1
|
Wu P, Zhang T, Zhao D, Xie Y, Huang D, Li Z, Huang Y. Microneedle-Enabled Breakthroughs in Nucleic Acid Therapeutics. Adv Healthc Mater 2025:e2501015. [PMID: 40370139 DOI: 10.1002/adhm.202501015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/30/2025] [Indexed: 05/16/2025]
Abstract
Nucleic acid therapy demonstrates great potential in cancer treatment, infectious disease prevention, and vaccine development due to its advantages, such as rapid production, long-lasting effects, and high target specificity. Although nucleic acid therapy is considered ideal for the development of novel therapeutic strategies, its clinical application still faces numerous challenges, including the lack of efficient delivery systems, insufficient drug formulation stability, and the limitations imposed by the skin barrier on drug dosage delivery. Microneedles, as an innovative transdermal drug delivery technology, can penetrate the stratum corneum and directly access the skin's microcirculation, enabling the efficient delivery of genes and drugs. This technology offers several advantages, such as ease of operation, minimally invasive and painless application, and high safety. Combining microneedle technology with nucleic acid therapy fully leverages the strengths of both approaches, significantly enhancing therapeutic efficacy and bioavailability while maximizing treatment potential. This review explores the application prospects and advantages of combining microneedle delivery systems with nucleic acid therapy.
Collapse
Affiliation(s)
- Pengfei Wu
- School of Life Science, School of Interdisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical, Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
- School of Medical Engineering, School of Interdisciplinary Science, Affiliated Zhuhai People's Hospital, Beijing Institute of Technology, Zhuhai, 519088, P.R. China
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan, 250100, P.R. China
- Zhengzhou Research Institute, Beijing Institute of Technology, Zhengzhou, 450040, P. R. China
| | - Tian Zhang
- School of Life Science, School of Interdisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical, Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
- School of Medical Engineering, School of Interdisciplinary Science, Affiliated Zhuhai People's Hospital, Beijing Institute of Technology, Zhuhai, 519088, P.R. China
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan, 250100, P.R. China
- Zhengzhou Research Institute, Beijing Institute of Technology, Zhengzhou, 450040, P. R. China
| | - Deyao Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Erqi, Zhengzhou, 450000, P. R. China
| | - Yingqiu Xie
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Astana, 010000, Kazakhstan
| | - Dong Huang
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, School of Integrated Circuits, Peking University, Beijing, 100871, P. R. China
- AciMicro Medical Technology, Guangzhou, 510700, P. R. China
| | - Zhihong Li
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, School of Integrated Circuits, Peking University, Beijing, 100871, P. R. China
| | - Yuanyu Huang
- School of Life Science, School of Interdisciplinary Science, Aerospace Center Hospital, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical, Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, P. R. China
- School of Medical Engineering, School of Interdisciplinary Science, Affiliated Zhuhai People's Hospital, Beijing Institute of Technology, Zhuhai, 519088, P.R. China
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan, 250100, P.R. China
- Zhengzhou Research Institute, Beijing Institute of Technology, Zhengzhou, 450040, P. R. China
| |
Collapse
|
2
|
Hulimane Shivaswamy R, Binulal P, Benoy A, Lakshmiramanan K, Bhaskar N, Pandya HJ. Microneedles as a Promising Technology for Disease Monitoring and Drug Delivery: A Review. ACS MATERIALS AU 2025; 5:115-140. [PMID: 39802146 PMCID: PMC11718548 DOI: 10.1021/acsmaterialsau.4c00125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025]
Abstract
The delivery of molecules, such as DNA, RNA, peptides, and certain hydrophilic drugs, across the epidermal barrier poses a significant obstacle. Microneedle technology has emerged as a prominent area of focus in biomedical research because of its ability to deliver a wide range of biomolecules, vaccines, medicines, and other substances through the skin. Microneedles (MNs) form microchannels by disrupting the skin's structure, which compromises its barrier function, and facilitating the easy penetration of drugs into the skin. These devices enhance the administration of many therapeutic substances to the skin, enhancing their stability. Transcutaneous delivery of medications using a microneedle patch offers advantages over conventional drug administration methods. Microneedles containing active substances can be stimulated by different internal and external factors to result in the regulated release of the substances. To achieve efficient drug administration to the desired location, it is necessary to consider the design of needles with appropriate optimized characteristics. The choice of materials for developing and manufacturing these devices is vital in determining the pharmacodynamics and pharmacokinetics of drug delivery. This article provides the most recent update and overview of the numerous microneedle systems that utilize different activators to stimulate the release of active components from the microneedles. Further, it discusses the materials utilized for producing microneedles and the design strategies important in managing the release of drugs. An explanation of the commonly employed fabrication techniques in biomedical applications and electronics, particularly for integrated microneedle drug delivery systems, is discussed. To successfully implement microneedle technology in clinical settings, it is essential to comprehensively assess several factors, such as biocompatibility, drug stability, safety, and production cost. Finally, an in-depth review of these criteria and the difficulties and potential future direction of microneedles in delivering drugs and monitoring diseases is explored.
Collapse
Affiliation(s)
| | - Pranav Binulal
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| | - Aloysious Benoy
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| | - Kaushik Lakshmiramanan
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| | - Nitu Bhaskar
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| | - Hardik Jeetendra Pandya
- Department of Electronic
Systems Engineering, Indian Institute of
Science, Bangalore 560012, India
| |
Collapse
|
3
|
Koenitz L, Crean A, Vucen S. Pharmacokinetic differences between subcutaneous injection and intradermal microneedle delivery of protein therapeutics. Eur J Pharm Biopharm 2024; 204:114517. [PMID: 39349073 DOI: 10.1016/j.ejpb.2024.114517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
Protein therapeutics are essential in the treatment of various diseases, but most of them require parenteral administration. Since intravenous and subcutaneous injections are associated with discomfort and pain, other routes have been investigated including intradermal microneedle delivery. Microneedles are shorter than hypodermic needles and therefore minimize contact with pain receptors in deeper skin layers. But the differences in anatomical and physiological characteristics of dermis and subcutis can potentially result in varying protein penetration through the skin, absorption, and metabolism. This review summarizes pharmacokinetic studies that compare the administration of protein therapeutics by subcutaneous injections and different types of microneedles intradermally including hollow, dissolvable, coated, and hydrogel-forming microneedles. Across animal and human studies, hollow microneedle delivery resulted in quicker and higher peak plasma levels of proteins and comparable bioavailability to subcutaneous injections potentially due to the extensive network of lymphatic and blood vessels in the dermis. In case of dissolvable and coated microneedles, drug release kinetics depend on component materials. The dissolution of polymer excipients can slow the release and permeation of protein therapeutics at the administration site and thereby delay absorption. The understanding of drug penetration through different skin layers, its absorption into blood capillaries or lymphatics, and dermal metabolism remains limited. Additionally, the effects of these processes on the differences in pharmacokinetic profiles of proteins following intradermal microneedle administration are not well understood. Greater insights are required for the development of the next generation of intradermal microneedle biotherapeutics.
Collapse
Affiliation(s)
- Laura Koenitz
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland.
| | - Abina Crean
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| | - Sonja Vucen
- SSPC, the SFI Research Centre for Pharmaceuticals, School of Pharmacy, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
4
|
Rajesh NU, Luna Hwang J, Xu Y, Saccone MA, Hung AH, Hernandez RAS, Coates IA, Driskill MM, Dulay MT, Jacobson GB, Tian S, Perry JL, DeSimone JM. 3D-Printed Latticed Microneedle Array Patches for Tunable and Versatile Intradermal Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404606. [PMID: 39221508 DOI: 10.1002/adma.202404606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/29/2024] [Indexed: 09/04/2024]
Abstract
Using high-resolution 3D printing, a novel class of microneedle array patches (MAPs) is introduced, called latticed MAPs (L-MAPs). Unlike most MAPs which are composed of either solid structures or hollow needles, L-MAPs incorporate tapered struts that form hollow cells capable of trapping liquid droplets. The lattice structures can also be coated with traditional viscous coating formulations, enabling both liquid- and solid-state cargo delivery, on a single patch. Here, a library of 43 L-MAP designs is generated and in-silico modeling is used to down-select optimal geometries for further characterization. Compared to traditionally molded and solid-coated MAPs, L-MAPs can load more cargo with fewer needles per patch, enhancing cargo loading and drug delivery capabilities. Further, L-MAP cargo release kinetics into the skin can be tuned based on formulation and needle geometry. In this work, the utility of L-MAPs as a platform is demonstrated for the delivery of small molecules, mRNA lipid nanoparticles, and solid-state ovalbumin protein. In addition, the production of programmable L-MAPs is demonstrated with tunable cargo release profiles, enabled by combining needle geometries on a single patch.
Collapse
Affiliation(s)
- Netra U Rajesh
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Jihyun Luna Hwang
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Yue Xu
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Max A Saccone
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Andy H Hung
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Rosa A S Hernandez
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Ian A Coates
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Madison M Driskill
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Maria T Dulay
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | | | - Shaomin Tian
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jillian L Perry
- Center for Nanotechnology in Drug Delivery and Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joseph M DeSimone
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
5
|
Anjani QK, Nainggolan ADC, Li H, Miatmoko A, Larrañeta E, Donnelly RF. Parafilm® M and Strat-M® as skin simulants in in vitro permeation of dissolving microarray patches loaded with proteins. Int J Pharm 2024; 655:124071. [PMID: 38554738 DOI: 10.1016/j.ijpharm.2024.124071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
In vitro permeation studies play a crucial role in early formulation optimisation before extensive animal model investigations. Biological membranes are typically used in these studies to mimic human skin conditions accurately. However, when focusing on protein and peptide transdermal delivery, utilising biological membranes can complicate analysis and quantification processes. This study aims to explore Parafilm®M and Strat-M® as alternatives to dermatomed porcine skin for evaluating protein delivery from dissolving microarray patch (MAP) platforms. Initially, various MAPs loaded with different model proteins (ovalbumin, bovine serum albumin and amniotic mesenchymal stem cell metabolite products) were prepared. These dissolving MAPs underwent evaluation for insertion properties and in vitro permeation profiles when combined with different membranes, dermatomed porcine skin, Parafilm®M, and Strat-M®. Insertion profiles indicated that both Parafilm®M and Strat-M® showed comparable insertion depths to dermatomed porcine skin (in range of 360-430 µm), suggesting promise as membrane substitutes for insertion studies. In in vitro permeation studies, synthetic membranes such as Parafilm®M and Strat-M® demonstrated the ability to bypass protein-derived skin interference, providing more reliable results compared to dermatomed neonatal porcine skin. Consequently, these findings present valuable tools for preliminary screening across various MAP formulations, especially in the transdermal delivery of proteins and peptides.
Collapse
Affiliation(s)
- Qonita Kurnia Anjani
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK; Fakultas Farmasi, Universitas Megarezky, Jl. Antang Raya No. 43, Makassar 90234, Indonesia
| | | | - Huanhuan Li
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Andang Miatmoko
- Faculty of Pharmacy, Airlangga University, Nanizar Zaman Joenoes Building, Campus C, Mulyorejo, Surabaya 60115, Indonesia; Stem Cell Research and Development Center, Airlangga University, Institute of Tropical Disease Building, Campus C, Mulyorejo, Surabaya 60115, Indonesia
| | - Eneko Larrañeta
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
6
|
Qi Z, Yan Z, Tan G, Kundu SC, Lu S. Smart Responsive Microneedles for Controlled Drug Delivery. Molecules 2023; 28:7411. [PMID: 37959830 PMCID: PMC10649748 DOI: 10.3390/molecules28217411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
As an emerging technology, microneedles offer advantages such as painless administration, good biocompatibility, and ease of self-administration, so as to effectively treat various diseases, such as diabetes, wound repair, tumor treatment and so on. How to regulate the release behavior of loaded drugs in polymer microneedles is the core element of transdermal drug delivery. As an emerging on-demand drug-delivery technology, intelligent responsive microneedles can achieve local accurate release of drugs according to external stimuli or internal physiological environment changes. This review focuses on the research efforts in smart responsive polymer microneedles at home and abroad in recent years. It summarizes the response mechanisms based on various stimuli and their respective application scenarios. Utilizing innovative, responsive microneedle systems offers a convenient and precise targeted drug delivery method, holding significant research implications in transdermal drug administration. Safety and efficacy will remain the key areas of continuous efforts for research scholars in the future.
Collapse
Affiliation(s)
- Zhenzhen Qi
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China; (Z.Q.); (Z.Y.); (G.T.)
| | - Zheng Yan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China; (Z.Q.); (Z.Y.); (G.T.)
| | - Guohongfang Tan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China; (Z.Q.); (Z.Y.); (G.T.)
| | - Subhas C. Kundu
- 3Bs Research Group, I3Bs Research Institute on Biomaterials, Biodegrabilities, and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, AvePark, Guimaraes, 4805-017 Barco, Portugal;
| | - Shenzhou Lu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China; (Z.Q.); (Z.Y.); (G.T.)
| |
Collapse
|
7
|
Dong H, Li Q, Zhang Y, Ding M, Teng Z, Mou Y. Biomaterials Facilitating Dendritic Cell-Mediated Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301339. [PMID: 37088780 PMCID: PMC10288267 DOI: 10.1002/advs.202301339] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/22/2023] [Indexed: 05/03/2023]
Abstract
Dendritic cell (DC)-based cancer immunotherapy has exhibited remarkable clinical prospects because DCs play a central role in initiating and regulating adaptive immune responses. However, the application of traditional DC-mediated immunotherapy is limited due to insufficient antigen delivery, inadequate antigen presentation, and high levels of immunosuppression. To address these challenges, engineered biomaterials have been exploited to enhance DC-mediated immunotherapeutic effects. In this review, vital principal components that can enhance DC-mediated immunotherapeutic effects are first introduced. The parameters considered in the rational design of biomaterials, including targeting modifications, size, shape, surface, and mechanical properties, which can affect biomaterial optimization of DC functions, are further summarized. Moreover, recent applications of various engineered biomaterials in the field of DC-mediated immunotherapy are reviewed, including those serve as immune component delivery platforms, remodel the tumor microenvironment, and synergistically enhance the effects of other antitumor therapies. Overall, the present review comprehensively and systematically summarizes biomaterials related to the promotion of DC functions; and specifically focuses on the recent advances in biomaterial designs for DC activation to eradicate tumors. The challenges and opportunities of treatment strategies designed to amplify DCs via the application of biomaterials are discussed with the aim of inspiring the clinical translation of future DC-mediated cancer immunotherapies.
Collapse
Affiliation(s)
- Heng Dong
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Qiang Li
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Yu Zhang
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Meng Ding
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information DisplaysJiangsu Key Laboratory for BiosensorsInstitute of Advanced MaterialsJiangsu National Synergetic Innovation Centre for Advanced MaterialsNanjing University of Posts and Telecommunications9 Wenyuan RoadNanjingJiangsu210023P. R. China
| | - Yongbin Mou
- Nanjing Stomatological HospitalAffiliated Hospital of Medical School, Nanjing University30 Zhongyang RoadNanjingJiangsu210008P. R. China
| |
Collapse
|
8
|
Singh AK, Malviya R, Prajapati B, Singh S, Goyal P. Utilization of Stimuli-Responsive Biomaterials in the Formulation of Cancer Vaccines. J Funct Biomater 2023; 14:jfb14050247. [PMID: 37233357 DOI: 10.3390/jfb14050247] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Immunology research has focused on developing cancer vaccines to increase the number of tumor-specific effector cells and their ability to fight cancer over the last few decades. There is a lack of professional success in vaccines compared to checkpoint blockade and adoptive T-cell treatment. The vaccine's inadequate delivery method and antigen selection are most likely to blame for the poor results. Antigen-specific vaccines have recently shown promising results in preclinical and early clinical investigations. To target particular cells and trigger the best immune response possible against malignancies, it is necessary to design a highly efficient and secure delivery method for cancer vaccines; however, enormous challenges must be overcome. Current research is focused on developing stimulus-responsive biomaterials, which are a subset of the range of levels of materials, to enhance therapeutic efficacy and safety and better regulate the transport and distribution of cancer immunotherapy in vivo. A concise analysis of current developments in the area of biomaterials that respond to stimuli has been provided in brief research. Current and anticipated future challenges and opportunities in the sector are also highlighted.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India
| | - Bhupendra Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva 384012, India
| | - Sudarshan Singh
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Priyanshi Goyal
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, India
| |
Collapse
|
9
|
Novel Pharmaceutical Strategies for Enhancing Skin Penetration of Biomacromolecules. Pharmaceuticals (Basel) 2022; 15:ph15070877. [PMID: 35890174 PMCID: PMC9317023 DOI: 10.3390/ph15070877] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022] Open
Abstract
Skin delivery of biomacromolecules holds great advantages in the systemic and local treatment of multiple diseases. However, the densely packed stratum corneum and the tight junctions between keratinocytes stand as formidable skin barriers against the penetration of most drug molecules. The large molecular weight, high hydrophilicity, and lability nature of biomacromolecules pose further challenges to their skin penetration. Recently, novel penetration enhancers, nano vesicles, and microneedles have emerged as efficient strategies to deliver biomacromolecules deep into the skin to exert their therapeutic action. This paper reviews the potential application and mechanisms of novel skin delivery strategies with emphasis on the pharmaceutical formulations.
Collapse
|
10
|
Cho H, Jeon SI, Ahn CH, Shim MK, Kim K. Emerging Albumin-Binding Anticancer Drugs for Tumor-Targeted Drug Delivery: Current Understandings and Clinical Translation. Pharmaceutics 2022; 14:728. [PMID: 35456562 PMCID: PMC9028280 DOI: 10.3390/pharmaceutics14040728] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/20/2022] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Albumin has shown remarkable promise as a natural drug carrier by improving pharmacokinetic (PK) profiles of anticancer drugs for tumor-targeted delivery. The exogenous or endogenous albumin enhances the circulatory half-lives of anticancer drugs and passively target the tumors by the enhanced permeability and retention (EPR) effect. Thus, the albumin-based drug delivery leads to a potent antitumor efficacy in various preclinical models, and several candidates have been evaluated clinically. The most successful example is Abraxane, an exogenous human serum albumin (HSA)-bound paclitaxel formulation approved by the FDA and used to treat locally advanced or metastatic tumors. However, additional clinical translation of exogenous albumin formulations has not been approved to date because of their unexpectedly low delivery efficiency, which can increase the risk of systemic toxicity. To overcome these limitations, several prodrugs binding endogenous albumin covalently have been investigated owing to distinct advantages for a safe and more effective drug delivery. In this review, we give account of the different albumin-based drug delivery systems, from laboratory investigations to clinical applications, and their potential challenges, and the outlook for clinical translation is discussed. In addition, recent advances and progress of albumin-binding drugs to move more closely to the clinical settings are outlined.
Collapse
Affiliation(s)
- Hanhee Cho
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Korea
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Seong Ik Jeon
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Cheol-Hee Ahn
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Korea
| | - Man Kyu Shim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Kwangmeyung Kim
- Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| |
Collapse
|
11
|
Faraji Rad Z, Prewett PD, Davies GJ. An overview of microneedle applications, materials, and fabrication methods. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2021; 12:1034-1046. [PMID: 34621614 PMCID: PMC8450954 DOI: 10.3762/bjnano.12.77] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/30/2021] [Indexed: 05/19/2023]
Abstract
Microneedle-based microdevices promise to expand the scope for delivery of vaccines and therapeutic agents through the skin and withdrawing biofluids for point-of-care diagnostics - so-called theranostics. Unskilled and painless applications of microneedle patches for blood collection or drug delivery are two of the advantages of microneedle arrays over hypodermic needles. Developing the necessary microneedle fabrication processes has the potential to dramatically impact the health care delivery system by changing the landscape of fluid sampling and subcutaneous drug delivery. Microneedle designs which range from sub-micron to millimetre feature sizes are fabricated using the tools of the microelectronics industry from metals, silicon, and polymers. Various types of subtractive and additive manufacturing processes have been used to manufacture microneedles, but the development of microneedle-based systems using conventional subtractive methods has been constrained by the limitations and high cost of microfabrication technology. Additive manufacturing processes such as 3D printing and two-photon polymerization fabrication are promising transformative technologies developed in recent years. The present article provides an overview of microneedle systems applications, designs, material selection, and manufacturing methods.
Collapse
Affiliation(s)
- Zahra Faraji Rad
- School of Mechanical and Electrical Engineering, University of Southern Queensland, Springfield Central, QLD 4300, Australia
| | - Philip D Prewett
- Department of Mechanical Engineering, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Oxacus Ltd, Dorchester-on-Thames, OX10 7HN, United Kingdom
| | - Graham J Davies
- Faculty of Engineering, UNSW Australia, NSW 2052, Australia
- College of Engineering & Physical Sciences, School of Engineering, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| |
Collapse
|
12
|
Ameri M, Ao Y, Lewis H. Formulation Approach that Enables the Coating of a Stable Influenza Vaccine on a Transdermal Microneedle Patch. AAPS PharmSciTech 2021; 22:175. [PMID: 34114100 DOI: 10.1208/s12249-021-02044-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/10/2021] [Indexed: 11/30/2022] Open
Abstract
A trivalent influenza split vaccine was formulated at high concentration for coating on the transdermal microneedle system. Monovalent vaccine bulks of three influenza strains, two influenza A strains, and one B strain were diafiltrated, concentrated, and lyophilized. The lyophilized powder of each vaccine strain was separately reconstituted and subsequently combined into a coating formulation of high concentration trivalent vaccine. The formulation process converted the monovalent vaccine bulks with low hemagglutinin (HA) concentrations 0.1 mg/mL into a viscous, emulsion containing HA at ~50 mg/mL. This physically stable emulsion demonstrated viscosity 1 poise and 30° contact angle for effective, homogeneous coating on each microneedle. Evaluation of the vaccine antigen HA by SRID and SDS-PAGE/Western blot showed that HA remained stable throughout the vaccine transdermal microneedle system manufacturing process and 1-year ambient storage (25°C). Anti-influenza antibody responses were evaluated by ELISA and hemagglutination inhibition (HAI) assay after primary and booster immunization with the vaccine-coated transdermal microneedle systems at either 25-μg or 40-μg total HA. The results showed the induction of serum anti-influenza IgG and anti-HA neutralizing antibodies after primary immunization and significant titer rises after booster immunization for both doses, indicating the dry-coated trivalent vaccine delivered by transdermal microneedle system elicited both primary and recall antibody responses against all three antigen strains. The study demonstrates that the transdermal microneedle system provides an attractive alternative for influenza vaccine delivery with key advantages such as preservative-free and room-temperature storage.
Collapse
|
13
|
Lee J, van der Maaden K, Gooris G, O'Mahony C, Jiskoot W, Bouwstra J. Engineering of an automated nano-droplet dispensing system for fabrication of antigen-loaded dissolving microneedle arrays. Int J Pharm 2021; 600:120473. [PMID: 33737094 DOI: 10.1016/j.ijpharm.2021.120473] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/28/2021] [Accepted: 03/05/2021] [Indexed: 12/13/2022]
Abstract
Dissolving microneedle arrays (dMNAs) are promising devices for intradermal vaccine delivery. The aim of this study was to develop a reproducible fabrication method for dMNAs based on an automated nano-droplet dispensing system that minimizes antigen waste. First, a polymer formulation was selected to dispense sufficiently small droplets (<18 nL) that can enter the microneedle cavities (base diameter 330 µm). Besides, three linear stages were assembled to align the dispenser with the cavities, and a vacuum chamber was designed to fill the cavities with dispensed droplets without entrapped air. Lastly, the dispenser and stages were incorporated to build a fully automated system. To examine the function of dMNAs as a vaccine carrier, ovalbumin was loaded in dMNAs by dispensing a mixture of ovalbumin and polymer formulation, followed by determining the ovalbumin loading and release into the skin. The results demonstrate that functional dMNAs which can deliver antigen into the skin were successfully fabricated via the automatic fabrication system, and hardly any antigen waste was encountered. Compared to the method that centrifuges the mould, it resulted in a 98.5% volume reduction of antigen/polymer solution and a day shorter production time. This system has potential for scale-up of manufacturing to an industrial scale.
Collapse
Affiliation(s)
- Jihui Lee
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 2300, Einsteinweg 55, 2333 CC Leiden, the Netherlands
| | - Koen van der Maaden
- Tumor Immunology Group, Department of Immunology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; TECO Development GmbH, 53359 Rheinbach, Germany
| | - Gerrit Gooris
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 2300, Einsteinweg 55, 2333 CC Leiden, the Netherlands
| | - Conor O'Mahony
- Tyndall National Institute, University College Cork, Cork T12 R5CP, Ireland
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 2300, Einsteinweg 55, 2333 CC Leiden, the Netherlands
| | - Joke Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, P.O. Box 2300, Einsteinweg 55, 2333 CC Leiden, the Netherlands.
| |
Collapse
|
14
|
Chen Y, Chen N, Feng X. The role of internal and external stimuli in the rational design of skin-specific drug delivery systems. Int J Pharm 2021; 592:120081. [PMID: 33189810 DOI: 10.1016/j.ijpharm.2020.120081] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/15/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022]
Abstract
The concept of skin-specific drug delivery with a spatio-temporal control has just recently received concerns in dermatology. Inspired by the progress in smart materials and their perspective application in medicine science, development of stimuli responsive drug delivery systems with skin-specificity has become possible, which has led to a new era in the localized treatment of skin diseases. This review highlights both the internal and external stimuli that have been employed in this field, with a focus on their implication on the rational design of pharmaceutical formulations, especially those nanoscale drug carriers that are able to provide release of payloads with a precise spatio-temporal control in response to specific stimuli. Also, the strategy of dual stimuli responsive drug delivery systems will be discussed for further improvement of the efficacy of skin drug delivery. The prominent examples of the established approaches are described as comprehensive and current as possible. The review is expected to provide some inspiration for utilizing different stimuli for realizing the site-specific and on-demand drug delivery to the skin.
Collapse
Affiliation(s)
- Yang Chen
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China.
| | - Naiying Chen
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang, No.77 Puhe Road, Shenyang North New Area, Shenyang 110122, China
| | - Xun Feng
- Department of Sanitary Inspection, School of Public Health, Shenyang Medical College, No.146 Yellow River North Street, Shenyang 110034, China
| |
Collapse
|
15
|
Li D, Hu D, Xu H, Patra HK, Liu X, Zhou Z, Tang J, Slater N, Shen Y. Progress and perspective of microneedle system for anti-cancer drug delivery. Biomaterials 2020; 264:120410. [PMID: 32979655 DOI: 10.1016/j.biomaterials.2020.120410] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023]
Abstract
Transdermal drug delivery exhibited encouraging prospects, especially through superficial drug administration routes. However, only a few limited lipophilic drug molecules could cross the skin barrier, those are with low molecular weight and rational Log P value. Microneedles (MNs) can overcome these limitations to deliver numerous drugs into the dermal layer by piercing the outermost skin layer of the body. In the case of superficial cancer treatments, topical drug administration faces severely low transfer efficiency, and systemic treatments are always associated with side effects and premature drug degradation. MN-based systems have achieved excellent technical capabilities and been tested for pre-clinical chemotherapy, photothermal therapy, photodynamic therapy, and immunotherapy. In this review, we will focus on the features, progress, and opportunities of MNs in the anticancer drug delivery system. Then, we will discuss the strategies and advantages in these works and summarize challenges, perspectives, and translational potential for future applications.
Collapse
Affiliation(s)
- Dongdong Li
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Doudou Hu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Hongxia Xu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Hirak K Patra
- Wolfson College, University of Cambridge, Cambridge, CB3 9BB, United Kingdom; Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, CB3 0AS, United Kingdom
| | - Xiangrui Liu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhuxian Zhou
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jianbin Tang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Nigel Slater
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, CB3 0AS, United Kingdom
| | - Youqing Shen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
16
|
|
17
|
Ingrole RSJ, Gill HS. Microneedle Coating Methods: A Review with a Perspective. J Pharmacol Exp Ther 2019; 370:555-569. [PMID: 31175217 PMCID: PMC6806358 DOI: 10.1124/jpet.119.258707] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 05/31/2019] [Indexed: 02/06/2023] Open
Abstract
A coated microneedle array comprises sharp micrometer-sized needle shafts attached to a base substrate and coated with a drug on their surfaces. Coated microneedles are under investigation for drug delivery into the skin and other tissues, and a broad assortment of active materials, including small molecules, peptides, proteins, deoxyribonucleic acids, and viruses, have been coated onto microneedles. To coat the microneedles, different methods have been developed. Some coating methods achieve selective coating of just the microneedle shafts, whereas other methods coat not only microneedle shafts but also the array base substrate. Selective coating of just the microneedle shafts is more desirable since it provides control over drug dosage, prevents drug waste, and offers high delivery efficiency. Different excipients are added to the coating liquid to modulate its viscosity and surface tension in order to achieve uniform coatings on microneedles. Coated microneedles have been used in a broad range of biomedical applications. To highlight these different applications, a table summarizing the different active materials and the amounts coated on microneedles is provided. We also discuss factors that should be considered when deciding suitability of coated microneedles for new-drug delivery applications. In recent years, many coated microneedles have been investigated in human clinical trials, and there is now a strong effort to bring the first coated microneedle-based product to market.
Collapse
Affiliation(s)
- Rohan S J Ingrole
- Department of Chemical Engineering, Texas Tech University, Lubbock, Texas
| | | |
Collapse
|
18
|
Enhanced anti-tumor immunotherapy by dissolving microneedle patch loaded ovalbumin. PLoS One 2019; 14:e0220382. [PMID: 31386690 PMCID: PMC6684091 DOI: 10.1371/journal.pone.0220382] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 07/15/2019] [Indexed: 01/04/2023] Open
Abstract
The skin is a very suitable organ for the induction of immune responses to vaccine antigens. Antigen delivery systems to the skin by needle and syringe directly deposit the antigen into the epidermal-dermal compartment, one of the most immunocompetent sites due to the presence of professional antigen-presenting cells aimed at the induction of antigen-specific T cells. In this study, we analyzed the amount of ovalbumin as an antigen delivered to the skin by a microneedle. When ovalbumin protein as an antigen was delivered to the skin of mice using a dissolving microneedle, it induced an immune response through the enhanced proliferation and cytokines production by the splenocytes and lymph nodes. Also, it effectively increased the ovalbumin-specific CD8+ T cell and CD4+ T cell population and induced an ovalbumin-specific CTL response against the graft of ovalbumin-expressing EG7 tumor cells in the immunized mice. Also, we identified the inhibition of tumor growth and prevention of tumor formation in the context of the therapeutic and prophylactic vaccine, respectively through EG-7 tumor mouse model. Finally, these data show the potential of patches as attractive antigen delivery vehicles.
Collapse
|
19
|
van der Maaden K, Schipper P, Jiskoot W, Bouwstra JA. Chemical Modifications of Gold Surfaces with Basic Groups and a Fluorescent Nanoparticle Adhesion Assay To Determine Their Surface p K a. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:7121-7128. [PMID: 31045370 DOI: 10.1021/acs.langmuir.9b00139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
For pharmaceutical, biological, and biomedical applications, the functionalization of gold surfaces with pH-sensitive groups has great potential. The aim of this work was to modify gold surfaces with pH-sensitive groups and to determine the p Ka of the modified gold surfaces using a fluorescent nanoparticle adhesion assay. To introduce pH-sensitive groups onto gold surfaces, we modified gold-coated silicon slides with four different bases: 4-mercaptopyridine (4-MP), 4-pyridylethylmercaptan (4-PEM), 4-aminothiophenol (4-ATP), and 2-mercaptoethylamine (2-MEA). To screen whether the modifications were successful, the binding of negatively charged fluorescently labeled nanoparticles to the positively charged surfaces was visualized by fluorescence microscopy and atomic force microscopy. Next, the p Ka of the modified surfaces was determined by quantifying the pH-dependent adhesion of the fluorescently labeled nanoparticles with fluorescence spectroscopy. Fluorescence microscopy showed that the gold surfaces were successfully modified with the four different basic molecules. Moreover, fluorescence spectroscopy revealed that fluorescently labeled negatively charged nanoparticles bound onto gold surfaces that were modified with one of the four bases in a pH-dependent manner. By quantifying the adsorption of negatively charged fluorescently labeled nanoparticles onto the functionalized gold surfaces and using the Henderson-Hasselbalch equation, the p Ka of these surfaces was determined to be 3.7 ± 0.1 (4-MP), 5.0 ± 0.1 (4-PEM), 5.4 ± 0.1 (4-ATP), and 7.4 ± 0.3 (2-MEA). We successfully functionalized gold surfaces with four different basic molecules, yielding modified surfaces with different p Ka values, as determined with a fluorescent nanoparticle adhesion assay.
Collapse
Affiliation(s)
- K van der Maaden
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR) , Leiden University , 2300 RA Leiden , The Netherlands
| | - P Schipper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR) , Leiden University , 2300 RA Leiden , The Netherlands
| | - W Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR) , Leiden University , 2300 RA Leiden , The Netherlands
| | - J A Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR) , Leiden University , 2300 RA Leiden , The Netherlands
| |
Collapse
|
20
|
Du G, Woythe L, van der Maaden K, Leone M, Romeijn S, Kros A, Kersten G, Jiskoot W, Bouwstra JA. Coated and Hollow Microneedle-Mediated Intradermal Immunization in Mice with Diphtheria Toxoid Loaded Mesoporous Silica Nanoparticles. Pharm Res 2018; 35:189. [PMID: 30105542 PMCID: PMC6096895 DOI: 10.1007/s11095-018-2476-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/06/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE To examine the immunogenicity of diphtheria toxoid (DT) loaded mesoporous silica nanoparticles (MSNs) after coated and hollow microneedle-mediated intradermal immunization in mice. METHODS DT was loaded into MSNs and the nanoparticle surface was coated with a lipid bilayer (LB-MSN-DT). To prepare coated microneedles, alternating layers of negatively charged LB-MSN-DT and positively charged N-trimethyl chitosan (TMC) were coated onto pH-sensitive microneedle arrays via a layer-by-layer approach. Microneedle arrays coated with 5 or 3 layers of LB-MSN-DT were used to immunize mice and the elicited antibody responses were compared with those induced by hollow microneedle-injected liquid formulation of LB-MSN-DT. Liquid DT formulation with and without TMC (DT/TMC) injected by a hollow microneedle were used as controls. RESULTS LB-MSN-DT had an average size of about 670 nm and a zeta potential of -35 mV. The encapsulation efficiency of DT in the nanoparticles was 77%. The amount of nano-encapsulated DT coated onto the microneedle array increased linearly with increasing number of the coating layers. Nano-encapsulated DT induced stronger immune responses than DT solution when delivered intradermally via hollow microneedles, but not when delivered via coated microneedles. CONCLUSION Both the nano-encapsulation of DT and the type of microneedles affect the immunogenicity of the antigen.
Collapse
Affiliation(s)
- Guangsheng Du
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Laura Woythe
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Koen van der Maaden
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mara Leone
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Stefan Romeijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Gideon Kersten
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Institute for Translational Vaccinology (Intravacc), Bilthoven, The Netherlands
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Joke A Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
21
|
Courtenay AJ, McCrudden MTC, McAvoy KJ, McCarthy HO, Donnelly RF. Microneedle-Mediated Transdermal Delivery of Bevacizumab. Mol Pharm 2018; 15:3545-3556. [DOI: 10.1021/acs.molpharmaceut.8b00544] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Aaron J. Courtenay
- School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, U.K
| | - Maelíosa T. C. McCrudden
- School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, U.K
| | - Kathryn J. McAvoy
- School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, U.K
| | - Helen O. McCarthy
- School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, U.K
| | - Ryan F. Donnelly
- School of Pharmacy, Queen’s University Belfast, Medical Biology Centre, 97 Lisburn Road, Belfast BT9 7BL, U.K
| |
Collapse
|
22
|
Vallhov H, Xia W, Engqvist H, Scheynius A. Bioceramic microneedle arrays are able to deliver OVA to dendritic cells in human skin. J Mater Chem B 2018; 6:6808-6816. [DOI: 10.1039/c8tb01476k] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bioceramic arrays with high-aspect-ratio microneedles were able to penetrate human ex vivo skin and deliver their cargo to dendritic cells.
Collapse
Affiliation(s)
- Helen Vallhov
- Department of Clinical Science and Education
- Karolinska Institutet
- and Sachs' Children and Youth Hospital
- SE-118 83 Stockholm
- Sweden
| | - Wei Xia
- Division for Applied Materials Science
- Department of Engineering Sciences
- The Ångström Laboratory
- Uppsala University
- SE-751 21 Uppsala
| | - Håkan Engqvist
- Division for Applied Materials Science
- Department of Engineering Sciences
- The Ångström Laboratory
- Uppsala University
- SE-751 21 Uppsala
| | - Annika Scheynius
- Department of Clinical Science and Education
- Karolinska Institutet
- and Sachs' Children and Youth Hospital
- SE-118 83 Stockholm
- Sweden
| |
Collapse
|
23
|
Hollow microneedle-mediated intradermal delivery of model vaccine antigen-loaded PLGA nanoparticles elicits protective T cell-mediated immunity to an intracellular bacterium. J Control Release 2017; 266:27-35. [PMID: 28917531 DOI: 10.1016/j.jconrel.2017.09.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 02/08/2023]
Abstract
The skin is an attractive organ for immunization due to the presence of a large number of epidermal and dermal antigen-presenting cells. Hollow microneedles allow for precise and non-invasive intradermal delivery of vaccines. In this study, ovalbumin (OVA)-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles with and without TLR3 agonist poly(I:C) were prepared and administered intradermally by hollow microneedles. The capacity of the PLGA nanoparticles to induce a cytotoxic T cell response, contributing to protection against intracellular pathogens, was examined. We show that a single injection of OVA-loaded PLGA nanoparticles, compared to soluble OVA, primed both adoptively transferred antigen-specific naïve transgenic CD8+ and CD4+ T cells with markedly high efficiency. Applying a triple immunization protocol, PLGA nanoparticles primed also endogenous OVA-specific CD8+ T cells. Immune response, following immunization with in particular anionic PLGA nanoparticles co-encapsulated with OVA and poly(I:C), provided protection against a recombinant strain of the intracellular bacterium Listeria monocytogenes, secreting OVA. Taken together, we show that PLGA nanoparticle formulation is an excellent delivery system for protein antigen into the skin and that protective cellular immune responses can be induced using hollow microneedles for intradermal immunizations.
Collapse
|
24
|
Bhatnagar S, Dave K, Venuganti VVK. Microneedles in the clinic. J Control Release 2017; 260:164-182. [DOI: 10.1016/j.jconrel.2017.05.029] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 05/21/2017] [Accepted: 05/23/2017] [Indexed: 12/16/2022]
|
25
|
Tu J, Du G, Reza Nejadnik M, Mönkäre J, van der Maaden K, Bomans PHH, Sommerdijk NAJM, Slütter B, Jiskoot W, Bouwstra JA, Kros A. Mesoporous Silica Nanoparticle-Coated Microneedle Arrays for Intradermal Antigen Delivery. Pharm Res 2017; 34:1693-1706. [PMID: 28536970 PMCID: PMC5498618 DOI: 10.1007/s11095-017-2177-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/09/2017] [Indexed: 11/27/2022]
Abstract
PURPOSE To develop a new intradermal antigen delivery system by coating microneedle arrays with lipid bilayer-coated, antigen-loaded mesoporous silica nanoparticles (LB-MSN-OVA). METHODS Synthesis of MSNs with 10-nm pores was performed and the nanoparticles were loaded with the model antigen ovalbumin (OVA), and coated with a lipid bilayer (LB-MSN-OVA). The uptake of LB-MSN-OVA by bone marrow-derived dendritic cells (BDMCs) was studied by flow cytometry. The designed LB-MSN-OVA were coated onto pH-sensitive pyridine-modified microneedle arrays and the delivery of LB-MSN-OVA into ex vivo human skin was studied. RESULTS The synthesized MSNs demonstrated efficient loading of OVA with a maximum loading capacity of about 34% and the lipid bilayer enhanced the colloidal stability of the MSNs. Uptake of OVA loaded in LB-MSN-OVA by BMDCs was higher than that of free OVA, suggesting effective targeting of LB-MSN-OVA to antigen-presenting cells. Microneedles were readily coated with LB-MSN-OVA at pH 5.8, yielding 1.5 μg of encapsulated OVA per microneedle array. Finally, as a result of the pyridine modification, LB-MSN-OVA were effectively released from the microneedles upon piercing the skin. CONCLUSION Microneedle arrays coated with LB-MSN-OVA were successfully developed and shown to be suitable for intradermal delivery of the encapsulated protein antigen.
Collapse
Affiliation(s)
- Jing Tu
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, Leiden, 2300, RA, The Netherlands
| | - Guangsheng Du
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300, RA, The Netherlands
| | - M Reza Nejadnik
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300, RA, The Netherlands
| | - Juha Mönkäre
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300, RA, The Netherlands
| | - Koen van der Maaden
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300, RA, The Netherlands
| | - Paul H H Bomans
- Laboratory of Materials and Interface Chemistry & Center of Multiscale Electron Microscopy, Department of Chemical Engineering and Chemistry, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600, MB, The Netherlands
| | - Nico A J M Sommerdijk
- Laboratory of Materials and Interface Chemistry & Center of Multiscale Electron Microscopy, Department of Chemical Engineering and Chemistry, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5600, MB, The Netherlands
| | - Bram Slütter
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300, RA, The Netherlands
- Division of Biopharmaceutics, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300, RA, The Netherlands
| | - Wim Jiskoot
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300, RA, The Netherlands
| | - Joke A Bouwstra
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research (LACDR), Leiden University, Leiden, 2300, RA, The Netherlands.
| | - Alexander Kros
- Department of Supramolecular & Biomaterials Chemistry, Leiden Institute of Chemistry (LIC), Leiden University, Leiden, 2300, RA, The Netherlands.
| |
Collapse
|
26
|
Leone M, Mönkäre J, Bouwstra JA, Kersten G. Dissolving Microneedle Patches for Dermal Vaccination. Pharm Res 2017; 34:2223-2240. [PMID: 28718050 PMCID: PMC5643353 DOI: 10.1007/s11095-017-2223-2] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 06/26/2017] [Indexed: 12/31/2022]
Abstract
The dermal route is an attractive route for vaccine delivery due to the easy skin accessibility and a dense network of immune cells in the skin. The development of microneedles is crucial to take advantage of the skin immunization and simultaneously to overcome problems related to vaccination by conventional needles (e.g. pain, needle-stick injuries or needle re-use). This review focuses on dissolving microneedles that after penetration into the skin dissolve releasing the encapsulated antigen. The microneedle patch fabrication techniques and their challenges are discussed as well as the microneedle characterization methods and antigen stability aspects. The immunogenicity of antigens formulated in dissolving microneedles are addressed. Finally, the early clinical development is discussed.
Collapse
Affiliation(s)
- M Leone
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, P.O. Box 9502, 2300 RA, Leiden, the Netherlands
| | - J Mönkäre
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, P.O. Box 9502, 2300 RA, Leiden, the Netherlands
| | - J A Bouwstra
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, P.O. Box 9502, 2300 RA, Leiden, the Netherlands.
| | - G Kersten
- Division of Drug Delivery Technology, Cluster BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, P.O. Box 9502, 2300 RA, Leiden, the Netherlands.,Department of Analytical Development and Formulation, Intravacc, Bilthoven, the Netherlands
| |
Collapse
|
27
|
Bhatnagar S, Chawla SR, Kulkarni OP, Venuganti VVK. Zein Microneedles for Transcutaneous Vaccine Delivery: Fabrication, Characterization, and in Vivo Evaluation Using Ovalbumin as the Model Antigen. ACS OMEGA 2017; 2:1321-1332. [PMID: 30023631 PMCID: PMC6044761 DOI: 10.1021/acsomega.7b00343] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 05/28/2023]
Abstract
Transcutaneous antigen administration provides an alternative to invasive syringe injections. The objective of this study was to investigate the feasibility of fabrication and antigen delivery using microneedles made from corn protein, zein. Micromolding technique was used to cast cone-shaped zein microneedles (ZMNs). The insertion of ZMNs and the delivery of the model antigen, ovalbumin (OVA), into the skin was confirmed by histological examination and confocal microscopy. In addition, a significantly (p < 0.05) lower bacterial skin penetration was observed after ZMN application compared with hypodermic syringe application. OVA coated on ZMNs was stable after storage under ambient and refrigerator conditions. Transcutaneous immunization studies showed significantly (p < 0.001) greater antibody titers (total IgG, IgG1, and IgG2a) after the application of OVA-coated ZMNs and OVA intradermal injection compared with the control group. Taken together, antigen-coated ZMNs can be developed for transcutaneous vaccine delivery.
Collapse
Affiliation(s)
- Shubhmita Bhatnagar
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Shameerpet, Hyderabad 500078, Telangana, India
| | | | - Onkar Prakash Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Shameerpet, Hyderabad 500078, Telangana, India
| | - Venkata Vamsi Krishna Venuganti
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Shameerpet, Hyderabad 500078, Telangana, India
| |
Collapse
|
28
|
Ali AA, McCrudden CM, McCaffrey J, McBride JW, Cole G, Dunne NJ, Robson T, Kissenpfennig A, Donnelly RF, McCarthy HO. DNA vaccination for cervical cancer; a novel technology platform of RALA mediated gene delivery via polymeric microneedles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 13:921-932. [DOI: 10.1016/j.nano.2016.11.019] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 10/11/2016] [Accepted: 11/30/2016] [Indexed: 11/30/2022]
|
29
|
Zeng Q, Gammon JM, Tostanoski LH, Chiu YC, Jewell CM. In Vivo Expansion of Melanoma-Specific T Cells Using Microneedle Arrays Coated with Immune-Polyelectrolyte Multilayers. ACS Biomater Sci Eng 2016; 3:195-205. [PMID: 28286864 PMCID: PMC5338335 DOI: 10.1021/acsbiomaterials.6b00414] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 09/01/2016] [Indexed: 02/07/2023]
Abstract
![]()
Microneedles
(MNs) are micron-scale polymeric or metallic structures
that offer distinct advantages for vaccines by efficiently targeting
skin-resident immune cells, eliminating injection-associated pain,
and improving patient compliance. These advantages, along with recent
studies showing therapeutic benefits achieved using traditional intradermal
injections in human cancer patients, suggest MN delivery might enhance
cancer vaccines and immunotherapies. We recently developed a new class
of polyelectrolyte multilayers based on the self-assembly of model
peptide antigens and molecular toll-like receptor agonists (TLRa)
into ultrathin, conformal coatings. Here, we reasoned that these immune
polyelectrolyte multilayers (iPEMs) might be a useful platform for
assembling cancer vaccine components on MN arrays for intradermal
delivery from these substrates. Using conserved human melanoma antigens
and a potent TLRa vaccine adjuvant, CpG, we show that iPEMs can be
assembled on MNs in an automated fashion. These films, prepared with
up to 128 layers, are approximately 200 nm thick but provide cancer
vaccine cargo loading >225 μg/cm2. In cell culture,
iPEM cargo released from MNs is internalized by primary dendritic
cells, promotes activation of these cells, and expands T cells during
coculture. In mice, application of iPEM-coated MNs results in the
codelivery of tumor antigen and CpG through the skin, expanding tumor-specific
T cells during initial MN applications and resulting in larger memory
recall responses during a subsequent booster MN application. This
study support MNs coated with PEMs built from tumor vaccine components
as a well-defined, modular system for generating tumor-specific immune
responses, enabling new approaches that can be explored in combination
with checkpoint blockade or other combination cancer therapies.
Collapse
Affiliation(s)
- Qin Zeng
- Fischell Department of Bioengineering, University of Maryland, College Park , 8228 Paint Branch Drive, 2212 Jeong H. Kim Building, College Park, Maryland 20742, United States
| | - Joshua M Gammon
- Fischell Department of Bioengineering, University of Maryland, College Park , 8228 Paint Branch Drive, 2212 Jeong H. Kim Building, College Park, Maryland 20742, United States
| | - Lisa H Tostanoski
- Fischell Department of Bioengineering, University of Maryland, College Park , 8228 Paint Branch Drive, 2212 Jeong H. Kim Building, College Park, Maryland 20742, United States
| | - Yu-Chieh Chiu
- Fischell Department of Bioengineering, University of Maryland, College Park , 8228 Paint Branch Drive, 2212 Jeong H. Kim Building, College Park, Maryland 20742, United States
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, 8228 Paint Branch Drive, 2212 Jeong H. Kim Building, College Park, Maryland 20742, United States; Department of Microbiology and Immunology, University of Maryland Medical School, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, Maryland 21201, United States; Marlene and Stewart Greenebaum Cancer Center, 22 S. Greene Street, Suite N9E17, Baltimore, Maryland 21201, United States
| |
Collapse
|
30
|
Zhao X, Birchall JC, Coulman SA, Tatovic D, Singh RK, Wen L, Wong FS, Dayan CM, Hanna SJ. Microneedle delivery of autoantigen for immunotherapy in type 1 diabetes. J Control Release 2016; 223:178-187. [DOI: 10.1016/j.jconrel.2015.12.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 12/23/2015] [Indexed: 11/24/2022]
|
31
|
Rejinold NS, Shin JH, Seok HY, Kim YC. Biomedical applications of microneedles in therapeutics: recent advancements and implications in drug delivery. Expert Opin Drug Deliv 2015; 13:109-31. [DOI: 10.1517/17425247.2016.1115835] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
32
|
van der Maaden K, Luttge R, Vos PJ, Bouwstra J, Kersten G, Ploemen I. Microneedle-based drug and vaccine delivery via nanoporous microneedle arrays. Drug Deliv Transl Res 2015; 5:397-406. [PMID: 26044672 PMCID: PMC4529475 DOI: 10.1007/s13346-015-0238-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the literature, several types of microneedles have been extensively described. However, porous microneedle arrays only received minimal attention. Hence, only little is known about drug delivery via these microneedles. However, porous microneedle arrays may have potential for future microneedle-based drug and vaccine delivery and could be a valuable addition to the other microneedle-based drug delivery approaches. To gain more insight into porous microneedle technologies, the scientific and patent literature is reviewed, and we focus on the possibilities and constraints of porous microneedle technologies for dermal drug delivery. Furthermore, we show preliminary data with commercially available porous microneedles and describe future directions in this field of research.
Collapse
|