51
|
Kimoto T, Kim H, Sakai S, Takahashi E, Kido H. Oral vaccination with influenza hemagglutinin combined with human pulmonary surfactant-mimicking synthetic adjuvant SF-10 induces efficient local and systemic immunity compared with nasal and subcutaneous vaccination and provides protective immunity in mice. Vaccine 2018; 37:612-622. [PMID: 30553569 DOI: 10.1016/j.vaccine.2018.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Revised: 12/03/2018] [Accepted: 12/06/2018] [Indexed: 12/23/2022]
Abstract
We reported previously that a synthetic mucosal adjuvant SF-10, which mimics human pulmonary surfactant, delivers antigen to mucosal dendritic cells in the nasal cavity and promotes induction of humoral and cellular immunity. The aim of the present study was to determine the effects of oral administration of antigen combined with SF-10 (antigen-SF-10) on systemic and local immunity. Oral administration of ovalbumin, a model antigen, combined with SF-10 enhanced ovalbumin uptake into intestinal antigen presenting MHC II+CD11c+ cells and their CD11b+CD103+ and CD11b+CD103- subtype dendritic cells, which are the major antigen presenting subsets of the intestinal tract, more efficiently compared to without SF-10. Oral vaccination with influenza hemagglutinin vaccine (HAv)-SF-10 induced HAv-specific IgA and IgG in the serum, and HAv-specific secretory IgA and IgG in bronchoalveolar lavage fluid, nasal washes, gastric extracts and fecal material; their levels were significantly higher than those induced by subcutaneous HAv or intranasal HAv and HAv-SF-10 vaccinations. Enzyme-linked immunospot assay showed high numbers of HAv-specific IgA and IgG antibody secreting cells in the gastrointestinal and respiratory mucosal lymphoid tissues after oral vaccination with HAv-SF-10, but no or very low induction following oral vaccination with HAv alone. Oral vaccination with HAv-SF-10 provided protective immunity against severe influenza A virus infection, which was significantly higher than that induced by HAv combined with cholera toxin. Oral vaccination with HAv-SF-10 was associated with unique cytokine production patterns in the spleen after HAv stimulation; including marked induction of HAv-responsive Th17 cytokines (e.g., IL-17A and IL-22), high induction of Th1 cytokines (e.g., IL-2 and IFN-γ) and moderate induction of Th2 cytokines (e.g., IL-4 and IL-5). These results indicate that oral vaccination with HAv-SF-10 induces more efficient systemic and local immunity than nasal or subcutaneous vaccination with characteristically high levels of secretory HAv-specific IgA in various mucosal organs and protective immunity.
Collapse
Affiliation(s)
- Takashi Kimoto
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Tokushima, Japan
| | - Hyejin Kim
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Tokushima, Japan
| | - Satoko Sakai
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Tokushima, Japan
| | - Etsuhisa Takahashi
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Tokushima, Japan
| | - Hiroshi Kido
- Division of Enzyme Chemistry, Institute for Enzyme Research, Tokushima University, Tokushima, Japan.
| |
Collapse
|
52
|
Robust mucosal and systemic responses against HTLV-1 by delivery of multi-epitope vaccine in PLGA nanoparticles. Eur J Pharm Biopharm 2018; 133:321-330. [PMID: 30408519 DOI: 10.1016/j.ejpb.2018.11.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 10/28/2018] [Accepted: 11/04/2018] [Indexed: 12/18/2022]
Abstract
In this investigation, the immunogenicity of HTLV-1 fusion epitope-loaded PLGA nanoparticles (NPs) was assessed in the absence or presence of co-encapsulated CpG ODN adjuvant, in a mice model. For this purpose, the multi-epitope chimera including Tax, env, and gag immunodominant HTLV-1 epitopes was encapsulated in biodegradable PLGA NPs with or without CpG adjuvant. PLGA nanospheres produced by a double emulsion method had a size of <200 nm, and encapsulation efficiency of chimera antigen was 85%. The release profile of radiolabeled chimera indicated that only 17.4% and 20.1% of chimera were released from PLGA NPs without or with co-encapsulated CPG ODN during one month, respectively. The PLGA formulations significantly elevated titers of IgG1, IgG2a, and sIgA antibodies, as well as IL-10, and IFN-γ cytokines and also reduced the amount of TGF-β1 production relative to the other vaccines. Additionally, co-delivery of chimera and CpG ODN in PLGA NPs significantly promoted cellular and mucosal responses compared to the incorporation of CpG and chimera antigen. In summary, these results revealed that the sustained release of chimera from PLGA as an efficient polymeric system elicited potent cell-mediated and mucosal immunity without inflammatory responses against HTLV-1. Therefore, the proper design of vaccine formulation and immunization strategy are crucial factors to construct an efficient vaccine.
Collapse
|
53
|
Diaz-Dinamarca DA, Soto DA, Leyton YY, Altamirano-Lagos MJ, Avendaño MJ, Kalergis AM, Vasquez AE. Oral vaccine based on a surface immunogenic protein mixed with alum promotes a decrease in Streptococcus agalactiae vaginal colonization in a mouse model. Mol Immunol 2018; 103:63-70. [PMID: 30205305 DOI: 10.1016/j.molimm.2018.08.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/07/2018] [Accepted: 08/31/2018] [Indexed: 01/15/2023]
Abstract
The Surface Immunogenic Protein (SIP) of Group B Streptococcus (GBS) had been described as a good target for vaccine development. To date, SIP has been reported as a highly conserved protein, and in a mouse model it induces protection against lethal GBS challenge. Also, similar effects have been described by intranasal immunization with a SIP-based vaccine. In this study, we show the immune response induced by an oral SIP-based vaccine formulated on alum in a mouse model. Our vaccine can reduce vaginal GBS colonization and induce specific SIP-antibodies with opsonophagocytosis activities against GBS. Moreover, we observed the activation of T-cells producing IFN-γ, TNF-α, IL-10, IL-2, and increased expression of the transcription factor T-bet, suggesting a Th1-type humoral response. The oral SIP-based vaccine is a novel alternative in the development of a vaccine against GBS.
Collapse
Affiliation(s)
- D A Diaz-Dinamarca
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile; Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - D A Soto
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile
| | - Y Y Leyton
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile
| | - M J Altamirano-Lagos
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile; Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M J Avendaño
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile; Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A M Kalergis
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A E Vasquez
- Sección de Biotecnología, Instituto de Salud Pública de Chile, Chile; Universidad San Sebastián, Facultad de Medicina y Ciencia, Escuela de Bioquímica, Providencia, Santiago, Chile.
| |
Collapse
|
54
|
Kang SH, Hong SJ, Lee YK, Cho S. Oral Vaccine Delivery for Intestinal Immunity-Biological Basis, Barriers, Delivery System, and M Cell Targeting. Polymers (Basel) 2018; 10:E948. [PMID: 30960873 PMCID: PMC6403562 DOI: 10.3390/polym10090948] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/21/2018] [Accepted: 08/22/2018] [Indexed: 12/19/2022] Open
Abstract
Most currently available commercial vaccines are delivered by systemic injection. However, needle-free oral vaccine delivery is currently of great interest for several reasons, including the ability to elicit mucosal immune responses, ease of administration, and the relatively improved safety. This review summarizes the biological basis, various physiological and immunological barriers, current delivery systems with delivery criteria, and suggestions for strategies to enhance the delivery of oral vaccines. In oral vaccine delivery, basic requirements are the protection of antigens from the GI environment, targeting of M cells and activation of the innate immune response. Approaches to address these requirements aim to provide new vaccines and delivery systems that mimic the pathogen's properties, which are capable of eliciting a protective mucosal immune response and a systemic immune response and that make an impact on current oral vaccine development.
Collapse
Affiliation(s)
- Sung Hun Kang
- Department of Medical Sciences, College of Medicine, Hallym University, Chuncheon 24252, Korea.
| | - Seok Jin Hong
- Department of Otorhinolaryngology-Head and Neck Surgery, Hallym University, Dongtan Sacred Heart Hospital, Hwaseong 18450, Korea.
| | - Yong-Kyu Lee
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 27469, Korea.
- 4D Biomaterials Center, Korea National University of Transportation, Jeungpyeong 27909, Korea.
| | - Sungpil Cho
- 4D Biomaterials Center, Korea National University of Transportation, Jeungpyeong 27909, Korea.
| |
Collapse
|
55
|
The novel immunogenic chimeric peptide vaccine to elicit potent cellular and mucosal immune responses against HTLV-1. Int J Pharm 2018; 549:404-414. [PMID: 30075250 DOI: 10.1016/j.ijpharm.2018.07.069] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/28/2018] [Accepted: 07/30/2018] [Indexed: 12/15/2022]
Abstract
This study reports on the immunogenicity assessment of a novel chimeric peptide vaccine including Tax, gp21, gp46, and gag immunodominant epitopes of human T-cell lymphotropic virus type 1 (HTLV-1) to induce immunity against HTLV-1 after subcutaneous (SC) or intranasal administration in a mice model. Additionally, to elevate the efficacy of the HTLV-1 vaccine, the chimera was physically mixed with monophosphoryl lipid A (MPLA) or ISCOMATRIX (IMX) adjuvants. For this purpose, the ISCOMATRIX with a size range of 40-60 nm were prepared using lipid film hydration method. Our investigation revealed that the mixture of IMX and chimera could significantly increase antibody titers containing IgG2a, and mucosal IgA, as well as IFN-γ and IL-10 cytokines and decrease the level of TGF-β1, compared to other vaccine formulations. The intranasal delivery of chimera vaccine in the absence or presence adjuvants stimulated potent mucosal sIgA titer relative to subcutaneous immunization. Furthermore, the SC or nasal delivery of various vaccine formulations could shift the immunity toward cell-mediated responses, as evident by higher IgG2a and IFN-γ, as well as suppressed TGF-β1 level. Our findings suggest that proper design, construction, and immunization of multi-epitope vaccine are essential for developing an effective HTLV-1 vaccine.
Collapse
|
56
|
de Oliveira Santos FA, Lincopan N, De Gaspari E. Evaluation of intranasal and subcutaneous route of immunization in neonatal mice using DODAB-BF as adjuvant with outer membrane vesicles of Neisseria meningitis B. Immunobiology 2018; 223:750-760. [PMID: 30055864 DOI: 10.1016/j.imbio.2018.07.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 07/17/2018] [Accepted: 07/18/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND The Neisseria meningitidis bacterium is a Gram-negative diplococcus that can be classified into different serogroups according to the capsular structure. Six of them (A, B, C, W, X, Y) are responsible for causing Invasive Meningococcal Disease (IMD). The strategies for the development of a vaccine for serogroup B have been directed to the use of outer membrane vesicles (OMVs). The aim of this study was to evaluate the immunogenicity of antigenic determinants from OMVs of N. meningitidis B complexed with two different adjuvants: DODAB-BF and aluminum hydroxide (alum), comparing the evaluation of intranasal and subcutaneous route of immunization. METHODS We used prime-boost immunization for the first time in outbred neonatal mice evaluating the cellular and humoral immune response. RESULTS Immunoblot, ELISA DOT-ELISA and ELISpot were used universal methods of antibody detection, in order to detect the humoral and cellular immune response in male and female mice. Immunoblot analyzes the specificity of antibodies with the homologous N. meningitidis strain. ELISA served to quantify and compare the titers of antibodies in the serum of mice immunized with DODAB-BF + OMVs and alum + OMVs for IgG, IgG1, and IgG2a. Intranasal immunization produced a mixed response in the T helper cells Th1 and Th2, while subcutaneous immunization exhibited a Th1 profile. The DOT-ELISA identified cross-reactivity with DODAB-BF to different serogroups of N. meningitidis (B, C, W, and Y) that was not observed with alum. ELISpot analyzed IFN-γ- and IL-4 and the results showed the response directly to Th1 and Th2 profile. CONCLUSION Our findings indicate that DODAB-BF can be an alternative adjuvant for mucosal cell activation with OMVs of N. meningitidis B and that DODAB-BF was similar to aluminum hydroxide as an adjuvant for subcutaneous immunization.
Collapse
Affiliation(s)
- Fernanda Ayane de Oliveira Santos
- Departamento de Imunologia, Av. Dr. Arnaldo 355, 11 andar, 01246902 São Paulo, SP, Brazil; Programa de Pós-Graduação Interunidades em Biotecnologia, Instituto de Ciências Biomédicas, USP, São Paulo, SP, Brazil
| | - Nilton Lincopan
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Elizabeth De Gaspari
- Departamento de Imunologia, Av. Dr. Arnaldo 355, 11 andar, 01246902 São Paulo, SP, Brazil; Programa de Pós-Graduação Interunidades em Biotecnologia, Instituto de Ciências Biomédicas, USP, São Paulo, SP, Brazil.
| |
Collapse
|
57
|
Sun Y, Qian J, Xu X, Tang Y, Xu W, Yang W, Jiang Y, Yang G, Ding Z, Cong Y, Wang C. Dendritic cell-targeted recombinantLactobacilli induce DC activation and elicit specific immune responses against G57 genotype of avian H9N2 influenza virus infection. Vet Microbiol 2018; 223:9-20. [PMID: 30173758 DOI: 10.1016/j.vetmic.2018.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/11/2018] [Accepted: 07/15/2018] [Indexed: 12/21/2022]
Abstract
H9N2 avian influenza viruses are of significance in poultry and public health for the past two decades. Vaccination plays an important role in preventing the infection in domestic poultry. Current H9N2 vaccines have not yet offered ideal protection and eliminated shedding of G57 genotype viruses responsible for H9N2 outbreaks during 2010-2013. Targeted vaccination is a promising strategy to improve vaccine effectiveness. Such a vaccine strategy can be achieved if it is targeted to dendritic cells (DCs) that directly elicit mucosal and adaptive immune responses against microbe challenge. For this purpose, we develop a DC-targeted mucosal vaccine for the oral delivery of the HA protein fused to a DCpep by using Lactobacillus plantarum as an antigen delivery system against G57 virus infection. It showed that Lactobacillus plantarum expressing HA-DCpep confers efficient protection against G57 H9N2 infection, due to have the potential to activate DCs by the TLR-induced NF-κB pathway, to promote DC migration by the CCR7-CCL19/CCL21 axis, thereby enhancing the presentation of immunogen to T and B lymphocytes, resulting in skewing T cells polarization towards Th1, Th2 and Treg cells and evoking more efficient mucosal and adaptive immunity responses. The presented oral mucosal vaccine strategy illustrates the feasibility and efficacy of antigen targeting to DCs through genetic fusion of vaccines to DC-targeting peptides and aids in the design and selection of indications that could be used with this oral vaccine platform against influenza.
Collapse
Affiliation(s)
- Yixue Sun
- Engineering Research Center of Jilin Province for Animals Probiotics, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Jing Qian
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun, China; Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China
| | - Xiaohong Xu
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun, China; Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China
| | - Yubo Tang
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun, China; Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China
| | - Wenzhang Xu
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun, China; Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China
| | - Wentao Yang
- Engineering Research Center of Jilin Province for Animals Probiotics, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yanlong Jiang
- Engineering Research Center of Jilin Province for Animals Probiotics, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Guilian Yang
- Engineering Research Center of Jilin Province for Animals Probiotics, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Zhuang Ding
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun, China; Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China.
| | - Yanlong Cong
- Laboratory of Infectious Diseases, College of Veterinary Medicine, Jilin University, Changchun, China; Key Laboratory of Zoonosis Research, Ministry of Education, Jilin University, Changchun, China.
| | - Chunfeng Wang
- Engineering Research Center of Jilin Province for Animals Probiotics, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
58
|
Hwang HS, Puth S, Tan W, Verma V, Jeong K, Lee SE, Rhee JH. More robust gut immune responses induced by combining intranasal and sublingual routes for prime-boost immunization. Hum Vaccin Immunother 2018; 14:2194-2202. [PMID: 29781755 PMCID: PMC6183199 DOI: 10.1080/21645515.2018.1472185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Norovirus causes acute and debilitating gastroenteritis, characterized by vomiting and diarrhea. We recently reported a recombinant GII. 4 P domain particle (Pd) vaccine adjuvanted with a flagellin, Vibrio vulnificus FlaB, effectively promoting both humoral and cell-mediated immune responses. In the previous study, we found that sublingual (SL) immunization induced higher fecal secretory IgA (SIgA) responses while intranasal (IN) route provided higher amplitude of humoral and cellular immune responses in the systemic compartment. We hypothesized that the combination of IN and SL routes should induce more potent and sustained SIgA responses in the gut. In this study, we have tried combinatorial prime-boost immunization employing both IN and SL routes. The IN priming and SL boosting with the Pd+FlaB vaccine enhanced highest SIgA responses in feces, accompanying increased Pd-specific memory B cells and plasma cells in spleen and bone marrow, respectively. Notably, the strongest long-lasting SIgA response in feces was induced by combined IN prime and SL boost vaccination, which was sustained for more than 3 months. Significantly enhanced gut-homing B cell and follicular helper T cell responses in mesenteric lymph nodes (mLNs) were observed in the IN prime and SL boost combination. IN priming was a requisite for the robust induction of Pd-specific IFNγ, IL-2, IL-4 and IL-5 cytokine responses in the systemic immune compartment. Collectively, the IN prime and SL boost combination was the best option for inducing balanced long-lasting immune responses against the norovirus antigen in both enteric and systemic compartments. These results suggest that immune responses in specific mucosal compartments may be programmed by employing different prime-boost immunization routes.
Collapse
Affiliation(s)
- Hye Suk Hwang
- a Clinical Vaccine R&D Center, Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea
| | - Sao Puth
- a Clinical Vaccine R&D Center, Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea
| | - Wenzhi Tan
- a Clinical Vaccine R&D Center, Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea
| | - Vivek Verma
- a Clinical Vaccine R&D Center, Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea
| | - Kwangjoon Jeong
- a Clinical Vaccine R&D Center, Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Shee Eun Lee
- a Clinical Vaccine R&D Center, Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,c Department of Pharmacology and Dental Therapeutics , School of Dentistry, Chonnam National University , Gwangju , Republic of Korea
| | - Joon Haeng Rhee
- a Clinical Vaccine R&D Center, Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| |
Collapse
|
59
|
Matoo JJ, Bashir K, Kumar A, Krishnaswamy N, Dey S, Chellappa MM, Ramakrishnan S. Resiquimod enhances mucosal and systemic immunity against avian infectious bronchitis virus vaccine in the chicken. Microb Pathog 2018; 119:119-124. [PMID: 29635053 PMCID: PMC7127065 DOI: 10.1016/j.micpath.2018.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/15/2018] [Accepted: 04/06/2018] [Indexed: 12/04/2022]
Abstract
Adjuvant enhancing mucosal immune response is preferred in controlling many pathogens at the portal of entry. Earlier, we reported that a toll-like-receptor 7 (TLR7) agonist, resiquimod (R-848), stimulated the systemic immunity when adjuvanted with the inactivated Newcastle disease virus vaccine in the chicken. Here, we report the effect of R-848 when adjuvanted with live or inactivated avian infectious bronchitis virus (IBV) vaccines with special emphasis on mucosal immunity. Specific pathogen free (SPF) chicks (n = 60) were equally divided into six groups at two weeks of age and immunized with either inactivated or live IBV vaccine adjuvanted with or without R-848. Groups that received either PBS or R-848 served as control. A booster was given on 14 days post-immunization (dpi). R-848 enhanced the antigen specific humoral and cellular immune responses when co-administered with the vaccines as evidenced by an increase in the antibody titre in ELISA and stimulation index in lymphocyte transformation test (LTT) till 35 dpi and increased proportion of CD4+ and CD8+ T cells on 21 dpi in the flow cytometry. Interestingly, it potentiated the IgA responses in the tear and intestinal secretions when used with both live and inactivated IBV vaccines. The combination of IBV vaccine with R-848 significantly up-regulated the transforming growth factor beta 4 (TGFβ4) transcripts in the peripheral blood mononuclear cells (PBMCs) than that of the respective vaccine per se. An enhanced secretory IgA response is likely due to the up-regulation of TGFβ4, which is responsible for class switching to IgA. In conclusion, co-administration of R-848 with inactivated or live IBV vaccine enhanced the systemic as well as mucosal immune responses in the chicken.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Viral/blood
- CD4-Positive T-Lymphocytes
- CD8-Positive T-Lymphocytes
- Chickens/immunology
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/virology
- Disease Models, Animal
- Imidazoles/pharmacology
- Immunity/drug effects
- Immunity/immunology
- Immunity, Cellular/drug effects
- Immunity, Humoral/drug effects
- Immunity, Mucosal/drug effects
- Immunity, Mucosal/immunology
- Immunization
- Immunoglobulin A
- Infectious bronchitis virus/drug effects
- Infectious bronchitis virus/pathogenicity
- Leukocytes, Mononuclear/immunology
- Poultry Diseases/immunology
- Poultry Diseases/prevention & control
- Poultry Diseases/virology
- Specific Pathogen-Free Organisms
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
- Vaccination
- Vaccines, Attenuated/immunology
- Vaccines, Inactivated/administration & dosage
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Javaid Jeelani Matoo
- Immunology Section, ICAR - Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India
| | - Khalid Bashir
- Immunology Section, ICAR - Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India
| | - Ajay Kumar
- Division of Animal Biochemistry, ICAR - Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India
| | - Narayanan Krishnaswamy
- Division of Animal Reproduction, ICAR - Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India
| | - Sohini Dey
- Division of Veterinary Biotechnology, ICAR - Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India
| | - Madhan Mohan Chellappa
- Division of Veterinary Biotechnology, ICAR - Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India
| | - Saravanan Ramakrishnan
- Immunology Section, ICAR - Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243 122, India.
| |
Collapse
|
60
|
Abstract
INTRODUCTION Bacterial flagellin, as a pathogen-associated molecular pattern (PAMP), can activate both innate and adaptive immunity. Its unique structural characteristics endow an effective and flexible adjuvant activity, which allow the design of different types of vaccine strategies to prevent various diseases. This review will discuss recent progress in the mechanism of action of flagellin and its prospects for use as a vaccine adjuvant. AREAS COVERED Herein we summarize various types of information related to flagellin adjuvants from PubMed, including structures, signaling pathways, natural immunity, and extensive applications in vaccines, and it discusses the immunogenicity, safety, and efficacy of flagellin-adjuvanted vaccines in clinical trials. EXPERT COMMENTARY It is widely accepted that as an adjuvant, flagellin can induce an enhanced antigen-specific immune response. Flagellin adjuvants will allow more effective flagellin-based vaccines to enter clinical trials. Furthermore, vaccine formulations containing PAMPs are crucial to exert the maximum potential of vaccine antigens. Therefore, combinations of flagellin-adjuvanted vaccines with other adjuvants that act in a synergistic manner, particularly TLR ligands, represent a promising method for tailoring targeted vaccines to meet specific requirements.
Collapse
Affiliation(s)
- Baofeng Cui
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Xinsheng Liu
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Yuzhen Fang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Peng Zhou
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Yongguang Zhang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| | - Yonglu Wang
- a State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture , Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences , Lanzhou , China.,b Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses , Yangzhou , China
| |
Collapse
|
61
|
Ma S, Wang L, Huang X, Wang X, Chen S, Shi W, Qiao X, Jiang Y, Tang L, Xu Y, Li Y. Oral recombinant Lactobacillus vaccine targeting the intestinal microfold cells and dendritic cells for delivering the core neutralizing epitope of porcine epidemic diarrhea virus. Microb Cell Fact 2018; 17:20. [PMID: 29426335 PMCID: PMC5807822 DOI: 10.1186/s12934-018-0861-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 01/15/2018] [Indexed: 01/02/2023] Open
Abstract
Background Porcine epidemic diarrhea caused by porcine epidemic diarrhea virus (PEDV) has led to serious economic losses to the swine industry worldwide. In this study, an oral recombinant Lactobacillus casei vaccine against PEDV infection targeting the intestinal microfold (M) cells and dendritic cells (DCs) for delivering the core neutralizing epitope (COE) of PEDV spike protein was developed with M cell-targeting peptide (Col) and dendritic cell-targeting peptide (DCpep). The immunogenicity of the orally administered recombinant strains was evaluated. Results After immunization, significantly higher levels of anti-PEDV specific IgG antibodies with PEDV neutralizing activity in the sera and mucosal sIgA antibodies in the tractus genitalis, intestinal mucus, and stools were detected in mice orally administered with the recombinant strain pPG-COE-Col-DCpep/L393, which expressed DCpep and Col targeting ligands fused with the PEDV COE antigen, compared to mice orally immunized with the recombinant strain pPG-COE/L393 without the DCpep and Col targeting ligands. Moreover, in response to restimulation with the PEDV COE antigen in vitro, a significant difference in splenocyte proliferation response and Th2-associated cytokine IL-4 level was observed in the group of mice orally immunized with pPG-COE-Col-DCpep/L393 (p < 0.05) compared to the groups of mice that received pPG-COE-Col/L393 and pPG-COE-DCpep/L393, respectively. Conclusions The intestinal M cells- and DCs-targeting oral delivery of genetically engineered Lactobacillus expressing the COE antigen of PEDV can efficiently induce anti-PEDV mucosal, humoral, and cellular immune responses via oral administration, suggesting a promising vaccine strategy against PEDV infection.![]() Electronic supplementary material The online version of this article (10.1186/s12934-018-0861-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sunting Ma
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China
| | - Li Wang
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China
| | - Xuewei Huang
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China
| | - Xiaona Wang
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China
| | - Su Chen
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China
| | - Wen Shi
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China
| | - Xinyuan Qiao
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, People's Republic of China
| | - Yanping Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China
| | - Lijie Tang
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China.,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, People's Republic of China
| | - Yigang Xu
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China. .,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, People's Republic of China.
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University, Mu Cai Street No. 59, Xiang Fang District, Harbin, People's Republic of China. .,Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Harbin, People's Republic of China.
| |
Collapse
|
62
|
Dehghan S, Kheiri MT, Abnous K, Eskandari M, Tafaghodi M. Preparation, characterization and immunological evaluation of alginate nanoparticles loaded with whole inactivated influenza virus: Dry powder formulation for nasal immunization in rabbits. Microb Pathog 2017; 115:74-85. [PMID: 29223454 DOI: 10.1016/j.micpath.2017.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 12/21/2022]
Abstract
It has become important to explore more efficient and feasible influenza vaccines, since epidemics of influenza virus cause hundreds of thousands of deaths all around the world. Improving immunogenicity of parentral influenza vaccines has given rise to mucosal delivery routes. In this study, alginate nanoparticles (NPs) were efficiently synthetized by ionic gelation method and influenza virus and CpG ODN or Quillaja Saponin (QS) adjuvants were actively incorporated into alginate NPs. The prepared particles were evaluated for both humoral and cellular immune responses in rabbits' nostrils. The vaccination started with a prime dose and followed by three boosters (two intranasal (IN) on days 45 and 60 and the last dose, intramuscular (IM) on day 75). HAI titer had increased in all the samples; although, only in the group received WV + CPG suspension reached to the protective HAI titer. All the immunized rabbits elicited significantly high sIgA levels on day 75, compared to the negative and the IM groups. At the end of the study, IN administration of CpG ODN adjuvant with virus antigen induced higher IgG level than the groups vaccinated with alginate NPs with or without CpG ODN (P < 0.001). As for the cellular immunity, CpG ODN was capable of inducing significant levels of IL-4 and TNF-α, either through inoculation along with the virus suspension or as incorporated in alginate NPs. According to the obtained data, CpG ODN adjuvant showed higher immunogenic potential as part of a vaccine delivery system than QS. Moreover, applying alginate polymer as a nasal delivery system carrier was not deemed immunogenic against influenza whole virus.
Collapse
Affiliation(s)
- Solmaz Dehghan
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Khalil Abnous
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Sciences Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Eskandari
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Tafaghodi
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
63
|
Thermostability of the coating, antigen and immunostimulator in an adjuvanted oral capsule vaccine formulation. Int J Pharm 2017; 534:60-70. [PMID: 29024788 DOI: 10.1016/j.ijpharm.2017.10.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/05/2017] [Accepted: 10/07/2017] [Indexed: 12/17/2022]
Abstract
Oral vaccines present an attractive alternative to injectable vaccines for enteric diseases due to ease of delivery and the induction of intestinal immunity at the site of infection. However, susceptibility to gastrointestinal proteolysis, limited transepithelial uptake and a lack of clinically acceptable adjuvants present significant challenges. A further challenge to mass vaccination in developing countries is the very expensive requirement to maintain the cold chain. We recently described the effectiveness of a Single Multiple Pill® (SmPill®) adjuvanted capsule approach to enhance the effectiveness of a candidate enterotoxigenic Escherichia coli (ETEC) oral vaccine. Here it was demonstrated that this delivery system maintains the antigenicity of ETEC colonisation factor antigen I (CFA/I) and the immunostimulatory activity of the orally active α-Galactosylceramide (α-GalCer) adjuvant after storage of SmPill® minispheres under room temperature and extreme storage conditions for several months. In addition, the internal structure of the cores of SmPill® minispheres and antigen release features at intestinal pH were found to be preserved under all these conditions. However, changes in the surface morphology of SmPill® minispheres leading to the antigen release at gastric pH were observed after a few weeks of storage under extreme conditions. Those modifications were prevented by the introduction of an Opadry® White film coating layer between the core of SmPill® minispheres and the enteric coating. Under these conditions, protection against antigen release at gastric pH was maintained even under high temperature and humidity conditions. These results support the potential of the SmPill® minisphere approach to maintain the stability of an adjuvanted whole cell killed oral vaccine formulation.
Collapse
|
64
|
White JA, Haghighi C, Brunner J, Estrada M, Lal M, Chen D. Preformulation studies with the Escherichia coli double mutant heat-labile toxin adjuvant for use in an oral vaccine. J Immunol Methods 2017; 451:83-89. [PMID: 28939395 PMCID: PMC5703769 DOI: 10.1016/j.jim.2017.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/12/2017] [Accepted: 09/12/2017] [Indexed: 11/25/2022]
Abstract
Double mutant heat-labile toxin (dmLT) is a promising adjuvant for oral vaccine administration. The aims of our study were to develop sensitive methods to detect low concentrations of dmLT and to use the assays in preformulation studies to determine whether dmLT remains stable under conditions encountered by an oral vaccine. We developed a sandwich ELISA specific for intact dmLT and a sensitive SDS-PAGE densitometry method, and tested stability of dmLT in glass and plastic containers, in saliva, at the pH of stomach fluid, and in high-osmolarity buffers. The developed ELISA has a quantification range of 62.5 to 0.9 ng/mL and lower limit of detection of 0.3 ng/mL; the limit of quantification of the SDS-PAGE is 10 μg/mL. This work demonstrates the application of dmLT assays in preformulation studies to development of an oral vaccine containing dmLT. Assays reported here will facilitate the understanding and use of dmLT as an adjuvant.
Collapse
|
65
|
Bailey BA, Desai KGH, Ochyl LJ, Ciotti SM, Moon JJ, Schwendeman SP. Self-encapsulating Poly(lactic-co-glycolic acid) (PLGA) Microspheres for Intranasal Vaccine Delivery. Mol Pharm 2017; 14:3228-3237. [PMID: 28726424 PMCID: PMC5642922 DOI: 10.1021/acs.molpharmaceut.7b00586] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Herein we describe a formulation of self-encapsulating poly(lactic-co-glycolic acid) (PLGA) microspheres for vaccine delivery. Self-healing encapsulation is a novel encapsulation method developed by our group that enables the aqueous loading of large molecules into premade PLGA microspheres. Calcium phosphate (CaHPO4) adjuvant gel was incorporated into the microspheres as a protein-trapping agent for improved encapsulation of antigen. Microspheres were found to have a median size of 7.05 ± 0.31 μm, with a w/w loading of 0.60 ± 0.05% of ovalbumin (OVA) model antigen. The formulation demonstrated continuous release of OVA over a 49-day period. Released OVA maintained its antigenicity over the measured period of >21 days of release. C57BL/6 mice were immunized via the intranasal route with prime and booster doses of OVA (10 μg) loaded into microspheres or coadministered with cholera toxin B (CTB), the gold standard of mucosal adjuvants. Microspheres generated a Th2-type response in both serum and local mucosa, with IgG antibody responses approaching those generated by CTB. The results suggest that this formulation of self-encapsulating microspheres shows promise for further study as a vaccine delivery system.
Collapse
Affiliation(s)
- Brittany A. Bailey
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kashappa-Goud H. Desai
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Lukasz J. Ochyl
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Susan M. Ciotti
- NanoBio Corporation, 2311 Green Road, Ann Arbor, Michigan 48105, United States
| | - James J. Moon
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Biomedical Engineering and the Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Steven P. Schwendeman
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
66
|
Moreno-Mendieta S, Barrios-Payán J, Mata-Espinosa D, Sánchez S, Hernández-Pando R, Rodríguez-Sanoja R. Raw starch microparticles have immunostimulant activity in mice vaccinated with BCG and challenged with Mycobacterium tuberculosis. Vaccine 2017; 35:5123-5130. [PMID: 28818565 DOI: 10.1016/j.vaccine.2017.08.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/11/2017] [Accepted: 08/07/2017] [Indexed: 12/20/2022]
Abstract
The main challenge for vaccine development or improvement is the lack of safe adjuvants or immunostimulants that induce protective immune responses and can be used for mucosal immunization, which is a highly desirable strategy for vaccination against infectious diseases acquired by oral or intranasal routes. One promising alternative is the use of biodegradable and biocompatible polymeric microparticles. Recently, we developed an immobilization and delivery system with starch microparticles (SMPs) and a starch-binding domain (SBDtag) suitable for the mucosal administration of antigens and the induction of antigen-specific immune responses. Here, we explore the immunostimulant and reinforcing potential of the system using BALB/c mice with progressive pulmonary tuberculosis (PPT). The heat shock protein alpha-crystallin from Mycobacterium tuberculosis immobilized on SMPs (µAcr-SBDtag) or SMPs alone were administered nasally as boosters to BCG-vaccinated mice without any extra adjuvant. The mice were challenged intratracheally with either moderately virulent or highly virulent M. tuberculosis strains. Our results showed that the administration of either the immobilized antigen or SMPs asa booster for the BCG vaccination induced a significant reduction of bacterial loads in the lungs of mice, even more than in mice that received the BCG vaccination alone. Since no difference was observed in pulmonary bacillary burdens between the two reinforced groups, the obtained effect was most likely primarily caused by the starch. As determined by histological study, the administration of boosters did not contribute to the progress of pneumonia, which diminishes the safety concerns related to the administration of SMPs intranasally. Taken together, our findings suggest that this system may be considered asa new carbohydrate-based adjuvant suitable for mucosal vaccines against tuberculosis and other infectious diseases, and more generally, they highlight the potential of particulate α-glucans as immune response modifiers.
Collapse
Affiliation(s)
- Silvia Moreno-Mendieta
- CONACYT, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Jorge Barrios-Payán
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico
| | - Dulce Mata-Espinosa
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico
| | - Sergio Sánchez
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, Mexico
| | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Vasco de Quiroga 15, Delegación Tlalpan, Ciudad de México, Mexico.
| | - Romina Rodríguez-Sanoja
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), A.P. 70228, Ciudad Universitaria, Ciudad de México 04510, Mexico.
| |
Collapse
|
67
|
Li B, Yuan H, Chen L, Sun H, Hu J, Wei S, Zhao Z, Zou Q, Wu C. The influence of adjuvant on UreB protection against Helicobacter pylori through the diversity of CD4+ T-cell epitope repertoire. Oncotarget 2017; 8:68138-68152. [PMID: 28978104 PMCID: PMC5620244 DOI: 10.18632/oncotarget.19248] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/20/2017] [Indexed: 02/06/2023] Open
Abstract
Adjuvants are widely used to enhance the effects of vaccines against pathogen infections. Interestingly, different adjuvants and vaccination routes usually induce dissimilar immune responses, and can even have completely opposite effects. The mechanism remains unclear. In this study, urease B subunit (UreB), an antigen of Helicobacter pylori (H. pylori) that can induce protective immune responses, was used as a model to vaccinate mice. We investigated the effects of different adjuvants and routes on consequent T cell epitope-specific targeting and protection against H. pylori infection. Comparison of the protective effects of UreB, administered either subcutaneously (sc) or intranasally (in), with the adjuvants AddaVax (sc), Complete Freund’s adjuvant (CFA; sc), or CpG oligonucleotide (CpG; sc or in), indicated that only CFA (sc) and CpG (in) were protective. Protective vaccines induced T cells targeting epitopes that differed from that targeted by control vaccination. Subsequent peptide vaccination demonstrated that only two of the identified epitopes were protective: UreB373–385 and UreB317–329. Overall, we found that both adjuvant and vaccination route affected the T cell response repertoire to antigen epitopes. The data obtained in this study contribute to improved characterization of the relationship between adjuvants, routes of vaccination, and epitope-specific T cell response repertoires.
Collapse
Affiliation(s)
- Bin Li
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Hanmei Yuan
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Li Chen
- Department of Blood Transfusion, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Heqiang Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Jian Hu
- Department of Intensive Care Unit, Chengdu Military General Hospital, Chengdu, PR China.,Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Shanshan Wei
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | - Zhuo Zhao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| | - Chao Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Third Military Medical University, Chongqing, PR China
| |
Collapse
|
68
|
Schulze K, Ebensen T, Riese P, Prochnow B, Lehr CM, Guzmán CA. New Horizons in the Development of Novel Needle-Free Immunization Strategies to Increase Vaccination Efficacy. Curr Top Microbiol Immunol 2017; 398:207-234. [PMID: 27370343 DOI: 10.1007/82_2016_495] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The young twenty-first century has already brought several medical advances, such as a functional artificial human liver created from stem cells, improved antiviral (e.g., against HIV) and cancer (e.g., against breast cancer) therapies, interventions controlling cardiovascular diseases, and development of new and optimized vaccines (e.g., HPV vaccine). However, despite this substantial progress and the achievements of the last century, humans still suffer considerably from diseases, especially from infectious diseases. Thus, almost one-fourth of all deaths worldwide are caused directly or indirectly by infectious agents. Although vaccination has led to the control of many diseases, including smallpox, diphtheria, and tetanus, emerging diseases are still not completely contained. Furthermore, pathogens such as Bordetella pertussis undergo alterations making adaptation of the respective vaccine necessary. Moreover, insufficient implementation of vaccination campaigns leads to re-emergence of diseases which were believed to be already under control (e.g., poliomyelitis). Therefore, novel vaccination strategies need to be developed in order to meet the current challenges including lack of compliance, safety issues, and logistic constraints. In this context, mucosal and transdermal approaches constitute promising noninvasive vaccination strategies able to match these demands.
Collapse
Affiliation(s)
- Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.
| | - Peggy Riese
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Blair Prochnow
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Braunschweig, Germany.,Department of Pharmacy, Helmholtz Centre for Infection Research (HZI), Saarland University, Saarbrücken, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| |
Collapse
|
69
|
Puth S, Hong SH, Park MJ, Lee HH, Lee YS, Jeong K, Kang IC, Koh JT, Moon B, Park SC, Rhee JH, Lee SE. Mucosal immunization with a flagellin-adjuvanted Hgp44 vaccine enhances protective immune responses in a murine Porphyromonas gingivalis infection model. Hum Vaccin Immunother 2017; 13:2794-2803. [PMID: 28604268 PMCID: PMC5718812 DOI: 10.1080/21645515.2017.1327109] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Chronic periodontitis is caused by interactions between the oral polymicrobial community and host factors. Periodontal diseases are associated with dysbiotic shift in oral microbiota. Vaccination against periodontopathic bacteria could be a fundamental therapeutic to modulate polymicrobial biofilms. Because oral cavity is the site of periodontopathic bacterial colonization, mucosal vaccines should provide better protection than vaccines administered systemically. We previously reported that bacterial flagellin is an excellent mucosal adjuvant. In this study, we investigated whether mucosal immunization with a flagellin-adjuvanted polypeptide vaccine induces protective immune responses using a Porphyromonas gingivalis infection model. We used the Hgp44 domain polypeptide of Arg-gingipain A (RgpA) as a mucosal antigen. Intranasal (IN) immunization induced a significantly higher Hgp44-specific IgG titer in the serum of mice than sublingual (SL) administration. The co-administration of flagellin potentiated serum IgG responses for both the IN and SL vaccinations. On the other hand, the anti-Hgp44-specific IgA titer in the saliva was comparable between IN and SL vaccinations, suggesting SL administration as more compliant vaccination route for periodontal vaccines. The co-administration of flagellin significantly potentiated the secretory IgA response in saliva also. Furthermore, mice administered a mixture of Hgp44 and flagellin via the IN and SL routes exhibited significant reductions in alveolar bone loss induced by live P. gingivalis infections. An intranasally administered Hgp44-flagellin fusion protein induced a comparable level of Hgp44-specific antibody responses to the mixture of Hgp44 and flagellin. Overall, a flagellin-adjuvanted Hgp44 antigen would serve an important component for a multivalent mucosal vaccine against polymicrobial periodontitis.
Collapse
Affiliation(s)
- Sao Puth
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Seol Hee Hong
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Mi Jin Park
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Hye Hwa Lee
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,c Department of Pharmacology and Dental Therapeutics, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea ; Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Youn Suhk Lee
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,c Department of Pharmacology and Dental Therapeutics, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea ; Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Kwangjoon Jeong
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - In-Chol Kang
- d Department of Oral Microbiology, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea
| | - Jeong Tae Koh
- c Department of Pharmacology and Dental Therapeutics, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea ; Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Byounggon Moon
- e Well Aging Research Center, Samsung Adv. Inst. of Technology (SAIT) , Samsung Electronics Co., Ltd. Suwon-si , Gyeonggi-do , Republic of Korea
| | - Sang Chul Park
- e Well Aging Research Center, Samsung Adv. Inst. of Technology (SAIT) , Samsung Electronics Co., Ltd. Suwon-si , Gyeonggi-do , Republic of Korea
| | - Joon Haeng Rhee
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,b Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| | - Shee Eun Lee
- a Clinical Vaccine R&D Center , Chonnam National University , Hwasun-gun , Jeonnam , Republic of Korea.,c Department of Pharmacology and Dental Therapeutics, School of Dentistry , Chonnam National University , Gwangju , Republic of Korea ; Department of Microbiology , Chonnam National University Medical School , Hwasun-gun , Jeonnam , Republic of Korea
| |
Collapse
|
70
|
Potential of glucans as vaccine adjuvants: A review of the α-glucans case. Carbohydr Polym 2017; 165:103-114. [DOI: 10.1016/j.carbpol.2017.02.030] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 02/08/2017] [Accepted: 02/08/2017] [Indexed: 01/06/2023]
|
71
|
Rajan B, Løkka G, Koppang EO, Austbø L. Passive Immunization of Farmed Fish. THE JOURNAL OF IMMUNOLOGY 2017; 198:4195-4202. [DOI: 10.4049/jimmunol.1700154] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 03/16/2017] [Indexed: 11/19/2022]
|
72
|
Gomez-Samblas M, García-Rodríguez JJ, Trelis M, Bernal D, Lopez-Jaramillo FJ, Santoyo-Gonzalez F, Vilchez S, Espino AM, Bolás-Fernández F, Osuna A. Self-adjuvanting C18 lipid vinil sulfone-PP2A vaccine: study of the induced immunomodulation against Trichuris muris infection. Open Biol 2017; 7:rsob.170031. [PMID: 28404797 PMCID: PMC5413912 DOI: 10.1098/rsob.170031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/14/2017] [Indexed: 12/11/2022] Open
Abstract
Despite the importance of the adjuvant in the immunization process, very few adjuvants merge with the antigens in vaccines. A synthetic self-adjuvant oleic-vinyl sulfone (OVS) linked to the catalytic region of recombinant serine/threonine phosphatase 2A from the nematode Angiostrongylus costaricensis (rPP2A) was used for intranasal immunization in mice previously infected with Trichuris muris. The animal intranasal immunization with rPP2A-OVS showed a reduction of 99.01% in the number of the nematode eggs and 97.90% in adult. The immunohistochemical analysis of the intestinal sections showed that in immunized animals with lipopeptide the mucus was significantly higher than in the other experimental groups. Also, these animals presented significantly different chemokine, CCL20 and CCL11, levels. However, although the number and size of Tuft cells did not vary between groups, the intensity of fluorescence per cell was significant in the group immunized with the rPP2A-OVS. The results of the present study suggest that mice immunized with the lipopeptide are capable of activating a combined Th17/Th9 response. This strategy of immunization may be of great applicability not only in immunotherapy and immunoprophylaxis to control diseases caused by nematodes but also in pathologies necessitating action at the level of the Th9 response in the intestinal mucosa.
Collapse
Affiliation(s)
- M Gomez-Samblas
- Instituto de Biotecnología, Grupo de Bioquímica y Parasitología Molecular, Departamento de Parasitología, Universidad de Granada, Campus Universitario Fuentenueva, 18071 Granada, Spain
| | - J J García-Rodríguez
- Departamento de Parasitología, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal s/n. Ciudad Universitaria, 28040 Madrid, Spain
| | - M Trelis
- Àrea de Parasitologia, Departament de Farmàcia i Tecnologia Farmacèutica i Parasitologia, Universitat de València, Av. V.A. Estellés, s/n, 46100 Burjassot (Valencia), Spain.,Joint Research Unit on Endocrinology, Nutrition and Clinical Dietetics, Health Research Institute-La Fe, Universitat de Valencia, Av. Fdo. Abril Martorell, 106, 46026 Valencia, Spain
| | - D Bernal
- Departament de Bioquímica i Biologia Molecular, Universitat de València, C/ Dr Moliner, 50, 46100 Burjassot (Valencia), Spain
| | - F J Lopez-Jaramillo
- Departamento de Química Orgánica, Facultad de Ciencias, Instituto de Biotecnología, Universidad de Granada, 18071 Granada, Spain
| | - F Santoyo-Gonzalez
- Departamento de Química Orgánica, Facultad de Ciencias, Instituto de Biotecnología, Universidad de Granada, 18071 Granada, Spain
| | - S Vilchez
- Instituto de Biotecnología, Grupo de Bioquímica y Parasitología Molecular, Departamento de Bioquímica, Universidad de Granada, Campus Universitario Fuentenueva, 18071 Granada, Spain
| | - A M Espino
- Laboratory of Immunology and Molecular Parasitology, Department of Microbiology, University of Puerto Rico, School of Medicine. PO Box 365067, San Juan 00936-5067, Puerto Rico
| | - F Bolás-Fernández
- Departamento de Parasitología, Facultad de Farmacia, Universidad Complutense, Plaza de Ramón y Cajal s/n. Ciudad Universitaria, 28040 Madrid, Spain
| | - A Osuna
- Instituto de Biotecnología, Grupo de Bioquímica y Parasitología Molecular, Departamento de Parasitología, Universidad de Granada, Campus Universitario Fuentenueva, 18071 Granada, Spain
| |
Collapse
|
73
|
Qian F, Yin J, Li M, Guo A, Li T, Zhou L, Wu X, Xu H. Intranasal immunization with a peptide conjugated to Salmonella flagellin induces both systemic and mucosal peptide-specific antibody responses in mice. Microbiol Immunol 2017; 60:497-500. [PMID: 27301339 DOI: 10.1111/1348-0421.12396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/06/2016] [Accepted: 06/02/2016] [Indexed: 11/30/2022]
Abstract
In this study, the mucosal adjuvant activity of Salmonella flagellin as a carrier in a conjugate of EXP153-rFliC was investigated. EXP153-rFliC was made by conjugation of a synthetic B-cell epitope peptide derived from Plasmodium falciparum exported protein-1(EXP153) to recombinant phase 1 flagellin of Salmonella enterica serovar Typhimurium expressed in Escherichia coli (rFliC), and used to immunize BALB/c mice via intranasal instillation. It was found that robust EXP153-specific serum IgG antibodies were induced without additional adjuvant. EXP153-specific sIgA antibodies were also induced, these being detected in bronchoalveolar, nasal, vaginal and intestinal washes. These observations demonstrate that Salmonella flagellin as a carrier is an effective mucosal adjuvant in that its conjugated peptide induces antibody responses.
Collapse
Affiliation(s)
- Feng Qian
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Jian Yin
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Mengmeng Li
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Aihua Guo
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Ting Li
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Ling Zhou
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Xin Wu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| | - Huji Xu
- Department of Rheumatology and Immunology, Shanghai Changzheng Hospital, Second Military Medical University, 415 Fengyang Road, Shanghai 200003, China
| |
Collapse
|
74
|
Aran K, Chooljian M, Paredes J, Rafi M, Lee K, Kim AY, An J, Yau JF, Chum H, Conboy I, Murthy N, Liepmann D. An oral microjet vaccination system elicits antibody production in rabbits. Sci Transl Med 2017; 9:eaaf6413. [PMID: 28275153 DOI: 10.1126/scitranslmed.aaf6413] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 08/16/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022]
Abstract
Noninvasive immunization technologies have the potential to revolutionize global health by providing easy-to-administer vaccines at low cost, enabling mass immunizations during pandemics. Existing technologies such as transdermal microneedles are costly, deliver drugs slowly, and cannot generate mucosal immunity, which is important for optimal immunity against pathogens. We present a needle-free microjet immunization device termed MucoJet, which is a three-dimensional microelectromechanical systems-based drug delivery technology. MucoJet is administered orally, placed adjacent to the buccal tissue within the oral cavity, and uses a self-contained gas-generating chemical reaction within its two-compartment plastic housing to produce a high-pressure liquid jet of vaccine. We show that the vaccine jet ejected from the MucoJet device is capable of penetrating the buccal mucosal layer in silico, in porcine buccal tissue ex vivo, and in rabbits in vivo. Rabbits treated with ovalbumin by MucoJet delivery have antibody titers of anti-ovalbumin immunoglobulins G and A in blood serum and buccal tissue, respectively, that are three orders of magnitude higher than rabbits receiving free ovalbumin delivered topically by a dropper in the buccal region. MucoJet has the potential to accelerate the development of noninvasive oral vaccines, given its ability to elicit antibody production that is detectable locally in the buccal tissue and systemically via the circulation.
Collapse
Affiliation(s)
- Kiana Aran
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA.
- School of Applied Life Sciences, Keck Graduate Institute, Claremont, CA 91711, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Marc Chooljian
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
| | - Jacobo Paredes
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
- Center of Studies and Technical Research of Gipuzkoa and Tecnun (Technological Campus of the University of Navarra), 20018 San Sebastián, Spain
| | - Mohammad Rafi
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Kunwoo Lee
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
| | - Allison Y Kim
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Jeanny An
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Jennifer F Yau
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
| | - Helen Chum
- Office of Laboratory Animal Care, UC Berkeley, Berkeley, CA 94720, USA
| | - Irina Conboy
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Niren Murthy
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA.
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
| | - Dorian Liepmann
- Department of Bioengineering, University of California (UC), Berkeley, Berkeley, CA 94720, USA.
- UC Berkeley-UC San Francisco Graduate Program in Bioengineering, Berkeley and San Francisco, CA 94158, USA
- Berkeley Sensor and Actuator Center, Berkeley, CA 94720, USA
| |
Collapse
|
75
|
Shakya AK, Chowdhury MYE, Tao W, Gill HS. Mucosal vaccine delivery: Current state and a pediatric perspective. J Control Release 2016; 240:394-413. [PMID: 26860287 PMCID: PMC5381653 DOI: 10.1016/j.jconrel.2016.02.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 01/21/2016] [Accepted: 02/05/2016] [Indexed: 12/30/2022]
Abstract
Most childhood infections occur via the mucosal surfaces, however, parenterally delivered vaccines are unable to induce protective immunity at these surfaces. In contrast, delivery of vaccines via the mucosal routes can allow antigens to interact with the mucosa-associated lymphoid tissue (MALT) to induce both mucosal and systemic immunity. The induced mucosal immunity can neutralize the pathogen on the mucosal surface before it can cause infection. In addition to reinforcing the defense at mucosal surfaces, mucosal vaccination is also expected to be needle-free, which can eliminate pain and the fear of vaccination. Thus, mucosal vaccination is highly appealing, especially for the pediatric population. However, vaccine delivery across mucosal surfaces is challenging because of the different barriers that naturally exist at the various mucosal surfaces to keep the pathogens out. There have been significant developments in delivery systems for mucosal vaccination. In this review we provide an introduction to the MALT, highlight barriers to vaccine delivery at different mucosal surfaces, discuss different approaches that have been investigated for vaccine delivery across mucosal surfaces, and conclude with an assessment of perspectives for mucosal vaccination in the context of the pediatric population.
Collapse
Affiliation(s)
| | | | - Wenqian Tao
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Harvinder Singh Gill
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|
76
|
Tsai HC, Wu R. Mechanisms of Cholera Toxin in the Modulation of TH17 Responses. Crit Rev Immunol 2016; 35:135-52. [PMID: 26351147 DOI: 10.1615/critrevimmunol.2015012295] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Numerous studies have shown that TH17 cells and their signature cytokine IL-17A are critical to host defense against various bacterial and fungal infections. The protective responses mediated by TH17 cells and IL-17A include the recruitment of neutrophils, release of antimicrobial peptides and chemokines, and enhanced tight junction of epithelial cells. Due to the importance of TH17 cells in infections, efforts have been made to develop TH17-based vaccines. The goal of vaccination is to establish a protective immunological memory. Most currently approved vaccines are antibody-based and have limited protection against stereotypically different strains. Studies show that T-cell-based vaccines may overcome this limitation and protect hosts against infection of different strains. Two main strategies are used to develop TH17 vaccines: identification of TH17-specific antigens and TH17-skewing adjuvants. Studies have revealed that cholera toxin (CT) induces a potent Th17 response following vaccination. Antigen vaccination along with CT induces a robust TH17 response, which is sometimes accompanied by TH1 responses. Due to the toxicity of CT, it is hard to apply CT in a clinical setting. Thus, understanding how CT modulates TH17 responses may lead to the development of successful TH17-based vaccines.
Collapse
Affiliation(s)
- Hsing-Chuan Tsai
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Reen Wu
- Center for Comparative Respiratory Biology and Medicine, University of California, USA
| |
Collapse
|
77
|
Adjuvants: Classification, Modus Operandi, and Licensing. J Immunol Res 2016; 2016:1459394. [PMID: 27274998 PMCID: PMC4870346 DOI: 10.1155/2016/1459394] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/02/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023] Open
Abstract
Vaccination is one of the most efficient strategies for the prevention of infectious diseases. Although safer, subunit vaccines are poorly immunogenic and for this reason the use of adjuvants is strongly recommended. Since their discovery in the beginning of the 20th century, adjuvants have been used to improve immune responses that ultimately lead to protection against disease. The choice of the adjuvant is of utmost importance as it can stimulate protective immunity. Their mechanisms of action have now been revealed. Our increasing understanding of the immune system, and of correlates of protection, is helping in the development of new vaccine formulations for global infections. Nevertheless, few adjuvants are licensed for human vaccines and several formulations are now being evaluated in clinical trials. In this review, we briefly describe the most well known adjuvants used in experimental and clinical settings based on their main mechanisms of action and also highlight the requirements for licensing new vaccine formulations.
Collapse
|
78
|
Hedegaard CJ, Heegaard PMH. Passive immunisation, an old idea revisited: Basic principles and application to modern animal production systems. Vet Immunol Immunopathol 2016; 174:50-63. [PMID: 27185263 PMCID: PMC7127230 DOI: 10.1016/j.vetimm.2016.04.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 04/11/2016] [Accepted: 04/13/2016] [Indexed: 12/19/2022]
Abstract
Immunisation by administration of antibodies (immunoglobulins) has been known for more than one hundred years as a very efficient means of obtaining immediate, short-lived protection against infection and/or against the disease-causing effects of toxins from microbial pathogens and from other sources. Thus, due to its rapid action, passive immunisation is often used to treat disease caused by infection and/or toxin exposure. However immunoglobulins may also be administered prior to exposure to infection and/or toxin, although they will not provide long-lasting protection as is seen with active immunisation (vaccination) in which an immunological memory is established by controlled exposure of the host to the pathogen in question. With multi-factorial infectious diseases in production animals, especially those that have proven hard to control by vaccination, the potential of passive immunisation remains big. This review highlights a number of examples on the use of passive immunisation for the control of infectious disease in the modern production of a range of animals, including pigs, cattle, sheep, goat, poultry and fish. Special emphasis is given on the enablement of passive immunisation strategies in these production systems through low cost and ease of use as well as on the sources, composition and purity of immunoglobulin preparations used and their benefits as compared to current measures, including vaccination (also comprising maternal vaccination), antibiotics and feed additives such as spray-dried plasma. It is concluded that provided highly efficient, relatively low-price immunoglobulin products are available, passive immunisation has a clear role in the modern animal production sector as a means of controlling infectious diseases, importantly with a very low risk of causing development of bacterial resistance, thus constituting a real and widely applicable alternative to antibiotics.
Collapse
Affiliation(s)
- Chris J Hedegaard
- National Veterinary Institute, Technical University of Denmark, Section for Immunology and Vaccinology, The innate immunology Group, Denmark.
| | - Peter M H Heegaard
- National Veterinary Institute, Technical University of Denmark, Section for Immunology and Vaccinology, The innate immunology Group, Denmark
| |
Collapse
|
79
|
Samuelson DR, de la Rua NM, Charles TP, Ruan S, Taylor CM, Blanchard EE, Luo M, Ramsay AJ, Shellito JE, Welsh DA. Oral Immunization of Mice with Live Pneumocystis murina Protects against Pneumocystis Pneumonia. THE JOURNAL OF IMMUNOLOGY 2016; 196:2655-65. [PMID: 26864029 DOI: 10.4049/jimmunol.1502004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/06/2016] [Indexed: 01/01/2023]
Abstract
Pneumocystis pneumonia is a major cause of morbidity and mortality in immunocompromised patients, particularly those infected with HIV. In this study, we evaluated the potential of oral immunization with live Pneumocystis to elicit protection against respiratory infection with Pneumocystis murina. C57BL/6 mice vaccinated with live P. murina using a prime-boost vaccination strategy were protected from a subsequent lung challenge with P. murina at 2, 7, 14, and 28 d postinfection even after CD4(+) T cell depletion. Specifically, vaccinated immunocompetent mice had significantly faster clearance than unvaccinated immunocompetent mice and unvaccinated CD4-depleted mice remained persistently infected with P. murina. Vaccination also increased numbers of CD4(+) T cells, CD8(+) T cells, CD19(+) B cells, and CD11b(+) macrophages in the lungs following respiratory infection. In addition, levels of lung, serum, and fecal P. murina-specific IgG and IgA were increased in vaccinated animals. Furthermore, administration of serum from vaccinated mice significantly reduced Pneumocystis lung burden in infected animals compared with control serum. We also found that the diversity of the intestinal microbial community was altered by oral immunization with P. murina. To our knowledge, our data demonstrate for the first time that an oral vaccination strategy prevents Pneumocystis infection.
Collapse
Affiliation(s)
- Derrick R Samuelson
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Nicholas M de la Rua
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Tysheena P Charles
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Sanbao Ruan
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Christopher M Taylor
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112; and
| | - Eugene E Blanchard
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112; and
| | - Meng Luo
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112; and
| | - Alistair J Ramsay
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112; and Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Judd E Shellito
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112; Louisiana Vaccine Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - David A Welsh
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112;
| |
Collapse
|
80
|
Shamriz S, Ofoghi H. Design, structure prediction and molecular dynamics simulation of a fusion construct containing malaria pre-erythrocytic vaccine candidate, PfCelTOS, and human interleukin 2 as adjuvant. BMC Bioinformatics 2016; 17:71. [PMID: 26851942 PMCID: PMC4744421 DOI: 10.1186/s12859-016-0918-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/29/2016] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Malaria infection is still widespread in some parts of the world and threatens the lives of millions of people every year. Vaccines, especially oral vaccines are considered to be effective in reducing the burden of malaria morbidity and mortality. By using recombinant technology, suitable oral hosts could serve as antigen delivering vehicles in developing oral vaccines. This study was aimed towards designing and computational analysis of a fusion protein consisting of Plasmodium falciparum cell-traversal protein for ookinetes and sporozoites (PfCelTOS) fused to human interleukin-2 (IL-2) and M cell-specific peptide ligand (Co1), as a step toward developing a vaccine candidate. RESULTS To our best knowledge, the three dimensional (3D) structure of CelTOS is not reported in protein database. Therefore, we carried out computational modeling and simulation in the hope of understanding the properties and structure of PfCelTOS. Then we fused IL-2 to PfCelTOS by a flexible linker and did in silico analysis to confirm the proper folding of each domain in the designed fusion protein. In the last step, Co1 ligand was added to the confirmed fusion structure using a rigid linker and computational analysis was performed to evaluate the final fusion construct. One structure out of five predicted by I-TASSER for PfCelTOS and fusion constructs was selected based on the highest value for C-score. Molecular dynamics (MD) simulation analysis indicated that predicted structures are stable during the simulation. Ramchandran Plot analysis of PfCelTOS and fusion constructs before and after MD simulation also represented that most residues were fallen in favorable regions. CONCLUSION In silico study showed that Co1-(AEEEK)3- IL-2-(GGGGS)3-PfCelTOS construct has a constant structure and the selected linkers are effectively able to separate the domains. Therefore, data reported in this paper represents the first step toward developing of an oral vaccine candidate against malaria infection.
Collapse
Affiliation(s)
- Shabnam Shamriz
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran.
| | - Hamideh Ofoghi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran.
| |
Collapse
|
81
|
Natural Pig Plasma Immunoglobulins Have Anti-Bacterial Effects: Potential for Use as Feed Supplement for Treatment of Intestinal Infections in Pigs. PLoS One 2016; 11:e0147373. [PMID: 26824607 PMCID: PMC4744083 DOI: 10.1371/journal.pone.0147373] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 12/15/2015] [Indexed: 12/22/2022] Open
Abstract
There is an increasing demand for non-antibiotics solutions to control infectious disease in intensive pig production. Here, one such alternative, namely pig antibodies purified from slaughterhouse blood was investigated in order to elucidate its potential usability to control post-weaning diarrhoea (PWD), which is one of the top indications for antibiotics usage in the pig production. A very cost-efficient and rapid one-step expanded bed adsorption (EBA) chromatography procedure was used to purify pig immunoglobulin G from slaughterhouse pig plasma (more than 100 litres), resulting in >85% pure pig IgG (ppIgG). The ppIgG thus comprised natural pig immunoglobulins and was subsequently shown to contain activity towards four pig-relevant bacterial strains (three different types of Escherichia coli and one type of Salmonella enterica) but not towards a fish pathogen (Yersinia ruckeri), and was demonstrated to inhibit the binding of the four pig relevant bacteria to a pig intestinal cell line (IPEC-J2). Finally it was demonstrated in an in vivo weaning piglet model for intestinal colonization with an E. coli F4+ challenge strain that ppIgG given in the feed significantly reduced shedding of the challenge strain, reduced the proportion of the bacterial family Enterobacteriaceae, increased the proportion of families Enterococcoceae and Streptococcaceae and generally increased ileal microbiota diversity. Conclusively, our data support the idea that natural IgG directly purified from pig plasma and given as a feed supplement can be used in modern swine production as an efficient and cost-effective means for reducing both occurrence of PWD and antibiotics usage and with a potential for the prevention and treatment of other intestinal infectious diseases even if the causative agent might not be known.
Collapse
|
82
|
Singh S, Nehete PN, Yang G, He H, Nehete B, Hanley PW, Barry MA, Sastry KJ. Enhancement of Mucosal Immunogenicity of Viral Vectored Vaccines by the NKT Cell Agonist Alpha-Galactosylceramide as Adjuvant. Vaccines (Basel) 2015; 2:686-706. [PMID: 25553254 PMCID: PMC4278383 DOI: 10.3390/vaccines2040686] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Gene-based vaccination strategies, specifically viral vectors encoding vaccine immunogens are effective at priming strong immune responses. Mucosal routes offer practical advantages for vaccination by ease of needle-free administration, and immunogen delivery at readily accessible oral/nasal sites to efficiently induce immunity at distant gut and genital tissues. However, since mucosal tissues are inherently tolerant for induction of immune responses, incorporation of adjuvants for optimal mucosal vaccination strategies is important. We report here the effectiveness of alpha-galactosylceramide (α-GalCer), a synthetic glycolipid agonist of natural killer T (NKT) cells, as an adjuvant for enhancing immunogenicity of vaccine antigens delivered using viral vectors by mucosal routes in murine and nonhuman primate models. Significant improvement in adaptive immune responses in systemic and mucosal tissues was observed by including α-GalCer adjuvant for intranasal immunization of mice with vesicular stomatitis virus vector encoding the model antigen ovalbumin and adenoviral vectors expressing HIV env and Gag antigens. Activation of NKT cells in systemic and mucosal tissues along with significant increases in adaptive immune responses were observed in rhesus macaques immunized by intranasal and sublingual routes with protein or adenovirus vectored antigens when combined with α-GalCer adjuvant. These results support the utility of α-GalCer adjuvant for enhancing immunogenicity of mucosal vaccines delivered using viral vectors.
Collapse
Affiliation(s)
- Shailbala Singh
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; E-Mails: (S.S.); (G.Y.)
| | - Pramod N. Nehete
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Bastrop, TX 78602, USA; E-Mails: (P.N.N.); b (B.N.)
| | - Guojun Yang
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; E-Mails: (S.S.); (G.Y.)
| | - Hong He
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; E-Mail:
| | - Bharti Nehete
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Bastrop, TX 78602, USA; E-Mails: (P.N.N.); b (B.N.)
| | - Patrick W. Hanley
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health Rocky Mountain Laboratories, Hamilton, MT 59840, USA; E-Mail:
| | - Michael A. Barry
- Department of Internal Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, MN 55902, USA; E-Mail:
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55902, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55902, USA
- Translational Immunovirology and Biodefense Program, Mayo Clinic, Rochester, MN 55902, USA
| | - K. Jagannadha Sastry
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; E-Mails: (S.S.); (G.Y.)
- Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Bastrop, TX 78602, USA; E-Mails: (P.N.N.); b (B.N.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +1-713-563-3304; Fax: +1-713-563-3357
| |
Collapse
|
83
|
Lee SE, Hong SH, Verma V, Lee YS, Duong TMN, Jeong K, Uthaman S, Sung YC, Lee JT, Park IK, Min JJ, Rhee JH. Flagellin is a strong vaginal adjuvant of a therapeutic vaccine for genital cancer. Oncoimmunology 2015; 5:e1081328. [PMID: 27057462 PMCID: PMC4801456 DOI: 10.1080/2162402x.2015.1081328] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 08/03/2015] [Accepted: 08/04/2015] [Indexed: 01/21/2023] Open
Abstract
Cervical cancer is a high-incidence female cancer most commonly caused by human papilloma virus (HPV) infection of the genital mucosa. Immunotherapy targeting HPV-derived tumor antigens (TAs) has been widely studied in animal models and in patients. Because the female genital tract is a portal for the entry of HPV and a highly compartmentalized system, the development of topical vaginal immunotherapy in an orthotopic cancer model would provide an ideal therapeutic. Thus, we examined whether flagellin, a potent mucosal immunomodulator, could be used as an adjuvant for a topical therapeutic vaccine for female genital cancer. Intravaginal (IVAG) co-administration of the E6/E7 peptides with flagellin resulted in tumor suppression and long-term survival of tumor-bearing mice. In contrast to IVAG vaccination, intranasal (IN) or subcutaneous (SC) immunization did not induce significant tumor suppression in the same model. The vaginal adjuvant effect of the flagellin was completely abolished in Toll-like receptor-5 (TLR5) knock-out mice. IVAG immunization with the E6/E7 peptides plus flagellin induced the accumulation of CD4+ and CD8+ cells and the expression of T cell activation-related genes in the draining genital lymph nodes (gLNs). The co-administered flagellin elicited antigen-specific IFNγ production in the gLNs and spleen. The intravaginally administered flagellin was found in association with CD11c+ cells in the gLNs. Moreover, after immunization with a flagellin and the E6/E7 peptides, the TLR5 expression in gLN cells was significantly upregulated. These results suggest that flagellin serves as a potent vaginal adjuvant for a therapeutic peptide cancer vaccine through the activation of TLR5 signaling.
Collapse
Affiliation(s)
- Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Seol Hee Hong
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Vivek Verma
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Youn Suhk Lee
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Republic of Korea; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Republic of Korea
| | - Tra-My Nu Duong
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Republic of Korea; Department of Molecular Medicine, Graduate School, Chonnam National University, Gwangju, Republic of Korea
| | - Kwangjoon Jeong
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Saji Uthaman
- Department of Biomedical Sciences, Chonnam National University Medical School , Gwangju, Republic of Korea
| | - Young Chul Sung
- Department of Life Sciences, POSTECH , Gyeongbuk, Republic of Korea
| | - Jae-Tae Lee
- Department of Nuclear Medicine, Kyungpook National University School of Medicine , Daegu, Republic of Korea
| | - In-Kyu Park
- Department of Molecular Medicine, Graduate School, Chonnam National University , Gwangju, Republic of Korea
| | - Jung-Joon Min
- Department of Nuclear Medicine, Chonnam National University Medical School , Gwangju, Republic of Korea
| | - Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University, Gwangju, Republic of Korea; Department of Microbiology, Chonnam National University Medical School, Gwangju, Republic of Korea
| |
Collapse
|
84
|
Noh HJ, Chowdhury MYE, Cho S, Kim JH, Park HS, Kim CJ, Poo H, Sung MH, Lee JS, Lim YT. Programming of Influenza Vaccine Broadness and Persistence by Mucoadhesive Polymer-Based Adjuvant Systems. THE JOURNAL OF IMMUNOLOGY 2015. [PMID: 26216889 DOI: 10.4049/jimmunol.1500492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The development of an anti-influenza vaccine with the potential for cross-protection against seasonal drift variants as well as occasionally emerging reassortant viruses is essential. In this study, we successfully generated a novel anti-influenza vaccine system combining conserved matrix protein 2 (sM2) and stalk domain of hemagglutinin (HA2) fusion protein (sM2HA2) and poly-γ-glutamic acid (γ-PGA)-based vaccine adjuvant systems that can act as a mucoadhesive delivery vehicle of sM2HA2 as well as a robust strategy for the incorporation of hydrophobic immunostimulatory 3-O-desacyl-4'-monophosphoryl lipid A (MPL) and QS21. Intranasal coadministration of sM2HA2 and the combination adjuvant γ-PGA/MPL/QS21 (CA-PMQ) was able to induce a high degree of protective mucosal, systemic, and cell-mediated immune responses. The sM2HA2/CA-PMQ immunization was able to prevent disease symptoms, confering complete protection against lethal infection with divergent influenza subtypes (H5N1, H1N1, H5N2, H7N3, and H9N2) that lasted for at least 6 mo. Therefore, our data suggest that mucosal administration of sM2HA2 in combination with CA-PMQ could be a potent strategy for a broad cross-protective influenza vaccine, and CA-PMQ as a mucosal adjuvant could be used for effective mucosal vaccines.
Collapse
Affiliation(s)
- Hyun Jong Noh
- Department of Chemical Engineering, Sungkyunkwan University Advanced Institute of Nanotechnology, Suwon 440-746, South Korea
| | - Mohammed Y E Chowdhury
- College of Veterinary Medicine (BK21 Plus Program), Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, South Korea; Faculty of Veterinary Medicine, Chittagong Veterinary and Animal Sciences University, Chittagong 4202, Bangladesh
| | - Seonghun Cho
- College of Veterinary Medicine (BK21 Plus Program), Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, South Korea
| | - Jae-Hoon Kim
- College of Veterinary Medicine (BK21 Plus Program), Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, South Korea
| | - Hye Sun Park
- Korea Basic Science Institute, Chungbuk 363-883, South Korea
| | - Chul-Joong Kim
- College of Veterinary Medicine (BK21 Plus Program), Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, South Korea
| | - Haryoung Poo
- Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, South Korea; and
| | - Moon-Hee Sung
- Department of Advanced Fermentation Fusion Science and Technology, Kookmin University, Seoul 136-702, South Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine (BK21 Plus Program), Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, South Korea;
| | - Yong Taik Lim
- Department of Chemical Engineering, Sungkyunkwan University Advanced Institute of Nanotechnology, Suwon 440-746, South Korea;
| |
Collapse
|
85
|
Nizard M, Diniz MO, Roussel H, Tran T, Ferreira LC, Badoual C, Tartour E. Mucosal vaccines: novel strategies and applications for the control of pathogens and tumors at mucosal sites. Hum Vaccin Immunother 2015; 10:2175-87. [PMID: 25424921 DOI: 10.4161/hv.29269] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mucosal immune system displays several adaptations reflecting the exposure to the external environment. The efficient induction of mucosal immune responses also requires specific approaches, such as the use of appropriate administration routes and specific adjuvants and/or delivery systems. In contrast to vaccines delivered via parenteral routes, experimental, and clinical evidences demonstrated that mucosal vaccines can efficiently induce local immune responses to pathogens or tumors located at mucosal sites as well as systemic response. At least in part, such features can be explained by the compartmentalization of mucosal B and T cell populations that play important roles in the modulation of local immune responses. In the present review, we discuss molecular and cellular features of the mucosal immune system as well as novel immunization approaches that may lead to the development of innovative and efficient vaccines targeting pathogens and tumors at different mucosal sites.
Collapse
Affiliation(s)
- Mevyn Nizard
- a INSERM U970; Universite Paris Descartes; Sorbonne Paris-Cité; Paris, France
| | | | | | | | | | | | | |
Collapse
|
86
|
Karuturi BVK, Tallapaka SB, Phillips JA, Sanderson SD, Vetro JA. Preliminary evidence that the novel host-derived immunostimulant EP67 can act as a mucosal adjuvant. Clin Immunol 2015; 161:251-9. [PMID: 26111481 DOI: 10.1016/j.clim.2015.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2015] [Revised: 06/11/2015] [Accepted: 06/12/2015] [Indexed: 12/13/2022]
Abstract
EP67 is a complement component 5a (C5a)-derived peptide agonist of the C5a receptor (CD88) that selectively activates DCs over neutrophils. Systemic administration of EP67 covalently attached to peptides, proteins, or attenuated pathogens generates TH1-biased immunogen-specific humoral and cellular immune responses with little inflammation. Furthermore, intranasal administration of EP67 alone increases the proportion of activated APCs in the airways. As such, we hypothesized that EP67 can act as a mucosal adjuvant. Intranasal immunization with an EP67-conjugated CTL peptide vaccine against protective MCMV epitopes M84 and pp89 increased protection of naïve female BALB/c mice against primary respiratory infection with salivary gland-derived MCMV and generated higher proportions of epitope responsive and long-lived memory precursor effector cells (MPEC) in the lungs and spleen compared to an inactive, scrambled EP67-conjugated CTL peptide vaccine and vehicle alone. Thus, EP67 may be an effective adjuvant for mucosal vaccines and warrants further study.
Collapse
Affiliation(s)
- Bala Vamsi K Karuturi
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical, Omaha, NE, USA
| | - Shailendra B Tallapaka
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical, Omaha, NE, USA
| | - Joy A Phillips
- Donald P. Shiley BioScience Center, San Diego State University, San Diego, CA, USA
| | - Sam D Sanderson
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical, Omaha, NE, USA
| | - Joseph A Vetro
- Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical, Omaha, NE, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical, Omaha, NE, USA.
| |
Collapse
|
87
|
Newsted D, Fallahi F, Golshani A, Azizi A. Advances and challenges in mucosal adjuvant technology. Vaccine 2015; 33:2399-405. [PMID: 25865473 DOI: 10.1016/j.vaccine.2015.03.096] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 03/17/2015] [Accepted: 03/26/2015] [Indexed: 12/23/2022]
Abstract
Adjuvants play attractive roles in enhancement of immune response during vaccination; however, due to several challenges, only a limited number of adjuvants are licensed by health authorities. The lack of an effective mucosal adjuvant is even more significant as none of the licensed adjuvants revealed a strong enhancement in immune system after mucosal administration. Over the past two decades, several mucosal adjuvants have been developed to deliver antigens to the target cells in the mucosal immune system and increase specific immune responses. However, the safety and efficacy of these adjuvants for testing in human trials is still an important issue, requiring further study. In this article, we briefly review the challenges associated with most common mucosal adjuvants and discuss potential strategies for targeting the mucosal immune system.
Collapse
Affiliation(s)
- Daniel Newsted
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main St W, Hamilton, ON, Canada
| | | | - Ashkan Golshani
- Department of Biology, Carleton University, 1125 Colonel by Drive, Ottawa, ON, Canada
| | - Ali Azizi
- Department of Pathology and Laboratory Medicine, University of Ottawa, 451 Smyth Rd, Ottawa, ON, Canada; Department of Biotechnology, University of Ontario Institute of Technology, Toronto, ON, Canada.
| |
Collapse
|
88
|
Larena M, Holmgren J, Lebens M, Terrinoni M, Lundgren A. Cholera toxin, and the related nontoxic adjuvants mmCT and dmLT, promote human Th17 responses via cyclic AMP-protein kinase A and inflammasome-dependent IL-1 signaling. THE JOURNAL OF IMMUNOLOGY 2015; 194:3829-39. [PMID: 25786687 DOI: 10.4049/jimmunol.1401633] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 01/25/2015] [Indexed: 11/19/2022]
Abstract
We have examined the molecular pathways involved in the adjuvant action of cholera toxin (CT) and two novel nontoxic molecules, multiple-mutated CT (mmCT) and double-mutant heat-labile toxin (dmLT) on human T cell responses. Human PBMCs or isolated monocytes were stimulated in vitro with CT, mmCT, or dmLT plus a polyclonal stimulus (staphylococcal enterotoxin B) or specific bacterial Ags, and effects on expression of cytokines and signaling molecules were determined. CT, mmCT, and dmLT strongly enhanced IL-17A and to a lesser extent IL-13 responses, but had little effect on IFN-γ production or cell proliferation. Intracellular cytokine staining revealed that the enhanced IL-17A production was largely confined to CD4(+) T cells and coculture experiments showed that the IL-17A promotion was effectively induced by adjuvant-treated monocytes. Relative to CT, mmCT and dmLT induced at least 100-fold lower levels of cAMP, yet this cAMP was enough and essential for the promotion of Th17 responses. Thus, inhibition of cAMP-dependent protein kinase A was abolished, and stimulation with a cAMP analog mimicked the adjuvant effect. Furthermore, CT, mmCT, and dmLT induced IL-1β production and caspase-1 activation in monocytes, which was associated with increased expression of key proinflammatory and inflammasome-related genes, including NLRP1, NLRP3, and NLRC4. Inflammasome inhibition with a specific caspase-1 inhibitor, or blocking of IL-1 signaling by IL-1 receptor antagonist, abrogated the Th17-promoting effect. We conclude that CT, mmCT, and dmLT promote human Th17 responses via cAMP-dependent protein kinase A and caspase-1/inflammasome-dependent IL-1 signaling.
Collapse
Affiliation(s)
- Maximilian Larena
- University of Gothenburg Vaccine Research Institute (GUVAX), Department of Microbiology and Immunology, University of Gothenburg, 405 30 Sweden
| | - Jan Holmgren
- University of Gothenburg Vaccine Research Institute (GUVAX), Department of Microbiology and Immunology, University of Gothenburg, 405 30 Sweden
| | - Michael Lebens
- University of Gothenburg Vaccine Research Institute (GUVAX), Department of Microbiology and Immunology, University of Gothenburg, 405 30 Sweden
| | - Manuela Terrinoni
- University of Gothenburg Vaccine Research Institute (GUVAX), Department of Microbiology and Immunology, University of Gothenburg, 405 30 Sweden
| | - Anna Lundgren
- University of Gothenburg Vaccine Research Institute (GUVAX), Department of Microbiology and Immunology, University of Gothenburg, 405 30 Sweden
| |
Collapse
|
89
|
Savelkoul HFJ, Ferro VA, Strioga MM, Schijns VEJC. Choice and Design of Adjuvants for Parenteral and Mucosal Vaccines. Vaccines (Basel) 2015; 3:148-71. [PMID: 26344951 PMCID: PMC4494243 DOI: 10.3390/vaccines3010148] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 10/11/2014] [Accepted: 02/24/2015] [Indexed: 11/16/2022] Open
Abstract
The existence of pathogens that escape recognition by specific vaccines, the need to improve existing vaccines and the increased availability of therapeutic (non-infectious disease) vaccines necessitate the rational development of novel vaccine concepts based on the induction of protective cell-mediated immune responses. For naive T-cell activation, several signals resulting from innate and adaptive interactions need to be integrated, and adjuvants may interfere with some or all of these signals. Adjuvants, for example, are used to promote the immunogenicity of antigens in vaccines, by inducing a pro-inflammatory environment that enables the recruitment and promotion of the infiltration of phagocytic cells, particularly antigen-presenting cells (APC), to the injection site. Adjuvants can enhance antigen presentation, induce cytokine expression, activate APC and modulate more downstream adaptive immune reactions (vaccine delivery systems, facilitating immune Signal 1). In addition, adjuvants can act as immunopotentiators (facilitating Signals 2 and 3) exhibiting immune stimulatory effects during antigen presentation by inducing the expression of co-stimulatory molecules on APC. Together, these signals determine the strength of activation of specific T-cells, thereby also influencing the quality of the downstream T helper cytokine profiles and the differentiation of antigen-specific T helper populations (Signal 3). New adjuvants should also target specific (innate) immune cells in order to facilitate proper activation of downstream adaptive immune responses and homing (Signal 4). It is desirable that these adjuvants should be able to exert such responses in the context of mucosal administered vaccines. This review focuses on the understanding of the potential working mechanisms of the most well-known classes of adjuvants to be used effectively in vaccines.
Collapse
Affiliation(s)
- Huub F J Savelkoul
- Cell Biology and Immunology, Wageningen University, Wageningen, P.O. Box 338, 6700 AH Wageningen, The Netherlands.
| | - Valerie A Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - Marius M Strioga
- Department of Immunology, Center of Oncosurgery, National Cancer Institute, P. Baublio Str. 3b-321, LT-08406 Vilnius, Lithuania.
| | - Virgil E J C Schijns
- Cell Biology and Immunology, Wageningen University, Wageningen, P.O. Box 338, 6700 AH Wageningen, The Netherlands.
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
- ERC-Belgium and ERC-The Netherlands, 5374 RE Schaijk, The Netherlands.
| |
Collapse
|
90
|
Lee SE, Nguyen CT, Kim SY, Thi TN, Rhee JH. Tetanus toxin fragment C fused to flagellin makes a potent mucosal vaccine. Clin Exp Vaccine Res 2015; 4:59-67. [PMID: 25649002 PMCID: PMC4313110 DOI: 10.7774/cevr.2015.4.1.59] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Revised: 10/20/2014] [Accepted: 10/27/2014] [Indexed: 11/15/2022] Open
Abstract
PURPOSE Recombinant subunit vaccines provide safe and targeted protection against microbial infections. However, the protective efficacy of recombinant subunit vaccines tends to be less potent than the whole cell vaccines, especially when they are administered through mucosal routes. We have reported that a bacterial flagellin has strong mucosal adjuvant activity to induce protective immune responses. In this study, we tested whether FlaB could be used as a fusion partner of subunit vaccine for tetanus. MATERIALS AND METHODS We constructed fusion proteins consisted with tetanus toxin fragment C (TTFC), the nontoxic C-terminal portion of tetanus toxin, and a Toll-like receptor 5 agonist from Vibrio vulnificus (FlaB). Mice were intranasally administered with fusion protein and protective immune responses of the vaccinated mice were analyzed. RESULTS FlaB-TTFC recombinant protein induced strong tetanus-specific antibody responses in both systemic and mucosal compartments and prolonged the survival of mice after challenge with a supra-lethal dose of tetanus toxin. CONCLUSION This study establishes FlaB as a successful fusion partner for recombinant subunit tetanus vaccine applicable through mucosal route, and it further endorses our previous observations that FlaB could be a stable adjuvant partner for mucosal vaccines.
Collapse
Affiliation(s)
- Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea. ; Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, Korea
| | - Chung Truong Nguyen
- Clinical Vaccine R&D Center, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea. ; Department of Microbiology, Chonnam National University Medical School, Gwangju, Korea
| | - Soo Young Kim
- Clinical Vaccine R&D Center, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea. ; Department of Microbiology, Chonnam National University Medical School, Gwangju, Korea
| | - Thinh Nguyen Thi
- Clinical Vaccine R&D Center, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea. ; Department of Microbiology, Chonnam National University Medical School, Gwangju, Korea
| | - Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University Hwasun Hospital, Chonnam National University Medical School, Hwasun, Korea. ; Department of Microbiology, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
91
|
Srivastava A, Gowda DV, Madhunapantula SV, Shinde CG, Iyer M. Mucosal vaccines: a paradigm shift in the development of mucosal adjuvants and delivery vehicles. APMIS 2015; 123:275-88. [PMID: 25630573 DOI: 10.1111/apm.12351] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 11/05/2014] [Indexed: 12/25/2022]
Abstract
Mucosal immune responses are the first-line defensive mechanisms against a variety of infections. Therefore, immunizations of mucosal surfaces from which majority of infectious agents make their entry, helps to protect the body against infections. Hence, vaccinization of mucosal surfaces by using mucosal vaccines provides the basis for generating protective immunity both in the mucosal and systemic immune compartments. Mucosal vaccines offer several advantages over parenteral immunization. For example, (i) ease of administration; (ii) non-invasiveness; (iii) high-patient compliance; and (iv) suitability for mass vaccination. Despite these benefits, to date, only very few mucosal vaccines have been developed using whole microorganisms and approved for use in humans. This is due to various challenges associated with the development of an effective mucosal vaccine that can work against a variety of infections, and various problems concerned with the safe delivery of developed vaccine. For instance, protein antigen alone is not just sufficient enough for the optimal delivery of antigen(s) mucosally. Hence, efforts have been made to develop better prophylactic and therapeutic vaccines for improved mucosal Th1 and Th2 immune responses using an efficient and safe immunostimulatory molecule and novel delivery carriers. Therefore, in this review, we have made an attempt to cover the recent advancements in the development of adjuvants and delivery carriers for safe and effective mucosal vaccine production.
Collapse
Affiliation(s)
- Atul Srivastava
- Department of Pharmaceutics, JSS College of Pharmacy, JSS University, Mysore, India
| | | | | | | | | |
Collapse
|
92
|
Singh S, Yang G, Byrareddy SN, Barry MA, Sastry KJ. Natural killer T cell and TLR9 agonists as mucosal adjuvants for sublingual vaccination with clade C HIV-1 envelope protein. Vaccine 2014; 32:6934-6940. [PMID: 25444819 DOI: 10.1016/j.vaccine.2014.10.051] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 07/22/2014] [Accepted: 10/23/2014] [Indexed: 02/06/2023]
Abstract
The vast majority of HIV-1 infections occur at mucosa during sexual contact. It may therefore be advantageous to provide mucosal barrier protection against this entry by mucosal vaccination. While a number of mucosal routes of vaccination are possible, many like enteric oral vaccines or intranasal vaccines have significant impediments that limit vaccine efficacy or pose safety risks. In contrast, immunogens applied to the sublingual region of the mouth could provide a simple route for mucosal vaccination. While sublingual immunization is appealing, this site does not always drive strong immune responses, particularly when using protein antigens. To address this issue, we have tested the ability of two mucosal adjuvants: alpha-galactosylceramide (αGalCer) that is a potent stimulator of natural killer T cells and CpG-oligodeoxynucleotide (CpG-ODN) a TLR9 agonist for their ability to amplify immune responses against clade C gp140 HIV-1 envelope protein antigen. Immunization with envelope protein alone resulted in a weak T cell and antibody responses. In contrast, CD4(+) and CD8(+) T cells responses in systemic and mucosal tissues were significantly higher in mice immunized with gp140 in the presence of either αGalCer or CpG-ODN and these responses were further augmented when the two adjuvants were used together. While both the adjuvants effectively increased gp140-specific serum IgG and vaginal IgA antibody levels, combining both significantly improved these responses. Memory T cell responses 60 days after immunization revealed αGalCer to be more potent than CpG-ODN and the combination of the αGalCer and CpG-ODN adjuvants was more effective than either alone. Serum and vaginal washes collected 60 days after immunization with gp140 with both αGalCer and CpG-ODN adjuvants had significant neutralization activity against Tier 1 and Tier 2 SHIVs. These data support the utility of the sublingual route for mucosal vaccination particularly in combination with αGalCer and CpG-ODN adjuvants.
Collapse
Affiliation(s)
- Shailbala Singh
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Guojun Yang
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States
| | - Siddappa N Byrareddy
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA, United States
| | - Michael A Barry
- Department of Internal Medicine, Division of Infectious Diseases, Translational Immunovirology Program, Department of Immunology, Department of Molecular Medicine, Mayo Clinic, Rochester, MN, United States
| | - K Jagannadha Sastry
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, United States; Department of Veterinary Sciences, The University of Texas M.D. Anderson Cancer Center, Bastrop, TX, United States.
| |
Collapse
|
93
|
Ou J, Shi W, Xu Y, Tao Z. Intranasal immunization with DNA vaccine coexpressing Der p 1 and ubiquitin in an allergic rhinitis mouse model. Ann Allergy Asthma Immunol 2014; 113:658-665.e1. [PMID: 25240330 DOI: 10.1016/j.anai.2014.08.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 08/12/2014] [Accepted: 08/16/2014] [Indexed: 11/29/2022]
Abstract
BACKGROUND The worldwide prevalence of allergic rhinitis (AR) is increasing, whereas treatments for AR remain limited in effect. Therefore, a new type of effective drug is eagerly in demand. OBJECTIVE To create a hypoallergenic vaccine by forced ubiquitination. METHODS In the present study, we constructed a DNA vaccine coexpressing Der p 1 allergen and murine ubiquitin, which used chitosan as a carrier. Through the vitro and vivo experiments, we evaluated its protective efficacy against AR. RESULTS The results indicated that the DNA vaccine pVAX1-Ub-Derp1/CS had been successfully constructed. This nanoparticle could not only transfect 293T cells in vitro but also transform cells in vivo. The inflammation of nasal mucosa in an AR murine model via immunization with pVAX1-Ub-Derp1/CS was less severe than those without treatments. Furthermore, it found that mice immunized with pVAX1-Ub-Derp1/CS generated a high level of specific IgG but a low level of specific IgE (P < .01). The significantly increased levels of interferon-γ and the significantly decreased levels of interleukins 4, 10, and 17 indicated that a TH1-type response was elicited by immunization with pVAX1-Ub-Derp1/CS (P < .01). This effect was especially stronger through intranasal immunization. CONCLUSION Nasal mucosal immunization and ubiquitination are efficacious strategies to enhance the efficiency and safety of DNA vaccine. The nanoparticle pVAX1-Ub-Derp1/CS is expected to be a new kind of effective vaccine for AR.
Collapse
Affiliation(s)
- Jing Ou
- Department of Otorhinolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wendan Shi
- Department of Otorhinolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Zezhang Tao
- Department of Otorhinolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
94
|
Monteiro-Maia R, de Pinho RT. Oral bacillus Calmette-Guérin vaccine against tuberculosis: why not? Mem Inst Oswaldo Cruz 2014; 109:838-45. [PMID: 25317714 PMCID: PMC4238780 DOI: 10.1590/0074-0276140091] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 06/16/2014] [Indexed: 12/31/2022] Open
Abstract
The bacillus Calmette-Guérin (BCG) vaccine is the only licensed vaccine for human use against tuberculosis (TB). Although controversy exists about its efficacy, the BCG vaccine is able to protect newborns and children against disseminated forms of TB, but fails to protect adults against active forms of TB. In the last few years, interest in the mucosal delivery route for the vaccine has been increasing owing to its increased capacity to induce protective immune responses both in the mucosal and the systemic immune compartments. Here, we show the importance of this route of vaccination in newly developed vaccines, especially for vaccines against TB.
Collapse
Affiliation(s)
| | - Rosa Teixeira de Pinho
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz-Fiocruz, Rio de
Janeiro, RJ, Brasil
| |
Collapse
|
95
|
Ye T, Yue Y, Fan X, Dong C, Xu W, Xiong S. M cell-targeting strategy facilitates mucosal immune response and enhances protection against CVB3-induced viral myocarditis elicited by chitosan-DNA vaccine. Vaccine 2014; 32:4457-4465. [PMID: 24958702 DOI: 10.1016/j.vaccine.2014.06.050] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/21/2014] [Accepted: 06/11/2014] [Indexed: 11/20/2022]
Abstract
Efficient delivery of antigen to mucosal associated lymphoid tissue is a first and critical step for successful induction of mucosal immunity by vaccines. Considering its potential transcytotic capability, M cell has become a more and more attractive target for mucosal vaccines. In this research, we designed an M cell-targeting strategy by which mucosal delivery system chitosan (CS) was endowed with M cell-targeting ability via conjugating with a CPE30 peptide, C terminal 30 amino acids of clostridium perfringens enterotoxin (CPE), and then evaluated its immune-enhancing ability in the context of coxsackievirus B3 (CVB3)-specific mucosal vaccine consisting of CS and a plasmid encoding CVB3 predominant antigen VP1. It had shown that similar to CS-pVP1, M cell-targeting CPE30-CS-pVP1 vaccine appeared a uniform spherical shape with about 300 nm diameter and +22 mV zeta potential, and could efficiently protect DNA from DNase I digestion. Mice were orally immunized with 4 doses of CPE30-CS-pVP1 containing 50 μg pVP1 at 2-week intervals and challenged with CVB3 4 weeks after the last immunization. Compared with CS-pVP1 vaccine, CPE30-CS-pVP1 vaccine had no obvious impact on CVB3-specific serum IgG level and splenic T cell immune responses, but significantly increased specific fecal SIgA level and augmented mucosal T cell immune responses. Consequently, much milder myocarditis and lower viral load were witnessed in CPE30-CS-pVP1 immunized group. The enhanced immunogenicity and immunoprotection were associated with the M cell-targeting ability of CPE30-CS-pVP1 which improved its mucosal uptake and transcytosis. Our findings indicated that CPE30-CS-pVP1 may represent a novel prophylactic vaccine against CVB3-induced myocarditis, and this M cell-targeting strategy indeed could be applied as a promising and universal platform for mucosal vaccine development.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Oral
- Animals
- Antibodies, Viral/analysis
- Antibodies, Viral/blood
- Chitosan/administration & dosage
- Coxsackievirus Infections/pathology
- Coxsackievirus Infections/prevention & control
- Disease Models, Animal
- Enterovirus B, Human/immunology
- Immunity, Mucosal
- Immunoglobulin A, Secretory/analysis
- Immunoglobulin G/blood
- Male
- Mice, Inbred BALB C
- Myocarditis/pathology
- Myocarditis/prevention & control
- T-Lymphocytes/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Viral Load
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Ting Ye
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou 215123, Jiangsu, PR China
| | - Yan Yue
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou 215123, Jiangsu, PR China
| | - Xiangmei Fan
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou 215123, Jiangsu, PR China
| | - Chunsheng Dong
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou 215123, Jiangsu, PR China
| | - Wei Xu
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou 215123, Jiangsu, PR China
| | - Sidong Xiong
- Jiangsu Provincial Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, 199 Ren-Ai Road, Suzhou 215123, Jiangsu, PR China.
| |
Collapse
|
96
|
PMA induces vaccine adjuvant activity by the modulation of TLR signaling pathway. Mediators Inflamm 2014; 2014:406514. [PMID: 24948847 PMCID: PMC4052173 DOI: 10.1155/2014/406514] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 03/31/2014] [Accepted: 04/01/2014] [Indexed: 02/08/2023] Open
Abstract
Toll-like receptor (TLR) ligands are being developed for use as vaccine adjuvants and as immunomodulators because of their ability to stimulate innate and adaptive immune responses. Flagellin, a TLR5 ligand, was reported to show potent mucosal vaccine adjuvant activity. To identify ligands that potentiate the adjuvant activity of flagellin, we screened a plant library using HEK293T cells transiently cotransfected with phTLR5 and pNF-κB-SEAP plasmids. The 90% EtOH extract from Croton tiglium showed significant NF-κB transactivation in a TLR5-independent manner along with the increase of a flagellin activity. We have studied to characterize an active component from Croton tiglium and to elucidate the action mechanisms. Phorbol 12-myristate 13-acetate (PMA) was isolated as an active component of Croton tiglium by activity-guided fractionation, column chromatography, HPLC, NMR, and MS. PMA at a range of nM induced PKC-dependent NF-κB activation and IL-8 production in both TLR5− and TLR5+ assay systems. In in vivo mouse vaccination model, PMA induced antigen-specific IgG and IgA antibody responses and increased IL-12 production corresponding to T cell responses in spleen lymphocytes. These results suggest that PMA would serve as an efficacious mucosal vaccine adjuvant.
Collapse
|
97
|
Rosales-Mendoza S, Rubio-Infante N, Zarazúa S, Govea-Alonso DO, Martel-Gallegos G, Moreno-Fierros L. Plant-based vaccines for Alzheimer's disease: an overview. Expert Rev Vaccines 2014; 13:429-41. [PMID: 24405291 DOI: 10.1586/14760584.2014.874948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Plants are considered advantageous platforms for biomanufacturing recombinant vaccines. This constitutes a field of intensive research and some plant-derived vaccines are expected to be marketed in the near future. In particular, plant-based production of immunogens targeting molecules with implications on the pathology of Alzheimer's has been explored over the last decade. These efforts involve targeting amyloid beta and β-secretase with several immunogen configurations that have been evaluated in test animals. The results of these developments are analyzed in this review. Perspectives on the topic are identified, such as exploring additional antigen configurations and adjuvants in order to improve immunization schemes, characterizing in detail the elicited immune responses, and immunological considerations in the achievement of therapeutic humoral responses via mucosal immunization. Safety concerns related to these therapies will also be discussed.
Collapse
Affiliation(s)
- Sergio Rosales-Mendoza
- Laboratorio de Biofarmacéuticos recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí , Av. Dr. Manuel Nava 6, SLP, 78210 , México
| | | | | | | | | | | |
Collapse
|
98
|
Cheng C, Pal S, Tifrea D, Jia Z, de la Maza LM. A vaccine formulated with a combination of TLR-2 and TLR-9 adjuvants and the recombinant major outer membrane protein elicits a robust immune response and significant protection against a Chlamydia muridarum challenge. Microbes Infect 2014; 16:244-52. [PMID: 24291713 PMCID: PMC3965591 DOI: 10.1016/j.micinf.2013.11.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 12/17/2022]
Abstract
Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen in the World and there is a need for a vaccine. To enhance the immunogenicity of a vaccine formulated with the Chlamydia muridarum (Cm) mouse pneumonitis recombinant major outer membrane protein (MOMP), we used combinations of Pam2CSK4 + CpG-1826 and Montanide ISA 720 VG + CpG-1826 as adjuvants. Neisseria gonorrhoeae recombinant porin B (Ng-PorB) was used as the antigen control with the same adjuvants. Female BALB/c mice were immunized twice in the nares (i.n.) or in the colon (cl.) and were boosted twice by the intramuscular plus subcutaneous (i.m. + s.c.) routes. Based on the IgG2a/IgG1 ratio in sera, mice immunized with MOMP + Pam2CSK4 + CpG-1826 showed a strong Th2 response while animals vaccinated with MOMP + Montanide ISA 720 VG + CpG-1826 had a Th1 response. Both groups of mice also developed robust Cm-specific T cell proliferation and high levels of IFN-γ. Four weeks after the last immunization, the mice were challenged i.n. with 10(4) inclusion-forming units (IFU) of Cm. Using changes in body weight and number of IFU recovered from the lungs at 10 days post-challenge mice immunized i.n. + i.m./s.c. with MOMP + Pam2CSK4 + CpG-1826 were better protected than other groups. In conclusion, MOMP adjuvanted with Pam2CSK4 + CpG-1826, elicits strong humoral and cellular immune responses and induces significant protection against Chlamydia.
Collapse
Affiliation(s)
- Chunmei Cheng
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Sukumar Pal
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Delia Tifrea
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Zhenyu Jia
- Translational Cancer Biology, Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Luis M de la Maza
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA.
| |
Collapse
|
99
|
Rhee JH. Towards Vaccine 3.0: new era opened in vaccine research and industry. Clin Exp Vaccine Res 2013; 3:1-4. [PMID: 24427757 PMCID: PMC3890443 DOI: 10.7774/cevr.2014.3.1.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 11/20/2013] [Accepted: 11/25/2013] [Indexed: 11/15/2022] Open
Affiliation(s)
- Joon Haeng Rhee
- Department of Microbiology and Clinical Vaccine R&D Center, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
100
|
Smith A, Perelman M, Hinchcliffe M. Chitosan: a promising safe and immune-enhancing adjuvant for intranasal vaccines. Hum Vaccin Immunother 2013; 10:797-807. [PMID: 24346613 PMCID: PMC4130252 DOI: 10.4161/hv.27449] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 11/27/2013] [Accepted: 12/06/2013] [Indexed: 11/19/2022] Open
Abstract
The nasal route is attractive for the delivery of vaccines in that it not only offers an easy to use, non-invasive, needle-free alternative to more conventional parenteral injection, but it also creates an opportunity to elicit both systemic and (crucially) mucosal immune responses which may increase the capability of controlling pathogens at the site of entry. Immune responses to "naked" antigens are often modest and it is widely accepted that incorporation of an adjuvant is a prerequisite for the achievement of clinically effective nasal vaccines. Many existing adjuvants are sub-optimal or unsuitable because of local toxicity or poor enhancement of immunogenicity. Chitosan, particularly chitosan salts, have now been used in several preclinical and clinical studies with good tolerability, excellent immune stimulation and positive clinical results across a number of infections. Particularly significant evidence supporting chitosan as an adjuvant for nasal vaccination comes from clinical investigations on a norovirus vaccine; this demonstrated the ability of chitosan (ChiSys®), when combined with monophosphoryl lipid, to evoke robust immunological responses and confer protective immunity following (enteral) norovirus challenge. This article summarizes the totality of the meaningful information (including key unpublished data) supporting the development of chitosan-adjuvanted vaccines.
Collapse
Affiliation(s)
- Alan Smith
- Archimedes Development Limited; Albert Einstein Centre; Nottingham Science Park; Nottingham, UK
| | - Michael Perelman
- Archimedes Development Limited; Albert Einstein Centre; Nottingham Science Park; Nottingham, UK
| | | |
Collapse
|