1
|
Li QH, Zhao QY, Yang WJ, Jiang AF, Ren CE, Meng YH. Beyond Immune Balance: The Pivotal Role of Decidual Regulatory T Cells in Unexplained Recurrent Spontaneous Abortion. J Inflamm Res 2024; 17:2697-2710. [PMID: 38707955 PMCID: PMC11070170 DOI: 10.2147/jir.s459263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Recurrent spontaneous abortion (RSA) is defined as two or more consecutive pregnancy failures, which brings tremendous stress to women of childbearing age and seriously affects family well-being. However, the reason in about 50% of cases remains unknown and is defined as unexplained recurrent spontaneous abortion (URSA). The immunological perspective in URSA has attracted widespread attention in recent years. The embryo is regarded as a semi-allogeneic graft to the mother. A successful pregnancy requires transition to an immune environment conducive to embryo survival at the maternal-fetal interface. As an important member of regulatory immunity, regulatory T (Treg) cells play a key role in regulating immune tolerance at the maternal-fetal interface. This review will focus on the phenotypic plasticity and lineage stability of Treg cells to illustrate its relationship with URSA.
Collapse
Affiliation(s)
- Qing-Hui Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Qiu-Yan Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Wei-Jing Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Ai-Fang Jiang
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Chun-E Ren
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Yu-Han Meng
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| |
Collapse
|
2
|
Chakravarti AR, Groer CE, Gong H, Yudistyra V, Forrest ML, Berkland CJ. Design of a Tumor Binding GMCSF as Intratumoral Immunotherapy of Solid Tumors. Mol Pharm 2023; 20:1975-1989. [PMID: 36825806 DOI: 10.1021/acs.molpharmaceut.2c00897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023]
Abstract
Next-generation cancer immunotherapies may utilize immunostimulants to selectively activate the host immune system against tumor cells. Checkpoint inhibitors (CPIs) like anti-PD1/PDL-1 that inhibit immunosuppression have shown unprecedented success but are only effective in the 20-30% of patients that possess an already "hot" (immunogenic) tumor. In this regard, intratumoral (IT) injection of immunostimulants is a promising approach since they can work synergistically with CPIs to overcome the resistance to immunotherapies by inducing immune stimulation in the tumor. One such immunostimulant is granulocyte macrophage-colony-stimulating factor (GMCSF) that functions by recruiting and activating antigen-presenting cells (dendritic cells) in the tumor, thereby initiating anti-tumor immune responses. However, key problems with GMCSF are lack of efficacy and the risk of systemic toxicity caused by the leakage of GMCSF from the tumor tissue. We have designed tumor-retentive versions of GMCSF that are safe yet potent immunostimulants for the local treatment of solid tumors. The engineered GMCSFs (eGMCSF) were synthesized by recombinantly fusing tumor-ECM (extracellular matrix) binding peptides to GMCSF. The eGMCSFs exhibited enhanced tumor binding and potent immunological activity in vitro and in vivo. Upon IT administration, the tumor-retentive eGMCSFs persisted in the tumor, thereby alleviating systemic toxicity, and elicited localized immune activation to effectively turn an unresponsive immunologically "cold" tumor "hot".
Collapse
Affiliation(s)
| | - Chad E Groer
- HylaPharm, LLC, Lawrence, Kansas 66047, United States.,Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Huan Gong
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Vivian Yudistyra
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| | - Marcus Laird Forrest
- HylaPharm, LLC, Lawrence, Kansas 66047, United States.,Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States
| | - Cory J Berkland
- Bioengineering Program, The University of Kansas, Lawrence, Kansas 66045, United States.,Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, Kansas 66047, United States.,Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas 66045, United States
| |
Collapse
|
3
|
Cagigi A, Douradinha B. Have mRNA vaccines sentenced DNA vaccines to death? Expert Rev Vaccines 2023; 22:1154-1167. [PMID: 37941101 DOI: 10.1080/14760584.2023.2282065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/07/2023] [Indexed: 11/10/2023]
Abstract
INTRODUCTION After receiving emergency approval during the COVID-19 pandemic, mRNA vaccines have taken center stage in the quest to enhance future vaccination strategies for both infectious diseases and cancer. Indeed, they have significantly overshadowed another facet of genetic vaccination, specifically DNA vaccines. Nevertheless, it is important to acknowledge that both types of genetic vaccines have distinct advantages and disadvantages that set them apart from each other. AREAS COVERED In this work, we delve extensively into the history of genetic vaccines, their mechanisms of action, their strengths, and limitations, and ultimately highlight ongoing research in key areas for potential enhancement of both DNA and mRNA vaccines. EXPERT OPINION Here, we assess the significance of the primary benefits and drawbacks associated with DNA and mRNA vaccination. We challenge the current lines of thought by highlighting that the existing drawbacks of DNA vaccination could potentially be more straightforward to address compared to those linked with mRNA vaccination. In our view, this suggests that DNA vaccines should remain viable contenders in the pursuit of the future of vaccination.
Collapse
Affiliation(s)
- Alberto Cagigi
- Nykode Therapeutics ASA, Oslo Science Park, Oslo, Norway
| | | |
Collapse
|
4
|
Yan WL, Wu CC, Shen KY, Liu SJ. Activation of GM-CSF and TLR2 signaling synergistically enhances antigen-specific antitumor immunity and modulates the tumor microenvironment. J Immunother Cancer 2021; 9:jitc-2021-002758. [PMID: 34599024 PMCID: PMC8488721 DOI: 10.1136/jitc-2021-002758] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 01/21/2023] Open
Abstract
Background The major challenge of antitumor immunotherapy is dealing with the immunosuppressive tumor microenvironment, which involves immature myeloid cell accumulation that results in T cell dysfunction. Myeloid cell activation is induced by Toll-like receptor agonists. Additionally, granulocyte/macrophage colony stimulating factor (GM-CSF) promotes myelopoiesis and recruits myeloid cells. Here, we combined the Toll-like receptor 2 (TLR2) agonist lipoprotein and GM-CSF to assess whether this bifunctional immunotherapy has synergistic effects on myeloid cells and could be further developed as a therapeutic intervention that enhances the antitumor response. Methods We investigated the synergistic effects of biadjuvanted tumor antigen on antigen-presenting cell (APC) activation in bone marrow-derived dendritic cells. Furthermore, therapeutic efficacy was monitored in different tumor models treated via intratumoral or subcutaneous administration routes. The immune effects of the bifunctional fusion protein on myeloid cells in the tumor mass and draining lymph nodes were analyzed by flow cytometry. The induction of cytotoxic T lymphocytes was evaluated via intracellular cytokine levels, perforin/granzyme B staining and an in vivo killing assay. Results The TLR2 agonist lipoprotein combined with GM-CSF synergistically induced DC maturation, which subsequently enhanced antitumor immunity. In addition, rlipoE7m-MoGM modulated tumor-infiltrating myeloid cell populations. Vaccination with rlipoE7m-MoGM therapy increased the number of CCR7+CD103+ cDC1s, whereas the number of suppressive tumor-associated macrophages was reduced in the tumor lesions. Consistent with this observation, proliferating antigen-specific CD8+ T cells are highly infiltrated within the tumor, and the expression of IFN-r and perforin was most pronounced within antigen-specific CD8+ T cells in mice administered rlipoE7m-MoGM therapy. This finding corresponded with observation that the combination of a TLR2 agonist and GM-CSF provides increased antitumor activity by inhibiting established tumor outgrowth and protecting against metastatic cancer compared with a TLR2 agonist alone. Importantly, tumor growth inhibition was not due to the direct effects of the TLR2 agonist or GM-CSF but was instead due to the induction of antigen-specific immunity. Conclusions The combination of a TLR2 agonist and GM-CSF has synergistic effects that inhibit tumor growth and modulate tumor-infiltrating APCs. This therapeutic approach could be applied to other tumor antigens to treat different cancers.
Collapse
Affiliation(s)
- Wan-Lun Yan
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Chiao-Chieh Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Kuan-Yin Shen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan.,School of Dentistry, Tri-Service General Hospital and National Defense Medical Center, Taipei, Taiwan
| | - Shih-Jen Liu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan .,National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.,Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Chen YP, Lin CC, Xie YX, Chen CY, Qiu JT. Enhancing immunogenicity of HPV16 E 7 DNA vaccine by conjugating codon-optimized GM-CSF to HPV16 E 7 DNA. Taiwan J Obstet Gynecol 2021; 60:700-705. [PMID: 34247810 DOI: 10.1016/j.tjog.2021.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2020] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE To generate immunity against human papillomavirus (HPV), the use of a recombinant DNA vaccine to carry an appropriate target gene is a promising and cost-effective approach. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a potent immunomodulatory cytokine that enhances the efficacy of vaccines by promoting the development and prolongation of humoral and cellular immunity. In this study, we linked codon-optimized GM-CSF (cGM-CSF) to the HPV16 E7 sequence as fused protein and evaluated the immunogenic potential of this DNA vaccine. MATERIALS AND METHODS We have demonstrated that cGM-CSF enhanced immunity against tumor challenges by generating and promoting the proliferation of HPV16 E7-specific CD8+ T cells, which secrete IFN-γ in the murine model. In this study, we aimed to evaluate the immunogenic potential of DNA vaccine that constructed by linking codon-optimized GM-CSF to HPV16 E7 sequence in the animal model. We study the half-life of RNA decay and cellular location of HPV16 E7 by Q-PCR and Western blot. We also assess immune response in the animal model by flow cytometry and ELISA. RESULTS The cGM-CSF-E7 sequence increased and extended the expression of E7 mRNA, in comparison with the E7 sequence alone. Mice vaccinated with the cGM-CSF-E7 DNA vaccine exhibited a slower rate of tumor growth than those vaccinated with the unconjugated E7 DNA vaccine. We also found that the CD4 and CD8+ T cells from these mice showed strong secretion of IFN-γ. CONCLUSION Through in vivo antibody depletion experiments, we demonstrated that both CD4+ and CD8+ T cells play an important role in the suppression of tumor growth.
Collapse
Affiliation(s)
- Yi-Pin Chen
- Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan, ROC; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chu-Chi Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC; Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan, ROC
| | - Yu-Xin Xie
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - Chia-Yuan Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan, ROC
| | - J Timothy Qiu
- Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei, Taiwan, ROC; College of Medicine, Taipei Medical University, Taipei, Taiwan, ROC.
| |
Collapse
|
6
|
Curci C, Picerno A, Chaoul N, Stasi A, De Palma G, Franzin R, Pontrelli P, Castellano G, Pertosa GB, Macchia L, Di Lorenzo VF, Sabbà C, Gallone A, Gesualdo L, Sallustio F. Adult Renal Stem/Progenitor Cells Can Modulate T Regulatory Cells and Double Negative T Cells. Int J Mol Sci 2020; 22:274. [PMID: 33383950 PMCID: PMC7795073 DOI: 10.3390/ijms22010274] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/22/2020] [Accepted: 12/25/2020] [Indexed: 01/22/2023] Open
Abstract
Adult Renal Stem/Progenitor Cells (ARPCs) have been recently identified in the human kidney and several studies show their active role in kidney repair processes during acute or chronic injury. However, little is known about their immunomodulatory properties and their capacity to regulate specific T cell subpopulations. We co-cultured ARPCs activated by triggering Toll-Like Receptor 2 (TLR2) with human peripheral blood mononuclear cells for 5 days and 15 days and studied their immunomodulatory capacity on T cell subpopulations. We found that activated-ARPCs were able to decrease T cell proliferation but did not affect CD8+ and CD4+ T cells. Instead, Tregs and CD3+ CD4- CD8- double-negative (DN) T cells decreased after 5 days and increased after 15 days of co-culture. In addition, we found that PAI1, MCP1, GM-CSF, and CXCL1 were significantly expressed by TLR2-activated ARPCs alone and were up-regulated in T cells co-cultured with activated ARPCs. The exogenous cocktail of cytokines was able to reproduce the immunomodulatory effects of the co-culture with activated ARPCs. These data showed that ARPCs can regulate immune response by inducing Tregs and DN T cells cell modulation, which are involved in the balance between immune tolerance and autoimmunity.
Collapse
Affiliation(s)
- Claudia Curci
- Nephrology, Dialysis and Transplantation Unit, DETO, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (A.P.); (A.S.); (R.F.); (P.P.); (G.B.P.); (L.G.)
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Angela Picerno
- Nephrology, Dialysis and Transplantation Unit, DETO, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (A.P.); (A.S.); (R.F.); (P.P.); (G.B.P.); (L.G.)
| | - Nada Chaoul
- Allergology Unit, DETO, University of Bari “Aldo Moro”, 70124 Bari, Italy; (N.C.); (L.M.)
| | - Alessandra Stasi
- Nephrology, Dialysis and Transplantation Unit, DETO, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (A.P.); (A.S.); (R.F.); (P.P.); (G.B.P.); (L.G.)
| | - Giuseppe De Palma
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Rossana Franzin
- Nephrology, Dialysis and Transplantation Unit, DETO, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (A.P.); (A.S.); (R.F.); (P.P.); (G.B.P.); (L.G.)
| | - Paola Pontrelli
- Nephrology, Dialysis and Transplantation Unit, DETO, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (A.P.); (A.S.); (R.F.); (P.P.); (G.B.P.); (L.G.)
| | - Giuseppe Castellano
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Science, University of Foggia, 71122 Foggia, Italy;
| | - Giovanni B. Pertosa
- Nephrology, Dialysis and Transplantation Unit, DETO, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (A.P.); (A.S.); (R.F.); (P.P.); (G.B.P.); (L.G.)
| | - Luigi Macchia
- Allergology Unit, DETO, University of Bari “Aldo Moro”, 70124 Bari, Italy; (N.C.); (L.M.)
| | | | - Carlo Sabbà
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Anna Gallone
- Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Loreto Gesualdo
- Nephrology, Dialysis and Transplantation Unit, DETO, University of Bari “Aldo Moro”, 70124 Bari, Italy; (C.C.); (A.P.); (A.S.); (R.F.); (P.P.); (G.B.P.); (L.G.)
| | - Fabio Sallustio
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| |
Collapse
|
7
|
Abdolalipour E, Mahooti M, Gorji A, Ghaemi A. Synergistic Therapeutic Effects of Probiotic Lactobacillus casei TD-2 Consumption on GM-CSF-Induced Immune Responses in a Murine Model of Cervical Cancer. Nutr Cancer 2020; 74:372-382. [PMID: 33356596 DOI: 10.1080/01635581.2020.1865419] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We perceive the potential of combined immunotherapy for the synergistic treatment of human papillomavirus (HPV)-associated tumors. So, the tumor inhibiting effects of combination of L. casei TD2a and GM-CSF on the TC-1 growth were evaluated In Vivo using lymphocyte proliferation, lymphocyte cytotoxicity, splenocyte, and tumor cytokine assays. The results showed that tumor inhibition in transplanted mice in the GM-CSF combined with probiotic L. casei group was significantly higher than that observed in the other groups excluding GM-CSF group whose tumor inhibition effect was considerable. The findings also indicated that the combined group could generate tumor-specific cytolytic and splenocyte proliferative responses. The levels of IFN-γ, IL-4, and IL-12 after treating with GM-CSF combined with probiotic L. casei were significantly higher than those of other groups. The intratumoral Tumor Necrosis Factor Related Apoptosis-Inducing Ligand (TRAIL) was also significantly increased in the combined group. Tumor analysis further showed that the combined group decreased the accumulation of IL-10 in the tumor microenvironment of treated mice. Furthermore, tumor volume analysis demonstrated that combination group and even GM-CSF suppress tumor growth. Our findings showed that the combination of GM-CSF and probiotic results in improved tumor suppression against HPV-associated tumors and stimulates enhancement of specific antitumor immune responses.
Collapse
Affiliation(s)
| | - Mehran Mahooti
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Ali Gorji
- Department of Neurosurgery and Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Amir Ghaemi
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
8
|
Russo MA, Georgius P, Pires AS, Heng B, Allwright M, Guennewig B, Santarelli DM, Bailey D, Fiore NT, Tan VX, Latini A, Guillemin GJ, Austin PJ. Novel immune biomarkers in complex regional pain syndrome. J Neuroimmunol 2020; 347:577330. [PMID: 32731051 DOI: 10.1016/j.jneuroim.2020.577330] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023]
Abstract
We investigated serum levels of 29 cytokines and immune-activated kynurenine and tetrahydrobiopterin pathway metabolites in 15 complex regional pain syndrome (CRPS) subjects and 14 healthy controls. Significant reductions in interleukin-37 and tryptophan were found in CRPS subjects, along with positive correlations between kynurenine/tryptophan ratio and TNF-α levels with kinesiophobia, tetrahydrobiopterin levels with McGill pain score, sRAGE, and xanthurenic acid and neopterin levels with depression, anxiety and stress scores. Using machine learning, we identified a set of binary variables, including IL-37 and GM-CSF, capable of distinguishing controls from established CRPS subjects. These results suggest possible involvement of various inflammatory markers in CRPS pathogenesis.
Collapse
Affiliation(s)
- Marc A Russo
- Hunter Pain Specialists, 91 Chatham Street, Broadmeadow, NSW, 2292, Australia; Genesis Research Services, 220 Denison St, Broadmeadow, NSW, 2292, Australia
| | - Peter Georgius
- Pain Rehab, Suite 4 Noosa Central, 6 Bottlebrush Avenue, Sunshine Coast, QLD, 4567, Australia
| | - Ananda Staats Pires
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia; Laboratório de Bioenergética e Estresse Oxidativo, LABOX; Departamento de Bioquímica, CCB; Universidade Federal de Santa Catarina; Florianópolis / SC, Brazil
| | - Benjamin Heng
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia
| | - Michael Allwright
- ForeFront, Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | - Boris Guennewig
- ForeFront, Brain & Mind Centre, The University of Sydney, NSW, 2006, Australia
| | | | - Dominic Bailey
- Genesis Research Services, 220 Denison St, Broadmeadow, NSW, 2292, Australia
| | - Nathan T Fiore
- Discipline of Anatomy & Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia
| | - Vanessa X Tan
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia
| | - Alexandra Latini
- Laboratório de Bioenergética e Estresse Oxidativo, LABOX; Departamento de Bioquímica, CCB; Universidade Federal de Santa Catarina; Florianópolis / SC, Brazil
| | - Gilles J Guillemin
- Neuroinflammation Group; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences; Macquarie University, Sydney, NSW, 2109, Australia
| | - Paul J Austin
- Discipline of Anatomy & Histology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, NSW, 2006, Australia.
| |
Collapse
|
9
|
Manara C, Brazzoli M, Piccioli D, Taccone M, D'Oro U, Maione D, Frigimelica E. Co-administration of GM-CSF expressing RNA is a powerful tool to enhance potency of SAM-based vaccines. Vaccine 2019; 37:4204-4213. [PMID: 31227353 DOI: 10.1016/j.vaccine.2019.04.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/21/2022]
Abstract
Self-amplifying mRNAs (SAM)-based vaccines have been shown to induce a robust immune response in various animal species against both viral and bacterial pathogens. Due to their synthetic nature and to the versatility of the manufacturing process, SAM technology may represent an attractive solution for rapidly producing novel vaccines, which is particularly critical in case of pandemic infections or diseases mediated by newly emerging pathogens. Recent published data support the hypothesis that Antigen Presenting Cells (APCs) are responsible for CD8+ T-cell priming after SAM vaccination, suggesting cross-priming as the key mechanism for antigen presentation by SAM vaccines. In our study we investigated the possibility to enhance the immune response induced in mice by a single immunization with SAM by increasing the recruitment of APCs at the site of injection. To enhance SAM immunogenicity, we selected murine granulocyte-macrophage colony-stimulating factor (GM-CSF) as a model chemoattractant for APCs, and developed a SAM-GM-CSF vector. We evaluated whether the use of SAM-GM-CSF in combination with a SAM construct encoding the Influenza A virus nucleoprotein (NP) would lead to an increase of APC recruitment and NP-specific immune response. We indeed observed that the administration of SAM-GM-CSF enhances the recruitment of APCs at the injection site. Consistently with our hypothesis, co-administration of SAM-GM-CSF with SAM-NP significantly improved the magnitude of NP-specific CD8+ T-cell response both in terms of frequency of cytotoxic antigen-specific CD8+ T-cells and their functional activity in vivo. Furthermore, co-immunization with SAM-GM-CSF and SAM-NP provided an increase in protection against a lethal challenge with influenza virus. In conclusion, we demonstrated that increased recruitment of APCs at the site of injection is associated with an enhanced effectiveness of SAM vaccination and might be a powerful tool to potentiate the efficacy of RNA vaccination.
Collapse
Affiliation(s)
| | | | - Diego Piccioli
- GSK Vaccines S.r.l., Via Fiorentina 1, 53100 Siena, Italy
| | | | - Ugo D'Oro
- GSK Vaccines S.r.l., Via Fiorentina 1, 53100 Siena, Italy
| | | | | |
Collapse
|
10
|
Bashaw AA, Teoh SM, Tuong ZK, Leggatt GR, Frazer IH, Chandra J. HPV16 E7-Driven Epithelial Hyperplasia Promotes Impaired Antigen Presentation and Regulatory T-Cell Development. J Invest Dermatol 2019; 139:2467-2476.e3. [PMID: 31207230 DOI: 10.1016/j.jid.2019.03.1162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/15/2019] [Accepted: 03/31/2019] [Indexed: 01/21/2023]
Abstract
Human papillomaviruses infect keratinocytes and can lead to hyperproliferative dysplasia and malignant transformation if not cleared by the immune system. Human papillomavirus has evolved an array of mechanisms to evade and manipulate the immune system to improve replication efficiency and promote persistent infection. We here demonstrate that hyperproliferative skin expressing the high-risk human papillomavirus 16 E7 oncogene as a transgene drives immunomodulation of dendritic cells (DCs), resulting in reduced capacity to take up antigen and prime effector CD4+ T cell responses. The phenotype of DCs in the E7-expressing hyperproliferative skin was not reversible by activation through intradermal immunization. Naïve CD4+ T cells primed by E7-driven hyperproliferative skin acquired FoxP3 expression and an anergic phenotype. DC and T help modulation was dependent on E7-retinoblastoma protein interaction-driven epithelial hyperproliferation, rather than on expression of E7. Inhibition of binding of E7 to retinoblastoma protein, and of consequent epithelial hyperplasia, was associated with normal skin DC phenotype, and T helper type 1 effector responses to immunization were restored. We conclude that human papillomavirus-induced epithelial hyperplasia modulates epithelial DCs and inhibits T helper type 1 immunity while polarizing T-cell differentiation to a regulatory or anergic phenotype.
Collapse
Affiliation(s)
- Abate Assefa Bashaw
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Siok M Teoh
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Zewen K Tuong
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia.
| | - Janin Chandra
- The University of Queensland Diamantina Institute, Translational Research Institute, Woolloongabba, Queensland, Australia
| |
Collapse
|
11
|
Saxena M, Bhardwaj N. Re-Emergence of Dendritic Cell Vaccines for Cancer Treatment. Trends Cancer 2018; 4:119-137. [PMID: 29458962 DOI: 10.1016/j.trecan.2017.12.007] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/19/2017] [Accepted: 12/21/2017] [Indexed: 02/07/2023]
Abstract
Dendritic cells (DCs) are essential in immunity owing to their role in activating T cells, thereby promoting antitumor responses. Tumor cells, however, hijack the immune system, causing T cell exhaustion and DC dysfunction. Tumor-induced T cell exhaustion may be reversed through immune checkpoint blockade (ICB); however, this treatment fails to show clinical benefit in many patients. While ICB serves to reverse T cell exhaustion, DCs are still necessary to prime, activate, and direct the T cells to target tumor cells. In this review we provide a brief overview of DC function, describe mechanisms by which DC functions are disrupted by the tumor microenvironment, and highlight recent developments in DC cancer vaccines.
Collapse
Affiliation(s)
- Mansi Saxena
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Nina Bhardwaj
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA; Parker Institute of Cancer Immunotherapy, San Francisco, CA 94129, USA.
| |
Collapse
|
12
|
Megele R, Riemenschneider MJ, Dodoo-Schittko F, Feyrer M, Kleindienst A. Intra-tumoral treatment with oxygen-ozone in glioblastoma: A systematic literature search and results of a case series. Oncol Lett 2018; 16:5813-5822. [PMID: 30344733 PMCID: PMC6176341 DOI: 10.3892/ol.2018.9397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 08/30/2018] [Indexed: 12/20/2022] Open
Abstract
Despite progress in surgery and radiochemotherapy, the prognosis of glioblastoma (GB) remains poor. GB cells exhibit a preference for hypoxia to maintain their tumor-forming capacity. Treatment strategies utilizing oxygen (O2) or ozone (O3) and generating reactive oxygen species induce cell growth inhibition and apoptosis. The anti-tumorigenic properties of O2-O3 are accompanied by a key role in regulating immunogenicity. The present study reported a case series of an intra-tumoral O2-O3 application in recurrent GB. Following surgery in combination with standard radiochemotherapy, O2-O3 (5 ml at 40 µg/ml) was applied every four weeks into the tumor vicinity. The patients received a median of 27 (range, 3–44) O2-O3 applications. In addition, a systematic literature search was performed in order to evaluate the role of O3 in the treatment of malignancies. The median overall survival rate was 40 (range, 16–53) months. The median survival rate following the first recurrence or the initiation of the O2-O3 treatment, respectively, was 34 (range, 12–53) months. In one patient, a local infection and in another, hemorrhage occurred, necessitating in both the temporary removal of the reservoir. The data from the present study support the potential benefit of an intra-tumoral O2-O3 application in recurrent GB. The scientific literature revealed by the bibliographic search suggests that O3 may be considered a viable adjuvant therapy in oncological patients. The present study may serve as a starting point for further observational and clinical studies elucidating the cellular and systemic effects of O2 and/or O3 and demonstrating their efficacy and safety in larger patient samples.
Collapse
Affiliation(s)
- Richard Megele
- Department of Neurosurgery, Klinikum St. Marien, D-92224 Amberg, Germany
| | | | - Frank Dodoo-Schittko
- Medical Sociology, Institute of Epidemiology and Preventative Medicine, University of Regensburg, D-93053 Regensburg, Germany
| | - Matthias Feyrer
- Department of Radiology, Klinikum St. Marien, D-92224 Amberg, Germany
| | - Andrea Kleindienst
- Department of Neurosurgery, Klinikum St. Marien, D-92224 Amberg, Germany.,Department of Neurosurgery, University of Erlangen-Nürnberg, D-91054 Erlangen, Germany
| |
Collapse
|
13
|
Barros MR, de Melo CML, Barros MLCMGR, de Cássia Pereira de Lima R, de Freitas AC, Venuti A. Activities of stromal and immune cells in HPV-related cancers. J Exp Clin Cancer Res 2018; 37:137. [PMID: 29976244 PMCID: PMC6034319 DOI: 10.1186/s13046-018-0802-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/19/2018] [Indexed: 02/07/2023] Open
Abstract
The immune system is composed of immune as well as non-immune cells. As this system is a well-established component of human papillomavirus- (HPV)-related carcinogenesis, high risk human papillomavirus (hrHPV) prevents its routes and mechanisms in order to cause the persistence of infection. Among these mechanisms are those originated from stromal cells, which include the cancer-associated fibroblasts (CAFs), the myeloid-derived suppressor cells (MDSCs) and the host infected cells themselves, i.e. the keratinocytes. These types of cells play central role since they modulate immune cells activities to create a prosperous milieu for cancer development, and the knowledge how such interactions occur are essential for prognostic assessment and development of preventive and therapeutic approaches. Nevertheless, the precise mechanisms are not completely understood, and this lack of knowledge precluded the development of entirely efficient immunotherapeutic strategies for HPV-associated tumors. As a result, an intense work for attaining how host immune response works, and developing of effective therapies has been applied in the last decade. Based on this, this review aims to discuss the major mechanisms of immune and non-immune cells modulated by hrHPV and the potential and existing immunotherapies involving such mechanisms in HPV-related cancers. It is noticed that the combination of immunotherapies has been demonstrated to be essential for obtaining better results, especially because the possibility of increasing the modulating capacity of the HPV-tumor microenvironment has been shown to be central in strengthening the host immune system.
Collapse
Affiliation(s)
- Marconi Rego Barros
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Moraes Rego, 1235, Recife, PE CEP-50670-901 Brazil
| | - Cristiane Moutinho Lagos de Melo
- Laboratory of Immunological and Antitumor Analysis (LAIA), Department of Antibiotics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Artur de Sá, s/n, Recife, PE CEP-50740-525 Brazil
| | | | - Rita de Cássia Pereira de Lima
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Moraes Rego, 1235, Recife, PE CEP-50670-901 Brazil
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy (LEMTE), Department of Genetics, Center of Biological Sciences, Federal University of Pernambuco, Cidade Universitária, Av. Prof Moraes Rego, 1235, Recife, PE CEP-50670-901 Brazil
| | - Aldo Venuti
- HPV-Unit, Tumor Immunology and Immunotherapy Unit, Department of Research, Advanced Diagnostic and Technological Innovation, IRCCS Regina Elena National Cancer Institute, Via Elio Chianesi 53, 00144 Rome, Italy
| |
Collapse
|
14
|
Grasse M, Meryk A, Miggitsch C, Grubeck-Loebenstein B. GM-CSF improves the immune response to the diphtheria-component in a multivalent vaccine. Vaccine 2018; 36:4672-4680. [PMID: 29961602 PMCID: PMC7116485 DOI: 10.1016/j.vaccine.2018.06.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/07/2018] [Accepted: 06/14/2018] [Indexed: 12/24/2022]
Abstract
Multivalent tetanus and diphtheria toxoid containing vaccines belong to the most frequently applied vaccines. However, there is an imbalance in the degree of protection against the two antigens with insufficient long-term protection against diphtheria, particularly in the elderly population. We have previously reported a positive correlation between granulocyte macrophage-colony stimulating factor (GM-CSF) and the production of diphtheria-specific antibodies. Therefore, in the present study we analyzed the effects of in vivo applied recombinant GM-CSF on immunization with multivalent tetanus/diphtheria vaccine in mice of different age. In vivo application of GM-CSF lead to enhanced production of diphtheria-specific antibodies as well as more diphtheria-specific CD4+ T cells following vaccination with multivalent tetanus/diphtheria vaccine. In contrast, the humoral and cellular immune response to the tetanus component was unaltered. Furthermore, application of GM-CSF resulted in more splenic CD11b+ dendritic cells (DCs) with a higher MHC-II expression. GM-CSF also induced a stronger recruitment of CD11b+ DCs to the injected muscle. Most remarkably, GM-CSF was able to boost the diphtheria-specific immune response to the multivalent vaccine in aged mice. This study demonstrates that local administration of GM-CSF is able to improve immune responsiveness to the diphtheria component of multivalent tetanus/diphtheria vaccine in young and old mice. This information could be useful for the future design of vaccines for the elderly.
Collapse
Affiliation(s)
- Marco Grasse
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
| | - Andreas Meryk
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
| | - Carina Miggitsch
- Institute for Biomedical Aging Research, Universität Innsbruck, Innsbruck, Austria
| | | |
Collapse
|
15
|
Gogesch P, Schülke S, Scheurer S, Mühlebach MD, Waibler Z. Modular MLV-VLPs co-displaying ovalbumin peptides and GM-CSF effectively induce expansion of CD11b + APC and antigen-specific T cell responses in vitro. Mol Immunol 2018; 101:19-28. [PMID: 29852456 DOI: 10.1016/j.molimm.2018.05.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 04/27/2018] [Accepted: 05/22/2018] [Indexed: 12/30/2022]
Abstract
The development of novel vaccination strategies is a persistent challenge to provide effective prophylactic treatments to encounter viral infections. In general, the physical conjugation of selected vaccine components, e.g. antigen and adjuvant, has been shown to enhance the immunogenicity and hence, can increase effectiveness of the vaccine. In our proof-of-concept study, we generated non-infectious, replication deficient Murine Leukemia Virus (MLV)-derived virus-like particles (VLPs) that physically link antigen and adjuvant in a modular fashion by co-displaying them on their surface. For this purpose, we selected the immunodominant peptides of the model antigen ovalbumin (OVA) and the cytokine granulocyte macrophage-colony stimulating factor (GM-CSF) as non-classical adjuvant. Our results show that murine GM-CSF displayed on MLV-VLPs mediates expansion and proliferation of CD11b+ cells within murine bone marrow and total spleen cells. Moreover, we show increased immunogenicity of modular VLPs co-displaying OVA peptides and GM-CSF by their elevated capacity to induce OVA-specific T cell-activation and -proliferation within OT-I and OT-II splenocyte cultures. These enhanced effects were not achieved by using an equimolar mixture of VLPs displaying either OVA or GM-CSF. Taken together, OVA and GM-CSF co-displaying MLV-VLPs are able to target and expand antigen presenting cells which in turn results in enhanced antigen-specific T cell activation and proliferation in vitro. These data suggest MLV-VLPs to be an attractive platform to flexibly combine antigen and adjuvant for novel modular vaccination approaches.
Collapse
Affiliation(s)
- Patricia Gogesch
- Section "Product Testing of Immunological Biomedicines", Paul-Ehrlich-Institut, D-63225, Langen, Germany
| | - Stefan Schülke
- Section Molecular Allergology, Paul-Ehrlich-Institut, D-63225, Langen, Germany
| | - Stephan Scheurer
- Section Molecular Allergology, Paul-Ehrlich-Institut, D-63225, Langen, Germany
| | - Michael D Mühlebach
- Section Product Testing of IVMP, Paul-Ehrlich-Institut, Langen, Paul-Ehrlich-Institut, D-63225, Langen, Germany.
| | - Zoe Waibler
- Section "Product Testing of Immunological Biomedicines", Paul-Ehrlich-Institut, D-63225, Langen, Germany.
| |
Collapse
|
16
|
Cheng MA, Farmer E, Huang C, Lin J, Hung CF, Wu TC. Therapeutic DNA Vaccines for Human Papillomavirus and Associated Diseases. Hum Gene Ther 2018; 29:971-996. [PMID: 29316817 DOI: 10.1089/hum.2017.197] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human papillomavirus (HPV) has long been recognized as the causative agent of cervical cancer. High-risk HPV types 16 and 18 alone are responsible for over 70% of all cases of cervical cancers. More recently, HPV has been identified as an etiological factor for several other forms of cancers, including oropharyngeal, anogenital, and skin. Thus, the association of HPV with these malignancies creates an opportunity to control these HPV lesions and HPV-associated malignancies through immunization. Strategies to prevent or to therapeutically treat HPV infections have been developed and are still pushing innovative boundaries. Currently, commercial prophylactic HPV vaccines are widely available, but they are not able to control established infections or lesions. As a result, there is an urgent need for the development of therapeutic HPV vaccines, to treat existing infections, and to prevent the development of HPV-associated cancers. In particular, DNA vaccination has emerged as a promising form of therapeutic HPV vaccine. DNA vaccines have great potential for the treatment of HPV infections and HPV-associated cancers due to their safety, stability, simplicity of manufacturability, and ability to induce antigen-specific immunity. This review focuses on the current state of therapeutic HPV DNA vaccines, including results from recent and ongoing clinical trials, and outlines different strategies that have been employed to improve their potencies. The continued progress and improvements made in therapeutic HPV DNA vaccine development holds great potential for innovative ways to effectively treat HPV infections and HPV-associated diseases.
Collapse
Affiliation(s)
- Max A Cheng
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - Emily Farmer
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - Claire Huang
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - John Lin
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - Chien-Fu Hung
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,2 Department of Oncology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| | - T-C Wu
- 1 Department of Pathology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,2 Department of Oncology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,3 Department of Obstetrics and Gynecology, Johns Hopkins Medical Institutions , Baltimore, Maryland.,4 Department of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions , Baltimore, Maryland
| |
Collapse
|
17
|
Mechanistic insights into the efficacy of cell penetrating peptide-based cancer vaccines. Cell Mol Life Sci 2018; 75:2887-2896. [PMID: 29508006 PMCID: PMC6061156 DOI: 10.1007/s00018-018-2785-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 11/15/2017] [Accepted: 02/22/2018] [Indexed: 02/07/2023]
Abstract
Immunotherapies are increasingly used to treat cancer, with some outstanding results. Immunotherapy modalities include therapeutic vaccination to eliminate cancer cells through the activation of patient’s immune system against tumor-derived antigens. Nevertheless, the full potential of therapeutic vaccination has yet to be demonstrated clinically because many early generation vaccines elicited low-level immune responses targeting only few tumor antigens. Cell penetrating peptides (CPPs) are highly promising tools to advance the field towards clinical success. CPPs efficiently penetrate cell membranes, even when linked to antigenic cargos, which can induce both CD8 and CD4 T-cell responses. Pre-clinical studies demonstrated that targeting multiple tumor antigens, even those considered to be poorly immunogenic, led to tumor regression. Therefore, CPP-based cancer vaccines represent a flexible and powerful means to extend therapeutic vaccination to many cancer indications. Here, we review recent findings in CPP development and discuss their use in next generation immunotherapies.
Collapse
|
18
|
Bashaw AA, Leggatt GR, Chandra J, Tuong ZK, Frazer IH. Modulation of antigen presenting cell functions during chronic HPV infection. PAPILLOMAVIRUS RESEARCH (AMSTERDAM, NETHERLANDS) 2017; 4:58-65. [PMID: 29179871 PMCID: PMC5883240 DOI: 10.1016/j.pvr.2017.08.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/14/2017] [Accepted: 08/15/2017] [Indexed: 12/12/2022]
Abstract
High-risk human papillomaviruses (HR-HPV) infect basal keratinocytes, where in some individuals they evade host immune responses and persist. Persistent HR-HPV infection of the cervix causes precancerous neoplasia that can eventuate in cervical cancer. Dendritic cells (DCs) are efficient in priming/cross-priming antigen-specific T cells and generating antiviral and antitumor cytotoxic CD8+ T cells. However, HR-HPV have adopted various immunosuppressive strategies, with modulation of DC function crucial to escape from the host adaptive immune response. HPV E6 and E7 oncoproteins alter recruitment and localization of epidermal DCs, while soluble regulatory factors derived from HPV-induced hyperplastic epithelium change DC development and influence initiation of specific cellular immune responses. This review focuses on current evidence for HR-HPV manipulation of antigen presentation in dendritic cells and escape from host immunity.
Collapse
Affiliation(s)
- Abate Assefa Bashaw
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Graham R Leggatt
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Janin Chandra
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Zewen K Tuong
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia
| | - Ian H Frazer
- The University of Queensland Diamantina Institute, Translational Research Institute, Princess Alexandra Hospital, 37 Kent Street, Woolloongabba, Queensland 4102, Australia.
| |
Collapse
|
19
|
Sun Y, Peng S, Yang A, Farmer E, Wu TC, Hung CF. Coinjection of IL2 DNA enhances E7-specific antitumor immunity elicited by intravaginal therapeutic HPV DNA vaccination with electroporation. Gene Ther 2017; 24:408-415. [PMID: 28492521 PMCID: PMC5510480 DOI: 10.1038/gt.2017.38] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 02/16/2017] [Accepted: 05/04/2017] [Indexed: 01/19/2023]
Abstract
The generation and use of therapeutic human papillomavirus (HPV) DNA vaccines represent an appealing treatment method against HPV-associated cervical cancer owing to their safety and durability. Previously, we created a therapeutic HPV DNA vaccine candidate by linking the HPV16-E7 DNA sequence to calreticulin (CRT/E7), which we showed could generate significant E7-specific cytotoxic T lymphocyte (CTL)-mediated antitumor immune responses against HPV16 oncogenes expressing murine tumor model TC-1. Here we assess the therapeutic efficacy of intravaginal immunization with pcDNA3-CRT/E7 followed by electroporation. In addition, we examined whether coadministration of DNA-encoding interleukin 2 (IL2) with the pcDNA3-CRT/E7 could improve the T-cell responses elicited by pcDNA3-CRT/E7. TC-1 tumor-bearing mice vaccinated intravaginally with both pcDNA3-CRT/E7 and IL2 DNA followed by electroporation induced stronger local antitumor CTL response in comparison to mice that received other treatment regimens. Additionally, we found that coadministration of IL2 DNA with pcDNA3-CRT/E7 modified the tumor microenvironment by decreasing the population of regulatory T cells and myeloid-derived suppressor cells relative to that of CTLs. Our data demonstrate the translational potential of local administration of IL2 and pcDNA3-CRT/E7 followed by electroporation in treating cervicovaginal tumors.
Collapse
Affiliation(s)
- Yunyan Sun
- Department of Obstetrics and Gynecology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Shiwen Peng
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Andrew Yang
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Emily Farmer
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - T.-C. Wu
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
- Departments of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
- Departments of Molecular Microbiology and Immunology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
- Departments of Obstetrics and Gynecology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| | - Chien-Fu Hung
- Departments of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
- Departments of Oncology, Johns Hopkins Medical Institutions, Baltimore, MD, United States
| |
Collapse
|
20
|
Mahdavi M, Tajik AH, Ebtekar M, Rahimi R, Adibzadeh MM, Moozarmpour HR, Beikverdi MS, Olfat S, Hassan ZM, Choopani M, Kameli M, Hartoonian C. Granulocyte-macrophage colony-stimulating factor, a potent adjuvant for polarization to Th-17 pattern: an experience on HIV-1 vaccine model. APMIS 2017; 125:596-603. [PMID: 28493367 DOI: 10.1111/apm.12660] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2016] [Accepted: 12/21/2016] [Indexed: 10/19/2022]
Abstract
Cytokines are mediators for polarization of immune response in vaccines. Studies show that co-immunization of DNA vaccines with granulocyte-macrophage colony-stimulating factor (GM-CSF) can increase immune responses. Here, experimental mice were immunized with HIV-1tat/pol/gag/env DNA vaccine with GM-CSF and boosted with recombinant vaccine. Lymphocyte proliferation with Brdu and CTL activity, IL-4, IFN-γ, IL-17 cytokines, total antibody, and IgG1 and IgG2a isotypes were assessed with ELISA. Results show that GM-CSF as adjuvant in DNA immunization significantly increased lymphocyte proliferation and IFN-γ cytokines, but CTL response was tiny increased. Also GM-CSF as adjuvant decreased IL-4 cytokine vs mere vaccine group. IL-17 in the group that immunized with mixture of DNA vaccine/GM-CSF was significantly increased vs DNA vaccine group. Result of total antibody shows that GM-CSF increased antibody response in which both IgG1 and IgG2a increased. Overall, results confirmed the beneficial effect of GM-CSF as adjuvant to increase vaccine immunogenicity. The hallmark result of this study was to increase IL-17 cytokine with DNA vaccine/GM-CSF immunized group. This study for the first time provides the evidence of the potency of GM-CSF in the induction of IL-17 in response to a vaccine, which is important for control of infection such as HIV-1.
Collapse
Affiliation(s)
- Mehdi Mahdavi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Amir Hossein Tajik
- Department of Clinical Biochemistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Roghieh Rahimi
- Department of Immunology, Tarbiat Modares University, Tehran, Iran
| | | | - Hamid Reza Moozarmpour
- Department of Microbiology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Sadegh Beikverdi
- Department of Biology, Faculty of Basic Science, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Soophie Olfat
- Department of Life Science Engineering, Faculty of new Science and Technologies, University of Tehran, Tehran, Iran
| | | | - Mohammad Choopani
- Department of Biology, College of Basic Science, Karaj Branch, Islamic Azad University, Alborz, Iran
| | - Morteza Kameli
- Department of Biology, College of Basic Science, Karaj Branch, Islamic Azad University, Alborz, Iran
| | - Christine Hartoonian
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Biotechnology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Zanotta N, Tornesello ML, Annunziata C, Stellato G, Buonaguro FM, Comar M. Candidate Soluble Immune Mediators in Young Women with High-Risk Human Papillomavirus Infection: High Expression of Chemokines Promoting Angiogenesis and Cell Proliferation. PLoS One 2016; 11:e0151851. [PMID: 26990868 PMCID: PMC4798492 DOI: 10.1371/journal.pone.0151851] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/04/2016] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND The causal interpretation of cervical immune response to Human Papillomavirus (HPV) infection is complex and poorly characterized mainly due to the delicate balance that exists between viral infection, increase of inflammatory cytokines and host risk factors. This study aims to explore the significance of cervical immune mediators associated to cell survival, angiogenesis and interaction with immune response, in predicting the risk to develop HPV-related intraepithelial lesions. METHODS A panel of 48 cytokines and growth factors were explored in a selected cohort of 168 immunocompetent women including 88 diagnosed with low (LSIL) or high (HSIL) squamous intraepithelial lesions of the cervix and 80 with normal cervical cytology (NIL). HPV genotyping was performed by Linear Array HPV test and the soluble concentration of 48 immune molecules was analyzed using the Bio-Plex platform. RESULTS The prevalence of single HR-HPV infection was 30% in NIL and 100% in LSIL and HSIL women. The expression of 13 cytokines, including interleukins IL-6, IL-3, IL-12p40, IL-12p70, IL-16, IL-18, LIF, of chemokines CCL7 (MCP-3), CXCL9 (MIG), CXCL12 (SDF-1α) and of the tropic factors VEGF, G-CSF, M-CSF were significantly associated with the presence of infection, with levels being higher in women with precancerous lesions compared to NIL HPV negative women. Only the growth factor GM-CSF was positively associated with the cytological abnormalities. CONCLUSIONS The ability of HR-HPV to escape from innate immune recognition and to orchestrate the production of specific inflammatory and growth factors, involved in early inflammatory response and in the cell-proliferating phase of intraepithelial damage, was documented in women before the development of cervical lesions.
Collapse
Affiliation(s)
- Nunzia Zanotta
- Institute for Maternal and Child Health – IRCCS “Burlo Garofolo”, Trieste, Italy
| | - Maria Lina Tornesello
- Molecular Biology and Viral Oncology Division, Istituto Nazionale Tumori IRCCS – “Fond. Pascale,” Napoli, Italy
| | - Clorinda Annunziata
- Molecular Biology and Viral Oncology Division, Istituto Nazionale Tumori IRCCS – “Fond. Pascale,” Napoli, Italy
| | - Giovanni Stellato
- Gynecology Oncology Division, Istituto Nazionale Tumori IRCCS – “Fond. Pascale,” Napoli, Italy
| | - Franco Maria Buonaguro
- Molecular Biology and Viral Oncology Division, Istituto Nazionale Tumori IRCCS – “Fond. Pascale,” Napoli, Italy
| | - Manola Comar
- Institute for Maternal and Child Health – IRCCS “Burlo Garofolo”, Trieste, Italy
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
22
|
Yang A, Jeang J, Cheng K, Cheng T, Yang B, Wu TC, Hung CF. Current state in the development of candidate therapeutic HPV vaccines. Expert Rev Vaccines 2016; 15:989-1007. [PMID: 26901118 DOI: 10.1586/14760584.2016.1157477] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The identification of human papillomavirus (HPV) as an etiological factor for HPV-associated malignancies creates the opportunity to control these cancers through vaccination. Currently, available preventive HPV vaccines have not yet demonstrated strong evidences for therapeutic effects against established HPV infections and lesions. Furthermore, HPV infections remain extremely common. Thus, there is urgent need for therapeutic vaccines to treat existing HPV infections and HPV-associated diseases. Therapeutic vaccines differ from preventive vaccines in that they are aimed at generating cell-mediated immunity rather than neutralizing antibodies. The HPV-encoded early proteins, especially oncoproteins E6 and E7, form ideal targets for therapeutic HPV vaccines since they are consistently expressed in HPV-associated malignancies and precancerous lesions, playing crucial roles in the generation and maintenance of HPV-associated disease. Our review will cover various therapeutic vaccines in development for the treatment of HPV-associated lesions and cancers. Furthermore, we review strategies to enhance vaccine efficacy and the latest clinical trials on therapeutic HPV vaccines.
Collapse
Affiliation(s)
- Andrew Yang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Jessica Jeang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Kevin Cheng
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Ting Cheng
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - Benjamin Yang
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA
| | - T-C Wu
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA.,b Department of Obstetrics and Gynecology , Johns Hopkins University , Baltimore , MD , USA.,c Department of Molecular Microbiology and Immunology , Johns Hopkins University , Baltimore , MD , USA.,d Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| | - Chien-Fu Hung
- a Department of Pathology , Johns Hopkins University , Baltimore , MD , USA.,d Department of Oncology , Johns Hopkins University , Baltimore , MD , USA
| |
Collapse
|