1
|
Joffe A. The Promise and Predicament of Combining Adjuvants in Vaccines. VACCINE INSIGHTS 2025; 4:27-30. [PMID: 40051535 PMCID: PMC11884258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Subscribe] [Scholar Register] [Indexed: 03/09/2025]
Affiliation(s)
- Ari Joffe
- Division of Allergy, Immunology, and Transplantation, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
2
|
Pan JM, Liang Y, Zhu KC, Guo HY, Liu BS, Zhang N, Zhang DC. Identification of the NOD-like receptor family of golden pompano and expression in response to bacterial and parasitic exposure reveal its key role in innate immunity. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 152:105123. [PMID: 38135022 DOI: 10.1016/j.dci.2023.105123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/01/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
This study presents a genome-wide identification of NOD-like receptors (NLRs) in the golden pompano, key to its innate immunity. We identified 30 ToNLRs, analyzing their chromosomal positions, characteristics, evolutionary relationships, evidence of positive selection, and synteny with the yellowtail kingfish. Our findings categorize these NLRs into three main subgroups: NLRA, NLRC, and the distinct ToNLRX1. Post-exposure to Streptococcus agalactiae, most ToNLRs increased expression in the spleen, whereas NLRC3like13, NLRC3like16, and NLRC3like19 so in the kidneys. Upon Cryptocaryon irritans exposure, we categorized our groups based on the site of infection into the control group (BFS), the trophont-attached skin (TAS), and the nearby region skin (NRS). ToAPAF1 and ToNOD1 expressions rose in the NRS, in contrast to decreased expressions of ToNLRC5, ToNWD1 and ToCIITA. Other ToNLRs showed variable expressions in the TAS. Overall, this research lays the groundwork for further exploration of innate immunity in the golden pompano.
Collapse
Affiliation(s)
- Jin-Min Pan
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, Guangdong Province, PR China
| | - Yu Liang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, Guangdong Province, PR China
| | - Ke-Cheng Zhu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, Guangdong Province, PR China; Sanya Tropical Fisheries Research Institute, Sanya, 572018, PR China; Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, PR China
| | - Hua-Yang Guo
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, Guangdong Province, PR China; Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, PR China
| | - Bao-Suo Liu
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, Guangdong Province, PR China; Sanya Tropical Fisheries Research Institute, Sanya, 572018, PR China; Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, PR China
| | - Nan Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, Guangdong Province, PR China; Sanya Tropical Fisheries Research Institute, Sanya, 572018, PR China; Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, PR China
| | - Dian-Chang Zhang
- Key Laboratory of South China Sea Fishery Resources Exploitation and Utilization, Ministry of Agriculture and Rural Affairs, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, 510300, Guangzhou, Guangdong Province, PR China; Sanya Tropical Fisheries Research Institute, Sanya, 572018, PR China; Guangdong Provincial Engineer Technology Research Center of Marine Biological Seed Industry, Guangzhou, Guangdong Province, PR China.
| |
Collapse
|
3
|
Zhao T, Cai Y, Jiang Y, He X, Wei Y, Yu Y, Tian X. Vaccine adjuvants: mechanisms and platforms. Signal Transduct Target Ther 2023; 8:283. [PMID: 37468460 PMCID: PMC10356842 DOI: 10.1038/s41392-023-01557-7] [Citation(s) in RCA: 236] [Impact Index Per Article: 118.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/21/2023] Open
Abstract
Adjuvants are indispensable components of vaccines. Despite being widely used in vaccines, their action mechanisms are not yet clear. With a greater understanding of the mechanisms by which the innate immune response controls the antigen-specific response, the adjuvants' action mechanisms are beginning to be elucidated. Adjuvants can be categorized as immunostimulants and delivery systems. Immunostimulants are danger signal molecules that lead to the maturation and activation of antigen-presenting cells (APCs) by targeting Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs) to promote the production of antigen signals and co-stimulatory signals, which in turn enhance the adaptive immune responses. On the other hand, delivery systems are carrier materials that facilitate antigen presentation by prolonging the bioavailability of the loaded antigens, as well as targeting antigens to lymph nodes or APCs. The adjuvants' action mechanisms are systematically summarized at the beginning of this review. This is followed by an introduction of the mechanisms, properties, and progress of classical vaccine adjuvants. Furthermore, since some of the adjuvants under investigation exhibit greater immune activation potency than classical adjuvants, which could compensate for the deficiencies of classical adjuvants, a summary of the adjuvant platforms under investigation is subsequently presented. Notably, we highlight the different action mechanisms and immunological properties of these adjuvant platforms, which will provide a wide range of options for the rational design of different vaccines. On this basis, this review points out the development prospects of vaccine adjuvants and the problems that should be paid attention to in the future.
Collapse
Affiliation(s)
- Tingmei Zhao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yulong Cai
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yujie Jiang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yifan Yu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
4
|
Singleton KL, Joffe A, Leitner WW. Review: Current trends, challenges, and success stories in adjuvant research. Front Immunol 2023; 14:1105655. [PMID: 36742311 PMCID: PMC9892189 DOI: 10.3389/fimmu.2023.1105655] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 01/04/2023] [Indexed: 01/20/2023] Open
Abstract
Vaccine adjuvant research is being fueled and driven by progress in the field of innate immunity that has significantly advanced in the past two decades with the discovery of countless innate immune receptors and innate immune pathways. Receptors for pathogen-associated molecules (PAMPs) or host-derived, danger-associated molecules (DAMPs), as well as molecules in the signaling pathways used by such receptors, are a rich source of potential targets for agonists that enable the tuning of innate immune responses in an unprecedented manner. Targeted modulation of immune responses is achieved not only through the choice of immunostimulator - or select combinations of adjuvants - but also through formulation and systematic modifications of the chemical structure of immunostimulatory molecules. The use of medium and high-throughput screening methods for finding immunostimulators has further accelerated the identification of promising novel adjuvants. However, despite the progress that has been made in finding new adjuvants through systematic screening campaigns, the process is far from perfect. A major bottleneck that significantly slows the process of turning confirmed or putative innate immune receptor agonists into vaccine adjuvants continues to be the lack of defined in vitro correlates of in vivo adjuvanticity. This brief review discusses recent developments, exciting trends, and notable successes in the adjuvant research field, albeit acknowledging challenges and areas for improvement.
Collapse
|
5
|
Abstract
Self-adjuvanting vaccines, covalent conjugates between antigens and adjuvants, are chemically well-defined compared with conventional vaccines formulated through mixing antigens with adjuvants. Innate immune receptor ligands effectively induce acquired immunity through the activation of innate immunity, thereby enhancing host immune responses. Thus, innate immune receptor ligands are often used as adjuvants in self-adjuvanting vaccines. In a self-adjuvanting vaccine, the covalent linkage of antigen and adjuvant enables their simultaneous uptake into immune cells where the adjuvant consequently induces antigen-specific immune responses. Importantly, self-adjuvanting vaccines do not require immobilization to carrier proteins or co-administration of additional adjuvants and thus avoid inducing undesired immune responses. Because of these excellent properties, self-adjuvanting vaccines are expected to be candidates for next-generation vaccines. Here, we take an overview of vaccine adjuvants, mainly focusing on those utilized in self-adjuvanting vaccines and then we review recent reports on self-adjuvanting conjugate vaccines.
Collapse
|
6
|
TLR2 agonistic lipopeptide enriched PLGA nanoparticles as combinatorial drug delivery vehicle. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
7
|
Kaur A, Kanwar R, Kaushik D, Sakala IG, Honda-Okubo Y, Petrovsky N, Salunke DB, Mehta SK. Combined delivery of TLR2 and TLR7 agonists by Nanostructured lipid carriers induces potent vaccine adjuvant activity in mice. Int J Pharm 2021; 613:121378. [PMID: 34915144 DOI: 10.1016/j.ijpharm.2021.121378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/12/2021] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
Toll-like receptor (TLR) agonists are promising adjuvants and the combination of TLR agonists enhance immune responses by providing synergistic immune activity via triggering different signalling pathways. However, systematic cytotoxicity due to the immediate release of such immune potentiators from the site of injection hampers its clinical performance. Nanostructured lipid carriers (NLCs) offer a possibility to incorporate multiple TLR agonists with high encapsulation efficiency and slow drug release. Herein, we synthesized NLCs from didodecyldimethylammonium bromide (D12DAB) and oleic acid and used these to co-encapsulate a Pam2CS derivative (T-2, TLR2 agonist) with an imidazoquinoline derivative (T-7, TLR7 agonist) as a combination vaccine adjuvant. Hydrodynamic diameter and zeta potential of the prepared NLCs were found to be in the range of 200-500 nm and 23-27 mV, respectively. Spherical shape and size of prepared NLCs were also assessed through Field Emission Scanning Electron Microscopy (FE-SEM) and Transmission Electron Microscopy (TEM) analysis. In-vitro release studies of T-7 demonstrated sustained release and the addition of lipopeptide T-2 augmented encapsulation efficiency (from 84 to 92.9%) with a slight trigger in the release percentage. All NLC formulations were screened in TLR2/1, TLR2/6, TLR7 and TLR8 reporter cell lines and loaded NLC formulation showed high TLR2 and TLR7 agonistic activity. Adjuvant potency was evaluated through intramuscular immunization of female C57BL/6 mice with recombinant hepatitis B surface antigen and influenza hemagglutinin protein. T-2 and T-7 loaded NLCs induced good protective efficacy in mice challenged with a lethal dose of influenza virus.
Collapse
Affiliation(s)
- Arshpreet Kaur
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India
| | - Rohini Kanwar
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; Mehr Chand Mahajan DAV College for Women, Chandigarh, India
| | - Deepender Kaushik
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India
| | - Isaac G Sakala
- Vaxine Pty Ltd, Warradale, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Yoshikazu Honda-Okubo
- Vaxine Pty Ltd, Warradale, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Warradale, Australia; College of Medicine and Public Health, Flinders University, Adelaide, Australia.
| | - Deepak B Salunke
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India; National Interdisciplinary Centre of Vaccines, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh, India.
| | - Surinder K Mehta
- Department of Chemistry and Centre for Advanced Studies, Panjab University, Chandigarh, India.
| |
Collapse
|
8
|
Bencze D, Fekete T, Pázmándi K. Type I Interferon Production of Plasmacytoid Dendritic Cells under Control. Int J Mol Sci 2021; 22:ijms22084190. [PMID: 33919546 PMCID: PMC8072550 DOI: 10.3390/ijms22084190] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
One of the most powerful and multifaceted cytokines produced by immune cells are type I interferons (IFNs), the basal secretion of which contributes to the maintenance of immune homeostasis, while their activation-induced production is essential to effective immune responses. Although, each cell is capable of producing type I IFNs, plasmacytoid dendritic cells (pDCs) possess a unique ability to rapidly produce large amounts of them. Importantly, type I IFNs have a prominent role in the pathomechanism of various pDC-associated diseases. Deficiency in type I IFN production increases the risk of more severe viral infections and the development of certain allergic reactions, and supports tumor resistance; nevertheless, its overproduction promotes autoimmune reactions. Therefore, the tight regulation of type I IFN responses of pDCs is essential to maintain an adequate level of immune response without causing adverse effects. Here, our goal was to summarize those endogenous factors that can influence the type I IFN responses of pDCs, and thus might serve as possible therapeutic targets in pDC-associated diseases. Furthermore, we briefly discuss the current therapeutic approaches targeting the pDC-type I IFN axis in viral infections, cancer, autoimmunity, and allergy, together with their limitations defined by the Janus-faced nature of pDC-derived type I IFNs.
Collapse
Affiliation(s)
- Dóra Bencze
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary; (D.B.); (T.F.)
- Doctoral School of Molecular Cell and Immune Biology, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary
| | - Tünde Fekete
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary; (D.B.); (T.F.)
| | - Kitti Pázmándi
- Department of Immunology, Faculty of Medicine, University of Debrecen, 1 Egyetem Square, H-4032 Debrecen, Hungary; (D.B.); (T.F.)
- Correspondence: ; Tel./Fax: +36-52-417-159
| |
Collapse
|
9
|
Fukase K. Glycoconjugates for Adjuvants and Self-Adjuvanting Vaccines. COMPREHENSIVE GLYCOSCIENCE 2021:166-184. [DOI: 10.1016/b978-0-12-819475-1.00099-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
10
|
Kaur A, Kaushik D, Piplani S, Mehta SK, Petrovsky N, Salunke DB. TLR2 Agonistic Small Molecules: Detailed Structure-Activity Relationship, Applications, and Future Prospects. J Med Chem 2020; 64:233-278. [PMID: 33346636 DOI: 10.1021/acs.jmedchem.0c01627] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) are the pattern recognition receptors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs) in microbial species. Among the various TLRs, TLR2 has a special place due to its ability to sense the widest repertoire of PAMPs owing to its heterodimerization with either TLR1 or TLR6, broadening its ligand diversity against pathogens. Various scaffolds are reported to activate TLR2, which include naturally occurring lipoproteins, synthetic lipopeptides, and small heterocyclic molecules. We described a detailed SAR in TLR2 agonistic scaffolds and also covered the design and chemistry for the conjugation of TLR2 agonists to antigens, carbohydrates, polymers, and fluorophores. The approaches involved in delivery of TLR2 agonists such as lipidation of antigen, conjugation to polymers, phosphonic acids, and other linkers to achieve surface adsorption, liposomal formulation, and encapsulating nanoparticles are elaborated. The crystal structure analysis and computational modeling are also included with the structural features that facilitate TLR2 activation.
Collapse
Affiliation(s)
- Arshpreet Kaur
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Deepender Kaushik
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Sakshi Piplani
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale, Australia 5046.,College of Medicine and Public Health, Flinders University, Bedford Park, Australia, 5042
| | - Surinder K Mehta
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, 11 Walkley Avenue, Warradale, Australia 5046.,College of Medicine and Public Health, Flinders University, Bedford Park, Australia, 5042
| | - Deepak B Salunke
- Department of Chemistry and Centre of Advanced Studies in Chemistry, Panjab University, Chandigarh 160014, India.,National Interdisciplinary Centre of Vaccine, Immunotherapeutics and Antimicrobials, Panjab University, Chandigarh 160014, India
| |
Collapse
|
11
|
Manabe Y, Chang TC, Fukase K. Recent advances in self-adjuvanting glycoconjugate vaccines. DRUG DISCOVERY TODAY. TECHNOLOGIES 2020; 37:61-71. [PMID: 34895656 DOI: 10.1016/j.ddtec.2020.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/21/2020] [Accepted: 11/26/2020] [Indexed: 01/02/2023]
Abstract
Compared to traditional vaccines that are formulated into mixtures of an adjuvant and an antigen, a self-adjuvanting vaccine consists of an antigen that is covalently conjugated to a well-defined adjuvant. In self-adjuvanting vaccines, innate immune receptor ligands are usually used as adjuvants. Innate immune receptor ligands effectively trigger acquired immunity through the activation of innate immunity to enhance host immune responses to antigens. When a self-adjuvanting vaccine is used, immune cells simultaneously uptake the antigen and the adjuvant because they are covalently linked. Consequently, the adjuvant can specifically induce immune responses against the conjugated antigen. Importantly, self-adjuvanting vaccines do not require co-administration of additional adjuvants or immobilization to carrier proteins, which enables avoidance of the use of highly toxic adjuvants or the induction of undesired immune responses. Given these excellent properties, self-adjuvanting vaccines are expected to serve as candidates for the next generation of vaccines. Herein, we review vaccine adjuvants, with a focus on the adjuvants used in self-adjuvanting vaccines, and then overview recent advances made with self-adjuvanting conjugate vaccines.
Collapse
Affiliation(s)
- Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, Japan; Core for Medicine and Science Collaborative Research and Education, Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, Japan.
| | - Tsung-Che Chang
- Department of Chemistry, Graduate School of Science, Osaka University, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, Japan; Core for Medicine and Science Collaborative Research and Education, Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, Japan.
| |
Collapse
|
12
|
Aiga T, Manabe Y, Ito K, Chang T, Kabayama K, Ohshima S, Kametani Y, Miura A, Furukawa H, Inaba H, Matsuura K, Fukase K. Immunological Evaluation of Co‐Assembling a Lipidated Peptide Antigen and Lipophilic Adjuvants: Self‐Adjuvanting Anti‐Breast‐Cancer Vaccine Candidates. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202007999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Taku Aiga
- Department of Chemistry Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
| | - Yoshiyuki Manabe
- Department of Chemistry Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
- Core for Medicine and Science Collaborative Research and Education Project Research Center for Fundamental Sciences Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
| | - Keita Ito
- Department of Chemistry Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
- Core for Medicine and Science Collaborative Research and Education Project Research Center for Fundamental Sciences Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
| | - Tsung‐Che Chang
- Department of Chemistry Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
| | - Kazuya Kabayama
- Department of Chemistry Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
- Core for Medicine and Science Collaborative Research and Education Project Research Center for Fundamental Sciences Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
| | - Shino Ohshima
- School of Medicine Tokai University Isehara Kanagawa 259-1193 Japan
| | - Yoshie Kametani
- School of Medicine Tokai University Isehara Kanagawa 259-1193 Japan
| | - Ayane Miura
- Department of Chemistry Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
| | - Hiroto Furukawa
- Department of Chemistry and Biotechnology Graduate School of Engineering Center for Research on Green Sustainable Chemistry Tottori University 4-101 Koyama-Minami Tottori 680-8552 Japan
| | - Hiroshi Inaba
- Department of Chemistry and Biotechnology Graduate School of Engineering Center for Research on Green Sustainable Chemistry Tottori University 4-101 Koyama-Minami Tottori 680-8552 Japan
| | - Kazunori Matsuura
- Department of Chemistry and Biotechnology Graduate School of Engineering Center for Research on Green Sustainable Chemistry Tottori University 4-101 Koyama-Minami Tottori 680-8552 Japan
| | - Koichi Fukase
- Department of Chemistry Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
- Core for Medicine and Science Collaborative Research and Education Project Research Center for Fundamental Sciences Graduate School of Science Osaka University 1-1 Machikaneyama, Toyonaka Osaka 560-0043 Japan
| |
Collapse
|
13
|
Aiga T, Manabe Y, Ito K, Chang TC, Kabayama K, Ohshima S, Kametani Y, Miura A, Furukawa H, Inaba H, Matsuura K, Fukase K. Immunological Evaluation of Co-Assembling a Lipidated Peptide Antigen and Lipophilic Adjuvants: Self-Adjuvanting Anti-Breast-Cancer Vaccine Candidates. Angew Chem Int Ed Engl 2020; 59:17705-17711. [PMID: 32583549 DOI: 10.1002/anie.202007999] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Indexed: 12/22/2022]
Abstract
Co-assembling vaccines composed of a lipidated HER2-derived antigenic CH401 peptide and either a lipophilic adjuvant, Pam3 CSK4 , α-GalCer, or lipid A 506, were evaluated as breast cancer vaccine candidates. This vaccine design was aimed to inherit both antigen multivalency and antigen-specific immunostimulation properties, observed in reported self-adjuvanting vaccine candidates, by using self-assembly and adjuvant-conjugated antigens. Under vaccination concentrations, respective lipophilic adjuvants underwent co-assembly with lipidated CH401, which boosted the anti-CH401 IgG and IgM production. In particular, α-GalCer was responsible for the most significant immune activation. Therefore, the newly developed vaccine design enabled the optimization of adjuvants against the antigenic CH401 peptide in a simple preparatory manner. Overall, the co-assembling vaccine design opens the door for efficient and practical self-adjuvanting vaccine development.
Collapse
Affiliation(s)
- Taku Aiga
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan.,Core for Medicine and Science Collaborative Research and Education, Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Keita Ito
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan.,Core for Medicine and Science Collaborative Research and Education, Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Tsung-Che Chang
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan.,Core for Medicine and Science Collaborative Research and Education, Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Shino Ohshima
- School of Medicine, Tokai University, Isehara, Kanagawa, 259-1193, Japan
| | - Yoshie Kametani
- School of Medicine, Tokai University, Isehara, Kanagawa, 259-1193, Japan
| | - Ayane Miura
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Hiroto Furukawa
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Center for Research on Green Sustainable Chemistry, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8552, Japan
| | - Hiroshi Inaba
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Center for Research on Green Sustainable Chemistry, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8552, Japan
| | - Kazunori Matsuura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Center for Research on Green Sustainable Chemistry, Tottori University, 4-101 Koyama-Minami, Tottori, 680-8552, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan.,Core for Medicine and Science Collaborative Research and Education, Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| |
Collapse
|
14
|
Sui Y, Berzofsky JA. Myeloid Cell-Mediated Trained Innate Immunity in Mucosal AIDS Vaccine Development. Front Immunol 2020; 11:315. [PMID: 32184782 PMCID: PMC7058986 DOI: 10.3389/fimmu.2020.00315] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
Trained innate immunity has recently emerged as a novel concept of innate immune cells, such as myeloid cells, exhibiting immune memory, and nonspecific heterologous immunity to protect against infections. The memory and specificity are mediated by epigenetic, metabolic, and functional reprogramming of the myeloid cells and myeloid progenitors (and/or NK cells) in the bone marrow and peripheral tissues such as gut and lung mucosa. A variety of agents, such as BCG, viruses, and their components, as well as TLR agonists, and cytokines have been shown to be involved in the induction of trained immunity. Since these agents have been widely used in AIDS vaccine development as antigen delivery vectors or adjuvants, myeloid cell mediated trained immunity might also play an important role in protecting against mucosal AIDS virus transmission or in control of virus replication in the major gut mucosal reservoir. Here we review the trained innate immunity induced by these vectors/adjuvants that have been used in AIDS vaccine studies and discuss their role in mucosal vaccine efficacy and possible utilization in AIDS vaccine development. Delineating the protective effect of the trained innate immunity mediated by myeloid cells will guide the design of novel AIDS vaccines.
Collapse
Affiliation(s)
- Yongjun Sui
- Vaccine Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Jay A Berzofsky
- Vaccine Branch, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
15
|
Albin T, Tom JK, Manna S, Gilkes AP, Stetkevich SA, Katz BB, Supnet M, Felgner J, Jain A, Nakajima R, Jasinskas A, Zlotnik A, Pearlman E, Davies DH, Felgner PL, Burkhardt AM, Esser-Kahn AP. Linked Toll-Like Receptor Triagonists Stimulate Distinct, Combination-Dependent Innate Immune Responses. ACS CENTRAL SCIENCE 2019; 5:1137-1145. [PMID: 31403067 PMCID: PMC6661867 DOI: 10.1021/acscentsci.8b00823] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Indexed: 05/04/2023]
Abstract
Traditional vaccination strategies have failed to generate effective vaccines for many infections like tuberculosis and HIV. New approaches are needed for each type of disease. The protective immunity and distinct responses of many successful vaccines come from activating multiple Toll-like receptors (TLRs). Vaccines with multiple TLRs as adjuvants have proven effective in preclinical studies, but current research has not explored two important elements. First, few multi-TLR systems explore spatial organization-a critical feature of whole-cell vaccines. Second, no multi-TLR systems to date provide systematic analysis of the combinatorial space of three TLR agonists. Here, we present the first examination of the combinatorial space of several spatially defined triple-TLR adjuvants, by synthesizing a series of five triple-TLR agonists and testing their innate activity both in vitro and in vivo. The combinations were evaluated by measuring activation of immune stimulatory genes (Nf-κB, ISGs), cytokine profiles (IL12-p70, TNF-α, IL-6, IL-10, CCL2, IFN-α, IFN-β, IFN-γ), and in vivo cytokine serum levels (IL-6, TNF-α, IL12-p40, IFN-α, IFN-β). We demonstrate that linking TLR agonists substantially alters the resulting immune response compared to their unlinked counterparts and that each combination results in a distinct immune response, particularly between linked combinations. We show that combinations containing a TLR9 agonist produce more Th1 biasing immune response profiles, and that the effect is amplified upon conjugation. However, combinations containing TLR2/6 agonist are skewed toward TH2 biasing profiles despite the presence of TLR9. These results demonstrate the profound effects that conjugation and combinatorial administration of TLR agonists can have on immune responses, a critical element of vaccine development.
Collapse
Affiliation(s)
- Tyler
J. Albin
- Department
of Chemistry, University of California, Irvine, California 92617, United States
| | - Janine K. Tom
- Department
of Chemistry, University of California, Irvine, California 92617, United States
| | - Saikat Manna
- Department
of Chemistry, University of California, Irvine, California 92617, United States
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
| | - Adrienne P. Gilkes
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - Samuel A. Stetkevich
- Department
of Chemistry, University of California, Irvine, California 92617, United States
| | - Benjamin B. Katz
- Department
of Chemistry, University of California, Irvine, California 92617, United States
| | - Medalyn Supnet
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - Jiin Felgner
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - Aarti Jain
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - Rie Nakajima
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - Algis Jasinskas
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - Albert Zlotnik
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - Eric Pearlman
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - D. Huw Davies
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - Phillip L. Felgner
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
| | - Amanda M. Burkhardt
- Vaccine
Research and Development Center, Department of Physiology & Biophysics, University of California, Irvine, California 92617, United States
- School
of Medicine, Institute for Immunology, University
of California, Irvine, California 92617, United States
- E-mail:
| | - Aaron P. Esser-Kahn
- Department
of Chemistry, University of California, Irvine, California 92617, United States
- Pritzker
School of Molecular Engineering, University
of Chicago, Chicago, Illinois 60637, United States
- E-mail:
| |
Collapse
|
16
|
Lee Y, Ko EJ, Kim KH, Lee YT, Hwang HS, Kwon YM, Graham BS, Kang SM. A unique combination adjuvant modulates immune responses preventing vaccine-enhanced pulmonary histopathology after a single dose vaccination with fusion protein and challenge with respiratory syncytial virus. Virology 2019; 534:1-13. [PMID: 31163351 DOI: 10.1016/j.virol.2019.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 10/26/2022]
Abstract
Alum adjuvanted formalin-inactivated respiratory syncytial virus (RSV) vaccination resulted in enhanced respiratory disease in young children upon natural infection. Here, we investigated the adjuvant effects of monophosphoryl lipid A (MPL) and oligodeoxynucleotide CpG (CpG) on vaccine-enhanced respiratory disease after fusion (F) protein prime vaccination and RSV challenge in infant and adult mouse models. Combination CpG + MPL adjuvant in RSV F protein single dose priming of infant and adult age mice was found to promote the induction of IgG2a isotype antibodies and neutralizing activity, and lung viral clearance after challenge. CpG + MPL adjuvanted F protein (Fp) priming of infant and adult age mice was effective in avoiding lung histopathology, in reducing interleukin-4+ CD4 T cells and cellular infiltration of monocytes and neutrophils after RSV challenge. This study suggests that combination CpG and MPL adjuvant in RSV subunit vaccination might contribute to priming protective immune responses and preventing inflammatory RSV disease after infection.
Collapse
Affiliation(s)
- Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA; Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, South Korea
| | - Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sang Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
17
|
Applications of Immunomodulatory Immune Synergies to Adjuvant Discovery and Vaccine Development. Trends Biotechnol 2018; 37:373-388. [PMID: 30470547 DOI: 10.1016/j.tibtech.2018.10.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 01/01/2023]
Abstract
Pathogens comprise a diverse set of immunostimulatory molecules that activate the innate immune system during infection. The immune system recognizes distinct combinations of pathogenic molecules leading to multiple immune activation events that cooperate to produce enhanced immune responses, known as 'immune synergies'. Effective immune synergies are essential for the clearance of pathogens, thus inspiring novel adjuvant design to improve vaccines. We highlight current vaccine adjuvants and the importance of immune synergies to adjuvant and vaccine design. The focus is on new technologies used to study and apply immune synergies to adjuvant and vaccine development. Finally, we discuss how recent findings can be applied to the future design and characterization of synergistic adjuvants and vaccines.
Collapse
|
18
|
Astill J, Alkie T, Yitbarek A, Taha-Abdelaziz K, Shojadoost B, Petrik JJ, Nagy É, Sharif S. A Comparison of Toll-Like Receptor 5 and 21 Ligands as Adjuvants for a Formaldehyde Inactivated H9N2 Avian Influenza Virus Vaccine in Chickens. Viral Immunol 2018; 31:605-612. [PMID: 30222508 DOI: 10.1089/vim.2018.0072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Low pathogenic avian influenza virus (AIV) infection in chickens can result in economic losses and has impacts on human health. Poultry vaccination is a tool that can be used to decrease infection and transmission of AIVs. Prior research has demonstrated that Toll-like receptor (TLR) ligands can act as vaccine adjuvants and their addition to inactivated AIV vaccines can enhance immune responses elicited in chickens. The objective of this study was to compare the adjuvant capabilities of TLR5 ligand (flagellin) and TLR21 ligand (CpG ODN 2007) administered either alone or in combination with an intramuscular formaldehyde inactivated H9N2 whole virus vaccine in chickens. Along with the inactivated virus, chickens were administered either a single dose of CpG ODN 2007 (2 or 10 μg), flagellin (0.4 or 2 μg), or a combination of both ligands. An additional group received AddaVax™, an oil emulsion style adjuvant. Chickens were vaccinated twice and serum and lachrymal samples were collected weekly following the primary vaccination, and antibody-mediated immune responses were quantified. Results showed that vaccines containing CpG ODN 2007 induce significantly greater systemic and lachrymal antibody responses than vaccines containing flagellin or AddaVax. Combinations of flagellin and CpG ODN 2007 did not demonstrate inhibitory, additive, or synergistic effects on systemic or lachrymal antibody-mediated immune responses. Additionally, for both flagellin and CpG ODN 2007, a fivefold higher dose of each did not induce significantly higher antibody-mediated immune responses compared with the lesser dose. Future studies should examine the induction of cell-mediated immune responses when flagellin, CpG ODN 2007, or other TLR ligands are administered either alone or combined as adjuvants for inactivated H9N2 AIV vaccines.
Collapse
Affiliation(s)
- Jake Astill
- 1 Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| | - Tamiru Alkie
- 1 Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| | - Alexander Yitbarek
- 1 Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| | - Khaled Taha-Abdelaziz
- 1 Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada .,2 Pathology Department, Faculty of Veterinary Medicine, Beni-Suef University , Beni-Suef, Egypt
| | - Bahram Shojadoost
- 1 Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| | - James John Petrik
- 3 Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| | - Éva Nagy
- 1 Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| | - Shayan Sharif
- 1 Department of Pathobiology, Ontario Veterinary College, University of Guelph , Guelph, Ontario, Canada
| |
Collapse
|
19
|
Tada R, Yamanaka D, Ogasawara M, Saito M, Ohno N, Kiyono H, Kunisawa J, Aramaki Y. Polymeric Caffeic Acid Is a Safer Mucosal Adjuvant That Augments Antigen-Specific Mucosal and Systemic Immune Responses in Mice. Mol Pharm 2018; 15:4226-4234. [PMID: 30107747 DOI: 10.1021/acs.molpharmaceut.8b00648] [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] [Indexed: 01/08/2023]
Abstract
Infections remain a major threat to human lives. To overcome the threat caused by pathogens, mucosal vaccines are considered a promising strategy. However, no inactivated and/or subunit mucosal vaccine has been approved for human use, largely because of the lack of a safe and effective mucosal adjuvant. Here, we show that enzymatically synthesized polymeric caffeic acid (pCA) can act as a potent mucosal adjuvant in mice. Intranasal administration of ovalbumin (OVA) in combination with pCA resulted in the induction of OVA-specific mucosal IgA and serum IgG, especially IgG1. Importantly, pCA was synthesized from caffeic acid and horseradish peroxidase from coffee beans and horseradish, respectively, which are commonly consumed. Therefore, pCA is believed to be a highly safe material. In fact, administration of pCA did not show distinct toxicity in mice. These data indicate that pCA has merit for use as a mucosal adjuvant for nasal vaccine formulations.
Collapse
Affiliation(s)
| | | | | | | | | | - Hiroshi Kiyono
- Division of Mucosal Immunology and International Research and Development Center for Mucosal Vaccines, Department of Microbiology and Immunology, The Institute of Medical Science , The University of Tokyo , Tokyo 108-8639 , Japan
| | - Jun Kunisawa
- Division of Mucosal Immunology and International Research and Development Center for Mucosal Vaccines, Department of Microbiology and Immunology, The Institute of Medical Science , The University of Tokyo , Tokyo 108-8639 , Japan.,Laboratory of Vaccine Materials , National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN) , Osaka 567-0085 , Japan
| | | |
Collapse
|
20
|
Gammon JM, Dold NM, Jewell CM. Improving the clinical impact of biomaterials in cancer immunotherapy. Oncotarget 2017; 7:15421-43. [PMID: 26871948 PMCID: PMC4941251 DOI: 10.18632/oncotarget.7304] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 01/29/2016] [Indexed: 12/20/2022] Open
Abstract
Immunotherapies for cancer have progressed enormously over the past few decades, and hold great promise for the future. The successes of these therapies, with some patients showing durable and complete remission, demonstrate the power of harnessing the immune system to eradicate tumors. However, the effectiveness of current immunotherapies is limited by hurdles ranging from immunosuppressive strategies employed by tumors, to inadequate specificity of existing therapies, to heterogeneity of disease. Further, the vast majority of approved immunotherapies employ systemic delivery of immunomodulators or cells that make addressing some of these challenges more difficult. Natural and synthetic biomaterials–such as biocompatible polymers, self-assembled lipid particles, and implantable biodegradable devices–offer unique potential to address these hurdles by harnessing the benefits of therapeutic targeting, tissue engineering, co-delivery, controlled release, and sensing. However, despite the enormous investment in new materials and nanotechnology, translation of these ideas to the clinic is still an uncommon outcome. Here we review the major challenges facing immunotherapies and discuss how the newest biomaterials and nanotechnologies could help overcome these challenges to create new clinical options for patients.
Collapse
Affiliation(s)
- Joshua M Gammon
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Neil M Dold
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA.,Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, USA.,Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA
| |
Collapse
|
21
|
Ko EJ, Lee YT, Lee Y, Kim KH, Kang SM. Distinct Effects of Monophosphoryl Lipid A, Oligodeoxynucleotide CpG, and Combination Adjuvants on Modulating Innate and Adaptive Immune Responses to Influenza Vaccination. Immune Netw 2017; 17:326-342. [PMID: 29093654 PMCID: PMC5662782 DOI: 10.4110/in.2017.17.5.326] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/31/2022] Open
Abstract
Monophosphoryl lipid A (MPL) and oligodeoxynucleotide CpG are toll-like receptor (TLR) 4 and 9 agonist, respectively. Here, we investigated the effects of MPL, CpG, and combination adjuvants on stimulating in vitro dendritic cells (DCs), in vivo innate and adaptive immune responses, and protective efficacy of influenza vaccination. Combination of MPL and CpG was found to exhibit distinct effects on stimulating DCs in vitro to secrete IL-12p70 and tumor necrosis factor (TNF)-α and proliferate allogeneic CD8 T cells. Prime immunization of mice with inactivated split influenza vaccine in the presence of low dose MPL+CpG adjuvants increased the induction of virus-specific IgG and IgG2a isotype antibodies. MPL and CpG adjuvants contribute to improving the efficacy of prime influenza vaccination against lethal influenza challenge as determined by body weight monitoring, lung function, viral titers, and histology. A combination of MPL and CpG adjuvants was effective in improving vaccine efficacy as well as in reducing inflammatory immune responses locally and in inducing cellular immune responses upon lethal influenza virus challenge. This study demonstrates unique adjuvant effects of MPL, CpG, and combination adjuvants on modulating innate and adaptive immune responses to influenza prime vaccination.
Collapse
Affiliation(s)
- Eun-Ju Ko
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
22
|
Gause KT, Wheatley AK, Cui J, Yan Y, Kent SJ, Caruso F. Immunological Principles Guiding the Rational Design of Particles for Vaccine Delivery. ACS NANO 2017; 11:54-68. [PMID: 28075558 DOI: 10.1021/acsnano.6b07343] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Despite the immense public health successes of immunization over the past century, effective vaccines are still lacking for globally important pathogens such as human immunodeficiency virus, malaria, and tuberculosis. Exciting recent advances in immunology and biotechnology over the past few decades have facilitated a shift from empirical to rational vaccine design, opening possibilities for improved vaccines. Some of the most important advancements include (i) the purification of subunit antigens with high safety profiles, (ii) the identification of innate pattern recognition receptors (PRRs) and cognate agonists responsible for inducing immune responses, and (iii) developments in nano- and microparticle fabrication and characterization techniques. Advances in particle engineering now allow highly tunable physicochemical properties of particle-based vaccines, including composition, size, shape, surface characteristics, and degradability. Enhanced collaborative efforts between researchers in immunology and materials science are expected to rise to next-generation vaccines. This process will be significantly aided by a greater understanding of the immunological principles guiding vaccine antigenicity, immunogenicity, and efficacy. With specific emphasis on PRR-targeted adjuvants and particle physicochemical properties, this review aims to provide an overview of the current literature to guide and focus rational particle-based vaccine design efforts.
Collapse
Affiliation(s)
- Katelyn T Gause
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Adam K Wheatley
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity , Parkville, Victoria 3010, Australia
| | - Jiwei Cui
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Yan Yan
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Stephen J Kent
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity , Parkville, Victoria 3010, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| |
Collapse
|
23
|
Akinbobuyi B, Wang L, Upchurch KC, Byrd MR, Chang CA, Quintana JM, Petersen RE, Seifert ZJ, Boquin JR, Oh S, Kane RR. Synthesis and immunostimulatory activity of substituted TLR7 agonists. Bioorg Med Chem Lett 2016; 26:4246-9. [PMID: 27476423 DOI: 10.1016/j.bmcl.2016.07.049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 11/18/2022]
Abstract
Fifteen new substituted adenines were synthesized as potential TLR7 agonists. These compounds, along with 9 previously reported compounds, were analyzed for TLR7 activity and for the selective stimulation of B cell proliferation. Several functionalized derivatives exhibit significant activity, suggesting their potential for use as vaccine adjuvants.
Collapse
Affiliation(s)
- Babatope Akinbobuyi
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - Lei Wang
- Baylor Institute for Immunology Research, Baylor Research Institute, 3434 Live Oak Street, Dallas, TX 75204, USA
| | - Katherine C Upchurch
- Institute of Biomedical Studies, Baylor University, One Bear Place #97224, Waco, TX 76798, USA; Baylor Institute for Immunology Research, Baylor Research Institute, 3434 Live Oak Street, Dallas, TX 75204, USA
| | - Matthew R Byrd
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - Charles A Chang
- Institute of Biomedical Studies, Baylor University, One Bear Place #97224, Waco, TX 76798, USA
| | - Jeremy M Quintana
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - Rachel E Petersen
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - Zacharie J Seifert
- Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA
| | - José R Boquin
- Department of Chemistry, Augustana College, 639 38th Street, Rock Island, IL 61201, USA
| | - SangKon Oh
- Institute of Biomedical Studies, Baylor University, One Bear Place #97224, Waco, TX 76798, USA; Baylor Institute for Immunology Research, Baylor Research Institute, 3434 Live Oak Street, Dallas, TX 75204, USA
| | - Robert R Kane
- Institute of Biomedical Studies, Baylor University, One Bear Place #97224, Waco, TX 76798, USA; Baylor Institute for Immunology Research, Baylor Research Institute, 3434 Live Oak Street, Dallas, TX 75204, USA; Department of Chemistry and Biochemistry, Baylor University, One Bear Place #97348, Waco, TX 76798, USA.
| |
Collapse
|
24
|
Targeted Programming of the Lymph Node Environment Causes Evolution of Local and Systemic Immunity. Cell Mol Bioeng 2016; 9:418-432. [PMID: 27547269 PMCID: PMC4978773 DOI: 10.1007/s12195-016-0455-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/16/2016] [Indexed: 01/21/2023] Open
Abstract
Biomaterial vaccines offer cargo protection, targeting, and co-delivery of signals to immune organs such as lymph nodes (LNs), tissues that coordinate adaptive immunity. Understanding how individual vaccine components impact immune response has been difficult owing to the systemic nature of delivery. Direct intra-lymph node (i.LN.) injection offers a unique opportunity to dissect how the doses, kinetics, and combinations of signals reaching LNs influence the LN environment. Here, i.LN. injection was used as a tool to study the local and systemic responses to vaccines comprised of soluble antigen and degradable polymer particles encapsulating toll-like receptor agonists as adjuvants. Microparticle vaccines increased antigen presenting cells and lymphocytes in LNs, enhancing activation of these cells. Enumeration of antigen-specific CD8+ T cells in blood revealed expansion over 7 days, followed by a contraction period over 1 month as memory developed. Extending this strategy to conserved mouse and human tumor antigens resulted in tumor antigen-specific primary and recall responses by CD8+ T cells. During challenge with an aggressive metastatic melanoma model, i.LN. delivery of depots slowed tumor growth more than a potent human vaccine adjuvant, demonstrating local treatment of a target immunological site can promote responses that are potent, systemic, and antigen-specific.
Collapse
|
25
|
Acharya AP, Carstens MR, Lewis JS, Dolgova N, Xia CQ, Clare-Salzler MJ, Keselowsky BG. A cell-based microarray to investigate combinatorial effects of microparticle-encapsulated adjuvants on dendritic cell activation. J Mater Chem B 2016; 4:1672-1685. [PMID: 26985393 PMCID: PMC4790840 DOI: 10.1039/c5tb01754h] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Experimental vaccine adjuvants are being designed to target specific toll-like receptors (TLRs) alone or in combination, expressed by antigen presenting cells, notably dendritic cells (DCs). There is a need for high-content screening (HCS) platforms to explore how DC activation is affected by adjuvant combinations. Presented is a cell-based microarray approach, "immunoarray", exposing DCs to a large number of adjuvant combinations. Microparticles encapsulating TLR ligands are printed onto arrays in a range of doses for each ligand, in all possible dose combinations. Dendritic cells are then co-localized with physisorbed microparticles on the immunoarray, adherent to isolated islands surrounded by a non-fouling background, and DC activation is quantified. Delivery of individual TLR ligands was capable of eliciting high levels of specific DC activation markers. For example, either TLR9 ligand, CpG, or TLR3 ligand, poly I:C, was capable of inducing among the highest 10% expression levels of CD86. In contrast, MHC-II expression in response to TLR4 agonist MPLA was among the highest, whereas either MPLA or poly I:C, was capable of producing among the highest levels of CCR7 expression, as well as inflammatory cytokine IL-12. However, in order to produce robust responses across all activation markers, adjuvant combinations were required, and combinations were more represented among the high responders. The immunoarray also enables investigation of interactions between adjuvants, and each TLR ligand suggested antagonism to other ligands, for various markers. Altogether, this work demonstrates feasibility of the immunoarray platform to screen microparticle-encapsulated adjuvant combinations for the development of improved and personalized vaccines.
Collapse
Affiliation(s)
- Abhinav P. Acharya
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 130 BME/PO Box 116131, Gainesville, Florida, 32611-6131, USA
- Department of Materials Science and Engineering, University of Florida, USA
| | - Matthew R. Carstens
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 130 BME/PO Box 116131, Gainesville, Florida, 32611-6131, USA
| | - Jamal S. Lewis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 130 BME/PO Box 116131, Gainesville, Florida, 32611-6131, USA
- Department of Biomedical Engineering, University of California, Davis, US
| | - Natalia Dolgova
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 130 BME/PO Box 116131, Gainesville, Florida, 32611-6131, USA
| | - C. Q. Xia
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, USA
| | | | - Benjamin G. Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 130 BME/PO Box 116131, Gainesville, Florida, 32611-6131, USA
| |
Collapse
|
26
|
Zhou L, Cao X, Fang J, Li Y, Fan M. Macrophages polarization is mediated by the combination of PRR ligands and distinct inflammatory cytokines. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:10964-10974. [PMID: 26617814 PMCID: PMC4637629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 08/21/2015] [Indexed: 06/05/2023]
Abstract
Macrophages recognize microbes through Pattern Recognition Receptors (PRRs), and then release pro-inflammatory and anti-inflammatory cytokines. Recent studies have highlighted that collaboration between different PRRs. However, these studies have neglected the crosstalk between various PRRs on macrophages. In the present study, we investigated the interplay of nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs) (NOD1, NOD2) and TLRs (TLR1, 2, 3, 4, 5, 6, 7, 8) in terms of macrophage activation, the expression and production of cytokines. The macrophages were stimulated with a single PRR ligand or a combination of TLR and NOD ligands. After 8 h of incubation, the mRNA expression of interleukin-1β (IL-1β), IL-4, IL-6, IL-10, IL-12p35, IL-12p40, IL-13, and interferon-γ (IFN-γ) was evaluated. The production of these cytokines was also measured. NOD2 synergized with TLR3 agonists on enhancement of IL-10 release. However, the combination of NOD1 with TLR3 ligands showed little effect on IL-10 production. Moreover, NOD2 inhibited the percentages of CD11b + F4/80 + cells activated by TLR3 agonist.
Collapse
Affiliation(s)
- Lili Zhou
- Key Laboratory for Oral Biomedical Engineering of Ministry of Education, School and Hospital of Stomatology, Wuhan University Luoyu Road 237, Wuhan 430079, Hubei, China
| | - Xixi Cao
- Key Laboratory for Oral Biomedical Engineering of Ministry of Education, School and Hospital of Stomatology, Wuhan University Luoyu Road 237, Wuhan 430079, Hubei, China
| | - Jie Fang
- Key Laboratory for Oral Biomedical Engineering of Ministry of Education, School and Hospital of Stomatology, Wuhan University Luoyu Road 237, Wuhan 430079, Hubei, China
| | - Yuhong Li
- Key Laboratory for Oral Biomedical Engineering of Ministry of Education, School and Hospital of Stomatology, Wuhan University Luoyu Road 237, Wuhan 430079, Hubei, China
| | - Mingwen Fan
- Key Laboratory for Oral Biomedical Engineering of Ministry of Education, School and Hospital of Stomatology, Wuhan University Luoyu Road 237, Wuhan 430079, Hubei, China
| |
Collapse
|
27
|
Pavot V, Berthet M, Rességuier J, Legaz S, Handké N, Gilbert SC, Paul S, Verrier B. Poly(lactic acid) and poly(lactic-co-glycolic acid) particles as versatile carrier platforms for vaccine delivery. Nanomedicine (Lond) 2015; 9:2703-18. [PMID: 25529572 DOI: 10.2217/nnm.14.156] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The development of safe and effective vaccines for cancer and infectious diseases remains a major goal in public health. Over the last two decades, controlled release of vaccine antigens and immunostimulant molecules has been achieved using nanometer or micron-sized delivery vehicles synthesized using biodegradable polymers. In addition to achieving a depot effect, enhanced vaccine efficacy using such delivery vehicles has been attributed to efficient targeting of antigen presenting cells such as dendritic cells. Biodegradable and biocompatible poly(lactic acid) and poly(lactic-co-glycolic acid) polymers belong to one such family of polymers that have been a popular choice of material used in the design of these delivery vehicles. This review summarizes research findings from ourselves and others highlighting the promise of poly(lactic acid)- and poly(lactic-co-glycolic acid)-based vaccine carriers in enhancing immune responses.
Collapse
Affiliation(s)
- Vincent Pavot
- The Jenner Institute, University of Oxford, Oxford, UK
| | | | | | | | | | | | | | | |
Collapse
|
28
|
A Multiantigenic DNA Vaccine That Induces Broad Hepatitis C Virus-Specific T-Cell Responses in Mice. J Virol 2015; 89:7991-8002. [PMID: 26018154 DOI: 10.1128/jvi.00803-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 05/15/2015] [Indexed: 12/23/2022] Open
Abstract
UNLABELLED There are 3 to 4 million new hepatitis C virus (HCV) infections annually around the world, but no vaccine is available. Robust T-cell mediated responses are necessary for effective clearance of the virus, and DNA vaccines result in a cell-mediated bias. Adjuvants are often required for effective vaccination, but during natural lytic viral infections damage-associated molecular patterns (DAMPs) are released, which act as natural adjuvants. Hence, a vaccine that induces cell necrosis and releases DAMPs will result in cell-mediated immunity (CMI), similar to that resulting from natural lytic viral infection. We have generated a DNA vaccine with the ability to elicit strong CMI against the HCV nonstructural (NS) proteins (3, 4A, 4B, and 5B) by encoding a cytolytic protein, perforin (PRF), and the antigens on a single plasmid. We examined the efficacy of the vaccines in C57BL/6 mice, as determined by gamma interferon enzyme-linked immunosorbent spot assay, cell proliferation studies, and intracellular cytokine production. Initially, we showed that encoding the NS4A protein in a vaccine which encoded only NS3 reduced the immunogenicity of NS3, whereas including PRF increased NS3 immunogenicity. In contrast, the inclusion of NS4A increased the immunogenicity of the NS3, NS4B, andNS5B proteins, when encoded in a DNA vaccine that also encoded PRF. Finally, vaccines that also encoded PRF elicited similar levels of CMI against each protein after vaccination with DNA encoding NS3, NS4A, NS4B, and NS5B compared to mice vaccinated with DNA encoding only NS3 or NS4B/5B. Thus, we have developed a promising "multiantigen" vaccine that elicits robust CMI. IMPORTANCE Since their development, vaccines have reduced the global burden of disease. One strategy for vaccine development is to use commercially viable DNA technology, which has the potential to generate robust immune responses. Hepatitis C virus causes chronic liver infection and is a leading cause of liver cancer. To date, no vaccine is currently available, and treatment is costly and often results in side effects, limiting the number of patients who are treated. Despite recent advances in treatment, prevention remains the key to efficient control and elimination of this virus. Here, we describe a novel DNA vaccine against hepatitis C virus that is capable of inducing robust cell-mediated immune responses in mice and is a promising vaccine candidate for humans.
Collapse
|
29
|
Wu J, Zhang Y, Xin Z, Wu X. The crosstalk between TLR2 and NOD2 in Aspergillus fumigatus keratitis. Mol Immunol 2015; 64:235-43. [DOI: 10.1016/j.molimm.2014.11.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 11/10/2014] [Accepted: 11/30/2014] [Indexed: 01/12/2023]
|
30
|
Quayle K, Coy C, Standish L, Lu H. The TLR2 agonist in polysaccharide-K is a structurally distinct lipid which acts synergistically with the protein-bound β-glucan. J Nat Med 2014; 69:198-208. [PMID: 25510899 DOI: 10.1007/s11418-014-0879-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 11/19/2014] [Indexed: 01/21/2023]
Abstract
Protein-bound polysaccharide-K (Krestin; PSK) is a hot-water extract of Trametes versicolor with immune stimulatory activity. It has been used for the past 30 years and has demonstrated anti-tumor efficacy in multiple types of cancer. The ability of PSK to activate dendritic cells and T cells is dependent on its ability to stimulate Toll-like receptor 2 (TLR2), yet it remains unknown which structural component within PSK activates TLR2. The purpose of this study was to identify the TLR2 agonist within PSK and understand its role in the overall mechanism of PSK's immunogenic activity. TLR2 activity was eliminated by treatment with lipoprotein lipase but not by trypsin or lyticase. Rapid centrifugation of PSK can separate the fraction with TLR2 agonist activity from the soluble β-glucan fraction. To study the potential interaction between the β-glucan component and the lipid component, we labeled the soluble β-glucan with fluorescein. Uptake of the labeled β-glucan by J774A macrophages and JAWSII dendritic cells was inhibited by anti-Dectin-1 antibody but not by anti-TLR2 antibody, confirming that Dectin-1 is the receptor for β-glucan. Interestingly, pre-treatment of JAWSII cells with the TLR2-active lipid fraction significantly enhanced the uptake of the soluble β-glucan, indicating the synergy between the TLR2 agonist component and the β-glucan component. Altogether, these results present evidence that PSK has two active components-the well-characterized protein-bound β-glucan and a previously unreported lipid-which work synergistically via the Dectin-1 and TLR2 receptors.
Collapse
Affiliation(s)
- Kenneth Quayle
- Tumor Vaccine Group, Department of Medicine, University of Washington, 850 Republican Street, Seattle, WA, 98109, USA,
| | | | | | | |
Collapse
|
31
|
Pavot V, Rochereau N, Rességuier J, Gutjahr A, Genin C, Tiraby G, Perouzel E, Lioux T, Vernejoul F, Verrier B, Paul S. Cutting edge: New chimeric NOD2/TLR2 adjuvant drastically increases vaccine immunogenicity. THE JOURNAL OF IMMUNOLOGY 2014; 193:5781-5. [PMID: 25392526 DOI: 10.4049/jimmunol.1402184] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
TLR ligands are critical activators of innate immunity and are being developed as vaccine adjuvants. However, their usefulness in conjunction with NOD-like receptor agonists remains poorly studied. In this study, we evaluated a new ligand that targets both TLR2 and NOD2 receptors. We assessed its ability to enhance dendritic cell maturation in vitro in addition to improving systemic and mucosal immune responses in mice. The chimeric NOD2/TLR2 ligand induced synergistic upregulation of dendritic cell maturation markers, costimulatory molecules, and secretion of proinflammatory cytokines compared with combinations of separate ligands. Furthermore, when coadministered with biodegradable nanoparticles carrying a model Ag, the ligand was able to induce high Ag-specific IgA and IgG titers at both systemic and mucosal sites after parenteral immunizations. These findings point out the potential utility of chimeric molecules TLR/NOD as adjuvants for vaccines to induce systemic and mucosal immune responses.
Collapse
Affiliation(s)
- Vincent Pavot
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5305, Université de Lyon, Lyon F-69007, France; Groupe Immunité des Muqueuses et Agents Pathogènes, INSERM Centre d'Investigation Clinique en Vaccinologie 1408, Université de Lyon, Saint-Etienne F-42023, France; and
| | - Nicolas Rochereau
- Groupe Immunité des Muqueuses et Agents Pathogènes, INSERM Centre d'Investigation Clinique en Vaccinologie 1408, Université de Lyon, Saint-Etienne F-42023, France; and
| | - Julien Rességuier
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5305, Université de Lyon, Lyon F-69007, France
| | - Alice Gutjahr
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5305, Université de Lyon, Lyon F-69007, France; Groupe Immunité des Muqueuses et Agents Pathogènes, INSERM Centre d'Investigation Clinique en Vaccinologie 1408, Université de Lyon, Saint-Etienne F-42023, France; and Cayla-InvivoGen, Toulouse F-31000, France
| | - Christian Genin
- Groupe Immunité des Muqueuses et Agents Pathogènes, INSERM Centre d'Investigation Clinique en Vaccinologie 1408, Université de Lyon, Saint-Etienne F-42023, France; and
| | | | | | | | | | - Bernard Verrier
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5305, Université de Lyon, Lyon F-69007, France
| | - Stéphane Paul
- Groupe Immunité des Muqueuses et Agents Pathogènes, INSERM Centre d'Investigation Clinique en Vaccinologie 1408, Université de Lyon, Saint-Etienne F-42023, France; and
| |
Collapse
|
32
|
Mancini RJ, Stutts L, Ryu KA, Tom JK, Esser-Kahn AP. Directing the immune system with chemical compounds. ACS Chem Biol 2014; 9:1075-85. [PMID: 24690004 PMCID: PMC5674983 DOI: 10.1021/cb500079s] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Agonists of immune cell receptors direct innate and adaptive immunity. These agonists range in size and complexity from small molecules to large macromolecules. Here, agonists of a class of immune cell receptors known as the Toll-like receptors (TLRs) are highlighted focusing on the distinctive molecular moieties that pertain to receptor binding and activation. How the structure and combined chemical signals translate into a variety of immune responses remain major questions in the field. In this structure-focused review, we outline potential areas where the tools of chemical biology could help decipher the emerging molecular codes that direct immune stimulation.
Collapse
Affiliation(s)
- Rock J Mancini
- Department of Chemistry, Chemical Engineering and Materials Science, and Biomedical Engineering, University of California Irvine , 1102 Natural Sciences 2, Irvine, California 92697-2025, United States
| | | | | | | | | |
Collapse
|
33
|
Labuda LA, de Jong SE, Meurs L, Amoah AS, Mbow M, Ateba-Ngoa U, van der Ham AJ, Knulst AC, Yazdanbakhsh M, Adegnika AA. Differences in innate cytokine responses between European and African children. PLoS One 2014; 9:e95241. [PMID: 24743542 PMCID: PMC3990610 DOI: 10.1371/journal.pone.0095241] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/24/2014] [Indexed: 01/22/2023] Open
Abstract
Although differences in immunological responses between populations have been found in terms of vaccine efficacy, immune responses to infections and prevalence of chronic inflammatory diseases, the mechanisms responsible for these differences are not well understood. Therefore, innate cytokine responses mediated by various classes of pattern-recognition receptors including Toll-like receptors (TLR), C-type lectin receptors (CLRs) and nucleotide-binding oligomerisation domain-like receptors (NLRs) were compared between Dutch (European), semi-urban and rural Gabonese (African) children. Whole blood was stimulated for 24 hours and the pro-inflammatory tumor necrosis factor (TNF) and the anti-inflammatory/regulatory interleukin-10 (IL-10) cytokines in culture supernatant were measured by enzyme-linked immunosorbent assay (ELISA). Gabonese children had a lower pro-inflammatory response to poly(I:C) (TLR3 ligand), but a higher pro-inflammatory response to FSL-1 (TLR2/6 ligand), Pam3 (TLR2/1 ligand) and LPS (TLR4 ligand) compared to Dutch children. Anti-inflammatory responses to Pam3 were also higher in Gabonese children. Non-TLR ligands did not induce substantial cytokine production on their own. Interaction between various TLR and non-TLR receptors was further assessed, but no differences were found between the three populations. In conclusion, using a field applicable assay, significant differences were observed in cytokine responses between European and African children to TLR ligands, but not to non-TLR ligands.
Collapse
Affiliation(s)
- Lucja A. Labuda
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany
| | - Sanne E. de Jong
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
| | - Lynn Meurs
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Abena S. Amoah
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Accra, Ghana
| | - Moustapha Mbow
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Immunology Department of the Laboratory of Bacteriology and Virology of Aristide Le Dantec University Hospital, Dakar, Senegal
| | - Ulysse Ateba-Ngoa
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany
| | - Alwin J. van der Ham
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - André C. Knulst
- Department of Dermatology/Allergology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Maria Yazdanbakhsh
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ayola A. Adegnika
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
- Centre de Recherches Médicales de Lambaréné, Lambaréné, Gabon
- Institut für Tropenmedizin, Universität Tübingen, Tübingen, Germany
| |
Collapse
|
34
|
Bergmann-Leitner ES, Leitner WW. Adjuvants in the Driver's Seat: How Magnitude, Type, Fine Specificity and Longevity of Immune Responses Are Driven by Distinct Classes of Immune Potentiators. Vaccines (Basel) 2014; 2:252-96. [PMID: 26344620 PMCID: PMC4494256 DOI: 10.3390/vaccines2020252] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 03/20/2014] [Accepted: 03/28/2014] [Indexed: 12/16/2022] Open
Abstract
The mechanism by which vaccine adjuvants enhance immune responses has historically been considered to be the creation of an antigen depot. From here, the antigen is slowly released and provided to immune cells over an extended period of time. This "depot" was formed by associating the antigen with substances able to persist at the injection site, such as aluminum salts or emulsions. The identification of Pathogen-Associated Molecular Patterns (PAMPs) has greatly advanced our understanding of how adjuvants work beyond the simple concept of extended antigen release and has accelerated the development of novel adjuvants. This review focuses on the mode of action of different adjuvant classes in regards to the stimulation of specific immune cell subsets, the biasing of immune responses towards cellular or humoral immune response, the ability to mediate epitope spreading and the induction of persistent immunological memory. A better understanding of how particular adjuvants mediate their biological effects will eventually allow them to be selected for specific vaccines in a targeted and rational manner.
Collapse
Affiliation(s)
- Elke S Bergmann-Leitner
- US Military Malaria Research Program, Malaria Vaccine Branch, 503 Robert Grant Ave, 3W65, Silver Spring, MD 20910, USA.
| | - Wolfgang W Leitner
- Division on Allergy, Immunology and Transplantation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 6610 Rockledge Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
35
|
Zuidscherwoude M, de Winde CM, Cambi A, van Spriel AB. Microdomains in the membrane landscape shape antigen-presenting cell function. J Leukoc Biol 2013; 95:251-63. [PMID: 24168856 DOI: 10.1189/jlb.0813440] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The plasma membrane of immune cells is a highly organized cell structure that is key to the initiation and regulation of innate and adaptive immune responses. It is well-established that immunoreceptors embedded in the plasma membrane have a nonrandom spatial distribution that is important for coupling to components of intracellular signaling cascades. In the last two decades, specialized membrane microdomains, including lipid rafts and TEMs, have been identified. These domains are preformed structures ("physical entities") that compartmentalize proteins, lipids, and signaling molecules into multimolecular assemblies. In APCs, different microdomains containing immunoreceptors (MHC proteins, PRRs, integrins, among others) have been reported that are imperative for efficient pathogen recognition, the formation of the immunological synapse, and subsequent T cell activation. In addition, recent work has demonstrated that tetraspanin microdomains and lipid rafts are involved in BCR signaling and B cell activation. Research into the molecular mechanisms underlying membrane domain formation is fundamental to a comprehensive understanding of membrane-proximal signaling and APC function. This review will also discuss the advances in the microscopy field for the visualization of the plasma membrane, as well as the recent progress in targeting microdomains as novel, therapeutic approach for infectious and malignant diseases.
Collapse
Affiliation(s)
- Malou Zuidscherwoude
- 1.Nijmegen Centre for Molecular Life Sciences/278 TIL, Radboud University Medical Centre, Geert Grooteplein 28, 6525GA, Nijmegen, The Netherlands.
| | | | | | | |
Collapse
|
36
|
Tom JK, Mancini RJ, Esser-Kahn AP. Covalent modification of cell surfaces with TLR agonists improves & directs immune stimulation. Chem Commun (Camb) 2013; 49:9618-20. [PMID: 24022092 PMCID: PMC4399865 DOI: 10.1039/c3cc45468a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We present a primary example of a cell surface modified with a synergistic combination of agonists to tune immune stimulation. A model cell line, Lewis Lung Carcinoma, was covalently modified with CpG-oligonucleotides and lipoteichoic acid, both Toll-like receptor (TLR) agonists. The immune-stimulating constructs provided greater stimulation of NF-κB in a model cell line and bone marrow-derived dendritic cells than the components unconjugated in solution.
Collapse
Affiliation(s)
- Janine K Tom
- Department of Chemistry, University of California Irvine, Irvine, CA 92697, USA.
| | | | | |
Collapse
|
37
|
Willcocks S, Offord V, Seyfert HM, Coffey TJ, Werling D. Species-specific PAMP recognition by TLR2 and evidence for species-restricted interaction with Dectin-1. J Leukoc Biol 2013; 94:449-58. [DOI: 10.1189/jlb.0812390] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|