1
|
Bhadouria N, Yadav S, Bukke SPN, Narapureddy BR. Advancements in vaccine delivery: harnessing 3D printing for microneedle patch technology. Ann Med Surg (Lond) 2025; 87:2059-2067. [PMID: 40212146 PMCID: PMC11981410 DOI: 10.1097/ms9.0000000000003060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 01/31/2025] [Indexed: 04/13/2025] Open
Abstract
The development of 3D-printed microneedle (MN) technology is a significant step in vaccine delivery, providing a painless, effective, and adaptable substitute for conventional injection-based techniques. Direct transdermal vaccination distribution without the need for needles is made possible by microneedle patches, which employ a variety of tiny needles that dissolve when they penetrate the skin. By using 3D printing to precisely customise microneedles' size, shape, and density to meet particular vaccine requirements, administration control can be improved and vaccine efficiency may even be increased. Furthermore, rapid prototyping made possible by 3D printing speeds up the development process, enabling quicker testing and improvement of vaccines. Additionally, this scalable technology can greatly increase vaccine accessibility, particularly in environments with limited resources. Research indicates that by directly interacting with the skin's immune-rich layers, microneedle patches enhance antigen delivery and elicit a strong immune response. Because MN technology offers a useful, self-administrable vaccination approach with little waste, it has significant potential for use in public health applications, notably during pandemics. This study emphasises how 3D-printed microneedle patches have the potential to revolutionise vaccination procedures and increase vaccine accessibility globally.
Collapse
Affiliation(s)
- Namrata Bhadouria
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Shikha Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Sarad Pawar Naik Bukke
- Department of Pharmaceutics and Pharmaceutical Technology, Kampala International University, Western Campus, Ishaka-Bushenyi, Uganda
| | - Bayapa Reddy Narapureddy
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
2
|
Han S, Lee P, Choi HJ. Non-Invasive Vaccines: Challenges in Formulation and Vaccine Adjuvants. Pharmaceutics 2023; 15:2114. [PMID: 37631328 PMCID: PMC10458847 DOI: 10.3390/pharmaceutics15082114] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Given the limitations of conventional invasive vaccines, such as the requirement for a cold chain system and trained personnel, needle-based injuries, and limited immunogenicity, non-invasive vaccines have gained significant attention. Although numerous approaches for formulating and administrating non-invasive vaccines have emerged, each of them faces its own challenges associated with vaccine bioavailability, toxicity, and other issues. To overcome such limitations, researchers have created novel supplementary materials and delivery systems. The goal of this review article is to provide vaccine formulation researchers with the most up-to-date information on vaccine formulation and the immunological mechanisms available, to identify the technical challenges associated with the commercialization of non-invasive vaccines, and to guide future research and development efforts.
Collapse
Affiliation(s)
| | | | - Hyo-Jick Choi
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, AB T6G 1H9, Canada; (S.H.); (P.L.)
| |
Collapse
|
3
|
Synthesis and Characterization of Innovative Microgels Based on Polyacrylic Acid and Microalgae Cell Wall and Their Potential as Antigen Delivery Vehicles. Pharmaceutics 2022; 15:pharmaceutics15010133. [PMID: 36678762 PMCID: PMC9863243 DOI: 10.3390/pharmaceutics15010133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/11/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
In this study, hybrid polyacrylic acid and Schizochytrium sp. microalgae (PAA/Schizo) microgels were synthesized by inverse emulsion assisted by ultrasound using the cell wall fraction as crosslinker. Physicochemical characterization of PAA/Schizo microgels revealed polymeric spherical particles (288 ± 39 nm) and were deemed stable and negatively charged. The produced microgels are not inherently toxic as cell viability was sustained above 80% when mice splenocytes were exposed to concentrations ranging 10-900 µg/mL. PAA/Schizo microgels were evaluated as antigen delivery nanovehicle by adsorbing bovine serum albumin (BSA); with a loading efficiency of 72% and loading capacity of 362 µg/mg. Overall, intranasally-immunized BALB/c mice showed null IgG or IgA responses against PAA/Schizo microgel-BSA, whereas soluble BSA induced significant humoral responses in systemic and mucosal compartments. Splenocytes proliferation assay upon BSA stimulus revealed positive CD4+ T cells-proliferation response in PAA/Schizo microgels-BSA group. Thus, PAA/Schizo microgels constitute functional antigen delivery vehicles of simple and ecofriendly synthesis. Moreover, the use of cell wall fraction as cross-linker agent provides an alternative use for the generation of high-value products using residual algae biomass from the oil industry. Our data suggests that the PAA/Schizo microgels are potential antigen delivery vehicles for immunotherapy development.
Collapse
|
4
|
Chaikhumwang P, Madapong A, Saeng-Chuto K, Nilubol D, Tantituvanont A. Intranasal delivery of inactivated PRRSV loaded cationic nanoparticles coupled with enterotoxin subunit B induces PRRSV-specific immune responses in pigs. Sci Rep 2022; 12:3725. [PMID: 35260663 PMCID: PMC8904483 DOI: 10.1038/s41598-022-07680-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 02/08/2022] [Indexed: 12/18/2022] Open
Abstract
This study was conducted to evaluate the induction of systemic and mucosal immune responses and protective efficacy following the intranasal administration of inactivated porcine reproductive and respiratory syndrome virus (PRRSV) loaded in polylactic acid (PLA) nanoparticles coupled with heat-labile enterotoxin subunit B (LTB) and dimethyldioctadecylammonium bromide (DDA). Here, 42- to 3-week-old PRRSV-free pigs were randomly allocated into 7 groups of 6 pigs each. Two groups represented the negative (nonvaccinated pigs/nonchallenged pigs, NoVacNoChal) and challenge (nonvaccinated/challenged, NoVacChal) controls. The pigs in the other 5 groups, namely, PLA nanoparticles/challenged (blank NPs), LTB-DDA coupled with PLA nanoparticles/challenged (adjuvant-blank NPs), PLA nanoparticles-encapsulating inactivated PRRSV/challenged (KNPs), LTB-DDA coupled with PLA nanoparticles loaded with inactivated PRRSV/challenged pigs (adjuvant-KNPs) and inactivated PRRSV/challenged pigs (inactivated PRRSV), were intranasally vaccinated with previously described vaccines at 0, 7 and 14 days post-vaccination (DPV). Serum and nasal swab samples were collected weekly and assayed by ELISA to detect the presence of IgG and IgA, respectively. Viral neutralizing titer (VNT) in sera, IFN-γ-producing cells and IL-10 secretion in stimulated peripheral blood mononuclear cells (PBMCs) were also measured. The pigs were intranasally challenged with PRRSV-2 at 28 DPV and necropsied at 35 DPV, and then macro- and microscopic lung lesions were evaluated. The results demonstrated that following vaccination, adjuvant-KNP-vaccinated pigs had significantly higher levels of IFN-γ-producing cells, VNT and IgG in sera, and IgA in nasal swab samples and significantly lower IL-10 levels than the other vaccinated groups. Following challenge, the adjuvant-KNP-vaccinated pigs had significantly lower PRRSV RNA and macro- and microscopic lung lesions than the other vaccinated groups. In conclusion, the results of the study demonstrated that adjuvant-KNPs are effective in eliciting immune responses against PRRSV and protecting against PRRSV infections over KNPs and inactivated PRRSV and can be used as an adjuvant for intranasal PRRSV vaccines.
Collapse
Affiliation(s)
- Puwich Chaikhumwang
- Division of Pharmaceutical Sciences, Department of Pharmaceutical Care, Faculty of Pharmaceutical Sciences, University of Phayao, Phayao, 56000, Thailand
| | - Adthakorn Madapong
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Kepalee Saeng-Chuto
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Dachrit Nilubol
- Swine Viral Evolution and Vaccine Development Research Unit, Department of Veterinary Microbiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Angkana Tantituvanont
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
5
|
Pushparajah D, Jimenez S, Wong S, Alattas H, Nafissi N, Slavcev RA. Advances in gene-based vaccine platforms to address the COVID-19 pandemic. Adv Drug Deliv Rev 2021; 170:113-141. [PMID: 33422546 PMCID: PMC7789827 DOI: 10.1016/j.addr.2021.01.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/23/2020] [Accepted: 01/01/2021] [Indexed: 01/07/2023]
Abstract
The novel betacoronavirus, SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), has spread across the globe at an unprecedented rate since its first emergence in Wuhan City, China in December 2019. Scientific communities around the world have been rigorously working to develop a potent vaccine to combat COVID-19 (coronavirus disease 2019), employing conventional and novel vaccine strategies. Gene-based vaccine platforms based on viral vectors, DNA, and RNA, have shown promising results encompassing both humoral and cell-mediated immune responses in previous studies, supporting their implementation for COVID-19 vaccine development. In fact, the U.S. Food and Drug Administration (FDA) recently authorized the emergency use of two RNA-based COVID-19 vaccines. We review current gene-based vaccine candidates proceeding through clinical trials, including their antigenic targets, delivery vehicles, and route of administration. Important features of previous gene-based vaccine developments against other infectious diseases are discussed in guiding the design and development of effective vaccines against COVID-19 and future derivatives.
Collapse
Affiliation(s)
- Deborah Pushparajah
- School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada
| | - Salma Jimenez
- School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada; Theraphage, 151 Charles St W Suite # 199, Kitchener, ON, N2G 1H6, Canada
| | - Shirley Wong
- School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada
| | - Hibah Alattas
- School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada
| | - Nafiseh Nafissi
- Mediphage Bioceuticals, 661 University Avenue, Suite 1300, Toronto, ON, M5G 0B7, Canada
| | - Roderick A Slavcev
- School of Pharmacy, University of Waterloo, 10A Victoria St S, Kitchener N2G 1C5, Canada; Mediphage Bioceuticals, 661 University Avenue, Suite 1300, Toronto, ON, M5G 0B7, Canada; Theraphage, 151 Charles St W Suite # 199, Kitchener, ON, N2G 1H6, Canada.
| |
Collapse
|
6
|
Maina TW, Grego EA, Boggiatto PM, Sacco RE, Narasimhan B, McGill JL. Applications of Nanovaccines for Disease Prevention in Cattle. Front Bioeng Biotechnol 2020; 8:608050. [PMID: 33363134 PMCID: PMC7759628 DOI: 10.3389/fbioe.2020.608050] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Vaccines are one of the most important tools available to prevent and reduce the incidence of infectious diseases in cattle. Despite their availability and widespread use to combat many important pathogens impacting cattle, several of these products demonstrate variable efficacy and safety in the field, require multiple doses, or are unstable under field conditions. Recently, nanoparticle-based vaccine platforms (nanovaccines) have emerged as promising alternatives to more traditional vaccine platforms. In particular, polymer-based nanovaccines provide sustained release of antigen payloads, stabilize such payloads, and induce enhanced antibod- and cell-mediated immune responses, both systemically and locally. To improve vaccine administrative strategies and efficacy, they can be formulated to contain multiple antigenic payloads and have the ability to protect fragile proteins from degradation. Nanovaccines are also stable at room temperature, minimizing the need for cold chain storage. Nanoparticle platforms can be synthesized for targeted delivery through intranasal, aerosol, or oral administration to induce desired mucosal immunity. In recent years, several nanovaccine platforms have emerged, based on biodegradable and biocompatible polymers, liposomes, and virus-like particles. While most nanovaccine candidates have not yet advanced beyond testing in rodent models, a growing number have shown promise for use against cattle infectious diseases. This review will highlight recent advancements in polymeric nanovaccine development and the mechanisms by which nanovaccines may interact with the bovine immune system. We will also discuss the positive implications of nanovaccines use for combating several important viral and bacterial disease syndromes and consider important future directions for nanovaccine development in beef and dairy cattle.
Collapse
Affiliation(s)
- Teresia W. Maina
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Elizabeth A. Grego
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Paola M. Boggiatto
- Infectious Bacterial Diseases Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Randy E. Sacco
- Ruminant Diseases and Immunology Research Unit, National Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Ames, IA, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA, United States
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
7
|
Chen Y, Jie K, Li B, Yu H, Ruan H, Wu J, Huang X, Liu Q. Immunization With Outer Membrane Vesicles Derived From Major Outer Membrane Protein-Deficient Salmonella Typhimurium Mutants for Cross Protection Against Salmonella Enteritidis and Avian Pathogenic Escherichia coli O78 Infection in Chickens. Front Microbiol 2020; 11:588952. [PMID: 33329465 PMCID: PMC7720508 DOI: 10.3389/fmicb.2020.588952] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/12/2020] [Indexed: 11/19/2022] Open
Abstract
Colibacillosis is an economically important infectious disease in poultry, caused by avian pathogenic Escherichia coli (APEC). Salmonella enterica serovar Enteritidis (S. Enteritidis) is a major cause of food-borne diseases in human circulated through poultry-derived products, including meat and chicken eggs. Vaccine control is the mainstream approach for combating these infections, but it is difficult to create a vaccine for the broad-spectrum protection of poultry due to multiple serotypes of these pathogens. Our previous studies have shown that outer membrane vesicles (OMVs) derived from S. enterica serovar Typhimurium mutants with a remodeled outer membrane could induce cross-protection against heteroserotypic Salmonella infection. Therefore, in this study, we further evaluated the potential of broad-spectrum vaccines based on major outer membrane protein (OMP)-deficient OMVs, including ΔompA, ΔompC, and ΔompD, and determined the protection effectiveness of these candidate vaccines in murine and chicken infection models. The results showed that ΔompA led to an increase in the production of OMVs. Notably, ΔompAΔompCΔompD OMVs showed significantly better cross-protection against S. enterica serovar Choleraesuis, S. Enteritidis, APEC O78, and Shigella flexneri 2a than did other omp-deficient OMVs, with the exception of ΔompA OMVs. Subsequently, we verified the results in the chicken model, in which ΔompAΔompCΔompD OMVs elicited significant cross-protection against S. Enteritidis and APEC O78 infections. These findings further confirmed the feasibility of improving the immunogenicity of OMVs by remodeling the outer membrane and provide a new perspective for the development of broad-spectrum vaccines based on OMVs.
Collapse
Affiliation(s)
- Yuxuan Chen
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China.,The First Clinical Medical College, Nanchang University, Nanchang, China
| | - Kaiwen Jie
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China.,The First Clinical Medical College, Nanchang University, Nanchang, China
| | - Biaoxian Li
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Haiyan Yu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Huan Ruan
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Jing Wu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China.,Key Laboratory of Tumor Pathogenesis and Molecular Pathology, School of Medicine, Nanchang University, Nanchang, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, China.,Key Laboratory of Tumor Pathogenesis and Molecular Pathology, School of Medicine, Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Chaikhumwang P, Kitsongsermthon J, Manopakdee K, Chongcharoen W, Nilubol D, Chanvorachote P, Somparn P, Tantituvanont A. Cationic Polylactic Acid-Based Nanoparticles Improve BSA-FITC Transport Across M Cells and Engulfment by Porcine Alveolar Macrophages. AAPS PharmSciTech 2020; 21:134. [PMID: 32415347 DOI: 10.1208/s12249-020-01689-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/13/2020] [Indexed: 01/14/2023] Open
Abstract
This work described the development of a cationic polylactic acid (PLA)-based nanoparticles (NPs) as an antigen delivery system using dimethyldioctadecylammonium bromide (DDA) to facilitate the engulfment of BSA-FITC by porcine alveolar macrophages (3D4/2 cells) and heat-labile enterotoxin subunit B (LTB) to enhance the transport of BSA-FITC across M cells. The experimental design methodology was employed to study the influence of PLA, polyvinyl alcohol (PVA), DDA, and LTB on the physical properties of the PLA-based NPs. The size of selected cationic PLA NPs comprising 5% PLA, 5% PVA, and 0.6% DDA with or without LTB absorption was range from 367 to 390 nm with a polydispersity index of 0.26, a zeta potential of + 26.00 to + 30.55 mV, and entrapment efficiency of 41.43%. Electron micrographs revealed NPs with spherical shape. The release kinetic of BSA from the NPs followed the Korsmeyer-Peppas kinetics. The cationic PLA NPs with LTB surface absorption showed 3-fold increase in BSA-FITC transported across M cells compared with the NPs without LTB absorption. The uptake studies demonstrated 2-fold increase in BSA-FITC intensity in 3D4/2 cells with cationic NPs as compared with anionic NPs. Overall, the results suggested that LTB decreased the retention time of BSA-FITC loaded in the cationic PLA NPs within the M cells, thus promoting the transport of BSA-FITC across the M cells, and cationic NPs composed of DDA help facilitate the uptake of BSA-FITC in the 3D4/2 cells. Further studies in pigs with respiratory antigens will provide information on the efficacy of cationic PLA NPs as a nasal antigen carrier system.
Collapse
|
9
|
Renu S, Feliciano-Ruiz N, Ghimire S, Han Y, Schrock J, Dhakal S, Patil V, Krakowka S, Renukaradhya GJ. Poly(I:C) augments inactivated influenza virus-chitosan nanovaccine induced cell mediated immune response in pigs vaccinated intranasally. Vet Microbiol 2020; 242:108611. [PMID: 32122615 DOI: 10.1016/j.vetmic.2020.108611] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 02/03/2023]
Abstract
To improve the innate and adaptive immune responses elicited by a killed/inactivated swine influenza virus antigen (KAg)-loaded chitosan nanoparticles (CS NPs-KAg), we used the adjuvant, poly(I:C). The formulated CS NPs-KAg and CS NPs-poly(I:C) had a net surface charge of +30.7 mV and +25.1 mV, respectively. The CS NPs-KAg was coadministered with CS NPs-poly(I:C) (chitosan nanovaccine) as intranasal mist. Vaccinations enhanced homologous (H1N2-OH10) and heterologous (H1N1-OH7) hemagglutination inhibition (HI) titers in both vaccinated and virus-challenged animals compared to the control soluble poly(I:C) vaccinated pigs. In addition, the chitosan nanovaccine induced the proliferation of antigen-specific IFNγ secreting T-helper/memory and γδ T cells compared to control poly(I:C) group; and an increased Th1 (IFNγ, IL-6 and IL-2) and Th2 (IL-10 and IL-13) cytokines mRNA expression in the tracheobronchial lymph nodes compared to lymphoid tissues obtained from pigs given commercial influenza vaccine. The virus load in nasal passages and microscopic lung lesions were partially reduced by both chitosan nanovaccine and commercial vaccine. The HA gene homology between the vaccine and challenge viruses indicated that the chitosan nanovaccine induced a cross-protective immune response. In conclusion, coadministration of CS NPs-poly(I:C) with CS NPs-KAg augmented the cross-reactive specific HI titers and the cell-mediated immune responses in pigs.
Collapse
Affiliation(s)
- Sankar Renu
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH, 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Ninoshkaly Feliciano-Ruiz
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH, 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Shristi Ghimire
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH, 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Yi Han
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH, 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Jennifer Schrock
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH, 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Santosh Dhakal
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH, 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Veerupaxagouda Patil
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH, 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Steven Krakowka
- Emeritus Professor, Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Gourapura J Renukaradhya
- Food Animal Health Research Program, Ohio Agricultural Research and Development Center, 1680 Madison Avenue, Wooster, OH, 44691, USA; Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
10
|
Vemireddy S, Madhurantakam PP, Talati MN, Sampath Kumar HM. Cationic pH-Responsive Polycaprolactone Nanoparticles as Intranasal Antigen Delivery System for Potent Humoral and Cellular Immunity against Recombinant Tetravalent Dengue Antigen. ACS APPLIED BIO MATERIALS 2019; 2:4837-4846. [DOI: 10.1021/acsabm.9b00597] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Sravanthi Vemireddy
- Vaccine Immunology Laboratory, OSPC Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, CSIR−Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Preethi Pallavi Madhurantakam
- Vaccine Immunology Laboratory, OSPC Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, CSIR−Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Mamta N Talati
- Vaccine Immunology Laboratory, OSPC Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, CSIR−Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Halmuthur M. Sampath Kumar
- Vaccine Immunology Laboratory, OSPC Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
- Academy of Scientific and Innovative Research, CSIR−Indian Institute of Chemical Technology, Hyderabad 500007, India
| |
Collapse
|
11
|
Evaluation of CpG-ODN-adjuvanted polyanhydride-based intranasal influenza nanovaccine in pigs. Vet Microbiol 2019; 237:108401. [PMID: 31585639 DOI: 10.1016/j.vetmic.2019.108401] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 01/01/2023]
Abstract
Influenza results in significant economic loss in the swine industry each year. A broadly protective swine influenza vaccine would have the dual benefit of protecting pigs from influenza A viruses (IAVs) and limiting their possible zoonotic transmission to humans. In this study, we developed polyanhydride nanoparticles-based swine influenza vaccine (KAg + CpG-nanovaccine) co-encapsulating inacticated/killed soluble antigen (KAg) and Toll-like receptor (TLR)-9 agonist (CpG-ODN). The immunogenicity and protective efficacy of KAg + CpG-nanovaccine was compared with KAg vaccine containing five-times greater quantity of antigens following heterologous virus challenge. Prime-boost intranasally delivered KAg + CpG-nanovaccine induced significantly higher levels of cross-reactive antigen-specific IgA antibody responses in the nasal cavity, greater lymphoproliferative response in peripheral blood mononuclear cells (PBMCs), and higher IFN-γ secretion during antigen-induced recall responses of PBMCs and tracheobronchial lymph nodes cells compared to those immunized with KAg alone. Importantly, KAg + CpG-nanovaccine provided better protective efficacy through a significant reduction in influenza-induced fever, 16-fold reduction of nasal virus shedding and 80-fold reduction in lung virus titers compared to those immunized with soluble KAg. Our results indicated that CpG-ODN-adjuvanted polyanhydride nanovaccine can induce higher mucosal antibody and cellular immune responses in pigs; and provide better protection as compared with intranasally delivered soluble KAg.
Collapse
|
12
|
Mucosal Vaccination via the Respiratory Tract. Pharmaceutics 2019; 11:pharmaceutics11080375. [PMID: 31374959 PMCID: PMC6723941 DOI: 10.3390/pharmaceutics11080375] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/12/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
Vaccine delivery via mucosal surfaces is an interesting alternative to parenteral vaccine administration, as it avoids the use of a needle and syringe. Mucosal vaccine administration also targets the mucosal immune system, which is the largest lymphoid tissue in the human body. The mucosal immune response involves systemic, antigen-specific humoral and cellular immune response in addition to a local response which is characterised by a predominantly cytotoxic T cell response in combination with secreted IgA. This antibody facilitates pathogen recognition and deletion prior to entrance into the body. Hence, administration via the respiratory mucosa can be favoured for all pathogens which use the respiratory tract as entry to the body, such as influenza and for all diseases directly affecting the respiratory tract such as pneumonia. Additionally, the different mucosal tissues of the human body are interconnected via the so-called “common mucosal immune system”, which allows induction of an antigen-specific immune response in distant mucosal sites. Finally, mucosal administration is also interesting in the area of therapeutic vaccination, in which a predominant cellular immune response is required, as this can efficiently be induced by this route of delivery. The review gives an introduction to respiratory vaccination, formulation approaches and application strategies.
Collapse
|
13
|
Namvarpour M, Tebianian M, Mansouri R, Ebrahimi SM, Kashkooli S. Comparison of different immunization routes on the immune responses induced by Mycobacterium tuberculosis ESAT-6/CFP-10 recombinant protein. Biologicals 2019; 59:6-11. [PMID: 31014910 DOI: 10.1016/j.biologicals.2019.04.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 12/20/2018] [Accepted: 04/14/2019] [Indexed: 01/09/2023] Open
Abstract
According to some difficulties against tuberculosis (TB) vaccination, development of new TB vaccines has been noted in recent years. Selection of proper route for vaccination is one of the most important factors for induction of good immune responses. Hence, in this study, the effects of different administration routes, including intranasal (I.N), subcutaneous (S.C) and intramuscular (I.M) on immune responses against Mycobacterium tuberculosis ESAT-6/CFP-10 recombinant protein has been considered. Recombinant ESAT-6/CFP-10 protein with or without adjuvant (MF59 or cholera toxin B (CTB)) was administered by three routes of I.M, I.N and S.C to mice for three times. Then, the levels of specific antibodies, lymphocyte proliferation and IFN-γ/IL-5 cytokine profile have been carried out to evaluate the humoral and cellular responses. The results showed that the titers of specific antibodies were quickly elevated in S.C and I.M groups after first immunization. Otherwise, the raise of antibody has delay in the I.N immunized animals. The levels of IFN-γ and lymphocyte proliferation have been increased in all of vaccinated groups. However, the I.N immunized mice have lower levels of IL-5 production. Based on our finding, the ESAT-6/CFP-10 recombinant protein is a potent stimulator of immune responses in all of three immunization strategies. However intranasal administration of this antigen has tended to reinforcement of cellular immune responses.
Collapse
Affiliation(s)
- Mozhdeh Namvarpour
- Department of Immunology, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Majid Tebianian
- Department of Biotechnology, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran.
| | - Reza Mansouri
- Department of Immunology, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Seyyed Mahmoud Ebrahimi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, P.O. Box 14155-3651, Tehran, Iran
| | - Shiva Kashkooli
- - Department of Biotechnology, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, (IAUPS), Tehran, Iran
| |
Collapse
|
14
|
Nakahashi-Ouchida R, Yuki Y, Kiyono H. Development of a nanogel-based nasal vaccine as a novel antigen delivery system. Expert Rev Vaccines 2017; 16:1231-1240. [PMID: 29053938 DOI: 10.1080/14760584.2017.1395702] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Nasal vaccination is one of the most effective immunization methods because it can induce effective antigen-specific immune responses not only at the mucosal site of administration but also at distant mucosal surfaces, as well as in the systemic compartment. Based on this advantage, many nasal vaccines are being developed and some have been licensed and marketed for clinical use. However, some have been withdrawn because of unacceptable adverse events such as inactivated influenza vaccine administrated with a heat-labile enterotoxin of Escherichia coli as an adjuvant. Thus, it is important to consider both the efficacy and safety of nasal vaccines. Areas covered: This review describes the benefits of cholesteryl group-bearing pullulan (CHP) nanogels for nasal vaccine delivery and vaccine development identified on Pubmed database with the term 'Nanogel-based nasal vaccine'. Expert commentary: CHP nanogels have been developed as novel drug delivery system, and a cationic CHP nanogels have been demonstrated to induce effective immunity as a nasal vaccine antigen carrier. Since vaccine antigens incorporated into CHP nanogels have exhibited no brain deposition after nasal administration in mice and nonhuman primates, the vaccine seems safe, and could be a promising new delivery system.
Collapse
Affiliation(s)
- Rika Nakahashi-Ouchida
- a Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science , University of Tokyo , Tokyo , Japan
| | - Yoshikazu Yuki
- a Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science , University of Tokyo , Tokyo , Japan
| | - Hiroshi Kiyono
- a Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science , University of Tokyo , Tokyo , Japan.,b International Research and Development Center for Mucosal Vaccine, The Institute of Medical Science , The University of Tokyo , Tokyo , Japan.,c Department of Immunology, Graduate School of Medicine , Chiba University , Chiba , Japan
| |
Collapse
|
15
|
Li Y, Li M, Gong T, Zhang Z, Sun X. Antigen-loaded polymeric hybrid micelles elicit strong mucosal and systemic immune responses after intranasal administration. J Control Release 2017; 262:151-158. [PMID: 28756271 DOI: 10.1016/j.jconrel.2017.07.034] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 06/26/2017] [Accepted: 07/25/2017] [Indexed: 02/01/2023]
Abstract
Increasing attention has been paid to nasal delivery. Subunit vaccines based on antigenic proteins or polypeptides offer good safety. However, lack of delivery efficiency, particularly for nasal immunization, is a big issue. Here we designed a highly tunable polymeric hybrid micelle (PHM) system offering good vaccine efficacy after nasal administration. PHMs are formulated from two amphiphilic diblock copolymers, polycaprolactone-polyethylenimine (PCL-PEI) and polycaprolactone-polyethyleneglycol (PCL-PEG), the ratio of which determines PHM physicochemical properties. Citraconic anhydride-modified ovalbumin (Cit-OVA), as model antigen, was incorporated into PHMs via electrostatic interaction, giving antigen-loaded micelles of around 150nm in size. Their surface characteristics which are found closely related to their in vivo kinetics can be modulated by adjusting the mass ratio of PCL-PEG and PCL-PEI. PHM/Cit-OVA complexes containing PCL-PEI and PCL-PEG in a 1:1 mass ratio induced strong immune responses in nasal mucosa and serum in vivo without causing obvious toxicity, and Cit-OVA was efficiently internalized by dendritic cells. These results demonstrate the promise of this multifunctional polymeric delivery system for nasal vaccination.
Collapse
Affiliation(s)
- You Li
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Man Li
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Tao Gong
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China
| | - Xun Sun
- Key Laboratory of Drug Targeting, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, People's Republic of China.
| |
Collapse
|
16
|
Giebink GS, Bakaletz LO, Barenkamp SJ, Green B, Gu XX, Heikkinen T, Hotomi M, Karma P, Kurono Y, Kyd JM, Murphy TF, Ogra PL, Patel JA, Pelton SI. 6. Vaccine. Ann Otol Rhinol Laryngol 2016. [DOI: 10.1177/00034894051140s110] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
17
|
Liu Q, Liu Q, Yi J, Liang K, Hu B, Zhang X, Curtiss R, Kong Q. Outer membrane vesicles from flagellin-deficient Salmonella enterica serovar Typhimurium induce cross-reactive immunity and provide cross-protection against heterologous Salmonella challenge. Sci Rep 2016; 6:34776. [PMID: 27698383 PMCID: PMC5048178 DOI: 10.1038/srep34776] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 09/16/2016] [Indexed: 01/12/2023] Open
Abstract
Outer membrane vesicles (OMVs) isolated from Salmonella Typhimurium are potentially useful for developing subunit vaccines because of high immunogenicity and protective efficacy. However, flagella might remain in OMV pellets following OMV purification, resulting in non-essential immune responses and counteraction of bacterial protective immune responses when developing a vaccine against infection of multiple serotypes Salmonella. In this study, a flagellin-deficient S. Typhimurium mutant was constructed. Lipopolysaccharide profiles, protein profiles and cryo-electron microscopy revealed that there were no significant differences between the wild-type and mutant OMVs, with the exception of a large amount of flagellin in the wild-type OMVs. Neither the wild-type OMVs nor the non-flagellin OMVs were toxic to macrophages. Mice immunized with the non-flagellin OMVs produced high concentrations of IgG. The non-flagellin OMVs elicited strong mucosal antibody responses in mice when administered via the intranasal route in addition to provoking higher cross-reactive immune responses against OMPs isolated from S. Choleraesuis and S. Enteritidis. Both intranasal and intraperitoneal immunization with the non-flagellin OMVs provided efficient protection against heterologous S. Choleraesuis and S. Enteritidis challenge. Our results indicate that the flagellin-deficient OMVs may represent a new vaccine platform that could be exploited to facilitate the production of a broadly protective vaccine.
Collapse
Affiliation(s)
- Qiong Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.,Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287-5401, USA.,Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, 330006, China
| | - Qing Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jie Yi
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Kang Liang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China
| | - Bo Hu
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Xiangmin Zhang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, 48202, USA
| | - Roy Curtiss
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287-5401, USA
| | - Qingke Kong
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, China.,Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ, 85287-5401, USA
| |
Collapse
|
18
|
Liu Q, Liu Q, Yi J, Liang K, Liu T, Roland KL, Jiang Y, Kong Q. Outer membrane vesicles derived from Salmonella Typhimurium mutants with truncated LPS induce cross-protective immune responses against infection of Salmonella enterica serovars in the mouse model. Int J Med Microbiol 2016; 306:697-706. [PMID: 27578609 DOI: 10.1016/j.ijmm.2016.08.004] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 08/21/2016] [Accepted: 08/24/2016] [Indexed: 11/25/2022] Open
Abstract
Salmonella enterica cause diarrheal and systemic diseases and are of considerable concern worldwide. Vaccines that are cross-protective against multiple serovars could provide effective control of Salmonella-mediated diseases. Bacteria-derived outer membrane vesicles (OMVs) are highly immunogenic and are capable of eliciting protective immune responses. Alterations in lipopolysaccharide (LPS) length can result in outer membrane remodeling and composition of outer membrane proteins (OMPs) changing. In this study, we investigated the impact of truncated LPS on both the production and immunogenicity of Salmonella OMVs, including the ability of OMVs to elicit cross-protection against challenge by heterologous Salmonella strains. We found that mutations in waaJ and rfbP enhanced vesiculation, while mutations in waaC, waaF and waaG inhibited this process. Animal experiments indicated that OMVs from waaC, rfaH and rfbP mutants induced stronger serum immune responses compared to OMVs from the parent strain, while all elicited protective responses against the wild-type S. Typhimurium challenge. Furthermore, intranasal or intraperitoneal immunization with OMVs derived from the waaC and rfbP mutants elicited significantly higher cross-reactive IgG responses and provided enhanced cross-protection against S. Choleraesuis and S. Enteritidis challenge than the wild-type OMVs. These results indicate that truncated-LPS OMVs are capable of conferring cross protection against multiple serotypes of Salmonella infection.
Collapse
Affiliation(s)
- Qiong Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, 611130 Chengdu, China; Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, USA; Department of Medical Microbiology, School of Medicine, Nanchang University, 330006 Nanchang, China
| | - Qing Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, 611130 Chengdu, China
| | - Jie Yi
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, 611130 Chengdu, China
| | - Kang Liang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, 611130 Chengdu, China
| | - Tian Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, 611130 Chengdu, China
| | - Kenneth L Roland
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, USA
| | - Yanlong Jiang
- College of Animal Science and Technology, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Agricultural University, 130118 Changchun, China
| | - Qingke Kong
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, 611130 Chengdu, China; Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ 85287-5401, USA.
| |
Collapse
|
19
|
Trows S, Scherließ R. Carrier-based dry powder formulation for nasal delivery of vaccines utilizing BSA as model drug. POWDER TECHNOL 2016. [DOI: 10.1016/j.powtec.2016.01.042] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
20
|
Liu Q, Zhang C, Zheng X, Shao X, Zhang X, Zhang Q, Jiang X. Preparation and evaluation of antigen/N-trimethylaminoethylmethacrylate chitosan conjugates for nasal immunization. Vaccine 2014; 32:2582-90. [PMID: 24681230 DOI: 10.1016/j.vaccine.2014.03.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 02/22/2014] [Accepted: 03/13/2014] [Indexed: 11/18/2022]
Abstract
The frequent outbreak of respiratory infectious diseases such as influenza and pulmonary tuberculosis calls for new immunization strategies with high effectiveness. Nasal immunization is one of the most potential methods to prevent the diseases infected through the respiratory tract. In this study, we designed a water-soluble system based on antigen/N-trimethylaminoethylmethacrylate chitosan conjugates for nasal immunization. N-trimethylaminoethylmethacrylate chitosan (TMC) was synthesized by free radical polymerization of chitosan and N-trimethylaminoethylmethacrylate chloride and identified by (1)H NMR and FT-IR. Thiolated ovalbumin (OVA) was covalently conjugated to maleimide modified TMC with high conjugation efficiency. OVA conjugated TMC (OVA-TMC) significantly increased uptake of OVA by Raw 264.7 cells, which was 2.38 times higher than that of OVA/TMC physical mixture (OVA+TMC) at 4h. After nasal administration, OVA-TMC showed higher transport efficiency to superficial and deep cervical lymph nodes than OVA+TMC or OVA alone. Balb/C mice were intranasally given with OVA-TMC three times at 2-week internals to evaluate the immunological effect. The serum IgG, IgG1 and IgG2a levels of the OVA-TMC group were 17.9-87.9 times higher than that of the OVA+TMC group and comparable to that of the intramuscular group. The secretory IgA levels in nasal wash and saliva of the OVA-TMC group were 5.2-7.1 times higher than that of the OVA+TMC group while the secretory IgA levels of the intramuscular alum-precipitated OVA group were not increased. After immunofluorescence staining of nasal cavity, IgA antibody secreting cells were mainly observed in the lamina propria regions and glands of nasal mucosa. OVA-TMC showed little toxicity to the nasal epithelia or cilia of rats after nasal administration for three consecutive days. These results demonstrated that antigen conjugated TMC can induce both systemic and mucosal immune responses after nasal administration and may serve as a convenient, safe and effective vaccine for preventing respiratory infectious diseases.
Collapse
Affiliation(s)
- Qingfeng Liu
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, 826 ZhangHeng Rd., Shanghai 201203, PR China
| | - Chi Zhang
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, 826 ZhangHeng Rd., Shanghai 201203, PR China
| | - Xiaoyao Zheng
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, 826 ZhangHeng Rd., Shanghai 201203, PR China
| | - Xiayan Shao
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, 826 ZhangHeng Rd., Shanghai 201203, PR China
| | - Xi Zhang
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, 826 ZhangHeng Rd., Shanghai 201203, PR China
| | - Qizhi Zhang
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, 826 ZhangHeng Rd., Shanghai 201203, PR China.
| | - Xinguo Jiang
- Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, 826 ZhangHeng Rd., Shanghai 201203, PR China
| |
Collapse
|
21
|
Vyas SP, Gupta PN. Implication of nanoparticles/microparticles in mucosal vaccine delivery. Expert Rev Vaccines 2014; 6:401-18. [PMID: 17542755 DOI: 10.1586/14760584.6.3.401] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Although polymeric nanoparticles/microparticles are well established for the mucosal administration of conventional drugs, they have not yet been developed commercially for vaccine delivery. The limitation of the mucosal (particularly oral) route of delivery, including low pH, gastric enzymes, rapid transit and poor absorption of large molecules, has made mucosal vaccine delivery challenging. Nevertheless, several polymeric delivery systems for mucosal vaccine delivery are currently being evaluated. The polymer-based approaches are designed to protect the antigen in the gut, to target the antigen to the gut-associated lymphoid tissue or to increase the residence time of the antigen in the gut through bioadhesion. M-cell targeting is a potential approach for mucosal vaccine delivery, which can be achieved using M-cell-specific lectins, microbial adhesins or immunoglobulins. While many hurdles must be overcome before targeted mucosal vaccine delivery becomes a practical reality, this is a potential area of research that has important implications for future vaccine development. This review comprises various aspects that could be decisive in the development of polymer based mucosal vaccine delivery systems.
Collapse
Affiliation(s)
- Suresh P Vyas
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar-470003 (M.P.), India.
| | | |
Collapse
|
22
|
Sasaki H, Ishikawa H, Kojima K, Itoh M, Matsumoto T, Itoh T, Hosomi O, Kawamoto E. Intranasal immunization with a non-adjuvanted adhesive protein descended from Pasteurella pneumotropica and its preventive efficacy against opportunistic infection in mice. Vaccine 2013; 31:5729-35. [DOI: 10.1016/j.vaccine.2013.09.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 08/25/2013] [Accepted: 09/19/2013] [Indexed: 12/14/2022]
|
23
|
Zhao K, Chen G, Shi XM, Gao TT, Li W, Zhao Y, Zhang FQ, Wu J, Cui X, Wang YF. Preparation and efficacy of a live newcastle disease virus vaccine encapsulated in chitosan nanoparticles. PLoS One 2012; 7:e53314. [PMID: 23285276 PMCID: PMC3532065 DOI: 10.1371/journal.pone.0053314] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 11/30/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Newcastle disease (ND) is a highly contagious viral disease of poultry caused by pathogenic strains of the Newcastle disease virus (NDV). Live NDV vaccines are administered by drinking water, eyedrops or coarse aerosol spray. To further enhance mucosal immune responses, chitosan nanoparticles were developed for the mucosal delivery of a live NDV vaccine. METHODOLOGY/PRINCIPAL FINDINGS A lentogenic live-virus vaccine (strain LaSota) against NDV encapsulated in chitosan nanoparticles were developed using an ionic crosslinking method. Chitosan nanoparticles containing the lentogenic live-virus vaccine against NDV (NDV-CS-NPs) were produced with good morphology, high stability, a mean diameter of 371.1 nm, an encapsulation rate of 77% and a zeta potential of +2.84 mV. The Western blotting analysis showed that NDV structural proteins were detected in NDV-CS-NPs. The virus release assay results of NDV-CS-NPs indicated that NDV was released from NDV-CS-NPs. Chickens immunized orally or intranasally with NDV-CS-NPs were fully protected whereas one out of five chickens immunized with the LaSota live NDV vaccine and three out of five chickens immunized with the inactivated NDV vaccine were dead after challenge with the highly virulent NDV strain F48E9. CONCLUSIONS/SIGNIFICANCE NDV-CS-NPs induced better protection of immunized specific pathogen free chickens compared to the live NDV vaccine strain LaSota and the inactivated NDV vaccine. This study lays a foundation for the further development of mucosal vaccines and drugs encapsulated in chitosan nanoparticles.
Collapse
Affiliation(s)
- Kai Zhao
- Laboratory of Microbiology, College of Life Science, Heilongjiang University, Harbin, China
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS, Harbin, China
- * E-mail: (YFW); (KZ); (XC)
| | - Gang Chen
- Laboratory of Microbiology, College of Life Science, Heilongjiang University, Harbin, China
| | - Xing-ming Shi
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Ting-ting Gao
- Laboratory of Microbiology, College of Life Science, Heilongjiang University, Harbin, China
| | - Wei Li
- Laboratory of Microbiology, College of Life Science, Heilongjiang University, Harbin, China
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Yan Zhao
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS, Harbin, China
| | - Feng-qiang Zhang
- Laboratory of Microbiology, College of Life Science, Heilongjiang University, Harbin, China
| | - Jin Wu
- Laboratory of Microbiology, College of Life Science, Heilongjiang University, Harbin, China
| | - Xianlan Cui
- Animal Health Laboratory, Department of Primary Industries, Parks, Water and Environment, Tasmania, Australia
- * E-mail: (YFW); (KZ); (XC)
| | - Yun-Feng Wang
- Division of Avian Infectious Diseases, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, CAAS, Harbin, China
- * E-mail: (YFW); (KZ); (XC)
| |
Collapse
|
24
|
Intranasal administration of an inactivated Yersinia pestis vaccine with interleukin-12 generates protective immunity against pneumonic plague. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1925-35. [PMID: 21880856 DOI: 10.1128/cvi.05117-11] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Inhalation of Yersinia pestis causes pneumonic plague, which rapidly progresses to death. A previously licensed killed whole-cell vaccine is presently unavailable due to its reactogenicity and inconclusive evidence of efficacy. The present study now shows that vaccination intranasally (i.n.) with inactivated Y. pestis CO92 (iYp) adjuvanted with interleukin-12 (IL-12) followed by an i.n. challenge with a lethal dose of Y. pestis CO92 prevented bacterial colonization and protected 100% of mice from pneumonic plague. Survival of the vaccinated mice correlated with levels of systemic and lung antibodies, reduced pulmonary pathology and proinflammatory cytokines, and the presence of lung lymphoid cell aggregates. Protection against pneumonic plague was partially dependent upon Fc receptors and could be transferred to naïve mice with immune mouse serum. On the other hand, protection was not dependent upon complement, and following vaccination, depletion of CD4 and/or CD8 T cells before challenge did not affect survival. In summary, the results demonstrate the safety, immunogenicity, and protective efficacy of i.n. administered iYp plus IL-12 in a mouse model of pneumonic plague.
Collapse
|
25
|
Advances in effective vaccine development against hepatitis B: focus on mucosal vaccine delivery strategies. Ther Deliv 2010; 1:397-410. [DOI: 10.4155/tde.10.19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hepatitis B virus causes chronic necroinflammatory liver disease, which is known as hepatitis B. This inflammatory condition may further aggravate liver cirrhosis or hepatocellular carcinoma. Currently available conventional hepatitis B vaccine contains one of the viral envelope proteins, hepatitis B surface antigen, which develops a humoral immune response and hence protects against the infection. However, it fails in developing the desired cellular immune response, which is one of the most important bioresponses contributing to virus elimination from infected hepatocytes. At the same time, moderate humoral response developed following conventional vaccination do not protect the mucosal surfaces through serosal response. The mucosa is a predominant entry site for most of the infectious pathogens. Several strategies, including the use of adjuvants, development of surface functionalized novel antigen carriers and mucosal immunization for example, have been explored to investigate their role in addressing the limitations associated with the current hepatitis B vaccine. This review focuses on recent advances that have been made in order to develop an effective vaccine against hepatitis B.
Collapse
|
26
|
Hall LJ, Clare S, Dougan G. NK cells influence both innate and adaptive immune responses after mucosal immunization with antigen and mucosal adjuvant. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:4327-37. [PMID: 20220095 PMCID: PMC3517975 DOI: 10.4049/jimmunol.0903357] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
NK cells were found to be recruited in a temporally controlled manner to the nasal-associated lymphoid tissue and the cervical lymph nodes of mice after intranasal immunization with Ag85B-early secreted antigenic target 6 kDa from Mycobacterium tuberculosis mixed with Escherichia coli heat-labile toxin as adjuvant. These NK cells were activated and secreted a diverse range of cytokines and other immunomodulators. Using Ab depletion targeting anti-asialo GM1, we found evidence for altered trafficking, impaired activation, and cytokine secretion of dendritic cells, macrophages, and neutrophils in immunized NK cell-depleted mice compared with control animals. Analysis of Ag-specific immune responses revealed an attenuated Ab and cytokine response in immunized NK cell-depleted animals. Systemic administration of rIL-6 but not rIFN-gamma significantly restored immune responses in mice depleted of NK cells. In conclusion, cytokine production, particularly IL-6, via NK cells and NK cell-activated immune populations plays an important role in the establishment of local innate immune responses and the consequent development of adaptive immunity after mucosal immunization.
Collapse
Affiliation(s)
- Lindsay J Hall
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, United Kingdom.
| | | | | |
Collapse
|
27
|
NALT (nasal cavity-associated lymphoid tissue) in the rabbit. Vet Immunol Immunopathol 2010; 133:212-8. [DOI: 10.1016/j.vetimm.2009.08.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2009] [Revised: 08/06/2009] [Accepted: 08/10/2009] [Indexed: 11/19/2022]
|
28
|
Kim SB, Han YW, Rahman MM, Kim SJ, Yoo DJ, Kang SH, Kim K, Eo SK. Modulation of protective immunity against herpes simplex virus via mucosal genetic co-transfer of DNA vaccine with beta2-adrenergic agonist. Exp Mol Med 2010; 41:812-23. [PMID: 19641376 DOI: 10.3858/emm.2009.41.11.087] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Cholera toxin, which has been frequently used as mucosal adjuvant, leads to an irreversible activation of adenylyl cyclase, thereby accumulating cAMP in target cells. Here, it was assumed that beta(2)-adrenergic agonist salbutamol may have modulatory functions of immunity induced by DNA vaccine, since beta(2)-adrenergic agonists induce a temporary cAMP accumulation. To test this assumption, the present study evaluated the modulatory functions of salbutamol co-administered with DNA vaccine expressing gB of herpes simplex virus (HSV) via intranasal (i.n.) route. We found that the i.n. co-administration of salbutamol enhanced gB-specific IgG and IgA responses in both systemic and mucosal tissues, but optimal dosages of co-administered salbutamol were required to induce maximal immune responses. Moreover, the mucosal co-delivery of salbutamol with HSV DNA vaccine induced Th2-biased immunity against HSV antigen, as evidenced by IgG isotypes and Th1/Th2-type cytokine production. The enhanced immune responses caused by co-administration of salbutamol provided effective and rapid responses to HSV mucosal challenge, thereby conferring prolonged survival and reduced inflammation against viral infection. Therefore, these results suggest that salbutamol may be an attractive adjuvant for mucosal genetic transfer of DNA vaccine.
Collapse
Affiliation(s)
- Seong Bum Kim
- Laboratory of Microbiology, College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Jeonju 561-756, Korea
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Liposome in situ gelling system: Novel carrier based vaccine adjuvant for intranasal delivery of recombinant protein vaccine. ACTA ACUST UNITED AC 2009. [DOI: 10.1016/j.provac.2009.07.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
30
|
Plasmid DNA loaded chitosan nanoparticles for nasal mucosal immunization against hepatitis B. Int J Pharm 2008; 354:235-41. [DOI: 10.1016/j.ijpharm.2007.11.027] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2007] [Revised: 11/01/2007] [Accepted: 11/17/2007] [Indexed: 11/19/2022]
|
31
|
Khatri K, Goyal AK, Gupta PN, Mishra N, Mehta A, Vyas SP. Surface modified liposomes for nasal delivery of DNA vaccine. Vaccine 2008; 26:2225-33. [PMID: 18396362 DOI: 10.1016/j.vaccine.2008.02.058] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 02/14/2008] [Accepted: 02/21/2008] [Indexed: 10/22/2022]
Abstract
The aim of the present work was to investigate the potential utility of glycol chitosan coated liposomes as nasal vaccine delivery vehicle for eliciting viral specific humoral mucosal and cellular immune responses. Plasmid pRc/CMV-HBs(S) encapsulated liposomes were prepared by dehydration-rehydration method and subsequently coated with glycol chitosan by simple incubation method. Liposomes were then characterized for their size, surface charge, entrapment efficiency, and ability to protect encapsulated DNA against nuclease digestion and for their mucoadhesiveness. The liposomes were then administered to mice in order to study their feasibility as nasal vaccine carriers. The developed liposomes possessed +9.8 mV zeta potential and an average vesicle size less than 1 microm and entrapment efficiency of approximately 53%. Following intranasal administration, glycol chitosan coated liposomes elicited humoral mucosal and cellular immune responses that were significant as compared to naked DNA justifying the potential advantage of mucosal vaccination in the production of local antibodies at the sites where pathogens enters the body.
Collapse
Affiliation(s)
- Kapil Khatri
- Drug Delivery Research Laboratory, Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya, Sagar 470003, M.P., India
| | | | | | | | | | | |
Collapse
|
32
|
Alcón VL, Baca-Estrada M, Vega-López MA, Willson P, Babiuk LA, Kumar P, Foldvari M. Intranasal immunization using biphasic lipid vesicles as delivery systems for OmlA bacterial protein antigen and CpG oligonucleotides adjuvant in a mouse model. J Pharm Pharmacol 2006; 57:955-62. [PMID: 16102250 DOI: 10.1211/0022357056695] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The nasal mucosa is an important arm of the mucosal system since it is often the first point of contact for inhaled antigens. The ineffectiveness of the simple delivery of soluble antigens to mucosal membranes for immunization has stimulated extensive studies in appropriate delivery systems and adjuvants. We have evaluated biphasic lipid vesicles as a novel intranasal (i.n.) delivery system (designated as vaccine targeting adjuvant, VTA) containing bacterial antigens and CpG oligodeoxynucleotides (ODNs). Results show that administration of antigen and CpG ODNs in biphasic lipid vesicles resulted in greater induction of IgA levels in serum (P< 0.05) and mucosal antibody responses such as IgA in nasal secretions and lung (P< 0.01) after immunization with a combined subcutaneous (s.c.)/i.n. as compared to s.c./s.c. approach. Based on antibody responses, VTA formulations were found to be suitable as delivery systems for antigens and CpG ODNs by the intranasal route, resulting in a Th2-type of immune response, characterized by IgG1 and IL-4 production at the systemic level.
Collapse
Affiliation(s)
- V L Alcón
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Canada
| | | | | | | | | | | | | |
Collapse
|
33
|
Vila A, Sánchez A, Janes K, Behrens I, Kissel T, Vila Jato JL, Alonso MJ. Low molecular weight chitosan nanoparticles as new carriers for nasal vaccine delivery in mice. Eur J Pharm Biopharm 2005; 57:123-31. [PMID: 14729088 DOI: 10.1016/j.ejpb.2003.09.006] [Citation(s) in RCA: 287] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
High molecular weight (Mw) chitosan (CS) solutions have already been proposed as vehicles for nasal immunization. The aim of the present work was to investigate the potential utility of low Mw CS in the form of nanoparticles as new long-term nasal vaccine delivery vehicles. For this purpose, CS of low Mws (23 and 38 kDa) was obtained previously by a depolymerization process of the commercially available CS (70 kDa). Tetanus toxoid (TT), used as a model antigen, was entrapped within CS nanoparticles by an ionic cross-linking technique. TT-loaded nanoparticles were first characterized for their size, electrical charge, loading efficiency and in vitro release of antigenically active toxoid. The nanoparticles were then administered intranasally to conscious mice in order to study their feasibility as vaccine carriers. CS nanoparticles were also labeled with FITC-BSA and their interaction with the rat nasal mucosa examined by confocal laser scanning microcopy (CLSM). Irrespective of the CS Mw, the nanoparticles were in the 350 nm size range, and exhibited a positive electrical charge (+40 mV) and associated TT quite efficiently (loading efficiency: 50-60%). In vitro release studies showed an initial burst followed by an extended release of antigenically active toxoid. Following intranasal administration, TT-loaded nanoparticles elicited an increasing and long-lasting humoral immune response (IgG concentrations) as compared to the fluid vaccine. Similarly, the mucosal response (IgA levels) at 6 months post-administration of TT-loaded CS nanoparticles was significantly higher than that obtained for the fluid vaccine. The CLSM images indicated that CS nanoparticles can cross the nasal epithelia and, hence, transport the associated antigen. Interestingly, the ability of these nanoparticles to provide improved access to the associated antigen to the immune system was not significantly affected by the CS Mw. Indeed, high and long-lasting responses could be obtained using low Mw CS molecules. Furthermore, the response was not influenced by the CS dose (70-200 microg), achieving a significant response for a very low CS dose. In conclusion, nanoparticles made of low Mw CS are promising carriers for nasal vaccine delivery.
Collapse
Affiliation(s)
- Ana Vila
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Santiago de Compostela, Spain
| | | | | | | | | | | | | |
Collapse
|
34
|
Vila A, Sánchez A, Evora C, Soriano I, Vila Jato JL, Alonso MJ. PEG-PLA nanoparticles as carriers for nasal vaccine delivery. ACTA ACUST UNITED AC 2004; 17:174-85. [PMID: 15294069 DOI: 10.1089/0894268041457183] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
This report presents an overview of the potential of nanoparticles as nasal carriers for drug/vaccine administration. In addition, this report shows, for the first time, the efficacy of polylactic acid nanoparticles coated with a hydrophilic polyethyleneglycol coating (PEG-PLA nanoparticles) as carriers for the nasal transport of bioactive compounds. For this purpose, tetanus toxoid (TT), a high molecular weight protein (Mw 150,000 Da), was chosen as a model antigen and encapsulated in the PEG-PLA nano- and microparticles (200 nm and 1.5 microm respectively). These nanosystems were first characterized for their stability in the presence of lysozyme and also for their size, electrical charge, loading efficiency, in vitro release of antigenically active toxoid and afterwards, these formulations were administered intranasally to mice and the systemic and mucosal anti-tetanus responses were evaluated for up to 24 weeks. Additionally, PEG-PLA particles labeled with rhodamine 6G were administered intranasally to rats in order to visualize their interaction with the nasal mucosae by fluorescence microscopy. Their behavior was compared with that of the well known PLA nanoparticles (200 nm). The results showed that PLA nanoparticles suffered an immediate aggregation upon incubation with lysozyme, whereas the PEG-coated nanoparticles remained totally stable. The antibody levels elicited following i.n. administration of PEG-coated nanoparticles were significantly higher than those corresponding to PLA nanoparticles. Furthermore, PEG-PLA nanoparticles generated an increasing and a long lasting response. The qualitative fluorescence microscopy studies revealed that PEG-PLA particles are able to cross the rat nasal epithelium. These studies indicate that the PEG coating around the particles has a role in stabilizing PLA particles in mucosal fluids and that it facilitates the transport of the nanoencapsulated antigen, hence eliciting a high and long lasting immune response.
Collapse
Affiliation(s)
- A Vila
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, Santiago de Compostela, Spain
| | | | | | | | | | | |
Collapse
|
35
|
Rial A, Lens D, Betancor L, Benkiel H, Silva JS, Chabalgoity JA. Intranasal immunization with a colloid-formulated bacterial extract induces an acute inflammatory response in the lungs and elicits specific immune responses. Infect Immun 2004; 72:2679-88. [PMID: 15102776 PMCID: PMC387843 DOI: 10.1128/iai.72.5.2679-2688.2004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nonspecific stimulation of lung defenses by repeated oral administration of immunomodulators, such as bacterial extracts, has shown potential for the prevention of respiratory tract infections. Here, we show that intranasal (i.n.) immunization with a bacterial extract formulated as a colloid induces an acute inflammatory response in the lungs characterized by increased production of CCL and CXCL chemokines and a major influx of dendritic cells (DCs) and neutrophils, with a higher proportion of DCs showing an activated phenotype (high CD80/CD86 expression). Cytokine levels measured in bronchoalveolar-lavage samples showed a small increase in the production of tumor necrosis factor alpha and similar levels of the other cytokines measured (interleukin 10 [IL-10], IL-12, and gamma interferon [IFN-gamma]) in immunized mice compared with control mice. However, the recall response of primed animals after antigenic challenge induced increased expression of IL-12 and IFN-gamma mRNAs in lung homogenates. Overall, all these effects were not due to the lipopolysaccharide content in the bacterial extract. Furthermore, we found that three i.n. doses administered 2 to 3 weeks apart were enough to elicit long-lasting specific serum immunoglobulin G (IgG) and secretory IgA antibody responses. Assessment of IgG subclasses showed a balanced pattern of IgG1-IgG2a responses. The serum total IgE concentrations were also elevated in immunized mice 2 weeks after the third dose, but they significantly decreased soon afterwards. Our results suggest that simple formulations of bacterial extracts administered i.n. are highly immunogenic, eliciting local and systemic immune responses, and may serve as the basis for cost-effective immunotherapies for the prevention and treatment of respiratory infections.
Collapse
Affiliation(s)
- A Rial
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Montevideo, Uruguay
| | | | | | | | | | | |
Collapse
|
36
|
Jurcisek JA, Durbin JE, Kusewitt DF, Bakaletz LO. Anatomy of the nasal cavity in the chinchilla. Cells Tissues Organs 2004; 174:136-52. [PMID: 12835577 DOI: 10.1159/000071154] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2003] [Indexed: 11/19/2022] Open
Abstract
There is currently great interest worldwide in developing noninvasive methods for the delivery of vaccines for upper respiratory tract diseases, including middle ear infection (otitis media, OM). One such noninvasive approach believed to have great potential for the prevention of diseases of the airway is to deliver vaccines by the intranasal (i.n.) route. Induction of a local, mucosal immune response in the upper respiratory tract, and particularly in the nasopharynx, would be a highly efficacious approach to prevention of OM. The chinchilla is the preferred rodent host for studying OM. However, although the anatomy of the chinchilla vomeronasal organ, inner ear, middle ear and Eustachian tube have been well-studied, to date there have been no reports in the literature of a similar complete analysis of the nasopharynx and nasal cavities of the chinchilla. In order to develop a relevant animal model of i.n. delivery as a potential immunization approach for the prevention of OM and to use these models for preclinical assessments of various vaccine candidates, it was important that we better understand the anatomy of the chinchilla nasal cavities and nasopharynx. Our anatomical studies revealed that the naso- and maxilloturbinates of the chinchilla nasal cavity more closely resemble the simple turbinates found in other rodents rather than the branched or complex turbinates seen in dogs, cats, and rabbits thus facilitating the i.n. delivery of vaccine candidates. The chinchilla nasal mucosa also contains numerous lymphoid aggregates like that of other rodents. Our findings thus suggest that we will be able to deliver i.n. vaccines effectively to chinchillas and that these vaccines will likely be able to induce specific immune responses.
Collapse
Affiliation(s)
- Joseph A Jurcisek
- Department of Pediatrics, Ohio State University College of Medicine and Public Health, Columbus, Ohio 43205-2696, USA
| | | | | | | |
Collapse
|
37
|
Sfeir RM, Dubarry M, Boyaka PN, Rautureau M, Tomé D. The mode of oral bovine lactoferrin administration influences mucosal and systemic immune responses in mice. J Nutr 2004; 134:403-9. [PMID: 14747680 DOI: 10.1093/jn/134.2.403] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Food protein intake interacts with the immune system. In earlier nutritional and immunological studies, nutrients, particularly milk whey proteins, were generally administered in soluble form and by gavage. However, orogastric intubation does not represent a natural way of ingesting nutrients such as lactoferrin (Lf). We examined how different modes of oral administration of Lf could affect the regulatory effect of this molecule on intestinal and systemic immune responses. Groups of 10 female BALB/c mice were administered Lf daily for 6 wk. To address the influence of the oral modes of administration, mice were given Lf either in solution, by gastric intubation or in the drinking water, or as a powder, by buccal deposition or in the diet. Mucosal and systemic immune responses, including specific immunoglobulin (Ig) secretion, cell proliferation, and cytokine production, were analyzed and compared with those of naïve mice given water under the same conditions or positive control mice that were administered Lf by i.m. injection. The addition of Lf to the drinking water had no visible effect on the immune status. Gastric intubation, single buccal doses, and continuous doses of Lf in the diet stimulated transient systemic and intestinal antibody responses against Lf. All of these oral modes of Lf exposure biased mucosal and systemic T-cell responses toward Thelper (Th)2-types and elevated IgA production by mucosal cells. However, the less natural gastric intubation also promoted Th1-type responses as evidenced by serum IgG(2a) antibodies and the secretion of Th1 cytokine by mucosal and systemic T cells in vitro. Thus, one should carefully consider the oral mode of administration for understanding regulation of immune responses by food proteins such as Lf.
Collapse
Affiliation(s)
- Rose Mary Sfeir
- Unité INRA 914 Physiologie de la Nutrition et du Comportement Alimentaire, Institut National Agronomique Paris-Grignon, F-75231 Paris Cedex 05, France
| | | | | | | | | |
Collapse
|
38
|
Beignon AS, Briand JP, Rappuoli R, Muller S, Partidos CD. The LTR72 mutant of heat-labile enterotoxin of Escherichia coli enhances the ability of peptide antigens to elicit CD4(+) T cells and secrete gamma interferon after coapplication onto bare skin. Infect Immun 2002; 70:3012-9. [PMID: 12010992 PMCID: PMC128003 DOI: 10.1128/iai.70.6.3012-3019.2002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2001] [Revised: 01/23/2002] [Accepted: 03/11/2002] [Indexed: 01/08/2023] Open
Abstract
Application of antigens with an adjuvant onto bare skin is a needle-free and pain-free immunization procedure that delivers antigens to the immunocompetent cells of the epidermis. We tested here the immunogenicity and adjuvanticity of two mutants of heat-labile enterotoxin (LT) of Escherichia coli, LTK63 and LTR72. Both mutants were shown to be immunogenic, inducing serum and mucosal antibody responses. The application of LTK63 and LTR72 to bare skin induced significant protection against intraperitoneal challenge with a lethal dose of LT. In addition, both LT mutants enhanced the capacity of peptides TT:830-843 and HA:307-319 (representing T-helper epitopes from tetanus toxin and influenza virus hemagglutinin, respectively) to elicit antigen-specific CD4(+) T cells after coapplication onto bare skin. However, only mutant LTR72 was capable of stimulating the secretion of high levels of gamma interferon. These findings demonstrate that successful skin immunization protocols require the selection of the right adjuvant in order to induce the appropriate type of antigen-specific immune responses in a selective and reliable way. Moreover, the use of adjuvants such the LTK63 and LTR72 mutants, with no or low residual toxicity, holds a lot of promise for the future application of vaccines to the bare skin of humans.
Collapse
Affiliation(s)
- A.-S. Beignon
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| | - J.-P. Briand
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| | - R. Rappuoli
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| | - S. Muller
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| | - C. D. Partidos
- UPR 9021, Institut de Biologie Moléculaire et Cellulaire, CNRS, F-67084 Strasbourg, France, IRIS, Chiron, SpA, 53100 Siena, Italy
| |
Collapse
|
39
|
Abstract
Drug delivery has metamorphosed from the concept of a pill to molecular medicine in the past 100 years. Better appreciation and integration of pharmacokinetic and pharmacodynamic principles in design of drug delivery systems has led to improved therapeutic efficacy. A greater understanding of the molecular transport in relation to physico-chemical properties has led to the evolution of a biopharmaceutics classification system, which should be a future road map, governing drug design, development and delivery. While drugs belonging to class I and II will be delivered by established platform technologies, novel delivery strategies will evolve and mature to realize the potential of 'new generation' biotech and non biotech drugs belonging to class III and IV, respectively.
Collapse
Affiliation(s)
- O Pillai
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Sector 67, Ph X, - SAS Nagar -- 160 062 (Punjab), India
| | | | | |
Collapse
|
40
|
Beignon AS, Briand JP, Muller S, Partidos CD. Immunization onto bare skin with heat-labile enterotoxin of Escherichia coli enhances immune responses to coadministered protein and peptide antigens and protects mice against lethal toxin challenge. Immunology 2001; 102:344-51. [PMID: 11298834 PMCID: PMC1783185 DOI: 10.1046/j.1365-2567.2001.01183.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2000] [Revised: 11/08/2000] [Accepted: 11/14/2000] [Indexed: 11/20/2022] Open
Abstract
In this study, the potential of the bare skin as a non-invasive route for vaccination was examined. Following application of heat-labile enterotoxin (LT) of Escherichia coli onto bare skin of BALB/c mice, strong serum anti-LT antibody responses were observed, and mucosal immunoglobulin A (IgA) and IgG antibodies were measured in vagina washes. In addition, LT enhanced the serum and mucosal antibody and proliferative T-cell responses to the model protein antigen beta-galactosidase (beta-gal) when coadministered onto bare skin, highlighting its potential to exert an adjuvant effect. When a peptide representing a T-helper epitope (aa 307-319) from the haemagglutinin of influenza virus was applied onto bare skin with LT or cholera toxin (CT), it primed effectively peptide- and virus-specific T cells, as measured in vitro by the interleukin-2 (IL-2) secretion assay. LT was shown to be as immunogenic as CT. Binding activity to GM1 gangliosides was essential for effective induction of anti-CT serum and mucosal antibody responses. Finally, mice immunized onto bare skin with LT were protected against intraperitoneal challenge with a lethal dose of the homologous toxin. These findings give further support to a growing body of evidence on the potential of skin as a non-invasive route for vaccine delivery. This immunization strategy might be advantageous for vaccination programmes in Third World countries, because administration by this route is simple, painless and economical.
Collapse
Affiliation(s)
- A S Beignon
- Institut de Biologie Moléculaire et Cellulaire, CNRS, Strasbourg, France
| | | | | | | |
Collapse
|