1
|
Choo JJY, McMillan CLD, Young PR, Muller DA. Microarray patches: scratching the surface of vaccine delivery. Expert Rev Vaccines 2023; 22:937-955. [PMID: 37846657 DOI: 10.1080/14760584.2023.2270598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
INTRODUCTION Microneedles are emerging as a promising technology for vaccine delivery, with numerous advantages over traditional needle and syringe methods. Preclinical studies have demonstrated the effectiveness of MAPs in inducing robust immune responses over traditional needle and syringe methods, with extensive studies using vaccines targeted against different pathogens in various animal models. Critically, the clinical trials have demonstrated safety, immunogenicity, and patient acceptance for MAP-based vaccines against influenza, measles, rubella, and SARS-CoV-2. AREAS COVERED This review provides a comprehensive overview of the different types of microarray patches (MAPs) and analyses of their applications in preclinical and clinical vaccine delivery settings. This review also covers additional considerations for microneedle-based vaccination, including adjuvants that are compatible with MAPs, patient safety and factors for global vaccination campaigns. EXPERT OPINION MAP vaccine delivery can potentially be a game-changer for vaccine distribution and coverage in both high-income and low- and middle-income countries. For MAPs to reach this full potential, many critical hurdles must be overcome, such as large-scale production, regulatory compliance, and adoption by global health authorities. However, given the considerable strides made in recent years by MAP developers, it may be possible to see the first MAP-based vaccines in use within the next 5 years.
Collapse
Affiliation(s)
- Jovin J Y Choo
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Christopher L D McMillan
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - Paul R Young
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| | - David A Muller
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
2
|
Kumar P, Bird C, Holland D, Joshi SB, Volkin DB. Current and next-generation formulation strategies for inactivated polio vaccines to lower costs, increase coverage, and facilitate polio eradication. Hum Vaccin Immunother 2022; 18:2154100. [PMID: 36576132 PMCID: PMC9891683 DOI: 10.1080/21645515.2022.2154100] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/08/2022] [Accepted: 11/29/2022] [Indexed: 12/29/2022] Open
Abstract
Implementation of inactivated polio vaccines (IPV) containing Sabin strains (sIPV) will further enable global polio eradication efforts by improving vaccine safety during use and containment during manufacturing. Moreover, sIPV-containing vaccines will lower costs and expand production capacity to facilitate more widespread use in low- and middle-income countries (LMICs). This review focuses on the role of vaccine formulation in these efforts including traditional Salk IPV vaccines and new sIPV-containing dosage forms. The physicochemical properties and stability profiles of poliovirus antigens are described. Formulation approaches to lower costs include developing multidose and combination vaccine formats as well as improving storage stability. Formulation strategies for dose-sparing and enhanced mucosal immunity include employing adjuvants (e.g. aluminum-salt and newer adjuvants) and/or novel delivery systems (e.g. ID administration with microneedle patches). The potential for applying these low-cost formulation development strategies to other vaccines to further improve vaccine access and coverage in LMICs is also discussed.
Collapse
Affiliation(s)
- Prashant Kumar
- 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
| | - David Holland
- 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
| | - David B. Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
3
|
Wijesundara YH, Herbert FC, Trashi O, Trashi I, Brohlin OR, Kumari S, Howlett T, Benjamin CE, Shahrivarkevishahi A, Diwakara SD, Perera SD, Cornelius SA, Vizuet JP, Balkus KJ, Smaldone RA, De Nisco NJ, Gassensmith JJ. Carrier gas triggered controlled biolistic delivery of DNA and protein therapeutics from metal-organic frameworks. Chem Sci 2022; 13:13803-13814. [PMID: 36544734 PMCID: PMC9710232 DOI: 10.1039/d2sc04982a] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/24/2022] [Indexed: 12/24/2022] Open
Abstract
The efficacy and specificity of protein, DNA, and RNA-based drugs make them popular in the clinic; however, these drugs are often delivered via injection, requiring skilled medical personnel, and producing biohazardous waste. Here, we report an approach that allows for their controlled delivery, affording either a burst or slow release without altering the formulation. We show that when encapsulated within zeolitic-imidazolate framework eight (ZIF-8), the biomolecules are stable in powder formulations and can be inoculated with a low-cost, gas-powered "MOF-Jet" into living animal and plant tissues. Additionally, their release profiles can be modulated through judicious selection of the carrier gas used in the MOF-Jet. Our in vitro and in vivo studies reveal that when CO2 is used, it creates a transient and weakly acidic local environment that causes a near-instantaneous release of the biomolecules through an immediate dissolution of ZIF-8. Conversely, when air is used, ZIF-8 biodegrades slowly, releasing the biomolecules over a week. This is the first example of controlled-biolistic delivery of biomolecules using ZIF-8, which provides a powerful tool for fundamental and applied science research.
Collapse
Affiliation(s)
- Yalini H. Wijesundara
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Fabian C. Herbert
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Orikeda Trashi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Ikeda Trashi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Olivia R. Brohlin
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Sneha Kumari
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Thomas Howlett
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Candace E. Benjamin
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Arezoo Shahrivarkevishahi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Shashini D. Diwakara
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Sachini D. Perera
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Samuel A. Cornelius
- Department of Biological Sciences, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Juan P. Vizuet
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Kenneth J. Balkus
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Ronald A. Smaldone
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Nicole J. De Nisco
- Department of Biological Sciences, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| | - Jeremiah J. Gassensmith
- Department of Chemistry and Biochemistry, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA,Department of Biomedical Engineering, The University of Texas at Dallas800 West Campbel RdRichardson 75080TXUSA
| |
Collapse
|
4
|
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
|
5
|
Henricson J, Muller DA, Baker SB, Iredahl F, Togö T, Anderson CD. Micropuncture closure following high density microarray patch application in healthy subjects. Skin Res Technol 2022; 28:305-310. [PMID: 35064694 PMCID: PMC9907643 DOI: 10.1111/srt.13131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/18/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND The high-density microarray patch (HD-MAP) promises to be a robust vaccination platform with clear advantages for future global societal demands for health care management. The method of action has its base not only in efficient delivery of vaccine but also in the reliable induction of a local innate physical inflammatory response to adjuvant the vaccination process. The application process needs to induce levels of reactivity, which are acceptable to the vaccine, and from which the skin promptly recovers. MATERIALS AND METHODS 1 × 1 cm HD-MAP patches containing 5000, 250-μm long microprojections were applied to the skin in 12 healthy volunteers. The return of skin barrier function was assessed by transepidermal water loss (TEWL) and reaction to topical histamine challenge. RESULTS Skin barrier recovery by 48 h was confirmed for all HD-MAP sites by recovered resistance to the effects of topical histamine application. CONCLUSIONS Our previous observation, that the barrier disruption indicator TEWL returns to normal by 48 h, is supported by this paper's demonstration of return of skin resistance to topical histamine challenge in twelve healthy subjects.
Collapse
Affiliation(s)
- Joakim Henricson
- Department of Biomedical and Clinical Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden.,Department of Emergency Medicine, Local Health Care Services in Central Östergötland, Linkoping, Sweden
| | - David A Muller
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Queensland, Australia
| | - S Ben Baker
- Vaxxas Pty Ltd, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Fredrik Iredahl
- Department of Primary health care, Region Östergötland, Linköping, Sweden.,Department of Medical and Health Sciences, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Totte Togö
- Allergy Centre, Region Östergötland, Linkoping, Sweden
| | - Chris D Anderson
- Division of Cell Biology, Faculty of Health Sciences, Linköping University, Linkoping, Sweden
| |
Collapse
|
6
|
Alotaibi BS, Buabeid M, Ibrahim NA, Kharaba ZJ, Ijaz M, Murtaza G. Recent strategies driving oral biologic administration. Expert Rev Vaccines 2021; 20:1587-1601. [PMID: 34612121 DOI: 10.1080/14760584.2021.1990044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION High patient compliance, noninvasiveness, and self-administration are the leading features of vaccine delivery through the oral route. The implementation of swift mass vaccination campaigns in pandemic outbreaks fascinates the use of oral vaccination. This approach can elicit both mucosal and systemic immune responses to protect against infection at the surface of the mucosa. AREA COVERED As pathogen entry and spread mainly occurs through the gastrointestinal tract (GIT) mucosal surfaces, oral vaccination may protect and limit disease spread. Oral vaccines target various potential mucosal inductive sites in the GIT, such as the oral cavity, gastric area, and small intestine. Orally delivered vaccines having subunit and nucleic acid pass through various GIT-associated risks, such as the biodegradation of biologics and their reduced absorption. This article presents a summarized review of the existing technologies and prospects for oral vaccination. EXPERT OPINION The intestinal mucosa focuses on current approaches, while future strategies target new mucosal sites, i.e. oral cavity and stomach. Recent developments in biologic delivery through the oral route and their potential use in future oral vaccination are mainly considered.
Collapse
Affiliation(s)
- Badriyah Shadid Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Manal Buabeid
- Department of Clinical Sciences, Ajman University, Ajman, 346, UAE.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Nihal Abdalla Ibrahim
- Department of Clinical Sciences, Ajman University, Ajman, 346, UAE.,Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
| | - Zelal Jaber Kharaba
- Department of Clinical Sciences, College of Pharmacy, Al-Ain University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Munazza Ijaz
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore, 54000, Pakistan
| |
Collapse
|
7
|
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
|
8
|
Depelsenaire ACI, Witham K, Veitch M, Wells JW, Anderson CD, Lickliter JD, Rockman S, Bodle J, Treasure P, Hickling J, Fernando GJP, Forster AH. Cellular responses at the application site of a high-density microarray patch delivering an influenza vaccine in a randomized, controlled phase I clinical trial. PLoS One 2021; 16:e0255282. [PMID: 34329337 PMCID: PMC8323919 DOI: 10.1371/journal.pone.0255282] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 06/09/2021] [Indexed: 12/23/2022] Open
Abstract
Microarray patches (MAPs) have the potential to be a safer, more acceptable, easier to use and more cost-effective method for administration of vaccines when compared to the needle and syringe. Since MAPs deliver vaccine to the dermis and epidermis, a degree of local immune response at the site of application is expected. In a phase 1 clinical trial (ACTRN 12618000112268), the Vaxxas high-density MAP (HD-MAP) was used to deliver a monovalent, split inactivated influenza virus vaccine into the skin. HD-MAP immunisation led to significantly enhanced humoral responses on day 8, 22 and 61 compared with IM injection of a quadrivalent commercial seasonal influenza vaccine (Afluria Quadrivalent®). Here, the aim was to analyse cellular responses to HD-MAPs in the skin of trial subjects, using flow cytometry and immunohistochemistry. HD-MAPs were coated with a split inactivated influenza virus vaccine (A/Singapore/GP1908/2015 [H1N1]), to deliver 5 μg haemagglutinin (HA) per HD-MAP. Three HD-MAPs were applied to the volar forearm (FA) of five healthy volunteers (to achieve the required 15 μg HA dose), whilst five control subjects received three uncoated HD-MAPs (placebo). Local skin response was recorded for over 61 days and haemagglutination inhibition antibody titres (HAI) were assessed on days 1, 4, 8, 22, and 61. Skin biopsies were taken before (day 1), and three days after HD-MAP application (day 4) and analysed by flow-cytometry and immunohistochemistry to compare local immune subset infiltration. HD-MAP vaccination with 15 μg HA resulted in significant HAI antibody titres compared to the placebo group. Application of uncoated placebo HD-MAPs resulted in mild erythema and oedema in most subjects, that resolved by day 4 in 80% of subjects. Active, HA-coated HD-MAP application resulted in stronger erythema responses on day 4, which resolved between days 22-61. Overall, these erythema responses were accompanied by an influx of immune cells in all subjects. Increased cell infiltration of CD3+, CD4+, CD8+ T cells as well as myeloid CD11b+ CD11c+ and non-myeloid CD11b- dendritic cells were observed in all subjects, but more pronounced in active HD-MAP groups. In contrast, CD19+/CD20+ B cell counts remained unchanged. Key limitations include the use of an influenza vaccine, to which the subjects may have had previous exposure. Different results might have been obtained with HD-MAPs inducing a primary immune response. In conclusion, influenza vaccine administered to the forearm (FA) using the HD-MAP was well-tolerated and induced a mild to moderate skin response with lymphocytic infiltrate at the site of application.
Collapse
Affiliation(s)
| | | | - Margaret Veitch
- The University of Queensland Diamantina Institute, Woolloongabba, Queensland, Australia
| | - James W. Wells
- The University of Queensland Diamantina Institute, Woolloongabba, Queensland, Australia
| | | | | | - Steve Rockman
- Seqirus Pty Ltd, Parkville, Victoria, Australia
- Department of Microbiology and Immunology, University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jesse Bodle
- Seqirus Pty Ltd, Parkville, Victoria, Australia
| | - Peter Treasure
- Peter Treasure Statistical Services Ltd, Kings Lynn, United Kingdom
| | | | - Germain J. P. Fernando
- Vaxxas Pty Ltd, Brisbane, Queensland, Australia
- The University of Queensland, School of Chemistry & Molecular Biosciences, Faculty of Science, Brisbane, Queensland, Australia
| | | |
Collapse
|
9
|
Clustering and Erratic Movement Patterns of Syringe-Injected versus Mosquito-Inoculated Malaria Sporozoites Underlie Decreased Infectivity. mSphere 2021; 6:6/2/e00218-21. [PMID: 33827910 PMCID: PMC8546700 DOI: 10.1128/msphere.00218-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Malaria vaccine candidates based on live, attenuated sporozoites have led to high levels of protection. However, their efficacy critically depends on the sporozoites' ability to reach and infect the host liver. Administration via mosquito inoculation is by far the most potent method for inducing immunity but highly impractical. Here, we observed that intradermal syringe-injected Plasmodium berghei sporozoites (syrSPZ) were 3-fold less efficient in migrating to and infecting mouse liver than mosquito-inoculated sporozoites (msqSPZ). This was related to a clustered dermal distribution (2-fold-decreased median distance between syrSPZ and msqSPZ) and, more importantly, a 1.4-fold (significantly)-slower and more erratic movement pattern. These erratic movement patterns were likely caused by alteration of dermal tissue morphology (>15-μm intercellular gaps) due to injection of fluid and may critically decrease sporozoite infectivity. These results suggest that novel microvolume-based administration technologies hold promise for replicating the success of mosquito-inoculated live, attenuated sporozoite vaccines.IMPORTANCE Malaria still causes a major burden on global health and the economy. The efficacy of live, attenuated malaria sporozoites as vaccine candidates critically depends on their ability to migrate to and infect the host liver. This work sheds light on the effect of different administration routes on sporozoite migration. We show that the delivery of sporozoites via mosquito inoculation is more efficient than syringe injection; however, this route of administration is highly impractical for vaccine purposes. Using confocal microscopy and automated imaging software, we demonstrate that syringe-injected sporozoites do cluster, move more slowly, and display more erratic movement due to alterations in tissue morphology. These findings indicate that microneedle-based engineering solutions hold promise for replicating the success of mosquito-inoculated live, attenuated sporozoite vaccines.
Collapse
|
10
|
Coffey JW, Gaiha GD, Traverso G. Oral Biologic Delivery: Advances Toward Oral Subunit, DNA, and mRNA Vaccines and the Potential for Mass Vaccination During Pandemics. Annu Rev Pharmacol Toxicol 2021; 61:517-540. [PMID: 32466690 PMCID: PMC8057107 DOI: 10.1146/annurev-pharmtox-030320-092348] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Oral vaccination enables pain-free and self-administrable vaccine delivery for rapid mass vaccination during pandemic outbreaks. Furthermore, it elicits systemic and mucosal immune responses. This protects against infection at mucosal surfaces, which may further enhance protection and minimize the spread of disease. The gastrointestinal (GI) tract presents a number of prospective mucosal inductive sites for vaccine targeting, including the oral cavity, stomach, and small intestine. However, currently available oral vaccines are effectively limited to live-attenuated and inactivated vaccines against enteric diseases. The GI tract poses a number of challenges,including degradative processes that digest biologics and mucosal barriers that limit their absorption. This review summarizes the approaches currently under development and future opportunities for oral vaccine delivery to established (intestinal) and relatively new (oral cavity, stomach) mucosal targets. Special consideration is given to recent advances in oral biologic delivery that offer promise as future platforms for the administration of oral vaccines.
Collapse
Affiliation(s)
- Jacob William Coffey
- Department of Chemical Engineering and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunology, University of Melbourne, Victoria, 3000, Australia
| | - Gaurav Das Gaiha
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts 02139, USA
- Gastrointestinal Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Giovanni Traverso
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA;
| |
Collapse
|
11
|
Muller DA, Henricson J, Baker SB, Togö T, Jayashi Flores CM, Lemaire PA, Forster A, Anderson CD. Innate local response and tissue recovery following application of high density microarray patches to human skin. Sci Rep 2020; 10:18468. [PMID: 33116241 PMCID: PMC7595201 DOI: 10.1038/s41598-020-75169-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/09/2020] [Indexed: 12/15/2022] Open
Abstract
The development of microarray patches for vaccine application has the potential to revolutionise vaccine delivery. Microarray patches (MAP) reduce risks of needle stick injury, do not require reconstitution and have the potential to enhance immune responses using a fractional vaccine dose. To date, the majority of research has focused on vaccine delivery with little characterisation of local skin response and recovery. Here we study in detail the immediate local skin response and recovery of the skin post high density MAP application in 12 individuals receiving 3 MAPs randomly assigned to the forearm and upper arm. Responses were characterised by clinical scoring, dermatoscopy, evaporimetry and tissue viability imaging (TiVi). MAP application resulted in punctures in the epidermis, a significant transepidermal water loss (TEWL), the peak TEWL being concomitant with peak erythema responses visualised by TiVi. TEWL and TiVi responses reduced over time, with TEWL returning to baseline by 48 h and erythema fading over the course of a 7 day period. As MAPs for vaccination move into larger clinical studies more variation of individual subject phenotypic or disease propensity will be encountered which will require consideration both in regard to reliability of dose delivery and degree of inherent skin response.
Collapse
Affiliation(s)
- David A Muller
- School of Chemistry and Molecular Biosciences, The University of Queensland, Building 76 Cooper road, St. Lucia, QLD, 4072, Australia.
| | - Joakim Henricson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Department of Emergency Medicine, Local Health Care Services in Central Östergötland, Linköping, Sweden
| | - S Ben Baker
- Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Totte Togö
- Allergy Center Linköping, Region Östergötland, Sweden
| | - Cesar M Jayashi Flores
- Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Pierre A Lemaire
- Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Angus Forster
- Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
| | - Chris D Anderson
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden. .,Division of Cell Biology, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
12
|
Pan J, Cui Z. Self-Assembled Nanoparticles: Exciting Platforms for Vaccination. Biotechnol J 2020; 15:e2000087. [PMID: 33411412 DOI: 10.1002/biot.202000087] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/25/2020] [Indexed: 12/14/2022]
Abstract
Vaccination is successfully advanced to control several fatal diseases and improve human life expectancy. However, additional innovations are required in this field because there are no effective vaccines to prevent some infectious diseases. The shift from the attenuated or inactivated pathogens to safer but less immunogenic protein or peptide antigens has led to a search for effective antigen delivery carriers that can function as both antigen vehicles and intrinsic adjuvants. Among these carriers, self-assembled nanoparticles (SANPs) have shown great potential to be the best representative. For the nanoscale and multiple presentation of antigens, with accurate control over size, geometry, and functionality, these nanoparticles are assembled spontaneously and mimic pathogens, resulting in enhanced antigen presentation and increased cellular and humoral immunity responses. In addition, they may be applied through needle-free routes due to their adhesive ability, which gives them a great future in vaccination applications. This review provides an overview of various SANPs and their applications in prophylactic vaccines.
Collapse
Affiliation(s)
- Jingdi Pan
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
13
|
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
|
14
|
Efficient Delivery of Dengue Virus Subunit Vaccines to the Skin by Microprojection Arrays. Vaccines (Basel) 2019; 7:vaccines7040189. [PMID: 31756967 PMCID: PMC6963636 DOI: 10.3390/vaccines7040189] [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: 10/09/2019] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 11/20/2022] Open
Abstract
Dengue virus is the most important arbovirus impacting global human health, with an estimated 390 million infections annually, and over half the world’s population at risk of infection. While significant efforts have been made to develop effective vaccines to mitigate this threat, the task has proven extremely challenging, with new approaches continually being sought. The majority of protective, neutralizing antibodies induced during infection are targeted by the envelope (E) protein, making it an ideal candidate for a subunit vaccine approach. Using truncated, recombinant, secreted E proteins (sE) of all 4 dengue virus serotypes, we have assessed their immunogenicity and protective efficacy in mice, with or without Quil-A as an adjuvant, and delivered via micropatch array (MPA) to the skin in comparison with more traditional routes of immunization. The micropatch contains an ultra-high density array (21,000/cm2) of 110 μm microprojections. Mice received 3 doses of 1 μg (nanopatch, intradermal, subcutaneous, or intra muscular injection) or 10 μg (intradermal, subcutaneous, or intra muscular injection) of tetravalent sE spaced 4 weeks apart. When adjuvanted with Quil-A, tetravalent sE vaccination delivered via MPA resulted in earlier induction of virus-neutralizing IgG antibodies for all four serotypes when compared with all of the other vaccination routes. Using the infectious dengue virus AG129 mouse infectious dengue model, these neutralizing antibodies protected all mice from lethal dengue virus type 2 D220 challenge, with protected animals showing no signs of disease or circulating virus. If these results can be translated to humans, MPA-delivered sE represents a promising approach to dengue virus vaccination.
Collapse
|
15
|
Haridass IN, Wei JCJ, Mohammed YH, Crichton ML, Anderson CD, Henricson J, Sanchez WY, Meliga SC, Grice JE, Benson HAE, Kendall MAF, Roberts MS. Cellular metabolism and pore lifetime of human skin following microprojection array mediation. J Control Release 2019; 306:59-68. [PMID: 31121279 DOI: 10.1016/j.jconrel.2019.05.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 04/11/2019] [Accepted: 05/14/2019] [Indexed: 01/13/2023]
Abstract
Skin-targeting microscale medical devices are becoming popular for therapeutic delivery and diagnosis. We used cryo-SEM, fluorescence lifetime imaging microscopy (FLIM), autofluorescence imaging microscopy and inflammatory response to study the puncturing and recovery of human skin ex vivo and in vivo after discretised puncturing by a microneedle array (Nanopatch®). Pores induced by the microprojections were found to close by ~25% in diameter within the first 30 min, and almost completely close by ~6 h. FLIM images of ex vivo viable epidermis showed a stable fluorescence lifetime for unpatched areas of ~1000 ps up to 24 h. Only the cells in the immediate puncture zones (in direct contact with projections) showed a reduction in the observed fluorescence lifetimes to between ~518-583 ps. The ratio of free-bound NAD(P)H (α1/α2) in unaffected areas of the viable epidermis was ~2.5-3.0, whereas the ratio at puncture holes was almost double at ~4.2-4.6. An exploratory pilot in vivo study also suggested similar closure rate with histamine administration to the forearms of human volunteers after Nanopatch® treatment, although a prolonged inflammation was observed with Tissue Viability Imaging. Overall, this work shows that the pores created by the microneedle-type medical device, Nanopatch®, are transient, with the skin recovering rapidly within 1-2 days in the epidermis after application.
Collapse
Affiliation(s)
- Isha N Haridass
- Curtin Health Innovation Research Institute, School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, WA 6102, Australia; Therapeutics Research Centre, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Jonathan C J Wei
- Therapeutics Research Centre, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia; Department of Biomechanical Engineering, Faculty of Mechanical, Maritime and Materials Engineering, Delft University of Technology, 2628, CD, Delft, the Netherlands
| | - Yousuf H Mohammed
- Therapeutics Research Centre, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Michael L Crichton
- Institute of Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh EH14 4AS, United Kingdom
| | - Christopher D Anderson
- Department of Clinical and Experimental Medicine, Linköping University, 581 83 Linköping, Sweden
| | - Joakim Henricson
- Division of Drug Research, Department of Medical and Health Sciences, Faculty of Health Sciences Linköping University, Department of Emergency Medicine Local Health Care Services in Central Östergötland, Region Östergötland, Sweden
| | - Washington Y Sanchez
- Therapeutics Research Centre, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Stefano C Meliga
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Jeffrey E Grice
- Therapeutics Research Centre, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Heather A E Benson
- Curtin Health Innovation Research Institute, School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, WA 6102, Australia
| | - Mark A F Kendall
- Australian National University, Canberra, ACT 0200, Australia; Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital, Herston, QLD 4006, Australia
| | - Michael S Roberts
- Therapeutics Research Centre, Faculty of Medicine, The University of Queensland, Translational Research Institute, Woolloongabba, QLD 4102, Australia; School of Pharmacy and Medical Sciences, University of South Australia, Basil Hetzel Institute for Translational Health Research, Adelaide, SA 5011, Australia.
| |
Collapse
|
16
|
Wei JCJ, Haridass IN, Crichton ML, Mohammed YH, Meliga SC, Sanchez WY, Grice JE, Benson HAE, Roberts MS, Kendall MAF. Space- and time-resolved investigation on diffusion kinetics of human skin following macromolecule delivery by microneedle arrays. Sci Rep 2018; 8:17759. [PMID: 30531828 PMCID: PMC6288161 DOI: 10.1038/s41598-018-36009-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/09/2018] [Indexed: 12/16/2022] Open
Abstract
Microscale medical devices are being developed for targeted skin delivery of vaccines and the extraction of biomarkers, with the potential to revolutionise healthcare in both developing and developed countries. The effective clinical development of these devices is dependent on understanding the macro-molecular diffusion properties of skin. We hypothesised that diffusion varied according to specific skin layers. Using three different molecular weights of rhodamine dextran (RD) (MW of 70, 500 and 2000 kDa) relevant to the vaccine and therapeutic scales, we deposited molecules to a range of depths (0-300 µm) in ex vivo human skin using the Nanopatch device. We observed significant dissipation of RD as diffusion with 70 and 500 kDa within the 30 min timeframe, which varied with MW and skin layer. Using multiphoton microscopy, image analysis and a Fick's law analysis with 2D cartesian and axisymmetric cylindrical coordinates, we reported experimental trends of epidermal and dermal diffusivity values ranging from 1-8 µm2 s-1 to 1-20 µm2 s-1 respectively, with a significant decrease in the dermal-epidermal junction of 0.7-3 µm2 s-1. In breaching the stratum corneum (SC) and dermal-epidermal junction barriers, we have demonstrated practical application, delivery and targeting of macromolecules to both epidermal and dermal antigen presenting cells, providing a sound knowledge base for future development of skin-targeting clinical technologies in humans.
Collapse
Affiliation(s)
- Jonathan C J Wei
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, 4102, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Isha N Haridass
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, 4102, Australia
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, 6102, Australia
| | - Michael L Crichton
- Institute of Mechanical, Process and Energy Engineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, EH14 4AS, United Kingdom
| | - Yousuf H Mohammed
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Stefano C Meliga
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Washington Y Sanchez
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Jeffrey E Grice
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, 4102, Australia
| | - Heather A E Benson
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, 6102, Australia
| | - Michael S Roberts
- Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD, 4102, Australia.
- Basil Hetzel Institute for Translational Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, 5011, Australia.
| | | |
Collapse
|
17
|
Coffey JW, Corrie SR, Kendall MAF. Rapid and selective sampling of IgG from skin in less than 1 min using a high surface area wearable immunoassay patch. Biomaterials 2018; 170:49-57. [PMID: 29649748 DOI: 10.1016/j.biomaterials.2018.03.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 11/24/2022]
Abstract
Microprojection array (MPA) patches are an attractive approach to selectively capture circulating proteins from the skin with minimal invasiveness for diagnostics at the point-of-care or in the home. A key challenge to develop this technology is to extract sufficient quantities of specific proteins from within the skin to enable high diagnostic sensitivity within a convenient amount of time. To achieve this, we investigated the effect of MPA geometry (i.e. projection density, length and array size) on protein capture. We hypothesised that the penetrated surface area of MPAs is a major determinant of protein capture however it was not known if simultaneously increasing projection density, length and array size is possible without adversely affecting penetration and/or tolerability. We show that increasing the projection density (5000-30,000 proj. cm-2) and array size (4-36 mm2) significantly increases biomarker capture whilst maintaining of a similar level tolerability, which supports previous literature for projection length (40-190 μm). Ultimately, we designed a high surface area MPA (30,000 proj. cm-2, 36 mm2, 140 μm) with a 4.5-fold increase in penetrated surface area compared to our standard MPA design (20,408 proj. cm-2, 16 mm2, 100 μm). The high surface area MPA captured antigen-specific IgG from mice in 30 s with 100% diagnostic sensitivity compared with 10-30 min for previous MPA immunoassay patches, which is over an order of magnitude reduction in wear time. This demonstrates for the first time that MPAs may be used for ultra-rapid (<1 min) protein capture from skin in a time competitive with standard clinical procedures like the needle and lancet, which has broad implications for minimally invasive and point-of-care diagnostics.
Collapse
Affiliation(s)
- Jacob W Coffey
- Australian Institute for Bioengineering and Nanotechnology, Delivery of Drugs and Genes Group (D2G2), The University of Queensland, St Lucia, Queensland 4072, Australia; Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia
| | - Simon R Corrie
- Australian Institute for Bioengineering and Nanotechnology, Delivery of Drugs and Genes Group (D2G2), The University of Queensland, St Lucia, Queensland 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia; Department of Chemical Engineering, Monash University, Clayton, Victoria, 3800, Australia; Australian Infectious Diseases Research Centre, St. Lucia, Queensland, 4067, Australia
| | - Mark A F Kendall
- Australian Institute for Bioengineering and Nanotechnology, Delivery of Drugs and Genes Group (D2G2), The University of Queensland, St Lucia, Queensland 4072, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The University of Queensland, St Lucia, Queensland 4072, Australia; Australian Infectious Diseases Research Centre, St. Lucia, Queensland, 4067, Australia; The Australian National University, Canberra, Australian Capital Territory 2600, Australia.
| |
Collapse
|
18
|
Wan Y, Hickey JM, Bird C, Witham K, Fahey P, Forster A, Joshi SB, Volkin DB. Development of Stabilizing Formulations of a Trivalent Inactivated Poliovirus Vaccine in a Dried State for Delivery in the Nanopatch™ Microprojection Array. J Pharm Sci 2018; 107:1540-1551. [PMID: 29421219 PMCID: PMC5959271 DOI: 10.1016/j.xphs.2018.01.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 01/22/2018] [Accepted: 01/26/2018] [Indexed: 12/20/2022]
Abstract
The worldwide switch to inactivated polio vaccines (IPVs) is a key component of the overall strategy to achieve and maintain global polio eradication. To this end, new IPV vaccine delivery systems may enhance patient convenience and compliance. In this work, we examine Nanopatch™ (a solid, polymer microprojection array) which offers potential advantages over standard needle/syringe administration including intradermal delivery and reduced antigen doses. Using trivalent IPV (tIPV) and a purpose-built evaporative dry-down system, candidate tIPV formulations were developed to stabilize tIPV during the drying process and on storage. Identifying conditions to minimize tIPV potency losses during rehydration and potency testing was a critical first step. Various classes and types of pharmaceutical excipients (∼50 total) were then evaluated to mitigate potency losses (measured through D-antigen ELISAs for IPV1, IPV2, and IPV3) during drying and storage. Various concentrations and combinations of stabilizing additives were optimized in terms of tIPV potency retention, and 2 candidate tIPV formulations containing cyclodextrin and a reducing agent (e.g., glutathione), maintained ≥80% D-antigen potency during drying and subsequent storage for 4 weeks at 4°C, and ≥60% potency for 3 weeks at room temperature with the majority of losses occurring within the first day of storage.
Collapse
Affiliation(s)
- Ying Wan
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047
| | - John M Hickey
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047
| | - Christopher Bird
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047
| | - Katey Witham
- Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Brisbane, Queensland 4102, Australia
| | - Paul Fahey
- Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Brisbane, Queensland 4102, Australia
| | - Angus Forster
- Vaxxas Pty Ltd, Translational Research Institute, 37 Kent Street, Brisbane, Queensland 4102, Australia
| | - Sangeeta B Joshi
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Macromolecule and Vaccine Stabilization Center, University of Kansas, Lawrence, Kansas 66047.
| |
Collapse
|
19
|
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
|
20
|
Griffin P, Elliott S, Krauer K, Davies C, Rachel Skinner S, Anderson CD, Forster A. Safety, acceptability and tolerability of uncoated and excipient-coated high density silicon micro-projection array patches in human subjects. Vaccine 2017; 35:6676-6684. [DOI: 10.1016/j.vaccine.2017.10.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 10/09/2017] [Accepted: 10/11/2017] [Indexed: 11/16/2022]
|
21
|
Abstract
Microneedle patches (MNPs) contain arrays of solid needles measuring hundreds of microns in length that deliver drugs and vaccines into skin in a painless, easy-to-use manner. Optimal MNP design balances multiple interdependent parameters that determine mechanical strength, skin-insertion reliability, drug delivery efficiency, painlessness, manufacturability, and other features of MNPs that affect their performance. MNPs can be made by adapting various microfabrication technologies for delivery of small-molecule drugs, biologics, and vaccines targeted to the skin, which can have pharmacokinetic and immunologic advantages. A small number of human clinical trials, as well as a large and growing market for MNP products for cosmetics, indicate that MNPs can be used safely, efficaciously, and with strong patient acceptance. More advanced clinical trials and commercial-scale manufacturing will facilitate development of MNPs to realize their potential to dramatically increase patient access to otherwise-injectable drugs and to improve drug performance via skin delivery.
Collapse
Affiliation(s)
- Mark R Prausnitz
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332-0100;
| |
Collapse
|