1
|
Wang Z, Zhai B, Sun J, Zhang X, Zou J, Shi Y, Guo D. Recent advances of injectable in situ-forming hydrogels for preventing postoperative tumor recurrence. Drug Deliv 2024; 31:2400476. [PMID: 39252545 PMCID: PMC11389645 DOI: 10.1080/10717544.2024.2400476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/17/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
The unavoidable residual tumor tissue from surgery and the strong aggressiveness of tumor cells pose challenges to the postoperative treatment of tumor patients, accompanied by in situ tumor recurrence and decreased quality of life. Therefore, there is an urgent need to explore appropriate postoperative therapeutic strategies to remove residual tumor cells after surgery to inhibit tumor recurrence and metastasis after surgery. In recent years, with the rapid development of biomedical materials, the study of local delivery systems as postoperative delivery of therapeutic agents has gradually attracted the attention of researchers. Injectable in situ-forming hydrogel is a locally administered agent injected in situ as a solution that can be loaded with various therapeutic agents and rapidly gels to form a semi-solid gel at the treatment site. This type of hydrogel tightly fills the surgical site and covers irregular excision surfaces. In this paper, we review the recent advances in the application of injectable in situ-forming hydrogels in postoperative therapy, focusing on the matrix materials of this type of hydrogel and its application in the postoperative treatment of different types of tumors, as well as discussing the challenges and prospects of its clinical application.
Collapse
Affiliation(s)
- Zhanpeng Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Bingtao Zhai
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Jing Sun
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Xiaofei Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Junbo Zou
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Yajun Shi
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Dongyan Guo
- School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| |
Collapse
|
2
|
Yang W, Yan K, Feng Y, Zhao X. Charge reversible hyaluronic acid-based drug delivery system with pH-responsive dissociation for enhanced drug delivery. Eur J Pharm Biopharm 2024:114560. [PMID: 39447775 DOI: 10.1016/j.ejpb.2024.114560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/26/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Improving the efficiency of drug delivery is one of the most important goals in the field of drug delivery. One strategy for drug delivery efficiency is to make the drug delivery system capable of charge reversal. In this study, we used hyaluronic acid (HA) as the skeleton to anchor dimethylmaleic anhydride-modified polylysine (PLL-DMMA) and N-(3-Aminopropyl)-imidazole (IMI) to construct a pH-sensitive (IMI/Zn2+)-HA-PLL-DMMA system via Zn coordination. The (IMI/Zn2+)-HA-PLL-DMMA system can detach DMMA moieties and expose PLL with a positive charge in the acidic tumor microenvironment (TME), which enhances cellular uptake in cancer cells through charge reversal. Once the drug-loaded (IMI/Zn2+)-HA-PLL-DMMA enters cancer cells, it specifically responds and disassembles in the acidic TME, resulting in drug release and inhibition of cancer cell viability. The (IMI/Zn2+)-HA-PLL-DMMA system is designed to regulate drug release behavior with Zn2+ and IMI groups as control units. The HA-based system shows synergistic selective drug delivery in suppressing tumor cells and has potential in cancer therapy.
Collapse
Affiliation(s)
- Wenjing Yang
- Department of Anesthesiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Ke Yan
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Yecheng Feng
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Xubo Zhao
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
3
|
Lin Q, Chee PL, Pang JJM, Loh XJ, Kai D, Lim JYC. Sustainable Polymeric Biomaterials from Alternative Feedstocks. ACS Biomater Sci Eng 2024. [PMID: 39382551 DOI: 10.1021/acsbiomaterials.4c01154] [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: 10/10/2024]
Abstract
As materials engineered to interact with biological systems for medical purposes, polymeric biomedical materials have revolutionized and are indispensable in modern healthcare. However, aging populations and improving healthcare standards worldwide have resulted in ever-increasing demands for such biomaterials. Currently, many clinically used polymers are derived from nonrenewable petroleum resources, thus spurring the need for exploring alternatives for the next generation of sustainable biomaterials. Other than biomass, this Perspective also spotlights carbon dioxide and postuse plastics as viable resources potentially suitable for biomaterial production. For each alternative feedstock, key recent developments and practical considerations are discussed, including emerging biomaterial applications, possible feedstock sources, and hindrances toward translation and practical adoption. Other than replacements for petroleum-derived polymers, we explore how utilization of these alternatives capitalizes on their intrinsic physiochemical and material properties to achieve their desired therapeutic effects. We hope that this Perspective can stimulate further development in sustainable biomaterials to achieve practical therapeutic benefits as part of a circular materials economy with minimal environmental impact.
Collapse
Affiliation(s)
- Qianyu Lin
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Pei Lin Chee
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Jaime J M Pang
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore (NUS), 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Dan Kai
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE2), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, 138634, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 62 Nanyang Dr, 637459, Singapore
| | - Jason Y C Lim
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore (NUS), 9 Engineering Drive 1, Singapore 117576, Singapore
| |
Collapse
|
4
|
Modi D, Hussain MS, Ainampudi S, Prajapati BG. Long acting injectables for the treatment of prostate cancer. J Drug Deliv Sci Technol 2024; 100:105996. [DOI: 10.1016/j.jddst.2024.105996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
|
5
|
Abulateefeh SR, Abuhamdan RM, Saed H, Alsalem M, Shnewer K. In vitro and in vivo evaluation of in situ forming polyester implants for the extended release of carvedilol. Drug Deliv Transl Res 2024:10.1007/s13346-024-01706-7. [PMID: 39313736 DOI: 10.1007/s13346-024-01706-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 09/25/2024]
Abstract
Polyester based in situ forming implants (ISFIs) are injectable long-acting drug delivery systems that offer a wide range of unique advantages. As a result of these advantages, two relatively high molecular weight, ester terminated grades of poly (D,L-lactide-co-glycolide) (PLGA) and poly(D,L-lactide) (PLA) were evaluated for their ability (i) to form ISFIs loaded with carvedilol, and (ii) to control its release both in vitro and in vivo. At a polymeric concentration of 40% w/w, implant solutions were syringeable, injectable, and able to encapsulate carvedilol to a high degree (encapsulated drug% > 97%). When visualized using scanning electron microscopy (SEM), implants were found to have a dense thin surface atop porous sublayers. As for their in vitro evaluation, PLGA and PLA implants were able to maintain drug release over the course of 49 and 84 days, respectively. On the other hand, in vivo drug release from both implants was almost identical and lasted for only 42 days. This may be due to the overriding effect of the similar host environment at the injection site that diminished the effect of polymeric physiochemistry on phase inversion and drug release. Lastly, while the polymer-free drug/NMP solution completely released its drug content within the initial half hour in vitro, the formulation extended drug release in vivo. This could be due to a yet to be investigated interaction between carvedilol and NMP under in vivo conditions. These results cement the significance of formulating carvedilol loaded ISFIs for the management of chronic conditions.
Collapse
Affiliation(s)
- Samer R Abulateefeh
- School of Pharmacy, The University of Jordan, Amman, 11942, Jordan.
- Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, 11733, Jordan.
| | | | - Husam Saed
- School of Pharmacy, The University of Jordan, Amman, 11942, Jordan
| | - Mohammad Alsalem
- School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | | |
Collapse
|
6
|
Biswas A, Kumar S, Choudhury AD, Bisen AC, Sanap SN, Agrawal S, Mishra A, Verma SK, Kumar M, Bhatta RS. Polymers and their engineered analogues for ocular drug delivery: Enhancing therapeutic precision. Biopolymers 2024; 115:e23578. [PMID: 38577865 DOI: 10.1002/bip.23578] [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: 01/26/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024]
Abstract
Ocular drug delivery is constrained by anatomical and physiological barriers, necessitating innovative solutions for effective therapy. Natural polymers like hyaluronic acid, chitosan, and gelatin, alongside synthetic counterparts such as PLGA and PEG, have gained prominence for their biocompatibility and controlled release profiles. Recent strides in polymer conjugation strategies have enabled targeted delivery through ligand integration, facilitating tissue specificity and cellular uptake. This versatility accommodates combined drug delivery, addressing diverse anterior (e.g., glaucoma, dry eye) and posterior segment (e.g., macular degeneration, diabetic retinopathy) afflictions. The review encompasses an in-depth exploration of each natural and synthetic polymer, detailing their individual advantages and disadvantages for ocular drug delivery. By transcending ocular barriers and refining therapeutic precision, these innovations promise to reshape the management of anterior and posterior segment eye diseases.
Collapse
Affiliation(s)
- Arpon Biswas
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Shivansh Kumar
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Abhijit Deb Choudhury
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Amol Chhatrapati Bisen
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sachin Nashik Sanap
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sristi Agrawal
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Anjali Mishra
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sarvesh Kumar Verma
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Mukesh Kumar
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
- Jawaharlal Nehru University, New Delhi, India
| | - Rabi Sankar Bhatta
- Pharmaceutics and Pharmacokinetic Division, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
7
|
Li Y, Jie C, Wang J, Zhang W, Wang J, Deng Y, Liu Z, Hou X, Bi X. Global research trends and future directions in diabetic macular edema research: A bibliometric and visualized analysis. Medicine (Baltimore) 2024; 103:e38596. [PMID: 38905408 PMCID: PMC11191902 DOI: 10.1097/md.0000000000038596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 05/24/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Diabetic Macular Edema (DME) significantly impairs vision in diabetics, with varied patient responses to current treatments like anti-vascular endothelial growth factor (VEGF) therapy underscoring the necessity for continued research into more effective strategies. This study aims to evaluate global research trends and identify emerging frontiers in DME to guide future research and clinical management. METHODS A qualitative and quantitative analysis of publications related to diabetic macular edema retrieved from the Web of Science Core Collection (WoSCC) between its inception and September 4, 2023, was conducted. Microsoft Excel, CiteSpace, VOSviewer, Bibliometrix Package, and Tableau were used for the bibliometric analysis and visualization. This encompasses an examination of the overall distribution of annual output, major countries, regions, institutions, authors, core journals, co-cited references, and keyword analyses. RESULTS Overall, 5624 publications were analyzed, indicating an increasing trend in DME research. The United States was identified as the leading country in DME research, with the highest h-index of 135 and 91,841 citations. Francesco Bandello emerged as the most prolific author with 97 publications. Neil M. Bressler has the highest h-index and highest total citation count of 46 and 9692, respectively. The journals "Retina - the Journal of Retinal and Vitreous Diseases" and "Ophthalmology" were highlighted as the most prominent in this field. "Retina" leads with 354 publications, a citation count of 11,872, and an h-index of 59. Meanwhile, "Ophthalmology" stands out with the highest overall citation count of 31,558 and the highest h-index of 90. The primary research focal points in diabetic macular edema included "prevalence and risk factors," "pathological mechanisms," "imaging modalities," "treatment strategies," and "clinical trials." Emerging research areas encompassed "deep learning and artificial intelligence," "novel treatment modalities," and "biomarkers." CONCLUSION Our bibliometric analysis delineates the leading role of the United States in DME research. We identified current research hotspots, including epidemiological studies, pathophysiological mechanisms, imaging advancements, and treatment innovations. Emerging trends, such as the integration of artificial intelligence and novel therapeutic approaches, highlight future directions. These insights underscore the importance of collaborative and interdisciplinary approaches in advancing DME research and clinical management.
Collapse
Affiliation(s)
- Yuanyuan Li
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Chuanhong Jie
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianwei Wang
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Weiqiong Zhang
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Jingying Wang
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Deng
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Ziqiang Liu
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoyu Hou
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| | - Xuqi Bi
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
8
|
Dedeloudi A, Martinez-Marcos L, Quinten T, Andersen S, Lamprou DA. Biopolymeric 3D printed implantable scaffolds as a potential adjuvant treatment for acute post-operative pain management. Expert Opin Drug Deliv 2024:1-13. [PMID: 38555481 DOI: 10.1080/17425247.2024.2336492] [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: 12/01/2023] [Accepted: 03/09/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Pain is characterized as a major symptom induced by tissue damage occurring from surgical procedures, whose potency is being experienced subjectively, while current pain relief strategies are not always efficient in providing individualized treatment. 3D printed implantable devices hold the potential to offer a precise and customized medicinal approach, targeting both tissue engineering and drug delivery. RESEARCH DESIGN AND METHODS Polycaprolactone (PCL) and PCL - chitosan (CS) composite scaffolds loaded with procaine (PRC) were fabricated by bioprinting. Geometrical features including dimensions, pattern, and infill of the scaffolds were mathematically optimized and digitally determined, aiming at developing structurally uniform 3D printed models. Printability studies based on thermal imaging of the bioprinting system were performed, and physicochemical, surface, and mechanical attributes of the extruded scaffolds were evaluated. The release rate of PRC was examined at different time intervals up to 1 week. RESULTS Physicochemical stability and mechanical integrity of the scaffolds were studied, while in vitro drug release studies revealed that CS contributes to the sustained release dynamic of PRC. CONCLUSIONS The printing extrusion process was capable of developing implantable devices for a local and sustained delivery of PRC as a 7-day adjuvant regimen in post-operative pain management.
Collapse
Affiliation(s)
| | - Laura Martinez-Marcos
- Janssen Pharmaceutica, Oral Solids Development (OSD) Research & Development Department, Beerse, Belgium
| | - Thomas Quinten
- Janssen Pharmaceutica, Oral Solids Development (OSD) Research & Development Department, Beerse, Belgium
| | - Sune Andersen
- Janssen Pharmaceutica, Oral Solids Development (OSD) Research & Development Department, Beerse, Belgium
| | | |
Collapse
|
9
|
Ebrahimnia M, Alavi S, Vaezi H, Karamat Iradmousa M, Haeri A. Exploring the vast potentials and probable limitations of novel and nanostructured implantable drug delivery systems for cancer treatment. EXCLI JOURNAL 2024; 23:143-179. [PMID: 38487087 PMCID: PMC10938236 DOI: 10.17179/excli2023-6747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/08/2024] [Indexed: 03/17/2024]
Abstract
Conventional cancer chemotherapy regimens, albeit successful to some extent, suffer from some significant drawbacks, such as high-dose requirements, limited bioavailability, low therapeutic indices, emergence of multiple drug resistance, off-target distribution, and adverse effects. The main goal of developing implantable drug delivery systems (IDDS) is to address these challenges and maintain anti-cancer drugs directly at the intended sites of therapeutic action while minimizing inevitable side effects. IDDS possess numerous advantages over conventional drug delivery, including controlled drug release patterns, one-time drug administration, as well as loading and stabilizing poorly water-soluble chemotherapy drugs. Here, we summarized conventional and novel (three-dimensional (3D) printing and microfluidic) preparation techniques of different IDDS, including nanofibers, films, hydrogels, wafers, sponges, and osmotic pumps. These systems could be designed with high biocompatibility and biodegradability features using a wide variety of natural and synthetic polymers. We also reviewed the published data on these systems in cancer therapy with a particular focus on their release behavior. Various release profiles could be attained in IDDS, which enable predictable, adjustable, and sustained drug releases. Furthermore, multi-step or stimuli-responsive drug release could be obtained in these systems. The studies mentioned in this article have proven the effectiveness of IDDS for treating different cancer types with high prevalence, including breast cancer, and aggressive cancer types, such as glioblastoma and liver cancer. Additionally, the challenges in applying IDDS for efficacious cancer therapy and their potential future developments are also discussed. Considering the high potential of IDDS for further advancements, such as programmable release and degradation features, further clinical trials are needed to ensure their efficiency. The overall goal of this review is to expand our understanding of the behavior of commonly investigated IDDS and to identify the barriers that should be addressed in the pursuit of more efficient therapies for cancer. See also the graphical abstract(Fig. 1).
Collapse
Affiliation(s)
- Maryam Ebrahimnia
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sonia Alavi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- College of Pharmacy, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Hamed Vaezi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdieh Karamat Iradmousa
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Nayan MU, Panja S, Sultana A, Zaman LA, Vora LK, Sillman B, Gendelman HE, Edagwa B. Polymer Delivery Systems for Long-Acting Antiretroviral Drugs. Pharmaceutics 2024; 16:183. [PMID: 38399244 PMCID: PMC10892262 DOI: 10.3390/pharmaceutics16020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
The success of long-acting (LA) drug delivery systems (DDSs) is linked to their biocompatible polymers. These are used for extended therapeutic release. For treatment or prevention of human immune deficiency virus type one (HIV-1) infection, LA DDSs hold promise for improved regimen adherence and reduced toxicities. Current examples include Cabenuva, Apretude, and Sunlenca. Each is safe and effective. Alternative promising DDSs include implants, prodrugs, vaginal rings, and microarray patches. Each can further meet patients' needs. We posit that the physicochemical properties of the formulation chemical design can optimize drug release profiles. We posit that the strategic design of LA DDS polymers will further improve controlled drug release to simplify dosing schedules and improve regimen adherence.
Collapse
Affiliation(s)
- Mohammad Ullah Nayan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Sudipta Panja
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Ashrafi Sultana
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Lubaba A. Zaman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Lalitkumar K. Vora
- School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK;
| | - Brady Sillman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA; (M.U.N.); (S.P.); (A.S.); (L.A.Z.); (B.S.)
| |
Collapse
|
11
|
Ashique S, Mishra N, Mohanto S, Gowda BJ, Kumar S, Raikar AS, Masand P, Garg A, Goswami P, Kahwa I. Overview of processed excipients in ocular drug delivery: Opportunities so far and bottlenecks. Heliyon 2024; 10:e23810. [PMID: 38226207 PMCID: PMC10788286 DOI: 10.1016/j.heliyon.2023.e23810] [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: 08/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 01/17/2024] Open
Abstract
Ocular drug delivery presents a unique set of challenges owing to the complex anatomy and physiology of the eye. Processed excipients have emerged as crucial components in overcoming these challenges and improving the efficacy and safety of ocular drug delivery systems. This comprehensive overview examines the opportunities that processed excipients offer in enhancing drug delivery to the eye. By analyzing the current landscape, this review highlights the successful applications of processed excipients, such as micro- and nano-formulations, sustained-release systems, and targeted delivery strategies. Furthermore, this article delves into the bottlenecks that have impeded the widespread adoption of these excipients, including formulation stability, biocompatibility, regulatory constraints, and cost-effectiveness. Through a critical evaluation of existing research and industry practices, this review aims to provide insights into the potential avenues for innovation and development in ocular drug delivery, with a focus on addressing the existing challenges associated with processed excipients. This synthesis contributes to a deeper understanding of the promising role of processed excipients in improving ocular drug delivery systems and encourages further research and development in this rapidly evolving field.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur 713212, West Bengal, India
| | - Neeraj Mishra
- Amity Institute of Pharmacy, Amity University Madhya Pradesh, Gwalior, 474005, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - B.H. Jaswanth Gowda
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, Belfast BT9 7BL, UK
| | - Shubneesh Kumar
- Department of Pharmaceutics, Bharat Institute of Technology, School of Pharmacy, Meerut 250103, UP, India
| | - Amisha S. Raikar
- Department of Pharmaceutics, PES Rajaram and Tarabai Bandekar College of Pharmacy, Ponda, Goa 403401, India
| | - Priya Masand
- Department of Pharmaceutical Technology, Meerut Institute of Engineering & Technology, (MIET), NH-58, Delhi-Roorkee Highway, Meerut, Uttar Pradesh 250005, India
| | - Ashish Garg
- Department of Pharmaceutics, Guru Ramdas Khalsa Institute of Science and Technology (Pharmacy), Jabalpur, Madhya Pradesh, India
| | - Priyanka Goswami
- Department of Pharmacognosy, Saraswati Institute of Pharmaceutical Sciences, Gandhinagar 382355, Gujarat, India
- Maharashtra Educational Society's H.K. College of Pharmacy, Mumbai: 400102.India
| | - Ivan Kahwa
- Department of Pharmacy, Faculty of Medicine, Mbarara University of Science and Technology, P.O Box 1410, Mbarara, Uganda
- Pharm-Bio Technology and Traditional Medicine Centre, Mbarara University of Science and Technology, P. O Box 1410, Mbarara, Uganda
| |
Collapse
|
12
|
Wang Y, Chen Z, Davis B, Lipman W, Xing S, Zhang L, Wang T, Hafiz P, Xie W, Yan Z, Huang Z, Song J, Bai W. Digital automation of transdermal drug delivery with high spatiotemporal resolution. Nat Commun 2024; 15:511. [PMID: 38218967 PMCID: PMC10787768 DOI: 10.1038/s41467-023-44532-0] [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: 08/19/2023] [Accepted: 12/18/2023] [Indexed: 01/15/2024] Open
Abstract
Transdermal drug delivery is of vital importance for medical treatments. However, user adherence to long-term repetitive drug delivery poses a grand challenge. Furthermore, the dynamic and unpredictable disease progression demands a pharmaceutical treatment that can be actively controlled in real-time to ensure medical precision and personalization. Here, we report a spatiotemporal on-demand patch (SOP) that integrates drug-loaded microneedles with biocompatible metallic membranes to enable electrically triggered active control of drug release. Precise control of drug release to targeted locations (<1 mm2), rapid drug release response to electrical triggers (<30 s), and multi-modal operation involving both drug release and electrical stimulation highlight the novelty. Solution-based fabrication ensures high customizability and scalability to tailor the SOP for various pharmaceutical needs. The wireless-powered and digital-controlled SOP demonstrates great promise in achieving full automation of drug delivery, improving user adherence while ensuring medical precision. Based on these characteristics, we utilized SOPs in sleep studies. We revealed that programmed release of exogenous melatonin from SOPs improve sleep of mice, indicating potential values for basic research and clinical treatments.
Collapse
Affiliation(s)
- Yihang Wang
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zeka Chen
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Brayden Davis
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Will Lipman
- Department of Psychology and Neuroscience, University of North Carolina at chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sicheng Xing
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Lin Zhang
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Tian Wang
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Priyash Hafiz
- UNC/NCSU Joint Department of Biomedical Engineering, Chapel Hill, NC, 27599, USA
| | - Wanrong Xie
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zijie Yan
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zhili Huang
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China
| | - Juan Song
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Wubin Bai
- Department of Applied Physical Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
13
|
Jiang X, Wu H, Xiao A, Huang Y, Yu X, Chang L. Recent Advances in Bioelectronics for Localized Drug Delivery. SMALL METHODS 2024; 8:e2301068. [PMID: 37759393 DOI: 10.1002/smtd.202301068] [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: 08/14/2023] [Revised: 09/12/2023] [Indexed: 09/29/2023]
Abstract
The last decade has witnessed remarkable advancements in bioelectronics, ushering in a new era of wearable and implantable devices for drug delivery. By utilizing miniaturized system design and/or flexible materials, bioelectronics illustrates ideal integration with target organs and tissues, making them ideal platforms for localized drug delivery. Furthermore, the development of electrically assisted drug delivery systems has enhanced the efficiency and safety of therapeutic administration, particularly for the macromolecules that encounter additional challenges in penetrating biological barriers. In this review, a concise overview of recent progress in bioelectronic devices for in vivo localized drug delivery, with highlights on the latest trends in device design, working principles, and their corresponding functionalities, is provided. The reported systems based on their targeted delivery locations as wearable systems, ingestible systems, and implantable systems are categorized. Each category is introduced in detail by highlighting the special requirements for devices and the corresponding solutions. The remaining challenges in this field and future directions are also discussed.
Collapse
Affiliation(s)
- Xinran Jiang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Han Wu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Ao Xiao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Ya Huang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, 999077, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, 999077, China
| | - Xinge Yu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, 999077, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong, 999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, 518057, China
| | - Lingqian Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| |
Collapse
|
14
|
He Y, Zang M, Zhang J, Cheng H, Cui Y, Wang D, Zhang H, Guan X, Wang S, Yuan Y, Gao Y. A universal powder-laden crosslinked chitosan microneedle patch for high-dose controllable drug delivery. Int J Biol Macromol 2024; 255:127988. [PMID: 37956809 DOI: 10.1016/j.ijbiomac.2023.127988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/23/2023] [Accepted: 11/07/2023] [Indexed: 11/15/2023]
Abstract
In this study, we constructed a novel powder-laden core-shell crosslinked chitosan microneedle patch for high-dose and controllable delivery of various drugs, including both macromolecular biological drugs and small-molecule chemical drugs. Direct loading of drug powders greatly improved drug loading capacity and minimized degradation. The results of the in vitro drug release study suggested that the release behaviors of the most tested drugs (both macromolecular drugs and small-molecule drugs) can be tuned by adjusting the crosslink density of the microneedle shell to achieve either rapid or sustained release of the loaded drug. The in vivo hypoglycemic efficacy test in streptozotocin-induced diabetic mice further proved that the onset and duration of the insulin-laden patch can be customized by adjusting the crosslink density. Furthermore, a combination of microneedle patches with different crosslink densities not only rapidly reduced blood glucose levels to normoglycemic levels (within 1 h) but also maintained normoglycemia for up to 36 h. The insulin loaded in the patch also showed good stability during storage at 40 °C for 6 months. Our results suggest that this powder-laden patch represents a strong candidate for addressing the multiple challenges in the preparation and application of polymeric microneedles and shows promise in clinical applications.
Collapse
Affiliation(s)
- Ye He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mingming Zang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jinting Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Hui Cheng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yong Cui
- Department of Biomedical Engineering, School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Da Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Haotian Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinyao Guan
- Experimental Teaching Center, Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yue Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yikun Gao
- Department of Biomedical Engineering, School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
15
|
Jiang Z, Song Z, Cao C, Yan M, Liu Z, Cheng X, Wang H, Wang Q, Liu H, Chen S. Multiple Natural Polymers in Drug and Gene Delivery Systems. Curr Med Chem 2024; 31:1691-1715. [PMID: 36927424 DOI: 10.2174/0929867330666230316094540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 03/18/2023]
Abstract
Natural polymers are organic compounds produced by living organisms. In nature, they exist in three main forms, including proteins, polysaccharides, and nucleic acids. In recent years, with the continuous research on drug and gene delivery systems, scholars have found that natural polymers have promising applications in drug and gene delivery systems due to their excellent properties such as biocompatibility, biodegradability, low immunogenicity, and easy modification. However, since the structure, physicochemical properties, pharmacological properties and biological characteristics of biopolymer molecules have not yet been entirely understood, further studies are required before large-scale clinical application. This review focuses on recent advances in the representative natural polymers such as proteins (albumin, collagen, elastin), polysaccharides (chitosan, alginate, cellulose) and nucleic acids. We introduce the characteristics of various types of natural polymers, and further outline the characterization methods and delivery forms of these natural polymers. Finally, we discuss possible challenges for natural polymers in subsequent experimental studies and clinical applications. It provides an important strategy for the clinical application of natural polymers in drug and gene delivery systems.
Collapse
Affiliation(s)
- Zhengfa Jiang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zongmian Song
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Chen Cao
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, PR China
| | - Miaoheng Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Zhendong Liu
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, PR China
| | - Xingbo Cheng
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, PR China
| | - Hongbo Wang
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou, PR China
| | - Qingnan Wang
- Department of Orthopedics, Zhengzhou University People's Hospital, Zhengzhou, 450003, PR China
- Department of Orthopedics, Henan Provincial People's Hospital, 450003, PR China
| | - Hongjian Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Songfeng Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| |
Collapse
|
16
|
Arora R, Baldi A. Revolutionizing Neurological Disorder Treatment: Integrating Innovations in Pharmaceutical Interventions and Advanced Therapeutic Technologies. Curr Pharm Des 2024; 30:1459-1471. [PMID: 38616755 DOI: 10.2174/0113816128284824240328071911] [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: 09/29/2023] [Revised: 01/31/2024] [Accepted: 02/12/2024] [Indexed: 04/16/2024]
Abstract
Neurological disorders impose a significant burden on individuals, leading to disabilities and a reduced quality of life. However, recent years have witnessed remarkable advancements in pharmaceutical interventions aimed at treating these disorders. This review article aims to provide an overview of the latest innovations and breakthroughs in neurological disorder treatment, with a specific focus on key therapeutic areas such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, epilepsy, and stroke. This review explores emerging trends in drug development, including the identification of novel therapeutic targets, the development of innovative drug delivery systems, and the application of personalized medicine approaches. Furthermore, it highlights the integration of advanced therapeutic technologies such as gene therapy, optogenetics, and neurostimulation techniques. These technologies hold promise for precise modulation of neural circuits, restoration of neuronal function, and even disease modification. While these advancements offer hopeful prospects for more effective and tailored treatments, challenges such as the need for improved diagnostic tools, identification of new targets for intervention, and optimization of drug delivery methods will remain. By addressing these challenges and continuing to invest in research and collaboration, we can revolutionize the treatment of neurological disorders and significantly enhance the lives of those affected by these conditions.
Collapse
Affiliation(s)
- Rimpi Arora
- Pharma Innovation Lab., Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda 151001, India
| | - Ashish Baldi
- Pharma Innovation Lab., Department of Pharmaceutical Sciences & Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda 151001, India
| |
Collapse
|
17
|
Salehi T, Raeisi Estabragh MA, Salarpour S, Ohadi M, Dehghannoudeh G. Absorption enhancer approach for protein delivery by various routes of administration: a rapid review. J Drug Target 2023; 31:950-961. [PMID: 37842966 DOI: 10.1080/1061186x.2023.2271680] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
As bioactive molecules, peptides and proteins are essential in living organisms, including animals and humans. Defects in their function lead to various diseases in humans. Therefore, the use of proteins in treating multiple diseases, such as cancers and hepatitis, is increasing. There are different routes to administer proteins, which have limitations due to their large and hydrophilic structure. Another limitation is the presence of biological and lipophilic membranes that do not allow proteins to pass quickly. There are different strategies to increase the absorption of proteins from these biological membranes. One of these strategies is to use compounds as absorption enhancers. Absorption enhancers are compounds such as surfactants, phospholipids and cyclodextrins that increase protein passage through the biological membrane and their absorption by different mechanisms. This review focuses on using other absorption enhancers and their mechanism in protein administration routes.
Collapse
Affiliation(s)
- Toktam Salehi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Raeisi Estabragh
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Soodeh Salarpour
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mandana Ohadi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Dehghannoudeh
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
18
|
Niloy KK, Lowe TL. Injectable systems for long-lasting insulin therapy. Adv Drug Deliv Rev 2023; 203:115121. [PMID: 37898336 DOI: 10.1016/j.addr.2023.115121] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
Insulin therapy is the mainstay to treat diabetes characterizedd by hyperglycemia. However, its short half-life of only 4-6 min limits its effectiveness in treating chronic diabetes. Advances in recombinant DNA technology and protein engineering have led to several insulin analogue products that have up to 42 h of glycemic control. However, these insulin analogues still require once- or twice-daily injections for optimal glycemic control and have poor patient compliance and adherence issues. To achieve insulin release for more than one day, different injectable delivery systems including microspheres, in situ forming depots, nanoparticles and composite systems have been developed. Several of these delivery systems have advanced to clinical trials for once-weekly insulin injection. This review comprehensively summarizes the developments of injectable insulin analogs and delivery systems covering the whole field of injectable long-lasting insulin technologies from prototype design, preclinical studies, clinical trials to marketed products for the treatment of diabetes.
Collapse
Affiliation(s)
- Kumar Kulldeep Niloy
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Tao L Lowe
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Maryland, Baltimore, MD 21201, USA; Fischell Department of Bioengineering, A. James Clark School of Engineering, University of Maryland, College Park, MD 20742, USA.
| |
Collapse
|
19
|
Wu L, Li J, Wang Y, Zhao X, He Y, Mao H, Tang W, Liu R, Luo K, Gu Z. Engineered Hierarchical Microdevices Enable Pre-Programmed Controlled Release for Postsurgical and Unresectable Cancer Treatment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305529. [PMID: 37549042 DOI: 10.1002/adma.202305529] [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: 06/09/2023] [Revised: 07/24/2023] [Indexed: 08/09/2023]
Abstract
Drug treatment is required for both resectable and unresectable cancers to strive for any meaningful improvement in patient outcomes. However, the clinical benefit of receiving conventional systemic administrations is often less than satisfactory. Drug delivery systems are preferable substitutes but still fail to meet diverse clinical demands due to the difficulty in programming drug release profiles. Herein, a microfabrication concept, termed "Hierarchical Multiple Polymers Immobilization" (HMPI), is introduced and biodegradable-polymer-based hierarchical microdevices (HMDs) that can pre-program any desired controlled release profiles are engineered. Based on the first-line medication of pancreatic and breast cancer, controlled release of single gemcitabine and the doxorubicin/paclitaxel combination in situ for multiple courses is implemented, respectively. Preclinical models of postsurgical pancreatic, postsurgical breast, and unresectable breast cancer are established, and the designed HMDs are demonstrated as well-tolerable and effective treatments for inhibiting tumor growth, recurrence, and metastasis. The proposed HMPI strategy allows the creation of tailorable and high-resolution hierarchical microstructures for pre-programming controlled release according to clinical medication schedules, which may provide promising alternative treatments for postsurgical and unresectable tumor control.
Collapse
Affiliation(s)
- Lihuang Wu
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Junhua Li
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Yuqi Wang
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Xinyue Zhao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Yiyan He
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
| | - Hongli Mao
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, 210009, China
| | - Wenbo Tang
- Faculty of Hepatopancreatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Rong Liu
- Faculty of Hepatopancreatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhongwei Gu
- Research Institute for Biomaterials, Tech Institute for Advanced Materials Bioinspired Biomedical Materials & Devices Center, College of Materials Science and Engineering, Jiangsu Collaborative Innovation Center for Advanced Inorganic Function Composites, Suqian Advanced Materials Industry Technology Innovation Center, Nanjing Tech University, Nanjing, 211816, China
- Faculty of Hepatopancreatobiliary Surgery, the First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China
- Department of Radiology, Huaxi MR Research Center (HMRRC), National Clinical Research Center for Geriatrics, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
20
|
Serna N, López-Laguna H, Aceituno P, Rojas-Peña M, Parladé E, Voltà-Durán E, Martínez-Torró C, Sánchez JM, Di Somma A, Carratalá JV, Livieri AL, Ferrer-Miralles N, Vázquez E, Unzueta U, Roher N, Villaverde A. Efficient Delivery of Antimicrobial Peptides in an Innovative, Slow-Release Pharmacological Formulation. Pharmaceutics 2023; 15:2632. [PMID: 38004610 PMCID: PMC10674355 DOI: 10.3390/pharmaceutics15112632] [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: 10/23/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Both nanostructure and multivalency enhance the biological activities of antimicrobial peptides (AMPs), whose mechanism of action is cooperative. In addition, the efficacy of a particular AMP should benefit from a steady concentration at the local place of action and, therefore, from a slow release after a dynamic repository. In the context of emerging multi-resistant bacterial infections and the urgent need for novel and effective antimicrobial drugs, we tested these concepts through the engineering of four AMPs into supramolecular complexes as pharmacological entities. For that purpose, GWH1, T22, Pt5, and PaD, produced as GFP or human nidogen-based His-tagged fusion proteins, were engineered as self-assembling oligomeric nanoparticles ranging from 10 to 70 nm and further packaged into nanoparticle-leaking submicron granules. Since these materials slowly release functional nanoparticles during their time-sustained unpacking, they are suitable for use as drug depots in vivo. In this context, a particular AMP version (GWH1-NIDO-H6) was selected for in vivo validation in a zebrafish model of a complex bacterial infection. The GWH1-NIDO-H6-secreting protein granules are protective in zebrafish against infection by the multi-resistant bacterium Stenotrophomonas maltophilia, proving the potential of innovative formulations based on nanostructured and slowly released recombinant AMPs in the fight against bacterial infections.
Collapse
Affiliation(s)
- Naroa Serna
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Hèctor López-Laguna
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Patricia Aceituno
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Biologia Cel·lular, Fisiologia Animal i Immunologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Mauricio Rojas-Peña
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Biologia Cel·lular, Fisiologia Animal i Immunologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Eloi Parladé
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Eric Voltà-Durán
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Carlos Martínez-Torró
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Julieta M. Sánchez
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
- Instituto de Investigaciones Biológicas y Tecnológicas (IIBYT), (CONICET-Universidad Nacional de Córdoba), ICTA, FCEFyN, UNC. Av. Velez Sarsfield 1611, Córdoba X 5016GCA, Argentina
| | - Angela Di Somma
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
| | - Jose Vicente Carratalá
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Andrea L. Livieri
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
| | - Neus Ferrer-Miralles
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
| | - Ugutz Unzueta
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
- Biomedical Research Institute Sant Pau (IIB Sant Pau), 08041 Barcelona, Spain
| | - Nerea Roher
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
- Departament de Biologia Cel·lular, Fisiologia Animal i Immunologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina (IBB), Universitat Autònoma de Barcelona, 08193 Barcelona, Spain; (N.S.); (P.A.); (M.R.-P.); (E.P.); (E.V.-D.); (C.M.-T.); (J.M.S.); (A.D.S.); (J.V.C.); (A.L.L.); (N.F.-M.); (E.V.); (N.R.)
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, 28029 Barcelona, Spain
| |
Collapse
|
21
|
Al Fatease A, Abdelkader H. Recent Advances in Long-Acting Drug Delivery and Formulations. Pharmaceutics 2023; 15:2519. [PMID: 38004499 PMCID: PMC10674492 DOI: 10.3390/pharmaceutics15112519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/18/2023] [Indexed: 11/26/2023] Open
Abstract
Conventional immediate-release delivery systems are simple, industrially reproducible, acceptable, and easy-to-use by most patients [...].
Collapse
Affiliation(s)
| | - Hamdy Abdelkader
- Pharmaceutics Department, College of Pharmacy, King Khalid University, Abha 62223, Saudi Arabia;
| |
Collapse
|
22
|
Zhang Q, Li S, Tong R, Zhu Y. Sialylation: An alternative to designing long-acting and targeted drug delivery system. Biomed Pharmacother 2023; 166:115353. [PMID: 37611437 DOI: 10.1016/j.biopha.2023.115353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 08/25/2023] Open
Abstract
Long-acting and specific targeting are two important properties of excellent drug delivery systems. Currently, the long-acting strategies based on polyethylene glycol (PEG) are controversial, and PEGylation is incapable of simultaneously possessing targeting ability. Thus, it is crucial to identify and develop approaches to produce long-acting and targeted drug delivery systems. Sialic acid (SA) is an endogenous, negatively charged, nine-carbon monosaccharide. SA not only mediates immune escape in the body but also binds to numerous disease related targets. This suggests a potential strategy, namely "sialylation," for preparing long-acting and targeted drug delivery systems. This review focuses on the application status of SA-based long-acting and targeted agents as a reference for subsequent research.
Collapse
Affiliation(s)
- Qixiong Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Shanshan Li
- College of Pharmacy, Southwest Minzu University, Chengdu 610041, China
| | - Rongsheng Tong
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yuxuan Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China.
| |
Collapse
|
23
|
Pandya AK, Vora LK, Umeyor C, Surve D, Patel A, Biswas S, Patel K, Patravale VB. Polymeric in situ forming depots for long-acting drug delivery systems. Adv Drug Deliv Rev 2023; 200:115003. [PMID: 37422267 DOI: 10.1016/j.addr.2023.115003] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/10/2023]
Abstract
Polymeric in situ forming depots have emerged as highly promising drug delivery systems for long-acting applications. Their effectiveness is attributed to essential characteristics such as biocompatibility, biodegradability, and the ability to form a stable gel or solid upon injection. Moreover, they provide added versatility by complementing existing polymeric drug delivery systems like micro- and nanoparticles. The formulation's low viscosity facilitates manufacturing unit operations and enhances delivery efficiency, as it can be easily administered via hypodermic needles. The release mechanism of drugs from these systems can be predetermined using various functional polymers. To enable unique depot design, numerous strategies involving physiological and chemical stimuli have been explored. Important assessment criteria for in situ forming depots include biocompatibility, gel strength and syringeability, texture, biodegradation, release profile, and sterility. This review focuses on the fabrication approaches, key evaluation parameters, and pharmaceutical applications of in situ forming depots, considering perspectives from academia and industry. Additionally, insights about the future prospects of this technology are discussed.
Collapse
Affiliation(s)
- Anjali K Pandya
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400 019, India; School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Chukwuebuka Umeyor
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400 019, India; Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Nnamdi Azikiwe University, Awka 422001, Anambra State, Nigeria
| | - Dhanashree Surve
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA
| | - Akanksha Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Swati Biswas
- Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad, Telangana 500078, India
| | - Ketankumar Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Vandana B Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400 019, India.
| |
Collapse
|
24
|
Tao X, Liu Y, Ding Z, Xie S, Cao W, Li X. Injectable cell-targeting fiber rods to promote lipolysis and regulate inflammation for obesity treatment. Biomater Sci 2023; 11:5663-5673. [PMID: 37432672 DOI: 10.1039/d3bm00619k] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2023]
Abstract
Obesity has become a worldwide public health problem and continues to be one of the leading causes of chronic diseases. Obesity treatment is challenged by large drug doses, high administration frequencies and severe side effects. Herein, we propose an antiobesity strategy through the local administration of HaRChr fiber rods loaded with chrysin and grafted with hyaluronic acid and AtsFRk fiber fragments loaded with raspberry ketone and grafted with adipocyte target sequences (ATSs). The hyaluronic acid grafts double the uptake levels of HaRChr by M1 macrophages to promote phenotype transformation from M1 to M2 through upregulating CD206 and downregulating CD86 expressions. ATS-mediated targeting and sustained release of raspberry ketone from AtsFRk increase the secretion of glycerol and adiponectin, and Oil red O staining shows much fewer lipid droplets in adipocytes. The combination treatment with AtsFRk and the conditioned media from HaRChr-treated macrophages elevates adiponectin levels, suggesting that M2 macrophages may secrete anti-inflammatory factors to stimulate adipocytes to produce adiponectin. Diet-induced obese mice showed significant weight losses of inguinal (49.7%) and epididymal (32.5%) adipose tissues after HaRChr/AtsFRk treatment, but no effect was observed on food intake. HaRChr/AtsFRk treatment reduces adipocyte volumes, lowers serum levels of triglycerides and total cholesterol and restores adiponectin levels to those of normal mice. In the meantime, HaRChr/AtsFRk treatment significantly elevates the gene expression of adiponectin and interleukin-10 and downregulates tissue necrosis factor-α expression in the inguinal adipose tissues. Thus, local injection of cell-targeting fiber rods and fragments demonstrates a feasible and effective antiobesity strategy through improving lipid metabolism and normalizing the inflammatory microenvironment.
Collapse
Affiliation(s)
- Xinyan Tao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China.
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Yuan Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Zhenhua Ding
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Shuang Xie
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China.
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Wenxiong Cao
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China.
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Xiaohong Li
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, P.R. China.
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China
| |
Collapse
|
25
|
Herdiana Y, Sriwidodo S, Sofian FF, Wilar G, Diantini A. Nanoparticle-Based Antioxidants in Stress Signaling and Programmed Cell Death in Breast Cancer Treatment. Molecules 2023; 28:5305. [PMID: 37513179 PMCID: PMC10384004 DOI: 10.3390/molecules28145305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer (BC) is a complex and heterogeneous disease, and oxidative stress is a hallmark of BC. Oxidative stress is characterized by an imbalance between the production of reactive oxygen species (ROS) and antioxidant defense mechanisms. ROS has been implicated in BC development and progression by inducing DNA damage, inflammation, and angiogenesis. Antioxidants have been shown to scavenge ROS and protect cells from oxidative damage, thereby regulating signaling pathways involved in cell growth, survival, and death. Plants contain antioxidants like ascorbic acid, tocopherols, carotenoids, and flavonoids, which have been found to regulate stress signaling and PCD in BC. Combining different antioxidants has shown promise in enhancing the effectiveness of BC treatment. Antioxidant nanoparticles, when loaded with antioxidants, can effectively target breast cancer cells and enhance their cellular uptake. Notably, these nanoparticles have shown promising results in inducing PCD and sensitizing breast cancer cells to chemotherapy, even in cases where resistance is observed. This review aims to explore how nanotechnology can modulate stress signaling and PCD in breast cancer. By summarizing current research, it underscores the potential of nanotechnology in enhancing antioxidant properties for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Sriwidodo Sriwidodo
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Ferry Ferdiansyah Sofian
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Ajeng Diantini
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| |
Collapse
|
26
|
Rahmani F, Naderpour S, Nejad BG, Rahimzadegan M, Ebrahimi ZN, Kamali H, Nosrati R. The recent insight in the release of anticancer drug loaded into PLGA microspheres. Med Oncol 2023; 40:229. [PMID: 37410278 DOI: 10.1007/s12032-023-02103-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 06/21/2023] [Indexed: 07/07/2023]
Abstract
Cancer is a series of diseases leading to a high rate of death worldwide. Microspheres display specific characteristics that make them appropriate for a variety of biomedical purposes such as cancer therapy. Newly, microspheres have the potentials to be used as controlled drug release carriers. Recently, PLGA-based microspheres have attracted exceptional attention relating to effective drug delivery systems (DDS) because of their distinctive properties for a simple preparation, biodegradability, and high capability of drug loading which might be increased drug delivery. In this line, the mechanisms of controlled drug release and parameters that influence the release features of loaded agents from PLGA-based microspheres should be mentioned. The current review is focused on the new development of the release features of anticancer drugs, which are loaded into PLGA-based microspheres. Consequently, future perspective and challenges of anticancer drug release from PLGA-based microspheres are mentioned concisely.
Collapse
Affiliation(s)
- Farzad Rahmani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saghi Naderpour
- Faculty of Pharmacy, Eastern Mediterranean University, Famagusta, Cyprus
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behnam Ghorbani Nejad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Milad Rahimzadegan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zivar Nejad Ebrahimi
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Hossein Kamali
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Rahim Nosrati
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
27
|
Ren S, Liu C, Sun Y, Zhang Y, Ruan J, Fang L. Formulation Development and Molecular Mechanism Characterization of Long-Acting Patches of Asenapine for Efficient Delivery by Combining API-ILs Strategy and Controlled-Release Polymers. J Pharm Sci 2023; 112:1850-1862. [PMID: 36858176 DOI: 10.1016/j.xphs.2023.02.002] [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: 09/27/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 03/02/2023]
Abstract
The objective of our study, which combined API-ILs strategy and controlled-release polymers, was to prepare a 72 h long-acting drug-in-adhesive patch for optimum delivery of asenapine (ASE). Special attention was paid to the permeation promotion mechanism and the controlled release behavior of ASE-ILs in pressure sensitive adhesives (PSA). Formulation factors were investigated by ex vivo transdermal experiments. The optimized patch was evaluated by pharmacokinetics study and skin irritation test. The obtained formulation was as follows, 15% w/w ASE-MA (about 1136 μg/cm2 ASE, 413 μg/cm2 MA), AACONH2 (Amide adhesive) as the matrix, 80 μm thickness, backing film of CoTran™ 9733. The optimized patch displayed satisfactory ex vivo and in vivo performance with Q 72 h of 620 ± 44 µg/cm2 and Fabs of 62.4%, which utilization rate (54.6%) was significantly higher than the control group (38.3%). By using the classical shake flask method, 13C NMR, DSC, and FTIR, the physicochemical properties and structure of ILs were characterized. log Do/w, ATR-FTIR, Raman, and molecular dynamics simulation results confirmed that ASE-MA (MA: 3-Methoxypropionic acid) had appropriate lipophilicity, and affected lipid fluidity as well as the conformation of keratin to improve the skin permeation. The FTIR, MDSC, rheology, and molecular docking results revealed that hydrogen bond (H-bond), were formed between ASE-MA and PSA, and the drug increased the molecular mobility of polymer chains. In summary, the 72 h long-acting patch of ASE was successfully prepared and it supplied a reference for the design of long-acting patches with ASE.
Collapse
Affiliation(s)
- Shoujun Ren
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| | - Chao Liu
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| | - Yutong Sun
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| | - Yang Zhang
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| | - Jiuheng Ruan
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China
| | - Liang Fang
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, China.
| |
Collapse
|
28
|
Xu D, Wan Y, Xie Z, Du C, Wang Y. Hierarchically Structured Hydroxyapatite Particles Facilitate the Enhanced Integration and Selective Anti-Tumor Effects of Amphiphilic Prodrug for Osteosarcoma Therapy. Adv Healthc Mater 2023; 12:e2202668. [PMID: 36857811 DOI: 10.1002/adhm.202202668] [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: 10/16/2022] [Revised: 02/22/2023] [Indexed: 03/03/2023]
Abstract
Efficient delivery of cargo into target cells is a formidable challenge in modern medicine. Despite the great promise of biomimetic hydroxyapatite (HA) particles in tissue engineering, their potential applications in bone tumor therapy, particularly their structure-function relationships in cargo delivery to target cells, have not yet been well explored. In this study, biomimetic multifunctional composite microparticles (Bm-cMPs) are developed by integrating an amphiphilic prodrug of curcumin with hierarchically structured HA microspheres (Hs-hMPs). Then, the effects of the hierarchical structure of vehicles on the integration and delivery of cargo as well as the anti-osteosarcoma (OS) effect of the composite are determined. Different hierarchical structures of the vehicles strongly influence the self-assembly behavior of the prodrug. The flake-like crystals of Hs-hMPs enable the highest loading capacity and enhance the stability of the cargo. Compared to the normal cells, OS cells exhibit 3.56-times better uptake of flake-like Hs-hMPs, facilitating the selective anti-tumor effect of the prodrug. Moreover, Bm-cMPs suppress tumor growth and metastasis by promoting apoptosis and inhibiting cell proliferation and tumor vascularization. The findings shed light on the potential application of Bm-cMPs and suggest a feasible strategy for developing an effective targeted therapy platform using hierarchically structured minerals for OS treatment.
Collapse
Affiliation(s)
- Dong Xu
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yuxin Wan
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Zhenze Xie
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Chang Du
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, and Innovation Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yingjun Wang
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Key Laboratory of Biomedical Engineering of Guangdong Province, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
29
|
Al-Qaysi ZK, Beadham IG, Schwikkard SL, Bear JC, Al-Kinani AA, Alany RG. Sustained release ocular drug delivery systems for glaucoma therapy. Expert Opin Drug Deliv 2023; 20:905-919. [PMID: 37249548 DOI: 10.1080/17425247.2023.2219053] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 05/24/2023] [Indexed: 05/31/2023]
Abstract
INTRODUCTION Glaucoma is a group of progressive optic neuropathies resulting in irreversible blindness. It is associated with an elevation of intraocular pressure (>21 mm Hg) and optic nerve damage. Reduction of the intraocular pressure (IOP) through the administration of ocular hypotensive eye drops is one of the most common therapeutic strategies. Patient adherence to conventional eye drops remains a major obstacle in preventing glaucoma progression. Additional problems emerge from inadequate patient education as well as local and systemic side effects associated with adminstering ocular hypotensive drugs. AREAS COVERED Sustained-release drug delivery systems for glaucoma treatment are classified into extraocular systems including wearable ocular surface devices or multi-use (immediate-release) eye formulations (such as aqueous solutions, gels; ocular inserts, contact lenses, periocular rings, or punctual plugs) and intraocular drug delivery systems (such as intraocular implants, and microspheres for supraciliary drug delivery). EXPERT OPINION Sustained release platforms for the delivery of ocular hypotensive drugs (small molecules and biologics) may improve patient adherence and prevent vision loss. Such innovations will only be widely adopted when efficacy and safety has been established through large-scale trials. Sustained release drug delivery can improve glaucoma treatment adherence and reverse/prevent vision deterioration. It is expected that these approaches will improve clinical management and prognosis of glaucoma.
Collapse
Affiliation(s)
- Zinah K Al-Qaysi
- Drug Discovery, Delivery and Patient Care (DDDPC) Theme, Department of Pharmacy, Kingston University London, Kingston Upon Thames, UK
| | - Ian G Beadham
- Drug Discovery, Delivery and Patient Care (DDDPC) Theme, Department of Pharmacy, Kingston University London, Kingston Upon Thames, UK
| | - Sianne L Schwikkard
- Department of Chemical and Pharmaceutical Sciences, Kingston University, Kingston Upon Thames, UK
| | - Joseph C Bear
- Department of Chemical and Pharmaceutical Sciences, Kingston University, Kingston Upon Thames, UK
| | - Ali A Al-Kinani
- Drug Discovery, Delivery and Patient Care (DDDPC) Theme, Department of Pharmacy, Kingston University London, Kingston Upon Thames, UK
| | - Raid G Alany
- Drug Discovery, Delivery and Patient Care (DDDPC) Theme, Department of Pharmacy, Kingston University London, Kingston Upon Thames, UK
- School of Pharmacy, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
30
|
Fathalla Z, Fatease AA, Abdelkader H. Formulation and In-Vitro/Ex-Vivo Characterization of Pregelled Hybrid Alginate-Chitosan Microparticles for Ocular Delivery of Ketorolac Tromethamine. Polymers (Basel) 2023; 15:2773. [PMID: 37447419 DOI: 10.3390/polym15132773] [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: 05/20/2023] [Revised: 06/13/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Innovative hybrid chitosan-sodium alginate (Ch-Ag) microparticles (MPs) were fabricated using both the ionic gelation method as well as the pre-gelation technique. The hybrid Ch-Ag MPs were studied for size, zeta potential, morphology, mucoadhesion, in-vitro release, corneal permeation, and ocular irritation using lens and corneal epithelial cell lines. The average particle size ranged from 1322 nm to 396 nm. The zeta potential for the prepared formulations showed an increase with increasing Ch concentrations up to a value of >35 mV; the polydispersity index (PDI) of some optimized MPs was around 0.1. Compared to drug-free MPs, ketorolac-loaded Ch-Ag MPs demonstrated a drug proportion-dependent increase in their size. SEM, as well as TEM of KT-loaded MPs, confirmed that the formed particles were quasi-spherical to elliptical in shape. The KT release from the MPs demonstrated a prolonged release profile in comparison to the control KT solution. Further, mucoadhesion studies with porcine mucin revealed that the KT-loaded MPs had effective mucoadhesive properties, and polymeric particles were stable in the presence of mucin. Corneal permeation was studied on bovine eyes, and the results revealed that Ch-based MPs were capable of showing more sustained KT release across the cornea compared with that for the control drug solution. Conclusively, the cytotoxicity assay confirmed that the investigated MPs were non-irritant and could confer protection from direct drug irritation of KT on the ocular surface. The MTT cytotoxicity assay confirmed that KT-loaded MPs showed acceptable and reasonable tolerability with both human lens and corneal epithelial cell lines compared to the control samples.
Collapse
Affiliation(s)
- Zeinab Fathalla
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia 61519, Egypt
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62223, Saudi Arabia
| | - Hamdy Abdelkader
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62223, Saudi Arabia
| |
Collapse
|
31
|
Mostafa M, Al Fatease A, Alany RG, Abdelkader H. Recent Advances of Ocular Drug Delivery Systems: Prominence of Ocular Implants for Chronic Eye Diseases. Pharmaceutics 2023; 15:1746. [PMID: 37376194 PMCID: PMC10302848 DOI: 10.3390/pharmaceutics15061746] [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: 05/16/2023] [Revised: 06/02/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Chronic ocular diseases can seriously impact the eyes and could potentially result in blindness or serious vision loss. According to the most recent data from the WHO, there are more than 2 billion visually impaired people in the world. Therefore, it is pivotal to develop more sophisticated, long-acting drug delivery systems/devices to treat chronic eye conditions. This review covers several drug delivery nanocarriers that can control chronic eye disorders non-invasively. However, most of the developed nanocarriers are still in preclinical or clinical stages. Long-acting drug delivery systems, such as inserts and implants, constitute the majority of the clinically used methods for the treatment of chronic eye diseases due to their steady state release, persistent therapeutic activity, and ability to bypass most ocular barriers. However, implants are considered invasive drug delivery technologies, especially those that are nonbiodegradable. Furthermore, in vitro characterization approaches, although useful, are limited in mimicking or truly representing the in vivo environment. This review focuses on long-acting drug delivery systems (LADDS), particularly implantable drug delivery systems (IDDS), their formulation, methods of characterization, and clinical application for the treatment of eye diseases.
Collapse
Affiliation(s)
- Mahmoud Mostafa
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minya 61519, Egypt;
| | - Adel Al Fatease
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62223, Saudi Arabia;
| | - Raid G. Alany
- School of Pharmacy, Kingston University London, Kingston Upon Tames KT1 2EE, UK;
- School of Pharmacy, The University of Auckland, Auckland 1010, New Zealand
| | - Hamdy Abdelkader
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 62223, Saudi Arabia;
| |
Collapse
|
32
|
Wu D, Chen Q, Chen X, Han F, Chen Z, Wang Y. The blood-brain barrier: structure, regulation, and drug delivery. Signal Transduct Target Ther 2023; 8:217. [PMID: 37231000 PMCID: PMC10212980 DOI: 10.1038/s41392-023-01481-w] [Citation(s) in RCA: 201] [Impact Index Per Article: 201.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/27/2023] Open
Abstract
Blood-brain barrier (BBB) is a natural protective membrane that prevents central nervous system (CNS) from toxins and pathogens in blood. However, the presence of BBB complicates the pharmacotherapy for CNS disorders as the most chemical drugs and biopharmaceuticals have been impeded to enter the brain. Insufficient drug delivery into the brain leads to low therapeutic efficacy as well as aggravated side effects due to the accumulation in other organs and tissues. Recent breakthrough in materials science and nanotechnology provides a library of advanced materials with customized structure and property serving as a powerful toolkit for targeted drug delivery. In-depth research in the field of anatomical and pathological study on brain and BBB further facilitates the development of brain-targeted strategies for enhanced BBB crossing. In this review, the physiological structure and different cells contributing to this barrier are summarized. Various emerging strategies for permeability regulation and BBB crossing including passive transcytosis, intranasal administration, ligands conjugation, membrane coating, stimuli-triggered BBB disruption, and other strategies to overcome BBB obstacle are highlighted. Versatile drug delivery systems ranging from organic, inorganic, and biologics-derived materials with their synthesis procedures and unique physio-chemical properties are summarized and analyzed. This review aims to provide an up-to-date and comprehensive guideline for researchers in diverse fields, offering perspectives on further development of brain-targeted drug delivery system.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang Chinese Medical University, 310053, Hangzhou, China.
| | - Qi Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, 310053, Hangzhou, China.
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang Chinese Medical University, 310053, Hangzhou, China.
| |
Collapse
|
33
|
Ismail A, Sial N, Rehman R, Abid S, Ismail MS. Survival, growth, behavior, hematology and serum biochemistry of mice under different concentrations of orally administered amorphous silica nanoparticle. Toxicol Rep 2023; 10:659-668. [PMID: 37274627 PMCID: PMC10238806 DOI: 10.1016/j.toxrep.2023.05.006] [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: 03/16/2023] [Revised: 05/11/2023] [Accepted: 05/13/2023] [Indexed: 06/06/2023] Open
Abstract
Silica nanoparticles (SiNPs) are used extensively in consumer products and biomedical research basically due to ease of production and low cost. However, insufficient literature is reported regarding the toxicity and biocompatibility of SiNPs. The present study aimed to investigate the potential role of amorphous SiNPs on survival, growth, behavioral alterations, hematology and serum biochemistry of mice at four concentrations (control, 50, 100 and 150 mg/kg/day) of an oral supplementation for a period of 3 months. Signs of toxicity (lethargy, nausea, coma, tremors, vomiting and diarrhea, etc.) were noted at 9:00 am and 9:00 pm (twice a day) and the body weight of each of these mice was measured every week. The data were subjected to mean, standard deviation (S.D). Moreover, One-Way Analysis of Variance (ANOVA) and Dunnett's test were applied for analysis of statistical significance between groups by using SPSS software, version 20. All the mice survived with minor alterations in behavior and no significant weight changes were observed during the stipulated time period. Complete blood count (CBC) analysis indicated non-significant (P ≥ 0.05) systemic dysfunctions of organ systems. However, there was elevation in the level of AST and ALT in the analysis of serum biochemistry, while the values of all other examined parameters were not-significant (P ≥ 0.05). The study concluded that orally administered large silica nanoparticles up to the dose level of 150 mg/kg/day are nontoxic for the in vivo use in mice.
Collapse
Affiliation(s)
- Amna Ismail
- Department of Zoology, The Islamia University of Bahawalpur, Pakistan
| | - Nuzhat Sial
- Department of Zoology, The Islamia University of Bahawalpur, Pakistan
| | - Rakhshanda Rehman
- Department of Zoology, The Islamia University of Bahawalpur, Pakistan
| | - Sobia Abid
- Department of Zoology, The Islamia University of Bahawalpur, Pakistan
| | - Muhammad Shoaib Ismail
- Department of Agronomy, Muhammad Nawaz Shareef University of Agriculture Multan, Pakistan
| |
Collapse
|
34
|
Jindal AB, Bhide AR, Salave S, Rana D, Benival D. Long-acting Parenteral Drug Delivery Systems for the Treatment of Chronic Diseases. Adv Drug Deliv Rev 2023; 198:114862. [PMID: 37160247 DOI: 10.1016/j.addr.2023.114862] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/12/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
The management of chronic conditions often requires patients to take daily medication for an extended duration. However, the need for daily dosing can lead to nonadherence to the therapy, which can result in the recurrence of the disease. Long-acting parenteral drug delivery systems have the potential to improve the treatment of chronic conditions. These systems use various technologies, such as oil-based injectables, PLGA-based microspheres, and in situ forming gel-based depots, to deliver different types of drugs. The use of long-acting parenteral formulations for the treatment of chronic infections such as HIV/AIDS and tuberculosis is a recent development in the field. Researchers are also exploring the use of long-acting parenteral formulations for the treatment of malaria, with the aim of reducing dosing frequency and improving adherence to treatment. This review discusses various aspects of long-acting formulation development, including the impact of the physicochemical properties of the drug, the type of long-acting formulation, and the route of administration. The clinical significance of long-acting formulations and recent advances in the field, such as long-acting nanoformulations and long-acting products currently in clinical trials, have also been highlighted.
Collapse
Affiliation(s)
- Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Jhunjhunu, Rajasthan - 333031, India.
| | - Atharva R Bhide
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Jhunjhunu, Rajasthan - 333031, India
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research - Ahmedabad (NIPER-A) An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar - 382355, Gujarat, India
| | - Dhwani Rana
- National Institute of Pharmaceutical Education and Research - Ahmedabad (NIPER-A) An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar - 382355, Gujarat, India
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research - Ahmedabad (NIPER-A) An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Opp. Airforce Station, Palaj, Gandhinagar - 382355, Gujarat, India
| |
Collapse
|
35
|
Talebian S, Mendes B, Conniot J, Farajikhah S, Dehghani F, Li Z, Bitoque D, Silva G, Naficy S, Conde J, Wallace GG. Biopolymeric Coatings for Local Release of Therapeutics from Biomedical Implants. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207603. [PMID: 36782094 PMCID: PMC10131825 DOI: 10.1002/advs.202207603] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Indexed: 06/18/2023]
Abstract
The deployment of structures that enable localized release of bioactive molecules can result in more efficacious treatment of disease and better integration of implantable bionic devices. The strategic design of a biopolymeric coating can be used to engineer the optimal release profile depending on the task at hand. As illustrative examples, here advances in delivery of drugs from bone, brain, ocular, and cardiovascular implants are reviewed. These areas are focused to highlight that both hard and soft tissue implants can benefit from controlled localized delivery. The composition of biopolymers used to achieve appropriate delivery to the selected tissue types, and their corresponding outcomes are brought to the fore. To conclude, key factors in designing drug-loaded biopolymeric coatings for biomedical implants are highlighted.
Collapse
Affiliation(s)
- Sepehr Talebian
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Bárbara Mendes
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - João Conniot
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Syamak Farajikhah
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Fariba Dehghani
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - Zhongyan Li
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
| | - Diogo Bitoque
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Gabriela Silva
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Sina Naficy
- School of Chemical and Biomolecular EngineeringThe University of SydneySydneyNSW2006Australia
- Nano Institute (Sydney Nano)The University of SydneySydneyNSW2006Australia
| | - João Conde
- ToxOmicsNOVA Medical School|Faculdade de Ciências MédicasNMS|FCMUniversidade Nova de LisboaLisboa1169‐056Portugal
| | - Gordon G. Wallace
- Intelligent Polymer Research InstituteARC Centre of Excellence for Electromaterials ScienceAIIM FacilityUniversity of WollongongSydneyNSW2522Australia
| |
Collapse
|
36
|
Woodring RN, Gurysh EG, Bachelder EM, Ainslie KM. Drug Delivery Systems for Localized Cancer Combination Therapy. ACS APPLIED BIO MATERIALS 2023; 6:934-950. [PMID: 36791273 PMCID: PMC10373430 DOI: 10.1021/acsabm.2c00973] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
With over 2 million cancer cases and over 600,000 cancer-associated deaths predicted in the U.S. for 2022, this life-debilitating disease continuously impacts the lives of people across the nation every day. Therapeutic treatment options for cancer have historically involved chemotherapies to eradicate tumors with cytotoxic mechanisms which can negatively affect the efficacy versus toxicity ratio of treatment. With a need for more directed and therapeutically active options, targeted small-molecule inhibitors and immunotherapies have since emerged to mitigate treatment-associated toxicities. However, aggressive tumors can employ a wide range of defense mechanisms to evade monotherapy treatment altogether, resulting in the recurrence of therapeutically resistant tumors. Therefore, many clinical routines have included combination therapy in which anticancer agents are combined to provide a synergistic attack on tumors. Even with this approach, maximizing the efficacy of cancer treatment is contingent upon the dose of drug that reaches the site of the tumor, so often therapy is administered at the site of a tumor via localized delivery platforms. Commonly used platforms for localized drug delivery include polymeric wafers, nanofibrous scaffolds, and hydrogels where drug combinations can be loaded and delivered synchronously. Attaining synergistic activity from these localized systems is dependent on proper material selection and fabrication methods. Herein, we describe these important considerations for enhancing the efficacy of cancer combination therapy through biodegradable, localized delivery systems.
Collapse
Affiliation(s)
- Ryan N. Woodring
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Elizabeth G. Gurysh
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
37
|
Simón JA, Utomo E, Pareja F, Collantes M, Quincoces G, Otero A, Ecay M, Domínguez-Robles J, Larrañeta E, Peñuelas I. Radiolabeled Risperidone microSPECT/CT Imaging for Intranasal Implant Studies Development. Pharmaceutics 2023; 15:pharmaceutics15030843. [PMID: 36986704 PMCID: PMC10054269 DOI: 10.3390/pharmaceutics15030843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
The use of intranasal implantable drug delivery systems has many potential advantages for the treatment of different diseases, as they can provide sustained drug delivery, improving patient compliance. We describe a novel proof-of-concept methodological study using intranasal implants with radiolabeled risperidone (RISP) as a model molecule. This novel approach could provide very valuable data for the design and optimization of intranasal implants for sustained drug delivery. RISP was radiolabeled with 125I by solid supported direct halogen electrophilic substitution and added to a poly(lactide-co-glycolide) (PLGA; 75/25 D,L-Lactide/glycolide ratio) solution that was casted on top of 3D-printed silicone molds adapted for intranasal administration to laboratory animals. Implants were intranasally administered to rats, and radiolabeled RISP release followed for 4 weeks by in vivo non-invasive quantitative microSPECT/CT imaging. Percentage release data were compared with in vitro ones using radiolabeled implants containing either 125I-RISP or [125I]INa and also by HPLC measurement of drug release. Implants remained in the nasal cavity for up to a month and were slowly and steadily dissolved. All methods showed a fast release of the lipophilic drug in the first days with a steadier increase to reach a plateau after approximately 5 days. The release of [125I]I− took place at a much slower rate. We herein demonstrate the feasibility of this experimental approach to obtain high-resolution, non-invasive quantitative images of the release of the radiolabeled drug, providing valuable information for improved pharmaceutical development of intranasal implants.
Collapse
Affiliation(s)
- Jon Ander Simón
- Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, University of Navarra, IdiSNA, 31008 Pamplona, Spain
| | - Emilia Utomo
- School of Pharmacy, Queen’s University Belfast, Lisburn Road 97, Belfast BT9 7BL, UK
| | - Félix Pareja
- Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, University of Navarra, IdiSNA, 31008 Pamplona, Spain
| | - María Collantes
- Translational Molecular Imaging Unit (UNIMTRA), Department of Nuclear Medicine, Clinica Universidad de Navarra, 31008 Pamplona, Spain
- Correspondence: (M.C.); (E.L.)
| | - Gemma Quincoces
- Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, University of Navarra, IdiSNA, 31008 Pamplona, Spain
| | - Aarón Otero
- Translational Molecular Imaging Unit (UNIMTRA), Department of Nuclear Medicine, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Margarita Ecay
- Translational Molecular Imaging Unit (UNIMTRA), Department of Nuclear Medicine, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| | - Juan Domínguez-Robles
- School of Pharmacy, Queen’s University Belfast, Lisburn Road 97, Belfast BT9 7BL, UK
- Department of Pharmacy and Pharmaceutical Technology, University of Seville, 41012 Seville, Spain
| | - Eneko Larrañeta
- School of Pharmacy, Queen’s University Belfast, Lisburn Road 97, Belfast BT9 7BL, UK
- Correspondence: (M.C.); (E.L.)
| | - Iván Peñuelas
- Radiopharmacy Unit, Department of Nuclear Medicine, Clinica Universidad de Navarra, University of Navarra, IdiSNA, 31008 Pamplona, Spain
- Translational Molecular Imaging Unit (UNIMTRA), Department of Nuclear Medicine, Clinica Universidad de Navarra, 31008 Pamplona, Spain
| |
Collapse
|
38
|
Pacheco C, Baião A, Ding T, Cui W, Sarmento B. Recent advances in long-acting drug delivery systems for anticancer drug. Adv Drug Deliv Rev 2023; 194:114724. [PMID: 36746307 DOI: 10.1016/j.addr.2023.114724] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/20/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The use of systemic anticancer chemotherapy is intrinsically limited by its toxicity. Whether dealing with small molecules or biopharmaceuticals, after systemic administration, small doses fail to reach effective intratumoral concentrations, while high doses with significant tumor inhibition effects may also drive the death of healthy cells, endangering the patients. Therefore, strategies based on drug delivery systems (DDSs) for avoiding the systemic toxicity have been designed. Due to their ability to protect drugs from early elimination and control drug release, DDSs can foster tumor exposure to anticancer therapeutics by extending their circulation time or steadily releasing drugs into the tumor sites. However, approval of tailored DDSs systems for clinical use is minimal as the safety and the in vivo activity still need to be ameliorated by manipulating their physicochemical characteristics. During the last few years, several strategies have been described to improve their safety, stability, and fine-tune pharmaceuticals release kinetics. Herein, we reviewed the main DDSs, namely polymeric conjugates, nano or microparticles, hydrogels, and microneedles, explored for long-acting anticancer treatments, highlighting recently proposed modifications and their potential advantages for different anticancer therapies. Additionally, important limitations of long-acting anticancer therapies and future technology directions were also covered.
Collapse
Affiliation(s)
- Catarina Pacheco
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana Baião
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Tao Ding
- 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, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Wenguo Cui
- 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, 197 Ruijin 2nd Road, Shanghai 200025, China
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal; IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, 4585-116 Gandra, Portugal; 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, 197 Ruijin 2nd Road, Shanghai 200025, China.
| |
Collapse
|
39
|
Ghate V, Renjith A, Badnikar K, Pahal S, Jayadevi SN, Nayak MM, Vemula PK, Subramanyam DN. Single step fabrication of hollow microneedles and an experimental package for controlled drug delivery. Int J Pharm 2023; 632:122546. [PMID: 36574913 DOI: 10.1016/j.ijpharm.2022.122546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/26/2022]
Abstract
Hollow microneedle arrays (HMNs) are an excellent choice for managing chronic diseases requiring the administration of multiple drug doses over a prolonged duration. However, HMNs have gained partial success due to limitations in their manufacturing capabilities, and cumbersome processes. In the present study, polymeric HMNs were fabricated using a novel single-step drop-casting process without needing cleanroom facilities, and sophisticated instrumentation. When drop casted on the pyramidal tip stainless steel needles, the optimized polymer solution allowed the reproducible formation of desired height HMMs on a detachable acrylic base. To enable broader applications, the base with HMNs was integrated into an experimental package built to deliver a dose of ∼ 5 µL per 30° clockwise rotation of the actuator, allowing multiple metered drug dose administrations. The fabricated HMNs were optically imaged, and tested for mechanical integrity and stability. The working and functional utility of the HMNs package in delivering metered drug doses was demonstrated by delivering vitamin B12 (ex vivo) and insulin (in vivo), respectively. The optimized process can be used for the large-scale manufacturing of HMNs and the experimental package shows the potential to be further developed into a wearable device.
Collapse
Affiliation(s)
- Vivek Ghate
- Department of Electronic Systems Engineering, Indian Institute of Science, Bengaluru, Karnataka 560012, India.
| | - Anu Renjith
- Department of Electronic Systems Engineering, Indian Institute of Science, Bengaluru, Karnataka 560012, India.
| | - Kedar Badnikar
- Department of Electronic Systems Engineering, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Suman Pahal
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, Karnataka 560065, India
| | - Shreyas N Jayadevi
- Department of Electronic Systems Engineering, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Manjunatha M Nayak
- Centre for Nano Science and Engineering (CeNSE), Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Praveen K Vemula
- Institute for Stem Cell Science and Regenerative Medicine (inStem), Bengaluru, Karnataka 560065, India.
| | - Dinesh N Subramanyam
- Department of Electronic Systems Engineering, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| |
Collapse
|
40
|
Self-intensified synergy of a versatile biomimetic nanozyme and doxorubicin on electrospun fibers to inhibit postsurgical tumor recurrence and metastasis. Biomaterials 2023; 293:121942. [PMID: 36512863 DOI: 10.1016/j.biomaterials.2022.121942] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/26/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
Tumor-positive resection margins after surgery can result in tumor recurrence and metastasis. Although adjuvant postoperative radiotherapy and chemotherapy have been adopted in clinical practice, they lack efficacy and result in unavoidable side effects. Herein, a self-intensified in-situ therapy approach using electrospun fibers loaded with a biomimetic nanozyme and doxorubicin (DOX) is developed. The fabricated PEG-coated zeolite imidazole framework-67 (PZIF67) is demonstrated as a versatile nanozyme triggering reactions in cancer cells based on endogenous H2O2 and •O2-. The PZIF67-generated •OH induces reactive oxygen species (ROS) overload, implementing chemodynamic therapy (CDT). The O2 produced by PZIF67 inhibits the expression of hypoxia-up-regulated proteins, thereby suppressing tumor progression. PZIF67 also catalyzes the degradation of glutathione, further disturbing the intracellular redox homeostasis and enhancing CDT. Furthermore, the introduced DOX not only kills cancer cells individually, but also replenishes the continuously consumed substrates for PZIF67-catalyzed reactions. The PZIF67-weakened drug resistance strengthens the cytotoxicity of DOX. The combined application of PZIF67 and DOX also suppresses metastasis-associated genes. Both in vitro and in vivo results demonstrate that the self-intensified synergy of PZIF67 and DOX on electrospun fibers efficiently prevents postsurgical tumor recurrence and metastasis, offering a feasible therapeutic regimen for operable malignant tumors.
Collapse
|
41
|
Guller A, Igrunkova A. Engineered Microenvironments for 3D Cell Culture and Regenerative Medicine: Challenges, Advances, and Trends. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 10:bioengineering10010017. [PMID: 36671589 PMCID: PMC9854955 DOI: 10.3390/bioengineering10010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022]
Abstract
The overall goal of regenerative medicine is to restore the functional performance of the tissues and organs that have been severely damaged or lost due to traumas and diseases [...].
Collapse
Affiliation(s)
- Anna Guller
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
- Correspondence:
| | - Alexandra Igrunkova
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
- World-Class Research Centre “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow 119992, Russia
| |
Collapse
|
42
|
Ghosal K, Chatterjee S, Thomas S, Roy P. A Detailed Review on Synthesis, Functionalization, Application, Challenges, and Current Status of Magnetic Nanoparticles in the Field of Drug Delivery and Gene Delivery System. AAPS PharmSciTech 2022; 24:25. [PMID: 36550283 DOI: 10.1208/s12249-022-02485-5] [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: 08/18/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
For progression of health care system, it has always been a challenge to the researchers for formulation to a type of advanced drug delivery system which will have less toxicity, targeted delivery and will be highly biodegradable. Nano science or nanotechnology has been validated to be a successful method as of targeting the drug to its active site be due to its special physicochemical properties and size thereby reducing the dose of administration, increasing bioavailability, and also reducing toxicity. Magnetic nanoparticles recently in few decades have proved as an effective advanced drug delivery system for its elevated magnetic responsiveness, biocompatibility, elevated targeted drug delivery effectiveness, etc. The drug can be easily targeted to active site by application of external magnetic field. Among the various elements, nanoparticles prepared with magnetically active iron oxide or other iron-based spinel oxide nanoparticles are widely used due to its high electrical resistivity, mechanical hardness, chemical stability, etc. Owing to their easy execution towards drug delivery application, extensive research has been carried out in this area. This review paper has summarized all recent modifications of iron-based magnetically active nanoparticle based drug delivery system along with their synthesis, characterization, and applications.
Collapse
Affiliation(s)
- Kajal Ghosal
- Division of Pharmaceutics, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India.
| | - Shreya Chatterjee
- Division of Pharmaceutics, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Sabu Thomas
- Mahatma Gandhi University, Kottayam, Kerala, India
| | - Poulomi Roy
- Materials Processing & Microsystems Laboratory, CSIR-Central Mechanical Engineering Research Institute (CMERI), Mahatma Gandhi Avenue, Durgapur, 713209, West Bengal, India.,Academy of Scientific and Innovative Research (AcSIR), Uttar Pradesh, Ghaziabad, 201002, India
| |
Collapse
|
43
|
Markowicz-Piasecka M, Darłak P, Markiewicz A, Sikora J, Kumar Adla S, Bagina S, Huttunen KM. Current approaches to facilitate improved drug delivery to the central nervous system. Eur J Pharm Biopharm 2022; 181:249-262. [PMID: 36372271 DOI: 10.1016/j.ejpb.2022.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
Although many pharmaceuticals have therapeutic potentials for central nervous system (CNS) diseases, few of these agents have been effectively administered. It is due to the fact that the blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSF) restrict them from crossing the brain to exert biological activity. This article reviews the current approaches aiming to improve penetration across these barriers for effective drug delivery to the CNS. These issues are summarized into direct systemic delivery and invasive delivery, including the BBB disruption and convection enhanced delivery. Furthermore, novel drug delivery systems used at the nanoscale, including polymeric nanoparticles, liposomes, nanoemulsions, dendrimers, and micelles are discussed. These nanocarriers could contribute to a breakthrough in the treatment of many different CNS diseases. However, further broadened studies are needed to assess the biocompatibility and safety of these medical devices.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland.
| | - Patrycja Darłak
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland.
| | - Agata Markiewicz
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland.
| | - Joanna Sikora
- Department of Bioinorganic Chemistry, Medical University of Lodz, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland.
| | - Santosh Kumar Adla
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland; Institute of Organic Chemistry and Biochemistry (IOCB), Czech Academy of Sciences, Flemingovo Namesti 542/2, 160 00 Prague, Czech Republic.
| | - Sreelatha Bagina
- Charles River Discovery Research Services Finland Oy, Neulaniementie 4, 70210 Kuopio, Finland
| | - Kristiina M Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland.
| |
Collapse
|
44
|
Kong X, Feng M, Wu L, He Y, Mao H, Gu Z. Biodegradable gemcitabine-loaded microdevice with sustained local drug delivery and improved tumor recurrence inhibition abilities for postoperative pancreatic tumor treatment. Drug Deliv 2022; 29:1595-1607. [PMID: 35612309 PMCID: PMC9176693 DOI: 10.1080/10717544.2022.2075984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/27/2022] [Accepted: 05/01/2022] [Indexed: 11/05/2022] Open
Abstract
At present, the 10-year survival rate of patients with pancreatic cancer is still less than 4%, mainly due to the high cancer recurrence rate caused by incomplete surgery and lack of effective postoperative adjuvant treatment. Systemic chemotherapy remains the only choice for patients after surgery; however, it is accompanied by off-target effects and server systemic toxicity. Herein, we proposed a biodegradable microdevice for local sustained drug delivery and postoperative pancreatic cancer treatment as an alternative and safe option. Biodegradable poly(l-lactic-co-glycolic acid) (P(L)LGA) was developed as the matrix material, gemcitabine hydrochloride (GEM·HCl) was chosen as the therapeutic drug and polyethylene glycol (PEG) was employed as the drug release-controlled regulator. Through adjusting the amount and molecular weight of PEG, the controllable degradation of matrix and the sustained release of GEM·HCl were obtained, thus overcoming the unstable drug release properties of traditional microdevices. The drug release mechanism of microdevice and the regulating action of PEG were studied in detail. More importantly, in the treatment of the postoperative recurrence model of subcutaneous pancreatic tumor in mice, the microdevice showed effective inhibition of postoperative in situ recurrences of pancreatic tumors with excellent biosafety and minimum systemic toxicity. The microdevice developed in this study provides an option for postoperative adjuvant pancreatic treatment, and greatly broadens the application prospects of traditional chemotherapy drugs.
Collapse
Affiliation(s)
- Xiangming Kong
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
| | - Miao Feng
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
| | - Lihuang Wu
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
| | - Yiyan He
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, PR China
- Suqian Advanced Materials Industry Technology Innovation Center of Nanjing Tech University, Nanjing, PR China
| | - Hongli Mao
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, PR China
- Suqian Advanced Materials Industry Technology Innovation Center of Nanjing Tech University, Nanjing, PR China
| | - Zhongwei Gu
- College of Materials Science and Engineering, Research Institute for Biomaterials, Tech Institute for Advanced Materials, Nanjing Tech University, Nanjing, PR China
- NJTech-BARTY Joint Research Center for Innovative Medical Technology, Nanjing Tech University, Nanjing, PR China
- Suqian Advanced Materials Industry Technology Innovation Center of Nanjing Tech University, Nanjing, PR China
| |
Collapse
|
45
|
Malek-Khatabi A, Tabandeh Z, Nouri A, Mozayan E, Sartorius R, Rahimi S, Jamaledin R. Long-Term Vaccine Delivery and Immunological Responses Using Biodegradable Polymer-Based Carriers. ACS APPLIED BIO MATERIALS 2022; 5:5015-5040. [PMID: 36214209 DOI: 10.1021/acsabm.2c00638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Biodegradable polymers are largely employed in the biomedical field, ranging from tissue regeneration to drug/vaccine delivery. The biodegradable polymers are highly biocompatible and possess negligible toxicity. In addition, biomaterial-based vaccines possess adjuvant properties, thereby enhancing immune responses. This Review introduces the use of different biodegradable polymers and their degradation mechanism. Different kinds of vaccines, as well as the interaction between the carriers with the immune system, then are highlighted. Natural and synthetic biodegradable micro-/nanoplatforms, hydrogels, and scaffolds for local or targeted and controlled vaccine release are subsequently discussed.
Collapse
Affiliation(s)
- Atefeh Malek-Khatabi
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - Zahra Tabandeh
- Department of Physical Chemistry, Faculty of Chemistry, University of Kashan, Kashan 8731753153, Iran
| | - Akram Nouri
- School of Chemistry, College of Science, University of Tehran, Tehran 141556455, Iran
| | - Elaheh Mozayan
- Department of Cell and Molecular Biology, University of Kashan, Kashan 8731753153, Iran
| | | | - Shahnaz Rahimi
- School of Chemistry, College of Science, University of Tehran, Tehran 141556455, Iran
| | - Rezvan Jamaledin
- Department of Chemical, Materials & Industrial Production Engineering, University of Naples Federico II, Naples 80125, Italy
| |
Collapse
|
46
|
Bhandari M, Nguyen S, Yazdani M, Utheim TP, Hagesaether E. The Therapeutic Benefits of Nanoencapsulation in Drug Delivery to the Anterior Segment of the Eye: A Systematic Review. Front Pharmacol 2022; 13:903519. [PMID: 35645827 PMCID: PMC9136980 DOI: 10.3389/fphar.2022.903519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/25/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Although numerous nanoparticle formulations have been developed for ocular administration, concerns are being raised about a possible mismatch between potential promises made by the field of nanoparticle research and demonstration of actual therapeutic benefit. Therefore, the primary focus of this present review was to critically assess to what extent nanoencapsulation of ocular drugs improved the therapeutic outcome when treating conditions in the anterior segment of the eye. Methods: A systematic search was conducted using Medline, PubMed, and Embase databases as well as Google Scholar for published peer-reviewed articles in English focusing on conventional nanoparticles used as drug delivery systems to the anterior segment of the eye in in vivo studies. The major therapeutic outcomes were intraocular pressure, tear secretion, number of polymorphonuclear leucocytes and pupil size. The outcome after encapsulation was compared to the non-encapsulated drug. Results: From the search, 250 results were retrieved. Thirty-eight studies met the inclusion criteria. Rabbits were used as study subjects in all but one study, and the number of animals ranged from 3 to 10. Coated and uncoated liposomes, lipid-based and polymeric nanoparticles, as well as micelles, were studied, varying in both particle size and surface charge, and encapsulating a total of 24 different drugs, including 6 salts. The majority of the in vivo studies demonstrated some improvement after nanoencapsulation, but the duration of the benefit varied from less than 1 h to more than 20 h. The most common in vitro methods performed in the studies were drug release, transcorneal permeation, and mucin interaction. Discussion: Nanoparticles that are small and mucoadhesive, often due to positive surface charge, appeared beneficial. Although in vitro assays can unravel more of the hidden and sophisticated interplay between the encapsulated drug and the nanoparticle structure, they suffered from a lack of in vitro—in vivo correlation. Therefore, more research should be focused towards developing predictive in vitro models, allowing rational design and systematic optimization of ocular nanoparticles with minimal animal experimentation.
Collapse
Affiliation(s)
- Madhavi Bhandari
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
- *Correspondence: Madhavi Bhandari,
| | - Sanko Nguyen
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| | - Mazyar Yazdani
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Tor Paaske Utheim
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
- Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
| | - Ellen Hagesaether
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway
| |
Collapse
|
47
|
Madamsetty VS, Mohammadinejad R, Uzieliene I, Nabavi N, Dehshahri A, García-Couce J, Tavakol S, Moghassemi S, Dadashzadeh A, Makvandi P, Pardakhty A, Aghaei Afshar A, Seyfoddin A. Dexamethasone: Insights into Pharmacological Aspects, Therapeutic Mechanisms, and Delivery Systems. ACS Biomater Sci Eng 2022; 8:1763-1790. [PMID: 35439408 PMCID: PMC9045676 DOI: 10.1021/acsbiomaterials.2c00026] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Dexamethasone (DEX) has been widely used to treat a variety of diseases, including autoimmune diseases, allergies, ocular disorders, cancer, and, more recently, COVID-19. However, DEX usage is often restricted in the clinic due to its poor water solubility. When administered through a systemic route, it can elicit severe side effects, such as hypertension, peptic ulcers, hyperglycemia, and hydro-electrolytic disorders. There is currently much interest in developing efficient DEX-loaded nanoformulations that ameliorate adverse disease effects inhibiting advancements in scientific research. Various nanoparticles have been developed to selectively deliver drugs without destroying healthy cells or organs in recent years. In the present review, we have summarized some of the most attractive applications of DEX-loaded delivery systems, including liposomes, polymers, hydrogels, nanofibers, silica, calcium phosphate, and hydroxyapatite. This review provides our readers with a broad spectrum of nanomedicine approaches to deliver DEX safely.
Collapse
Affiliation(s)
- Vijay Sagar Madamsetty
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Jacksonville, Florida 32224, United States
| | - Reza Mohammadinejad
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman 7618866749, Iran
| | - Ilona Uzieliene
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Santariskiu 5, LT-08406 Vilnius, Lithuania
| | - Noushin Nabavi
- Department of Urologic Sciences, Vancouver Prostate Centre, Vancouver, British Columbia, Canada V6H 3Z6
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
| | - Jomarien García-Couce
- Department of Radiology, Division of Translational Nanobiomaterials and Imaging, Leiden University Medical Center, Leiden 2333 ZA, The Netherlands
- Department of Polymeric Biomaterials, Biomaterials Center (BIOMAT), University of Havana, Havana 10600, Cuba
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1417755469, Iran
| | - Saeid Moghassemi
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Micro-BioRobotics, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Abbas Pardakhty
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 7618866748, Iran
| | - Abbas Aghaei Afshar
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman 7618866749, Iran
| | - Ali Seyfoddin
- Drug Delivery Research Group, Auckland University of Technology (AUT), School of Science, Auckland 1010, New Zealand
| |
Collapse
|
48
|
Polymeric Coatings and Antimicrobial Peptides as Efficient Systems for Treating Implantable Medical Devices Associated-Infections. Polymers (Basel) 2022; 14:polym14081611. [PMID: 35458361 PMCID: PMC9024559 DOI: 10.3390/polym14081611] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 02/04/2023] Open
Abstract
Many infections are associated with the use of implantable medical devices. The excessive utilization of antibiotic treatment has resulted in the development of antimicrobial resistance. Consequently, scientists have recently focused on conceiving new ways for treating infections with a longer duration of action and minimum environmental toxicity. One approach in infection control is based on the development of antimicrobial coatings based on polymers and antimicrobial peptides, also termed as “natural antibiotics”.
Collapse
|
49
|
Thanongsak W, Boongird A, Nasongkla N. Nanocoating and biological evaluation of clindamycin- and rifampicin-loaded nanospheres impregnated Silicone tube for antibacterial application. Pharm Dev Technol 2022; 27:372-378. [DOI: 10.1080/10837450.2022.2063890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Watunyu Thanongsak
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakorn Pathom, 73170, Thailand
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Mahidol University, Bangkok, 10400, Thailand
| | - Atthaporn Boongird
- Department of Surgery, Neurosurgical Unit, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Norased Nasongkla
- Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakorn Pathom, 73170, Thailand
- Department of Chemistry and Center of Excellence for Innovation in Chemistry, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
50
|
Park H, Otte A, Park K. Evolution of drug delivery systems: From 1950 to 2020 and beyond. J Control Release 2022; 342:53-65. [PMID: 34971694 PMCID: PMC8840987 DOI: 10.1016/j.jconrel.2021.12.030] [Citation(s) in RCA: 138] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 12/21/2021] [Indexed: 02/03/2023]
Abstract
Modern drug delivery technology began in 1952 with the advent of the Spansule® sustained-release capsule technology, which can deliver a drug for 12 h after oral administration through an initial immediate dose followed by the remaining released gradually. Until the 1980s, oral and transdermal formulations providing therapeutic durations up to 24 h for small molecules dominated the drug delivery field and the market. The introduction of Lupron Depot® in 1989 opened the door for long-acting injectables and implantables, extending the drug delivery duration from days to months and occasionally years. Notably, the new technologies allowed long-term delivery of peptide and protein drugs, although limited to parenteral administration. The introduction of the first PEGylated protein, Adagen®, in 1990 marked the new era of PEGylation, resulting in Doxil® (doxorubicin in PEGylated liposome) in 1995, Movantik® (PEGylated naloxone - naloxegol) in 2014, and Onpattro® (Patisiran - siRNA in PEGylated lipid nanoparticle) in 2018. Drug-polymer complexes were introduced, e.g., InFed® (iron-dextran complex injection) in 1974 and Abraxane® (paclitaxel-albumin complex) in 2005. In 2000, both Mylotarg™ (antibody-drug conjugate - gemtuzumab ozogamicin) and Rapamune® (sirolimus nanocrystal formulation) were introduced. The year 2000 also marked the launching of the National Nanotechnology Initiative by the U.S. government, which was soon followed by the rest of the world. Extensive work on nanomedicine, particularly formulations designed to escape from endosomes after being taken by tumor cells, along with PEGylation technology, ultimately resulted in the timely development of lipid nanoparticle formulations for COVID-19 vaccine delivery in 2020. While the advances in drug delivery technologies for the last seven decades are breathtaking, they are only the tip of an iceberg of technologies that have yet to be utilized in an approved formulation or even to be discovered. As life expectancy continues to increase, more people require long-term care for various diseases. Filling the current and future unmet needs requires innovative drug delivery technologies to overcome age-old familiar hurdles, e.g., improving water-solubility of poorly soluble drugs, overcoming biological barriers, and developing more efficient long-acting depot formulations. The lessons learned from the past are essential assets for developing future drug delivery technologies implemented into products. As the development of COVID-19 vaccines demonstrated, meeting the unforeseen crisis of the uncertain future requires continuous cumulation of failures (as learning experiences), knowledge, and technologies. Conscious efforts of supporting diversified research topics in the drug delivery field are urgently needed more than ever.
Collapse
Affiliation(s)
- Haesun Park
- Akina, Inc., West Lafayette, IN 47906, United States of America
| | - Andrew Otte
- Purdue University, Departments of Biomedical Engineering and Pharmaceutics, West Lafayette, IN 47907, United States of America
| | - Kinam Park
- Akina, Inc., West Lafayette, IN 47906, United States of America; Purdue University, Departments of Biomedical Engineering and Pharmaceutics, West Lafayette, IN 47907, United States of America.
| |
Collapse
|