1
|
Pahal S, Huang F, Singh P, Sharma N, Pham HP, Tran TBT, Sakhrie A, Akbaba H, Duc Nguyen T. Enhancing vaccine stability in transdermal microneedle platforms. Drug Deliv Transl Res 2025:10.1007/s13346-025-01854-4. [PMID: 40240731 DOI: 10.1007/s13346-025-01854-4] [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] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
Micron-scale needles, so-called microneedles (MNs) offer a minimally invasive, nearly painless, and user-friendly method for effective intradermal immunization. Maintaining the stability of antigens and therapeutics is the primary challenge in producing vaccine or drug-loaded MNs. The manufacturing of MNs patches involves processes at ambient or higher temperatures and various physio-mechanical stresses that can impact the therapeutic efficacy of sensitive biologics or vaccines. Therefore, it is crucial to develop techniques that safeguard vaccines and other biological payloads within MNs. Despite growing research interest in deploying MNs as an efficient tool for delivering vaccines, there is no comprehensive review that integrates the strategies and efforts to preserve the thermostability of vaccine payloads to ensure compatibility with MNs fabrication. The discussion delves into various physical and chemical approaches for stabilizing antigens in vaccine formulations, which are subsequently integrated into the MNs matrix. The primary focus is to comprehensively examine the challenges associated with the translation of thermostable vaccine MNs for clinical applications while considering a safe, cost-effective approach with a regulatory roadmap. The recent cutting-edge advances facilitating flexible and scalable manufacturing of stabilized MNs patches have been emphasized. In conclusion, the ability to stabilize vaccines and therapeutics for MNs applications could bolster the effectiveness, safety and user-compliance for various drugs and vaccines, potentially offering a substantial impact on global public health.
Collapse
Affiliation(s)
- Suman Pahal
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| | - Feifei Huang
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA
| | - Parbeen Singh
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Nidhi Sharma
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Hoang-Phuc Pham
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA
| | - Thi Bao Tram Tran
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Aseno Sakhrie
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Hasan Akbaba
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Ege University, Izmir, 35100, Turkey
| | - Thanh Duc Nguyen
- Institute of Materials Science, Polymer Program, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
- Department of Mechanical Engineering, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
2
|
Chakraborty C, Bhattacharya M, Lee SS. Current Status of Microneedle Array Technology for Therapeutic Delivery: From Bench to Clinic. Mol Biotechnol 2024; 66:3415-3437. [PMID: 37987985 DOI: 10.1007/s12033-023-00961-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/23/2023] [Indexed: 11/22/2023]
Abstract
In recent years, microneedle (MN) patches have emerged as an alternative technology for transdermal delivery of various drugs, therapeutics proteins, and vaccines. Therefore, there is an urgent need to understand the status of MN-based therapeutics. The article aims to illustrate the current status of microneedle array technology for therapeutic delivery through a comprehensive review. However, the PubMed search was performed to understand the MN's therapeutics delivery status. At the same time, the search shows the number no of publications on MN is increasing (63). The search was performed with the keywords "Coated microneedle," "Hollow microneedle," "Dissolvable microneedle," and "Hydrogel microneedle," which also shows increasing trend. Similarly, the article highlighted the application of different microneedle arrays for treating different diseases. The article also illustrated the current status of different phases of MN-based therapeutics clinical trials. It discusses the delivery of different therapeutic molecules, such as drug molecule delivery, using microneedle array technology. The approach mainly discusses the delivery of different therapeutic molecules. The leading pharmaceutical companies that produce the microneedle array for therapeutic purposes have also been discussed. Finally, we discussed the limitations and future prospects of this technology.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal, 700126, India.
| | - Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore, Odisha, 756020, India
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, Gangwon-do, 24252, Republic of Korea
| |
Collapse
|
3
|
Nguyen TD, Nguyen TH, Vo VT, Nguyen TQ. Panoramic review on polymeric microneedle arrays for clinical applications. Biomed Microdevices 2024; 26:41. [PMID: 39312013 DOI: 10.1007/s10544-024-00724-z] [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] [Accepted: 09/12/2024] [Indexed: 11/01/2024]
Abstract
Transdermal drug delivery (TDD) has significantly advanced medical practice in recent years due to its ability to prevent the degradation of substances in the gastrointestinal tract and avoid hepatic metabolism. Among different available approaches, microneedle arrays (MNAs) technology represents a fascinating delivery tool for enhancing TDD by penetrating the stratum corneum painless and minimally invasive for delivering antibacterial, antifungal, and antiviral medications. Polymeric MNAs are extensively utilized among many available materials due to their biodegradability, biocompatibility, and low toxicity. Therefore, this review provides a comprehensive discussion of polymeric MNAs, starting with understanding stratum corneum and developing MNA technology. Furthermore, the engineering concepts, fundamental considerations, challenges, and future perspectives of polymeric MNAs in clinical applications are properly outlined, offering a comprehensive and unique overview of polymeric MNAs and their potential for a broad spectrum of clinical applications.
Collapse
Affiliation(s)
- Tien Dat Nguyen
- School of Biomedical Engineering, International University, Ho Chi Minh City, 700000 HCMC, Viet Nam
- Vietnam National University, Ho Chi Minh City, 700000 HCMC, Vietnam
| | - Thi-Hiep Nguyen
- School of Biomedical Engineering, International University, Ho Chi Minh City, 700000 HCMC, Viet Nam
- Vietnam National University, Ho Chi Minh City, 700000 HCMC, Vietnam
| | - Van Toi Vo
- School of Biomedical Engineering, International University, Ho Chi Minh City, 700000 HCMC, Viet Nam
- Vietnam National University, Ho Chi Minh City, 700000 HCMC, Vietnam
| | - Thanh-Qua Nguyen
- School of Biomedical Engineering, International University, Ho Chi Minh City, 700000 HCMC, Viet Nam.
- Vietnam National University, Ho Chi Minh City, 700000 HCMC, Vietnam.
| |
Collapse
|
4
|
Battah B, Shbibe L, Ahmad O, Soukkarieh C, Al Okla SM, Chianese T, Rosati L, Vora LK, Zhao L, Marrazzo A, Ferrari M, Li L, Donnelly RF, Zanetti S, Mazzarello V, Donadu MG. Juniperus oxycedrus L. ssp. Essential Oil Microneedles: A Promising Antimicrobial and Wound Healing Activity. Pharmaceuticals (Basel) 2023; 17:40. [PMID: 38256874 PMCID: PMC10821373 DOI: 10.3390/ph17010040] [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: 11/20/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/24/2024] Open
Abstract
The use of essential oil (EO) in treating infected wounds is still challenging. A lot of effort has been made to make such an application more convenient. Recently, microneedles (MNDs) have been considered as a smart dermal delivery system to overcome the poor absorption and distribution, low bioavailability, and skin penetration of some drugs. The aim of our study is to evaluate the wound healing activity of juniper-EO-loaded MNDs (EO MNDs) against wounds with bacterial and fungal infection. The Polyvinylpyrrolidone (PVP) MNDs were prepared using the gel-filled mold technique and loaded with juniper EO. In vivo models were created and wounds on rats were infected with two clinically isolated bacterial strains Pseudomonas aeruginosa and Staphylococcus aureus. Furthermore, Candida albicans was used to mimic fungal infection and juniper EO MNDs were tested. The obtained results showed an improvement in wound healing which started from the third day after application of the juniper EO MNDs, and at the sixth day post-infection, the treated wounds were significantly smaller than untreated wounds. A complete healing was shown by the 12th day after infection. Furthermore, our cytotoxicity results showed a cytotoxic effect of juniper EO MNDs on epithelial cells, which explained the faster wound healing in rats. Our study showed that juniper EO MNDs represent a novel strategy in EO delivery with minimal invasion. Juniper EO MNDs demonstrated significant antimicrobial activity against both the bacterial strains Pseudomonas aeruginosa and Staphylococcus aureus and against one fungal strain, Candida albicans. Finally, application of juniper EO MNDs exerted promising activity in the treatment and healing of wound infection.
Collapse
Affiliation(s)
- Basem Battah
- Department of Biochemistry and Microbiology, Faculty of Pharmacy, Antioch Syrian Private University, Maaret Saidnaya 22734, Syria
| | - Lama Shbibe
- Faculty of Science, Damascus University, Damascus P.O. Box 30621, Syria; (L.S.); (C.S.)
| | - Osama Ahmad
- Faculty of Medicine, Syrian Private University (SPU), Daraa International Highway, Damascus 36822, Syria;
- Faculty of Medicine, Kalamoon University, Damascus 222, Syria
| | - Chadi Soukkarieh
- Faculty of Science, Damascus University, Damascus P.O. Box 30621, Syria; (L.S.); (C.S.)
| | - Souad Mahmoud Al Okla
- College of Medicine and Health Sciences, National University of Science, Sohar 321, Oman;
| | - Teresa Chianese
- Department of Biology, University Federico II, Via Cintia 21, 80126 Napoli, Italy; (T.C.); (L.R.)
| | - Luigi Rosati
- Department of Biology, University Federico II, Via Cintia 21, 80126 Napoli, Italy; (T.C.); (L.R.)
| | - Lalitkumar K. Vora
- Medical Biology Centre, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (L.K.V.); (L.Z.); (L.L.); (R.F.D.)
| | - Li Zhao
- Medical Biology Centre, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (L.K.V.); (L.Z.); (L.L.); (R.F.D.)
| | - Alessandra Marrazzo
- Hospital Pharmacy, Azienda Ospedaliero Universitaria di Sassari, 07100 Sassari, Italy;
| | - Marco Ferrari
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Via dell’Istria, 65, 34137 Trieste, Italy;
| | - Linlin Li
- Medical Biology Centre, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (L.K.V.); (L.Z.); (L.L.); (R.F.D.)
| | - Ryan F. Donnelly
- Medical Biology Centre, School of Pharmacy, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK; (L.K.V.); (L.Z.); (L.L.); (R.F.D.)
| | - Stefania Zanetti
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (V.M.)
| | - Vittorio Mazzarello
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (V.M.)
| | - Matthew Gavino Donadu
- Scuola di Specializzazione in Farmacia Ospedaliera, Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy;
- Hospital Pharmacy, Giovanni Paolo II Hospital, ASL Gallura, 07026 Olbia, Italy
| |
Collapse
|
5
|
Park CO, Kim HL, Park JW. Microneedle Transdermal Drug Delivery Systems for Allergen-Specific Immunotherapy, Skin Disease Treatment, and Vaccine Development. Yonsei Med J 2022; 63:881-891. [PMID: 36168240 PMCID: PMC9520048 DOI: 10.3349/ymj.2022.0092] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/05/2022] [Accepted: 08/14/2022] [Indexed: 11/27/2022] Open
Abstract
Transdermal drug delivery systems (TDDSs) overcome the hurdle of an intact skin barrier by penetrating the skin to allow molecules through. These systems reduce side effects associated with conventional hypodermic needles. Here, we introduce novel microneedle (MN) TDDSs that enhance drug delivery by creating micron-sized pores across the skin. Many MN TDDSs designed to deliver a diverse array of therapeutics, including allergen-specific immunotherapy, skin disease treatments, and vaccines, are under pre-clinical and clinical trials. Although epicutaneous approaches are emerging as new options for treating food allergy in many clinical trials, MN TDDSs could provide a more efficient and convenient route to deliver macromolecules. Furthermore, MN TDDSs may allow for safe vaccine delivery without permanent scars. MN TDDSs are a major emerging strategy for delivering novel vaccines and treatments for diseases, including skin diseases, allergic diseases, and so on.
Collapse
Affiliation(s)
- Chang Ook Park
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Korea
| | - Hye Li Kim
- Department of Dermatology, Yonsei University College of Medicine, Seoul, Korea
| | - Jung-Won Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
- Institute of Allergy, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
6
|
Gallovic MD, Junkins RD, Sandor AM, Pena ES, Sample CJ, Mason AK, Arwood LC, Sahm RA, Bachelder EM, Ainslie KM, Sempowski GD, Ting JPY. STING agonist-containing microparticles improve seasonal influenza vaccine efficacy and durability in ferrets over standard adjuvant. J Control Release 2022; 347:356-368. [PMID: 35569585 PMCID: PMC10136936 DOI: 10.1016/j.jconrel.2022.05.017] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 04/18/2022] [Accepted: 05/08/2022] [Indexed: 12/13/2022]
Abstract
The current pandemic highlights the need for effective vaccines against respiratory viruses. An ideal vaccine should induce robust and long-lasting responses with high manufacturing scalability. We use an adjuvant comprised of a Stimulator of Interferon Genes (STING) agonist incorporated in a scalable microparticle platform to achieve durable protection against the influenza virus. This formulation overcomes the challenges presented by the cytosolic localization of STING and the hydrophilicity of its agonists. We evaluated a monoaxial formulation of polymeric acetalated dextran microparticles (MPs) to deliver the STING agonist cyclic GMP-AMP (cGAMP) which achieved >10× dose-sparing effects compared to other published work. Efficacy was evaluated in ferrets, a larger animal model of choice for influenza vaccines. cGAMP MPs with recombinant hemagglutinin reduced viral shedding and improved vaccine outcomes compared to a seasonal influenza vaccine. Importantly, sustained protection against a lethal influenza infection was detected a year after a single dose of the vaccine-adjuvant.
Collapse
Affiliation(s)
- Matthew D Gallovic
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Robert D Junkins
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Adam M Sandor
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Erik S Pena
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Christopher J Sample
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Ariel K Mason
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Leslee C Arwood
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Rebecca A Sahm
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Eric M Bachelder
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kristy M Ainslie
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, USA
| | - Gregory D Sempowski
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Jenny P-Y Ting
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Institute for Inflammatory Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Center for Translational Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
7
|
Hassan J, Haigh C, Ahmed T, Uddin MJ, Das DB. Potential of Microneedle Systems for COVID-19 Vaccination: Current Trends and Challenges. Pharmaceutics 2022; 14:1066. [PMID: 35631652 PMCID: PMC9144974 DOI: 10.3390/pharmaceutics14051066] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/27/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
To prevent the coronavirus disease 2019 (COVID-19) pandemic and aid restoration to prepandemic normality, global mass vaccination is urgently needed. Inducing herd immunity through mass vaccination has proven to be a highly effective strategy for preventing the spread of many infectious diseases, which protects the most vulnerable population groups that are unable to develop immunity, such as people with immunodeficiencies or weakened immune systems due to underlying medical or debilitating conditions. In achieving global outreach, the maintenance of the vaccine potency, transportation, and needle waste generation become major issues. Moreover, needle phobia and vaccine hesitancy act as hurdles to successful mass vaccination. The use of dissolvable microneedles for COVID-19 vaccination could act as a major paradigm shift in attaining the desired goal to vaccinate billions in the shortest time possible. In addressing these points, we discuss the potential of the use of dissolvable microneedles for COVID-19 vaccination based on the current literature.
Collapse
Affiliation(s)
- Jasmin Hassan
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
| | - Charlotte Haigh
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK;
| | - Tanvir Ahmed
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
| | - Md Jasim Uddin
- Drug Delivery & Therapeutics Lab, Dhaka 1212, Bangladesh; (J.H.); (T.A.)
- Faculty of Engineering and Science, University of Greenwich, Chatham Maritime, Kent ME4 4TB, UK
- Department of Pharmacy, Brac University, 66 Mohakhali, Dhaka 1212, Bangladesh
| | - Diganta B. Das
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK;
| |
Collapse
|
8
|
Coffey JW, van der Burg NMD, Rananakomol T, Ng HI, Fernando GJP, Kendall MAF. An Ultrahigh‐Density Microneedle Array for Skin Vaccination: Inducing Epidermal Cell Death by Increasing Microneedle Density Enhances Total IgG and IgG1 Immune Responses. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202100151] [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] Open
Affiliation(s)
- Jacob W. Coffey
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
- Department of Chemical Engineering David H. Koch Institute for Integrative Cancer Research Massachusetts Institute of Technology Cambridge MA 02139 USA
- Division of Gastroenterology Brigham and Women's Hospital Harvard Medical School Boston MA 02115 USA
- Department of Microbiology and Immunology Peter Doherty Institute for Infection and Immunology University of Melbourne Melbourne VIC 3000 Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of Queensland St Lucia QLD 4072 Australia
| | - Nicole M. D. van der Burg
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of Queensland St Lucia QLD 4072 Australia
| | - Thippayawan Rananakomol
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
| | - Hwee-Ing Ng
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
| | - Germain J. P. Fernando
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
- The University of Queensland School of Chemistry and Molecular Biosciences Brisbane QLD 4072 Australia
- Vaxxas Pty Translational Research Institute Woolloongabba QLD 4102 Australia
| | - Mark A. F. Kendall
- The Delivery of Drugs and Genes Group (D2G) Australian Institute for Bioengineering and Nanotechnology University of Queensland St. Lucia QLD 4072 Australia
- ARC Centre of Excellence in Convergent Bio‐Nano Science and Technology The University of Queensland St Lucia QLD 4072 Australia
- The University of Queensland School of Chemistry and Molecular Biosciences Brisbane QLD 4072 Australia
| |
Collapse
|
9
|
Niu Y, Zheng W, Wang S, Zhao Q, Wei L, Zhao Y, Wang B, Ju Y, Wang F. Efficacy and safety of acupuncture for patients with pseudomyopia: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2022; 101:e28859. [PMID: 35147133 PMCID: PMC8830834 DOI: 10.1097/md.0000000000028859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Myopia are common health problems that people experience in daily life. Pediatric myopia has become a major international public health concern that has a negative impact on physical, mental health of patients, and quality of life. Currently, there is no cure available. Press needle as an adjuvant therapy is currently undergoing clinical trials in different medical centers. However, no relevant systematic review or meta-analysis has been designed to evaluate the effects of press needle patching on early myopia. There is also a lack of systematic evaluation and analysis of acupoints and thumbtack needle. METHODS We will electronically search Medline, Embase, PubMed, Web of Science, the Cochrane Central Register of Controlled Trials, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, Chinese Scientific Journal Database, and Wan-Fang Database from their inception to October 2021. In addition, we will manually retrieve other resources including the reference lists of identified publications, conference articles, and gray literature. The clinical randomized controlled trials or quasi-randomized controlled trials related to press needle for the treatment of early myopia will be included in the study. The language is limited to Chinese and English. Research selection, data extraction, and research quality assessment will be independently completed by 2 researchers. Data were synthesized by using afixed effect model or random effect model depend on the heterogeneity test. The total effective rate was the primary outcomes. RevMan V.5.3 statistical software will be used for meta-analysis. If it is not appropriate for a meta-analysis, then a descriptive analysis will be conducted. Data synthesis will use the risk ratio and the standardized or weighted average difference of continuous data to represent the results. RESULTS This study will analyze the clinical effective rate, pesudomyopia outcomes, quality of life, improvement of clinical symptoms of pesudomyopia, and validity of thumbtack needle for patients with pesudomyopia. CONCLUSION This systematic review will provide evidence to judge whether thumbtack needle is an effective intervention for patients with early myopia. SYSTEMATIC REVIEW REGISTRATION PROSPERO, CRD42021243151.
Collapse
Affiliation(s)
- Ye Niu
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun City, Jilin Province, China
| | - Wei Zheng
- Affiliated Hospital of Changchun University of Chinese Medicine, 1478 Gongnong Road, Changchun City, Jilin Province, China
| | - Shan Wang
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun City, Jilin Province, China
| | - Qi Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun City, Jilin Province, China
| | - Lijuan Wei
- Affiliated Hospital of Changchun University of Chinese Medicine, 1478 Gongnong Road, Changchun City, Jilin Province, China
| | - Yang Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun City, Jilin Province, China
| | - Bo Wang
- Affiliated Hospital of Changchun University of Chinese Medicine, 1478 Gongnong Road, Changchun City, Jilin Province, China
| | - Yuan Ju
- Affiliated Hospital of Changchun University of Chinese Medicine, 1478 Gongnong Road, Changchun City, Jilin Province, China
| | - Fuchun Wang
- Department of Acupuncture, The Affiliated Hospital of Changchun University of Chinese Medicine, 1478 Gongnong Road, Changchun City, Jilin Province, China
| |
Collapse
|
10
|
Nguyen TT, Nguyen TTD, Tran NMA, Nguyen HT, Vo GV. Microneedles enable the development of skin-targeted vaccines against coronaviruses and influenza viruses. Pharm Dev Technol 2021; 27:83-94. [PMID: 34802372 DOI: 10.1080/10837450.2021.2008967] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Throughout the COVID-19 pandemic, many have seriously worried that the plus burden of seasonal influenza that might create a destructive scenario, resulting in overwhelmed healthcare capacities and onwards loss of life. Many efforts to develop a safe and efficacious vaccine to prevent infection by coronavirus and influenza, highlight the importance of vaccination to combat infectious pathogens. While vaccines are traditionally given as injections into the muscle, microneedle (MN) patches designed to precisely deliver cargos into the cutaneous microenvironment, rich in immune cells, provide a noninvasive and self-applicable vaccination approach, reducing overall costs and improving access to vaccines in places with limited supply. The current review aimed to highlight advances in research on the development of MNs-mediated cutaneous vaccine delivery. Concluding remarks and challenges on MNs-based skin immunization are also provided to contribute to the rational development of safe and effective MN-delivered vaccines against these emerging infectious diseases.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Pharmacy, Ho Chi Minh City University of Technology (HUTECH), Ho Chi Minh City, Vietnam
| | - Thi Thuy Dung Nguyen
- Faculty of Environmental and Food Engineering, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Nguyen-Minh-An Tran
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Huy Truong Nguyen
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University - Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, Vietnam.,Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University - Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, Vietnam.,Vietnam National University - Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, Vietnam
| |
Collapse
|
11
|
Cordeiro AS, Patil-Sen Y, Shivkumar M, Patel R, Khedr A, Elsawy MA. Nanovaccine Delivery Approaches and Advanced Delivery Systems for the Prevention of Viral Infections: From Development to Clinical Application. Pharmaceutics 2021; 13:2091. [PMID: 34959372 PMCID: PMC8707864 DOI: 10.3390/pharmaceutics13122091] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Viral infections causing pandemics and chronic diseases are the main culprits implicated in devastating global clinical and socioeconomic impacts, as clearly manifested during the current COVID-19 pandemic. Immunoprophylaxis via mass immunisation with vaccines has been shown to be an efficient strategy to control such viral infections, with the successful and recently accelerated development of different types of vaccines, thanks to the advanced biotechnological techniques involved in the upstream and downstream processing of these products. However, there is still much work to be done for the improvement of efficacy and safety when it comes to the choice of delivery systems, formulations, dosage form and route of administration, which are not only crucial for immunisation effectiveness, but also for vaccine stability, dose frequency, patient convenience and logistics for mass immunisation. In this review, we discuss the main vaccine delivery systems and associated challenges, as well as the recent success in developing nanomaterials-based and advanced delivery systems to tackle these challenges. Manufacturing and regulatory requirements for the development of these systems for successful clinical and marketing authorisation were also considered. Here, we comprehensively review nanovaccines from development to clinical application, which will be relevant to vaccine developers, regulators, and clinicians.
Collapse
Affiliation(s)
- Ana Sara Cordeiro
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Yogita Patil-Sen
- Wrightington, Wigan and Leigh Teaching Hospitals NHS Foundation Trust, National Health Service, Wigan WN6 0SZ, UK;
| | - Maitreyi Shivkumar
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| | - Ronak Patel
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK;
| | - Abdulwahhab Khedr
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Mohamed A. Elsawy
- Leicester Institute for Pharmaceutical Innovation, Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK; (A.S.C.); (M.S.); (A.K.)
| |
Collapse
|
12
|
Vazquez-Pagan A, Schultz-Cherry S. Serological Responses to Influenza Vaccination during Pregnancy. Microorganisms 2021; 9:microorganisms9112305. [PMID: 34835431 PMCID: PMC8619416 DOI: 10.3390/microorganisms9112305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/25/2021] [Accepted: 11/03/2021] [Indexed: 11/25/2022] Open
Abstract
Pregnant women, newborns, and infants under six months old are at the highest risk of developing severe and even fatal influenza. This risk is compounded by the inability to vaccinate infants under six months, highlighting the importance of vertically transferred immunity. This review identifies novel insights that have emerged from recent studies using animal models of pregnancy and vaccination. We also discuss the knowledge obtained using existing clinical trials that have evaluated influenza-specific serological responses in pregnant women and how these responses may impact early life immunity. We delineate the mechanisms involved in transferring specific maternal antibodies and discuss the consequences for early life immunity. Most importantly, we highlight the need for continued research using pregnant animal models and the inclusion of pregnant women, a commonly neglected population, when evaluating novel vaccine platforms to better serve and treat communicable diseases.
Collapse
Affiliation(s)
- Ana Vazquez-Pagan
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Stacey Schultz-Cherry
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Correspondence:
| |
Collapse
|
13
|
Wan Y, Gupta V, Bird C, Pullagurla SR, Fahey P, Forster A, Volkin DB, Joshi SB. Formulation Development and Improved Stability of a Combination Measles and Rubella Live-Viral Vaccine Dried for Use in the Nanopatch TM Microneedle Delivery System. Hum Vaccin Immunother 2021; 17:2501-2516. [PMID: 33957843 PMCID: PMC8475600 DOI: 10.1080/21645515.2021.1887692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/18/2021] [Accepted: 02/03/2021] [Indexed: 12/27/2022] Open
Abstract
Measles (Me) and rubella (Ru) viral diseases are targeted for elimination by ensuring a high level of vaccination coverage worldwide. Less costly, more convenient MeRu vaccine delivery systems should improve global vaccine coverage, especially in low - and middle - income countries (LMICs). In this work, we examine formulating a live, attenuated Me and Ru combination viral vaccine with Nanopatch™, a solid polymer micro-projection array for intradermal delivery. First, high throughput, qPCR-based viral infectivity and genome assays were established to enable formulation development to stabilize Me and Ru in a scaled-down, custom-built evaporative drying system to mimic the Nanopatch™ vaccine coating process. Second, excipient screening and optimization studies identified virus stabilizers for use during the drying process and upon storage in the dried state. Finally, a series of real-time and accelerated stability studies identified eight candidate formulations that met a target thermal stability criterion for live vaccines (<1 log10 loss after 1 week storage at 37°C). Compared to -80°C control samples, the top candidate formulations resulted in minimal viral infectivity titer losses after storage at 2-8°C for 6 months (i.e., <0.1 log10 for Me, and ~0.4 log10 for Ru). After storage at 25°C over 6 months, ~0.3-0.5 and ~1.0-1.4 log10 titer losses were observed for Me and Ru, respectively, enabling the rank-ordering of the stability of candidate formulations. These results are discussed in the context of future formulation challenges for developing microneedle-based dosage forms containing stabilized live, attenuated viral vaccines for use in LMICs.
Collapse
Affiliation(s)
- Ying Wan
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Vineet Gupta
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Christopher Bird
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Swathi R. Pullagurla
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Paul Fahey
- Vaxxas Pty Ltd, Translational Research Institute, Brisbane, QLD, Australia
| | - Angus Forster
- Vaxxas Pty Ltd, Translational Research Institute, Brisbane, QLD, Australia
| | - David B. Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| | - Sangeeta B. Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
14
|
Shin JH, Lee JH, Jeong SD, Noh JY, Lee HW, Song CS, Kim YC. C-di-GMP with influenza vaccine showed enhanced and shifted immune responses in microneedle vaccination in the skin. Drug Deliv Transl Res 2021; 10:815-825. [PMID: 32141036 DOI: 10.1007/s13346-020-00728-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A microneedle is a biomedical device which consists of multiple micron scale needles. It is widely used in various fields to deliver drugs and vaccines to the skin effectively. However, when considering improved vaccine efficacy in microneedle vaccination, it is important to find an appropriate adjuvant that is able to be used in transdermal delivery. Herein, we demonstrated the applicability of c-di-GMP, which is a stimulator of interferon genes (STING) agonist, as an adjuvant for influenza microneedle vaccination. Thus, 2 and 10 μg of GMP with the influenza vaccine were coated onto a microneedle, and then, BALB/c mice were immunized with the coated microneedle to investigate the immunogenicity and protection efficacy of the influenza microneedle vaccination. As a result, the adjuvant groups had an enhanced IgG response, IgG subtypes and HI titer compared to the vaccine only group. In addition to the humoral immunity, the use of an adjuvant has also been shown to improve the cellular immune response. In a challenge study, adjuvant groups had a 100% survival rate and rapid weight recovery. Taken together, this study confirms that GMP is an effective adjuvant for influenza microneedle vaccination. Graphical abstract.
Collapse
Affiliation(s)
- Ju-Hyung Shin
- Department of Chemical and Biomolecular engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji-Ho Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul, Republic of Korea
| | - Seong Dong Jeong
- Department of Chemical and Biomolecular engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Jin-Yong Noh
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul, Republic of Korea
| | - Hyo Won Lee
- Department of Chemical and Biomolecular engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Chang-Seon Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul, Republic of Korea.
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
| |
Collapse
|
15
|
Novel formulations and drug delivery systems to administer biological solids. Adv Drug Deliv Rev 2021; 172:183-210. [PMID: 33705873 DOI: 10.1016/j.addr.2021.02.011] [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: 11/30/2020] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Recent advances in formulation sciences have expanded the previously limited design space for biological modalities, including peptide, protein, and vaccine products. At the same time, the discovery and application of new modalities, such as cellular therapies and gene therapies, have presented formidable challenges to formulation scientists. We explore these challenges and highlight the opportunities to overcome them through the development of novel formulations and drug delivery systems as biological solids. We review the current progress in both industry and academic laboratories, and we provide expert perspectives in those settings. Formulation scientists have made a tremendous effort to accommodate the needs of these novel delivery routes. These include stability-preserving formulations and dehydration processes as well as dosing regimes and dosage forms that improve patient compliance.
Collapse
|
16
|
Korkmaz E, Balmert SC, Sumpter TL, Carey CD, Erdos G, Falo LD. Microarray patches enable the development of skin-targeted vaccines against COVID-19. Adv Drug Deliv Rev 2021; 171:164-186. [PMID: 33539853 PMCID: PMC8060128 DOI: 10.1016/j.addr.2021.01.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/10/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022]
Abstract
The COVID-19 pandemic is a serious threat to global health and the global economy. The ongoing race to develop a safe and efficacious vaccine to prevent infection by SARS-CoV-2, the causative agent for COVID-19, highlights the importance of vaccination to combat infectious pathogens. The highly accessible cutaneous microenvironment is an ideal target for vaccination since the skin harbors a high density of antigen-presenting cells and immune accessory cells with broad innate immune functions. Microarray patches (MAPs) are an attractive intracutaneous biocargo delivery system that enables safe, reproducible, and controlled administration of vaccine components (antigens, with or without adjuvants) to defined skin microenvironments. This review describes the structure of the SARS-CoV-2 virus and relevant antigenic targets for vaccination, summarizes key concepts of skin immunobiology in the context of prophylactic immunization, and presents an overview of MAP-mediated cutaneous vaccine delivery. Concluding remarks on MAP-based skin immunization are provided to contribute to the rational development of safe and effective MAP-delivered vaccines against emerging infectious diseases, including COVID-19.
Collapse
Affiliation(s)
- Emrullah Korkmaz
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | - Stephen C Balmert
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Tina L Sumpter
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Cara Donahue Carey
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Geza Erdos
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Louis D Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; The McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
17
|
Vassilieva EV, Li S, Korniychuk H, Taylor DM, Wang S, Prausnitz MR, Compans RW. cGAMP/Saponin Adjuvant Combination Improves Protective Response to Influenza Vaccination by Microneedle Patch in an Aged Mouse Model. Front Immunol 2021; 11:583251. [PMID: 33603732 PMCID: PMC7884748 DOI: 10.3389/fimmu.2020.583251] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
Current strategies for improving protective response to influenza vaccines during immunosenescence do not adequately protect individuals over 65 years of age. Here, we used an aged mouse model to investigate the potential of co-delivery of influenza vaccine with the recently identified combination of a saponin adjuvant Quil-A and an activator of the STING pathway, 2’3 cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) via dissolving microneedle patches (MNPs) applied to skin. We demonstrate that synergy between the two adjuvant components is observed after their incorporation with H1N1 vaccine into MNPs as revealed by analysis of the immune responses in adult mice. Aged 21-month-old mice were found to be completely protected against live influenza challenge after vaccination with the MNPs adjuvanted with the Quil-A/cGAMP combination (5 µg each) and demonstrated significantly reduced morbidity compared to the observed responses in these mice vaccinated with unadjuvanted MNPs. Analysis of the lung lysates of the surviving aged mice post challenge revealed the lowest level of residual inflammation in the adjuvanted groups. We conclude that combining influenza vaccine with a STING pathway activator and saponin-based adjuvant in MNPs is a novel option for skin vaccination of the immunosenescent population, which is at high risk for influenza.
Collapse
Affiliation(s)
- Elena V Vassilieva
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Song Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Heorhiy Korniychuk
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Dahnide M Taylor
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Shelly Wang
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Richard W Compans
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
18
|
Vora LK, Moffatt K, Tekko IA, Paredes AJ, Volpe-Zanutto F, Mishra D, Peng K, Raj Singh Thakur R, Donnelly RF. Microneedle array systems for long-acting drug delivery. Eur J Pharm Biopharm 2021; 159:44-76. [DOI: 10.1016/j.ejpb.2020.12.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/25/2020] [Accepted: 12/08/2020] [Indexed: 12/31/2022]
|
19
|
Hossain MK, Ahmed T, Bhusal P, Subedi RK, Salahshoori I, Soltani M, Hassanzadeganroudsari M. Microneedle Systems for Vaccine Delivery: the story so far. Expert Rev Vaccines 2021; 19:1153-1166. [PMID: 33427523 DOI: 10.1080/14760584.2020.1874928] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Vaccine delivery via a microneedle (MN) system has been identified as a potential alternative to conventional vaccine delivery. MN can be self-administered, is pain-free and is capable of producing superior immunogenicity. Over the last few decades, significant research has been carried out in this area, and this review aims to provide a comprehensive picture on the progress of this delivery platform. AREAS COVERED This review highlights the potential role of skin as a vaccine delivery route using a microneedle system, examines recent advancements in microneedle fabrication techniques, and provides an update on potential preclinical and clinical studies on vaccine delivery through microneedle systems against various infectious diseases. Articles for the review study were searched electronically in PubMed, Google, Google Scholar, and Science Direct using specific keywords to cover the scope of the article. The advanced search strategy was employed to identify the most relevant articles. EXPERT OPINION A significant number of MN mediated vaccine candidates have shown promising results in preclinical and clinical trials. The recent emergence of cleanroom free, 3D or additive manufacturing of MN systems and stability, together with the dose-sparing capacity of the Nanopatch® system, have made this platform, commercially, highly lucrative.
Collapse
Affiliation(s)
- Md Kamal Hossain
- Institute for Health and Sport, Victoria University , Melbourne, VIC, Australia
| | - Taksim Ahmed
- Leslie Dan Faculty of Pharmacy, University of Toronto , Toronto, Ontario, Canada
| | - Prabhat Bhusal
- School of Pharmacy, University of Otago , Dunedin New Zealand
| | | | - Iman Salahshoori
- Department of Chemical Engineering, Science and Research Branch, Islamic Azad University , Tehran, Iran
| | - M Soltani
- Department of Mechanical Engineering, K. N. Toosi University of Technology , Tehran, Iran.,Department of Electrical and Computer Engineering, Faculty of Engineering, School of Optometry and Vision Science, Faculty of Science, University of Waterloo , Waterloo, Ontario, Canada.,Centre for Biotechnology and Bioengineering (CBB), University of Waterloo , Waterloo, Ontario, Canada.,Advanced Bioengineering Initiative Center, Multidisciplinary International Complex, K. N. Toosi University of Technology , Tehran, Iran
| | - Majid Hassanzadeganroudsari
- Institute for Health and Sport, Victoria University , Melbourne, VIC, Australia.,Department of Chemical Engineering, Science and Research Branch, Islamic Azad University , Tehran, Iran
| |
Collapse
|
20
|
Lima AF, Amado IR, Pires LR. Poly(d,l-lactide- co-glycolide) (PLGA) Nanoparticles Loaded with Proteolipid Protein (PLP)-Exploring a New Administration Route. Polymers (Basel) 2020; 12:polym12123063. [PMID: 33371329 PMCID: PMC7767393 DOI: 10.3390/polym12123063] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
The administration of specific antigens is being explored as a mean to re-establish immunological tolerance, namely in the context of multiple sclerosis (MS). PLP139-151 is a peptide of the myelin's most abundant protein, proteolipid protein (PLP), which has been identified as a potent tolerogenic molecule in MS. This work explored the encapsulation of the peptide into poly(lactide-co-glycolide) nanoparticles and its subsequent incorporation into polymeric microneedle patches to achieve efficient delivery of the nanoparticles and the peptide into the skin, a highly immune-active organ. Different poly(d,l-lactide-co-glycolide) (PLGA) formulations were tested and found to be stable and to sustain a freeze-drying process. The presence of trehalose in the nanoparticle suspension limited the increase in nanoparticle size after freeze-drying. It was shown that rhodamine can be loaded in PLGA nanoparticles and these into poly(vinyl alcohol)-poly(vinyl pyrrolidone) microneedles, yielding fluorescently labelled structures. The incorporation of PLP into the PLGA nanoparticles resulted in nanoparticles in a size range of 200 µm and an encapsulation efficiency above 20%. The release of PLP from the nanoparticles occurred in the first hours after incubation in physiological media. When loading the nanoparticles into microneedle patches, structures were obtained with 550 µm height and 180 µm diameter. The release of PLP was detected in PLP-PLGA.H20 nanoparticles when in physiological media. Overall, the results show that this strategy can be explored to integrate a new antigen-specific therapy in the context of multiple sclerosis, providing minimally invasive administration of PLP-loaded nanoparticles into the skin.
Collapse
|
21
|
Ramalheiro A, Paris JL, Silva BFB, Pires LR. Rapidly dissolving microneedles for the delivery of cubosome-like liquid crystalline nanoparticles with sustained release of rapamycin. Int J Pharm 2020; 591:119942. [PMID: 33039495 DOI: 10.1016/j.ijpharm.2020.119942] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 09/15/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022]
Abstract
In this study, we developed a system for the transdermal delivery and controlled release of the hydrophobic immunosuppressive drug rapamycin, foreseeing an application in psoriasis treatment. To do so, rapamycin was encapsulated in phytantriol-based cubosome-like liquid crystalline nanoparticles stabilized with pluronic F127. The final mass percent composition of the lipid nanoparticles was 0.25% phytantriol, 0.1% pluronic F127, 4.75% ethanol and 94.9% water. These particles showed a rapamycin encapsulation efficiency above 95% and a sustained in vitrodrug release profile throughout 14 days. Subsequently the rapamycin-carrying particles were incorporated into rapidly dissolving microneedle patches composed of a polymeric matrix of poly(vinylpyrrolidone) and poly(vinyl alcohol). Confocal microscopy allowed to infer the preferential distribution of the cubosome-like particles at the tip and baseplate of the microneedles. The fabricated microneedles showed successful piercing and deposition of the loaded cubosome-like particles on a skin-mimicking agarose gel. Finally, the rapamycin-loaded cubosome-like particles showed antiproliferative activity in natural killer cells in vitro. The results here presented show the potential of the developed system to deliver cubosome-like particles into the skin and promote the sustained release of rapamycin in the context of immunomodulation.
Collapse
Affiliation(s)
- Ana Ramalheiro
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal; Instituto Superior Técnico, Lisboa, Portugal
| | - Juan L Paris
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - Bruno F B Silva
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal.
| | - Liliana R Pires
- International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal.
| |
Collapse
|
22
|
Beaver JT, Mills LK, Swieboda D, Lelutiu N, Esser ES, Antao OQ, Scountzou E, Williams DT, Papaioannou N, Littauer EQ, Romanyuk A, Compans RW, Prausnitz MR, Skountzou I. Cutaneous vaccination ameliorates Zika virus-induced neuro-ocular pathology via reduction of anti-ganglioside antibodies. Hum Vaccin Immunother 2020; 16:2072-2091. [PMID: 32758106 PMCID: PMC7553697 DOI: 10.1080/21645515.2020.1775460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Zika virus (ZIKV) causes moderate to severe neuro-ocular sequelae, with symptoms ranging from conjunctivitis to Guillain-Barré Syndrome (GBS). Despite the international threat ZIKV poses, no licensed vaccine exists. As ZIKV and DENV are closely related, antibodies against one virus have demonstrated the ability to enhance the other. To examine if vaccination can confer robust, long-term protection against ZIKV, preventing neuro-ocular pathology and long-term inflammation in immune-privileged compartments, BALB/c mice received two doses of unadjuvanted inactivated whole ZIKV vaccine (ZVIP) intramuscularly (IM) or cutaneously with dissolving microneedle patches (MNP). MNP immunization induced significantly higher B and T cell responses compared to IM vaccination, resulting in increased antibody titers with greater avidity for ZPIV as well as increased numbers of IFN-γ, TNF-α, IL- and IL-4 secreting T cells. When compared to IM vaccination, antibodies generated by cutaneous vaccination demonstrated greater neutralization activity, increased cross-reactivity with Asian and African lineage ZIKV strains (PRVABC59, FLR, and MR766) and Dengue virus (DENV) serotypes, limited ADE, and lower reactivity to GBS-associated gangliosides. MNP vaccination effectively controlled viremia and inflammation, preventing neuro-ocular pathology. Conversely, IM vaccination exacerbated ocular pathology, resulting in uncontrolled, long-term inflammation. Importantly, neuro-ocular pathology correlated with anti-ganglioside antibodies implicated in demyelination and GBS. This study highlights the importance of longevity studies in ZIKV immunization, and the need of exploring alternative vaccination platforms to improve the quality of vaccine-induced immune responses.
Collapse
Affiliation(s)
- Jacob T Beaver
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Lisa K Mills
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Dominika Swieboda
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Nadia Lelutiu
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Edward S Esser
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Olivia Q Antao
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | | | - Dahnide T Williams
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Nikolaos Papaioannou
- Faculty of Veterinary Medicine, Laboratory of Pathologic Anatomy, Aristotle University of Thessaloniki , Greece
| | - Elizabeth Q Littauer
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Andrey Romanyuk
- Department of Biomedical Engineering, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology , Atlanta, GA, USA
| | - Richard W Compans
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Mark R Prausnitz
- Department of Biomedical Engineering, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology , Atlanta, GA, USA
| | - Ioanna Skountzou
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| |
Collapse
|
23
|
Nguyen TT, Oh Y, Kim Y, Shin Y, Baek SK, Park JH. Progress in microneedle array patch (MAP) for vaccine delivery. Hum Vaccin Immunother 2020; 17:316-327. [PMID: 32667239 PMCID: PMC7872046 DOI: 10.1080/21645515.2020.1767997] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A microneedle array patch (MAP) has been developed as a new delivery system for vaccines. Preclinical and clinical trials with a vaccine MAP showed improved stability, safety, and immunological efficacy compared to conventional vaccine administration. Various vaccines can be delivered with a MAP. Currently, microneedle manufacturers can mass-produce pharmaceutical MAP and cosmetic MAP and this mass-production system can be adapted to produce a vaccine MAP. Clinical trials with a vaccine MAP have shown comparable efficacy with conventional administration, and discussions about regulations for a vaccine MAP are underway. However, there are concerns of reasonable cost, mass production, efficacy, and safety standards that meet FDA approval, as well as the need for feedback regarding the best method of administration. Currently, microneedles have been studied for the delivery of many kinds of vaccines, and preclinical and clinical studies of vaccine microneedles are in progress. For the foreseeable future, some vaccines will continue to be administered with syringes and needles while the use of a vaccine MAP continues to be improved because of the advantages of less pain, self-administration, improved stability, convenience, and safety.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Pharmacy, Ho Chi Minh City University of Technology-HUTECH , Ho Chi Minh, Vietnam
| | - Yujeong Oh
- Department of BioNano Technology, Gachon BioNano Research Institute, Gachon University , Seongnam, Republic of Korea
| | - Yunseo Kim
- Department of BioNano Technology, Gachon BioNano Research Institute, Gachon University , Seongnam, Republic of Korea
| | - Yura Shin
- Department of BioNano Technology, Gachon BioNano Research Institute, Gachon University , Seongnam, Republic of Korea
| | - Seung-Ki Baek
- QuadMedicine R&D Centre, QuadMedicine Inc , Seongnam, Republic of Korea
| | - Jung-Hwan Park
- Department of BioNano Technology, Gachon BioNano Research Institute, Gachon University , Seongnam, Republic of Korea
| |
Collapse
|
24
|
Shields CW, Wang LLW, Evans MA, Mitragotri S. Materials for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901633. [PMID: 31250498 DOI: 10.1002/adma.201901633] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/17/2019] [Indexed: 05/20/2023]
Abstract
Breakthroughs in materials engineering have accelerated the progress of immunotherapy in preclinical studies. The interplay of chemistry and materials has resulted in improved loading, targeting, and release of immunomodulatory agents. An overview of the materials that are used to enable or improve the success of immunotherapies in preclinical studies is presented, from immunosuppressive to proinflammatory strategies, with particular emphasis on technologies poised for clinical translation. The materials are organized based on their characteristic length scale, whereby the enabling feature of each technology is organized by the structure of that material. For example, the mechanisms by which i) nanoscale materials can improve targeting and infiltration of immunomodulatory payloads into tissues and cells, ii) microscale materials can facilitate cell-mediated transport and serve as artificial antigen-presenting cells, and iii) macroscale materials can form the basis of artificial microenvironments to promote cell infiltration and reprogramming are discussed. As a step toward establishing a set of design rules for future immunotherapies, materials that intrinsically activate or suppress the immune system are reviewed. Finally, a brief outlook on the trajectory of these systems and how they may be improved to address unsolved challenges in cancer, infectious diseases, and autoimmunity is presented.
Collapse
Affiliation(s)
- C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael A Evans
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
25
|
Liu Y, Wen Z, Carrion R, Nunneley J, Staples H, Ticer A, Patterson JL, Compans RW, Ye L, Yang C. Intradermal Immunization of EBOV VLPs in Guinea Pigs Induces Broader Antibody Responses Against GP Than Intramuscular Injection. Front Microbiol 2020; 11:304. [PMID: 32174901 PMCID: PMC7056717 DOI: 10.3389/fmicb.2020.00304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 02/11/2020] [Indexed: 11/20/2022] Open
Abstract
Ebolavirus (EBOV) infection in humans causes severe hemorrhagic fevers with high mortality rates that range from 30 to 80% as shown in different outbreaks. Thus the development of safe and efficacious EBOV vaccines remains an important goal for biomedical research. We have shown in early studies that immunization with insect cell-produced EBOV virus-like particles (VLPs) is able to induce protect vaccinated mice against lethal EBOV challenge. In the present study, we investigated immune responses induced by Ebola VLPs via two different routes, intramuscular and intradermal immunizations, in guinea pigs. Analyses of antibody responses revealed that similar levels of total IgG antibodies against the EBOV glycoprotein (GP) were induced by the two different immunization methods. However, further characterization showed that the EBOV GP-specific antibodies induced by intramuscular immunization were mainly of the IgG2 subtype whereas both IgG1 and IgG2 antibodies against EBOV GP were induced by intradermal immunization. In contrast, antibody responses against the EBOV matrix protein VP40 induced by intramuscular or intradermal immunizations exhibited similar IgG1 and IgG2 profiles. More interestingly, we found that the sites that the IgG1 antibodies induced by intradermal immunizations bind to in GP are different from those that bind to the IgG2 antibodies induced by intramuscular immunization. Further analyses revealed that sera from all vaccinated guinea pigs exhibited neutralizing activity against Ebola GP-mediated HIV pseudovirion infection at high levels. Moreover, all EBOV VLP-vaccinated guinea pigs survived the challenge by a high dose (1000 pfu) of guinea pig-adapted EBOV, while all control guinea pigs immunized with irrelevant VLPs succumbed to the challenge. The induction of both IgG1 and IgG2 antibody responses that recognized broader sites in GP by intradermal immunization of EBOV VLPs indicates that this approach may represent a more advantageous route of vaccination against virus infection.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Food Nutrition and Safety, Institute of Health Biotechnology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Department of Microbiology and Immunology and Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, United States
| | - Zhiyuan Wen
- Department of Microbiology and Immunology and Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, United States
- Harbin Veterinary Research Institute, Harbin, China
| | - Ricardo Carrion
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jerritt Nunneley
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Hilary Staples
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Anysha Ticer
- Texas Biomedical Research Institute, San Antonio, TX, United States
| | | | - Richard W. Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, United States
| | - Ling Ye
- Department of Microbiology and Immunology and Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, United States
| | - Chinglai Yang
- Department of Microbiology and Immunology and Emory Vaccine Center, School of Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
26
|
Badizadegan K, Goodson JL, Rota PA, Thompson KM. The potential role of using vaccine patches to induce immunity: platform and pathways to innovation and commercialization. Expert Rev Vaccines 2020; 19:175-194. [PMID: 32182145 PMCID: PMC7814398 DOI: 10.1080/14760584.2020.1732215] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/12/2020] [Indexed: 01/14/2023]
Abstract
Introduction: In the last two decades, the evidence related to using vaccine patches with multiple short projections (≤1 mm) to deliver vaccines through the skin increased significantly and demonstrated their potential as an innovative delivery platform.Areas covered: We review the vaccine patch literature published in English as of 1 March 2019, as well as available information from key stakeholders related to vaccine patches as a platform. We identify key research topics related to basic and translational science on skin physical properties and immunobiology, patch development, and vaccine manufacturing.Expert opinion: Currently, vaccine patch developers continue to address some basic science and other platform issues in the context of developing a potential vaccine patch presentation for an existing or new vaccine. Additional clinical data and manufacturing experience could shift the balance toward incentivizing existing vaccine manufactures to further explore the use of vaccine patches to deliver their products. Incentives for innovation of vaccine patches differ for developed and developing countries, which will necessitate different strategies (e.g. public-private partnerships, push, or pull mechanisms) to support the basic and applied research needed to ensure a strong evidence base and to overcome translational barriers for vaccine patches as a delivery platform.
Collapse
Affiliation(s)
| | - James L Goodson
- Global Immunization Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Paul A Rota
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | |
Collapse
|
27
|
Vassilieva EV, Taylor DW, Compans RW. Combination of STING Pathway Agonist With Saponin Is an Effective Adjuvant in Immunosenescent Mice. Front Immunol 2019; 10:3006. [PMID: 31921219 PMCID: PMC6935580 DOI: 10.3389/fimmu.2019.03006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022] Open
Abstract
There is an urgent need to improve protective responses to influenza vaccination in the elderly population, which is at especially high risk for adverse outcomes from influenza infection. Currently available inactivated vaccines provide limited protection, even when a 4-fold higher dose of the vaccine is administered. Adjuvants are often added to vaccines to boost protective efficacy. Here we describe a novel combination of an activator of the STING pathway, 2′,3′-cyclic guanosine monophosphate–adenosine monophosphate (cGAMP) with a saponin adjuvant, that we found to be highly effective in boosting protective immunity from vaccination in an aged mouse model. Using this combination with a subunit influenza vaccine, we observed that survival of vaccinated 20 month-old mice after lethal challenge increased from 0 to 20% with unadjuvanted vaccine to 80–100%, depending on the vaccination route. Compared to unadjuvanted vaccine, the levels of vaccine-specific IgG and IgG2a increased by almost two orders of magnitude as early as 2 weeks after a single immunization with the adjuvanted formulation. By analyzing phosphorylation of interferon regulatory factor 3 (IRF3) in cell culture, we provide evidence that the saponin component increases access of exogenous cGAMP to the intracellular STING pathway. Our findings suggest that combining a STING activator with a saponin-based adjuvant increases the effectiveness of influenza vaccine in aged hosts, without having to increase dose or perform additional vaccinations. This study reports a novel adjuvant combination that (a) is more effective than current methods of boosting vaccine efficacy, (b) can be used to enhance efficacy of licensed influenza vaccines, and (c) results in effective protection using a single vaccine dose.
Collapse
Affiliation(s)
- Elena V Vassilieva
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Dahnide W Taylor
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Richard W Compans
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
28
|
Uppu DSSM, Turvey ME, Sharif ARM, Bidet K, He Y, Ho V, Tambe AD, Lescar J, Tan EY, Fink K, Chen J, Hammond PT. Temporal release of a three-component protein subunit vaccine from polymer multilayers. J Control Release 2019; 317:130-141. [PMID: 31756392 DOI: 10.1016/j.jconrel.2019.11.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 10/14/2019] [Accepted: 11/18/2019] [Indexed: 12/11/2022]
Abstract
Sustained antigen and adjuvant availability have been shown to improve antiviral immune responses following vaccination. Transcutaneous delivery of vaccines using microneedles has also shown promise and may be particularly relevant for mosquito-borne viruses. We aim to combine these traits to create a three-component Protein Subunit vaccine on Microneedle Arrays (PSMNs) for transcutaneous delivery using layer-by-layer (LbL) assembly. Polymer multilayer thin films were generated to co-deliver a model combination of three chemically distinct vaccine components, a dengue virus Envelope protein Domain III (EDIII) subunit antigen and two adjuvants, a double-stranded RNA (Poly (inosinic:cytidylic acid) (PolyI:C)) and an amphiphilic hexapeptide, Pam3CSK4. Following application of PSMNs to the skin, implanted thin films facilitated sustained and temporal release of individual vaccine components from polymer multilayers. By modulating LbL composition and architecture, component release profiles in the skin could be independently tuned to allow release of adjuvants and antigen from days up to two weeks. Uptake of antigen and adjuvant from implanted vaccine films by antigen-presenting cells was demonstrated using in vivo mouse and ex vivo human skin models. Overall, we believe that such modular vaccine strategies offer design principles for enhancing the immunogenicity of protein subunit vaccines.
Collapse
Affiliation(s)
- Divakara S S M Uppu
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore
| | - Michelle E Turvey
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore
| | - Abdul Rahim Mohammed Sharif
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore
| | - Katell Bidet
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore
| | - Yanpu He
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Victor Ho
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore
| | - Anagha D Tambe
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore
| | - Julien Lescar
- School of Biological Sciences, Nanyang Technology University, Singapore
| | - Ern Yu Tan
- Department of General Surgery, Tan Tock Seng Hospital, Singapore
| | - Katja Fink
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore
| | - Jianzhu Chen
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Paula T Hammond
- Interdisciplinary Research Group in Infectious Diseases, Singapore-MIT Alliance for Research and Technology, Singapore; Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
29
|
Microprojection arrays applied to skin generate mechanical stress, induce an inflammatory transcriptome and cell death, and improve vaccine-induced immune responses. NPJ Vaccines 2019; 4:41. [PMID: 31632742 PMCID: PMC6789026 DOI: 10.1038/s41541-019-0134-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/11/2019] [Indexed: 11/08/2022] Open
Abstract
Chemical adjuvants are typically used to improve immune responses induced by immunisation with protein antigens. Here we demonstrate an approach to enhance immune responses that does not require chemical adjuvants. We applied microprojection arrays to the skin, producing a range of controlled mechanical energy to invoke localised inflammation, while administering influenza split virus protein antigen. We used validated computational modelling methods to identify links between mechanical stress and energy generated within the skin strata and resultant cell death. We compared induced immune responses to those induced by needle-based intradermal antigen delivery and used a systems biology approach to examine the nature of the induced inflammatory response, and correlated this with markers of cell stress and death. Increasing the microprojection array application energy and the addition of QS-21 adjuvant were each associated with enhanced antibody response to delivered antigen and with induction of gene transcriptions associated with TNF and NF-κB signalling pathways. We concluded that microprojection intradermal antigen delivery inducing controlled local cell death could potentially replace chemical adjuvants to enhance the immune response to protein antigen. Adjuvants play an essential function in vaccine formulations by boosting immune responses to the delivered antigen. Mark A. F. Kendall and colleagues investigate the efficacy of vaccine delivered intradermally via NanopatchTM—a 16 mm2 ultra-high-density array of 100 micron needles. Systems analysis comparisons demonstrate that depending on the force applied, the NanopatchTM triggers skin transcriptomic changes similar to that elicited by the adjuvant QS-21, including evidence of localised cell death and inflammatory gene expression. Flu vaccine delivered by NanopatchTM elicits anti-hemagglutinin titers equivalent to that observed with conventional intradermal syringe delivery of vaccine plus adjuvant. Sterile mechanical stress elicited by the application of microneedles might therefore be a viable replacement to the use of conventional needles.
Collapse
|
30
|
Jeong HR, Park S, Park JH, Bae JY, Kim GY, Baek SK, Park MS, Park JH. Preparation of H1N1 microneedles by a low-temperature process without a stabilizer. Eur J Pharm Biopharm 2019; 143:1-7. [DOI: 10.1016/j.ejpb.2019.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 01/18/2023]
|
31
|
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: 118] [Impact Index Per Article: 19.7] [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
|
32
|
Cheeseman HM, Day S, McFarlane LR, Fleck S, Miller A, Cole T, Sousa-Santos N, Cope A, Cizmeci D, Tolazzi M, Hwekwete E, Hannaman D, Kratochvil S, McKay PF, Chung AW, Kent SJ, Cook A, Scarlatti G, Abraham S, Combadiere B, McCormack S, Lewis DJ, Shattock RJ. Combined Skin and Muscle DNA Priming Provides Enhanced Humoral Responses to a Human Immunodeficency Virus Type 1 Clade C Envelope Vaccine. Hum Gene Ther 2019; 29:1011-1028. [PMID: 30027768 PMCID: PMC6214652 DOI: 10.1089/hum.2018.075] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Intradermal (i.d.) and intramuscular (i.m.) injections when administered with or without electroporation (EP) have the potential to tailor the immune response to DNA vaccination. This Phase I randomized controlled clinical trial in human immunodeficiency virus type 1–negative volunteers investigated whether the site and mode of DNA vaccination influences the quality of induced cellular and humoral immune responses following the DNA priming phase and subsequent protein boost with recombinant clade C CN54 gp140. A strategy of concurrent i.d. and i.m. DNA immunizations administered with or without EP was adopted. Subtle differences were observed in the shaping of vaccine-induced virus-specific CD4+ and CD8+ T cell–mediated immune responses between groups receiving: i.d.EP + i.m., i.d. + i.m.EP, and i.d.EP + i.m.EP regimens. The DNA priming phase induced 100% seroconversion in all of the groups. A single, non-adjuvanted protein boost induced a rapid and profound increase in binding antibodies in all groups, with a trend for higher responses in i.d.EP + i.m.EP. The magnitude of antigen-specific binding immunoglobulin G correlated with neutralization of closely matched clade C 93MW965 virus and Fc-dimer receptor binding (FcγRIIa and FcγRIIIa). These results offer new perspectives on the use of combined skin and muscle DNA immunization in priming humoral and cellular responses to recombinant protein.
Collapse
Affiliation(s)
- Hannah Mary Cheeseman
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Suzanne Day
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Leon Robert McFarlane
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sue Fleck
- 2 Medical Research Council Clinical Trials Unit at UCL, University College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Aleisha Miller
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Tom Cole
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Nelson Sousa-Santos
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Alethea Cope
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Deniz Cizmeci
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Monica Tolazzi
- 4 Viral Evolution and Transmission Unit, Division of Immunology, Transplant and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Edith Hwekwete
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Drew Hannaman
- 5 Ichor Medical Systems, Inc., San Diego, California; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sven Kratochvil
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Paul Francis McKay
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Amy W Chung
- 6 Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, and UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Stephen J Kent
- 6 Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, and UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France .,7 ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, Australia; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France .,8 Melbourne Sexual Health Centre, Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University , Melbourne, Australia; and UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Adrian Cook
- 2 Medical Research Council Clinical Trials Unit at UCL, University College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Gabriella Scarlatti
- 4 Viral Evolution and Transmission Unit, Division of Immunology, Transplant and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sonya Abraham
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Behazine Combadiere
- 9 Sorbonne Universités, UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sheena McCormack
- 2 Medical Research Council Clinical Trials Unit at UCL, University College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - David John Lewis
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Robin John Shattock
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| |
Collapse
|
33
|
Meyer BK, Kendall MAF, Williams DM, Bett AJ, Dubey S, Gentzel RC, Casimiro D, Forster A, Corbett H, Crichton M, Baker SB, Evans RK, Bhambhani A. Immune response and reactogenicity of an unadjuvanted intradermally delivered human papillomavirus vaccine using a first generation Nanopatch™ in rhesus macaques: An exploratory, pre-clinical feasibility assessment. Vaccine X 2019; 2:100030. [PMID: 31384745 PMCID: PMC6668242 DOI: 10.1016/j.jvacx.2019.100030] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 05/21/2019] [Accepted: 06/07/2019] [Indexed: 12/26/2022] Open
Abstract
The human papillomavirus (HPV) 9-valent, recombinant vaccine (Gardasil™9) helps protect young adults (males and females) against anogenital cancers and genital warts caused by certain HPV genotypes (ref. Gardasil™9 insert). This vaccine is administered intramuscularly (IM). The aim of this study was to determine preclinically whether intradermal (ID) vaccination with an unadjuvanted 9-valent recombinant HPV vaccine using a first-generation ID delivery device, the Nanopatch™, could enhance vaccine immunogenicity compared with the traditional ID route (Mantoux technique). IM injection of HPV VLPs formulated with Merck & Co., Inc., Kenilworth, NJ, USA Alum Adjuvant (MAA) were included in the rhesus study for comparison. The Nanopatch™ prototype contains a high-density array comprised of 10,000 microprojections/cm2, each 250 µm long. It was hypothesized the higher density array with shallower ID delivery may be superior to the Mantoux technique. To test this hypothesis, HPV VLPs without adjuvant were coated on the Nanopatch™, stability of the Nanopatch™ with unadjuvanted HPV VLPs were evaluated under accelerated conditions, skin delivery was verified using radiolabelled VLPs or FluoSpheres®, and the immune response and skin site reaction with the Nanopatch™ was evaluated in rhesus macaques. The immune response induced by Nanopatch™ administration, measured as HPV-specific binding antibodies, was similar to that induced using the Mantoux technique. It was also observed that a lower dose of unadjuvanted HPV VLPs delivered with the first-generation Nanopatch™ and applicator or Mantoux technique resulted in an immune response that was significantly lower compared to a higher-dose of alum adjuvanted HPV VLPs delivered IM in rhesus macaques. The study also indicated unadjuvanted HPV VLPs could be delivered with the first-generation Nanopatch™ and applicator to the skin in 15 s with a transfer efficiency of approximately 20%. This study is the first demonstration of patch administration in non-human primates with a vaccine composed of HPV VLPs.
Collapse
Affiliation(s)
- Brian K Meyer
- New Technologies, Vaccine Drug Product Development, Vaccine Process Research and Development, MRL, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Mark A F Kendall
- Delivery of Drugs and Genes Group (DG), Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland QLD 4072, Australia.,Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Brisbane, QLD 4102, Australia
| | - Donna M Williams
- New Technologies, Vaccine Drug Product Development, Vaccine Process Research and Development, MRL, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Andrew J Bett
- Infectious Disease and Vaccines, MRL, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Sheri Dubey
- Infectious Disease and Vaccines, MRL, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Renee C Gentzel
- Movement Disorders and Translation, MRL, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Danilo Casimiro
- Infectious Disease and Vaccines, MRL, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Angus Forster
- Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Brisbane, QLD 4102, Australia
| | - Holly Corbett
- Delivery of Drugs and Genes Group (DG), Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland QLD 4072, Australia
| | - Michael Crichton
- Delivery of Drugs and Genes Group (DG), Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland QLD 4072, Australia.,Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Brisbane, QLD 4102, Australia
| | - S Ben Baker
- Delivery of Drugs and Genes Group (DG), Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland QLD 4072, Australia.,Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Brisbane, QLD 4102, Australia
| | - Robert K Evans
- New Technologies, Vaccine Drug Product Development, Vaccine Process Research and Development, MRL, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| | - Akhilesh Bhambhani
- New Technologies, Vaccine Drug Product Development, Vaccine Process Research and Development, MRL, Merck & Co., Inc., 770 Sumneytown Pike, West Point, PA 19486, USA
| |
Collapse
|
34
|
Turvey ME, Uppu DS, Mohamed Sharif AR, Bidet K, Alonso S, Ooi EE, Hammond PT. Microneedle-based intradermal delivery of stabilized dengue virus. Bioeng Transl Med 2019; 4:e10127. [PMID: 31249877 PMCID: PMC6584444 DOI: 10.1002/btm2.10127] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/24/2019] [Accepted: 01/24/2019] [Indexed: 12/26/2022] Open
Abstract
Current live-attenuated dengue vaccines require strict cold chain storage. Methods to preserve dengue virus (DENV) viability, which enable vaccines to be transported and administered at ambient temperatures, will be decisive towards the implementation of affordable global vaccination schemes with broad immunization coverage in resource-limited areas. We have developed a microneedle (MN)-based vaccine platform for the stabilization and intradermal delivery of live DENV from minimally invasive skin patches. Dengue virus-stabilized microneedle arrays (VSMN) were fabricated using saccharide-based formulation of virus and could be stored dry at ambient temperature up to 3 weeks with maintained virus viability. Following intradermal vaccination, VSMN-delivered DENV was shown to elicit strong neutralizing antibody responses and protection from viral challenge, comparable to that of the conventional liquid vaccine administered subcutaneously. This work supports the potential for MN-based dengue vaccine technology and the progression towards cold chain-independence. Dengue virus can be stabilized using saccharide-based formulations and coated on microneedle array vaccine patches for storage in dry state with preserved viability at ambient temperature (VSMN; virus-stabilized microneedle arrays).
Collapse
Affiliation(s)
- Michelle E. Turvey
- Infectious Diseases IRGSingapore‐MIT Alliance for Research and TechnologySingapore
| | - Divakara S.S.M. Uppu
- Infectious Diseases IRGSingapore‐MIT Alliance for Research and TechnologySingapore
| | | | - Katell Bidet
- Infectious Diseases IRGSingapore‐MIT Alliance for Research and TechnologySingapore
| | - Sylvie Alonso
- Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, Immunology ProgrammeLife Sciences Institute, National University of SingaporeSingapore
| | - Eng Eong Ooi
- Infectious Diseases IRGSingapore‐MIT Alliance for Research and TechnologySingapore
- Emerging Infectious DiseasesDuke‐NUS Graduate Medical SchoolSingapore
| | - Paula T. Hammond
- Infectious Diseases IRGSingapore‐MIT Alliance for Research and TechnologySingapore
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of TechnologyCambridgeMA
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA
| |
Collapse
|
35
|
Stable incorporation of GM-CSF into dissolvable microneedle patch improves skin vaccination against influenza. J Control Release 2018; 276:1-16. [PMID: 29496540 PMCID: PMC5967648 DOI: 10.1016/j.jconrel.2018.02.033] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/17/2018] [Accepted: 02/19/2018] [Indexed: 11/20/2022]
Abstract
The widely used influenza subunit vaccine would benefit from increased protection rates in vulnerable populations. Skin immunization by microneedle (MN) patch can increase vaccine immunogenicity, as well as increase vaccination coverage due to simplified administration. To further increase immunogenicity, we used granulocyte-macrophage colony stimulating factor (GM-CSF), an immunomodulatory cytokine already approved for skin cancer therapy and cancer support treatment. GM-CSF has been shown to be upregulated in skin following MN insertion. The GM-CSF-adjuvanted vaccine induced robust and long-lived antibody responses cross-reactive to homosubtypic and heterosubtypic influenza viruses. Addition of GM-CSF resulted in increased memory B cell persistence relative to groups given influenza vaccine alone and led to rapid lung viral clearance following lethal infection with homologous virus in the mouse model. Here we demonstrate that successful incorporation of the thermolabile cytokine GM-CSF into MN resulted in improved vaccine-induced protective immunity holding promise as a novel approach to improved influenza vaccination. To our knowledge, this is the first successful incorporation of a cytokine adjuvant into dissolvable MNs, thus advancing and diversifying the rapidly developing field of MN vaccination technology.
Collapse
|
36
|
Inactivated rotavirus vaccine by parenteral administration induces mucosal immunity in mice. Sci Rep 2018; 8:561. [PMID: 29330512 PMCID: PMC5766576 DOI: 10.1038/s41598-017-18973-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/19/2017] [Indexed: 01/10/2023] Open
Abstract
To improve the safety and efficacy of oral rotavirus vaccines, we developed an inactivated rotavirus vaccine (IRV) for parenteral administration. Since it remains unknown whether parenteral vaccination can induce mucosal immunity, we performed a comprehensive assessment of immune responses to IRV in mice with an adjuvant-free dissolving polymer MN patch or by alum-adjuvanted IM injection. We demonstrated that IRV induced the expression of the gut homing receptor LPAM-1 on T and B cells in spleen and mLN of vaccinated mice. MN patch IRV vaccination induced a slight Th1 phenotype while IM vaccination induced a balanced Th1/Th2 phenotype. In addition, a dose-sparing effect was seen for rotavirus-specific serum IgG and neutralizing activity for both vaccination routes. Our study is the first to show that parenterally administered IRV can induce mucosal immunity in the gut, in addition to strong serum antibody response, and is a promising candidate vaccine in achieving global immunization against rotavirus.
Collapse
|
37
|
Vassilieva EV, Wang S, Li S, Prausnitz MR, Compans RW. Skin immunization by microneedle patch overcomes statin-induced suppression of immune responses to influenza vaccine. Sci Rep 2017; 7:17855. [PMID: 29259264 PMCID: PMC5736694 DOI: 10.1038/s41598-017-18140-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/06/2017] [Indexed: 01/02/2023] Open
Abstract
Recent studies indicated that in elderly individuals, statin therapy is associated with a reduced response to influenza vaccination. The present study was designed to determine effects on the immune response to influenza vaccination induced by statin administration in a mouse model, and investigate potential approaches to improve the outcome of vaccination on the background of statin therapy. We fed middle aged BALB/c mice a high fat “western” diet (WD) alone or supplemented with atorvastatin (AT) for 14 weeks, and control mice were fed with the regular rodent diet. Mice were immunized with a single dose of subunit A/Brisbane/59/07 (H1N1) vaccine, either systemically or with dissolving microneedle patches (MNPs). We observed that a greater age-dependent decline in the hemagglutinin inhibition titers occurred in systemically-immunized mice than in MNP- immunized mice. AT dampened the antibody response in the animals vaccinated by either route of vaccine delivery. However, the MNP-vaccinated AT-treated animals had ~20 times higher total antibody levels to the influenza vaccine than the systemically vaccinated group one month postvaccination. We propose that microneedle vaccination against influenza provides an approach to ameliorate the immunosuppressive effect of statin therapy observed with systemic immunization.
Collapse
Affiliation(s)
- Elena V Vassilieva
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Shelly Wang
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, Georgia
| | - Song Li
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, Georgia
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, Georgia
| | - Richard W Compans
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, Georgia.
| |
Collapse
|
38
|
Lau S, Fei J, Liu H, Chen W, Liu R. Multilayered pyramidal dissolving microneedle patches with flexible pedestals for improving effective drug delivery. J Control Release 2017; 265:113-119. [DOI: 10.1016/j.jconrel.2016.08.031] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/23/2016] [Accepted: 08/25/2016] [Indexed: 11/17/2022]
|
39
|
Mistilis MJ, Joyce JC, Esser ES, Skountzou I, Compans RW, Bommarius AS, Prausnitz MR. Long-term stability of influenza vaccine in a dissolving microneedle patch. Drug Deliv Transl Res 2017; 7:195-205. [PMID: 26926241 DOI: 10.1007/s13346-016-0282-2] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study tested the hypothesis that optimized microneedle patch formulations can stabilize trivalent subunit influenza vaccine during long-term storage outside the cold chain and when exposed to potential stresses found during manufacturing and storage. Formulations containing combinations of trehalose/sucrose, sucrose/arginine, and arginine/heptagluconate were successful at retaining most or all vaccine activity during storage at 25 °C for up to 24 months as determined by ELISA assay. The best formulation of microneedle patches contained arginine/heptagluconate, which showed no significant loss of vaccine activity during the study. To validate these in vitro findings, mice were immunized using trivalent influenza vaccine stored in microneedle patches for more than 1 year at 25 °C, which elicited antibody titers greater than or equal to fresh liquid vaccine delivered by intradermal injection, indicating the retention of immunogenicity during storage. Finally, influenza vaccine in microneedle patches lost no significant activity during exposure to 60 °C for 4 months, multiple freeze-thaw cycles, or electron beam irradiation. We conclude that optimally formulated microneedle patches can retain influenza vaccine activity during extended storage outside the cold chain and during other environmental stresses, which suggests the possibility of microneedle patch storage on pharmacy shelves without refrigeration.
Collapse
Affiliation(s)
- Matthew J Mistilis
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive, Atlanta, GA, 30332-0100, USA
| | - Jessica C Joyce
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332-0100, USA
| | - E Stein Esser
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA
| | - Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA
| | - Richard W Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA
| | - Andreas S Bommarius
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive, Atlanta, GA, 30332-0100, USA
- School of Chemistry and Biochemistry, Georgia Institute of Technology, 901 Atlantic Drive, Atlanta, GA, 30332-0400, USA
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 311 Ferst Drive, Atlanta, GA, 30332-0100, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Tech and Emory University, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332-0100, USA.
| |
Collapse
|
40
|
Shin JH, Noh JY, Kim KH, Park JK, Lee JH, Jeong SD, Jung DY, Song CS, Kim YC. Effect of zymosan and poly (I:C) adjuvants on responses to microneedle immunization coated with whole inactivated influenza vaccine. J Control Release 2017; 265:83-92. [PMID: 28890214 DOI: 10.1016/j.jconrel.2017.09.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 12/14/2022]
Abstract
Microneedles are the micrometer size devices used for the delivery of vaccines and biotherapeutics. In order to increase the vaccine efficacy and reduce the antigen dose, there is a significant need to find some adjuvants for the microneedle vaccination. In this study, zymosan, which is the cell wall preparation of Saccharomyces cerevisiae, or poly (I:C) was coated on a microneedle with inactivated influenza virus, and then immunized into BALB/c mouse to determine the immunogenicity, protection and synergetic effect between two adjuvants. As a result, the group administered with zymosan and vaccine antigen showed significantly stronger IgG response, HI titer and IgG subtypes without any adverse effects, compared to the group immunized with the vaccine antigen alone. Also, there were enhanced cellular immune responses in the group received adjuvant with vaccine antigen. In addition, they showed superior protection and lung viral reduction against lethal viral challenge. Taken together, this study confirms that zymosan can be used as an immunostimulant for microneedle vaccination.
Collapse
Affiliation(s)
- Ju-Hyung Shin
- Department of Chemical and Biomolecular engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Jin-Yong Noh
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Kwon-Ho Kim
- Department of Chemical and Biomolecular engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Jae-Keun Park
- Viral Pathogenesis and Evolution Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ji-Ho Lee
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Seong Dong Jeong
- Department of Chemical and Biomolecular engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Dae-Yoon Jung
- Department of Chemical and Biomolecular engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea
| | - Chang-Seon Song
- Avian Disease Laboratory, College of Veterinary Medicine, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 305-701, Republic of Korea.
| |
Collapse
|
41
|
Schipper P, van der Maaden K, Groeneveld V, Ruigrok M, Romeijn S, Uleman S, Oomens C, Kersten G, Jiskoot W, Bouwstra J. Diphtheria toxoid and N -trimethyl chitosan layer-by-layer coated pH-sensitive microneedles induce potent immune responses upon dermal vaccination in mice. J Control Release 2017; 262:28-36. [DOI: 10.1016/j.jconrel.2017.07.017] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/02/2017] [Accepted: 07/10/2017] [Indexed: 01/18/2023]
|
42
|
Esser ES, Pulit-Penaloza JA, Kalluri H, McAllister D, Vassilieva EV, Littauer EQ, Lelutiu N, Prausnitz MR, Compans RW, Skountzou I. Microneedle patch delivery of influenza vaccine during pregnancy enhances maternal immune responses promoting survival and long-lasting passive immunity to offspring. Sci Rep 2017; 7:5705. [PMID: 28720851 PMCID: PMC5515933 DOI: 10.1038/s41598-017-05940-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 06/06/2017] [Indexed: 11/25/2022] Open
Abstract
Influenza virus causes life-threatening infections in pregnant women and their newborns. Immunization during pregnancy is the most effective means of preventing maternal and infant mortality/morbidity; however, influenza vaccination rates of pregnant women remain under 50%. Furthermore, the availability of vaccines in low-resource populations is limited. Skin immunization with microneedle patches (MN) is a novel and safe vaccination platform featuring thermostable vaccine formulations. Cold-chain independence and the potential for self-administration can expand influenza vaccination coverage in developing countries. In this study of pregnant BALB/c mice immunized with subunit H1N1 influenza vaccine, we demonstrate the advantage of skin vaccination over intramuscular delivery of a two-fold higher vaccine dose. MN vaccine induced superior humoral immune responses and conferred protective immunity against a lethal challenge dose of homologous influenza virus. Importantly, MN vaccination of mice at mid-gestation resulted in enhanced and long-lasting passive immunity of the offspring, measured by neutralizing antibody titers and survival rates after virus challenge. We conclude that skin vaccination using MN is a superior immunization approach with the potential to overcome immune tolerance observed in pregnancy, and lower vaccination costs through antigen dose-sparing, which is especially relevant in underserved countries.
Collapse
Affiliation(s)
- E Stein Esser
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Joanna A Pulit-Penaloza
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Haripriya Kalluri
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Devin McAllister
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Elena V Vassilieva
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Elizabeth Q Littauer
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Nadia Lelutiu
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Richard W Compans
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ioanna Skountzou
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
43
|
Hsueh KJ, Chen MC, Cheng LT, Lee JW, Chung WB, Chu CY. Transcutaneous immunization of Streptococcus suis bacterin using dissolving microneedles. Comp Immunol Microbiol Infect Dis 2016; 50:78-87. [PMID: 28131383 DOI: 10.1016/j.cimid.2016.12.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 11/14/2016] [Accepted: 12/08/2016] [Indexed: 11/17/2022]
Abstract
Vaccine delivery using microneedle (MN) patches is an easy, safe and painless alternative to traditional needle injections. In this study, we examined whether MN patches can enhance the efficacy of a Streptococcus suis serotype 2 (S. suis 2) vaccine in a mouse model. Results showed that MNs can reach 200-250μm into the skin, a depth beneficial for targeted delivery of antigens to antigen-presenting cells in the epidermis and dermis. Vaccination with prime-boost of MN induced higher levels of IgG2a antibody titer, T cell proliferation, and TH1 cytokines (IFN-γ and IL-12) as compared to intramuscular (IM) injection. In addition, single dose MN vaccination better protected mice against lethal challenge than IM vaccination. MN vaccination also conferred long-term IgG2a antibody against S. suis 2 bacteria presence for up to 7 months. Taken together, these data showed that vaccine delivery by MNs results in superior immune response and protection rate when compared to IM injections.
Collapse
Affiliation(s)
- Kai-Jen Hsueh
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, 91201 Taiwan; Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, 91201 Taiwan
| | - Mei-Chin Chen
- Department of Chemical Engineering, National Cheng Kung University, Tainan, 701 Taiwan
| | - Li-Ting Cheng
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, 91201 Taiwan
| | - Jai-Wei Lee
- Department of Tropical Agriculture and International Cooperation, National Pingtung University of Science and Technology, Pingtung, 91201 Taiwan
| | - Wen-Bin Chung
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, 91201 Taiwan
| | - Chun-Yen Chu
- Graduate Institute of Animal Vaccine Technology, College of Veterinary Medicine, National Pingtung University of Science and Technology, Pingtung, 91201 Taiwan.
| |
Collapse
|
44
|
Scavenging nucleic acid debris to combat autoimmunity and infectious disease. Proc Natl Acad Sci U S A 2016; 113:9728-33. [PMID: 27528673 DOI: 10.1073/pnas.1607011113] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Nucleic acid-containing debris released from dead and dying cells can be recognized as damage-associated molecular patterns (DAMPs) or pattern-associated molecular patterns (PAMPs) by the innate immune system. Inappropriate activation of the innate immune response can engender pathological inflammation and autoimmune disease. To combat such diseases, major efforts have been made to therapeutically target the pattern recognition receptors (PRRs) such as the Toll-like receptors (TLRs) that recognize such DAMPs and PAMPs, or the downstream effector molecules they engender, to limit inflammation. Unfortunately, such strategies can limit the ability of the immune system to combat infection. Previously, we demonstrated that nucleic acid-binding polymers can act as molecular scavengers and limit the ability of artificial nucleic acid ligands to activate PRRs. Herein, we demonstrate that nucleic acid scavengers (NASs) can limit pathological inflammation and nucleic acid-associated autoimmunity in lupus-prone mice. Moreover, we observe that such NASs do not limit an animal's ability to combat viral infection, but rather their administration improves survival when animals are challenged with lethal doses of influenza. These results indicate that molecules that scavenge extracellular nucleic acid debris represent potentially safer agents to control pathological inflammation associated with a wide range of autoimmune and infectious diseases.
Collapse
|
45
|
Esser ES, Romanyuk A, Vassilieva EV, Jacob J, Prausnitz MR, Compans RW, Skountzou I. Tetanus vaccination with a dissolving microneedle patch confers protective immune responses in pregnancy. J Control Release 2016; 236:47-56. [PMID: 27327766 DOI: 10.1016/j.jconrel.2016.06.026] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/24/2016] [Accepted: 06/16/2016] [Indexed: 11/25/2022]
Abstract
Maternal and neonatal tetanus claim tens of thousands lives every year in developing countries, but could be prevented by hygienic practices and improved immunization of pregnant women. This study tested the hypothesis that skin vaccination can overcome the immunologically transformed state of pregnancy and enhance protective immunity to tetanus in mothers and their newborns. To achieve this goal, we developed microneedle patches (MNPs) that efficiently delivered unadjuvanted tetanus toxoid to skin of pregnant mice and demonstrated that this route induced superior immune responses in female mice conferring 100% survival to tetanus toxin challenge when compared to intramuscular vaccination. Mice born to MNP-vaccinated mothers showed detectable tetanus-specific IgG antibodies up to 12weeks of age and complete protection to tetanus toxin challenge up at 6weeks of age. In contrast, none of the 6-week old mice born to intramuscularly vaccinated mothers survived challenge. Although pregnant mice vaccinated with unadjuvanted tetanus toxoid had 30% lower IgG and IgG1 titers than mice vaccinated intramuscularly with Alum®-adjuvanted tetanus toxoid vaccine, IgG2a titers and antibody affinity maturation were similar between these groups. We conclude that skin immunization with MNPs containing unadjuvanted tetanus toxoid can confer potent protective efficacy to mothers and their offspring using a delivery method well suited for expanding vaccination coverage in developing countries.
Collapse
Affiliation(s)
- E Stein Esser
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta 30322, Georgia
| | - AndreyA Romanyuk
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta 30332, Georgia
| | - Elena V Vassilieva
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta 30322, Georgia
| | - Joshy Jacob
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta 30322, Georgia
| | - Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta 30332, Georgia
| | - Richard W Compans
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta 30322, Georgia
| | - Ioanna Skountzou
- Department of Microbiology and Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta 30322, Georgia.
| |
Collapse
|
46
|
Tomar J, Born PA, Frijlink HW, Hinrichs WLJ. Dry influenza vaccines: towards a stable, effective and convenient alternative to conventional parenteral influenza vaccination. Expert Rev Vaccines 2016; 15:1431-1447. [DOI: 10.1080/14760584.2016.1182869] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
47
|
Vassilieva EV, Kalluri H, McAllister D, Taherbhai MT, Esser ES, Pewin WP, Pulit-Penaloza JA, Prausnitz MR, Compans RW, Skountzou I. Improved immunogenicity of individual influenza vaccine components delivered with a novel dissolving microneedle patch stable at room temperature. Drug Deliv Transl Res 2016; 5:360-71. [PMID: 25895053 DOI: 10.1007/s13346-015-0228-0] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Prevention of seasonal influenza epidemics and pandemics relies on widespread vaccination coverage to induce protective immunity. In addition to a good antigenic match with the circulating viruses, the effectiveness of individual strains represented in the trivalent vaccines depends on their immunogenicity. In this study, we evaluated the immunogenicity of H1N1, H3N2, and B seasonal influenza virus vaccine strains delivered individually with a novel dissolving microneedle patch and the stability of this formulation during storage at 25 °C. Our data demonstrate that all strains retained their antigenic activity after incorporation in the dissolving patches as measured by single radial diffusion (SRID) assay and immune responses to vaccination in BALB/c mice. After a single immunization, all three antigens delivered with microneedle patches induced superior neutralizing antibody titers compared to intramuscular immunization. Cutaneous antigen delivery was especially beneficial for the less immunogenic B strain. Mice immunized with dissolving microneedle patches encapsulating influenza A/Brisbane/59/07 (H1N1) vaccine were fully protected against lethal challenge by homologous mouse-adapted influenza virus. All vaccine components retained activity during storage at room temperature for at least 3 months as measured in vitro by SRID assay and in vivo by mouse immunization studies. Our data demonstrate that dissolving microneedle patches are a promising advance for influenza cutaneous vaccination due to improved immune responses using less immunogenic influenza antigens and enhanced stability.
Collapse
Affiliation(s)
- Elena V Vassilieva
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, 1518 Clifton Road, Atlanta, GA, 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Marshall S, Sahm LJ, Moore AC. The success of microneedle-mediated vaccine delivery into skin. Hum Vaccin Immunother 2016; 12:2975-2983. [PMID: 27050528 DOI: 10.1080/21645515.2016.1171440] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Microneedles (MNs) are designed to specifically target the outermost, skin barrier layer, the stratum corneum, creating transient pathways for minimally invasive transcutaneous delivery. It is reported that MNs can facilitate delivery without stimulating the pain receptors or damaging blood vessels that lie beneath, thus being perceived as painless and associated with reduced bleeding. This immunocompetence of the skin, coupled with its ease of access, makes this organ an attractive vaccination site. The purpose of this review was to collate primary scientific literature pertaining to MN-mediated in vivo vaccination programmes. A total of 62 original research articles are presented, compiling vaccination strategies in 6 different models (mouse, rat, guinea pig, rabbit, pig, macaque and human). Vaccines tested span a wide range of viral, bacterial and protozoan pathogens and includes 7 of the 13 vaccine-preventable diseases, as defined by the WHO. This review highlights the paucity of available clinical trial data. MN-delivered vaccines have demonstrated safety and immunogenicity in pre-clinical models and boast desirable attributes such as painless administration, thermostability, dose-sparing capacity and the potential for self-administration. These advantages should contribute to enhanced global vaccine access.
Collapse
Affiliation(s)
- Sarah Marshall
- a School of Pharmacy, University College Cork , Cork , Ireland
| | - Laura J Sahm
- a School of Pharmacy, University College Cork , Cork , Ireland.,b Department of Pharmacy , Mercy University Hospital , Cork , Ireland
| | - Anne C Moore
- a School of Pharmacy, University College Cork , Cork , Ireland.,c Department of Pharmacology and Therapeutics , University College Cork , Cork , Ireland
| |
Collapse
|
49
|
Kim YC, Lee SH, Choi WH, Choi HJ, Goo TW, Lee JH, Quan FS. Microneedle delivery of trivalent influenza vaccine to the skin induces long-term cross-protection. J Drug Target 2016; 24:943-951. [PMID: 26957023 DOI: 10.3109/1061186x.2016.1159213] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A painless self-immunization method with effective and broad cross-protection is urgently needed to prevent infections against newly emerging influenza viruses. In this study, we investigated the cross-protection efficacy of trivalent influenza vaccine containing inactivated A/PR/8/34 (H1N1), A/Hong Kong/68 (H3N2) and B/Lee/40 after skin vaccination using microneedle patches coated with this vaccine. Microneedle vaccination of mice in the skin provided 100% protection against lethal challenges with heterologous pandemic strain influenza A/California/04/09, heterogeneous A/Philippines/2/82 and B/Victoria/287 viruses 8 months after boost immunization. Cross-reactive serum IgG antibody responses against heterologous influenza viruses A/California/04/09, A/Philippines/2/82 and B/Victoria/287 were induced at high levels. Hemagglutination inhibition titers were also maintained at high levels against these heterogeneous viruses. Microneedle vaccination induced substantial levels of cross-reactive IgG antibody responses in the lung and cellular immune responses, as well as cross-reactive antibody-secreting plasma cells in the spleen. Viral loads in the lung were significantly (p < 0.05) reduced. All mice survived after viral challenges. These results indicate that skin vaccination with trivalent vaccine using a microneedle array could provide protection against seasonal epidemic or new pandemic strain of influenza viruses.
Collapse
Affiliation(s)
- Yeu-Chun Kim
- a Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST) , Daejeon, Korea
| | - Su-Hwa Lee
- b Department of Biomedical Science, Graduate School, Kyung Hee University , Seoul, Korea
| | - Won-Hyung Choi
- c Department of Medical Zoology, Kyung Hee University School of Medicine , Seoul, Korea
| | - Hyo-Jick Choi
- d Department of Chemical and Materials Engineering, University of Alberta , Edmonton, Alberta, Canada
| | - Tae-Won Goo
- e Department of Biochemistry, Dongguk University College of Medicine , Gyeongju, Korea
| | - Ju-Hie Lee
- f Department of Pathology, Kyung Hee University Medical Center , Seoul, Korea
| | - Fu-Shi Quan
- c Department of Medical Zoology, Kyung Hee University School of Medicine , Seoul, Korea
| |
Collapse
|
50
|
Ramezanli T, Karry K, Zhang Z, Shah K, Michniak-Kohn B. Transdermal Delivery of Drugs Using Patches and Patchless Delivery Systems. Drug Deliv 2016. [DOI: 10.1002/9781118833322.ch11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023] Open
|