1
|
Abbasi K, Tahamtan Y, Moazamian E, Hosseini MH. Formalin and ferric chloride inactivated Pasteurella multocida type a adjuvanted with bacterial DNA and alum as a new vaccine candidate in sheep pasteurellosis. Microb Pathog 2023; 183:106282. [PMID: 37591320 DOI: 10.1016/j.micpath.2023.106282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/01/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
The aim of the present study was to investigate humoral and cellular immune responses in sheep inoculated with inactivated P. multocida antigen with alum and bacterial DNA adjuvant by identifying IgG and cytokines from serum and cell culture. Sheep were immunized with iron and formalin-inactivated antigens at an interval of 2 weeks. These immunogens were mixed with alum adjuvant and P. multocida type A DNA (AbDNA). After injection and blood sampling, the serum antibody titer and cellular immune responses (IL-4, IFN-γ, and TNF-α) on serum samples and lymphocyte cell were tested by ELISA. The ELISA results showed a higher antibody titer in the bDNA adjuvant group compared to the alum adjuvant group and the control group. In general, the level of IgG in the serum of immunized animals was significantly increased compared to the control group. The peak antibody titer (1.794) was observed on the 28th day of injection in the IIV-AbDNA group. After immunization, inactivation with iron and bDNA adjuvant increased cytokine production compared to other experimental and control groups. High levels of lymphocyte and serum titers of IL-4, IFN-γ, and TNF-α were also obtained in the IIV-AbDNA group. The findings showed that killed P. multocida type A antigens formulated with bacterial DNA as an adjuvant are candidates for new immunogens against P. multocida infections in sheep. The inactivation of bacteria with iron also enhanced proper immune responses.
Collapse
Affiliation(s)
- Keivandokht Abbasi
- Department of Microbiology, College of Science, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Yahya Tahamtan
- Department of Microbiology, Shiraz Branch, Razi Vaccine and Serum Research Institute, Agriculture Research, Education and Extension Organization, Shiraz, Iran.
| | - Elham Moazamian
- Department of Microbiology, College of Science, Agriculture and Modern Technology, Shiraz Branch, Islamic Azad University, Shiraz, Iran
| | - Mohammad Hossein Hosseini
- Department of Microbiology, Shiraz Branch, Razi Vaccine and Serum Research Institute, Agriculture Research, Education and Extension Organization, Shiraz, Iran
| |
Collapse
|
2
|
Corripio-Miyar Y, MacLeod CL, Mair I, Mellanby RJ, Moore BD, McNeilly TN. Self-Adjuvanting Calcium-Phosphate-Coated Microcrystal-Based Vaccines Induce Pyroptosis in Human and Livestock Immune Cells. Vaccines (Basel) 2023; 11:1229. [PMID: 37515044 PMCID: PMC10385459 DOI: 10.3390/vaccines11071229] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Successful vaccines require adjuvants able to activate the innate immune system, eliciting antigen-specific immune responses and B-cell-mediated antibody production. However, unwanted secondary effects and the lack of effectiveness of traditional adjuvants has prompted investigation into novel adjuvants in recent years. Protein-coated microcrystals modified with calcium phosphate (CaP-PCMCs) in which vaccine antigens are co-immobilised within amino acid crystals represent one of these promising self-adjuvanting vaccine delivery systems. CaP-PCMCs has been shown to enhance antigen-specific IgG responses in mouse models; however, the exact mechanism of action of these microcrystals is currently unclear. Here, we set out to investigate this mechanism by studying the interaction between CaP-PCMCs and mammalian immune cells in an in vitro system. Incubation of cells with CaP-PCMCs induced rapid pyroptosis of peripheral blood mononuclear cells and monocyte-derived dendritic cells from cattle, sheep and humans, which was accompanied by the release of interleukin-1β and the activation of Caspase-1. We show that this pyroptotic event was cell-CaP-PCMCs contact dependent, and neither soluble calcium nor microcrystals without CaP (soluble PCMCs) induced pyroptosis. Our results corroborate CaP-PCMCs as a promising delivery system for vaccine antigens, showing great potential for subunit vaccines where the enhancement or find tuning of adaptive immunity is required.
Collapse
Affiliation(s)
| | - Clair Lyle MacLeod
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1XQ, UK
| | - Iris Mair
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian EH25 9RG, UK
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Richard J Mellanby
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian EH25 9RG, UK
| | - Barry D Moore
- Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow G1 1XQ, UK
| | - Tom N McNeilly
- Moredun Research Institute, Pentlands Science Park, Penicuik EH26 0PZ, UK
| |
Collapse
|
3
|
Sasaki E, Asanuma H, Momose H, Furuhata K, Mizukami T, Matsumura T, Takahashi Y, Hamaguchi I. Systemically inoculated adjuvants stimulate pDC-dependent IgA response in local site. Mucosal Immunol 2023; 16:275-286. [PMID: 36935091 DOI: 10.1016/j.mucimm.2023.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/25/2023] [Accepted: 03/08/2023] [Indexed: 03/19/2023]
Abstract
The stimulation of local immunity by vaccination is desirable for controlling virus replication in the respiratory tract. However, the local immune stimulatory effects of adjuvanted vaccines administered through the non-mucosal route are poorly understood. Here, we clarify the mechanisms by which non-mucosal inoculation of adjuvants stimulates the plasmacytoid dendritic cell (pDC)-dependent immunoglobulin (Ig)A response in the lungs. After systemic inoculation with type 1 interferon (IFN)-inducing adjuvants, type 1 IFN promotes CXCL9/10/11 release from alveolar endothelial and epithelial cells and recruits CXCR3-expressing pDCs into the lungs. Because adjuvant-activated pulmonary pDCs highly express major histocompatibility complex II, cluster of differentiation 80, and cluster of differentiation 86, transplantation of such cells into the lungs successfully enhances antigen-specific IgA production by the intranasally sensitized vaccine. In contrast, pDC accumulation in the lungs and subsequent IgA production are impaired in pDC-depleted mice and Ifnar1-/- mice. Notably, the combination of systemic inoculation with type 1 IFN-inducing adjuvants and intranasal antigen sensitization protects mice against influenza virus infection due to the pDC-dependent IgA response and type I IFN response. Our results provide insights into the novel mucosal vaccine strategies using non-mucosal inoculated adjuvants.
Collapse
Affiliation(s)
- Eita Sasaki
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan.
| | - Hideki Asanuma
- Center for Influenza and Respiratory Virus Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Haruka Momose
- Research Center for Biological Products in the Next Generation, National Institute of Infectious Diseases, Tokyo, Japan
| | - Keiko Furuhata
- Research Center for Biological Products in the Next Generation, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takuo Mizukami
- Research Center for Biological Products in the Next Generation, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takayuki Matsumura
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yoshimasa Takahashi
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, Japan
| | - Isao Hamaguchi
- Research Center for Biological Products in the Next Generation, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
4
|
Vaccines and Vaccine Adjuvants for Infectious Diseases and Autoimmune Diseases. Vaccines (Basel) 2023; 11:vaccines11020202. [PMID: 36851080 PMCID: PMC9963674 DOI: 10.3390/vaccines11020202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
A dynamic association of specific microbiota during different stages of human life is well documented [...].
Collapse
|
5
|
Lee J, Ahn SY, Le CTT, Lee DH, Jung J, Ko EJ. Protective and vaccine dose-sparing efficacy of Poly I:C-functionalized calcium phosphate nanoparticle adjuvants in inactivated influenza vaccination. Int Immunopharmacol 2022; 112:109240. [PMID: 36115278 DOI: 10.1016/j.intimp.2022.109240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022]
Abstract
Adjuvants are required to increase the immunogenicity and efficacy of vaccination and enable vaccine dose sparing. Polyinosinic-polycytidylic acid (Poly I:C), a toll-like receptor 3 agonist, is a promising adjuvant candidate that can induce cell-mediated immune responses; however, it remains unlicensed owing to its low stability and toxicity. Calcium phosphate (CaP), a biocompatible and biodegradable nanoparticle, is widely used in biomedicine for stable and targeted drug delivery. In this study, we developed Poly I:C-functionalized CaP (Poly-CaP) and evaluated its vaccine adjuvant efficacy in vitro and in vivo. A half dose of Poly-CaP nanoparticles showed similar efficacy to a full dose of soluble Poly I:C in stimulating bone marrow-derived dendritic cells and macrophages to secrete proinflammatory cytokines and express their activation markers. Immunization with a half dose of inactivated influenza vaccine in the presence of Poly I:C or Poly-CaP adjuvants induced sufficient antigen-specific humoral responses after boost immunization. Immunization with Poly I:C, CaP, or Poly-CaP-adjuvanted with a half dose of influenza vaccine showed comparable protective efficacy against lethal virus infection, with lower weight loss and virus titer than a full dose of influenza vaccine. The Poly-CaP adjuvant was effective in stimulating antigen-specific CD4+ T cell proliferation in the lungs. Collectively, our results showed that the Poly-CaP adjuvant enhanced antigen-specific cell-mediated immunity and humoral immune responses with vaccine dose-sparing effects, suggesting its potential as a novel vaccine adjuvant candidate.
Collapse
Affiliation(s)
- Jueun Lee
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - So Yeon Ahn
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Chau Thuy Tien Le
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Dong-Ha Lee
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jaehan Jung
- Department of Materials Science and Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea.
| | - Eun-Ju Ko
- College of Veterinary Medicine, Jeju National University, Jeju 63243, Republic of Korea; Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju 63243, Republic of Korea; Veterinary Medical Research Institute, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
6
|
Xu S, Carpenter MC, Spreng RL, Neidich SD, Sarkar S, Tenney D, Goodman D, Sawant S, Jha S, Dunn B, Juliana McElrath M, Bekker V, Mudrak SV, Flinko R, Lewis GK, Ferrari G, Tomaras GD, Shen X, Ackerman ME. Impact of adjuvants on the biophysical and functional characteristics of HIV vaccine-elicited antibodies in humans. NPJ Vaccines 2022; 7:90. [PMID: 35927399 PMCID: PMC9352797 DOI: 10.1038/s41541-022-00514-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/01/2022] [Indexed: 01/14/2023] Open
Abstract
Adjuvants can alter the magnitude, characteristics, and persistence of the humoral response to protein vaccination. HIV vaccination might benefit from tailored adjuvant choice as raising a durable and protective response to vaccination has been exceptionally challenging. Analysis of trials of partially effective HIV vaccines have identified features of the immune response that correlate with decreased risk, including high titers of V1V2-binding IgG and IgG3 responses with low titers of V1V2-binding IgA responses and enhanced Fc effector functions, notably antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). However, there has been limited opportunity to compare the effect of different adjuvants on these activities in humans. Here, samples from the AVEG015 study, a phase 1 trial in which participants (n = 112) were immunized with gp120SF-2 and one of six different adjuvants or combinations thereof were assessed for antibody titer, biophysical features, and diverse effector functions. Three adjuvants, MF59 + MTP-PE, SAF/2, and SAF/2 + MDP, increased the peak magnitude and durability of antigen-specific IgG3, IgA, FcγR-binding responses and ADCP activity, as compared to alum. While multiple adjuvants increased the titer of IgG, IgG3, and IgA responses, none consistently altered the balance of IgG to IgA or IgG3 to IgA. Linear regression analysis identified biophysical features including gp120-specific IgG and FcγR-binding responses that could predict functional activity, and network analysis identified coordinated aspects of the humoral response. These analyses reveal the ability of adjuvants to drive the character and function of the humoral response despite limitations of small sample size and immune variability in this human clinical trial.
Collapse
Affiliation(s)
- Shiwei Xu
- Quantitative Biomedical Science Program, Dartmouth College, Hanover, NH, USA
| | | | - Rachel L Spreng
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Scott D Neidich
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sharanya Sarkar
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - DeAnna Tenney
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Derrick Goodman
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sheetal Sawant
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Shalini Jha
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Brooke Dunn
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA
- Departments of Laboratory Medicine and Medicine, University of Washington, Seattle, WA, USA
| | - Valerie Bekker
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sarah V Mudrak
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Robin Flinko
- Division of Vaccine Research, The Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - George K Lewis
- Division of Vaccine Research, The Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Guido Ferrari
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Georgia D Tomaras
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA.
| | - Margaret E Ackerman
- Quantitative Biomedical Science Program, Dartmouth College, Hanover, NH, USA.
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
7
|
An efficient and safe MUC1-dendritic cell-derived exosome conjugate vaccine elicits potent cellular and humoral immunity and tumor inhibition in vivo. Acta Biomater 2022; 138:491-504. [PMID: 34757230 DOI: 10.1016/j.actbio.2021.10.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/20/2021] [Accepted: 10/24/2021] [Indexed: 12/15/2022]
Abstract
Antitumor vaccines are a promising strategy for preventing or treating cancers by eliciting antitumor immune responses and inducing protective immunity against specific antigens expressed on tumor cells. Vaccine formulations that enhance the humoral and cellular immune responses of vaccine candidates would be highly beneficial but are still limited. Here we developed an antitumor vaccine candidate by conjugating a MUC1 glycopeptide antigen to dendritic cell-derived exosomes (Dex). In vivo, the MUC1-Dex construct induced high MUC1-specific IgG antibody titers with strong binding affinities for MUC1-positive tumor cells and promoted cytokine secretion. Moreover, CD8+ T cells from immunized mice exhibited strong cytotoxicity against MUC1-positive tumor cells. Importantly, in both preventative and therapeutic tumor-bearing mouse models, the construct inhibited tumor growth and prolonged survival. Collectively, these results demonstrate that Dex is a promising vaccine carrier that can be used as adjuvant to enhance the immunological efficacy of tumor vaccines. STATEMENT OF SIGNIFICANCE.
Collapse
|
8
|
Recent Progress in Dendritic Cell-Based Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13102495. [PMID: 34065346 PMCID: PMC8161242 DOI: 10.3390/cancers13102495] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Cancer immunotherapy has now attracted much attention because of the recent success of immune checkpoint inhibitors. However, they are only beneficial in a limited fraction of patients most probably due to lack of sufficient CD8+ cytotoxic T-lymphocytes against tumor antigens in the host. In this regard, dendritic cells are useful tools to induce host immune responses against exogenous antigens. In particular, recently characterized cross-presenting dendritic cells are capable of inducing CD8+ cytotoxic T-lymphocytes against exogenous antigens such as tumor antigens and uniquely express the chemokine receptor XCR1. Here we focus on the recent progress in DC-based cancer vaccines and especially the use of the XCR1 and its ligand XCL1 axis for the targeted delivery of cancer vaccines to cross-presenting dendritic cells. Abstract Cancer immunotherapy aims to treat cancer by enhancing cancer-specific host immune responses. Recently, cancer immunotherapy has been attracting much attention because of the successful clinical application of immune checkpoint inhibitors targeting the CTLA-4 and PD-1/PD-L1 pathways. However, although highly effective in some patients, immune checkpoint inhibitors are beneficial only in a limited fraction of patients, possibly because of the lack of enough cancer-specific immune cells, especially CD8+ cytotoxic T-lymphocytes (CTLs), in the host. On the other hand, studies on cancer vaccines, especially DC-based ones, have made significant progress in recent years. In particular, the identification and characterization of cross-presenting DCs have greatly advanced the strategy for the development of effective DC-based vaccines. In this review, we first summarize the surface markers and functional properties of the five major DC subsets. We then describe new approaches to induce antigen-specific CTLs by targeted delivery of antigens to cross-presenting DCs. In this context, the chemokine receptor XCR1 and its ligand XCL1, being selectively expressed by cross-presenting DCs and mainly produced by activated CD8+ T cells, respectively, provide highly promising molecular tools for this purpose. In the near future, CTL-inducing DC-based cancer vaccines may provide a new breakthrough in cancer immunotherapy alone or in combination with immune checkpoint inhibitors.
Collapse
|
9
|
Belizário J. Immunity, virus evolution, and effectiveness of SARS-CoV-2 vaccines. Braz J Med Biol Res 2021; 54:e10725. [PMID: 33729394 PMCID: PMC7959154 DOI: 10.1590/1414-431x202010725] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/29/2020] [Indexed: 12/15/2022] Open
Abstract
Phylogenetic and pathogenesis studies of the severe acute respiratory syndrome-related coronaviruses (SARS-CoVs) strains have highlighted some specific mutations that could confer the RNA genome fitness advantages and immunological resistance for their rapid spread in the human population. The analyses of 30 kb RNA SARS-CoVs genome sequences, protein structures, and functions have provided us a perspective of how host-virus protein-protein complexes act to mediate virus infection. The open reading frame (ORF)1a and ORF1b translation yields 16 non-structural (nsp1-16) and 6 accessory proteins (p6, p7a, p8ab, p9b) with multiple functional domains. Viral proteins recruit over 300 host partners forming hetero-oligomeric complexes enabling the viral RNA synthesis, packing, and virion release. Many cellular host factors and the innate immune cells through pattern-recognition receptors and intracellular RNA sensor molecules act to inhibit virus entry and intracellular replication. However, non-structural ORF proteins hijack them and suppress interferon synthesis and its antiviral effects. Pro-inflammatory chemokines and cytokines storm leads to dysfunctional inflammation, lung injury, and several clinical symptoms in patients. During the global pandemic, COVID-19 patients were identified with non-synonymous substitution of G614D in the spike protein, indicating virus co-evolution in host cells. We review findings that suggest that host RNA editing and DNA repair systems, while carrying on recombination, mutation, and repair of viral RNA intermediates, may facilitate virus evolution. Understanding how the host cell RNA replication process may be driven by SARS-CoV-2 RNA genome fitness will help the testing of vaccines effectiveness to multiple independent mutated coronavirus strains that will emerge.
Collapse
Affiliation(s)
- J.E. Belizário
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
10
|
Seya T, Shime H, Takaki H, Azuma M, Oshiumi H, Matsumoto M. TLR3/TICAM-1 signaling in tumor cell RIP3-dependent necroptosis. Oncoimmunology 2021; 1:917-923. [PMID: 23162759 PMCID: PMC3489747 DOI: 10.4161/onci.21244] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The engagement of Toll-like receptor 3 (TLR3) leads to the oligomerization of the adaptor TICAM-1 (TRIF), which can induces either of three acute cellular responses, namely, cell survival coupled to Type I interferon production, or cell death, via apoptosis or necrosis. The specific response elicited by TLR3 determines the fate of affected cells, although the switching mechanism between the two cell death pathways in TLR3-stimulated cells remains molecularly unknown. Tumor necrosis factor α (TNFα)-mediated cell death can proceed via apoptosis or via a non-apoptotic pathway, termed necroptosis or programmed necrosis, which have been described in detail. Interestingly, death domain-containing kinases called receptor-interacting protein kinases (RIPs) are involved in the signaling pathways leading to these two cell death pathways. Formation of the RIP1/RIP3 complex (called necrosome) in the absence of caspase 8 activity is crucial for the induction of necroptosis in response to TNFα signaling. On the other hand, RIP1 is known to interact with the C-terminal domain of TICAM-1 and to modulate TLR3 signaling. In macrophages and perhaps tumor cell lines, RIP1/RIP3-mediated necroptotic cell death can ensue the administration of the TLR agonist polyI:C. If this involved the TLR3/TICAM-1 pathway, the innate sensing of viral dsRNA would be linked to cytopathic effects and to persistent inflammation, in turn favoring the release of damage-associated molecular patterns (DAMPs) in the microenvironment. Here, we review accumulating evidence pointing to the involvement of the TLR3/TICAM-1 axis in tumor cell necroptosis and the subsequent release of DAMPs.
Collapse
Affiliation(s)
- Tsukasa Seya
- Department of Microbiology and Immunology; Hokkaido University Graduate School of Medicine; Sapporo, Japan
| | | | | | | | | | | |
Collapse
|
11
|
Hyder Pottoo F, Abu-Izneid T, Mohammad Ibrahim A, Noushad Javed M, AlHajri N, Hamrouni AM. Immune system response during viral Infections: Immunomodulators, cytokine storm (CS) and Immunotherapeutics in COVID-19. Saudi Pharm J 2021; 29:173-187. [PMID: 33519271 PMCID: PMC7833973 DOI: 10.1016/j.jsps.2020.12.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022] Open
Abstract
Coronaviruses are non-segmented and single stranded positive-sense RNA (+ssRNA) viruses. To date, 06 human coronaviruses (HCoVs) are reported; α-CoVs (HCoVs-NL63 and HCoVs-229E) and β-CoVs (HCoVs-OC43, HCoVs-HKU1, SARS-CoV, MERS-CoV). While, novel coronavirus (SARS-CoV-2) is the most recent member. The genome sequence of SARS-CoV-2 is 82% similar to SARS-COV-1. The compelling evidences link the progression of viral infection of SARS-CoV-2 with excessive inflammation as a result of the exaggerated immune response and elevated production of "immunocytokines" resulting in cytokine storm (CS); followed by a series of events, like acute organ damage, acute respiratory distress syndrome (ARDS) as well as death. Hence attempts to reduce cytokine storm are now being considered as a new paradigm shift in the clinical management of SARS-CoV-2. Tocilizumab (IL-6 blocker), Baricitinib (JAKs and AAK1 inhibitor), TNFα inhibitors (Infliximab, Adalimumab, Certolizumab) are currently being evaluated for possible block of the CS. Hence, rationalizing anti-inflammatory therapeutics would be the most judicious approach for significant reduction in COVID-19 mortality. In order to elucidate optimized and rationaled use of different therapeutics in COVID-19, we collated latest available information from emerging scientific evidences, integrated previous attempts as well as clinical successes, and various adopted approaches to mitigate past outbreaks with of SARS-CoV and MERS CoV.
Collapse
Affiliation(s)
- Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam 31441, Saudi Arabia
| | - Tareq Abu-Izneid
- Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Abdallah Mohammad Ibrahim
- Fundamentals of Nursing Department, College of Nursing, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam 31441, Saudi Arabia
| | - Md. Noushad Javed
- Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New-Delhi, India
| | - Noora AlHajri
- Department of Epidemiology and Population Health, College of Medicine, Khalifa University, United Arab Emirates
| | - Amar M. Hamrouni
- Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, Abu Dhabi, United Arab Emirates
| |
Collapse
|
12
|
Feng X, Yu W, Cao L, Meng F, Cong M. A novel chrysin thiazole derivative polarizes macrophages to an M1 phenotype via targeting TLR4. Int Immunopharmacol 2020; 88:106986. [PMID: 33182070 DOI: 10.1016/j.intimp.2020.106986] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022]
Abstract
Tumor-associated macrophages (TAMs) are an important cause of tumorigenesis and tumor development. M2 macrophages can promote tumor growth while M1 macrophages kill tumor cells, therefore, polarizing macrophages to achieve a functional M1 phenotype could effectively play its anti-tumor role. In the current study, we synthesized a novel chrysin derivative which is termed as ChR-TD. And we found ChR-TD might be a ligand of TLR4 that polarized the TAMs towards M1 phenotype and played its anti-tumor role. Further study indicated that ChR-TD reprogrammed the macrophages into an M1 phenotype via TLR4 activation. Moreover, ChR-TD activated TLR4/NF-κB signaling pathway and promoted the NF-κB/p65 translocated into the nuclear, leading to the activation of NF-κB and proinflammatory cytokines release. In addition, type I interferon signaling was also activated by ChR-TD, leading to the expressions of IFN-α and IFN-β and its targeted genes NOS2, MCP-1 and IP-10 were significantly increased in macrophages. Importantly, these effects were disturbed in TLR4-/- macrophages, which are constructed by using CRISPR/Cas9 system. And the molecule docking simulation further indicated that ChR-TD could bind to TLR4 and might be a ligand of TLR4. Hence, these findings suggested that ChR-TD might be a ligand of TLR4 and can be used as a potential lead compound for tumors treatment.
Collapse
Affiliation(s)
- Xiujing Feng
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China.
| | - Wen Yu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210046, China
| | - Lingsen Cao
- Key Lab of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210093, China
| | - Fanda Meng
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China
| | - Mulin Cong
- Institute of Basic Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong First Medical University, Jinan 250021, China
| |
Collapse
|
13
|
Hu Y, Smith D, Zhao Z, Harmon T, Pentel PR, Ehrich M, Zhang C. Alum as an adjuvant for nanoparticle based vaccines: A case study with a hybrid nanoparticle-based nicotine vaccine. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2019; 20:102023. [PMID: 31181264 PMCID: PMC6702048 DOI: 10.1016/j.nano.2019.102023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 05/13/2019] [Accepted: 05/16/2019] [Indexed: 12/14/2022]
Abstract
The treatment efficacy of a nicotine vaccine largely relies on its ability to induce high titers of nicotine-specific antibodies. Due to its strong immune-potentiating effects, aluminum salt (Alum) has been commonly used as an adjuvant in various nicotine vaccine formulations. In this study, we attempted to improve the immunological performance of a hybrid nanoparticle-based nicotine vaccine (NanoNicVac) by co-administering it with Alum. It was found that Alum severely restricted the release of NanoNicVac at the site of injection. Moreover, Alum damaged the hybrid structure of the vaccine. In the animal trial, mice immunized with NanoNicVac alone achieved an anti-nicotine IgG titer of 3.5 ± 0.2 × 104 after three injections. Unexpectedly, Alum with quantities of 125, 250, 500, and 1000 μg did not enhance the immunogenicity of NanoNicVac. In addition, Alum did not improve the ability of the vaccine to reduce the entry of nicotine into the brain.
Collapse
Affiliation(s)
- Yun Hu
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Daniel Smith
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Zongmin Zhao
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, USA
| | - Theresa Harmon
- Minneapolis Medical Research Foundation, Minneapolis, MN, USA
| | - Paul R Pentel
- Minneapolis Medical Research Foundation, Minneapolis, MN, USA
| | - Marion Ehrich
- Department of Biomedical Sciences and Pathobiology, Virginia Tech, Blacksburg, VA, USA
| | - Chenming Zhang
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
14
|
Lee MSJ, Natsume-Kitatani Y, Temizoz B, Fujita Y, Konishi A, Matsuda K, Igari Y, Tsukui T, Kobiyama K, Kuroda E, Onishi M, Marichal T, Ise W, Inoue T, Kurosaki T, Mizuguchi K, Akira S, Ishii KJ, Coban C. B cell-intrinsic MyD88 signaling controls IFN-γ-mediated early IgG2c class switching in mice in response to a particulate adjuvant. Eur J Immunol 2019; 49:1433-1440. [PMID: 31087643 DOI: 10.1002/eji.201848084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/27/2019] [Accepted: 05/13/2019] [Indexed: 02/01/2023]
Abstract
Adjuvants improve the potency of vaccines, but the modes of action (MOAs) of most adjuvants are largely unknown. TLR-dependent and -independent innate immune signaling through the adaptor molecule MyD88 has been shown to be pivotal to the effects of most adjuvants; however, MyD88's involvement in the TLR-independent MOAs of adjuvants is poorly understood. Here, using the T-dependent antigen NIPOVA and a unique particulate adjuvant called synthetic hemozoin (sHZ), we show that MyD88 is required for early GC formation and enhanced antibody class-switch recombination (CSR) in mice. Using cell-type-specific MyD88 KO mice, we found that IgG2c class switching, but not IgG1 class switching, was controlled by B cell-intrinsic MyD88 signaling. Notably, IFN-γ produced by various cells including T cells, NK cells, and dendritic cells was the primary cytokine for IgG2c CSR and B-cell intrinsic MyD88 is required for IFN-γ production. Moreover, IFN-γ receptor (IFNγR) deficiency abolished sHZ-induced IgG2c production, while recombinant IFN-γ administration successfully rescued IgG2c CSR impairment in mice lacking B-cell intrinsic MyD88. Together, our results show that B cell-intrinsic MyD88 signaling is involved in the MOA of certain particulate adjuvants and this may enhance our specific understanding of how adjuvants and vaccines work.
Collapse
Affiliation(s)
- Michelle Sue Jann Lee
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Yayoi Natsume-Kitatani
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Burcu Temizoz
- Laboratory of Vaccine Science, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Yukiko Fujita
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Aki Konishi
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Kyoko Matsuda
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Yoshikatsu Igari
- ZENOAQ, Nippon Zenyaku Kogyo Co. Ltd., Koriyama, Fukushima, Japan
| | - Toshihiro Tsukui
- ZENOAQ, Nippon Zenyaku Kogyo Co. Ltd., Koriyama, Fukushima, Japan
| | - Kouji Kobiyama
- Laboratory of Vaccine Science, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan.,Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Etsushi Kuroda
- Laboratory of Vaccine Science, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan.,Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Motoyasu Onishi
- Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Thomas Marichal
- Laboratory of Cellular and Molecular Immunology, GIGA Institute, and Faculty of Veterinary Medicine, Liege University, Liège, Belgium
| | - Wataru Ise
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Takeshi Inoue
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Ken J Ishii
- Laboratory of Vaccine Science, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan.,Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Cevayir Coban
- Laboratory of Malaria Immunology, Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| |
Collapse
|
15
|
Lee H, Park H, Yu HS, Na K, Oh KT, Lee ES. Dendritic Cell-Targeted pH-Responsive Extracellular Vesicles for Anticancer Vaccination. Pharmaceutics 2019; 11:pharmaceutics11020054. [PMID: 30691225 PMCID: PMC6410067 DOI: 10.3390/pharmaceutics11020054] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/20/2022] Open
Abstract
Immunotherapy can potentially treat cancers on a patient-dependent manner. Most of the efforts expended on anticancer vaccination parallel the efforts expended on prototypical immunization in infectious diseases. In this study, we designed and synthesized pH-responsive extracellular vesicles (EVs) coupled with hyaluronic acid (HA), 3-(diethylamino)propylamine (DEAP), monophosphoryl lipid A (MPLA), and mucin 1 peptide (MUC1), referred to as HDEA@EVAT. HDEA@EVAT potentiated the differentiation and maturation of monocytes into dendritic cells (DCs) and the priming of CD8+ T-cells for cancer therapy. MPLA and HA enabled HDEA@EVAT to interact with the toll-like receptor 4 and the CD44 receptor on DCs, followed by endosomal escape, owing to the protonation of pH-sensitive DEAP on the EV in conjunction with MUC1 release. The MUC1 was then processed and presented to DCs to activate CD8+ T-cells for additional anticancer-related immune reactions. Our findings support the anticancer vaccine activity by which HDEA@EVAT expedites the interaction between DCs and CD8+ T-cells by inducing DC-targeted maturation and by presenting the cancer-associated peptide MUC1.
Collapse
Affiliation(s)
- Hyuk Lee
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Korea.
| | - Hongsuk Park
- Division of Endocrinology, Metabolism & Lipid Research, Washington University School of Medicine, Saint Louis, MO 63110, USA.
| | - Hyeong Sup Yu
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Korea.
| | - Kun Na
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Korea.
| | - Kyung Taek Oh
- College of Pharmacy, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul 06974, Korea.
| | - Eun Seong Lee
- Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon-si, Gyeonggi-do 14662, Korea.
| |
Collapse
|
16
|
Matsuo K, Kitahata K, Kawabata F, Kamei M, Hara Y, Takamura S, Oiso N, Kawada A, Yoshie O, Nakayama T. A Highly Active Form of XCL1/Lymphotactin Functions as an Effective Adjuvant to Recruit Cross-Presenting Dendritic Cells for Induction of Effector and Memory CD8 + T Cells. Front Immunol 2018; 9:2775. [PMID: 30542351 PMCID: PMC6277777 DOI: 10.3389/fimmu.2018.02775] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/12/2018] [Indexed: 12/21/2022] Open
Abstract
The chemokine receptor XCR1 is known to be selectively expressed by cross-presenting dendritic cells (DCs), while its ligand XCL1/lymphotactin is mainly produced by activated CD8+ T cells and natural killer cells. Recent studies have shown that XCL1-antigen fusion proteins efficiently induce CD8+ T cell responses by preferentially delivering antigens to XCR1+ DCs. However, XCL1 per se was found to be a poor adjuvant for induction of CD8+ T cell responses. XCL1 is unique because of its lack of one of the two disulfide bonds commonly conserved in all other chemokines and thus has an unstable structure with a relatively weak chemokine activity. In the present study, we generated a variant form of murine XCL1 termed mXCL1-V21C/A59C that contained a second disulfide bond to stabilize its chemokine structure. We confirmed that mXCL1-V21C/A59C had much more potent chemotactic and calcium mobilization activities than the wild type XCL1 (mXCL1-WT). Intradermal injection of mXCL1-V21C/A59C, but not that of mXCL1-WT, significantly increased the accumulation of XCR1+CD103+ DCs in the injection site, and most of the accumulated XCR1+CD103+ DCs were found to take up co-injected ovalbumin (OVA). Furthermore, recruited XCR1+CD103+ DCs efficiently migrated to the draining lymph nodes and stayed for a prolonged period of time. Consequently, mXCL1-V21C/A59C strongly induced OVA-specific CD8+ T cells. The combination of OVA and mXCL1-V21C/A59C well protected mice from E.G7-OVA tumor growth in both prophylactic and therapeutic protocols. Finally, memory CTL responses were efficiently induced in mice immunized with OVA and mXCL1-V21C/A59C. Although intradermal injection of OVA and polyinosinic-polycytidylic acid (poly(I:C)) as an adjuvant also induced CD8+ T cell responses to OVA, poly (I:C) poorly recruited XCR1+CD103+ DCs in the injection site and failed to induce significant memory CTL responses to OVA. Collectively, our findings demonstrate that a highly active form of XCL1 is a promising vaccine adjuvant for cross-presenting DCs to induce antigen-specific effector and memory CD8+ T cells.
Collapse
Affiliation(s)
- Kazuhiko Matsuo
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Osaka, Japan
| | - Kosuke Kitahata
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Osaka, Japan
| | - Fumika Kawabata
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Osaka, Japan
| | - Momo Kamei
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Osaka, Japan
| | - Yuta Hara
- Laboratory of Cell Biology, Kindai University Faculty of Pharmacy, Osaka, Japan
| | - Shiki Takamura
- Department of Immunology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Naoki Oiso
- Department of Dermatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Akira Kawada
- Department of Dermatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Osamu Yoshie
- Kindai University, Osaka, Japan.,The Health and Kampo Institute, Miyagi, Japan
| | - Takashi Nakayama
- Division of Chemotherapy, Kindai University Faculty of Pharmacy, Osaka, Japan
| |
Collapse
|
17
|
Malik A, Gupta M, Gupta V, Gogoi H, Bhatnagar R. Novel application of trimethyl chitosan as an adjuvant in vaccine delivery. Int J Nanomedicine 2018; 13:7959-7970. [PMID: 30538470 PMCID: PMC6260144 DOI: 10.2147/ijn.s165876] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The application of natural carbohydrate polysaccharides for antigen delivery and its adjuvanation potential has garnered interest in the scientific community in the recent years. These biomaterials are considered favorable candidates for adjuvant development due to their desirable properties like enormous bioavailability, non-toxicity, biodegradability, stability, affordability, and immunostimulating ability. Chitosan is the one such extensively studied natural polymer which has been appreciated for its excellent applications in pharmaceuticals. Trimethyl chitosan (TMC), a derivative of chitosan, possesses these properties. In addition it has the properties of high aqueous solubility, high charge density, mucoadhesive, permeation enhancing (ability to cross tight junction), and stability over a range of ionic conditions which makes the spectrum of its applicability much broader. It has also been seen to perform analogously to alum, complete Freund’s adjuvant, incomplete Freund’s adjuvant, and cyclic guanosine monophosphate adjuvanation, which justifies its role as a potent adjuvant. Although many review articles detailing the applications of chitosan in vaccine delivery are available, a comprehensive review of the applications of TMC as an adjuvant is not available to date. This article provides a comprehensive overview of structural and chemical properties of TMC which affect its adjuvant characteristics; the efficacy of various delivery routes for TMC antigen combination; and the recent advances in the elucidation of its mechanism of action.
Collapse
Affiliation(s)
- Anshu Malik
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Manish Gupta
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Vatika Gupta
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Himanshu Gogoi
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| |
Collapse
|
18
|
Li Z, Ding W, Guo Q, Liu Z, Zhu Z, Song S, Li W, Liao G. Analysis of the dose-sparing effect of adjuvanted Sabin-inactivated poliovirus vaccine (sIPV). Hum Vaccin Immunother 2018; 14:1987-1994. [PMID: 29601259 PMCID: PMC6150041 DOI: 10.1080/21645515.2018.1454571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Sabin-based inactivated poliovirus vaccine(sIPV) is gradually replacing live-attenuated oral polio vaccine(OPV). Sabin-inactivated poliovirus vaccine(sIPV) has played a vital role in reducing economic burden of poliomyelitis and maintaining appropriate antibody levels in the population. However, due to its high cost and limited manufacturing capacity, sIPV cannot reach its full potential for global poliovirus eradication in developing countries. Therefore, to address this situation, we designed this study to evaluate the dose-sparing effects of AS03, CpG oligodeoxynucleotides (CpG-ODN) and polyinosinic:polycytidylic acid (PolyI:C) admixed with sIPV in rats. Our results showed that a combination of 1/4-dose sIPV adjuvanted with AS03 or AS03 with BW006 provides a seroconversion rate similar to that of full-dose sIPV without adjuvant and that, this rate is 5-fold higher than that of 1/4-dose sIPV without adjuvant after the first immunization. The combination of AS03 or AS03 with BW006 as an adjuvant effectively reduced sIPV dose by at least 4-fold and induced both humoral and cellular immune responses. Therefore, our study revealed that the combination of AS03 or AS03 with BW006 is a promising adjuvant for sIPV development.
Collapse
Affiliation(s)
- Zhuofan Li
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Wenting Ding
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Qi Guo
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Ze Liu
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Zhe Zhu
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Shaohui Song
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Weidong Li
- b The Department of Production Administration , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| | - Guoyang Liao
- a The fifth Department of Biological products , Institute of Medical Biology, Chinese Academy of Medical Science and Peking Union Medical College , Kunming , People's Republic of China
| |
Collapse
|
19
|
Haug M, Brede G, Håkerud M, Nedberg AG, Gederaas OA, Flo TH, Edwards VT, Selbo PK, Høgset A, Halaas Ø. Photochemical Internalization of Peptide Antigens Provides a Novel Strategy to Realize Therapeutic Cancer Vaccination. Front Immunol 2018; 9:650. [PMID: 29670624 PMCID: PMC5893651 DOI: 10.3389/fimmu.2018.00650] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 03/16/2018] [Indexed: 12/30/2022] Open
Abstract
Effective priming and activation of tumor-specific CD8+ cytotoxic T lymphocytes (CTLs) is crucial for realizing the potential of therapeutic cancer vaccination. This requires cytosolic antigens that feed into the MHC class I presentation pathway, which is not efficiently achieved with most current vaccination technologies. Photochemical internalization (PCI) provides an emerging technology to route endocytosed material to the cytosol of cells, based on light-induced disruption of endosomal membranes using a photosensitizing compound. Here, we investigated the potential of PCI as a novel, minimally invasive, and well-tolerated vaccination technology to induce priming of cancer-specific CTL responses to peptide antigens. We show that PCI effectively promotes delivery of peptide antigens to the cytosol of antigen-presenting cells (APCs) in vitro. This resulted in a 30-fold increase in MHC class I/peptide complex formation and surface presentation, and a subsequent 30- to 100-fold more efficient activation of antigen-specific CTLs compared to using the peptide alone. The effect was found to be highly dependent on the dose of the PCI treatment, where optimal doses promoted maturation of immature dendritic cells, thus also providing an adjuvant effect. The effect of PCI was confirmed in vivo by the successful induction of antigen-specific CTL responses to cancer antigens in C57BL/6 mice following intradermal peptide vaccination using PCI technology. We thus show new and strong evidence that PCI technology holds great potential as a novel strategy for improving the outcome of peptide vaccines aimed at triggering cancer-specific CD8+ CTL responses.
Collapse
Affiliation(s)
- Markus Haug
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway.,Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, Trondheim, Norway.,Department of Infection, St. Olavs University Hospital, Trondheim, Norway
| | - Gaute Brede
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| | - Monika Håkerud
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway
| | - Anne Grete Nedberg
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway
| | - Odrun A Gederaas
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway.,Department of Chemistry, Faculty of Natural Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Trude H Flo
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway.,Centre of Molecular Inflammation Research (CEMIR), Norwegian University of Science and Technology, Trondheim, Norway.,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, Oslo University Hospital, Oslo, Norway
| | - Victoria T Edwards
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway.,PCI Biotech AS, Oslo, Norway
| | - Pål K Selbo
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital - The Norwegian Radium Hospital, Oslo, Norway
| | | | - Øyvind Halaas
- Department of Clinical and Molecular Medicine (IKOM), Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
20
|
Lapenta F, Aupič J, Strmšek Ž, Jerala R. Coiled coil protein origami: from modular design principles towards biotechnological applications. Chem Soc Rev 2018; 47:3530-3542. [DOI: 10.1039/c7cs00822h] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
This review illustrates the current state in designing coiled-coil-based proteins with an emphasis on coiled coil protein origami structures and their potential.
Collapse
Affiliation(s)
- Fabio Lapenta
- Department of Synthetic Biology and Immunology
- National Institute of Chemistry
- Ljubljana
- Slovenia
| | - Jana Aupič
- Department of Synthetic Biology and Immunology
- National Institute of Chemistry
- Ljubljana
- Slovenia
| | - Žiga Strmšek
- Department of Synthetic Biology and Immunology
- National Institute of Chemistry
- Ljubljana
- Slovenia
| | - Roman Jerala
- Department of Synthetic Biology and Immunology
- National Institute of Chemistry
- Ljubljana
- Slovenia
- EN-FIST Centre of Excellence
| |
Collapse
|
21
|
Abstract
Vaccination is a biological process that administrates antigenic materials to stimulate an individual's immune system to develop immunity to a specific pathogen. It is the most effective tool to prevent illness and death from infectious diseases or diseases leading to cancers. Because many recombinant and synthetic antigens are poorly immunogenic, adjuvant is essentially added to vaccine formula that can potentiate the immune responses, offer better protection against pathogens and reduce the amount of antigens needed for protective immunity. To date, there are nearly 100 different types of adjuvants associated with about 400 vaccines that are either commercially available or under development. Among these adjuvants, many of them are particulates and nano-scale in nature. Nanoparticles represent a wide range of materials with novel physicochemical properties that exhibit immunostimulatory effects. However, the mechanistic understandings on how their physicochemical properties affect immunopotentiation remain elusive. In this article, we aim to review current development status of nanomaterial-based vaccine adjuvants, and further discuss their acting mechanisms, understanding of which will benefit the rational design of effective vaccine adjuvants with improved immunogenicity for prevention of infectious disease as well as therapeutic cancer treatment.
Collapse
Affiliation(s)
- Bingbing Sun
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, California, 90095, United States
- Center for Environmental Implications of Nanotechnology (CEIN), California NanoSystems Institute (CNSI), University of California, Los Angeles, California, 90095, United States
- School of Chemical Engineering, Dalian University of Technology, 2 Linggong Road, 116024, Dalian, China
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine; University of California, Los Angeles, California, 90095, United States
- Center for Environmental Implications of Nanotechnology (CEIN), California NanoSystems Institute (CNSI), University of California, Los Angeles, California, 90095, United States
| |
Collapse
|
22
|
Santone M, Aprea S, Wu TYH, Cooke MP, Mbow ML, Valiante NM, Rush JS, Dougan S, Avalos A, Ploegh H, De Gregorio E, Buonsanti C, D'Oro U. A new TLR2 agonist promotes cross-presentation by mouse and human antigen presenting cells. Hum Vaccin Immunother 2016; 11:2038-50. [PMID: 26024409 DOI: 10.1080/21645515.2015.1027467] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cross-presentation is the process by which professional APCs load peptides from an extracellularly derived protein onto class I MHC molecules to trigger a CD8(+) T cell response. The ability to enhance this process is therefore relevant for the development of antitumor and antiviral vaccines. We investigated a new TLR2-based adjuvant, Small Molecule Immune Potentiator (SMIP) 2.1, for its ability to stimulate cross-presentation. Using OVA as model antigen, we demonstrated that a SMIP2.1-adjuvanted vaccine formulation induced a greater CD8(+) T cell response, in terms of proliferation, cytokine production and cytolytic activity, than a non-adjuvanted vaccine. Moreover, using an OVA-expressing tumor model, we showed that the CTLs induced by the SMIP2.1 formulated vaccine inhibits tumor growth in vivo. Using a BCR transgenic mouse model we found that B cells could cross-present the OVA antigen when stimulated with SMIP2.1. We also used a flow cytometry assay to detect activation of human CD8(+) T cells isolated from human PBMCs of cytomegalovirus-seropositive donors. Stimulation with SMIP2.1 increased the capacity of human APCs, pulsed in vitro with the pp65 CMV protein, to activate CMV-specific CD8(+) T cells. Therefore, vaccination with an exogenous antigen formulated with SMIP2.1 is a successful strategy for the induction of a cytotoxic T cell response along with antibody production.
Collapse
Key Words
- APC, antigen presenting cell
- B cells
- BCR, B cell receptor
- CMV, cytomegalovirus
- CTL, cytotoxic t lymphocyte
- DC, dendritic cell
- HCMV, human CMV
- KO, knock out
- LN, lymph node
- MHC, major histocompatibility complex
- OVA, avalbumin
- PBMC, peripheral blood mononuclear cell
- SMIP, Small Molecule Immune Potentiator
- TLR, toll like receptor
- cross presentation/priming
- cytotoxic T cells
- dendritic cells
- vaccination
Collapse
Affiliation(s)
- Melissa Santone
- a Novartis Vaccines and Diagnostics s.r.l. (a GlaxoSmithKline Company) ; Siena , Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
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
|
24
|
Wang H, Luo Z, Wang Y, He T, Yang C, Ren C, Ma L, Gong C, Li X, Yang Z. Enzyme-Catalyzed Formation of Supramolecular Hydrogels as Promising Vaccine Adjuvants. ADVANCED FUNCTIONAL MATERIALS 2016. [DOI: 10.1002/adfm.201505188] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Huaimin Wang
- State Key Laboratory of Medicinal Chemical Biology; Key Laboratory of Bioactive MaterialsMinistry of Education; College of Life Sciences; Nankai University and Collaborative Innovation Centerof Chemical Science and Engineering; Tianjin 300071 P. R. China
| | - Zichao Luo
- Institute of Biomaterials and Engineering; Wenzhou Medical University; Wenzhou 325035 P. R. China
- Wenzhou Institute of Biomaterials and Engineering; Wenzhou 325035 P. R. China
| | - Youzhi Wang
- State Key Laboratory of Medicinal Chemical Biology; Key Laboratory of Bioactive MaterialsMinistry of Education; College of Life Sciences; Nankai University and Collaborative Innovation Centerof Chemical Science and Engineering; Tianjin 300071 P. R. China
| | - Tao He
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University, and Collaborative Innovation Center for Biotherapy; Chengdu 610041 P. R. China
| | - Chengbiao Yang
- State Key Laboratory of Medicinal Chemical Biology; Key Laboratory of Bioactive MaterialsMinistry of Education; College of Life Sciences; Nankai University and Collaborative Innovation Centerof Chemical Science and Engineering; Tianjin 300071 P. R. China
| | - Chunhua Ren
- State Key Laboratory of Medicinal Chemical Biology; Key Laboratory of Bioactive MaterialsMinistry of Education; College of Life Sciences; Nankai University and Collaborative Innovation Centerof Chemical Science and Engineering; Tianjin 300071 P. R. China
| | - Linsha Ma
- State Key Laboratory of Medicinal Chemical Biology; Key Laboratory of Bioactive MaterialsMinistry of Education; College of Life Sciences; Nankai University and Collaborative Innovation Centerof Chemical Science and Engineering; Tianjin 300071 P. R. China
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center; West China Hospital; Sichuan University, and Collaborative Innovation Center for Biotherapy; Chengdu 610041 P. R. China
| | - Xingyi Li
- Institute of Biomaterials and Engineering; Wenzhou Medical University; Wenzhou 325035 P. R. China
- Wenzhou Institute of Biomaterials and Engineering; Wenzhou 325035 P. R. China
| | - Zhimou Yang
- State Key Laboratory of Medicinal Chemical Biology; Key Laboratory of Bioactive MaterialsMinistry of Education; College of Life Sciences; Nankai University and Collaborative Innovation Centerof Chemical Science and Engineering; Tianjin 300071 P. R. China
| |
Collapse
|
25
|
Nishida S, Sugiyama H. Immunotherapy Targeting WT1: Designing a Protocol for WT1 Peptide-Based Cancer Vaccine. Methods Mol Biol 2016; 1467:221-232. [PMID: 27417973 DOI: 10.1007/978-1-4939-4023-3_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
There is much current excitement about the potential of cancer immunotherapy. WT1 is high on the National Cancer Institute's list of priority antigens for immune therapy. In this chapter we describe a protocol for a clinical trial using a WT1 peptide-based cancer vaccine.
Collapse
Affiliation(s)
- Sumiyuki Nishida
- Department of Respiratory Medicine, Allergy and Rheumatic Disease, Graduate School of Medicine, Osaka University, 2-2, Yamada-Oka, Suita-City, Osaka, 565-0871, Japan.
| | - Haruo Sugiyama
- Department of Functional Diagnostic Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
26
|
Mammadov R, Cinar G, Gunduz N, Goktas M, Kayhan H, Tohumeken S, Topal AE, Orujalipoor I, Delibasi T, Dana A, Ide S, Tekinay AB, Guler MO. Virus-like nanostructures for tuning immune response. Sci Rep 2015; 5:16728. [PMID: 26577983 PMCID: PMC4649742 DOI: 10.1038/srep16728] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/19/2015] [Indexed: 11/10/2022] Open
Abstract
Synthetic vaccines utilize viral signatures to trigger immune responses. Although the immune responses raised against the biochemical signatures of viruses are well characterized, the mechanism of how they affect immune response in the context of physical signatures is not well studied. In this work, we investigated the ability of zero- and one-dimensional self-assembled peptide nanostructures carrying unmethylated CpG motifs (signature of viral DNA) for tuning immune response. These nanostructures represent the two most common viral shapes, spheres and rods. The nanofibrous structures were found to direct immune response towards Th1 phenotype, which is responsible for acting against intracellular pathogens such as viruses, to a greater extent than nanospheres and CpG ODN alone. In addition, nanofibers exhibited enhanced uptake into dendritic cells compared to nanospheres or the ODN itself. The chemical stability of the ODN against nuclease-mediated degradation was also observed to be enhanced when complexed with the peptide nanostructures. In vivo studies showed that nanofibers promoted antigen-specific IgG production over 10-fold better than CpG ODN alone. To the best of our knowledge, this is the first report showing the modulation of the nature of an immune response through the shape of the carrier system.
Collapse
Affiliation(s)
- Rashad Mammadov
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey
| | - Goksu Cinar
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey
| | - Nuray Gunduz
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey
| | - Melis Goktas
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey
| | - Handan Kayhan
- Adult Hematology Laboratory, School of Medicine, Gazi University, Ankara 06500, Turkey
| | - Sehmus Tohumeken
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey
| | - Ahmet E Topal
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey
| | - Ilghar Orujalipoor
- Department of Physics Engineering, Hacettepe University, 06800, Ankara, Turkey
| | - Tuncay Delibasi
- Hacettepe University and Diskapi Research and Training Hospital, 06800, Ankara, Turkey
| | - Aykutlu Dana
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey
| | - Semra Ide
- Department of Physics Engineering, Hacettepe University, 06800, Ankara, Turkey
| | - Ayse B Tekinay
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey
| | - Mustafa O Guler
- Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center (UNAM), Bilkent University, Ankara 06800, Turkey
| |
Collapse
|
27
|
Salmonella flagellin is a potent carrier-adjuvant for peptide conjugate to induce peptide-specific antibody response in mice. Vaccine 2015; 33:2038-44. [PMID: 25765964 DOI: 10.1016/j.vaccine.2015.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 02/13/2015] [Accepted: 03/02/2015] [Indexed: 11/24/2022]
Abstract
As an agonist to innate immune system, Salmonella flagellin has been proven to be a potent adjuvant either admixed or genetically fused with antigens and applied to a variety of vaccines against infectious diseases. However, relatively little is known about its carrier-adjuvant effect for conjugate vaccine. Conjugation is an effective approach often used to make haptens such as some peptides and polysaccharides immunogenic and in some cases used to make poor immunogens more immunogenic. In the current study, Salmonella flagellin was tested for its carrier-adjuvant effect in a peptide conjugation. The recombinant Salmonella flagellin (rFliC) purified from Escherichia coli was firstly modified by maleimide groups, then coupled with a synthetic peptide (EXP153:CDNNLVSGP) that is a B-cell epitope derived from Plasmodium falciparum exported protein-1 to generate the conjugate of EXP153-rFliC. Bioactivity assay showed that both chemical modification and conjugation did not apparently impair the TLR5-ligand activity of rFliC. EXP153-rFliC was used to immunize BALB/c mice via subcutaneous route, and the sera obtained from immunized mice were examined by ELISA and IFA. While no detectable antibody responses were induced by the peptide admixed with rFliC, the robust peptide-specific antibody responses were observed in mice immunized with the peptide conjugated to rFliC in the absence of any additional adjuvant. The immune sera induced by the conjugate recognized the native protein of malaria parasite. The data obtained from this study demonstrate the carrier-adjuvant activity of Salmonella flagellin in peptide conjugate immunization and indicate its promising application for conjugate vaccine research and development.
Collapse
|
28
|
Quiroga D, Aldhamen YA, Appledorn DM, Godbehere S, Amalfitano A. Strengthened tumor antigen immune recognition by inclusion of a recombinant Eimeria antigen in therapeutic cancer vaccination. Cancer Immunol Immunother 2015; 64:479-91. [PMID: 25655760 DOI: 10.1007/s00262-015-1659-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Accepted: 01/16/2015] [Indexed: 12/17/2022]
Abstract
The need for novel, effective adjuvants that are capable of eliciting stronger cellular and humoral adaptive immune responses to antigenic targets is well understood in the vaccine development field. Unfortunately, many adjuvants investigated thus far are either too toxic for human application or too weak to induce a substantial response against difficult antigens, such as tumor-associated antigens (TAAs). In spite of this trend, clinical investigations of recombinant Eimeria antigen (rEA) have revealed this protein to be a non-toxic immunogenic agent with the ability to trigger a Th1-predominant response in both murine and human subjects. Our past studies have shown that the injection of a rEA-encoding adenovirus (rAd5-rEA) alongside an HIV antigen-encoding adenovirus greatly improves the adaptive immune response against this pathogen-derived transgene. In this report, we investigated whether rAd5-rEA could promote and/or alter cytotoxic memory responses toward carcinoembryonic antigen (CEA), a colorectal cancer-related TAA. We found that the addition of rAd5-rEA to an Ad-based CEA vaccine induced a dose-dependent increase in several anti-CEA T and B cell responses. Moreover, inclusion of rAd5-rEA increased the number of CEA-derived antigenic epitopes that elicited significant cell-mediated and IgG-mediated recognition. These enhanced anti-CEA immune responses also translated into superior CEA-targeted cell killing, as evaluated by an in vivo cytotoxic T lymphocyte assay. Overall, these results suggest that co-administration of rAd5-rEA with a tumor antigen vaccine can substantially boost and broaden the TAA-specific adaptive memory response, thereby validating the potential of rAd5-rEA to be a beneficial adjuvant during therapeutic cancer vaccination.
Collapse
Affiliation(s)
- Dionisia Quiroga
- Department of Microbiology and Molecular Genetics, Michigan State University, 567 Wilson Road, 4194 Biomedical and Physical Sciences Building, East Lansing, MI, 48824, USA
| | | | | | | | | |
Collapse
|
29
|
Rodriguez Garcia M, Patel MV, Shen Z, Fahey JV, Biswas N, Mestecky J, Wira CR. Mucosal Immunity in the Human Female Reproductive Tract. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00108-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Basto AP, Badenes M, Almeida SCP, Martins C, Duarte A, Santos DM, Leitão A. Immune response profile elicited by the model antigen ovalbumin expressed in fusion with the bacterial OprI lipoprotein. Mol Immunol 2014; 64:36-45. [PMID: 25467796 DOI: 10.1016/j.molimm.2014.10.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/23/2014] [Accepted: 10/25/2014] [Indexed: 12/11/2022]
Abstract
The use of immunogenic formulations targeting pattern recognition receptors towards modulation of immune responses is a promising strategy to develop better vaccines against infectious and malignant diseases. Molecules targeting TLR2 offer interesting properties that are relevant for vaccine development, including the possibility to covalently attach the lipidic ligands to the antigens. However, the type of immune response elicited by these formulations is still controversial. In this work, we used the model antigen ovalbumin (OVA) expressed in fusion with the bacterial lipoprotein OprI in order to characterize the immunomodulatory properties of this TLR ligand. Murine bone marrow-derived dendritic cells stimulated with OprI-OVA fusion lipoprotein produced high levels of the pro-inflammatory cytokines TNF-α and IL-6 and also IL-10, IL-12(p70) and IL-27, while TGF-β and IL-23 were not detected. Using OT-II and OT-I mice, an enhancement of MHC class II and class I antigen presentation was observed for the OVA antigen in fusion with OprI. Mice immunized by intraperitoneal route with this fusion lipoprotein in prime-boost protocols developed strong specific antibody responses including IgG1, IgG2c, IgG2b, IgG3 and IgE. These results, together with data obtained by restimulation of splenocytes from the immunized mice, point to an immune response profile that does not correspond to a strict Th1 or Th2 polarization. Finally, in a challenge experiment using a melanoma syngeneic mouse model (B16-OVA), prophylactic inoculation with OprI fused with the unrelated antigen eGFP increased the tumor growth, while the fusion with the tumor-associated antigen OVA delayed the tumor growth and increased mice survival.
Collapse
Affiliation(s)
- Afonso P Basto
- CIISA, Faculdade de Medicina Veterinária, ULisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal.
| | - Marina Badenes
- CIISA, Faculdade de Medicina Veterinária, ULisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
| | - Sílvia C P Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Carlos Martins
- CIISA, Faculdade de Medicina Veterinária, ULisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - António Duarte
- CIISA, Faculdade de Medicina Veterinária, ULisboa, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; Instituto Gulbenkian de Ciência, 2781-901 Oeiras, Portugal
| | - Dulce M Santos
- Instituto de Investigação Científica Tropical, CVZ, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; CIISA, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Alexandre Leitão
- Instituto de Investigação Científica Tropical, CVZ, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal; CIISA, Avenida da Universidade Técnica, 1300-477 Lisboa, Portugal.
| |
Collapse
|
31
|
Neeland MR, Elhay MJ, Meeusen ENT, de Veer MJ. Vaccination with liposomal poly(I:C) induces discordant maturation of migratory dendritic cell subsets and anti-viral gene signatures in afferent lymph cells. Vaccine 2014; 32:6183-92. [PMID: 25280435 DOI: 10.1016/j.vaccine.2014.09.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 09/18/2014] [Accepted: 09/19/2014] [Indexed: 01/14/2023]
Abstract
Vaccine formulations administered in the periphery must activate naive immune cells within the lymph node. In this study, we have directly cannulated the ovine lymphatic vessels to investigate the cellular and molecular mechanisms that transfer information from the periphery into the local draining lymph node via the afferent lymph. Inclusion of poly(I:C) into a liposomal vaccine formulation enhances the neutrophil-associated inflammatory immune response in afferent lymph and increases antigen uptake by migratory dendritic cells (DCs). Interestingly, antigen positive migratory DCs undergo discordant maturation, with peak expression of CD86 at 4 h and CD80 at 48-72 h post vaccination. Afferent lymph monocytes up-regulate expression of genes related to inflammatory and anti-viral immune phenotypes following vaccination however show no differentiation into APCs prior to their migration to the local lymph node as measured by surface MHC II expression. Finally, this study reveals the addition of poly(I:C) increases systemic antigen-specific humoral immunity. These findings provide a detailed understanding of the real time in vivo immune response induced by liposomes incorporating the innate immune agonist poly(I:C) utilising a vaccination setting comparable to that administered in humans.
Collapse
Affiliation(s)
- Melanie R Neeland
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Wellington Road Clayton VIC 3800, Australia
| | - Martin J Elhay
- Zoetis Research and Manufacturing Australia P/L, 45 Poplar Road, Parkville VIC 3052, Australia
| | - Els N T Meeusen
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Wellington Road Clayton VIC 3800, Australia
| | - Michael J de Veer
- Biotechnology Research Laboratories, Department of Physiology, Monash University, Wellington Road Clayton VIC 3800, Australia.
| |
Collapse
|
32
|
DNA nanotechnology-based development of delivery systems for bioactive compounds. Eur J Pharm Sci 2014; 58:26-33. [DOI: 10.1016/j.ejps.2014.03.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/04/2014] [Accepted: 03/12/2014] [Indexed: 12/25/2022]
|
33
|
Targeting TLR2 for vaccine development. J Immunol Res 2014; 2014:619410. [PMID: 25057505 PMCID: PMC4098989 DOI: 10.1155/2014/619410] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/16/2014] [Accepted: 06/04/2014] [Indexed: 02/07/2023] Open
Abstract
Novel and more effective immunization strategies against many animal diseases may profit from the current knowledge on the modulation of specific immunity through stimulation of innate immune receptors. Toll-like receptor (TLR)2-targeting formulations, such as synthetic lipopeptides and antigens expressed in fusion with lipoproteins, have been shown to have built-in adjuvant properties and to be effective at inducing cellular and humoral immune mechanisms in different animal species. However, contradictory data has arisen concerning the profile of the immune response elicited. The benefits of targeting TLR2 for vaccine development are thus still debatable and more studies are needed to rationally explore its characteristics. Here, we resume the main features of TLR2 and TLR2-induced immune responses, focusing on what has been reported for veterinary animals.
Collapse
|
34
|
Kennedy RB, Ovsyannikova IG, Lambert ND, Haralambieva IH, Poland GA. The personal touch: strategies toward personalized vaccines and predicting immune responses to them. Expert Rev Vaccines 2014; 13:657-69. [PMID: 24702429 DOI: 10.1586/14760584.2014.905744] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The impact of vaccines on public health and wellbeing has been profound. Smallpox has been eradicated, polio is nearing eradication, and multiple diseases have been eliminated from certain areas of the world. Unfortunately, we now face diseases such as hepatitis C, malaria or tuberculosis, as well as new and re-emerging pathogens for which we lack effective vaccines. Empirical approaches to vaccine development have been successful in the past, but may not be up to the current infectious disease challenges facing us. New, directed approaches to vaccine design, development, and testing need to be developed. Ideally these approaches will capitalize on cutting-edge technologies, advanced analytical and modeling strategies, and up-to-date knowledge of both pathogen and host. These approaches will pay particular attention to the causes of inter-individual variation in vaccine response in order to develop new vaccines tailored to the unique needs of individuals and communities within the population.
Collapse
|
35
|
Neeland MR, Meeusen EN, de Veer MJ. Afferent lymphatic cannulation as a model system to study innate immune responses to infection and vaccination. Vet Immunol Immunopathol 2014; 158:86-97. [DOI: 10.1016/j.vetimm.2013.01.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 01/09/2013] [Accepted: 01/10/2013] [Indexed: 12/28/2022]
|
36
|
Moser C, Amacker M, Zurbriggen R. Influenza virosomes as a vaccine adjuvant and carrier system. Expert Rev Vaccines 2014; 10:437-46. [DOI: 10.1586/erv.11.15] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
37
|
In vivo and in vitro characterization of the immune stimulating activity of the Neisserial porin PorB. PLoS One 2013; 8:e82171. [PMID: 24349212 PMCID: PMC3859593 DOI: 10.1371/journal.pone.0082171] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 10/30/2013] [Indexed: 12/22/2022] Open
Abstract
Vaccines play a vital role in modern medicine. The development of novel vaccines for emerging and resistant pathogens has been aided in recent years by the use of novel adjuvants in subunit vaccines. A deeper understanding of the molecular pathways behind adjuvanticity is required to better select immunostimulatory molecules for use in individual vaccines. To this end, we have undertaken a study of the essential signaling pathways involved in the innate and adaptive immune responses to the Neisseria meningitidis outer membrane protein Porin B (PorB). We have previously demonstrated that PorB is an agonist of Toll-Like Receptor 2 (TLR2) and acts as an adjuvant in vaccines for protein, carbohydrate and lipopolysaccharide antigens using murine models. Here we demonstrate NFκB translocation following stimulation with PorB only occurs in the presence of TLR2. IL-6 and TNF-α secretion was shown to be MAPK dependent. Surface expression of activation markers on macrophages, including CD40, CD69, and CD86, was increased following PorB stimulation in vitro. Interestingly, some upregulation of CD54 and CD69 was still observed in macrophages obtained from TLR2 KO mice, indicating a possible non-TLR2 mediated activation pathway induced by PorB. In a murine vaccination model, using ovalbumin as the antigen and PorB as the adjuvant, a decreased antigen-specific IgG production was observed in TLR2 KO mice; adjuvant-dependent increased IgG production was entirely ablated in MyD88 KO mice. These observations demonstrate the importance of the above pathways to the adjuvant activity of PorB. The potential TLR2 independent effect is currently being explored.
Collapse
|
38
|
Zhou W, Moguche AO, Chiu D, Murali-Krishna K, Baneyx F. Just-in-time vaccines: Biomineralized calcium phosphate core-immunogen shell nanoparticles induce long-lasting CD8(+) T cell responses in mice. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 10:571-8. [PMID: 24275478 DOI: 10.1016/j.nano.2013.11.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 11/09/2013] [Accepted: 11/13/2013] [Indexed: 11/16/2022]
Abstract
UNLABELLED Distributed and on-demand vaccine production could be game-changing for infectious disease treatment in the developing world by providing new therapeutic opportunities and breaking the refrigeration "cold chain". Here, we show that a fusion protein between a calcium phosphate binding domain and the model antigen ovalbumin can mineralize a biocompatible adjuvant in a single step. The resulting 50 nm calcium phosphate core-immunogen shell particles are comparable to soluble protein in inducing ovalbumin-specific antibody response and class switch recombination in mice. However, single dose vaccination with nanoparticles leads to higher expansion of ovalbumin-specific CD8(+) T cells upon challenge with an influenza virus bearing the ovalbumin-derived SIINFEKL peptide, and these cells produce high levels of IFN-γ. Furthermore, mice exhibit a robust antigen-specific CD8(+) T cell recall response when challenged with virus 8 months post-immunization. These results underscore the promise of immunogen-controlled adjuvant mineralization for just-in-time manufacturing of effective T cell vaccines. FROM THE CLINICAL EDITOR This paper reports that a fusion protein between a calcium phosphate binding domain and the model antigen ovalbumin can mineralize into a biocompatible adjuvant in a single step, enabling distributed and on-demand vaccine production and eliminating the need for refrigeration of vaccines. The findings highlight the possibility of immunogen-controlled adjuvant mineralization for just-in-time manufacturing of effective T cell vaccines.
Collapse
Affiliation(s)
- Weibin Zhou
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Albanus O Moguche
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - David Chiu
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | | | - François Baneyx
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA.
| |
Collapse
|
39
|
Vaccine efficacy of transcutaneous immunization with amyloid β using a dissolving microneedle array in a mouse model of Alzheimer's disease. J Neuroimmunol 2013; 266:1-11. [PMID: 24315156 DOI: 10.1016/j.jneuroim.2013.11.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 11/02/2013] [Accepted: 11/05/2013] [Indexed: 12/26/2022]
Abstract
Vaccine therapy for Alzheimer's disease (AD) based on the amyloid cascade hypothesis has recently attracted attention for treating AD. Injectable immunization using amyloid β peptide (Aβ) comprising 1-42 amino-acid residues (Aβ1-42) as antigens showed therapeutic efficacy in mice; however, the clinical trial of this injected Aβ1-42 vaccine was stopped due to the incidence of meningoencephalitis caused by excess activation of Th1 cells infiltrating the brain as a serious adverse reaction. Because recent studies have suggested that transcutaneous immunization (TCI) is likely to elicit Th2-dominant immune responses, TCI is expected to be effective in treating AD without inducing adverse reactions. Previously reported TCI procedures employed complicated and impractical vaccination procedures; therefore, a simple, easy-to-use, and novel TCI approach needs to be established. In this study, we investigated the vaccine efficacy of an Aβ1-42-containing TCI using our novel dissolving microneedle array (MicroHyala; MH) against AD. MH-based TCI induced anti-Aβ1-42 immune responses by simple and low-invasive application of Aβ1-42-containing MH to the skin. Unfortunately, this TCI system resulted in little significant improvement in cognitive function and Th2-dominant immune responses, suggesting the need for further modification.
Collapse
|
40
|
Padte NN, Boente-Carrera M, Andrews CD, McManus J, Grasperge BF, Gettie A, Coelho-dos-Reis JG, Li X, Wu D, Bruder JT, Sedegah M, Patterson N, Richie TL, Wong CH, Ho DD, Vasan S, Tsuji M. A glycolipid adjuvant, 7DW8-5, enhances CD8+ T cell responses induced by an adenovirus-vectored malaria vaccine in non-human primates. PLoS One 2013; 8:e78407. [PMID: 24205224 PMCID: PMC3808339 DOI: 10.1371/journal.pone.0078407] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/11/2013] [Indexed: 01/12/2023] Open
Abstract
A key strategy to a successful vaccine against malaria is to identify and develop new adjuvants that can enhance T-cell responses and improve protective immunity. Upon co-administration with a rodent malaria vaccine in mice, 7DW8-5, a recently identified novel analog of α-galactosylceramide (α-GalCer), enhances the level of malaria-specific protective immune responses more strongly than the parent compound. In this study, we sought to determine whether 7DW8-5 could provide a similar potent adjuvant effect on a candidate human malaria vaccine in the more relevant non-human primate (NHP) model, prior to committing to clinical development. The candidate human malaria vaccine, AdPfCA (NMRC-M3V-Ad-PfCA), consists of two non-replicating recombinant adenoviral (Ad) vectors, one expressing the circumsporozoite protein (CSP) and another expressing the apical membrane antigen-1 (AMA1) of Plasmodium falciparum. In several phase 1 clinical trials, AdPfCA was well tolerated and demonstrated immunogenicity for both humoral and cell-mediated responses. In the study described herein, 25 rhesus macaques received prime and boost intramuscular (IM) immunizations of AdPfCA alone or with an ascending dose of 7DW8-5. Our results indicate that 7DW8-5 is safe and well-tolerated and provides a significant enhancement (up to 9-fold) in malaria-specific CD8+ T-cell responses after both priming and boosting phases, supporting further clinical development.
Collapse
Affiliation(s)
- Neal N. Padte
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Mar Boente-Carrera
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Chasity D. Andrews
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Jenny McManus
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Brooke F. Grasperge
- Tulane National Primate Research Center, Tulane University Medical Center, Covington, Louisiana, United States of America
| | - Agegnehu Gettie
- Tulane National Primate Research Center, Tulane University Medical Center, Covington, Louisiana, United States of America
| | - Jordana G. Coelho-dos-Reis
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Xiangming Li
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Douglass Wu
- Department of Chemistry, the Scripps Research Institute, La Jolla, California, United States of America
| | - Joseph T. Bruder
- Research, GenVec, Inc., Gaithersburg, Maryland, United States of America
| | - Martha Sedegah
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Noelle Patterson
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, Maryland, United States of America
| | - Thomas L. Richie
- US Military Malaria Vaccine Program, Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Chi-Huey Wong
- Department of Chemistry, the Scripps Research Institute, La Jolla, California, United States of America
| | - David D. Ho
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
| | - Sandhya Vasan
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
- * E-mail: (SV); (MT)
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, United States of America
- * E-mail: (SV); (MT)
| |
Collapse
|
41
|
Demberg T, Brocca-Cofano E, Kuate S, Aladi S, Vargas-Inchaustegui DA, Venzon D, Kalisz I, Kalyanaraman V, Lee EM, Pal R, DiPasquale J, Ruprecht RM, Montefiori DC, Srivastava I, Barnett SW, Robert-Guroff M. Impact of antibody quality and anamnestic response on viremia control post-challenge in a combined Tat/Env vaccine regimen in rhesus macaques. Virology 2013; 440:210-21. [PMID: 23528732 PMCID: PMC3744165 DOI: 10.1016/j.virol.2013.02.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 12/03/2012] [Accepted: 02/27/2013] [Indexed: 11/18/2022]
Abstract
Previously, priming rhesus macaques with Adenovirus type 5 host range mutant-recombinants encoding Tat and Env and boosting with Tat and Env protein in MPL-SE controlled chronic viremia by 4 logs following homologous intravenous SHIV89.6P challenge. Here we evaluated Tat, Env, and Tat/Env regimens for immunogenicity and protective efficacy using clade C Env, alum adjuvant, and a heterologous intrarectal SHIV1157ipd3N4 challenge. Despite induction of strong cellular and humoral immunity, Tat/Env group T and B-cell memory responses were not significantly enhanced over Tat- or Env-only groups. Lack of viremia control post-challenge was attributed to lower avidity Env antibodies and no anamnestic ADCC response or SHIV1157ipd3N4 neutralizing antibody development post-challenge. Poor biologic activity of the Tat immunogen may have impaired Tat immunity. In the absence of sterilizing immunity, strong anamnestic responses to heterologous virus can help control viremia. Both antibody breadth and optimal adjuvanticity are needed to elicit high-quality antibody for protective efficacy.
Collapse
Affiliation(s)
- Thorsten Demberg
- Vaccine Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Seraphin Kuate
- Vaccine Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Stanley Aladi
- Vaccine Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - David Venzon
- Biostatistics and Data Management Section, National Cancer Institute, Bethesda, MD 20892, USA
| | - Irene Kalisz
- Advanced BioScience Laboratories, Inc., Kensington, MD 20895, USA
| | | | - Eun Mi Lee
- Advanced BioScience Laboratories, Inc., Kensington, MD 20895, USA
| | - Ranajit Pal
- Advanced BioScience Laboratories, Inc., Kensington, MD 20895, USA
| | - Janet DiPasquale
- Vaccine Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | |
Collapse
|
42
|
Chuai X, Chen H, Wang W, Deng Y, Wen B, Ruan L, Tan W. Poly(I:C)/alum mixed adjuvant priming enhances HBV subunit vaccine-induced immunity in mice when combined with recombinant adenoviral-based HBV vaccine boosting. PLoS One 2013; 8:e54126. [PMID: 23335993 PMCID: PMC3545998 DOI: 10.1371/journal.pone.0054126] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 12/06/2012] [Indexed: 12/30/2022] Open
Abstract
Background Virus-specific cellular immune responses play a critical role in virus clearance during acute or chronic HBV infection. Currently, the commercially available HBV vaccine is combined with alum adjuvant, which stimulates mainly Th2 immune responses. Therefore, development of new therapeutic HBV vaccine adjuvants and immune strategies that also promote Th1 and CTL responses is urgently needed. Methodology/Principal findings To improve the immunity induced by the novel HBSS1 HBV vaccine, we evaluated the ability of adjuvants, including alum, CpG and polyriboinosinic polyribocytidylic acid [poly(I:C)], to enhance the response when boosted with the recombinant adenoviral vector vaccine rAdSS1. The immune responses to different adjuvant combinations were assessed in C57BL/6 mice by enzyme-linked immunosorbent assay (ELISA), ELISpot and cytokine release assays. Among the combinations tested, a HBV protein particle vaccine with CpG/alum and poly(I:C)/alum priming combinations accelerated specific seroconversion and produced high antibody (anti-PreS1, anti-S antibody) titres with a Th1 bias. After boosting with recombinant adenoviral vector vaccine rAdSS1, both groups produced a strong multi-antigen (S and PreS1)-specific cellular immune response. HBSS1 immunisation with poly(I:C)/alum priming also generated high-level CD4+ and CD8+ T cell responses in terms of Th1 cytokines (IFN-γand IL-2). Conclusions The protein-vaccine HBSS1 with mixed poly(I:C)/alum adjuvant priming, followed by a rAdSS1 vaccine boost, maximises specific antibody and Th1-biased cellular immune responses. This regime might prove useful in the development of HBV therapeutic vaccines. Furthermore, this promising strategy might be applied to vaccines against other persistent infections, such as human immunodeficiency virus and tuberculosis.
Collapse
Affiliation(s)
- Xia Chuai
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
- Department of Microbiology, Hebei Medical University, Shijiazhuang, Heibei Province, People’s Republic of China
| | - Hong Chen
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Wen Wang
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Yao Deng
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Bo Wen
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
- College of Life Science, Jilin University, Changchun,Jinlin Province, People’s Republic of China
| | - Li Ruan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
| | - Wenjie Tan
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, People’s Republic of China
- * E-mail:
| |
Collapse
|
43
|
Alum-adjuvanted H5N1 whole virion inactivated vaccine (WIV) induced IgG1 and IgG4 antibody responses in young children. Vaccine 2012; 30:7662-6. [DOI: 10.1016/j.vaccine.2012.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
44
|
Rask C, Lund L, Lund G, Heydenreich B, Wurtzen P, Bellinghausen I, Saloga J, Lund K. An alternative allergen:adjuvant formulation potentiates the immunogenicity and reduces allergenicity of a novel subcutaneous immunotherapy product for treatment of grass-pollen allergy. Clin Exp Allergy 2012; 42:1356-68. [DOI: 10.1111/j.1365-2222.2012.04026.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- C. Rask
- Department of Pharmacology; ALK-Abelló A/S; Hoersholm; Denmark
| | - L. Lund
- Department of CMC Research; ALK-Abelló A/S; Hoersholm; Denmark
| | - G. Lund
- Department of Pharmacology; ALK-Abelló A/S; Hoersholm; Denmark
| | - B. Heydenreich
- Universitätsmedizin der Johannes Gutenberg-Universität; Hautklinik und Poliklinik ; Mainz; Germany
| | - P. Wurtzen
- Department of Pharmacology; ALK-Abelló A/S; Hoersholm; Denmark
| | - I. Bellinghausen
- Universitätsmedizin der Johannes Gutenberg-Universität; Hautklinik und Poliklinik ; Mainz; Germany
| | - J. Saloga
- Universitätsmedizin der Johannes Gutenberg-Universität; Hautklinik und Poliklinik ; Mainz; Germany
| | - K. Lund
- Department of Global Innovation Management; ALK-Abelló A/S; Hoersholm; Denmark
| |
Collapse
|
45
|
Pentamers not found in the universal proteome can enhance antigen specific immune responses and adjuvant vaccines. PLoS One 2012; 7:e43802. [PMID: 22937099 PMCID: PMC3427150 DOI: 10.1371/journal.pone.0043802] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/26/2012] [Indexed: 12/22/2022] Open
Abstract
Certain short peptides do not occur in humans and are rare or non-existent in the universal proteome. Antigens that contain rare amino acid sequences are in general highly immunogenic and may activate different arms of the immune system. We first generated a list of rare, semi-common, and common 5-mer peptides using bioinformatics tools to analyze the UniProtKB database. Experimental observations indicated that rare and semi-common 5-mers generated stronger cellular responses in comparison with common-occurring sequences. We hypothesized that the biological process responsible for this enhanced immunogenicity could be used to positively modulate immune responses with potential application for vaccine development. Initially, twelve rare 5-mers, 9-mers, and 13-mers were incorporated in frame at the end of an H5N1 hemagglutinin (HA) antigen and expressed from a DNA vaccine. The presence of some 5-mer peptides induced improved immune responses. Adding one 5-mer peptide exogenously also offered improved clinical outcome and/or survival against a lethal H5N1 or H1N1 influenza virus challenge in BALB/c mice and ferrets, respectively. Interestingly, enhanced anti-HBsAg antibody production by up to 25-fold in combination with a commercial Hepatitis B vaccine (Engerix-B, GSK) was also observed in BALB/c mice. Mechanistically, NK cell activation and dependency was observed with enhancing peptides ex vivo and in NK-depleted mice. Overall, the data suggest that rare or non-existent oligopeptides can be developed as immunomodulators and supports the further evaluation of some 5-mer peptides as potential vaccine adjuvants.
Collapse
|
46
|
Koizumi SI, Masuko K, Wakita D, Tanaka S, Mitamura R, Kato Y, Tabata H, Nakahara M, Kitamura H, Nishimura T. Extracts of Larix Leptolepis effectively augments the generation of tumor antigen-specific cytotoxic T lymphocytes via activation of dendritic cells in TLR-2 and TLR-4-dependent manner. Cell Immunol 2012; 276:153-61. [PMID: 22677561 DOI: 10.1016/j.cellimm.2012.05.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 05/01/2012] [Accepted: 05/10/2012] [Indexed: 10/28/2022]
Abstract
Type-1 immunity plays a crucial role in host defense against various tumors and infectious diseases. Here, we first demonstrated that extract of Larix Leptolepis (ELL), one of the most popular timbers at Hokkaido area in Japan, strongly activated Type-1 immunity. ELL induced production of Type-1 cytokines such as IL-12 and TNF-α from bone marrow-derived dendritic cells (BMDCs) in TLR2- and TLR4-dependent manner and remarkably up-regulated the expression of MHC and co-stimulatory molecules. In addition, antigen-specific CTLs were significantly augmented by the combined administration of ELL, antigen and BMDCs. Finally, we revealed that combination therapy using ELL, antigen and BMDCs significantly inhibited the growth of established tumor in mouse model. Thus, these findings suggested that ELL would be a novel adjuvant for inducing an activation of Type-1-dependent immunity including activation of BMDCs and induction of tumor-specific CTLs, which is applicable to the therapy of cancer and infectious diseases.
Collapse
Affiliation(s)
- Shin-ichi Koizumi
- Division of Immunoregulation, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Protective immunization of hamsters against Opisthorchis viverrini infection is associated with the reduction of TGF-β expression. Acta Trop 2012; 122:189-95. [PMID: 22266215 DOI: 10.1016/j.actatropica.2012.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 01/05/2012] [Accepted: 01/06/2012] [Indexed: 11/21/2022]
Abstract
Opisthorchis viverrini infection is a significant health problem in Thailand and other countries in Southeast Asia. There is little known about the mechanisms of the immune response to O. viverrini in immunoprotection. However, it has been reported that this parasite can suppress both cell and antibody mediated immune responses. The TGF-β and IL-10 are immunosuppressive cytokines that play an important role in inhibition of host immune response leading to worm survival. In this study, we immunized hamsters to protect against O. viverrini infection and the IL-4, IL-10, TGF-β and IFN-γ expression in spleen was investigated by real time PCR analysis. An O. viverrini-crude somatic antigen preparation (CSAg) administered with complete Freund's adjuvant (CFA) or with alum was used to stimulate immune responses in O. viverrini-primed hamsters. The greatest percent protection (48.4%) was seen following immunization with CSAg plus alum. The mean number±SD of worms recovered in the PBS control, CFA alone, CSAg plus CFA, alum alone and CSAg plus alum was 17.4±2.3, 17.1±3.3, 14.5±3.8, 14.5±2.3 and 9±2.7, respectively. Significant protection correlated with the reduction of TGF-β and IL-10, but not IL-4, IFN-γ expressions. Since TGF-β expression is significantly increased in the spleens of hamsters with opisthorchiasis, stimulation of this cytokine by parasite antigens was confirmed by using CSAg and primary hamster spleen cells. Antigen fractions with molecular masses of 81-92, 64-72 and 19-21.4kDa were found to significantly induce TGF-β production. Our results suggested that TGF-β induction by O. viverrini may have an important role in parasite survival.
Collapse
|
48
|
Nakayama T, Kashiwagi Y, Kawashima H, Kumagai T, Ishii KJ, Ihara T. Alum-adjuvanted H5N1 whole virion inactivated vaccine (WIV) enhanced inflammatory cytokine productions. Vaccine 2012; 30:3885-90. [PMID: 22507655 DOI: 10.1016/j.vaccine.2012.04.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Revised: 03/29/2012] [Accepted: 04/01/2012] [Indexed: 10/28/2022]
Abstract
Alum-adjuvanted H5 whole virion inactivated vaccine (WIV) was licensed for adults in Japan but induced marked febrile reactions with significantly stronger antibody responses in children. In this study, the mechanisms behind the different responses were investigated. Lymphocytes were obtained from 25 healthy subjects who were not immunized with H5 vaccine, to examine the innate immune impact of the various vaccine formulations, analyzing the cytokine production profile stimulated with alum adjuvant alone, alum-adjuvanted H5 WIIV, plain H5 WIV, and H5 split vaccine. Alum adjuvant did not induce cytokine production, but H5 split induced IFN-γ and TNF-α. H5 WIV induced IL-6, IL-17, TNF-α, MCP-1, IFN-γ, and IFN-α. An extremely low level of IL-1β was produced in response to H5 WIV, and alum-adjuvanted H5 WIV enhanced IL-1β production, with similar levels of other cytokines stimulated with H5 WIV. Enhanced production of cytokines induced by alum-adjuvanted H5 WIV may be related to the higher incidence of febrile reactions with stronger immune responses in children but it should be further investigated why efficient immune responses with febrile illness were observed only in young children.
Collapse
Affiliation(s)
- Tetsuo Nakayama
- Kitasato Institute for Life Sciences, Laboratory of Viral Infection, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- Tetsuo NAKAYAMA
- Kitasato Institute for Life Sciences, Laboratory of Viral Infection I
| |
Collapse
|
50
|
Pinto VV, Salanti A, Joergensen LM, Dahlbäck M, Resende M, Ditlev SB, Agger EM, Arnot DE, Theander TG, Nielsen MA. The effect of adjuvants on the immune response induced by a DBL4ɛ-ID4 VAR2CSA based Plasmodium falciparum vaccine against placental malaria. Vaccine 2011; 30:572-9. [PMID: 22122859 DOI: 10.1016/j.vaccine.2011.11.068] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 10/15/2011] [Accepted: 11/16/2011] [Indexed: 10/15/2022]
Abstract
A vaccine protecting women against placental malaria could be based on the sub-domains of the VAR2CSA antigen, since antibodies against the DBL4ɛ-ID4 subunit of the VAR2CSA protein can inhibit parasite binding to the placental ligand chondroitin sulphate A (CSA). Here we tested the ability of DBL4ɛ-ID4 to induce binding-inhibitory antibodies when formulated with adjuvants approved for human use. We have characterized the immune response of DBL4ɛ-ID4 in combination with Freund's complete and incomplete adjuvant and with three adjuvants currently being used in clinical trials: Montanide(®) ISA 720, Alhydrogel(®) and CAF01. Antibodies induced against DBL4ɛ-ID4 in combination with these adjuvants inhibited parasite binding to CSA from 82% to 99%. Although, different epitope recognition patterns were obtained for the different formulations, all adjuvant combinations induced strong Th1 and Th2 type responses, resulting in IgG with similar binding strength, with to the DBL4ɛ-ID4 antigen. These results demonstrate that the DBL4ɛ-ID4 antigen is highly immunogenic and that binding inhibitory antibodies are induced when formulated with any of the tested adjuvants.
Collapse
Affiliation(s)
- V V Pinto
- Centre for Medical Parasitology, Department of International Health, University of Copenhagen, CSS, Øster Farimagsgade 5 A, DK-1014 Copenhagen K, Denmark.
| | | | | | | | | | | | | | | | | | | |
Collapse
|