1
|
Gao Y, Wang N, Qi Y, Wang X, Zhang K, Zhang Y, Cao Y, Zang T, Wang T. The aluminum nanoparticle-encircled SQ-in-water emulsions (ANSWE) as a vaccine adjuvant-delivery system (VADS) for developing robust mucosal subunit vaccines. BIOMATERIALS ADVANCES 2025; 166:214076. [PMID: 39490192 DOI: 10.1016/j.bioadv.2024.214076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
The aluminum nanoparticle-encircled squalene (SQ)-in-water emulsions (ANSWE) were engineered as a VADS (vaccine adjuvant-delivery system) using a simple procedure for carrying antigens (Ag) to develop subunit vaccines. In vitro, due to possessing the synergistic adjuvanticity of both AN and SQ, ANSWE were efficiently taken up by APC (antigen-presenting cells) and triggered them to mature and make extra ROS (reactive oxygen species) and multiple cytokines, such as IL-12, IL-1β and IFN-β, which favor balanced Th1/Th2 immunoresponses. Within APC, ANSWE managed lysosome escape and consequently enhanced proteasome activities to facilitate Ag cross-presentation. Mice given twice ovalbumin-ANSWE via intrapulmonary vaccination (IPV) produced high levels of anti-Ag antibodies as well as cytotoxic T lymphocytes, which efficiently erased cells bearing cognate Ag. Thus, ANSWE as a potent VADS may be feasible for developing mucosal subunit vaccines that can elicit comprehensive immunity against infectious diseases, including especially the respiratory infections, and even aggressive cancers.
Collapse
Affiliation(s)
- Yuhao Gao
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 420 Jade Road, Hefei, Anhui Province 230601, China
| | - Yuanyuan Qi
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Xiujuan Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Keyi Zhang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Yuxi Zhang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Yachen Cao
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Tairan Zang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China.
| |
Collapse
|
2
|
Wang N, Wang C, Wei C, Chen M, Gao Y, Zhang Y, Wang T. Constructing the cGAMP-Aluminum Nanoparticles as a Vaccine Adjuvant-Delivery System (VADS) for Developing the Efficient Pulmonary COVID-19 Subunit Vaccines. Adv Healthc Mater 2024; 13:e2401650. [PMID: 39319481 DOI: 10.1002/adhm.202401650] [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: 05/04/2024] [Revised: 09/05/2024] [Indexed: 09/26/2024]
Abstract
The cGAMP-aluminum nanoparticles (CAN) are engineered as a vaccine adjuvant-delivery system to carry mixed RBD (receptor-binding domain) of the original severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its new variant for developing bivalent pulmonary coronavirus disease 2019 (COVID-19) vaccines (biRBD-CAN). High phosphophilicity/adsorptivity made intrapulmonary CAN instantly form the pulmonary ingredient-coated CAN (piCAN) to possess biomimetic features enhancing biocompatibility. In vitro biRBD-CAN sparked APCs (antigen-presenting cells) to mature and make extra reactive oxygen species, engendered lysosome escape effects and enhanced proteasome activities. Through activating the intracellular stimulator of interferon genes (STING) and nucleotide-binding domain and leucine-rich repeat and pyrin domain containing proteins 3 (NALP3) inflammasome pathways to exert synergy between cGAMP and AN, biRBD-CAN stimulated APCs to secret cytokines favoring mixed Th1/Th2 immunoresponses. Mice bearing twice intrapulmonary biRBD-CAN produced high levels of mucosal antibodies, the long-lasting systemic antibodies, and potent cytotoxic T lymphocytes which efficiently erased cells displaying cognate epitopes. Notably, biRBD-CAN existed in mouse lungs and different lymph nodes for at least 48 h, unveiling their sustained immunostimulatory activity as the main mechanism underlying the long-lasting immunity and memory. Hamsters bearing twice intrapulmonary biRBD-CAN developed high resistance to pseudoviral challenges performed using different recombinant strains including the ones with distinct SARS-CoV-2-spike mutations. Thus, biRBD-CAN as a broad-spectrum pulmonary COVID-19 vaccine candidate may provide a tool for controlling the emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 420 Jade Road, Hefei, Anhui Province, 230601, China
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Can Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
- Department of Pharmacy, The Second People's Hospital of Lianyungang, 41 Hailian East Road, Lianyungang, Jiangsu Province, 222006, China
| | - Chunliu Wei
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Minnan Chen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Yuhao Gao
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Yuxi Zhang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province, 230032, China
| |
Collapse
|
3
|
Zheng Z, Li M, Yang J, Zhou X, Chen Y, Silli EK, Tang J, Gong S, Yuan Y, Zong Y, Kong J, Chen P, Yu L, Luo S, Wang Y, Tan C. Growth inhibition of pancreatic cancer by targeted delivery of gemcitabine via fucoidan-coated pH-sensitive liposomes. Int J Biol Macromol 2024; 277:134517. [PMID: 39111497 DOI: 10.1016/j.ijbiomac.2024.134517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 08/10/2024]
Abstract
Fucoidan-coated pH sensitive liposomes were designed for targeted delivery of gemcitabine (FU-GEM PSL) to treat pancreatic cancer (PC). FU-GEM PSL had a particle size of 175.3 ± 4.9 nm, zeta potential of -19.0 ± 3.7 mV, encapsulation efficiency (EE) of 74.05 ± 0.17 %, and drug loading (DL) of 21.27 ± 0.05 %. Cell experiments in vitro showed that FU-GEM PSL could increase the release of GEM and drug concentration, and could inhibit tumor cell proliferation by affecting the cell cycle. FU-GEM PSL entered cells through macropinocytosis and caveolin-mediated endocytosis to exert effects. Meanwhile, the expression of P-selectin was detected in human tissues, demonstrating the feasibility of targeting FU. Moreover, combined with animal experiments in vivo, FU-GEM PSL could inhibit the development of PC. Furthermore, anti-tumor experiments in vivo carried on BALB/c mice indicated that FU-GEM PSL had tumor suppression abilities and safety. Therefore, FU-GEM PSL is a promising formulation for PC therapy.
Collapse
Affiliation(s)
- Zhenjiang Zheng
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Mengfei Li
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jianchen Yang
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Xintao Zhou
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Yonghua Chen
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Epiphane K Silli
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jiali Tang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Songlin Gong
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Yuan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yihao Zong
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Jianping Kong
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Pu Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Lingxi Yu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Shujun Luo
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu 211198, China
| | - Ying Wang
- School of Life Sciences and Technology, China Pharmaceutical University, Nanjing, Jiangsu 211198, China.
| | - Chunlu Tan
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
4
|
Barati M, Hashemi S, Sayed Tabatabaei M, Zarei Chamgordani N, Mortazavi SM, Moghimi HR. Protein-based microneedles for biomedical applications: A systematic review. Biomed Microdevices 2024; 26:19. [PMID: 38430398 DOI: 10.1007/s10544-024-00701-6] [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] [Accepted: 02/06/2024] [Indexed: 03/03/2024]
Abstract
Microneedles are minimally-invasive devices with the unique capability of bypassing physiological barriers. Hence, they are widely used for different applications from drug/vaccine delivery to diagnosis and cosmetic fields. Recently, natural biopolymers (particularly carbohydrates and proteins) have garnered attention as safe and biocompatible materials with tailorable features for microneedle construction. Several review articles have dealt with carbohydrate-based microneedles. This review aims to highlight the less-noticed role of proteins through a systematic search strategy based on the PRISMA guideline from international databases of PubMed, Science Direct, Scopus, and Google Scholar. Original English articles with the keyword "microneedle(s)" in their titles along with at least one of the keywords "biopolymers, silk, gelatin, collagen, zein, keratin, fish-scale, mussel, and suckerin" were collected and those in which the proteins undertook a structural role were screened. Then, we focused on the structures and applications of protein-based microneedles. Also, the unique features of some protein biopolymers that make them ideal for microneedle construction (e.g., excellent mechanical strength, self-adhesion, and self-assembly), as well as the challenges associated with them were reviewed. Altogether, the proteins identified so far seem not only promising for the fabrication of "better" microneedles in the future but also inspiring for designing biomimetic structural biopolymers with ideal characteristics.
Collapse
Affiliation(s)
- Maedeh Barati
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shiva Hashemi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahsa Sayed Tabatabaei
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasrin Zarei Chamgordani
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Maryam Mortazavi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Moghimi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Loh JM, Lim YJL, Tay JT, Cheng HM, Tey HL, Liang K. Design and fabrication of customizable microneedles enabled by 3D printing for biomedical applications. Bioact Mater 2024; 32:222-241. [PMID: 37869723 PMCID: PMC10589728 DOI: 10.1016/j.bioactmat.2023.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/22/2023] [Accepted: 09/30/2023] [Indexed: 10/24/2023] Open
Abstract
Microneedles (MNs) is an emerging technology that employs needles ranging from 10 to 1000 μm in height, as a minimally invasive technique for various procedures such as therapeutics, disease monitoring and diagnostics. The commonly used method of fabrication, micromolding, has the advantage of scalability, however, micromolding is unable to achieve rapid customizability in dimensions, geometries and architectures, which are the pivotal factors determining the functionality and efficacy of the MNs. 3D printing offers a promising alternative by enabling MN fabrication with high dimensional accuracy required for precise applications, leading to improved performance. Furthermore, enabled by its customizability and one-step process, there is propitious potential for growth for 3D-printed MNs especially in the field of personalized and on-demand medical devices. This review provides an overview of considerations for the key parameters in designing MNs, an introduction on the various 3D-printing techniques for fabricating this new generation of MNs, as well as highlighting the advancements in biomedical applications facilitated by 3D-printed MNs. Lastly, we offer some insights into the future prospects of 3D-printed MNs, specifically its progress towards translation and entry into market.
Collapse
Affiliation(s)
- Jia Min Loh
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Yun Jie Larissa Lim
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Jin Ting Tay
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore
| | | | - Hong Liang Tey
- National Skin Centre (NSC), Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Yong Loo Ling School of Medicine, National University of Singapore, Singapore
- Skin Research Institute of Singapore, Singapore
| | - Kun Liang
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore
- Skin Research Institute of Singapore, Singapore
| |
Collapse
|
6
|
Wang N, Wang T. Innovative translational platforms for rapid developing clinical vaccines against COVID-19 and other infectious disease. Biotechnol J 2024; 19:e2300658. [PMID: 38403469 DOI: 10.1002/biot.202300658] [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: 11/23/2023] [Revised: 12/18/2023] [Accepted: 12/28/2023] [Indexed: 02/27/2024]
Abstract
A vaccine is a biological preparation that contains the antigen capable of stimulating the immune system to form the defense against pathogens. Vaccine development often confronts big challenges, including time/energy-consuming, low efficacy, lag to pathogen emergence and mutation, and even safety concern. However, these seem now mostly conquerable through constructing the advanced translational platforms that can make innovative vaccines, sometimes, potentiated with a distinct multifunctional VADS (vaccine adjuvant delivery system), as evidenced by the development of various vaccines against the covid-19 pandemic at warp speed. Particularly, several covid-19 vaccines, such as the viral-vectored vaccines, mRNA vaccines and DNA vaccines, regarded as the innovative ones that are rapidly made via the high technology-based translational platforms. These products have manifested powerful efficacy while showing no unacceptable safety profile in clinics, allowing them to be approved for massive vaccination at also warp speed. Now, the proprietary translational platforms integrated with the state-of-the-art biotechnologies, and even the artificial intelligence (AI), represent an efficient mode for rapid making innovative clinical vaccines against infections, thus increasingly attracting interests of vaccine research and development. Herein, the advanced translational platforms for making innovative vaccines, together with their design principles and immunostimulatory efficacies, are comprehensively elaborated.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Biological engineering, Hefei University of Technology, Hefei, Anhui Province, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
7
|
Zhi Chen B, Ting He Y, Qiang Zhao Z, Hao Feng Y, Liang L, Peng J, Yu Yang C, Uyama H, Shahbazi MA, Dong Guo X. Strategies to develop polymeric microneedles for controlled drug release. Adv Drug Deliv Rev 2023; 203:115109. [PMID: 39492421 DOI: 10.1016/j.addr.2023.115109] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 11/05/2024]
Abstract
The remarkable appeal of microneedle controlled-release systems has captivated both the academic community and pharmaceutical industry due to their great potential for achieving spatiotemporally controlled release, coupled with their the minimally invasive nature and ease of application. Over the years, scientists have dedicated their efforts to advancing microneedle systems by manipulating the physicochemical properties of matrix materials, refining microneedle designs, and interfacing with external devices to provide tailored drug release profiles in a spatiotemporally controllable manner. Expanding upon our understanding of drug release mechanisms from polymeric microneedles, which include diffusion, swelling, degradation, triggering, and targeting, there is a growing focus on manipulating the location and rate of drug release through innovative microneedle designs. This burgeoning field of microneedle-based drug delivery systems offers further prospects for precise control over drug release. The design strategies of polymeric microneedle systems for temporally controlled and locally targeted release, as well as the delivery mechanisms by which drugs can be released from a microneedle system are critically reviewed in this work. Furthermore, this review also puts forward some perspectives on the potential and challenges involved in translating these microneedle-based delivery systems into the next generation therapies.
Collapse
Affiliation(s)
- Bo Zhi Chen
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China; Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, Japan
| | - Yu Ting He
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ze Qiang Zhao
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yun Hao Feng
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ling Liang
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Juan Peng
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Chen Yu Yang
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Hiroshi Uyama
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, Japan.
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands; Department of Pharmaceutics, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran.
| | - Xin Dong Guo
- State Key Laboratory of Organic-Inorganic Composites, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
8
|
Mbituyimana B, Adhikari M, Qi F, Shi Z, Fu L, Yang G. Microneedle-based cell delivery and cell sampling for biomedical applications. J Control Release 2023; 362:692-714. [PMID: 37689252 DOI: 10.1016/j.jconrel.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/16/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023]
Abstract
Cell-based therapeutics are novel therapeutic strategies that can potentially treat many presently incurable diseases through novel mechanisms of action. Cell therapies may benefit from the ease, safety, and efficacy of administering therapeutic cells. Despite considerable recent technological and biological advances, several barriers remain to the clinical translation and commercialization of cell-based therapies, including low patient compliance, personal handling inconvenience, poor biosafety, and limited biocompatibility. Microneedles (MNs) are emerging as a promising biomedical device option for improved cell delivery with little invasion, pain-free administration, and simplicity of disposal. MNs have shown considerable promise in treating a wide range of diseases and present the potential to improve cell-based therapies. In this review, we first summarized the latest advances in the various types of MNs developed for cell delivery and cell sampling. Emphasis was given to the design and fabrication of various types of MNs based on their structures and materials. Then we focus on the recent biomedical applications status of MNs-mediated cell delivery and sampling, including tissue repair (wound healing, heart repair, and endothelial repair), cancer treatment, diabetes therapy, cell sampling, and other applications. Finally, the current status of clinical application, potential perspectives, and the challenges for clinical translation are also highlighted.
Collapse
Affiliation(s)
- Bricard Mbituyimana
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Manjila Adhikari
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Fuyu Qi
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zhijun Shi
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Lina Fu
- College of Medicine, Huanghuai University, Zhumadian, Henan 463000, China; Zhumadian Central Hospital, Zhumadian, Henan 463000, China.
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
9
|
Freire Haddad H, Roe EF, Collier JH. Expanding opportunities to engineer mucosal vaccination with biomaterials. Biomater Sci 2023; 11:1625-1647. [PMID: 36723064 DOI: 10.1039/d2bm01694j] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mucosal vaccines are receiving increasing interest both for protecting against infectious diseases and for inducing therapeutic immune responses to treat non-infectious diseases. However, the mucosal barriers of the lungs, gastrointestinal tract, genitourinary tract, nasal, and oral tissues each present unique challenges for constructing efficacious vaccines. Vaccination through each of these mucosae requires transport through the mucus and across specialized epithelia to reach tissue-specific immune cells and lymphoid structures, necessitating finely tuned and multifunctional strategies. Serving as inspiration for mucosal vaccine design, pathogens have evolved elaborate, diverse, and multipronged approaches to penetrate and infect mucosae. This review is focused on biomaterials-based strategies, many inspired by pathogens, for designing mucosal vaccine platforms. Passive and active technologies are discussed, along with the microbial processes that they seek to mimic.
Collapse
Affiliation(s)
- Helena Freire Haddad
- Theodore Kennedy Professor of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| | - Emily F Roe
- Theodore Kennedy Professor of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| | - Joel H Collier
- Theodore Kennedy Professor of Biomedical Engineering, Duke University, 101 Science Drive, Durham, NC 27708, USA.
| |
Collapse
|
10
|
Castañeda-Montes MA, Cuevas-Romero JS, Cerriteño-Sánchez JL, de María Ávila-De la Vega L, García-Cambrón JB, Ramírez-Álvarez H. Small ruminant lentivirus capsid protein (SRLV-p25) antigenic structural prediction and immunogenicity to recombinant SRLV-rp25-coupled to immunostimulatory complexes based on glycyrrhizinic acid. Biosci Biotechnol Biochem 2023; 87:267-278. [PMID: 36535645 DOI: 10.1093/bbb/zbac206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
Small ruminant lentiviruses (SRLV) infect sheep and goats resulting in significant economic losses. This study evaluated for the first time the predicted conformational structure of the SRLV-capsid-protein 25 (SRLV-p25) and analyzed the antigenicity of recombinant protein (SRLV-rp25) in mice by coupling to an immunostimulatory complexes based on glycyrrhizinic acid liposomes (GAL) and tested plasma from goats and sheep naturally infected. Analysis in silico and conformational structure of SRLV-p25 (genotype B-FESC-752) showed similar characteristics to other lentiviral capsids. The efficient expression of SRLV-rp25 was confirmed by Western blot. The humoral immune responses in mice showed an increased level of antibodies from day 21 to 35 of the SRLV-rp25-GAL and SRLV-rp25-ISCOM® groups and the cellular immune response showed no significant difference in IL-10 levels (P >.05), however, a significant difference (P <.001) was observed when comparing SRLV-rp25-GAL with SRLV-rp25 groups. Immunoreactivity toward SRLV-rp25 revealed 61% of positive samples from naturally infected goats and sheep.
Collapse
Affiliation(s)
- María Azucena Castañeda-Montes
- Virology, Genetics, and Molecular Biology Laboratory. Faculty of Higher Education, Cuautitlán, Veterinary Medicine, Campus 4. National Autonomous University of Mexico. Km. 2.5 ctra. Cuautitlán-Teoloyucan, San Sebastián Xhala. Cuautitlán Izcalli Estado de México, México
| | - Julieta Sandra Cuevas-Romero
- Laboratorio de Virología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad (CENID-SAI), INIFAP, KM. 15.5 Carretera México-Toluca, Col. Palo Alto, Cuajimalpa, Ciudad de México, México
| | - José Luis Cerriteño-Sánchez
- Laboratorio de Virología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad (CENID-SAI), INIFAP, KM. 15.5 Carretera México-Toluca, Col. Palo Alto, Cuajimalpa, Ciudad de México, México
| | - Lucero de María Ávila-De la Vega
- Virology, Genetics, and Molecular Biology Laboratory. Faculty of Higher Education, Cuautitlán, Veterinary Medicine, Campus 4. National Autonomous University of Mexico. Km. 2.5 ctra. Cuautitlán-Teoloyucan, San Sebastián Xhala. Cuautitlán Izcalli Estado de México, México
| | - José Bryan García-Cambrón
- Laboratorio de Virología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad (CENID-SAI), INIFAP, KM. 15.5 Carretera México-Toluca, Col. Palo Alto, Cuajimalpa, Ciudad de México, México
| | - Hugo Ramírez-Álvarez
- Virology, Genetics, and Molecular Biology Laboratory. Faculty of Higher Education, Cuautitlán, Veterinary Medicine, Campus 4. National Autonomous University of Mexico. Km. 2.5 ctra. Cuautitlán-Teoloyucan, San Sebastián Xhala. Cuautitlán Izcalli Estado de México, México
| |
Collapse
|
11
|
Castañeda-Montes MA, Cuevas-Romero JS, Cerriteño-Sánchez JL, de María Ávila-De la Vega L, García-Cambrón JB, Ramírez-Álvarez H. Small ruminant lentivirus capsid protein (SRLV-p25) antigenic structural prediction and immunogenicity to recombinant SRLV- rp25-coupled to immunostimulatory complexes based on glycyrrhizinic acid. Biosci Biotechnol Biochem 2022. [DOI: https:/doi.org/10.1093/bbb/zbac206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
ABSTRACT
Small ruminant lentiviruses (SRLV) infect sheep and goats resulting in significant economic losses. This study evaluated for the first time the predicted conformational structure of the SRLV-capsid-protein 25 (SRLV-p25) and analyzed the antigenicity of recombinant protein (SRLV-rp25) in mice by coupling to an immunostimulatory complexes based on glycyrrhizinic acid liposomes (GAL) and tested plasma from goats and sheep naturally infected. Analysis in silico and conformational structure of SRLV-p25 (genotype B-FESC-752) showed similar characteristics to other lentiviral capsids. The efficient expression of SRLV-rp25 was confirmed by Western blot. The humoral immune responses in mice showed an increased level of antibodies from day 21 to 35 of the SRLV-rp25-GAL and SRLV-rp25-ISCOM® groups and the cellular immune response showed no significant difference in IL-10 levels (P >.05), however, a significant difference (P <.001) was observed when comparing SRLV-rp25-GAL with SRLV-rp25 groups. Immunoreactivity toward SRLV-rp25 revealed 61% of positive samples from naturally infected goats and sheep.
Collapse
Affiliation(s)
- María Azucena Castañeda-Montes
- Virology, Genetics, and Molecular Biology Laboratory. Faculty of Higher Education, Cuautitlán, Veterinary Medicine , Campus 4. National Autonomous University of Mexico. Km. 2.5 ctra. Cuautitlán-Teoloyucan, San Sebastián Xhala. Cuautitlán Izcalli Estado de México , México
| | - Julieta Sandra Cuevas-Romero
- Laboratorio de Virología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad (CENID-SAI), INIFAP , KM. 15.5 Carretera México-Toluca, Col. Palo Alto, Cuajimalpa, Ciudad de México , México
| | - José Luis Cerriteño-Sánchez
- Laboratorio de Virología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad (CENID-SAI), INIFAP , KM. 15.5 Carretera México-Toluca, Col. Palo Alto, Cuajimalpa, Ciudad de México , México
| | - Lucero de María Ávila-De la Vega
- Virology, Genetics, and Molecular Biology Laboratory. Faculty of Higher Education, Cuautitlán, Veterinary Medicine , Campus 4. National Autonomous University of Mexico. Km. 2.5 ctra. Cuautitlán-Teoloyucan, San Sebastián Xhala. Cuautitlán Izcalli Estado de México , México
| | - José Bryan García-Cambrón
- Laboratorio de Virología, Centro Nacional de Investigación Disciplinaria en Salud Animal e Inocuidad (CENID-SAI), INIFAP , KM. 15.5 Carretera México-Toluca, Col. Palo Alto, Cuajimalpa, Ciudad de México , México
| | - Hugo Ramírez-Álvarez
- Virology, Genetics, and Molecular Biology Laboratory. Faculty of Higher Education, Cuautitlán, Veterinary Medicine , Campus 4. National Autonomous University of Mexico. Km. 2.5 ctra. Cuautitlán-Teoloyucan, San Sebastián Xhala. Cuautitlán Izcalli Estado de México , México
| |
Collapse
|
12
|
Mbituyimana B, Ma G, Shi Z, Yang G. Polymer-based microneedle composites for enhanced non-transdermal drug delivery. APPLIED MATERIALS TODAY 2022; 29:101659. [DOI: 10.1016/j.apmt.2022.101659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
13
|
Ren H, Li H, Cao L, Wang Z, Zhou Y, Guo J, Zhang Y, Liu H, Xu W. Intranasal immunization with HRSV prefusion F protein and CpG adjuvant elicits robust protective effects in mice. Vaccine 2022; 40:6830-6838. [PMID: 36253219 DOI: 10.1016/j.vaccine.2022.09.071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/09/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
Human respiratory syncytial virus (HRSV) is a leading cause of lower respiratory tract infections in elderly individuals and young children/infants and can cause bronchiolitis and even death. There is no licensed HRSV vaccine. An ideal vaccine should induce high titers of neutralizing antibodies and a Th1-biased immune response. In this study, we used EXPI293 cells to express the fusion (F) protein with a prefusion conformation (PrF) and compared the safety and efficacy of intranasal immunization with PrF in combination with two mucosal adjuvants (CpG ODN and liposomes) in mice. After two intranasal administrations, mice in the PrF + CpG group produced high titers of neutralizing antibodies (4961) and a Th1-biased immune response compared with the PrF + Lipo group. The lung viral load of mice in the PrF + CpG group was significantly reduced (3.5 log) compared with that in the adjuvant control group, and the survival rate was 100 %, while the survival rate of mice in the PrF + Lipo group was only 67 %. At the same time, this immunization strategy reduced the pathological damage to the lungs in mice. In conclusion, the combination of PrF and CpG adjuvant is immunogenic, elicits a Th1 type immune response, and completely protects mice from a lethal HRSV challenge. It is worthy of further evaluation as an HRSV vaccine in clinical trials. Clinical trial registration. This study was not related to human participation or experimentation.
Collapse
Affiliation(s)
- Hu Ren
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hai Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lei Cao
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhan Wang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yangzi Zhou
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jinyuan Guo
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Zhang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hongtu Liu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China.
| | - Wenbo Xu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China; Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Cao X, Chen G. Advances in microneedles for non-transdermal applications. Expert Opin Drug Deliv 2022; 19:1081-1097. [DOI: 10.1080/17425247.2022.2118711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Xiaona Cao
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- School of Nursing, Tianjin Medical University, Tianjin, China
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, Montreal, Quebec, Canada
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
15
|
Recent progress in application of nanovaccines for enhancing mucosal immune responses. Acta Pharm Sin B 2022. [DOI: 10.1016/j.apsb.2022.08.010] [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] Open
|
16
|
Mucosal vaccine delivery: A focus on the breakthrough of specific barriers. Acta Pharm Sin B 2022; 12:3456-3474. [PMID: 35818435 PMCID: PMC9259023 DOI: 10.1016/j.apsb.2022.07.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/03/2022] [Accepted: 06/30/2022] [Indexed: 12/30/2022] Open
Abstract
Mucosal vaccines can effectively induce an immune response at the mucosal site and form the first line of defense against microbial invasion. The induced mucosal immunity includes the proliferation of effector T cells and the production of IgG and IgA antibodies, thereby effectively blocking microbial infection and transmission. However, after a long period of development, the transformation of mucosal vaccines into clinical use is still relatively slow. To date, fewer than ten mucosal vaccines have been approved. Only seven mucosal vaccines against coronavirus disease 2019 (COVID-19) are under investigation in clinical trials. A representative vaccine is the adenovirus type-5 vectored COVID-19 vaccine (Ad5-nCoV) developed by Chen and coworkers, which is currently in phase III clinical trials. The reason for the limited progress of mucosal vaccines may be the complicated mucosal barriers. Therefore, this review summarizes the characteristics of mucosal barriers and highlights strategies to overcome these barriers for effective mucosal vaccine delivery.
Collapse
|
17
|
Ali M, Namjoshi S, Benson HAE, Mohammed Y, Kumeria T. Dissolvable polymer microneedles for drug delivery and diagnostics. J Control Release 2022; 347:561-589. [PMID: 35525331 DOI: 10.1016/j.jconrel.2022.04.043] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 10/18/2022]
Abstract
Dissolvable transdermal microneedles (μND) are promising micro-devices used to transport a wide selection of active compounds into the skin. To provide an effective therapeutic outcome, μNDs must pierce the human stratum corneum (~10 to 20 μm), without rupturing or bending during penetration, then release their cargo at the predetermined area and time. The ability of dissolvable μND arrays/patches to sufficiently pierce the skin is a crucial requirement, which depends on the material composition, μND geometry and fabrication techniques. This comprehensive review not only provides contemporary knowledge on the μND design approaches, but also the materials science facilitating these delivery systems and the opportunities these advanced materials can provide to enhance clinical outcomes.
Collapse
Affiliation(s)
- Masood Ali
- Therapeutics Research Group, The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD 4102, Australia
| | - Sarika Namjoshi
- Therapeutics Research Group, The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD 4102, Australia; Vaxxas Pty Ltd, Brisbane, Woolloongabba, QLD 4102, Australia
| | - Heather A E Benson
- Curtin Medical School, Curtin University, Bentley, WA 6102, Australia; UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA 5001, Australia; Basil Hetzel institute for Translational Health Research, Adelaide, SA 5001, Australia.
| | - Yousuf Mohammed
- Therapeutics Research Group, The University of Queensland Diamantina Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD 4102, Australia.
| | - Tushar Kumeria
- School of Materials Science and Engineering, The University of New South Wales, Sydney. NSW 2052, Australia; Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW 2052, Australia; School of Pharmacy, The University of Queensland, Brisbane, QLD 4102, Australia.
| |
Collapse
|
18
|
Abstract
A favorable outcome of the COVID-19 crisis might be achieved with massive vaccination. The proposed vaccines contain several different vaccine active principles (VAP), such as inactivated virus, antigen, mRNA, and DNA, which are associated with either standard adjuvants or nanomaterials (NM) such as liposomes in Moderna's and BioNTech/Pfizer's vaccines. COVID-19 vaccine adjuvants may be chosen among liposomes or other types of NM composed for example of graphene oxide, carbon nanotubes, micelles, exosomes, membrane vesicles, polymers, or metallic NM, taking inspiration from cancer nano-vaccines, whose adjuvants may share some of their properties with those of viral vaccines. The mechanisms of action of nano-adjuvants are based on the facilitation by NM of targeting certain regions of immune interest such as the mucus, lymph nodes, and zones of infection or blood irrigation, the possible modulation of the type of attachment of the VAP to NM, in particular VAP positioning on the NM external surface to favor VAP presentation to antigen presenting cells (APC) or VAP encapsulation within NM to prevent VAP degradation, and the possibility to adjust the nature of the immune response by tuning the physico-chemical properties of NM such as their size, surface charge, or composition. The use of NM as adjuvants or the presence of nano-dimensions in COVID-19 vaccines does not only have the potential to improve the vaccine benefit/risk ratio, but also to reduce the dose of vaccine necessary to reach full efficacy. It could therefore ease the overall spread of COVID-19 vaccines within a sufficiently large portion of the world population to exit the current crisis.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France. .,Nanobacterie SARL, 36 Boulevard Flandrin, 75116, Paris, France.,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| |
Collapse
|
19
|
Panda A, Matadh VA, Suresh S, Shivakumar HN, Murthy SN. Non-dermal applications of microneedle drug delivery systems. Drug Deliv Transl Res 2022; 12:67-78. [PMID: 33629222 DOI: 10.1007/s13346-021-00922-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2021] [Indexed: 12/12/2022]
Abstract
Microneedles (MNs) are micron-scaled needles measuring 100 to 1000 μm that were initially explored for delivery of therapeutic agents across the skin. Considering the success in transcutaneous drug delivery, the application of microneedles has been extended to different tissues and organs. The review captures the application of microneedles to the oral mucosa, the eye, vagina, gastric mucosa, nail, scalp, and vascular tissues for delivery of vaccines, biologics, drugs, and diagnostic agents. The technology has created easy access to the poorly accessible segments of eye to facilitate delivery of monoclonal antibodies and therapeutic agents in management of neovascular disease. Microporation has been reported to drastically improve the drug delivery through the poorly permeable nail plate. Curved microneedles and spatially designed microneedle cuffs have been found to be capable of delivering stem cells and therapeutic macromolecules directly to the cardiac tissue and the vascular smooth muscle cells, respectively. Besides being minimally invasive and patient compliant, the technology has the potential to offer viable solutions to deliver drugs through impermeable barriers owing to the ability to penetrate several biological barriers. The technology has been successful to overcome the delivery hurdles and enable direct delivery of drug to the target sites, thus maximizing the efficacy thereby reducing the required dose. This review is an attempt to capture the non-dermatological applications of microneedles being explored and provides an insight on the future trends in the field of microneedle technology. Pictorial representation of different microneedle application.
Collapse
Affiliation(s)
- Apoorva Panda
- The University of Mississippi School of Pharmacy, Oxford, MS, USA
| | - V Anusha Matadh
- Institute for Drug Delivery and Biomedical Research, Bengaluru, India
| | - Sarasija Suresh
- Institute for Drug Delivery and Biomedical Research, Bengaluru, India
| | - H N Shivakumar
- Institute for Drug Delivery and Biomedical Research, Bengaluru, India
- Department of Pharmaceutics, K.L.E. College of Pharmacy, Bengaluru, India
| | - S Narasimha Murthy
- The University of Mississippi School of Pharmacy, Oxford, MS, USA.
- Institute for Drug Delivery and Biomedical Research, Bengaluru, India.
| |
Collapse
|
20
|
VanBenschoten HM, Woodrow KA. Vaginal delivery of vaccines. Adv Drug Deliv Rev 2021; 178:113956. [PMID: 34481031 PMCID: PMC8722700 DOI: 10.1016/j.addr.2021.113956] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/06/2021] [Accepted: 08/28/2021] [Indexed: 11/22/2022]
Abstract
Recent estimates suggest that one in two sexually active individuals will acquire a sexually transmitted infection by age 25, an alarming statistic that amounts to over 1 million new infections per day worldwide. Vaccination against STIs is highly desirable for alleviating this global burden of disease. Vaginal immunization is a promising strategy to combat transmission via the vaginal mucosa. The vagina is typically considered a poor inductive site for common correlates of adaptive immunity. However, emerging evidence suggests that immune tolerance may be overcome by precisely engineered vaccination schemes that orchestrate cell-mediated immunity and establish tissue resident memory immune cells. In this review, we will discuss the unique immunological milieu of the vaginal mucosa and our current understanding of correlates of pathogenesis and protection for several common STIs. We then present a summary of recent vaginal vaccine studies and explore the role that mucosal adjuvants and delivery systems play in enhancing protection according to requisite features of immunity. Finally, we offer perspectives on the challenges and future directions of vaginal vaccine delivery, discussing remaining physiological barriers and innovative vaccine formulations that may overcome them.
Collapse
Affiliation(s)
- Hannah M VanBenschoten
- 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
|
21
|
Wang J, Zhang Y, Pi J, Xing D, Wang C. Localized delivery of immunotherapeutics: A rising trend in the field. J Control Release 2021; 340:149-167. [PMID: 34699871 DOI: 10.1016/j.jconrel.2021.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/11/2021] [Indexed: 02/08/2023]
Abstract
Immunotherapy is becoming a new standard of care for multiple cancers, while several limitations are impending its further clinical success. Immunotherapeutic agents often have inappropriate pharmacokinetics on their own and/or exhibit limited specificity to tumor cells, leading to severe immuno-related adverse effects and limited efficacy. Suitable formulating strategies that confer prolonged contact with or efficient proliferation in tumors while reducing exposure to normal tissues are highly worthy to explore. With the assistance of biomaterial carriers, targeted therapy can be achieved artificially by implanting or injecting drug depots into desired sites, about which the wisdoms in literature have been rich. The relevant results have suggested a "local but systemic" effect, that is, local replenishment of immune modulators achieves a high treatment efficacy that also governs distant metastases, thereby building another rationale for localized delivery. Particularly, implantable scaffolds have been further engineered to recruit disseminated tumor cells with an efficiency high enough to reduce tumor burdens at typical metastatic organs, and simultaneously provide diagnostic signals. This review introduces recent advances in this emerging area along with a perspective on the opportunities and challenges in the way to clinical application.
Collapse
Affiliation(s)
- Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Yukun Zhang
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Jiuchan Pi
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Chao Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
22
|
Christodoulides M, Humbert MV, Heckels JE. The potential utility of liposomes for Neisseria vaccines. Expert Rev Vaccines 2021; 20:1235-1256. [PMID: 34524062 DOI: 10.1080/14760584.2021.1981865] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Species of the genus Neisseria are important global pathogens. Neisseria gonorrhoeae (gonococcus) causes the sexually transmitted disease gonorrhea and Neisseria meningitidis (meningococcus) causes meningitis and sepsis. Liposomes are self-assembled spheres of phospholipid bilayers enclosing a central aqueous space, and they have attracted much interest and use as a delivery vehicle for Neisseria vaccine antigens. AREAS COVERED A brief background on Neisseria infections and the success of licensed meningococcal vaccines are provided. The absence of a gonococcal vaccine is highlighted. The use of liposomes for delivering Neisseria antigens and adjuvants, for the purposes of generating specific immune responses, is reviewed. The use of other lipid-based systems for antigen and adjuvant delivery is examined briefly. EXPERT OPINION With renewed interest in developing a gonococcal vaccine, liposomes remain an attractive option for delivering antigens. The discipline of nanotechnology provides additional nanoparticle-based options for gonococcal vaccine development. Future work would be needed to tailor the composition of liposomes and other nanoparticles to the specific vaccine antigen(s), in order to generate optimal anti-gonococcal immune responses. The potential use of liposomes and other nanoparticles to deliver anti-gonococcal compounds to treat infections also should be explored further.
Collapse
Affiliation(s)
- Myron Christodoulides
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - Maria Victoria Humbert
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| | - John E Heckels
- Neisseria Research Group, Molecular Microbiology, School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, UK
| |
Collapse
|
23
|
Bilal M, Mehmood S, Raza A, Hayat U, Rasheed T, Iqbal HM. Microneedles in Smart Drug Delivery. Adv Wound Care (New Rochelle) 2021; 10:204-219. [PMID: 32320365 PMCID: PMC7906867 DOI: 10.1089/wound.2019.1122] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Significance: In biomedical setup, at large, and drug delivery, in particular, transdermal patches, hypodermal needles, and/or dermatological creams with the topical appliance are among the most widely practiced routes for transdermal drug delivery. Owing to the stratum corneum layer of the skin, traditional drug delivery methods are inefficient, and the effect of the administered therapeutic cues is limited. Recent Advances: The current advancement at the microlevel and nanolevel has revolutionized the drug delivery sector. Particularly, various types of microneedles (MNs) are becoming popular for drug delivery applications because of safety, patient compliance, and smart action. Critical Issues: Herein, we reviewed state-of-the-art MNs as a smart and sophisticated drug delivery approach. Following a brief introduction, the drug delivery mechanism of MNs is discussed. Different types of MNs, that is, solid, hollow, coated, dissolving, and hydrogel forming, are discussed with suitable examples. The latter half of the work is focused on the applied perspective and clinical translation of MNs. Furthermore, a detailed overview of clinical applications and future perspectives is also included in this review. Future Directions: Regardless of ongoing technological and clinical advancement, the focus should be diverted to enhance the efficacy and strength of MNs. Besides, the possible immune response or interference should also be avoided for successful clinical translation of MNs as an efficient drug delivery system.
Collapse
Affiliation(s)
- Muhammad Bilal
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Correspondence: Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam.
| | - Shahid Mehmood
- Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ali Raza
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Uzma Hayat
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tahir Rasheed
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hafiz M.N. Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, Monterrey, Mexico
| |
Collapse
|
24
|
O’Shea J, Prausnitz MR, Rouphael N. Dissolvable Microneedle Patches to Enable Increased Access to Vaccines against SARS-CoV-2 and Future Pandemic Outbreaks. Vaccines (Basel) 2021; 9:320. [PMID: 33915696 PMCID: PMC8066809 DOI: 10.3390/vaccines9040320] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/25/2021] [Accepted: 03/30/2021] [Indexed: 01/02/2023] Open
Abstract
Vaccines are an essential component of pandemic preparedness but can be limited due to challenges in production and logistical implementation. While vaccine candidates were rapidly developed against severe acute respiratory syndrome coronavirus 2 (SARS-COV-2), immunization campaigns remain an obstacle to achieving herd immunity. Dissolvable microneedle patches are advantageous for many possible reasons: improved immunogenicity; dose-sparing effects; expected low manufacturing cost; elimination of sharps; reduction of vaccine wastage; no need for reconstitution; simplified supply chain, with reduction of cold chain supply through increased thermostability; ease of use, reducing the need for healthcare providers; and greater acceptability compared to traditional hypodermic injections. When applied to coronavirus disease 2019 (COVID-19) and future pandemic outbreaks, microneedle patches have great potential to improve vaccination globally and save many lives.
Collapse
Affiliation(s)
- Jesse O’Shea
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, Atlanta, GA 30030, USA;
| | - Mark R. Prausnitz
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, 500 Irvin Court, Suite 200, Decatur, Atlanta, GA 30030, USA;
| |
Collapse
|
25
|
Aksit A, Rastogi S, Nadal ML, Parker AM, Lalwani AK, West AC, Kysar JW. Drug delivery device for the inner ear: ultra-sharp fully metallic microneedles. Drug Deliv Transl Res 2021; 11:214-226. [PMID: 32488817 PMCID: PMC8649787 DOI: 10.1007/s13346-020-00782-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Drug delivery into the inner ear is a significant challenge due to its inaccessibility as a fluid-filled cavity within the temporal bone of the skull. The round window membrane (RWM) is the only delivery portal from the middle ear to the inner ear that does not require perforation of bone. Recent advances in microneedle fabrication enable the RWM to be perforated safely with polymeric microneedles as a means to enhance the rate of drug delivery from the middle ear to the inner ear. However, the polymeric material is not biocompatible and also lacks the strength of other materials. Herein we describe the design and development of gold-coated metallic microneedles suitable for RWM perforation. When developing microneedle technology for drug delivery, we considered three important general attributes: (1) high strength and ductility material, (2) high accuracy and precision of fabrication, and (3) broad design freedom. We developed a hybrid additive manufacturing method using two-photon lithography and electrochemical deposition to fabricate ultra-sharp gold-coated copper microneedles with these attributes. We refer to the microneedle fabrication methodology as two-photon templated electrodeposition (2PTE). We demonstrate the use of these microneedles by inducing a perforation with a minimal degree of trauma in a guinea pig RWM while the microneedle itself remains undamaged. Thus, this microneedle has the potential literally of opening the RWM for enhanced drug delivery into the inner ear. Finally, the 2PTE methodology can be applied to many different classes of microneedles for other drug delivery purposes as well the fabrication of small scale structures and devices for non-medical applications. Graphical Abstract Fully metallic ultra-sharp microneedle mounted at end of a 24-gauge stainless steel blunt syringe needle tip: (left) Size of microneedle shown relative to date stamp on U.S. one-cent coin; (right) Perforation through guinea pig round window membrane introduced with microneedle.
Collapse
Affiliation(s)
- Aykut Aksit
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY, 10027, USA
| | - Shruti Rastogi
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY, 10027, USA
| | - Maria L Nadal
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY, 10027, USA
| | - Amber M Parker
- Department of Otolaryngology - Head & Neck Surgery, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Anil K Lalwani
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY, 10027, USA
- Department of Otolaryngology - Head & Neck Surgery, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Alan C West
- Department of Chemical Engineering, Columbia University, 500 W. 120th St., New York, NY, 10027, USA
| | - Jeffrey W Kysar
- Department of Mechanical Engineering, Columbia University, 500 West 120th Street, New York, NY, 10027, USA.
- Department of Otolaryngology - Head & Neck Surgery, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA.
| |
Collapse
|
26
|
Aluminum Nanoparticles Acting as a Pulmonary Vaccine Adjuvant-Delivery System (VADS) Able to Safely Elicit Robust Systemic and Mucosal Immunity. J Inorg Organomet Polym Mater 2020; 30:4203-4217. [PMID: 32395098 PMCID: PMC7210793 DOI: 10.1007/s10904-020-01572-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 04/29/2020] [Indexed: 12/17/2022]
Abstract
Abstract Vulnerability of respiratory mucosa to invasions of airborne pathogens, such as SARS-CoV, MERS-CoV and avian viruses which sometimes cause a life-threatening epidemic and even pandemic, underscores significance of developing a pulmonary vaccine adjuvant-delivery system (VADS). Herein, 30-nm aluminum nanoparticles (ANs), unlike the mostly used adjuvant alum which is unsuitable for delivering pulmonary vaccines due to side effects, proved able to act as a VADS fitting inhalation immunization to elicit wide-spread anti-antigen immunity. In vitro ANs facilitated cellular uptake of their cargos and, after pulmonary vaccination, induced mouse production of high levels of anti-antigen IgG in serum and IgA in saliva, nasal, bronchoalveolar and also vaginal fluids. Besides, IFN-γ and anti-antigen IgG2a enriched in immunized mice which meanwhile showed no obvious lung inflammation indicated balanced Th1/Th2 responses were safely induced. These outcomes suggest ANs may be an efficient pulmonary VADS for defending against pathogens, especially, the ones invading hosts via respiratory system. Graphic Abstract
Aluminum nanoparticles can safely induce humoral and cellular immunity at systemic and mucosal level through pulmonary vaccination to contrast the conventional adjuvant alum.![]()
Collapse
|
27
|
Zhang X, Wang Y, Chi J, Zhao Y. Smart Microneedles for Therapy and Diagnosis. RESEARCH (WASHINGTON, D.C.) 2020; 2020:7462915. [PMID: 33623910 PMCID: PMC7877383 DOI: 10.34133/2020/7462915] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/26/2020] [Indexed: 01/26/2023]
Abstract
Microneedles represent a cutting-edge and idea-inspiring technology in biomedical engineering, which have attracted increasing attention of scientific researchers and medical staffs. Over the past decades, numerous great achievements have been made. The fabrication process of microneedles has been simplified and becomes more precise, easy-to-operate, and reusable. Besides, microneedles with various features have been developed and the microneedle materials have greatly expanded. In recent years, efforts have been focused on generating smart microneedles by endowing them with intriguing functions such as adhesion ability, responsiveness, and controllable drug release. Such improvements enable the microneedles to take an important step in practical applications including household drug delivery devices, wearable biosensors, biomedical assays, cell culture, and microfluidic chip analysis. In this review, the fabrication strategies, distinctive properties, and typical applications of the smart microneedles are discussed. Recent accomplishments, remaining challenges, and future prospects are also presented.
Collapse
Affiliation(s)
- Xiaoxuan Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yuetong Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Junjie Chi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Yuanjin Zhao
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
28
|
Lee K, Goudie MJ, Tebon P, Sun W, Luo Z, Lee J, Zhang S, Fetah K, Kim HJ, Xue Y, Darabi MA, Ahadian S, Sarikhani E, Ryu W, Gu Z, Weiss PS, Dokmeci MR, Ashammakhi N, Khademhosseini A. Non-transdermal microneedles for advanced drug delivery. Adv Drug Deliv Rev 2019; 165-166:41-59. [PMID: 31837356 PMCID: PMC7295684 DOI: 10.1016/j.addr.2019.11.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
Abstract
Microneedles (MNs) have been used to deliver drugs for over two decades. These platforms have been proven to increase transdermal drug delivery efficiency dramatically by penetrating restrictive tissue barriers in a minimally invasive manner. While much of the early development of MNs focused on transdermal drug delivery, this technology can be applied to a variety of other non-transdermal biomedical applications. Several variations, such as multi-layer or hollow MNs, have been developed to cater to the needs of specific applications. The heterogeneity in the design of MNs has demanded similar variety in their fabrication methods; the most common methods include micromolding and drawing lithography. Numerous materials have been explored for MN fabrication which range from biocompatible ceramics and metals to natural and synthetic biodegradable polymers. Recent advances in MN engineering have diversified MNs to include unique shapes, materials, and mechanical properties that can be tailored for organ-specific applications. In this review, we discuss the design and creation of modern MNs that aim to surpass the biological barriers of non-transdermal drug delivery in ocular, vascular, oral, and mucosal tissue.
Collapse
Affiliation(s)
- KangJu Lee
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marcus J Goudie
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Peyton Tebon
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wujin Sun
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zhimin Luo
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Junmin Lee
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Shiming Zhang
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kirsten Fetah
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Han-Jun Kim
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yumeng Xue
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mohammad Ali Darabi
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samad Ahadian
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Einollah Sarikhani
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - WonHyoung Ryu
- Department of Mechanical Engineering, Yonsei University, Seoul 03722, South Korea
| | - Zhen Gu
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90024, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC 27695, USA
| | - Paul S Weiss
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mehmet R Dokmeci
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Radiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nureddin Ashammakhi
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Radiology, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Ali Khademhosseini
- Department of Bioengineering and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA 90024, USA; Department of Radiology, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
29
|
Pires LR, Vinayakumar KB, Turos M, Miguel V, Gaspar J. A Perspective on Microneedle-Based Drug Delivery and Diagnostics in Paediatrics. J Pers Med 2019; 9:jpm9040049. [PMID: 31731656 PMCID: PMC6963643 DOI: 10.3390/jpm9040049] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/04/2019] [Accepted: 11/12/2019] [Indexed: 12/24/2022] Open
Abstract
Microneedles (MNs) have been extensively explored in the literature as a means to deliver drugs in the skin, surpassing the stratum corneum permeability barrier. MNs are potentially easy to produce and may allow the self-administration of drugs without causing pain or bleeding. More recently, MNs have been investigated to collect/assess the interstitial fluid in order to monitor or detect specific biomarkers. The integration of these two concepts in closed-loop devices holds the promise of automated and minimally invasive disease detection/monitoring and therapy. These assure low invasiveness and, importantly, open a window of opportunity for the application of population-specific and personalised therapies.
Collapse
Affiliation(s)
- Liliana R Pires
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal; (L.R.P.); (J.G.)
| | - KB Vinayakumar
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal; (L.R.P.); (J.G.)
- Correspondence: or
| | - Maria Turos
- University of Oviedo, 33006 Asturias, Spain;
| | - Verónica Miguel
- Department of Cell Biology and Immunology, Centro de Biología Molecular “Severo Ochoa”, 28049 Madrid, Spain;
| | - João Gaspar
- International Iberian Nanotechnology Laboratory, 4715-330 Braga, Portugal; (L.R.P.); (J.G.)
| |
Collapse
|
30
|
Mandal A, Boopathy AV, Lam LKW, Moynihan KD, Welch ME, Bennett NR, Turvey ME, Thai N, Van JH, Love JC, Hammond PT, Irvine DJ. Cell and fluid sampling microneedle patches for monitoring skin-resident immunity. Sci Transl Med 2019; 10:10/467/eaar2227. [PMID: 30429353 DOI: 10.1126/scitranslmed.aar2227] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 04/24/2018] [Accepted: 10/15/2018] [Indexed: 12/18/2022]
Abstract
Important cell populations reside within tissues and are not accessed by traditional blood draws used to monitor the immune system. To address this issue at an essential barrier tissue, the skin, we created a microneedle-based technology for longitudinal sampling of cells and interstitial fluid, enabling minimally invasive parallel monitoring of immune responses. Solid microneedle projections were coated by a cross-linked biocompatible polymer, which swells upon skin insertion, forming a porous matrix for local leukocyte infiltration. By embedding molecular adjuvants and specific antigens encapsulated in nanocapsules within the hydrogel coating, antigen-specific lymphocytes can be enriched in the recovered cell population, allowing for subsequent detailed phenotypic and functional analysis. We demonstrate this approach in mice immunized with a model protein antigen or infected in the skin with vaccinia virus. After vaccination or infection, sampling microneedles allowed tissue-resident memory T cells (TRMs) to be longitudinally monitored in the skin for many months, during which time the antigen-specific T cell population in systemic circulation contracted to low or undetectable counts. Sampling microneedles did not change the immune status of naïve or antigen-exposed animals. We also validated the ability of cell sampling using human skin samples. This approach may be useful in vaccines and immunotherapies to temporally query TRM populations or as a diagnostic platform to sample for biomarkers in chronic inflammatory and autoimmune disorders, allowing information previously accessible only via invasive biopsies to be obtained in a minimally invasive manner from the skin or other mucosal tissues.
Collapse
Affiliation(s)
- Anasuya Mandal
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA.,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Institute for Soldier Nanotechnologies, MIT, Cambridge, MA 02139, USA
| | - Archana V Boopathy
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Lionel K W Lam
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA.,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Kelly D Moynihan
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - Mary E Welch
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Nitasha R Bennett
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Michelle E Turvey
- Infectious Diseases IRG, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Nikki Thai
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA
| | - Jenny H Van
- Department of Biological Engineering, MIT, Cambridge, MA 02139, USA
| | - J Christopher Love
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA.,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Institute for Soldier Nanotechnologies, MIT, Cambridge, MA 02139, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Paula T Hammond
- Department of Chemical Engineering, MIT, Cambridge, MA 02139, USA. .,Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA.,Institute for Soldier Nanotechnologies, MIT, Cambridge, MA 02139, USA.,Infectious Diseases IRG, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA 02139, USA. .,Institute for Soldier Nanotechnologies, MIT, Cambridge, MA 02139, USA.,Department of Biological Engineering, MIT, Cambridge, MA 02139, USA.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA.,Department of Materials Science and Engineering, MIT, Cambridge, MA 02139, USA.,Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| |
Collapse
|
31
|
Rodgers AM, Cordeiro AS, Donnelly RF. Technology update: dissolvable microneedle patches for vaccine delivery. MEDICAL DEVICES-EVIDENCE AND RESEARCH 2019; 12:379-398. [PMID: 31572025 PMCID: PMC6756839 DOI: 10.2147/mder.s198220] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/08/2019] [Indexed: 12/17/2022] Open
Abstract
Despite vaccination representing one of the greatest advances of modern preventative medicine, there remain significant challenges in vaccine distribution, delivery and compliance. Dissolvable microarray patches or dissolving microneedles (DMN) have been proposed as an innovative vaccine delivery platform that could potentially revolutionize vaccine delivery and circumvent many of the challenges faced with current vaccine strategies. DMN, due to their ease of use, lack of elicitation of pain response, self-disabling nature and ease of transport and distribution, offer an attractive delivery option for vaccines. Additionally, as DMN inherently targets the uppermost skin layers, they facilitate improved vaccine efficacy, due to direct targeting of skin antigen-presenting cells. A plethora of publications have demonstrated the efficacy of DMN vaccination for a range of vaccines, with influenza receiving particular attention. However, before the viable adoption of DMN for vaccination purposes in a clinical setting, a number of fundamental questions must be addressed. Accordingly, this review begins by introducing some of the key barriers faced by current vaccination approaches and how DMN can overcome these challenges. We introduce some of the recent advances in the field of DMN technology, highlighting the potential impact DMN could have, particularly in countries of the developing world. We conclude by reflecting on some of the key questions that remain unanswered and which warrant further investigation before DMNs can be utilized in clinical settings.
Collapse
Affiliation(s)
- Aoife M Rodgers
- School of Pharmacy, Queen’s University Belfast, Belfast, BT9 7BL, UK
| | - Ana Sara Cordeiro
- School of Pharmacy, Queen’s University Belfast, Belfast, BT9 7BL, UK
| | - Ryan F Donnelly
- School of Pharmacy, Queen’s University Belfast, Belfast, BT9 7BL, UK
| |
Collapse
|
32
|
Covering Aluminum Oxide Nanoparticles with Biocompatible Materials to Efficiently Deliver Subunit Vaccines. Vaccines (Basel) 2019; 7:vaccines7020052. [PMID: 31212955 PMCID: PMC6631575 DOI: 10.3390/vaccines7020052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 06/05/2019] [Accepted: 06/07/2019] [Indexed: 12/12/2022] Open
Abstract
Subunit vaccines have advantages of good safety, minimal reactogenicity, and high specificity. However, subunit vaccines also show a crucial disadvantage of poor immunogenicity and, therefore, are often formulated with an adjuvant carrier to form a vaccine adjuvant-delivery system (VADS) to enhance their efficacies. Alums, the coarse aggregates of the insoluble aluminum salts, are the conventional adjuvants and have been widely used in clinical vaccines for a long time. Unfortunately, alums also show two main drawbacks of low potency in eliciting cellular immunity, and high reactogenicity to cause unwanted inflammations. Therefore, herein the phospholipid bilayer-coated aluminum oxide nanoparticles (PLANs) and the PEGylated PLANs (PEG-PLANs) were engineered as a VADS to overcome the drawbacks of both subunit vaccines and coarse alums, while synergizing their functions. In vitro experiments demonstrated that, unlike the micron-sized alums, the nanosized PLANs and PEG-PLANs loaded with model antigen of ovalbumin (OVA) showed a high safety profile and were able to promote APC (antigen-presenting cell) uptake and engender lysosome escape for enhancing the MHC (major histocompatibility complex)-I-antigen display. Subcutaneously administered to mice, PLANs and, especially, PEG-PLANs smoothly trafficked into the draining lymph nodes, wherein the densely clustered immune cells were activated in substantial numbers, leading to robust immunoresponses and efficient production of the anti-antigen antibodies and CD8+ T cells. Thus, the aluminum-based nanocarriers, especially the PEG-PLANs, are a promising VADS possessing the potential of eliciting strong and comprehensive immunity against pathogens.
Collapse
|
33
|
Wang N, Chen M, Wang T. Liposomes used as a vaccine adjuvant-delivery system: From basics to clinical immunization. J Control Release 2019; 303:130-150. [PMID: 31022431 PMCID: PMC7111479 DOI: 10.1016/j.jconrel.2019.04.025] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2019] [Revised: 04/17/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022]
Abstract
Liposomes are widely utilized as a carrier to improve therapeutic efficacy of agents thanks to their merits of high loading capacity, targeting delivery, reliable protection of agents, good biocompatibility, versatile structure modification and adjustable characteristics, such as size, surface charge, membrane flexibility and the agent loading mode. In particular, in recent years, through modification with immunopotentiators and targeting molecules, and in combination with innovative immunization devices, liposomes are rapidly developed as a multifunctional vaccine adjuvant-delivery system (VADS) that has a high capability in inducing desired immunoresponses, as they can target immune cells and even cellular organelles, engender lysosome escape, and promote Ag cross-presentation, thus enormously enhancing vaccination efficacy. Moreover, after decades of development, several products developed on liposome VADS have already been authorized for clinical immunization and are showing great advantages over conventional vaccines. This article describes in depth some critical issues relevant to the development of liposomes as a VADS, including principles underlying immunization, physicochemical properties of liposomes as the immunity-influencing factors, functional material modification to enhance immunostimulatory functions, the state-of-the-art liposome VADSs, as well as the marketed vaccines based on a liposome VADS. Therefore, this article provides a comprehensive reference to the development of novel liposome vaccines.
Collapse
Affiliation(s)
- Ning Wang
- School of Food and Bioengineering, Hefei University of Technology, 193 Tun Brook Road, Hefei, Anhui Province 230009, China
| | - Minnan Chen
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China
| | - Ting Wang
- School of Pharmacy, Anhui Medical University, 81 Plum Hill Road, Hefei, Anhui Province 230032, China.
| |
Collapse
|
34
|
Duarah S, Sharma M, Wen J. Recent advances in microneedle-based drug delivery: Special emphasis on its use in paediatric population. Eur J Pharm Biopharm 2019; 136:48-69. [DOI: 10.1016/j.ejpb.2019.01.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/24/2018] [Accepted: 01/07/2019] [Indexed: 12/12/2022]
|
35
|
Challenges of using lipopolysaccharides for cancer immunotherapy and potential delivery-based solutions thereto. Ther Deliv 2019; 10:165-187. [DOI: 10.4155/tde-2018-0076] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Despite being one of the earliest Toll-like receptor (TLR)-based cancer immunotherapeutics discovered and investigated, the full extent of lipopolysaccharide (LPS) potentials within this arena remains hitherto unexploited. In this review, we will debate the challenges that have complicated the improvement of LPS-based immunotherapeutic approaches in cancer therapy. Based on their nature, those will be discussed with a focus on side effect-related, tolerance-related and in vivo model-related challenges. We will then explore how drug delivery strategies can be integrated within this domain to address such challenges in order to improve the therapeutic outcome, and will present a summary of the studies that have been dedicated thereto. This paper may inspire further developments based on reconciling the advantages of drug delivery and LPS-based cancer immunotherapy.
Collapse
|
36
|
Yu R, Mai Y, Zhao Y, Hou Y, Liu Y, Yang J. Targeting strategies of liposomal subunit vaccine delivery systems to improve vaccine efficacy. J Drug Target 2018; 27:780-789. [PMID: 30589361 DOI: 10.1080/1061186x.2018.1547734] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Liposomes are versatile delivery systems and immunological adjuvants that not only can load various antigens, such as proteins, peptides, nucleic acids and carbohydrates, but also can combine them with immunostimulators. Liposomes have great potential in the development of new types of vaccines, and much effort has been devoted to enhancing vaccine efficacy in recent years. Different types of immune cells such as macrophages and dendritic cells play an important role in the immune response and in preventing or treating cancer, allergy or many other infectious diseases. Targeting liposome-based delivery systems to certain immune cells and organs is one of the most effective measures in such treatments. Extensive research has shown that liposomes combined with immunostimulators or modified with pattern recognition receptor ligands can target various immune cells and the lymphatic system, thus not only inducing and promoting the desired immune response but also decreasing adverse effects throughout the body and avoiding targeting irrelevant cell types or tissues. Therefore, in this review, we outline some targeting strategies that can be adopted in the design of liposomal vaccines to improve vaccine efficacy, and we summarise the related liposome-based vaccine applications in several diseases. These applications have great potential to treat or prevent some infectious and intractable diseases.
Collapse
Affiliation(s)
- Rui Yu
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , People's Republic of China
| | - Yaping Mai
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , People's Republic of China
| | - Yue Zhao
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , People's Republic of China
| | - Yanhui Hou
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , People's Republic of China
| | - Yanhua Liu
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , People's Republic of China
| | - Jianhong Yang
- a Department of Pharmaceutics, School of Pharmacy , Ningxia Medical University , Yinchuan , People's Republic of China
| |
Collapse
|
37
|
Kour P, Rath G, Sharma G, Goyal AK. Recent advancement in nanocarriers for oral vaccination. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:S1102-S1114. [DOI: 10.1080/21691401.2018.1533842] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Preeti Kour
- Department of Pharmaceutics, I.S.F. College of Pharmacy, Moga, India
| | - Goutam Rath
- Department of Pharmaceutics, I.S.F. College of Pharmacy, Moga, India
| | - Gazal Sharma
- Department of Food Engineering,Inder Kumar Gujral Punjab Technical University, Kapurthala, India
| | - Amit Kumar Goyal
- Department of Pharmaceutics, I.S.F. College of Pharmacy, Moga, India
| |
Collapse
|
38
|
Lee JW, Prausnitz MR. Drug delivery using microneedle patches: not just for skin. Expert Opin Drug Deliv 2018; 15:541-543. [DOI: 10.1080/17425247.2018.1471059] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Jeong Woo Lee
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Mark R. Prausnitz
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
39
|
Du B, Jia S, Wang Q, Ding X, Liu Y, Yao H, Zhou J. A Self-Targeting, Dual ROS/pH-Responsive Apoferritin Nanocage for Spatiotemporally Controlled Drug Delivery to Breast Cancer. Biomacromolecules 2018; 19:1026-1036. [PMID: 29455519 DOI: 10.1021/acs.biomac.8b00012] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this study, an intelligent pH and ROS dual-responsive drug delivery system based on an apoferritin (AFt) nanocage was prepared. This therapeutic system can specifically self-target 4T1 breast cancer cells by exploiting L-apoferritin receptor SCARA 5, avoiding the nonspecific binding or aggregation of nanoparticles due to the chemical functionalization for targeting. The characteristics of AFt were utilized for the simultaneous delivery of anticancer drug doxorubicin (DOX) and photosensitizer rose bengal (RB). RB exhibited efficient reactive oxygen species (ROS) generation, which can be applied to photodynamic therapy. Meanwhile, the AFt nanocage was prone to undergoing peptide backbone cleavage when oxidized by ROS. Therefore, by combining the intrinsic pH-responsive property of AFt, the dual ROS/pH-responsive system was developed. The time and location of drug release can be controlled by the combination of internal and external stimulus, which avoids the incomplete drug release under single stimulus response. The drug release rate increased significantly (from 26.1% to 92.0%) under low-pH condition (pH 5.0) and laser irradiation. More DOX from AFt entered the nucleus and killed the tumor cells, and the cell inhibition rate was up to ∼83% (DOX concentration: 5 μg/mL) after 48 h incubation. In addition, the biodistribution and the in vivo antitumor efficacy (within 14 d treatment) of the nanosystem were investigated in 4T1 breast cancer BALB/c mice. The results indicated that the system is a promising therapeutic agent involving ROS/pH dual response, self-targeting, and chemo-photodynamic therapy.
Collapse
Affiliation(s)
- Bin Du
- School of Pharmaceutical Sciences , Zhengzhou University , 100 Science Road , Zhengzhou 450001 , China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation , Henan Province 100 Science Road , Zhengzhou 450001 , China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases , Henan Province 100 Science Road , Zhengzhou 450001 , China
| | - Shaona Jia
- School of Pharmaceutical Sciences , Zhengzhou University , 100 Science Road , Zhengzhou 450001 , China
| | - Qinghui Wang
- School of Pharmaceutical Sciences , Zhengzhou University , 100 Science Road , Zhengzhou 450001 , China
| | - Xiaoyu Ding
- School of Pharmaceutical Sciences , Zhengzhou University , 100 Science Road , Zhengzhou 450001 , China
| | - Ying Liu
- School of Pharmaceutical Sciences , Zhengzhou University , 100 Science Road , Zhengzhou 450001 , China
| | - Hanchun Yao
- School of Pharmaceutical Sciences , Zhengzhou University , 100 Science Road , Zhengzhou 450001 , China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation , Henan Province 100 Science Road , Zhengzhou 450001 , China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases , Henan Province 100 Science Road , Zhengzhou 450001 , China
| | - Jie Zhou
- School of Pharmaceutical Sciences , Zhengzhou University , 100 Science Road , Zhengzhou 450001 , China.,Collaborative Innovation Center of New Drug Research and Safety Evaluation , Henan Province 100 Science Road , Zhengzhou 450001 , China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases , Henan Province 100 Science Road , Zhengzhou 450001 , China
| |
Collapse
|
40
|
Microneedles as the technique of drug delivery enhancement in diverse organs and tissues. J Control Release 2018; 270:184-202. [DOI: 10.1016/j.jconrel.2017.11.048] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/22/2017] [Accepted: 11/29/2017] [Indexed: 11/24/2022]
|
41
|
Wan WL, Chung MF, Shih PC, Sung HW. Response to Comment on "A Liposomal System Capable of Generating CO 2 Bubbles to Induce Transient Cavitation, Lysosomal Rupturing and Cell Necrosis". Angew Chem Int Ed Engl 2017; 56:11690-11692. [PMID: 28815902 DOI: 10.1002/anie.201706509] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Wei-Lin Wan
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan ROC
| | - Min-Fan Chung
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan ROC
| | - Po-Chien Shih
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan ROC
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan ROC
| |
Collapse
|
42
|
Wan WL, Chung MF, Shih PC, Sung HW. Response to Comment on “A Liposomal System Capable of Generating CO 2Bubbles to Induce Transient Cavitation, Lysosomal Rupturing and Cell Necrosis”. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201706509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Wei-Lin Wan
- Department of Chemical Engineering and Institute of Biomedical Engineering; National Tsing Hua University; Hsinchu 30013 Taiwan ROC
| | - Min-Fan Chung
- Department of Chemical Engineering and Institute of Biomedical Engineering; National Tsing Hua University; Hsinchu 30013 Taiwan ROC
| | - Po-Chien Shih
- Department of Chemical Engineering and Institute of Biomedical Engineering; National Tsing Hua University; Hsinchu 30013 Taiwan ROC
| | - Hsing-Wen Sung
- Department of Chemical Engineering and Institute of Biomedical Engineering; National Tsing Hua University; Hsinchu 30013 Taiwan ROC
| |
Collapse
|