1
|
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
|
2
|
Ding X, Sun S, Zhang J, Zhao H, Lun F, Liu X, Zhen Y, Dong J, Wu J. Ferric citrate for the treatment of hyperphosphatemia and iron deficiency anaemia in patients with NDD-CKD: a systematic review and meta-analysis. Front Pharmacol 2024; 15:1285012. [PMID: 38515853 PMCID: PMC10955115 DOI: 10.3389/fphar.2024.1285012] [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: 08/29/2023] [Accepted: 02/27/2024] [Indexed: 03/23/2024] Open
Abstract
Background: The application of ferric citrate therapy has yielded unexpected benefits in recent years for Chronic kidney disease patients suffering from hyperphosphatemia and iron deficiency -anaemia. Despite this, earlier research on the impact of ferric citrate on NDD-CKD has been contentious. Objective: The goal of the meta-analysis is to evaluate the evidence regarding the advantages and dangers of ferric citrate for the treatment of hyperphosphatemia and iron deficiency anaemia in NDD-CKD patients. Methods: Between the start of the study and June 2022, we searched PubMed, Embase, Cochrane, EBSCO, Scopus, Web of Science, Wan Fang Data, CNKI, and VIP databases for randomised controlled trials of iron citrate for hyperphosphatemia and anaemia in patients with NDD-CKD. For binary categorical data, risk ratios (OR) were employed, and for continuous variables, weighted mean differences The effect sizes for both count and measurement data were expressed using 95% confidence intervals Results: The meta-analysis includes eight trials with a total of 1281 NDD-CKD patients. The phosphorus-lowering effect of ferric citrate was greater compared to the control group (WMD, -0.55, 95% CI, -0.81 to -0.28; I2 = 86%, p < 0.001). Calcium (WMD, 0.092; 95% CI, -0.051 to 0.234; p > 0.05; I2 = 61.9%), PTH (WMD, -0.10; 95% CI, -0.44 to 0.23; I2 = 75%, p > 0.05) and iFGF23 (WMD, -7.62; 95% CI, -21.18 to 5.94; I2 = 20%, p > 0.05) levels were not statistically different after ferric citrate treatment compared to control treatment. Furthermore, ferric citrate increased iron reserves and haemoglobin. The ferric citrate group had considerably greater levels than the controls. Ferric citrate, on the other hand, may raise the risk of constipation, diarrhoea, and nausea. Conclusion: This meta-analysis found that ferric citrate had a beneficial effect in the treatment of NDD-CKD, particularly in reducing blood phosphorus levels when compared to a control intervention. It also shown that ferric citrate has a favourable effect on iron intake and anaemia management. In terms of safety, ferric citrate may increase the likelihood of gastrointestinal side effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jinping Dong
- Medical College, Weifang University of Science and Technology, Weifang, China
| | - Jingliang Wu
- Medical College, Weifang University of Science and Technology, Weifang, China
| |
Collapse
|
3
|
Moens F, Larsson A, De Blaiser A, Vandevijver G, Spreafico F, Nicolas JM, Lacombe L, Segregur D, Flanagan T, Berben P. Contribution of the Dynamic Intestinal Absorption Model (Diamod) to the Development of a Patient-Centric Drug Formulation. Mol Pharm 2023; 20:6197-6212. [PMID: 37955627 DOI: 10.1021/acs.molpharmaceut.3c00607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Compound X is a weak basic drug targeting the early stages of Parkinson's disease, for which a theoretical risk assessment has indicated that elevated gastric pH conditions could potentially result in reduced plasma concentrations. Different in vitro dissolution methodologies varying in level of complexity and a physiologically based pharmacokinetic (PBPK) absorption model demonstrated that the dissolution, solubility, and intestinal absorption of compound X was indeed reduced under elevated gastric pH conditions. These observations were confirmed in a crossover pharmacokinetic study in Beagle dogs. As a result, the development of a formulation resulting in robust performance that is not sensitive to the exposed gastric pH levels is of crucial importance. The dynamic intestinal absorption MODel (Diamod), an advanced in vitro gastrointestinal transfer tool that allows to study the gastrointestinal dissolution and interconnected permeation of drugs, was selected as an in vitro tool for the formulation optimization activities given its promising predictive capacity and its capability to generate insights into the mechanisms driving formulation performance. Different pH-modifiers were screened for their potential to mitigate the pH-effect by decreasing the microenvironmental pH at the dissolution surface. Finally, an optimized formulation containing a clinically relevant dose of the drug and a functional amount of the selected pH-modifier was evaluated for its performance in the Diamod. This monolayer tablet formulation resulted in rapid gastric dissolution and supersaturation, inducing adequate intestinal supersaturation and permeation of compound X, irrespective of the gastric acidity level in the stomach. In conclusion, this study describes the holistic biopharmaceutics approach driving the development of a patient-centric formulation of compound X.
Collapse
Affiliation(s)
| | - Adam Larsson
- ProDigest BV, Technologiepark 82, 9052 Ghent, Belgium
| | | | | | | | - Jean-Marie Nicolas
- UCB Pharma SA, Early Solutions, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| | - Lucie Lacombe
- UCB Pharma SA, Product Design & Performance, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| | - Domagoj Segregur
- UCB Pharma SA, Product Design & Performance, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| | - Talia Flanagan
- UCB Pharma SA, Product Design & Performance, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| | - Philippe Berben
- UCB Pharma SA, Product Design & Performance, Chemin du Foriest 1, 1420 Braine l'Alleud, Belgium
| |
Collapse
|
4
|
Raines K, Agarwal P, Augustijns P, Alayoubi A, Attia L, Bauer-Brandl A, Brandl M, Chatterjee P, Chen H, Yu YC, Coutant C, Coutinho AL, Curran D, Dressman J, Ericksen B, Falade L, Gao Y, Gao Z, Ghosh D, Ghosh T, Govada A, Gray E, Guo R, Hammell D, Hermans A, Jaini R, Li H, Mandula H, Men S, Milsmann J, Moldthan H, Moody R, Moseson DE, Müllertz A, Patel R, Paudel K, Reppas C, Savkur R, Schaefer K, Serajuddin A, Taylor LS, Valapil R, Wei K, Weitschies W, Yamashita S, Polli JE. Drug Dissolution in Oral Drug Absorption: Workshop Report. AAPS J 2023; 25:103. [PMID: 37936002 DOI: 10.1208/s12248-023-00865-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/02/2023] [Indexed: 11/09/2023] Open
Abstract
The in-person workshop "Drug Dissolution in Oral Drug Absorption" was held on May 23-24, 2023, in Baltimore, MD, USA. The workshop was organized into lectures and breakout sessions. Three common topics that were re-visited by various lecturers were amorphous solid dispersions (ASDs), dissolution/permeation interplay, and in vitro methods to predict in vivo biopharmaceutics performance and risk. Topics that repeatedly surfaced across breakout sessions were the following: (1) meaning and assessment of "dissolved drug," particularly of poorly water soluble drug in colloidal environments (e.g., fed conditions, ASDs); (2) potential limitations of a test that employs sink conditions for a poorly water soluble drug; (3) non-compendial methods (e.g., two-stage or multi-stage method, dissolution/permeation methods); (4) non-compendial conditions (e.g., apex vessels, non-sink conditions); and (5) potential benefit of having both a quality control method for batch release and a biopredictive/biorelevant method for biowaiver or bridging scenarios. An identified obstacle to non-compendial methods is the uncertainty of global regulatory acceptance of such methods.
Collapse
Affiliation(s)
- Kimberly Raines
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Payal Agarwal
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, ON2 Herestraat 49-Box 921, 3000, Leuven, Belgium
| | - Alaadin Alayoubi
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Lucas Attia
- Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, Massachusetts, 02139, USA
| | | | - Martin Brandl
- University of Southern Denmark, Campusvej 55, 5230, Odense, Denmark
| | - Parnali Chatterjee
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Hansong Chen
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Yuly Chiang Yu
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Carrie Coutant
- Eli Lilly and Company, 893 Delaware St, Indianapolis, Indiana, 46225, USA
| | | | - David Curran
- GlaxoSmithKline, 1250 S. Collegeville Road, Collegeville, Pennsylvania, 19046, USA
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Pharmacology and Medicine, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Bryan Ericksen
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Leah Falade
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Yi Gao
- AbbVie Inc, 1 North Waukegan Road, North Chicago, Illinois, 60064, USA
| | - Zongming Gao
- Food and Drug Administration, Center for Drug Evaluation and Research, St. Louis, Missouri, USA
| | - Debasis Ghosh
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Tapash Ghosh
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Anitha Govada
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Elizabeth Gray
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Ruiqiong Guo
- Takeda Pharmaceuticals, 650 E Kendall St, Cambridge, Massachusetts, 02142, USA
| | - Dana Hammell
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Andre Hermans
- Merck & Co. Inc., 2025 E Scott Ave, Rahway, New Jersey, 07065, USA
| | - Rohit Jaini
- Pfizer Inc., 1 Portland St, Cambridge, Massachusetts, 02139, USA
| | - Hanlin Li
- Vertex Pharmaceuticals, 50 Northern Ave, Boston, Massachusetts, 02210, USA
| | - Haritha Mandula
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Shuaiqian Men
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Johanna Milsmann
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400, Biberach an der Riss, Germany
| | - Huong Moldthan
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Rebecca Moody
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Dana E Moseson
- Pfizer Inc., 558 Eastern Point Rd., Groton, Connecticut, 06340, USA
| | - Anette Müllertz
- University of Copenhagen, Nørregade 10, 1165, København, Denmark
| | - Roshni Patel
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Kalpana Paudel
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Christos Reppas
- National and Kapodistrian University of Athens, 157 72, Athens, Greece
| | - Rajesh Savkur
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | - Kerstin Schaefer
- Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Str. 65, 88400, Biberach an der Riss, Germany
| | - Abu Serajuddin
- Department of Pharmaceutical Sciences, St John's University, 8000 Utopia Parkway, Queens, New York, USA
| | - Lynne S Taylor
- Purdue University, 610 Purdue Mall, West Lafayette, Indiana, 47907, USA
| | - Rutu Valapil
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Kevin Wei
- Food and Drug Administration, Center for Drug Evaluation and Research, White Oak, Maryland, USA
| | | | - Shinji Yamashita
- Ritsumeikan University, 56-1 Tojiin Kitamachi, Kita Ward, Kyoto, 603-8577, Japan
| | - James E Polli
- University of Maryland School of Pharmacy, Baltimore, Maryland, USA.
| |
Collapse
|
5
|
Hens B, Sarcevica I, Tomaszewska I, McAllister M. Digitalizing the TIM-1 Model Using Computational Approaches─Part Two: Digital TIM-1 Model in GastroPlus. Mol Pharm 2023; 20:5429-5439. [PMID: 37878668 DOI: 10.1021/acs.molpharmaceut.3c00423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
A TIM-1 model is an in vitro gastrointestinal (GI) simulator considering crucial physiological parameters that will affect the in vivo drug release process. The outcome of these experiments can indicate the critical bioavailability attributes (CBAs) that will impact the fraction absorbed in vivo. The model is widely used in the nonclinical stage of drug product development to assess the bioaccessible fraction of drugs for numerous candidate formulations. In this work, we developed a digital TIM-1 model in the GastroPlus platform. In a first step, we performed validation experiments to assess the luminal concentrations and bioaccessible fractions for two marker compounds. The digital TIM-1 was able to adequately reflect the luminal concentrations and bioaccessible fractions of these markers under different prandial conditions, confirming the appropriate integration of mass transfer in the TIM-1 model. In a second set of experiments, a case example with PF-07059013 was performed, where luminal concentrations and bioaccessible fractions were predicted for 200 and 1000 mg doses under fasted and achlorhydric conditions. Experimental and simulated data pointed out that the achlorhydric effect was more pronounced at the 1000 mg dose, showing a solubility-limited dissolution and, consequently, decreased bioaccessible fraction. Toward future applications, the digital TIM-1 model will be thoroughly applied to explore a link between in vitro and in vivo outcomes based on more case examples with model compounds with the access of TIM-1 and plasma data. Ideally, this digital TIM-1 can be directly used in GastroPlus to explore an in vitro-in vivo correlation (IVIVC) between the fraction dissolved (digital TIM-1 settings) and the fraction absorbed (human PBPK settings).
Collapse
Affiliation(s)
- Bart Hens
- Drug Product Design, Pfizer, Discovery Park, Ramsgate Road, Sandwich CT13 9ND, U.K
| | - Inese Sarcevica
- Drug Product Design, Pfizer, Discovery Park, Ramsgate Road, Sandwich CT13 9ND, U.K
| | - Irena Tomaszewska
- Drug Product Design, Pfizer, Discovery Park, Ramsgate Road, Sandwich CT13 9ND, U.K
| | - Mark McAllister
- Drug Product Design, Pfizer, Discovery Park, Ramsgate Road, Sandwich CT13 9ND, U.K
| |
Collapse
|
6
|
Zhao L, Sun D, Tan ML, Xu M, Kinjo M, Feng K, Wang H, Lionberger R. Effect of Omeprazole Administration on the Pharmacokinetics of Oral Extended-Release Nifedipine in Healthy Subjects. Clin Pharmacol Ther 2023; 114:1134-1141. [PMID: 37669218 DOI: 10.1002/cpt.3043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 08/05/2023] [Indexed: 09/07/2023]
Abstract
Oral extended-release (ER) dosage forms have been used to sustain blood drug levels, reduce adverse events, and improve patient compliance. We investigated potential effects of comedication on pharmacokinetic exposure of nifedipine ER products with different formulation designs and manufacturing processes. A clinical study compared a generic version of nifedipine ER tablet with pH-dependent dissolution behavior with an osmotic pump product with pH independent drug release under fasting condition. In this study, two nifedipine tablet products were tested with or without short-term omeprazole comedication in healthy subjects. Seven-day administration of omeprazole before nifedipine dosing significantly increased the gastric pH, and subsequently increased the geometric least square (LS) means of area under the concentration-time curve from time zero to the last measurable timepoint (AUC0-t ) and maximum plasma concentration (Cmax ) of nifedipine to 132.6% (90% confidence interval (CI): 120.6-145.7%) and 112.8% (90% CI: 100.8-126.3%) for pH-dependent ER tablets, and 120.6% (90% CI: 109.7-132.5%) and 122.5% (90% CI: 113.7-131.9%) for the pH-independent ER tablets, respectively. Similar extent of increase in AUC0-t and Cmax was confirmed in the subpopulations whose gastric pH was ≥ 4 or ≤ 3 in subjects with or without omeprazole administration. Given that similar increases in drug exposures were observed for both pH-dependent and pH-independent nifedipine formulations and the geometric LS mean ratios were between 112% and 133% with and without short-term omeprazole comedication, the gastric pH may have limited effects on omeprazole-induced nifedipine PK changes on the tested formulations. The inhibition of cytochrome P450 3A4 activity may play a significant role causing nifedipine exposure changes for both formulations, which would warrant additional assessment.
Collapse
Affiliation(s)
- Liang Zhao
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Dajun Sun
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Ming-Liang Tan
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Mingjiang Xu
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Minori Kinjo
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Kairui Feng
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hezhen Wang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Robert Lionberger
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| |
Collapse
|
7
|
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
|
8
|
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: 8.0] [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
|
9
|
Kuminek G, Salehi N, Waltz NM, Sperry DC, Greenwood DE, Hate SS, Amidon GE. Use of Gastrointestinal Simulator, Mass Transport Analysis, and Absorption Simulation to Investigate the Impact of pH Modifiers in Mitigating Weakly Basic Drugs' Performance Issues Related to Gastric pH: Palbociclib Case Study. Mol Pharm 2023; 20:147-158. [PMID: 36367432 DOI: 10.1021/acs.molpharmaceut.2c00545] [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: 11/13/2022]
Abstract
It is well known that reduced gastric acidity, for example with concomitant administration of acid reducing agents, can result in variable pharmacokinetics and decreased absorption of weakly basic drugs. It is important to identify the risk of reduced and variable absorption early in development, so that product design options to address the risk can be considered. This article describes the utilization of in vitro and in silico tools to predict the effect of gastric pH, as well as the impact of adding pH modifiers, in mitigating the effect of acid reducing agents on weak base drugs' dissolution and absorption. Palbociclib, a weakly basic drug, was evaluated in low and high gastric pH conditions in a multicompartmental dissolution apparatus referred to as a gastrointestinal simulator (GIS). The GIS permits the testing of pharmaceutical products in a way that better assesses dissolution under physiologically relevant conditions of pH, buffer concentration, formulation additives, and physiological variations including GI pH, buffer concentrations, secretions, stomach emptying rate, residence time in the GI, and aqueous luminal volume. To predict drug dissolution in the GIS, a hierarchical mass transport model was used and validated using in vitro experimental data. Dissolution results were then compared to observed human clinical plasma data with and without proton pump inhibitors using a GastroPlus absorption model to predict palbociclib plasma profiles and pharmacokinetic parameters. The results showed that the in silico model successfully predicted palbociclib dissolution in the GIS under low and high gastric pH conditions with and without pH modifiers. Furthermore, the GIS data coupled with the in silico tools anticipated (1) the reduced palbociclib exposure due to proton pump inhibitor coadministration and (2) the mitigating effect of a pH-modifying agent. This study provides tools to help in the development of orally administered formulations to overcome the effect of elevated gastric pH, especially when formulating with pH modifiers.
Collapse
Affiliation(s)
- Gislaine Kuminek
- Synthetic Molecule Design & Development, Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana46285, United States.,Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan48109, United States
| | - Niloufar Salehi
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan48109, United States.,Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan48109, United States
| | - Nicholas M Waltz
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan48109, United States.,College of Pharmacy, Ohio State University, Columbus, Ohio43210, United States
| | - David C Sperry
- Synthetic Molecule Design & Development, Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Dale E Greenwood
- Synthetic Molecule Design & Development, Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Siddhi S Hate
- Synthetic Molecule Design & Development, Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, Indiana46285, United States
| | - Gregory E Amidon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan48109, United States
| |
Collapse
|
10
|
The impact of viscosity on the dissolution of naproxen immediate-release tablets. J Taibah Univ Med Sci 2022; 18:687-695. [PMID: 36852245 PMCID: PMC9957778 DOI: 10.1016/j.jtumed.2022.12.009] [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: 08/02/2022] [Revised: 10/12/2022] [Accepted: 12/17/2022] [Indexed: 12/31/2022] Open
Abstract
Objectives The increase in viscosity of gastric fluid as a result of food ingestion is one criterion that can negatively impact the dissolution and solubility of orally administered medications. Consequently, it is crucial to address this issue in the pharmacokinetic profile assessment of oral formulations. In this scientific work, various kinds of viscosity enhancers, namely carboxy methylcellulose, pectin, guar gum, and xanthan, were applied to the preparation of different media similar to the biological condition after a meal, and their impacts on the rate of naproxen dissolution and its saturation solubility were evaluated. Methods A Brookfield viscometer was used to assess the rheological features of two potencies of each viscosity booster dissolved in fed state simulated intestinal fluid (FeSSIF). After 24 h of samples shaking, the saturation solubility of the selected medicine in the assessed media was measured using an ultraviolet spectrophotometer, and investigation of the drug dissolution profile was performed with a paddle dissolution apparatus in 200 mL of fluid. Results Great reduction in the saturation solubility of naproxen was detected when the viscosity of the tested media was increased and the highest reduction of solubility was observed with pectin in FeSSIF. Similarly, the dissolution profile of naproxen decrease with enhancement of the viscosity of investigated media. Conclusion A polymer structure not only enhances the viscosity of media but also interferes with drug solubilization. As a result, it is essential to address the rheological aspect in designing in vitro media during the assessment of drug dissolution profiles.
Collapse
|
11
|
Sharma S, Pepin X, Burri H, Zheng L, Kuptsova-Clarkson N, de Jong A, Yu T, MacArthur HL, Majewski M, Byrd JC, Furman RR, Ware JA, Mann J, Ramies D, Munugalavadla V, Sheridan L, Tomkinson H. Bioequivalence and Relative Bioavailability Studies to Assess a New Acalabrutinib Formulation That Enables Coadministration With Proton-Pump Inhibitors. Clin Pharmacol Drug Dev 2022; 11:1294-1307. [PMID: 36029150 DOI: 10.1002/cpdd.1153] [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: 03/29/2022] [Accepted: 07/11/2022] [Indexed: 01/27/2023]
Abstract
Acalabrutinib is a Bruton tyrosine kinase (BTK) inhibitor approved to treat adults with chronic lymphocytic leukemia, small lymphocytic lymphoma, or previously treated mantle cell lymphoma. As the bioavailability of the acalabrutinib capsule (AC) depends on gastric pH for solubility and is impaired by acid-suppressing therapies, coadministration with proton-pump inhibitors (PPIs) is not recommended. Three studies in healthy subjects (N = 30, N = 66, N = 20) evaluated the pharmacokinetics (PKs), pharmacodynamics (PDs), safety, and tolerability of acalabrutinib maleate tablet (AT) formulated with pH-independent release. Subjects were administered AT or AC (orally, fasted state), AT in a fed state, or AT in the presence of a PPI, and AT or AC via nasogastric (NG) route. Acalabrutinib exposures (geometric mean [% coefficient of variation, CV]) were comparable for AT versus AC (AUCinf 567.8 ng h/mL [36.9] vs 572.2 ng h/mL [38.2], Cmax 537.2 ng/mL [42.6] vs 535.7 ng/mL [58.4], respectively); similar results were observed for acalabrutinib's active metabolite (ACP-5862) and for AT-NG versus AC-NG. The geometric mean Cmax for acalabrutinib was lower when AT was administered in the fed versus the fasted state (Cmax 255.6 ng/mL [%CV, 46.5] vs 504.9 ng/mL [49.9]); AUCs were similar. For AT + PPI, geometric mean Cmax was lower (371.9 ng/mL [%CV, 81.4] vs 504.9 ng/mL [49.9]) and AUCinf was higher (AUCinf 694.1 ng h/mL [39.7] vs 559.5 ng h/mL [34.6]) than AT alone. AT and AC were similar in BTK occupancy. Most adverse events were mild with no new safety concerns. Acalabrutinib formulations were comparable and AT could be coadministered with PPIs, food, or via NG tube without affecting the PKs or PDs.
Collapse
Affiliation(s)
| | - Xavier Pepin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, England, UK
| | - Harini Burri
- AstraZeneca, South San Francisco, California, USA
| | | | | | | | - Ting Yu
- AstraZeneca, South San Francisco, California, USA
| | | | | | - John C Byrd
- Department of Internal Medicine and University of Cincinnati Cancer Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Richard R Furman
- New York-Presbyterian/Weill Cornell Medicine, New York, New York, USA
| | | | - James Mann
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, England, UK
| | - David Ramies
- AstraZeneca, South San Francisco, California, USA
| | | | - Louise Sheridan
- Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, England, UK
| | | |
Collapse
|
12
|
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: 3.0] [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
|
13
|
Lex TR, Rodriguez JD, Zhang L, Jiang W, Gao Z. Development of In Vitro Dissolution Testing Methods to Simulate Fed Conditions for Immediate Release Solid Oral Dosage Forms. AAPS J 2022; 24:40. [PMID: 35277760 DOI: 10.1208/s12248-022-00690-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 02/10/2022] [Indexed: 11/30/2022] Open
Abstract
In vitro dissolution testing is widely used to mimic and predict in vivo performance of oral drug products in the gastrointestinal (GI) tract. This literature review assesses the current in vitro dissolution methodologies being employed to simulate and predict in vivo drug dissolution under fasted and fed conditions, with emphasis on immediate release (IR) solid oral dosage forms. Notable human GI physiological conditions under fasted and fed states have been reviewed and summarized. Literature results showed that dissolution media, mechanical forces, and transit times are key dissolution test parameters for simulating specific postprandial conditions. A number of biorelevant systems, including the fed stomach model (FSM), GastroDuo device, dynamic gastric model (DGM), simulated gastrointestinal tract models (TIM), and the human gastric simulator (HGS), have been developed to mimic the postprandial state of the stomach. While these models have assisted in expanding physiological relevance of in vitro dissolution tests, in general, these models lack the ability to fully replicate physiological conditions/processes. Furthermore, the translatability of in vitro data to an in vivo system remains challenging. Additionally, physiologically based pharmacokinetic (PBPK) modeling has been employed to evaluate the effect of food on drug bioavailability and bioequivalence. Here, we assess the current status of in vitro dissolution methodologies and absorption PBPK modeling approaches to identify knowledge gaps and facilitate further development of in vitro dissolution methods that factor in fasted and fed states. Prediction of in vivo drug performance under fasted and fed conditions via in vitro dissolution testing and modeling may potentially help efforts in harmonizing global regulatory recommendations regarding in vivo fasted and fed bioequivalence studies for solid oral IR products.
Collapse
Affiliation(s)
- Timothy R Lex
- Division of Complex Drug Analysis, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, St. Louis, Missouri, 63110, USA
| | - Jason D Rodriguez
- Division of Complex Drug Analysis, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, St. Louis, Missouri, 63110, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20993, USA
| | - Wenlei Jiang
- Office of Research and Standards, Office of Generic Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, 20993, USA.
| | - Zongming Gao
- Division of Complex Drug Analysis, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, St. Louis, Missouri, 63110, USA.
| |
Collapse
|
14
|
O'Dwyer PJ, Box KJ, Imanidis G, Vertzoni M, Reppas C. On the usefulness of four in vitro methods in assessing the intraluminal performance of poorly soluble, ionisable compounds in the fasted state. Eur J Pharm Sci 2021; 168:106034. [PMID: 34628003 PMCID: PMC8665220 DOI: 10.1016/j.ejps.2021.106034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/03/2021] [Accepted: 10/05/2021] [Indexed: 01/12/2023]
Abstract
A small-scale two-stage biphasic system, a small-scale two-stage dissolution-permeation system, the Erweka mini-paddle apparatus, and the BioGIT system were evaluated for their usefulness in assessing the intraluminal performance of two low solubility drugs in the fasted state, one with weakly acidic properties (tested in a salt form, diclofenac potassium) and one with weakly alkaline properties [ritonavir, tested as an amorphous solid dispersion (ASD) formulation]. In all in vitro methods, an immediate-release tablet and a powder formulation of diclofenac potassium were both rapidly dissolved in Level II biorelevant media simulating the conditions in the upper small intestine. Physiologically based biopharmaceutics (PBB) modelling for the tablet formulation resulted in a successful simulation of the average plasma profile in adults, whereas for the powder formulation modelling indicated that gastric emptying and transport through the intestinal epithelium limit the absorption rates. Detailed information on the behaviour of the ritonavir ASD formulation under both simulated gastric and upper small intestinal conditions were crucial for understanding the luminal performance. PBB modelling showed that the dissolution and precipitation parameters, estimated from the Erweka mini-paddle apparatus data and the small-scale two-stage biphasic system data, respectively, were necessary to adequately simulate the average plasma profile after administration of the ritonavir ASD formulation. Simulation of the gastrointestinal transfer process from the stomach to the small intestine was necessary to evaluate the effects of hypochlorhydric conditions on the luminal performance of the ritonavir ASD formulation. Based on this study, the selection of the appropriate in vitro method for evaluating the intraluminal performance of ionisable lipophilic drugs depends on the characteristics of the drug substance. The results suggest that for (salts of) acidic drugs (e.g., diclofenac potassium) it is only an issue of availability and ease of operation of the apparatus. For weakly alkaline substances (e.g., ritonavir), the results indicate that the dynamic dissolution process needs to be simulated, with the type of requested information (e.g., dissolution parameters, precipitation parameters, luminal concentrations) being key for selecting the most appropriate method. Regardless of the ionisation characteristics, early in the drug development process the use of small-scale systems may be inevitable, due to the limited quantities of drug substance available.
Collapse
Affiliation(s)
- Patrick J O'Dwyer
- Pion Inc. (UK) Ltd., Forest Row, East Sussex, United Kingdom; Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece; School of Pharmacy, University College Cork, College Road, Cork, Ireland
| | - Karl J Box
- Pion Inc. (UK) Ltd., Forest Row, East Sussex, United Kingdom
| | - Georgios Imanidis
- University of Applied Sciences Northwest. Switzerland. School of Life Sciences, Institute of Pharma Technology, Hofackerstrasse 30, 4132 Muttenz, Switzerland; University of Basel, Department of Pharmaceutical Sciences, Basel, Switzerland
| | - 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
|
15
|
Segregur D, Mann J, Moir A, Karlsson EM, Dressman J. Biorelevant in vitro Tools and in silico Modeling to Assess pH-Dependent Drug-drug Interactions for Salts of Weak Acids: Case Example Potassium Raltegravir. J Pharm Sci 2021; 111:517-528. [PMID: 34597624 DOI: 10.1016/j.xphs.2021.09.037] [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: 05/18/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Early assessment of pH-dependent drug-drug-interactions (DDIs) for salts of poorly soluble weakly acidic compounds offers various advantages for patient safety, the pharmaceutical industry, and regulatory bodies. Biorelevant media and tests reflecting physiological changes during acid-reducing agent (ARA) co-administration can be used to explore and predict the extent of the pH effect during therapy with ARAs. METHODS Solubility, one-stage and two-stage dissolution of tablets containing potassium raltegravir, the marketed salt form of this poorly soluble, weakly acidic drug, was investigated using biorelevant media specially designed to reflect administration without and during ARA co-therapy. The dissolution data were then converted into parameters suitable for input into an in silico model (Simcyp™) and the simulated plasma profiles were compared with available pharmacokinetic (PK) data from the literature. RESULTS Dissolution of the potassium raltegravir formulation in media reflecting ARA co-administration, and thus elevated gastric pH, was faster and more complete than in experiments reflecting the low gastric pH observed in the absence of ARA co-administration. Simulations using data from dissolution experiments with ARA media appropriately bracketed the in vivo data for ARA co-administration in healthy volunteers. CONCLUSION Dissolution data from in vitro experiments in biorelevant media reflecting physiological changes due to ARA co-administration provide valuable information about potassium raltegravir's behavior during concomitant ARA therapy. The approach may also be suitable for salts forms of other poorly soluble, weakly acidic drugs.
Collapse
Affiliation(s)
- Domagoj Segregur
- Institute of Pharmaceutical Technology, J. W. Goethe University, 9 Max von Laue St., 60438 Frankfurt am Main, Germany (now employed at Product Design and Performance, UCB Pharma, Braine-l'Alleud, Belgium)
| | - James Mann
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, United Kingdom
| | - Andrea Moir
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, United Kingdom
| | - Eva M Karlsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Jennifer Dressman
- Institute of Pharmaceutical Technology, J. W. Goethe University, 9 Max von Laue St., 60438 Frankfurt am Main, Germany (now employed at Product Design and Performance, UCB Pharma, Braine-l'Alleud, Belgium); Fraunhofer Institute of Translational Medicine and Pharmacology, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
16
|
Polybetaines in Biomedical Applications. Int J Mol Sci 2021; 22:ijms22179321. [PMID: 34502230 PMCID: PMC8430529 DOI: 10.3390/ijms22179321] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 12/28/2022] Open
Abstract
Polybetaines, that have moieties bearing both cationic (quaternary ammonium group) and anionic groups (carboxylate, sulfonate, phosphate/phosphinate/phosphonate groups) situated in the same structural unit represent an important class of smart polymers with unique and specific properties, belonging to the family of zwitterionic materials. According to the anionic groups, polybetaines can be divided into three major classes: poly(carboxybetaines), poly(sulfobetaines) and poly(phosphobetaines). The structural diversity of polybetaines and their special properties such as, antifouling, antimicrobial, strong hydration properties and good biocompatibility lead to their use in nanotechnology, biological and medical fields, water remediation, hydrometallurgy and the oil industry. In this review we aimed to highlight the recent developments achieved in the field of biomedical applications of polybetaines such as: antifouling, antimicrobial and implant coatings, wound healing and drug delivery systems.
Collapse
|
17
|
Segregur D, Barker R, Mann J, Moir A, Karlsson EM, Turner DB, Arora S, Dressman J. Evaluating the impact of acid-reducing agents on drug absorption using biorelevant in vitro tools and PBPK modeling - case example dipyridamole. Eur J Pharm Sci 2021; 160:105750. [PMID: 33581261 DOI: 10.1016/j.ejps.2021.105750] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND In vitro and in silico methods have become an essential tool in assessing metabolic drug-drug interactions (DDI) and avoiding reduced efficacy and increased side-effects. Another important type of DDI is the impact of acid-reducing agent (ARA) co-therapy on drug pharmacokinetics due to changes in gastric pH, especially for poorly soluble weakly basic drugs. METHODS One-stage, two-stage and transfer dissolution experiments with dipyridamole tablets using novel biorelevant media representing the ARA effect were conducted and the results were coupled with a PBPK model. Clinical pharmacokinetic data were compared with the simulations from the PBPK model and with output from TIM-1 experiments, an evolved in vitro system which aims to simulate the physiology in the upper GI tract. RESULTS Two-stage and transfer experiments confirmed that these in vitro set-ups tend to overestimate the extent of dipyridamole precipitation occurring in the intestines in vivo. Consequently, data from one-stage dissolution testing under elevated gastric pH conditions were used as an input for PBPK modeling of the ARA/dipyridamole interaction. Using media representing the ARA effect in conjunction with the PBPK model, the ARA effect observed in vivo was successfully bracketed. As an alternative, the TIM-1 system with gastric pH values adjusted to simulate ARA pre-treatment can be used to forecast the ARA effect on dipyridamole pharmacokinetics. CONCLUSION Drug-drug interactions of dipyridamole with ARA were simulated well with a combination of dissolution experiments using biorelevant media representing the gastric environment after an ARA treatment together with the PBPK model. Adjustment of the TIM-1 model to reflect ARA-related changes in gastric pH was also successful in forecasting the interaction. Further testing of both approaches for predicting ARA-related DDIs using a wider range of drugs should be conducted to verify their utility for this purpose.
Collapse
Affiliation(s)
- Domagoj Segregur
- Institute of Pharmaceutical Technology, J. W. Goethe University, 9 Max von Laue St., 60438, Frankfurt am Main, Germany
| | - Richard Barker
- New Modalities & Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, United Kingdom
| | - James Mann
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, United Kingdom
| | - Andrea Moir
- New Modalities & Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, United Kingdom
| | - Eva M Karlsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - David B Turner
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom
| | - Sumit Arora
- Certara UK Limited, Simcyp Division, Sheffield, United Kingdom
| | - Jennifer Dressman
- Institute of Pharmaceutical Technology, J. W. Goethe University, 9 Max von Laue St., 60438, Frankfurt am Main, Germany; Fraunhofer Institute of Translational Medicine and Pharmacology, Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.
| |
Collapse
|
18
|
Liu J, Nagapudi K, Dolton MJ, Chiang PC. Utilizing Tiny-TIM to Assess the Effect of Acid-Reducing Agents on the Absorption of Orally Administered Drugs. J Pharm Sci 2021; 110:3020-3026. [PMID: 33940027 DOI: 10.1016/j.xphs.2021.04.017] [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: 03/25/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 10/21/2022]
Abstract
Acid-reducing agents (ARAs) are the most commonly used medicines to treat patients with gastric acid-related disorders. ARA administration results in an elevation of intragastric pH and eases symptoms such as acid reflux. However, this effect could also lead to a reduction in the absorption of some co-administered oral medications (i.e. weakly basic drugs) by decreasing their gastric solubility. This in turn can result in a significant reduction of the efficacy of the co-administered oral medications. In order to address this problem, substantial efforts in translational modeling and the development of predictive in-vitro assays to better forecast the effect of ARA on oral absorption are conducted in the pharmaceutical industry. Despite these efforts, it remains challenging to predict the impact of ARAs on co-administered drugs. In this study, we evaluated the utility of Triskelion's Gastro-Intestinal Model (Tiny-TIM) in predicting ARA effect on twelve model drugs whose in-vivo data are available. The Tiny-TIM prediction of the ARA effect matched the observed effect of ARA co-administration in humans for the 12 model compounds. In summary, Tiny-TIM is a very reliable and promising GI model to successfully predict the nature of DDI when ARAs are co-administered with the drug of interest.
Collapse
Affiliation(s)
- Jia Liu
- Small Molecule Pharmaceutical Sciences, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Karthik Nagapudi
- Small Molecule Pharmaceutical Sciences, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Michael J Dolton
- Roche Products Australia Pty Ltd, Level 8, 30-34 Hickson Road, Sydney, NSW 2000 Australia
| | - Po-Chang Chiang
- Small Molecule Pharmaceutical Sciences, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
19
|
Segregur D, Mann J, Moir A, Karlsson EM, Dressman J. Prediction of plasma profiles of a weakly basic drug after administration of omeprazole using PBPK modeling. Eur J Pharm Sci 2021; 158:105656. [PMID: 33253885 DOI: 10.1016/j.ejps.2020.105656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/24/2020] [Accepted: 11/24/2020] [Indexed: 11/27/2022]
Abstract
BACKGROUND Oral medicines must release the drug appropriately in the GI tract in order to assure adequate and reproducible absorption. Disease states and co-administration of drugs may alter GI physiology and therefore the release profile of the drug. Acid-reducing agents (ARAs), especially proton pump inhibitors (PPIs), are frequently co-administered during various therapies. As orally administered drugs are frequently poorly soluble weak bases, PPI co-administration raises the risk of pH-induced drug-drug interactions (DDIs) and the potential for changes in the therapeutic outcome. METHODS This research compared the dissolution data of a poorly soluble weakly basic drug ("PSWB 001") from capsules in standard fasted state biorelevant media (FaSSGF, FaSSIF V1 and FaSSIF V2), water and recently devised media representing gastric conditions under various levels of PPI co-administration. An in silico simulation model, based on Simcyp software, was developed to compare simulated plasma profiles with clinical data. RESULTS PSWB 001 capsules showed rapid and complete dissolution in acidic conditions representing gastric fluids, whereas limited dissolution was observed in deionized water, media representing PPI co-administration and in two biorelevant media representing fluids in the upper small intestine. Buffer capacity and the presence of native surfactants were shown to be important factors in the in vitro dissolution of PSWB 001. The data from in vitro experiments were used in conjunction with the in silico simulation model, which correctly predicted the plasma profiles of PSWB 001 when administered without PPIs, as well as bracketing the PPI effect observed in vivo. CONCLUSIONS Recently developed biorelevant media representing gastric conditions under PPI therapy, combined with PBPK modeling, were able to bracket the observed plasma profiles of PSWB 001. These media may also be useful for predicting PPI effects for other poorly soluble, weakly basic drugs.
Collapse
Affiliation(s)
- Domagoj Segregur
- Institute of Pharmaceutical Technology, J. W. Goethe University, 9 Max von Laue St., 60438 Frankfurt am Main, Germany
| | - James Mann
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Andrea Moir
- New Modalities & Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Macclesfield, UK
| | - Eva M Karlsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Jennifer Dressman
- Institute of Pharmaceutical Technology, J. W. Goethe University, 9 Max von Laue St., 60438 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology, ITMP, Theodor Stern Kai 7, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
20
|
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
|
21
|
Zhou LY, Zhu YH, Wang XY, Shen C, Wei XW, Xu T, He ZY. Novel zwitterionic vectors: Multi-functional delivery systems for therapeutic genes and drugs. Comput Struct Biotechnol J 2020; 18:1980-1999. [PMID: 32802271 PMCID: PMC7403891 DOI: 10.1016/j.csbj.2020.07.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/17/2020] [Accepted: 07/18/2020] [Indexed: 02/05/2023] Open
Abstract
Zwitterions consist of equal molar cationic and anionic moieties and thus exhibit overall electroneutrality. Zwitterionic materials include phosphorylcholine, sulfobetaine, carboxybetaine, zwitterionic amino acids/peptides, and other mix-charged zwitterions that could form dense and stable hydration shells through the strong ion-dipole interaction among water molecules and zwitterions. As a result of their remarkable hydration capability and low interfacial energy, zwitterionic materials have become ideal choices for designing therapeutic vectors to prevent undesired biosorption especially nonspecific biomacromolecules during circulation, which was termed antifouling capability. And along with their great biocompatibility, low cytotoxicity, negligible immunogenicity, systematic stability and long circulation time, zwitterionic materials have been widely utilized for the delivery of drugs and therapeutic genes. In this review, we first summarized the possible antifouling mechanism of zwitterions briefly, and separately introduced the features and advantages of each type of zwitterionic materials. Then we highlighted their applications in stimuli-responsive "intelligent" drug delivery systems as well as tumor-targeting carriers and stressed the multifunctional role they played in therapeutic gene delivery.
Collapse
Affiliation(s)
- Ling-Yan Zhou
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Yang-Hui Zhu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Xiao-Yu Wang
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Chao Shen
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
| | - Xia-Wei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China
| | - Ting Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhi-Yao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan 610041, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
22
|
de Waal T, Rubbens J, Grimm M, Vandecaveye V, Tack J, Weitschies W, Brouwers J, Augustijns P. Exploring the Effect of Esomeprazole on Gastric and Duodenal Fluid Volumes and Absorption of Ritonavir. Pharmaceutics 2020; 12:pharmaceutics12070670. [PMID: 32708859 PMCID: PMC7408179 DOI: 10.3390/pharmaceutics12070670] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Proton-pump inhibitors (PPIs), frequently prescribed to lower gastric acid secretion, often exert an effect on the absorption of co-medicated drug products. A previous study showed decreased plasma levels of the lipophilic drug ritonavir after co-administration with the PPI Nexium (40 mg esomeprazole), even though duodenal concentrations were not affected. The present study explored if a PPI-induced decrease in gastrointestinal (GI) fluid volume might contribute to the reduced absorption of ritonavir. In an exploratory cross-over study, five volunteers were given a Norvir tablet (100 mg ritonavir) orally, once without PPI pre-treatment and once after a three-day pre-treatment with the PPI esomeprazole. Blood samples were collected for eight hours to assess ritonavir absorption and magnetic resonance imaging (MRI) was used to determine the gastric and duodenal fluid volumes during the first three hours after administration of the tablet. The results confirmed that PPI intake reduced ritonavir plasma concentrations by 40%. The gastric residual volume and gastric fluid volume decreased by 41% and 44% respectively, while the duodenal fluid volume was reduced by 33%. These data suggest that the PPI esomeprazole lowers the available fluid volume for dissolution, which may limit the amount of ritonavir that can be absorbed. Although additional factors may play a role, the effect of PPI intake on the GI fluid volume should be considered when simulating the absorption of poorly soluble drugs like ritonavir in real-life conditions.
Collapse
Affiliation(s)
- Tom de Waal
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49—Box 921, 3000 Leuven, Belgium; (T.d.W.); (J.R.); (J.B.)
| | - Jari Rubbens
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49—Box 921, 3000 Leuven, Belgium; (T.d.W.); (J.R.); (J.B.)
| | - Michael Grimm
- Center of Drug Absorption and Transport, Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany; (M.G.); (W.W.)
| | | | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders, TARGID, KU Leuven, 3000 Leuven, Belgium;
| | - Werner Weitschies
- Center of Drug Absorption and Transport, Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany; (M.G.); (W.W.)
| | - Joachim Brouwers
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49—Box 921, 3000 Leuven, Belgium; (T.d.W.); (J.R.); (J.B.)
| | - Patrick Augustijns
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49—Box 921, 3000 Leuven, Belgium; (T.d.W.); (J.R.); (J.B.)
- Correspondence:
| |
Collapse
|
23
|
Parrott N, Stillhart C, Lindenberg M, Wagner B, Kowalski K, Guerini E, Djebli N, Meneses-Lorente G. Physiologically Based Absorption Modelling to Explore the Impact of Food and Gastric pH Changes on the Pharmacokinetics of Entrectinib. AAPS JOURNAL 2020; 22:78. [PMID: 32458089 DOI: 10.1208/s12248-020-00463-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/29/2020] [Indexed: 02/07/2023]
Abstract
Entrectinib is a potent and selective tyrosine kinase inhibitor (TKI) of TRKA/B/C, ROS1, and ALK with both systemic and CNS activities, which has recently received FDA approval for ROS1 fusion-positive non-small cell lung cancer and NTRK fusion-positive solid tumors. This paper describes the application of a physiologically based biophamaceutics modeling (PBBM) during clinical development to understand the impact of food and gastric pH changes on absorption of this lipophilic, basic, molecule with reasonable permeability but strongly pH-dependent solubility. GastroPlus™ was used to develop a physiologically based pharmacokinetics (PBPK) model integrating in vitro and in silico data and dissolution studies and in silico modelling in DDDPlus™ were used to understand the role of self-buffering and acidulant on formulation performance. Models were verified by comparison of simulated pharmacokinetics for acidulant and non-acidulant containing formulations to clinical data from a food effect study and relative bioavailability studies with and without the gastric acid-reducing agent lansoprazole. A negligible food effect and minor pH-dependent drug-drug interaction for the market formulation were predicted based on biorelevant in vitro measurements, dissolution studies, and in silico modelling and were confirmed in clinical studies. These outcomes were explained as due to the acidulant counteracting entrectinib self-buffering and greatly reducing the effect of gastric pH changes. Finally, sensitivity analyses with the verified model were applied to support drug product quality. PBBM has great potential to streamline late-stage drug development and may have impact on regulatory questions.
Collapse
Affiliation(s)
- Neil Parrott
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Cordula Stillhart
- Pharmaceutical Research & Development, Formulation & Process Sciences, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marc Lindenberg
- Pharmaceutical Research & Development, Analytical, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Bjoern Wagner
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | | | - Elena Guerini
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Nassim Djebli
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Georgina Meneses-Lorente
- Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| |
Collapse
|
24
|
Prediction of pH-Dependent Drug-Drug Interactions for Basic Drugs Using Physiologically Based Biopharmaceutics Modeling: Industry Case Studies. J Pharm Sci 2020; 109:1380-1394. [DOI: 10.1016/j.xphs.2019.11.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/15/2019] [Accepted: 11/19/2019] [Indexed: 01/16/2023]
|
25
|
Auch C, Jede C, Harms M, Wagner C, Mäder K. Impact of amorphization and GI physiology on supersaturation and precipitation of poorly soluble weakly basic drugs using a small-scale in vitro transfer model. Int J Pharm 2020; 574:118917. [PMID: 31811926 DOI: 10.1016/j.ijpharm.2019.118917] [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: 09/28/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/26/2022]
Abstract
Formulation of amorphous solid dispersions (ASD) is one possibility to improve poor aqueous drug solubility by creating supersaturation. In case of weakly basic drugs like ketoconazole (KTZ), supersaturation can also be generated during the gastrointestinal (GI) transfer from the stomach to the intestine due to pH-dependent solubility. In both cases, the supersaturation during dissolution can be stabilized by polymeric precipitation inhibitors. A small-scale GI transfer model was used to compare the dissolution performance of ASD versus crystalline KTZ with the polymeric precipitation inhibitor HPMCAS. Similar in vitro AUCs were found for the transfer from SGF pH2 into FaSSIF. Moreover, the impact of variability in gastric pH on drug dissolution was assessed. Here, the ASD performed significantly better at a simulated hypochlorhydric gastric pHof 4. Last, the importance of drug-polymer interactions for precipitation inhibition was evaluated. HPMCAS HF and LF grades with and without the basic polymer Eudragit EPO were used. However, EPO caused a faster precipitation probably due to competition for the interaction sites between KTZ and HPMCAS. Thus, the results are suited to assess the benefits of amorphous formulations vs. precipitation inhibitors under different gastrointestinal conditions to optimize the design of such drug delivery systems.
Collapse
Affiliation(s)
- Carolin Auch
- Institute of Pharmacy, Faculty I of Natural Sciences, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany; Department of Pharmaceutical Technologies, Merck Healthcare KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Christian Jede
- Department of Analytical Development, Merck Healthcare KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Meike Harms
- Department of Pharmaceutical Technologies, Merck Healthcare KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Christian Wagner
- Department of Pharmaceutical Technologies, Merck Healthcare KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Karsten Mäder
- Institute of Pharmacy, Faculty I of Natural Sciences, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany.
| |
Collapse
|