1
|
Varanasi SK, Jaggi U, Hay N, Rouse BT. Hexokinase II may be dispensable for CD4 T cell responses against a virus infection. PLoS One 2018; 13:e0191533. [PMID: 29352298 PMCID: PMC5774810 DOI: 10.1371/journal.pone.0191533] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 01/05/2018] [Indexed: 11/18/2022] Open
Abstract
Activation of CD4 T cells leads to their metabolic reprogramming which includes enhanced glycolysis, catalyzed through hexokinase enzymes. Studies in some systems indicate that the HK2 isoform is the most up regulated isoform in activated T cells and in this report the relevance of this finding is evaluated in an infectious disease model. Genetic ablation of HK2 was achieved in only T cells and the outcome was evaluated by measures of T cell function. Our results show that CD4 T cells from both HK2 depleted and WT animals displayed similar responses to in vitro stimulation and yielded similar levels of Th1, Treg or Th17 subsets when differentiated in vitro. A modest increase in the levels of proliferation was observed in CD4 T cells lacking HK2. Deletion of HK2 led to enhanced levels of HK1 indicative of a compensatory mechanism. Finally, CD4 T cell mediated immuno-inflammatory responses to a virus infection were similar between WT and HK2 KO animals. The observations that the expression of HK2 appears non-essential for CD4 T cell responses against virus infections is of interest since it suggests that targeting HK2 for cancer therapy may not have untoward effects on CD4 T cell mediated immune response against virus infections.
Collapse
Affiliation(s)
- Siva Karthik Varanasi
- Department of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Ujjaldeep Jaggi
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Nissim Hay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Barry T. Rouse
- Department of Genome Science and Technology, University of Tennessee, Knoxville, Tennessee, United States of America
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
2
|
Kelly M, McNeel D, Fisch P, Malkovsky M. Immunological considerations underlying heat shock protein-mediated cancer vaccine strategies. Immunol Lett 2017; 193:1-10. [PMID: 29129721 DOI: 10.1016/j.imlet.2017.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/01/2017] [Accepted: 11/05/2017] [Indexed: 12/31/2022]
Abstract
The success of active immunotherapies in the prevention of many infectious diseases over the course of over 200 years has lead scientists to wonder if the same principles could be applied to cancer. Antigen-specific active immunotherapies for the treatment of cancer have been researched for over two decades, however, the overwhelming majority of these studies have failed to stimulate robust clinical responses. It is clear that current active immunotherapy research should incorporate methods to increase the immunostimulatory capacity of these therapies. To directly address this need, we propose the addition of the immunostimulatory heat shock proteins (HSPs) to active immunotherapeutic strategies to augment their efficacy. Heat shock proteins are a family of highly conserved intracellular chaperone proteins, and are the most abundant family proteins inside cells. This ubiquity, and their robust immunostimulatory capacity, points to their importance in regulation of intracellular processes and, therefore, indicators of loss of cellular integrity if found extracellularly. Thus, we emphasize the importance of taking into consideration the location of vaccine-derived HSP/tumor-antigen complexes when designing active immunotheraputic strategies.
Collapse
Affiliation(s)
- Matthew Kelly
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Douglas McNeel
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA; Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Paul Fisch
- Universitätsklinikum Freiburg, Institut für Pathologie, Freiburg, Germany
| | - Miroslav Malkovsky
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA; Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
3
|
Wald A, Koelle DM, Fife K, Warren T, Leclair K, Chicz RM, Monks S, Levey DL, Musselli C, Srivastava PK. Safety and immunogenicity of long HSV-2 peptides complexed with rhHsc70 in HSV-2 seropositive persons. Vaccine 2011; 29:8520-9. [PMID: 21945262 DOI: 10.1016/j.vaccine.2011.09.046] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 09/07/2011] [Accepted: 09/12/2011] [Indexed: 12/27/2022]
Abstract
HSV-2, the primary causative agent of genital herpes, establishes latency in sensory ganglia and reactivates causing recurrent lesions and viral shedding. Induction or expansion of CD4(+) and CD8(+) T cell responses are expected to be important for a successful therapeutic vaccine against HSV-2. A candidate vaccine consisting of 32 synthetic 35mer HSV-2 peptides non-covalently complexed with recombinant human Hsc70 protein (named HerpV, formerly AG-707) was tested for safety and immunogenicity in a Phase I study. These peptides are derived from 22 HSV-2 proteins representative of all phases of viral replication. Thirty-five HSV-2 infected participants were randomized and treated in one of four groups: HerpV+QS-21 (saponin adjuvant), HerpV, QS-21, or vehicle. The vaccine was well tolerated and safe. All seven participants with evaluable samples who were administered HerpV with QS-21 demonstrated a statistically significant CD4(+) T cell response to HSV-2 antigens, and the majority of such participants demonstrated a statistically significant CD8(+) T cell response as well. To our knowledge, this is the first candidate vaccine against HSV-2 to demonstrate a broad CD4(+) and CD8(+) T cell response in HSV-2(+) participants, and the first HSP-based vaccine to show immune responses against viral antigens in humans.
Collapse
Affiliation(s)
- Anna Wald
- Department of Medicine, University of Washington, Seattle, WA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Mo A, Musselli C, Chen H, Pappas J, Leclair K, Liu A, Chicz RM, Truneh A, Monks S, Levey DL, Srivastava PK. A heat shock protein based polyvalent vaccine targeting HSV-2: CD4(+) and CD8(+) cellular immunity and protective efficacy. Vaccine 2011; 29:8530-41. [PMID: 21767588 DOI: 10.1016/j.vaccine.2011.07.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 06/24/2011] [Accepted: 07/04/2011] [Indexed: 11/25/2022]
Abstract
Efforts to develop a subunit vaccine against genital herpes have been hampered by lack of knowledge of the protective antigens of HSV-2, the causative agent of the disease. Vaccines based either on selected antigens or attenuated live virus approaches have not demonstrated meaningful clinical activity. We present here results of a therapeutic vaccine candidate, HerpV (formerly called AG-707), consisting of 32 HSV-2 peptides derived from 22 HSV-2 proteins, complexed non-covalently to the HSP70 chaperone and formulated with QS-21 saponin adjuvant. HerpV is observed to be immunogenic, generating CD4(+) and CD8(+) T cell responses in three mouse strains including HLA-A2 transgenic mice. Optimal T cell stimulation was dependent on the synergistic adjuvant properties of QS-21 with hsp70. The vaccine provided significant protection from viral challenge in a mouse prophylaxis model and showed signals of activity in a guinea pig therapeutic model of existing infection. Peripheral blood mononuclear cells from human HSV-2(+) subjects also showed reactivity in vitro to a subset of individual peptides and to the pool of all 32 peptides. Recombinant human Hsc70 complexed with the 32 peptides also stimulated the expansion of CD8(+) T cells from HSV-2(+) subjects in vitro. These studies demonstrate that HerpV is a promising immunotherapy candidate for genital herpes, and provide a foundation for evaluating HerpV in human HSV-2(+) subjects with the intent of eliciting CD4(+) and CD8(+) T cell responses to a broad array of viral antigens.
Collapse
|
5
|
Takemoto S, Nishikawa M, Guan X, Ohno Y, Yata T, Takakura Y. Enhanced Generation of Cytotoxic T Lymphocytes by Heat Shock Protein 70 Fusion Proteins Harboring Both CD8+ T Cell and CD4+ T Cell Epitopes. Mol Pharm 2010; 7:1715-23. [DOI: 10.1021/mp1001069] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Seiji Takemoto
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Makiya Nishikawa
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Xin Guan
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuji Ohno
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tomoya Yata
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
6
|
Leao IC, Ganesan P, Armstrong TD, Jaffee EM. Effective depletion of regulatory T cells allows the recruitment of mesothelin-specific CD8 T cells to the antitumor immune response against a mesothelin-expressing mouse pancreatic adenocarcinoma. Clin Transl Sci 2010; 1:228-39. [PMID: 20357913 DOI: 10.1111/j.1752-8062.2008.00070.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Vaccine-induced CD8(+) T-cell responses can eradicate developing tumors in vivo in mouse models. Translating these successes into approved treatments for cancer patients has been challenging, since many of these models lack expression of clinically proven/relevant tumor antigens. We have shown that mesothelin is a clinically relevant CD8(+) T-cell target in human pancreas cancer, which is also highly conserved among species. Here, we utilize the murine mesothelin-expressing pancreatic tumor model (Panc02) to identify the immune-relevant mesothelin-derived peptides and study interventions that enhance the antitumor response. We first screened overlapping peptides of the entire murine mesothelin protein to identify two new CD8(+) mesothelin-restricted epitopes. These peptides were then evaluated for recognition by vaccine-induced T cells from mice treated with vaccine in sequence with low-dose cyclophosphamide (CY) and an anti-CD25 IL-2Ralpha monoclonal antibody (PC61). These treatments are both known to deplete subpopulations of T regulatory cells (Tregs). Our findings demonstrate that combined Treg-depleting therapies synergize to enhance vaccine efficacy. Furthermore, our data supports mesothelin as a relevant antigen in murine and clinical models and the use of Panc02 as a clinically relevant murine model of pancreatic cancer for evaluating antigen-targeted immunotherapies in immune-tolerant hosts.
Collapse
Affiliation(s)
- Ihid C Leao
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|
7
|
Yen HR, Harris TJ, Wada S, Grosso JF, Getnet D, Goldberg MV, Liang KL, Bruno TC, Pyle KJ, Chan SL, Anders RA, Trimble CL, Adler AJ, Lin TY, Pardoll DM, Huang CT, Drake CG. Tc17 CD8 T cells: functional plasticity and subset diversity. THE JOURNAL OF IMMUNOLOGY 2009; 183:7161-8. [PMID: 19917680 DOI: 10.4049/jimmunol.0900368] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
IL-17-secreting CD8 T cells (Tc17) have been described in several settings, but little is known regarding their functional characteristics. While Tc1 cells produced IFN-gamma and efficiently killed targets, Tc17 cells lacked lytic function in vitro. Interestingly, the small numbers of IFN-gamma-positive or IL-17/IFN-gamma-double-positive cells generated under Tc17 conditions also lacked lytic activity and expressed a similar pattern of cell surface proteins to IL-17-producing cells. As is the case for Th17 (CD4) cells, STAT3 is important for Tc17 polarization, both in vitro and in vivo. Adoptive transfer of highly purified, Ag-specific IL-17-secreting Tc17 cells into Ag-bearing hosts resulted in near complete conversion to an IFN-gamma-secreting phenotype and substantial pulmonary pathology, demonstrating functional plasticity. Tc17 also accumulated to a greater extent than did Tc1 cells, suggesting that adoptive transfer of CD8 T cells cultured in Tc17 conditions may have therapeutic potential for diseases in which IFN-gamma-producing cells are desired.
Collapse
Affiliation(s)
- Hung-Rong Yen
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Abstract
Heat shock proteins (HSPs) are immunogenic, with the specificity of the immune response provided by the peptides that they chaperone. Binding of cell surface receptors by HSPs is central to the elicitation of the innate and adaptive immune responses obtained after vaccination and also plays a physiologic role in cross-priming. These effects of HSPs have been exploited in prophylaxis and therapy of cancer and infectious disease. The data obtained from murine studies have been translated into ongoing clinical trials of cancer of which the most recent results are provided here.
Collapse
Affiliation(s)
- Robert J Binder
- Center for Immunotherapy of Cancer and Infectious Diseases, Department of Immunology, University of Connecticut School of Medicine, Farmington, Connecticut 06030-1920, USA.
| |
Collapse
|
9
|
Aloysius MM, Mc Kechnie AJ, Robins RA, Verma C, Eremin JM, Farzaneh F, Habib NA, Bhalla J, Hardwick NR, Satthaporn S, Sreenivasan T, El-Sheemy M, Eremin O. Generation in vivo of peptide-specific cytotoxic T cells and presence of regulatory T cells during vaccination with hTERT (class I and II) peptide-pulsed DCs. J Transl Med 2009; 7:18. [PMID: 19298672 PMCID: PMC2674878 DOI: 10.1186/1479-5876-7-18] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2009] [Accepted: 03/19/2009] [Indexed: 12/23/2022] Open
Abstract
Background Optimal techniques for DC generation for immunotherapy in cancer are yet to be established. Study aims were to evaluate: (i) DC activation/maturation milieu (TNF-α +/- IFN-α) and its effects on CD8+ hTERT-specific T cell responses to class I epitopes (p540 or p865), (ii) CD8+ hTERT-specific T cell responses elicited by vaccination with class I alone or both class I and II epitope (p766 and p672)-pulsed DCs, prepared without IFN-α, (iii) association between circulating T regulatory cells (Tregs) and clinical responses. Methods Autologous DCs were generated from 10 patients (HLA-0201) with advanced cancer by culturing CD14+ blood monocytes in the presence of GM-CSF and IL-4 supplemented with TNF-α [DCT] or TNF-α and IFN-α [DCTI]. The capacity of the DCs to induce functional CD8+ T cell responses to hTERT HLA-0201 restricted nonapeptides was assessed by MHC tetramer binding and peptide-specific cytotoxicity. Each DC preparation (DCT or DCTI) was pulsed with only one type of hTERT peptide (p540 or p865) and both preparations were injected into separate lymph node draining regions every 2–3 weeks. This vaccination design enabled comparison of efficacy between DCT and DCTI in generating hTERT peptide specific CD8+ T cells and comparison of class I hTERT peptide (p540 or p865)-loaded DCT with or without class II cognate help (p766 and p672) in 6 patients. T regulatory cells were evaluated in 8 patients. Results (i) DCTIs and DCTs, pulsed with hTERT peptides, were comparable (p = 0.45, t-test) in inducing peptide-specific CD8+ T cell responses. (ii) Class II cognate help, significantly enhanced (p < 0.05, t-test) peptide-specific CD8+T cell responses, compared with class I pulsed DCs alone. (iii) Clinical responders had significantly lower (p < 0.05, Mann-Whitney U test) T regs, compared with non-responders. 4/16 patients experienced partial but transient clinical responses during vaccination. Vaccination was well tolerated with minimal toxicity. Conclusion Addition of IFN-α to ex vivo monocyte-derived DCs, did not significantly enhance peptide-specific T cell responses in vivo, compared with TNF-α alone. Class II cognate help significantly augments peptide-specific T cell responses. Clinically favourable responses were seen in patients with low levels of circulating T regs.
Collapse
Affiliation(s)
- Mark M Aloysius
- Section of Surgery, Biomedical Research Unit, Nottingham Digestive Diseases Centre, University of Nottingham, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Pack CD, Gierynska M, Rouse BT. An intranasal heat shock protein based vaccination strategy confers protection against mucosal challenge with herpes simplex virus. HUMAN VACCINES 2008; 4:360-4. [PMID: 18382144 DOI: 10.4161/hv.4.5.5978] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Herpes simplex virus-1 (HSV-1) represents a significant obstacle for vaccine designers, despite decades of investigation. The virus primarily infects the host at vulnerable mucosal surfaces that progresses to lesion development, latency in nervous tissue, and possible reactivation. Therefore, protection at the site of infection is crucial. Mucosal adjuvants are critical for the development of an effective vaccine approach and heat-shock protein 70 (Hsp70) represents an attractive candidate for this purpose. This study demonstrates that Hsp70 coupled to gB498-505 from HSV-1 induced mucosal and systemic priming of CD8(+) T cells capable of protecting C57BL/6 mice against a lethal vaginal challenge. Elevated gB-specific cytotoxicity was observed in the spleen of mice immunized with conjugated Hsp70 and gB498-505. In addition, both vaginal IFNgamma levels and viral clearance were enhanced in mice mucosally immunized with Hsp70 and gB peptide versus peptide only control mice or mice receiving Hsp70 and a control peptide. These studies demonstrate that Hsp70 can be used as an effective mucosal adjuvant capable of generating a protective cell-mediated immune response against HSV-1.
Collapse
Affiliation(s)
- Christopher D Pack
- Department of Microbiology, The University of Tennessee, Knoxville, Tennessee 37996, USA
| | | | | |
Collapse
|
11
|
Natural killer cells as novel helpers in anti-herpes simplex virus immune response. J Virol 2008; 82:10820-31. [PMID: 18715907 DOI: 10.1128/jvi.00365-08] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Innate defenses help to eliminate infection, but some of them also play a major role in shaping the magnitude and efficacy of the adaptive immune response. With regard to influencing subsequent adaptive immunity, NK cells aided by dendritic cells may be the most relevant components of the innate reaction to herpes simplex virus (HSV) infection. We confirm that mice lacking or depleted of NK cells are susceptible to HSV-induced lesions. The quantity and quality of CD8(+) cytotoxic T lymphocytes generated in the absence of NK cells were diminished, thereby contributing to susceptibility to HSV-induced encephalitis. We demonstrate a novel helper role for NK cells, in that NK cells compensate for the loss of CD4 helper T cells and NK cell supplementation enhances the function of wild type anti-HSV CD8 T cells. In addition, NK cells were able to partially rescue the dysfunctional CD8(+) T cells generated in the absence of CD4 T helper cells, thereby performing a novel rescue function. Hence, NK cells may well be exploited for enhancing and rescuing the T-cell response in situations where the CD4 helper response is affected.
Collapse
|
12
|
Abstract
Almost 60 years ago, the pioneering work of George Klein and others showed that cancers could be made targets for the immune system. Identification of the tumor targets, known as tumor antigens, became a focus in cancer biology that led to the discovery of the immunological properties of heat-shock proteins (HSPs) in 1986 by Pramod Srivastava and colleagues. Since then, the use of HSPs in the therapeutics of cancer and infectious disease in several clinical trials has been guided by our understanding of the role and effects of HSPs in adaptive and innate immune responses, investigated primarily in mice. This review will highlight the immunological properties of HSPs as we understand them today and review the clinical work on human cancers. Several Phase I and II clinical trials in different types of cancer that have been completed, as well as ongoing Phase III trials, will be summarized.
Collapse
Affiliation(s)
- Robert J Binder
- University of Pittsburgh, E1051, BSTWR, 200 Lothrop Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
13
|
Wong RM, Scotland RR, Lau RL, Wang C, Korman AJ, Kast WM, Weber JS. Programmed death-1 blockade enhances expansion and functional capacity of human melanoma antigen-specific CTLs. Int Immunol 2008; 19:1223-34. [PMID: 17898045 DOI: 10.1093/intimm/dxm091] [Citation(s) in RCA: 174] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Negative co-stimulatory signaling mediated via cell surface programmed death (PD)-1 expression modulates T and B cell activation and is involved in maintaining peripheral tolerance. In this study, we examined the effects of a fully human PD-1-abrogating antibody on the in vitro expansion and function of human vaccine-induced CD8+ T cells (CTLs) specific for the melanoma-associated antigens glycoprotein 100 (gp100) and melanoma antigen recognized by T cells (MART)-1. PD-1 blockade during peptide stimulation augmented the absolute numbers of CD3+, CD4+, CD8+ and gp100/MART-1 MHC:peptide tetramer+ CTLs. This correlated with increased frequencies of IFN-gamma-secreting antigen-specific cells and augmented lysis of gp100+/MART-1+ melanoma targets. PD-1 blockade also increased the fraction of antigen-specific CTLs that recognized melanoma targets by degranulation, suggesting increased recognition efficiency for cognate peptide. The increased frequencies and absolute numbers of antigen-specific CTLs by PD-1 blockade resulted from augmented proliferation, not decreased apoptosis. Kinetic analysis of cytokine secretion demonstrated that PD-1 blockade increased both type-1 and type-2 cytokine accumulation in culture without any apparent skewing of the cytokine repertoire. These findings have implications for developing new cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Raymond M Wong
- Department of Medicine, University of Southern California, 1441 Eastlake Avenue, Room 6428, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Radcliffe JN, Roddick JS, Stevenson FK, Thirdborough SM. Prolonged Antigen Expression following DNA Vaccination Impairs Effector CD8+ T Cell Function and Memory Development. THE JOURNAL OF IMMUNOLOGY 2007; 179:8313-21. [DOI: 10.4049/jimmunol.179.12.8313] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
15
|
Cao Q, Wang L, Du F, Sheng H, Zhang Y, Wu J, Shen B, Shen T, Zhang J, Li D, Li N. Downregulation of CD4+CD25+ regulatory T cells may underlie enhanced Th1 immunity caused by immunization with activated autologous T cells. Cell Res 2007; 17:627-37. [PMID: 17563757 DOI: 10.1038/cr.2007.46] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Regulatory T cells (Treg) play important roles in immune system homeostasis, and may also be involved in tumor immunotolerance by suppressing Th1 immune response which is involved in anti-tumor immunity. We have previously reported that immunization with attenuated activated autologous T cells leads to enhanced anti-tumor immunity and upregulated Th1 responses in vivo. However, the underlying molecular mechanisms are not well understood. Here we show that Treg function was significantly downregulated in mice that received immunization of attenuated activated autologous T cells. We found that Foxp3 expression decreased in CD4+CD25+ T cells from the immunized mice. Moreover, CD4+CD25+Foxp3+ Treg obtained from immunized mice exhibited diminished immunosuppression ability compared to those from naïve mice. Further analysis showed that the serum of immunized mice contains a high level of anti-CD25 antibody (about 30 ng/ml, p<0.01 vs controls). Consistent with a role of anti-CD25 response in the downregulation of Treg, adoptive transfer of serum from immunized mice to naïve mice led to a significant decrease in Treg population and function in recipient mice. The triggering of anti-CD25 response in immunized mice can be explained by the fact that CD25 was induced to a high level in the ConA activated autologous T cells used for immunization. Our results demonstrate for the first time that immunization with attenuated activated autologous T cells evokes anti-CD25 antibody production, which leads to impeded CD4+CD25+Foxp3+ Treg expansion and function in vivo. We suggest that dampened Treg function likely contributes to enhanced Th1 response in immunized mice and is at least part of the mechanism underlying the boosted anti-tumor immunity.
Collapse
Affiliation(s)
- Qi Cao
- Department of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Zeng R, Qi X, Gong W, Mei X, Wei L, Ma C, Yin X. Long-lasting balanced immunity and protective efficacy against respiratory syncytial virus in mice induced by a recombinant protein G1F/M2. Vaccine 2007; 25:7422-8. [PMID: 17850930 DOI: 10.1016/j.vaccine.2007.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2007] [Revised: 08/03/2007] [Accepted: 08/07/2007] [Indexed: 11/18/2022]
Abstract
Respiratory syncytial virus (RSV) is the primary cause of serious lower respiratory tract illness in young children. We have engineered a recombinant candidate vaccine G1F/M2, consisting of a cytotoxic T lymphocyte (CTL) epitope of RSV-M2 protein and a domain of RSV-G protein. In this study, the long-term immunogenicity and protective effect were evaluated. In G1F/M2-immunized mice, special antibodies lasted for more than 19 weeks, and the IgG1/IgG2a ratio remained a balanced level till the end of the study, suggesting mixed Th1/Th2 type of responses. Concomitantly, G1F/M2 elicited long-lived RSV-specific CTL activity that was detectable at 12 weeks after the final immunization. Stronger CTL responses were induced with immunization once more at 13 weeks after the last immunization in G1F/M2-primed mice than those in F/M2-primed mice. These results suggest that G1F/M2-induced long-lasting balanced humoral and cellular immunity responses, and immunological memory in mice. Furthermore, following RSV challenge, long-term protective efficacy was observed. RSV replication in lungs of G1F/M2-primed mice elicited also mixed Th1/Th2 responses, a property that is considered advantageous for the safety of an RSV vaccine. Therefore, G1F/M2 is a promising RSV subunit vaccine.
Collapse
Affiliation(s)
- Ruihong Zeng
- Department of Immunology, Hebei Medical University, Shijiazhuang 050017, China.
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The effectiveness of T-cell-mediated immunotherapy of cancer depends on both an optimal immunostimulatory context of the therapy and the proper selection with respect to quality and quantity of the targeted tumor-associated antigens (TAA), and, more precisely, the T-cell epitopes contained in these tumor proteins. Our progressing insight in human leukocyte antigen (HLA) class I and class II antigen processing and presentation mechanisms has improved the prediction by reverse immunology of novel cytotoxic T lymphocyte and T-helper cell epitopes within known antigens. Computer algorithms that in silico predict HLA class I and class II binding, proteasome cleavage patterns and transporter associated with antigen processing translocation are now available to expedite epitope identification. The advent of genomics allows a high-throughput screening for tumor-specific transcripts and mutations, with that identifying novel shared and unique TAA. The increasing power of mass spectrometry and proteomics will lead to the direct identification from the tumor cell surface of numerous novel tumor-specific HLA class I and class II presented ligands. Together, the expanded repertoire of tumor-specific T-cell epitopes will enable more precise immunomonitoring and the development of effective epitope-defined adoptive T-cell transfer and multi-epitope-based vaccination strategies targeting epitopes derived from a wider diversity of TAA presented in a broader array of HLA molecules.
Collapse
Affiliation(s)
- J H Kessler
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| | | |
Collapse
|
18
|
de Alencar BCG, Araújo AFS, Penido MLO, Gazzinelli RT, Rodrigues MM. Cross-priming of long lived protective CD8+ T cells against Trypanosoma cruzi infection: importance of a TLR9 agonist and CD4+ T cells. Vaccine 2007; 25:6018-27. [PMID: 17629597 DOI: 10.1016/j.vaccine.2007.05.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2007] [Revised: 04/09/2007] [Accepted: 05/13/2007] [Indexed: 11/26/2022]
Abstract
We recently described that vaccination of mice with a glutathione S transferase fusion protein representing amino acids 261-500 of the Amastigote Surface Protein-2 efficiently cross-primed protective CD8+ T cells against a lethal challenge with the human protozoan parasite Trypanosoma cruzi. In this study, we initially established that this protective immunity was long lived. Subsequently, we studied the importance of TLR9 agonist CpG ODN 1826, TLR4 and CD4+ T cells for the generation of these protective CD8+ T cells. We found that: (i) the TLR9 agonist CpG ODN 1826 improved the efficiency of protective immunity; (ii) TLR4 is not relevant for priming of specific CD8+ T cells; (iii) CD4+ T cells are critical for priming of memory/protective CD8+ T cells.
Collapse
Affiliation(s)
- Bruna C G de Alencar
- Centro Interdisciplinar de Terapia Gênica, Universidade Federal de São Paulo, Escola Paulista de Medicina, Brazil
| | | | | | | | | |
Collapse
|
19
|
Goldberg MV, Maris CH, Hipkiss EL, Flies AS, Zhen L, Tuder RM, Grosso JF, Harris TJ, Getnet D, Whartenby KA, Brockstedt DG, Dubensky TW, Chen L, Pardoll DM, Drake CG. Role of PD-1 and its ligand, B7-H1, in early fate decisions of CD8 T cells. Blood 2007; 110:186-92. [PMID: 17392506 PMCID: PMC1896112 DOI: 10.1182/blood-2006-12-062422] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Expression of the PD-1 receptor on T cells has been shown to provide an important inhibitory signal that down-modulates peripheral effector responses in normal tissues and tumors. Furthermore, PD-1 up-regulation on chronically activated T cells can maintain them in a partially reversible inactive state. The function of PD-1 in the very early stages of T-cell response to antigen in vivo has not been fully explored. In this study, we evaluate the role of PD-1 and its 2 B7 family ligands, B7-H1 (PD-L1) and B7-DC (PD-L2), in early fate decisions of CD8 T cells. We show that CD8 T cells specific for influenza hemagglutinin (HA) expressed as a self-antigen become functionally tolerized and express high levels of surface PD-1 by the time of their first cell division. Blockade of PD-1 or B7-H1, but not B7-DC, at the time of self-antigen encounter mitigates tolerance induction and results in CD8 T-cell differentiation into functional cytolytic T lymphocytes (CTLs). These findings demonstrate that, in addition to modulating effector functions in the periphery, B7-H1:PD-1 interactions regulate early T-cell-fate decisions.
Collapse
Affiliation(s)
- Monica V Goldberg
- Department of Oncology, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Srivastava PK. Immunotherapy for human cancer using heat shock protein-peptide complexes. Curr Oncol Rep 2006; 7:104-8. [PMID: 15717943 DOI: 10.1007/s11912-005-0035-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Heat shock proteins (HSPs) are primordial and abundant molecules expressed in all cells. Publications starting in 1984 have shown that immunization of mice, rats, and frogs with purified preparations of selected HSPs isolated from cancers leads to protective immunity against the cancer used as the source of the HSP. The basis of the tumor-specific immunogenicity of these molecules lies not in the molecules themselves but in the array of peptides, including antigenic peptides chaperoned by them. These experiments and the ideas derived from them form the basis of an approach to immunotherapy for human cancer that began in 1995 and is now in full swing.
Collapse
Affiliation(s)
- Pramod K Srivastava
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut School of Medicine, Farmington, CT 06030-1601, USA.
| |
Collapse
|
21
|
Durrant LG, Ramage JM. Development of cancer vaccines to activate cytotoxic T lymphocytes. Expert Opin Biol Ther 2006; 5:555-63. [PMID: 15934833 DOI: 10.1517/14712598.5.4.555] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cancer vaccines have been shown to stimulate cytotoxic T lymphocyte (CTL) responses in a variety of cancer patients. However, the response is often of low frequency and moderate avidity, and does not result in objective clinical responses. This is related to the target antigens, which are usually over-expressed self-antigens that elicit tolerogenic and regulatory immune responses, resulting in deletion or inactivation of high-avidity T cells. Although moderate-avidity T cells can be efficient killers, tumours are often poor targets as they express a variety of molecules to protect them from cell-mediated immunity. Adoptive transfer of large numbers of high-avidity T cells has been shown to induce regression of bulky disease, proving that immune responses can effectively eradicate tumours. New approaches that target activated dendritic cells in vivo, resulting in cross-presentation of CTL epitopes and release of cytokines that suppress regulatory T cells, have resulted in the production of T cells with sufficient avidity to kill tumour target cells. These approaches in combination with regimes, such as cytokine therapy, chemotherapy or radiotherapy, that modulate effector costimulatory expression on tumour targets may result in more effective second-generation cancer vaccines.
Collapse
Affiliation(s)
- L G Durrant
- University of Nottingham, Institute of Infections, Immunity and Inflammation, Department of Clinical Oncology, City Hospital, Hucknall Road, NG5 1PB, UK.
| | | |
Collapse
|
22
|
Kumaraguru U, Banerjee K, Rouse BT. In vivo rescue of defective memory CD8+ T cells by cognate helper T cells. J Leukoc Biol 2005; 78:879-87. [PMID: 16081600 DOI: 10.1189/jlb.0105007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The magnitude and efficacy of CD8(+) T cell memory may notably regress, especially if immune induction occurs in the absence of adequate CD4(+) help. This report demonstrates that this CD8(+) memory malfunction could be remedied if a source of cognate antigen-recognizing helper cells were provided during recall. The inability of adoptive transfer of memory SIINFEKL-specific CD8 cells to reject tumors was overcome if recipients were primed for ovalbumin-specific helper cell responses. Additionally, animals primed for a SIINFEKL-specific memory response and incapable of rejecting the tumor could regain protective immunity if given helper cells. This pattern of CD8(+) T cell functional rescue or reprogramming by helper cell transfer was replicated using a Herpes simplex virus antiviral immunity system. Our results could mean that therapeutic vaccine approaches could be designed to compensate situations that have defective CD8(+) T cell function.
Collapse
|
23
|
Fan CF, Zeng RH, Sun CY, Mei XG, Wang YF, Liu Y. Fusion of DsbA to the N-terminus of CTL chimeric epitope, F/M2:81-95, of respiratory syncytial virus prolongs protein- and virus-specific CTL responses in Balb/c mice. Vaccine 2005; 23:2869-75. [PMID: 15780735 DOI: 10.1016/j.vaccine.2004.11.066] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2004] [Accepted: 11/24/2004] [Indexed: 11/15/2022]
Abstract
In an effort to seek a means of inducing long lasting respiratory syncytial virus-specific CTL responses in mice, we constructed a new recombinant protein, DsbA-F/M2:81-95, by fusing carrier protein DsbA (disulfide bond isomerase) to the N-terminus of CTL chimeric epitope F/M2:81-95 of this virus. DsbA-F/M2:81-95 can induce effectively virus-specific CTL responses as well as protective immunity without association with enhanced disease. Furthermore, compared with F/M2:81-95 alone, it increases the longevity of CTL responses in vivo up to 2.93 folds. Our study emphasizes that appropriate stimulation of non-antigen-specific T helper cells is essential to induce long lasting CD8+ CTL, and also implies DsbA-F/M2:81-95 may be a promising candidate for RSV vaccine development since it is an efficacious and safe immunogen.
Collapse
Affiliation(s)
- Chang-Fa Fan
- Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, PR China.
| | | | | | | | | | | |
Collapse
|
24
|
Pack CD, Kumaraguru U, Suvas S, Rouse BT. Heat-shock protein 70 acts as an effective adjuvant in neonatal mice and confers protection against challenge with Herpes Simplex Virus. Vaccine 2005; 23:3526-34. [PMID: 15855011 DOI: 10.1016/j.vaccine.2005.01.152] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2004] [Revised: 12/17/2004] [Accepted: 01/31/2005] [Indexed: 11/21/2022]
Abstract
Immunization of the neonate is a highly desirable goal for vaccine developers, since the neonate is profoundly susceptible to a number of viral and bacterial pathogens. The neonatal immune system tends to generate Th2 recall responses, known as neonatal tolerance, which may not protect against viral challenge later in life. In this study we demonstrate that a potent immune proinflammatory stimulator, heat-shock protein 70 (hsp70), can act as an effective and safe adjuvant in neonates. Priming of neonates with hsp70 coupled to a viral MHC Class I-restricted epitope (gB498-505) and injection with recombinant gB generated strong cytotoxic T lymphocyte (CTL) responses and a Th1 primary T helper cell response during the neonatal period. In addition, enhanced CTL and predominant Th1 recall responses to viral antigens were observed following secondary challenge as adults. These responses were sufficient to allow protection against a lethal challenge with Herpes Simplex Virus Type-1 (HSV-1). Therefore, hsp70 in conjunction with viral epitopes and recombinant viral protein can perhaps prime protective immune responses to herpes viruses early in life when infection, which can be life-threatening, and the establishment of latency frequently occur.
Collapse
Affiliation(s)
- Christopher D Pack
- Department of Microbiology, The University of Tennessee, Knoxville, TN 37996, USA
| | | | | | | |
Collapse
|