1
|
Garrett M, Curry S, Feris S, Lu Y, Gu Q, Clark A, Martin SF, Kastellorizios M. Delivery of a novel neuroprotective compound to the retina in rat and rabbit animal models. J Control Release 2025; 382:113659. [PMID: 40139393 DOI: 10.1016/j.jconrel.2025.113659] [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: 01/16/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Posterior segment-related diseases are among the leading causes of irreversible blindness and loss of vision globally. These diseases are extremely difficult to treat due to the drug delivery barriers posed by the eye, among other challenges. One delivery method that bypasses many of these obstacles, albeit not without risk, is ocular injections, and long-acting formulations such as implants can improve patient compliance by allowing for longer intervals between injections. Here, we report our development of a preclinical in situ-forming implant dosage form that provides sustained release of a novel compound, DKR-1677, with a target in the retina. An in situ-forming implant based on polylactic co glycolic acid (PLGA) was chosen in this preclinical stage because it is readily translatable to a preformed implant product. The formulations were tested in vitro, in rat and rabbit animal models for drug release and pharmacokinetics. A two-step in vitro dissolution method with implant formation in a biorelevant gel followed by incubation in release media showed a 30-day three-phase release profile with an initial burst release of 36.04 ± 4.23 %, a plateau, and a controlled release up to 93.75 ± 4.68 % at day 30, typical of PLGA-based implant formulations. Immediate and controlled-release formulations were tested in rat and rabbit animal models and confirmed that DKR-1677 is taken up by the retina after intravitreal administration. Furthermore, the in situ-forming implant was found to prolong drug presence in the retina to 30 days following a single administration, confirming that a PLGA-based implant is a viable approach for this drug candidate.
Collapse
Affiliation(s)
- Meredith Garrett
- Department of Pharmaceutical Sciences, University of North Texas System College of Pharmacy 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Stacy Curry
- North Texas Eye Research Institute, The University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Sherri Feris
- North Texas Eye Research Institute, The University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Yan Lu
- Department of Chemistry, College of Natural Sciences, The University of Texas at Austin, 105 E 24(th) St., Austin, TX 78712-1224, USA
| | - Qi Gu
- Department of Chemistry, College of Natural Sciences, The University of Texas at Austin, 105 E 24(th) St., Austin, TX 78712-1224, USA
| | - Abe Clark
- North Texas Eye Research Institute, The University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Stephen F Martin
- Department of Chemistry, College of Natural Sciences, The University of Texas at Austin, 105 E 24(th) St., Austin, TX 78712-1224, USA
| | - Michail Kastellorizios
- Department of Pharmaceutical Sciences, University of North Texas System College of Pharmacy 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA.
| |
Collapse
|
2
|
Yarlagadda DL, Kawakami K, Samavedi S. Leveraging Molecular Interactions to Develop a Generalized Design Framework for Coamorphous Drug-Drug Mixtures Exhibiting Elevated Glass Transition Temperatures. Mol Pharm 2025. [PMID: 40377977 DOI: 10.1021/acs.molpharmaceut.5c00006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Coamorphous mixtures (CAMs) prepared with two drugs have the potential to enhance the oral absorption of poorly soluble drugs and achieve combination therapy. From a practical standpoint, improving the glass transition temperature (Tg) of CAMs is desirable as it enhances stability and extends shelf life during storage. Toward the eventual goal of developing highly stable CAMs, this study establishes a generalized framework that systematically relates elevated Tg values of CAMs to intermolecular interactions based on specific functional groups. CAMs were prepared via quench-cooling using various combinations of indomethacin, ketoprofen, flurbiprofen, flufenamic acid, aripiprazole, bifonazole, and clotrimazole. CAMs prepared with drugs containing the COOH group exhibited significant positive deviations from the Tg values predicted by the Gordon-Taylor equation (i.e., ideal mixing behavior). COOH-associated hydrogen bonding was determined to be a key factor for Tg elevation, with synergistic contributions from π-π interactions and halogen bonding. In CAMs exhibiting the largest Tg deviations, contributions from ionic bonding were crucial, and were likely favored by differences in the pKa values of the constituent drugs. Continuity in Tg as a function of varying molar ratios indicated that stoichiometric pairing had a relatively minor contribution, while a decrease in the width of the glass transition suggested enhancement of molecular cooperativity as a possible mechanism for CAM stabilization. In contrast, non-COOH hydrogen bonding, π-π interactions, and halogen bonding on their own did not result in any meaningful Tg deviations from theoretical predictions. Systematic correlations between Tg deviations and molecular interactions reported in this study can lead to generalized design rules for the development of stable CAMs.
Collapse
Affiliation(s)
- Dani Lakshman Yarlagadda
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, IITH Main Road, Near NH 65, Kandi, Sangareddy, Telangana 502285, India
| | - Kohsaku Kawakami
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Satyavrata Samavedi
- Department of Chemical Engineering, Indian Institute of Technology Hyderabad, IITH Main Road, Near NH 65, Kandi, Sangareddy, Telangana 502285, India
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| |
Collapse
|
3
|
Dayo Owoyemi BC, Zeller M, Pereira da Silva B, Akinyemi AO, Ando RA, de Araujo GLB, Byrn SR. Drug Property Optimization: Design, Synthesis, and Characterization of Novel Pharmaceutical Salts and Cocrystal-Salt of Lumefantrine. Mol Pharm 2025; 22:1042-1060. [PMID: 39804247 DOI: 10.1021/acs.molpharmaceut.4c01244] [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: 02/04/2025]
Abstract
Lumefantrine (LMF) is a low-solubility antimalarial drug that cures acute, uncomplicated malaria. It exerts its pharmacological effects against erythrocytic stages of Plasmodium spp. and prevents malaria pathogens from producing nucleic acid and protein, thereby eliminating the parasites. Modifying the structure of a drug through the formation of a pharmaceutical cocrystal or salt presents an avenue through which its physicochemical properties can be optimized. In this work, we report the design/synthesis and solid-state characterization of four new salts and cocrystal-salt forms of LMF; an LMF-ADP salt, monoclinic space group P21/n; an LMF-FUM cocrystal-salt, monoclinic space group P21/c; an LMF-TAR solvate salt, monoclinic space group P21/n; and an LMF-SUC salt, triclinic, space group P1̅ (ADP, dianion of adipic acid; FUM, monoanion of fumaric acid; TAR, dianion of tartaric acid; SUC, dianion of succinic acid). These salts can be obtained by solution as well as by mechanochemical cocrystallization methods. The multicomponent systems gain their stability from hydrogen and partial ionic bonding interactions (N-H···O, O-H···O, N+-H···O-, and O-H+···O-) originating from both the dibutyl ammonium (N+-H) site and the alcohol hydroxyl (-OH) site of LMF toward the carboxylate (-C(O-)═O) functional groups of the coformer anions. SCXRD indicates for LMF-ADP, LMF-TAR, and LMF-SUC complete transfer of all carboxylic acid protons (H+) toward the LMF nitrogen, while for LMF-FUM, one of the protons is transferred (leaving a hydrofumarate monoanion). Using salicylic and acetylsalicylic acids as coformers yielded coamorphous solids. Solid-state characterization using powder X-ray diffraction (XRD) and thermal techniques (DSC and TGA) support and confirm the structures obtained from single-crystal XRD. LMF-ADP and LMF-FUM present superior stability under standard conditions (40 ± 2 °C, 75 ± 5% RH, and 3 months) compared to the amorphous samples and the other two salts. LMF-SUC showed poor thermal stability by DSC/TGA, and powder XRD patterns for LMF-TAR showed substantial change after the 3-month stability test. Finally, the calculated equilibrium solubilities for the cocrystal salts indicate an increase of more than twofold compared to LMF's solubility.
Collapse
Affiliation(s)
- Bolaji C Dayo Owoyemi
- Department of Pharmaceutical Science, University of São Paulo - USP, Av. Professor Lineu Prestes, 580 - Cidade Universitária São Paulo, São Paulo CEP 05508-000, Brazil
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Matthias Zeller
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, Indiana 47907, United States
| | - Brenda Pereira da Silva
- Department of Pharmaceutical Science, University of São Paulo - USP, Av. Professor Lineu Prestes, 580 - Cidade Universitária São Paulo, São Paulo CEP 05508-000, Brazil
| | - Amos O Akinyemi
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington 40536-0298, United States
| | - Romulo A Ando
- Institute of Chemistry, University of São Paulo - USP, Av. Professor Lineu Prestes, 748 - Cidade Universitária São Paulo, São Paulo CEP 05508-000, Brazil
| | - Gabriel L Barros de Araujo
- Department of Pharmaceutical Science, University of São Paulo - USP, Av. Professor Lineu Prestes, 580 - Cidade Universitária São Paulo, São Paulo CEP 05508-000, Brazil
| | - Stephen R Byrn
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, 575 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
4
|
Juneja M, Mehtre K, Saini V, Singh R, Amate P, Kashyap M, Sangamwar AT. Synergistic effect of polymers in stabilizing amorphous pretomanid through high drug loaded amorphous solid dispersion. Drug Deliv Transl Res 2025; 15:717-731. [PMID: 38837117 DOI: 10.1007/s13346-024-01630-w] [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] [Accepted: 05/13/2024] [Indexed: 06/06/2024]
Abstract
Pretomanid (PTM), an oral antibiotic used in the treatment of adults with pulmonary extensively drug-resistant, nonresponsive multidrug-resistant tuberculosis (MDR-TB). It is a poor glass former, that shows high recrystallization tendency from the amorphous and supersaturated state, resulting in low aqueous solubility and suboptimal absorption through the gastrointestinal tract. The present investigation aimed to develop high drug loaded ternary amorphous solid dispersions (ASDs) of PTM with improved stability and enhanced biopharmaceutical performance by utilizing a combination of polymers. The polymers were comprehensively screened based on drug-polymer miscibility and saturation solubility analysis. A combination of Hydroxypropyl Methylcellulose Acetate Succinate (HPMCAS-HF) and Polyvinylpyrrolidone K-30 (PVP K-30) showed synergism in drug-polymer miscibility as evidenced through pronounced depression in the melting endotherm of PTM. The Powder X-ray Diffraction (P-XRD) diffractograms of 30% w/w PTM loaded ternary ASDs displayed the halo pattern, contrary to the binary ASDs. Drug-polymer interactions (hydrophobic forces) involved between PTM and polymers were detected through Fourier Transform Infrared Spectroscopy (FT-IR) and Nuclear Magnetic Resonance Spectroscopy (13C-NMR) which contributed to the synergistic enhancement in solubility and dissolution of ternary ASDs with sustained release over 12 h. Ternary ASDs demonstrated better in-vivo performance compared to the binary ASDs, showing a 4.63-fold increase in maximum plasma concentration. All ASDs remained stable and resisted phase separation during short-term stability studies for 3 months at ambient conditions. It was concluded that the hydrophobic and hydrophilic polymeric combination (HPMCAS-HF and PVP K-30, respectively) effectively prevented the crystallization and ensured sustained drug release with improved in-vivo absorption of PTM.
Collapse
Affiliation(s)
- Mehak Juneja
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Krishna Mehtre
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Vanshul Saini
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Ridhima Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Prakash Amate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Mahesh Kashyap
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062
| | - Abhay T Sangamwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, S.A.S. Nagar, Punjab, India, 160062.
| |
Collapse
|
5
|
Deac A, Que C, Cousineau ML, Indulkar AS, Gao Y, Zhang GGZ, Taylor LS. Dissolution mechanisms of amorphous solid dispersions: Role of polymer molecular weight and identification of a new failure mode. J Pharm Sci 2025; 114:486-496. [PMID: 39461502 DOI: 10.1016/j.xphs.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024]
Abstract
The mechanisms of drug release from amorphous solid dispersions (ASDs) are complex and not fully explored, making it difficult to optimize for in vivo performance. A recurring behavior has been the limit of congruency (LoC), a drug loading above which the ASD surface forms an amorphous drug-rich barrier in the presence of water, which hinders release, especially in non-sink conditions. Drug-polymer interactions and drug glass transition temperature were reported to affect the LoC. However, the effect of polymer molecular weight has not been explored. ASDs of clotrimazole and different molecular weight grades of poly (vinylpyrrolidone) (PVP) were studied for their release to obtain their LoC drug loadings. Failure modes underpinning the LoC were investigated using fluorescence confocal microscopy to analyze the ASD/solution interface and phase behavior of ASD films at high relative humidity. ASDs with good release formed stable drug-rich nanodroplets at the ASD/solution interface, while ASDs with poor release were limited by one of two failure modes, depending on PVP molecular weight. In Failure Mode I the nanodroplets quickly agglomerated, while in Failure Mode II the system underwent phase inversion. This work highlights the importance of identifying the mechanisms underlying the LoC to improve the release of higher drug loading ASDs.
Collapse
Affiliation(s)
- Alexandru Deac
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Chailu Que
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Michelle L Cousineau
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States
| | - Yi Gao
- Formulation Development, Drug Product Science & Technology, AbbVie Inc., North Chicago, IL 60064, United States.
| | - Geoff G Z Zhang
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States; Development Sciences, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States; ProPhysPharm LLC, Lincolnshire, IL 60069, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|
6
|
Moseson DE, Li N, Rantanen J, Ueda K, Zhang GGZ. Professor Lynne S. Taylor: Scientist, educator, and adventurer. J Pharm Sci 2025; 114:2-9. [PMID: 39426563 DOI: 10.1016/j.xphs.2024.10.015] [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: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
This special edition of the Journal of Pharmaceutical Sciences is dedicated to Professor Lynne S. Taylor (Retter Distinguished Professor of Pharmacy, Department of Industrial and Molecular Pharmaceutics, Purdue University), to honor her distinguished career as a pharmaceutical scientist and educator. The goal of this commentary is to provide an overview of Professor Taylor's career path, summarize her key research contributions, and provide some insight into her personal and professional contributions as an educator, mentor, wife, mother, friend, and adventurer.
Collapse
Affiliation(s)
- Dana E Moseson
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States.
| | - Na Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269, United States
| | - Jukka Rantanen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Keisuke Ueda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Geoff G Z Zhang
- ProPhysPharm LLC, Lincolnshire, Illinois 60069, United States; Department of Industrial and Molecular Pharmaceutics, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
7
|
Hiew TN, Solomos MA, Kafle P, Polyzois H, Zemlyanov DY, Punia A, Smith D, Schenck L, Taylor LS. The importance of surface composition and wettability on the dissolution performance of high drug loading amorphous dispersion formulations. J Pharm Sci 2025; 114:289-303. [PMID: 39349295 DOI: 10.1016/j.xphs.2024.09.020] [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: 06/27/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/02/2024]
Abstract
One of the limitations with an amorphous solid dispersion (ASD) formulation strategy is low drug loading. Hydrophobic drugs have poor wettability and require a substantial amount of polymer to stabilize the amorphous drug and facilitate release. Using grazoprevir and hypromellose acetate succinate as model drug and polymer, respectively, the interplay between particle surface composition, particle wettability, and release performance was investigated. A hierarchical particle approach was used where the surfaces of high drug loading ASDs generated by either solvent evaporation or co-precipitation were further modified with a secondary excipient (i.e., polymer or wetting agent). The surface-modified particles were characterized for drug release, wettability, morphology, and surface composition using two-stage dissolution studies, contact angle measurements, scanning electron microscopy, and X-ray photoelectron spectroscopy, respectively. Despite surface modification with hydrophilic polymers, the hierarchical particles did not consistently exhibit good release performance. Contact angle measurements showed that the secondary excipient had a profound impact on particle wettability. Particles with good wettability showed improved drug release relative to particles that did not wet well, even with similar drug loadings. These observations underscore the intricate interplay between particle wettability and performance in amorphous dispersion formulations and illustrate a promising hierarchical particle approach to formulate high drug loading amorphous dispersions with improved dissolution performance.
Collapse
Affiliation(s)
- Tze Ning Hiew
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Marina A Solomos
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Prapti Kafle
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Hector Polyzois
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ashish Punia
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Daniel Smith
- Analytical Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Luke Schenck
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States.
| |
Collapse
|
8
|
Song Y, Chen Y, Heng W, Hu E, Shi Y, Gao Y, Zhang J, Wei Y, Qian S. The potential of supramolecular synthon to develop coamorphous systems with tailored physical stability: Mechanistic insights integrating kinetics and thermodynamics. Int J Pharm 2024; 667:124857. [PMID: 39442766 DOI: 10.1016/j.ijpharm.2024.124857] [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: 07/22/2024] [Revised: 10/08/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
Coamorphous drug delivery systems have received increasing interest owing to their potential to improve the solubility, dissolution and bioavailability of poorly water-soluble drugs. However, the crystallization risk is one of major limitations in their application. It has been widely recognized that the coformer plays a vital role in physical stability of coamorphous formulation. Unfortunately, the screen of optimal coformer still adopts a trial-and-error method, which is time-consuming and expensive. Herein, a supramolecular synthon approach based on the interaction between functional groups, was exploited to design coamorphous systems (CMs) consisting of lurasidone hydrochloride (LH) and three coformers, saccharin (SAC), L-tryptophan (TRP), and L-cysteine hydrochloride (CYS). X-ray powder diffraction suggested the order of physical stability of the coamorphous systems was ranked as LH-CYS CM > LH-TRP CM > LH-SAC CM. The charge-assisted hydrogen bond between LH and coformer was confirmed by infrared spectroscopy and solid-state 13C NMR. Moreover, structural, electronic, and molecular interaction information, especially hydrogen bonding interactions, were quantified by theoretical calculations, including miscibility calculations, molecular dynamics simulations and quantum chemical calculations. It was revealed that LH-CYS CM exhibited the best miscibility, strongest binding energy and strongest H-bond with partially covalent character, demonstrating the significant role of supramolecular synthon in stabilizing coamorphous formulations. Interestingly, LH-TRP CM, not LH-CYS CM, exhibited the lowest molecular mobility among three coamorphous systems, which was inconsistent with their physical stability. But from thermodynamic perspective, the order of configurational entropy and physical stability of coamorphous systems was completely consistent. We shed light on the comprehensive effects of molecular mobility and configurational entropy on physical stability of coamorphous systems. Importantly, the relationship between supramolecular synthon and kinetic/thermodynamic mechanisms was also discussed, and the positive correlation between configurational entropy and intermolecular interactions was proposed in this paper. Our findings demonstrated the great potential of supramolecular synthon in designing coamorphous systems with tailored physical stability, and further provided a deeper insight into the mechanisms of physical stability of coamorphous systems.
Collapse
Affiliation(s)
- Yutong Song
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China; School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Yu Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Weili Heng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, PR China
| | - Enshi Hu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Yunyi Shi
- Department of Biology, Emory University, Atlanta, GA 30332, United States
| | - Yuan Gao
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China
| | - Jianjun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Yuanfeng Wei
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Shuai Qian
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
9
|
Luo C, Li R, Tang M, Gao Y, Zhang J, Qian S, Wei Y, Shen P. Amorphous solid dispersion to facilitate the delivery of poorly water-soluble drugs: recent advances on novel preparation processes and technology coupling. Expert Opin Drug Deliv 2024; 21:1807-1822. [PMID: 39484838 DOI: 10.1080/17425247.2024.2423813] [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: 07/29/2024] [Revised: 10/13/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024]
Abstract
INTRODUCTION Amorphous solid dispersion (ASD) technique has recently been used as an effective formulation strategy to significantly improve the bioavailability of insoluble drugs. The main industrialized preparation methods for ASDs are mainly hot melt extrusion and spray drying techniques; however, they face the limitations of being unsuitable for heat-sensitive materials and organic reagent residues, respectively, and therefore novel preparation processes and technology coupling for developing ASDs have received increasing attention. AREAS COVERED This paper reviews recent advances in ASD and provides an overview of novel preparation methods, mechanisms for improving drug bioavailability, and especially technology coupling. EXPERT COVERED As a mature pharmaceutical technology, ASD has broad application prospects and values. During the period from 2012 to 2024, the FDA has approved 49 formulation products containing ASDs. However, with the diversification of drug types and clinical needs, the traditional formulation technology of ASDs is gradually no longer sufficient to meet the needs of clinical medication. Therefore, this review summarizes the studies on both novel preparation processes and technology combinations; and provides a comprehensive overview of the mechanisms of ASD to improve drug bioavailability, in order to better select appropriate preparation methods for the development of ASD formulations.
Collapse
Affiliation(s)
- Chengxiang Luo
- School of Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Ruipeng Li
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Mi Tang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
- Jiangsu Litaier Pharma Ltd, Nanjing, China
| | - Yuan Gao
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Jianjun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Shuai Qian
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Yuanfeng Wei
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Peiya Shen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
10
|
Helmy AM, Lu A, Duggal I, Rodrigues KP, Maniruzzaman M. Electromagnetic drop-on-demand (DoD) technology as an innovative platform for amorphous solid dispersion production. Int J Pharm 2024; 658:124185. [PMID: 38703932 DOI: 10.1016/j.ijpharm.2024.124185] [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: 01/12/2024] [Revised: 04/25/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
Production of amorphous solid dispersions (ASDs) is an effective strategy to promote the solubility and bioavailability of poorly water soluble medicinal substances. In general, ASD is manufactured using a variety of classic and modern techniques, most of which rely on either melting or solvent evaporation. This proof-of-concept study is the first ever to introduce electromagnetic drop-on-demand (DoD) technique as an alternative solvent evaporation-based method for producing ASDs. Herein 3D printing of ASDs for three drug-polymer combinations (efavirenz-Eudragit L100-55, lumefantrine-hydroxypropyl methylcellulose acetate succinate, and favipiravir-polyacrylic acid) was investigated to ascertain the reliability of this technique. Polarized light microscopy, differential scanning calorimetry (DSC), X-ray powder diffraction (XRPD), and Fourier Transform Infrared (FTIR) spectroscopy results supported the formation of ASDs for the three drugs by means of DoD 3D printing, which significantly increases the equilibrium solubility of efavirenz from 0.03 ± 0.04 µg/ml to 21.18 ± 4.20 µg/ml, and the equilibrium solubility of lumefantrine from 1.26 ± 1.60 µg/ml to 20.21 ± 6.91 µg/ml. Overall, the reported findings show how this new electromagnetic DoD technology can have a potential to become a cutting-edge 3D printing solvent-evaporation technique for on-demand and continuous manufacturing of ASDs for a variety of drugs.
Collapse
Affiliation(s)
- Abdelrahman M Helmy
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Deraya University, Minya, Egypt
| | - Anqi Lu
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ishaan Duggal
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Kristina P Rodrigues
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Mohammed Maniruzzaman
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA; Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, University, MS 38677-1848, USA.
| |
Collapse
|
11
|
Moreira GG, Taveira SF, Martins FT, Wagner KG, Marreto RN. Multivariate Analysis of Solubility Parameters for Drug-Polymer Miscibility Assessment in Preparing Raloxifene Hydrochloride Amorphous Solid Dispersions. AAPS PharmSciTech 2024; 25:127. [PMID: 38844724 DOI: 10.1208/s12249-024-02844-4] [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: 03/26/2024] [Accepted: 05/21/2024] [Indexed: 09/05/2024] Open
Abstract
The success of obtaining solid dispersions for solubility improvement invariably depends on the miscibility of the drug and polymeric carriers. This study aimed to categorize and select polymeric carriers via the classical group contribution method using the multivariate analysis of the calculated solubility parameter of RX-HCl. The total, partial, and derivate parameters for RX-HCl were calculated. The data were compared with the results of excipients (N = 36), and a hierarchical clustering analysis was further performed. Solid dispersions of selected polymers in different drug loads were produced using solvent casting and characterized via X-ray diffraction, infrared spectroscopy and scanning electron microscopy. RX-HCl presented a Hansen solubility parameter (HSP) of 23.52 MPa1/2. The exploratory analysis of HSP and relative energy difference (RED) elicited a classification for miscible (n = 11), partially miscible (n = 15), and immiscible (n = 10) combinations. The experimental validation followed by a principal component regression exhibited a significant correlation between the crystallinity reduction and calculated parameters, whereas the spectroscopic evaluation highlighted the hydrogen-bonding contribution towards amorphization. The systematic approach presented a high discrimination ability, contributing to optimal excipient selection for the obtention of solid solutions of RX-HCl.
Collapse
Affiliation(s)
- Guilherme G Moreira
- Laboratory of Nanosystems and Drug Delivery Devices (NanoSYS), School of Pharmacy, Universidade Federal de Goiás, Goiânia, Goiás, 74.605-170, Brazil
| | - Stephânia F Taveira
- Laboratory of Nanosystems and Drug Delivery Devices (NanoSYS), School of Pharmacy, Universidade Federal de Goiás, Goiânia, Goiás, 74.605-170, Brazil
| | - Felipe T Martins
- Institute of Chemistry, Universidade Federal de Goiás, Goiânia, 74.001-970, Brazil
| | - Karl G Wagner
- Department of Pharmaceutics, Pharmaceutical Institute, University of Bonn, 53121, Bonn, Germany
| | - Ricardo N Marreto
- Laboratory of Nanosystems and Drug Delivery Devices (NanoSYS), School of Pharmacy, Universidade Federal de Goiás, Goiânia, Goiás, 74.605-170, Brazil.
| |
Collapse
|
12
|
Moseson DE, Tran TB, Karunakaran B, Ambardekar R, Hiew TN. Trends in amorphous solid dispersion drug products approved by the U.S. Food and Drug Administration between 2012 and 2023. Int J Pharm X 2024; 7:100259. [PMID: 38974024 PMCID: PMC11225173 DOI: 10.1016/j.ijpx.2024.100259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 07/09/2024] Open
Abstract
Forty-eight (48) drug products (DPs) containing amorphous solid dispersions (ASDs) have been approved by the U.S. Food and Drug Administration in the 12-year period between 2012 and 2023. These DPs comprise 36 unique amorphous drugs. Ten (10) therapeutic categories are represented, with most DPs containing antiviral and antineoplastic agents. The most common ASD polymers are copovidone (49%) and hypromellose acetate succinate (30%), while spray drying (54%) and hot melt extrusion (35%) are the most utilized manufacturing processes to prepare the ASD drug product intermediate (DPI). Tablet dosage forms are the most common, with several capsule products available. Line extensions of several DPs based on flexible oral solids and powders for oral suspension have been approved which provide patient-centric dosing to pediatric and other patient populations. The trends in the use of common excipients and film coating types are discussed. Eighteen (18) DPs are fixed-dose combinations, and some contain a mixture of amorphous and crystalline drugs. The DPs have dose/unit of amorphous drug ranging from <5 mg up to 300 mg, with the majority being ≤100 mg/unit. This review details several aspects of DPI and DP formulation and manufacturing of ASDs, as well as trends related to therapeutic category, dose, and patient-centricity.
Collapse
Affiliation(s)
- Dana E. Moseson
- Worldwide Research and Development, Pfizer, Inc., Groton, CT 06340, USA
| | - Trong Bien Tran
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa 52242, USA
| | - Bharathi Karunakaran
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa 52242, USA
| | - Rohan Ambardekar
- Worldwide Research and Development, Pfizer, Inc., Sandwich CT13 9NJ, UK
| | - Tze Ning Hiew
- Department of Pharmaceutical Sciences and Experimental Therapeutics, College of Pharmacy, University of Iowa, Iowa 52242, USA
| |
Collapse
|
13
|
Deac A, Luebbert C, Qi Q, Courtney RM, Indulkar AS, Gao Y, Zhang GGZ, Sadowski G, Taylor LS. Dissolution Mechanisms of Amorphous Solid Dispersions: Application of Ternary Phase Diagrams To Explain Release Behavior. Mol Pharm 2024; 21:1900-1918. [PMID: 38469754 DOI: 10.1021/acs.molpharmaceut.3c01179] [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: 03/13/2024]
Abstract
The use of amorphous solid dispersions (ASDs) in commercial drug products has increased in recent years due to the large number of poorly soluble drugs in the pharmaceutical pipeline. However, the release behavior of ASDs is complex and remains not well understood. Often, the drug release from ASDs is rapid and complete at lower drug loadings (DLs) but becomes slow and incomplete at higher DLs. The DL where release becomes hindered is termed the limit of congruency (LoC). Currently, there are no approaches to predict the LoC. However, recent findings show that one potential cause leading to the LoC is a change in phase morphology after water-induced phase separation at the ASD/solution interface. In this study, the phase behavior of ASDs in contact with aqueous solutions was described thermodynamically by constructing experimental and computational ternary phase diagrams, and these were used to predict morphology changes and ultimately the LoC. Experimental ternary phase diagrams were obtained by equilibrating ASD/water mixtures over time. Computational ternary phase diagrams were obtained by Perturbed Chain Statistical Associating Fluid Theory (PC-SAFT). The morphology of the hydrophobic phase was studied with fluorescence confocal microscopy. It was demonstrated that critical point (plait point) composition approximately corresponded to the ASD DL, where the hydrophobic phase, formed during phase separation, became interconnected and hindered ASD release. This work provides mechanistic insights into the ASD release behavior and highlights the potential of in silico ASD design using phase diagrams.
Collapse
Affiliation(s)
- Alexandru Deac
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | | | - Qingqing Qi
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Reagan M Courtney
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Yi Gao
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Geoff G Z Zhang
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | | | - Lynne S Taylor
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
14
|
Hasegawa K, Goto S, Kataoka H, Chatani H, Kinoshita T, Yokoyama H, Tsuchida T. Quantification of crystallinity during indomethacin crystalline transformation from α- to γ-polymorphic forms and of the thermodynamic contribution to dissolution in aqueous buffer and solutions of solubilizer. RSC Adv 2024; 14:4129-4141. [PMID: 38292264 PMCID: PMC10825737 DOI: 10.1039/d3ra08481g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/19/2024] [Indexed: 02/01/2024] Open
Abstract
The thermodynamic properties and dissolution of indomethacin (INM) were analyzed as models for poorly water-soluble drugs. Physical mixtures of the most stable γ-form and metastable α-form of INM at various proportions were prepared, and their individual signal intensities proportional to their mole fractions were observed using X-ray powder diffraction and Fourier transform infrared spectrometry at standard temperature. The endothermic signals of the α-form, with a melting point of 426 K, and that of the γ-form, with a melting point of 433 K, were obtained by differential scanning calorimetry (DSC). Furthermore, an exothermic DSC peak of the α/γ-phase transition at approximately 428 K was obtained. As we computed the melting entropy of the α-form and that of its transformation, the frequency of the transition was quantitatively determined, which indicated the maximum of the α/γ-phase transition at an α-form proportion of 68%. Subsequently, the thermodynamic contributions of the α- and γ-forms were analyzed using a Van't Hoff plot for solubility in aqueous solutions at pH 6.8. The dissolution enthalpies for α- and γ-forms were 28.2 and 31.2 kJ mol-1, respectively, which are in agreement with the quantitative contribution predicted by the product of the temperature and melting entropy. The contribution of melting entropy was conserved in different dissolution processes with aqueous solvents containing lidocaine, diltiazem, l-carnosine, and aspartame as solubilizers; their γ-form Setschenow coefficients were -39.6, +82.9, -17.3, and +23.2, whereas those of the α-form were -39.7, +80.4, -16.7, and +22.7, respectively. We conclude that the dissolution ability of the solid state and solubilizers indicate their additivity independently.
Collapse
Affiliation(s)
- Kanji Hasegawa
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Satoru Goto
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Hikaru Kataoka
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Hitoshi Chatani
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Takatoshi Kinoshita
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Hideshi Yokoyama
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| | - Tomohiro Tsuchida
- Faculty of Pharmaceutical Sciences, Tokyo University of Science 2641 Yamazaki, Noda Chiba 278-8510 Japan
| |
Collapse
|
15
|
Tripathi D, B H MP, Sahoo J, Kumari J. Navigating the Solution to Drug Formulation Problems at Research and Development Stages by Amorphous Solid Dispersion Technology. RECENT ADVANCES IN DRUG DELIVERY AND FORMULATION 2024; 18:79-99. [PMID: 38062659 DOI: 10.2174/0126673878271641231201065151] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 08/30/2024]
Abstract
Amorphous Solid Dispersions (ASDs) have indeed revolutionized the pharmaceutical industry, particularly in drug solubility enhancement. The amorphous state of a drug, which is a highenergy metastable state, can lead to an increase in the apparent solubility of the drug. This is due to the absence of a long-range molecular order, which results in higher molecular mobility and free volume, and consequently, higher solubility. The success of ASD preparation depends on the selection of appropriate excipients, particularly polymers that play a crucial role in drug solubility and physical stability. However, ASDs face challenges due to their thermodynamic instability or tendency to recrystallize. Measuring the crystallinity of the active pharmaceutical ingredient (API) and drug solubility is a complex process that requires a thorough understanding of drug-polymer miscibility and molecular interactions. Therefore, it is important to monitor drug solids closely during preparation, storage, and application. Techniques such as solid-state nuclear magnetic resonance (ssNMR), attenuated total reflectance Fourier transform infrared spectroscopy (ATR-FTIR), Raman spectroscopy, and dielectric spectroscopy have been successful in understanding the mechanism of drug crystallization. In addition, the continuous downstream processing of drug-loaded ASDs has introduced new automated methods for consistent ASD production. Advanced techniques such as hot melt extrusion, KinetiSol, electro spraying, and electrospinning have gained popularity. This review provides a comprehensive overview of Amorphous Solid Dispersions (ASDs) for oral drug delivery. It highlights the critical challenges faced during formulation, the impact of manufacturing variables, theoretical aspects of drug-polymer interaction, and factors related to drug-polymer miscibility. ASDs have been recognized as a promising strategy to improve the oral bioavailability of poorly water-soluble drugs. However, the successful development of an ASD-based drug product is not straightforward due to the complexity of the ASD systems. The formulation and process parameters can significantly influence the performance of the final product. Understanding the interactions between the drug and polymer in ASDs is crucial for predicting their stability and performance.
Collapse
Affiliation(s)
- Devika Tripathi
- Pranveer Singh Institute of Technology (Pharmacy), Uttar Pradesh, Kanpur, India
| | - Manjunatha Prabhu B H
- Department of Food Protection and Infestation Control, CSIR-CFTRI, Central Food Technological Research Institute, Mysore, India
| | - Jagannath Sahoo
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, NIMMS, Mumbai, India
| | - Jyoti Kumari
- Pranveer Singh Institute of Technology (Pharmacy), Uttar Pradesh, Kanpur, India
| |
Collapse
|
16
|
Li C, Li N, Chen X, Li X, Liu C, Abbas A, Wang Y, Qi S, Zhang Y, Li D, Zhang W, Shu G, Lin J, Li H, Xu F, Peng G, Fu H. Enhancement of dissolution rate and oral bioavailability of poorly soluble drug florfenicol by using solid dispersion and effervescent disintegration technology. Drug Dev Ind Pharm 2024; 50:45-54. [PMID: 38095592 DOI: 10.1080/03639045.2023.2295488] [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: 10/05/2023] [Accepted: 12/01/2023] [Indexed: 12/29/2023]
Abstract
OBJECTIVE Florfenicol(FF) is an excellent veterinary antibiotic, limited by poor solubility and poor bioavailability. SIGNIFICANCE Here in, we aimed to explore the applicability of fast disintegrating tablets compressed from Florfenicol-loaded solid dispersions (FF-SD-FDTs) to improve the dissolution rate and oral bioavailability of Florfenicol. METHODS Utilizing selecting appropriate preparation methods and carriers, the solid dispersions of Florfenicol (FF-SDs) were prepared by solvent evaporation and the fast disintegrating tablets (FF-SD-FDTs) were prepared by the direct compression (DC) method. RESULTS The tablet properties including hardness, friability, disintegration time, weight variation, etc. all met the specifications of Chinese Veterinary Pharmacopeia(CVP). FF-SD-FDTs significantly improved drug dissolution and dispersion of FF in vitro compared to florfenicol conventional tablets (FF-CTs). A pharmacokinetics study in German shepherd dogs proved the AUC0-∞ and Cmax values of FF-SD-FDTs are 1.38 and 1.38 times more than FF-CTs, respectively. CONCLUSIONS Overall, it can be concluded that FF-SD-FDTs with excellent disintegration and dissolution properties were successfully produced, which greatly improved the oral bioavailability of the poorly soluble drug FF, and the study provided a new idea for a broader role of FF in pet clinics.
Collapse
Affiliation(s)
- Chao Li
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Nanxin Li
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xingyu Chen
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Xiaojuan Li
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Chang Liu
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Awn Abbas
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yueli Wang
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Shuangcai Qi
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yifan Zhang
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Dongbo Li
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Wei Zhang
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Gang Shu
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Juchun Lin
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Haohuan Li
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Funeng Xu
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Guangneng Peng
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| | - Hualin Fu
- Department of Pharmacy, College of Vet Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
17
|
Chiang CW, Tang S, Mao C, Chen Y. Effect of Buffer pH and Concentration on the Dissolution Rates of Sodium Indomethacin-Copovidone and Indomethacin-Copovidone Amorphous Solid Dispersions. Mol Pharm 2023; 20:6451-6462. [PMID: 37917181 DOI: 10.1021/acs.molpharmaceut.3c00827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
The incorporation of counterions into amorphous solid dispersions (ASDs) has been proven to be effective for improving the dissolution rates of ionizable drugs in ASDs. In this work, the effect of dissolution buffer pH and concentration on the dissolution rate of indomethacin-copovidone 40:60 (IMC-PVPVA, w/w) ASD with or without incorporated sodium hydroxide (NaOH) was studied by surface area-normalized dissolution to provide further mechanistic understanding of this phenomenon. Buffer pH from 4.7 to 7.2 and concentration from 20 to 100 mM at pH 5.5 were investigated. As the buffer pH decreased, the IMC dissolution rate from both ASDs decreased. Compared to IMC-PVPVA ASD, the dissolution rate decrease from IMCNa-PVPVA ASD was more resistant to the decrease of buffer pH. In contrast, while buffer concentration had a negligible impact on the IMC dissolution rate from IMC-PVPVA ASD, the increase of buffer concentration significantly reduced the IMC dissolution rate from IMCNa-PVPVA ASD. Surrogate evaluation of microenvironment pH modification by the dissolution of IMCNa-PVPVA ASD demonstrated the successful elevation of buffer microenvironment pH and the suppression of such pH elevation by the increase of buffer concentration. These results are consistent with the hypothesis that the dissolution rate enhancement by the incorporation of counterions originates from the enhanced drug solubility by ionization and the modification of diffusion layer pH in favor of drug dissolution. At the studied drug loading (∼40%), relatively congruent release between IMC and PVPVA was observed when IMC was ionized in ASD or in solution, highlighting the importance of studying the ionization effect on the congruent release of ASDs, especially when drug ionization is expected in vivo. Overall, this work further supports the application of incorporating counterions into ASDs for improving the dissolution rates of ionizable drugs when enabling formulation development is needed.
Collapse
Affiliation(s)
- Cheng W Chiang
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Shijia Tang
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Chen Mao
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yinshan Chen
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
18
|
Bookwala M, Wildfong PLD. The Implications of Drug-Polymer Interactions on the Physical Stability of Amorphous Solid Dispersions. Pharm Res 2023; 40:2963-2981. [PMID: 37389801 DOI: 10.1007/s11095-023-03547-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/03/2023] [Indexed: 07/01/2023]
Abstract
Amorphous solid dispersions (ASDs) are a formulation and development strategy that can be used to increase the apparent aqueous solubility of poorly water-soluble drugs. Their implementation, however, can be hindered by destabilization of the amorphous form, as the drug recrystallizes from its metastable state. Factors such as the drug-polymer solubility, miscibility, mobility, and nucleation/crystal growth rates are all known to impact the physical stability of an ASD. Non-covalent interactions (NCI) between the drug and polymer have also been widely reported to influence product shelf-life. In this review, the relationship between thermodynamic/kinetic factors and adhesive NCI is assessed. Various types of NCIs reported to stabilize ASDs are described, and their role in affecting physical stability is examined. Finally, NCIs that have not yet been widely explored in ASD formulations, but may potentially impact their physical stability are also briefly described. This review aims to stimulate further theoretical and practical exploration of various NCIs and their applications in ASD formulations in the future.
Collapse
Affiliation(s)
- Mustafa Bookwala
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, 422C Mellon Hall, Pittsburgh, PA, 15282, USA
| | - Peter L D Wildfong
- School of Pharmacy and Graduate School of Pharmaceutical Sciences, Duquesne University, 600 Forbes Avenue, 422C Mellon Hall, Pittsburgh, PA, 15282, USA.
| |
Collapse
|
19
|
Mukesh S, Mukherjee G, Singh R, Steenbuck N, Demidova C, Joshi P, Sangamwar AT, Wade RC. Comparative analysis of drug-salt-polymer interactions by experiment and molecular simulation improves biopharmaceutical performance. Commun Chem 2023; 6:201. [PMID: 37749228 PMCID: PMC10519957 DOI: 10.1038/s42004-023-01006-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 09/14/2023] [Indexed: 09/27/2023] Open
Abstract
The propensity of poorly water-soluble drugs to aggregate at supersaturation impedes their bioavailability. Supersaturated amorphous drug-salt-polymer systems provide an emergent approach to this problem. However, the effects of polymers on drug-drug interactions in aqueous phase are largely unexplored and it is unclear how to choose an optimal salt-polymer combination for a particular drug. Here, we describe a comparative experimental and computational characterization of amorphous solid dispersions containing the drug celecoxib, and a polymer, polyvinylpyrrolidone vinyl acetate (PVP-VA) or hydroxypropyl methylcellulose acetate succinate, with or without Na+/K+ salts. Classical models for drug-polymer interactions fail to identify the best drug-salt-polymer combination. In contrast, more stable drug-polymer interaction energies computed from molecular dynamics simulations correlate with prolonged stability of supersaturated amorphous drug-salt-polymer systems, along with better dissolution and pharmacokinetic profiles. The celecoxib-salt-PVP-VA formulations exhibit excellent biopharmaceutical performance, offering the prospect of a low-dosage regimen for this widely used anti-inflammatory, thereby increasing cost-effectiveness, and reducing side-effects.
Collapse
Affiliation(s)
- Sumit Mukesh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector-67, Mohali, Punjab, 160062, India
| | - Goutam Mukherjee
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Im Neuenheimer Feld 282, 69120, Heidelberg, Germany
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany
| | - Ridhima Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector-67, Mohali, Punjab, 160062, India
| | - Nathan Steenbuck
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Heidelberg University, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany
| | - Carolina Demidova
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany
- Faculty of Chemistry, Heidelberg University, Im Neuenheimer Feld 364, 69120, Heidelberg, Germany
| | - Prachi Joshi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector-67, Mohali, Punjab, 160062, India
| | - Abhay T Sangamwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Sector-67, Mohali, Punjab, 160062, India.
| | - Rebecca C Wade
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Im Neuenheimer Feld 282, 69120, Heidelberg, Germany.
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies, Schloss-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany.
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Im Neuenheimer Feld 205, Heidelberg, Germany.
| |
Collapse
|
20
|
Li J, Wang Y, Yu D. Effects of Additives on the Physical Stability and Dissolution of Polymeric Amorphous Solid Dispersions: a Review. AAPS PharmSciTech 2023; 24:175. [PMID: 37603110 DOI: 10.1208/s12249-023-02622-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
Polymeric amorphous solid dispersion (ASD) is a popular approach for enhancing the solubility of poorly water-soluble drugs. However, achieving both physical stability and dissolution performance in an ASD prepared with a single polymer can be challenging. Therefore, a secondary excipient can be added. In this paper, we review three classes of additives that can be added internally to ASDs: (i) a second polymer, to form a ternary drug-polymer-polymer ASD, (ii) counterions, to facilitate in situ salt formation, and (iii) surfactants. In an ASD prepared with a combination of polymers, each polymer exerts a unique function, such as a stabilizer in the solid state and a crystallization inhibitor during dissolution. In situ salt formation in ASD usually leads to substantial increases in the glass transition temperature, contributing to improved physical stability. Surfactants can enhance the wettability of ASD particles, thereby promoting rapid drug release. However, their potential adverse effects on physical stability and dissolution, resulting from enhanced molecular mobility and competitive molecular interaction with the polymer, respectively, warrant careful consideration. Finally, we discuss the impact of magnesium stearate and inorganic salts, excipients added externally upon downstream processing, on the solid-state stability as well as the dissolution of ASD tablets.
Collapse
Affiliation(s)
- Jinghan Li
- Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, 55455, USA
| | - Yihan Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, 20 North Pine Street, Baltimore, Maryland, 21201, USA
| | - Dongyue Yu
- Pharmaceutical Candidate Optimization, Bristol Myers Squibb, Route 206 and Province Line Road, Princeton, New Jersey, 08540, USA.
| |
Collapse
|
21
|
Zhang J, Guo M, Luo M, Cai T. Advances in the development of amorphous solid dispersions: The role of polymeric carriers. Asian J Pharm Sci 2023; 18:100834. [PMID: 37635801 PMCID: PMC10450425 DOI: 10.1016/j.ajps.2023.100834] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/26/2023] [Accepted: 07/23/2023] [Indexed: 08/27/2023] Open
Abstract
Amorphous solid dispersion (ASD) is one of the most effective approaches for delivering poorly soluble drugs. In ASDs, polymeric materials serve as the carriers in which the drugs are dispersed at the molecular level. To prepare the solid dispersions, there are many polymers with various physicochemical and thermochemical characteristics available for use in ASD formulations. Polymer selection is of great importance because it influences the stability, solubility and dissolution rates, manufacturing process, and bioavailability of the ASD. This review article provides a comprehensive overview of ASDs from the perspectives of physicochemical characteristics of polymers, formulation designs and preparation methods. Furthermore, considerations of safety and regulatory requirements along with the studies recommended for characterizing and evaluating polymeric carriers are briefly discussed.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- College of Biological and Chemical Engineering, Changsha University, Changsha 410022, China
| | - Minshan Guo
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Minqian Luo
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ting Cai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
22
|
Deac A, Qi Q, Indulkar AS, Gao Y, Zhang GGZ, Taylor LS. Dissolution Mechanisms of Amorphous Solid Dispersions: A Close Look at the Dissolution Interface. Mol Pharm 2023; 20:2217-2234. [PMID: 36926898 DOI: 10.1021/acs.molpharmaceut.3c00020] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Despite the recent success of amorphous solid dispersions (ASDs) at enabling the delivery of poorly soluble small molecule drugs, ASD-based dosage forms are limited by low drug loading. This is partially due to a sharp decline in drug release from the ASD at drug loadings surpassing the 'limit of congruency' (LoC). In some cases, the LoC is as low as 5% drug loading, significantly increasing the risk of pill burden. Despite efforts to understand the mechanism responsible for the LoC, a clear picture of the molecular processes occurring at the ASD/solution interface remains elusive. In this study, the ASD/solution interface was studied for two model compounds formulated as ASDs with copovidone. The evolution of a gel layer and its phase behavior was captured in situ with fluorescence confocal microscopy, where fluorescent probes were added to label the hydrophobic and hydrophilic phases. Phase separation was detected in the gel layer for most of the ASDs. The morphology of the hydrophobic phase was found to correlate with the release behavior, where a discrete phase resulted in good release and a continuous phase formed a barrier leading to poor release. The continuous phase formed at a lower drug loading for the system with stronger drug-polymer interactions. This was due to incorporation of the polymer into the hydrophobic phase. The study highlights the complex molecular and phase behavior at the ASD/solution interface of copovidone-based ASDs and provides a thermodynamic argument for qualitatively predicting the release behavior based on drug-polymer interactions.
Collapse
Affiliation(s)
- Alexandru Deac
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Qingqing Qi
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Anura S Indulkar
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Yi Gao
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Geoff G Z Zhang
- Development Sciences, Research and Development, AbbVie Inc., North Chicago, Illinois 60064, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
23
|
Hiew TN, Saboo S, Zemlyanov DY, Punia A, Wang M, Smith D, Lowinger M, Solomos MA, Schenck L, Taylor LS. Improving Dissolution Performance and Drug Loading of Amorphous Dispersions Through a Hierarchical Particle Approach. J Pharm Sci 2022:S0022-3549(22)00583-4. [PMID: 36574837 DOI: 10.1016/j.xphs.2022.12.019] [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: 10/12/2022] [Revised: 12/01/2022] [Accepted: 12/18/2022] [Indexed: 12/26/2022]
Abstract
Co-precipitation is an emerging manufacturing strategy for amorphous solid dispersions (ASDs). Herein, the interplay between processing conditions, surface composition, and release performance was evaluated using grazoprevir and hypromellose acetate succinate as the model drug and polymer, respectively. Co-precipitated amorphous dispersion (cPAD) particles were produced in the presence and absence of an additional polymer that was either dissolved or dispersed in the anti-solvent. This additional polymer in the anti-solvent was deposited on the surfaces of the cPAD particles during isolation and drying to create hierarchical particles, which we define here as a core ASD particle with an additional water soluble component that is coating the particle surfaces. The resultant hierarchical particles were characterized using X-ray powder diffraction, differential scanning calorimetry, scanning electron microscopy, and X-ray photoelectron spectroscopy (XPS). Release performance was evaluated using a two-stage dissolution test. XPS analysis revealed a trend whereby cPAD particles with a lower surface drug concentration showed improved release relative to particles with a higher surface drug concentration, for nominally similar drug loadings. This surface drug concentration could be impacted by whether the secondary polymer was dissolved in the anti-solvent or dispersed in the anti-solvent prior to isolating final dried hierarchical cPAD powders. Grazoprevir exposure in dogs was higher when the hierarchical cPAD was dosed, with ∼1.8 fold increase in AUC compared to the binary cPAD. These observations highlight the important interplay between processing conditions and ASD performance in the context of cPAD particles and illustrate a hierarchical particle design as a successful approach to alter ASD surface chemistry to improve dissolution performance.
Collapse
Affiliation(s)
- Tze Ning Hiew
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States
| | - Sugandha Saboo
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Dmitry Y Zemlyanov
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN 47907, United States
| | - Ashish Punia
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Michael Wang
- Biopharmaceutics, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Daniel Smith
- Analytical Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Michael Lowinger
- Oral Formulation Sciences, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Marina A Solomos
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Luke Schenck
- Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, IN 47907, United States.
| |
Collapse
|