1
|
Current view on novel vaccine technologies to combat human infectious diseases. Appl Microbiol Biotechnol 2022; 106:25-56. [PMID: 34889981 PMCID: PMC8661323 DOI: 10.1007/s00253-021-11713-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/19/2021] [Accepted: 11/24/2021] [Indexed: 02/06/2023]
Abstract
Inactivated and live attenuated vaccines have improved human life and significantly reduced morbidity and mortality of several human infectious diseases. However, these vaccines have faults, such as reactivity or suboptimal efficacy and expensive and time-consuming development and production. Additionally, despite the enormous efforts to develop vaccines against some infectious diseases, the traditional technologies have not been successful in achieving this. At the same time, the concerns about emerging and re-emerging diseases urge the need to develop technologies that can be rapidly applied to combat the new challenges. Within the last two decades, the research of vaccine technologies has taken several directions to achieve safe, efficient, and economic platforms or technologies for novel vaccines. This review will give a brief overview of the current state of the novel vaccine technologies, new vaccine candidates in clinical trial phases 1-3 (listed by European Medicines Agency (EMA) and Food and Drug Administration (FDA)), and vaccines based on the novel technologies which have already been commercially available (approved by EMA and FDA) with the special reference to pandemic COVID-19 vaccines. KEY POINTS: • Vaccines of the new generation follow the minimalist strategy. • Some infectious diseases remain a challenge for the vaccine development. • The number of new vaccine candidates in the late phase clinical trials remains low.
Collapse
|
2
|
Sufyan M, Shahid F, Irshad F, Javaid A, Qasim M, Ashfaq UA. Implementation of Vaccinomics and In-Silico Approaches to Construct Multimeric Based Vaccine Against Ovarian Cancer. Int J Pept Res Ther 2021; 27:2845-2859. [PMID: 34690620 PMCID: PMC8524215 DOI: 10.1007/s10989-021-10294-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2021] [Indexed: 11/03/2022]
Abstract
One of the most common gynecologic cancers is ovarian cancer and ranked third after the other two most common cancers: cervical and uterine. The highest mortality rate has been observed in the case of ovarian cancer. To treat ovarian cancer, an immune-informatics approach was used to design a multi-epitope vaccine (MEV) structure. Epitopes prediction of the cancer testis antigens (NY-ESO-1), A-Kinase anchor protein (AKAP4), Acrosin binding protein (ACRBP), Piwi-like protein (PIWIL3), and cancer testis antigen 2 (LAGE-1) was done. Non-toxic, highly antigenic, non-allergenic, and overlapping epitopes were shortlisted for vaccine construction. Chosen T-cell epitopes displayed a robust binding attraction with their corresponding Human Leukocyte Antigen (HLA) alleles demonstrated 97.59% of population coverage. The vaccine peptide was established by uniting three key constituents, comprising the 14 epitopes of CD8 + cytotoxic T lymphocytes (CTLs), 5 helper epitopes, and the adjuvant. For the generation of the effective response of CD4 + cells towards the T-helper cells, granulocyte–macrophage-colony-stimulating factor (GM-CSF) was applied. With the addition of adjuvants and linkers, the construct size was 547 amino acids. The developed MEV structure was predicted to be antigenic, non-toxic, non-allergenic, and firm in nature. I-tasser anticipated the 3D construction of MEV. Moreover, disulfide engineering further enhanced the stability of the final vaccine protein. In-silico cloning and vaccine codon optimization were done to analyze the up-regulation of its expression. The outcomes established the vaccine’s immunogenicity and safety profile, besides its aptitude to encourage both humoral and cellular immune responses. The offered vaccine, grounded on our in-silico investigation, may be considered for ovarian cancer immunotherapy.
Collapse
Affiliation(s)
- Muhammad Sufyan
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Farah Shahid
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Faiza Irshad
- Environment Biotechnology Lab, Institute of Botany, University of the Punjab, Lahore, Pakistan
| | - Anam Javaid
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Muhammad Qasim
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| |
Collapse
|
3
|
Liu Q, Sun Z, Chen L. Memory T cells: strategies for optimizing tumor immunotherapy. Protein Cell 2020; 11:549-564. [PMID: 32221812 PMCID: PMC7381543 DOI: 10.1007/s13238-020-00707-9] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/08/2020] [Indexed: 12/15/2022] Open
Abstract
Several studies have demonstrated that memory T cells including stem cell memory (Tscm) T cells and central memory (Tcm) T cells show superior persistence and antitumor immunity compared with effector memory T (Tem) cells and effector T (Teff) cells. Furthermore, the Tcm/Teff ratio has been reported to be a predictive biomarker of immune responses against some tumors. Thus, a system-level understanding of the mechanisms underlying the differentiation of effector and memory T cells is of increasing importance for developing immunological strategies against various tumors. This review focuses on recent advances in efficacy against tumors, the origin, formation mechanisms of memory T cells, and the role of the gut microbiota in memory T cell formation. Furthermore, we summarize strategies to generate memory T cells in (ex) vivo that, might be applicable in clinical practice.
Collapse
Affiliation(s)
- Qingjun Liu
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China.,Newish Technology (Beijing) Co., Ltd., Xihuan South Road 18, Economic & Technical Development Zone, Beijing, 100176, China.,Moon (Guangzhou) Biotech Co., Ltd., Room 301, Building B5, Enterprise Accelerator, No. 11 Kaiyuan Avenue, Huangpu District, Guangzhou, 510000, China
| | - Zhongjie Sun
- Newish Technology (Beijing) Co., Ltd., Xihuan South Road 18, Economic & Technical Development Zone, Beijing, 100176, China.
| | - Ligong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China. .,Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100088, China.
| |
Collapse
|
4
|
Graham BS. Immunological goals for respiratory syncytial virus vaccine development. Curr Opin Immunol 2019; 59:57-64. [PMID: 31029910 DOI: 10.1016/j.coi.2019.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 03/20/2019] [Accepted: 03/20/2019] [Indexed: 12/26/2022]
Abstract
Defining the immunological goals for respiratory syncytial virus (RSV) vaccination requires understanding of RSV biology and tropism, mechanisms of cell-to-cell spread and immunity, epidemiology, and transmission dynamics. The immunological goals for a particular vaccine would be product-specific based on antigen selection, delivery approach, and target population. There are many ways to achieve immunity against RSV infection involving innate and adaptive responses, humoral, and cellular effector mechanisms, and mucosal and systemic responses. Both protective and pathological immune response patterns have been demonstrated in animal models and humans. In this short commentary, the entire information matrix that may inform the design of particular vaccine candidates cannot be fully reviewed, but the rationale behind the major vaccine approaches in key target populations will be discussed.
Collapse
Affiliation(s)
- Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
5
|
Parvizpour S, Razmara J, Pourseif MM, Omidi Y. In silico design of a triple-negative breast cancer vaccine by targeting cancer testis antigens. ACTA ACUST UNITED AC 2018; 9:45-56. [PMID: 30788259 PMCID: PMC6378095 DOI: 10.15171/bi.2019.06] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/10/2018] [Accepted: 04/14/2018] [Indexed: 12/14/2022]
Abstract
Introduction: Triple-negative breast cancer (TNBC) is an important subtype of breast cancer, which occurs in the absence of estrogen, progesterone and HER-2 receptors. According to the recent studies, TNBC may be a cancer testis antigen (CTA)-positive tumor, indicating that the CTA-based cancer vaccine can be a treatment option for the patients bearing such tumors. Of these antigens (Ags), the MAGE-A family and NY-ESO-1 as the most immunogenic CTAs are the potentially relevant targets for the development of an immunotherapeutic way of the breast cancer treatment. Methods: In the present study, immunoinformatics approach was used to design a multi-epitope peptide vaccine to combat the TNBC. The vaccine peptide was constructed by the fusion of three crucial components, including the CD8+ cytotoxic T lymphocytes (CTLs) epitopes, helper epitopes and adjuvant. The epitopes were predicted from the MAGE-A and NY-ESO-1 Ags. In addition, the granulocyte-macrophage-colony-stimulating factor (GM-CSF) was used as an adjuvant to promote the CD4+ T cells towards the T-helper for more strong induction of CTL responses. The components were conjugated by proper linkers. Results: The vaccine peptide was examined for different physiochemical characteristics to confirm the safety and immunogenic behavior. Furthermore, the 3D-structure of the vaccine peptide was predicted based on the homology modeling approach using the MODELLER v9.17 program. The vaccine structure was also subjected to the molecular dynamics simulation study for structure refinement. The results verified the immunogenicity and safety profile of the constructed vaccine as well as its capability for stimulating both the cellular and humoral immune responses. Conclusion: Based on our in-silico analyses, the proposed vaccine may be considered for the immunotherapy of TNBC.
Collapse
Affiliation(s)
- Sepideh Parvizpour
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Razmara
- Department of Computer Science, Faculty of Mathematical Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad M Pourseif
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Suschak JJ, Williams JA, Schmaljohn CS. Advancements in DNA vaccine vectors, non-mechanical delivery methods, and molecular adjuvants to increase immunogenicity. Hum Vaccin Immunother 2017. [PMID: 28604157 DOI: 10.1080/21645515.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
A major advantage of DNA vaccination is the ability to induce both humoral and cellular immune responses. DNA vaccines are currently used in veterinary medicine, but have not achieved widespread acceptance for use in humans due to their low immunogenicity in early clinical studies. However, recent clinical data have re-established the value of DNA vaccines, particularly in priming high-level antigen-specific antibody responses. Several approaches have been investigated for improving DNA vaccine efficacy, including advancements in DNA vaccine vector design, the inclusion of genetically engineered cytokine adjuvants, and novel non-mechanical delivery methods. These strategies have shown promise, resulting in augmented adaptive immune responses in not only mice, but also in large animal models. Here, we review advancements in each of these areas that show promise for increasing the immunogenicity of DNA vaccines.
Collapse
Affiliation(s)
- John J Suschak
- a U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | | | - Connie S Schmaljohn
- a U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
7
|
Suschak JJ, Williams JA, Schmaljohn CS. Advancements in DNA vaccine vectors, non-mechanical delivery methods, and molecular adjuvants to increase immunogenicity. Hum Vaccin Immunother 2017; 13:2837-2848. [PMID: 28604157 PMCID: PMC5718814 DOI: 10.1080/21645515.2017.1330236] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
A major advantage of DNA vaccination is the ability to induce both humoral and cellular immune responses. DNA vaccines are currently used in veterinary medicine, but have not achieved widespread acceptance for use in humans due to their low immunogenicity in early clinical studies. However, recent clinical data have re-established the value of DNA vaccines, particularly in priming high-level antigen-specific antibody responses. Several approaches have been investigated for improving DNA vaccine efficacy, including advancements in DNA vaccine vector design, the inclusion of genetically engineered cytokine adjuvants, and novel non-mechanical delivery methods. These strategies have shown promise, resulting in augmented adaptive immune responses in not only mice, but also in large animal models. Here, we review advancements in each of these areas that show promise for increasing the immunogenicity of DNA vaccines.
Collapse
Affiliation(s)
- John J Suschak
- a U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| | | | - Connie S Schmaljohn
- a U.S. Army Medical Research Institute of Infectious Diseases , Fort Detrick , MD , USA
| |
Collapse
|
8
|
Effect of HSV-IL12 Loaded Tumor Cell-Based Vaccination in a Mouse Model of High-Grade Neuroblastoma. J Immunol Res 2016; 2016:2568125. [PMID: 27610392 PMCID: PMC5005549 DOI: 10.1155/2016/2568125] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/01/2016] [Accepted: 07/03/2016] [Indexed: 12/18/2022] Open
Abstract
We designed multimodal tumor vaccine that consists of irradiated tumor cells infected with the oncolytic IL-12-expressing HSV-1 virus, M002. This vaccine was tested against the syngeneic neuroblastoma mouse model Neuro 2a injected into the right caudate nucleus of the immunocompetent A/J mice. Mice were vaccinated via intramuscular injection of multimodal vaccine or uninfected irradiated tumor cells at seven and 14 days after tumor establishment. While there was no survival difference between groups vaccinated with cell-based vaccine applied following tumor injection, a premunition prime/boost vaccination strategy produced a significant survival advantage in both groups and sustained immune response to an intracranial rechallenge of the same tumor. The syngeneic but unrelated H6 hepatocellular tumor cell line grew unrestricted in vaccinated mice, indicative of vaccine-mediated specific immunity to Neuro 2a tumors. Longitudinal analyses of tumor-infiltrating lymphocytes revealed a primary adaptive T cell response involving both CD4+ and CD8+ T cell subsets. Spleen cell mononuclear preparations from vaccinated mice were significantly more cytotoxic to Neuro 2a tumor cells than spleen cells from control mice as demonstrated in a four-hour in vitro cytotoxicity assay. These results strongly suggest that an irradiated whole cell tumor vaccine incorporating IL-12-expressing M002 HSV can produce a durable, specific immunization in a murine model of intracranial tumor.
Collapse
|
9
|
Santana VC, Almeida RR, Ribeiro SP, Ferreira LCDS, Kalil J, Rosa DS, Cunha-Neto E. Co-administration of plasmid-encoded granulocyte-macrophage colony-stimulating factor increases human immunodeficiency virus-1 DNA vaccine-induced polyfunctional CD4+ T-cell responses. Mem Inst Oswaldo Cruz 2015; 110:1010-6. [PMID: 26602876 PMCID: PMC4708021 DOI: 10.1590/0074-02760150283] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 10/20/2015] [Indexed: 02/04/2023] Open
Abstract
T-cell based vaccines against human immunodeficiency virus (HIV) generate specific
responses that may limit both transmission and disease progression by controlling
viral load. Broad, polyfunctional, and cytotoxic CD4+T-cell responses have
been associated with control of simian immunodeficiency virus/HIV-1 replication,
supporting the inclusion of CD4+ T-cell epitopes in vaccine formulations.
Plasmid-encoded granulocyte-macrophage colony-stimulating factor (pGM-CSF)
co-administration has been shown to induce potent CD4+ T-cell responses
and to promote accelerated priming and increased migration of antigen-specific
CD4+ T-cells. However, no study has shown whether co-immunisation with
pGM-CSF enhances the number of vaccine-induced polyfunctional CD4+
T-cells. Our group has previously developed a DNA vaccine encoding conserved,
multiple human leukocyte antigen (HLA)-DR binding HIV-1 subtype B peptides, which
elicited broad, polyfunctional and long-lived CD4+ T-cell responses. Here,
we show that pGM-CSF co-immunisation improved both magnitude and quality of
vaccine-induced T-cell responses, particularly by increasing proliferating
CD4+ T-cells that produce simultaneously interferon-γ, tumour necrosis
factor-α and interleukin-2. Thus, we believe that the use of pGM-CSF may be helpful
for vaccine strategies focused on the activation of anti-HIV CD4+ T-cell
immunity.
Collapse
Affiliation(s)
- Vinicius Canato Santana
- Divisão de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Rafael Ribeiro Almeida
- Divisão de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | - Susan Pereira Ribeiro
- Divisão de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| | | | - Jorge Kalil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia, São Paulo, SP, Brasil
| | - Daniela Santoro Rosa
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia, São Paulo, SP, Brasil
| | - Edecio Cunha-Neto
- Divisão de Imunologia Clínica e Alergia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
10
|
Alum Adjuvant Enhances Protection against Respiratory Syncytial Virus but Exacerbates Pulmonary Inflammation by Modulating Multiple Innate and Adaptive Immune Cells. PLoS One 2015; 10:e0139916. [PMID: 26468884 PMCID: PMC4607166 DOI: 10.1371/journal.pone.0139916] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 09/18/2015] [Indexed: 12/15/2022] Open
Abstract
Respiratory syncytial virus (RSV) is well-known for inducing vaccine-enhanced respiratory disease after vaccination of young children with formalin-inactivated RSV (FI-RSV) in alum formulation. Here, we investigated alum adjuvant effects on protection and disease after FI-RSV immunization with or without alum in comparison with live RSV reinfections. Despite viral clearance, live RSV reinfections caused weight loss and substantial pulmonary inflammation probably due to high levels of RSV specific IFN-γ+IL4-, IFN-γ-TNF-α+, IFN-γ+TNF-α- effector CD4 and CD8 T cells. Alum adjuvant significantly improved protection as evidenced by effective viral clearance compared to unadjuvanted FI-RSV. However, in contrast to unadjuvanted FI-RSV, alum-adjuvanted FI-RSV (FI-RSV-A) induced severe vaccine-enhanced RSV disease including weight loss, eosinophilia, and lung histopathology. Alum adjuvant in the FI-RSV-A was found to be mainly responsible for inducing high levels of RSV-specific IFN-γ-IL4+, IFN-γ-TNF-α+ CD4+ T cells, and proinflammatory cytokines IL-6 and IL-4 as well as B220+ plasmacytoid and CD4+ dendritic cells, and inhibiting the induction of IFN-γ+CD8 T cells. This study suggests that alum adjuvant in FI-RSV vaccines increases immunogenicity and viral clearance but also induces atypical T helper CD4+ T cells and multiple inflammatory dendritic cell subsets responsible for vaccine-enhanced severe RSV disease.
Collapse
|
11
|
Robertson FC, Berzofsky JA, Terabe M. NKT cell networks in the regulation of tumor immunity. Front Immunol 2014; 5:543. [PMID: 25389427 PMCID: PMC4211539 DOI: 10.3389/fimmu.2014.00543] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 10/13/2014] [Indexed: 12/31/2022] Open
Abstract
CD1d-restricted natural killer T (NKT) cells lie at the interface between the innate and adaptive immune systems and are important mediators of immune responses and tumor immunosurveillance. These NKT cells uniquely recognize lipid antigens, and their rapid yet specific reactions influence both innate and adaptive immunity. In tumor immunity, two NKT subsets (type I and type II) have contrasting roles in which they not only cross-regulate one another, but also impact innate immune cell populations, including natural killer, dendritic, and myeloid lineage cells, as well as adaptive populations, especially CD8+ and CD4+ T cells. The extent to which NKT cells promote or suppress surrounding cells affects the host’s ability to prevent neoplasia and is consequently of great interest for therapeutic development. Data have shown the potential for therapeutic use of NKT cell agonists and synergy with immune response modifiers in both pre-clinical studies and preliminary clinical studies. However, there is room to improve treatment efficacy by further elucidating the biological mechanisms underlying NKT cell networks. Here, we discuss the progress made in understanding NKT cell networks, their consequent role in the regulation of tumor immunity, and the potential to exploit that knowledge in a clinical setting.
Collapse
Affiliation(s)
- Faith C Robertson
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| |
Collapse
|
12
|
Rossey I, Sedeyn K, De Baets S, Schepens B, Saelens X. CD8+ T cell immunity against human respiratory syncytial virus. Vaccine 2014; 32:6130-7. [PMID: 25223272 DOI: 10.1016/j.vaccine.2014.08.063] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/08/2014] [Accepted: 08/27/2014] [Indexed: 12/11/2022]
Abstract
Human respiratory syncytial virus (HRSV) was first discovered in the 1950s, but despite decades of research, a licensed vaccine against it is not available. Epidemiological studies indicate that antibodies directed against the fusion protein (F) partially correlate with protection. In addition, an F-specific monoclonal antibody is licensed as a prophylactic treatment in children who are at high risk of developing complications following HRSV infection. Therefore, most HRSV-oriented vaccination strategies focus on inducing a humoral immune response against F. In the quest for the development of a safe HRSV vaccine, the induction of a T cell immune response has received a lot less attention. T cell immunity directed against HRSV has not been associated unequivocally with protection against HRSV and CD4(+) T helper cell responses may even worsen disease due to HRSV. However, many studies support a protective role for CD8(+) T cells in clearance of HRSV from the lungs. In this review we highlight the clinical and experimental evidence in favor of a CD8(+) T lymphocyte-based vaccination strategy to protect against HRSV. First, we describe how T cell responses and T cell memory are induced in the lungs upon respiratory viral infection. HRSV has evolved mechanisms that hamper CD8(+) T cell priming and effector functions. We appraise the information on HRSV-specific CD8(+) T cell immunity gained from laboratory mouse studies, taking into account the advantages and limitations of this animal model and, where possible, the accordance with clinical evidence. Finally, we focus on recent efforts to develop T cell based vaccines against HRSV.
Collapse
Affiliation(s)
- Iebe Rossey
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium
| | - Koen Sedeyn
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium
| | - Sarah De Baets
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium
| | - Bert Schepens
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium
| | - Xavier Saelens
- Inflammation Research Center, VIB, Technologiepark 927, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Technologiepark 927, Ghent 9052, Belgium.
| |
Collapse
|
13
|
Jackson RJ, Worley M, Trivedi S, Ranasinghe C. Novel HIV IL-4R antagonist vaccine strategy can induce both high avidity CD8 T and B cell immunity with greater protective efficacy. Vaccine 2014; 32:5703-14. [PMID: 25151041 DOI: 10.1016/j.vaccine.2014.08.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Revised: 05/07/2014] [Accepted: 08/11/2014] [Indexed: 12/18/2022]
Abstract
We have established that the efficacy of a heterologous poxvirus vectored HIV vaccine, fowlpox virus (FPV)-HIV gag/pol prime followed by attenuated vaccinia virus (VV)-HIV gag/pol booster immunisation, is strongly influenced by the cytokine milieu at the priming vaccination site, with endogenous IL-13 detrimental to the quality of the HIV specific CD8+ T cell response induced. We have now developed a novel HIV vaccine that co-expresses a C-terminal deletion mutant of the mouse IL-4, deleted for the essential tyrosine (Y119) required for signalling. In our vaccine system, the mutant IL-4C118 can bind to IL-4 type I and II receptors with high affinity, and transiently prevent the signalling of both IL-4 and IL-13 at the vaccination site. When this IL-4C118 adjuvanted vaccine was used in an intranasal rFPV/intramuscular rVV prime-boost immunisation strategy, greatly enhanced mucosal/systemic HIV specific CD8+ T cells with higher functional avidity, expressing IFN-γ, TNF-α and IL-2 and greater protective efficacy were detected. Surprisingly, the IL-4C118 adjuvanted vaccines also induced robust long-lived HIV gag-specific serum antibody responses, specifically IgG1 and IgG2a. The p55-gag IgG2a responses induced were of a higher magnitude relative to the IL-13Rα2 adjuvant vaccine. More interestingly, our recently tested IL-13Rα2 adjuvanted vaccine which only inhibited IL-13 activity, even though induced excellent high avidity HIV-specific CD8+ T cells, had a detrimental impact on the induction of gag-specific IgG2a antibody immunity. Our observations suggest that (i) IL-4 cell-signalling in the absence of IL-13 retarded gag-specific antibody isotype class switching, or (ii) IL-13Rα2 signalling was involved in inducing good gag-specific B cell immunity. Thus, we believe our novel IL-4R antagonist adjuvant strategy offers great promise not only for HIV-1 vaccines, but also against a range of chronic infections where sustained high quality mucosal and systemic T and B cell immunity are required for protection.
Collapse
Affiliation(s)
- Ronald J Jackson
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia
| | - Matthew Worley
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia
| | - Shubhanshi Trivedi
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia
| | - Charani Ranasinghe
- Molecular Mucosal Vaccine Immunology Group, Department of Immunology, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 0200, Australia.
| |
Collapse
|
14
|
Ranasinghe C, Trivedi S, Wijesundara DK, Jackson RJ. IL-4 and IL-13 receptors: Roles in immunity and powerful vaccine adjuvants. Cytokine Growth Factor Rev 2014; 25:437-42. [DOI: 10.1016/j.cytogfr.2014.07.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/15/2014] [Indexed: 01/01/2023]
|
15
|
Abu-Eid R, Samara RN, Ozbun L, Abdalla MY, Berzofsky JA, Friedman KM, Mkrtichyan M, Khleif SN. Selective inhibition of regulatory T cells by targeting the PI3K-Akt pathway. Cancer Immunol Res 2014; 2:1080-9. [PMID: 25080445 DOI: 10.1158/2326-6066.cir-14-0095] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Despite the strides that immunotherapy has made in mediating tumor regression, the clinical effects are often transient, and therefore more durable responses are still needed. The temporary nature of the therapy-induced immune response can be attributed to tumor immune evasion mechanisms, mainly the effect of suppressive immune cells and, in particular, regulatory T cells (Treg). Although the depletion of Tregs has been shown to be effective in enhancing immune responses, selective depletion of these suppressive cells without affecting other immune cells has not been very successful, and new agents are sought. We found that PI3K-Akt pathway inhibitors selectively inhibit Tregs with minimal effect on conventional T cells (Tconv). Our results clearly show selective in vitro inhibition of activation (as represented by a decrease in downstream signaling) and proliferation of Tregs in comparison with Tconvs when treated with different Akt and PI3K inhibitors. This effect has been observed in both human and murine CD4 T cells. In vivo treatment with these inhibitors resulted in a significant and selective reduction in Tregs in both naïve and tumor-bearing mice. Furthermore, these PI3K-Akt inhibitors led to a significant therapeutic antitumor effect, which was shown to be Treg dependent. Here, we report the use of PI3K-Akt pathway inhibitors as potent agents for the selective depletion of suppressive Tregs. We show that these inhibitors are able to enhance the antitumor immune response and are therefore promising clinical reagents for Treg depletion.
Collapse
Affiliation(s)
- Rasha Abu-Eid
- Georgia Regents University Cancer Center, Augusta, Georgia
| | - Raed N Samara
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Laurent Ozbun
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maher Y Abdalla
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jay A Berzofsky
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | - Samir N Khleif
- Georgia Regents University Cancer Center, Augusta, Georgia.
| |
Collapse
|
16
|
Sui Y, Hogg A, Wang Y, Frey B, Yu H, Xia Z, Venzon D, McKinnon K, Smedley J, Gathuka M, Klinman D, Keele BF, Langermann S, Liu L, Franchini G, Berzofsky JA. Vaccine-induced myeloid cell population dampens protective immunity to SIV. J Clin Invest 2014; 124:2538-49. [PMID: 24837435 PMCID: PMC4038576 DOI: 10.1172/jci73518] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Vaccines are largely evaluated for their ability to promote adaptive immunity, with little focus on the induction of negative immune regulators. Adjuvants facilitate and enhance vaccine-induced immune responses and have been explored for mediating protection against HIV. Using a regimen of peptide priming followed by a modified vaccinia Ankara (MVA) boost in a nonhuman primate model, we found that an SIV vaccine incorporating molecular adjuvants mediated partial protection against rectal SIVmac251 challenges. Animals treated with vaccine and multiple adjuvants exhibited a reduced viral load (VL) compared with those treated with vaccine only. Surprisingly, animals treated with adjuvant alone had reduced VLs that were comparable to or better than those of the vaccine-treated group. VL reduction was greatest in animals with the MHC class I allele Mamu-A*01 that were treated with adjuvant only and was largely dependent on CD8+ T cells. Early VLs correlated with Ki67+CCR5+CD4+ T cell frequency, while set-point VL was associated with expansion of a myeloid cell population that was phenotypically similar to myeloid-derived suppressor cells (MDSCs) and that suppressed T cell responses in vitro. MDSC expansion occurred in animals receiving vaccine and was not observed in the adjuvant-only group. Collectively, these results indicate that vaccine-induced MDSCs inhibit protective cellular immunity and suggest that preventing MDSC induction may be critical for effective AIDS vaccination.
Collapse
Affiliation(s)
- Yongjun Sui
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Alison Hogg
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Yichuan Wang
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Blake Frey
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Huifeng Yu
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Zheng Xia
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - David Venzon
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Katherine McKinnon
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Jeremy Smedley
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Mercy Gathuka
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Dennis Klinman
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Brandon F. Keele
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Sol Langermann
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Linda Liu
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Genoveffa Franchini
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| | - Jay A. Berzofsky
- Vaccine Branch, Biostatistics and Data Management Section, Laboratory Animal Sciences Program, and Laboratory of Experimental Immunology, National Cancer Institute, NIH, Bethesda, Maryland, USA. AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA. Amplimmune Inc., Gaithersburg, Maryland, USA
| |
Collapse
|
17
|
Jorquera PA, Oakley KE, Tripp RA. Advances in and the potential of vaccines for respiratory syncytial virus. Expert Rev Respir Med 2014; 7:411-27. [PMID: 23964629 DOI: 10.1586/17476348.2013.814409] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of serious lower respiratory track illness causing bronchiolitis and some mortality in infants and the elderly. Despite decades of research there is no licensed RSV vaccine. To enable the development of RSV vaccines, several major obstacles must be overcome including immature and waning immunity to RSV infection, the capacity of RSV to evade immunity and the failure of RSV infection to induce robust enduring immunity. Since the failure of the formalin-inactivated RSV vaccine trial, more cautious and deliberate progress has been made toward RSV vaccine development using a variety of experimental approaches. The scientific rational and the state of development of these approaches are reviewed in this article.
Collapse
Affiliation(s)
- Patricia A Jorquera
- College of Veterinary Medicine, Department of Infectious Disease, Animal Health Research Center, 111 Carlton Street, University of Georgia, Athens, GA 30602, USA
| | | | | |
Collapse
|
18
|
Fraser CK, Diener KR, Brown MP, Hayball JD. Improving vaccines by incorporating immunological coadjuvants. Expert Rev Vaccines 2014; 6:559-78. [PMID: 17669010 DOI: 10.1586/14760584.6.4.559] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
While vaccination continues to be the most successful interventionist health policy to date, infectious disease remains a significant cause of death worldwide. A primary reason that vaccination is not able to generate effective immunity is a lack of appropriate adjuvants capable of initiating the desired immune response. Adjuvant combinations can potentially overcome this problem; however, the possible permutations to consider, which include the route and kinetics of vaccination, as well as combinations of adjuvants, are practically limitless. This review aims to summarize the current understanding of adjuvants and related immunological processes and how this knowledge can and has been applied to the strategic selection of adjuvant combinations as components of vaccines against human infectious disease.
Collapse
Affiliation(s)
- Cara K Fraser
- Experimental Therapeutics Laboratory, Hanson Institute, and School of Pharmacy and Medical Sciences, Sansom Institute, University of South Australia, Australia.
| | | | | | | |
Collapse
|
19
|
Ranasinghe C, Ramshaw IA. Genetic heterologous prime–boost vaccination strategies for improved systemic and mucosal immunity. Expert Rev Vaccines 2014; 8:1171-81. [DOI: 10.1586/erv.09.86] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
20
|
Unique IL-13Rα2-based HIV-1 vaccine strategy to enhance mucosal immunity, CD8(+) T-cell avidity and protective immunity. Mucosal Immunol 2013; 6:1068-80. [PMID: 23403475 DOI: 10.1038/mi.2013.1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 12/18/2012] [Indexed: 02/04/2023]
Abstract
We have established that mucosal immunization can generate high-avidity human immunodeficiency virus (HIV)-specific CD8(+) T cells compared with systemic immunization, and interleukin (IL)-13 is detrimental to the functional avidity of these T cells. We have now constructed two unique recombinant HIV-1 vaccines that co-express soluble or membrane-bound forms of the IL-13 receptor α2 (IL-13Rα2), which can "transiently" block IL-13 activity at the vaccination site causing wild-type animals to behave similar to an IL-13 KO animal. Following intranasal/intramuscular prime-boost immunization, these IL-13Rα2-adjuvanted vaccines have shown to induce (i) enhanced HIV-specific CD8(+) T cells with higher functional avidity, with broader cytokine/chemokine profiles and greater protective immunity using a surrogate mucosal HIV-1 challenge, and also (ii) excellent multifunctional mucosal CD8(+) T-cell responses, in the lung, genito-rectal nodes (GN), and Peyer's patch (PP). Data revealed that intranasal delivery of these IL-13Rα2-adjuvanted HIV vaccines recruited large numbers of unique antigen-presenting cell subsets to the lung mucosae, ultimately promoting the induction of high-avidity CD8(+) T cells. We believe our novel IL-13R cytokine trap vaccine strategy offers great promise for not only HIV-1, but also as a platform technology against range of chronic infections that require strong sustained high-avidity mucosal/systemic immunity for protection.
Collapse
|
21
|
Berzofsky JA, Wood LV, Terabe M. Cancer vaccines: 21st century approaches to harnessing an ancient modality to fight cancer. Expert Rev Vaccines 2013; 12:1115-8. [PMID: 24124874 PMCID: PMC6326573 DOI: 10.1586/14760584.2013.836906] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 41-Room D702D, 41 Medlars Drive, Bethesda, MD 20892, USA +1 301 496 6874 +1 301 480 0681
| | | | | |
Collapse
|
22
|
Wijesundara DK, Jackson RJ, Tscharke DC, Ranasinghe C. IL-4 and IL-13 mediated down-regulation of CD8 expression levels can dampen anti-viral CD8+ T cell avidity following HIV-1 recombinant pox viral vaccination. Vaccine 2013; 31:4548-55. [DOI: 10.1016/j.vaccine.2013.07.062] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 07/02/2013] [Accepted: 07/25/2013] [Indexed: 01/23/2023]
|
23
|
Doyle MC, Tremblay S, Dumais N. 15-Deoxy-Δ(12,14)-prostaglandin J2 inhibits IL-13 production in T cells via an NF-κB-dependent mechanism. Biochem Biophys Res Commun 2013; 431:472-7. [PMID: 23333326 DOI: 10.1016/j.bbrc.2013.01.036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 01/09/2013] [Indexed: 11/18/2022]
Abstract
Interleukin (IL)-13 is a cytokine produced by activated CD4(+) T cells that plays a critical role in promoting allergic responses and tumor cell growth. The 15-deoxy-Δ(12,14)-prostaglandin J(2) (15d-PGJ(2)) is a natural ligand for the nuclear receptor peroxisome proliferator-activated receptor gamma (PPAR-γ), a known regulator of anti-inflammatory activities. We determined the effects of 15d-PGJ(2) on IL-13 expression in the Jurkat E6.1 T-cell line and in peripheral blood mononuclear cells. Semi-quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay revealed that treatment of activated T cells with 15d-PGJ(2) significantly decreased IL-13 mRNA transcription and secretion, respectively. This inhibition by 15d-PGJ(2) was independent of PPAR-γ since treatment with GW9662, an irreversible antagonist of the nuclear receptor, produced no effect. Our data also revealed the involvement of nuclear factor-κB in mediating 15d-PGJ(2)-dependent down regulation of IL-13 expression. Collectively, these results demonstrate the potential of 15d-PGJ(2) in attenuating expression and production of IL-13 in activated T cells.
Collapse
Affiliation(s)
- Marie-Christine Doyle
- Département de Biologie, Faculté des Sciences, Université de Sherbrooke, Sherbrooke (QC), Canada J1K 2R1
| | | | | |
Collapse
|
24
|
|
25
|
Mkrtichyan M, Najjar YG, Raulfs EC, Liu L, Langerman S, Guittard G, Ozbun L, Khleif SN. B7-DC-Ig enhances vaccine effect by a novel mechanism dependent on PD-1 expression level on T cell subsets. THE JOURNAL OF IMMUNOLOGY 2012; 189:2338-47. [PMID: 22837483 DOI: 10.4049/jimmunol.1103085] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Programmed death receptor 1 (PD-1) is an important signaling molecule often involved in tumor-mediated suppression of activated immune cells. Binding of this receptor to its ligands, B7-H1 (PD-L1) and B7-DC (PD-L2), attenuates T cell activation, reduces IL-2 and IFN-γ secretion, decreases proliferation and cytotoxicity, and induces apoptosis. B7-DC-Ig is a recombinant protein that binds and targets PD-1. It is composed of an extracellular domain of murine B7-DC fused to the Fc portion of murine IgG2a. In this study, we demonstrate that B7-DC-Ig can enhance the therapeutic efficacy of vaccine when combined with cyclophosphamide. We show that this combination significantly enhances Ag-specific immune responses and leads to complete eradication of established tumors in 60% of mice and that this effect is CD8 dependent. We identified a novel mechanism by which B7-DC-Ig exerts its therapeutic effect that is distinctly different from direct blocking of the PD-L1-PD-1 interaction. In this study, we demonstrate that there are significant differences between levels and timing of surface PD-1 expression on different T cell subsets. We found that these differences play critical roles in anti-tumor immune effect exhibited by B7-DC-Ig through inhibiting proliferation of PD-1(high) CD4 T cells, leading to a significant decrease in the level of these cells, which are enriched for regulatory T cells, within the tumor. In addition, it also leads to a decrease in PD-1(high) CD8 T cells, tipping the balance toward nonexhausted functional PD-1(low) CD8 T cells. We believe that the PD-1 expression level on T cells is a crucial factor that needs to be considered when designing PD-1-targeting immune therapies.
Collapse
Affiliation(s)
- Mikayel Mkrtichyan
- Cancer Vaccine Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Berzofsky JA. A push-pull vaccine strategy using Toll-like receptor ligands, IL-15, and blockade of negative regulation to improve the quality and quantity of T cell immune responses. Vaccine 2012; 30:4323-7. [PMID: 22115635 PMCID: PMC3319860 DOI: 10.1016/j.vaccine.2011.11.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 10/25/2011] [Accepted: 11/10/2011] [Indexed: 01/17/2023]
Abstract
We have developed a strategy to optimize the efficacy of vaccines to induce T-cell immunity against chronic viral infections and cancer based on a "push-pull" approach in which we first optimize the antigen structure by increasing the affinity of epitopes for major histocompatibility complex molecules ("epitope enhancement"), then push the response not only in magnitude but also in quality toward the desired response phenotype, using synergistic combinations of cytokines, Toll-like receptor ligands, and costimulatory molecules, and then pull the response by removing the brakes exerted by negative regulatory mechanisms, including regulatory cells, cell surface molecules, and cytokines. Components of this approach show promise in macaque models of AIDS virus infection and in murine models of cancer, and are being developed for human clinical trials.
Collapse
Affiliation(s)
- Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Building 10, Room 6B-04 (MSC#1578) NIH, Bethesda, MD 20892-1578, USA.
| |
Collapse
|
27
|
Abstract
Cancers so much resemble self that they prove difficult for the immune system to eliminate, and those that have already escaped natural immunosurveillance have gotten past the natural immune barriers to malignancy. A successful therapeutic cancer vaccine must overcome these escape mechanisms. Our laboratory has focused on a multistep "push-pull" approach in which we combine strategies to overcome each of the mechanisms of escape. If tumor epitopes are insufficiently immunogenic, we increase their immunogenicity by epitope enhancement, improving their binding affinity to major histocompatibility complex (MHC) molecules. If the anti-tumor response is too weak or of the wrong phenotype, we use cytokines, costimulatory molecules, Toll-like receptor ligands, and other molecular adjuvants to increase not only the quantity of the response but also its quality, to push the response in the right direction. Finally, the tumor invokes multiple immunosuppressive mechanisms to defend itself, so we need to overcome those as well, including blocking or depleting regulatory cells or inhibiting regulatory molecules, to pull the response by removing the brakes. Some of these strategies individually have now been translated into human clinical trials in cancer patients. Combinations of these in a push-pull approach are promising for the successful immunotherapy of cancer.
Collapse
Affiliation(s)
- Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
28
|
Isakov D, Dzutsev A, Berzofsky JA, Belyakov IM. Lack of IL-7 and IL-15 signaling affects interferon-γ production by, more than survival of, small intestinal intraepithelial memory CD8+ T cells. Eur J Immunol 2012; 41:3513-28. [PMID: 21928282 DOI: 10.1002/eji.201141453] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Survival of antigen-specific CD8(+) T cells in peripheral lymphoid organs during viral infection is known to be dependent predominantly on IL-7 and IL-15. However, little is known about a possible influence of tissue environmental factors on this process. To address this question, we studied survival of memory antigen-specific CD8(+) T cells in the small intestine. Here, we show that 2 months after vaccinia virus infection, B8R(20-27) /H2-K(b) tetramer(+) CD8(+) T cells in the small intestinal intraepithelial (SI-IEL) layer are found in mice deficient in IL-15 expression. Moreover, SI-IEL and lamina propria lymphocytes do not express the receptor for IL-7 (IL-7Rα/CD127). In addition, after in vitro stimulation with B8R(20-27) peptide, SI-IEL cells do not produce high amounts of IFN-γ neither at 5 days nor at 2 months postinfection (p.i.). Importantly, the lack of IL-15 was found to shape the functional activity of antigen-specific CD8(+) T cells, by narrowing the CTL avidity repertoire. Taken together, these results reveal that survival factors, as well as the functional activity, of antigen-specific CD8(+) T cells in the SI-IEL compartments may markedly differ from their counterparts in peripheral lymphoid tissues.
Collapse
Affiliation(s)
- Dmitry Isakov
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
29
|
Stathopoulos A, Pretto C, Devillers L, Pierre D, Hofman FM, Epstein AL, Farghadani H, Kruse CA, Jadus MR, Chen TC, Schijns VEJC. Exploring the Therapeutic Efficacy of Glioma Vaccines Based on Allo- and Syngeneic Antigens and Distinct Immunological Costimulation Activators. ACTA ACUST UNITED AC 2012; Suppl 5:004. [PMID: 24955288 PMCID: PMC4062195 DOI: 10.4172/2155-9899.s5-004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The efficacy of a various immunotherapeutic immunisation strategies for malignant glioma brain cancer was evaluated in the syngeneic CNS-1 Lewis rat glioma model. A prototype glioma cancer vaccine, which was composed of multivalent antigens derived from allogeneic and syngeneic cells and lysates, formed the prototype preparation of antigens. These antigens reflect the autologous antigens derived from the patient’s surgically removed tumor tissue, as well as allogeneic antigens form glioma tumor tissue surgically removed from donor patients. This antigen mixture provides a broad spectrum of tumor associated antigens (TAA) and helps to prevent escape of tumor immune surveillance when given as a vaccine. This antigen preparation was administered in a therapeutic setting with distinct single or multiple co-stimulation-favouring immunostimulants and evaluated for inhibition of tumor growth. Our prototype vaccine was able to arrest progression of tumor growth when co-delivered in a specific regimen together with the costimulating multi-TLR agonist, Bacille Calmette Guerin (BCG) and interleukin-2, or with the Toll-Like receptor (TLR) 7/8 activator resiquimod.
Collapse
Affiliation(s)
- Apostolos Stathopoulos
- Department of Neurosurgery, Arlon Hospital, Arlon, Belgium ; Epitopoietic Research Corporation (ERC), Namur, Belgium ; Department of Neurosurgery, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | | | | | - Denis Pierre
- Epitopoietic Research Corporation (ERC), Namur, Belgium
| | - Florence M Hofman
- Department of Pathology, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | - Alan L Epstein
- Department of Pathology, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | | | - Carol A Kruse
- Department of Neurosurgery, University of California, Los Angeles, California and the Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA
| | - Martin R Jadus
- Veterans Affairs Medical Center, Long Beach, CA 90822, box 113, 5901 E7th St. and Chao Cancer Center, University of California, Irvine, Orange CA, USA
| | - Thomas C Chen
- Epitopoietic Research Corporation (ERC), Namur, Belgium ; Department of Neurosurgery, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | - Virgil E J C Schijns
- Epitopoietic Research Corporation (ERC), Namur, Belgium ; Cell Biology & Immunology Group, Wageningen University, PO Box 338, 6700 AH Wageningen, The Netherlands
| |
Collapse
|
30
|
Mkrtichyan M, Najjar YG, Raulfs EC, Abdalla MY, Samara R, Rotem-Yehudar R, Cook L, Khleif SN. Anti-PD-1 synergizes with cyclophosphamide to induce potent anti-tumor vaccine effects through novel mechanisms. Eur J Immunol 2011; 41:2977-86. [PMID: 21710477 DOI: 10.1002/eji.201141639] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 05/16/2011] [Accepted: 06/20/2011] [Indexed: 11/12/2022]
Abstract
Programmed death-1 receptor (PD-1) is expressed on T cells following TCR activation. Binding of this receptor to its cognate ligands, programmed death ligand (PDL)-1 and PDL-2, down-regulates signals by the TCR, promoting T-cell anergy and apoptosis, thus leading to immune suppression. Here, we find that using an anti-PD-1 antibody (CT-011) with Treg-cell depletion by low-dose cyclophosphamide (CPM), combined with a tumor vaccine, induces synergistic antigen-specific immune responses and reveals novel activities of each agent in this combination. This strategy led to complete regression of established tumors in a significant percentage of treated animals, with survival prolongation. We show for the first time that combining CT-011 and CPM significantly increases the number of vaccine-induced tumor-infiltrating CD8(+) T cells, with simultaneous decrease in infiltrating Treg cells. Interestingly, we find that CT-011 prolongs Treg-cell inhibition induced by CPM, leading to a sustainable significant synergistic decrease of splenic and tumor-infiltrated Treg cells. Surprisingly, we find that the anti-tumor effect elicited by the combination of CT-011 and CPM is dependent on both CD8(+) and CD4(+) T-cell responses, although the antigen we used is a class I MHC-restricted peptide. Thus, we describe a novel and effective therapeutic approach by combining multiple strategies to target several tumor-mediated immune inhibitory mechanisms.
Collapse
Affiliation(s)
- Mikayel Mkrtichyan
- Cancer Vaccine Section, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Gene Therapy against Murine Melanoma B16F10-Nex2 Using IL-13Ralpha2-Fc Chimera and Interleukin 12 in Association with a Cyclopalladated Drug. Transl Oncol 2011; 1:110-20. [PMID: 18795121 DOI: 10.1593/tlo.08115] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Revised: 06/20/2008] [Accepted: 06/23/2008] [Indexed: 02/02/2023] Open
Abstract
Interleukin 13 (IL-13) is immunoregulatory in many diseases, including cancer. The protective or suppressive role of CD1-restricted natural killer T cells (NKT cells) in tumor immunosurveillance and immunity is well documented. Interleukin 12 (IL-12) can activate type I NKT cells to produce interferon-gamma (IFN-gamma), whereas type II NKT cells may produce IL-13. The high-affinity chain of IL-13Ralpha2 may act as negative inhibitor, suppressing the action of IL-13 and helping to maintain tumor immunosurveillance. We constructed an mIL-13Ralpha2-Fc chimera in a eukaryotic expression vector and confirmed the identity of the recombinant protein by immunoblot analysis and binding to IL-13 in chemiluminescent ELISA. Such DNA vaccine was tested against syngeneic B16F10-Nex2 murine melanoma. In vivo experiments showed a protective effect mediated by high production of IFN-gamma and down-regulation of anti-inflammatory interleukins mainly by NKT 1.1(+) T cells. Biochemoterapy in vivo with plasmid encoding mIL-13Ralpha2-Fc in association with plasmid encoding IL-12 and the 7A cyclopalladated drug led to a significant reduction in the tumor evolution with 30% tumor-free mice. We conclude that IL-12 gene therapy, followed by continuous administration of IL-13Ralpha2-Fc gene along with 7A-drug has antitumor activity involving the high production of proinflammatory cytokines and low immune suppression, specifically by NK1.1(+)T cells producing IL-13 and IL-10.
Collapse
|
32
|
Graham BS. Biological challenges and technological opportunities for respiratory syncytial virus vaccine development. Immunol Rev 2011; 239:149-66. [PMID: 21198670 PMCID: PMC3023887 DOI: 10.1111/j.1600-065x.2010.00972.x] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Respiratory syncytial virus (RSV) is an important cause of respiratory disease causing high rates of hospitalizations in infants, significant morbidity in children and adults, and excess mortality in the elderly. Major barriers to vaccine development include early age of RSV infection, capacity of RSV to evade innate immunity, failure of RSV-induced adaptive immunity to prevent reinfection, history of RSV vaccine-enhanced disease, and lack of an animal model fully permissive to human RSV infection. These biological challenges, safety concerns, and practical issues have significantly prolonged the RSV vaccine development process. One great advantage compared to other difficult viral vaccine targets is that passively administered neutralizing monoclonal antibody is known to protect infants from severe RSV disease. Therefore, the immunological goals for vaccine development are to induce effective neutralizing antibody to prevent infection and to avoid inducing T-cell response patterns associated with enhanced disease. Live-attenuated RSV and replication-competent chimeric viruses are in advanced clinical trials. Gene-based strategies, which can control the specificity and phenotypic properties of RSV-specific T-cell responses utilizing replication-defective vectors and which may improve on immunity from natural infection, are progressing through preclinical testing. Atomic level structural information on RSV envelope glycoproteins in complex with neutralizing antibodies is guiding design of new vaccine antigens that may be able to elicit RSV-specific antibody responses without induction of RSV-specific T-cell responses. These new technologies may allow development of vaccines that can protect against RSV-mediated disease in infants and establish a new immunological paradigm in the host to achieve more durable protection against reinfection.
Collapse
Affiliation(s)
- Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-3017, USA.
| |
Collapse
|
33
|
Mucosal immunity and HIV-1 infection: applications for mucosal AIDS vaccine development. Curr Top Microbiol Immunol 2011; 354:157-79. [PMID: 21203884 DOI: 10.1007/82_2010_119] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Natural transmission of human immunodeficiency virus type 1 (HIV-1) occurs through gastrointestinal and vaginal mucosa. These mucosal tissues are major reservoirs for initial HIV replication and amplification, and the sites of rapid CD4(+) T cell depletion. In both HIV-infected humans and SIV-infected macaques, massive loss of CD4(+) CCR5(+) memory T cells occurs in the gut and vaginal mucosa within the first 10-14 days of infection. Induction of local HIV-specific immune responses by vaccines may facilitate effective control of HIV or SIV replication at these sites. Vaccines that induce mucosal responses, in particular CD8(+) cytotoxic T lymphocytes (CTL), have controlled viral replication at mucosal sites and curtailed systemic dissemination. Thus, there is strong justification for development of next generation vaccines that induce mucosal immune effectors against HIV-1 including CD8(+) CTL, CD4(+) T helper cells and secretory IgA. In addition, further understanding of local innate mechanisms that impact early viral replication will greatly inform future vaccine development. In this review, we examine the current knowledge concerning mucosal AIDS vaccine development. Moreover, we propose immunization strategies that may be able to elicit an effective immune response that can protect against AIDS as well as other mucosal infections.
Collapse
|
34
|
Ahlers JD, Belyakov IM. Molecular pathways regulating CD4+ T cell differentiation, anergy and memory with implications for vaccines. Trends Mol Med 2010; 16:478-91. [DOI: 10.1016/j.molmed.2010.07.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Revised: 07/18/2010] [Accepted: 07/19/2010] [Indexed: 12/23/2022]
|
35
|
Ahlers JD, Belyakov IM. Lessons learned from natural infection: focusing on the design of protective T cell vaccines for HIV/AIDS. Trends Immunol 2010; 31:120-30. [PMID: 20089450 DOI: 10.1016/j.it.2009.12.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 12/08/2009] [Accepted: 12/11/2009] [Indexed: 01/26/2023]
Abstract
CD8(+) cytotoxic T lymphocyte (CTL) responses are crucial in establishing the control of persistent virus infections. Population studies of HIV-1-infected individuals suggest that CD8(+) CTL responses targeting epitopes that take the greatest toll on virus replication are instrumental in immune control. A major question for vaccine design is whether incorporating epitopes responsible for controlling a persistent virus will translate into protection from natural infection or serve solely as a fail-safe mechanism to prevent overt disease in infected individuals. Here, we discuss qualitative parameters of the CD8(+) CTL response and mechanisms operative in the control of persistent virus infections and suggest new strategies for design and delivery of HIV vaccines.
Collapse
|
36
|
Abstract
For acute self-limiting infections a vaccine is successful if it elicits memory at least as good as the natural experience; however, for persistent and chronic infections such as HIV, hepatitis C virus (HCV), human papillomavirus (HPV), and human herpes viruses, this paradigm is not applicable. At best, during persistent virus infection the person must be able to maintain the integrity of the immune system in equilibrium with controlling replicating virus. New vaccine strategies are required that elicit both potent high-avidity CD8(+) T-cell effector/memory and central memory responses that can clear the nidus of initial virus-infected cells at mucosal surfaces to prevent mucosal transmission or significantly curtail development of disease. The objective of an HIV-1 T-cell vaccine is to generate functional CD8(+) effector memory cells at mucosal portals of virus entry to prevent viral transmission. In addition, long-lived CD8(+) and CD4(+) central memory cells circulating through secondary lymphoid organs and resident in bone marrow, respectively, are needed to provide a concerted second wave of defense that can contain virus at mucosal surfaces and prevent systemic dissemination. Further understanding of factors which can influence long-lived effector and central memory cell differentiation will significantly contribute to development of effective T-cell vaccines. In this review we will focus on discussing mechanisms involved in T-cell memory and provide promising new approaches toward expanding current vaccine strategies to enhance antiviral memory.
Collapse
|
37
|
Ranasinghe C, Ramshaw IA. Immunisation route-dependent expression of IL-4/IL-13 can modulate HIV-specific CD8(+) CTL avidity. Eur J Immunol 2009; 39:1819-30. [PMID: 19582753 DOI: 10.1002/eji.200838995] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
All HIV-1 'systemic vaccine trials' in humans have yielded poor outcomes. Thus, it is important to understand whether the route of delivery influences the quality of protective CTL immunity. Using heterologous poxvirus immunisation we have shown that systemically (i.m./i.m.) immunised CD8(+) T cells generated higher levels of IL-4/IL-13 compared to mucosal delivery and expression also correlated with i.m./i.m. immunised mice eliciting CTL of lower avidity. Studies using IL-4(-/-) and IL-13(-/-) KO mice have shown that the capacity to express IFN-gamma, IL-4 and/or IL-13 by K(d)Gag(197-205)-specific CTL differed between these groups and was inversely correlated with CTL avidity (IL-13(-/-)>IL-4(-/-)>BALB/c), although no significant differences in the magnitude of CTL responses were observed between IL-13(-/-) and wild type mice. When IL-13 was reconstituted in IL-13(-/-) splenocytes in vitro, their ability to bind tetramers also decreased significantly. Our data reveal that total absence of IL-13 can greatly enhance CTL avidity. In contrast, extracellular IL-4 appears to be important in maintaining long-term Th1/Th2 balance in CTL, even though expression of IL-4 by CTL markedly reduced avidity. STAT6(-/-) mice also showed memory CTL of higher avidity. Furthermore, CCL5 expression in K(d)Gag(197-205)-specific CTL was also regulated by IL-4/IL-13.
Collapse
|
38
|
Silbermann K, Schneider G, Grassmann R. Stimulation of interleukin-13 expression by human T-cell leukemia virus type 1 oncoprotein Tax via a dually active promoter element responsive to NF-kappaB and NFAT. J Gen Virol 2009; 89:2788-2798. [PMID: 18931077 DOI: 10.1099/vir.0.2008/003699-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The human T-cell leukemia virus type 1 (HTLV-1) Tax oncoprotein transforms human lymphocytes and is critical for the pathogenesis of HTLV-1-induced adult T-cell leukaemia. In HTLV-transformed cells, Tax upregulates interleukin (IL)-13, a cytokine with proliferative and anti-apoptotic functions that is linked to leukaemogenesis. Tax-stimulated IL-13 is thought to result in autocrine stimulation of HTLV-infected cells and thus may be relevant to their growth. The causal transactivation of the IL-13 promoter by Tax is predominantly dependent on a nuclear factor of activated T cells (NFAT)-binding P element. Here, it was shown that the isolated IL-13 Tax-responsive element (IL13TaxRE) was sufficient to mediate IL-13 transactivation by Tax and NFAT1. However, cyclosporin A, a specific NFAT inhibitor, revealed that Tax transactivation of IL13TaxRE or wild-type IL-13 promoter was independent of NFAT and that NFAT did not contribute to IL-13 upregulation in HTLV-transformed cells. By contrast, Tax stimulation was repressible by an efficient nuclear factor (NF)-kappaB inhibitor (IkBaDN), indicating the requirement for NF-kappaB. The capacity of NF-kappaB to stimulate IL13TaxRE was demonstrated by a strong response to NF-kappaB in reporter assays and by direct binding of NF-kappaB to IL13TaxRE. Thus, IL13TaxRE in the IL-13 promoter represents a dually active promoter element responsive to NF-kappaB and NFAT. Together, these results indicate that Tax causes IL-13 upregulation in HTLV-1-infected cells via NF-kappaB.
Collapse
Affiliation(s)
- Katrin Silbermann
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Grit Schneider
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ralph Grassmann
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
39
|
Abstract
The clinical course of ovarian cancer is often marked by periods of relapse and remission until chemo-resistance develops. Patients in remission with minimal disease burdens are ideally suited for the evaluation of immune-based strategies. Major obstacles to the development of successful immune strategies include the identification of tumor-restricted immunogenic targets, generation of a sufficient immune response to cause tumor rejection, and approaches to overcome evasion of immune attack. Many questions remain as optimal strategies are developed, which include: (i) What is the best antigen form (e.g. peptides, proteins or tumor lysates)? (ii) What are the appropriate adjuvants? (iii) Are mono-valent or multi-valent vaccines likely to be more effective? (iv) What is the optimal frequency and duration of vaccination? (v) How should antigen-specific responses be monitored? and (vi) How should the anti-cancer response be maintained? In this review, we explore representative examples of immune strategies under investigation for patients with ovarian carcinoma which illustrate many of these issues. Basic principles generic to all these immunotherapeutic approaches will also be discussed.
Collapse
Affiliation(s)
- Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA.
| | | |
Collapse
|
40
|
Palucka AK, Ueno H, Fay JW, Banchereau J. Taming cancer by inducing immunity via dendritic cells. Immunol Rev 2008; 220:129-50. [PMID: 17979844 DOI: 10.1111/j.1600-065x.2007.00575.x] [Citation(s) in RCA: 153] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immunotherapy seeks to mobilize a patient's immune system for therapeutic benefit. It can be passive, i.e. transfer of immune effector cells (T cells) or proteins (antibodies), or active, i.e. vaccination. In cancer, passive immunotherapy can lead to some objective clinical responses, thus demonstrating that the immune system can reject tumors. However, passive immunotherapy is not expected to yield long-lived memory T cells that might control tumor outgrowth. Active immunotherapy with dendritic cell (DC)-based vaccines has the potential to induce both tumor-specific effector and memory T cells. Early clinical trials testing vaccination with ex vivo-generated DCs pulsed with tumor antigens provide a proof-of-principle that therapeutic immunity can be elicited. Yet, there is a need to improve their efficacy. The next generation of DC vaccines is expected to generate large numbers of high-avidity effector CD8(+) T cells and to overcome regulatory T cells. Therapeutic vaccination protocols will combine improved ex vivo DC vaccines with therapies that offset the suppressive environment established by tumors.
Collapse
Affiliation(s)
- A Karolina Palucka
- Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, TX, USA.
| | | | | | | |
Collapse
|
41
|
Pinzon-Charry A, Maxwell T, López JA. Dendritic cell dysfunction in cancer: a mechanism for immunosuppression. Immunol Cell Biol 2008; 83:451-61. [PMID: 16174093 DOI: 10.1111/j.1440-1711.2005.01371.x] [Citation(s) in RCA: 226] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Several reports have demonstrated that tumours are not intrinsically resistant to the immune response. However, neoplasias commonly fail to initiate and maintain adequate immunity. A number of factors have been implicated in causing the failure, including aberrant antigen processing by tumour cells, anergy or deletion of T cells, and recruitment of inhibitory/regulatory cell types. It has been suggested that dysfunction of dendritic cells (DC) induced by the tumour is one of the critical mechanisms to escape immune surveillance. As a minor subset of leucocytes, DC are the key APC for initiating immune responses. DC are poised at the boundaries of the periphery and the inner tissues, sampling antigens of diverse origin. Following their encounter with antigen or danger signals, DC migrate to lymph nodes, where they activate effector cells essential for tumour clearance. Although the DC system is highly heterogeneous, the differentiation and function of DC populations is largely regulated by exogenous factors. Malignancies appear to exploit this by producing a plethora of immunosuppressive factors capable of affecting DC, thus exerting systemic effects on immune function. This review examines recent findings on the effects of tumour-derived factors inducing DC dysfunction and in particular examines the findings on alteration of DC differentiation, maturation and longevity as a potent mechanism for immune suppression in cancer.
Collapse
Affiliation(s)
- Alberto Pinzon-Charry
- Dendritic Cell and Cancer Laboratory, Queensland Institute of Medical Research, Brisbane, Queensland, Australia
| | | | | |
Collapse
|
42
|
Abstract
NKT cells are a relatively newly recognized member of the immune community, with profound effects on the rest of the immune system despite their small numbers. They are true T cells with a T cell receptor (TCR), but unlike conventional T cells that detect peptide antigens presented by conventional major histocompatibility (MHC) molecules, NKT cells recognize lipid antigens presented by CD1d, a nonclassical MHC molecule. As members of both the innate and adaptive immune systems, they bridge the gap between these, and respond rapidly to set the tone for subsequent immune responses. They fill a unique niche in providing the immune system a cellular arm to recognize lipid antigens. They play both effector and regulatory roles in infectious and autoimmune diseases. Furthermore, subsets of NKT cells can play distinct and sometimes opposing roles. In cancer, type I NKT cells, defined by their invariant TCR using Valpha14Jalpha18 in mice and Valpha24Jalpha18 in humans, are mostly protective, by producing interferon-gamma to activate NK and CD8(+) T cells and by activating dendritic cells to make IL-12. In contrast, type II NKT cells, characterized by more diverse TCRs recognizing lipids presented by CD1d, primarily inhibit tumor immunity. Moreover, type I and type II NKT cells counter-regulate each other, forming a new immunoregulatory axis. Because NKT cells respond rapidly, the balance along this axis can greatly influence other immune responses that follow. Therefore, learning to manipulate the balance along the NKT regulatory axis may be critical to devising successful immunotherapies for cancer.
Collapse
Affiliation(s)
- Masaki Terabe
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
43
|
Belyakov IM, Kozlowski S, Mage M, Ahlers JD, Boyd LF, Margulies DH, Berzofsky JA. Role of alpha3 domain of class I MHC molecules in the activation of high- and low-avidity CD8+ CTLs. Int Immunol 2007; 19:1413-20. [PMID: 17981793 DOI: 10.1093/intimm/dxm111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
CD8 can serve as a co-receptor or accessory molecule on the surface of CTL. As a co-receptor, CD8 can bind to the alpha3 domain of the same MHC class I molecules as the TCR to facilitate TCR signaling. To evaluate the role of the MHC class I molecule alpha3 domain in the activation of CD8(+) CTL, we have produced a soluble 227 mutant of H-2D(d), with a point mutation in the alpha3 domain (Glu227 --> Lys). 227 mutant class I-peptide complexes were not able to effectively activate H-2D(d)-restricted CD8 T cells in vitro, as measured by IFN-gamma production by an epitope-specific CD8(+) CTL line. However, the 227 mutant class I-peptide complexes in the presence of another MHC class I molecule (H-2K(b)) (that cannot present the peptide) with a normal alpha3 domain can induce the activation of CD8(+) CTL. Therefore, in order to activate CD8(+) CTL, the alpha3 domain of MHC class I does not have to be located on the same molecule with the alpha1 and alpha2 domains of MHC class I. A low-avidity CD8(+) CTL line was significantly less sensitive to stimulation by the 227 mutant class I-peptide complexes in the presence of the H-2K(b) molecule. Thus, low-avidity CTL may not be able to take advantage of the interaction between CD8 and the alpha3 domain of non-presenting class I MHC molecules, perhaps because of a shorter dwell time for the TCR-MHC interaction.
Collapse
Affiliation(s)
- Igor M Belyakov
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
44
|
Biragyn A, Schiavo R, Olkhanud P, Sumitomo K, King A, McCain M, Indig FE, Almanzar G, Baatar D. Tumor-associated embryonic antigen-expressing vaccines that target CCR6 elicit potent CD8+ T cell-mediated protective and therapeutic antitumor immunity. THE JOURNAL OF IMMUNOLOGY 2007; 179:1381-8. [PMID: 17617631 PMCID: PMC2365706 DOI: 10.4049/jimmunol.179.2.1381] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Despite its potency, the wider use of immunotherapy for B cell malignancies is hampered by the lack of well-defined tumor-specific Ags. In this study, we demonstrate that an evolutionarily conserved 37-kDa immature laminin receptor protein (OFA-iLRP), a nonimmunogenic embryonic Ag expressed by a variety of tumors, is rendered immunogenic if targeted to the APCs using the CCR6 ligands MIP3alpha/CCL20 and mDF2beta. The CCR6 targeting facilitated efficient Ag cross-presentation and induction of tumor-neutralizing CTLs. Although the Ag targeting alone, without activation of dendritic cells (DCs), is proposed to induce tolerance, and MIP3alpha does not directly activate DCs, the MIP3alpha-based vaccine efficiently induced protective and therapeutic antitumor responses. The responses were as strong as those elicited by the OFA-iLRP fusions with moieties that activated DCs and Th1-type cytokine responses, mDF2beta, or mycobacterial Hsp70 Ag. Although the same cDNA encodes the dimerized high-affinity mature 67-kDa mLRP that is expressed in normal tissues to stabilize the binding of laminin to cell surface integrins, the vaccines expressing OFA-iLRP elicited long-term protective CD8(+) T cell-mediated memory responses against syngeneic B cell lymphoma, indicating the potential application of these simple vaccines as preventive and therapeutic formulations for human use.
Collapse
MESH Headings
- Animals
- Antigen Presentation/immunology
- Antigen-Presenting Cells/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Chemokine CCL20
- Chemokines, CC/immunology
- Cloning, Molecular
- Cytotoxicity, Immunologic
- Female
- Lymphoma, B-Cell/immunology
- Lymphoma, B-Cell/therapy
- Macrophage Inflammatory Proteins/immunology
- Mice
- Microscopy, Confocal
- Receptors, CCR6
- Receptors, Chemokine/immunology
- Receptors, Laminin/genetics
- Receptors, Laminin/immunology
- Vaccination
Collapse
Affiliation(s)
- Arya Biragyn
- Laboratory of Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The clinical course of ovarian cancer is often marked by periods of relapse and remission until chemotherapy resistance develops. Patients in remission with minimal disease burdens are ideally suited for the evaluation of immune-based strategies. The role of immune surveillance in improving outcome has been supported by the correlation of increased survival with the presence or absence of tumor-infiltrating lymphocytes in a given patient. Major obstacles to the development of successful immune strategies include the identification of tumor-restricted immunogenic targets, generation of a sufficient immune response to cause tumor rejection, and approaches to overcome evasion of immune attack. As optimal strategies are being developed, many questions remain. Some of the questions are as follows: What is the best antigen form (eg, peptides, proteins, or tumor lysates)? What are the appropriate adjuvants? Are monovalent or multivalent vaccines likely to be more effective? What is the optimal frequency and duration of vaccination? How should antigen-specific responses be monitored? How should the anticancer response be maintained? In this review, we will explore representative examples of immune strategies under investigation for patients with ovarian carcinoma that illustrate many of these issues. We will review ongoing phase III studies for patients in first clinical remission. Basic principles generic to all these immunotherapeutic approaches will be discussed in the hopes of yielding the most promising results as the field continues to evolve.
Collapse
Affiliation(s)
- Paul Sabbatini
- Medical Gynecologic Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York 10021, USA.
| | | |
Collapse
|
46
|
Affiliation(s)
- Jay A Berzofsky
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
47
|
Busick RY, Aguilera C, Quinn A. Dominant CTL-inducing epitopes on GAD65 are adjacent to or overlap with dominant Th-inducing epitopes. Clin Immunol 2007; 122:298-311. [PMID: 17174605 DOI: 10.1016/j.clim.2006.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 10/16/2006] [Accepted: 10/16/2006] [Indexed: 12/22/2022]
Abstract
Immune responses to GAD65 are associated with progression to T1D in NOD mice and humans. Our previous data suggested that dominant CTL-inducing and Th-inducing determinants might preferentially occur in proximal GAD65 sequences. Using a panel of 192 GAD65 peptides we discovered that four of the eight CTL-inducing peptides, including those most biologically relevant, were proximal to previously described I-A(g7)-restricted determinants that characterize natural islet autoimmunity in NOD mice. The CTL determinants 546-554 and 88-98 were presented by GAD65-expressing cells and were displayed on pancreatic LNC, along with 268-278, following beta cell damage. p546-554-specific CTL were detectable in young naive mice and transferred significant islet inflammation into NOD.scid mice. These findings demonstrate that unique regions of GAD65 may be favored during antigen processing, such that diverse dominant epitopes are produced from overlapping sequences, which can engage distinct T cell subsets. Additionally, cross-presentation may enhance GAD65-specific CTL responses in T1D.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cells, Cultured
- Cytotoxicity, Immunologic
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/immunology
- Epitopes, T-Lymphocyte/analysis
- Epitopes, T-Lymphocyte/immunology
- Glutamate Decarboxylase/genetics
- Glutamate Decarboxylase/immunology
- H-2 Antigens/immunology
- H-2 Antigens/metabolism
- Immunodominant Epitopes/analysis
- Immunodominant Epitopes/immunology
- Isoenzymes/genetics
- Isoenzymes/immunology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Molecular Sequence Data
- Peptide Mapping
- T-Lymphocytes, Cytotoxic/enzymology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
- Rhea Y Busick
- Department of Biological Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43606, USA
| | | | | |
Collapse
|
48
|
Ranasinghe C, Turner SJ, McArthur C, Sutherland DB, Kim JH, Doherty PC, Ramshaw IA. Mucosal HIV-1 Pox Virus Prime-Boost Immunization Induces High-Avidity CD8+ T Cells with Regime-Dependent Cytokine/Granzyme B Profiles. THE JOURNAL OF IMMUNOLOGY 2007; 178:2370-9. [PMID: 17277143 DOI: 10.4049/jimmunol.178.4.2370] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The quality of virus-specific CD8(+) CTL immune responses generated by mucosal and systemic poxvirus prime-boost vaccines were evaluated in terms of T cell avidity and single-cell analysis of effector gene expression. Intranasal (I.N.) immunization regimes generated higher avidity CTL responses specific for HIV K(d)Gag(197-205) (amino acid sequence AMQMLKETI; H-2K(d) binding) compared with i.m. immunization regime. Single-cell RT-PCR of K(d)Gag(197-205)-specific mucosal and systemic CTL revealed that the cytokine and granzyme B expression profiles were dependent on both the route and time after immunization. The I.N./i.m.-immunized group elicited elevated number of CTL-expressing granzyme B mRNA from the genitomucosal sites compared with the i.m./i.m. regime. Interestingly, CTL generated after both I.N. or i.m. immunization demonstrated expression of Th2 cytokine IL-4 mRNA that was constitutively expressed over time, although lower numbers were observed after I.N./I.N. immunization. Results suggest that after immunization, Ag-specific CTL expression of IL-4 may be an inherent property of the highly evolved poxvirus vectors. Current observations indicate that the quality of CTL immunity generated after immunization can be influenced by the inherent property of vaccine vectors and route of vaccine delivery. A greater understanding of these factors will be crucial for the development of effective vaccines in the future.
Collapse
Affiliation(s)
- Charani Ranasinghe
- Division of Immunology and Genetics, John Curtin School of Medical Research, Australian National University, Canberra, Australia.
| | | | | | | | | | | | | |
Collapse
|
49
|
Zhou W, Hashimoto K, Moore ML, Elias JA, Zhu Z, Durbin J, Colasurdo G, Rutigliano JA, Chiappetta CL, Goleniewska K, O'Neal JF, Graham BS, Stokes Peebles R. IL-13 is associated with reduced illness and replication in primary respiratory syncytial virus infection in the mouse. Microbes Infect 2006; 8:2880-9. [PMID: 17110149 PMCID: PMC1811125 DOI: 10.1016/j.micinf.2006.09.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2005] [Revised: 09/07/2006] [Accepted: 09/15/2006] [Indexed: 01/20/2023]
Abstract
The role of IL-13 in respiratory syncytial virus (RSV) immunopathogenesis is incompletely described. To assess the effect of IL-13 on primary RSV infection, transgenic mice which either overexpress IL-13 in the lung (IL-13 OE) or non-transgenic littermates (IL-13 NT) were challenged intranasally with RSV. IL-13 OE mice had significantly decreased peak viral titers four days after infection compared to non-transgenic littermates. In addition, IL-13 OE mice had significantly lower RSV-induced weight loss and reduced lung IFN-γ protein expression compared with IL-13 NT mice. In contrast, primary RSV challenge of IL-13 deficient mice resulted in a small, but statistically significant increase in viral titers on day four after infection, no difference in RSV-induced weight loss compared to wild type mice, and augmented IFN-γ production on day 6 after infection. In STAT1-deficient (STAT1 KO) mice, where primary RSV challenge produced high levels of IL-13 production in the lungs, treatment with an IL-13 neutralizing protein resulted in greater peak viral titers both four and six days after RSV and greater RSV-induced weight loss compared to mice treated with a control protein. These results suggest that IL-13 modulates illness from RSV-infection.
Collapse
Affiliation(s)
- Weisong Zhou
- Department of Medicine, T-1218 MCN, Vanderbilt University Medical Center, Nashville, TN 37232-2650, USA
| | - Koichi Hashimoto
- Department of Microbiology, Fukushima Medical University, 1st Hikariga-oka Fukushima, Fukushima 960-1295, Japan
| | - Martin L. Moore
- Department of Medicine, T-1218 MCN, Vanderbilt University Medical Center, Nashville, TN 37232-2650, USA
| | - Jack A. Elias
- Department of Medicine, Yale University, 333 Cedar Street/105LCI, New Haven, CT 06520-8057, USA
| | - Zhou Zhu
- Department of Medicine, Johns Hopkins University, 5501 Hopkins Bayview Boulevard, Baltimore, MD 21224, USA
| | - Joan Durbin
- Department of Pediatrics, Ohio State University, Columbus Children's Research Institute, 700 Children's Drive, Columbus, OH 43205, USA
| | - Giuseppe Colasurdo
- Department of Pediatrics, University of Texas-Houston Medical School, 6431 Fannin MSB 3 228, Houston, TX 77030, USA
| | - John A. Rutigliano
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 40, 40 Convent Drive, MSC 3017, Bethesda, MD 20892-3017, USA
| | - Constance L. Chiappetta
- Department of Pediatrics, University of Texas-Houston Medical School, 6431 Fannin MSB 3 228, Houston, TX 77030, USA
| | - Kasia Goleniewska
- Department of Medicine, T-1218 MCN, Vanderbilt University Medical Center, Nashville, TN 37232-2650, USA
| | - Jamye F. O'Neal
- Department of Medicine, T-1218 MCN, Vanderbilt University Medical Center, Nashville, TN 37232-2650, USA
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Building 40, 40 Convent Drive, MSC 3017, Bethesda, MD 20892-3017, USA
| | - R. Stokes Peebles
- Department of Medicine, T-1218 MCN, Vanderbilt University Medical Center, Nashville, TN 37232-2650, USA
- Corresponding author. Present address: Center for Lung Research, T-1217 MCN, Vanderbilt University Medical Center, Nashville, TN 37232-2650, USA. Tel.: +1 615 322 3412; fax: +1 615 343 7448.
| |
Collapse
|
50
|
Kornbluth RS, Stone GW. Immunostimulatory combinations: designing the next generation of vaccine adjuvants. J Leukoc Biol 2006; 80:1084-102. [PMID: 16931603 DOI: 10.1189/jlb.0306147] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Agents that activate dendritic cells are essential components for vaccines and can be conceptualized as molecular adjuvants. Other molecular adjuvants affect downstream factors that shape the resulting immune response. This review provides a compendium of recently studied molecular adjuvants, focusing on CD8+ T cell responses, which have important roles in HIV vaccines. Reference is also made to CD8+ T cell antitumor responses, where parallel studies of molecular adjuvants are being pursued. Molecular adjuvants can be considered in the following groups: TNF superfamily molecules such as CD40 ligand; agonists for TLRs; agonists for NAIP, CIITA, HET-E, TP-1-leucine-rich repeat pathway receptors, such as nucleotide-binding and oligomerization domain (NOD)1, NOD2, and cryopyrin; chemokines; ILs; CSFs; IFNs; alarmins; and purinergic P2X7 receptor agonists. Complementing these positively acting agents are strategies to reduce the immunosuppressive effects of CD4+CD25+ regulatory T cells and negatively acting factors such as TGF-beta, IL-10, suppressor of cytokine signaling 1, and programmed cell death-1 using neutralizing antibodies, antisense, and small interfering RNA. Especially effective are combinations of molecular adjuvants, which can elicit a massive expansion of antigen-specific CD8+ T cells and show unprecedented efficacy in vaccine and tumor models. Taken together, these new approaches provide significant incremental progress in the development of vaccines to elicit cell-mediated immunity against HIV and other pathogens.
Collapse
Affiliation(s)
- Richard S Kornbluth
- Department of Medicine, University of California San Diego, 9500 Gilman Dr., #0679, La Jolla, CA 92093-0679, USA.
| | | |
Collapse
|