacokinetic (PBPK) modeling to support the drug product quality attributes, also known as physiologically based biopharmaceutics modeling (PBBM) is an evolving field and the interest in using PBBM is increasing. The US-FDA has emphasized on the use of patient centric quality standards and clinically relevant drug product specifications over the years. Establishing an in vitro in vivo link is an important step towards achieving the goal of patient centric quality standard. Such a link can aid in constructing a bioequivalence safe space and establishing clinically relevant drug product specifications. PBBM is an important tool to construct a safe space which can be used during the drug product development and lifecycle management. There are several advantages of using the PBBM approach, though there are also a few challenges, both with in vitro methods and in vivo understanding of drug absorption and disposition, that preclude using this approach and therefore further improvements are needed. In this review we have provided an overview of experience gained so far and the current perspective from regulatory and industry point of view. Collaboration between scientists from regulatory, industry and academic fields can further help to advance this field and deliver on promises that PBBM can offer towards establishing patient centric quality standards.
Collapse
Affiliation(s)
- Om Anand
- 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, Maryland, USA.
| | - Xavier J H Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Vidula Kolhatkar
- 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, Maryland, USA
| | - Paul Seo
- Office of Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| |
Collapse
|
9
|
Wanasathop A, Murawsky M, Kevin Li S. Modification of Small Dissolution Chamber System for Long-acting Periodontal Drug Product Evaluation. Int J Pharm 2022; 618:121646. [PMID: 35259441 PMCID: PMC9136688 DOI: 10.1016/j.ijpharm.2022.121646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
Conventional dissolution testing methods may not be suitable for long-acting periodontal drug products due to the small volume, slow fluid flow rate, and environment in the periodontal pocket. The objective of this study was to evaluate a 3D-printed small volume flow-through dissolution chamber system (modified from a previous study) for biorelevant and dose-discriminating testing. Three periodontal drug products with different dosage forms were tested: Atridox, Arestin, and PerioChip. Modifications were made to suit the specific characteristics of these dosage forms. No significant differences were observed between the % drug release profiles in vitro and in vivo except for Atridox. The differences observed with Atridox could be related to the exposing surface area of the drug product. Similar differences were observed from this effect in COMSOL model simulations. Overall, the drugs show reasonable in vitro-in vivo correlations (R2 ≥ 0.91) with linear regression slopes close to unity. For dose discrimination between 75% and full dosing, significant differences were observed in the drug release data at specific time points of the products (p ≤ 0.05). The present results suggest that a small volume dissolution chamber with slow flow rate could potentially provide biologically relevant and dose-discriminating evaluations for periodontal drug products.
Collapse
|
10
|
In Silico Modeling and Simulation to Guide Bioequivalence Testing for Oral Drugs in a Virtual Population. Clin Pharmacokinet 2021; 60:1373-1385. [PMID: 34191255 DOI: 10.1007/s40262-021-01045-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 12/18/2022]
Abstract
Model-informed drug discovery and development (MID3) shows great advantages in facilitating drug development. A physiologically based pharmacokinetic model is one of the powerful computational approaches of MID3, and the emerging field of virtual bioequivalence is well recognized to be the future of the physiologically based pharmacokinetic model. Based on the translational link between in vitro, in silico, and in vivo, virtual bioequivalence study can evaluate the similarity and potential difference of pharmacokinetic and clinical performance between test and reference formulations. With the aid of virtual bioequivalence study, the pivotal information of clinical trials can be provided to streamline the development for both new and generic drugs. However, a regulatory framework of virtual bioequivalence study has not reached its full maturity. Therefore, this article aims to present an overview of the current status of bioequivalence study, identify the framework of virtual bioequivalence studies for oral drugs, and also discuss the future opportunities of virtual bioequivalence in supporting the waiver and optimization of in vivo clinical trials.
Collapse
|
11
|
Jereb R, Opara J, Bajc A, Petek B. Evaluating the Impact of Physiological Properties of the Gastrointestinal Tract On Drug In Vivo Performance Using Physiologically Based Biopharmaceutics Modeling and Virtual Clinical Trials. J Pharm Sci 2021; 110:3069-3081. [PMID: 33878322 DOI: 10.1016/j.xphs.2021.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 12/13/2022]
Abstract
The physiological properties of the gastrointestinal tract, such as pH, fluid volume, bile salt concentration, and gastrointestinal transit time, are highly variable in vivo. These properties can affect the dissolution and absorption of a drug, depending on its properties and formulation. The effect of gastrointestinal physiology on the bioperformance of a drug was studied in silico for a delayed-release pantoprazole tablet and an immediate-release dolutegravir tablet. Physiologically based absorption models were developed and virtual clinical trials were performed. Reasons for the variability in drug bioperformance between subjects were investigated, taking into account differences in gastrointestinal tract characteristics, pharmacokinetic parameters, and additional parameters (e.g., permeability). Default software parameters describing gastrointestinal physiology in the fasted and fed states, and variation in these parameters, were altered to match variability in these parameters reported in vivo. The altered model physiologies better described the variability of gastrointestinal conditions, and therefore the results of virtual trials using these physiologies are likely to be more relevant in vivo. With such altered gastrointestinal physiologies used to develop models, it is possible to obtain additional knowledge and improve the understanding of subject-formulation interactions.
Collapse
Affiliation(s)
- Rebeka Jereb
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia; Lek Pharmaceuticals d.d., a Sandoz Company, Verovškova ulica 57, 1526 Ljubljana, Slovenia.
| | - Jerneja Opara
- Lek Pharmaceuticals d.d., a Sandoz Company, Verovškova ulica 57, 1526 Ljubljana, Slovenia
| | - Aleksander Bajc
- Lek Pharmaceuticals d.d., a Sandoz Company, Verovškova ulica 57, 1526 Ljubljana, Slovenia
| | - Boštjan Petek
- Lek Pharmaceuticals d.d., a Sandoz Company, Verovškova ulica 57, 1526 Ljubljana, Slovenia
| |
Collapse
|
12
|
Loisios-Konstantinidis I, Dressman J. Physiologically Based Pharmacokinetic/Pharmacodynamic Modeling to Support Waivers of In Vivo Clinical Studies: Current Status, Challenges, and Opportunities. Mol Pharm 2020; 18:1-17. [PMID: 33320002 DOI: 10.1021/acs.molpharmaceut.0c00903] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Physiologically based pharmacokinetic/pharmacodynamic (PBPK/PD) modeling has been extensively applied to quantitatively translate in vitro data, predict the in vivo performance, and ultimately support waivers of in vivo clinical studies. In the area of biopharmaceutics and within the context of model-informed drug discovery and development (MID3), there is a rapidly growing interest in applying verified and validated mechanistic PBPK models to waive in vivo clinical studies. However, the regulatory acceptance of PBPK analyses for biopharmaceutics and oral drug absorption applications, which is also referred to variously as "PBPK absorption modeling" [Zhang et al. CPT: Pharmacometrics Syst. Pharmacol. 2017, 6, 492], "physiologically based absorption modeling", or "physiologically based biopharmaceutics modeling" (PBBM), remains rather low [Kesisoglou et al. J. Pharm. Sci. 2016, 105, 2723] [Heimbach et al. AAPS J. 2019, 21, 29]. Despite considerable progress in the understanding of gastrointestinal (GI) physiology, in vitro biopharmaceutic and in silico tools, PBPK models for oral absorption often suffer from an incomplete understanding of the physiology, overparameterization, and insufficient model validation and/or platform verification, all of which can represent limitations to their translatability and predictive performance. The complex interactions of drug substances and (bioenabling) formulations with the highly dynamic and heterogeneous environment of the GI tract in different age, ethnic, and genetic groups as well as disease states have not been yet fully elucidated, and they deserve further research. Along with advancements in the understanding of GI physiology and refinement of current or development of fully mechanistic in silico tools, we strongly believe that harmonization, interdisciplinary interaction, and enhancement of the translational link between in vitro, in silico, and in vivo will determine the future of PBBM. This Perspective provides an overview of the current status of PBBM, reflects on challenges and knowledge gaps, and discusses future opportunities around PBPK/PD models for oral absorption of small and large molecules to waive in vivo clinical studies.
Collapse
Affiliation(s)
| | - Jennifer Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main 60438, Germany.,Fraunhofer Institute of Translational Pharmacology and Medicine (ITMP), Carl-von-Noorden Platz 9, Frankfurt am Main 60438, Germany
| |
Collapse
|