1
|
Ojuka P, Ochieng CO, Ndarawit W, Nyongesa DW, Mukavi J, Nyabuga Nyariki J, Apollo S, Santos CBR, Kimani NM. Alkaloids Isolated from Vepris glandulosa with Antidiabetic Properties: An In Vitro and In Silico Analysis. Chem Biodivers 2024:e202401515. [PMID: 39495611 DOI: 10.1002/cbdv.202401515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 11/06/2024]
Abstract
Diabetes is a major global health issue and as current treatments fail, the search for new antidiabetic drugs is crucial. This investigation, focusing on identifying potential antidiabetic compounds from the endangered plant species Vepris glandulosa, led to the isolation of two known alkaloids, choisyine acetate (1) and choisyine (2). The study established the in vitro inhibitory activities and in silico molecular interaction of the two alkaloids with α-amylase based on IC50 values, Linewaever-Burk/Dixon plot kinetic analyses and Molecular docking, respectively. The α-amylase inhibition assay revealed noncompetitive inhibition for both compounds with IC50 and Ki values of 4.74±0.17 and 4.75 mM for compound 1, and 11.29±0.44 and 12.37 mM for compound 2, respectively. In comparison, the standard drug acarbose displayed a competitive mode of inhibition, with IC50 and Ki values of 11.99±0.02 and 12.68 mM, respectively. The binding affinities with α-amylase were -6.42 and -6.07 kcal/mol for compounds 1 and 2, respectively relative to acarbose -8.03 Kcal/mol. Moreover, these two compounds' predicted physicochemical and ADMET properties justified their potential as lead compounds for drug discovery. These compounds demonstrated remarkable inhibition potential comparable to the standard drug, highlighting their potential as viable alternatives in managing diabetes.
Collapse
Affiliation(s)
- Prince Ojuka
- Department of Physical Science, University of Embu, P.O Box 6-60100, Embu, Kenya
| | - Charles O Ochieng
- Department of Chemistry, Maseno University, Private Bag, Maseno, Kenya
| | - Wilberforce Ndarawit
- Department of Physical Science, University of Embu, P.O Box 6-60100, Embu, Kenya
| | - Daniel W Nyongesa
- Department of Chemistry, Maseno University, Private Bag, Maseno, Kenya
| | - Justus Mukavi
- Institute of Pharmaceutical Biology and Phytochemistry (IPBP), University of Münster, PharmaCampus Corrensstrasse 48, Muenster, D-48149, Germany
| | - James Nyabuga Nyariki
- Department of Biochemistry and Biotechnology, Technical of University of Kenya, P.O Box 52428-00200, Nairobi, Kenya
| | - Seth Apollo
- Department of Physical Science, University of Embu, P.O Box 6-60100, Embu, Kenya
| | - Cleydson B R Santos
- Laboratory of Modeling and Computational Chemistry, Department of Biological Sciences and Health, Federal University of Amapá, Macapá, Amapá, Brazil
- Graduate Program in Medicinal Chemistry and Molecular Modelling, Health Science Institute, Federal University of Pará, 66075-110, Belém, PA, Brazil
| | - Njogu M Kimani
- Department of Physical Science, University of Embu, P.O Box 6-60100, Embu, Kenya
- Natural Product Chemistry and Computational Drug Discovery Laboratory, Embu, Kenya
| |
Collapse
|
2
|
D’Ambrosio A, Itaj F, Cacace F, Piemonte V. Mathematical Modeling of the Gastrointestinal System for Preliminary Drug Absorption Assessment. Bioengineering (Basel) 2024; 11:813. [PMID: 39199771 PMCID: PMC11352181 DOI: 10.3390/bioengineering11080813] [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: 06/01/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
The objective of this study is to demonstrate the potential of a multicompartmental mathematical model to simulate the activity of the gastrointestinal system after the intake of drugs, with a limited number of parameters. The gastrointestinal system is divided into five compartments, modeled as both continuous systems with discrete events (stomach and duodenum) and systems with delay (jejunum, ileum, and colon). The dissolution of the drug tablet occurs in the stomach and is described through the Noyes-Whitney equation, with pH dependence expressed through the Henderson-Hasselbach relationship. The boluses resulting from duodenal activity enter the jejunum, ileum, and colon compartments, where drug absorption takes place as blood flows countercurrent. The model includes only three parameters with assigned physiological meanings. It was tested and validated using data from in vivo experiments. Specifically, the model was tested with the concentration profiles of nine different drugs and validated using data from two drugs with varying initial concentrations. Overall, the outputs of the model are in good agreement with experimental data, particularly with regard to the time of peak concentration. The primary sources of discrepancy were identified in the concentration decay. The model's main strength is its relatively low computational cost, making it a potentially excellent tool for in silico assessment and prediction of drug adsorption in the intestine.
Collapse
Affiliation(s)
- Antonio D’Ambrosio
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Department of Science and Technology for Sustainable Development and One Health, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; (F.I.); (V.P.)
| | - Fatjon Itaj
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Department of Science and Technology for Sustainable Development and One Health, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; (F.I.); (V.P.)
| | - Filippo Cacace
- Research Unit of Computer Systems and Bioinformatics, Department of Engineering, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy;
| | - Vincenzo Piemonte
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Department of Science and Technology for Sustainable Development and One Health, University Campus Bio-Medico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy; (F.I.); (V.P.)
| |
Collapse
|
3
|
Zhang SY, Ong WSY, Subelzu N, Gleeson JP. Validation of a Caco-2 microfluidic Chip model for predicting intestinal absorption of BCS Class I-IV drugs. Int J Pharm 2024; 656:124089. [PMID: 38599444 DOI: 10.1016/j.ijpharm.2024.124089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/12/2024]
Abstract
Oral delivery is considered the most patient preferred route of drug administration, however, the drug must be sufficiently soluble and permeable to successfully formulate an oral formulation. There have been advancements in the development of more predictive solubility and dissolution tools, but the tools that has been developed for permeability assays have not been validated as extensively as the gold-standard Caco-2 Transwell assay. Here, we evaluated Caco-2 intestinal permeability assay in Transwells and a commercially available microfluidic Chip using 19 representative Biopharmaceutics Classification System (BCS) Class I-IV compounds. For each selected compound, we performed a comprehensive viability test, quantified its apparent permeability (Papp), and established an in vitro in vivo correlation (IVIVC) to the human fraction absorbed (fa) in both culture conditions. Permeability differences were observed across the models as demonstrated by antipyrine (Transwell Papp: 38.5 ± 6.1 × 10-8 cm/s vs Chip Papp: 32.9 ± 11.3 × 10-8 cm/s) and nadolol (Transwell Papp: 0.6 ± 0.1 × 10-7 cm/s vs Chip Papp: 3 ± 1.2 × 10-7 cm/s). The in vitro in vivo correlation (IVIVC; Papp vs. fa) of the Transwell model (r2 = 0.59-0.83) was similar to the Chip model (r2 = 0.41-0.79), highlighting similar levels of predictivity. Comparing to historical data, our Chip Papp data was more closely aligned to native tissues assessed in Ussing chambers. This is the first study to comprehensively validate a commercial Gut-on-a-Chip model as a predictive tool for assessing oral absorption to further reduce our reliance on animal models.
Collapse
Affiliation(s)
- Stephanie Y Zhang
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Whitney S Y Ong
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - Natalia Subelzu
- Analytical Research and Development, Merck & Co., Inc., Rahway, NJ 07065, USA
| | - John P Gleeson
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, NJ 07065, USA.
| |
Collapse
|
4
|
Kibet S, Kimani NM, Mwanza SS, Mudalungu CM, Santos CBR, Tanga CM. Unveiling the Potential of Ent-Kaurane Diterpenoids: Multifaceted Natural Products for Drug Discovery. Pharmaceuticals (Basel) 2024; 17:510. [PMID: 38675469 PMCID: PMC11054903 DOI: 10.3390/ph17040510] [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: 03/15/2024] [Revised: 03/29/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Natural products hold immense potential for drug discovery, yet many remain unexplored in vast libraries and databases. In an attempt to fill this gap and meet the growing demand for effective drugs, this study delves into the promising world of ent-kaurane diterpenoids, a class of natural products with huge therapeutic potential. With a dataset of 570 ent-kaurane diterpenoids obtained from the literature, we conducted an in silico analysis, evaluating their physicochemical, pharmacokinetic, and toxicological properties with a focus on their therapeutic implications. Notably, these natural compounds exhibit drug-like properties, aligning closely with those of FDA-approved drugs, indicating a high potential for drug development. The ranges of the physicochemical parameters were as follows: molecular weights-288.47 to 626.82 g/mol; number of heavy atoms-21 to 44; the number of hydrogen bond donors and acceptors-0 to 8 and 1 to 11, respectively; the number of rotatable bonds-0 to 11; fraction Csp3-0.65 to 1; and TPSA-20.23 to 189.53 Ų. Additionally, the majority of these molecules display favorable safety profiles, with only 0.70%, 1.40%, 0.70%, and 46.49% exhibiting mutagenic, tumorigenic, reproduction-enhancing, and irritant properties, respectively. Importantly, ent-kaurane diterpenoids exhibit promising biopharmaceutical properties. Their average lipophilicity is optimal for drug absorption, while over 99% are water-soluble, facilitating delivery. Further, 96.5% and 28.20% of these molecules exhibited intestinal and brain bioavailability, expanding their therapeutic reach. The predicted pharmacological activities of these compounds encompass a diverse range, including anticancer, immunosuppressant, chemoprotective, anti-hepatic, hepatoprotectant, anti-inflammation, antihyperthyroidism, and anti-hepatitis activities. This multi-targeted profile highlights ent-kaurane diterpenoids as highly promising candidates for further drug discovery endeavors.
Collapse
Affiliation(s)
- Shadrack Kibet
- Department of Physical Sciences, University of Embu, Embu P.O. Box 6-60100, Kenya; (S.K.); (S.S.M.)
- International Centre of Insects Physiology and Ecology, Nairobi P.O. Box 30772-00100, Kenya;
| | - Njogu M. Kimani
- Department of Physical Sciences, University of Embu, Embu P.O. Box 6-60100, Kenya; (S.K.); (S.S.M.)
- Natural Product Chemistry and Computational Drug Discovery Laboratory, Embu P.O. Box 6-60100, Kenya
| | - Syombua S. Mwanza
- Department of Physical Sciences, University of Embu, Embu P.O. Box 6-60100, Kenya; (S.K.); (S.S.M.)
- International Centre of Insects Physiology and Ecology, Nairobi P.O. Box 30772-00100, Kenya;
| | - Cynthia M. Mudalungu
- International Centre of Insects Physiology and Ecology, Nairobi P.O. Box 30772-00100, Kenya;
- School of Chemistry and Material Science, The Technical University of Kenya, Nairobi P.O. Box 52428-00200, Kenya
| | - Cleydson B. R. Santos
- Graduate Program in Medicinal Chemistry and Molecular Modelling, Health Science Institute, Federal University of Pará, Belém 66075-110, Brazil;
- Laboratory of Modelling and Computational Chemistry, Department of Biological and Health Sciences, Federal University of Amapá, Macapá 68902-280, Brazil
| | - Chrysantus M. Tanga
- International Centre of Insects Physiology and Ecology, Nairobi P.O. Box 30772-00100, Kenya;
| |
Collapse
|
5
|
Weiss D, Baylon JL, Evans ED, Paiva A, Everlof G, Cutrone J, Broccatelli F. Balanced Permeability Index: A Multiparameter Index for Improved In Vitro Permeability. ACS Med Chem Lett 2024; 15:457-462. [PMID: 38628792 PMCID: PMC11017404 DOI: 10.1021/acsmedchemlett.3c00542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
The optimization of passive permeability is a key objective for orally available small molecule drug candidates. For drugs targeting the central nervous system (CNS), minimizing P-gp-mediated efflux is an additional important target for optimization. The physicochemical properties most strongly associated with high passive permeability and lower P-gp efflux are size, polarity, and lipophilicity. In this study, a new metric called the Balanced Permeability Index (BPI) was developed that combines these three properties. The BPI was found to be more effective than any single property in classifying molecules based on their permeability and efflux across a diverse range of chemicals and assays. BPI is easy to understand, allowing researchers to make decisions about which properties to prioritize during the drug development process.
Collapse
Affiliation(s)
- Dahlia
R. Weiss
- Bristol-Myers
Squibb Company, Redwood
City, California 94063, United States
| | - Javier L. Baylon
- Bristol-Myers
Squibb Company, San Diego, California 92121, United States
| | - Ethan D. Evans
- Bristol-Myers
Squibb Company, Redwood
City, California 94063, United States
| | - Anthony Paiva
- Bristol-Myers
Squibb Company, Lawrence Township, New Jersey 08648, United States
| | - Gerry Everlof
- Bristol-Myers
Squibb Company, Lawrence Township, New Jersey 08648, United States
| | - Jingfang Cutrone
- Bristol-Myers
Squibb Company, Lawrence Township, New Jersey 08648, United States
| | - Fabio Broccatelli
- Bristol-Myers
Squibb Company, San Diego, California 92121, United States
| |
Collapse
|
6
|
Zhang F, Erskine TC, McClymont EL, Moore LM, LeBaron MJ, McNett D, Marty SS. Predictions of tissue concentrations of myclobutanil, oxyfluorfen, and pronamide in rat and human after oral exposures via GastroPlus TM physiologically based pharmacokinetic modelling. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2024; 35:285-307. [PMID: 38588502 DOI: 10.1080/1062936x.2024.2333878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/15/2024] [Indexed: 04/10/2024]
Abstract
Heritage agrochemicals like myclobutanil, oxyfluorfen, and pronamide, are extensively used in agriculture, with well-established studies on their animal toxicity. Yet, human toxicity assessment relies on conventional human risk assessment approaches including the utilization of animal-based ADME (Absorption, Distribution, Metabolism, and Excretion) data. In recent years, Physiologically Based Pharmacokinetic (PBPK) modelling approaches have played an increasing role in human risk assessment of many chemicals including agrochemicals. This study addresses the absence of PBPK-type data for myclobutanil, oxyfluorfen, and pronamide by generating in vitro data for key input PBPK parameters (Caco-2 permeability, rat plasma binding, rat blood to plasma ratio, and rat liver microsomal half-life), followed by generation of PBPK models for these three chemicals via the GastroPlusTM software. Incorporating these experimental input parameters into PBPK models, the prediction accuracy of plasma AUC (area under curve) was significantly improved. Validation against rat oral administration data demonstrated substantial enhancement. Steady-state plasma concentrations (Css) of pronamide aligned well with published data using measured PBPK parameters. Following validation, parent-based tissue concentrations for these agrochemicals were predicted in humans and rats after single or 30-day repeat exposure of 10 mg/kg/day. These predicted concentrations contribute valuable information for future human toxicity risk assessments of these agrochemicals.
Collapse
Affiliation(s)
- F Zhang
- Toxicology & Environmental Research & Consulting, The Dow Chemical Company, Midland, MI, USA
| | - T C Erskine
- Toxicology & Environmental Research & Consulting, The Dow Chemical Company, Midland, MI, USA
| | - E L McClymont
- Toxicology & Environmental Research & Consulting, The Dow Chemical Company, Midland, MI, USA
| | - L M Moore
- Toxicology & Environmental Research & Consulting, The Dow Chemical Company, Midland, MI, USA
| | - M J LeBaron
- Toxicology & Environmental Research & Consulting, The Dow Chemical Company, Midland, MI, USA
| | - D McNett
- Toxicology & Environmental Research & Consulting, The Dow Chemical Company, Midland, MI, USA
| | - S S Marty
- Toxicology & Environmental Research & Consulting, The Dow Chemical Company, Midland, MI, USA
| |
Collapse
|
7
|
Djuris J, Cvijic S, Djekic L. Model-Informed Drug Development: In Silico Assessment of Drug Bioperformance following Oral and Percutaneous Administration. Pharmaceuticals (Basel) 2024; 17:177. [PMID: 38399392 PMCID: PMC10892858 DOI: 10.3390/ph17020177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/23/2023] [Accepted: 12/29/2023] [Indexed: 02/25/2024] Open
Abstract
The pharmaceutical industry has faced significant changes in recent years, primarily influenced by regulatory standards, market competition, and the need to accelerate drug development. Model-informed drug development (MIDD) leverages quantitative computational models to facilitate decision-making processes. This approach sheds light on the complex interplay between the influence of a drug's performance and the resulting clinical outcomes. This comprehensive review aims to explain the mechanisms that control the dissolution and/or release of drugs and their subsequent permeation through biological membranes. Furthermore, the importance of simulating these processes through a variety of in silico models is emphasized. Advanced compartmental absorption models provide an analytical framework to understand the kinetics of transit, dissolution, and absorption associated with orally administered drugs. In contrast, for topical and transdermal drug delivery systems, the prediction of drug permeation is predominantly based on quantitative structure-permeation relationships and molecular dynamics simulations. This review describes a variety of modeling strategies, ranging from mechanistic to empirical equations, and highlights the growing importance of state-of-the-art tools such as artificial intelligence, as well as advanced imaging and spectroscopic techniques.
Collapse
Affiliation(s)
- Jelena Djuris
- Department of Pharmaceutical Technology and Cosmetology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (S.C.); (L.D.)
| | | | | |
Collapse
|
8
|
Wang K, Amidon GL, Smith DE. Physiological Dynamics in the Upper Gastrointestinal Tract and the Development of Gastrointestinal Absorption Models for the Immediate-Release Oral Dosage Forms in Healthy Adult Human. Pharm Res 2023; 40:2607-2626. [PMID: 37783928 DOI: 10.1007/s11095-023-03597-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/26/2023] [Indexed: 10/04/2023]
Abstract
This review is a revisit of various oral drug absorption models developed in the past decades, focusing on how to incorporate the physiological dynamics in the upper gastrointestinal (GI) tract. For immediate-release oral drugs, GI absorption is a critical input of drug exposure and subsequent human body response, yet difficult to model largely due to the complex GI environment. One of the biggest hurdles lies at capturing the high within-subject variability (WSV) of bioavailability measures, which can be mechanistically explained by the GI physiological dynamics. A thorough summary of how GI dynamics is handled in the absorption models would promote the development of mechanism-based oral drug absorption models, aid in the design of clinical studies regarding dosing regimens and bioequivalence studies based on WSV, and advance the decision-making on formulation selection.
Collapse
Affiliation(s)
- Kai Wang
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Gordon L Amidon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David E Smith
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
9
|
Hussain A, Afzal O, Yasmin S, Haider N, Altamimi AS, Martinez F, Acree WE, Ramzan M. Preferential Solvation Study of Rosuvastatin in the {PEG400 (1) + Water (2)} Cosolvent Mixture and GastroPlus Software-Based In Vivo Predictions. ACS OMEGA 2023; 8:12761-12772. [PMID: 37065087 PMCID: PMC10099431 DOI: 10.1021/acsomega.2c07968] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
Rosuvastatin (RST) is a poorly water-soluble drug responsible for limited in vivo dissolution and subsequently low oral systemic absorption (poor bioavailability). The mole fraction solubility values of RST in various ratios of binary mixtures "{PEG400 (1) + water (2)}" at 298.15 K were employed to investigate the preferential solvation (PS) of RST (3) by the binary components. Moreover, the GastroPlus program predicted the drug dissolution/absorption rates, plasma drug concentration, and compartmental regional drug absorbed from a conventional tablet as compared to the RST-loaded (PEG400 + water) mixture (at x 1 = 0.5) in healthy subjects (considering the fast condition). Fedors' method was adopted to estimate the values of molar volume (314.8 cm3·mol-1) and Hildebrand solubility parameter (28.08 MPa1/2) of RST. The results of inverse Kirkwood-Buff integrals showed the PS of RST by PEG400 as observed in all studied ratios of the binary mixture. The highest PS value (δx 1,3 = 1.65 × 10-2) for RST by PEG400 was attained at x 1 = 0.5. Finally, the GastroPlus program predicted the maximum dissolution rate [20 mg within 15 min as compared to pure RST (1.5 mg within 15 min)]. Moreover, the program predicted increased in vivo oral absorption (1.2 μg/mL) and enhanced regional absorption (95.3%) of RST from upper segments of the gastrointestinal tract for the RST-loaded PEG400 + water mixture in humans as compared to conventional tablets (87.5% as total regional absorption and 0.88 μg/mL as in vivo absorption). Hence, the present binary system ferrying RST can be a promising strategy to control systemic dyslipidemia after oral or subcutaneous administration.
Collapse
Affiliation(s)
- Afzal Hussain
- Department
of Pharmaceutics, College of Pharmacy, King
Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Obaid Afzal
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Sabina Yasmin
- Department
of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Nazima Haider
- Department
of Pathology, College of Medicine, King
Khalid University, Abha 61421, Saudi Arabia
| | | | - Fleming Martinez
- Grupo
de Investigaciones Farmacéutico-Fisicoquímicas, Departamento
de Farmacia, Universidad Nacional de Colombia,
Sede Bogotá, Cra
30 No. 45-03, Bogotá D. C. 111321, Colombia
| | - William E. Acree
- Department
of Chemistry, University of North Texas, Denton, Texas 76203-5070, United States
| | - Mohhammad Ramzan
- School
of Pharmacy, Lovely Professional University, Jalandhar-Delhi, Grand Trunk Road, Phagwara, Punjab 144001, India
| |
Collapse
|
10
|
Tannergren C, Jadhav H, Eckernäs E, Fagerberg J, Augustijns P, Sjögren E. Physiologically Based Biopharmaceutics Modeling of regional and colon absorption in humans. Eur J Pharm Biopharm 2023; 186:144-159. [PMID: 37028605 DOI: 10.1016/j.ejpb.2023.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/24/2023] [Accepted: 03/25/2023] [Indexed: 04/08/2023]
Abstract
Colon absorption is a key determinant for successful development of extended release and colon targeted drug products. This is the first systematic evaluation of the ability to predict in vivo regional differences in absorption and the extent of colon absorption in humans using mechanistic physiologically based biopharmaceutics modeling (PBBM). A new dataset, consisting of 19 drugs with a wide range of biopharmaceutics properties and extent of colon absorption in humans, was established. Mechanistic predictions of the extent of absorption and plasma exposure after oral, or jejunal and direct colon administration were performed in GastroPlus and GI-Sim using an a priori approach. Two new colon models developed in GI-Sim, were also evaluated to assess if the prediction performance could be improved. Both GastroPlus and GI-Sim met the pre-defined criteria for accurate predictions of regional and colon absorption for high permeability drugs irrespective of formulation type, while the prediction performance was poor for low permeability drugs. For solutions, the two new GI-Sim colon models improved the colon absorption prediction performance for the low permeability drugs while maintaining the accurate prediction performance for the high permeability drugs. In contrast, the prediction performance decreased for non-solutions using the two new colon models. In conclusion, PBBM can be used with sufficient accuracy to predict regional and colon absorption in humans for high permeability drugs in candidate selection as well as early design and development of extended release or colon targeted drug products. The prediction performance of the current models needs to be improved to allow high accuracy predictions for commercial drug product applications including highly accurate predictions of the entire plasma concentration-time profiles as well as for low permeability drugs.
Collapse
|
11
|
Supersaturation and Precipitation Applicated in Drug Delivery Systems: Development Strategies and Evaluation Approaches. Molecules 2023; 28:molecules28052212. [PMID: 36903470 PMCID: PMC10005129 DOI: 10.3390/molecules28052212] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Supersaturation is a promising strategy to improve gastrointestinal absorption of poorly water-soluble drugs. Supersaturation is a metastable state and therefore dissolved drugs often quickly precipitate again. Precipitation inhibitors can prolong the metastable state. Supersaturating drug delivery systems (SDDS) are commonly formulated with precipitation inhibitors, hence the supersaturation is effectively prolonged for absorption, leading to improved bioavailability. This review summarizes the theory of and systemic insight into supersaturation, with the emphasis on biopharmaceutical aspects. Supersaturation research has developed from the generation of supersaturation (pH-shift, prodrug and SDDS) and the inhibition of precipitation (the mechanism of precipitation, the character of precipitation inhibitors and screening precipitation inhibitors). Then, the evaluation approaches to SDDS are discussed, including in vitro, in vivo and in silico studies and in vitro-in vivo correlations. In vitro aspects involve biorelevant medium, biomimetic apparatus and characterization instruments; in vivo aspects involve oral absorption, intestinal perfusion and intestinal content aspiration and in silico aspects involve molecular dynamics simulation and pharmacokinetic simulation. More physiological data of in vitro studies should be taken into account to simulate the in vivo environment. The supersaturation theory should be further completed, especially with regard to physiological conditions.
Collapse
|
12
|
Prieto Garcia L, Lundahl A, Ahlström C, Vildhede A, Lennernäs H, Sjögren E. Does the choice of applied physiologically‐based pharmacokinetics platform matter? A case study on simvastatin disposition and drug–drug interaction. CPT Pharmacometrics Syst Pharmacol 2022; 11:1194-1209. [PMID: 35722750 PMCID: PMC9469690 DOI: 10.1002/psp4.12837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022] Open
Abstract
Physiologically‐based pharmacokinetic (PBPK) models have an important role in drug discovery/development and decision making in regulatory submissions. This is facilitated by predefined PBPK platforms with user‐friendly graphical interface, such as Simcyp and PK‐Sim. However, evaluations of platform differences and the potential implications for disposition‐related applications are still lacking. The aim of this study was to assess how PBPK model development, input parameters, and model output are affected by the selection of PBPK platform. This is exemplified via the establishment of simvastatin PBPK models (workflow, final models, and output) in PK‐Sim and Simcyp as representatives of established whole‐body PBPK platforms. The major finding was that the choice of PBPK platform influenced the model development strategy and the final model input parameters, however, the predictive performance of the simvastatin models was still comparable between the platforms. The main differences between the structure and implementation of Simcyp and PK‐Sim were found in the absorption and distribution models. Both platforms predicted equally well the observed simvastatin (lactone and acid) pharmacokinetics (20–80 mg), BCRP and OATP1B1 drug–gene interactions (DGIs), and drug–drug interactions (DDIs) when co‐administered with CYP3A4 and OATP1B1 inhibitors/inducers. This study illustrates that in‐depth knowledge of established PBPK platforms is needed to enable an assessment of the consequences of PBPK platform selection. Specifically, this work provides insights on software differences and potential implications when bridging PBPK knowledge between Simcyp and PK‐Sim users. Finally, it provides a simvastatin model implemented in both platforms for risk assessment of metabolism‐ and transporter‐mediated DGIs and DDIs.
Collapse
Affiliation(s)
- Luna Prieto Garcia
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and Development Uppsala University Uppsala Sweden
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Anna Lundahl
- Clinical Pharmacology and Quantitative Pharmacology, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Christine Ahlström
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Anna Vildhede
- DMPK, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D AstraZeneca Gothenburg Sweden
| | - Hans Lennernäs
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and Development Uppsala University Uppsala Sweden
| | - Erik Sjögren
- Department of Pharmaceutical Bioscience, Translational Drug Discovery and Development Uppsala University Uppsala Sweden
| |
Collapse
|
13
|
Berg S, Suljovic D, Kärrberg L, Englund M, Bönisch H, Karlberg I, Van Zuydam N, Abrahamsson B, Hugerth AM, Davies N, Bergström CAS. Intestinal Absorption of FITC-Dextrans and Macromolecular Model Drugs in the Rat Intestinal Instillation Model. Mol Pharm 2022; 19:2564-2572. [PMID: 35642793 PMCID: PMC9257752 DOI: 10.1021/acs.molpharmaceut.2c00261] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
![]()
In this work, we
studied the intestinal absorption of a peptide
with a molecular weight of 4353 Da (MEDI7219) and a protein having
a molecular weight of 11 740 Da (PEP12210) in the rat intestinal
instillation model and compared their absorption to fluorescein isothiocyanate
(FITC)-labeled dextrans of similar molecular weights (4 and 10 kDa).
To increase the absorption of the compounds, the permeation enhancer
sodium caprate (C10) was included in the liquid formulations at concentrations
of 50 and 300 mM. All studied compounds displayed an increased absorption
rate and extent when delivered together with 50 mM C10 as compared
to control formulations not containing C10. The time period during
which the macromolecules maintained an increased permeability through
the intestinal epithelium was approximately 20 min for all studied
compounds at 50 mM C10. For the formulations containing 300 mM C10,
it was noted that the dextrans displayed an increased absorption rate
(compared to 50 mM C10), and their absorption continued for at least
60 min. The absorption rate of MEDI7219, on the other hand, was similar
at both studied C10 concentrations, but the duration of absorption
was extended at the higher enhancer concentration, leading to an increase
in the overall extent of absorption. The absorption of PEP12210 was
similar in terms of the rate and duration at both studied C10 concentrations.
This is likely caused by the instability of this molecule in the intestinal
lumen. The degradation decreases the luminal concentrations over time,
which in turn limits absorption at time points beyond 20 min. The
results from this study show that permeation enhancement effects cannot
be extrapolated between different types of macromolecules. Furthermore,
to maximize the absorption of a macromolecule delivered together with
C10, prolonging the duration of absorption appears to be important.
In addition, the macromolecule needs to be stable enough in the intestinal
lumen to take advantage of the prolonged absorption time window enabled
by the permeation enhancer.
Collapse
Affiliation(s)
- Staffan Berg
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, BMC P.O. Box 580, Uppsala SE-751 23, Sweden.,Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Denny Suljovic
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, BMC P.O. Box 580, Uppsala SE-751 23, Sweden
| | - Lillevi Kärrberg
- Animal Sciences and Technologies, Clinical Pharmacology and Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Maria Englund
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | | | | | - Natalie Van Zuydam
- Data Science and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg 431 83, Sweden
| | - Andreas Martin Hugerth
- Ferring Pharmaceuticals A/S, Product Development and Drug Delivery, Global Pharmaceutical R&D, Amager Strandvej 405, Kastrup 2770, Denmark
| | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg 431 83, Sweden
| | - Christel A S Bergström
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, BMC P.O. Box 580, Uppsala SE-751 23, Sweden
| |
Collapse
|
14
|
Madny MA, Deshpande P, Tumuluri V, Borde P, Sangana R. Physiologically Based Biopharmaceutics Model of Vildagliptin Modified Release (MR) Tablets to Predict In Vivo Performance and Establish Clinically Relevant Dissolution Specifications. AAPS PharmSciTech 2022; 23:108. [PMID: 35386066 DOI: 10.1208/s12249-022-02264-2] [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: 01/07/2022] [Accepted: 03/21/2022] [Indexed: 11/30/2022] Open
Abstract
The objective of the study was to predict pharmacokinetic (PK) and pharmacodynamic (PD) parameters of matrix-based modified release (MR) drug product of vildagliptin. Physiologically based biopharmaceutics modeling (PBBM) was developed using GastroPlus™ based on the available data including immediate-release (IR) drug product of vildagliptin. In vitro-in vivo correlation (IVIVC) was developed using mechanistic deconvolution to predict plasma concentration-time profile and PK parameters for a MR drug product planned for clinical use. Both methods, i.e., PBBM and IVIVC, were compared for the predicted PK parameters. Integration of DDDPlus™ and GastroPlus™ modeling was performed to explore clinically relevant dissolution specifications for vildagliptin MR tablets. The bioequivalence (BE) between batches with different dissolution specifications was evaluated using virtual clinical trials. The PD effect of dipeptidyl peptidase-IV (DPP-IV) inhibition was simulated utilizing PDPlus™ model in GastroPlus™. The results indicated that IVIVC best correlated the simulated PK parameters with those observed with the clinical study. The outcomes highlight the importance of integration of in vitro and in silico tools towards predictability of PK and PD parameters for a MR drug product. However, the post absorptive phase was found to be more dependent on the demographics of the healthy subjects.
Collapse
|
15
|
Emami Riedmaier A. Predicting Food Effects: Are We There Yet? AAPS J 2022; 24:25. [PMID: 35006448 DOI: 10.1208/s12248-021-00674-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/08/2021] [Indexed: 01/06/2023] Open
Affiliation(s)
- Arian Emami Riedmaier
- Nonclinical Disposition and Bioanalysis, Nonclinical R&D, Bristol Myers Squibb, Rt. 206 & Province Line Roads, Princeton, NJ, 08543, USA.
| |
Collapse
|
16
|
Berg S, Kärrberg L, Suljovic D, Seeliger F, Söderberg M, Perez-Alcazar M, Van Zuydam N, Abrahamsson B, Hugerth AM, Davies N, Bergström CAS. Impact of Intestinal Concentration and Colloidal Structure on the Permeation-Enhancing Efficiency of Sodium Caprate in the Rat. Mol Pharm 2022; 19:200-212. [PMID: 34928160 PMCID: PMC8728734 DOI: 10.1021/acs.molpharmaceut.1c00724] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
![]()
In this work, we
set out to better understand how the permeation
enhancer sodium caprate (C10) influences the intestinal absorption
of macromolecules. FITC-dextran 4000 (FD4) was selected as a model
compound and formulated with 50–300 mM C10. Absorption was
studied after bolus instillation of liquid formulation to the duodenum
of anesthetized rats and intravenously as a reference, whereafter
plasma samples were taken and analyzed for FD4 content. It was found
that the AUC and Cmax of FD4 increased
with increasing C10 concentration. Higher C10 concentrations were
associated with an increased and extended absorption but also increased
epithelial damage. Depending on the C10 concentration, the intestinal
epithelium showed significant recovery already at 60–120 min
after administration. At the highest studied C10 concentrations (100
and 300 mM), the absorption of FD4 was not affected by the colloidal
structures of C10, with similar absorption obtained when C10 was administered
as micelles (pH 8.5) and as vesicles (pH 6.5). In contrast, the FD4
absorption was lower when C10 was administered at 50 mM formulated
as micelles as compared to vesicles. Intestinal dilution of C10 and
FD4 revealed a trend of decreasing FD4 absorption with increasing
intestinal dilution. However, the effect was smaller than that of
altering the total administered C10 dose. Absorption was similar when
the formulations were prepared in simulated intestinal fluids containing
mixed micelles of bile salts and phospholipids and in simple buffer
solution. The findings in this study suggest that in order to optimally
enhance the absorption of macromolecules, high (≥100 mM) initial
intestinal C10 concentrations are likely needed and that both the
concentration and total dose of C10 are important parameters.
Collapse
Affiliation(s)
- Staffan Berg
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, BMC P.O. Box 580, SE-751 23 Uppsala, Sweden.,Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Lillevi Kärrberg
- Animal Sciences and Technologies, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Denny Suljovic
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, BMC P.O. Box 580, SE-751 23 Uppsala, Sweden
| | - Frank Seeliger
- Cardiovascular, Renal and Metabolism Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Magnus Söderberg
- Cardiovascular, Renal and Metabolism Safety, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Marta Perez-Alcazar
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Natalie Van Zuydam
- Data Science and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Andreas M Hugerth
- Ferring Pharmaceuticals A/S Global Pharmaceutical R&D, 2300 Copenhagen, Denmark
| | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Christel A S Bergström
- The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, BMC P.O. Box 580, SE-751 23 Uppsala, Sweden
| |
Collapse
|
17
|
Wilson CG, Aarons L, Augustijns P, Brouwers J, Darwich AS, De Waal T, Garbacz G, Hansmann S, Hoc D, Ivanova A, Koziolek M, Reppas C, Schick P, Vertzoni M, García-Horsman JA. Integration of advanced methods and models to study drug absorption and related processes: An UNGAP perspective. Eur J Pharm Sci 2021; 172:106100. [PMID: 34936937 DOI: 10.1016/j.ejps.2021.106100] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 12/14/2021] [Accepted: 12/16/2021] [Indexed: 01/09/2023]
Abstract
This collection of contributions from the European Network on Understanding Gastrointestinal Absorption-related Processes (UNGAP) community assembly aims to provide information on some of the current and newer methods employed to study the behaviour of medicines. It is the product of interactions in the immediate pre-Covid period when UNGAP members were able to meet and set up workshops and to discuss progress across the disciplines. UNGAP activities are divided into work packages that cover special treatment populations, absorption processes in different regions of the gut, the development of advanced formulations and the integration of food and pharmaceutical scientists in the food-drug interface. This involves both new and established technical approaches in which we have attempted to define best practice and highlight areas where further research is needed. Over the last months we have been able to reflect on some of the key innovative approaches which we were tasked with mapping, including theoretical, in silico, in vitro, in vivo and ex vivo, preclinical and clinical approaches. This is the product of some of us in a snapshot of where UNGAP has travelled and what aspects of innovative technologies are important. It is not a comprehensive review of all methods used in research to study drug dissolution and absorption, but provides an ample panorama of current and advanced methods generally and potentially useful in this area. This collection starts from a consideration of advances in a priori approaches: an understanding of the molecular properties of the compound to predict biological characteristics relevant to absorption. The next four sections discuss a major activity in the UNGAP initiative, the pursuit of more representative conditions to study lumenal dissolution of drug formulations developed independently by academic teams. They are important because they illustrate examples of in vitro simulation systems that have begun to provide a useful understanding of formulation behaviour in the upper GI tract for industry. The Leuven team highlights the importance of the physiology of the digestive tract, as they describe the relevance of gastric and intestinal fluids on the behaviour of drugs along the tract. This provides the introduction to microdosing as an early tool to study drug disposition. Microdosing in oncology is starting to use gamma-emitting tracers, which provides a link through SPECT to the next section on nuclear medicine. The last two papers link the modelling approaches used by the pharmaceutical industry, in silico to Pop-PK linking to Darwich and Aarons, who provide discussion on pharmacometric modelling, completing the loop of molecule to man.
Collapse
Affiliation(s)
- Clive G Wilson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, Glasgow, U.K.
| | | | | | | | | | | | | | | | | | | | - Mirko Koziolek
- NCE Formulation Sciences, Abbvie Deutschland GmbH & Co. KG, Germany
| | | | - Philipp Schick
- Department of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, Germany
| | | | | |
Collapse
|
18
|
Cacace F, Menci M, Papi M, Piemonte V. In-Silico Prediction of Oral Drug Bioavailability: A multi-boluses approach. Med Eng Phys 2021; 98:140-150. [PMID: 34848033 DOI: 10.1016/j.medengphy.2021.11.004] [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: 05/05/2021] [Revised: 11/02/2021] [Accepted: 11/03/2021] [Indexed: 10/19/2022]
Abstract
This work focuses on a new mathematical model able to describe in a simple manner the intestinal physiology, in order to better study drug absorption and bioavailability. The aim of our model is to overcome the limitations of physiological pharmacokinetics models of the literature, introducing a different modelling approach. The core of the new proposed model is a Discrete-Continuous Approach (DCA): a sequence of boluses travels in the investigated portion of the intestine, in counter-current with blood that flows in continuous mode. No empirical equations are implemented in this model. Simulation results show an excellent correlation between the predicted and experimental concentration profile used to validate our model. Our new approach provides a simple tool, with a good reliability, to analyze a very complex phenomenon, using only few parameters.
Collapse
Affiliation(s)
| | - Marta Menci
- Istituto per le Applicazioni del Calcolo, Consiglio Nazionale delle Ricerche, Rome, Italy.
| | - Marco Papi
- Universitá Campus Bio-Medico di Roma, Rome, Italy.
| | | |
Collapse
|
19
|
Sadighi A, Leggio L, Akhlaghi F. Development of a Physiologically Based Pharmacokinetic Model for Prediction of Ethanol Concentration-Time Profile in Different Organs. Alcohol Alcohol 2021; 56:401-414. [PMID: 33316031 DOI: 10.1093/alcalc/agaa129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/25/2020] [Accepted: 11/07/2020] [Indexed: 11/15/2022] Open
Abstract
AIMS A physiologically based pharmacokinetic (PBPK) modeling approach was used to simulate the concentration-time profile of ethanol (EtOH) in stomach, duodenum, plasma and other tissues upon consumption of beer and whiskey under fasted and fed conditions. METHODS A full PBPK model was developed for EtOH using the advanced dissolution, absorption and metabolism (ADAM) model fully integrated into the Simcyp Simulator® 15 (Simcyp Ltd., Sheffield, UK). The prediction performance of the developed model was verified and the EtOH concentration-time profile in different organs was predicted. RESULTS Simcyp simulation showed ≤ 2-fold difference in values of EtOH area under the concentration-time curve (AUC) in stomach and duodenum as compared to the observed values. Moreover, the simulated EtOH maximum concentration (Cmax), time to reach Cmax (Tmax) and AUC in plasma were comparable to the observed values. We showed that liver is exposed to the highest EtOH concentration, faster than other organs (Cmax = 839.50 mg/L and Tmax = 0.53 h), while brain exposure of EtOH (AUC = 1139.43 mg·h/L) is the highest among all other organs. Sensitivity analyses (SAs) showed direct proportion of EtOH rate and extent of absorption with administered EtOH dose and inverse relationship with gastric emptying time (GE) and steady-state volume of distribution (Vss). CONCLUSIONS The current PBPK model approach might help with designing in vitro experiments in the area of alcohol organ damage or alcohol-drug interaction studies.
Collapse
Affiliation(s)
- Armin Sadighi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 7 Greenhouse Road, Kingston, RI 02881, USA
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, 10 Center Drive (10CRC/15330), Bethesda, MD 20892, USA.,Medication Development Program, National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd., Baltimore, MD 21224, USA.,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, 121 South Main Street, Providence, RI 02912, USA.,Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.,Department of Neuroscience, Georgetown University Medical Center, 4000 Reservoir Road, Washington D.C., DC 20007, USA
| | - Fatemeh Akhlaghi
- Clinical Pharmacokinetics Research Laboratory, Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, 7 Greenhouse Road, Kingston, RI 02881, USA
| |
Collapse
|
20
|
Xu H, Zhang Y, Wang P, Zhang J, Chen H, Zhang L, Du X, Zhao C, Wu D, Liu F, Yang H, Liu C. A comprehensive review of integrative pharmacology-based investigation: A paradigm shift in traditional Chinese medicine. Acta Pharm Sin B 2021; 11:1379-1399. [PMID: 34221858 PMCID: PMC8245857 DOI: 10.1016/j.apsb.2021.03.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/12/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Over the past decade, traditional Chinese medicine (TCM) has widely embraced systems biology and its various data integration approaches to promote its modernization. Thus, integrative pharmacology-based traditional Chinese medicine (TCMIP) was proposed as a paradigm shift in TCM. This review focuses on the presentation of this novel concept and the main research contents, methodologies and applications of TCMIP. First, TCMIP is an interdisciplinary science that can establish qualitative and quantitative pharmacokinetics-pharmacodynamics (PK-PD) correlations through the integration of knowledge from multiple disciplines and techniques and from different PK-PD processes in vivo. Then, the main research contents of TCMIP are introduced as follows: chemical and ADME/PK profiles of TCM formulas; confirming the three forms of active substances and the three action modes; establishing the qualitative PK-PD correlation; and building the quantitative PK-PD correlations, etc. After that, we summarize the existing data resources, computational models and experimental methods of TCMIP and highlight the urgent establishment of mathematical modeling and experimental methods. Finally, we further discuss the applications of TCMIP for the improvement of TCM quality control, clarification of the molecular mechanisms underlying the actions of TCMs and discovery of potential new drugs, especially TCM-related combination drug discovery.
Collapse
|
21
|
Abstract
Colon absorption is a key determinant for the successful development of modified-release (MR) formulations, and the risk that colon absorption may limit the in vivo performance of an MR product can be assessed early by various in vitro tests or by preclinical in vivo regional absorption studies in dogs. Mechanistic physiologically based biopharmaceutics modeling (PBBM) is becoming increasingly accepted to predict in vivo performance and guide formulation development; however, no evaluation of the ability to predict colon absorption has been performed. The purpose of this study was to investigate if regional and colon absorption of drugs in dogs could be predicted with sufficient accuracy using PBBM to enable the replacement of in vivo dog studies in the early assessment of colon absorption limitation risks. This was done by predicting the regional and colon absorption and plasma exposure of 14 drugs after administration to the dog colon according to an a priori approach using the in silico absorption models GI-Sim and GastroPlus. Predictive performance was primarily assessed by comparing observed and predicted plasma concentration-time profiles, AUC0-t, and the relative bioavailability in the colon (Frel,colon) as compared to an oral/duodenal reference. Trends in dependency of prediction performance on predicted fraction absorbed, permeability, and solubility/dissolution rate were also investigated. For GI-Sim, the absolute average fold error (AAFE) values for AUC0-t and Frel,colon were within a 2-fold prediction error for both solutions (1.88 and 1.51, respectively) and suspensions (1.58 and 1.99, respectively). For GastroPlus, the AAFE values for AUC0-t and Frel,colon were outside the set 2-fold prediction error limit for accurate predictions for both solutions (3.63 and 2.98, respectively) and suspensions (2.94 and 2.09, respectively). No trends for over- or underprediction were observed for GI-Sim, whereas GastroPlus showed a slight trend for underprediction of both AUC0-t and Frel,colon for compounds with low permeability. In addition, regional differences in the plasma profiles were qualitatively predicted in the majority of cases for both software. Despite the differences in prediction performance, both models can be considered to predict regional differences in absorption as well as AUC0-t and Frel,colon with acceptable accuracy in an early development setting. The results of this study indicate that it is acceptable to replace in vivo regional absorption studies in dogs with the evaluated models as a method for the early assessment of the risk for colon absorption limitation of MR drug product candidates.
Collapse
Affiliation(s)
- Emma Eckernäs
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, S-431 83 Mölndal, Sweden
| | - Christer Tannergren
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, S-431 83 Mölndal, Sweden
| |
Collapse
|
22
|
Pandya AK, Patravale VB. Computational avenues in oral protein and peptide therapeutics. Drug Discov Today 2021; 26:1510-1520. [PMID: 33684525 DOI: 10.1016/j.drudis.2021.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/12/2020] [Accepted: 03/02/2021] [Indexed: 12/19/2022]
Abstract
Proteins and peptides are amongst the most sought-after biomolecules because of their exceptional potential to cater to a vast range of diseases. Although widely studied and researched, the oral delivery of these biomolecules remains a challenge. Alongside formulation strategies, approaches to overcome the inherent barriers for peptide absorption are being designed at the molecular level to establish a sound rationale and to achieve higher bioavailability. Computer-aided drug design (CADD) is a modern in silico approach for developing successful bio-formulations. CADD enables intricate study of the biomolecules in conjunction with their target sites or receptors at the molecular level. Knowledge of the molecular interactions of proteins and peptides makes way for the pre-screening of suitable formulation components and facilitates their delivery.
Collapse
Affiliation(s)
- Anjali K Pandya
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India
| | - Vandana B Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400019, India.
| |
Collapse
|
23
|
O'Dwyer PJ, Box KJ, Dressman J, Griffin BT, Henze LJ, Litou C, Pentafragka C, Statelova M, Vertzoni M, Reppas C. Oral biopharmaceutics tools: recent progress from partnership through the Pharmaceutical Education and Research with Regulatory Links collaboration. J Pharm Pharmacol 2021; 73:437-446. [PMID: 33793836 DOI: 10.1093/jpp/rgaa055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To summarise key contributions of the Pharmaceutical Education and Research with Regulatory Links (PEARRL) project (2016-2020) to the optimisation of existing and the development of new biopharmaceutics tools for evaluating the in vivo performance of oral drug products during the development of new drugs and at the regulatory level. KEY FINDINGS Optimised biopharmaceutics tools: Based on new clinical data, the composition of biorelevant media for simulating the fed state conditions in the stomach was simplified. Strategies on how to incorporate biorelevant in vitro data of bio-enabling drug products into physiologically based pharmacokinetic (PBPK) modelling were proposed. Novel in vitro biopharmaceutics tools: Small-scale two-stage biphasic dissolution and dissolution-permeation setups were developed to facilitate understanding of the supersaturation effects and precipitation risks of orally administered drugs. A porcine fasted state simulated intestinal fluid was developed to improve predictions and interpretation of preclinical results using in vitro dissolution studies. Based on new clinical data, recommendations on the design of in vitro methodologies for evaluating the GI drug transfer process in the fed state were suggested. The optimized design of in vivo studies for investigating food effects: A food effect study protocol in the pig model was established which successfully predicted the food-dependent bioavailability of two model compounds. The effect of simulated infant fed state conditions in healthy adults on the oral absorption of model drugs was evaluated versus the fasted state and the fed state conditions, as defined by regulatory agencies for adults. Using PBPK modelling, the extrapolated fasted and infant fed conditions data appeared to be more useful to describe early drug exposure in infants, while extrapolation of data collected under fed state conditions, as defined by regulators for adults, failed to capture in vivo infant drug absorption. SUMMARY Substantial progress has been made in developing an advanced suite of biopharmaceutics tools for streamlining drug formulation screening and supporting regulatory applications. These advances in biopharmaceutics were achieved through networking opportunities and research collaborations provided under the H2020 funded PEARRL project.
Collapse
Affiliation(s)
- Patrick J O'Dwyer
- School of Pharmacy, University College Cork, Cork, Ireland.,Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK.,Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Karl J Box
- Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK
| | - Jennifer Dressman
- Institute of Translational Medicine and Pharmacology (ITMP), Fraunhofer Gesellschaft, Frankfurt am Main, Germany
| | | | - Laura J Henze
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Chara Litou
- Institute of Translational Medicine and Pharmacology (ITMP), Fraunhofer Gesellschaft, Frankfurt am Main, Germany
| | - Christina Pentafragka
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Marina Statelova
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Maria Vertzoni
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Christos Reppas
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| |
Collapse
|
24
|
Kesisoglou F. Can PBPK Modeling Streamline Food Effect Assessments? J Clin Pharmacol 2020; 60 Suppl 1:S98-S104. [PMID: 33205433 DOI: 10.1002/jcph.1678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 05/27/2020] [Indexed: 11/10/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is routinely used to study drug-drug interactions, replace some dedicated clinical studies, and inform product labeling. More recently, there has been increased application of PBPK models in the oral absorption field around drug product quality. Given the success of the models to characterize absorption of several orally administered drug products, a question arises whether PBPK could be used in a clinical setting to model food-drug interactions and thus streamline food effect assessments. Multiple publications have reported food effect predictions and comparisons with clinical data, primarily focusing on 2 food effect mechanisms: slowing down of gastric emptying and luminal solubilization by bile salts. Based on the available literature, this commentary proposes a workflow that PBPK model could be used to streamline food effect assessment during clinical development for different Biopharmaceutics Classification System classes.
Collapse
Affiliation(s)
- Filippos Kesisoglou
- Biopharmaceutics, Pharmaceutical Sciences, Merck & Co., Inc., Kenilworth, New Jersey, USA
| |
Collapse
|
25
|
Goracci L, Desantis J, Valeri A, Castellani B, Eleuteri M, Cruciani G. Understanding the Metabolism of Proteolysis Targeting Chimeras (PROTACs): The Next Step toward Pharmaceutical Applications. J Med Chem 2020; 63:11615-11638. [PMID: 33026811 PMCID: PMC8015227 DOI: 10.1021/acs.jmedchem.0c00793] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Indexed: 12/15/2022]
Abstract
Hetero-bifunctional PROteolysis TArgeting Chimeras (PROTACs) represent a new emerging class of small molecules designed to induce polyubiquitylation and proteasomal-dependent degradation of a target protein. Despite the increasing number of publications about the synthesis, biological evaluation, and mechanism of action of PROTACs, the characterization of the pharmacokinetic properties of this class of compounds is still minimal. Here, we report a study on the metabolism of a series of 40 PROTACs in cryopreserved human hepatocytes at multiple time points. Our results indicated that the metabolism of PROTACs could not be predicted from that of their constituent ligands. Their linkers' chemical nature and length resulted in playing a major role in the PROTACs' liability. A subset of compounds was also tested for metabolism by human cytochrome P450 3A4 (CYP3A4) and human aldehyde oxidase (hAOX) for more in-depth data interpretation, and both enzymes resulted in active PROTAC metabolism.
Collapse
Affiliation(s)
- Laura Goracci
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Jenny Desantis
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | | | - Beatrice Castellani
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| | - Michela Eleuteri
- Montelino
Therapeutics, LLC, 7
Powdermill Lane, Southborough, Massachusetts 01772 Unites States
| | - Gabriele Cruciani
- Department
of Chemistry, Biology and Biotechnology, University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
26
|
Wright L, Barnes TJ, Prestidge CA. Oral delivery of protein-based therapeutics: Gastroprotective strategies, physiological barriers and in vitro permeability prediction. Int J Pharm 2020; 585:119488. [PMID: 32504774 DOI: 10.1016/j.ijpharm.2020.119488] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023]
Abstract
The number of biological molecules emerging as therapeutics is growing exponentially due to their higher specificity and tolerability profiles compared to small molecules. Despite this, their traditionally parenteral delivery often results in poor patient compliance and incomplete treatment. Current research is focussed on developing effective oral delivery strategies to facilitate administration of these biomolecules, however no universal method exists to simultaneously provide gastric protection as well as enhance transport across the gastrointestinal epithelium. Furthermore, for efficient formulation development it is imperative that we can reliably analyse permeability of biomolecules through the gastrointestinal tract, highlighting the importance of the continual development and ongoing evaluation of in vitro predictive permeability tools. Here, we review the physiological obstacles associated with peptide and protein delivery throughout the gastrointestinal tract. Furthermore, we highlight methods utilised to circumvent these barriers and promote improved intestinal permeability. Lastly, we explore in vitro models employed to predict epithelial transport. Key findings highlight the need to carefully understand gastrointestinal physiology, allowing specific engineering of oral delivery systems for biomolecules. Significant importance is placed upon understanding enzymatic degradation susceptibility as well as uptake mechanisms for particulate and protein-based therapeutics for the development of successful oral protein delivery platforms.
Collapse
Affiliation(s)
- Leah Wright
- School of Pharmacy and Medical Science, University of South Australia, 5001, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UniSA, Australia
| | - Timothy J Barnes
- School of Pharmacy and Medical Science, University of South Australia, 5001, Australia
| | - Clive A Prestidge
- School of Pharmacy and Medical Science, University of South Australia, 5001, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UniSA, Australia.
| |
Collapse
|
27
|
Davies M, Jones RDO, Grime K, Jansson-Löfmark R, Fretland AJ, Winiwarter S, Morgan P, McGinnity DF. Improving the Accuracy of Predicted Human Pharmacokinetics: Lessons Learned from the AstraZeneca Drug Pipeline Over Two Decades. Trends Pharmacol Sci 2020; 41:390-408. [PMID: 32359836 DOI: 10.1016/j.tips.2020.03.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 03/20/2020] [Accepted: 03/25/2020] [Indexed: 01/15/2023]
Abstract
During drug discovery and prior to the first human dose of a novel candidate drug, the pharmacokinetic (PK) behavior of the drug in humans is predicted from preclinical data. This helps to inform the likelihood of achieving therapeutic exposures in early clinical development. Once clinical data are available, the observed human PK are compared with predictions, providing an opportunity to assess and refine prediction methods. Application of best practice in experimental data generation and predictive methodologies, and a focus on robust mechanistic understanding of the candidate drug disposition properties before nomination to clinical development, have led to maximizing the probability of successful PK predictions so that 83% of AstraZeneca drug development projects progress in the clinic with no PK issues; and 71% of key PK parameter predictions [64% of area under the curve (AUC) predictions; 78% of maximum concentration (Cmax) predictions; and 70% of half-life predictions] are accurate to within twofold. Here, we discuss methods to predict human PK used by AstraZeneca, how these predictions are assessed and what can be learned from evaluating the predictions for 116 candidate drugs.
Collapse
Affiliation(s)
- Michael Davies
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK.
| | - Rhys D O Jones
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Ken Grime
- DMPK, Research and Early Development, Respiratory, Inflammation and Autoimmune, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rasmus Jansson-Löfmark
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Adrian J Fretland
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Boston, MA, USA
| | - Susanne Winiwarter
- DMPK, Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Paul Morgan
- Mechanistic Safety and ADME Sciences, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Dermot F McGinnity
- DMPK, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
28
|
The Segregated Intestinal Flow Model (SFM) for Drug Absorption and Drug Metabolism: Implications on Intestinal and Liver Metabolism and Drug-Drug Interactions. Pharmaceutics 2020; 12:pharmaceutics12040312. [PMID: 32244748 PMCID: PMC7238003 DOI: 10.3390/pharmaceutics12040312] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 03/26/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022] Open
Abstract
The properties of the segregated flow model (SFM), which considers split intestinal flow patterns perfusing an active enterocyte region that houses enzymes and transporters (<20% of the total intestinal blood flow) and an inactive serosal region (>80%), were compared to those of the traditional model (TM), wherein 100% of the flow perfuses the non-segregated intestine tissue. The appropriateness of the SFM model is important in terms of drug absorption and intestinal and liver drug metabolism. Model behaviors were examined with respect to intestinally (M1) versus hepatically (M2) formed metabolites and the availabilities in the intestine (FI) and liver (FH) and the route of drug administration. The %contribution of the intestine to total first-pass metabolism bears a reciprocal relation to that for the liver, since the intestine, a gateway tissue, regulates the flow of substrate to the liver. The SFM predicts the highest and lowest M1 formed with oral (po) and intravenous (iv) dosing, respectively, whereas the extent of M1 formation is similar for the drug administered po or iv according to the TM, and these values sit intermediate those of the SFM. The SFM is significant, as this drug metabolism model explains route-dependent intestinal metabolism, describing a higher extent of intestinal metabolism with po versus the much reduced or absence of intestinal metabolism with iv dosing. A similar pattern exists for drug–drug interactions (DDIs). The inhibitor or inducer exerts its greatest effect on victim drugs when both inhibitor/inducer and drug are given po. With po dosing, more drug or inhibitor/inducer is brought into the intestine for DDIs. The bypass of flow and drug to the enterocyte region of the intestine after intravenous administration adds complications to in vitro–in vivo extrapolations (IVIVE).
Collapse
|
29
|
O’Dwyer PJ, Imanidis G, Box KJ, Reppas C. On the Usefulness of Two Small-Scale In Vitro Setups in the Evaluation of Luminal Precipitation of Lipophilic Weak Bases in Early Formulation Development. Pharmaceutics 2020; 12:pharmaceutics12030272. [PMID: 32188116 PMCID: PMC7151110 DOI: 10.3390/pharmaceutics12030272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/08/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023] Open
Abstract
A small-scale biphasic dissolution setup and a small-scale dissolution-permeation (D-P) setup were evaluated for their usefulness in simulating the luminal precipitation of three lipophilic weak bases—dipyridamole, ketoconazole and itraconazole. The transition from the gastric to intestinal environment was incorporated into both experimental procedures. Emulsification during the biphasic dissolution experiments had a minimal impact on the data, when appropriate risk mitigation steps were incorporated. Precipitation parameters estimated from the in vitro data were inputted into the Simcyp® physiologically based pharmacokinetic (PBPK) modelling software and simulated human plasma profiles were compared with previously published pharmacokinetic data. Average Cmax and AUC values estimated using experimentally derived precipitation parameters from the biphasic experiments deviated from corresponding published actual values less than values estimated using the default simulator parameters for precipitation. The slow rate of transport through the biomimetic membrane in the D-P setup limited its usefulness in forecasting the rates of in vivo precipitation used in the modelling of average plasma profiles.
Collapse
Affiliation(s)
- Patrick J. O’Dwyer
- Pion Inc. (UK) Ltd., Forest Row, East Sussex RH18 5DW, UK; (P.J.O.); (K.J.B.)
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, GR 157 84 Zografou, Greece
| | - Georgios Imanidis
- School of Life Sciences, Institute of Pharma Technology, University of Applied Sciences Northwestern Switzerland, Hofackerstrasse 30, 4132 Muttenz, Switzerland;
- Department of Pharmaceutical Sciences, University of Basel, CH 4056 Basel, Switzerland
| | - Karl J. Box
- Pion Inc. (UK) Ltd., Forest Row, East Sussex RH18 5DW, UK; (P.J.O.); (K.J.B.)
| | - Christos Reppas
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, GR 157 84 Zografou, Greece
- Correspondence: ; Tel.: +30-210-727-4678; Fax: +30-210-727-4027
| |
Collapse
|
30
|
Rathaur P, SR KJ. Metabolism and Pharmacokinetics of Phytochemicals in the Human Body. Curr Drug Metab 2020; 20:1085-1102. [DOI: 10.2174/1389200221666200103090757] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/27/2019] [Accepted: 11/14/2019] [Indexed: 12/12/2022]
Abstract
Background:Phytochemicals are obtained from various plants and used for the treatment of diseases as both traditional and modern medicines. Poor bioavailability of phytochemicals is a major concern in applying phytochemicals as a therapeutic agent. It is, therefore, necessary to understand the metabolism and pharmacokinetics of phytochemicals for its implication as a therapeutic agent.Methods:Articles on the metabolism of phytochemicals from the PubMed database. The articles were classified into the digestion, absorption, metabolism, excretion, toxicity, and bioavailability of phytochemicals and the effect of gut microbiota on the metabolism of phytochemicals.Results:The metabolism of each phytochemical is largely dependent on the individual's digestive ability, membrane transporters, metabolizing enzymes and gut microbiota. Further, the form of the phytochemical and genetic make-up of the individual greatly influences the metabolism of phytochemicals.Conclusion:The metabolism of phytochemicals is mostly depended on the form of phytochemicals and individualspecific variations in the metabolism of phytochemicals. Understanding the metabolism and pharmacokinetics of phytochemicals might help in applying plant-based medicines for the treatment of various diseases.
Collapse
Affiliation(s)
- Pooja Rathaur
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad, India
| | - Kaid Johar SR
- Department of Zoology, Biomedical Technology and Human Genetics, School of Sciences, Gujarat University, Ahmedabad, India
| |
Collapse
|
31
|
Bransford P, Cook J, Gupta M, Haertter S, He H, Ju R, Kanodia J, Lennernäs H, Lindley D, Polli JE, Wenning L, Wu Y. ICH M9 Guideline in Development on Biopharmaceutics Classification System-Based Biowaivers: An Industrial Perspective from the IQ Consortium. Mol Pharm 2020; 17:361-372. [DOI: 10.1021/acs.molpharmaceut.9b01062] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Philip Bransford
- Vertex Pharmaceuticals Incorporated, 50 Northern Avenue, Boston, Massachusetts 02210, United States
| | - Jack Cook
- Clinical Pharmacology Department, Global Product Development, Pfizer, Inc., Groton, Connecticut 06320, United States
| | - Manish Gupta
- Biopharmaceutics, Product Development and Supply, GlaxoSmithKline, Collegeville, Pennsylvania 19426, United States
| | - Sebastian Haertter
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut 06877, United States
| | - Handan He
- Department of Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, East Hanover, New Jersey 07936, United States
| | - Rob Ju
- Drug Product Development, Abbvie, North Chicago, Illinois 60064, United States
| | - Jitendra Kanodia
- Theravance Biopharma US, Inc., South San Francisco, California 94080, United States
| | - Hans Lennernäs
- Department of Pharmacy, Uppsala University, Box 580, 751 23 Uppsala, Sweden
| | - David Lindley
- AbbVie Inc., North Chicago, Illinois 60064, United States
| | - James E. Polli
- Department of Pharmaceutical Sciences, University of Maryland, Baltimore, Maryland 21201, United States
| | - Larissa Wenning
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Yunhui Wu
- Pharmaceutical Sciences, MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
32
|
In Vitro Dissolution and in Silico Modeling Shortcuts in Bioequivalence Testing. Pharmaceutics 2020; 12:pharmaceutics12010045. [PMID: 31947944 PMCID: PMC7022479 DOI: 10.3390/pharmaceutics12010045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/31/2019] [Accepted: 01/02/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose: To review in vitro testing and simulation platforms that are in current use to predict in vivo performances of generic products as well as other situations to provide evidence for biowaiver and support drug formulations development. Methods: Pubmed and Google Scholar databases were used to review published literature over the past 10 years. The terms used were “simulation AND bioequivalence” and “modeling AND bioequivalence” in the title field of databases, followed by screening, and then reviewing. Results: A total of 22 research papers were reviewed. Computer simulation using software such as GastroPlus™, PK-Sim® and SimCyp® find applications in drug modeling. Considering the wide use of optimization for in silico predictions to fit observed data, a careful review of publications is required to validate the reliability of these platforms. For immediate release (IR) drug products belonging to the Biopharmaceutics Classification System (BCS) classes I and III, difference factor (ƒ1) and similarity factor (ƒ2) are calculated from the in vitro dissolution data of drug formulations to support biowaiver; however, this method can be more discriminatory and may not be useful for all dissolution profiles. Conclusions: Computer simulation platforms need to improve their mechanistic physiologically based pharmacokinetic (PBPK) modeling, and if prospectively validated within a small percentage of error from the observed clinical data, they can be valuable tools in bioequivalence (BE) testing and formulation development.
Collapse
|
33
|
Matsumura N, Hayashi S, Akiyama Y, Ono A, Funaki S, Tamura N, Kimoto T, Jiko M, Haruna Y, Sarashina A, Ishida M, Nishiyama K, Fushimi M, Kojima Y, Yoneda K, Nakanishi M, Kim S, Fujita T, Sugano K. Prediction Characteristics of Oral Absorption Simulation Software Evaluated Using Structurally Diverse Low-Solubility Drugs. J Pharm Sci 2019; 109:1403-1416. [PMID: 31863733 DOI: 10.1016/j.xphs.2019.12.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 12/11/2019] [Accepted: 12/11/2019] [Indexed: 02/06/2023]
Abstract
The purpose of the present study was to characterize current biopharmaceutics modeling and simulation software regarding the prediction of the fraction of a dose absorbed (Fa) in humans. As commercial software products, GastroPlus™ and Simcyp® were used. In addition, the gastrointestinal unified theoretical framework, a simple and publicly accessible model, was used as a benchmark. The Fa prediction characteristics for a total of 96 clinical Fa data of 27 model drugs were systematically evaluated using the default settings of each software product. The molecular weight, dissociation constant, octanol-water partition coefficient, solubility in biorelevant media, dose, and particle size of model drugs were used as input data. Although the same input parameters were used, GastroPlus™, Simcyp®, and the gastrointestinal unified theoretical framework showed different Fa prediction characteristics depending on the rate-limiting steps of oral drug absorption. The results of the present study would be of great help for the overall progression of physiologically based absorption models.
Collapse
Affiliation(s)
- Naoya Matsumura
- Minase Research Institute, Ono Pharmaceutical Co., Ltd., 3-1-1, Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan.
| | - Shun Hayashi
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugadenaka, Konohana-ku, Osaka 554-0022, Japan
| | - Yoshiyuki Akiyama
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Asami Ono
- Laboratory for Chemistry, Manufacturing and Control Pharmaceuticals Research Center, Asahi Kasei Pharma Corporation, 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Satoko Funaki
- Drug Metabolism & Pharmacokinetics, Research Laboratory for Development, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Naomi Tamura
- Drug Metabolism & Pharmacokinetics, Research Laboratory for Development, Shionogi & Co., Ltd., 3-1-1, Futaba-cho, Toyonaka-shi, Osaka 561-0825, Japan
| | - Takahiro Kimoto
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka 569-1125, Japan
| | - Maiko Jiko
- Medical Analysis Research Department, Towa Pharmaceutical Co., Ltd., 134 Chudoji Minami-machi, Shimogyo-ku, Kyoto 600-8813, Japan
| | - Yuka Haruna
- Medical Analysis Research Department, Towa Pharmaceutical Co., Ltd., 134 Chudoji Minami-machi, Shimogyo-ku, Kyoto 600-8813, Japan
| | - Akiko Sarashina
- Clinical PK/PD Department, Nippon Boehringer Ingelheim Co., Ltd., 6-7-5 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Masahiro Ishida
- Clinical PK/PD Department, Nippon Boehringer Ingelheim Co., Ltd., 6-7-5 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Kotaro Nishiyama
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., 6-7-5 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | - Masahiro Fushimi
- Biological Research Department, Sawai Pharmaceutical Co., Ltd., 5-2-30, Miyahara, Yodogawa-ku, Osaka 532-0003, Japan
| | - Yukiko Kojima
- Biological Research Department, Sawai Pharmaceutical Co., Ltd., 5-2-30, Miyahara, Yodogawa-ku, Osaka 532-0003, Japan
| | - Kazuhiro Yoneda
- Minase Research Institute, Ono Pharmaceutical Co., Ltd., 3-1-1, Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - Misato Nakanishi
- Minase Research Institute, Ono Pharmaceutical Co., Ltd., 3-1-1, Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - Soonih Kim
- Minase Research Institute, Ono Pharmaceutical Co., Ltd., 3-1-1, Sakurai, Shimamoto-cho, Mishima-gun, Osaka 618-8585, Japan
| | - Takuya Fujita
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| | - Kiyohiko Sugano
- Molecular Pharmaceutics Laboratory, College of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-higashi, Kusatsu, Shiga 525-8577, Japan
| |
Collapse
|
34
|
|
35
|
Preformulation Studies and Enabling Formulation Selection for an Insoluble Compound at Preclinical Stage-From In Vitro, In Silico to In Vivo. J Pharm Sci 2019; 109:950-958. [PMID: 31647952 DOI: 10.1016/j.xphs.2019.10.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/19/2019] [Accepted: 10/11/2019] [Indexed: 01/15/2023]
Abstract
The objective of this work was to identify an enabling formulation for an insoluble compound ZL006 with potency of boosting leukocytes after chemotherapy. The low oral bioavailability (<1%) of its conventional suspension was the hurdle for the preclinical evaluation via oral administration. Preformulation studies including physical form screening and physicochemical properties determination were performed. Polymorphism was observed, and the more thermodynamically stable form was selected for further studies. ZL006 showed certain supersaturation solubility, although the thermodynamic solubility in FaSSIF was low, which indicated the supersaturating formulation might work. Parameter sensitivity analysis by in silico simulation predicted that in vivo exposure was sensitive to solubility, while particle size reduction would have limited impact on exposure. Based on in silico prediction and the understanding of the molecule from preformulation studies, solid dispersion approach was selected. A preliminary dose escalation pharmacokinetic study in rats demonstrated that in vivo exposure increased in dose-proportional manner from 12.5 mg/kg to 50 mg/kg with around 50% oral bioavailability after oral dosing of the solid dispersion. This work showed that combination of preformulation studies and in silico simulation could efficiently guide the selection of enabling formulation, which could save resources at preclinical stage.
Collapse
|
36
|
Combining biorelevant in vitro and in silico tools to simulate and better understand the in vivo performance of a nano-sized formulation of aprepitant in the fasted and fed states. Eur J Pharm Sci 2019; 138:105031. [DOI: 10.1016/j.ejps.2019.105031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/01/2019] [Accepted: 08/01/2019] [Indexed: 11/21/2022]
|
37
|
Dahlgren D, Lennernäs H. Intestinal Permeability and Drug Absorption: Predictive Experimental, Computational and In Vivo Approaches. Pharmaceutics 2019; 11:pharmaceutics11080411. [PMID: 31412551 PMCID: PMC6723276 DOI: 10.3390/pharmaceutics11080411] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023] Open
Abstract
The main objective of this review is to discuss recent advancements in the overall investigation and in vivo prediction of drug absorption. The intestinal permeability of an orally administered drug (given the value Peff) has been widely used to determine the rate and extent of the drug’s intestinal absorption (Fabs) in humans. Preclinical gastrointestinal (GI) absorption models are currently in demand for the pharmaceutical development of novel dosage forms and new drug products. However, there is a strong need to improve our understanding of the interplay between pharmaceutical, biopharmaceutical, biochemical, and physiological factors when predicting Fabs and bioavailability. Currently, our knowledge of GI secretion, GI motility, and regional intestinal permeability, in both healthy subjects and patients with GI diseases, is limited by the relative inaccessibility of some intestinal segments of the human GI tract. In particular, our understanding of the complex and highly dynamic physiology of the region from the mid-jejunum to the sigmoid colon could be significantly improved. One approach to the assessment of intestinal permeability is to use animal models that allow these intestinal regions to be investigated in detail and then to compare the results with those from simple human permeability models such as cell cultures. Investigation of intestinal drug permeation processes is a crucial biopharmaceutical step in the development of oral pharmaceutical products. The determination of the intestinal Peff for a specific drug is dependent on the technique, model, and conditions applied, and is influenced by multiple interactions between the drug molecule and the biological membranes.
Collapse
Affiliation(s)
- David Dahlgren
- Department of Pharmacy, Uppsala University, Box 580 SE-751 23 Uppsala, Sweden
| | - Hans Lennernäs
- Department of Pharmacy, Uppsala University, Box 580 SE-751 23 Uppsala, Sweden.
| |
Collapse
|
38
|
Effect of Ticagrelor, a Cytochrome P450 3A4 Inhibitor, on the Pharmacokinetics of Tadalafil in Rats. Pharmaceutics 2019; 11:pharmaceutics11070354. [PMID: 31330787 PMCID: PMC6680770 DOI: 10.3390/pharmaceutics11070354] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023] Open
Abstract
Tadalafil is a cytochrome P450 (CYP) 3A4 substrate. Because there are few data on drug-drug interactions, it is advisable to take sufficient consideration when co-administering tadalafil with CYP3A4 inducers or inhibitors. This study was conducted to assess the effect of ticagrelor, a CYP3A4 inhibitor, on the pharmacokinetic properties of tadalafil after oral administration to rats. A total of 20 Sprague–Dawley male rats were randomly divided into the non-pretreated group and ticagrelor-pretreated group, and tadalafil was orally administered to each group after pretreatment with or without ticagrelor. Blood samples were collected at predetermined time points after oral administration of tadalafil. As a result, systemic exposure of tadalafil in the ticagrelor-pretreated group was significantly increased compared to the non-pretreated group (1.61-fold), and the clearance of tadalafil in the ticagrelor-pretreated group was significantly reduced than the non-pretreated group (37%). The prediction of the drug profile through the one-compartment model could explain the differences of pharmacokinetic properties of tadalafil in the non-pretreated and ticagrelor-pretreated groups. This study suggests that ticagrelor reduces a CYP3A-mediated tadalafil metabolism and that tadalafil and a combination regimen with tadalafil and ticagrelor requires dose control and specific pharmacotherapy.
Collapse
|
39
|
O'Driscoll CM, Bernkop-Schnürch A, Friedl JD, Préat V, Jannin V. Oral delivery of non-viral nucleic acid-based therapeutics - do we have the guts for this? Eur J Pharm Sci 2019; 133:190-204. [PMID: 30946964 DOI: 10.1016/j.ejps.2019.03.027] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/25/2019] [Accepted: 03/28/2019] [Indexed: 12/22/2022]
Abstract
Gene therapy with RNA and pDNA-based drugs is limited by poor enzymatic stability and poor cellular permeation. The delivery of nucleic acids, in particular by the oral route, remains a major hurdle. This review will focus on the barriers to the oral delivery of nucleic acids and the strategies, in particular formulation strategies, which have been developed to overcome these barriers. Due to their very low oral bioavailability, the most obvious and most investigated biomedical applications for their oral delivery are related to the local treatment of inflammatory bowel diseases and colorectal cancers. Preclinical data but not yet clinical studies support the potential use of the oral route for the local delivery of formulated nucleic acid-based drugs.
Collapse
Affiliation(s)
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria.
| | - Julian D Friedl
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, 6020 Innsbruck, Austria
| | - Véronique Préat
- Universite catholique de Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier, 73 bte B1.73.12, 1200 Brussels, Belgium.
| | - Vincent Jannin
- Gattefossé SAS, 36 chemin de Genas, 69804 Saint-Priest cedex, France.
| |
Collapse
|
40
|
Hens B, Bolger MB. Application of a Dynamic Fluid and pH Model to Simulate Intraluminal and Systemic Concentrations of a Weak Base in GastroPlus™. J Pharm Sci 2019; 108:305-315. [DOI: 10.1016/j.xphs.2018.10.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/19/2018] [Accepted: 10/22/2018] [Indexed: 10/27/2022]
|
41
|
Macroscopic Chiral Recognition by Calix[4]arene‐Based Host–Guest Interactions. Chemistry 2018; 24:15502-15506. [DOI: 10.1002/chem.201803564] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 07/30/2018] [Indexed: 01/12/2023]
|
42
|
Boland JW, Johnson M, Ferreira D, Berry DJ. In silico (computed) modelling of doses and dosing regimens associated with morphine levels above international legal driving limits. Palliat Med 2018; 32:1222-1232. [PMID: 29724154 PMCID: PMC6041735 DOI: 10.1177/0269216318773956] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Morphine can cause central nervous system side effects which impair driving skills. The legal blood morphine concentration limit for driving is 20 µg/L in France/Poland/Netherlands and 80 µg/L in England/Wales. There is no guidance as to the morphine dose leading to this concentration. AIM The in silico (computed) relationship of oral morphine dose and plasma concentration was modelled to provide dose estimates for a morphine plasma concentration above 20 and 80 µg/L in different patient groups. DESIGN A dose-concentration model for different genders, ages and oral morphine formulations, validated against clinical pharmacokinetic data, was generated using Simcyp®, a population-based pharmacokinetic simulator. SETTING/PARTICIPANTS Healthy Northern European population parameters were used with age, gender and renal function being varied in the different simulation groups. In total, 36,000 simulated human subjects (100 per modelled group of different ages and gender) received repeated simulated morphine dosing with modified-release or immediate-release formulations. RESULTS Older age, women, modified-release formulation and worse renal function were associated with higher plasma concentrations. Across all groups, morphine doses below 20 mg/day were unlikely to result in a morphine plasma concentration above 20 µg/L; this was 80 mg/day with the 80 µg/L limit. CONCLUSION This novel study provides predictions of the in silico (computed) dose-concentration relationship for international application. Individualised morphine prescribing decisions by clinicians must be informed by clinical judgement considering the individual patient's level of impairment and insight irrespective of the blood morphine concentration as people who have impaired driving will be breaking the law. Taking into account expected morphine concentrations enables improved individualised decision making.
Collapse
Affiliation(s)
- Jason W Boland
- 1 Wolfson Palliative Care Research Centre, Hull York Medical School, University of Hull, Hull, UK
| | - Miriam Johnson
- 1 Wolfson Palliative Care Research Centre, Hull York Medical School, University of Hull, Hull, UK
| | | | | |
Collapse
|
43
|
O'Dwyer PJ, Litou C, Box KJ, Dressman JB, Kostewicz ES, Kuentz M, Reppas C. In vitro methods to assess drug precipitation in the fasted small intestine – a PEARRL review. J Pharm Pharmacol 2018; 71:536-556. [DOI: 10.1111/jphp.12951] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/28/2018] [Indexed: 12/12/2022]
Abstract
Abstract
Objectives
Drug precipitation in vivo poses a significant challenge for the pharmaceutical industry. During the drug development process, the impact of drug supersaturation or precipitation on the in vivo behaviour of drug products is evaluated with in vitro techniques. This review focuses on the small and full scale in vitro methods to assess drug precipitation in the fasted small intestine.
Key findings
Many methods have been developed in an attempt to evaluate drug precipitation in the fasted state, with varying degrees of complexity and scale. In early stages of drug development, when drug quantities are typically limited, small-scale tests facilitate an early evaluation of the potential precipitation risk in vivo and allow rapid screening of prototype formulations. At later stages of formulation development, full-scale methods are necessary to predict the behaviour of formulations at clinically relevant doses. Multicompartment models allow the evaluation of drug precipitation after transfer from stomach to the upper small intestine. Optimisation of available biopharmaceutics tools for evaluating precipitation in the fasted small intestine is crucial for accelerating the development of novel breakthrough medicines and reducing the development costs.
Summary
Despite the progress from compendial quality control dissolution methods, further work is required to validate the usefulness of proposed setups and to increase their biorelevance, particularly in simulating the absorption of drug along the intestinal lumen. Coupling results from in vitro testing with physiologically based pharmacokinetic modelling holds significant promise and requires further evaluation.
Collapse
Affiliation(s)
- Patrick J O'Dwyer
- Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| | - Chara Litou
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Karl J Box
- Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK
| | - Jennifer B Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Edmund S Kostewicz
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Martin Kuentz
- University of Applied Sciences and Arts Northwestern Switzerland, Muttenz, Switzerland
| | - Christos Reppas
- Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Zografou, Greece
| |
Collapse
|
44
|
Madsen CM, Feng KI, Leithead A, Canfield N, Jørgensen SA, Müllertz A, Rades T. Effect of composition of simulated intestinal media on the solubility of poorly soluble compounds investigated by design of experiments. Eur J Pharm Sci 2018; 111:311-319. [DOI: 10.1016/j.ejps.2017.10.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 09/14/2017] [Accepted: 10/02/2017] [Indexed: 11/29/2022]
|
45
|
Samant TS, Dhuria S, Lu Y, Laisney M, Yang S, Grandeury A, Mueller‐Zsigmondy M, Umehara K, Huth F, Miller M, Germa C, Elmeliegy M. Ribociclib Bioavailability Is Not Affected by Gastric pH Changes or Food Intake: In Silico and Clinical Evaluations. Clin Pharmacol Ther 2017; 104:374-383. [PMID: 29134635 PMCID: PMC6099197 DOI: 10.1002/cpt.940] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/30/2017] [Accepted: 11/05/2017] [Indexed: 02/06/2023]
Abstract
Ribociclib (KISQALI), a cyclin‐dependent kinase 4/6 inhibitor approved for the first‐line treatment of HR+/HER2– advanced breast cancer with an aromatase inhibitor, is administered with no restrictions on concomitant gastric pH‐elevating agents or food intake. The influence of proton pump inhibitors (PPIs) on ribociclib bioavailability was assessed using 1) biorelevant media solubility, 2) physiologically based pharmacokinetic (PBPK) modeling, 3) noncompartmental analysis (NCA) of clinical trial data, and 4) population PK (PopPK) analysis. This multipronged approach indicated no effect of gastric pH changes on ribociclib PK and served as a platform for supporting ribociclib labeling language, stating no impact of gastric pH‐altering agents on the absorption of ribociclib, without a dedicated drug–drug interaction trial. The bioequivalence of ribociclib exposure with or without a high‐fat meal was demonstrated in a clinical trial. Lack of restrictions on ribociclib dosing may facilitate better patient compliance and therefore clinical benefit.
Collapse
Affiliation(s)
| | | | - Yasong Lu
- Novartis PharmaceuticalsEast HanoverNew JerseyUSA
| | | | - Shu Yang
- Novartis PharmaceuticalsEast HanoverNew JerseyUSA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Harloff-Helleberg S, Nielsen LH, Nielsen HM. Animal models for evaluation of oral delivery of biopharmaceuticals. J Control Release 2017; 268:57-71. [DOI: 10.1016/j.jconrel.2017.09.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 09/06/2017] [Accepted: 09/15/2017] [Indexed: 12/20/2022]
|
47
|
Yu A, Jackson T, Tsume Y, Koenigsknecht M, Wysocki J, Marciani L, Amidon GL, Frances A, Baker JR, Hasler W, Wen B, Pai A, Sun D. Mechanistic Fluid Transport Model to Estimate Gastrointestinal Fluid Volume and Its Dynamic Change Over Time. AAPS JOURNAL 2017; 19:1682-1690. [DOI: 10.1208/s12248-017-0145-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 09/01/2017] [Indexed: 12/22/2022]
|
48
|
Kambayashi A, Dressman JB. Forecasting gastrointestinal precipitation and oral pharmacokinetics of dantrolene in dogs using an in vitro precipitation testing coupled with in silico modeling and simulation. Eur J Pharm Biopharm 2017; 119:107-113. [PMID: 28619610 DOI: 10.1016/j.ejpb.2017.06.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/06/2017] [Accepted: 06/11/2017] [Indexed: 11/26/2022]
|
49
|
Nagar S, Korzekwa RC, Korzekwa K. Continuous Intestinal Absorption Model Based on the Convection-Diffusion Equation. Mol Pharm 2017; 14:3069-3086. [PMID: 28712300 DOI: 10.1021/acs.molpharmaceut.7b00286] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Prediction of the rate and extent of drug absorption upon oral dosing needs models that capture the complexities of both the drug molecule and intestinal physiology. We report here the development of a continuous intestinal absorption model based on the convection-diffusion equation. The model includes explicit enterocyte apical membrane and intracellular lipid radial compartments along the length of the intestine. Physiologic functions along length x are built into the model and include velocity, diffusion, surface areas, and pH of the intestine. Also included are expression levels of the intestinal active uptake transporter OATP2B1 and efflux transporter P-gp. Oral dosing of solution as well as solid (with a dissolution function) was modeled for several drugs. The fraction absorbed (FA) and concentration-time (C-t) profiles were predicted and compared with clinical data. Overall, FA was well predicted upon oral (n = 21) or colonic dosing (n = 11), with four outliers. The overall accuracy (prediction of the correct bin) was 81% with outliers and 90% without outliers. Of the nine solution dosing data sets, six drugs were very well predicted with an exposure overlap coefficient (EOC) > 0.9 and predicted Cmax and Tmax values similar to those observed. Of the six solid dose formulations evaluated, the EOC values were > 0.9 for all drugs except budesonide. The observed precipitation of nifedipine at high doses was predicted by the model. Most of the poor predictions were for drugs that are known to be transporter substrates. As proof of concept, incorporating OATP2B1 and P-gp markedly improved the EOC and predicted Cmax and Tmax for fexofenadine. Finally, the continuous intestinal model accurately recapitulated the known relationships between drug absorption and permeability, solubility, and particle size. Together, these results indicate that this preliminary intestinal absorption model offers a simple and straightforward framework to build in complexities such as drug permeability, lipid partitioning, solubility, metabolism, and transport for improved prediction of the rate and extent of drug absorption.
Collapse
Affiliation(s)
- Swati Nagar
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy , Philadelphia, Pennsylvania 19140, United States
| | - Richard C Korzekwa
- Department of Physics, University of Texas , Austin, Texas 78712, United States
| | - Ken Korzekwa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy , Philadelphia, Pennsylvania 19140, United States
| |
Collapse
|
50
|
Abstract
Drug discovery is a multidisciplinary and multivariate optimization endeavor. As such, in silico screening tools have gained considerable importance to archive, analyze and exploit the vast and ever-increasing amount of experimental data generated throughout the process. The current review will focus on the computer-aided prediction of the numerous properties that need to be controlled during the discovery of a preliminary hit and its promotion to a viable clinical candidate. It does not pretend to the almost impossible task of an exhaustive report but will highlight a few key points that need to be collectively addressed both by chemists and biologists to fuel the drug discovery pipeline with innovative and safe drug candidates.
Collapse
Affiliation(s)
- Didier Rognan
- Laboratoire d'Innovation Thérapeutique, UMR 7200 CNRS-Université de Strasbourg, 74 route du Rhin, 67400 Illkirch, France.
| |
Collapse
|