1
|
Ueda K, Takemoto S, Higashi K, Moribe K. Impact of colloidal drug-rich droplet size and amorphous solubility on drug membrane permeability: A comprehensive analysis. J Pharm Sci 2025; 114:136-144. [PMID: 38942292 DOI: 10.1016/j.xphs.2024.06.017] [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: 05/20/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
This study aimed to investigate the impact of amorphous solubility and colloidal drug-rich droplets on drug absorption. The amorphous solubility of cilnidipine (CND) in AS-HF grade of hypromellose acetate succinate (HPMC-AS) solution was significantly reduced compared to that in non-polymer solution due to AS-HF partitioning into the CND-rich phase. In contrast, AS-LF grade of HPMC-AS has minimal effect on the amorphous solubility. The size of colloidal CND-rich droplets formed in the CND-supersaturated solution was less than 100 nm in the presence of AS-HF, while 200-450 nm in the presence of AS-LF. When the CND concentrations were near the amorphous solubility, CND membrane flux was reduced in the presence of AS-HF due to the decrease in the amorphous solubility of CND. However, the CND flux increased with the increase in CND-rich droplets, especially in the AS-HF solution. The size reduction of the CND-rich droplets led to their effective diffusion into the unstirred water layer, enhancing CND flux. In higher CND concentration regions, the CND flux became higher in the AS-HF solution than in the AS-LF solution. Thus, it is essential to elucidate the drug concentration-dependent impact of the colloidal drug-rich droplets on the drug absorption performance to optimize supersaturating formulations.
Collapse
Affiliation(s)
- Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan.
| | - Shiryu Takemoto
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kenjirou Higashi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kunikazu Moribe
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
2
|
Patel RP, Taylor LS, Polli JE. Impact of drug incorporation into micelle on reduced griseofulvin and meloxicam permeation across a hollow fiber membrane. J Pharm Sci 2025; 114:402-415. [PMID: 39426562 DOI: 10.1016/j.xphs.2024.10.017] [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: 06/08/2024] [Revised: 10/10/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
A hollow fiber membrane (HFM) was previously characterized as a potential permeation component of a dissolution/permeation system. Two objectives were to assess the impact of micellization on drug permeation across HFM and identify a preferred permeation model from three models: permeation from only free drug, permeation from both free drug and micelle-bound drug, and permeation with enhancement from micelle shuttling. HFM studies were conducted under unsaturated drug conditions, using griseofulvin and the more hydrophilic drug meloxicam, with and without surfactant [sodium lauryl sulfate, polysorbate 80, and polyoxyethylene (10) lauryl ether]. Griseofulvin was micelle incorporated to a greater extent than meloxicam, such that griseofulvin flux decreased to a greater extent than for meloxicam. The griseofulvin permeation model from only free drug was rejected, since griseofulvin flux required free drug to be about 5-20 fold higher in HFM flux studies than supported by solubility studies, depending on surfactant. Permeation from both free griseofulvin and micelle-bound griseofulvin successfully accommodated observed flux, where micelle permeability was about 5-fold lower than free drug permeability for HFM with 10 KDa MWCO. Permeation with enhancement from micelle shuttling was not the preferred explanation, although the model accommodated flux data and provided aqueous boundary layer thicknesses similar to other setups.
Collapse
Affiliation(s)
- Roshni P Patel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - James E Polli
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA.
| |
Collapse
|
3
|
Ueda K, Moseson DE, Taylor LS. Amorphous solubility advantage: Theoretical considerations, experimental methods, and contemporary relevance. J Pharm Sci 2025; 114:18-39. [PMID: 39222748 DOI: 10.1016/j.xphs.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Twenty-five years ago, Hancock and Parks asked a provocative question: "what is the true solubility advantage for amorphous pharmaceuticals?" Difficulties in determining the amorphous solubility have since been overcome due to significant advances in theoretical understanding and experimental methods. The amorphous solubility is now understood to be the concentration after the drug undergoes liquid-liquid or liquid-glass phase separation, forming a water-saturated drug-rich phase in metastable equilibrium with an aqueous phase containing molecularly dissolved drug. While crystalline solubility is an essential parameter impacting the absorption of crystalline drug formulations, amorphous solubility is a vital factor for considering absorption from supersaturating formulations. However, the amorphous solubility of drugs is complex, especially in the presence of formulation additives and gastrointestinal components, and concentration-based measurements may not indicate the maximum drug thermodynamic activity. This review discusses the concept of the amorphous solubility advantage, including a historical perspective, theoretical considerations, experimental methods for amorphous solubility measurement, and the contribution of supersaturation and amorphous solubility to drug absorption. Leveraging amorphous solubility and understanding the associated physicochemical principles can lead to more effective development strategies for poorly water-soluble drugs, ultimately benefiting therapeutic outcomes.
Collapse
Affiliation(s)
- Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Dana E Moseson
- Worldwide Research and Development, Pfizer, Inc., Groton, CT 06340, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
4
|
Yang DH, Najafian S, Chaudhuri B, Li N. The Particle Drifting Effect: A Combined Function of Colloidal and Drug Properties. Mol Pharm 2024; 21:5510-5528. [PMID: 39332024 DOI: 10.1021/acs.molpharmaceut.4c00751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/29/2024]
Abstract
The particle drifting effect, where nanosized colloidal drug particles overcome the diffusional resistance of the aqueous boundary layer adjacent to the intestinal wall and increase drug absorption rates, is drawing increasing attention in pharmaceutical research. However, mechanistic understanding and accurate prediction of the particle drifting effect remain lacking. In this study, we systematically evaluated the extent of the particle drifting effect affected by drug and colloidal properties, including the size, number, and type of the moving species using biphasic diffusion experiments combined with computational fluid dynamics simulations and mass transport analyses. The results showed that the particle drifting effect is a sequential reaction of particle dissolution/dissociation in the diffusional boundary layer, followed by absorption of the free drug. Therefore, factors affecting the rate-limiting step, which can be either process or both under different circumstances, alter the particle drifting effect. Experimental results also agree with the theory that the particle dissolution rate is dependent on particle size, concentration, and drug solubility. In addition, rapid bile micelle dissociation and bile salt absorption facilitated drug absorption by the particle drifting effect. Our findings explain the highly dynamic nature of the particle drifting effect and will contribute to rational formulation development and better bioavailability prediction for formulations containing colloidal particles.
Collapse
Affiliation(s)
- Da Hye Yang
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States
| | - Saeed Najafian
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States
- Department of Chemical & Biomolecular Engineering, University of Connecticut, 191 Auditorium Road, Unit 3222, Storrs, Connecticut 06269, United States
| | - Bodhisattwa Chaudhuri
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States
- Institute of Materials Science, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, Connecticut 06269, United States
- Department of Chemical & Biomolecular Engineering, University of Connecticut, 191 Auditorium Road, Unit 3222, Storrs, Connecticut 06269, United States
| | - Na Li
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States
- Institute of Materials Science, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, Connecticut 06269, United States
- Department of Chemical & Biomolecular Engineering, University of Connecticut, 191 Auditorium Road, Unit 3222, Storrs, Connecticut 06269, United States
| |
Collapse
|
5
|
Pepin X, Arora S, Borges L, Cano-Vega M, Carducci T, Chatterjee P, Chen G, Cristofoletti R, Dallmann A, Delvadia P, Dressman J, Fotaki N, Gray E, Heimbach T, Holte Ø, Kijima S, Kotzagiorgis E, Lennernäs H, Lindahl A, Loebenberg R, Mackie C, Malamatari M, McAllister M, Mitra A, Moody R, Mudie D, Musuamba Tshinanu F, Polli JE, Rege B, Ren X, Rullo G, Scherholz M, Song I, Stillhart C, Suarez-Sharp S, Tannergren C, Tsakalozou E, Veerasingham S, Wagner C, Seo P. Parameterization of Physiologically Based Biopharmaceutics Models: Workshop Summary Report. Mol Pharm 2024; 21:3697-3731. [PMID: 38946085 PMCID: PMC11304397 DOI: 10.1021/acs.molpharmaceut.4c00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 07/02/2024]
Abstract
This Article shares the proceedings from the August 29th, 2023 (day 1) workshop "Physiologically Based Biopharmaceutics Modeling (PBBM) Best Practices for Drug Product Quality: Regulatory and Industry Perspectives". The focus of the day was on model parametrization; regulatory authorities from Canada, the USA, Sweden, Belgium, and Norway presented their views on PBBM case studies submitted by industry members of the IQ consortium. The presentations shared key questions raised by regulators during the mock exercise, regarding the PBBM input parameters and their justification. These presentations also shed light on the regulatory assessment processes, content, and format requirements for future PBBM regulatory submissions. In addition, the day 1 breakout presentations and discussions gave the opportunity to share best practices around key questions faced by scientists when parametrizing PBBMs. Key questions included measurement and integration of drug substance solubility for crystalline vs amorphous drugs; impact of excipients on apparent drug solubility/supersaturation; modeling of acid-base reactions at the surface of the dissolving drug; choice of dissolution methods according to the formulation and drug properties with a view to predict the in vivo performance; mechanistic modeling of in vitro product dissolution data to predict in vivo dissolution for various patient populations/species; best practices for characterization of drug precipitation from simple or complex formulations and integration of the data in PBBM; incorporation of drug permeability into PBBM for various routes of uptake and prediction of permeability along the GI tract.
Collapse
Affiliation(s)
- Xavier Pepin
- Regulatory
Affairs, Simulations Plus Inc., 42505 10th Street West, Lancaster, California 93534-7059, United States
| | - Sumit Arora
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Luiza Borges
- ANVISA, SIA Trecho 5́, Guara, Brasília, Federal District 71205-050, Brazil
| | - Mario Cano-Vega
- Drug
Product Technologies, Amgen Inc., Thousand Oaks, California 91320-1799, United
States
| | - Tessa Carducci
- Analytical
Commercialization Technology, Merck & Co., Inc., 126 E. Lincoln Ave., Rahway, New Jersey 07065, United States
| | - Parnali Chatterjee
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Grace Chen
- Takeda
Development Center Americas Inc., 300 Shire Way, Lexington, Massachusetts 02421, United States
| | - Rodrigo Cristofoletti
- College
of Pharmacy, University of Florida, 6550 Sanger Rd., Orlando, Florida 32827, United States
| | - André Dallmann
- Bayer
HealthCare SAS, 59000 Lille, France, on behalf of Bayer
AG, Pharmacometrics/Modeling and Simulation, Systems Pharmacology
& Medicine, PBPK, Leverkusen, Germany
| | - Poonam Delvadia
- Office
of Translational Science, Office of Clinical Pharmacology (OCP), Center
for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| | - Jennifer Dressman
- Fraunhofer Institute of Translational Medicine and Pharmacology, Frankfurt am Main 60596, Germany
| | - Nikoletta Fotaki
- University of Bath, Claverton Down, Bath BA2
7AY, United Kingdom
| | - Elizabeth Gray
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Tycho Heimbach
- Pharmaceutical Sciences and Clinical Supply, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Øyvind Holte
- Norwegian Medical Products Agency, Oslo 0213, Norway
| | - Shinichi Kijima
- Office
of New Drug V, Pharmaceuticals and Medical
Devices Agency (PMDA), Tokyo 100-0013, Japan
| | - Evangelos Kotzagiorgis
- European Medicines Agency (EMA), Domenico Scarlattilaan 6, Amsterdam 1083 HS, The Netherlands
| | - Hans Lennernäs
- Translational
Drug Discovery and Development, Department of Pharmaceutical Bioscience, Uppsala University, Uppsala 751 05, Sweden
| | | | - Raimar Loebenberg
- Faculty
of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmontonton T6G 2E1, Canada
| | - Claire Mackie
- Janssen
Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium
| | - Maria Malamatari
- Medicines & Healthcare Products Regulatory Agency, 10 S Colonnade, London SW1W 9SZ, United Kingdom
| | - Mark McAllister
- Global
Biopharmaceutics, Drug Product Design, Pfizer, Sandwich CT13 9NJ, United Kingdom
| | - Amitava Mitra
- Clinical
Pharmacology, Kura Oncology Inc., Boston, Massachusetts 02210, United States
| | - Rebecca Moody
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Deanna Mudie
- Global
Research and Development, Small Molecules, Lonza, 63045 NE Corporate
Pl., Bend, Oregon 97701, United States
| | - Flora Musuamba Tshinanu
- Belgian Federal Agency for Medicines and Health Products, Galileelaan 5/03, Brussel 1210, Belgium
| | - James E. Polli
- School
of Pharmacy, University of Maryland, Baltimore, Maryland 21201, United States
| | - Bhagwant Rege
- Office
of
Pharmaceutical Quality (OPQ), Center for Drug Evaluation and Research
(CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United
States
| | - Xiaojun Ren
- PK
Sciences/Translational Medicine, BioMedical Research, Novartis, One Health Plaza, East Hanover, New Jersey 07936, United States
| | - Gregory Rullo
- Regulatory
CMC, AstraZeneca, 1 Medimmune Way, Gaithersburg, Maryland 20878, United States
| | - Megerle Scherholz
- Pharmaceutical
Development, Bristol Myers Squibb, Route 206 & Province Line Road, Princeton, New Jersey 08543, United States
| | - Ivy Song
- Takeda
Development Center Americas Inc., 300 Shire Way, Lexington, Massachusetts 02421, United States
| | - Cordula Stillhart
- Pharmaceutical
R&D, F. Hoffmann-La Roche Ltd., Basel 4070, Switzerland
| | - Sandra Suarez-Sharp
- Regulatory
Affairs, Simulations Plus Inc., 42505 10th Street West, Lancaster, California 93534-7059, United States
| | - Christer Tannergren
- Biopharmaceutics
Science, New Modalities & Parenteral Product Development, Pharmaceutical
Technology & Development, Operations, AstraZeneca, Gothenburg 431 50, Sweden
| | - Eleftheria Tsakalozou
- Division
of Quantitative Methods and Modeling, Office of Research and Standards,
Office of Generic Drugs, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20903-1058, United
States
| | - Shereeni Veerasingham
- Pharmaceutical
Drugs Directorate (PDD), Health Canada, 1600 Scott St., Ottawa K1A 0K9, Canada
| | - Christian Wagner
- Global
Drug Product Development, Global CMC Development, the Healthcare Business of Merck KGaA, Darmstadt D-64293, Germany
| | - Paul Seo
- Office
of Translational Science, Office of Clinical Pharmacology (OCP), Center
for Drug Evaluation and Research (CDER), Food and Drug Administration (FDA), Silver Spring, Maryland 20903-1058, United States
| |
Collapse
|
6
|
Jangid AK, Kim S, Kim K. Delivery of piperlongumine via hyaluronic acid/phenylboronic acid-mediated dual targetable polymersome for enhanced anticancer functionality against pancreatic tumor. Int J Biol Macromol 2024; 275:133738. [PMID: 38992536 DOI: 10.1016/j.ijbiomac.2024.133738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/05/2024] [Accepted: 07/06/2024] [Indexed: 07/13/2024]
Abstract
Pancreatic cancer cells highly resistance to conventional chemo drugs, resulting low survival rates. The aim of the study was to design and develop dual targeting polymersomes (DTPS) loaded with phyto alkaloid agent i.e., piperlongumine (PL) for effective pancreatic cancer treatment. Here, hyaluronic acid (HA) was functionalized with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] (DSPEPEG-NH2), poly(ethylene glycol) bis (amine) (PEG), and phenylboronic acid (PBA) moieties. The designed DTPS could selectively recognize CD44/sialic acid (SA) and deliver PL to MIA PaCa-2 pancreatic cancer cells, facilitated via HA-CD44 and PBA-SA interactions. Drug release and stability results implied sustained PL release profile and pH sensitivity. DTPS could be more efficiently bound with SA than other sugars based on fluorescence spectroscopy. The anticancer efficacy of designed polymersomes was tested with H6C7 normal pancreas cells and SA/CD44-overexpressed MIA PaCa-2 pancreatic cancer cells. DTPS showed both SA and CD44-mediated higher cellular uptake while single-targeted polymersomes showed CD44-mediated cellular uptake. The PL-loaded DTPS efficiently uptake by MIA PaCa-2 cancer cells, causing up to 80 % cell growth inhibition, reduced cell spheroids volume and increased dead cells by 58.3 %. These results indicate that the newly developed DTPS can effectively serve as a pH-responsive drug delivery system for efficient treatment of cancer.
Collapse
Affiliation(s)
- Ashok Kumar Jangid
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul, South Korea
| | - Sungjun Kim
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul, South Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, College of Engineering, Dongguk University, Seoul, South Korea.
| |
Collapse
|
7
|
Purohit HS, Zhou D, Yu M, Zaroudi M, Oberoi H, López ADLR, Kelkar MS, He Y, Gates B, Nere N, Law D. Proof-of-Concept in Developing a 45% Drug Loaded Amorphous Nanoparticle Formulation. J Pharm Sci 2024; 113:1007-1019. [PMID: 37832919 DOI: 10.1016/j.xphs.2023.10.012] [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: 06/26/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 10/15/2023]
Abstract
Amorphous solid dispersion (ASD) is an enabling approach utilized to deliver poorly soluble compounds. ASDs can spontaneously generate drug-rich amorphous nanoparticles upon dissolution, which can act as a reservoir for maintaining supersaturation during oral absorption. But, conventional ASDs are often limited in drug loadings to < 20 %. For indications where the dose is high, this can translate into a significant pill burden. The aim of this research was to develop a high drug loading (DL) amorphous nanoparticle (ANP) formulation that can release the drug-rich nanoparticles into solution upon contact with aqueous environment. Nanoparticles were directly engineered using solvent/anti-solvent precipitation. The obtained nanoparticle suspension was then concentrated followed by solidification to a re-dispersible amorphous dosage form using spray drying or lyophilization. The impact of process variables was studied using dynamic light scattering (DLS), scanning electron microscopy (SEM), high performance liquid chromatography (HPLC), nuclear magnetic resonance (NMR) and differential scanning calorimetry (DSC). It was observed that spray drying led to a non-re-dispersible formulation. Sucrose and trehalose containing lyocakes resulted in re-dispersible formulations. The trehalose containing lyocakes, in a dog study, gave comparable performance to the reference tablet in the fasted state but lower area under the curve (AUC) in fed state.
Collapse
Affiliation(s)
- Hitesh S Purohit
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL, USA.
| | - Deliang Zhou
- Small Molecule Drug Product Development, BeiGene, Beijing, China
| | - Mengqi Yu
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL, USA
| | | | - Hardeep Oberoi
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL, USA
| | | | - Manish S Kelkar
- Small molecule CMC development, Process Engineering, AbbVie Inc., North Chicago, IL, USA
| | - Yan He
- Small molecule CMC development, Analytical Research and Development, AbbVie Inc., North Chicago, IL, USA
| | - Bradley Gates
- Small molecule CMC development, Process Chemistry, AbbVie Inc., North Chicago, IL, USA
| | - Nandkishor Nere
- Small molecule CMC development, Process Engineering, AbbVie Inc., North Chicago, IL, USA
| | - Devalina Law
- Small molecule CMC development, Drug Product Development, AbbVie Inc., North Chicago, IL, USA.
| |
Collapse
|
8
|
Yoshikawa E, Ueda K, Hakata R, Higashi K, Moribe K. Quantitative Investigation of Intestinal Drug Absorption Enhancement by Drug-Rich Nanodroplets Generated via Liquid-Liquid Phase Separation. Mol Pharm 2024; 21:1745-1755. [PMID: 38501717 DOI: 10.1021/acs.molpharmaceut.3c01078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
Drug-rich droplets formed through liquid-liquid phase separation (LLPS) have the potential to enhance the oral absorption of drugs. This can be attributed to the diffusion of these droplets into the unstirred water layer (UWL) of the gastrointestinal tract and their reservoir effects on maintaining drug supersaturation. However, a quantitative understanding of the effect of drug-rich droplets on intestinal drug absorption is still lacking. In this study, the enhancement of intestinal drug absorption through the formation of drug-rich droplets was quantitatively evaluated on a mechanistic basis. To obtain fenofibrate (FFB)-rich droplets, an amorphous solid dispersion (ASD) of FFB/hypromellose (HPMC) was dispersed in an aqueous medium. Physicochemical characterization confirmed the presence of nanosized FFB-rich droplets in the supercooled liquid state within the FFB/HPMC ASD dispersion. An in situ single-pass intestinal perfusion (SPIP) assay in rats demonstrated that increased quantities of FFB-rich nanodroplets enhanced the intestinal absorption of FFB. The effective diffusion of FFB-rich nanodroplets through UWL would partially contribute to the improved FFB absorption. Additionally, confocal laser scanning microscopy (CLSM) of cross sections of the rat intestine after the administration of fluorescently labeled FFB-rich nanodroplets showed that these nanodroplets were directly taken up by small intestinal epithelial cells. Therefore, the direct uptake of drug-rich nanodroplets by the small intestine is a potential mechanism for improving FFB absorption in the intestine. To quantitatively evaluate the impact of FFB-rich droplets on the FFB absorption enhancement, we determined the apparent permeabilities of the FFB-rich nanodroplets and dissolved FFB based on the SPIP results. The apparent permeability of the FFB-rich nanodroplets was 110-130 times lower than that of dissolved FFB. However, when the FFB-rich nanodroplet concentration was several hundred times higher than that of dissolved FFB, the FFB-rich nanodroplets contributed significantly to FFB absorption improvement. The present study highlights that drug-rich nanodroplets play a direct role in enhancing drug absorption in the gastrointestinal tract, indicating their potential for further improvement of oral absorption from ASD formulations.
Collapse
Affiliation(s)
- Etsushi Yoshikawa
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Rei Hakata
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kenjirou Higashi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kunikazu Moribe
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
9
|
Lale AS, Sirvi A, Debaje S, Patil S, Sangamwar AT. Supersaturable diacyl phospholipid dispersion for improving oral bioavailability of brick dust molecule: A case study of Aprepitant. Eur J Pharm Biopharm 2024; 197:114241. [PMID: 38432600 DOI: 10.1016/j.ejpb.2024.114241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/28/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
This study aims to investigate the potential use of polymer inclusion in the phospholipid-based solid dispersion approach for augmenting the biopharmaceutical performance of Aprepitant (APT). Initially, different polymers were screened using the microarray plate method to assess their ability to inhibit drug precipitation in the supersaturated solution and HPMCAS outperformed the others. Later, the binary (BD) and ternary (TD) phospholipid dispersions were prepared using the co-solvent evaporation method. Solid-state characterization was performed using SEM and PXRD to examine the physical properties, while molecular interactions were probed through FTIR and NMR analysis. In vitro dissolution studies were performed in both fasted and fed state biorelevant media. The results demonstrated a substantial increase in drug release from BD and TD, approximately 4.8 and 9.9 times higher compared to crystalline APT in FaSSIF. Notably, TD also showed a lowered dissolution difference between fed and fasted states in comparison to crystalline APT, indicating a reduction in the positive food effect of APT. Moreover, we assessed the impact of polymer inclusion on permeation under in vitro biomimetic conditions. In comparison with the crystalline APT suspension, both BD and TD demonstrated approximately 3.3 times and 14 times higher steady-state flux (Jss values), respectively. This can be ascribed to the supersaturation and presence of drug-rich submicron particles (nanodroplets) along with the multiple aggregates of drug with phospholipids and polymer in the donor compartment, consequently resulting in a more substantial driving force for passive diffusion. Lastly, in vivo pharmacokinetic evaluation demonstrated the enhanced absorption of both TD and BD over the free drug suspension in the fasted state. This enhancement was evident through a 2.1-fold and 1.3-fold increase in Cmax and a 2.3-fold and 1.4-fold increase in AUC0-t, respectively. Overall, these findings emphasize the potential of polymer-based phospholipid dispersion in enhancing the overall biopharmaceutical performance of APT.
Collapse
Affiliation(s)
- Ajay Sanjay Lale
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Punjab, India
| | - Arvind Sirvi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Punjab, India
| | - Shubham Debaje
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Punjab, India
| | - Sadhana Patil
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Punjab, India
| | - Abhay T Sangamwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector-67, S.A.S Nagar, Punjab, India.
| |
Collapse
|
10
|
Kambayashi A. In Silico Modeling Approaches Coupled with In Vitro Characterization in Predicting In Vivo Performance of Drug Delivery System Formulations. Mol Pharm 2023; 20:4344-4353. [PMID: 37523273 DOI: 10.1021/acs.molpharmaceut.3c00184] [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] [Indexed: 08/02/2023]
Abstract
Optimization of the in vivo performance of dosage forms in humans is essential in developing not only conventional formulations but also drug delivery system (DDS) formulations. Although animal experiments are still useful for these formulations, in silico approaches have become increasingly important for DDS formulations with regard to species-specific differences in physiology that can affect the in vivo performance of dosage forms between animals and humans. Furthermore, it is also important to couple in vitro characterizations with in silico models to predict in vivo performance in humans precisely. In this review article, I summarized in vitro-in silico approaches to predicting the in vivo performance of oral DDS formulations (amorphous solid dispersions, lipid-based formulations, nanosized formulations, cyclodextrins-based formulations, sustained release products, enteric coat products, and orally disintegrating tablets) and parenteral DDS formulations (cyclodextrins-based formulations, liposomes, and inhaled formulations).
Collapse
Affiliation(s)
- Atsushi Kambayashi
- Pharmaceutical Research and Technology Laboratories, Astellas Pharma Incorporated, 180 Ozumi, Yaizu, Shizuoka 425-0072, Japan
| |
Collapse
|
11
|
Pires FQ, Gross IP, Sa-Barreto LL, Gratieri T, Gelfuso GM, Bao SN, Cunha-Filho M. In-situ formation of nanoparticles from drug-loaded 3D polymeric matrices. Eur J Pharm Sci 2023; 188:106517. [PMID: 37406970 DOI: 10.1016/j.ejps.2023.106517] [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: 04/18/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/07/2023]
Abstract
The in-situ formation of nanoparticles from polymer-based solid medicines, although previously described, has been overlooked despite its potential to interfere with oral drug bioavailability. Such polymeric pharmaceuticals are becoming increasingly common on the market and can become even more popular due to the dizzying advance of 3D printing medicines. Hence, this work aimed to study this phenomenon during the dissolution of 3D printed tablets produced with three different polymers, hydroxypropylmethylcellulose acetate succinate (HPMCAS), polyvinyl alcohol (PVA), and Eudragit RL PO® (EUD RL) combined with plasticizers and the model drug naringenin (NAR). The components' interaction, dissolution behavior, and characteristics of the formed particles were investigated employing thermal, spectroscopic, mechanical, and chromatographic assays. All the systems generated stable spherical-shaped particles throughout 24 h, encapsulating over 25% of NAR. Results suggest encapsulation efficiencies variations may depend on interactions between polymer-drug, drug-plasticizer, and polymer-plasticizer, which formed stable nanoparticles even in the drug absence, as observed with the HPMCAS and EUD RL formulations. Additionally, components solubility in the medium and previous formulation treatments are also a decisive factor for nanoparticle formation. In particular, the treatment provided by hot-melt extrusion and FDM 3D printing affected the dissolution efficiency enhancing the interaction between the components, reverberating on particle size and particle formation kinetics mainly for HPMCAS and EUD RL. In conclusion, the 3D printing process influences the in-situ formation of nanoparticles, which can directly affect oral drug bioavailability and needs to be monitored.
Collapse
Affiliation(s)
- Felipe Q Pires
- University of Brasilia, School of Health Sciences, Laboratory of Food, Drugs and Cosmetics (LTMAC), 70.910-900, Brasília, DF, Brazil
| | - Idejan P Gross
- University of Brasilia, School of Health Sciences, Laboratory of Food, Drugs and Cosmetics (LTMAC), 70.910-900, Brasília, DF, Brazil
| | - Livia L Sa-Barreto
- University of Brasilia, Faculty of Ceilandia, 72220-900, Brasília, DF, Brazil
| | - Tais Gratieri
- University of Brasilia, School of Health Sciences, Laboratory of Food, Drugs and Cosmetics (LTMAC), 70.910-900, Brasília, DF, Brazil
| | - Guilherme M Gelfuso
- University of Brasilia, School of Health Sciences, Laboratory of Food, Drugs and Cosmetics (LTMAC), 70.910-900, Brasília, DF, Brazil
| | - Sonia N Bao
- University of Brasilia, Institute of Biological Sciences, Laboratório de Microscopia e Microanálise. 70910-900, Brasília, DF, Brazil
| | - Marcilio Cunha-Filho
- University of Brasilia, School of Health Sciences, Laboratory of Food, Drugs and Cosmetics (LTMAC), 70.910-900, Brasília, DF, Brazil.
| |
Collapse
|
12
|
Andrews GP, Qian K, Jacobs E, Jones DS, Tian Y. High drug loading nanosized amorphous solid dispersion (NASD) with enhanced in vitro solubility and permeability: Benchmarking conventional ASD. Int J Pharm 2023; 632:122551. [PMID: 36581107 DOI: 10.1016/j.ijpharm.2022.122551] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/06/2022] [Accepted: 12/23/2022] [Indexed: 12/27/2022]
Abstract
Through liquid-liquid phase separation (LLPS), it is possible to generate drug-rich nanoparticles during the dissolution of conventional amorphous solid dispersions (ASDs). These self-generated nanoparticles may improve the oral absorption of poorly water-soluble drugs by enhancing the drug's apparent solubility and effective membrane permeability. However, due to the high concentration threshold required for LLPS, conventional ASDs that can consistently generate drug-rich nanoparticles during dissolution are rare. More importantly, the quality of these meta-stable drug-rich nanoparticles is hard to control during dissolution, leading to inconsistency in formulation performances. This work has described a continuous twin-screw extrusion process capable of producing nanosized ASD (NASD) formulations that can offer better solubility and permeability enhancements over conventional ASD formulations. Two polymeric carriers, polyvinylpyrrolidone-co-vinyl acetate (PVPVA) and hydroxypropyl methylcellulose acetate succinate (HPMCAS), with a model hydrophobic drug celecoxib (BCS II), were formulated into both ASD and NASD formulations. Compared to the conventional ASD formulation, the prefabricated NASD (sizes ranging between 40 and 200 nm) embedded within a polyol matrix can be rapidly dispersed into a nanoparticle suspension in the presence of aqueous media. The resulting NASDs achieved drug loadings up to 80 % w/w and a maximum of 98 % encapsulation efficiency. Because of the TSE platform's high drug-loading capacity and high scalability, the developed method may be useful for continuously producing personalized nanomedicines.
Collapse
Affiliation(s)
- Gavin P Andrews
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - Kaijie Qian
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - Esther Jacobs
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - David S Jones
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom
| | - Yiwei Tian
- School of Pharmacy, Queen's University Belfast, BT9 7BL, United Kingdom.
| |
Collapse
|
13
|
Enhancement of itraconazole solubility and release by hot-melt extrusion with Soluplus®. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
14
|
Zhao P, Han W, Shu Y, Li M, Sun Y, Sui X, Liu B, Tian B, Liu Y, Fu Q. Liquid-liquid phase separation drug aggregate: Merit for oral delivery of amorphous solid dispersions. J Control Release 2023; 353:42-50. [PMID: 36414193 DOI: 10.1016/j.jconrel.2022.11.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/23/2022]
Abstract
As a promising strategy, amorphous solid dispersion has been extensively employed in improving the oral bioavailability of insoluble drugs. Despite the numerous advantages, the problems associated with supersaturation stability limit its further application. Recently, the formation and stability of the liquid-liquid phase separation drug aggregate (LLPS-DA) have been found to be vital for supersaturation maintenance. An in-depth review of LLPS-DA was required to further explore the supersaturation maintenance mechanism in vivo. Hence, this study aimed to present a short review to introduce the LLPS-DA, highlight the in vivo advantages for oral administration, and discuss the prospects to help understand the in vivo behavior of LLPS-DA.
Collapse
Affiliation(s)
- Peixu Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Wen Han
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Yecheng Shu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Mo Li
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Yichi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Xiaofan Sui
- Liaoning Institute for Drug Control, No. 7 Chongshan West Road, Shenyang 110016, China
| | - Bingyang Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Baocheng Tian
- School of Pharmacy, Binzhou Medical University, No. 346, Guanhai Road, Yantai 264003, China
| | - Yanhua Liu
- Department of Pharmaceutics, Ningxia Medical University, 1160 Shengli Street, Yinchuan 750004, China
| | - Qiang Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| |
Collapse
|
15
|
Verma V, Patel P, Ryan KM, Hudson S, Padrela L. Production of hydrochlorothiazide nanoparticles with increased permeability using top-spray coating process. J Supercrit Fluids 2023. [DOI: 10.1016/j.supflu.2022.105788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
16
|
Kutumova EO, Akberdin IR, Kiselev IN, Sharipov RN, Egorova VS, Syrocheva AO, Parodi A, Zamyatnin AA, Kolpakov FA. Physiologically Based Pharmacokinetic Modeling of Nanoparticle Biodistribution: A Review of Existing Models, Simulation Software, and Data Analysis Tools. Int J Mol Sci 2022; 23:12560. [PMID: 36293410 PMCID: PMC9604366 DOI: 10.3390/ijms232012560] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer treatment and pharmaceutical development require targeted treatment and less toxic therapeutic intervention to achieve real progress against this disease. In this scenario, nanomedicine emerged as a reliable tool to improve drug pharmacokinetics and to translate to the clinical biologics based on large molecules. However, the ability of our body to recognize foreign objects together with carrier transport heterogeneity derived from the combination of particle physical and chemical properties, payload and surface modification, make the designing of effective carriers very difficult. In this scenario, physiologically based pharmacokinetic modeling can help to design the particles and eventually predict their ability to reach the target and treat the tumor. This effort is performed by scientists with specific expertise and skills and familiarity with artificial intelligence tools such as advanced software that are not usually in the "cords" of traditional medical or material researchers. The goal of this review was to highlight the advantages that computational modeling could provide to nanomedicine and bring together scientists with different background by portraying in the most simple way the work of computational developers through the description of the tools that they use to predict nanoparticle transport and tumor targeting in our body.
Collapse
Affiliation(s)
- Elena O. Kutumova
- Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, 354340 Sochi, Russia
- Federal Research Center for Information and Computational Technologies, 630090 Novosibirsk, Russia
- BIOSOFT.RU, Ltd., 630058 Novosibirsk, Russia
| | - Ilya R. Akberdin
- Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, 354340 Sochi, Russia
- BIOSOFT.RU, Ltd., 630058 Novosibirsk, Russia
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Ilya N. Kiselev
- Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, 354340 Sochi, Russia
- Federal Research Center for Information and Computational Technologies, 630090 Novosibirsk, Russia
- BIOSOFT.RU, Ltd., 630058 Novosibirsk, Russia
| | - Ruslan N. Sharipov
- Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, 354340 Sochi, Russia
- Federal Research Center for Information and Computational Technologies, 630090 Novosibirsk, Russia
- BIOSOFT.RU, Ltd., 630058 Novosibirsk, Russia
- Specialized Educational Scientific Center, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Vera S. Egorova
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anastasiia O. Syrocheva
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Alessandro Parodi
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Andrey A. Zamyatnin
- Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340 Sochi, Russia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Fedor A. Kolpakov
- Scientific Center for Information Technologies and Artificial Intelligence, Sirius University of Science and Technology, 354340 Sochi, Russia
- Federal Research Center for Information and Computational Technologies, 630090 Novosibirsk, Russia
- BIOSOFT.RU, Ltd., 630058 Novosibirsk, Russia
| |
Collapse
|
17
|
Abstract
![]()
Formulations containing nanosized drug particles such
as nanocrystals
and nanosized amorphous drug aggregates recently came into light as
promising strategies to improve the bioavailability of poorly soluble
drugs. However, the increased solubility due to the reduction in particle
size cannot adequately explain the enhanced bioavailability. In this
study, the mechanisms and extent of enhanced passive permeation by
drug particles were investigated using atazanavir, lopinavir, and
clotrimazole as model drugs. Franz diffusion cells with lipid-infused
membranes were utilized to evaluate transmembrane flux. The impact
of stirring rate, receiver buffer condition, and particle size was
investigated, and mass transport analyses were conducted to calculate
transmembrane flux. Flux enhancement by particles was found to be
dependent on particle size as well as the partitioning behavior of
the drug between the receiver solution and the membrane, which is
determined by both the drug and buffer used. A flux plateau was observed
at high particle concentrations above amorphous solubility, confirming
that mass transfer of amorphous drug particles from the aqueous solution
to the membrane occurs only through the molecularly dissolved drug.
Mass transport models were used to calculate flux enhancement by particles
for various drugs at different conditions. Good agreements were obtained
between experimental and predicted values. These results should contribute
to improved bioavailability prediction of nanosized drug particles
and better design of formulations containing colloidal drug particles.
Collapse
Affiliation(s)
- Akshay Narula
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road Unit 3092, Storrs, Connecticut 06269, United States
| | - Rayan Sabra
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road Unit 3092, Storrs, Connecticut 06269, United States
| | - Na Li
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road Unit 3092, Storrs, Connecticut 06269, United States.,Institute of Materials Science, University of Connecticut, 97 North Eagleville Road Unit 3136, Storrs, Connecticut 06269, United States
| |
Collapse
|
18
|
Sabra R, Narula A, Taylor LS, Li N. Comparisons of in Vitro Models to Evaluate the Membrane Permeability of Amorphous Drug Nanoparticles. Mol Pharm 2022; 19:3412-3428. [PMID: 35972995 DOI: 10.1021/acs.molpharmaceut.2c00565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The spontaneous formation of amorphous drug nanoparticles following the release of a drug from a supersaturating formulation is gaining increasing attention due to their potential contribution to increased oral bioavailability. The formation of nanosized drug particles also has considerable implications for the interpretation of in vitro and in vivo data. However, the membrane transport properties of these drug particles remain less well understood. Herein, the membrane permeation of nanosized amorphous drug particles of a model drug atazanavir was evaluated using different artificial membrane-based, cell-based, and animal tissue-based models. Results showed that flux enhancement by particles was different for the various systems used. Generally, good agreement was obtained among experiments performed using the same apparatus with different model membranes, with the exception of the Madin-Darby canine kidney cell monolayer and the Long-Evans rat intestine tissue, which showed lower flux enhancements. Franz cell-based models showed slightly higher flux enhancements by particles compared to Transwell and intestinal tissue sac models. Mass transport analysis suggested that the extent of flux enhancement by particles is dependent on the geometry of the apparatus as well as the properties of the membrane and buffer used, whereas the flux plateau concentration is dependent on the unstirred water later (UWL) asymmetry. These results highlight the complexity in characterizing the permeability advantage of these nonmembrane permeable drug particles and suggest that caution should be used in selecting the appropriate in vitro model to evaluate the overall permeability of colloidal drug particles.
Collapse
Affiliation(s)
- Rayan Sabra
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States
| | - Akshay Narula
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Na Li
- Department of Pharmaceutical Sciences, University of Connecticut, 69 North Eagleville Road, Unit 3092, Storrs, Connecticut 06269, United States.,Institute of Materials Science, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, Connecticut 06269, United States
| |
Collapse
|
19
|
Ciprofloxacin-Loaded Zein/Hyaluronic Acid Nanoparticles for Ocular Mucosa Delivery. Pharmaceutics 2022; 14:pharmaceutics14081557. [PMID: 35893813 PMCID: PMC9332751 DOI: 10.3390/pharmaceutics14081557] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 07/24/2022] [Indexed: 01/03/2023] Open
Abstract
Bacterial conjunctivitis is a worldwide problem that, if untreated, can lead to severe complications, such as visual impairment and blindness. Topical administration of ciprofloxacin is one of the most common treatments for this infection; however, topical therapeutic delivery to the eye is quite challenging. To tackle this, nanomedicine presents several advantages compared to conventional ophthalmic dosage forms. Herein, the flash nanoprecipitation technique was applied to produce zein and hyaluronic acid nanoparticles loaded with ciprofloxacin (ZeinCPX_HA NPs). ZeinCPX_HA NPs exhibited a hydrodynamic diameter of <200 nm and polydispersity index of <0.3, suitable for ocular drug delivery. In addition, the freeze-drying of the nanoparticles was achieved by using mannitol as a cryoprotectant, allowing their resuspension in water without modifying the physicochemical properties. Moreover, the biocompatibility of nanoparticles was confirmed by in vitro assays. Furthermore, a high encapsulation efficiency was achieved, and a release profile with an initial burst was followed by a prolonged release of ciprofloxacin up to 24 h. Overall, the obtained results suggest ZeinCPX_HA NPs as an alternative to the common topical dosage forms available on the market to treat conjunctivitis.
Collapse
|
20
|
Martinez MN, Wu F, Sinko B, Brayden DJ, Grass M, Kesisoglou F, Stewart A, Sugano K. A Critical Overview of the Biological Effects of Excipients (Part II): Scientific Considerations and Tools for Oral Product Development. AAPS J 2022; 24:61. [DOI: 10.1208/s12248-022-00713-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 04/16/2022] [Indexed: 11/30/2022] Open
|
21
|
Lu X, Li M, Arce FA, Ling J, Setiawan N, Wang Y, Shi X, Campbell HR, Nethercott MJ, Xu W, Munson EJ, Marsac PJ, Su Y. Mechanistic Investigation of Drug Supersaturation in the Presence of Polysorbates as Solubilizing Additives by Solution Nuclear Magnetic Resonance Spectroscopy. Mol Pharm 2021; 18:4310-4321. [PMID: 34761934 DOI: 10.1021/acs.molpharmaceut.1c00477] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The introduction of solubilizing additives has historically been an attractive approach to address the ever-growing proportion of poorly water-soluble drug (PWSD) compounds within the modern drug discovery pipeline. Lipid-formulations, and more specifically micelle formulations, have garnered particular interest because of their simplicity, size, scalability, and avoidance of solid-state limitations. Although micelle formulations have been widely utilized, the molecular mechanism of drug solubilization in surfactant micelles is still poorly understood. In this study, a series of modern nuclear magnetic resonance (NMR) methods are utilized to gain a molecular-level understanding of intermolecular interactions and kinetics in a model system. This approach enabled the understanding of how a PWSD, 17β-Estradiol (E2), solubilizes within a nonionic micelle system composed of polysorbate 80 (PS80). Based on one-dimensional (1D) 1H chemical shift differences of E2 in PS80 solutions, as well as intermolecular correlations established from 1D selective nuclear Overhauser effect (NOE) and two-dimensional NOE spectroscopy experiments, E2 was found to accumulate within the palisade layer of PS80 micelles. A potential hydrogen-bonding interaction between a hydroxyl group of E2 and a carbonyl group of PS80 alkane chains may allow for stabilizing E2-PS80 mixed micelles. Diffusion and relaxation NMR analysis and particle size measurements using dynamic light scattering indicate a slight increase in the micellar size with increasing degrees of supersaturation, resulting in slower mobility of the drug molecule. Based on these structural findings, a theoretical orientation model of E2 molecules with PS80 molecules was developed and validated by computational docking simulations.
Collapse
Affiliation(s)
- Xingyu Lu
- Analytical Research and Development, Merck & Co., Rahway, New Jersey 07065, United States.,Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Mingyue Li
- Analytical Research and Development, Merck & Co., Rahway, New Jersey 07065, United States
| | - Freddy A Arce
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Jing Ling
- Pharmaceutical Sciences, Merck & Co., South San Francisco, California 94080, United States
| | - Nico Setiawan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Yaqiang Wang
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California 90095 United States
| | - Xiaohuo Shi
- Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Heather R Campbell
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | | | - Wei Xu
- Analytical Research and Development, Merck & Co., Rahway, New Jersey 07065, United States
| | - Eric J Munson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Patrick J Marsac
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Yongchao Su
- Analytical Research and Development, Merck & Co., Rahway, New Jersey 07065, United States.,Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States.,Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
22
|
Gupta R, Chen Y, Xie H. In vitro dissolution considerations associated with nano drug delivery systems. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1732. [PMID: 34132050 PMCID: PMC8526385 DOI: 10.1002/wnan.1732] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022]
Abstract
Nano drug delivery systems (NDDS) offer promising solution for the translation of future nanomedicines. As bioavailability and therapeutic outcomes can be improved by altering the drug release from these NDDS, it becomes essential to thoroughly understand their drug release kinetics. Moreover, U.S. Food and Drug Administration requires critical evaluation of potential safety, efficacy, and public health impacts of nanomaterials. Spiraling up market share of NDDS has also stimulated the pharmaceutical industry to develop their cost-effective generic versions after the expiry of patent and associated exclusivity. However, unlike the conventional dosage forms, the in vivo disposition of NDDS is highly intricate and different from their in vitro behavior. Significant challenges exist in the establishment of in vitro-in vivo correlation (IVIVC) due to incomplete understanding of nanoparticles' in vivo biofate and its impact on in vitro experimental protocols. A rational design of dissolution may serve as quality and quantity control tool and help develop a meaningful IVIVC for favorable economic implications. Clinically relevant drug product specifications (critical quality attributes) can be identified by establishing a link between in vitro performance and in vivo exposure. In vitro dissolution may also play a pivotal role to understand the dissolution-mediated clearance and safety of NDDS. Prevalent in vitro dissolution methods for NDDS and their limitations are discussed in this review, among which USP 4 is gaining more interest recently. Researchers are working diligently to develop biorelevant in vitro release assays to ensure optimal therapeutic performance of generic versions of these NDDS. This article focuses on these studies and presents important considerations for the future development of clinically relevant in vitro release methods. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Ritu Gupta
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA 77004
| | - Yuan Chen
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA 77004
| | - Huan Xie
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX, USA 77004
| |
Collapse
|
23
|
El Sayed M, Alhalaweh A, Bergström CAS. Impact of Simulated Intestinal Fluids on Dissolution, Solution Chemistry, and Membrane Transport of Amorphous Multidrug Formulations. Mol Pharm 2021; 18:4079-4089. [PMID: 34613730 PMCID: PMC8564758 DOI: 10.1021/acs.molpharmaceut.1c00480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
The solution behavior
and membrane transport of multidrug formulations
were herein investigated in a biorelevant medium simulating fasted
conditions. Amorphous multidrug formulations were prepared by the
solvent evaporation method. Combinations of atazanavir (ATV) and ritonavir
(RTV) and felodipine (FDN) and indapamide (IPM) were prepared and
stabilized by a polymer for studying their dissolution (under non-sink
conditions) and membrane transport in fasted state simulated intestinal
fluid (FaSSIF). The micellar solubilization by FaSSIF enhanced the
amorphous solubility of the drugs to different extents. Similar to
buffer, the maximum achievable concentration of drugs in combination
was reduced in FaSSIF, but the extent of reduction was affected by
the degree of FaSSIF solubilization. Dissolution studies of ATV and
IPM revealed that the amorphous solubility of these two drugs was
not affected by FaSSIF solubilization. In contrast, RTV was significantly
affected by FaSSIF solubilization with a 30% reduction in the maximum
achievable concentration upon combination to ATV, compared to 50%
reduction in buffer. This positive deviation by FaSSIF solubilization
was not reflected in the mass transport–time profiles. Interestingly,
FDN concentrations remain constant until the amount of IPM added was
over 1000 μg/mL. No decrease in the membrane transport of FDN
was observed for a 1:1 M ratio of FDN-IPM combination. This study
demonstrates the importance of studying amorphous multidrug formulations
under physiologically relevant conditions to obtain insights into
the performance of these formulations after oral administration.
Collapse
Affiliation(s)
- Mira El Sayed
- Department of Pharmacy, Biomedical Centre, Uppsala University, P.O. Box 580, Uppsala SE-751 23, Sweden.,Recipharm OT Chemistry AB, Uppsala SE-754 50, Sweden
| | | | - Christel A S Bergström
- Department of Pharmacy, Biomedical Centre, Uppsala University, P.O. Box 580, Uppsala SE-751 23, Sweden
| |
Collapse
|
24
|
Parodi A, Buzaeva P, Nigovora D, Baldin A, Kostyushev D, Chulanov V, Savvateeva LV, Zamyatnin AA. Nanomedicine for increasing the oral bioavailability of cancer treatments. J Nanobiotechnology 2021; 19:354. [PMID: 34717658 PMCID: PMC8557561 DOI: 10.1186/s12951-021-01100-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 10/21/2021] [Indexed: 12/28/2022] Open
Abstract
Abstract Oral administration is an appealing route of delivering cancer treatments. However, the gastrointestinal tract is characterized by specific and efficient physical, chemical, and biological barriers that decrease the bioavailability of medications, including chemotherapeutics. In recent decades, the fields of material science and nanomedicine have generated several delivery platforms with high potential for overcoming multiple barriers associated to oral administration. This review describes the properties of several nanodelivery systems that improve the bioavailability of orally administered therapeutics, highlighting their advantages and disadvantages in generating successful anticancer oral nanomedicines. Graphical Abstract ![]()
Collapse
Affiliation(s)
- Alessandro Parodi
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia. .,Sirius University of Science and Technology, 1 Olympic Ave, 354340, Sochi, Russia.
| | - Polina Buzaeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Daria Nigovora
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Alexey Baldin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russia
| | - Dmitry Kostyushev
- Sirius University of Science and Technology, 1 Olympic Ave, 354340, Sochi, Russia.,National Medical Research Center of Tuberculosis and Infectious Diseases, Ministry of Health, 127994, Moscow, Russia
| | - Vladimir Chulanov
- Sirius University of Science and Technology, 1 Olympic Ave, 354340, Sochi, Russia.,National Medical Research Center of Tuberculosis and Infectious Diseases, Ministry of Health, 127994, Moscow, Russia.,Department of Infectious Diseases, Sechenov University, 119991, Moscow, Russia
| | - Lyudmila V Savvateeva
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991, Moscow, Russia. .,Sirius University of Science and Technology, 1 Olympic Ave, 354340, Sochi, Russia. .,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russia. .,Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7X, UK.
| |
Collapse
|
25
|
Ramachandran G, Sudheesh MS. Role of Permeability on the Biopredictive Dissolution of Amorphous Solid Dispersions. AAPS PharmSciTech 2021; 22:243. [PMID: 34595565 DOI: 10.1208/s12249-021-02125-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 08/23/2021] [Indexed: 02/08/2023] Open
Abstract
An ideal dissolution test for amorphous solid dispersions (ASDs) should reflect physicochemical, physiological, and hydrodynamic conditions which accurately represent in vivo dissolution. However, this is confounded by the evolution of different molecular and colloidal species during dissolution, generating a supersaturated state of the drug. The supersaturated state of a drug is thermodynamically unstable which drives the process of precipitation resulting in a loss of solubility advantage. Maintaining a supersaturated state of the drug with the help of precipitation inhibiting excipients is a key component in the design of ASDs. Therefore, a biopredictive dissolution test is critical for proper risk assessment during the development of an optimal ASD formulation. One of the overlooked components of biopredictive dissolution is the role of drug permeability. The kinetic changes in the phase behavior of a drug during dissolution of ASDs are influenced by drug permeability across a membrane. Conventionally, drug dissolution and permeation are analyzed separately although they occur simultaneously in vivo. The kinetic phase changes occurring during dissolution of ASDs can influence the thermodynamic activity and membrane flux of a drug. The present review evaluates the feasibility, predictability, and practicability of permeability/dissolution for the optimal development and risk assessment of ASD formulations.
Collapse
|
26
|
In Vitro-In Silico Tools for Streamlined Development of Acalabrutinib Amorphous Solid Dispersion Tablets. Pharmaceutics 2021; 13:pharmaceutics13081257. [PMID: 34452217 PMCID: PMC8398755 DOI: 10.3390/pharmaceutics13081257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 11/17/2022] Open
Abstract
Amorphous solid dispersion (ASD) dosage forms can improve the oral bioavailability of poorly water-soluble drugs, enabling the commercialization of new chemical entities and improving the efficacy and patient compliance of existing drugs. However, the development of robust, high-performing ASD dosage forms can be challenging, often requiring multiple formulation iterations, long timelines, and high cost. In a previous study, acalabrutinib/hydroxypropyl methylcellulose acetate succinate (HPMCAS)-H grade ASD tablets were shown to overcome the pH effect of commercially marketed Calquence in beagle dogs. This study describes the streamlined in vitro and in silico approach used to develop those ASD tablets. HPMCAS-H and -M grade polymers provided the longest acalabrutinib supersaturation sustainment in an initial screening study, and HPMCAS-H grade ASDs provided the highest in vitro area under the curve (AUC) in gastric to intestinal transfer dissolution tests at elevated gastric pH. In silico simulations of the HPMCAS-H ASD tablet and Calquence capsule provided good in vivo study prediction accuracy using a bottom–up approach (absolute average fold error of AUC0-inf < 2). This streamlined approach combined an understanding of key drug, polymer, and gastrointestinal properties with in vitro and in silico tools to overcome the acalabrutinib pH effect without the need for reformulation or multiple studies, showing promise for reducing time and costs to develop ASD drug products.
Collapse
|
27
|
Yen CW, Kuhn R, Hu C, Zhang W, Chiang PC, Chen JZ, Hau J, Estevez A, Nagapudi K, Leung DH. Impact of surfactant selection and incorporation on in situ nanoparticle formation from amorphous solid dispersions. Int J Pharm 2021; 607:120980. [PMID: 34371147 DOI: 10.1016/j.ijpharm.2021.120980] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/15/2021] [Accepted: 08/04/2021] [Indexed: 01/26/2023]
Abstract
Spray dried amorphous solid dispersions (ASDs) stand as one of the most effective formulation strategies to address issues of low aqueous solubility when developing new chemical entities.An emerging research topic focusing on the formation of amorphous nanoparticles or nanodroplets from ASD formulations has attracted attention recently. These ASD nanoparticlescan be highly beneficial and able to further increase oral bioavailability. The incorporation of surfactants in ASD formulations has been shown to facilitate the formation of these nanoparticles. Therefore, understanding the mechanism of surfactant-promoted nanoparticle formation becomes critical for the rational design of ASD formulations. This work demonstrated the importance of inclusion of the surfactant within the ASD composition for nanoparticle formation. In contrast, when a surfactant is added externally (e.g., by inclusion in the dosing vehicle), only a limited degree of nanoparticle formation was observed even at the optimized surfactant-to-drug ratios. A variety of different surfactants were also assessed for understanding their impact on ASD nanoparticle formation. The spray drying systems containing nonionic surfactants, Tween 80 and Vitamin E TPGS, produced higher amounts of in situ ASD nanoparticles when compared to an anionic surfactant, sodium lauryl sulfate (SLS). The ASD nanoparticles produced by the Genentech developmental compound, GDC-0334, were highly stable and retained their original particle size and amorphous feature for at least 18 h under biorelevant conditions. The high degree of nanoparticle formation from spray dried GDC-0334 containing Tween 80 combined with the superior physical stability of the nanoparticles also translated to enhanced in vivo performance in a rat pharmacokinetics study.
Collapse
Affiliation(s)
- Chun-Wan Yen
- Small Molecule Pharmaceutical Sciences, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Robert Kuhn
- Small Molecule Pharmaceutical Sciences, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Chloe Hu
- Small Molecule Pharmaceutical Sciences, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wei Zhang
- Small Molecule Pharmaceutical Sciences, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Po-Chang Chiang
- Small Molecule Pharmaceutical Sciences, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jacob Z Chen
- Drug Metabolism and Pharmacokinetics, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jonathan Hau
- Small Molecule Pharmaceutical Sciences, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Alberto Estevez
- Structural Biology, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Karthik Nagapudi
- Small Molecule Pharmaceutical Sciences, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Dennis H Leung
- Small Molecule Pharmaceutical Sciences, 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
28
|
Thakore SD, Sirvi A, Joshi VC, Panigrahi SS, Manna A, Singh R, Sangamwar AT, Bansal AK. Biorelevant dissolution testing and physiologically based absorption modeling to predict in vivo performance of supersaturating drug delivery systems. Int J Pharm 2021; 607:120958. [PMID: 34332060 DOI: 10.1016/j.ijpharm.2021.120958] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 07/08/2021] [Accepted: 07/26/2021] [Indexed: 11/29/2022]
Abstract
Supersaturating drug delivery systems (SDDS) enhance the oral absorption of poorly water-soluble drugs by achieving a supersaturated state in the gastrointestinal tract. The maintenance of a supersaturated state is decided by the complex interplay among inherent properties of drug, excipients and physiological conditions of gastrointestinal tract. The biopharmaceutical advantage through SDDS can be mechanistically investigated by coupling biopredictive dissolution testing with physiologically based absorption modeling (PBAM). However, the development of biopredictive dissolution methods possess challenges due to concurrent dissolution, supersaturation, precipitation, and possible redissolution of precipitates during gastrointestinal transit of SDDS. In this comprehensive review, our effort is to critically assess the current state-of-knowledge and provide future directions for PBAM of SDDS. The review outlines various methods used to retrieve physiologically relevant values for input parameters like solubility, dissolution, precipitation, lipid-digestion and permeability of SDDS. SDDS-specific parameterization includes solubility values corresponding to apparent physical form, dissolution in physiologically relevant volumes with biorelevant media, and transfer experiments to incorporate precipitation kinetics. Interestingly, the lack of experimental permeability values and modification of absorption flux through SDDS possess the additional challenge for its PBAM. Supersaturation triggered permeability modifications are reported to fit the observed plasma concentration-time profile. Hence, the experimental insights on good fitting with modified permeability can be potential area of future research for the development of in vitro methods to reliably predict oral absorption of SDDS.
Collapse
Affiliation(s)
- Samarth D Thakore
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Arvind Sirvi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Vikram C Joshi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Sanjali S Panigrahi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Arijita Manna
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Ridhima Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Abhay T Sangamwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India
| | - Arvind K Bansal
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Mohali, Punjab 160062, India.
| |
Collapse
|
29
|
Sigfridsson K, Andreasson T, Fihn BM, Kearns M, Lindblom S. Supersaturated formulations of poorly soluble weak acid drugs evaluated in rodents; a case study. Int J Pharm 2021; 606:120883. [PMID: 34271156 DOI: 10.1016/j.ijpharm.2021.120883] [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: 04/01/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 10/20/2022]
Abstract
In the present study we describe a way of working to overcome oral administration challenges in an early preclinical project. As candidate drugs were obtained, the preclinical delivery route was replaced by the intended route of the product and resources were allocated to optimize the oral absorption. Two main approaches were followed in order to formulate a selected weak acid, AZ'403, for oral administration in large scale toxicological studies and the early clinical phases. Both approaches relies on the suppression of precipitation from obtained supersaturated solutions achieved either by amorphous solid dispersions (using hydroxypropyl methylcellulose acetate succinate, HPMC-AS) or crystalline salts (sodium and potassium salts). In vivo studies in rodents were performed to evaluate oral AZ'403 absorption from amorphous and crystalline formulations, using nano- and micro crystalline particles of the neutral form, as references. The oral absorption of AZ'403 formulated using both approaches was significantly higher compared with the references. The improvements in overall exposures were 7-100 times during the investigated conditions. The pharmacokinetic profiles implied that both solid dispersions and crystalline salts of AZ'403 generated supersaturation in the small intestine in rodents and indicated that both approaches may be ways forward for subsequent late stage product development.
Collapse
Affiliation(s)
- Kalle Sigfridsson
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden.
| | - Theresa Andreasson
- Bioscience, Research and Early Development, Respiratory & Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Britt-Marie Fihn
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Respiratory & Inflammation, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Martin Kearns
- Early Product Development and Manufacturing, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Sara Lindblom
- Early Product Development and Manufacturing, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
30
|
Qian K, Stella L, Jones DS, Andrews GP, Du H, Tian Y. Drug-Rich Phases Induced by Amorphous Solid Dispersion: Arbitrary or Intentional Goal in Oral Drug Delivery? Pharmaceutics 2021; 13:889. [PMID: 34203969 PMCID: PMC8232734 DOI: 10.3390/pharmaceutics13060889] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022] Open
Abstract
Among many methods to mitigate the solubility limitations of drug compounds, amorphous solid dispersion (ASD) is considered to be one of the most promising strategies to enhance the dissolution and bioavailability of poorly water-soluble drugs. The enhancement of ASD in the oral absorption of drugs has been mainly attributed to the high apparent drug solubility during the dissolution. In the last decade, with the implementations of new knowledge and advanced analytical techniques, a drug-rich transient metastable phase was frequently highlighted within the supersaturation stage of the ASD dissolution. The extended drug absorption and bioavailability enhancement may be attributed to the metastability of such drug-rich phases. In this paper, we have reviewed (i) the possible theory behind the formation and stabilization of such metastable drug-rich phases, with a focus on non-classical nucleation; (ii) the additional benefits of the ASD-induced drug-rich phases for bioavailability enhancements. It is envisaged that a greater understanding of the non-classical nucleation theory and its application on the ASD design might accelerate the drug product development process in the future.
Collapse
Affiliation(s)
- Kaijie Qian
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (K.Q.); (D.S.J.); (G.P.A.)
| | - Lorenzo Stella
- Atomistic Simulation Centre, School of Mathematics and Physics, Queen’s University Belfast, 7–9 College Park E, Belfast BT7 1PS, UK;
- David Keir Building, School of Chemistry and Chemical Engineering, Queen’s University Belfast, Stranmillis Road, Belfast BT9 5AG, UK
| | - David S. Jones
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (K.Q.); (D.S.J.); (G.P.A.)
| | - Gavin P. Andrews
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (K.Q.); (D.S.J.); (G.P.A.)
- School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Huachuan Du
- Laboratory of Applied Mechanobiology, Department of Health Sciences and Technology, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
- Simpson Querrey Institute, Northwestern University, 303 East Superior Street, 11th floor, Chicago, IL 60611, USA
| | - Yiwei Tian
- Pharmaceutical Engineering Group, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (K.Q.); (D.S.J.); (G.P.A.)
| |
Collapse
|
31
|
Vinarov Z, Abrahamsson B, Artursson P, Batchelor H, Berben P, Bernkop-Schnürch A, Butler J, Ceulemans J, Davies N, Dupont D, Flaten GE, Fotaki N, Griffin BT, Jannin V, Keemink J, Kesisoglou F, Koziolek M, Kuentz M, Mackie A, Meléndez-Martínez AJ, McAllister M, Müllertz A, O'Driscoll CM, Parrott N, Paszkowska J, Pavek P, Porter CJH, Reppas C, Stillhart C, Sugano K, Toader E, Valentová K, Vertzoni M, De Wildt SN, Wilson CG, Augustijns P. Current challenges and future perspectives in oral absorption research: An opinion of the UNGAP network. Adv Drug Deliv Rev 2021; 171:289-331. [PMID: 33610694 DOI: 10.1016/j.addr.2021.02.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/12/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Although oral drug delivery is the preferred administration route and has been used for centuries, modern drug discovery and development pipelines challenge conventional formulation approaches and highlight the insufficient mechanistic understanding of processes critical to oral drug absorption. This review presents the opinion of UNGAP scientists on four key themes across the oral absorption landscape: (1) specific patient populations, (2) regional differences in the gastrointestinal tract, (3) advanced formulations and (4) food-drug interactions. The differences of oral absorption in pediatric and geriatric populations, the specific issues in colonic absorption, the formulation approaches for poorly water-soluble (small molecules) and poorly permeable (peptides, RNA etc.) drugs, as well as the vast realm of food effects, are some of the topics discussed in detail. The identified controversies and gaps in the current understanding of gastrointestinal absorption-related processes are used to create a roadmap for the future of oral drug absorption research.
Collapse
Affiliation(s)
- Zahari Vinarov
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium; Department of Chemical and Pharmaceutical Engineering, Sofia University, Sofia, Bulgaria
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, Gothenburg, Sweden
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Hannah Batchelor
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Philippe Berben
- Pharmaceutical Development, UCB Pharma SA, Braine- l'Alleud, Belgium
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innsbruck, Austria
| | - James Butler
- GlaxoSmithKline Research and Development, Ware, United Kingdom
| | | | - Nigel Davies
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Gøril Eide Flaten
- Department of Pharmacy, UiT The Arctic University of Norway, Tromsø, Norway
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, University of Bath, Bath, United Kingdom
| | | | | | | | | | | | - Martin Kuentz
- Institute for Pharma Technology, University of Applied Sciences and Arts Northwestern Switzerland, Basel, Switzerland
| | - Alan Mackie
- School of Food Science & Nutrition, University of Leeds, Leeds, United Kingdom
| | | | | | - Anette Müllertz
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Petr Pavek
- Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
| | | | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Kiyohiko Sugano
- College of Pharmaceutical Sciences, Ritsumeikan University, Shiga, Japan
| | - Elena Toader
- Faculty of Medicine, University of Medicine and Pharmacy of Iasi, Romania
| | - Kateřina Valentová
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Saskia N De Wildt
- Department of Pharmacology and Toxicology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Clive G Wilson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
32
|
Ueda K, Taylor LS. Partitioning of surfactant into drug-rich nanodroplets and its impact on drug thermodynamic activity and droplet size. J Control Release 2021; 330:229-243. [DOI: 10.1016/j.jconrel.2020.12.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/16/2020] [Accepted: 12/13/2020] [Indexed: 02/08/2023]
|
33
|
Boullata JI. Enteral Medication for the Tube-Fed Patient: Making This Route Safe and Effective. Nutr Clin Pract 2020; 36:111-132. [PMID: 33373487 DOI: 10.1002/ncp.10615] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/15/2020] [Indexed: 12/26/2022] Open
Abstract
The administration of medication through an enteral access device requires important forethought. Meeting a patient's therapeutic needs requires achieving expected drug bioavailability without increasing the risk for toxicity, therapeutic failure, or feeding tube occlusion. Superimposing gut dysfunction, critical illness, or enteral nutrition-drug interaction further increases the need for a systematic approach to prescribing, evaluating, and preparing a drug for administration through an enteral access device. This review will explain the fundamental factors involved in drug bioavailability through the gut, address the influencing considerations for the enterally fed patient, and describe best practices for enteral drug preparation and administration.
Collapse
Affiliation(s)
- Joseph I Boullata
- Department of Clinical Nutrition Support Services, Penn Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
34
|
Arce FA, Setiawan N, Campbell HR, Lu X, Nethercott MJ, Bummer P, Su Y, Marsac PJ. Toward Developing Discriminating Dissolution Methods for Formulations Containing Nanoparticulates in Solution: The Impact of Particle Drift and Drug Activity in Solution. Mol Pharm 2020; 17:4125-4140. [PMID: 32965123 DOI: 10.1021/acs.molpharmaceut.0c00599] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Enabling formulations are an attractive approach to increase the dissolution rate, solubility, and oral bioavailability of poorly soluble compounds. With the growing prevalence of poorly soluble drug compounds in the pharmaceutical pipeline, supersaturating drug delivery systems (SDDS), a subset of enabling formulations, have grown in popularity due to their properties allowing for drug concentrations greater than the corresponding crystalline solubility. However, the extent of supersaturation generated as the enabling formulation traverses the gastrointestinal (GI) tract is dynamic and poorly understood. The dynamic nature of supersaturation is a result of several competing kinetic processes such as dissolution, solubilization by formulation and endogenous surfactants, crystallization, and absorption. Ultimately, the free drug concentration, which is equivalent to the drug's inherent thermodynamic activity amid these kinetic processes, defines the true driving force for drug absorption. However, in cases where solubilizing agents are present (i.e., surfactants and bile salts), drug molecules may associate with colloidal nanoscale species, complicating drug activity determination. These nanoscale species can drift into the aqueous boundary layer (ABL), increasing the local API activity at the membrane surface, resulting in increased bioavailability. Herein, a novel approach was developed to accurately measure thermodynamic drug activity in complex media containing drug distributed in nanoparticulate species. This approach captures the influence of the ABL on the observed flux and, ultimately, the predicted unbound drug concentration. The results demonstrate that this approach can help to (1) measure the true extent of local supersaturation in complex systems containing solubilizing excipients and (2) elucidate the mechanisms by which colloidal aggregates can modulate the drug activity in solution and potentially enhance the flux observed across a membrane. The utilization of these techniques may provide development scientists with a strategy to evaluate formulation sensitivity to nanospeciation and allow formulators to maximize the driving force for absorption in a complex environment, perhaps enabling the development of dissolution methods with greater discrimination and correlation to pre-clinical and clinical data sets.
Collapse
Affiliation(s)
- Freddy A Arce
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Nico Setiawan
- Albany Molecular Research Inc., West Lafayette, Indiana 47906, United States
| | - Heather R Campbell
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Xingyu Lu
- Pharmaceutical Sciences, Merck & Co., Kenilworth, New Jersey 07033, United States.,Key Laboratory of Precise Synthesis of Functional Molecules of Zhejiang Province, School of Science, Westlake University, Hangzhou, Zhejiang 310024, China
| | | | - Paul Bummer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| | - Yongchao Su
- Pharmaceutical Sciences, Merck & Co., Kenilworth, New Jersey 07033, United States.,Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, Texas 78712, United States.,Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Patrick J Marsac
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, United States
| |
Collapse
|
35
|
Ueda K, Hate SS, Taylor LS. Impact of Hypromellose Acetate Succinate Grade on Drug Amorphous Solubility and In Vitro Membrane Transport. J Pharm Sci 2020; 109:2464-2473. [DOI: 10.1016/j.xphs.2020.04.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/22/2022]
|
36
|
Ueda K, Taylor LS. Polymer Type Impacts Amorphous Solubility and Drug-Rich Phase Colloidal Stability: A Mechanistic Study Using Nuclear Magnetic Resonance Spectroscopy. Mol Pharm 2020; 17:1352-1362. [PMID: 32097023 DOI: 10.1021/acs.molpharmaceut.0c00061] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The polymer used in an amorphous solid dispersion (ASD) formulation plays a critical role in dosage form performance. Herein, drug-polymer interactions in aqueous solution were evaluated in order to better understand the dispersion stability of the colloidal drug-rich phase generated when the amorphous solubility is exceeded. The amorphous solubility (Sa,IBP) of ibuprofen (IBP) decreased when hypromellose (HPMC) or polyvinylpyrrolidone/vinyl acetate (PVP-VA) were present in solution. Solution nuclear magnetic resonance (NMR) spectroscopy revealed that a large amount of HPMC and PVP-VA distributed into the IBP-rich phase. The mixing of HPMC and PVP-VA with the IBP-rich phase led to the decreased Sa,IBP. In contrast, the charged amino methacrylate copolymer (Eudragit E PO, EUD-E) showed minimal mixing with the IBP-rich phase; however, this polymer did lead to solubilization of IBP in the bulk aqueous phase. Although the IBP-rich phase generated by dissolving IBP at concentrations above Sa,IBP rapidly coarsened followed by creaming in the absence of polymer, all of the polymers stabilized the IBP dispersion to some extent. The droplet size of the IBP-rich phase immediately after formation was around 300 nm in HPMC and PVP-VA solutions, and around 800 nm in the EUD-E solution. The mixing of the former two polymers with the drug-rich phase is thought to account for the smaller droplet size. Despite a smaller initial size, the dispersion stability of the IBP-rich droplets was relatively poor in the presence of PVP-VA. In contrast, the coalescence of the IBP-rich droplets was effectively suppressed by the steric repulsion and electrostatic repulsion derived from adsorbed HPMC and EUD-E, respectively. The present study highlights the complex effects of a polymer on the drug amorphous solubility and colloidal stability, which should be considered when optimizing ASD formulations with the goal of maximizing drug absorption.
Collapse
Affiliation(s)
- Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan.,Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|