1
|
Wakisaka R, Yamaki H, Kono M, Inoue T, Sato R, Komatsuda H, Ohara K, Kosaka A, Ohkuri T, Nagato T, Kishibe K, Nakayama K, Kobayashi H, Kumai T, Takahara M. Hypoxia-Targeted Immunotherapy with PD-1 Blockade in Head and Neck Cancer. Cancers (Basel) 2024; 16:3013. [PMID: 39272872 PMCID: PMC11394489 DOI: 10.3390/cancers16173013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Intratumoral hypoxia is associated with tumor progression, aggressiveness, and therapeutic resistance in several cancers. Hypoxia causes cancer cells to experience replication stress, thereby activating DNA damage and repair pathways. MutT homologue-1 (MTH1, also known as NUDT1), a member of the Nudix family, maintains the genomic integrity and viability of tumor cells in the hypoxic tumor microenvironment. Although hypoxia is associated with poor prognosis and can cause therapeutic resistance by regulating the microenvironment, it has not been considered a treatable target in cancer. This study aimed to investigate whether hypoxia-induced MTH1 is a useful target for immunotherapy and whether hypoxic conditions influence the antitumor activity of immune cells. Our results showed that MTH1 expression was elevated under hypoxic conditions in head and neck cancer cell lines. Furthermore, we identified a novel MTH1-targeting epitope peptide that can activate peptide-specific CD4+ helper T cells with cytotoxic activity. The proliferation and cytotoxic activity of T cells were maintained under hypoxic conditions, and PD-1 blockade further augmented the cytotoxicity. These results indicate that MTH1-targeted immunotherapy combined with checkpoint blockade can be an effective strategy for the treatment of hypoxic tumors.
Collapse
Affiliation(s)
- Risa Wakisaka
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Hidekiyo Yamaki
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Michihisa Kono
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Takahiro Inoue
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Ryosuke Sato
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Hiroki Komatsuda
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Kenzo Ohara
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
- Department of Innovative Head & Neck Cancer Research and Treatment (IHNCRT), Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Akemi Kosaka
- Department of Pathology, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Takayuki Ohkuri
- Department of Pathology, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Toshihiro Nagato
- Department of Pathology, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Kan Kishibe
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Koh Nakayama
- Department of Pharmacology, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Hiroya Kobayashi
- Department of Pathology, Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Takumi Kumai
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
- Department of Innovative Head & Neck Cancer Research and Treatment (IHNCRT), Asahikawa Medical University, Asahikawa 0788510, Japan
| | - Miki Takahara
- Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa 0788510, Japan
| |
Collapse
|
2
|
Wang P, Wang XY, Man CF, Gong DD, Fan Y. Advances in hyperbaric oxygen to promote immunotherapy through modulation of the tumor microenvironment. Front Oncol 2023; 13:1200619. [PMID: 37790761 PMCID: PMC10543083 DOI: 10.3389/fonc.2023.1200619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 08/25/2023] [Indexed: 10/05/2023] Open
Abstract
Hyperbaric oxygen therapy is a relatively safe treatment method that has been used for a long time in the clinic. It has been proven that it can enhance the sensitivity of radiotherapy and photodynamic therapy for cancer. However, there are few studies on hyperbaric oxygen and immunotherapy. In this article, we summarize that hyperbaric oxygen therapy regulates the tumor microenvironment through various pathways such as improving tumor hypoxia, targeting hypoxia-inducing factors, and generating reactive oxygen species. The change in the tumor microenvironment ultimately affects the curative effect of immunotherapy. Therefore, hyperbaric oxygen can influence immunotherapy by regulating the tumor microenvironment, providing a direction for the future development of immunotherapy.
Collapse
Affiliation(s)
- Pei Wang
- Cancer Institute, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiao-Yan Wang
- Department of Gastroenterology, The Affiliated Suqian First People’s Hospital of Xuzhou Medical University, Suqian, Jiangsu, China
| | - Chang-Feng Man
- Cancer Institute, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Dan-Dan Gong
- Cancer Institute, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu Fan
- Cancer Institute, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
3
|
Schindler NR, Braun DA. Antigenic targets in clear cell renal cell carcinoma. KIDNEY CANCER 2023; 7:81-91. [PMID: 38014393 PMCID: PMC10475986 DOI: 10.3233/kca-230006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/11/2023] [Indexed: 11/29/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the management of advanced renal cell carcinoma (RCC), but most patients still do not receive a long-term benefit from these therapies, and many experience off-target, immune-related adverse effects. RCC is also different from many other ICI-responsive tumors, as it has only a modest mutation burden, and total neoantigen load does not correlate with ICI response. In order to improve the efficacy and safety of immunotherapies for RCC, it is therefore critical to identify the antigens that are targeted in effective anti-tumor immunity. In this review, we describe the potential classes of target antigens, and provide examples of previous and ongoing efforts to investigate and target antigens in RCC, with a focus on clear cell histology. Ultimately, we believe that a concerted antigen discovery effort in RCC will enable an improved understanding of response and resistance to current therapies, and lay a foundation for the future development of "precision" antigen-directed immunotherapies.
Collapse
Affiliation(s)
- Nicholas R. Schindler
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - David A. Braun
- Section of Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Center of Molecular and Cellular Oncology, Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
4
|
Qu B, Han X, Zhao L, Zhang F, Gao Q. Relationship of HIF‑1α expression with apoptosis and cell cycle in bone marrow mesenchymal stem cells from patients with myelodysplastic syndrome. Mol Med Rep 2022; 26:239. [PMID: 35642674 PMCID: PMC9185697 DOI: 10.3892/mmr.2022.12755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 02/21/2022] [Indexed: 11/09/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a group of abnormal clonal disorders with ineffective hematopoiesis, which are incurable with conventional therapy. Of note, MDS features an abnormal bone marrow microenvironment, which is related to its incidence. The hypoxia-inducible factor-1α (HIF-1α) transcriptional signature is generally activated in bone marrow stem/progenitor cells of patients with MDS. To analyze the expression of HIF-1α in bone marrow mesenchymal stem cells (BM-MSCs) and the apoptosis and cell cycle features associated with the disease, BM-MSCs were obtained from 40 patients with a definitive diagnosis of MDS and 20 subjects with hemocytopenia but a negative diagnosis of MDS as a control group. Reverse transcription-quantitative PCR and western blot analyses were used to measure HIF-1α expression in cells from the two groups and apoptosis and cell cycle were also analyzed and compared between the groups using flow cytometry assays. BM-MSCs from both the control group and the MDS group exhibited a fibroblast-like morphology, had similar growth cycles and were difficult to passage stably. It was observed that BM-MSCs from the MDS group had significantly higher HIF-1α expression levels than the control group (P<0.05). Furthermore, the BM-MSCs from the MDS group had a higher proportion of cells in early apoptosis (5.22±1.34 vs. 2.04±0.08%; P<0.0001) and late apoptosis (3.38±0.43 vs. 1.23±0.11%; P<0.01) and exhibited cell cycle arrest. This may be a noteworthy aspect of the pathogenesis of MDS and may be related to high HIF-1α expression under a hypoxic state in the bone marrow microenvironment. Furthermore, the expression of HIF-1α in bone marrow tissue sections from patients with MDS in the International Prognostic Scoring System (IPSS) lower-risk group was higher than that from patients with MDS in the IPSS high-risk group. These results revealed the role of HIF-1α as a central pathobiology mediator of MDS and an effective therapeutic target for a broad spectrum of patients with MDS, particularly for patients in the lower-risk group.
Collapse
Affiliation(s)
- Beibei Qu
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Xiuhua Han
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Lan Zhao
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Feifei Zhang
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| | - Qingmei Gao
- Department of Hematology, Jiading District Central Hospital, Affiliated Shanghai University of Medicine and Health Sciences, Shanghai 201800, P.R. China
| |
Collapse
|
5
|
Mortezaee K, Majidpoor J. The impact of hypoxia on immune state in cancer. Life Sci 2021; 286:120057. [PMID: 34662552 DOI: 10.1016/j.lfs.2021.120057] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022]
Abstract
Hypoxia is a known feature of solid tumors and a critical promoter of tumor hallmarks. Hypoxia influences tumor immunity in a way favoring immune evasion and resistance. Extreme hypoxia and aberrant hypoxia-inducible factor-1 (HIF-1) activity in tumor microenvironment (TME) is a drawback for effective immunotherapy. Infiltration and activity of CD8+ T cells is reduced in such condition, whereas regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) show high activities. Highly hypoxic TME also impairs maturation and activity of dendritic cell (DCs) and natural killer (NK) cells. In addition, the hypoxic TME positively is linked positively with metabolic changes in cells of immune system. These alterations are indicative of a need for hypoxia modulation as a complementary targeting strategy to go with immune checkpoint inhibitor (ICI) therapy.
Collapse
Affiliation(s)
- Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Disease Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
6
|
Peng W, Zhang ML, Zhang J, Chen G. Potential role of hydrogen sulfide in central nervous system tumors: a narrative review. Med Gas Res 2021; 12:6-9. [PMID: 34472496 PMCID: PMC8447953 DOI: 10.4103/2045-9912.324590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Central nervous system tumors are classified as diseases of special clinical significance with high disability and high mortality. In addition to cerebrovascular diseases and craniocerebral injuries, tumors are the most common diseases of the central nervous system. Hydrogen sulfide, the third endogenous gas signaling molecule discovered in humans besides nitric oxide and carbon monoxide, plays an important role in the pathophysiology of human diseases. It is reported that hydrogen sulfide not only exerts a wide range of biological effects, but also develops a certain relationship with tumor development and neovascularization. A variety of studies have shown that hydrogen sulfide acts as a vasodilator and angiogenetic factor to facilitate growth, proliferation, migration and invasion of cancer cells. In this review, the pathological mechanisms and the effect of hydrogen sulfide on the central nervous system tumors are introduced.
Collapse
Affiliation(s)
- Wei Peng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Meng-Ling Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jian Zhang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
7
|
Harada M, Iida Y, Kotani H, Minami T, Komohara Y, Eto M, Yoshikawa K, Uemura H. T-cell responses and combined immunotherapy against human carbonic anhydrase 9-expressing mouse renal cell carcinoma. Cancer Immunol Immunother 2021; 71:339-352. [PMID: 34160685 DOI: 10.1007/s00262-021-02992-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 06/16/2021] [Indexed: 10/21/2022]
Abstract
Renal cell carcinoma (RCC) is known to respond to immune checkpoint blockade (ICB) therapy, whereas there has been limited analysis of T-cell responses to RCC. In this study, we utilized human carbonic anhydrase 9 (hCA9) as a model neoantigen of mouse RENCA RCC. hCA9-expressing RENCA RCC (RENCA/hCA9) cells were rejected in young mice but grew in aged mice. CD8+ T cells were the primary effector cells involved in rejection in young mice, whereas CD4+ T cells participated at the early stage. Screening of a panel of hCA9-derived peptides revealed that mouse CD8+ T cells responded to hCA9288-296 peptide. Mouse CD4+ T cells responded to lysates of RENCA/hCA9, but not RENCA cells, and showed reactivity to hCA9 276-290, which shares three amino acids with hCA9 288-296 peptide. Immunohistochemistry analysis revealed that few T cells infiltrated RENCA/hCA9 tissues in aged mice. ICB therapy of anti-PD-1/anti-CTLA-4 antibodies promoted T-cell infiltration into tumor tissues, whereas no definite antitumor effect was observed. However, additional combination with cyclophosphamide or axitinib, a vascular endothelial growth factor receptor inhibitor, induced complete regression in half of the RENCA/hCA9-bearing aged mice with increased expression of PD-L1 in tumor tissues. These results indicate that hCA9 can be a useful model neoantigen to investigate antitumor T-cell responses in mice with RCC, and that RENCA/hCA9 in aged mice can serve as a non-inflamed 'cold' tumor model facilitating the development of effective combined immunotherapies for RCC.
Collapse
Affiliation(s)
- Mamoru Harada
- Department of Immunology, Shimane University Faculty of Medicine, Izumo, Shimane, 693-8501, Japan.
| | - Yuichi Iida
- Department of Immunology, Shimane University Faculty of Medicine, Izumo, Shimane, 693-8501, Japan
| | - Hitoshi Kotani
- Department of Immunology, Shimane University Faculty of Medicine, Izumo, Shimane, 693-8501, Japan
| | - Takafumi Minami
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Kazuhiro Yoshikawa
- Division of Research Creation and Biobank, Research Creation Support Center, Aichi Medical University, Aichi, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| |
Collapse
|
8
|
Maiorano BA, Schinzari G, Ciardiello D, Rodriquenz MG, Cisternino A, Tortora G, Maiello E. Cancer Vaccines for Genitourinary Tumors: Recent Progresses and Future Possibilities. Vaccines (Basel) 2021; 9:623. [PMID: 34207536 PMCID: PMC8228524 DOI: 10.3390/vaccines9060623] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 06/04/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In the last years, many new treatment options have widened the therapeutic scenario of genitourinary malignancies. Immunotherapy has shown efficacy, especially in the urothelial and renal cell carcinomas, with no particular relevance in prostate cancer. However, despite the use of immune checkpoint inhibitors, there is still high morbidity and mortality among these neoplasms. Cancer vaccines represent another way to activate the immune system. We sought to summarize the most recent advances in vaccine therapy for genitourinary malignancies with this review. METHODS We searched PubMed, Embase and Cochrane Database for clinical trials conducted in the last ten years, focusing on cancer vaccines in the prostate, urothelial and renal cancer. RESULTS Various therapeutic vaccines, including DNA-based, RNA-based, peptide-based, dendritic cells, viral vectors and modified tumor cells, have been demonstrated to induce specific immune responses in a variable percentage of patients. However, these responses rarely corresponded to significant survival improvements. CONCLUSIONS Further preclinical and clinical studies will improve the knowledge about cancer vaccines in genitourinary malignancies to optimize dosage, select targets with a driver role for tumor development and growth, and finally overcome resistance mechanisms. Combination strategies represent possibly more effective and long-lasting treatments.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
| | - Giovanni Schinzari
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
- Medical Oncology Unit, Comprehensive Cancer Center, Foundation A. Gemelli Policlinic IRCCS, 00168 Rome, Italy
| | - Davide Ciardiello
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
- Medical Oncology, Department of Precision Medicine, Luigi Vanvitelli University of Campania, 80131 Naples, Italy
| | - Maria Grazia Rodriquenz
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
| | - Antonio Cisternino
- Urology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy;
| | - Giampaolo Tortora
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
- Medical Oncology Unit, Comprehensive Cancer Center, Foundation A. Gemelli Policlinic IRCCS, 00168 Rome, Italy
| | - Evaristo Maiello
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
| |
Collapse
|
9
|
Abstract
Renal cell carcinoma (RCC) is increasing in incidence and one third of newly diagnosed cases already are metastatic. The metastatic spread of solid tumors renders RCC incurable by surgical resection and consequently more difficult to treat. New molecular-targeted therapies have played a pivotal role in RCC treatment. Unfortunately, tumors frequently develop resistance to these targeted therapies by activating bypass pathways in which alternative signaling or biochemical pathways are activated in response to targeted inhibition of a signaling pathway, allowing cancer cells to continue to survive. Although the advent of immunotherapy with checkpoint inhibitors has led to significant changes in the treatment landscape for advanced RCC, many issues remain to be resolved. For these reasons, there is an urgent need to develop novel therapies and new treatment paradigms for patients with RCC. Much research has been performed thus far in identifying novel targets and treatment strategies in RCC and many of these currently are under investigation and/or in clinical trials. In this article, we discuss therapeutic options in the management of RCC with a focus on the new therapeutic approaches currently investigated in research and for use in the clinic. We divide these potential novel therapies into five groups: nonbiologics, small-molecule drugs, biologics, immunomodulatory therapies, and peptide drugs. We also present some therapeutics and treatment paradigms.
Collapse
Affiliation(s)
- David C Yang
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA
| | - Ching-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA.
| |
Collapse
|
10
|
Obara W, Kanehira M, Katagiri T, Kato R, Kato Y, Takata R. Present status and future perspective of peptide-based vaccine therapy for urological cancer. Cancer Sci 2018; 109:550-559. [PMID: 29345737 PMCID: PMC5834812 DOI: 10.1111/cas.13506] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/26/2017] [Accepted: 01/12/2018] [Indexed: 12/20/2022] Open
Abstract
Use of peptide‐based vaccines as therapeutics aims to elicit immune responses through antigenic epitopes derived from tumor antigens. Peptide‐based vaccines are easily synthesized and lack significant side‐effects when given in vivo. Peptide‐based vaccine therapy against several cancers including urological cancers has made progress for several decades, but there is no worldwide approved peptide vaccine. Peptide vaccines were also shown to induce a high frequency of immune response in patients accompanied by clinical efficacy. These data are discussed in light of the recent progression of immunotherapy caused by the addition of immune checkpoint inhibitors thus providing a general picture of the potential therapeutic efficacy of peptide‐based vaccines and their combination with other biological agents. In this review, we discuss the mechanism of the antitumor effect of peptide‐based vaccine therapy, development of our peptide vaccine, recent clinical trials using peptide vaccines for urological cancers, and perspectives of peptide‐based vaccine therapy.
Collapse
Affiliation(s)
- Wataru Obara
- Department of Urology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Mitsugu Kanehira
- Department of Urology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Toyomasa Katagiri
- Division of Genome Medicine, Institute for Genome Research, Tokushima University, Tokushima, Japan
| | - Renpei Kato
- Department of Urology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Yoichiro Kato
- Department of Urology, Iwate Medical University School of Medicine, Morioka, Japan
| | - Ryo Takata
- Department of Urology, Iwate Medical University School of Medicine, Morioka, Japan
| |
Collapse
|