1
|
Yi H, Yu H, Wang L, Wang Y, Ouyang C, Keshta BE. Microneedle transdermal drug delivery as a candidate for the treatment of gouty arthritis: Material structure, design strategies and prospects. Acta Biomater 2024; 187:20-50. [PMID: 39182801 DOI: 10.1016/j.actbio.2024.08.032] [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: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Gouty arthritis (GA) is caused by monosodium urate (MSU) crystals deposition. GA is difficult to cure because of its complex disease mechanism and the tendency to reoccur. GA patients require long-term uric acid-lowering and anti-inflammatory treatments. In the past ten years, as a painless, convenient and well-tolerated new drug transdermal delivery method, microneedles (MNs) administration has been continuously developed, which can realize various drug release modes to deal with various complex diseases. Compared with the traditional administration methods (oral and injection), MNs are more conducive to the long-term independent treatment of GA patients because of their safe, efficient and controllable drug delivery ability. In this review, the pathological mechanism of GA and common therapeutic drugs for GA are summarized. After that, MNs drug delivery mechanisms were summarized: dissolution release mechanism, swelling release mechanism and channel-assisted release mechanism. According to drug delivery patterns of MNs, the mechanisms and applications of rapid-release MNs, long-acting MNs, intelligent-release MNs and multiple-release MNs were reviewed. Additionally, existing problems and future trends of MNs in the treatment of GA were also discussed. STATEMENT OF SIGNIFICANCE: Gout is an arthritis caused by metabolic disease "hyperuricemia". Epidemiological studies show that the number of gouty patients is increasing rapidly worldwide. Due to the complex disease mechanism and recurrent nature of gout, gouty patients require long-term therapy. However, traditional drug delivery modes (oral and injectable) have poor adherence, low drug utilization, and lack of local localized targeting. They may lead to adverse effects such as rashes and gastrointestinal reactions. As a painless, convenient and well-tolerated new drug transdermal delivery method, microneedles have been continuously developed, which can realize various drug release modes to deal with gouty arthritis. In this review, the material structure, design strategy and future outlook of microneedles for treating gouty arthritis will be reviewed.
Collapse
Affiliation(s)
- Hong Yi
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Haojie Yu
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China; Zhejiang-Russia Joint Laboratory of Photo-Electron-Megnetic Functional Materials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China.
| | - Li Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China; Zhejiang-Russia Joint Laboratory of Photo-Electron-Megnetic Functional Materials, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China.
| | - Yu Wang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Chenguang Ouyang
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| | - Basem E Keshta
- State Key Laboratory of Chemical Engineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, PR China
| |
Collapse
|
2
|
Tang B, Xie X, Lu J, Huang W, Yang J, Tian J, Lei L. Designing biomaterials for the treatment of autoimmune diseases. APPLIED MATERIALS TODAY 2024; 39:102278. [DOI: 10.1016/j.apmt.2024.102278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
|
3
|
Jain KMH, Ho T, Hoe S, Wan B, Muthal A, Subramanian R, Foti C. Accelerated and Biopredictive In Vitro Release Testing Strategy for Single Agent and Combination Long-Acting Injectables. J Pharm Sci 2024; 113:1885-1897. [PMID: 38369022 DOI: 10.1016/j.xphs.2024.02.013] [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: 11/09/2023] [Revised: 02/12/2024] [Accepted: 02/12/2024] [Indexed: 02/20/2024]
Abstract
The purpose of this study was to develop an in vitro release testing (IVRT) strategy to predict the pre-clinical performance of single agent and combination long acting injectable (LAI) suspension products. Two accelerated IVRT methods were developed using USP apparatus 2 to characterize initial, intermediate, and terminal phases of drug release. Initial and intermediate phases were captured using a suspension cup with moderate agitation to ensure a constant, low surface area exposure of the LAI suspension to the release media. The terminal phase was obtained by exposing the LAI suspension to a high initial paddle speed. This resulted in smaller suspension particulates with high cumulative surface area that were dispersed throughout the release media, enabling rapid drug release. The in vitro release profiles obtained with these two methods in 48 h or less were independently time scaled to reflect the in vivo time scale of approximately 1800 h. Level-A in vitro in vivo correlations (IVIVCs) were separately developed for each method and active pharmaceutical ingredient (API) using in vivo absorption profiles obtained by deconvolution of rat plasma concentration-time profiles. The IVIVCs were successfully validated for each API. This work provides a framework for evaluating individual phases of drug release of complex LAIs to ultimately predict their in vivo performance.
Collapse
Affiliation(s)
- Krutika Meena Harish Jain
- Analytical Development and Operations, Gilead Sciences, 355 Lakeside Drive, Foster City, CA 94404, USA.
| | - Tien Ho
- Analytical Development and Operations, Gilead Sciences, 355 Lakeside Drive, Foster City, CA 94404, USA
| | - Susan Hoe
- Formulation and Process Development, Gilead Sciences, Foster City, CA 94404, USA
| | - Bo Wan
- Analytical Development and Operations, Gilead Sciences, 355 Lakeside Drive, Foster City, CA 94404, USA
| | - Anumeha Muthal
- Analytical Development and Operations, Gilead Sciences, 355 Lakeside Drive, Foster City, CA 94404, USA
| | - Raju Subramanian
- Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, CA 94404, USA
| | - Chris Foti
- Analytical Development and Operations, Gilead Sciences, 355 Lakeside Drive, Foster City, CA 94404, USA
| |
Collapse
|
4
|
Torres-Terán I, Venczel M, Klein S. Prediction of subcutaneous drug absorption - Development of novel simulated interstitial fluid media for predictive subcutaneous in vitro assays. Int J Pharm 2024; 658:124227. [PMID: 38750979 DOI: 10.1016/j.ijpharm.2024.124227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/10/2024] [Accepted: 05/11/2024] [Indexed: 05/20/2024]
Abstract
Media that mimic physiological fluids at the site of administration have proven to be valuable in vitro tools for predicting in vivo drug release, particularly for routes of administration where animal studies cannot accurately predict human performance. The objective of the present study was to develop simulated interstitial fluids (SISFs) that mimic the major components and physicochemical properties of subcutaneous interstitial fluids (ISFs) from preclinical species and humans, but that can be easily prepared in the laboratory and used in in vitro experiments to estimate in vivo drug release and absorption of subcutaneously administered formulations. Based on data from a previous characterization study of ISFs from different species, two media were developed: a simulated mouse-rat ISF and a simulated human-monkey ISF. The novel SISFs were used in initial in vitro diffusion studies with a commercial injectable preparation of liraglutide. Although the in vitro model used for this purpose still requires significant refinement, these two new media will undoubtedly contribute to a better understanding of the in vivo performance of subcutaneous injectables in different species and will help to reduce the number of unnecessary in vivo experiments in preclinical species by implementation in predictive in vitro models.
Collapse
Affiliation(s)
- Iria Torres-Terán
- University of Greifswald. Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, 3 Felix Hausdorff Street, 17489 Greifswald, Germany; Sanofi-Aventis Deutschland GmbH, R&D, Global CMC Development, Synthetics Platform. Industriepark Hoechst, H770, D-65926 Frankfurt am Main, Germany
| | - Márta Venczel
- University of Greifswald. Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, 3 Felix Hausdorff Street, 17489 Greifswald, Germany
| | - Sandra Klein
- Sanofi-Aventis Deutschland GmbH, R&D, Global CMC Development, Synthetics Platform. Industriepark Hoechst, H770, D-65926 Frankfurt am Main, Germany.
| |
Collapse
|
5
|
Block M, Sieger P, Truenkle C, Saal C, Simon R, Truebenbach I. Miniaturized screening and performance prediction of tailored subcutaneous extended-release formulations for preclinical in vivo studies. Eur J Pharm Sci 2024; 196:106733. [PMID: 38408709 DOI: 10.1016/j.ejps.2024.106733] [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: 12/04/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 02/28/2024]
Abstract
Microencapsulation of active pharmaceutical ingredients (APIs) for preparation of long acting injectable (LAI) formulations is an auspicious technique to enable preclinical characterization of a broad variety of APIs, ideally independent of their physicochemical and pharmacokinetic (PK) characteristics. During early API discovery, tunable LAI formulations may enable pharmacological proof-of-concept for the given variety of candidates by tailoring the level of plasma exposure over the duration of various timespans. Although numerous reports on small scale preparation methods for LAIs utilizing copolymers of lactic and glycolic acid (PLGA) and polymers of lactic acid (PLA) highlight their potential, application in formulation screening and use in preclinical in vivo studies is yet very limited. Transfer from downscale formulation preparation to in vivo experiments is hampered in early preclinical API screening by the large number of API candidates with simultaneously very limited available amount in the lower sub-gram scale, lack of formulation stability and deficient tunability of sustained release. We hereby present a novel comprehensive platform tool for tailored extended-release formulations, aiming to support a variety of preclinical in vivo experiments with ranging required plasma exposure levels and timespans. A novel small-scale spray drying process was successfully implemented by using an air brush based instrument for preparation of PLGA and PLA based formulations. Using Design of Experiments (DoE), required API amount of 250 mg was demonstrated to suffice for identification of dominant polymer characteristics with largest impact on sustained release capability for an individual API. BI-3231, a hydrophilic and weakly acidic small compound with good water solubility and permeability, but low metabolic stability, was used as an exemplary model for one of the many candidates during API discovery. Furthermore, an in vitro to in vivo correlation (IVIVC) of API release rate was established in mice, which enabled the prediction of in vivo plasma concentration plateaus after single subcutaneous injection, using only in vitro dissolution profiles of screened formulations. By tailoring LAI formulations and their doses for acute and sub-chronic preclinical experiments, we exemplary demonstrate the practical use for BI-3231. Pharmacological proof-of-concept could be enabled whilst circumventing the need of multiple administration as result of extensive hepatic metabolism and simultaneously superseding numerous in vivo experiments for formulation tailoring.
Collapse
Affiliation(s)
- Marco Block
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß 88397, Germany
| | - Peter Sieger
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß 88397, Germany
| | - Cornelius Truenkle
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß 88397, Germany
| | - Christoph Saal
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß 88397, Germany
| | - Roman Simon
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß 88397, Germany
| | - Ines Truebenbach
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß 88397, Germany.
| |
Collapse
|
6
|
Cheng D, Tian R, Pan T, Yu Q, Wei L, Liyin J, Dai Y, Wang X, Tan R, Qu H, Lu M. High-performance lung-targeted bio-responsive platform for severe colistin-resistant bacterial pneumonia therapy. Bioact Mater 2024; 35:517-533. [PMID: 38404643 PMCID: PMC10885821 DOI: 10.1016/j.bioactmat.2024.02.017] [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: 10/23/2023] [Revised: 02/12/2024] [Accepted: 02/14/2024] [Indexed: 02/27/2024] Open
Abstract
Polymyxins are the last line of defense against multidrug-resistant (MDR) Gram-negative bacterial infections. However, this last resort has been threatened by the emergence of superbugs carrying the mobile colistin resistance gene-1 (mcr-1). Given the high concentration of matrix metalloproteinase 3 (MMP-3) in bacterial pneumonia, limited plasma accumulation of colistin (CST) in the lung, and potential toxicity of ionic silver (Ag+), we designed a feasible clinical transformation platform, an MMP-3 high-performance lung-targeted bio-responsive delivery system, which we named "CST&Ag@CNMS". This system exhibited excellent lung-targeting ability (>80% in lungs), MMP-3 bio-responsive release property (95% release on demand), and synergistic bactericidal activity in vitro (2-4-fold minimum inhibitory concentration reduction). In the mcr-1+ CST-resistant murine pneumonia model, treatment with CST&Ag@CNMS improved survival rates (70% vs. 20%), reduced bacteria burden (2-3 log colony-forming unit [CFU]/g tissue), and considerably mitigated inflammatory response. In this study, CST&Ag@CNMS performed better than the combination of free CST and AgNO3. We also demonstrated the superior biosafety and biodegradability of CST&Ag@CNMS both in vitro and in vivo. These findings indicate the clinical translational potential of CST&Ag@CNMS for the treatment of lung infections caused by CST-resistant bacteria carrying mcr-1.
Collapse
Affiliation(s)
- Decui Cheng
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rui Tian
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tingting Pan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Qiang Yu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Li Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiaozhi Liyin
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Yunqi Dai
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Xiaoli Wang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Ruoming Tan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China
| | - Min Lu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
7
|
Yu CP, Lin SW, Tsai JC, Shyong YJ. Long acting tariquidar loaded stearic acid-modified hydroxyapatite enhances brain penetration and antitumor effect of temozolomide. Eur J Pharm Biopharm 2024; 197:114231. [PMID: 38382724 DOI: 10.1016/j.ejpb.2024.114231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/31/2024] [Accepted: 02/18/2024] [Indexed: 02/23/2024]
Abstract
Temozolomide (TMZ) is the first line chemotherapy for glioblastoma (GBM) treatment, but the P-glycoprotein (P-gp) expressed in blood-brain barrier (BBB) will pump out TMZ from the brain leading to decreased TMZ concentration. Tariquidar (TQD), a selective and potent P-gp inhibitor, may be suitable for combination therapy to increase concentration of TMZ in brain. Hydroxyapatite (HAP) is a biodegradable material with sustained release characteristics, and stearic acid surface-modified HAP (SA-HAP) can increase hydrophobicity to facilitate TQD loading. TQD-loaded stearic acid surface-modified HAP (SA-HAP-TQD) was prepared with optimal size and high TQD loading efficiency, and in vitro release and cellular uptake of SA-HAP-TQD showed that SA-HAP-TQD were taken up into lysosome and continuously released TQD from macrophages. In vivo studies have found that over 70 % of SA-HAP was degraded and 80 % of TQD was released from SA-HAP-TQD 28 days after administration. SA-HAP-TQD could increase brain penetration of TMZ, but it would not enhance adverse effects of TMZ in healthy mice. SA-HAP-TQD and TMZ combination had longer median survival than TMZ single therapy in GL261 orthotopic model. These results suggest that SA-HAP-TQD has sustained release characteristics and are potential for improving antitumor effect with TMZ treatment.
Collapse
Affiliation(s)
- Cheng-Ping Yu
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan.
| | - Shang-Wen Lin
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan.
| | - Jui-Chen Tsai
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan.
| | - Yan-Jye Shyong
- School of Pharmacy, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City 701, Taiwan.
| |
Collapse
|
8
|
David C, de Souza JF, Silva AF, Grazioli G, Barboza AS, Lund RG, Fajardo AR, Moraes RR. Cannabidiol-loaded microparticles embedded in a porous hydrogel matrix for biomedical applications. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2024; 35:14. [PMID: 38353746 PMCID: PMC10866797 DOI: 10.1007/s10856-023-06773-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 12/19/2023] [Indexed: 02/16/2024]
Abstract
In this study, poly (lactic-co-glycolic acid) (PLGA) microparticles loaded with cannabidiol (CBD) were synthesized (PLGA@CBD microparticles) and embedded up to 10 wt% in a chondroitin sulfate/polyvinyl alcohol hydrogel matrix. In vitro chemical, physical, and biological assays were carried out to validate the potential use of the modified hydrogels as biomaterials. The microparticles had spherical morphology and a narrow range of size distribution. CBD encapsulation efficiency was around 52%, loading was approximately 50%. Microparticle addition to the hydrogels caused minor changes in their morphology, FTIR and thermal analyses confirmed these changes. Swelling degree and total porosity were reduced in the presence of microparticles, but similar hydrophilic and degradation in phosphate buffer solution behaviors were observed by all hydrogels. Rupture force and maximum strain at rupture were higher in the modified hydrogels, whereas modulus of elasticity was similar across all materials. Viability of primary human dental pulp cells up to 21 days was generally not influenced by the addition of PLGA@CBD microparticles. The control hydrogel showed no antimicrobial activity against Staphylococcus aureus, whereas hydrogels with 5% and 10% PLGA@CBD microparticles showed inhibition zones. In conclusion, the PLGA@CBD microparticles were fabricated and successfully embedded in a hydrogel matrix. Despite the hydrophobic nature of CBD, the physicochemical and morphological properties were generally similar for the hydrogels with and without the CBD-loaded microparticles. The data reported in this study suggested that this original biomaterial loaded with CBD oil has characteristics that could enable it to be used as a scaffold for tissue/cellular regeneration.
Collapse
Affiliation(s)
- Carla David
- Biopathological Research Group, Faculty of Dentistry (GIBFO), University of the Andes, Mérida, Venezuela.
- Graduate Program in Dentistry, Universidade Federal de Pelotas, Pelotas, Brazil.
| | - Jaqueline F de Souza
- Laboratory of Technology and Development of Composites and Polymeric Materials-LaCoPol, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Adriana F Silva
- Graduate Program in Dentistry, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Guillermo Grazioli
- Department of Dental Materials, Universidad de la República, Montevideo, Uruguay
| | - Andressa S Barboza
- Graduate Program in Dentistry, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Rafael G Lund
- Graduate Program in Dentistry, Universidade Federal de Pelotas, Pelotas, Brazil
| | - André R Fajardo
- Laboratory of Technology and Development of Composites and Polymeric Materials-LaCoPol, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Rafael R Moraes
- Graduate Program in Dentistry, Universidade Federal de Pelotas, Pelotas, Brazil
| |
Collapse
|
9
|
Giolando PA, Hopkins K, Davis B, Vike N, Ahmadzadegan A, Ardekani A, Vlachos P, Rispoli J, Solorio L, Kinzer-Ursem TL. Mechanistic Computational Modeling of Implantable, Bioresorbable Drug Release Systems. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301698. [PMID: 37243452 PMCID: PMC10697660 DOI: 10.1002/adma.202301698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/09/2023] [Indexed: 05/28/2023]
Abstract
Implantable, bioresorbable drug delivery systems offer an alternative to current drug administration techniques; allowing for patient-tailored drug dosage, while also increasing patient compliance. Mechanistic mathematical modeling allows for the acceleration of the design of the release systems, and for prediction of physical anomalies that are not intuitive and may otherwise elude discovery. This study investigates short-term drug release as a function of water-mediated polymer phase inversion into a solid depot within hours to days, as well as long-term hydrolysis-mediated degradation and erosion of the implant over the next few weeks. Finite difference methods are used to model spatial and temporal changes in polymer phase inversion, solidification, and hydrolysis. Modeling reveals the impact of non-uniform drug distribution, production and transport of H+ ions, and localized polymer degradation on the diffusion of water, drug, and hydrolyzed polymer byproducts. Compared to experimental data, the computational model accurately predicts the drug release during the solidification of implants over days and drug release profiles over weeks from microspheres and implants. This work offers new insight into the impact of various parameters on drug release profiles, and is a new tool to accelerate the design process for release systems to meet a patient specific clinical need.
Collapse
Affiliation(s)
- Patrick A. Giolando
- Weldon School of Biomedical Engineering, Purdue University
- School of Mechanical Engineering, Purdue University
| | - Kelsey Hopkins
- Weldon School of Biomedical Engineering, Purdue University
| | - Barrett Davis
- Weldon School of Biomedical Engineering, Purdue University
| | - Nicole Vike
- Weldon School of Biomedical Engineering, Purdue University
| | | | | | - Pavlos Vlachos
- Weldon School of Biomedical Engineering, Purdue University
- School of Mechanical Engineering, Purdue University
| | - Joseph Rispoli
- Weldon School of Biomedical Engineering, Purdue University
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University
| | | |
Collapse
|
10
|
Jurić Simčić A, Erak I, Cetina Čižmek B, Hafner A, Filipović-Grčić J. Selection of Excipients for the Preparation of Vancomycin-Loaded Poly(D,L-lactide-co-glycolide) Microparticles with Extended Release by Emulsion Spray Drying. Pharmaceutics 2023; 15:2438. [PMID: 37896198 PMCID: PMC10610132 DOI: 10.3390/pharmaceutics15102438] [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: 09/24/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
The aim of this study was to relate the composition of the W/O emulsion used as a starting fluid in the spray-drying process to the quality of the dry polymer particles obtained in terms of physical-chemical properties, compatibility and drug release performance. Four W/O emulsions containing vancomycin hydrochloride (VAN), an encapsulating PLGA polymer and Poloxamer® 407, chitosan and/or sorbitan monooleate as stabilisers were spray-dried using an ultrasonic atomising nozzle. The microparticles obtained were micron-sized, with a volume mean diameter between 43.2 ± 0.3 and 64.0 ± 12.6 µm, and spherical with a mostly smooth, non-porous surface and with high drug loading (between 14.5 ± 0.6 and 17.1 ± 1.9% w/w). All formulations showed a prolonged and biphasic VAN release profile, with diffusion being the primary release mechanism. Microparticles prepared from the emulsions with Poloxamer® 407 and sorbitan monooleate released VAN rapidly and completely within one day. The release of VAN from microparticles prepared from the emulsion without additives or with chitosan in the inner aqueous phase was significantly decreased; after four days, a cumulative release of 65% and 61%, respectively, was achieved. Microparticles with encapsulated chitosan had the largest mean particle diameter and the slowest release of VAN.
Collapse
Affiliation(s)
- Ana Jurić Simčić
- R&D, PLIVA Croatia Ltd., TEVA Group Member, 10000 Zagreb, Croatia; (A.J.S.); (I.E.); (B.C.Č.)
| | - Iva Erak
- R&D, PLIVA Croatia Ltd., TEVA Group Member, 10000 Zagreb, Croatia; (A.J.S.); (I.E.); (B.C.Č.)
| | - Biserka Cetina Čižmek
- R&D, PLIVA Croatia Ltd., TEVA Group Member, 10000 Zagreb, Croatia; (A.J.S.); (I.E.); (B.C.Č.)
| | - Anita Hafner
- Faculty of Pharmacy and Biochemistry, University of Zagreb, 10000 Zagreb, Croatia;
| | | |
Collapse
|
11
|
Van der Veken M, Brouwers J, Ozbey AC, Umehara K, Stillhart C, Knops N, Augustijns P, Parrott NJ. Investigating Tacrolimus Disposition in Paediatric Patients with a Physiologically Based Pharmacokinetic Model Incorporating CYP3A4 Ontogeny, Mechanistic Absorption and Red Blood Cell Binding. Pharmaceutics 2023; 15:2231. [PMID: 37765200 PMCID: PMC10536648 DOI: 10.3390/pharmaceutics15092231] [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: 07/24/2023] [Revised: 08/06/2023] [Accepted: 08/22/2023] [Indexed: 09/29/2023] Open
Abstract
Tacrolimus is a crucial immunosuppressant for organ transplant patients, requiring therapeutic drug monitoring due to its variable exposure after oral intake. Physiologically based pharmacokinetic (PBPK) modelling has provided insights into tacrolimus disposition in adults but has limited application in paediatrics. This study investigated age dependency in tacrolimus exposure at the levels of absorption, metabolism, and distribution. Based on the literature data, a PBPK model was developed to predict tacrolimus exposure in adults after intravenous and oral administration. This model was then extrapolated to the paediatric population, using a unique reference dataset of kidney transplant patients. Selecting adequate ontogeny profiles for hepatic and intestinal CYP3A4 appeared critical to using the model in children. The best model performance was achieved by using the Upreti ontogeny in both the liver and intestines. To mechanistically evaluate the impact of absorption on tacrolimus exposure, biorelevant in vitro solubility and dissolution data were obtained. A relatively fast and complete release of tacrolimus from its amorphous formulation was observed when mimicking adult or paediatric dissolution conditions (dose, fluid volume). In both the adult and paediatric PBPK models, the in vitro dissolution profiles could be adequately substituted by diffusion-layer-based dissolution modelling. At the level of distribution, sensitivity analysis suggested that differences in blood plasma partitioning of tacrolimus may contribute to the variability in exposure in paediatric patients.
Collapse
Affiliation(s)
- Matthias Van der Veken
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; (M.V.d.V.); (J.B.); (P.A.)
| | - Joachim Brouwers
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; (M.V.d.V.); (J.B.); (P.A.)
| | - Agustos Cetin Ozbey
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, 4070 Basel, Switzerland; (A.C.O.); (K.U.)
| | - Kenichi Umehara
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, 4070 Basel, Switzerland; (A.C.O.); (K.U.)
| | - Cordula Stillhart
- Pharmaceutical R&D, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland;
| | - Noël Knops
- Laboratory for Pediatrics, Department of Development & Regeneration, KU Leuven, O&N3, Bus 817, 3000 Leuven, Belgium;
- Department of Pediatrics, Groene Hart Ziekenhuis, 2803 Gouda, The Netherlands
| | - Patrick Augustijns
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; (M.V.d.V.); (J.B.); (P.A.)
| | - Neil John Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, 4070 Basel, Switzerland; (A.C.O.); (K.U.)
| |
Collapse
|
12
|
Pastorin G, Benetti C, Wacker MG. From in vitro to in vivo: A comprehensive guide to IVIVC development for long-acting therapeutics. Adv Drug Deliv Rev 2023; 199:114906. [PMID: 37286087 DOI: 10.1016/j.addr.2023.114906] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/22/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Affiliation(s)
- Giorgia Pastorin
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore.
| | - Camillo Benetti
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Matthias G Wacker
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| |
Collapse
|
13
|
Dabke A, Ghosh S, Dabke P, Sawant K, Khopade A. Revisiting the in-vitro and in-vivo considerations for in-silico modelling of complex injectable drug products. J Control Release 2023; 360:185-211. [PMID: 37353161 DOI: 10.1016/j.jconrel.2023.06.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/24/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Complex injectable drug products (CIDPs) have often been developed to modulate the pharmacokinetics along with efficacy for therapeutic agents used for remediation of chronic disorders. The effective development of CIDPs has exhibited complex kinetics associated with multiphasic drug release from the prepared formulations. Consequently, predictability of pharmacokinetic modelling for such CIDPs has been difficult and there is need for advanced complex computational models for the establishment of accurate prediction models for in-vitro-in-vivo correlation (IVIVC). The computational modelling aims at supplementing the existing knowledge with mathematical equations to develop formulation strategies for generation of predictable and discriminatory IVIVC. Such an approach would help in reduction of the burden of effect of hidden factors on preclinical to clinical translations. Computational tools like physiologically based pharmacokinetics (PBPK) modelling have combined physicochemical and physiological properties along with IVIVC characteristics of clinically used formulations. Such techniques have helped in prediction and understanding of variability in pharmacodynamic parameters of potential generic products to clinically used formulations like Doxil®, Ambisome®, Abraxane® in healthy and diseased population using mathematical equations. The current review highlights the important formulation characteristics, in-vitro, preclinical in-vivo aspects which need to be considered while developing a stimulatory predictive PBPK model in establishment of an IVIVC and in-vitro-in-vivo relationship (IVIVR).
Collapse
Affiliation(s)
- Amit Dabke
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India; Formulation Research & Development- Biopharmaceutics, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat 390012, India
| | - Saikat Ghosh
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India
| | - Pallavi Dabke
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India
| | - Krutika Sawant
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India.
| | - Ajay Khopade
- Faculty of Pharmacy, Kalabhavan Campus, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat 390001, India; Formulation Research & Development- Novel Drug Delivery Systems, Sun Pharmaceutical Industries Ltd, Vadodara, Gujarat 390012, India.
| |
Collapse
|
14
|
Wan B, Bao Q, Burgess D. Long-acting PLGA microspheres: advances in excipient and product analysis toward improved product understanding. Adv Drug Deliv Rev 2023; 198:114857. [PMID: 37149041 DOI: 10.1016/j.addr.2023.114857] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/16/2023] [Accepted: 04/28/2023] [Indexed: 05/08/2023]
Abstract
Poly(lactic-co-glycolic acid) (PLGA) microspheres are a sustained-release drug delivery system with several successful commercial products used for the treatment of a variety of diseases. By utilizing PLGA polymers with different compositions, therapeutic agents can be released over durations varying from several weeks to several months. However, precise quality control of PLGA polymers and a fundamental understanding of all the factors associated with the performance of PLGA microsphere formulations remains challenging. This knowledge gap can hinder product development of both innovator and generic products. In this review, variability of the key release controlling excipient (PLGA), as well as advanced physicochemical characterization techniques for the PLGA polymer and PLGA microspheres are discussed. The relative merits and challenges of different in vitro release testing methods, in vivo pharmacokinetic studies, and in vitro-in vivo correlation development are also summarized. This review is intended to provide an in-depth understanding of long-acting microsphere products and consequently facilitate the development of these complex products.
Collapse
Affiliation(s)
- Bo Wan
- University of Connecticut, Department of Pharmaceutical Sciences, Storrs, CT 06269
| | - Quanying Bao
- University of Connecticut, Department of Pharmaceutical Sciences, Storrs, CT 06269
| | - Diane Burgess
- University of Connecticut, Department of Pharmaceutical Sciences, Storrs, CT 06269
| |
Collapse
|
15
|
Lou H, Hageman MJ. Development of Drug Release Model for Suspensions in ESCAR (Emulator of SubCutaneous Absorption and Release). AAPS J 2023; 25:29. [PMID: 36949301 PMCID: PMC10184886 DOI: 10.1208/s12248-023-00799-1] [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: 01/25/2023] [Accepted: 02/28/2023] [Indexed: 03/24/2023] Open
Abstract
We recently developed an in vitro testing system, namely, ESCAR (Emulator of SubCutaneous Absorption and Release). The objective of this work was to investigate drug release behaviors of unmilled and milled suspensions in ESCAR. A mass transport-based model was developed to describe the multi-step drug release process, including drug dissolution, particle settling, drug distribution/partition, and drug permeation through the membrane(s). To address the particle settling effect, a correction factor was included in the model and its value was obtained by data fitting. It was found that, for both suspensions, (i) the experimental data of various dose/formulation combinations could be fit by the developed model; (ii) the dose effect on drug release was offset by the particle settling effect. This model may help to reduce experimental efforts and facilitate subcutaneous suspension formulation development using ESCAR.
Collapse
Affiliation(s)
- Hao Lou
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA.
- Biopharmaceutical Innovation and Optimization Center, University of Kansas, Lawrence, KS, 66047, USA.
| | - Michael J Hageman
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA.
- Biopharmaceutical Innovation and Optimization Center, University of Kansas, Lawrence, KS, 66047, USA.
| |
Collapse
|
16
|
Lou H, Hageman MJ. Development of an In Vitro System To Emulate an In Vivo Subcutaneous Environment: Small Molecule Drug Assessment. Mol Pharm 2022; 19:4017-4025. [PMID: 36279508 DOI: 10.1021/acs.molpharmaceut.2c00490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A reliable in vitro system can support and guide the development of subcutaneous (SC) drug products. Although several in vitro systems have been developed, they have some limitations, which may hinder them from getting more engaged in SC drug product development. This study sought to develop a novel in vitro system, namely, Emulator of SubCutaneous Absorption and Release (ESCAR), to better emulate the in vivo SC environment and predict the fate of drugs in SC delivery. ESCAR was designed using computer-aided design (CAD) software and fabricated using the three-dimensional (3D) printing technique. ESCAR has a design of two acceptor chambers representing the blood uptake pathway and the lymphatic uptake pathway, respectively, although only the blood uptake pathway was investigated for small molecules in this study. Via conducting a DoE factor screening study using acetaminophen solution, the relationship of the output (drug release from the "SC" chamber to the "blood circulation" chamber) and the input parameters could be modeled using a variety of methods, including polynomial equations, machine learning methods, and Monte Carlo simulation-based methods. The results suggested that the hyaluronic acid (HA) concentration was a critical parameter, whereas the influence of the injection volume and injection position was not substantial. An in vitro-in vivo correlation (IVIVC) study was developed using griseofulvin suspension to explore the feasibility of applying ESCAR in formulation development and bioequivalence studies. The developed LEVEL A IVIVC model demonstrated that the in vivo PK profile could be correlated with the in vitro release profile. Therefore, using this model, for new formulations, only in vitro studies need to be conducted in ESCAR, and in vivo studies might be waived. In conclusion, ESCAR had important implications for research and development and quality control of SC drug products. Future work would be focused on further optimizing ESCAR and expanding its applications via assessing more types of molecules and formulations.
Collapse
Affiliation(s)
- Hao Lou
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas66047, United States
- Biopharmaceutical Innovation and Optimization Center, University of Kansas, Lawrence, Kansas66047, United States
| | - Michael J. Hageman
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas66047, United States
- Biopharmaceutical Innovation and Optimization Center, University of Kansas, Lawrence, Kansas66047, United States
| |
Collapse
|
17
|
Song JS, Kim SY, Nam JH, Lee J, Song SY, Seong H. IVIVC of Octreotide in PLGA-Glucose Microsphere Formulation, Sandostatin® LAR. AAPS PharmSciTech 2022; 23:258. [PMID: 36123513 DOI: 10.1208/s12249-022-02359-w] [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: 12/20/2021] [Accepted: 07/08/2022] [Indexed: 11/30/2022] Open
Abstract
In vitro-in vivo correlation (IVIVC) analysis reveals a relationship between in vitro release and in vivo pharmacokinetic response of the drug of interest. Sandostatin LAR Depot (SLD) for endocrine tumors and acromegaly is a sustained-release formulation of octreotide, a cyclic oligomer of 8 amino acids, which prolongs therapeutic efficacy and enhances medication compliance of octreotide. Since the efficacy of SLD is dependent on the pharmacokinetic characteristics of octreotide released from a biodegradable matrix polymer, poly(lactide-co-glycolide)-glucose, of SLD, the IVIVC of SLD is critical for predicting an in vivo behavior of the octreotide. In this study, in vitro release of octreotide from SLD was investigated using the release test media each containing 0.02% or 0.5% surfactant and having different pH values of 7.4 and 5.5. In vivo pharmacokinetic profiles of SLD were determined by LC-MS/MS analysis of the systemic blood concentration of octreotide after the SLD injection to rodents. In IVIVC analysis, the Weibull model was adopted as a drug release model for biodegradable microsphere formulation. The IVIVC analyses revealed the in vitro release test condition of SLD with the highest IVIV correlation coefficient. By applying the in vitro release data to the model derived from the IVIVC analysis, pharmacokinetic parameters of SLD could be predicted with the prediction error of ± 10 ~ 15%. IVIVC analysis and pharmacokinetic prediction model of SLD in our study can be an efficient tool for the development of long-acting pharmaceutical dosage forms.
Collapse
Affiliation(s)
- Jin-Sook Song
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - So-Yeon Kim
- General Technical Institute, Kolmar Korea, 61 (Naegok-dong) 8-gil, Heolleung-ro, Seocho-gu, Seoul, 06800, South Korea.,Department of Polymer Science & Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, South Korea
| | - Jae-Hyun Nam
- College of Pharmacy, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, South Korea
| | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, South Korea
| | - Sang-Yong Song
- Peptron Inc., 37-24, Yuseong-daero 1628 beon-gil, Yuseong-gu, Daejeon, 34054, South Korea
| | - Hasoo Seong
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea.
| |
Collapse
|
18
|
Villa Nova M, Gan K, Wacker MG. Biopredictive tools for the development of injectable drug products. Expert Opin Drug Deliv 2022; 19:671-684. [PMID: 35603724 DOI: 10.1080/17425247.2022.2081682] [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: 11/04/2022]
Abstract
INTRODUCTION Biopredictive release tests are commonly used in the evaluation of oral medicines. They support decision-making in formulation development and allow predictions of the expected in-vivo performances. So far, there is limited experience in the application of these methodologies to injectable drug products. AREAS COVERED Parenteral drug products cover a variety of dosage forms and administration sites including subcutaneous, intramuscular, and intravenous injections. In this area, developing biopredictive and biorelevant methodologies often confronts us with unique challenges and knowledge gaps. Here, we provide a formulation-centric approach and explain the key considerations and workflow when designing biopredictive assays. Also, we outline the key role of computational methods in achieving clinical relevance and put all considerations into context using liposomal nanomedicines as an example. EXPERT OPINION Biopredictive tools are the need of the hour to exploit the tremendous opportunities of injectable drug products. A growing number of biopharmaceuticals such as peptides, proteins, and nucleic acids require different strategies and a better understanding of the influences on drug absorption. Here, our design strategy must maintain the balance of robustness and complexity required for effective formulation development.
Collapse
Affiliation(s)
- Mônica Villa Nova
- State University of Maringá, Department of Pharmacy, Maringá, Paraná, Brazil
| | - Kennard Gan
- National University of Singapore, Department of Pharmacy, Singapore
| | | |
Collapse
|
19
|
Torres-Terán I, Venczel M, Klein S. Prediction of subcutaneous drug absorption - do we have reliable data to design a simulated interstitial fluid? Int J Pharm 2021; 610:121257. [PMID: 34737015 DOI: 10.1016/j.ijpharm.2021.121257] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/15/2021] [Accepted: 10/28/2021] [Indexed: 01/02/2023]
Abstract
For many years subcutaneous (SC) administration has represented the main route for delivering biopharmaceuticals. However, little information exists about the milieu in the subcutaneous tissue, especially about the properties/composition of the fluid present in this tissue, the interstitial fluid (ISF), which is one of the key elements for the drug release and absorption. Better knowledge on SC ISF composition, properties and dynamics may provide better insight into in vivo drug performance. In addition, a simulated SC ISF, which allows better prediction of in vivo absorption of drugs after subcutaneous administration based on in vitro release experiments, would help to improve formulation design, and reduce the number of animal studies and clinical trials required to obtain marketing authorization. To date, a universal medium for predicting drug solubility/release in the interstitial space does not exist. This review provides an overview of the currently available information on composition and physicochemical properties of SC ISF and critically discusses different isolation techniques in the context of information that could be gained from the isolated fluid. Moreover, it surveys current in vitro release media aiming to mimic SC ISF composition and highlights information gaps that need to be filled for designing a meaningful artificial SC ISF.
Collapse
Affiliation(s)
- Iria Torres-Terán
- Sanofi-Aventis Deutschland GmbH, R&D, Global CMC Development, Synthetics Platform. Industriepark Hoechst, H770, D-65926 Frankfurt Am Main, Germany; Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, 3 Felix Hausdorff Street, 17489 Greifswald, Germany
| | - Márta Venczel
- Sanofi-Aventis Deutschland GmbH, R&D, Global CMC Development, Synthetics Platform. Industriepark Hoechst, H770, D-65926 Frankfurt Am Main, Germany
| | - Sandra Klein
- Department of Pharmacy, Institute of Biopharmaceutics and Pharmaceutical Technology, Center of Drug Absorption and Transport, University of Greifswald, 3 Felix Hausdorff Street, 17489 Greifswald, Germany.
| |
Collapse
|
20
|
An Update to Dialysis-Based Drug Release Testing-Data Analysis and Validation Using the Pharma Test Dispersion Releaser. Pharmaceutics 2021; 13:pharmaceutics13122007. [PMID: 34959289 PMCID: PMC8708653 DOI: 10.3390/pharmaceutics13122007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 11/17/2022] Open
Abstract
Currently, a wide variety of complex non-oral dosage forms are entering the global healthcare market. Although many assays have been described in recent research, harmonized procedures and standards for testing their in vitro performance remain widely unexplored. Among others, dialysis-based techniques such as the Pharma Test Dispersion Releaser are developed for testing the release of drugs from nanoparticles, liposomes, or extracellular vesicle preparations. Here, we provide advanced strategies and practical advice for the development and validation of dialysis-based techniques, including documentation, analysis, and interpretation of the raw data. For this purpose, key parameters of the release assay, including the hydrodynamics in the device at different stirring rates, the selectivity for particles and molecules, as well as the effect of excipients on drug permeation were investigated. At the highest stirring rate, a more than twofold increase in the membrane permeation rate (from 0.99 × 10−3 to 2.17 × 10−3 cm2/h) was observed. Additionally, we designed a novel computer model to identify important quality parameters of the dialysis experiment and to calculate error-corrected release profiles. Two hydrophilic creams of diclofenac, Voltaren® Emulgel, and Olfen® gel, were tested and provide first-hand evidence of the robustness of the assay in the presence of semisolid dosage forms.
Collapse
|
21
|
Simulate SubQ: The Methods and the Media. J Pharm Sci 2021; 112:1492-1508. [PMID: 34728176 DOI: 10.1016/j.xphs.2021.10.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/25/2021] [Accepted: 10/26/2021] [Indexed: 11/21/2022]
Abstract
For decades, there has been a growing interest in injectable subcutaneous formulations to improve the absorption of drugs into the systemic circulation and to prolong their release over a longer period. However, fluctuations in the blood plasma levels together with bioavailability issues often limit their clinical success. This warrants a closer look at the performance of long-acting depots, for example, and their dependence on the complex interplay between the dosage form and the physiological microenvironment. For this, biopredictive performance testing is used for a thorough understanding of the biophysical processes affecting the absorption of compounds from the injection site in vivo and their simulation in vitro. In the present work, we discuss in vitro methodologies including methods and media developed for the subcutaneous route of administration on the background of the most relevant absorption mechanisms. Also, we highlight some important knowledge gaps and shortcomings of the existing methodologies to provide the reader with a better understanding of the scientific evidence underlying these models.
Collapse
|
22
|
Deng C, Jin Q, Wu Y, Li H, Yi L, Chen Y, Gao T, Wang W, Wang J, Lv Q, Yang Y, Xu J, Fu W, Zhang L, Xie M. Immunosuppressive effect of PLGA-FK506-NPs in treatment of acute cardiac rejection via topical subcutaneous injection. Drug Deliv 2021; 28:1759-1768. [PMID: 34463172 PMCID: PMC8409942 DOI: 10.1080/10717544.2021.1968978] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
FK506, a first-line immunosuppressant, is routinely administered orally and intravenously to inhibit activation and proliferation of T cells after heart transplantation (HT). Current administration route is not conducive enough to exert its efficacy in lymphatic system. Herein, we proposed that subcutaneous (SC) administration of FK506-loaded nanoparticles (PLGA-FK506-NPs) would be valuable for treating acute rejection after HT. The biodistribution and pharmacokinetic study revealed that it could effectively deliver FK506 to the lymph nodes (LNs) due to their suitable particle size, especially in inguinal LNs. Subsequently, the therapeutic efficacy of PLGA-FK506-NPs on the HT model was evaluated using intravenous (IV), intragastric (IG), or SC injection. Histopathological analysis revealed that 80% of allografts exhibited only grade 1R rejection with negligible lymphocyte infiltration in the SC group. The IV group exhibited 40% 1R rejection with mild lymphocyte infiltration and 20% grade 3R that require further intervention, and the IG group exhibited grades 40% 3R rejection with more lymphocyte infiltration. Moreover, the infiltration of T cells and the secretion of IL-2 and IFN-γ were significantly reduced in the SC group compared with the IG or IV group. The mean survival time (MST) further revealed that 50% of grafts treated with PLGA-FK506-NPs via SC injection survived longer than IG and IV groups. Moreover, the MST of single-dose SC injection of PLGA-FK506-NPs demonstrated that it would effectively reduce the required dose for a similar therapeutic effect. Overall, these results indicate that SC administration of PLGA-FK506-NPs is a more effective route for chronic FK506 treatment.
Collapse
Affiliation(s)
- Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Ya Wu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Huiling Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Luyang Yi
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Tang Gao
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Wenyuan Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Jia Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Wenpei Fu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|