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
|
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
|
3
|
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
|
4
|
Indulkar AS, Alex S, Zhang GGZ. Impact of dissolution medium pH and ionization state of the drug on the release performance of amorphous solid dispersions. J Pharm Sci 2025; 114:497-506. [PMID: 39454946 DOI: 10.1016/j.xphs.2024.10.028] [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: 07/31/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Amorphous solid dispersions (ASDs) are widely employed as a strategy to improve oral bioavailability of poorly water soluble compounds. Typically, optimal dissolution performance from a polyvinylpyrrolidone vinyl acetate (PVPVA) based ASD is observed at relatively low drug loading limit. Above a certain drug load, termed limit of congruency (LoC), the release from ASDs significantly decreases. So far, the majority of the dissolution behavior has been tested in conditions where the drug primarily exists in unionized form. In this work, the impact of pH of the dissolution environment on the release performance of ASDs of an ionizable drug was studied. Atazanavir (ATZ), a weakly basic drug with a pKa of 4.5 was used as a model compound and PVPVA was used as a non-ionizable matrix polymer. Dissolution rate was measured using Wood's apparatus which normalizes the surface area of the dissolving tablet. The pH of the dissolution media was varied between 1 and 6.8, to cover a range where ATZ exists as >99 % ionized or unionized species. At pH 6.8, near complete release was observed only when the drug load was ≤ 6 %. Unlike typically observed drastic decline in release behavior for PVPVA based ASDs above LoC, ATZ ASDs underwent gradual decline in dissolution behavior when the DL was increased to 8 %. This was attributed to potential formation of an ATZ-PVPVA associated phase with dissolution rate slower than neat PVPVA. However, the 10 % DL ASD showed negligible ATZ release. On another extreme (pH 1) where ATZ is ∼100 % ionized, the dissolution rate of ATZ was faster than that of PVPVA. ASD dissolution rate was found to be slower than that of the neat drug but faster than PVPVA and interestingly, did not change with DL. This can be attributed to formation of an ionized ATZ-PVPVA phase which controls the dissolution rate of the ASD. At pH 3, where the drug is ∼97 % ionized, near complete release was observed for drug loads ≤ 8 %. To observe significant increase in drug loading with near complete release, >98 % ionization of ATZ was required. At pH 2 where ATZ is ∼99.7 % ionized, near complete release was observed for drug loads up to 30 %. Furthermore, the deterioration in dissolution performance with an increase in drug load continued to be gradual at pH 2. The enhancement in dissolution performance did not correlate with solubility enhancement of ATZ due to ionization. We theorize that the enhancement in the dissolution performance due to ionization is the result of formation of an ionized ATZ-PVPVA phase which increases the hydrophilicity and the miscibility of the ASD. This can help resist water induced phase separation during ASD dissolution and therefore, result in continuous, and congruent dissolution of the drug and polymer.
Collapse
Affiliation(s)
- Anura S Indulkar
- Small Molecule CMC Development, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States.
| | - Samantha Alex
- Small Molecule CMC Development, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States
| | - Geoff G Z Zhang
- Small Molecule CMC Development, Research and Development, AbbVie Inc., North Chicago, IL 60064, United States
| |
Collapse
|
5
|
B S, Ghosh A. Mechanistic Insights into Amorphous Solid Dispersions: Bridging Theory and Practice in Drug Delivery. Pharm Res 2025; 42:1-23. [PMID: 39849216 DOI: 10.1007/s11095-024-03808-w] [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: 11/01/2024] [Accepted: 12/12/2024] [Indexed: 01/25/2025]
Abstract
Improving the bioavailability of poorly water-soluble drugs presents a significant challenge in pharmaceutical development. Amorphous solid dispersions (ASDs) have garnered substantial attention for their capability to augment the solubility and dissolution rate of poorly water-soluble drugs, thereby markedly enhancing their bioavailability. ASDs, characterized by a metastable equilibrium where the active pharmaceutical ingredient (API) is molecularly dispersed, offer enhanced absorption compared to crystalline forms. This review explores recent research advancements in ASD, emphasizing dissolution mechanisms, phase separation phenomena, and the importance of drug loading and congruency limits on ASD performance. Principal occurrences such as liquid-liquid phase separation (LLPS) and supersaturation are discussed, highlighting their impact on drug solubility, absorption and subsequent bioavailability. Additionally, it addresses the role of polymers in controlling supersaturation, stabilizing drug-rich nanodroplets, and inhibiting recrystallization. Recent advancements and emerging technologies offer new avenues for ASD characterization and production and demonstrate the potential of ASDs to enhance bioavailability and reduce variability, making possible for more effective and patient-friendly pharmaceutical formulations. Future research directions are proposed, focusing on advanced computational models for predicting ASD stability, use of novel polymeric carriers, and methods for successful preparations.
Collapse
Affiliation(s)
- Srividya B
- Solid State Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India
| | - Animesh Ghosh
- Solid State Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, 835215, Jharkhand, India.
| |
Collapse
|
6
|
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
|
7
|
Carnero Canales CS, Marquez Cazorla JI, Marquez Cazorla RM, Roque-Borda CA, Polinário G, Figueroa Banda RA, Sábio RM, Chorilli M, Santos HA, Pavan FR. Breaking barriers: The potential of nanosystems in antituberculosis therapy. Bioact Mater 2024; 39:106-134. [PMID: 38783925 PMCID: PMC11112550 DOI: 10.1016/j.bioactmat.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/17/2024] [Accepted: 05/05/2024] [Indexed: 05/25/2024] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, continues to pose a significant threat to global health. The resilience of TB is amplified by a myriad of physical, biological, and biopharmaceutical barriers that challenge conventional therapeutic approaches. This review navigates the intricate landscape of TB treatment, from the stealth of latent infections and the strength of granuloma formations to the daunting specters of drug resistance and altered gene expression. Amidst these challenges, traditional therapies often fail, contending with inconsistent bioavailability, prolonged treatment regimens, and socioeconomic burdens. Nanoscale Drug Delivery Systems (NDDSs) emerge as a promising beacon, ready to overcome these barriers, offering better drug targeting and improved patient adherence. Through a critical approach, we evaluate a spectrum of nanosystems and their efficacy against MTB both in vitro and in vivo. This review advocates for the intensification of research in NDDSs, heralding their potential to reshape the contours of global TB treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Cesar Augusto Roque-Borda
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Giulia Polinário
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | | | - Rafael Miguel Sábio
- School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, the Netherlands
| | - Marlus Chorilli
- School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| | - Hélder A. Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 AV, the Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Fernando Rogério Pavan
- Tuberculosis Research Laboratory, School of Pharmaceutical Science, Sao Paulo State University (UNESP), Araraquara, 14800-903, Brazil
| |
Collapse
|
8
|
Zhang HJ, Chiang CW, Maschmeyer-Tombs T, Conklin B, Napolitano JG, Lubach JW, Nagapudi K, Mao C, Chen Y. Generality of Enhancing the Dissolution Rates of Free Acid Amorphous Solid Dispersions by the Incorporation of Sodium Hydroxide. Mol Pharm 2024; 21:3395-3406. [PMID: 38836777 DOI: 10.1021/acs.molpharmaceut.4c00118] [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: 06/06/2024]
Abstract
The incorporation of a counterion into an amorphous solid dispersion (ASD) has been proven to be an attractive strategy to improve the drug dissolution rate. In this work, the generality of enhancing the dissolution rates of free acid ASDs by incorporating sodium hydroxide (NaOH) was studied by surface-area-normalized dissolution. A set of diverse drug molecules, two common polymer carriers (copovidone or PVPVA and hydroxypropyl methylcellulose acetate succinate or HPMCAS), and two sample preparation methods (rotary evaporation and spray drying) were investigated. When PVPVA was used as the polymer carrier for the drugs in this study, enhancements of dissolution rates from 7 to 78 times were observed by the incorporation of NaOH into the ASDs at a 1:1 molar ratio with respect to the drug. The drugs having lower amorphous solubilities showed greater enhancement ratios, providing a promising path to improve the drug release performance from their ASDs. Samples generated by rotary evaporation and spray drying demonstrated comparable dissolution rates and enhancements when NaOH was added, establishing a theoretical foundation to bridge the ASD dissolution performance for samples prepared by different solvent-removal processes. In the comparison of polymer carriers, when HPMCAS was applied in the selected system (indomethacin ASD), a dissolution rate enhancement of 2.7 times by the incorporated NaOH was observed, significantly lower than the enhancement of 53 times from the PVPVA-based ASD. This was attributed to the combination of a lower dissolution rate of HPMCAS and the competition for NaOH between IMC and HPMCAS. By studying the generality of enhancing ASD dissolution rates by the incorporation of counterions, this study provides valuable insights into further improving drug release from ASD formulations of poorly water-soluble drugs.
Collapse
Affiliation(s)
- Helen J Zhang
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
- Department of Molecular and Cell Biology, University of California, Berkeley, 142 Weill Hall #3200, Berkeley, California 94720, United States
| | - Cheng W Chiang
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Tristan Maschmeyer-Tombs
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Breanna Conklin
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jose G Napolitano
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Joseph W Lubach
- Small Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Karthik Nagapudi
- 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
|
9
|
Di R, Bansal KK, Rosenholm JM, Grohganz H, Rades T. Utilizing the allyl-terminated copolymer methoxy(poly(ethylene glycol))-block-poly(jasmine lactone) in the development of amorphous solid dispersions: A comparative study of functionalized and non-functionalized polymer. Int J Pharm 2024; 657:124175. [PMID: 38685442 DOI: 10.1016/j.ijpharm.2024.124175] [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: 03/18/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Molecular interactions are crucial to stabilize amorphous drugs in amorphous solid dispersions (ASDs). Most polymers, however, have only a limited ability to form strong molecular interactions with drugs. Polymers tailored to fit the physicochemical properties of the drug molecule to be incorporated, for instance by allowing the incorporation of specific functional groups, would be highly sought-for in this regard. For this purpose, the novel allyl-terminated polymer methoxy(polyethylene glycol)-block-poly(jasmine lactone) (mPEG-b-PJL) has been synthesized and functionalized to potentially enhance specific drug-polymer interactions. This study investigated the use of mPEG-b-PJL in ASDs, using carvedilol (CAR), a weakly basic model drug. The findings revealed that the acidic functionalized form of the polymer (mPEG-b-PJL-COOH) indeed established stronger molecular interactions with CAR compared to its non-functionalized counterpart mPEG-b-PJL. Evaluations on polymer effectiveness in forming ASDs demonstrated that mPEG-b-PJL-COOH outperformed its non-functionalized counterpart in miscibility, drug loading ability, and stability, inferred from reduced molecular mobility. However, dissolution tests indicated that ASDs with mPEG-b-PJL-COOH did not significantly improve the dissolution behaviour compared to amorphous CAR alone, despite potential solubility enhancement through micelle formation. Overall, this study confirms the potential of functionalized polymers in ASD formulations, while the challenge of improving dissolution performance in these ASDs remains an area of further development.
Collapse
Affiliation(s)
- Rong Di
- University of Copenhagen, Faculty of Health and Medical Sciences, Department of Pharmacy, Copenhagen, Denmark.
| | - Kuldeep K Bansal
- Åbo Akademi University, Faculty of Science and Engineering, Pharmaceutical Sciences Laboratory, Turku, Finland.
| | - Jessica M Rosenholm
- Åbo Akademi University, Faculty of Science and Engineering, Pharmaceutical Sciences Laboratory, Turku, Finland.
| | - Holger Grohganz
- University of Copenhagen, Faculty of Health and Medical Sciences, Department of Pharmacy, Copenhagen, Denmark.
| | - Thomas Rades
- University of Copenhagen, Faculty of Health and Medical Sciences, Department of Pharmacy, Copenhagen, Denmark.
| |
Collapse
|
10
|
Liu Y, Wu Z, Chen Y, Guan Y, Guo H, Yang M, Yue P. Rubusoside As a Multifunctional Stabilizer for Novel Nanocrystal-Based Solid Dispersions with a High Drug Loading: A Case Study. J Pharm Sci 2024; 113:699-710. [PMID: 37659720 DOI: 10.1016/j.xphs.2023.08.024] [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: 03/23/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/04/2023]
Abstract
The oral bioavailability of poorly soluble drugs has always been the focus of pharmaceutical researchers. We innovatively combined nanocrystal technology and solid dispersion technology to prepare novel nanocrystalline solid dispersions (NCSDs), which enable both the solidification and redispersion of nanocrystals, offering a promising new pathway for oral delivery of insoluble Chinese medicine ingredients. The rubusoside (Rub) was first used as the multifunctional stabilizer of novel apigenin nanocrystal-based solid dispersions (AP-NSD), improving the in vitro solubilization rate of the insoluble drug apigenin(AP). AP-NSD has been produced using a combination of homogenisation and spray-drying technology. The effects of stabilizer type and concentration on AP nanosuspensions (AP-NS) particles, span, and zeta potential were studied. And the effects of different types of protective agents on the yield and redispersibility of AP-NSD were also studied. Furthermore, AP-NSD was characterized by infrared spectroscopy (IR), scanning electron microscopy (SEM), transmission electron microscopy (TEM), differential scanning calorimetry (DSC), and powder X-ray diffraction (PXRD). Solubility was used to assess the in vitro dissolution of AP-NSD relative to APIs and amorphous solid dispersions (AP-ASD), and AP-ASD was prepared by the solvent method. The results showed that 20% Rub stabilized AP-NSD exhibited high drug-loading and good redispersibility and stability, and higher in vitro dissolution rate, which may be related to the presence of Rub on surface of drug. Therefore provides a natural and safe option for the development of formulations for insoluble drugs.
Collapse
Affiliation(s)
- Yang Liu
- Key Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang 330004, China
| | - Zhenfeng Wu
- Key Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang 330004, China
| | - Yingchong Chen
- Key Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang 330004, China
| | - Yongmei Guan
- Key Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang 330004, China
| | - Huiwen Guo
- Key Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang 330004, China
| | - Ming Yang
- Key Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang 330004, China
| | - Pengfei Yue
- Key Lab of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Chinese Medicine, 1688 Meiling Avenue, Nanchang 330004, China.
| |
Collapse
|
11
|
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
|
12
|
Caggiano N, Armstrong MS, Georgiou JS, Rawal A, Wilson BK, White CE, Priestley RD, Prud’homme RK. Formulation and Scale-up of Delamanid Nanoparticles via Emulsification for Oral Tuberculosis Treatment. Mol Pharm 2023; 20:4546-4558. [PMID: 37578286 PMCID: PMC10481377 DOI: 10.1021/acs.molpharmaceut.3c00240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023]
Abstract
Delamanid (DLM) is a hydrophobic small molecule therapeutic used to treat drug-resistant tuberculosis (DR-TB). Due to its hydrophobicity and resulting poor aqueous solubility, formulation strategies such as amorphous solid dispersions (ASDs) have been investigated to enhance its aqueous dissolution kinetics and thereby improve oral bioavailability. However, ASD formulations are susceptible to temperature- and humidity-induced phase separation and recrystallization under harsh storage conditions typically encountered in areas with high tuberculosis incidence. Nanoencapsulation represents an alternative formulation strategy to increase aqueous dissolution kinetics while remaining stable at elevated temperature and humidity. The stabilizer layer coating the nanoparticle drug core limits the formation of large drug domains by diffusion during storage, representing an advantage over ASDs. Initial attempts to form DLM-loaded nanoparticles via precipitation-driven self-assembly were unsuccessful, as the trifluoromethyl and nitro functional groups present on DLM were thought to interfere with surface stabilizer attachment. Therefore, in this work, we investigated the nanoencapsulation of DLM via emulsification, avoiding the formation of a solid drug core and instead keeping DLM dissolved in a dichloromethane dispersed phase during nanoparticle formation. Initial emulsion formulation screening by probe-tip ultrasonication revealed that a 1:1 mass ratio of lecithin and HPMC stabilizers formed 250 nm size-stable emulsion droplets with 40% DLM loading. Scale-up studies were performed to produce nearly identical droplet size distribution at larger scale using high-pressure homogenization, a continuous and industrially scalable technique. The resulting emulsions were spray-dried to form a dried powder, and in vitro dissolution studies showed dramatically enhanced dissolution kinetics compared to both as-received crystalline DLM and micronized crystalline DLM, owing to the increased specific surface area and partially amorphous character of the DLM-loaded nanoparticles. Solid-state NMR and dissolution studies showed good physical stability of the emulsion powders during accelerated stability testing (50 °C/75% RH, open vial).
Collapse
Affiliation(s)
- Nicholas
J. Caggiano
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Madeleine S. Armstrong
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Joanna S. Georgiou
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Aditya Rawal
- Mark
Wainwright Analytical Centre, University
of New South Wales, Sydney, NSW 2032, Australia
| | - Brian K. Wilson
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| | - Claire E. White
- Department
of Civil and Environmental Engineering, Princeton University, Princeton, New Jersey 08544, United States
- Andlinger
Center for Energy and the Environment, Princeton
University, Princeton, New Jersey 08544, United States
| | - Rodney D. Priestley
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
- Princeton
Materials Institute, Princeton University, Princeton, New Jersey 08544, United States
| | - Robert K. Prud’homme
- Department
of Chemical and Biological Engineering, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
13
|
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
|
14
|
Van Duong T, Diab S, Hodnett NS, Taylor LS. Kinetic Barriers to Disproportionation of Salts of Weakly Basic Drugs. Mol Pharm 2023; 20:3886-3894. [PMID: 37494545 DOI: 10.1021/acs.molpharmaceut.2c01034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Disproportionation is a major issue in formulations containing salts of weakly basic drugs. Despite considerable interest in risk assessment approaches for disproportionation, the prediction of salt-to-base conversion remains challenging. Recent studies have highlighted several confounding factors other than pHmax that appear to play an important role in salt disproportionation and have suggested that kinetic barriers need to be considered in addition to the thermodynamic driving force when assessing the risk of a salt to undergo conversion to parent free base. Herein, we describe the concurrent application of in situ Raman spectroscopy and pH monitoring to investigate the disproportionation kinetics of three model salts, pioglitazone hydrochloride, sorafenib tosylate, and atazanavir sulfate, in aqueous slurries. We found that even for favorable thermodynamic conditions (i.e., pH ≫ pHmax), disproportionation kinetics of the salts were very different despite each system having a similar pHmax. The importance of free base nucleation kinetics was highlighted by the observation that the disproportionation conversion time in the slurries showed the same trend as the free base nucleation induction time. Pioglitazone hydrochloride, with a free base induction time of <1 min, rapidly converted to the free base in slurry experiments. In contrast, atazanavir sulfate, where the free base induction time was much longer, took several hours to undergo disproportionation in the slurry for pH ≫ pHmax. Additionally, we altered an established thermodynamically based modeling framework to account for kinetic effects (representing the nucleation kinetic barrier) to estimate the solid-state stability of salt formulations. In conclusion, a solution-based thermodynamic model is mechanistically appropriate to predict salt disproportionation in a solid-state formulation, when kinetic barriers are also taken into consideration.
Collapse
Affiliation(s)
- Tu Van Duong
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana47907, United States
| | - Samir Diab
- GlaxoSmithKline, Park Road, Ware, SG12 0DP, U.K
| | - Neil S Hodnett
- GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, U.K
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, Indiana47907, United States
| |
Collapse
|
15
|
Manghnani PN, Schenck L, Khan SA, Doyle PS. Templated Reactive Crystallization of Active Pharmaceutical Ingredient in Hydrogel Microparticles Enabling Robust Drug Product Processing. J Pharm Sci 2023; 112:2115-2123. [PMID: 37160228 DOI: 10.1016/j.xphs.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/04/2023] [Accepted: 05/04/2023] [Indexed: 05/11/2023]
Abstract
Commercialization of most promising active pharmaceutical ingredients (APIs) is impeded either by poor bioavailability or challenging physical properties leading to costly manufacture. Bioavailability of ionizable hydrophobic APIs can be enhanced by its conversion to salt form. While salt form of the API presents higher solution concentration than the non-ionized form, poor physical properties resulting from particle anisotropy or non-ideal morphology (needles) and particle size distribution not meeting dissolution rate targets can still inhibit its commercial translation. In this regard, API physical properties can be improved through addition of non-active components (excipients or carriers) during API manufacture. In this work, a facile method to perform reactive crystallization of an API salt in presence of the microporous environment of a hydrogel microparticle is presented. Specifically, the reaction between acidic antiretroviral API, raltegravir and base potassium hydroxide is performed in the presence of polyethylene glycol diacrylamide hydrogel microparticles. In this bottom-up approach, the spherical template hydrogel microparticles for the reaction lead to monodisperse composites loaded with inherently micronized raltegravir-potassium crystals, thus improving API physical properties without hampering bioavailability. Overall, this technique provides a novel approach to reactive crystallization while maintaining the API polymorph and crystallinity.
Collapse
Affiliation(s)
- Purnima N Manghnani
- Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, #04-13/14 Enterprise Wing 138602, Singapore
| | - Luke Schenck
- Process Research and Development, Merck & Co., Inc., 126 E. Lincoln Ave Rahway NJ 07065, USA
| | - Saif A Khan
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore; Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, #04-13/14 Enterprise Wing 138602, Singapore.
| | - Patrick S Doyle
- Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, #04-13/14 Enterprise Wing 138602, Singapore; Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue Room E17-504F, Cambridge, MA, 02139 USA; Harvard Medical School Initiative for RNA Medicine, Boston, MA, 02115 USA.
| |
Collapse
|
16
|
Thuy Nguyen H, Van Duong T, Taylor YS. Enteric coating of tablets containing an amorphous solid dispersion of an enteric polymer and a weakly basic drug: a strategy to enhance in vitro release. Int J Pharm 2023:123139. [PMID: 37311499 PMCID: PMC10390825 DOI: 10.1016/j.ijpharm.2023.123139] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/15/2023]
Abstract
Recent work has highlighted that amorphous solid dispersions (ASDs) containing delamanid (DLM) and an enteric polymer, hypromellose phthalate (HPMCP), appear to be susceptible to crystallization during immersion in simulated gastric fluids. The goal of this study was to minimize contact of the ASD particles with the acidic media via application of an enteric coating to tablets containing the ASD intermediate, and improve the subsequent drug release at higher pH conditions. DLM ASDs were prepared with HPMCP and formulated into a tablet that was then coated with a methacrylic acid copolymer (Acryl EZE II®). Drug release was studied in vitro using a two-stage dissolution test where the pH of the gastric compartment was altered to reflect physiological variations. The medium was subsequently switched to simulated intestinal fluid. The gastric resistance time of the enteric coating was probed over the pH range of 1.6-5.0. The enteric coating was found to be effective at protecting the drug against crystallization in pH conditions where HPMCP was insoluble. Consequently, the variability in drug release following gastric immersion under pH conditions reflecting different prandial states was notably reduced when compared to the reference product. These findings support closer examination of the potential for drug crystallization from ASDs in the gastric environment where acid-insoluble polymers may be less effective as crystallization inhibitors. Further, addition of a protective enteric coating appears to provide a promising remediation strategy to prevent crystallization at low pH environments, and may mitigate variability associated with prandial state that arises due to pH changes.
Collapse
Affiliation(s)
- Hanh Thuy Nguyen
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tu Van Duong
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States.
| |
Collapse
|
17
|
Nguyen HT, Van Duong T, Jaw-Tsai S, Bruning-Barry R, Pande P, Taneja R, Taylor LS. Fed- and Fasted-State Performance of Pretomanid Amorphous Solid Dispersions Formulated with an Enteric Polymer. Mol Pharm 2023. [PMID: 37220082 DOI: 10.1021/acs.molpharmaceut.3c00174] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Weakly acid polymers with pH-responsive solubility are being used with increasing frequency in amorphous solid dispersion (ASD) formulations of drugs with low aqueous solubility. However, drug release and crystallization in a pH environment where the polymer is insoluble are not well understood. The aim of the current study was to develop ASD formulations optimized for release and supersaturation longevity of a rapidly crystallizing drug, pretomanid (PTM), and to evaluate a subset of these formulations in vivo. Following screening of several polymers for their ability to inhibit crystallization, hypromellose acetate succinate HF grade (HPMCAS-HF; HF) was selected to prepare PTM ASDs. In vitro release studies were conducted in simulated fasted- and fed-state media. Drug crystallization in ASDs following exposure to dissolution media was evaluated by powder X-ray diffraction, scanning electron microscopy, and polarized light microscopy. In vivo oral pharmacokinetic evaluation was conducted in male cynomolgus monkeys (n = 4) given 30 mg PTM under both fasted and fed conditions in a crossover design. Three HPMCAS-based ASDs of PTM were selected for fasted-state animal studies based on their in vitro release performance. Enhanced bioavailability was observed for each of these formulations relative to the reference product that contained crystalline drug. The 20% drug loading PTM-HF ASD gave the best performance in the fasted state, with subsequent dosing in the fed state. Interestingly, while food improved drug absorption of the crystalline reference product, the exposure of the ASD formulation was negatively impacted. The failure of the HPMCAS-HF ASD to enhance absorption in the fed state was hypothesized to result from poor release in the reduced pH intestinal environment resulting from the fed state. In vitro experiments confirmed a reduced release rate under lower pH conditions, which was attributed to reduced polymer solubility and an enhanced crystallization tendency of the drug. These findings emphasize the limitations of in vitro assessment of ASD performance using standardized media conditions. Future studies are needed for improved understanding of food effects on ASD release and how this variability can be captured by in vitro testing methodologies for better prediction of in vivo outcomes, in particular for ASDs formulated with enteric polymers.
Collapse
Affiliation(s)
- Hanh Thuy Nguyen
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tu Van Duong
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Sarah Jaw-Tsai
- Sarah Jaw-Tsai Consulting Services, 12279 Skyracer Drive, Las Vegas, Nevada 89138, United States
| | - Rebecca Bruning-Barry
- Global Health Technologies Program, RTI International, Research Triangle Park, North Carolina 27704, United States
| | - Poonam Pande
- Global Alliance for TB Drug Development (TB Alliance), 80 Pine Street, 20th Floor, New York, New York 10005, United States
| | - Rajneesh Taneja
- Global Alliance for TB Drug Development (TB Alliance), 80 Pine Street, 20th Floor, New York, New York 10005, United States
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
18
|
Nguyen HT, Van Duong T, Taylor LS. Impact of Gastric pH Variations on the Release of Amorphous Solid Dispersion Formulations Containing a Weakly Basic Drug and Enteric Polymers. Mol Pharm 2023; 20:1681-1695. [PMID: 36730186 PMCID: PMC9997068 DOI: 10.1021/acs.molpharmaceut.2c00895] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Enteric polymers are widely used in amorphous solid dispersion (ASD) formulations. The aim of the current study was to explore ASD failure mechanisms across a wide range of pH conditions that mimic in vivo gastric compartment variations where enteric polymers such as hydroxypropyl methylcellulose phthalate (HPMCP) and hydroxypropyl methylcellulose acetate succinate (HPMCAS) are largely insoluble. Delamanid (DLM), a weakly basic drug used to treat tuberculosis, was selected as the model compound. Both DLM free base and the edisylate salt were formulated with HPMCP, while DLM edisylate ASDs were also prepared with different grades of HPMCAS. Two-stage release testing was conducted with the gastric stage pH varied between pH 1.6 and 5.0, prior to transfer to intestinal conditions of pH 6.5. ASD particles were collected following suspension in the gastric compartment and evaluated using X-ray powder diffraction and scanning electron microscopy. Additional samples were also evaluated with polarized light microscopy. In general, ASDs with HPMCP showed improved overall release for all testing conditions, relative to ASDs with HPMCAS. ASDs with the edisylate salt likewise outperformed those with DLM free base. Impaired release for certain formulations at intestinal pH conditions was attributed to surface drug crystallization that initiated during suspension in the gastric compartment where the polymer is insoluble; crystallization appeared more extensive for HPMCAS ASDs. These findings suggest that gastric pH variations should be evaluated for ASD formulations containing weakly basic drugs and enteric polymers.
Collapse
Affiliation(s)
- Hanh Thuy Nguyen
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tu Van Duong
- 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
|
19
|
Dimiou S, McCabe J, Booth R, Booth J, Nidadavole K, Svensson O, Sparén A, Lindfors L, Paraskevopoulou V, Mead H, Coates L, Workman D, Martin D, Treacher K, Puri S, Taylor LS, Yang B. Selecting Counterions to Improve Ionized Hydrophilic Drug Encapsulation in Polymeric Nanoparticles. Mol Pharm 2023; 20:1138-1155. [PMID: 36653946 DOI: 10.1021/acs.molpharmaceut.2c00855] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Hydrophobic ion pairing (HIP) can successfully increase the drug loading and control the release kinetics of ionizable hydrophilic drugs, addressing challenges that prevent these molecules from reaching the clinic. Nevertheless, polymeric nanoparticle (PNP) formulation development requires trial-and-error experimentation to meet the target product profile, which is laborious and costly. Herein, we design a preformulation framework (solid-state screening, computational approach, and solubility in PNP-forming emulsion) to understand counterion-drug-polymer interactions and accelerate the PNP formulation development for HIP systems. The HIP interactions between a small hydrophilic molecule, AZD2811, and counterions with different molecular structures were investigated. Cyclic counterions formed amorphous ion pairs with AZD2811; the 0.7 pamoic acid/1.0 AZD2811 complex had the highest glass transition temperature (Tg; 162 °C) and the greatest drug loading (22%) and remained as phase-separated amorphous nanosized domains inside the polymer matrix. Palmitic acid (linear counterion) showed negligible interactions with AZD2811 (crystalline-free drug/counterion forms), leading to a significantly lower drug loading despite having similar log P and pKa with pamoic acid. Computational calculations illustrated that cyclic counterions interact more strongly with AZD2811 than linear counterions through dispersive interactions (offset π-π interactions). Solubility data indicated that the pamoic acid/AZD2811 complex has a lower organic phase solubility than AZD2811-free base; hence, it may be expected to precipitate more rapidly in the nanodroplets, thus increasing drug loading. Our work provides a generalizable preformulation framework, complementing traditional performance-indicating parameters, to identify optimal counterions rapidly and accelerate the development of hydrophilic drug PNP formulations while achieving high drug loading without laborious trial-and-error experimentation.
Collapse
Affiliation(s)
- Savvas Dimiou
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D AstraZeneca, Granta Park, CambridgeCB21 6GH, U.K
- UCL School of Pharmacy, 29-39 Brunswick Square, LondonWC1N 1AX, U.K
| | - James McCabe
- Early Product Development, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Rebecca Booth
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Jonathan Booth
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Kalyan Nidadavole
- Early Product Development, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Olof Svensson
- Pharmaceutical Technology & Development, Operations, AstraZeneca, GothenburgSE-43183, Sweden
| | - Anders Sparén
- Pharmaceutical Technology & Development, Operations, AstraZeneca, GothenburgSE-43183, Sweden
| | - Lennart Lindfors
- Advanced Drug Delivery, Pharmaceutical Science, R&D AstraZeneca, GothenburgSE-43183, Sweden
| | - Vasiliki Paraskevopoulou
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Heather Mead
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Lydia Coates
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - David Workman
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D AstraZeneca, Granta Park, CambridgeCB21 6GH, U.K
| | - Dave Martin
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Kevin Treacher
- New Modalities and Parenteral Development, Pharmaceutical Technology & Development, Operations, AstraZeneca, MacclesfieldSK10 2NA, U.K
| | - Sanyogitta Puri
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D AstraZeneca, Granta Park, CambridgeCB21 6GH, U.K
| | - Lynne S Taylor
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana47907, United States
| | - Bin Yang
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D AstraZeneca, Granta Park, CambridgeCB21 6GH, U.K
| |
Collapse
|
20
|
Patil SM, Barji DS, Chavan T, Patel K, Collazo AJ, Prithipaul V, Muth A, Kunda NK. Solubility Enhancement and Inhalation Delivery of Cyclodextrin-Based Inclusion Complex of Delamanid for Pulmonary Tuberculosis Treatment. AAPS PharmSciTech 2023; 24:49. [PMID: 36702977 DOI: 10.1208/s12249-023-02510-1] [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: 08/09/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
Tuberculosis (TB) is a contiguous airborne disease caused by Mycobacterium tuberculosis (M.tb), primarily affecting the human lungs. The progression of drug-susceptible TB to drug-resistant strains, MDR-TB and XDR-TB, has become a global challenge toward eradicating TB. Conventional TB treatment involves frequent dosing and prolonged treatment regimens predominantly by an oral or invasive route, leading to treatment-related systemic adverse effects and patient's noncompliance. Pulmonary delivery is an attractive option as we could reduce dose, limit systemic side-effects, and achieve rapid onset of action. Delamanid (DLD), an antituberculosis drug, has poor aqueous solubility, and in this study, we aim to improve its solubility using cyclodextrin complexation. We screened different cyclodextrins and found that HP-β-CD resulted in a 54-fold increase in solubility compared to a 27-fold and 13-fold increase by SBE-β-CD and HP-ɣ-CD, respectively. The stability constant (265 ± 15 M-1) and complexation efficiency (8.5 × 10-4) suggest the formation of a stable inclusion complex of DLD and HP-β-CD in a 2:1 ratio. Solid-state characterization studies (DSC, PXRD, and NMR) further confirmed successful complexation of DLD in HP-β-CD. The nebulized DLD-CD complex solution showed a mass median aerodynamic diameter of 4.42 ± 0.62 μm and fine particle fraction of 82.28 ± 2.79%, suggesting deposition in the respiratory airways. In bacterial studies, minimum inhibitory concentration of DLD-CD complex was significantly reduced (four-fold) compared to free DLD in M.tb (H37Ra strain). Furthermore, accelerated stability studies confirmed that the inclusion complex was stable for 4 weeks with 90%w/w drug content. In conclusion, we increased the aqueous solubility of DLD through cyclodextrin complexation and improved its efficacy in vitro.
Collapse
Affiliation(s)
- Suyash M Patil
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, New York, 11439, USA
| | - Druva Sarika Barji
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, New York, 11439, USA
| | - Tejashri Chavan
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, New York, 11439, USA
| | - Kinjal Patel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, New York, 11439, USA
| | - Andrew J Collazo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, New York, 11439, USA
| | - Vasudha Prithipaul
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, New York, 11439, USA
| | - Aaron Muth
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, New York, 11439, USA
| | - Nitesh K Kunda
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, New York, 11439, USA.
| |
Collapse
|
21
|
Moseson DE, Benson EG, Cao Z, Bhalla S, Wang F, Wang M, Zheng K, Narwankar PK, Simpson GJ, Taylor LS. Impact of Aluminum Oxide Nanocoating on Drug Release from Amorphous Solid Dispersion Particles. Mol Pharm 2023; 20:593-605. [PMID: 36346665 DOI: 10.1021/acs.molpharmaceut.2c00818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Atomic layer coating (ALC) is emerging as a particle engineering strategy to inhibit surface crystallization of amorphous solid dispersions (ASDs). In this study, we turn our attention to evaluating drug release behavior from ALC-coated ASDs, and begin to develop a mechanistic framework. Posaconazole/hydroxypropyl methylcellulose acetate succinate was used as a model system at both 25% and 50% drug loadings. ALC-coatings of aluminum oxide up to 40 nm were evaluated for water sorption kinetics and dissolution performance under a range of pH conditions. Scanning electron microscopy with energy dispersive X-ray analysis was used to investigate the microstructure of partially released ASD particles. Coating thickness and defect density (inferred from deposition rates) were found to impact water sorption kinetics. Despite reduced water sorption kinetics, the presence of a coating was not found to impact dissolution rates under conditions where rapid drug release was observed. Under slower releasing conditions, underlying matrix crystallization was reduced by the coating, enabling greater levels of drug release. These results demonstrate that water was able to penetrate through the ALC coating, hydrating the amorphous solid, which can initiate dissolution of drug and/or polymer (depending on pH conditions). Swelling of the ASD substrate subsequently occurs, disrupting and cracking the coating, which serves to facilitate rapid drug release. Water sorption kinetics are highlighted as a potential predictive tool to investigate the coating quality and its potential impact on dissolution performance. This study has implications for formulation design and evaluation of ALC-coated ASD particles.
Collapse
Affiliation(s)
- Dana E Moseson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Emily G Benson
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ziyi Cao
- Department of Chemistry, College of Science, Purdue University, West Lafayette, Indiana 47907, United States
| | - Shradha Bhalla
- Department of Industrial and Physical Pharmacy, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| | - Fei Wang
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Miaojun Wang
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Kai Zheng
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Pravin K Narwankar
- Applied Materials, Inc., 3100 Bowers Ave, Santa Clara, California 95054, United States
| | - Garth J Simpson
- Department of Chemistry, College of Science, 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
|
22
|
Recent Advances in Amorphous Solid Dispersions: Preformulation, Formulation Strategies, Technological Advancements and Characterization. Pharmaceutics 2022; 14:pharmaceutics14102203. [PMID: 36297638 PMCID: PMC9609913 DOI: 10.3390/pharmaceutics14102203] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/27/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022] Open
Abstract
Amorphous solid dispersions (ASDs) are among the most popular and widely studied solubility enhancement techniques. Since their inception in the early 1960s, the formulation development of ASDs has undergone tremendous progress. For instance, the method of preparing ASDs evolved from solvent-based approaches to solvent-free methods such as hot melt extrusion and Kinetisol®. The formulation approaches have advanced from employing a single polymeric carrier to multiple carriers with plasticizers to improve the stability and performance of ASDs. Major excipient manufacturers recognized the potential of ASDs and began introducing specialty excipients ideal for formulating ASDs. In addition to traditional techniques such as differential scanning calorimeter (DSC) and X-ray crystallography, recent innovations such as nano-tomography, transmission electron microscopy (TEM), atomic force microscopy (AFM), and X-ray microscopy support a better understanding of the microstructure of ASDs. The purpose of this review is to highlight the recent advancements in the field of ASDs with respect to formulation approaches, methods of preparation, and advanced characterization techniques.
Collapse
|
23
|
Bhanushali JS, Dhiman S, Nandi U, Bharate SS. Molecular interactions of niclosamide with hydroxyethyl cellulose in binary and ternary amorphous solid dispersions for synergistic enhancement of water solubility and oral pharmacokinetics in rats. Int J Pharm 2022; 626:122144. [PMID: 36029996 DOI: 10.1016/j.ijpharm.2022.122144] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/10/2022] [Accepted: 08/22/2022] [Indexed: 12/14/2022]
Abstract
The cellulose-based polymers are extensively employed in oral formulations for addressing ADMET issues of API. Herein, we report the synergistic effect of hydroxyethyl cellulose in solubility/dissolution enhancement of BCS class II, anthelmintic drug niclosamide. The low solubility and poor oral bioavailability are the primary reasons for its high daily dose. The amorphous solid dispersions (ASDs) developed herein demonstrated reproducible solubility and dissolution enhancement in smaller-to-pilot batches. The significant boost in niclosamide solubility in HEC-based binary SD was rationalized as a result of intermolecular H-bonding as indicated by in-silico studies and further supported by characterization data. HEC is plausibly inhibiting the precipitation of drug and thereby enabling high dissolution and permeation across the membrane. The comparative oral pharmacokinetics in Wistar rats at 25 mg/kg provided 4.4-fold higher plasma exposure of niclosamide in SD formulation SB-ASD-N2 over the plain drug. The results presented herein warrant validation of this ASD under clinical settings. Teaser Amorphous solid dispersions of niclosamide.
Collapse
Affiliation(s)
- Jigar S Bhanushali
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - Sumit Dhiman
- PK-PD Toxicology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Utpal Nandi
- PK-PD Toxicology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Sonali S Bharate
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India.
| |
Collapse
|