1
|
Bohley M, Leroux J. Gastrointestinal Permeation Enhancers Beyond Sodium Caprate and SNAC - What is Coming Next? ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400843. [PMID: 38884149 PMCID: PMC11434117 DOI: 10.1002/advs.202400843] [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: 01/23/2024] [Revised: 03/13/2024] [Indexed: 06/18/2024]
Abstract
Oral peptide delivery is trending again. Among the possible reasons are the recent approvals of two oral peptide formulations, which represent a huge stride in the field. For the first time, gastrointestinal (GI) permeation enhancers (PEs) are leveraged to overcome the main limitation of oral peptide delivery-low permeability through the intestinal epithelium. Despite some success, the application of current PEs, such as salcaprozate sodium (SNAC), sodium caprylate (C8), and sodium caprate (C10), is generally resulting in relatively low oral bioavailabilities (BAs)-even for carefully selected therapeutics. With several hundred peptide-based drugs presently in the pipeline, there is a huge unmet need for more effective PEs. Aiming to provide useful insights for the development of novel PEs, this review summarizes the biological hurdles to oral peptide delivery with special emphasis on the epithelial barrier. It describes the concepts and action modes of PEs and mentions possible new targets. It further states the benchmark that is set by current PEs, while critically assessing and evaluating emerging PEs regarding translatability, safety, and efficacy. Additionally, examples of novel PEs under preclinical and clinical evaluation and future directions are discussed.
Collapse
Affiliation(s)
- Marilena Bohley
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| | - Jean‐Christophe Leroux
- Institute of Pharmaceutical SciencesDepartment of Chemistry and Applied BiosciencesETH ZurichZurich8093Switzerland
| |
Collapse
|
2
|
Liu Y, Long M, Wang Y, Liang Z, Dong Y, Qu M, Ge X, Nan Y, Chen Y, Zhou X. Chitosan-alginate/R8 ternary polyelectrolyte complex as an oral protein-based vaccine candidate induce effective mucosal immune responses. Int J Biol Macromol 2024; 275:133671. [PMID: 38971274 DOI: 10.1016/j.ijbiomac.2024.133671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Vaccination is the most effective method for preventing infectious diseases. Oral vaccinations have attracted much attention due to the ability to boost intestinal and systemic immunity. The focus of this study was to develop a poly (lactide-co-glycolide) acid (PLGA)-based ternary polyelectrolyte complex (PEC) with chitosan, sodium alginate, and transmembrane peptides R8 for the delivery of antigen proteins. In this study, the antigen protein (HBf), consisting of the Mycobacterium avium subspecies paratuberculosis (MAP) antigens HBHA, Ag85B, and Bfra, was combined with R8 to generate self-assembled conjugates. The results showed that PEC presented a cross-linked reticular structure to protect the encapsulated proteins in the simulated gastric fluid. Then, the nanocomposite separated into individual nanoparticles after entering the simulated intestinal fluid. The ternary PEC with R8 promoted the in vivo uptake of antigens by intestinal lymphoid tissue. Moreover, the ternary PEC administered orally to mice promoted the secretion of specific antibodies and intestinal mucosal IgA. In addition, in the mouse models of MAP infection, the ternary PEC enhanced splenic T cell responses, thus reducing bacterial load and liver pathology score. These results suggested that this ternary electrolyte complex could be a promising delivery platform for oral subunit vaccine candidates, not limited to MAP infection.
Collapse
Affiliation(s)
- Yiduo Liu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Meizhen Long
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Yuanzhi Wang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Zhengmin Liang
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Yuhui Dong
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Mengjin Qu
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Xin Ge
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Yue Nan
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Yulan Chen
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China
| | - Xiangmei Zhou
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, PR China.
| |
Collapse
|
3
|
Wang L, Liu J. Dopamine Polymerization-Mediated Surface Functionalization toward Advanced Bacterial Therapeutics. Acc Chem Res 2024; 57:945-956. [PMID: 38422996 DOI: 10.1021/acs.accounts.3c00798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Bacteria-based therapy has spotlighted an unprecedented potential in treating a range of diseases, given that bacteria can be used as both drug vehicles and therapeutic agents. However, the use of bacteria for disease treatment often suffers from unsatisfactory outcomes, due largely to their suboptimal bioavailability, dose-dependent toxicity, and low targeting colonization. In the past few years, substantial efforts have been devoted to tackling these difficulties, among which methods capable of integrating bacteria with multiple functions have been extensively pursued. Different from conventional genetic engineering and modern synthetic bioengineering, surface modification of bacteria has emerged as a simple yet flexible strategy to introduce different functional motifs. Polydopamine, which can be easily formed via in situ dopamine oxidation and self-polymerization, is an appealing biomimetic polymer that has been widely applied for interfacial modification and functionalization. By virtue of its catechol groups, polydopamine can be efficiently codeposited with a multitude of functional elements on diverse surfaces.In this Account, we summarize the recent advances from our group with a focus on the interfacial polymerization-mediated functionalization of bacteria for advanced microbial therapy. First, we present the optimized strategy for bacterial surface modification under cytocompatible conditions by in situ dopamine polymerization. Taking advantage of the hydrogen bonding, π-π stacking, Michael addition, and Schiff base reaction with polydopamine, diverse functional small molecules and macromolecules are facilely codeposited onto the bacterial surface. Namely, monomodal, dual-modal, and multimodal surface modification of bacteria can be achieved by dopamine self-deposition, codeposition with a unitary composition, and codeposition with a set of multiple components, respectively. Second, we outline the regulation of bacterial functions by surface modification. The formed polydopamine surface endows bacteria with the ability to resist in vivo insults, such as gastrointestinal tract stressors and immune clearance, resulting in greatly improved bioavailability. Integration with specific ligands or therapeutic components enables the modified bacteria to increase targeting accumulation and colonization at lesion sites or play synergistic effects in disease treatment. Bacteria codeposited with different bioactive moieties, such as protein antigens, antibodies, and immunoadjuvants, are even able to actively interact with the host, particularly to elicit immune responses by either suppressing immune overactivation to promote the reversion of pathological inflammations or provoking protective innate and/or adaptive immunity to inhibit pathogenic invaders. Third, we highlight the applications of surface-modified bacteria as multifunctional living therapeutics in disease treatment, especially alleviating inflammatory bowel diseases via oral delivery and intervening in different types of cancer through systemic or intratumoral injection. Finally, we discuss the challenges and prospects of dopamine polymerization-mediated multifunctionalization for preparing advanced bacterial therapeutics as well as their bench to bedside translation. We anticipate that this Account can provide an insightful overview of bacterial therapy and inspire innovative thinking and new efforts to develop next-generation living therapeutics for treating various diseases.
Collapse
Affiliation(s)
- Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
4
|
Pinto SFT, Santos HA, Sarmento BFCC. New insights into nanomedicines for oral delivery of glucagon-like peptide-1 analogs. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1952. [PMID: 38500351 DOI: 10.1002/wnan.1952] [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: 08/12/2023] [Revised: 01/23/2024] [Accepted: 02/21/2024] [Indexed: 03/20/2024]
Abstract
Type 2 diabetes mellitus (T2DM) is a metabolic disorder that arises when the body cannot respond fully to insulin, leading to impaired glucose tolerance. Currently, the treatment embraces non-pharmacological actions (e.g., diet and exercise) co-associated with the administration of antidiabetic drugs. Metformin is the first-line treatment for T2DM; nevertheless, alternative therapeutic strategies involving glucagon-like peptide-1 (GLP-1) analogs have been explored for managing the disease. GLP-1 analogs trigger insulin secretion and suppress glucagon release in a glucose-dependent manner thereby, reducing the risk of hyperglycemia. Additionally, GLP-1 analogs have an extended plasma half-life compared to the endogenous peptide due to their high resistance to degradation by dipeptidyl peptidase-4. However, GLP-1 analogs are mainly administered via subcutaneous route, which can be inconvenient for the patients. Even considering an oral delivery approach, GLP-1 analogs are exposed to the harsh conditions of the gastrointestinal tract (GIT) and the intestinal barriers (mucus and epithelium). Hereupon, there is an unmet need to develop non-invasive oral transmucosal drug delivery strategies, such as the incorporation of GLP-1 analogs into nanoplatforms, to overcome the GIT barriers. Nanotechnology has the potential to shield antidiabetic peptides against the acidic pH and enzymatic activity of the stomach. In addition, the nanoparticles can be coated and/or surface-conjugated with mucodiffusive polymers and target intestinal ligands to improve their transport through the intestinal mucus and epithelium. This review focuses on the main hurdles associated with the oral administration of GLP-1 and GLP-1 analogs, and the nanosystems developed to improve the oral bioavailability of the antidiabetic peptides. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Soraia Filipa Tavares Pinto
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Hélder Almeida Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Bruno Filipe Carmelino Cardoso Sarmento
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- Instituto Universitário de Ciências da Saúde (IUCS-CESPU), Gandra, Portugal
| |
Collapse
|
5
|
Zhang E, Zhu H, Song B, Shi Y, Cao Z. Recent advances in oral insulin delivery technologies. J Control Release 2024; 366:221-230. [PMID: 38161033 DOI: 10.1016/j.jconrel.2023.12.045] [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: 09/01/2023] [Revised: 12/23/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
With the rise in diabetes mellitus cases worldwide, oral delivery of insulin is preferred over subcutaneous insulin administration due to its good patient compliance and non-invasiveness, simplicity, and versatility. However, oral insulin delivery is hampered by various gastrointestinal barriers that result in low drug bioavailability and insufficient therapeutic efficiency. Numerous strategies have been developed to overcome these barriers and increase the bioavailability of oral insulin. Yet, no commercial oral insulin product is available to address all clinical hurdles because of various substantial obstacles related to the structural organization and physiological function of the gastrointestinal tract. Herein, we discussed the significant physiological barriers (including chemical, enzymatic, and physical barriers) that hinder the transportation and absorption of orally delivered insulin. Then, we showcased recent significant and innovative advances in oral insulin delivery technologies. Finally, we concluded the review with remarks on future perspectives on oral insulin delivery technologies and potential challenges for forthcoming clinical translation of oral insulin delivery technologies.
Collapse
Affiliation(s)
- Ershuai Zhang
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Hui Zhu
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Boyi Song
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Yuanjie Shi
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA
| | - Zhiqiang Cao
- Department of Chemical Engineering and Materials Science, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
6
|
Gomte SS, Agnihotri TG, Khopade S, Jain A. Exploring the potential of pH-sensitive polymers in targeted drug delivery. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:228-268. [PMID: 37927045 DOI: 10.1080/09205063.2023.2279792] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/01/2023] [Indexed: 11/07/2023]
Abstract
The pH-sensitive polymers have attained significant attention in the arena of targeted drug delivery (TDD) because of their exceptional capability to respond to alteration in pH in various physiological environments. This attribute aids pH-sensitive polymers to act as smart carriers for therapeutic agents, transporting them precisely to target locations while curtailing the release of drugs in off-targeted sites, thereby diminishing side effects. Many pH-responsive polymers in TDD have revealed promising results, with increased therapeutic efficacy and decreased toxic effects. Several pH-sensitive polymers, including, hydroxy-propyl-methyl cellulose, poly (methacrylic acid) (Eudragit series), poly (acrylic acid), and chitosan, have been broadly studied for their myriad applications in the management of various types of diseases. Additionally, the amalgamation of pH-sensitive polymers with, additive manufacturing techniques like 3D printing, has resulted in the progression of novel drug delivery systems that regulate drug release in a controlled manner. Herein, types of pH-sensitive polymers in TDD are systemically reviewed. We have briefly discussed the nanocarriers employed for the delivery of various pH-sensitive polymers in TDD. Finally, miscellaneous applications of pH-sensitive polymers are discussed thoroughly with special attention to the implication of 3D printing in pH-sensitive polymers.
Collapse
Affiliation(s)
- Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, India
| | - Shivani Khopade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, India
| |
Collapse
|
7
|
Hahn J, Ding S, Im J, Harimoto T, Leong KW, Danino T. Bacterial therapies at the interface of synthetic biology and nanomedicine. NATURE REVIEWS BIOENGINEERING 2024; 2:120-135. [PMID: 38962719 PMCID: PMC11218715 DOI: 10.1038/s44222-023-00119-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/08/2023] [Indexed: 07/05/2024]
Abstract
Bacteria are emerging as living drugs to treat a broad range of disease indications. However, the inherent advantages of these replicating and immunostimulatory therapies also carry the potential for toxicity. Advances in synthetic biology and the integration of nanomedicine can address this challenge through the engineering of controllable systems that regulate spatial and temporal activation for improved safety and efficacy. Here, we review recent progress in nanobiotechnology-driven engineering of bacteria-based therapies, highlighting limitations and opportunities that will facilitate clinical translation.
Collapse
Affiliation(s)
- Jaeseung Hahn
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Suwan Ding
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Jongwon Im
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Tetsuhiro Harimoto
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Medical Center, New York, NY, USA
| | - Tal Danino
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
- Data Science Institute, Columbia University, New York, NY, USA
| |
Collapse
|
8
|
Nicze M, Borówka M, Dec A, Niemiec A, Bułdak Ł, Okopień B. The Current and Promising Oral Delivery Methods for Protein- and Peptide-Based Drugs. Int J Mol Sci 2024; 25:815. [PMID: 38255888 PMCID: PMC10815890 DOI: 10.3390/ijms25020815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/05/2024] [Accepted: 01/07/2024] [Indexed: 01/24/2024] Open
Abstract
Drugs based on peptides and proteins (PPs) have been widely used in medicine, beginning with insulin therapy in patients with diabetes mellitus over a century ago. Although the oral route of drug administration is the preferred one by the vast majority of patients and improves compliance, medications of this kind due to their specific chemical structure are typically delivered parenterally, which ensures optimal bioavailability. In order to overcome issues connected with oral absorption of PPs such as their instability depending on digestive enzymes and pH changes in the gastrointestinal (GI) system on the one hand, but also their limited permeability across physiological barriers (mucus and epithelium) on the other hand, scientists have been strenuously searching for novel delivery methods enabling peptide and protein drugs (PPDs) to be administered enterally. These include utilization of different nanoparticles, transport channels, substances enhancing permeation, chemical modifications, hydrogels, microneedles, microemulsion, proteolytic enzyme inhibitors, and cell-penetrating peptides, all of which are extensively discussed in this review. Furthermore, this article highlights oral PP therapeutics both previously used in therapy and currently available on the medical market.
Collapse
Affiliation(s)
- Michał Nicze
- Department of Internal Medicine and Clinical Pharmacology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Medyków 18, 40-752 Katowice, Poland (B.O.)
| | | | | | | | - Łukasz Bułdak
- Department of Internal Medicine and Clinical Pharmacology, Faculty of Medical Sciences, Medical University of Silesia in Katowice, Medyków 18, 40-752 Katowice, Poland (B.O.)
| | | |
Collapse
|
9
|
Wang Y, Zhang Y, Wang P, Jing T, Hu Y, Chen X. Research Progress on Antiviral Activity of Heparin. Curr Med Chem 2024; 31:7-24. [PMID: 36740803 DOI: 10.2174/0929867330666230203124032] [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: 04/02/2022] [Revised: 11/06/2022] [Accepted: 11/17/2022] [Indexed: 02/07/2023]
Abstract
Heparin, as a glycosaminoglycan, is known for its anticoagulant and antithrombotic properties for several decades. Heparin is a life-saving drug and is widely used for anticoagulation in medical practice. In recent years, there have been extensive studies that heparin plays an important role in non-anticoagulant diseases, such as anti-inflammatory, anti-viral, anti-angiogenesis, anti-neoplastic, anti-metastatic effects, and so on. Clinical observation and in vitro experiments indicate that heparin displays a potential multitarget effect. In this brief review, we will summarize heparin and its derivative's recently studied progress for the treatment of various viral infections. The aim is to maximize the benefits of drugs through medically targeted development, to meet the unmet clinical needs of serious viral diseases.
Collapse
Affiliation(s)
- Yi Wang
- Chinese Materia Medica Pharmacology, Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Yanqing Zhang
- Shandong VeriSign Test Detection Co., LTD, Jinan, China
| | - Ping Wang
- Chinese Materia Medica Pharmacology, Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Tianyuan Jing
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanan Hu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiushan Chen
- Zhenjiang Runjing High Purity Chemical Technology Co., Ltd., Zhenjiang, Jiangsu, China
| |
Collapse
|
10
|
Hu S, Zhao R, Xu Y, Gu Z, Zhu B, Hu J. Orally-administered nanomedicine systems targeting colon inflammation for the treatment of inflammatory bowel disease: latest advances. J Mater Chem B 2023; 12:13-38. [PMID: 38018424 DOI: 10.1039/d3tb02302h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic and idiopathic condition that results in inflammation of the gastrointestinal tract, leading to conditions such as ulcerative colitis and Crohn's disease. Commonly used treatments for IBD include anti-inflammatory drugs, immunosuppressants, and antibiotics. Fecal microbiota transplantation is also being explored as a potential treatment method; however, these drugs may lead to systemic side effects. Oral administration is preferred for IBD treatment, but accurately locating the inflamed area in the colon is challenging due to multiple physiological barriers. Nanoparticle drug delivery systems possess unique physicochemical properties that enable precise delivery to the target site for IBD treatment, exploiting the increased permeability and retention effect of inflamed intestines. The first part of this review comprehensively introduces the pathophysiological environment of IBD, covering the gastrointestinal pH, various enzymes in the pathway, transport time, intestinal mucus, intestinal epithelium, intestinal immune cells, and intestinal microbiota. The second part focuses on the latest advances in the mechanism and strategies of targeted delivery using oral nanoparticle drug delivery systems for colitis-related fields. Finally, we present challenges and potential directions for future IBD treatment with the assistance of nanotechnology.
Collapse
Affiliation(s)
- Shumeng Hu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
| | - Runan Zhao
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yu Xu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| | - Zelin Gu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
| | - Beiwei Zhu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| | - Jiangning Hu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| |
Collapse
|
11
|
Kopp KT, Saerens L, Voorspoels J, Van den Mooter G. Solidification and oral delivery of biologics to the colon- A review. Eur J Pharm Sci 2023; 190:106523. [PMID: 37429482 DOI: 10.1016/j.ejps.2023.106523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/16/2023] [Accepted: 07/08/2023] [Indexed: 07/12/2023]
Abstract
The oral delivery of biologics such as therapeutic proteins, peptides and oligonucleotides for the treatment of colon related diseases has been the focus of increasing attention over the last years. However, the major disadvantage of these macromolecules is their degradation propensity in liquid state which can lead to the undesirable and complete loss of function. Therefore, to increase the stability of the biologic and reduce their degradation propensity, formulation techniques such as solidification can be performed to obtain a stable solid dosage form for oral administration. Due to their fragility, stress exerted on the biologic during solidification has to be reduced with the incorporation of stabilizing excipients into the formulation. This review focuses on the state-of-the-art solidification techniques required to obtain a solid dosage form for the oral delivery of biologics to the colon and the use of suitable excipients for adequate stabilization upon solidification. The solidifying processes discussed within this review are spray drying, freeze drying, bead coating and also other techniques such as spray freeze drying, electro spraying, vacuum- and supercritical fluid drying. Further, the colon as site of absorption in both healthy and diseased state is critically reviewed and possible oral delivery systems for biologics are discussed.
Collapse
Affiliation(s)
- Katharina Tatjana Kopp
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium; Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49, 3000 Leuven, Belgium
| | - Lien Saerens
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium
| | - Jody Voorspoels
- Eurofins Amatsigroup, Industriepark-Zwijnaarde 7B, 9052 Gent, Belgium
| | - Guy Van den Mooter
- Drug Delivery and Disposition, KU Leuven, Department of Pharmaceutical and Pharmacological Sciences, Campus Gasthuisberg ON2, Herestraat 49, 3000 Leuven, Belgium.
| |
Collapse
|
12
|
You J, Yang C, Han J, Wang H, Zhang W, Zhang Y, Lu Z, Wang S, Cai R, Li H, Yu J, Gao J, Zhang Y, Gu Z. Ultrarapid-Acting Microneedles for Immediate Delivery of Biotherapeutics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304582. [PMID: 37547966 DOI: 10.1002/adma.202304582] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/19/2023] [Indexed: 08/08/2023]
Abstract
Subcutaneous (SC) injection is a common administration route for rapid and efficient delivery of biotherapeutics. However, syringe-based injections usually require professional assistance and are associated with pain and potential risks of infections, thus leading to undesired patient compliance and poor life quality. Herein, this work presents an ultrarapid-acting microneedle (URA-MN) patch for immediate transdermal delivery of therapeutics in a minimally invasive manner. Effervescent agents are incorporated into the tip of URA-MN for rapid generation of CO2 bubbles upon insertion into the skin, immediately powering the biotherapeutics release within a few minutes. The release kinetics of diverse agents including liraglutide (LRT), insulin, and heparin from the URA-MN patches are evaluated in three different mouse models, and the rapid release of biotherapeutics and potent therapeutic effects are achieved with only 5 min administration. Noteworthily, attributed to the short application duration and negligible residuals of MN matrix remaining in the skin, the URA-MN patch shows desirable biocompatibility after six-week administration.
Collapse
Affiliation(s)
- Jiahuan You
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Changwei Yang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jinpeng Han
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hao Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Wentao Zhang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ying Zhang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Burns and Wound Care Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Ziyi Lu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Shiqi Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ruisi Cai
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hongjun Li
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Jicheng Yu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Jianqing Gao
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Burns and Wound Care Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China
- Jinhua Institute of Zhejiang University, Jinhua, 321299, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
13
|
Malta R, Marques AC, da Costa PC, Amaral MH. Stimuli-Responsive Hydrogels for Protein Delivery. Gels 2023; 9:802. [PMID: 37888375 PMCID: PMC10606693 DOI: 10.3390/gels9100802] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/28/2023] Open
Abstract
Proteins and peptides are potential therapeutic agents, but their physiochemical properties make their use as drug substances challenging. Hydrogels are hydrophilic polymeric networks that can swell and retain high amounts of water or biological fluids without being dissolved. Due to their biocompatibility, their porous structure, which enables the transport of various peptides and proteins, and their protective effect against degradation, hydrogels have gained prominence as ideal carriers for these molecules' delivery. Particularly, stimuli-responsive hydrogels exhibit physicochemical transitions in response to subtle modifications in the surrounding environment, leading to the controlled release of entrapped proteins or peptides. This review is focused on the application of these hydrogels in protein and peptide delivery, including a brief overview of therapeutic proteins and types of stimuli-responsive polymers.
Collapse
Affiliation(s)
- Rafaela Malta
- CeNTI—Centre for Nanotechnology and Smart Materials, Rua Fernando Mesquita, 2785, 4760-034 Vila Nova de Famalicão, Portugal;
| | - Ana Camila Marques
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Paulo Cardoso da Costa
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Maria Helena Amaral
- UCIBIO—Applied Molecular Biosciences Unit, MEDTECH, Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal;
- Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, R. Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| |
Collapse
|
14
|
Afrin H, Geetha Bai R, Kumar R, Ahmad SS, Agarwal SK, Nurunnabi M. Oral delivery of RNAi for cancer therapy. Cancer Metastasis Rev 2023; 42:699-724. [PMID: 36971908 PMCID: PMC10040933 DOI: 10.1007/s10555-023-10099-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Cancer is a major health concern worldwide and is still in a continuous surge of seeking for effective treatments. Since the discovery of RNAi and their mechanism of action, it has shown promises in targeted therapy for various diseases including cancer. The ability of RNAi to selectively silence the carcinogenic gene makes them ideal as cancer therapeutics. Oral delivery is the ideal route of administration of drug administration because of its patients' compliance and convenience. However, orally administered RNAi, for instance, siRNA, must cross various extracellular and intracellular biological barriers before it reaches the site of action. It is very challenging and important to keep the siRNA stable until they reach to the targeted site. Harsh pH, thick mucus layer, and nuclease enzyme prevent siRNA to diffuse through the intestinal wall and thereby induce a therapeutic effect. After entering the cell, siRNA is subjected to lysosomal degradation. Over the years, various approaches have been taken into consideration to overcome these challenges for oral RNAi delivery. Therefore, understanding the challenges and recent development is crucial to offer a novel and advanced approach for oral RNAi delivery. Herein, we have summarized the delivery strategies for oral delivery RNAi and recent advancement towards the preclinical stages.
Collapse
Affiliation(s)
- Humayra Afrin
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
| | - Renu Geetha Bai
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Chair of Biosystems Engineering, Institute of Forestry and Engineering, Estonian University of Life Sciences, Kreutzwaldi 56/1, 51006, Tartu, Estonia
| | - Raj Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
| | - Sheikh Shafin Ahmad
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX, 79965, USA
| | - Sandeep K Agarwal
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Md Nurunnabi
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA.
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX, 79965, USA.
- Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA.
| |
Collapse
|
15
|
Stoilov B, Truong VK, Gronthos S, Vasilev K. Noninvasive and Microinvasive Nanoscale Drug Delivery Platforms for Hard Tissue Engineering. ACS APPLIED BIO MATERIALS 2023; 6:2925-2943. [PMID: 37565698 DOI: 10.1021/acsabm.3c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Bone tissue plays a crucial role in protecting internal organs and providing structural support and locomotion of the body. Treatment of hard tissue defects and medical conditions due to physical injuries, genetic disorders, aging, metabolic syndromes, and infections is more often a complex and drawn out process. Presently, dealing with hard-tissue-based clinical problems is still mostly conducted via surgical interventions. However, advances in nanotechnology over the last decades have led to shifting trends in clinical practice toward noninvasive and microinvasive methods. In this review article, recent advances in the development of nanoscale platforms for bone tissue engineering have been reviewed and critically discussed to provide a comprehensive understanding of the advantages and disadvantages of noninvasive and microinvasive methods for treating medical conditions related to hard tissue regeneration and repair.
Collapse
Affiliation(s)
- Borislav Stoilov
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia 5042, Australia
| | - Vi Khanh Truong
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia 5042, Australia
| | - Stan Gronthos
- School of Biomedicine, Faculty of Health and Medical Sciences, University of Adelaide/SAHMRI, North Terrace, Adelaide, South Australia 5001, Australia
| | - Krasimir Vasilev
- Biomedical Nanoengineering Laboratory, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, South Australia 5042, Australia
| |
Collapse
|
16
|
Miao YB, Xu T, Gong Y, Chen A, Zou L, Jiang T, Shi Y. Cracking the intestinal lymphatic system window utilizing oral delivery vehicles for precise therapy. J Nanobiotechnology 2023; 21:263. [PMID: 37559085 PMCID: PMC10413705 DOI: 10.1186/s12951-023-01991-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/09/2023] [Indexed: 08/11/2023] Open
Abstract
Oral administration is preferred over other drug delivery methods due to its safety, high patient compliance, ease of ingestion without discomfort, and tolerance of a wide range of medications. However, oral drug delivery is limited by the poor oral bioavailability of many drugs, caused by extreme conditions and absorption challenges in the gastrointestinal tract. This review thoroughly discusses the targeted drug vehicles to the intestinal lymphatic system (ILS). It explores the structure and physiological barriers of the ILS, highlighting its significance in dietary lipid and medication absorption and transport. The review presents various approaches to targeting the ILS using spatially precise vehicles, aiming to enhance bioavailability, achieve targeted delivery, and reduce first-pass metabolism with serve in clinic. Furthermore, the review outlines several methods for leveraging these vehicles to open the ILS window, paving the way for potential clinical applications in cancer treatment and oral vaccine delivery. By focusing on targeted drug vehicles to the ILS, this article emphasizes the critical role of these strategies in improving therapeutic efficacy and patient outcomes. Overall, this article emphasizes the critical role of targeted drug vehicles to the ILS and the potential impact of these strategies on improving therapeutic efficacy and patient outcomes.
Collapse
Affiliation(s)
- Yang-Bao Miao
- Department of Haematology, School of Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China.
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| | - Tianxing Xu
- Department of Haematology, School of Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ying Gong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, People's Republic of China
| | - Anmei Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, People's Republic of China
| | - Liang Zou
- School of Food and Biological Engineering, Chengdu University, Chengdu, Sichuan, 610106, China
| | - Tao Jiang
- Department of Haematology, School of Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China.
| | - Yi Shi
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
- Natural Products Research Center, Institute of Chengdu Biology, Sichuan Translational Medicine Hospital, Chinese Academy of Sciences, Chengdu, Sichuan, 610072, China.
- Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan, 610072, China.
| |
Collapse
|
17
|
Desai P, Dasgupta A, Sofias AM, Peña Q, Göstl R, Slabu I, Schwaneberg U, Stiehl T, Wagner W, Jockenhövel S, Stingl J, Kramann R, Trautwein C, Brümmendorf TH, Kiessling F, Herrmann A, Lammers T. Transformative Materials for Interfacial Drug Delivery. Adv Healthc Mater 2023; 12:e2301062. [PMID: 37282805 PMCID: PMC11468550 DOI: 10.1002/adhm.202301062] [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: 04/04/2023] [Revised: 05/25/2023] [Indexed: 06/08/2023]
Abstract
Drug delivery systems (DDS) are designed to temporally and spatially control drug availability and activity. They assist in improving the balance between on-target therapeutic efficacy and off-target toxic side effects. DDS aid in overcoming biological barriers encountered by drug molecules upon applying them via various routes of administration. They are furthermore increasingly explored for modulating the interface between implanted (bio)medical materials and host tissue. Herein, an overview of the biological barriers and host-material interfaces encountered by DDS upon oral, intravenous, and local administration is provided, and material engineering advances at different time and space scales to exemplify how current and future DDS can contribute to improved disease treatment are highlighted.
Collapse
Affiliation(s)
- Prachi Desai
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Anshuman Dasgupta
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Alexandros Marios Sofias
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
| | - Quim Peña
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
| | - Robert Göstl
- DWI – Leibniz Institute for Interactive Materials52074AachenGermany
| | - Ioana Slabu
- Institute of Applied Medical EngineeringHelmholtz InstituteMedical FacultyRWTH Aachen University52074AachenGermany
| | | | - Thomas Stiehl
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Institute for Computational Biomedicine – Disease ModelingRWTH Aachen University52074AachenGermany
| | - Wolfgang Wagner
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Helmholtz‐Institute for Biomedical EngineeringMedical Faculty of RWTH Aachen University52074AachenGermany
- Institute for Stem Cell BiologyUniversity Hospital of RWTH Aachen52074AachenGermany
| | - Stefan Jockenhövel
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz Institute AachenRWTH Aachen University52074AachenGermany
| | - Julia Stingl
- Institute of Clinical PharmacologyUniversity Hospital RWTH Aachen52074AachenGermany
| | - Rafael Kramann
- Division of Nephrology and Clinical ImmunologyRWTH Aachen University52074AachenGermany
- Institute of Experimental Medicine and Systems BiologyRWTH Aachen University52074AachenGermany
| | - Christian Trautwein
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Department of Medicine III (GastroenterologyMetabolic diseases and Intensive Care)University Hospital RWTH Aachen52074AachenGermany
| | - Tim H. Brümmendorf
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Department of HematologyOncologyHemostaseology and Stem Cell TransplantationRWTH Aachen University Medical School52074AachenGermany
| | - Fabian Kiessling
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Helmholtz‐Institute for Biomedical EngineeringMedical Faculty of RWTH Aachen University52074AachenGermany
| | - Andreas Herrmann
- DWI – Leibniz Institute for Interactive Materials52074AachenGermany
- Institute of Technical and Macromolecular ChemistryRWTH Aachen UniversityWorringerweg 152074AachenGermany
| | - Twan Lammers
- Institute for Experimental Molecular ImagingRWTH Aachen University Hospital52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO)52074AachenGermany
- Helmholtz‐Institute for Biomedical EngineeringMedical Faculty of RWTH Aachen University52074AachenGermany
| |
Collapse
|
18
|
Chien ST, Suydam IT, Woodrow KA. Prodrug approaches for the development of a long-acting drug delivery systems. Adv Drug Deliv Rev 2023; 198:114860. [PMID: 37160248 PMCID: PMC10498988 DOI: 10.1016/j.addr.2023.114860] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/01/2023] [Accepted: 05/02/2023] [Indexed: 05/11/2023]
Abstract
Long-acting formulations are designed to reduce dosing frequency and simplify dosing schedules by providing an extended duration of action. One approach to obtain long-acting formulations is to combine long-acting prodrugs (LA-prodrug) with existing or emerging drug delivery technologies (DDS). The design criteria for long-acting prodrugs are distinct from conventional prodrug strategies that alter absorption, distribution, metabolism, and excretion (ADME) parameters. Our review focuses on long-acting prodrug delivery systems (LA-prodrug DDS), which is a subcategory of long-acting formulations where prodrug design enables DDS formulation to achieve an extended duration of action that is greater than the parent drug. Here, we define LA-prodrugs as the conjugation of an active pharmaceutical ingredient (API) to a promoiety group via a cleavable covalent linker, where both the promoiety and linker are selected to enable formulation and administration from a drug delivery system (DDS) to achieve an extended duration of action. These LA-prodrug DDS results in an extended interval where the API is within a therapeutic range without necessarily altering ADME as is typical of conventional prodrugs. The conversion of the LA-prodrug to the API is dependent on linker cleavage, which can occur before or after release from the DDS. The requirement for linker cleavage provides an additional tool to prolong release from these LA-prodrug DDS. In addition, the physicochemical properties of drugs can be tuned by promoiety selection for a particular DDS. Conjugation with promoieties that are carriers or amenable to assembly into carriers can also provide access to formulations designed for extending duration of action. LA-prodrugs have been applied to a wide variety of drug delivery strategies and are categorized in this review by promoiety size and complexity. Small molecule promoieties (typically MW < 1000 Da) have been used to improve encapsulation or partitioning as well as broaden APIs for use with traditional long-acting formulations such as solid drug dispersions. Macromolecular promoieties (typically MW > 1000 Da) have been applied to hydrogels, nanoparticles, micelles, dendrimers, and polymerized prodrug monomers. The resulting LA-prodrug DDS enable extended duration of action for active pharmaceuticals across a wide range of applications, with target release timescales spanning days to years.
Collapse
Affiliation(s)
- Shin-Tian Chien
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Ian T Suydam
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States
| | - Kim A Woodrow
- Department of Bioengineering, University of Washington, Seattle, WA 98105, United States.
| |
Collapse
|
19
|
Liu S, Wen X, Zhang X, Mao S. Oral delivery of biomacromolecules by overcoming biological barriers in the gastrointestinal tract: an update. Expert Opin Drug Deliv 2023; 20:1333-1347. [PMID: 37439101 DOI: 10.1080/17425247.2023.2231343] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023]
Abstract
INTRODUCTION Biomacromolecules have proven to be an attractive choice for treating diseases due to their properties of strong specificity, high efficiency, and low toxicity. Besides greatly improving the patient's complaint, oral delivery of macromolecules also complies with hormone physiological secretion, which has become one of the most innovative fields of research in recent years. AREAS COVERED Oral delivery biological barriers for biomacromolecule, transport mechanisms, and various administration strategies were discussed in this review, including absorption enhancers, targeting nanoparticles, mucoadhesion nanoparticles, mucus penetration nanoparticles, and intelligent bionic drug delivery systems. EXPERT OPINION The oral delivery of biomacromolecules has important clinical implications; however, these are still facing the challenges of low bioavailability due to certain barriers. Various promising technologies have been developed to overcome the barriers and improve the therapeutic effect of oral biomacromolecules. By considering safety and efficacy comprehensively, the development of intelligent nanoparticles based on the GIT environment has demonstrated some promise in overcoming these barriers; however, a more comprehensive understanding of the oral fate of oral biomacromolecules is still required.
Collapse
Affiliation(s)
- Shiyun Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiangce Wen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xin Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
20
|
Spoorthi Shetty S, Halagali P, Johnson AP, Spandana KMA, Gangadharappa HV. Oral insulin delivery: Barriers, strategies, and formulation approaches: A comprehensive review. Int J Biol Macromol 2023:125114. [PMID: 37263330 DOI: 10.1016/j.ijbiomac.2023.125114] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
Diabetes Mellitus is characterized by a hyperglycemic condition which can either be caused by the destruction of the beta cells or by the resistance developed against insulin in the cells. Insulin is a peptide hormone that regulates the metabolism of carbohydrates, proteins, and fats. Type 1 Diabetes Mellitus needs the use of Insulin for efficient management. However invasive methods of administration may lead to reduced adherence by the patients. Hence there is a need for a non-invasive method of administration. Oral Insulin has several merits over the conventional method including patient compliance, and reduced cost, and it also mimics endogenous insulin and hence reaches the liver by the portal vein at a higher concentration and thereby showing improved efficiency. However oral Insulin must pass through several barriers in the gastrointestinal tract. Some strategies that could be utilized to bypass these barriers include the use of permeation enhancers, absorption enhancers, use of suitable polymers, use of suitable carriers, and other agents. Several formulation types have been explored for the oral delivery of Insulin like hydrogels, capsules, tablets, and patches which have been described briefly by the article. A lot of attempts have been made for developing oral insulin delivery however none of them have been commercialized due to numerous shortcomings. Currently, there are several formulations from the companies that are still in the clinical phase, the success or failure of some is yet to be seen in the future.
Collapse
Affiliation(s)
- S Spoorthi Shetty
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Praveen Halagali
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Asha P Johnson
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - K M Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - H V Gangadharappa
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India.
| |
Collapse
|
21
|
Zeng CH, Kim SH, Kang JM, Park Y, Won DS, Kim JW, Ryu DS, Shim S, Kim DK, Shin JH, Park JH. Different routes of administering EW-7197 versus EW-7197⋅HBr for preventing peritoneal adhesion in a rat model. Surgery 2023; 173:1008-1014. [PMID: 36577598 DOI: 10.1016/j.surg.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/16/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND The relatively low aqueous solubility of EW-7197 that was administered orally may have affected the desired concentration in the systemic circulation for treating peritoneal adhesion. This experimental study aimed to compare the efficacy of different routes of administering EW-7197 (2-fluoro-N-[(5-[6-methylpyridin-2-yl]-4-[(1,2,4)triazolo(1,5-a)pyridin-6-yl]-1H-imidazol-2-yl)methyl]aniline) and EW-7197·hydrobromide (HBr), with improved aqueous solubility, for inhibiting peritoneal adhesion in a rat model. METHODS After peritoneal adhesion induction, 30 male Sprague-Dawley rats were randomly divided into 5 groups with 6 rats in each: group A, sham control; group B, orally administered 25 mg/kg of EW-7197·HBr for 7 days; group C, locally administered 25 mg/kg of EW-7197·HBr; group D, orally administered 20 mg/kg of EW-7197 for 7 days; and group E, locally administered 20 mg/kg of EW-7197. Gross examination, histologic staining (hematoxylin and eosin and Masson's trichrome), and immunohistochemical analyses (Ki-67 and α-smooth muscle actin marker [α-SMA]) were performed to evaluate the efficacy of both drugs. RESULTS All procedures were technically successful. All treatment groups, except for group C, showed significantly reduced incidence, quality, tenacity, fibrosis, and collagen deposition scores and lowered expressions of Ki-67- and α-SMA-positive cells compared with group A. When comparing between groups, all scores were significantly lower in group B than in group C (all P < .001), whereas no significant difference was noted in any of the scores between groups D and E and groups B and E (all P > .05). CONCLUSION Orally administering EW-7197·HBr and both orally and locally administering EW-7197 significantly prevented peritoneal adhesion formation, and orally administering EW-7197·HBr was the most effective overall.
Collapse
Affiliation(s)
- Chu Hui Zeng
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea; Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Song Hee Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Jeon Min Kang
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Yubeen Park
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Dong-Sung Won
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Ji Won Kim
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Dae Sung Ryu
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Soyeon Shim
- EWHA DrugDesignHouse, Seoul, Republic of Korea
| | - Dae-Kee Kim
- EWHA DrugDesignHouse, Seoul, Republic of Korea
| | - Ji Hoon Shin
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Jung-Hoon Park
- Biomedical Engineering Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Yang Y, Zhou R, Wang Y, Zhang Y, Yu J, Gu Z. Recent Advances in Oral and Transdermal Protein Delivery Systems. Angew Chem Int Ed Engl 2023; 62:e202214795. [PMID: 36478123 DOI: 10.1002/anie.202214795] [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: 10/07/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Protein and peptide drugs are predominantly administered by injection to achieve high bioavailability, but this greatly compromises patient compliance. Oral and transdermal drug delivery with minimal invasiveness and high adherence represent attractive alternatives to injection administration. However, oral and transdermal administration of bioactive proteins must overcome biological barriers, namely the gastrointestinal and skin barriers, respectively. The rapid development of new materials and technologies promises to address these physiological obstacles. This review provides an overview of the latest advances in oral and transdermal protein delivery, including chemical strategies, synthetic nanoparticles, medical microdevices, and biomimetic systems for oral administration, as well as chemical enhancers, physical approaches, and microneedles in transdermal delivery. We also discuss challenges and future perspectives of the field with a focus on innovation and translation.
Collapse
Affiliation(s)
- Yinxian Yang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ruyi Zhou
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yanfang Wang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuqi Zhang
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Department of Burns and Wound Center, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jicheng Yu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.,Jinhua Institute of Zhejiang University, Jinhua, 321299, China.,Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Zhen Gu
- Zhejiang Provincial Key Laboratory for Advanced Drug Delivery Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, 311121, China.,Jinhua Institute of Zhejiang University, Jinhua, 321299, China.,Department of General Surgery, Sir Run Run Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.,MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
23
|
Kommineni N, Sainaga Jyothi VGS, Butreddy A, Raju S, Shapira T, Khan W, Angsantikul P, Domb AJ. SNAC for Enhanced Oral Bioavailability: An Updated Review. Pharm Res 2023; 40:633-650. [PMID: 36539668 DOI: 10.1007/s11095-022-03459-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
The delivery of proteins and peptides via an oral route poses numerous challenges to improve the oral bioavailability and patient compliance. To overcome these challenges, as well as to improve the permeation of proteins and peptides via intestinal mucosa, several chemicals have been studied such as surfactants, fatty acids, bile salts, pH modifiers, and chelating agents, amongst these medium chain fatty acid like C10 (sodium caprate) and Sodium N-[8-(2-hydroxybenzoyl) amino] caprylate (SNAC) and its derivatives that have been well studied from a clinical perspective. This current review enumerates the challenges involved in protein and peptide delivery via the oral route, i.e., non-invasive routes of protein and peptide administration. This review also covers the chemistry behind SNAC and toxicity as well as mechanisms to enhance the oral delivery of clinically proven molecules like simaglutide and other small molecules under clinical development, as well as other permeation enhancers for efficient delivery of proteins and peptides.
Collapse
Affiliation(s)
- Nagavendra Kommineni
- Center for Biomedical Research, Population Council, New York, NY, 10065, USA.
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India.
| | - Vaskuri G S Sainaga Jyothi
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Arun Butreddy
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, Oxford, MS, 38677, USA
| | - Saka Raju
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Tovi Shapira
- School of Pharmacy and Faculty of Medicine, The Hebrew University of Jerusalem, Hadassah Medical Center, Ein Kerem Campus, 91120, Jerusalem, Israel
| | - Wahid Khan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
- Natco Research Centre, NATCO Pharma Limited, Hyderabad, 500018, India
| | - Pavimol Angsantikul
- Center for Biomedical Research, Population Council, New York, NY, 10065, USA
| | - Abraham J Domb
- School of Pharmacy and Faculty of Medicine, The Hebrew University of Jerusalem, Hadassah Medical Center, Ein Kerem Campus, 91120, Jerusalem, Israel.
| |
Collapse
|
24
|
Bimbrawh S, Chopra S, Ansari MJ, Alrobaian M, Almalki WH, Alharbi KS, Alenezi SK, Kaur R, Beg S, Bhatia A. Biocompatible phospholipid-based nanovesicular drug delivery system of ketoprofen: Systematic development, optimization, and preclinical evaluation. Biotechnol Appl Biochem 2023; 70:51-67. [PMID: 35262954 DOI: 10.1002/bab.2328] [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: 12/15/2021] [Accepted: 01/21/2022] [Indexed: 11/10/2022]
Abstract
The present work involved development of phospholipid-based permeation enhancing nanovesicles (PENVs) for topical delivery of ketoprofen. Screening of phospholipids and process parameters was performed. Central composite design was used for optimization of factors, that is, amount (%, w/w) of phospholipid and ethanol at three levels. The optimized nanovesicles (NVs) were loaded with different terpenes and then incorporated into a gel base. Optimized NVs exhibited 69% entrapment efficiency, 51% transmittance, 328 nm mean vesicle size, and polydispersity index of 0.25. In vitro release kinetics evaluation indicated best fitting as per Korsemeyer-Peppa's model and drug release via Fickian-diffusion mechanism. The optimized NVs loaded with mint terpene showed minimal degree of deformability and maximal elasticity as compared with the conventional NVs and liposomes. Rheology and texture analysis indicated pseudoplastic flow and smooth texture of the vesicle gel formulation. Ex vivo permeation studies across Wistar rat skin indicated low penetration (0.43-fold decrease) and high skin retention (4.26-fold increase) of ketoprofen from the optimized PENVs gel vis-à-vis the conventional gel. Skin irritancy study indicated lower scores for PENVs gel construing its biocompatible nature. Stability studies confirmed cold storage is best suitable for vesicle gel, and optimized PENVs were found to be suitable for topical delivery of ketoprofen.
Collapse
Affiliation(s)
- Senha Bimbrawh
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Shruti Chopra
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.,Department of Pharmaceutical Chemistry, Amity Institute of Pharmacy, Amity University, Noida, Uttar Pradesh, India
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Majed Alrobaian
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Khalid S Alharbi
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakakah, Saudi Arabia
| | - Sattam K Alenezi
- Department of Pharmacology & Toxicology, Unaizah College of Pharmacy, Qassim University, Qassim, Saudi Arabia
| | - Ripandeep Kaur
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.,Pharmaceutics Division, University institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Sarwar Beg
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Amit Bhatia
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India.,Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda, Punjab, India
| |
Collapse
|
25
|
Fang H, Chen L, Deng Z, Gao Y, Yang Y, Chen Q, Liu Z. In Situ Polymerization of Zwitterions on Therapeutic Proteins to Enable Their Effective Oral Delivery. ACS NANO 2023; 17:1128-1143. [PMID: 36595442 DOI: 10.1021/acsnano.2c08434] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Oral administration of protein drugs has always been challenging owing to various intestinal barriers. Herein, we developed an efficient oral protein delivery strategy by using in situ polymerization of zwitterions to encapsulate proteins, which were then loaded into enteric coated capsules for oral feeding. After oral administration of such capsules, the enteric coating would be degraded once the capsule enters the intestine, releasing polyzwitterion/protein nanocomplexes. With the help of polyzwitterion modification, such nanocomplexes were able to pass through the mucus and cellular barriers, likely by the proton-assisted amino acid transporter 1 (PAT1) pathway. Such a polyzwitterion-based protein encapsulation strategy could allow for effective oral delivery of different proteins, including bovine serum albumin (BSA), insulin, and antibodies. Using this strategy, the oral bioavailabilities of insulin and immunoglobin G (IgG) were measured to be as high as 16.9% and 12.5%, respectively. Notably, oral feeding of polyzwitterion/insulin capsules could effectively lower the blood glucose level of diabetic animals (mice, rats, and pigs). Moreover, polyzwitterion/antiprogramed death-1 (αPD-1) capsules were able to induce efficient antitumor immune responses, showing significant tumor inhibition effects toward B16F10- and 4T1-tumor bearing mouse models after oral administration. No significant toxic effect was observed for such oral protein formulations in the treated animals. Our work presents a strategy for the efficient oral delivery of protein drugs, including those with large molecular weights (e.g., antibodies) that can hardly be orally delivered using existing technologies.
Collapse
Affiliation(s)
- Huapan Fang
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Linfu Chen
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Zheng Deng
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yunxuan Gao
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa 999078, China
| |
Collapse
|
26
|
Pharmacological Effects and Clinical Prospects of Cepharanthine. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248933. [PMID: 36558061 PMCID: PMC9782661 DOI: 10.3390/molecules27248933] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Cepharanthine is an active ingredient separated and extracted from Stephania cepharantha Hayata, a Menispermaceae plant. As a bisbenzylisoquinoline alkaloid, cepharanthine has various pharmacological properties, including antioxidant, anti-inflammatory, immunomodulatory, antitumoral, and antiviral effects. Following the emergence of coronavirus disease 2019 (COVID-19), cepharanthine has been found to have excellent anti-COVID-19 activity. In this review, the important physicochemical properties and pharmacological effects of cepharanthine, particularly the antiviral effect, are systematically described. Additionally, the molecular mechanisms and novel dosage formulations for the efficient, safe, and convenient delivery of cepharanthine are summarized.
Collapse
|
27
|
Saxena T, Sie C, Lin K, Ye D, Saatchi K, Häfeli UO. Potential of Nuclear Imaging Techniques to Study the Oral Delivery of Peptides. Pharmaceutics 2022; 14:2809. [PMID: 36559303 PMCID: PMC9780892 DOI: 10.3390/pharmaceutics14122809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Peptides are small biomolecules known to stimulate or inhibit important functions in the human body. The clinical use of peptides by oral delivery, however, is very limited due to their sensitive structure and physiological barriers present in the gastrointestinal tract. These barriers can be overcome with chemical and mechanical approaches protease inhibitors, permeation enhancers, and polymeric encapsulation. Studying the success of these approaches pre-clinically with imaging techniques such as fluorescence imaging (IVIS) and optical microscopy is difficult due to the lack of in-depth penetration. In comparison, nuclear imaging provides a better platform to observe the gastrointestinal transit and quantitative distribution of radiolabeled peptides. This review provides a brief background on the oral delivery of peptides and states examples from the literature on how nuclear imaging can help to observe and analyze the gastrointestinal transit of oral peptides. The review connects the fields of peptide delivery and nuclear medicine in an interdisciplinary way to potentially overcome the challenges faced during the study of oral peptide formulations.
Collapse
Affiliation(s)
- Tanya Saxena
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Claire Sie
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Kristine Lin
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Daisy Ye
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Katayoun Saatchi
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
| | - Urs O. Häfeli
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T1Z3, Canada
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
28
|
Tan JY, Li Y, Chamani F, Tharzeen A, Prakash P, Natarajan B, Sheth RA, Park WM, Kim A, Yoon D, Kim J. Experimental Validation of Diffraction Lithography for Fabrication of Solid Microneedles. MATERIALS (BASEL, SWITZERLAND) 2022; 15:8934. [PMID: 36556744 PMCID: PMC9787912 DOI: 10.3390/ma15248934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/05/2022] [Accepted: 12/11/2022] [Indexed: 06/17/2023]
Abstract
Microneedles are highly sought after for medicinal and cosmetic applications. However, the current manufacturing process for microneedles remains complicated, hindering its applicability to a broader variety of applications. As diffraction lithography has been recently reported as a simple method for fabricating solid microneedles, this paper presents the experimental validation of the use of ultraviolet light diffraction to control the liquid-to-solid transition of photosensitive resin to define the microneedle shape. The shapes of the resultant microneedles were investigated utilizing the primary experimental parameters including the photopattern size, ultraviolet light intensity, and the exposure time. Our fabrication results indicated that the fabricated microneedles became taller and larger in general when the experimental parameters were increased. Additionally, our investigation revealed four unique crosslinked resin morphologies during the first growth of the microneedle: microlens, first harmonic, first bell-tip, and second harmonic shapes. Additionally, by tilting the light exposure direction, a novel inclined microneedle array was fabricated for the first time. The fabricated microneedles were characterized with skin insertion and force-displacement tests. This experimental study enables the shapes and mechanical properties of the microneedles to be predicted in advance for mass production and wide practical use for biomedical or cosmetic applications.
Collapse
Affiliation(s)
- Jun Ying Tan
- Department of Electrical Engineering, University of North Texas, Denton, TX 76207, USA
| | - Yuankai Li
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Faraz Chamani
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Aabila Tharzeen
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Punit Prakash
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Balasubramaniam Natarajan
- Department of Electrical and Computer Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Rahul A. Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Won Min Park
- Tim Taylor Department of Chemical Engineering, Kansas State University, Manhattan, KS 66506, USA
| | - Albert Kim
- Department of Medical Engineering, The University of South Florida, Tampa, FL 33620, USA
| | - Donghoon Yoon
- College of Medicine, University of Arkansas for Medical Science, Little Rock, AR 72205, USA
| | - Jungkwun Kim
- Department of Electrical Engineering, University of North Texas, Denton, TX 76207, USA
| |
Collapse
|
29
|
Application of Nanoparticles: Diagnosis, Therapeutics, and Delivery of Insulin/Anti-Diabetic Drugs to Enhance the Therapeutic Efficacy of Diabetes Mellitus. LIFE (BASEL, SWITZERLAND) 2022; 12:life12122078. [PMID: 36556443 PMCID: PMC9783843 DOI: 10.3390/life12122078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/16/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder of carbohydrates, lipids, and proteins due to a deficiency of insulin secretion or failure to respond to insulin secreted from pancreatic cells, which leads to high blood glucose levels. DM is one of the top four noncommunicable diseases and causes of death worldwide. Even though great achievements were made in the management and treatment of DM, there are still certain limitations, mainly related to the early diagnosis, and lack of appropriate delivery of insulin and other anti-diabetic agents. Nanotechnology is an emerging field in the area of nanomedicine and NP based anti-diabetic agent delivery is reported to enhance efficacy by increasing bioavailability and target site accumulation. Moreover, theranostic NPs can be used as diagnostic tools for the early detection and prevention of diseases owing to their unique biological, physiochemical, and magnetic properties. NPs have been synthesized from a variety of organic and inorganic materials including polysaccharides, dendrimers, proteins, lipids, DNA, carbon nanotubes, quantum dots, and mesoporous materials within the nanoscale size. This review focuses on the role of NPs, derived from organic and inorganic materials, in the diagnosis and treatment of DM.
Collapse
|
30
|
Gelli HP, Vazquez-Uribe R, Sommer MOA. Screening for effective cell-penetrating peptides with minimal impact on epithelial cells and gut commensals in vitro. Front Pharmacol 2022; 13:1049324. [PMID: 36408245 PMCID: PMC9666501 DOI: 10.3389/fphar.2022.1049324] [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: 09/20/2022] [Accepted: 10/18/2022] [Indexed: 01/25/2023] Open
Abstract
One of the biggest challenges for oral drug absorption is the epithelial barrier of the gastrointestinal tract. The use of cell-penetrating peptides (CPPs) to modulate the epithelial barrier function is known to be an effective strategy to improve drug absorption and bioavailability. In this study we compare side-by-side, 9 most promising CPPs to study their cytotoxicity (Cytotox Red dye staining) and cell viability (AlamarBlue staining) on epithelial cells and their effects on paracellular permeability of the intestinal barrier in vitro in a differentiated Caco-2 epithelial monolayer model. The data revealed that 4 out of 9 well-studied CPPs significantly improved Caco-2 paracellular permeability without compromising on cellular health. To assess the impact of CPPs on the human microbiota we studied the antimicrobial effects of the 4 effective CPPs from our permeation studies against 10 representative strains of the gut microbiota in vitro using microbroth dilution. Our data revealed that these 4 CPPs affected the growth of almost all tested commensal strains. Interestingly, we found that two synthetic CPPs (Shuffle and Penetramax) outperformed all the other CPPs in their ability to increase intestinal paracellular permeability at 50 µM and had only a small to moderate effect on the tested gut commensal strains. Based on these data Shuffle and Penetramax represent relevant CPPs to be further characterized in vivo for safe delivery of poorly absorbed therapeutics while minimizing negative impacts on the gut microbiota.
Collapse
|
31
|
Han L, Liu XW, Zang T, Ren H, Liang DS, Bai SC, Li C, Liao XP, Liu YH, Zhang C, Sun J. H2S responsive PEGylated poly (lipoic acid) with ciprofloxacin for targeted therapy of Salmonella. J Control Release 2022; 351:896-906. [DOI: 10.1016/j.jconrel.2022.09.060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022]
|
32
|
Ghosh A, Liu W, Li L, Pahapale GJ, Choi SY, Xu L, Huang Q, Zhang R, Zhong Z, Selaru FM, Gracias DH. Autonomous Untethered Microinjectors for Gastrointestinal Delivery of Insulin. ACS NANO 2022; 16:16211-16220. [PMID: 36201302 PMCID: PMC9960177 DOI: 10.1021/acsnano.2c05098] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The delivery of macromolecular drugs via the gastrointestinal (GI) tract is challenging as these drugs display low stability as well as poor absorption across the intestinal epithelium. While permeation-enhancing drug delivery methods can increase the bioavailability of low molecular weight drugs, the effective delivery of high molecular weight drugs across the tight epithelial cell junctions remains a formidable challenge. Here, we describe autonomous microinjectors that are deployed in the GI tract, then efficiently penetrate the GI mucosa to deliver a macromolecular drug, insulin, to the systemic circulation. We performed in vitro studies to characterize insulin release and assess the penetration capability of microinjectors and we measured the in vivo release of insulin in live rats. We found that the microinjectors administered within the luminal GI tract could deliver insulin transmucosally to the systemic circulation at levels similar to those with intravenously administered insulin. Due to their small size, tunability in sizing and dosing, wafer-scale fabrication, and parallel, autonomous operation, we anticipate that these microinjectors will significantly advance drug delivery across the GI tract mucosa to the systemic circulation in a safe manner.
Collapse
Affiliation(s)
- Arijit Ghosh
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Wangqu Liu
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ling Li
- Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Gayatri J. Pahapale
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Si Young Choi
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Liyi Xu
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Qi Huang
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ruili Zhang
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Zijian Zhong
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Florin M. Selaru
- Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - David H. Gracias
- Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Sidney Kimmel Comprehensive Cancer Center (SKCCC), Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Center for Microphysiological Systems, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Chemistry, Johns Hopkins University, Baltimore, MD 21218, USA
- Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Laboratory for Computational Sensing and Robotics (LCSR), Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
33
|
Doolan JA, Williams GT, Hilton KLF, Chaudhari R, Fossey JS, Goult BT, Hiscock JR. Advancements in antimicrobial nanoscale materials and self-assembling systems. Chem Soc Rev 2022; 51:8696-8755. [PMID: 36190355 PMCID: PMC9575517 DOI: 10.1039/d1cs00915j] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Indexed: 11/21/2022]
Abstract
Antimicrobial resistance is directly responsible for more deaths per year than either HIV/AIDS or malaria and is predicted to incur a cumulative societal financial burden of at least $100 trillion between 2014 and 2050. Already heralded as one of the greatest threats to human health, the onset of the coronavirus pandemic has accelerated the prevalence of antimicrobial resistant bacterial infections due to factors including increased global antibiotic/antimicrobial use. Thus an urgent need for novel therapeutics to combat what some have termed the 'silent pandemic' is evident. This review acts as a repository of research and an overview of the novel therapeutic strategies being developed to overcome antimicrobial resistance, with a focus on self-assembling systems and nanoscale materials. The fundamental mechanisms of action, as well as the key advantages and disadvantages of each system are discussed, and attention is drawn to key examples within each field. As a result, this review provides a guide to the further design and development of antimicrobial systems, and outlines the interdisciplinary techniques required to translate this fundamental research towards the clinic.
Collapse
Affiliation(s)
- Jack A Doolan
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK.
| | - George T Williams
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Kira L F Hilton
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| | - Rajas Chaudhari
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| | - John S Fossey
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK.
| | - Jennifer R Hiscock
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| |
Collapse
|
34
|
Weller A, Hansen MB, Marie R, Hundahl AC, Hempel C, Kempen PJ, Frandsen HL, Parhamifar L, Larsen JB, Andresen TL. Quantifying the transport of biologics across intestinal barrier models in real-time by fluorescent imaging. Front Bioeng Biotechnol 2022; 10:965200. [PMID: 36159696 PMCID: PMC9500407 DOI: 10.3389/fbioe.2022.965200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Unsuccessful clinical translation of orally delivered biological drugs remains a challenge in pharmaceutical development and has been linked to insufficient mechanistic understanding of intestinal drug transport. Live cell imaging could provide such mechanistic insights by directly tracking drug transport across intestinal barriers at subcellular resolution, however traditional intestinal in vitro models are not compatible with the necessary live cell imaging modalities. Here, we employed a novel microfluidic platform to develop an in vitro intestinal epithelial barrier compatible with advanced widefield- and confocal microscopy. We established a quantitative, multiplexed and high-temporal resolution imaging assay for investigating the cellular uptake and cross-barrier transport of biologics while simultaneously monitoring barrier integrity. As a proof-of-principle, we use the generic model to monitor the transport of co-administrated cell penetrating peptide (TAT) and insulin. We show that while TAT displayed a concentration dependent difference in its transport mechanism and efficiency, insulin displayed cellular internalization, but was restricted from transport across the barrier. This illustrates how such a sophisticated imaging based barrier model can facilitate mechanistic studies of drug transport across intestinal barriers and aid in vivo and clinical translation in drug development.
Collapse
Affiliation(s)
- Arjen Weller
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Morten B. Hansen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Rodolphe Marie
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Adam C. Hundahl
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Casper Hempel
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Paul J. Kempen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- The National Centre for Nano Fabrication and Characterization, DTU Nanolab, Technical University of Denmark, Lyngby, Denmark
| | - Henrik L. Frandsen
- National Food Institute, Technical University of Denmark, Lyngby, Denmark
| | - Ladan Parhamifar
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Jannik B. Larsen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- *Correspondence: Jannik B. Larsen, ; Thomas L. Andresen,
| | - Thomas L. Andresen
- Center for Intestinal Absorption and Transport of Biopharmaceuticals, Technical University of Denmark, Lyngby, Denmark
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- *Correspondence: Jannik B. Larsen, ; Thomas L. Andresen,
| |
Collapse
|
35
|
An overview of recent advances in insulin delivery and wearable technology for effective management of diabetes. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
36
|
Moore TL, Cook AB, Bellotti E, Palomba R, Manghnani P, Spanò R, Brahmachari S, Di Francesco M, Palange AL, Di Mascolo D, Decuzzi P. Shape-specific microfabricated particles for biomedical applications: a review. Drug Deliv Transl Res 2022; 12:2019-2037. [PMID: 35284984 PMCID: PMC9242933 DOI: 10.1007/s13346-022-01143-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2022] [Indexed: 12/13/2022]
Abstract
The storied history of controlled the release systems has evolved over time; from degradable drug-loaded sutures to monolithic zero-ordered release devices and nano-sized drug delivery formulations. Scientists have tuned the physico-chemical properties of these drug carriers to optimize their performance in biomedical/pharmaceutical applications. In particular, particle drug delivery systems at the micron size regime have been used since the 1980s. Recent advances in micro and nanofabrication techniques have enabled precise control of particle size and geometry-here we review the utility of microplates and discoidal polymeric particles for a range of pharmaceutical applications. Microplates are defined as micrometer scale polymeric local depot devices in cuboid form, while discoidal polymeric nanoconstructs are disk-shaped polymeric particles having a cross-sectional diameter in the micrometer range and a thickness in the hundreds of nanometer range. These versatile particles can be used to treat several pathologies such as cancer, inflammatory diseases and vascular diseases, by leveraging their size, shape, physical properties (e.g., stiffness), and component materials, to tune their functionality. This review highlights design and fabrication strategies for these particles, discusses their applications, and elaborates on emerging trends for their use in formulations.
Collapse
Affiliation(s)
- Thomas L Moore
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy.
| | - Alexander B Cook
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Elena Bellotti
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Purnima Manghnani
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Raffaele Spanò
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Martina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Anna Lisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Daniele Di Mascolo
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano Di Tecnologia, Via Morego, 30, 16163, Genoa, Italy
| |
Collapse
|
37
|
Mahmood A, Haneef R, Al Meslamani AZ, Bostanudin MF, Sohail M, Sarfraz M, Arafat M. Papain-Decorated Mucopenetrating SEDDS: A Tentative Approach to Combat Absorption Issues of Acyclovir via the Oral Route. Pharmaceutics 2022; 14:pharmaceutics14081584. [PMID: 36015210 PMCID: PMC9412565 DOI: 10.3390/pharmaceutics14081584] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of the current study was to enhance the oral bioavailability of Acyclovir (ACV) based on the papain-functionalized self-emulsifying drug delivery systems (SEDDS). The optimum control SEDDS formulation comprised of kolliphore (40%), transcutol (30%), propylene glycol (20%) and oleoyl chloride (10%). However, in the targeted SEDDS formulation, oleoyl chloride was replaced with oleoyl chloride-papain (OC-PAP) conjugate that was synthesized via an amide bond formation between the acyl halide groups of oleoyl chloride and the amino group of papain. Prior to adding in the SEDDS formulation, the newly synthesized conjugate was evaluated quantitatively by a Bradford assay that demonstrated 45 µg of papain contents per mg of the conjugate. Moreover, the conjugate formation was qualitatively confirmed through FTIR analysis and thin layer chromatography. ACV (a BCS class III drug) was incorporated into the SEDDS formulations after being hydrophobically ion paired with sodium deoxycholate, thereby making it lipophilic. The drug-loaded formulations were emulsified in the 0.1 M phosphate buffer (pH 6.8) and evaluated in vitro with respect to drug release and rabbit mucosal permeation studies. Both the formulations illustrated a very comparable drug release over a period of 4 h, afterwards, the OC-PAP-based formulation demonstrated a more sustaining effect. The extent of mucus diffusion evaluated via the silicon tube method demonstrated a 4.92-fold and a 1.46-fold higher penetration of the drug, a 3.21-fold and a 1.56-fold higher permeation through the rabbit intestinal mucus layer, and a 22.94-fold and a 2.27-fold higher retention of the drug over the intact mucosa of rabbit intestine, illustrated by OC-PAP-based nanoemulsions compared to the drug-free solution and controlled nanoemulsion, respectively. According to these in vitro results, papain-functionalized SEDDS is a promising approach for the oral delivery of ACV and many other drugs with oral bioavailability issues, however, in vivo studies in this respect have to be employed before making a comprehensive conclusion.
Collapse
Affiliation(s)
- Arshad Mahmood
- College of Pharmacy, Al Ain University, Abu Dhabi Campus, Abu Dhabi P.O. Box 112612, United Arab Emirates; (A.Z.A.M.); (M.F.B.)
- AAU Health and Biomedical Research Center (HBRC), Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates; (M.S.); (M.A.)
- Correspondence:
| | - Rabbia Haneef
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan; (R.H.); (M.S.)
| | - Ahmad Z. Al Meslamani
- College of Pharmacy, Al Ain University, Abu Dhabi Campus, Abu Dhabi P.O. Box 112612, United Arab Emirates; (A.Z.A.M.); (M.F.B.)
- AAU Health and Biomedical Research Center (HBRC), Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates; (M.S.); (M.A.)
| | - Mohammad F. Bostanudin
- College of Pharmacy, Al Ain University, Abu Dhabi Campus, Abu Dhabi P.O. Box 112612, United Arab Emirates; (A.Z.A.M.); (M.F.B.)
- AAU Health and Biomedical Research Center (HBRC), Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates; (M.S.); (M.A.)
| | - Muhammad Sohail
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan; (R.H.); (M.S.)
| | - Muhammad Sarfraz
- AAU Health and Biomedical Research Center (HBRC), Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates; (M.S.); (M.A.)
- College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates
| | - Mosab Arafat
- AAU Health and Biomedical Research Center (HBRC), Al Ain University, Abu Dhabi P.O. Box 112612, United Arab Emirates; (M.S.); (M.A.)
- College of Pharmacy, Al Ain University, Al Ain P.O. Box 64141, United Arab Emirates
| |
Collapse
|
38
|
Li DF, Yang MF, Xu HM, Zhu MZ, Zhang Y, Tian CM, Nie YQ, Wang JY, Liang YJ, Yao J, Wang LS. Nanoparticles for oral delivery: targeted therapy for inflammatory bowel disease. J Mater Chem B 2022; 10:5853-5872. [PMID: 35876136 DOI: 10.1039/d2tb01190e] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As a group of chronic and idiopathic gastrointestinal (GI) disorders, inflammatory bowel disease (IBD) is characterized by recurrent intestinal mucosal inflammation. Oral administration is critical for the treatment of IBD. Unfortunately, it is difficult to target the bowel located in the GI tract due to multiple physical barriers. The unique physicochemical properties of nanoparticle-based drug delivery systems (DDSs) and their enhanced permeability and retention effects in the inflamed bowel, render nanomedicines to be used to implement precise drug delivery at diseased sites in IBD therapy. In this review, we described the pathophysiological features of IBD, and designed strategies to exploit these features for intestinal targeting. In addition, we introduced the types of currently developed nano-targeted carriers, including synthetic nanoparticle-based and emerging naturally derived nanoparticles (e.g., extracellular vesicles and plant-derived nanoparticles). Moreover, recent developments in targeted oral nanoparticles for IBD therapy were also highlighted. Finally, we presented challenges associated with nanotechnology and potential directions for future IBD treatment.
Collapse
Affiliation(s)
- De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen 518020, Guangdong, China.
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen 518020, Guangdong, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510030, China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510030, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou 516000, Guangdong, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Yu-Qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510030, China
| | - Jian-Yao Wang
- Department of General Surgery, Shenzhen Children's Hospital, No. 7019, Yitian Road, Futian District, Shenzhen 518026, Guangdong, China.
| | - Yu-Jie Liang
- Shenzhen Kangning Hospital, No. 1080, Cuizu Road, Luohu District, Shenzhen 518020, Guangdong, China.
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen 518020, Guangdong, China.
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen 518020, Guangdong, China.
| |
Collapse
|
39
|
Mohammed Y, Holmes A, Kwok PCL, Kumeria T, Namjoshi S, Imran M, Matteucci L, Ali M, Tai W, Benson HA, Roberts MS. Advances and future perspectives in epithelial drug delivery. Adv Drug Deliv Rev 2022; 186:114293. [PMID: 35483435 DOI: 10.1016/j.addr.2022.114293] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 04/09/2022] [Indexed: 12/12/2022]
Abstract
Epithelial surfaces protect exposed tissues in the body against intrusion of foreign materials, including xenobiotics, pollen and microbiota. The relative permeability of the various epithelia reflects their extent of exposure to the external environment and is in the ranking: intestinal≈ nasal ≥ bronchial ≥ tracheal > vaginal ≥ rectal > blood-perilymph barrier (otic), corneal > buccal > skin. Each epithelium also varies in their morphology, biochemistry, physiology, immunology and external fluid in line with their function. Each epithelium is also used as drug delivery sites to treat local conditions and, in some cases, for systemic delivery. The associated delivery systems have had to evolve to enable the delivery of larger drugs and biologicals, such as peptides, proteins, antibodies and biologicals and now include a range of physical, chemical, electrical, light, sound and other enhancement technologies. In addition, the quality-by-design approach to product regulation and the growth of generic products have also fostered advancement in epithelial drug delivery systems.
Collapse
|
40
|
Zhang JY, Liu XX, Lin JY, Bao XY, Peng JQ, Gong ZP, Luan X, Chen Y. Biomimetic engineered nanocarriers inspired by viruses for oral-drug delivery. Int J Pharm 2022; 624:121979. [DOI: 10.1016/j.ijpharm.2022.121979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 06/20/2022] [Accepted: 06/30/2022] [Indexed: 10/17/2022]
|
41
|
Chen H, Lu Y, Shi S, Zhang Q, Cao X, Sun L, An D, Zhang X, Kong X, Liu J. Design and Development of a New Glucagon-Like Peptide-1 Receptor Agonist to Obtain High Oral Bioavailability. Pharm Res 2022; 39:1891-1906. [PMID: 35698011 DOI: 10.1007/s11095-022-03265-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/18/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Semaglutide is the only oral GLP-1 RA in the market, but oral bioavailability is generally limited in range of 0.4-1%. In this study, a new GLP-1RA named SHR-2042 was developed to gain higher oral bioavailability than semaglutide. METHOD Self-association of SHR-2042, semaglutide and liraglutide were assessed using SEC-MALS. The intestinal perfusion test in SD rats was used to select permeation enhancers (PEs) including SNAC, C10 and LCC. ITC, CD and DLS were used to explore the interaction between SHR-2042 and SNAC. Gastric administrated test in SD rats was used to screen SHR-2042 granules with different SHR-2042/SNAC ratios. The oral bioavailability of SHR-2042 was studied in rats and monkeys. RESULT The designed GLP-1RA, SHR-2042, gives a better solubility and lipophilicity than semaglutide. While it forms a similar oligomer with that of semaglutide. During the selection of PEs, SNAC shows better exposure than the other competing PEs including C10 and LCC. SHR-2042 and SNAC bind quickly and exhibit hydrophobic interaction. SNAC could promote monomerization of SHR-2042 and form micelles to trap the monomerized SHR-2042. The oral bioavailability of SHR-2042 paired with SNAC is 0.041% (1:0, w/w), 0.083% (1:10, w/w), 0.32% (1:30, w/w) and 2.83% (1:60, w/w) in rats. And the oral bioavailability of SHR-2042 matched with SNAC is 3.39% (1:30, w/w) in monkeys, which is over 10 times higher than that of semaglutide. CONCLUSION We believe that the design and development of oral SHR-2042 will provide a new way to design more and more GLP-1RAs with high oral bioavailability in the future.
Collapse
Affiliation(s)
- Hao Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Yun Lu
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Shuai Shi
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Qiang Zhang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Xiaoli Cao
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Lei Sun
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Dong An
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Xiaojie Zhang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Xianglin Kong
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Lianyungang, 222000, People's Republic of China
| | - Jianping Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
42
|
Iyer G, Dyawanapelly S, Jain R, Dandekar P. An overview of oral insulin delivery strategies (OIDS). Int J Biol Macromol 2022; 208:565-585. [PMID: 35346680 DOI: 10.1016/j.ijbiomac.2022.03.144] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/07/2022] [Accepted: 03/22/2022] [Indexed: 02/07/2023]
Abstract
Despite tremendous efforts, the world continues its fight against the common chronic disease-diabetes. Diabetes is caused by elevated glucose levels in the blood, which can lead to several complications like glaucoma, cataract, kidney failure, diabetic ketoacidosis, heart attack, and stroke. According to recent statistics, China, India, and the US rank at the top three positions with regards to the number of patients affected by diabetes. Ever since its discovery, insulin is one of the major therapeutic molecules that is used to control the disease in the diabetic population, worldwide. The most common route of insulin administration has been the subcutaneous route. However, the limitations associated with this route have motivated global efforts to explore alternative strategies to deliver insulin, including pulmonary, transdermal, nasal, rectal, buccal, and oral routes. Oral insulin delivery is the most convenient and patient-centered route. However, the oral route is also associated with numerous drawbacks that present significant challenges to the scientific fraternity. The human physiological system acts as a formidable barrier to insulin, limiting its bioavailability. The present review covers the major barriers against oral insulin delivery and explains formulation strategies that have been adopted to overcome these barriers. The review focuses on oral insulin delivery strategies (OIDS) for increasing the bioavailability of oral insulin, including nanoparticles, microparticles, nano-in-microparticles, hydrogels, tablets, capsules, intestinal patches, and use of ionic liquids. It also highlights some of the notable recent advancements and clinical trials in oral insulin delivery. This formulation based OIDS may significantly improve patient compliance in the treatment of diabetes.
Collapse
Affiliation(s)
- Gayatri Iyer
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, NP Marg, Matunga, Mumbai 400019, India
| | - Sathish Dyawanapelly
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, NP Marg, Matunga, Mumbai 400019, India
| | - Ratnesh Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Matunga, Mumbai 400019, India.
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, NP Marg, Matunga, Mumbai 400019, India.
| |
Collapse
|
43
|
Kulkarni D, Damiri F, Rojekar S, Zehravi M, Ramproshad S, Dhoke D, Musale S, Mulani AA, Modak P, Paradhi R, Vitore J, Rahman MH, Berrada M, Giram PS, Cavalu S. Recent Advancements in Microneedle Technology for Multifaceted Biomedical Applications. Pharmaceutics 2022; 14:1097. [PMID: 35631683 PMCID: PMC9144002 DOI: 10.3390/pharmaceutics14051097] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/07/2022] [Accepted: 05/17/2022] [Indexed: 02/04/2023] Open
Abstract
Microneedle (MNs) technology is a recent advancement in biomedical science across the globe. The current limitations of drug delivery, like poor absorption, low bioavailability, inadequate skin permeation, and poor biodistribution, can be overcome by MN-based drug delivery. Nanotechnology made significant changes in fabrication techniques for microneedles (MNs) and design shifted from conventional to novel, using various types of natural and synthetic materials and their combinations. Nowadays, MNs technology has gained popularity worldwide in biomedical research and drug delivery technology due to its multifaceted and broad-spectrum applications. This review broadly discusses MN's types, fabrication methods, composition, characterization, applications, recent advancements, and global intellectual scenarios.
Collapse
Affiliation(s)
- Deepak Kulkarni
- Department of Pharmaceutics, Srinath College of Pharmacy, Bajajnagar, Aurangabad 431136, India;
| | - Fouad Damiri
- Laboratory of Biomolecules and Organic Synthesis (BIOSYNTHO), Department of Chemistry, Faculty of Sciences Ben M’Sick, University Hassan II of Casablanca, Casablanca 20000, Morocco; (F.D.); (M.B.)
| | - Satish Rojekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India;
- Departments of Medicine and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University, Alkharj 11942, Saudi Arabia;
| | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh;
| | - Dipali Dhoke
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India;
| | - Shubham Musale
- Department of Pharmaceutics, Dr. DY Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; (S.M.); (A.A.M.); (P.M.); (R.P.)
| | - Ashiya A. Mulani
- Department of Pharmaceutics, Dr. DY Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; (S.M.); (A.A.M.); (P.M.); (R.P.)
| | - Pranav Modak
- Department of Pharmaceutics, Dr. DY Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; (S.M.); (A.A.M.); (P.M.); (R.P.)
| | - Roshani Paradhi
- Department of Pharmaceutics, Dr. DY Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; (S.M.); (A.A.M.); (P.M.); (R.P.)
| | - Jyotsna Vitore
- National Institute of Pharmaceutical Education and Research, Ahmedabad 160062, India;
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea
| | - Mohammed Berrada
- Laboratory of Biomolecules and Organic Synthesis (BIOSYNTHO), Department of Chemistry, Faculty of Sciences Ben M’Sick, University Hassan II of Casablanca, Casablanca 20000, Morocco; (F.D.); (M.B.)
| | - Prabhanjan S. Giram
- Department of Pharmaceutics, Dr. DY Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune 411018, India; (S.M.); (A.A.M.); (P.M.); (R.P.)
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|
44
|
Wang L, Wang Z, Pan Y, Chen S, Fan X, Li X, Chen G, Ma Y, Cai Y, Zhang J, Yang H, Xiao W, Yu M. Polycatechol-Derived Mesoporous Polydopamine Nanoparticles for Combined ROS Scavenging and Gene Interference Therapy in Inflammatory Bowel Disease. ACS APPLIED MATERIALS & INTERFACES 2022; 14:19975-19987. [PMID: 35442639 DOI: 10.1021/acsami.1c25180] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Benefiting from the evolution of nanotechnology, the combination therapy by gene interference and reactive oxygen species (ROS) scavenging are expected, which holds great potential in inflammatory bowel disease (IBD) therapy. However, the functional integration of different therapeutic modules through interface modification of gene vectors for safe and efficient treatment is urgently needed. Herein, we present a catechol chemistry-mediated core-shell nanoplatform for ROS scavenging-mediated oxidative stress alleviation and siRNA-mediated gene interference in a dextran sulfate sodium (DSS)-induced colitis model. The nanoplatform is constructed by employing mesoporous polydopamine nanoparticles (MPDA NPs) with surface modification of amines as the porous core for TNF-α-siRNA loading (31 wt %) and exerts an antioxidant function, while PDA-induced biomineralization of the calcium phosphate (CaP) coating is used as the pH-sensitive protective shell to prevent siRNA from premature release. The CaP layer degraded under weakly acidic subcellular conditions (lysosomes); thus, the synergistic integration of catechol and cation moieties on the exposed surface of MPDA resulted in an efficient lysosomal escape. Subsequently, effective ROS scavenging caused by the electron-donating ability of MPDA and efficient knocking down (40.5%) of tumor necrosis factor-α (TNF-α) via sufficient cytosolic gene delivery resulted in a synergistic anti-inflammation therapeutic effect both in vitro and in vivo. This work establishes the first paradigm of synergistic therapy in IBD by ROS scavenging and gene interference.
Collapse
Affiliation(s)
- Liucan Wang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Zhenqiang Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing 400044, China
| | - Yiyang Pan
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Shuaishuai Chen
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Xin Fan
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Xiaolong Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Guoqing Chen
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Yuanhang Ma
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Yujiao Cai
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Jixi Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, No. 174 Shazheng Road, Chongqing 400044, China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Weidong Xiao
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| | - Min Yu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, No. 183 Xinqiao Road, Chongqing 400037, China
| |
Collapse
|
45
|
Ostadhossein F, Moitra P, Gunaseelan N, Nelappana M, Lowe C, Moghiseh M, Butler A, de Ruiter N, Mandalika H, Tripathi I, Misra SK, Pan D. Hitchhiking probiotic vectors to deliver ultra-small hafnia nanoparticles for 'Color' gastrointestinal tract photon counting X-ray imaging. NANOSCALE HORIZONS 2022; 7:533-542. [PMID: 35311837 DOI: 10.1039/d1nh00626f] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Gastrointestinal (GI) tract is one of the hard-to-reach target tissues for the delivery of contrast agents and drugs mediated by nanoparticles due to its harsh environment. Herein, we overcame this barrier by designing orally ingestible probiotic vectors for 'hitchhiking' ultrasmall hafnia (HfO2) (∼1-2 nm) nanoparticles. The minute-made synthesis of these nanoparticles is accomplished through a simple reduction reaction. These nanoparticles were incubated with probiotic bacteria with potential health benefits and were non-specifically taken up due to their small size. Subsequently, the bacteria were lyophilized and packed into a capsule to be administered orally as the radiopaque contrast agents for delineating the GI features. These nano-bio-hybrid entities could successfully be utilized as contrast agents in vivo in the conventional and multispectral computed tomography (CT). We demonstrated in 'color' the accumulated nanoparticles using advanced detectors of the photon counting CT. The enhanced nano-bio-interfacing capability achieved here can circumvent traditional nanoparticle solubility and delivery problems while offering a patient friendly approach for GI imaging to replace the currently practiced barium meal.
Collapse
Affiliation(s)
- Fatemeh Ostadhossein
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 611 West Park Street, Urbana, IL, USA.
- Beckman Institute of Advanced Science and Technology, 405 N. Mathews M/C 251, Urbana, IL 61801-2325, USA
| | - Parikshit Moitra
- Departments of Diagnostic Radiology and Nuclear Medicine and Pediatrics, University of Maryland Baltimore, 670 W Baltimore St., Baltimore, Maryland, 21201, USA
| | - Nivetha Gunaseelan
- Departments of Diagnostic Radiology and Nuclear Medicine and Pediatrics, University of Maryland Baltimore, 670 W Baltimore St., Baltimore, Maryland, 21201, USA
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, Maryland, 21250, USA
| | - Michael Nelappana
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 611 West Park Street, Urbana, IL, USA.
| | - Chiara Lowe
- University of Otago Christchurch, Christchurch, New Zealand
| | - Mahdieh Moghiseh
- University of Otago Christchurch, Christchurch, New Zealand
- MARS Bioimaging Limited, Christchurch, New Zealand
| | - Anthony Butler
- University of Otago Christchurch, Christchurch, New Zealand
- MARS Bioimaging Limited, Christchurch, New Zealand
- University of Canterbury, Christchurch, New Zealand
- European Organization for Nuclear Research (CERN), Geneva, Switzerland
- Human Interface Technology Laboratory New Zealand, University of Canterbury, Christchurch, New Zealand
| | - Niels de Ruiter
- University of Otago Christchurch, Christchurch, New Zealand
- MARS Bioimaging Limited, Christchurch, New Zealand
- University of Canterbury, Christchurch, New Zealand
- Human Interface Technology Laboratory New Zealand, University of Canterbury, Christchurch, New Zealand
| | - Harish Mandalika
- MARS Bioimaging Limited, Christchurch, New Zealand
- University of Canterbury, Christchurch, New Zealand
- Human Interface Technology Laboratory New Zealand, University of Canterbury, Christchurch, New Zealand
| | - Indu Tripathi
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 611 West Park Street, Urbana, IL, USA.
| | - Santosh K Misra
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 611 West Park Street, Urbana, IL, USA.
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, 611 West Park Street, Urbana, IL, USA.
- Beckman Institute of Advanced Science and Technology, 405 N. Mathews M/C 251, Urbana, IL 61801-2325, USA
- Departments of Diagnostic Radiology and Nuclear Medicine and Pediatrics, University of Maryland Baltimore, 670 W Baltimore St., Baltimore, Maryland, 21201, USA
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, Maryland, 21250, USA
- Department of Materials Science and Engineering, 201 Materials Science and Engineering Building, 1304 W. Green St. MC 246, Urbana, IL 61801, USA
| |
Collapse
|
46
|
Ionic Liquids: Promising Approach for Oral Drug Delivery. Pharm Res 2022; 39:2353-2365. [PMID: 35449344 DOI: 10.1007/s11095-022-03260-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/06/2022] [Indexed: 12/22/2022]
Abstract
Oral administration is the most preferred route for drug administration in clinic. However, due to unsatisfactory physicochemical properties of drugs and various physiological barriers, the oral bioavailability of most poorly water-soluble and macromolecules drugs is low and the therapeutic effect is unsatisfactory. Ionic liquids (ILs), molten salts with unique properties, show amazing potential for oral delivery. In addition to being able to form active pharmaceutical ingredients based ILs (API-ILs) to overcome drug solubility and polymorphism issues, ILs have also been used to enhance the solubility of poorly soluble drugs, enhance drug stability in the gastrointestinal environment, improve drug permeability in intestinal mucus, and facilitate drug penetration across the intestinal epithelial barrier. Furthermore, ILs were attempted as formulation components to develop novel oral drug delivery systems. This review focus on the application progress of ILs in oral drug delivery and the mechanisms. The challenges and perspectives of the development of ILs-based oral delivery systems are also discussed. This article reviews the latest advances of ionic liquids for oral drug delivery, focusing on the application and related mechanisms of ionic liquids in improving the drug physicochemical properties and enhancing drug delivery across physiological barriers.
Collapse
|
47
|
Kaffash E, Ali Shahbazi M, Hatami H, Nokhodchi A. An insight into gastrointestinal macromolecule delivery using physical oral devices. Drug Discov Today 2022; 27:2309-2321. [PMID: 35460891 DOI: 10.1016/j.drudis.2022.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/25/2022] [Accepted: 04/13/2022] [Indexed: 11/03/2022]
Abstract
Oral delivery is preferred over other routes of drug administration by both patients and physicians. The bioavailability of some therapeutics that are delivered via the oral route is restricted due to the protease- and bacteria-rich environment in the gastrointestinal tract, and by the pH variability along the delivery route. Given these harsh environments, the oral delivery of therapeutic macromolecules is complicated and remains challenging. Various formulation approaches, including the use of permeation enhancers and nanosized carriers, as well as chemical alteration of the drug structure, have been studied as ways to improve the oral absorption of macromolecular drugs. Nevertheless, the bioavailability of marketed oral peptide medicines is often relatively poor. This review highlights the most recent and promising physical methods for improving the oral bioavailability of macromolecules such as peptides. These methods include microneedle injections, high-speed stream injectors, magnetic drug targeting, expandable hydrogels, and iontophoresis. We highlight the potential and challenges of these new technologies, which may impact the future approaches used by pharmaceutical companies to create more efficient and safer orally administered macromolecules. Teaser: Despite substantial effort, the oral delivery of macromolecules remains challenging due to their low bioavailability. This review discusses the potential, challenges, and safety concerns associated with new technologies and devices for oral macromolecule delivery.
Collapse
Affiliation(s)
- Ehsan Kaffash
- 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
| | - Mohammad Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, 45139-56184 Zanjan, Iran.
| | - Hooman Hatami
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Nokhodchi
- Pharmaceutics Research Laboratory, School of Life Sciences, University of Sussex, Brighton, UK.
| |
Collapse
|
48
|
Kremsmayr T, Aljnabi A, Blanco-Canosa JB, Tran HNT, Emidio NB, Muttenthaler M. On the Utility of Chemical Strategies to Improve Peptide Gut Stability. J Med Chem 2022; 65:6191-6206. [PMID: 35420805 PMCID: PMC9059125 DOI: 10.1021/acs.jmedchem.2c00094] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
Inherent susceptibility
of peptides to enzymatic degradation in
the gastrointestinal tract is a key bottleneck in oral peptide drug
development. Here, we present a systematic analysis of (i) the gut
stability of disulfide-rich peptide scaffolds, orally administered
peptide therapeutics, and well-known neuropeptides and (ii) medicinal
chemistry strategies to improve peptide gut stability. Among a broad
range of studied peptides, cyclotides were the only scaffold class
to resist gastrointestinal degradation, even when grafted with non-native
sequences. Backbone cyclization, a frequently applied strategy, failed
to improve stability in intestinal fluid, but several site-specific
alterations proved efficient. This work furthermore highlights the
importance of standardized gut stability test conditions and suggests
defined protocols to facilitate cross-study comparison. Together,
our results provide a comparative overview and framework for the chemical
engineering of gut-stable peptides, which should be valuable for the
development of orally administered peptide therapeutics and molecular
probes targeting receptors within the gastrointestinal tract.
Collapse
Affiliation(s)
- Thomas Kremsmayr
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Währinger Straße 38, Vienna 1090, Austria
| | - Aws Aljnabi
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Währinger Straße 38, Vienna 1090, Austria
| | - Juan B Blanco-Canosa
- Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, Barcelona 08034, Spain
| | - Hue N T Tran
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Nayara Braga Emidio
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Markus Muttenthaler
- Faculty of Chemistry, Institute of Biological Chemistry, University of Vienna, Währinger Straße 38, Vienna 1090, Austria.,Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia
| |
Collapse
|
49
|
Kishimoto H, Ridley C, Thornton DJ. The lipophilic cyclic peptide cyclosporin A induces aggregation of gel-forming mucins. Sci Rep 2022; 12:6153. [PMID: 35418571 PMCID: PMC9008041 DOI: 10.1038/s41598-022-10125-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/04/2022] [Indexed: 12/05/2022] Open
Abstract
Cyclic peptides are good candidates for orally delivered therapeutics, however, issues remain in their development due to low intestinal permeability. Although some of the biological factors have been reported that regulate intestinal permeation of cyclic peptides, the influence of the mucus barrier, a major hurdle to epithelial drug delivery, on cyclic peptide bioavailability is unclear. In this study, we show that the lipophilic cyclic peptide, cyclosporin A (CsA), interacted with, and likely induced aggregation, of polymeric, gel-forming mucins (MUC2, MUC5AC and MUC5B) which underpin the mucus gel-networks in the gastrointestinal tract. Under similar conditions, two other cyclic peptides (daptomycin and polymyxin B) did not cause mucin aggregation. Using rate-zonal centrifugation, purified MUC2, MUC5AC and MUC5B mucins sedimented faster in the presence of CsA, with a significant increase in mucins in the pellet fraction. In contrast, mucin sedimentation profiles were largely unaltered after treatment with daptomycin or polymyxin B. CsA increased MUC5B sedimentation was concentration-dependent, and sedimentation studies using recombinant mucin protein domains suggests CsA most likely causes aggregation of the relatively non-O-glycosylated N-terminal and C-terminal regions of MUC5B. Furthermore, the aggregation of the N-terminal region, but not the C-terminal region, was affected by pH. CsA has partially N-methylated amide groups, this unique molecular structure, not present in daptomycin and polymyxin B, may potentially be involved in interaction with gel-forming mucin. Taken together, our results indicate that the interaction of gel-forming mucins with the cyclic peptide CsA is mediated at the N- and C-terminal domains of mucin polymers under physiological conditions. Our findings demonstrate that the mucus barrier is an important physiological factor regulating the intestinal permeation of cyclic peptides in vivo.
Collapse
Affiliation(s)
- Hisanao Kishimoto
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, 1432-1, Horinouchi, Hachioji, Tokyo, 192-0392, Japan. .,Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| | - Caroline Ridley
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - David J Thornton
- Wellcome Trust Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Sciences Centre, The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
50
|
Green Synthesis of Silver Nanoparticles Using Musa balbisiana and Their Cytotoxic Effect on HL-60 and SiHa Cancer Cells Through Clathrin-Mediated Endocytosis. BIONANOSCIENCE 2022. [DOI: 10.1007/s12668-022-00955-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|