101
|
Yüksel S, Pekcan M, Puralı N, Esendağlı G, Tavukçuoğlu E, Rivero-Arredondo V, Ontiveros-Padilla L, López-Macías C, Şenel S. Development and in vitro evaluation of a new adjuvant system containing Salmonella Typhi porins and chitosan. Int J Pharm 2020; 578:119129. [PMID: 32045689 DOI: 10.1016/j.ijpharm.2020.119129] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 12/17/2022]
Abstract
In order to improve the immunogenicity of the highly purified vaccine antigens, addition of an adjuvant to formulation, without affecting the safety of the vaccine, has been the key aim of the vaccine formulators. In recent years, adjuvants which are composed of a delivery system and immunopotentiators have been preferred to induce potent immune responses. In this study, we have combined Salmonella Typhi porins and chitosan to develop a new adjuvant system to enhance the immunogenicity of the highly purified antigens. Cationic gels, microparticle (1.69 ± 0.01 μm) and nanoparticles (337.7 ± 1.7 nm) based on chitosan were prepared with high loading efficiency of porins. Cellular uptake was examined by confocal laser scanning microscopy, and the macrophage activation was investigated by measuring the surface marker as well as the cytokine release in vitro in J774A.1 macrophage murine cells. Porins alone were not taken up by the macrophage cells whereas in combination with chitosan a significant uptake was obtained. Porins-chitosan combination systems were found to induce CD80, CD86 and MHC-II expressions at different levels by different formulations depending on the particle size. Similarly, TNF-α and IL-6 levels were found to increase with porins-chitosan combination. Our results demonstrated that combination of porins with chitosan as a particulate system exerts enhanced adjuvant effect, suggesting a promising adjuvant system for subunit vaccines with combined immunostimulating activity.
Collapse
Affiliation(s)
- Selin Yüksel
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Hacettepe University, 06100 Ankara, Turkey
| | - Mert Pekcan
- Faculty of Veterinary Medicine, Department of Biochemistry, Ankara University, Dışkapı, 06110 Ankara, Turkey
| | - Nuhan Puralı
- Faculty of Medicine, Department of Biophysics, Hacettepe University, 06100 Ankara, Turkey
| | - Güneş Esendağlı
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Ece Tavukçuoğlu
- Department of Basic Oncology, Hacettepe University Cancer Institute, Ankara, Turkey
| | - Vanessa Rivero-Arredondo
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades Centro Médico Nacional 'Siglo XXI', Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Luis Ontiveros-Padilla
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades Centro Médico Nacional 'Siglo XXI', Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Constantino López-Macías
- Unidad de Investigación Médica en Inmunoquímica, Hospital de Especialidades Centro Médico Nacional 'Siglo XXI', Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico.
| | - Sevda Şenel
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Hacettepe University, 06100 Ankara, Turkey.
| |
Collapse
|
102
|
Mechanisms of Mixed Th1/Th2 Responses in Mice Induced by Albizia julibrissin Saponin Active Fraction by i n Silico Analysis. Vaccines (Basel) 2020; 8:vaccines8010048. [PMID: 32012760 PMCID: PMC7158666 DOI: 10.3390/vaccines8010048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/22/2020] [Accepted: 01/23/2020] [Indexed: 12/28/2022] Open
Abstract
The purified active fraction of Albizia julibrissin saponin (AJSAF) is an ideal adjuvant candidate that improves antigen-specific both cellular and humoral immune responses and elicits mixed Th1/Th2 responses, but its mechanisms remain unclear. The key features of action of AJSAF were investigated in mice immunized with Newcastle disease virus-based recombinant influenza vaccine (rL-H5) and AJSAF at the same leg (AJSAF+rL-H5) or different legs (AJSAF/rL-H5). The adjuvant activity of AJSAF on rL-H5 is strictly dependent on their spatial colocalization. Serum H5 antigen (H5Ag)-specific IgG, IgG1, IgG2a, and IgG2b antibody titers in AJSAF+rL-H5 group were significantly higher than those in AJSAF/rL-H5 group. The mechanisms of selectivity of Th1 or Th2 in mice induced by AJSAF was explored by the transcriptomic and proteomic profiles of H5Ag-stimulated splenocytes from the immunized mice using gene microarray and two-dimensional difference gel electrophoresis coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. Compared to rL-H5 alone, AJSAF/rL-H5 induced more differentially expressed genes (DEGs) than AJSAF+rL-H5, whereas AJSAF+rL-H5 upregulated higher mRNA expression of Th1 (T-bet, IFN-γ, TNF-α, IL-12β, and IL-12Rβ1) and Th2 (IL-10 and AICDA) immune response genes. The neutrophil response and its derived S100A8 and S100A9 might be involved in the AJSAF-mediated Th1 response. Meanwhile, AJSAF might induce the adaptive immune responses by improving a local innate immune microenvironment. These findings expanded the current knowledge on the mechanisms of action of saponin-based adjuvants, and provided new insights into how adjuvants shape adaptive immune responses.
Collapse
|
103
|
Askarizadeh A, Badiee A, Khamesipour A. Development of nano-carriers for Leishmania vaccine delivery. Expert Opin Drug Deliv 2020; 17:167-187. [PMID: 31914821 DOI: 10.1080/17425247.2020.1713746] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Leishmaniasis is a neglected tropical infection caused by several species of intracellular protozoan parasites of the genus Leishmania. It is strongly believed that the development of vaccines is the most appropriate approach to control leishmaniasis. However, there is no vaccine available yet and the lack of an appropriate adjuvant delivery system is the main reason.Areas covered: Adjuvants are the utmost important part of a vaccine, to induce the immune response in the right direction. Limitations and drawbacks of conventional adjuvants have been necessitated the development of novel particulate delivery systems as adjuvants to obtain desirable protection against infectious diseases such as leishmaniasis. This review focused on particulate adjuvants especially nanoparticles that are in use to develop vaccines against leishmaniasis. The list of adjuvants includes generally lipids-, polymers-, or mineral-based delivery systems that target antigens specifically to the site of action within the host's body and enhance immune responses.Expert opinion: Over the past few years, there has been an increasing interest in developing particulate adjuvants as alternatives to immunostimulatory types. The composition of nano-carriers and particularly the physicochemical properties of nanoparticles have great potential to overcome challenges posed to leishmaniasis vaccine developments.
Collapse
Affiliation(s)
- Anis Askarizadeh
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Khamesipour
- Center for Research and Training in Skin Diseases and Leprosy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
104
|
Shirai S, Shibuya M, Kawai A, Tamiya S, Munakata L, Omata D, Suzuki R, Aoshi T, Yoshioka Y. Lipid Nanoparticles Potentiate CpG-Oligodeoxynucleotide-Based Vaccine for Influenza Virus. Front Immunol 2020; 10:3018. [PMID: 31998305 PMCID: PMC6962196 DOI: 10.3389/fimmu.2019.03018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
Current influenza vaccines are generally effective against highly similar (homologous) strains, but their effectiveness decreases markedly against antigenically mismatched (heterologous) strains. One way of developing a universal influenza vaccine with a broader spectrum of protection is to use appropriate vaccine adjuvants to improve a vaccine's effectiveness and change its immune properties. Oligodeoxynucleotides (ODNs) with unmethylated cytosine-phosphate-guanine (CpG) motifs (CpG ODNs), which are Toll-like-receptor 9 (TLR9) agonists, are among the most promising adjuvants and are already being used in humans. However, the development of novel delivery vehicles to improve adjuvant effects in vivo is highly desirable. Here, we assessed the potential of lipid nanoparticles (LNPs) as CpG ODN delivery vehicles in mice to augment the vaccine adjuvant effects of CpG ODN and enhance the protective spectrum of conventional influenza split vaccine (SV). In vitro, compared with CpG ODN, LNPs containing CpG ODNs (LNP-CpGs) induced significantly greater production of cytokines such as IL-12 p40 and IFN-α by mouse dendritic cells (DCs) and significantly greater expression of the co-stimulatory molecules CD80 and CD86 on DCs. In addition, after subcutaneous administration in mice, compared with CpG ODN, LNP-CpGs enhanced the expression of CD80 and CD86 on plasmacytoid DCs in draining lymph nodes. LNP-CpGs given with SV from H1N1 influenza A virus improved T-cell responses and gave a stronger not only SV-specific but also heterologous-virus-strain-specific IgG2c response than CpG ODN. Furthermore, immunization with SV plus LNP-CpGs protected against not only homologous strain challenge but also heterologous and heterosubtypic strain challenge, whereas immunization with SV plus CpG ODNs protected against homologous strain challenge only. We therefore demonstrated that LNP-CpGs improved the adjuvant effects of CpG ODN and broadened the protective spectrum of SV against influenza virus. We expect that this strategy will be useful in developing adjuvant delivery vehicles and universal influenza vaccines.
Collapse
Affiliation(s)
- Seiki Shirai
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Meito Shibuya
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Atsushi Kawai
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Shigeyuki Tamiya
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Lisa Munakata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Daiki Omata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Taiki Aoshi
- Vaccine Dynamics Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,BIKEN Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Vaccine Creation Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan.,BIKEN Center for Innovative Vaccine Research and Development, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan.,Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| |
Collapse
|
105
|
Kelly SM, Mitra A, Mathur S, Narasimhan B. Synthesis and Characterization of Rapidly Degrading Polyanhydrides as Vaccine Adjuvants. ACS Biomater Sci Eng 2020; 6:265-276. [PMID: 33463223 DOI: 10.1021/acsbiomaterials.9b01427] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
There is a currently a need to develop adjuvants that are best suited to simultaneously enhance immune responses, induce immunologic memory, improve patient compliance (i.e., reduce doses and inflammation), and provide vaccine shelf stability for stockpiling and global deployment to challenging environments. Biodegradable polyanhydrides have been investigated extensively to overcome such challenges. It has been shown that controlling copolymer composition can result in chemistry-dependent immunomodulatory capabilities. These studies have revealed that copolymers rich in sebacic acid (SA) are highly internalized by antigen presenting cells and confer improved shelf stability of encapsulated proteins, while copolymers rich in 1,8-bis(p-carboxyphenoxy)-3,6-dioxaoctane (CPTEG) also exhibit enhanced internalization by and activation of antigen presenting cells (APCs), in addition to providing superior retention of protein stability following encapsulation and release. However, to date, CPTEG:SA copolymers have not been synthesized and described. In this work, we hypothesized that new copolymers composed of CPTEG and SA would combine the advantages of both monomers in terms of enhanced thermal properties, maintaining antigenicity of encapsulated proteins following nanoparticle synthesis, and superior cellular internalization and activation by APCs, demonstrated by the upregulation of costimulatory markers CD80, CD86, and CD40, as well as the secretion of proinflammatory cytokines IL-6, IL-1β, and TNF-α. Herein, we describe the synthesis and design of novel CPTEG:SA nanoparticles with improved thermal properties, payload stability, and internalization by antigen presenting cells for applications in vaccine delivery. The performance of these new CPTEG:SA formulations was compared to that of traditional polyanhydride copolymers.
Collapse
Affiliation(s)
- Sean M Kelly
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Akash Mitra
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Srishti Mathur
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, Iowa 50011, United States.,Nanovaccine Institute, Iowa State University, Ames, Iowa 50011-1098, United States
| |
Collapse
|
106
|
Heydari S, Hedayati Ch M, Saadat F, Abedinzade M, Nikokar I, Aboutaleb E, Khafri A, Mokarram AR. Diphtheria toxoid nanoparticles improve learning and memory impairment in animal model of Alzheimer's disease. Pharmacol Rep 2019; 72:814-826. [PMID: 32048245 DOI: 10.1007/s43440-019-00017-w] [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: 04/21/2019] [Revised: 08/16/2019] [Accepted: 09/10/2019] [Indexed: 10/23/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder involving memory. The present study aimed at evaluating the effects of encapsulated diphtheria toxoid (DT) on behavioral learning impairment, and XBP1 mRNA splicing in AD. METHODS A DT-loaded nanoparticle (NP) carrier was prepared using the ionic gelation method. Sixty-three rats were divided into nine groups: (1) healthy, (2-4) sham, and (5-9) AD models: (5) AD was induced by intracerebroventricular injection of amyloid beta (Aβ) 1-42. (6) The rats received a subcutaneous diphtheria vaccine only 28 days before Aβ injection. (7) The rats received an intranasal diphtheria vaccine, in group 8, induced by administering empty chitosan NPs. 9) it was induced by administering chitosan NPs carrying DT. Morris water maze (MWM) test was used to examine the animals' learning and memory. Also, X-box binding protein 1 (XBP-1) mRNA gene splicing was studied in the hippocampus by reverse-transcription polymerase chain reaction (RT-PCR). RESULTS For the first time, chitosan NPs were prepared with an average diameter size of 40 nm and the effectiveness of approximately 70% during DT encapsulation. In comparison with the healthy group, the AD models exhibited significant impairment of learning and memory (P < 0.05), while DT-administrated animals showed significant improvements in learning and memory impairment (P < 0.05). XBP-1 mRNA gene splicing was only detected in an untreated AD group, while encapsulated DT completely inhibited splicing. CONCLUSION The therapeutic effects of DT chitosan NPs against learning and memory impairment were observed in this study, and XBP1 mRNA splicing was reported in the animal models.
Collapse
Affiliation(s)
- Samane Heydari
- Medical Biotechnology Research Center, School of Nursing, Midwifery and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mojtaba Hedayati Ch
- Department of Microbiology, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Farshid Saadat
- Department of Microbiology, Parasitology, and Immunology, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahmood Abedinzade
- Medical Biotechnology Research Center, School of Nursing, Midwifery and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Iraj Nikokar
- Medical Biotechnology Research Center, School of Nursing, Midwifery and Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ehsan Aboutaleb
- Department of Pharmaceutics, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Abolfazl Khafri
- Quality Control of Bacterial and Parasitic Vaccine Department, Quality Control Management, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| | - Ali Rezaei Mokarram
- Quality Control of Bacterial and Parasitic Vaccine Department, Quality Control Management, Razi Vaccine and Serum Research Institute, Agricultural Research Education and Extension Organization (AREEO), Karaj, Iran
| |
Collapse
|
107
|
Zhou M, Li Y, Chen X, Zhou H, Yang S, Qu X. Preparation and characterization of polyoxyethylene dehydrated mannitol mono oleate as hydrophilic emulsifier potentially used in w/o/w type adjuvants. J DISPER SCI TECHNOL 2019. [DOI: 10.1080/01932691.2019.1701488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Mengmeng Zhou
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, P.R. China
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, P.R. China
| | - Yantao Li
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, P.R. China
| | - Xiaoqi Chen
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, P.R. China
| | - Haijun Zhou
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, P.R. China
| | - Shulan Yang
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, P.R. China
| | - Xiongwei Qu
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, P.R. China
| |
Collapse
|
108
|
Li Q, Ning X, Wang Y, Zhu Q, Guo Y, Li H, Zhou Y, Kou Z. The Integrity of α-β-α Sandwich Conformation Is Essential for a Novel Adjuvant TFPR1 to Maintain Its Adjuvanticity. Biomolecules 2019; 9:biom9120869. [PMID: 31842458 PMCID: PMC6995627 DOI: 10.3390/biom9120869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/30/2022] Open
Abstract
TFPR1 is a novel peptide vaccine adjuvant we recently discovered. To define the structural basis and optimize its application as an adjuvant, we designed three different truncated fragments that have removed dominant B epitopes on TFPR1, and evaluated their capacity to activate bone marrow-derived dendritic cells and their adjuvanticity. Results demonstrated that the integrity of an α-β-α sandwich conformation is essential for TFPR1 to maintain its immunologic activity and adjuvanticity. We obtained a functional truncated fragment TFPR-ta ranging from 40-168 aa of triflin that has similar adjuvanticity as TFPR1 but with 2-log fold lower immunogenicity. These results demonstrated a novel approach to evaluate and improve the activity of protein-based vaccine adjuvant.
Collapse
Affiliation(s)
- Qiao Li
- Beijing Institute of Microbiology and Epidemiology, Anhui Medical University, Hefei 230032, China; (Q.L.); (Y.W.)
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; (X.N.); (Q.Z.); (Y.G.); (H.L.); (Y.Z.)
| | - Xiuzhe Ning
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; (X.N.); (Q.Z.); (Y.G.); (H.L.); (Y.Z.)
| | - Yuepeng Wang
- Beijing Institute of Microbiology and Epidemiology, Anhui Medical University, Hefei 230032, China; (Q.L.); (Y.W.)
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; (X.N.); (Q.Z.); (Y.G.); (H.L.); (Y.Z.)
| | - Qing Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; (X.N.); (Q.Z.); (Y.G.); (H.L.); (Y.Z.)
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; (X.N.); (Q.Z.); (Y.G.); (H.L.); (Y.Z.)
| | - Hao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; (X.N.); (Q.Z.); (Y.G.); (H.L.); (Y.Z.)
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; (X.N.); (Q.Z.); (Y.G.); (H.L.); (Y.Z.)
| | - Zhihua Kou
- Beijing Institute of Microbiology and Epidemiology, Anhui Medical University, Hefei 230032, China; (Q.L.); (Y.W.)
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; (X.N.); (Q.Z.); (Y.G.); (H.L.); (Y.Z.)
- Correspondence: ; Tel.: +86-10-63858045
| |
Collapse
|
109
|
Chemically engineered glycan-modified cancer vaccines to mobilize skin dendritic cells. Curr Opin Chem Biol 2019; 53:167-172. [DOI: 10.1016/j.cbpa.2019.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/27/2019] [Accepted: 10/02/2019] [Indexed: 12/15/2022]
|
110
|
Abstract
Vaccination is a critical tool in modern animal production and key to maintaining animal health. Adjuvants affect the immune response by increasing the rate, quantity, or quality of the protective response generated by the target antigens. Although adjuvant technology dates back to the nineteenth century, there was relatively little improvement in adjuvant technology before the late twentieth century. With the discovery of molecular pathways that regulate the timing, quantity, and quality of the immune response, new technologies are focused on bringing safer, more effective, and inexpensive adjuvants to commercial use.
Collapse
Affiliation(s)
- Alan J Young
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Box 2175, ARW168F, Brookings, SD 57006, USA.
| |
Collapse
|
111
|
Adjuvant effect of TLR7 agonist adsorbed on aluminum hydroxide (AS37): A phase I randomized, dose escalation study of an AS37-adjuvanted meningococcal C conjugated vaccine. Clin Immunol 2019; 209:108275. [PMID: 31669193 DOI: 10.1016/j.clim.2019.108275] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 08/02/2019] [Accepted: 10/17/2019] [Indexed: 02/06/2023]
Abstract
An adjuvant system (AS37) has been developed containing a synthetic toll-like receptor agonist (TLR7a). We conducted a phase I randomized, observer-blind, dose-escalation study to assess the safety and immunogenicity of an investigational AS37-adjuvanted meningococcus C (MenC) conjugate vaccine in healthy adults (NCT02639351). A control group received a licensed MenC conjugate alum-adjuvanted vaccine. Eighty participants were randomized to receive one dose of control or investigational vaccine containing AS37 (TLR7a dose 12.5, 25, 50, 100 μg). All vaccines were well tolerated, apart from in the TLR7a 100 μg dose group, which had three reports (18.8%) of severe systemic adverse events. Four weeks after vaccination, human complement serum bactericidal assay seroresponse rates against MenC were 56-81% in all groups, and ELISA seroresponses were ≥81% for all AS37-adjuvanted vaccine groups (100% in 50 and 100 μg dose groups) and 88% in the control group. Antibody responses were maintained at six months after vaccination.
Collapse
|
112
|
Sun W, Li Q, Ning X, Yang Y, Guo J, Zhu Q, Guo Y, Li H, Wang Y, Zhou Y, Kou Z. TFPR1 acts as an immune regulator and an efficient adjuvant for proteins and peptides by activating immune cells, primarily through TLR2. Vaccine 2019; 38:288-297. [PMID: 31629567 DOI: 10.1016/j.vaccine.2019.10.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 09/03/2019] [Accepted: 10/05/2019] [Indexed: 02/08/2023]
Abstract
Triflin, a non-toxic protein found in the venom of the Habu snake, belongs to the CRISP (cysteine-rich secretory protein) family, which comprises two domains: a C-terminal cysteine-rich domain (CRD) and an N-terminal pathogenesis-related-1 (PR-1) domain. The function of the highly structurally conserved PR-1 domain is unknown. Here, we successfully expressed the PR-1 domain of triflin (hereafter called TFPR1) in E. coli. Animal experiments showed that TFPR1 augmented Th1-biased antibody- and cell-mediated immune responses in mice immunized with two protein antigens (OVA and HBsAg) or a peptide antigen (HIV-1 pep). A flow cytometry-based binding assay and in vitro stimulation with TFPR1 showed that it triggered Th1-biased proinflammatory and immunoregulatory cytokine secretion primarily by binding to B cells and macrophages within the mouse splenocyte population. Quantitative RT-PCR, antibody blocking assays using a specific anti-mTLR2 antibody, and stimulatory experiments in vitro using splenocytes from TLR2-KO mice demonstrated that TFPR1 activated murine immune cells, primarily by stimulating toll-like receptor 2 (TLR2). These results suggest that TFPR1 acts as a novel immune modulator and potent adjuvant primarily by activating TLR2. Thus, the PR-1-based core domain might play a role in immune regulation.
Collapse
Affiliation(s)
- Weilai Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qiao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Graduate School, Anhui Medical University, Hefei 230032, China
| | - Xiuzhe Ning
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yi Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Jingjing Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Qing Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yan Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Hao Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China
| | - Yuepeng Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Graduate School, Anhui Medical University, Hefei 230032, China
| | - Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Graduate School, Anhui Medical University, Hefei 230032, China
| | - Zhihua Kou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China; Graduate School, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
113
|
Zhang Z, Zhang T, Cao L, Wang X, Cao J, Huang X, Cai Y, Lin Z, Pan H, Yuan Q, Fang M, Li S, Zhang J, Xia N, Zhao Q. Simultaneous in situ visualization and quantitation of dual antigens adsorbed on adjuvants using high content analysis. Nanomedicine (Lond) 2019; 14:2535-2548. [PMID: 31603382 DOI: 10.2217/nnm-2019-0016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Aim: Traditional antigenicity assay requires antigen recovery from the particulate adjuvants prior to analysis. An in situ method was developed for interrogating vaccine antigens with monoclonal antibodies while being adsorbed on adjuvants. Materials & methods: The fluorescence imaging-based high content analysis was used to visualize the antigen distribution on adjuvant agglomerates and to analyze the antigenicity for adsorbed antigens. Results: Simultaneous visualization and quantitation were achieved for dual antigens in a bivalent human papillomavirus vaccine with uniquely labeled antibodies. Good agreement was observed between the in situ multiplexed assays with well-established sandwich enzyme-linked immunosorbent assays. Conclusion: The streamlined procedures and the amenability for multiplexing make the in situ antigenicity analysis a favorable choice for in vitro functional assessment of bionanoparticles as vaccine antigens.
Collapse
Affiliation(s)
- Zhigang Zhang
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Tianying Zhang
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Lu Cao
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Xin Wang
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Jiali Cao
- National Institute of Diagnostics & Vaccine Development in Infectious Diseases, School of Life Sciences, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Xiaofen Huang
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Yashuang Cai
- National Institute of Diagnostics & Vaccine Development in Infectious Diseases, School of Life Sciences, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Zhijie Lin
- Xiamen Innovax Biotech Co., Ltd, Xiamen, Fujian 361022, PR China
| | - Huirong Pan
- Xiamen Innovax Biotech Co., Ltd, Xiamen, Fujian 361022, PR China
| | - Quan Yuan
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China.,National Institute of Diagnostics & Vaccine Development in Infectious Diseases, School of Life Sciences, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Mujin Fang
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China.,National Institute of Diagnostics & Vaccine Development in Infectious Diseases, School of Life Sciences, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China.,National Institute of Diagnostics & Vaccine Development in Infectious Diseases, School of Life Sciences, Xiamen University, Xiamen, Fujian 361105, PR China
| | - Qinjian Zhao
- State Key Laboratory of Molecular Vaccinology & Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, Fujian 361105, PR China
| |
Collapse
|
114
|
Alqarni AM, Dissanayake T, Nelson DJ, Parkinson JA, Dufton MJ, Ferro VA, Watson DG. Metabolomic Profiling of the Immune Stimulatory Effect of Eicosenoids on PMA-Differentiated THP-1 Cells. Vaccines (Basel) 2019; 7:vaccines7040142. [PMID: 31600945 PMCID: PMC6963534 DOI: 10.3390/vaccines7040142] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022] Open
Abstract
Honey bee venom has been established to have significant effect in immunotherapy. In the present study, (Z)-11-eicosenol-a major constituent of bee venom, along with its derivations methyl cis-11-eicosenoate and cis-11-eicosenoic acid, were synthesised to investigate their immune stimulatory effect and possible use as vaccine adjuvants. Stimuli that prime and activate the immune system have exerted profound effects on immune cells, particularly macrophages; however, the effectiveness of bee venom constituents as immune stimulants has not yet been established. Here, the abilities of these compounds to act as pro-inflammatory stimuli were assessed, either alone or in combination with lipopolysaccharide (LPS), by examining the secretion of tumour necrosis factor-α (TNF-α) and the cytokines interleukin-1β (IL-1β), IL-6 and IL-10 by THP-1 macrophages. The compounds clearly increased the levels of IL-1β and decreased IL-10, whereas a decrease in IL-6 levels suggested a complex mechanism of action. A more in-depth profile of macrophage behaviour was therefore obtained by comprehensive untargeted metabolic profiling of the cells using liquid chromatography mass spectrometry (LC-MS) to confirm the ability of the eicosanoids to trigger the immune system. The level of 358 polar and 315 non-polar metabolites were changed significantly (p < 0.05) by all treatments. The LPS-stimulated production of most of the inflammatory metabolite biomarkers in glycolysis, the tricarboxylic acid (TCA) cycle, the pentose phosphate pathway, purine, pyrimidine and fatty acids metabolism were significantly enhanced by all three compounds, and particularly by methyl cis-11-eicosenoate and cis-11-eicosenoic acid. These findings support the proposed actions of (Z)-11-eicosenol, methyl cis-11-eicosenoate and cis-11-eicosenoic acid as immune system stimulators.
Collapse
Affiliation(s)
- Abdulmalik M Alqarni
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University (University of Dammam), Dammam 31441, Saudi Arabia.
| | - Tharushi Dissanayake
- WestCHEM Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK.
| | - David J Nelson
- WestCHEM Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK.
| | - John A Parkinson
- WestCHEM Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK.
| | - Mark J Dufton
- WestCHEM Department of Pure and Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow G1 1XL, UK.
| | - Valerie A Ferro
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| | - David G Watson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK.
| |
Collapse
|
115
|
Crooke SN, Ovsyannikova IG, Poland GA, Kennedy RB. Immunosenescence and human vaccine immune responses. IMMUNITY & AGEING 2019; 16:25. [PMID: 31528180 PMCID: PMC6743147 DOI: 10.1186/s12979-019-0164-9] [Citation(s) in RCA: 332] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 08/27/2019] [Indexed: 12/11/2022]
Abstract
The age-related dysregulation and decline of the immune system-collectively termed "immunosenescence"-has been generally associated with an increased susceptibility to infectious pathogens and poor vaccine responses in older adults. While numerous studies have reported on the clinical outcomes of infected or vaccinated individuals, our understanding of the mechanisms governing the onset of immunosenescence and its effects on adaptive immunity remains incomplete. Age-dependent differences in T and B lymphocyte populations and functions have been well-defined, yet studies that demonstrate direct associations between immune cell function and clinical outcomes in older individuals are lacking. Despite these knowledge gaps, research has progressed in the development of vaccine and adjuvant formulations tailored for older adults in order to boost protective immunity and overcome immunosenescence. In this review, we will discuss the development of vaccines for older adults in light of our current understanding-or lack thereof-of the aging immune system. We highlight the functional changes that are known to occur in the adaptive immune system with age, followed by a discussion of current, clinically relevant pathogens that disproportionately affect older adults and are the central focus of vaccine research efforts for the aging population. We conclude with an outlook on personalized vaccine development for older adults and areas in need of further study in order to improve our fundamental understanding of adaptive immunosenescence.
Collapse
Affiliation(s)
- Stephen N Crooke
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| | - Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Mayo Clinic, Guggenheim Building 611D, 200 First Street SW, Rochester, MN 55905 USA
| |
Collapse
|
116
|
Park HJ, Ko HL, Won DH, Hwang DB, Shin YS, Kwak HW, Kim HJ, Yun JW, Nam JH. Comprehensive Analysis of the Safety Profile of a Single-Stranded RNA Nano-Structure Adjuvant. Pharmaceutics 2019; 11:E464. [PMID: 31500241 PMCID: PMC6781302 DOI: 10.3390/pharmaceutics11090464] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 01/01/2023] Open
Abstract
Adjuvants enhance the efficacy of vaccines by stimulating immune response-related gene expression and pathways. Although some adjuvants have been approved for commercial use in human vaccines (e.g., Alum, MF59, and AS03), they might elicit adverse side effects, such as autoimmune diseases. Recently, we developed a novel single-stranded RNA (ssRNA) nano-structure adjuvant, which can stimulate both Th1 and Th2 responses. In this study, we evaluated the safety and toxicological profiles of this ssRNA nano-structure adjuvant in vitro and in vivo. Mice were intramuscularly immunized with the ssRNA nano-structure adjuvant three times, once every 2 weeks. The results indicate no significant differences in hematological and serum biochemistry parameters between the ssRNA-treated groups and the control group. From a histopathological perspective, no evidence of tissue damage was found in any group. The levels of IgE and anti-nuclear antibodies, which are markers of autoimmune disease, were not different between the ssRNA-treated groups and the control group. The findings of this study suggest that the ssRNA nano-structure can be used as a safe adjuvant to increase vaccine efficacies.
Collapse
Affiliation(s)
- Hyeong-Jun Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Hae Li Ko
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Dong-Hoon Won
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Da-Bin Hwang
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Yoo-Sub Shin
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Hye-Won Kwak
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Hye-Jung Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Jun-Won Yun
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| |
Collapse
|
117
|
Rudicell RS, Garinot M, Kanekiyo M, Kamp HD, Swanson K, Chou TH, Dai S, Bedel O, Simard D, Gillespie RA, Yang K, Reardon M, Avila LZ, Besev M, Dhal PK, Dharanipragada R, Zheng L, Duan X, Dinapoli J, Vogel TU, Kleanthous H, Mascola JR, Graham BS, Haensler J, Wei CJ, Nabel GJ. Comparison of adjuvants to optimize influenza neutralizing antibody responses. Vaccine 2019; 37:6208-6220. [PMID: 31493950 DOI: 10.1016/j.vaccine.2019.08.030] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/26/2019] [Accepted: 08/17/2019] [Indexed: 12/14/2022]
Abstract
Seasonal influenza vaccines represent a positive intervention to limit the spread of the virus and protect public health. Yet continual influenza evolution and its ability to evade immunity pose a constant threat. For these reasons, vaccines with improved potency and breadth of protection remain an important need. We previously developed a next-generation influenza vaccine that displays the trimeric influenza hemagglutinin (HA) on a ferritin nanoparticle (NP) to optimize its presentation. Similar to other vaccines, HA-nanoparticle vaccine efficacy is increased by the inclusion of adjuvants during immunization. To identify the optimal adjuvants to enhance influenza immunity, we systematically analyzed TLR agonists for their ability to elicit immune responses. HA-NPs were compatible with nearly all adjuvants tested, including TLR2, TLR4, TLR7/8, and TLR9 agonists, squalene oil-in-water mixtures, and STING agonists. In addition, we chemically conjugated TLR7/8 and TLR9 ligands directly to the HA-ferritin nanoparticle. These TLR agonist-conjugated nanoparticles induced stronger antibody responses than nanoparticles alone, which allowed the use of a 5000-fold-lower dose of adjuvant than traditional admixtures. One candidate, the oil-in-water adjuvant AF03, was also tested in non-human primates and showed strong induction of neutralizing responses against both matched and heterologous H1N1 viruses. These data suggest that AF03, along with certain TLR agonists, enhance strong neutralizing antibody responses following influenza vaccination and may improve the breadth, potency, and ultimately vaccine protection in humans.
Collapse
Affiliation(s)
| | | | - Masaru Kanekiyo
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | - Rebecca A Gillespie
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | - John R Mascola
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
118
|
Huang CH, Huang CY, Huang MH. Impact of antigen-adjuvant associations on antigen uptake and antigen-specific humoral immunity in mice following intramuscular injection. Biomed Pharmacother 2019; 118:109373. [PMID: 31545268 DOI: 10.1016/j.biopha.2019.109373] [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: 06/04/2019] [Revised: 08/17/2019] [Accepted: 08/22/2019] [Indexed: 11/26/2022] Open
Abstract
The effect of antigen-adjuvant associations on antigen uptake and antigen-specific humoral immunity is studied in detail. After formulation with a squalene-based double emulsion (referred to as PELC), the protein ovalbumin (OVA) was intramuscularly injected in mice, in either a separation (OVA-PELCSE), a surface attachment (OVA-PELCSA) or an encapsulation (OVA-PELCEN) manner. As an antigen delivery system, a significant increase of OVA-loaded cells migrating into draining lymph nodes (LNs) was detected in the PELC-formulated OVA groups, attachment and encapsulation as well. Additionally, OVA-PELCEN allowed the mice to induce a delayed but long-lasting OVA-specific antibodies production compared to OVA-PELCSA. In the extreme case where no antigen-adjuvant association at all (i.e., OVA-PELCSE), we found that even with the presence of PELC at the contralateral limb, an elevated level of OVA uptake was detected in ipsilateral draining CD11c+ LN cells, which subsequently augmented the production of OVA-specific IgG antibodies during early vaccination. The mouse study allows us to find out the optimal vaccine formulation and deepens our understandings on how antigen-adjuvant associations can govern the cellular uptake and transportation of protein antigen into the draining LNs and prolong antigen-specific humoral immunity, even if the antigen and the adjuvant are given separately.
Collapse
Affiliation(s)
- Chung-Hsiung Huang
- Department of Food Science, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Chiung-Yi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Ming-Hsi Huang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 35053, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan; Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| |
Collapse
|
119
|
Wigton EJ, DeFranco AL, Ansel KM. Antigen Complexed with a TLR9 Agonist Bolsters c-Myc and mTORC1 Activity in Germinal Center B Lymphocytes. Immunohorizons 2019; 3:389-401. [PMID: 31427364 PMCID: PMC6738343 DOI: 10.4049/immunohorizons.1900030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/31/2019] [Indexed: 01/12/2023] Open
Abstract
The germinal center (GC) is the anatomical site where humoral immunity evolves. B cells undergo cycles of proliferation and selection to produce high-affinity Abs against Ag. Direct linkage of a TLR9 agonist (CpG) to a T-dependent Ag increases the number of GC B cells. We used a T-dependent Ag complexed with CpG and a genetic model for ablating the TLR9 signaling adaptor molecule MyD88 specifically in B cells (B-MyD88- mice) together with transcriptomics to determine how this innate pathway positively regulates the GC. GC B cells from complex Ag-immunized B-MyD88- mice were defective in inducing gene expression signatures downstream of c-Myc and mTORC1. In agreement with the latter gene signature, ribosomal protein S6 phosphorylation was increased in GC B cells from wild-type mice compared with B-MyD88- mice. However, GC B cell expression of a c-Myc protein reporter was enhanced by CpG attached to Ag in both wild-type and B-MyD88- mice, indicating a B cell-extrinsic effect on c-Myc protein expression combined with a B cell-intrinsic enhancement of gene expression downstream of c-Myc. Both mTORC1 activity and c-Myc are directly induced by T cell help, indicating that TLR9 signaling in GC B cells either enhances their access to T cell help or directly influences these pathways to further enhance the effect of T cell help. Taken together, these findings indicate that TLR9 signaling in the GC could provide a surrogate prosurvival stimulus, "TLR help," thus lowering the threshold for selection and increasing the magnitude of the GC response.
Collapse
Affiliation(s)
- Eric J Wigton
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143; and.,Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143
| | - Anthony L DeFranco
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143
| | - K Mark Ansel
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143; and .,Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|
120
|
Comberlato A, Paloja K, Bastings MMC. Nucleic acids presenting polymer nanomaterials as vaccine adjuvants. J Mater Chem B 2019; 7:6321-6346. [PMID: 31460563 DOI: 10.1039/c9tb01222b] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Most vaccines developed today include only the antigens that best stimulate the immune system rather than the entire virus or microbe, which makes vaccine production and use safer and easier, though they lack potency to induce acceptable immunity and long-term protection. The incorporation of additional immune stimulating components, named adjuvants, is required to generate a strong protective immune response. Nucleic acids (DNA and RNA) and their synthetic analogs are promising candidates as vaccine adjuvants activating Toll-like receptors (TLRs). Additionally, in the last few years several nanocarriers have emerged as platforms for targeted co-delivery of antigens and adjuvants. In this review, we focus on the recent developments in polymer nanomaterials presenting nucleic acids as vaccine adjuvants. We aim to compare the effectiveness of the various classes of polymers in immune modulating materials (nanoparticles, dendrimers, single-chain particles, nanogels, polymersomes and DNA-based architectures). In particular, we address the critical role of parameters such as size, shape, complexation and release of TLR ligands, cellular uptake, stability, toxicity and potential importance of spatial control in ligand presentation.
Collapse
Affiliation(s)
- Alice Comberlato
- IMX/IBI, EPFL, EPFL-STI-IMX-PBL MXC 340 Station 12, Lausanne, 1015, Switzerland.
| | - Kaltrina Paloja
- IMX/IBI, EPFL, EPFL-STI-IMX-PBL MXC 340 Station 12, Lausanne, 1015, Switzerland.
| | - Maartje M C Bastings
- IMX/IBI, EPFL, EPFL-STI-IMX-PBL MXC 340 Station 12, Lausanne, 1015, Switzerland.
| |
Collapse
|
121
|
Kwak HW, Park HJ, Ko HL, Park H, Cha MH, Lee SM, Kang KW, Kim RH, Ryu SR, Kim HJ, Kim JO, Song M, Kim H, Jeong DG, Shin EC, Nam JH. Cricket paralysis virus internal ribosome entry site-derived RNA promotes conventional vaccine efficacy by enhancing a balanced Th1/Th2 response. Vaccine 2019; 37:5191-5202. [PMID: 31371226 PMCID: PMC7115557 DOI: 10.1016/j.vaccine.2019.07.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/21/2019] [Accepted: 07/18/2019] [Indexed: 12/20/2022]
Abstract
RNA adjuvant was developed from the CrPV intergenic region IRES. The RNA adjuvant functioned as an adjuvant with protein-based vaccines. The RNA adjuvant increased vaccine efficacy and induced balanced Th1/Th2 response. The RNA adjuvant enhanced APC chemotaxis.
An ideal adjuvant should increase vaccine efficacy through balanced Th1/Th2 responses and be safe to use. Recombinant protein-based vaccines are usually formulated with aluminum (alum)-based adjuvants to ensure an adequate immune response. However, use of alum triggers a Th2-biased immune induction, and hence is not optimal. Although the adjuvanticity of RNA has been reported, a systematic and overall investigation on its efficacy is lacking. We found that single strand RNA (termed RNA adjuvant) derived from cricket paralysis virus intergenic region internal ribosome entry site induced the expression of various adjuvant-function-related genes, such as type 1 and 2 interferon (IFN) and toll-like receptor (TLR), T cell activation, and leukocyte chemotaxis in human peripheral blood mononuclear cells; furthermore, its innate and IFN transcriptome profile patterns were similar to those of a live-attenuated yellow fever vaccine. This suggests that protein-based vaccines formulated using RNA adjuvant function as live-attenuated vaccines. Application of the RNA adjuvant in mouse enhanced the efficacy of Middle East respiratory syndrome spike protein, a protein-subunit vaccine and human papillomavirus L1 protein, a virus-like particle vaccine, by activating innate immune response through TLR7 and enhancing pAPC chemotaxis, leading to a balanced Th1/Th2 responses. Moreover, the combination of alum and the RNA adjuvant synergistically induced humoral and cellular immune responses and endowed long-term immunity. Therefore, RNA adjuvants have broad applicability and can be used with all conventional vaccines to improve vaccine efficacy qualitatively and quantitively.
Collapse
Affiliation(s)
- Hye Won Kwak
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hyo-Jung Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hae Li Ko
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hyelim Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Min Ho Cha
- KM Application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
| | - Sang-Myeong Lee
- Division of Biotechnology, The Chonbuk National University, Iksan, Republic of Korea
| | - Kyung Won Kang
- Division of Biotechnology, The Chonbuk National University, Iksan, Republic of Korea
| | - Rhoon-Ho Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Seung Rok Ryu
- Division of Biotechnology, The Chonbuk National University, Iksan, Republic of Korea
| | - Hye-Jung Kim
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Jae-Ouk Kim
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, Republic of Korea
| | - Manki Song
- Clinical Research Lab, International Vaccine Institute, Seoul National University Research Park, Seoul, Republic of Korea
| | - Hun Kim
- Life Science Research Institute, SK Bioscience, Seongnam, Republic of Korea
| | - Dae Gwin Jeong
- Infectious Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea.
| |
Collapse
|
122
|
Trier NH, Güven E, Skogstrand K, Ciplys E, Slibinskas R, Houen G. Comparison of immunological adjuvants. APMIS 2019; 127:635-641. [DOI: 10.1111/apm.12976] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/19/2019] [Indexed: 01/28/2023]
Affiliation(s)
- Nicole H. Trier
- Department of Autoimmunology Statens Serum Institut Copenhagen S Denmark
| | - Esin Güven
- Department of Autoimmunology Statens Serum Institut Copenhagen S Denmark
| | - Kristin Skogstrand
- Department of Congenital Diseases Statens Serum Institut Copenhagen S Denmark
| | - Evaldas Ciplys
- Institute of Biotechnology University of Vilnius Vilnius Lithuania
| | | | - Gunnar Houen
- Department of Autoimmunology Statens Serum Institut Copenhagen S Denmark
| |
Collapse
|
123
|
Use of syngeneic cells expressing membrane-bound GM-CSF as an adjuvant to induce antibodies against native multi-pass transmembrane protein. Sci Rep 2019; 9:9931. [PMID: 31289297 PMCID: PMC6616555 DOI: 10.1038/s41598-019-45160-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/12/2019] [Indexed: 12/23/2022] Open
Abstract
Membrane antigens (mAgs) are important targets for the development of antibody (Ab) drugs. However, native mAgs are not easily prepared, causing difficulties in acquiring functional Abs. In this study, we present a platform in which human mAgs were expressed in native form on cell adjuvants made with membrane-bound cytokines that were then used immunize syngeneic mice directly. The membrane-bound cytokines were used as immune stimulators to enhance specific Ab responses against the desired mAgs. Then, mAgs-expressing xenogeneic cells were used for Ab characterization to reduce non-specific binding. We established cell adjuvants by expressing membrane-bound cytokines (mIL-2, mIL-18, or mGM-CSF) on BALB/3T3 cells, which were effective in stimulating splenocyte proliferation in vitro. We then transiently expressed ecotropic viral integration site 2B (EVI2B) on the adjuvants and used them to directly immunize BALB/c mice. We found that 3T3/mGM-CSF cells stimulated higher specific anti-EVI2B Ab response in the immunized mice than the other cell adjuvants. A G-protein coupled receptor (GPCR), CXCR2, was then transiently expressed on 3T3/mGM-CSF cell adjuvant to immunize mice. The immune serum exhibited relatively higher binding to xenogeneic 293 A/CXCR2 cells than 293 A cells (~3.5-fold). Several hybridoma clones also exhibited selective binding to 293 A/CXCR2 cells. Therefore, the cell adjuvant could preserve the native conformation of mAgs and exhibit anti-mAg Ab stimulatory ability, providing a more convenient and effective method to generate functional Abs, thus possibly accelerating Ab drug development.
Collapse
|
124
|
Shimizu K, Yamasaki S, Sakurai M, Yumoto N, Ikeda M, Mishima-Tsumagari C, Kukimoto-Niino M, Watanabe T, Kawamura M, Shirouzu M, Fujii SI. Granzyme A Stimulates pDCs to Promote Adaptive Immunity via Induction of Type I IFN. Front Immunol 2019; 10:1450. [PMID: 31293597 PMCID: PMC6606709 DOI: 10.3389/fimmu.2019.01450] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 06/10/2019] [Indexed: 01/01/2023] Open
Abstract
Granzyme A (GzmA), together with perforin, are well-known for their cytotoxic activity against tumor or virus-infected cells. In addition to this cytotoxic function, GzmA stimulates several immune cell types and induces inflammation in the absence of perforin, however, its effect on the dendritic cell (DC) is unknown. In the current study, we showed that recombinant GzmA induced the phenotypic maturation of plasmacytoid DCs (pDCs) and conventional DCs (cDCs), but not their apoptosis. Particularly, GzmA made pDCs more functional, thus leading to production of type I interferon (IFN) via the TLR9-MyD88 pathway. We also demonstrated that GzmA binds TLR9 and co-localizes with it in endosomes. When co-administered with antigen, GzmA acted as a powerful adjuvant for eliciting antigen-specific cytotoxic CD8+ T lymphocytes (CTLs) that protected mice from tumor challenge. The induction of CTL was completely abolished in XCR1+ DC-depleted mice, whereas it was reduced to less than half in pDC-depleted or IFN-α/β receptor knockout mice. Thus, CTL cross-priming was dependent on XCR1+cDC and also type I IFN, which was produced by GzmA-activated pDCs. These results indicate that GzmA -stimulated pDCs enhance the cross-priming activity of cDCs in situ. We also showed that the adjuvant effect of GzmA is superior to CpG-ODN and LPS. Our findings highlight the ability of GzmA to bridge innate and adaptive immune responses via pDC help and suggest that GzmA may be useful as a vaccine adjuvant.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Maki Sakurai
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Noriko Yumoto
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mariko Ikeda
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Chiemi Mishima-Tsumagari
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Mutsuko Kukimoto-Niino
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Takashi Watanabe
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masami Kawamura
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
125
|
Affiliation(s)
- Judith E. Allen
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Tara E. Sutherland
- Lydia Becker Institute for Immunology and Infection, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| |
Collapse
|
126
|
Crotty S. T Follicular Helper Cell Biology: A Decade of Discovery and Diseases. Immunity 2019; 50:1132-1148. [PMID: 31117010 PMCID: PMC6532429 DOI: 10.1016/j.immuni.2019.04.011] [Citation(s) in RCA: 1036] [Impact Index Per Article: 172.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/16/2019] [Accepted: 04/25/2019] [Indexed: 01/09/2023]
Abstract
Helping B cells and antibody responses is a major function of CD4+ T cells. It has been 10 years since the publication of Bcl6 as the lineage-defining transcription factor for T follicular helper (Tfh) differentiation and the requirement of Tfh cells as the specialized subset of CD4+ T cells needed for germinal centers (the microanatomical sites of B cell mutation and antibody affinity maturation) and related B cell responses. A great deal has been learned about Tfh cells in the past 10 years, particularly regarding their roles in a surprising range of diseases. Advances in the understanding of Tfh cell differentiation and function are discussed, as are the understanding of Tfh cells in infectious diseases, vaccines, autoimmune diseases, allergies, atherosclerosis, organ transplants, and cancer. This includes discussion of Tfh cells in the human immune system. Based on the discoveries to date, the next decade of Tfh research surely holds many more surprises. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Shane Crotty
- Division of Vaccine Discovery, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery (Scripps CHAVI-ID), Scripps Research, La Jolla, CA 92037, USA; Department of Medicine, University of California, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
127
|
Vreman S, McCaffrey J, Popma-de Graaf DJ, Nauwynck H, Savelkoul HFJ, Moore A, Rebel JMJ, Stockhofe-Zurwieden N. Toll-like receptor agonists as adjuvants for inactivated porcine reproductive and respiratory syndrome virus (PRRSV) vaccine. Vet Immunol Immunopathol 2019; 212:27-37. [PMID: 31213249 DOI: 10.1016/j.vetimm.2019.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/21/2019] [Accepted: 04/27/2019] [Indexed: 01/14/2023]
Abstract
Toll-like receptor (TLR) agonists can effectively stimulate antigen-presenting cells (APCs) and are anticipated to be promising adjuvants in combination with inactivated vaccines. In this study, the adjuvant potential of three different TLR-agonists were compared with an oil-in-water (O/W) adjuvant in combination with inactivated porcine reproductive and respiratory syndrome virus (iPRRSV) applied by different administration routes: intramuscular (i.m.) or into the skin using dissolving microneedle (DMN) patches. Pigs received a prime vaccination followed by a booster vaccination four weeks later. TLR1/2 (Pam3Cys), TLR7/8 (R848) or TLR9 (CpG ODN) agonists were used as adjuvant in combination with iPRRSV strain 07V063. O/W adjuvant (Montanide™) was used as reference control adjuvant and one group received a placebo vaccination containing diluent only. All animals received a homologous challenge with PRRSV three weeks after the booster vaccination. Antibody and IFN-γ production, serum cytokines and viremia were measured at several time-points after vaccination and/or challenge, and lung pathology at necropsy. Our results indicate that a TLR 1/2, 7/8 or 9 agonist as adjuvant with iPRRSV does not induce a detectable PRRSV-specific immune response, independent of the administration route. However, the i.m. TLR9 agonist group showed reduction of viremia upon challenge compared to the non-vaccinated animals, supported by a non-antigen-specific IFN-γ level after booster vaccination and an anamnestic antibody response after challenge. Montanide™-adjuvanted iPRRSV induced antigen-specific immunity after booster combined with reduction of vireamia. Skin application of TLR7/8 agonist, but not the other agonists, induced a local skin reaction. Further research is needed to explore the potential of TLR agonists as adjuvants for inactivated porcine vaccines with a preference for TLR9 agonists.
Collapse
Affiliation(s)
- Sandra Vreman
- Wageningen Bioveterinary Research, Wageningen University & Research, the Netherlands; Cell Biology & Immunology group, Wageningen University & Research, the Netherlands.
| | | | | | - Hans Nauwynck
- Laboratory of Virology, Faculty of Veterinary Medicine, Ghent University, Belgium
| | - Huub F J Savelkoul
- Cell Biology & Immunology group, Wageningen University & Research, the Netherlands
| | - Anne Moore
- School of Pharmacy, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Johanna M J Rebel
- Wageningen Livestock Research, Wageningen University & Research, the Netherlands
| | | |
Collapse
|
128
|
Vaccination against atherosclerosis. Curr Opin Immunol 2019; 59:15-24. [PMID: 30928800 DOI: 10.1016/j.coi.2019.02.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/11/2019] [Accepted: 02/22/2019] [Indexed: 12/30/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease that causes most heart attacks and strokes, making it the biggest killer in the world. Although cholesterol-lowering drugs have dramatically reduced these major adverse cardiovascular events, there remains a high residual risk called inflammatory risk. Atherosclerosis has an autoimmune component that can be manipulated by immunologic approaches including vaccination. Vaccination is attractive, because it is antigen-specific, does not impair host defense, and provides long-term protection. Several candidate antigens for atherosclerosis vaccine development have been identified and have been shown to reduce atherosclerosis in animal models. In this review, we focus on two different types of atherosclerosis vaccines: antibody-inducing and regulatory T cell-inducing.
Collapse
|
129
|
Inflammatory parameters associated with systemic reactogenicity following vaccination with adjuvanted hepatitis B vaccines in humans. Vaccine 2019; 37:2004-2015. [PMID: 30850240 DOI: 10.1016/j.vaccine.2019.02.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/14/2018] [Accepted: 02/05/2019] [Indexed: 01/17/2023]
Abstract
BACKGROUND Adjuvants like AS01B increase the immunogenicity of vaccines and generally cause increased transient reactogenicity compared with Alum. A phase II randomized trial was conducted to characterize the response to AS01B and Alum adjuvanted vaccines. A post-hoc analysis was performed to examine the associations between reactogenicity and innate immune parameters. METHODS The trial involved 60 hepatitis B-naïve adults aged 18-45 years randomized 1:1 to receive either two doses of HBsAg-AS01B on Day (D)0 and D30, or three doses of HBsAg-Alum on D0, D30, D180. Prior to vaccination, all subjects received placebo injection in order to differentiate the impact of injection process and the vaccination. Main outcomes included reactogenicity symptoms, vital signs, blood cytokines, biochemical and hematological parameters after vaccination. Associations were explored using linear regression. FINDINGS The vaccine with AS01B induced higher HBsAg-specific antibody levels than Alum. Local and systemic symptoms were more frequent in individuals who received HBsAg AS01B/Alum vaccine or placebo, but were mild and short-lived. Blood levels of C-reactive protein (CRP), bilirubin, leukocyte, monocyte and neutrophil counts increased rapidly and transiently after AS01B but not after Alum or placebo. Lymphocyte counts decreased in the AS01B group and lactate dehydrogenase levels decreased after Alum. Modelling revealed associations between systemic symptoms and increased levels of CRP and IL-6 after the first HBsAg-AS01B or HBsAg-Alum immunization. Following the second vaccine dose, CRP, IL-6, IP-10, IFN-γ, MIP-1β and MCP-2 were identified as key parameters associated with systemic symptoms. These observations were confirmed using an independent data set extracted from a previous study of the immune response to HBsAg-adjuvanted vaccines (NCT00805389). CONCLUSIONS IL-6 and IFN-γ signals were associated with systemic reactogenicity following administration of AS01B-adjuvanted vaccine. These signals were similar to those previously associated with antibody and T-cell responses induced by HBsAg-adjuvanted vaccines, suggesting that similar innate immune signals may underlie adjuvant reactogenicity and immunogenicity. TRIAL REGISTRATION www.clinicaltrials.gov NCT01777295.
Collapse
|
130
|
Morris C, Glennie SJ, Lam HS, Baum HE, Kandage D, Williams NA, Morgan DJ, Woolfson DN, Davidson AD. A Modular Vaccine Platform Combining Self-Assembled Peptide Cages and Immunogenic Peptides. ADVANCED FUNCTIONAL MATERIALS 2019; 29:1807357. [PMID: 32313545 PMCID: PMC7161841 DOI: 10.1002/adfm.201807357] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/18/2018] [Indexed: 05/11/2023]
Abstract
Subunit vaccines use delivery platforms to present minimal antigenic components for immunization. The benefits of such systems include multivalency, self-adjuvanting properties, and more specific immune responses. Previously, the design, synthesis, and characterization of self-assembling peptide cages (SAGEs) have been reported. In these, de novo peptides are combined to make hubs that assemble into nanoparticles when mixed in aqueous solution. Here it is shown that SAGEs are nontoxic particles with potential as accessible synthetic peptide scaffolds for the delivery of immunogenic components. To this end, SAGEs functionalized with the model antigenic peptides tetanus toxoid632-651 and ovalbumin323-339 drive antigen-specific responses both in vitro and in vivo, eliciting both CD4+ T cell and B cell responses. Additionally, SAGEs functionalized with the antigenic peptide hemagglutinin518-526 from the influenza virus are also able to drive a CD8+ T cell response in vivo. This work demonstrates the potential of SAGEs to act as a modular scaffold for antigen delivery, capable of inducing and boosting specific and tailored immune responses.
Collapse
Affiliation(s)
- Caroline Morris
- BrisSynBioUniversity of BristolBristolBS8 1TQUK
- School of ChemistryUniversity of BristolBristolBS8 1TSUK
| | - Sarah J. Glennie
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Hon S. Lam
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Holly E. Baum
- BrisSynBioUniversity of BristolBristolBS8 1TQUK
- School of ChemistryUniversity of BristolBristolBS8 1TSUK
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Dhinushi Kandage
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Neil A. Williams
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - David J. Morgan
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| | - Derek N. Woolfson
- BrisSynBioUniversity of BristolBristolBS8 1TQUK
- School of ChemistryUniversity of BristolBristolBS8 1TSUK
- School of BiochemistryUniversity of BristolBristolBS8 1TDUK
| | - Andrew D. Davidson
- School of Cellular and Molecular MedicineUniversity of BristolBristolBS8 1TDUK
| |
Collapse
|
131
|
Ebensen T, Delandre S, Prochnow B, Guzmán CA, Schulze K. The Combination Vaccine Adjuvant System Alum/c-di-AMP Results in Quantitative and Qualitative Enhanced Immune Responses Post Immunization. Front Cell Infect Microbiol 2019; 9:31. [PMID: 30838180 DOI: 10.3389/fcimb.2019.00031] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 01/31/2019] [Indexed: 11/13/2022] Open
Abstract
The development of new effective vaccines strongly depends on adjuvants and formulations able to stimulate not only strong humoral responses against a certain pathogen but also effector as well as memory CD4+ and CD8+ T cells (Dubensky et al., 2013). However, the majority of vaccines licensed for human use or currently under clinical investigation fail to stimulate efficient cellular responses. For example, vaccines against hepatitis B virus (HBV), human papillomavirus (HPV), diphtheria, tetanus and influenza are usually administered by intramuscular (i.m.) injection and contain aluminum salts (alum) as adjuvant. Alum has been shown to stimulate Th2 immune cells resulting in increased production of antigen-specific antibodies but to be incapable of stimulating robust Th1 or cytotoxic responses. To overcome such limitations recent research has focused on the development of adjuvant combinations (e.g., MF59, AS03 or AS04) to not only further strengthen antigen-specific immune responses but to also allow their modulation. We have shown previously that bis-(3',5')-cyclic dimeric adenosine monophosphate (c-di-AMP) constitutes a promising adjuvant candidate stimulating both effective Th1/Th2 and cytotoxic immune responses when included in mucosal or parenteral vaccine formulations. In the present work we demonstrate that c-di-AMP can be also combined with other adjuvants like alum resulting in increases in not only humoral responses but more striking also in cellular immune responses. This leads to improved vaccine efficacy against intracellular pathogens.
Collapse
Affiliation(s)
- Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Simon Delandre
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Blair Prochnow
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
132
|
Immunotherapeutic Potential of Mollusk Hemocyanins in Combination with Human Vaccine Adjuvants in Murine Models of Oral Cancer. J Immunol Res 2019; 2019:7076942. [PMID: 30847353 PMCID: PMC6362480 DOI: 10.1155/2019/7076942] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 11/06/2018] [Indexed: 02/07/2023] Open
Abstract
Mollusk hemocyanins have been used for decades in immunological and clinical applications as natural, nontoxic, nonpathogenic, and nonspecific immunostimulants for the treatment of superficial bladder cancer, as carriers/adjuvants of tumor-associated antigens in cancer vaccine development and as adjuvants to dendritic cell-based immunotherapy, because these glycoproteins induce a bias towards Th1 immunity. Here, we analyzed the preclinical therapeutic potential of the traditional keyhole limpet hemocyanin (KLH) and two new hemocyanins from Concholepas concholepas (CCH) and Fissurella latimarginata (FLH) in mouse models of oral squamous cell carcinoma. Due to the aggressiveness and deadly malignant potential of this cancer, the hemocyanins were applied in combination with adjuvants, such as alum, AddaVax, and QS-21, which have been shown to be safe and effective in human vaccines, to potentiate their antitumor activity. The immunogenic performance of the hemocyanins in combination with the adjuvants was compared, and the best formulation was evaluated for its antitumor effects in two murine models of oral cancer: MOC7 cells implanted in the flank (heterotopic) and bioluminescent AT-84 E7 Luc cells implanted in the floor of the mouth (orthotopic). The results demonstrated that the hemocyanins in combination with QS-21 showed the greatest immunogenicity, as reflected by a robust, specific humoral response predominantly characterized by IgG2a antibodies and a sustained cellular response manifesting as a delayed hypersensitivity reaction. The KLH- and FLH-QS-21 formulations showed reduced tumor development and greater overall survival. Hemocyanins, as opposed to QS-21, had no cytotoxic effect on either oral cancer cell line cultured in vitro, supporting the idea that the antitumor effects of hemocyanins are associated with their modulation of the immune response. Therefore, hemocyanin utilization would allow a lower QS-21 dosage to achieve therapeutic results. Overall, our study opens a new door to further investigation of the use of hemocyanins plus adjuvants for the development of immunotherapies against oral carcinoma.
Collapse
|
133
|
Zhang R, Billingsley MM, Mitchell MJ. Biomaterials for vaccine-based cancer immunotherapy. J Control Release 2018; 292:256-276. [PMID: 30312721 PMCID: PMC6355332 DOI: 10.1016/j.jconrel.2018.10.008] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/06/2018] [Accepted: 10/08/2018] [Indexed: 12/28/2022]
Abstract
The development of therapeutic cancer vaccines as a means to generate immune reactivity against tumors has been explored since the early discovery of tumor-specific antigens by Georg Klein in the 1960s. However, challenges including weak immunogenicity, systemic toxicity, and off-target effects of cancer vaccines remain as barriers to their broad clinical translation. Advances in the design and implementation of biomaterials are now enabling enhanced efficacy and reduced toxicity of cancer vaccines by controlling the presentation and release of vaccine components to immune cells and their microenvironment. Here, we discuss the rational design and clinical status of several classes of cancer vaccines (including DNA, mRNA, peptide/protein, and cell-based vaccines) along with novel biomaterial-based delivery technologies that improve their safety and efficacy. Further, strategies for designing new platforms for personalized cancer vaccines are also considered.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Margaret M Billingsley
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, United States; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
134
|
Singha S, Shao K, Ellestad KK, Yang Y, Santamaria P. Nanoparticles for Immune Stimulation Against Infection, Cancer, and Autoimmunity. ACS NANO 2018; 12:10621-10635. [PMID: 30481968 DOI: 10.1021/acsnano.8b05950] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Vaccination using nanocarrier-based delivery systems has recently emerged as a promising approach for meeting the continued challenge posed by infectious diseases and cancer. A diverse portfolio of nanocarriers of various sizes, compositions, and physical parameters have now been developed, and this diversity provides an opportunity for the rational design of vaccines that can mediate targeted delivery of various antigens and adjuvants or immune regulatory agents in ways unachievable with classical vaccination approaches. This flexibility allows control over the characteristics of vaccine-elicited immune responses such that they can be tailored to be effective in circumstances where classical vaccines have failed. Furthermore, the utility of nanocarrier-based immune modulation extends to the treatment of autoimmune disease where precisely targeted inhibition of immune responses is desirable. Clearly, the selection of appropriate nanocarriers, antigens, adjuvants, and other components underpins the efficacy of these nanoimmune interventions. Herein, we provide an overview of currently available nanocarriers of various types and their physical and pharmacological properties with the goal of providing a resource for researchers exploring nanomaterial-based approaches for immune modulation and identify some information gaps and unexplored questions to help guide future investigation.
Collapse
Affiliation(s)
- Santiswarup Singha
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Kun Shao
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Kristofor K Ellestad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Yang Yang
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine , University of Calgary , Calgary , Alberta T2N 4N1 , Canada
- Institut D'Investigacions Biomèdiques August Pi i Sunyer , Barcelona 08036 , Spain
| |
Collapse
|
135
|
Chiodetti AL, Sánchez Vallecillo MF, Dolina JS, Crespo MI, Marin C, Schoenberger SP, Allemandi DA, Palma SD, Pistoresi-Palencia MC, Morón G, Maletto BA. Class-B CpG-ODN Formulated With a Nanostructure Induces Type I Interferons-Dependent and CD4 + T Cell-Independent CD8 + T-Cell Response Against Unconjugated Protein Antigen. Front Immunol 2018; 9:2319. [PMID: 30364187 PMCID: PMC6192457 DOI: 10.3389/fimmu.2018.02319] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022] Open
Abstract
There is a need for new vaccine adjuvant strategies that offer both vigorous antibody and T-cell mediated protection to combat difficult intracellular pathogens and cancer. To this aim, we formulated class-B synthetic oligodeoxynucleotide containing unmethylated cytosine-guanine motifs (CpG-ODN) with a nanostructure (Coa-ASC16 or coagel) formed by self-assembly of 6-0-ascorbyl palmitate ester. Our previous results demonstrated that mice immunized with ovalbumin (OVA) and CpG-ODN formulated with Coa-ASC16 (OVA/CpG-ODN/Coa-ASC16) elicited strong antibodies (IgG1 and IgG2a) and Th1/Th17 cellular responses without toxic systemic effects. These responses were superior to those induced by a solution of OVA with CpG-ODN or OVA/CpG-ODN formulated with aluminum salts. In this study, we investigated the capacity of this adjuvant strategy (CpG-ODN/Coa-ASC16) to elicit CD8+ T-cell response and some of the underlying cellular and molecular mechanisms involved in adaptive response. We also analyzed whether this adjuvant strategy allows a switch from an immunization scheme of three-doses to one of single-dose. Our results demonstrated that vaccination with OVA/CpG-ODN/Coa-ASC16 elicited an antigen-specific long-lasting humoral response and importantly-high quality CD8+ T-cell immunity with a single-dose immunization. Moreover, Coa-ASC16 promoted co-uptake of OVA and CpG-ODN by dendritic cells. The CD8+ T-cell response induced by OVA/CpG-ODN/Coa-ASC16 was dependent of type I interferons and independent of CD4+ T-cells, and showed polyfunctionality and efficiency against an intracellular pathogen. Furthermore, the cellular and humoral responses elicited by the nanostructured formulation were IL-6-independent. This system provides a simple and inexpensive adjuvant strategy with great potential for future rationally designed vaccines.
Collapse
Affiliation(s)
- Ana L. Chiodetti
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - María F. Sánchez Vallecillo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Joseph S. Dolina
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - María I. Crespo
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Constanza Marin
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Stephen P. Schoenberger
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Daniel A. Allemandi
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Unidad de Tecnología Farmacéutica, Córdoba, Argentina
| | - Santiago D. Palma
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Unidad de Tecnología Farmacéutica, Córdoba, Argentina
| | - María C. Pistoresi-Palencia
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Gabriel Morón
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| | - Belkys A. Maletto
- Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Centro de Investigaciones en Bioquímica Clínica e Inmunología, Córdoba, Argentina
| |
Collapse
|
136
|
Raman spectroscopy-based identification of toxoid vaccine products. NPJ Vaccines 2018; 3:50. [PMID: 30323957 PMCID: PMC6172244 DOI: 10.1038/s41541-018-0088-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/29/2018] [Accepted: 09/11/2018] [Indexed: 12/19/2022] Open
Abstract
Vaccines are complex biomedicines. Manufacturing is time consuming and requires a high level of quality control (QC) to guarantee consistent safety and potency. An increasing global demand has led to the need to reduce time and cost of manufacturing. The evolving concepts for QC and the upcoming threat of falsification of biomedicines define a new need for methods that allow the fast and reliable identification of vaccines. Raman spectroscopy is a non-destructive technology already established in QC of classical medicines. We hypothesized that Raman spectroscopy could be used for identification and differentiation of vaccine products. Raman maps obtained from air-dried samples of combination vaccines containing antigens from tetanus, diphtheria and pertussis (DTaP vaccines) were summarized to compile product-specific Raman signatures. Sources of technical variance were emphasized to evaluate the robustness and sensitivity in downstream data analysis. The data management approach corrects for spatial inhomogeneities in the dried sample while offering a proper representation of the original samples inherent chemical signature. Reproducibility of the identification was validated by a leave-one-replicate-out cross-validation. The results highlighted the high specificity and sensitivity of Raman measurements in identifying DTaP vaccine products. The results pave the way for further exploitation of the Raman technology for identification of vaccines in batch release and cases of suspected falsification. A light-based identification method offers a fast, reliable and non-destructive method to analyze vaccines. Using vaccines for tetanus, diphtheria, and pertussis, a German research team led by the Paul Ehrlich Institute and University of Jena and Leibniz-IPHT showed that Raman spectroscopy — which identifies substances based on how they scatter laser light — is able to identify distinctive signatures of vaccines. In their experiments, Raman spectroscopy was sensitive enough to detect subtle differences in vaccine formulation, such as the specific combination of vaccine and adjuvant components. However, a data analytics technique was required to correct for sample quality variation caused by their preparation. Raman spectroscopy already sees use in classical medicines, and its application to vaccines could help to reduce the time and cost of quality control while benefitting the unmet need for rapid analysis of vaccine quality and identity.
Collapse
|
137
|
Del Giudice G, Rappuoli R, Didierlaurent AM. Correlates of adjuvanticity: A review on adjuvants in licensed vaccines. Semin Immunol 2018; 39:14-21. [DOI: 10.1016/j.smim.2018.05.001] [Citation(s) in RCA: 327] [Impact Index Per Article: 46.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/30/2022]
|
138
|
Jain S, George PJ, Deng W, Koussa J, Parkhouse K, Hensley SE, Jiang J, Lu J, Liu Z, Wei J, Zhan B, Bottazzi ME, Shen H, Lustigman S. The parasite-derived rOv-ASP-1 is an effective antigen-sparing CD4 + T cell-dependent adjuvant for the trivalent inactivated influenza vaccine, and functions in the absence of MyD88 pathway. Vaccine 2018; 36:3650-3665. [PMID: 29764680 DOI: 10.1016/j.vaccine.2018.05.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 04/26/2018] [Accepted: 05/04/2018] [Indexed: 12/18/2022]
Abstract
Vaccination remains the most cost-effective biomedical approach for controlling influenza disease. In times of pandemics, however, these vaccines cannot be produced in sufficient quantities for worldwide use by the current manufacturing capacities and practices. What is needed is the development of adjuvanted vaccines capable of inducing an adequate or better immune response at a decreased antigen dose. Previously we showed that the protein adjuvant rOv-ASP-1 augments influenza-specific antibody titers and survival after virus challenge in both young adult and old-age mice when administered with the trivalent inactivated influenza vaccine (IIV3). In this study we show that a reduced amount of rOv-ASP-1, with 40-times less IIV3 can also induce protection. Apparently the potency of the rOv-ASP-1 adjuvanted IIV3 vaccine is independent of the IIV3-specific Th1/Th2 associated antibody responses, and independent of the presence of HAI antibodies. However, CD4+ T helper cells were indispensable for the protection. Further, rOv-ASP-1 with or without IIV3 elicited the increased level of various chemokines, which are known chemoattractant for immune cells, into the muscle 4 h after immunization, and significantly induced the recruitment of monocytes, macrophages and neutrophils into the muscles. The recruited monocytes had higher expression of the activation marker MHCII on their surface as well as CXCR3 and CCR2; receptors for IP-10 and MCP-1, respectively. These results show that the rOv-ASP-1 adjuvant allows substantial antigen sparing of IIV3 by stimulating at the site of injection the accumulation of chemokines and the recruitment of immune cells that can augment the activation of CD4+ T cell immune responses, essential for the production of antibody responses. Protection elicited by the rOv-ASP-1 adjuvanted IIV3 vaccine also appears to function in the absence of MyD88-signaling. Future studies will attempt to delineate the precise mechanisms by which the rOv-ASP-1 adjuvanted IIV3 vaccine works.
Collapse
Affiliation(s)
- Sonia Jain
- Laboratory of Molecular Parasitology, Lindsley F Kimball Research Institute, New York Blood Center, New York, NY 10065, United States
| | - Parakkal Jovvian George
- Laboratory of Molecular Parasitology, Lindsley F Kimball Research Institute, New York Blood Center, New York, NY 10065, United States
| | - Wanyan Deng
- Institute of Modern Biopharmaceuticals, School of Life Sciences, Southwest University, Chongqing 100045, China; Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Joseph Koussa
- Center for Genomics and Systems Biology, Department of Biology, New York University, New York, NY 10003, United States; Department of Biology, New York University Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Kaela Parkhouse
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Jiu Jiang
- Department of Biology, Drexel University, Philadelphia, PA 19104, United States
| | - Jie Lu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing 400715, China
| | - Zhuyun Liu
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatric Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, United States
| | - Junfei Wei
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatric Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, United States
| | - Bin Zhan
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatric Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, United States
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Department of Pediatric Tropical Medicine, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, United States
| | - Hao Shen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Sara Lustigman
- Laboratory of Molecular Parasitology, Lindsley F Kimball Research Institute, New York Blood Center, New York, NY 10065, United States.
| |
Collapse
|
139
|
Gómez L, Alvarez F, Betancur D, Oñate A. Brucellosis vaccines based on the open reading frames from genomic island 3 of Brucella abortus. Vaccine 2018; 36:2928-2936. [PMID: 29685597 DOI: 10.1016/j.vaccine.2018.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/02/2018] [Accepted: 04/04/2018] [Indexed: 01/18/2023]
Abstract
Brucella abortus is the etiological agent of brucellosis, a zoonotic disease affecting cattle and humans. This disease has been partially controlled in cattle by immunization with live attenuated B. abortus S19 and RB51 strains. However, use of these vaccine strains has been associated with safety issues in animals and humans. New vaccines have since emerged in the prevention of brucellosis, particularly DNA vaccines, which have shown effectiveness and a good safety profile. Their protection efficacy in mice is associated with the induction of Th1 type and cytotoxic T cell mediated immune response against structural antigens and virulence factors expressed during B. abortus infection. Some antigenic candidate for vaccine design against brucellosis (mainly DNA vaccines) have been obtained from genomic island 3 (GI-3) of B. abortus, which encodes several open reading frames (ORFs) involved in the intracellular survival and virulence of this pathogen. The immunogenicity and protection conferred by these DNA vaccines in a murine model is reviewed in this article, suggesting that some of them could be safe and effective vaccine candidates against to prevent B. abortus infection.
Collapse
Affiliation(s)
- Leonardo Gómez
- Laboratory of Molecular Immunology, Faculty of Biological Sciences, Department of Microbiology, University of Concepción, Concepción, Chile
| | - Francisco Alvarez
- Laboratory of Molecular Immunology, Faculty of Biological Sciences, Department of Microbiology, University of Concepción, Concepción, Chile
| | - Daniel Betancur
- Laboratory of Molecular Immunology, Faculty of Biological Sciences, Department of Microbiology, University of Concepción, Concepción, Chile
| | - Angel Oñate
- Laboratory of Molecular Immunology, Faculty of Biological Sciences, Department of Microbiology, University of Concepción, Concepción, Chile.
| |
Collapse
|
140
|
DNA-loaded nano-adjuvant formed with a vitamin E-scaffold intracellular environmentally-responsive lipid-like material for cancer immunotherapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:2587-2597. [PMID: 30170077 DOI: 10.1016/j.nano.2018.08.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/26/2018] [Accepted: 08/14/2018] [Indexed: 12/30/2022]
Abstract
Cytoplasmic DNA triggers cellular immunity via activating the stimulator of interferon genes pathway. Since DNA is degradable and membrane impermeable, delivery system would permit cytoplasmic delivery by destabilizing the endosomal membrane for the use as an adjuvant. Herein, we report on the development of a plasmid DNA (pDNA)-encapsulating lipid nanoparticle (LNP). The structural components include an SS-cleavable and pH-activated lipid-like material that mounts vitamin E as a hydrophobic scaffold, and dual sensing motifs that are responsive to the intracellular environment (ssPalmE). The pDNA-encapsulating LNP (ssPalmE-LNP) induced a high interferon-β production in Raw 264.7 cells. The subcutaneous injection of ssPalmE-LNP strongly enhanced antigen-specific cytotoxic T cell activity. The ssPalmE-LNP treatment efficiently induced antitumor effects against E.G7-OVA tumor and B16-F10 melanoma metastasis. Furthermore, when combined with an anti-programmed death 1 antibody, an extensive therapeutic antitumor effect was observed. Therefore, the ssPalmE-LNP is a promising carrier of adjuvants for cancer immunotherapy.
Collapse
|
141
|
The novel immunogenic chimeric peptide vaccine to elicit potent cellular and mucosal immune responses against HTLV-1. Int J Pharm 2018; 549:404-414. [PMID: 30075250 DOI: 10.1016/j.ijpharm.2018.07.069] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/28/2018] [Accepted: 07/30/2018] [Indexed: 12/15/2022]
Abstract
This study reports on the immunogenicity assessment of a novel chimeric peptide vaccine including Tax, gp21, gp46, and gag immunodominant epitopes of human T-cell lymphotropic virus type 1 (HTLV-1) to induce immunity against HTLV-1 after subcutaneous (SC) or intranasal administration in a mice model. Additionally, to elevate the efficacy of the HTLV-1 vaccine, the chimera was physically mixed with monophosphoryl lipid A (MPLA) or ISCOMATRIX (IMX) adjuvants. For this purpose, the ISCOMATRIX with a size range of 40-60 nm were prepared using lipid film hydration method. Our investigation revealed that the mixture of IMX and chimera could significantly increase antibody titers containing IgG2a, and mucosal IgA, as well as IFN-γ and IL-10 cytokines and decrease the level of TGF-β1, compared to other vaccine formulations. The intranasal delivery of chimera vaccine in the absence or presence adjuvants stimulated potent mucosal sIgA titer relative to subcutaneous immunization. Furthermore, the SC or nasal delivery of various vaccine formulations could shift the immunity toward cell-mediated responses, as evident by higher IgG2a and IFN-γ, as well as suppressed TGF-β1 level. Our findings suggest that proper design, construction, and immunization of multi-epitope vaccine are essential for developing an effective HTLV-1 vaccine.
Collapse
|
142
|
Activation of the endoplasmic reticulum stress sensor IRE1α by the vaccine adjuvant AS03 contributes to its immunostimulatory properties. NPJ Vaccines 2018; 3:20. [PMID: 29977610 PMCID: PMC6023910 DOI: 10.1038/s41541-018-0058-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/24/2018] [Accepted: 05/01/2018] [Indexed: 01/31/2023] Open
Abstract
The oil-in-water emulsion Adjuvant System 03 (AS03) is one of the few adjuvants used in licensed vaccines. Previous work indicates that AS03 induces a local and transient inflammatory response that contributes to its adjuvant effect. However, the molecular mechanisms involved in its immunostimulatory properties are ill-defined. Upon intramuscular injection in mice, AS03 elicited a rapid and transient downregulation of lipid metabolism-related genes in the draining lymph node. In vitro, these modifications were associated with profound changes in lipid composition, alteration of endoplasmic reticulum (ER) morphology and activation of the unfolded protein response pathway. In vivo, treatment with a chemical chaperone or deletion of the ER stress sensor kinase IRE1α in myeloid cells decreased AS03-induced cytokine production and its capacity to elicit high affinity antigen-specific antibodies. In summary, our results indicate that IRE1α is a sensor for the metabolic changes induced by AS03 in monocytic cells and may constitute a canonical pathway that could be exploited for the design of novel vaccine adjuvants.
Collapse
|
143
|
Fisher DG, Coppock GM, López CB. Virus-derived immunostimulatory RNA induces type I IFN-dependent antibodies and T-cell responses during vaccination. Vaccine 2018; 36:4039-4045. [PMID: 29861183 PMCID: PMC6265594 DOI: 10.1016/j.vaccine.2018.05.100] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 05/17/2018] [Accepted: 05/25/2018] [Indexed: 01/04/2023]
Abstract
Adjuvants potentiate and direct the type of immunity elicited during vaccination. However, there is a shortage of adjuvants that elicit robust type-1 immunity required for the control of intracellular pathogens, including viruses. RNA derived from Sendai virus defective viral genomes (DVGs) stimulates RIG-I-like receptor signaling leading to type-1 immunity during infection. Here, we investigated whether a 268nt DVG-derived oligonucleotide (DDO) functions as a strong type-1 immunity-inducing adjuvant during vaccination against influenza virus. We show that DDO induces robust IgG2c antibody production when used in an inactivated influenza A virus (IAV) vaccine. Additionally, DDO induces Th1 and CD8+ T-cell responses able to protect against heterosubtypic IAV challenge. Interestingly, DDO synergized with AddaVax and skewed the immune response towards type-1 immunity. The adjuvancy of DDO alone and in synergy with AddaVax was heavily dependent on type I interferon signaling. Our data support a critical role for type I interferon in the induction of type-1 immune responses during vaccination and demonstrate that DDO is a type-1 immunity orienting vaccine adjuvant that can be used alone or in synergy with other adjuvants.
Collapse
Affiliation(s)
- Devin G Fisher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Gaia M Coppock
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Carolina B López
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States.
| |
Collapse
|
144
|
Abstract
The inability to elicit strong and durable cellular responses is a major obstacle in the development of successful vaccines, in particular those against malaria. In this regard, the generation of novel adjuvants that will potently boost cell-mediated immunity induced by candidate vaccines is helpful. We and others have found a glycolipid, called α-galactosylceramide (α-GalCer), which could be presented on CD1d expressed by antigen-presenting cells (APCs) and stimulate natural killer T (NKT) cells. This triggers the activation/maturation of APCs, particularly dendritic cells (DCs). By activating NKT cells and subsequently DCs, α-GalCer has been shown to enhance adaptive immune responses, particularly of CD8
+ T cells, induced by the vaccines. More recently, we identified an analogue of α-GalCer, which can display a potent adjuvant activity in conjunction with malaria vaccines in mice and non-human primates. It is anticipated that CD1d-binding, NKT cell-stimulating glycolipids will be tested as adjuvants in humans in the near future.
Collapse
Affiliation(s)
- Jordana Grazziela Coelho-Dos-Reis
- Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brazil.,Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY, USA
| | - Xiangming Li
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY, USA.,Sanofi, Cambridge, MA, USA
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, NY, USA
| |
Collapse
|
145
|
Latest development on RNA-based drugs and vaccines. Future Sci OA 2018; 4:FSO300. [PMID: 29796303 PMCID: PMC5961404 DOI: 10.4155/fsoa-2017-0151] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/19/2018] [Indexed: 12/25/2022] Open
Abstract
Drugs and vaccines based on mRNA and RNA viruses show great potential and direct translation in the cytoplasm eliminates chromosomal integration. Limitations are associated with delivery and stability issues related to RNA degradation. Clinical trials on RNA-based drugs have been conducted in various disease areas. Likewise, RNA-based vaccines for viral infections and various cancers have been subjected to preclinical and clinical studies. RNA delivery and stability improvements include RNA structure modifications, targeting dendritic cells and employing self-amplifying RNA. Single-stranded RNA viruses possess self-amplifying RNA, which can provide extreme RNA replication in the cytoplasm to support RNA-based drug and vaccine development. Although oligonucleotide-based approaches have demonstrated potential, the focus here is on mRNA- and RNA virus-based methods. Drug development has suffered from inefficiency, side effects and high costs. For this reason novel approaches for drug discovery are of great importance. RNA-based methods provide the advantage of targeting ‘production’ of drugs to diseased cells and vaccines to immune response-stimulating cells. RNA drugs have demonstrated therapeutic efficacy in eye and heart diseases and in various cancers in clinical trials. Likewise, RNA-based vaccines have provided protection against challenges with lethal doses of viruses such as Ebola and cancer cells in animal models.
Collapse
|
146
|
Muñoz-Wolf N, Lavelle EC. A Guide to IL-1 family cytokines in adjuvanticity. FEBS J 2018; 285:2377-2401. [PMID: 29656546 DOI: 10.1111/febs.14467] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 03/21/2018] [Accepted: 04/04/2018] [Indexed: 12/16/2022]
Abstract
Growing awareness of the multiplicity of roles for the IL-1 family in immune regulation has prompted research exploring these cytokines in the context of vaccine-induced immunity. While tightly regulated, cytokines of the IL-1 family are normally released in response to cellular stress and in combination with other danger-/damage-associated molecular patterns (DAMPs), triggering potent local and systemic immune responses. In the context of infection or autoimmunity, engagement of IL-1 family receptors links robust innate responses to adaptive immunity. Clinical and experimental evidence has revealed that many vaccine adjuvants induce the release of one or multiple IL-1 family cytokines. The coordinated release of IL-1 family members in response to adjuvant-induced damage or cell death may be a determining factor in the transition from local inflammation to the induction of an adaptive response. Here, we analyse the effects of IL-1 family cytokines on innate and adaptive immunity with a particular emphasis on activation of antigen-presenting cells and induction of T cell-mediated immunity, and we address in detail the contribution of these cytokines to the modes of action of vaccine adjuvants including those currently approved for human use.
Collapse
Affiliation(s)
- Natalia Muñoz-Wolf
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland.,Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Advanced Materials and BioEngineering Research (AMBER), Trinity College Dublin, Ireland
| |
Collapse
|
147
|
Laser Adjuvant-Assisted Peptide Vaccine Promotes Skin Mobilization of Dendritic Cells and Enhances Protective CD8 + T EM and T RM Cell Responses against Herpesvirus Infection and Disease. J Virol 2018; 92:JVI.02156-17. [PMID: 29437976 DOI: 10.1128/jvi.02156-17] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 02/01/2018] [Indexed: 01/14/2023] Open
Abstract
There is an urgent need for chemical-free and biological-free safe adjuvants to enhance the immunogenicity of vaccines against widespread viral pathogens, such as herpes simplex virus 2 (HSV-2), that infect a large proportion of the world human population. In the present study, we investigated the safety, immunogenicity, and protective efficacy of a laser adjuvant-assisted peptide (LAP) vaccine in the B6 mouse model of genital herpes. This LAP vaccine and its laser-free peptide (LFP) vaccine analog contain the immunodominant HSV-2 glycoprotein B CD8+ T cell epitope (HSV-gB498-505) covalently linked with the promiscuous glycoprotein D CD4+ T helper cell epitope (HSV-gD49-89). Prior to intradermal delivery of the LAP vaccine, the lower-flank shaved skin of B6 or CD11c/eYFP transgenic mice received a topical skin treatment with 5% imiquimod cream and then was exposed for 60 s to a laser, using the FDA-approved nonablative diode. Compared to the LFP vaccine, the LAP vaccine (i) triggered mobilization of dendritic cells (DCs) in the skin, which formed small spots along the laser-treated areas, (ii) induced phenotypic and functional maturation of DCs, (iii) stimulated long-lasting HSV-specific effector memory CD8+ T cells (TEM cells) and tissue-resident CD8+ T cells (TRM cells) locally in the vaginal mucocutaneous tissues (VM), and (iv) induced protective immunity against genital herpes infection and disease. As an alternative to currently used conventional adjuvants, the chemical- and biological-free laser adjuvant offers a well-tolerated, simple-to-produce method to enhance mass vaccination for widespread viral infections.IMPORTANCE Herpes simplex viruses 1 and 2 (HSV-1 and HSV-2) infect a large proportion of the world population. There is an urgent need for chemical-free and biological-free safe adjuvants that would advance mass vaccination against the widespread herpes infections. The present study demonstrates that immunization with a laser-assisted herpes peptide vaccine triggered skin mobilization of dendritic cells (DCs) that stimulated strong and long-lasting HSV-specific effector memory CD8+ T cells (TEM cells) and tissue-resident CD8+ T cells (TRM cells) locally in the vaginal mucocutaneous tissues. The induced local CD8+ T cell response was associated with protection against genital herpes infection and disease. These results draw attention to chemical- and biological-free laser adjuvants as alternatives to currently used conventional adjuvants to enhance mass vaccination for widespread viral infections, such as those caused by HSV-1 and HSV-2.
Collapse
|
148
|
New Technologies for Vaccine Development: Harnessing the Power of Human Immunology. J Indian Inst Sci 2018. [DOI: 10.1007/s41745-018-0064-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
149
|
Chen WC, Murawsky CM. Strategies for Generating Diverse Antibody Repertoires Using Transgenic Animals Expressing Human Antibodies. Front Immunol 2018; 9:460. [PMID: 29563917 PMCID: PMC5845867 DOI: 10.3389/fimmu.2018.00460] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 02/21/2018] [Indexed: 01/14/2023] Open
Abstract
Therapeutic molecules derived from antibodies have become a dominant class of drugs used to treat human disease. Increasingly, therapeutic antibodies are discovered using transgenic animal systems that have been engineered to express human antibodies. While the engineering details differ, these platforms share the ability to raise an immune response that is comprised of antibodies with fully human idiotypes. Although the predominant transgenic host species has been mouse, the genomes of rats, rabbits, chickens, and cows have also been modified to express human antibodies. The creation of transgenic animal platforms expressing human antibody repertoires has revolutionized therapeutic antibody drug discovery. The observation that the immune systems of these animals are able to recognize and respond to a wide range of therapeutically relevant human targets has led to a surge in antibody-derived drugs in current development. While the clinical success of fully human monoclonal antibodies derived from transgenic animals is well established, recent trends have seen increasingly stringent functional design goals and a shift in difficulty as the industry attempts to tackle the next generation of disease-associated targets. These challenges have been met with a number of novel approaches focused on the generation of large, high-quality, and diverse antibody repertoires. In this perspective, we describe some of the strategies and considerations we use for manipulating the immune systems of transgenic animal platforms (such as XenoMouse®) with a focus on maximizing the diversity of the primary response and steering the ensuing antibody repertoire toward a desired outcome.
Collapse
Affiliation(s)
- Weihsu C Chen
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, BC, Canada
| | - Christopher M Murawsky
- Biologics Discovery, Department of Therapeutic Discovery, Amgen British Columbia Inc., Burnaby, BC, Canada
| |
Collapse
|
150
|
Corthésy B, Bioley G. Lipid-Based Particles: Versatile Delivery Systems for Mucosal Vaccination against Infection. Front Immunol 2018; 9:431. [PMID: 29563912 PMCID: PMC5845866 DOI: 10.3389/fimmu.2018.00431] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/19/2018] [Indexed: 12/19/2022] Open
Abstract
Vaccination is the process of administering immunogenic formulations in order to induce or harness antigen (Ag)-specific antibody and T cell responses in order to protect against infections. Important successes have been obtained in protecting individuals against many deleterious pathological situations after parenteral vaccination. However, one of the major limitations of the current vaccination strategies is the administration route that may not be optimal for the induction of immunity at the site of pathogen entry, i.e., mucosal surfaces. It is now well documented that immune responses along the genital, respiratory, or gastrointestinal tracts have to be elicited locally to ensure efficient trafficking of effector and memory B and T cells to mucosal tissues. Moreover, needle-free mucosal delivery of vaccines is advantageous in terms of safety, compliance, and ease of administration. However, the quest for mucosal vaccines is challenging due to (1) the fact that Ag sampling has to be performed across the epithelium through a relatively limited number of portals of entry; (2) the deleterious acidic and proteolytic environment of the mucosae that affect the stability, integrity, and retention time of the applied Ags; and (3) the tolerogenic environment of mucosae, which requires the addition of adjuvants to elicit efficient effector immune responses. Until now, only few mucosally applicable vaccine formulations have been developed and successfully tested. In animal models and clinical trials, the use of lipidic structures such as liposomes, virosomes, immune stimulating complexes, gas-filled microbubbles and emulsions has proven efficient for the mucosal delivery of associated Ags and the induction of local and systemic immune reponses. Such particles are suitable for mucosal delivery because they protect the associated payload from degradation and deliver concentrated amounts of Ags via specialized sampling cells (microfold cells) within the mucosal epithelium to underlying antigen-presenting cells. The review aims at summarizing recent development in the field of mucosal vaccination using lipid-based particles. The modularity ensured by tailoring the lipidic design and content of particles, and their known safety as already established in humans, make the continuing appraisal of these vaccine candidates a promising development in the field of targeted mucosal vaccination.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Gilles Bioley
- R&D Laboratory, Division of Immunology and Allergy, Centre des Laboratoires d'Epalinges, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|