1
|
Kato Y, Kumanogoh A. The immune memory of innate immune systems. Int Immunol 2025; 37:195-202. [PMID: 39588905 DOI: 10.1093/intimm/dxae067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/24/2024] [Indexed: 11/27/2024] Open
Abstract
Immune memory has long been considered a function specific to adaptive immune systems; however, adaptive immune memory alone has not fully explained the mechanism by which vaccines exert their protective effects against nontarget pathogens. Recently, trained immunity, in which human monocytes vaccinated with bacillus Calmette-Guérin become highly responsive to pathogens other than Mycobacterium tuberculosis, has been reported. However, a phenomenon called endotoxin tolerance is also known, in which monocyte responsiveness is attenuated after the first lipopolysaccharide stimulation. These phenomena represent an altered innate immune response after the initial exposure to the stimulus, indicating that memories are formed in the innate immune system. In this review, we discuss trained immunity and endotoxin tolerance, known as innate immune memory, and innate immune memory formation by mRNA vaccines, which have been newly used in the coronavirus disease 2019 (COVID-19) pandemic and are considered important vaccine modalities in the future.
Collapse
Affiliation(s)
- Yasuhiro Kato
- Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Center for Infectious Diseases Education and Research, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
2
|
Muranishi K, Kinoshita M, Inoue K, Ohara J, Mihara T, Sudo K, Ishii KJ, Sawa T, Ishikura H. Antibody Response Following the Intranasal Administration of SARS-CoV-2 Spike Protein-CpG Oligonucleotide Vaccine. Vaccines (Basel) 2023; 12:5. [PMID: 38276664 PMCID: PMC10818492 DOI: 10.3390/vaccines12010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/13/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
The new coronavirus infection causes severe respiratory failure following respiratory tract infection with severe acute respiratory syndrome-related coronavirus (SARS-CoV-2). All currently approved vaccines are administered intramuscularly; however, intranasal administration enhances mucosal immunity, facilitating the production of a less invasive vaccine with fewer adverse events. Herein, a recombinant vaccine combining the SARS-CoV-2 spike protein receptor-binding domain (RBD), or S1 protein, with CpG-deoxyoligonucleotide (ODN) or aluminum hydroxide (alum) adjuvants was administered intranasally or subcutaneously to mice. Serum-specific IgG titers, IgA titers in the alveolar lavage fluid, and neutralizing antibody titers were analyzed. The nasal administration of RBD protein did not increase serum IgG or IgA titers in the alveolar lavage fluid. However, a significant increase in serum IgG was observed in the intranasal group administered with S1 protein with CpG-ODN and the subcutaneous group administered with S1 protein with alum. The IgA and IgG levels increased significantly in the alveolar lavage fluid only after the intranasal administration of the S1 protein with CpG-ODN. The neutralizing antibody titers in serum and bronchoalveolar lavage were significantly higher in the intranasal S1-CpG group than in every other group. Hence, the nasal administration of the S1 protein vaccine with CpG adjuvant might represent an effective vaccine candidate.
Collapse
Affiliation(s)
- Kentaro Muranishi
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka 814-0133, Japan; (K.M.); (H.I.)
| | - Mao Kinoshita
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (K.I.); (J.O.); (T.M.); (K.S.); (T.S.)
| | - Keita Inoue
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (K.I.); (J.O.); (T.M.); (K.S.); (T.S.)
| | - Junya Ohara
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (K.I.); (J.O.); (T.M.); (K.S.); (T.S.)
| | - Toshihito Mihara
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (K.I.); (J.O.); (T.M.); (K.S.); (T.S.)
| | - Kazuki Sudo
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (K.I.); (J.O.); (T.M.); (K.S.); (T.S.)
| | - Ken J. Ishii
- Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan;
| | - Teiji Sawa
- Department of Anesthesiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (K.I.); (J.O.); (T.M.); (K.S.); (T.S.)
| | - Hiroyasu Ishikura
- Department of Emergency and Critical Care Medicine, Faculty of Medicine, Fukuoka University, Fukuoka 814-0133, Japan; (K.M.); (H.I.)
| |
Collapse
|
3
|
Tzeng TT, Chai KM, Chen IH, Chang RY, Chiang JR, Liu SJ. A TLR9 agonist synergistically enhances protective immunity induced by an Alum-adjuvanted H7N9 inactivated whole-virion vaccine. Emerg Microbes Infect 2023; 12:2249130. [PMID: 37585273 PMCID: PMC10467522 DOI: 10.1080/22221751.2023.2249130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/10/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Antigen sparing is an important strategy for pandemic vaccine development because of the limitation of worldwide vaccine production during disease outbreaks. However, several clinical studies have demonstrated that the current aluminum (Alum)-adjuvanted influenza vaccines fail to sufficiently enhance immune responses to meet licensing criteria. Here, we used pandemic H7N9 as a model virus to demonstrate that a 10-fold lower amount of vaccine antigen combined with Alum and TLR9 agonist can provide stronger protective effects than using Alum as the sole adjuvant. We found that the Alum/CpG 1018 combination adjuvant could induce more robust virus-specific humoral immune responses, including higher total IgG production, hemagglutination-inhibiting antibody activity, and neutralizing antibody titres, than the Alum-adjuvanted formulation. Moreover, this combination adjuvant shifted the immune response toward a Th1-biased immune response. Importantly, the Alum/CpG 1018-formulated vaccine could confer better protective immunity against H7N9 challenge than that adjuvanted with Alum alone. Notably, the addition of CpG 1018 to the Alum-adjuvanted H7N9 whole-virion vaccine exhibited an antigen-sparing effect without compromising vaccine efficacy. These findings have significant implications for improving Alum-adjuvanted influenza vaccines using the approved adjuvant CpG 1018 for pandemic preparedness.
Collapse
Affiliation(s)
- Tsai-Teng Tzeng
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Kit Man Chai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - I-Hua Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Ray-Yuan Chang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Jen-Ron Chiang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
4
|
Harnessing Nasal Immunity with IgA to Prevent Respiratory Infections. IMMUNO 2022. [DOI: 10.3390/immuno2040036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The nasal cavity is a primary checkpoint for the invasion of respiratory pathogens. Numerous pathogens, including SARS-CoV-2, S. pneumoniae, S. aureus, etc., can adhere/colonize nasal lining to trigger an infection. Secretory IgA (sIgA) serves as the first line of immune defense against foreign pathogens. sIgA facilitates clearance of pathogenic microbes by intercepting their access to epithelial receptors and mucus entrapment through immune exclusion. Elevated levels of neutralizing IgA at the mucosal surfaces are associated with a high level of protection following intranasal immunizations. This review summarizes recent advances in intranasal vaccination technology and challenges in maintaining nominal IgA levels at the mucosal surface. Overall, the review emphasizes the significance of IgA-mediated nasal immunity, which holds a tremendous potential to mount protection against respiratory pathogens.
Collapse
|
5
|
Bentes GA, Lanzarini NM, Guimarães JR, Heinemann MB, Volotão EDM, da Silva ADS, Heneine LGD, de Oliveira JM, Pinto MA. Production and Evaluation of Chicken Egg Yolk Immunoglobulin (IgY) against Human and Simian Rotaviruses. Viruses 2022; 14:v14091995. [PMID: 36146801 PMCID: PMC9502344 DOI: 10.3390/v14091995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022] Open
Abstract
Producing specific antibodies in chickens is an attractive approach for diagnosis or therapeutic applications. Besides the high immunoglobulin Y (IgY) yield transferred to the egg yolk and its suitability for large-scale production, such an approach is more bioethical for animal maintenance. The IgY technology offers new possibilities for application in human and veterinary diagnostics and therapeutics, including strategies for treating severe intestinal diseases in children, particularly in emerging countries. Herein, we describe the production and purification of polyclonal antibodies against rotavirus group A (RVA) in immunised hens aiming at its application in prophylaxis and treatment of rotavirus-induced diarrhoea. For this purpose, we inoculated Rhodia laying chickens (Gallus gallus domesticus) with two or three doses of RVA combined with adjuvants or only adjuvants (control group). As the egg-laying period began, the yolk protein purification processes yielded a high concentration of specific IgY, the highest titre resulting from the group of hens that received three doses of the immunogen. The purified IgY blocked the functional activity of RVA in MA-104 cells, thus confirming the neutralisation ability. Therefore, anti-RVA IgY could be a promising candidate for pre- and post-exposure prevention or treatment of rotavirus-induced diarrhoea.
Collapse
Affiliation(s)
- Gentil Arthur Bentes
- Laboratório de Desenvolvimento Tecnológico em Virologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
- Correspondence: (G.A.B.); (J.M.d.O.)
| | - Natália Maria Lanzarini
- Laboratório de Desenvolvimento Tecnológico em Virologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
- Laboratório de Virologia Comparada e Ambiental, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Juliana Rodrigues Guimarães
- Laboratório de Desenvolvimento Tecnológico em Virologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Marcos Bryan Heinemann
- Departamento de Medicina Veterinária Preventiva e Saúde Animal, Faculdade de Medicina Veterinária e Zootecnia, Universidade de São Paulo, São Paulo 05508-270, Brazil
| | - Eduardo de Mello Volotão
- Laboratório de Virologia Comparada e Ambiental, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | - Alexandre dos Santos da Silva
- Laboratório de Desenvolvimento Tecnológico em Virologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| | | | - Jaqueline Mendes de Oliveira
- Laboratório de Desenvolvimento Tecnológico em Virologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
- Correspondence: (G.A.B.); (J.M.d.O.)
| | - Marcelo Alves Pinto
- Laboratório de Desenvolvimento Tecnológico em Virologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro 21040-360, Brazil
| |
Collapse
|
6
|
Cao X, Cordova AF, Li L. Therapeutic Interventions Targeting Innate Immune Receptors: A Balancing Act. Chem Rev 2021; 122:3414-3458. [PMID: 34870969 DOI: 10.1021/acs.chemrev.1c00716] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The innate immune system is an organism's first line of defense against an onslaught of internal and external threats. The downstream adaptive immune system has been a popular target for therapeutic intervention, while there is a relative paucity of therapeutics targeting the innate immune system. However, the innate immune system plays a critical role in many human diseases, such as microbial infection, cancer, and autoimmunity, highlighting the need for ongoing therapeutic research. In this review, we discuss the major innate immune pathways and detail the molecular strategies underpinning successful therapeutics targeting each pathway as well as previous and ongoing efforts. We will also discuss any recent discoveries that could inform the development of novel therapeutic strategies. As our understanding of the innate immune system continues to develop, we envision that therapies harnessing the power of the innate immune system will become the mainstay of treatment for a wide variety of human diseases.
Collapse
|
7
|
Xu H, Cai L, Hufnagel S, Cui Z. Intranasal vaccine: Factors to consider in research and development. Int J Pharm 2021; 609:121180. [PMID: 34637935 DOI: 10.1016/j.ijpharm.2021.121180] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 01/01/2023]
Abstract
Most existing vaccines for human use are administered by needle-based injection. Administering vaccines needle-free intranasally has numerous advantages over by needle-based injection, but there are only a few intranasal vaccines that are currently approved for human use, and all of them are live attenuated influenza virus vaccines. Clearly, there are immunological as well as non-immunological challenges that prevent vaccine developers from choosing the intranasal route of administration. We reviewed current approved intranasal vaccines and pipelines and described the target of intranasal vaccines, i.e. nose and lymphoid tissues in the nasal cavity. We then analyzed factors unique to intranasal vaccines that need to be considered when researching and developing new intranasal vaccines. We concluded that while the choice of vaccine formulations, mucoadhesives, mucosal and epithelial permeation enhancers, and ligands that target M-cells are important, safe and effective intranasal mucosal vaccine adjuvants are needed to successfully develop an intranasal vaccine that is not based on live-attenuated viruses or bacteria. Moreover, more effective intranasal vaccine application devices that can efficiently target a vaccine to lymphoid tissues in the nasal cavity as well as preclinical animal models that can better predict intranasal vaccine performance in clinical trials are needed to increase the success rate of intranasal vaccines in clinical trials.
Collapse
Affiliation(s)
- Haiyue Xu
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Lucy Cai
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Stephanie Hufnagel
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States
| | - Zhengrong Cui
- The University of Texas at Austin, College of Pharmacy, Division of Molecular Pharmaceutics and Drug Delivery, Austin, TX, United States.
| |
Collapse
|
8
|
Fiege JK, Block KE, Pierson MJ, Nanda H, Shepherd FK, Mickelson CK, Stolley JM, Matchett WE, Wijeyesinghe S, Meyerholz DK, Vezys V, Shen SS, Hamilton SE, Masopust D, Langlois RA. Mice with diverse microbial exposure histories as a model for preclinical vaccine testing. Cell Host Microbe 2021; 29:1815-1827.e6. [PMID: 34731647 DOI: 10.1016/j.chom.2021.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 08/30/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022]
Abstract
Laboratory mice comprise an expeditious model for preclinical vaccine testing; however, vaccine immunogenicity in these models often inadequately translates to humans. Reconstituting physiologic microbial experience to specific pathogen-free (SPF) mice induces durable immunological changes that better recapitulate human immunity. We examined whether mice with diverse microbial experience better model human responses post vaccination. We co-housed laboratory mice with pet-store mice, which have varied microbial exposures, and then assessed immune responses to influenza vaccines. Human transcriptional responses to influenza vaccination are better recapitulated in co-housed mice. Although SPF and co-housed mice were comparably susceptible to acute influenza infection, vaccine-induced humoral responses were dampened in co-housed mice, resulting in poor control upon challenge. Additionally, protective heterosubtypic T cell immunity was compromised in co-housed mice. Because SPF mice exaggerated humoral and T cell protection upon influenza vaccination, reconstituting microbial experience in laboratory mice through co-housing may better inform preclinical vaccine testing.
Collapse
Affiliation(s)
- Jessica K Fiege
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Katharine E Block
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mark J Pierson
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Hezkiel Nanda
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Frances K Shepherd
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Clayton K Mickelson
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - J Michael Stolley
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - William E Matchett
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sathi Wijeyesinghe
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - David K Meyerholz
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Vaiva Vezys
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Steven S Shen
- Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sara E Hamilton
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - David Masopust
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Ryan A Langlois
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
9
|
Hsieh MS, Hsu CW, Tu LL, Chai KM, Yu LL, Wu CC, Chen MY, Chiang CY, Liu SJ, Liao CL, Chen HW. Intranasal Vaccination With Recombinant Antigen-FLIPr Fusion Protein Alone Induces Long-Lasting Systemic Antibody Responses and Broad T Cell Responses. Front Immunol 2021; 12:751883. [PMID: 34707615 PMCID: PMC8543008 DOI: 10.3389/fimmu.2021.751883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022] Open
Abstract
A simple formulation is urgently needed for mucosal vaccine development. We employed formyl peptide receptor-like 1 inhibitory protein (FLIPr), an FcγR antagonist secreted by Staphylococcus aureus, as a vector to target ovalbumin (OVA) to dendritic cells (DCs) via intranasal administration. Our results demonstrate that intranasal administration of recombinant OVA-FLIPr fusion protein (rOVA-FLIPr) alone efficiently delivers OVA to DCs in nasal lymphoid tissue. Subsequently, OVA-specific IgG and IgA antibodies in the circulatory system and IgA antibodies in mucosal tissue were detected. Importantly, activation of OVA-specific CD4+ and CD8+ T cells and induction of a broad-spectrum cytokine secretion profile were detected after intranasal administration of rOVA-FLIPr alone in immunocompetent C57BL/6 mice. Furthermore, we employed immunodeficient AG129 mice as a Zika virus infection model and demonstrated that intranasal administration of recombinant Zika virus envelope protein domain III-FLIPr fusion protein induced protective immune responses against the Zika virus. These results suggest that antigen-FLIPr fusion protein alone via intranasal administration can be applied to mucosal vaccine development.
Collapse
Affiliation(s)
- Ming-Shu Hsieh
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chia-Wei Hsu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Ling-Ling Tu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Kit Man Chai
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Li-Lu Yu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chiao-Chieh Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Mei-Yu Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Chen-Yi Chiang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Liu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Hsin-Wei Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
10
|
Abstract
CpG Oligonucleotides (ODN) are immunomodulatory synthetic oligonucleotides specifically designed to stimulate Toll-like receptor 9. TLR9 is expressed on human plasmacytoid dendritic cells and B cells and triggers an innate immune response characterized by the production of Th1 and pro-inflammatory cytokines. This chapter reviews recent progress in understanding the mechanism of action of CpG ODN and provides an overview of human clinical trial results using CpG ODN to improve vaccines for the prevention/treatment of cancer, allergy, and infectious disease.
Collapse
Affiliation(s)
| | | | - Dennis M Klinman
- National Cancer Institute, NIH, Frederick, MD, USA.
- Leitman Klinman Consulting, Potomac, MD, USA.
| |
Collapse
|
11
|
Kataoka K, Kawabata S, Koyanagi K, Hashimoto Y, Miyake T, Fujihashi K. Respiratory FimA-Specific Secretory IgA Antibodies Upregulated by DC-Targeting Nasal Double DNA Adjuvant Are Essential for Elimination of Porphyromonas gingivalis. Front Immunol 2021; 12:634923. [PMID: 33717178 PMCID: PMC7948520 DOI: 10.3389/fimmu.2021.634923] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/18/2021] [Indexed: 11/13/2022] Open
Abstract
Our previous studies showed that a combination of a DNA plasmid encoding Flt3 ligand (pFL) and CpG oligodeoxynucleotides 1826 (CpG ODN) (FL/CpG) as a nasal adjuvant provoked antigen-specific immune responses. In this study, we investigated the efficacy of a nasal vaccine consisting of FimA as the structural subunit of Porphyromonas gingivalis (P. gingivalis) fimbriae and FL/CpG for the induction of FimA-specific antibody (Ab) responses and their protective roles against nasal and lung infection by P. gingivalis, a keystone pathogen in the etiology of periodontal disease. C57BL/6 mice were nasally immunized with recombinant FimA (rFimA) plus FL/CpG three times at weekly intervals. As a control, mice were given nasal rFimA alone. Nasal washes (NWs) and bronchoalveolar lavage fluid (BALF) of mice given nasal rFimA plus FL/CpG resulted in increased levels of rFimA-specific secretory IgA (SIgA) and IgG Ab responses when compared with those in controls. Significantly increased numbers of CD8- or CD11b-expressing mature-type dendritic cells (DCs) were detected in the respiratory inductive and effector tissues of mice given rFimA plus FL/CpG. Additionally, significantly upregulated Th1/Th2-type cytokine responses by rFimA-stimulated CD4+ T cells were noted in the respiratory effector tissues. When mice were challenged with live P. gingivalis via the nasal route, mice immunized nasally with rFimA plus FL/CpG inhibited P. gingivalis colonization in the nasal cavities and lungs. In contrast, controls failed to show protection. Of interest, when IgA-deficient mice given nasal rFimA plus FL/CpG were challenged with nasal P. gingivalis, the inhibition of bacterial colonization in the respiratory tracts was not seen. Taken together, these results show that nasal FL/CpG effectively enhanced DCs and provided balanced Th1- and Th2-type cytokine response-mediated rFimA-specific IgA protective immunity in the respiratory tract against P. gingivalis. A nasal administration with rFimA and FL/CpG could be a candidate for potent mucosal vaccines for the elimination of inhaled P. gingivalis in periodontal patients.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Antibodies, Bacterial/metabolism
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- Bacteroidaceae Infections/immunology
- Bacteroidaceae Infections/microbiology
- Bacteroidaceae Infections/prevention & control
- Disease Models, Animal
- Female
- Fimbriae Proteins/administration & dosage
- Fimbriae Proteins/genetics
- Fimbriae Proteins/immunology
- Immunity, Mucosal/drug effects
- Immunization Schedule
- Immunogenicity, Vaccine
- Immunoglobulin A, Secretory/metabolism
- Membrane Proteins/administration & dosage
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice, Inbred C57BL
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Porphyromonas gingivalis/immunology
- Porphyromonas gingivalis/pathogenicity
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Respiratory System/drug effects
- Respiratory System/immunology
- Respiratory System/metabolism
- Respiratory System/microbiology
- Time Factors
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Mice
Collapse
Affiliation(s)
- Kosuke Kataoka
- Department of Preventive and Community Dentistry, Faculty of Dentistry, Osaka Dental University, Hirakata, Japan
| | - Shigetada Kawabata
- Department of Oral and Molecular Microbiology, Graduate School of Dentistry, Osaka University, Suita, Japan
| | - Kayo Koyanagi
- Department of Preventive and Community Dentistry, Faculty of Dentistry, Osaka Dental University, Hirakata, Japan
| | - Yoshiya Hashimoto
- Department of Biomaterials, Faculty of Dentistry, Osaka Dental University, Hirakata, Japan
| | - Tatsuro Miyake
- Department of Preventive and Community Dentistry, Faculty of Dentistry, Osaka Dental University, Hirakata, Japan
| | - Kohtaro Fujihashi
- Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Pediatric Dentistry, School of Dentistry, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
12
|
Calzas C, Chevalier C. Innovative Mucosal Vaccine Formulations Against Influenza A Virus Infections. Front Immunol 2019; 10:1605. [PMID: 31379823 PMCID: PMC6650573 DOI: 10.3389/fimmu.2019.01605] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/27/2019] [Indexed: 12/11/2022] Open
Abstract
Despite efforts made to develop efficient preventive strategies, infections with influenza A viruses (IAV) continue to cause serious clinical and economic problems. Current licensed human vaccines are mainly inactivated whole virus particles or split-virion administered via the parenteral route. These vaccines provide incomplete protection against IAV in high-risk groups and are poorly/not effective against the constant antigenic drift/shift occurring in circulating strains. Advances in mucosal vaccinology and in the understanding of the protective anti-influenza immune mechanisms suggest that intranasal immunization is a promising strategy to fight against IAV. To date, human mucosal anti-influenza vaccines consist of live attenuated strains administered intranasally, which elicit higher local humoral and cellular immune responses than conventional parenteral vaccines. However, because of inconsistent protective efficacy and safety concerns regarding the use of live viral strains, new vaccine candidates are urgently needed. To prime and induce potent and long-lived protective immune responses, mucosal vaccine formulations need to ensure the immunoavailability and the immunostimulating capacity of the vaccine antigen(s) at the mucosal surfaces, while being minimally reactogenic/toxic. The purpose of this review is to compile innovative delivery/adjuvant systems tested for intranasal administration of inactivated influenza vaccines, including micro/nanosized particulate carriers such as lipid-based particles, virus-like particles and polymers associated or not with immunopotentiatory molecules including microorganism-derived toxins, Toll-like receptor ligands and cytokines. The capacity of these vaccines to trigger specific mucosal and systemic humoral and cellular responses against IAV and their (cross)-protective potential are considered.
Collapse
Affiliation(s)
- Cynthia Calzas
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| | - Christophe Chevalier
- VIM, UR892, Equipe Virus Influenza, INRA, University PARIS-SACLAY, Jouy-en-Josas, France
| |
Collapse
|
13
|
CpG enhances the immunogenicity of heterologous DNA-prime/protein-boost vaccination with the heavy chain myosin of Brugia malayi in BALB/c mice. Parasitol Res 2019; 118:1943-1952. [PMID: 31069533 DOI: 10.1007/s00436-019-06318-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 04/09/2019] [Indexed: 10/26/2022]
Abstract
The recombinant heavy chain myosin of Brugia malayi (Bm-Myo) has earlier been reported as a potent vaccine candidate in our lab. Subsequently, we further enhanced its efficacy employing heterologous DNA prime/protein boost (Myo-pcD+Bm-Myo) immunization approach that produced superior immune-protection than protein or DNA vaccination. In the present study, we evaluated the efficacy of heterologous prime boost vaccination in combination with CpG, synthetic oligodeoxynucleotides (ODN) adjuvant in BALB/c mice. The results showed that CpG/Myo-pcD+Bm-Myo conferred 84.5 ± 0.62% protection against B. malayi infective larval challenge which was considerably higher than Myo-pcD+Bm-Myo (75.6 ± 1.10%) following immunization. Although, both the formulations of immunization elicited robust production of specific IgG antibody and their isotypes (IgG1, IgG2a, IgG2b, and IgG3); however, CpG/Myo-pcD+Bm-Myo predominantly enhanced the level of IgG2a suggesting Th1 biased immune response in presence of CpG. Furthermore, spleen isolated from mice that immunized with CpG/Myo-pcD+Bm-Myo had greater accumulation of CD4+, CD8+, and CD19+ B cells and there was an augmented expression of co-stimulatory molecules CD40, CD86 on host dendritic cells (DCs). In contrast to Myo-pcD+Bm-Myo group, the splenocytes of CpG/Myo-pcD+Bm-Myo immunized mice developed comparatively higher pro-inflammatory cytokines IL-2 and IFN-γ leaving anti-inflammatory cytokine levels unchanged. Moreover, CpG formulation also upregulated the RNA expression of IL-12 and TNF-α in spleenocytes. The current findings suggest that the use of CpG would be more advantageous as an adjuvant predominantly in DNA/protein prime boost vaccine against Bm-Myo and presumably also for filarial infection.
Collapse
|
14
|
Naito Y, Hamaoka S, Kinoshita M, Kainuma A, Shimizu M, Katoh H, Moriyama K, Ishii KJ, Sawa T. The protective effects of nasal PcrV-CpG oligonucleotide vaccination against Pseudomonas aeruginosa pneumonia. Microbiol Immunol 2019; 62:774-785. [PMID: 30378708 DOI: 10.1111/1348-0421.12658] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/13/2018] [Accepted: 10/27/2018] [Indexed: 01/15/2023]
Abstract
An effective vaccine against Pseudomonas aeruginosa would be hugely beneficial to people who are susceptible to the serious infections it can cause. Vaccination against PcrV of the P. aeruginosa type III secretion system is a potential prophylactic strategy for improving the incidence and prognosis of P. aeruginosa pneumonia. Here, the effect of nasal PcrV adjuvanted with CpG oligodeoxynucleotide (CpG) was compared with a nasal PcrV/aluminum hydroxide gel (alum) vaccine. Seven groups of mice were vaccinated intranasally with one of the following: 1, PcrV-CpG; 2, PcrV-alum; 3, PcrV alone; 4, CpG alone; 5, alum alone; 6 and 7, saline control. Fifty days after the first immunization, anti-PcrV IgG, IgA and IgG isotype titers were measured; significant increases in these titers were detected only in the PcrV-CpG vaccinated mice. The vaccinated mice were then intratracheally infected with a lethal dose of P. aeruginosa and their body temperatures and survival monitored for 24 hr, edema, bacteria, myeloperoxidase activity and lung histology also being evaluated at 24 hr post-infection. It was found that 73% of the PcrV-CpG-vaccinated mice survived, whereas fewer than 30% of the mice vaccinated with PcrV-alum or adjuvant alone survived. Lung edema and other inflammation-related variables were less severe in the PcrV-CpG group. The significant increase in PcrV-specific IgA titers detected following PcrV-CpG vaccination is probably a component of the disease protection mechanism. Overall, our data show that intranasal PcrV-CpG vaccination has potential efficacy for clinical application against P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Yoshifumi Naito
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Saeko Hamaoka
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Mao Kinoshita
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Atsushi Kainuma
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Masaru Shimizu
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Hideya Katoh
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| | - Kiyoshi Moriyama
- Department of Anesthesiology, School of Medicine, Kyorin University, 6-20-2 Shinkawa, Mitaka, Tokyo, 181-8611, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Asagi, Saito, Ibaraki, Osaka, 567-0085, Japan.,Laboratory of Vaccine Science, Immunology Frontier Research Center, World Premier International Research Center, Osaka University, 1-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Teiji Sawa
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi Hirokoji, Kamigyo, Kyoto, 602-8566, Japan
| |
Collapse
|
15
|
Hayashi T, Momota M, Kuroda E, Kusakabe T, Kobari S, Makisaka K, Ohno Y, Suzuki Y, Nakagawa F, Lee MSJ, Coban C, Onodera R, Higashi T, Motoyama K, Ishii KJ, Arima H. DAMP-Inducing Adjuvant and PAMP Adjuvants Parallelly Enhance Protective Type-2 and Type-1 Immune Responses to Influenza Split Vaccination. Front Immunol 2018; 9:2619. [PMID: 30515151 PMCID: PMC6255964 DOI: 10.3389/fimmu.2018.02619] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/24/2018] [Indexed: 01/14/2023] Open
Abstract
Recently, it was reported that 2-hydroxypropyl-β-cyclodextrin (HP-β-CyD), a common pharmaceutical additive, can act as a vaccine adjuvant to enhance protective type-2 immunogenicity to co-administered seasonal influenza split vaccine by inducing host-derived damage-associated molecular patterns (DAMPs). However, like most other DAMP-inducing adjuvants such as aluminum hydroxide (Alum), HP-β-CyD may not be sufficient for the induction of protective type-1 (cellular) immune responses, thereby leaving room for improvement. Here, we demonstrate that a combination of HP-β-CyD with a humanized TLR9 agonist, K3 CpG-ODN, a potent pathogen-associated molecular pattern (PAMP), enhanced the protective efficacy of the co-administered influenza split vaccine by inducing antigen-specific type-2 and type-1 immune responses, respectively. Moreover, substantial antigen-specific IgE induction by HP-β-CyD, which can cause an allergic response to immunized antigen was completely suppressed by the addition of K3 CpG-ODN. Furthermore, HP-β-CyD- and K3 CpG-ODN-adjuvanted influenza split vaccination protected the mice against lethal challenge with high doses of heterologous influenza virus, which could not be protected against by single adjuvant vaccines. Further experiments using gene deficient mice revealed the unique immunological mechanism of action in vivo, where type-2 and type-1 immune responses enhanced by the combined adjuvants were dependent on TBK1 and TLR9, respectively, indicating their parallel signaling pathways. Finally, the analysis of immune responses in the draining lymph node suggested that HP-β-CyD promotes the uptake of K3 CpG-ODN by plasmacytoid dendritic cells and B cells, which may contributes to the activation of these cells and enhanced production of IgG2c. Taken together, the results above may offer potential clinical applications for the combination of DAMP-inducing adjuvant and PAMP adjuvant to improve vaccine immunogenicity and efficacy by enhancing both type-2 and type-1 immune responses in a parallel manner.
Collapse
Affiliation(s)
- Tomoya Hayashi
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Masatoshi Momota
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Laboratory of Vaccine Science, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Etsushi Kuroda
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Laboratory of Vaccine Science, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Takato Kusakabe
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Laboratory of Vaccine Science, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Shingo Kobari
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Kotaro Makisaka
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoshitaka Ohno
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Program for Leading Graduate Schools “Health Life Science: Interdisciplinary and Global Oriented Program”, Kumamoto University, Kumamoto, Japan
| | - Yusuke Suzuki
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Fumika Nakagawa
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Michelle S. J. Lee
- Laboratory of Malaria Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Cevayir Coban
- Laboratory of Malaria Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Risako Onodera
- Building Regional Innovation Ecosystems, School of Pharmacy, Kumamoto University, Kumamoto, Japan
| | - Taishi Higashi
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keiichi Motoyama
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Ken J. Ishii
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institute of Biomedical Innovation, Health and Nutrition, Osaka, Japan
- Laboratory of Vaccine Science, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hidetoshi Arima
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Program for Leading Graduate Schools “Health Life Science: Interdisciplinary and Global Oriented Program”, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
16
|
Lin YL, Chow YH, Huang LM, Hsieh SM, Cheng PY, Hu KC, Chiang BL. A CpG-adjuvanted intranasal enterovirus 71 vaccine elicits mucosal and systemic immune responses and protects human SCARB2-transgenic mice against lethal challenge. Sci Rep 2018; 8:10713. [PMID: 30013088 PMCID: PMC6048030 DOI: 10.1038/s41598-018-28281-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 05/15/2018] [Indexed: 12/11/2022] Open
Abstract
Enterovirus 71 (EV71) is an aetiological agent responsible for seasonal epidemics of hand-foot-and-mouth disease, which causes considerable mortality among young children. Mucosal vaccines can efficiently induce secretory IgA at mucosal surfaces and thereby prevent or limit infection at the site of virus entry. CpG oligodeoxynucleotides (ODNs), which resemble bacterial DNA, can induce the innate immune response through activation of Toll-like receptor 9. Here, we used CpG ODNs as adjuvants to investigate an EV71 mucosal vaccine in mice. In the EV71 + CpG group, the EV71-specific IgG and IgA titres in the serum, nasal wash, bronchoalveolar lavage fluid, and faeces were substantially higher than those in the EV71- and phosphate-buffered saline-treated groups. Moreover, the number of EV71-specific IgG- and IgA-producing cells was also higher in the EV71 + CpG group. Furthermore, T-cell proliferative responses and interleukin-17 secretion were markedly increased when CpG-adjuvanted EV71 was delivered intranasally. More importantly, the induced antibodies neutralised infection by EV71 of the C2 genotype and crossneutralised infection by EV71 of the B4 and B5 genotypes. Lastly, human scavenger receptor class B, member 2-transgenic mice intranasally immunised with the CpG-adjuvanted EV71 vaccine resisted a subsequent lethal challenge with EV71, indicating that CpG was an effective intranasal adjuvant for EV71 mucosal-vaccine development.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Bronchoalveolar Lavage Fluid/immunology
- Disease Models, Animal
- Enterovirus A, Human/immunology
- Enterovirus A, Human/pathogenicity
- Female
- Hand, Foot and Mouth Disease/blood
- Hand, Foot and Mouth Disease/immunology
- Hand, Foot and Mouth Disease/prevention & control
- Hand, Foot and Mouth Disease/virology
- Humans
- Immunity, Mucosal
- Immunogenicity, Vaccine
- Lysosomal Membrane Proteins/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Receptors, Scavenger/genetics
- Treatment Outcome
- Vaccines, Inactivated
- Viral Vaccines/administration & dosage
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Yu-Li Lin
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Hung Chow
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Li-Min Huang
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan
| | - Szu-Min Hsieh
- Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Pei-Yun Cheng
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Kai-Chieh Hu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Bor-Luen Chiang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
17
|
Takaki H, Ichimiya S, Matsumoto M, Seya T. Mucosal Immune Response in Nasal-Associated Lymphoid Tissue upon Intranasal Administration by Adjuvants. J Innate Immun 2018; 10:515-521. [PMID: 29860261 DOI: 10.1159/000489405] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 04/12/2018] [Indexed: 12/23/2022] Open
Abstract
The nasal administration of vaccines directed against diseases caused by upper respiratory tract infections of pathogens, such as the influenza virus, mimics the natural infection of pathogens and induces immunoglobulin A (IgA) production in the nasal cavity to effectively protect viral entry. Therefore, the development of a nasally administered vaccine is a research objective. Because the antigenicity of influenza split vaccines is low, nasal inoculation with the vaccine alone does not induce strong IgA production in the nasal cavity. However, the addition of adjuvants activates the innate immune response, enhancing antigen-specific IgA production and the T-cell response. Although the development of suitable adjuvants for nasal vaccinations is in progress, the mechanism by which adjuvants promote the immune response is still unclear. In this review, we discuss the mucosal immune response, especially in the nasal-associated lymphoid tissue, induced in response to the intranasal inoculation of an influenza vaccine and adjuvants in animal models.
Collapse
Affiliation(s)
- Hiromi Takaki
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, .,Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo,
| | - Shingo Ichimiya
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Misako Matsumoto
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tsukasa Seya
- Department of Vaccine Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
18
|
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: 2.0] [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
|
19
|
Kataoka K, Fukuyama Y, Briles DE, Miyake T, Fujihashi K. Dendritic cell-targeting DNA-based nasal adjuvants for protective mucosal immunity to Streptococcus pneumoniae. Microbiol Immunol 2017; 61:195-205. [PMID: 28463465 DOI: 10.1111/1348-0421.12487] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/15/2017] [Accepted: 04/25/2017] [Indexed: 12/27/2022]
Abstract
To develop safe vaccines for inducing mucosal immunity to major pulmonary bacterial infections, appropriate vaccine antigens (Ags), delivery systems and nontoxic molecular adjuvants must be considered. Such vaccine constructs can induce Ag-specific immune responses that protect against mucosal infections. In particular, it has been shown that simply mixing the adjuvant with the bacterial Ag is a relatively easy means of constructing adjuvant-based mucosal vaccine preparations; the resulting vaccines can elicit protective immunity. DNA-based nasal adjuvants targeting mucosal DCs have been studied in order to induce Ag-specific mucosal and systemic immune responses that provide essential protection against microbial pathogens that invade mucosal surfaces. In this review, initially a plasmid encoding the cDNA of Flt3 ligand (pFL), a molecule that is a growth factor for DCs, as an effective adjuvant for mucosal immunity to pneumococcal infections, is introduced. Next, the potential of adding unmethylated CpG oligodeoxynucleotide and pFL together with a pneumococcal Ag to induce protection from pneumococcal infections is discussed. Pneumococcal surface protein A has been used as vaccine for restoring mucosal immunity in older persons. Further, our nasal pFL adjuvant system with phosphorylcholine-keyhole limpet hemocyanin (PC-KLH) has also been used in pneumococcal vaccine development to induce complete protection from nasal carriage by Streptococcus pneumoniae. Finally, the possibility that anti-PC antibodies induced by nasal delivery of pFL plus PC-KLH may play a protective role in prevention of atherogenesis and thus block subsequent development of cardiovascular disease is discussed.
Collapse
Affiliation(s)
- Kosuke Kataoka
- Department of Preventive and Community Dentistry, Osaka Dental University, Hirakata, Osaka 573-1121, Japan
| | - Yoshiko Fukuyama
- Departments of Pediatric Dentistry and Microbiology, Immunobiology Vaccine Center, Institute for Oral Health Research, University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | - David E Briles
- Departments of Pediatric Dentistry and Microbiology, Immunobiology Vaccine Center, Institute for Oral Health Research, University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | - Tatsuro Miyake
- Department of Preventive and Community Dentistry, Osaka Dental University, Hirakata, Osaka 573-1121, Japan
| | - Kohtaro Fujihashi
- Departments of Pediatric Dentistry and Microbiology, Immunobiology Vaccine Center, Institute for Oral Health Research, University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| |
Collapse
|
20
|
Su H, Liao Z, Yuan G, Su J. A plasmid containing CpG ODN as vaccine adjuvant against grass carp reovirus in grass carp Ctenopharyngodon idella. Oncotarget 2017; 8:86576-86591. [PMID: 29156818 PMCID: PMC5689708 DOI: 10.18632/oncotarget.21245] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/17/2017] [Indexed: 02/07/2023] Open
Abstract
CpG oligodeoxynucleotides (ODNs) are proved to have strong immune stimulatory activity. Plasmids containing CpG ODNs could be conveniently and low-costly used as vaccine adjuvant. However, they are different among various plasmids, motif repeats, species, etc. In the present study, plasmid pcDNA3.1 (+) containing five repetitions of CpG ODN 1670A named pcDNA3.1-1670A*5 with strong immunostimulation was screened out from twelve recombinant plasmids and three empty vectors by cell proliferation activity, interferon promoter activities and immune related gene expressions in CIK cells. It works through TLR9-mediated signaling pathway, triggering the immune related genes expression. Furthermore, the potentiality of pcDNA3.1-1670A*5 as adjuvant was tested in vivo. pcDNA3.1-1670A*5 was co-inoculated with inactivated GCRV vaccine on grass carp fingerlings. Immunoglobulins (IgM, IgD, IgZ), TLR9, IFNγ2, IFN1, TNF-α, Mx2 and VP4 were examined. Ultimately, pcDNA3.1-1670A*5 significantly enhanced the expressions of IgM in serum, head kidney and spleen, recognition receptor TLR9 as well as antiviral effector molecule Mx2, and inhibited GCRV proliferation in head kidney and spleen tissues. The present study explored the application and mechanism of plasmid containing CpG ODN as high-efficient adjuvant to promote efficiency of vaccine and control disease in grass carp, which will contribute to the development of new type CpG ODN adjuvant in aquaculture industry.
Collapse
Affiliation(s)
- Hang Su
- College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| | - Zhiwei Liao
- College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| | - Gailing Yuan
- College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China
| | - Jianguo Su
- College of Fisheries, Huazhong Agricultural University, Wuhan, China.,Hubei Engineering Technology Research Center for Aquatic Animal Disease Control and Prevention, Wuhan, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
21
|
Pérez-Toledo M, Valero-Pacheco N, Pastelin-Palacios R, Gil-Cruz C, Perez-Shibayama C, Moreno-Eutimio MA, Becker I, Pérez-Tapia SM, Arriaga-Pizano L, Cunningham AF, Isibasi A, Bonifaz LC, López-Macías C. Salmonella Typhi Porins OmpC and OmpF Are Potent Adjuvants for T-Dependent and T-Independent Antigens. Front Immunol 2017; 8:230. [PMID: 28337196 PMCID: PMC5344031 DOI: 10.3389/fimmu.2017.00230] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 02/17/2017] [Indexed: 02/05/2023] Open
Abstract
Several microbial components, such as bacterial DNA and flagellin, have been used as experimental vaccine adjuvants because of their inherent capacity to efficiently activate innate immune responses. Likewise, our previous work has shown that the major Salmonella Typhi (S. Typhi) outer membrane proteins OmpC and OmpF (porins) are highly immunogenic protective antigens that efficiently stimulate innate and adaptive immune responses in the absence of exogenous adjuvants. Moreover, S. Typhi porins induce the expression of costimulatory molecules on antigen-presenting cells through toll-like receptor canonical signaling pathways. However, the potential of major S. Typhi porins to be used as vaccine adjuvants remains unknown. Here, we evaluated the adjuvant properties of S. Typhi porins against a range of experimental and clinically relevant antigens. Co-immunization of S. Typhi porins with ovalbumin (OVA), an otherwise poorly immunogenic antigen, enhanced anti-OVA IgG titers, antibody class switching, and affinity maturation. This adjuvant effect was dependent on CD4+ T-cell cooperation and was associated with an increase in IFN-γ, IL-17A, and IL-2 production by OVA-specific CD4+ T cells. Furthermore, co-immunization of S. Typhi porins with an inactivated H1N1 2009 pandemic influenza virus experimental vaccine elicited higher hemagglutinating anti-influenza IgG titers, antibody class switching, and affinity maturation. Unexpectedly, co-administration of S. Typhi porins with purified, unconjugated Vi capsular polysaccharide vaccine (Vi CPS)—a T-independent antigen—induced higher IgG antibody titers and class switching. Together, our results suggest that S. Typhi porins OmpC and OmpF are versatile vaccine adjuvants, which could be used to enhance T-cell immune responses toward a Th1/Th17 profile, while improving antibody responses to otherwise poorly immunogenic T-dependent and T-independent antigens.
Collapse
Affiliation(s)
- Marisol Pérez-Toledo
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute, Mexico City, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Nuriban Valero-Pacheco
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute, Mexico City, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | | | - Cristina Gil-Cruz
- Institute of Immunobiology, Kantonsspital St. Gallen , St. Gallen , Switzerland
| | | | - Mario A Moreno-Eutimio
- Immunity and Inflammation Research Unit, Hospital Juárez de México, Ministry of Health , Mexico City , Mexico
| | - Ingeborg Becker
- Facultad de Medicina, Departamento de Medicina Experimental, Universidad Nacional Autónoma de México , Mexico City , Mexico
| | - Sonia Mayra Pérez-Tapia
- Unit of R&D in Bioprocesses (UDIBI), Department of Immunology, National School of Biological Sciences, National Polytechnic Institute , Mexico City , Mexico
| | - Lourdes Arriaga-Pizano
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute , Mexico City , Mexico
| | - Adam F Cunningham
- MRC Centre for Immune Regulation, College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham , Birmingham , UK
| | - Armando Isibasi
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute , Mexico City , Mexico
| | - Laura C Bonifaz
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute , Mexico City , Mexico
| | - Constantino López-Macías
- Medical Research Unit on Immunochemistry, Specialties Hospital, National Medical Centre "Siglo XXI", Mexican Social Security Institute, Mexico City, Mexico; Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
22
|
Adachi N, Takayama E, Adachi M, Mizuno-Kamiya M, Kawaki H, Takeuchi H, Kubo S, Ishigami H, Kurachi M, Kondoh N. Promotion of Nickel (Ni) Allergy by Anamnestic Sensitization with a Bacterial Component, Lipopolysaccharide (LPS), in Mice. Open Dent J 2016; 10:531-537. [PMID: 27843506 PMCID: PMC5070425 DOI: 10.2174/1874210601610010531] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 07/20/2016] [Accepted: 09/07/2016] [Indexed: 11/25/2022] Open
Abstract
Background/Objective: Lipopolysaccharides (LPS) promote allergic responses to nickel (Ni) both in the sensitization and elicitation steps. In this study, we examine the effect of pre-sensitization to LPS on the occurrence of Ni allergy using a mouse model. Method: A 100 mg of LPS was injected into C57BL/6J mice intraperitoneally (ip). Three weeks later, the mice were subsequently injected with 0.3 μ moles of nickel dichloride (NiCl2) and 100 μg of CpG-DNA, which acted as an adjuvant. The mice were repeatedly immunized with the 0.3 μg of nickel sulfate (NiSO4), along with 300 μl of the adjuvant, Inject Alum (Pierce, USA). Then we examined the producing capabilities of T helper type 1 (Th1) and 2 (Th2) cytokines (interferon-gamma- (IFN)-γ and interleukin (IL)-10, respectively) from anti CD3 antibody-stimulated spleen cells. Results: Pre-treatment with LPS, followed by repeated challenges with Ni2+ and adjuvants significantly enhanced the IFN-γ-producing capability of spleen cells (n=5, p<0.01); however, that could not enhance the capability of spleen cells by a single challenge with Ni2+ and adjuvants (n=5). In contrast, without LPS treatment, single or even repeated challenges by Ni2+ could not enhance the IFN-γ-producing capability. On the other hand, the IL-10-producing capability of spleen cells was not enhanced even by LPS and repeated challenges with Ni2+ and adjuvants. Conclusion: The solitary pre-sensitization to LPS is essential for the onset of Ni allergy by shifting the Th1/Th2 immune balance toward a Th1 dominant.
Collapse
Affiliation(s)
- Norimasa Adachi
- Department of Oral Biochemistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan; Department of Prosthodontics, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Eiji Takayama
- Department of Oral Biochemistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Makoto Adachi
- Department of Oral and Maxillofacial Surgery, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Masako Mizuno-Kamiya
- Department of Oral Biochemistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Harumi Kawaki
- Department of Oral Biochemistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Hiroko Takeuchi
- Department of Oral Biochemistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan; Department of Periodontology, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Shuri Kubo
- Department of Oral Biochemistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan; Department of Periodontology, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Hajime Ishigami
- Department of Prosthodontics, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Masakazu Kurachi
- Department of Prosthodontics, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| | - Nobuo Kondoh
- Department of Oral Biochemistry, Asahi University School of Dentistry, 1851 Hozumi, Mizuho, Gifu, 501-0296, Japan
| |
Collapse
|
23
|
Cheng WK, Plumb AW, Lai JCY, Abraham N, Dutz JP. Topical CpG Oligodeoxynucleotide Adjuvant Enhances the Adaptive Immune Response against Influenza A Infections. Front Immunol 2016; 7:284. [PMID: 27524984 PMCID: PMC4965457 DOI: 10.3389/fimmu.2016.00284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/13/2016] [Indexed: 11/26/2022] Open
Abstract
Current influenza vaccines generate humoral immunity, targeting highly variable epitopes and thus fail to achieve long-term protection. T cells recognize and respond to several highly conserved epitopes across influenza serotypes. A strategy of raising strong cytotoxic T cell memory responses to epitopes conserved across serotypes would provide cross serotype protection, eliminating the need for annual vaccination. We explored the adjuvant potential of epicutaneous (ec) and subcutaneous (sc) delivery of CpG oligodeoxynucleotide in conjunction with sc protein immunization to improve protection against influenza A virus (IAV) infections using a mouse model. We found enhanced long-term protection with epicutaneous CpG ODN (ecCpG) compared to subcutaneous CpG ODN (scCpG) as demonstrated by reduced viral titers in the lungs. This correlated with increased antigen-specific CD8 T cells in the airways and the lungs. The memory T cell response after immunization with ecCpG adjuvant was comparable to memory response by priming with IAV infection in the lungs. In addition, ecCpG was more efficient than scCpG in inducing the generation of IFN-γ producing CD4 T cells. The adjuvant effect of ecCpG was accompanied with its ability to modulate tissue-homing molecules on T cells that may direct them to the site of infection. Together, this work provides evidence for using ecCpG to induce strong antibody and memory T cell responses to confer protection against IAV infection.
Collapse
Affiliation(s)
- Wing Ki Cheng
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia , Vancouver, BC , Canada
| | - Adam William Plumb
- Department of Microbiology and Immunology, Faculty of Science, Life Sciences Institute, The University of British Columbia , Vancouver, BC , Canada
| | - Jacqueline Cheuk-Yan Lai
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia , Vancouver, BC , Canada
| | - Ninan Abraham
- Department of Microbiology and Immunology, Faculty of Science, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada; Department of Zoology, Faculty of Science, Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada
| | - Jan Peter Dutz
- Department of Dermatology and Skin Science, Faculty of Medicine, Child and Family Research Institute, The University of British Columbia , Vancouver, BC , Canada
| |
Collapse
|
24
|
A critical role of T follicular helper cells in human mucosal anti-influenza response that can be enhanced by immunological adjuvant CpG-DNA. Antiviral Res 2016; 132:122-30. [PMID: 27247060 DOI: 10.1016/j.antiviral.2016.05.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/23/2016] [Accepted: 05/26/2016] [Indexed: 01/22/2023]
Abstract
T Follicular helper cells (TFH) are considered critical for B cell antibody response, and recent efforts have focused on promoting TFH in order to enhance vaccine efficacy. We studied the frequency and function of TFH in nasopharynx-associated lymphoid tissues (NALT) from children and adults, and its role in anti-influenza antibody response following stimulation by a live-attenuated influenza vaccine (LAIV) or an inactivated seasonal virus antigen (sH1N1). We further studied whether CpG-DNA promotes TFH and by which enhances anti-influenza response. We showed NALT from children aged 1.5-10 years contained abundant TFH, suggesting efficient priming of TFH during early childhood. Stimulation by LAIV induced a marked increase in TFH that correlated with a strong production of anti-hemagglutinin (HA) IgA/IgG/IgM antibodies in tonsillar cells. Stimulation by the inactivated sH1N1 antigen induced a small increase in TFH which was markedly enhanced by CpG-DNA, accompanied by enhanced anti-HA antibody responses. In B cell co-culture experiment, anti-HA responses were only seen in the presence of TFH, and addition of plasmacytoid dendritic cell to TFH-B cell co-culture enhanced the TFH-mediated antibody production following CpG-DNA and sH1N1 antigen stimulation. Induction of TFH differentiation from naïve T cells was also shown following the stimulation. Our results support a critical role of TFH in human mucosal anti-influenza antibody response. Use of an adjuvant such as CpG-DNA that has the capacity to promote TFH by which to enhance antigen-induced antibody responses in NALT tissue may have important implications for future vaccination strategies against respiratory pathogens.
Collapse
|
25
|
Tomar J, Born PA, Frijlink HW, Hinrichs WLJ. Dry influenza vaccines: towards a stable, effective and convenient alternative to conventional parenteral influenza vaccination. Expert Rev Vaccines 2016; 15:1431-1447. [DOI: 10.1080/14760584.2016.1182869] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
26
|
Vogel AJ, Brown DM. Single-Dose CpG Immunization Protects Against a Heterosubtypic Challenge and Generates Antigen-Specific Memory T Cells. Front Immunol 2015; 6:327. [PMID: 26161083 PMCID: PMC4479795 DOI: 10.3389/fimmu.2015.00327] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/11/2015] [Indexed: 01/08/2023] Open
Abstract
Despite extensive research, influenza A virus (IAV) remains a major cause of morbidity, mortality, and healthcare expenditure. Emerging pandemics from highly pathogenic IAV strains, such as H5N1 and pandemic H1N1, highlight the need for universal, cross-protective vaccines. Current vaccine formulations generate strain-specific neutralizing antibodies primarily against the outer coat proteins, hemagglutinin and neuraminidase. In contrast to these highly mutable proteins, internal proteins of IAV are more conserved and are a favorable target for developing vaccines that induce strong T cell responses in addition to humoral immunity. Here, we found that intranasal administration with a single dose of CpG and inactivated x31 (H3N2) reduced viral titers and partially protected mice from a heterosubtypic challenge with a lethal dose of PR8 (H1N1). Early after immunization, vaccinated mice showed increased innate immune activation with high levels of MHCII and CD86 expression on dendritic cells in both draining lymph nodes and lungs. Three days after immunization, CD4 and CD8 cells in the lung upregulated CD69, suggesting that activated lymphocytes are present at the site of vaccine administration. The ensuing effector Th1 responses were capable of producing multiple cytokines and were present at least 30 days after immunization. Furthermore, functional memory responses were observed, as antigen-specific IFN-γ+ and GrB+ cells were detected early after lethal infection. Together, this work provides evidence for using pattern recognition receptor agonists as a mucosal vaccine platform for inducing robust T cell responses capable of protecting against heterologous IAV challenges.
Collapse
Affiliation(s)
- Alexander J Vogel
- School of Biological Sciences, University of Nebraska-Lincoln , Lincoln, NE , USA ; Nebraska Center for Virology, University of Nebraska-Lincoln , Lincoln, NE , USA
| | - Deborah M Brown
- School of Biological Sciences, University of Nebraska-Lincoln , Lincoln, NE , USA ; Nebraska Center for Virology, University of Nebraska-Lincoln , Lincoln, NE , USA
| |
Collapse
|
27
|
Sun W, Li Q, Zhu D, Feng J, Zhuang Z, Sun X, Xiao G, Duan Y. Enhancement of immune response to a hepatitis C virus E2 DNA vaccine by an immunoglobulin Fc fusion tag. J Med Virol 2015; 87:2090-7. [PMID: 26010499 DOI: 10.1002/jmv.24277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2015] [Indexed: 11/11/2022]
Abstract
Neutralizing antibodies and cellular immune response both play essential roles in the clearance of Hepatitis C virus (HCV) infection. The envelope glycoprotein E2 is a major target for producing neutralizing antibodies against HCV. Here, we constructed a recombinant plasmid, termed pcDNA3.1-E2-Fc, to express HCV E2 with an immunoglobulin Fc fusion tag (E2-Fc). Importantly, we found that the titers of E2-specific IgG from mice immunized with pcDNA3.1-E2-Fc were significantly higher than that from mice immunized with pcDNA3.1-E2. Moreover, pcDNA3.1-E2-Fc immunization could boost E2-specific lymphocyte proliferation and enhance the secretion of IFN-γ by lymphocytes upon in vitro stimulation with soluble E2 compared to pcDNA3.1-E2 immunization. Neutralization assays showed that serum from pcDNA3.1-E2-Fc immunized mice exhibited more effective neutralizing capacity of HCVpp entry into Huh-7 cells compared with that from pcDNA3.1-E2 immunized mice, although both of the sera could inhibit the virus entry. Taken together, our results imply that pcDNA3.1-E2-Fc immunization could enhance E2-specific humoral and cellular immune response in mice and thus provide a promising candidate for the development of an HCV vaccine.
Collapse
Affiliation(s)
- Wei Sun
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Qun Li
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Dandan Zhu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jinrong Feng
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Zhong Zhuang
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Xiaolei Sun
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Gengfu Xiao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Yinong Duan
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
28
|
Comparison of adjuvants for a spray freeze-dried whole inactivated virus influenza vaccine for pulmonary administration. Eur J Pharm Biopharm 2015; 93:231-41. [PMID: 25896446 DOI: 10.1016/j.ejpb.2015.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 03/25/2015] [Accepted: 04/09/2015] [Indexed: 02/02/2023]
Abstract
Stable vaccines administered to the lungs by inhalation could circumvent many of the problems associated with current immunizations against respiratory infections. We earlier provided proof of concept in mice that pulmonary delivered whole inactivated virus (WIV) influenza vaccine formulated as a stable dry powder effectively elicits influenza-specific antibodies in lung and serum. Yet, mucosal IgA, considered particularly important for protection at the site of virus entry, was poorly induced. Here we investigate the suitability of various Toll-like receptor (TLR) ligands and the saponin-derived compound GPI-0100 to serve as adjuvant for influenza vaccine administered to the lungs as dry powder. The TLR ligands palmitoyl-3-cysteine-serine-lysine-4 (Pam3CSK4), monophosphoryl lipid A (MPLA) and CpG oligodeoxynucleotides (CpG ODN) as well as GPI-0100 tolerated the process of spray freeze-drying well. While Pam3CSK4 had no effect on systemic antibody titers, all the other adjuvants significantly increased influenza-specific serum and lung IgG titers. Yet, only GPI-0100 also enhanced mucosal IgA titers. Moreover, only GPI-0100-adjuvanted WIV provided partial protection against heterologous virus challenge. Pulmonary immunization with GPI-0100-adjuvanted vaccine did not induce an overt inflammatory response since influx of neutrophils and production of inflammatory cytokines were moderate and transient and lung histology was normal. Our results indicate that a GPI-0100-adjuvanted dry powder influenza vaccine is a safe and effective alternative to current parenteral vaccines.
Collapse
|
29
|
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: 4.5] [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
|
30
|
Murugappan S, Frijlink HW, Petrovsky N, Hinrichs WLJ. Enhanced pulmonary immunization with aerosolized inactivated influenza vaccine containing delta inulin adjuvant. Eur J Pharm Sci 2014; 66:118-22. [PMID: 25459531 DOI: 10.1016/j.ejps.2014.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 09/04/2014] [Accepted: 10/10/2014] [Indexed: 01/05/2023]
Abstract
Vaccination is the primary intervention to contain influenza virus spread during seasonal and pandemic outbreaks. Pulmonary vaccination is gaining increasing attention for its ability to induce both local mucosal and systemic immune responses without the need for invasive injections. However, pulmonary administration of whole inactivated influenza virus (WIV) vaccine induces a Th2 dominant systemic immune response while a more balanced Th1/Th2 vaccine response may be preferred and only induces modest nasal immunity. This study evaluated immunity elicited by pulmonary versus intramuscular (i.m.) delivery of WIV, and tested whether the immune response could be improved by co-administration of delta (δ)-inulin, a novel carbohydrate-based particulate adjuvant. After pulmonary administration both unadjuvanted and δ-inulin adjuvanted WIV induced a potent systemic immune response, inducing higher serum anti-influenza IgG titers and nasal IgA titers than i.m. administration. Moreover, the addition of δ-inulin induced a more balanced Th1/Th2 response and induced higher nasal IgA titers versus pulmonary WIV alone. Pulmonary WIV alone or with δ-inulin induced hemagglutination inhibition (HI) titers>40, titers which are considered protective against influenza virus. In conclusion, in this study we have shown that δ-inulin adjuvanted WIV induces a better immune response after pulmonary administration than vaccine alone.
Collapse
Affiliation(s)
- Senthil Murugappan
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Flinders Medical Centre, Bedford Park, Adelaide 5042, Australia; Department of Diabetes and Endocrinology, Flinders Medical Centre/Flinders University, Adelaide 5042, Australia
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
31
|
Dehghan S, Tafaghodi M, Bolourieh T, Mazaheri V, Torabi A, Abnous K, Tavassoti Kheiri M. Rabbit nasal immunization against influenza by dry-powder form of chitosan nanospheres encapsulated with influenza whole virus and adjuvants. Int J Pharm 2014; 475:1-8. [PMID: 25148732 DOI: 10.1016/j.ijpharm.2014.08.032] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 08/14/2014] [Accepted: 08/17/2014] [Indexed: 11/26/2022]
Abstract
Influenza virus is one of the main causes of respiratory diseases in human. Although different vaccines have been produced during past decades, there is still a huge demand for a safe influenza vaccine with the ability to induce mucosal immune responses and sufficient protection, especially in elderly patients. In this study, chitosan nanospheres were employed as the drug delivery system. Influenza virus, CpG oligodeoxynucleotide (CpG ODN) and Quillaja saponins (QS) were incorporated in this nanospheric system. Three doses of dry powder nanosphere vaccine were nasally administered to rabbits on days 0, 45 and 60, followed by a final booster injection on day 75. Both humoral and cellular immune responses were investigated. Hemagglutination inhibition (HI) antibody titer was elevated in all groups compared to the control group at the end of vaccination in rabbits receiving nanospheres loaded with virus and CpG, CH(WV+CpG) (P<0.001). Rabbit serum IgG raised significantly in all the vaccinated groups, with the highest responses in CH(WV+CpG) group. CH(WV+CpG) and CH(WV) induced significant sIgA titers (P<0.001). CpG adjuvant also showed a prominent role in the stimulation and secretion of of IL-2 and IFN-γ cytokines (3 and 3.5 fold increase, respectively). Finally, as CH(WV+CpG) depicted to be effective in induction of humoral and cellular immune responses after nasal administration, this nanoparticulate adjuvant could be identified as an efficient adjuvant/delivery system for mucosal immunization against influenza virus.
Collapse
Affiliation(s)
- Solmaz Dehghan
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Influenza Research Lab, Pasteur Institute of Iran, No. 358, 12th Farvardin Street, Jomhoori Avenue, Tehran 13169-43551, Iran
| | - Mohsen Tafaghodi
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tina Bolourieh
- Influenza Research Lab, Pasteur Institute of Iran, No. 358, 12th Farvardin Street, Jomhoori Avenue, Tehran 13169-43551, Iran
| | - Vahideh Mazaheri
- Influenza Research Lab, Pasteur Institute of Iran, No. 358, 12th Farvardin Street, Jomhoori Avenue, Tehran 13169-43551, Iran
| | - Ali Torabi
- Influenza Research Lab, Pasteur Institute of Iran, No. 358, 12th Farvardin Street, Jomhoori Avenue, Tehran 13169-43551, Iran
| | - Khalil Abnous
- Pharmaceutical Sciences Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoumeh Tavassoti Kheiri
- Influenza Research Lab, Pasteur Institute of Iran, No. 358, 12th Farvardin Street, Jomhoori Avenue, Tehran 13169-43551, Iran.
| |
Collapse
|
32
|
Scheiermann J, Klinman DM. Clinical evaluation of CpG oligonucleotides as adjuvants for vaccines targeting infectious diseases and cancer. Vaccine 2014; 32:6377-89. [PMID: 24975812 DOI: 10.1016/j.vaccine.2014.06.065] [Citation(s) in RCA: 250] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 05/28/2014] [Accepted: 06/12/2014] [Indexed: 12/13/2022]
Abstract
Synthetic oligonucleotides (ODN) that express unmethylated "CpG motifs" trigger cells that express Toll-like receptor 9. In humans this includes plasmacytoid dendritic cells and B cells. CpG ODN induce an innate immune response characterized by the production of Th1 and pro-inflammatory cytokines. Their utility as vaccine adjuvants was evaluated in a number of clinical trials. Results indicate that CpG ODN improve antigen presentation and the generation of vaccine-specific cellular and humoral responses. This work provides an up-to-date overview of the utility of CpG ODN as adjuvants for vaccines targeting infectious agents and cancer.
Collapse
Affiliation(s)
- Julia Scheiermann
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick MD 21702, United States
| | - Dennis M Klinman
- Cancer and Inflammation Program, National Cancer Institute, NIH, Frederick MD 21702, United States.
| |
Collapse
|
33
|
Manuja A, Manuja BK, Kaushik J, Singha H, Singh RK. Immunotherapeutic potential of CpG oligodeoxynucleotides in veterinary species. Immunopharmacol Immunotoxicol 2013; 35:535-44. [PMID: 23981003 DOI: 10.3109/08923973.2013.828743] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Innate immunity plays a critical role in host defense against infectious diseases by discriminating between self and infectious non-self. The recognition of infectious non-self involves germ-line encoded pattern recognition receptors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs). The PAMPs are the components of pathogenic microbes which include not only the cell wall constituents but also the unmethylated 2'-deoxy-ribo-cytosine-phosphate-guanosine (CpG) motifs. These CpG motifs present within bacterial and viral DNA are recognized by toll-like receptor 9 (TLR9), and signaling by this receptor triggers a proinflammatory cytokine response which, in turn, influences both innate and adaptive immune responses. The activation of TLR9 with synthetic CpG oligodeoxynucleotides (ODNs) induces powerful Th1-like immune responses. It has been shown to provide protection against infectious diseases, allergy and cancer in laboratory animal models and some domestic animal species. With better understanding of the basic biology and immune mechanisms, it would be possible to exploit the potential of CpG motifs for animal welfare. The research developments in the area of CpG and TLR9 and the potential applications in animal health have been reviewed in this article.
Collapse
Affiliation(s)
- Anju Manuja
- Department of Veterinary Medicine, National Research Centre on Equines, Hisar , Haryana , India
| | | | | | | | | |
Collapse
|
34
|
A novel influenza virus hemagglutinin-respiratory syncytial virus (RSV) fusion protein subunit vaccine against influenza and RSV. J Virol 2013; 87:10792-804. [PMID: 23903841 DOI: 10.1128/jvi.01724-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Influenza A virus and respiratory syncytial virus (RSV) cause substantial morbidity and mortality afflicting the ends of the age spectrum during the autumn through winter months in the United States. The benefit of vaccination against RSV and influenza using a subunit vaccine to enhance immunity and neutralizing antibody was investigated. Influenza virus hemagglutinin (HA) and RSV fusion (F) protein were tested as vaccine components alone and in combination to explore the adjuvant properties of RSV F protein on HA immunity. Mice vaccinated with HA and F exhibited robust immunity that, when challenged, had reduced viral burden for both influenza and RSV. These studies show an enhancing and cross-protective benefit of F protein for anti-HA immunity.
Collapse
|
35
|
Asokanathan C, Corbel M, Xing D. A CpG-containing oligodeoxynucleotide adjuvant for acellular pertussis vaccine improves the protective response against Bordetella pertussis. Hum Vaccin Immunother 2013; 9:325-31. [PMID: 23291942 PMCID: PMC3859755 DOI: 10.4161/hv.22755] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 10/26/2012] [Accepted: 11/02/2012] [Indexed: 11/19/2022] Open
Abstract
We investigated the adjuvant effect of CpG ODN alone or in combination with aluminum hydroxide on the immune response to the three main antigens presented in current acellular pertussis vaccines: pertussis toxoid, filamentous haemagglutinin and pertactin. The development of protection in mice was investigated for the intra-peritoneal and intra-nasal immunisation routes. The results showed that CpG ODN alone, or in combination with aluminum hydroxide, gave enhancement in anti-pertussis toxin, anti- filamentous haemagglutinin and especially anti-pertactin titers after mucosal immunisation. Higher macrophage NO levels indicating activation were found when the antigens were co-formulated with CpG ODN. Vaccines containing CpG ODN gave enhanced humoral and CMI responses with a shift toward Th-1 and increased protection against challenge infection with B.pertussis in mice.
Collapse
Affiliation(s)
| | - Michael Corbel
- National Institute for Biological Standards and Control (NIBSC); South Mimms, Potters Bar, UK
| | - Dorothy Xing
- National Institute for Biological Standards and Control (NIBSC); South Mimms, Potters Bar, UK
| |
Collapse
|
36
|
|
37
|
CpG underrepresentation and the bacterial CpG-specific DNA methyltransferase M.MpeI. Proc Natl Acad Sci U S A 2012; 110:105-10. [PMID: 23248272 DOI: 10.1073/pnas.1207986110] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cytosine methylation promotes deamination. In eukaryotes, CpG methylation is thought to account for CpG underrepresentation. Whether scarcity of CpGs in prokaryotic genomes is diagnostic for methylation is not clear. Here, we report that Mycoplasms tend to be CpG depleted and to harbor a family of constitutively expressed or phase variable CpG-specific DNA methyltransferases. The very CpG poor Mycoplasma penetrans and its constitutively active CpG-specific methyltransferase M.MpeI were chosen for further characterization. Genome-wide sequencing of bisulfite-converted DNA indicated that M.MpeI methylated CpG target sites both in vivo and in vitro in a locus-nonselective manner. A crystal structure of M.MpeI with DNA at 2.15-Å resolution showed that the substrate base was flipped and that its place in the DNA stack was taken by a glutamine residue. A phenylalanine residue was intercalated into the "weak" CpG step of the nonsubstrate strand, indicating mechanistic similarities in the recognition of the short CpG target sequence by prokaryotic and eukaryotic DNA methyltransferases.
Collapse
|
38
|
Gong M, Zhou J, Yang C, Deng Y, Zhao G, Zhang Y, Wang Y, Zhou Y, Tan W, Xu H. Insect cell-expressed hemagglutinin with CpG oligodeoxynucleotides plus alum as an adjuvant is a potential pandemic influenza vaccine candidate. Vaccine 2012; 30:7498-505. [PMID: 23116697 DOI: 10.1016/j.vaccine.2012.10.054] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 08/15/2012] [Accepted: 10/16/2012] [Indexed: 11/15/2022]
Abstract
Vaccination is the most effective method used to reduce the morbidity and mortality of influenza infections. However, as exemplified in the current swine-origin influenza virus (S-OIV) pandemic, the global manufacturing capacity of influenza vaccines is severely limited. In the present proof-of-concept study, we combined cell substrate selection and antigen engineering with adjuvant development to design a potential pandemic influenza vaccine candidate, in which CpG oligodeoxynucleotides (CpG-ODN) plus alum was used as a composite adjuvant to enhance the immunogenicity of insect cell-expressed recombinant hemagglutinin (rHA). Our candidate vaccine was found to be effective in inducing protective humoral as well as cellular immunity in mice and able to protect the immunized mice from related influenza virus challenge. If this candidate vaccine is validated in humans, vaccine development can be started immediately after the release of the first HA sequence of any pandemic influenza virus. Moreover, given the potential of large-scale manufacturing capacity of the recombinant antigen, in combination with the antigen-sparing effect of the composite adjuvant, this technology could be an invaluable asset in the fight against pandemic influenza.
Collapse
Affiliation(s)
- Minqing Gong
- Department of Virology, National Vaccine and Serum Institute, Beijing 100024, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Dang Z, Feng J, Yagi K, Sugimoto C, Li W, Oku Y. Mucosal adjuvanticity of fibronectin-binding peptide (FBP) fused with Echinococcus multilocularis tetraspanin 3: systemic and local antibody responses. PLoS Negl Trop Dis 2012; 6:e1842. [PMID: 23029596 PMCID: PMC3459843 DOI: 10.1371/journal.pntd.0001842] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 08/15/2012] [Indexed: 12/24/2022] Open
Abstract
Background Studies have shown that a bacterial fibronectin attachment protein (FAP) is able to stimulate strong systemic and mucosal antibody responses when it is used alone or co-administrated with other antigens (Ags). Thus, it has been suggested to be a promising adjuvant candidate for the development of efficient vaccines. However, the co-administered Ags and FAP were cloned, expressed and purified individually to date. In a recent study, we first evaluated the adjuvanticity of a fibronectin-binding peptide (FBP, 24 amino acids) of Mycobacterium avium FAP fused with Echinococcus multilocularis tetraspanin 3 (Em-TSP3) by detecting systemic and local antibody responses in intranasally (i.n.) immunized BALB/c mice. Methodology/Principal Findings Em-TSP3 and FBP fragments were linked with a GSGGSG linker and expressed as a single fusion protein (Em-TSP3-FBP) using the pBAD/Thio-TOPO expression vector. BALB/c mice were immunized i.n. with recombinant Em-TSP3-FBP (rEm-TSP3-FBP) and rEm-TSP3+CpG and the systemic and local antibody responses were detected by ELISA. The results showed that both rEm-TSP3-FBP and rEm-TSP3+CpG evoked strong serum IgG (p<0.001) and IgG1 responses (p<0.001), whereas only the latter induced a high level IgG2α production (p<0.001), compared to that of rEm-TSP3 alone without any adjuvant. There were no significant differences in IgG and IgG1 production between the groups. Low level of serum IgA and IgM were detected in both groups. The tendency of Th1 and Th2 cell immune responses were assessed via detecting the IgG1/IgG2α ratio after the second and third immunizations. The results indicated that i.n. immunization with rEm-TSP3-FBP resulted in an increased IgG1/IgG2α ratio (a Th2 tendency), while rEm-TSP3+CpG caused a rapid Th1 response that later shifted to a Th2 response. Immunization with rEm-TSP3-FBP provoked significantly stronger IgA antibody responses in intestine (p<0.05), lung (p<0.001) and spleen (p<0.001) compared to those by rEm-TSP3+CpG. Significantly high level IgA antibodies were detected in nasal cavity (p<0.05) and liver (p<0.05) samples from both groups when compared to rEm-TSP3 alone without any adjuvant, with no significant difference between them. Conclusions I.n. administration of rEm-TSP3-FBP can induce strong systemic and mucosal antibody responses in immunized BALB/c mice, suggesting that fusion of Em-TSP3 with FBP is a novel, prospective strategy for developing safe and efficient human mucosal vaccines against alveolar echinococcosis (AE). Echinococcus metacestodes form a laminated layer and develop strategies to escape host immune responses once the infection established on the liver of intermediated host. One of the most important strategies is thought to be immunoregulation, where some molecules (e.g., antigen B) impair dendritic cell (DC) differentiation and polarize immature DC maturation towards a non-protective Th2 cell response. Therefore, it is more feasible to kill Echinococcus oncospheres in the early stage of infection in the intestine and blood. Systemic and local immune responses are believed to play a crucial role on oncosphere exclusion. Among antigen delivery systems, i.n. administration is the most efficient one, inducing both systemic and a full-range of mucosal immune responses. FAP is necessary to M. avium and S. pyogenes to efficiently attach and invade epithelial cells, and has been suggested as a potent vaccine adjuvant. Mucosal immune responses are induced after FAP binds to the fibronectin protein of host microfold (M) cells and DCs are activated. We developed a one-step delivery system where FAP and other Ags can be expressed, purified and immunized as one protein. The systemic and, in particular, the mucosal antibody responses induced by the fusion protein were detected to evaluate the adjuvanticity of FBP.
Collapse
Affiliation(s)
- Zhisheng Dang
- College of Life and Environmental Sciences, Minzu University of China, Beijing, People's Republic of China
| | | | | | | | | | | |
Collapse
|
40
|
Luo Z, Shi H, Zhang H, Li M, Zhao Y, Zhang J, Guo F, Luo S, Sun P, Zhang D, Qian Z, Yang L. Plasmid DNA containing multiple CpG motifs triggers a strong immune response to hepatitis B surface antigen when combined with incomplete Freund's adjuvant but not aluminum hydroxide. Mol Med Rep 2012; 6:1309-14. [PMID: 22971976 DOI: 10.3892/mmr.2012.1079] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 07/04/2012] [Indexed: 02/05/2023] Open
Abstract
Adjuvants are important components of recombinant protein vaccines which are often poorly immunogenic. For decades, the search for new vaccine adjuvants has been predominantly empirical. In addition, combinations of more than one adjuvant plus antigen have been systematically studied. Plasmid DNA containing additional oligodeoxynucleotides with unmethylated CpG motifs (CpG ODN) entrapped in liposomes has been used as an adjuvant for DNA vaccines and has shown powerful immunostimulatory functions. In our study, the combination of plasmid DNA containing 16 additional CpG ODNs (pv-16CpG) and aluminum hydroxide (AL) or incomplete Freund's adjuvant (IFA) was used as an adjuvant for a hepatitis B surface antigen (HBsAg) vaccine to immunize C57BL/6J mice. ELISA and ELISPOT assays were used to analyze the immunological effects of the novel vaccine. A significant enhancement of the anti-HBs titer and seroconversion was observed when the CpG plasmid was combined with IFA, but not with AL. In addition, anti-HBs antibody isotype analysis revealed that the combination of CpG plasmid and IFA induced a strong HBsAg-specific IgG2a response. Moreover, the ELISPOT assays suggested that pv-16CpG suspended in IFA evoked a strong T helper 1 (Th1) immune response and high IFN-γ production. These results demonstrate that pv-16CpG suspended in IFA is able to induce cellular and humoral immune responses to HBsAg, and confirm its potential as an adjuvant for use in protein vaccines.
Collapse
Affiliation(s)
- Zichao Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The vast majority of human pathogens colonize and invade at the mucosal surfaces. Preventing infection at these sites via mucosally active vaccines is a promising and rational approach for vaccine development. However, it is only recently that the stimulation of local immunity at the mucosal surfaces has become a primary objective in addition to inducing systemic immunity. This review describes vaccine formulations designed for mucosal delivery to the nasal-associated lymphoid tissue, via intranasal administration. The association of antigens with mucosal adjuvants and delivery systems is emphasised.
Collapse
Affiliation(s)
- Mehfuz Zaman
- School of Chemistry and Molecular Biosciences (SCMB), The University of Queensland, St. Lucia, 4072 QLD Australia
| | - Saranya Chandrudu
- School of Chemistry and Molecular Biosciences (SCMB), The University of Queensland, St. Lucia, 4072 QLD Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences (SCMB), The University of Queensland, St. Lucia, 4072 QLD Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD Australia
| |
Collapse
|
42
|
Fujkuyama Y, Tokuhara D, Kataoka K, Gilbert RS, McGhee JR, Yuki Y, Kiyono H, Fujihashi K. Novel vaccine development strategies for inducing mucosal immunity. Expert Rev Vaccines 2012; 11:367-79. [PMID: 22380827 DOI: 10.1586/erv.11.196] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To develop protective immune responses against mucosal pathogens, the delivery route and adjuvants for vaccination are important. The host, however, strives to maintain mucosal homeostasis by responding to mucosal antigens with tolerance, instead of immune activation. Thus, induction of mucosal immunity through vaccination is a rather difficult task, and potent mucosal adjuvants, vectors or other special delivery systems are often used, especially in the elderly. By taking advantage of the common mucosal immune system, the targeting of mucosal dendritic cells and microfold epithelial cells may facilitate the induction of effective mucosal immunity. Thus, novel routes of immunization and antigen delivery systems also show great potential for the development of effective and safe mucosal vaccines against various pathogens. The purpose of this review is to introduce several recent approaches to induce mucosal immunity to vaccines, with an emphasis on mucosal tissue targeting, new immunization routes and delivery systems. Defining the mechanisms of mucosal vaccines is as important as their efficacy and safety, and in this article, examples of recent approaches, which will likely accelerate progress in mucosal vaccine development, are discussed.
Collapse
Affiliation(s)
- Yoshiko Fujkuyama
- Departments of Pediatric Dentistry and Microbiology, The Immunobiology Vaccine Center, The University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Raman VS, Duthie MS, Fox CB, Matlashewski G, Reed SG. Adjuvants for Leishmania vaccines: from models to clinical application. Front Immunol 2012; 3:144. [PMID: 22701453 PMCID: PMC3371596 DOI: 10.3389/fimmu.2012.00144] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/18/2012] [Indexed: 12/12/2022] Open
Abstract
Two million new cases of leishmaniasis occur every year, with the cutaneous leishmaniasis (CL) presentation accounting for approximately two-thirds of all cases. Despite the high incidence rates and geographic expansion of the disease, CL remains a neglected tropical disease without effective intervention strategies. Efforts to address this deficit have given rise to the experimental murine model of CL. By virtue of its simplicity and pliability, the CL model has been used to provide substantial information regarding cellular immunity, as well as in the discovery and evaluation of various vaccine adjuvants. The CL model has facilitated in vivo studies of the mechanism of action of many adjuvants, including the TLR4 agonist monophosphoryl lipid A, the TLR7/8 agonist imiquimod, the TLR9 agonist CpG, adenoviral vectors, and the immunostimulatory complexes. Together, these studies have helped to unveil the requirement for certain types of immune responses at specific stages of CL disease and provide a basis to aid the design of effective second-generation vaccines for human CL. This review focuses on adjuvants that have been tested in experimental CL, outlining how they have helped advance our understanding of the disease and ultimately, how they have performed when applied within clinical trials against human CL.
Collapse
Affiliation(s)
- Vanitha S Raman
- Pre-clinical Biology, Infectious Disease Research Institute, Seattle, WA, USA
| | | | | | | | | |
Collapse
|
44
|
Asanuma H, Zamri NB, Sekine SI, Fukuyama Y, Tokuhara D, Gilbert RS, Fukuiwa T, Sata T, Tashiro M, Fujihashi K. A novel combined adjuvant for nasal delivery elicits mucosal immunity to influenza in aging. Vaccine 2012; 30:803-12. [PMID: 22100889 PMCID: PMC3253905 DOI: 10.1016/j.vaccine.2011.10.093] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 10/24/2011] [Accepted: 10/30/2011] [Indexed: 10/15/2022]
Abstract
Since a combination of flt3 ligand plasmid (pFL) and CpG-oligodeoxynucleotides (ODN)(3) as a dendritic cell (DC)-targeting double mucosal adjuvant elicited ovalbumin-specific secretory IgA (S-IgA) antibody (Ab) responses, we examined whether this double adjuvant could induce influenza-specific protective immunity in aged mice. A double adjuvant plus A/Puerto Rico/8/34 (PR8) hemagglutinin (HA) induced increased numbers of CD11b(+) CD11c(+) DCs and both CD4(+) Th1- and Th2-type responses in the nasopharyngeal-associated lymphoreticular tissue, nasal passages and cervical lymph nodes. Further, increased levels of PR8 HA-specific S-IgA Ab responses were detected in the upper respiratory tact (URT) of aged and young adult mice given nasal PR8 HA with this double adjuvant. Thus, when mice were challenged with PR8 virus via the nasal route, both aged and young adult mice given nasal vaccine exhibited complete protection. Further, IgA-deficient mice nasally immunized with a double adjuvant influenza vaccine failed to provide protection against PR8 challenge. These results indicate that a nasal double adjuvant successfully induces PR8 HA-specific IgA Ab responses in both young adult and aged mice, which are essential for the prevention of influenza infection in the murine URT.
Collapse
Affiliation(s)
- Hideki Asanuma
- Influenza Virus Research Center, National Institute of Infectious Diseases, Musashimurayama-shi, Tokyo, JAPAN
- Department of Pediatric Dentistry, The Immunobiology Vaccine Center, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | - Normaiza Binti Zamri
- Department of Applied Biochemistry, School or Engineering, Tokai University, Hiratsuka-shi, Kanagawa, JAPAN
| | - Shin-ichi Sekine
- Department of Pediatric Dentistry, The Immunobiology Vaccine Center, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | - Yoshiko Fukuyama
- Department of Pediatric Dentistry, The Immunobiology Vaccine Center, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | - Daisuke Tokuhara
- Department of Pediatric Dentistry, The Immunobiology Vaccine Center, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | - Rebekah S. Gilbert
- Department of Pediatric Dentistry, The Immunobiology Vaccine Center, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | - Tatsuya Fukuiwa
- Department of Pediatric Dentistry, The Immunobiology Vaccine Center, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | - Tetsutaro Sata
- Department of Pathology, National Institute of Infectious Diseases, Shinjuku-ku, Tokyo, JAPAN
| | - Masato Tashiro
- Influenza Virus Research Center, National Institute of Infectious Diseases, Musashimurayama-shi, Tokyo, JAPAN
| | - Kohtaro Fujihashi
- Department of Pediatric Dentistry, The Immunobiology Vaccine Center, The University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| |
Collapse
|
45
|
Abstract
Two striking facts surround the practice of vaccination: It is the sole medical approach to have fully annihilated a disease, yet the development of most effective vaccines took place without considering the intricate cellular processes they wish to effectuate. While extremely potent vaccines have been developed that can protect practically a lifetime after a single dose, numerous other vaccines have utterly failed or provide only marginal protection. Here, we aim to illustrate why this difference in efficacy exists, and underline why specific cytotoxic T cell-inducing vaccines could combat persistent major diseases. Moreover, we discuss how the combinatorial use of nucleic acid adjuvants in vaccines could aid the development of the latter and move vaccine design from the empirical stage into an era of "educated design."
Collapse
|
46
|
Zhou S, Kawakami S, Higuchi Y, Yamashita F, Hashida M. The involvement of NK cell activation following intranasal administration of CpG DNA lipoplex in the prevention of pulmonary metastasis and peritoneal dissemination in mice. Clin Exp Metastasis 2011; 29:63-70. [PMID: 22052391 DOI: 10.1007/s10585-011-9429-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 10/07/2011] [Indexed: 10/15/2022]
Abstract
Synthetic oligodeoxynucleotides containing CpG motifs (CpG DNA) can activate immunocompetent cells, which may possess antitumor activity. Previously, we found that when the cationic liposomes complexes formed with CpG DNA (CpG DNA lipoplex) were administered intranasally, they could prevent pulmonary metastasis in mice. However, the mechanisms underlying this process are unknown. In the present study, we show that natural killer (NK) cells play an important role in preventing pulmonary metastasis and peritoneal dissemination in a mouse model of metastatic disease. Further, in vitro, the NK cells obtained from mice treated with CpG DNA lipoplex showed higher cytotoxicity compared with untreated mice and in vivo, depletion of NK cells (achieved through injection of rabbit anti-asialo GM1 serum), abolished the inhibitory effect of CpG DNA lipoplex on pulmonary metastasis and peritoneal dissemination. In contrast, macrophage elimination did not disrupt the effects of the CpG DNA lipoplex. These results suggest that intranasal administration of CpG DNA lipoplex could prevent pulmonary metastasis and peritoneal dissemination by activating NK cells.
Collapse
Affiliation(s)
- Shuwen Zhou
- Department of Drug Delivery Research, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | |
Collapse
|
47
|
Tweaking Innate Immunity: the Promise of Innate Immunologicals As Anti-infectives. CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY 2011; 17:307-14. [PMID: 18382644 DOI: 10.1155/2006/195957] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
New and exciting insights into the importance of the innate immune system are revolutionizing our understanding of immune defense against infections, pathogenesis, and the treatment and prevention of infectious diseases. The innate immune system uses multiple families of germline-encoded pattern recognition receptors (PRRs) to detect infection and trigger a variety of antimicrobial defense mechanisms. PRRs are evolutionarily highly conserved and serve to detect infection by recognizing pathogen-associated molecular patterns that are unique to microorganisms and essential for their survival. Toll-like receptors (TLRs) are transmembrane signalling receptors that activate gene expression programs that result in the production of proinflammatory cytokines and chemokines, type I interferons and antimicrobial factors. Furthermore, TLR activation facilitates and guides activation of adaptive immune responses through the activation of dendritic cells. TLRs are localized on the cell surface and in endosomal/lysosomal compartments, where they detect bacterial and viral infections. In contrast, nucleotide-binding oligomerization domain proteins and RNA helicases are located in the cell cytoplasm, where they serve as intracellular PRRs to detect cytoplasmic infections, particularly viruses. Due to their ability to enhance innate immune responses, novel strategies to use ligands, synthetic agonists or antagonists of PRRs (also known as 'innate immunologicals') can be used as stand-alone agents to provide immediate protection or treatment against bacterial, viral or parasitic infections. Furthermore, the newly appreciated importance of innate immunity in initiating and shaping adaptive immune responses is contributing to our understanding of vaccine adjuvants and promises to lead to improved next-generation vaccines.
Collapse
|
48
|
Lebre F, Borchard G, de Lima MCP, Borges O. Progress towards a needle-free hepatitis B vaccine. Pharm Res 2011; 28:986-1012. [PMID: 21088986 DOI: 10.1007/s11095-010-0314-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 10/27/2010] [Indexed: 12/11/2022]
Abstract
Hepatitis B virus (HBV) infection is a worldwide public health problem. Vaccination is the most efficient way to prevent hepatitis B. Despite the success of the currently available vaccine, there is a clear need for the development of new generation of HBV vaccines. Needle-free immunization is an attractive approach for mass immunization campaigns, since avoiding the use of needles reduces the risk of needle-borne diseases and prevents needle-stick injuries and pain, thus augmenting patient compliance and eliminating the need for trained medical personnel. Moreover, this kind of immunization was shown to induce good systemic as well as mucosal immunological responses, which is important for the creation of both a prophylactic and therapeutic vaccine. In order to produce a better, safer, more efficient and more suitable vaccine, adjuvants have been used. In this article, several adjuvants tested over the years for their potential to help create a needle-free vaccine against HBV are reviewed.
Collapse
Affiliation(s)
- Filipa Lebre
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | | | |
Collapse
|
49
|
Steinhagen F, Kinjo T, Bode C, Klinman DM. TLR-based immune adjuvants. Vaccine 2011; 29:3341-55. [PMID: 20713100 PMCID: PMC3000864 DOI: 10.1016/j.vaccine.2010.08.002] [Citation(s) in RCA: 378] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 07/27/2010] [Accepted: 08/01/2010] [Indexed: 12/29/2022]
Abstract
This work describes the nature and strength of the immune response induced by various Toll-like receptor ligands and their ability to act as vaccine adjuvants. It reviews the various ligands capable of triggering individual TLRs, and then focuses on the efficacy and safety of those agents for which clinical results are available.
Collapse
Affiliation(s)
- Folkert Steinhagen
- Cancer and Inflammation Program, National Cancer Institute, Frederick, MD 21702, United States
| | | | | | | |
Collapse
|
50
|
Fukuyama Y, King JD, Kataoka K, Kobayashi R, Gilbert RS, Hollingshead SK, Briles DE, Fujihashi K. A combination of Flt3 ligand cDNA and CpG oligodeoxynucleotide as nasal adjuvant elicits protective secretory-IgA immunity to Streptococcus pneumoniae in aged mice. THE JOURNAL OF IMMUNOLOGY 2011; 186:2454-61. [PMID: 21242514 DOI: 10.4049/jimmunol.1002837] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Our previous study showed that a combination of a plasmid-expressing Flt3 ligand (pFL) and CpG oligodeoxynucleotides (CpG ODN) as a combined nasal adjuvant elicited mucosal immune responses in aged (2-y-old) mice. In this study, we investigated whether a combination of pFL and CpG ODN as a nasal adjuvant for a pneumococcal surface protein A (PspA) would enhance PspA-specific secretory-IgA Ab responses, which could provide protective mucosal immunity against Streptococcus pneumoniae infection in aged mice. Nasal immunization with PspA plus a combination of pFL and CpG ODN elicited elevated levels of PspA-specific secretory-IgA Ab responses in external secretions and plasma in both young adult and aged mice. Significant levels of PspA-specific CD4(+) T cell proliferative and PspA-induced Th1- and Th2- type cytokine responses were noted in nasopharyngeal-associated lymphoreticular tissue, cervical lymph nodes, and spleen of aged mice, which were equivalent to those in young adult mice. Additionally, increased numbers of mature-type CD8, CD11b-expressing dendritic cells were detected in mucosal inductive and effector lymphoid tissues of aged mice. Importantly, aged mice given PspA plus a combination of pFL and CpG ODN showed protective immunity against nasal S. pneumoniae colonization. These results demonstrate that nasal delivery of a combined DNA adjuvant offers an attractive possibility for protection against S. pneumoniae in the elderly.
Collapse
Affiliation(s)
- Yoshiko Fukuyama
- Department of Pediatric Dentistry, Immunobiology Vaccine Center, Institute of Oral Health Research, University of Alabama at Birmingham, AL 35294-0007, USA
| | | | | | | | | | | | | | | |
Collapse
|