1
|
Tian Z, Jia W, Wang Z, Mao H, Zhang J, Shi Q, Li X, Song S, Zhang J, Zhu Y, Yang B, Huang C, Huang J. Clinical significance of immune-related antigen CD58 in gliomas and analysis of its potential core related gene clusters. Heliyon 2024; 10:e29275. [PMID: 38699747 PMCID: PMC11063413 DOI: 10.1016/j.heliyon.2024.e29275] [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: 03/23/2024] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
Background The clinical significance of immune-related antigen CD58 in gliomas remains uncertain. The aim of this study was to examine the clinical importance and possible core related genes of CD58 in gliomas. Methods Pan-cancer analysis was to observe the association between CD58 and different tumors, glioma RNA sequencing data and clinical sample analyses were used to observe the relationship between CD58 and glioma, shRNA interference models were to observe the impact of CD58 on glioma cell function, and four glioma datasets and two online analysis platforms were used to explore the core related genes affecting the correlation between CD58 and glioma. Results High CD58 expression was associated with worse prognosis in various tumors and higher malignancy in glioma. Down regulation of CD58 expression was linked to decreased proliferation, increased apoptosis, and reduced metastasis in glioma cells. The pathways involved in CD58-related effects were enriched for immune cell adhesion and immune factor activation, and the core genes were CASP1, CCL2, IL18, MYD88, PTPRC, and TLR2. The signature of CD58 and its core-related genes showed superior predictive power for glioma prognosis. Conclusion High CD58 expression is correlated with more malignant glioma types, and also an independent risk factor for mortality in glioma. CD58 and its core-related genes may serve as novel biomarkers for diagnosing and treating glioma.
Collapse
Affiliation(s)
- Zhi Tian
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Wei Jia
- Medical College of Jishou University, Jishou, Hunan, 416000 PR China
| | - Zhao Wang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Hui Mao
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Jingjing Zhang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Qiongya Shi
- Medical College of Jishou University, Jishou, Hunan, 416000 PR China
| | - Xing Li
- Medical College of Jishou University, Jishou, Hunan, 416000 PR China
| | - Shaoyu Song
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Jiao Zhang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Yingjie Zhu
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Bo Yang
- Department of Pathology, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Chunhai Huang
- Department of Neurosurgery, First Affiliated Hospital, Jishou University, Jishou, Hunan, 416000 PR China
| | - Jun Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008 PR China
| |
Collapse
|
2
|
Hikmet F, Rassy M, Backman M, Méar L, Mattsson JSM, Djureinovic D, Botling J, Brunnström H, Micke P, Lindskog C. Expression of cancer-testis antigens in the immune microenvironment of non-small cell lung cancer. Mol Oncol 2023; 17:2603-2617. [PMID: 37341056 DOI: 10.1002/1878-0261.13474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 05/15/2023] [Accepted: 06/19/2023] [Indexed: 06/22/2023] Open
Abstract
The antigenic repertoire of tumors is critical for successful anti-cancer immune response and the efficacy of immunotherapy. Cancer-testis antigens (CTAs) are targets of humoral and cellular immune reactions. We aimed to characterize CTA expression in non-small cell lung cancer (NSCLC) in the context of the immune microenvironment. Of 90 CTAs validated by RNA sequencing, eight CTAs (DPEP3, EZHIP, MAGEA4, MAGEB2, MAGEC2, PAGE1, PRAME, and TKTL1) were selected for immunohistochemical profiling in cancer tissues from 328 NSCLC patients. CTA expression was compared with immune cell densities in the tumor environment and with genomic, transcriptomic, and clinical data. Most NSCLC cases (79%) expressed at least one of the analyzed CTAs, and CTA protein expression correlated generally with RNA expression. CTA profiles were associated with immune profiles: high MAGEA4 expression was related to M2 macrophages (CD163) and regulatory T cells (FOXP3), low MAGEA4 was associated with T cells (CD3), and high EZHIP was associated with plasma cell infiltration (adj. P-value < 0.05). None of the CTAs correlated with clinical outcomes. The current study provides a comprehensive evaluation of CTAs and suggests that their association with immune cells may indicate in situ immunogenic effects. The findings support the rationale to harness CTAs as targets for immunotherapy.
Collapse
Affiliation(s)
- Feria Hikmet
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Marc Rassy
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Max Backman
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Loren Méar
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | | | - Dijana Djureinovic
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
- Department of Medicine (Medical Oncology), Yale University School of Medicine, New Haven, CT, USA
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Hans Brunnström
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| |
Collapse
|
3
|
Hawlina S, Zorec R, Chowdhury HH. Potential of Personalized Dendritic Cell-Based Immunohybridoma Vaccines to Treat Prostate Cancer. Life (Basel) 2023; 13:1498. [PMID: 37511873 PMCID: PMC10382052 DOI: 10.3390/life13071498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed cancer and the second most common cause of death due to cancer. About 30% of patients with PCa who have been castrated develop a castration-resistant form of the disease (CRPC), which is incurable. In the last decade, new treatments that control the disease have emerged, slowing progression and spread and prolonging survival while maintaining the quality of life. These include immunotherapies; however, we do not yet know the optimal combination and sequence of these therapies with the standard ones. All therapies are not always suitable for every patient due to co-morbidities or adverse effects of therapies or both, so there is an urgent need for further work on new therapeutic options. Advances in cancer immunotherapy with an immune checkpoint inhibition mechanism (e.g., ipilimumab, an anti-CTLA-4 inhibitor) have not shown a survival benefit in patients with CRPC. Other immunological approaches have also not given clear results, which has indirectly prevented breakthrough for this type of therapeutic strategy into clinical use. Currently, the only approved form of immunotherapy for patients with CRPC is a cell-based medicine, but it is only available to patients in some parts of the world. Based on what was gained from recently completed clinical research on immunotherapy with dendritic cell-based immunohybridomas, the aHyC dendritic cell vaccine for patients with CRPC, we highlight the current status and possible alternatives that should be considered in the future.
Collapse
Affiliation(s)
- Simon Hawlina
- Clinical Department of Urology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Surgery, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Helena H Chowdhury
- Laboratory of Cell Engineering, Celica Biomedical, 1000 Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
4
|
Jiang M, Chen W, Sun Y, Zeng J, Ma L, Gong J, Guan X, Lu K, Zhang W. Synergistically enhanced cancer immunotherapy by combining protamine-based nanovaccine with PD-L1 gene silence nanoparticle. Int J Biol Macromol 2023; 242:125223. [PMID: 37276908 DOI: 10.1016/j.ijbiomac.2023.125223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/01/2023] [Accepted: 06/02/2023] [Indexed: 06/07/2023]
Abstract
Tumor vaccine has brought a new dawn for cancer immunotherapy, but disillusionary therapeutic outcomes have been achieved due to the inefficient in vivo vaccine delivery. Moreover, tumor cells customarily resort to various wily tricks to circumvent the recognition and sweeping of the immune system, the immune escape effect has badly aggravated the difficulty of cancer management. With respect to the foregoing, in this study, a promising combinational strategy which cooperated nanovaccine with immune escape inhibition was developed for synergistically enhancing the oncotherapy efficiency. On the one hand, natural polycationic macromolecule protamine (PRT) was utilized as the carrier to construct an antigen and adjuvant co-packaged nanovaccine for facilitating the ingestion in antigen-presenting cells, amplifying antigen cross-presentation and optimizing in vivo delivery. On the other hand, PD-L1 silence gene was selected and hitchhiked in a pH-responsive nanoparticle developed in our previous study. The therapeutic gene could be successfully delivered into the tumors to down-regulate PD-L1 expression and cripple tumor immune escape. The combination of nanovaccine with PD-L1 gene silence nanoparticle could synchronously stimulate antitumor immune responses and reduce immune escape, synergistically enhance the therapeutic efficiency. This study will furnish the prospective tactics for the research of cancer immunotherapy.
Collapse
Affiliation(s)
- Mingxia Jiang
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Wenqiang Chen
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Yanju Sun
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Jun Zeng
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Lina Ma
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Jianping Gong
- College of Pharmacy, Weifang Medical University, Weifang 261053, China
| | - Xiuwen Guan
- College of Pharmacy, Weifang Medical University, Weifang 261053, China.
| | - Keliang Lu
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China.
| | - Weifen Zhang
- College of Pharmacy, Weifang Medical University, Weifang 261053, China.
| |
Collapse
|
5
|
Goulielmaki M, Stokidis S, Anagnostou T, Voutsas IF, Gritzapis AD, Baxevanis CN, Fortis SP. Frequencies of an Immunogenic HER-2/ neu Epitope of CD8+ T Lymphocytes Predict Favorable Clinical Outcomes in Prostate Cancer. Int J Mol Sci 2023; 24:ijms24065954. [PMID: 36983028 PMCID: PMC10058793 DOI: 10.3390/ijms24065954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/08/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
HER-2/neu is the human epidermal growth factor receptor 2, which is associated with the progression of prostate cancer (PCa). HER-2/neu-specific T cell immunity has been shown to predict immunologic and clinical responses in PCa patients treated with HER-2/neu peptide vaccines. However, its prognostic role in PCa patients receiving conventional treatment is unknown, and this was addressed in this study. The densities of CD8+ T cells specific for the HER-2/neu(780-788) peptide in the peripheral blood of PCa patients under standard treatments were correlated with TGF-β/IL-8 levels and clinical outcomes. We demonstrated that PCa patients with high frequencies of HER-2/neu(780-788)-specific CD8+ T lymphocytes had better progression-free survival (PFS) as compared with PCa patients with low frequencies. Increased frequencies of HER-2/neu(780-788)-specific CD8+ T lymphocytes were also associated with lower levels of TGF-β and IL-8. Our data provide the first evidence of the predictive role of HER-2/neu-specific T cell immunity in PCa.
Collapse
Affiliation(s)
- Maria Goulielmaki
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Savvas Stokidis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | | | - Ioannis F Voutsas
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Angelos D Gritzapis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Constantin N Baxevanis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Sotirios P Fortis
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| |
Collapse
|
6
|
Mukherjee AG, Wanjari UR, Prabakaran DS, Ganesan R, Renu K, Dey A, Vellingiri B, Kandasamy S, Ramesh T, Gopalakrishnan AV. The Cellular and Molecular Immunotherapy in Prostate Cancer. Vaccines (Basel) 2022; 10:vaccines10081370. [PMID: 36016257 PMCID: PMC9416492 DOI: 10.3390/vaccines10081370] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 12/13/2022] Open
Abstract
In recent history, immunotherapy has become a viable cancer therapeutic option. However, over many years, its tenets have changed, and it now comprises a range of cancer-focused immunotherapies. Clinical trials are currently looking into monotherapies or combinations of medicines that include immune checkpoint inhibitors (ICI), CART cells, DNA vaccines targeting viruses, and adoptive cellular therapy. According to ongoing studies, the discipline should progress by incorporating patient-tailored immunotherapy, immune checkpoint blockers, other immunotherapeutic medications, hormone therapy, radiotherapy, and chemotherapy. Despite significantly increasing morbidity, immunotherapy can intensify the therapeutic effect and enhance immune responses. The findings for the immunotherapy treatment of advanced prostate cancer (PCa) are compiled in this study, showing that is possible to investigate the current state of immunotherapy, covering new findings, PCa treatment techniques, and research perspectives in the field’s unceasing evolution.
Collapse
Affiliation(s)
- Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - D. S. Prabakaran
- Department of Radiation Oncology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju 28644, Korea
- Department of Biotechnology, Ayya Nadar Janaki Ammal College (Autonomous), Srivilliputhur Main Road, Sivakasi 626124, Tamil Nadu, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon 24252, Korea
| | - Kaviyarasi Renu
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata 700073, West Bengal, India
| | - Balachandar Vellingiri
- Human Molecular Cytogenetics and Stem Cell Laboratory, Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Sabariswaran Kandasamy
- Water-Energy Nexus Laboratory, Department of Environmental Engineering, University of Seoul, Seoul 02504, Korea
| | - Thiyagarajan Ramesh
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
- Correspondence:
| |
Collapse
|
7
|
Jiang C, Li J, Zhang W, Zhuang Z, Liu G, Hong W, Li B, Zhang X, Chao CC. Potential association factors for developing effective peptide-based cancer vaccines. Front Immunol 2022; 13:931612. [PMID: 35967400 PMCID: PMC9364268 DOI: 10.3389/fimmu.2022.931612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/29/2022] [Indexed: 11/26/2022] Open
Abstract
Peptide-based cancer vaccines have been shown to boost immune systems to kill tumor cells in cancer patients. However, designing an effective T cell epitope peptide-based cancer vaccine still remains a challenge and is a major hurdle for the application of cancer vaccines. In this study, we constructed for the first time a library of peptide-based cancer vaccines and their clinical attributes, named CancerVaccine (https://peptidecancervaccine.weebly.com/). To investigate the association factors that influence the effectiveness of cancer vaccines, these peptide-based cancer vaccines were classified into high (HCR) and low (LCR) clinical responses based on their clinical efficacy. Our study highlights that modified peptides derived from artificially modified proteins are suitable as cancer vaccines, especially for melanoma. It may be possible to advance cancer vaccines by screening for HLA class II affinity peptides may be an effective therapeutic strategy. In addition, the treatment regimen has the potential to influence the clinical response of a cancer vaccine, and Montanide ISA-51 might be an effective adjuvant. Finally, we constructed a high sensitivity and specificity machine learning model to assist in designing peptide-based cancer vaccines capable of providing high clinical responses. Together, our findings illustrate that a high clinical response following peptide-based cancer vaccination is correlated with the right type of peptide, the appropriate adjuvant, and a matched HLA allele, as well as an appropriate treatment regimen. This study would allow for enhanced development of cancer vaccines.
Collapse
Affiliation(s)
- Chongming Jiang
- Department of Medicine, Baylor College of Medicine, Houston TX, United States
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Chongming Jiang, ; Cheng-Chi Chao,
| | - Jianrong Li
- Department of Medicine, Baylor College of Medicine, Houston TX, United States
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, United States
| | - Wei Zhang
- Institute of Super Cell, BGI-Shenzhen, Shenzhen, China
| | | | - Geng Liu
- Institute of Super Cell, BGI-Shenzhen, Shenzhen, China
| | - Wei Hong
- Department of Medicine, Baylor College of Medicine, Houston TX, United States
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, United States
| | - Bo Li
- Institute of Super Cell, BGI-Shenzhen, Shenzhen, China
| | - Xiuqing Zhang
- Institute of Super Cell, BGI-Shenzhen, Shenzhen, China
| | - Cheng-Chi Chao
- Department of Pipeline Development, Biomap, Inc, San Francisco, CA, United States
- *Correspondence: Chongming Jiang, ; Cheng-Chi Chao,
| |
Collapse
|
8
|
Suekane S, Yutani S, Toh U, Yoshiyama K, Itoh K. Immune responses of patients without cancer recurrence after a cancer vaccine over a long term. Mol Clin Oncol 2022; 16:112. [PMID: 35620212 PMCID: PMC9112399 DOI: 10.3892/mco.2022.2545] [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: 02/10/2022] [Accepted: 04/26/2022] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to clarify the humoral and cellular immune responses of patients with cancer who experienced no recurrence over a long term after receiving a cancer vaccine. The immune kinetics were investigated in response to a personalized peptide vaccination (PPV) among 44 Japanese patients without an active tumor at entry to the vaccination: Lung adenocarcinoma (n=11); colon (n=18); and breast cancer (n=15) (9, 10, 12, 8 and 5 patients with stage I, II, III and IV recurrences, respectively). The patients' immunoglobulin G (IgG) and cytotoxic T lymphocyte (CTL) activities were measured using a multiplexed Luminex assay and an interferon-γ release assay, respectively. There were no severe adverse events related to the PPV other than a grade III injection site reaction. A potent boost in IgG or CTL at the end of the 1st vaccination cycle was observed in 77% of the patients (n=84). The IgG levels were sustained throughout the follow-up period, whereas the CTL levels declined and were transient. A total of 37 of the 44 patients (84%) had no recurrence, with a median follow-up of 67.6 months (interquartile range, 45.6-82.8 months). Overall, the PPV induced long-term humoral immunity with transient cellular immunity in the majority of patients with cancer without an active tumor at their entry to the PPV.
Collapse
Affiliation(s)
- Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Shigeru Yutani
- Kurume Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Uhi Toh
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Koichi Yoshiyama
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Kyogo Itoh
- Kurume Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
9
|
Development of Cancer Immunotherapies. Cancer Treat Res 2022; 183:1-48. [PMID: 35551655 DOI: 10.1007/978-3-030-96376-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cancer immunotherapy, or the utilization of components of the immune system to target and eliminate cancer, has become a highly active area of research in the past several decades and a common treatment strategy for several cancer types. The concept of harnessing the immune system for this purpose originated over 100 years ago when a physician by the name of William Coley successfully treated several of his cancer patients with a combination of live and attenuated bacteria, later known as "Coley's Toxins", after observing a subset of prior patients enter remission following their diagnosis with the common bacterial infection, erysipelas. However, it was not until late in the twentieth century that cancer immunotherapies were developed for widespread use, thereby transforming the treatment landscape of numerous cancer types. Pivotal studies elucidating molecular and cellular functions of immune cells, such as the discovery of IL-2 and production of monoclonal antibodies, fostered the development of novel techniques for studying the immune system and ultimately the development and approval of several cancer immunotherapies by the United States Food and Drug Association in the 1980s and 1990s, including the tuberculosis vaccine-Bacillus Calmette-Guérin, IL-2, and the CD20-targeting monoclonal antibody. Approval of the first therapeutic cancer vaccine, Sipuleucel-T, for the treatment of metastatic castration-resistant prostate cancer and the groundbreaking success and approval of immune checkpoint inhibitors and chimeric antigen receptor T cell therapy in the last decade, have driven an explosion of interest in and pursuit of novel cancer immunotherapy strategies. A broad range of modalities ranging from antibodies to adoptive T cell therapies is under investigation for the generalized treatment of a broad spectrum of cancers as well as personalized medicine. This chapter will focus on the recent advances, current strategies, and future outlook of immunotherapy development for the treatment of cancer.
Collapse
|
10
|
Liu T, Tan J, Wu M, Fan W, Wei J, Zhu B, Guo J, Wang S, Zhou P, Zhang H, Shi L, Li J. High-affinity neoantigens correlate with better prognosis and trigger potent antihepatocellular carcinoma (HCC) activity by activating CD39 +CD8 + T cells. Gut 2021; 70:1965-1977. [PMID: 33262196 PMCID: PMC8458084 DOI: 10.1136/gutjnl-2020-322196] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 11/03/2020] [Accepted: 11/04/2020] [Indexed: 12/27/2022]
Abstract
OBJECTIVE It remains controversial whether tumour mutational burden (TMB) or neoantigens are prognostic markers in hepatocellular carcinoma (HCC). This study aimed to define the function of TMB or neoantigens in antitumour immunotherapy. DESIGN Neoantigens of patients (n=56) were analysed by pVAC tools with major histocompatibility complex-1 (MHC-I) algorithms based on whole exome sequencing and neoantigens with mutant type IC50 <50 nM were defined as high-affinity neoantigens (HANs). Patients were segregated into HAN-high/low groups by median of HAN value, and overall survival (OS) was analysed. Autologous organoid killing model was developed to clarify the antitumour activity of HANs. RESULTS The value of HAN showed a better correlation with OS (p=0.0199) than TMB (p=0.7505) or neoantigens (p=0.2297) in patients with HCC and positively correlated with the frequency of CD39+CD8+ tumour infiltrating lymphocytes (TILs). Furthermore, HAN-specific CD8+ T cells were identified in CD39+CD8+ TILs, which showed better antitumour activity in HAN-high versus HAN-low group. In addition, more effective HAN peptides were identified in HAN-high versus HAN-low group. Besides, flow cytometry data showed that in fresh tumour, CD39+PD-1intCD8+ TILs displayed an effector phenotype and stronger antitumour activity in HAN-high versus HAN-low group. More importantly, patients in HAN-high versus HAN-low group showed a better prognosis after anti-PD-1 therapy. CONCLUSIONS Our study first demonstrates that HAN value positively correlates with better OS in patients with HCC. HANs trigger antitumour activity by activating tumour-reactive CD39+CD8+ T cells, and patients in HAN-high group benefited more from anti-PD-1 therapy than HAN-low group. These findings may provide a novel strategy for personalised antitumour therapies for HCC.
Collapse
Affiliation(s)
- Ting Liu
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jizhou Tan
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Minhao Wu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wenzhe Fan
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jialiang Wei
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Bowen Zhu
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jian Guo
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Shutong Wang
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Penghui Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hui Zhang
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Liangrong Shi
- Radiological Intervention Center, Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Kasperska A, Borowczak J, Szczerbowski K, Stec E, Ahmadi N, Szylber Ł. Current challenges in targeting tumor desmoplasia to improve the efficacy of immunotherapy. Curr Cancer Drug Targets 2021; 21:919-931. [PMID: 34525931 DOI: 10.2174/1568009621666210825101456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/31/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022]
Abstract
Desmoplasia is crucial for the development, progression and treatment of immune-resistant malignancies. and treatment of immune-resistant malignancies. Targeting desmoplasia-related metabolic pathways appears to be an interesting approach to expand our stock of disposable anti-tumor agents.CXCL12/CXCR4 axis inhibition reduces fibrosis, alleviates immunosuppression and significantly enhances the efficacy of PD-1 immunotherapy. CD40L substitute therapy may increase the activity of T-cells, downregulate CD40+, prolong patients' survival and prevent cancer progression. Although FAPα antagonists used in preclinical models did not lead to permanent cure, an alleviation of immune-resistance, modification of desmoplasia and a decrease in angiogenesis were observed. Targeting DDR2 may enhance the effect of anti-PD-1 treatment in multiple neoplasm cell lines and has the ability to overcome the adaptation to BRAF-targeted therapy in melanoma. Reprogramming desmoplasia could potentially cooperate not only with present treatment, but also other potential therapeutic targets. We present the most promising metabolic pathways related to desmoplasia and discuss the emerging strategies to improve the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Anna Kasperska
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| | - Jędrzej Borowczak
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| | - Krzysztof Szczerbowski
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| | - Ewa Stec
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| | - Navid Ahmadi
- Department of Cardiothoracic Surgery, Royal Papworth Hospital, Cambridge. United Kingdom
| | - Łukasz Szylber
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun. Poland
| |
Collapse
|
12
|
Zhang W, Yin Q, Huang H, Lu J, Qin H, Chen S, Zhang W, Su X, Sun W, Dong Y, Li Q. Personal Neoantigens From Patients With NSCLC Induce Efficient Antitumor Responses. Front Oncol 2021; 11:628456. [PMID: 33928024 PMCID: PMC8076796 DOI: 10.3389/fonc.2021.628456] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/23/2021] [Indexed: 12/26/2022] Open
Abstract
Objective To develop a neoantigen-targeted personalized cancer treatment for non-small cell lung cancer (NSCLC), neoantigens were obtained from collected human lung cancer samples, and the utility of neoantigen and neoantigen-reactive T cells (NRTs) was assessed. Methods Tumor specimens from three patients with NSCLC were obtained and analyzed by whole-exome sequencing, and neoantigens were predicted accordingly. Dendritic cells and T lymphocytes were isolated, NRTs were elicited and IFN-γ ELISPOT tests were conducted. HLA-A2.1/Kb transgenic mice were immunized with peptides from HLA-A*02:01+patient with high immunogenicity, and NRTs were subjected to IFN-γ, IL-2 and TNF-α ELISPOT as well as time-resolved fluorescence assay for cytotoxicity assays to verify the immunogenicity in vitro. The HLA-A*02:01+lung cancer cell line was transfected with minigene and inoculated into the flanks of C57BL/6nu/nu mice and the NRTs induced by the immunogenic polypeptides from autologous HLA-A2.1/Kb transgenic mice were adoptively transfused to verify their immunogenicity in vivo. Results Multiple putative mutation-associated neoantigens with strong affinity for HLA were selected from each patient. Immunogenic neoantigen were identified in all three NSCLC patients, the potency of ACAD8-T105I, BCAR1-G23V and PLCG1-M425L as effective neoantigen to active T cells in suppressing tumor growth was further proven both in vitro and in vivo using HLA-A2.1/Kb transgenic mice and tumor-bearing mouse models. Conclusion Neoantigens with strong immunogenicity can be screened from NSCLC patients through the whole-exome sequencing of patient specimens and machine-learning-based neoantigen predictions. NRTs shown efficient antitumor responses in transgenic mice and tumor-bearing mouse models. Our results indicate that the development of neoantigen-based personalized immunotherapies in NSCLC is possible. Precis Neoantigens with strong immunogenicity were screened from NSCLC patients. This research provides evidence suggesting that neoantigen-based therapy might serve as feasible treatment for NSCLC.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qi Yin
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Haidong Huang
- Department of Pulmonary and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jingjing Lu
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Hao Qin
- Department of Pulmonary and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Si Chen
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Wenjun Zhang
- Department of Emergency, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaoping Su
- School of Basic Medicine, Wenzhou Medical University, Wenzhou Tea Mountain Higher Education Park, Wenzhou, China
| | - Weihong Sun
- Biotherapy Center, Qingdao Central Hospital, The Second Affiliated Hospital, Qingdao University, Qingdao, China
| | - Yuchao Dong
- Department of Pulmonary and Critical Care Medicine, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Qiang Li
- Department of Pulmonary and Critical Care Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW Cholangiocarcinoma is an aggressive heterogeneous group of cancers of the biliary epithelium and most patients are detected with advanced metastatic disease with poor prognosis. The therapeutic options are limited, and the current standard care as systemic therapy is still cytotoxic chemotherapy. With the understanding of the complex immune microenvironment in the liver and these cancers arising in the milieu of chronic inflammation, recent advances in immune oncology have transformed the landscape of cancer management with breakthroughs in the treatment of several solid tumors. RECENT FINDINGS With the advances of genome sequencing, subgroups of cholangiocarcinoma with hyper mutated status and rich in cancer neoantigen production may be susceptible to immunotherapies like cancer vaccines and immune checkpoint inhibitors by eliciting a host immune response resulting in tumor rejection or overcoming the immunosuppressive local tumor microenvironment. SUMMARY In this review, we look at the most recent evidence behind immunotherapy and its application in the treatment of cholangiocarcinoma. Though its utility is still in early development it shows great promise in improving response rates that may translate to durable disease control and improve clinical outcomes in this aggressive disease.
Collapse
|
14
|
Lin D, He H, Sun J, He X, Long W, Cui X, Sun Y, Zhao S, Zheng X, Zeng Z, Zhang K, Wang H. Co-delivery of PSMA antigen epitope and mGM-CSF with a cholera toxin-like chimeric protein suppressed prostate tumor growth via activating dendritic cells and promoting CTL responses. Vaccine 2021; 39:1609-1620. [PMID: 33612342 DOI: 10.1016/j.vaccine.2021.02.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 01/09/2021] [Accepted: 02/02/2021] [Indexed: 01/22/2023]
Abstract
Subunit vaccines derived from tumor antigens play a role in tumor therapy because of their unique advantages. However, because of the weak immunogenicity of peptides in subunit vaccines, it is difficult to trigger an effective cytotoxic T lymphocyte (CTL) response, which is critical for cancer therapy. A requirement for the activation of CTL cells by exogenous antigens is the stimulation of antigen presenting cells (APC) with the help of adjuvants and cross-presentation to T lymphocytes. Standard nonconjugated adjuvant-peptide mixtures do not ensure co-targeting of the antigen and the adjuvant to the same APC, which limits the effects of adjuvants. In this study, a fusion protein consisting of murine granulocyte-macrophage colony stimulating factor (mGM-CSF) fused with CTA2 (A2 subunit of cholera toxin) was generated and assembled with CTB-PSMA624-632 (prostate specific membrane antigen (PSMA) peptide 624-632 fused to CTB) to obtain a cholera toxin-like protein. The chimeric protein retained the biological activity of mGM-CSF and had stronger GM1 binding activity than (CTB-PSMA624-632)5. C57BL/6J mice immunized with the CT-like chimeric protein exhibited delayed tumor growth following challenge with human PSMA-EGFP-expressing RM-1 cells. Experiment results showed that the CT-like chimeric protein could induce the maturation of DC cells and improve CTL responses. Overall, these results indicate that the nasal administration of a CT-like chimeric protein vaccine results in the development of effective immunity against prostate tumor cells and might be useful for future clinical anti-tumoral applications.
Collapse
Affiliation(s)
- Danmin Lin
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Huafeng He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Jiajie Sun
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Xianying He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Wei Long
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Xiping Cui
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Yunxiao Sun
- Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, PR China
| | - Suqing Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Xi Zheng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Zheng Zeng
- The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, PR China
| | - Kun Zhang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China; School of Biotechnology and Health, Wuyi University, Jiangmen 529020, PR China
| | - Huaqian Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
15
|
Michiyuki S, Tomita N, Mori Y, Kanda H, Tashiro K, Notomi T. Discrimination of a single nucleotide polymorphism in the haptoglobin promoter region, rs5472, using a competitive fluorophore-labeled probe hybridization assay following loop-mediated isothermal amplification. Biosci Biotechnol Biochem 2021; 85:359-368. [PMID: 33604636 DOI: 10.1093/bbb/zbaa012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022]
Abstract
Personalized peptide vaccination, which involves activation of the host immune system against cancer cells using personalized peptide vaccines (PPVs), can improve overall survival in multiple cancer types. However, the clinical efficacies of PPVs vary for unknown reasons. Recently, a single nucleotide polymorphism (NG_012651.1:g.4461_5460[4960A>G]) in the haptoglobin promoter region, rs5472, was significantly associated with clinical response of PPV. Therefore, rs5472 is expected to be a predictive biomarker for PPV therapy. Here, we described a single nucleotide discrimination method for rs5472 analysis by combining the loop-mediated isothermal amplification and quenching probe methods. In evaluation of saliva samples, this method showed high concordance with the results of Sanger sequencing (100%, n = 36). Importantly, this method did not require calculation of melting temperature for single nucleotide discrimination and could therefore be carried out on a simple instrument. Accordingly, this method may be more robust and applicable to near-patient testing.
Collapse
Affiliation(s)
- Satoru Michiyuki
- Biochemical Research Laboratory, Eiken Chemical Co., Ltd., Otawara, Tochigi, Japan
| | - Norihiro Tomita
- Biochemical Research Laboratory, Eiken Chemical Co., Ltd., Otawara, Tochigi, Japan
| | - Yasuyoshi Mori
- Biochemical Research Laboratory, Eiken Chemical Co., Ltd., Otawara, Tochigi, Japan
| | - Hidetoshi Kanda
- Biochemical Research Laboratory, Eiken Chemical Co., Ltd., Otawara, Tochigi, Japan
| | - Kosuke Tashiro
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Nishi-Ku, Fukuoka, Japan
| | - Tsugunori Notomi
- Biochemical Research Laboratory, Eiken Chemical Co., Ltd., Otawara, Tochigi, Japan
| |
Collapse
|
16
|
Bansal D, Reimers MA, Knoche EM, Pachynski RK. Immunotherapy and Immunotherapy Combinations in Metastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2021; 13:cancers13020334. [PMID: 33477569 PMCID: PMC7831137 DOI: 10.3390/cancers13020334] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/01/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
Although most prostate cancers are localized, and the majority are curable, recurrences occur in approximately 35% of men. Among patients with prostate-specific antigen (PSA) recurrence and PSA doubling time (PSADT) less than 15 months after radical prostatectomy, prostate cancer accounted for approximately 90% of the deaths by 15 years after recurrence. An immunosuppressive tumor microenvironment (TME) and impaired cellular immunity are likely largely responsible for the limited utility of checkpoint inhibitors (CPIs) in advanced prostate cancer compared with other tumor types. Thus, for immunologically "cold" malignancies such as prostate cancer, clinical trial development has pivoted towards novel approaches to enhance immune responses. Numerous clinical trials are currently evaluating combination immunomodulatory strategies incorporating vaccine-based therapies, checkpoint inhibitors, and chimeric antigen receptor (CAR) T cells. Other trials evaluate the efficacy and safety of these immunomodulatory agents' combinations with standard approaches such as androgen deprivation therapy (ADT), taxane-based chemotherapy, radiotherapy, and targeted therapies such as tyrosine kinase inhibitors (TKI) and poly ADP ribose polymerase (PARP) inhibitors. Here, we will review promising immunotherapies in development and ongoing trials for metastatic castration-resistant prostate cancer (mCRPC). These novel trials will build on past experiences and promise to usher a new era to treat patients with mCRPC.
Collapse
|
17
|
Uchino Y, Muroya D, Yoshitomi M, Shichijo S, Yamada A, Sasada T, Yamada T, Okuda K, Itoh K, Yutani S. Investigation of factors associated with reduced clinical benefits of personalized peptide vaccination for pancreatic cancer. Mol Clin Oncol 2020; 14:39. [PMID: 33437477 PMCID: PMC7788558 DOI: 10.3892/mco.2020.2201] [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: 02/20/2020] [Accepted: 12/12/2020] [Indexed: 11/29/2022] Open
Abstract
The aim of the present study was to determine the factors associated with reduced clinical benefits of personalized peptide vaccination (PPV) for pancreatic cancer. Phase II PPV clinical trials comprising 309 (8 non-advanced and 301 advanced-stage) patients with pancreatic cancer were conducted. Two to four peptides were selected among a set of 31 different peptides as vaccine candidates for personalized peptide vaccination based on human leukocyte antigen types and preexisting peptide-specific IgG levels, and subcutaneously injected. The selected peptides were subcutaneously injected. Of the 309 patients, 81 failed to complete the 1st PPV cycle due to rapid disease progression, and their median overall survival [2.1 months; 95% confidence interval (CI), 1.8-2.7] was significantly shorter than that of the remaining 228 patients (8.4 months; 95% CI, 8.4-9.9; P<0.01). ‘Immune boosting’ was defined when IgG levels before vaccination increased more than 2-fold after vaccination. Immune boosting was observed in the majority of patients with PPV irrespective of whether or not they received concomitant chemotherapy. Additionally, patients demonstrating immune boosting exhibited longer survival rates. Although the positive-response rates and peptide-specific IgG levels in pre- and post-vaccination samples differed among the 31 peptides, patients exhibiting immune boosting in response to each of the vaccinated peptides demonstrated longer survival times. Pre-vaccination factors associated with reduced clinical benefits were high c-reactive protein (CRP) levels, high neutrophil counts, lower lymphocyte and red blood cell counts, advanced disease stage and the greater number of chemotherapy courses prior to the PPV treatment. The post-vaccination factors associated with lower clinical benefits were PPV monotherapy and lower levels of immune boosting. In conclusion, pre-vaccination inflammatory signatures, rather than pre- or post-vaccination immunological signatures, were associated with reduced clinical benefits of personalized peptide vaccination (PPV) for pancreatic cancer.
Collapse
Affiliation(s)
- Yoshihiro Uchino
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839-0823, Japan.,Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Daisuke Muroya
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Munehiro Yoshitomi
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Shigeki Shichijo
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839-0823, Japan
| | - Akira Yamada
- Cancer Vaccine Development Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Tetsuro Sasada
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Kanagawa, Yokohama 241-8515, Japan
| | - Teppei Yamada
- Department of Gastroenterological Surgery, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Koji Okuda
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839-0823, Japan
| | - Shigeru Yutani
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839-0823, Japan
| |
Collapse
|
18
|
Suekane S, Yutani S, Yamada A, Sasada T, Matsueda S, Takamori S, Toh U, Kawano K, Yoshiyama K, Sakamoto S, Sugawara S, Komatsu N, Yamada T, Naito M, Terasaki M, Mine T, Itoh K, Shichijo S, Noguchi M. Identification of biomarkers for personalized peptide vaccination in 2,588 cancer patients. Int J Oncol 2020; 56:1479-1489. [PMID: 32236612 PMCID: PMC7170040 DOI: 10.3892/ijo.2020.5019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 03/05/2020] [Indexed: 01/08/2023] Open
Abstract
Peptide-based cancer vaccines have failed to provide sufficient clinical benefits in order to be approved in clinical trials since the 1990s. To understand the mechanisms underlying this failure, the present study investigated biomarkers associated with the lower overall survival (OS) among 2,588 patients receiving personalized peptide vaccination (PPV). Survival data were obtained from a database of 2,588 cancer patients including 399 patients with lung, 354 with prostate and 344 with colon cancer. They entered into phase II clinical trials of PPV in which 2 to 4 of 31 warehouse peptides were selected for vaccination on an individual patient basis based on human leukocyte antigen (HLA) class IA-types and pre-existing peptide-specific IgG levels. Higher pre-vaccination neutrophil, monocyte and platelet counts, and lower pre-vaccination lymphocyte and red blood cell counts were inversely associated with OS, with higher sensitivities in the proportions of neutrophils and lymphocytes, respectively. The most potent unfavorable and favorable factors for OS were the median percentage of neutrophils (>64.8%) or percentage of lymphocytes (>25.1%) with correlation coefficients (R2) of 0.98 and 0.92, respectively. Higher pre-vaccination levels of c-reactive protein and other inflammatory soluble factors were inversely associated with OS. Pre-vaccination peptide-specific immunity levels had no effect on OS, although lower immune boosting levels were inversely associated with OS. None of the 31 peptides was inversely associated with OS, although a few peptides were positively associated with it. On the whole, the findings of the present study suggested that pre-vaccination inflammatory signatures, but not those of post-vaccination immune induction, were associated with lower clinical benefits of PPV.
Collapse
Affiliation(s)
- Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Shigeru Yutani
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| | - Akira Yamada
- Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Tetsuro Sasada
- Cancer Vaccine Center, Kanagawa Cancer Center, Yokohama, Kanagawa 241‑8515, Japan
| | - Satoko Matsueda
- Center for Immunotherapy, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | - Shinzo Takamori
- Department of Surgery, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Uhi Toh
- Department of Surgery, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Kouichiro Kawano
- Department of Obstetrics and Gynecology, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Koichi Yoshiyama
- Department of Surgery, Kurume University, Kurume, Fukuoka 830‑0011, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine School of Medicine, Hiroshima University, Hiroshima, Hiroshima 734‑8551, Japan
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Miyagi 980‑0873, Japan
| | - Nobukazu Komatsu
- Department of Immunology, Kurume University School of Medicine, Kurume, Fukuoka 830‑0011, Japan
| | - Teppei Yamada
- Department of Gastroenterological Surgery, Fukuoka University School of Medicine, Fukuoka, Fukuoka 814‑0180, Japan
| | - Masayasu Naito
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| | | | - Takashi Mine
- Department of Clinical Oncology, Nagasaki Harbor Medical Center, Nagasaki, Nagasaki 850‑8555, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| | - Shigeki Shichijo
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| | - Masanori Noguchi
- Cancer Vaccine Center, Kurume University, Kurume, Fukuoka 839‑0863, Japan
| |
Collapse
|
19
|
Boettcher AN, Usman A, Morgans A, VanderWeele DJ, Sosman J, Wu JD. Past, Current, and Future of Immunotherapies for Prostate Cancer. Front Oncol 2019; 9:884. [PMID: 31572678 PMCID: PMC6749031 DOI: 10.3389/fonc.2019.00884] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/27/2019] [Indexed: 12/22/2022] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men, and the second leading cause of cancer related death in men in Western countries. The standard therapy for metastatic PCa is androgen suppression therapy (AST). Men undergoing AST eventually develop metastatic castration-resistant prostate cancer (mCRPC), of which there are limited treatment options available. Immunotherapy has presented substantial benefits for many types of cancer, but only a marginal benefit for mCRPC, at least in part, due to the immunosuppressive tumor microenvironment (TME). Current clinical trials are investigating monotherapies or combination therapies involving adoptive cellular therapy, viral, DNA vaccines, oncolytic viruses, and immune checkpoint inhibitors (ICI). Immunotherapies are also being combined with chemotherapy, radiation, and AST. Additionally, preclinical investigations show promise with the recent description of alternative ways to circumvent the immunosuppressive nature of the prostate tumor microenvironment, including harnessing the immune stimulatory NKG2D pathway, inhibiting myeloid derived suppressor cells, and utilizing immunomodulatory oncolytic viruses. Herein we provide an overview of recent preclinical and clinical developments in cancer immunotherapies and discuss the perspectives for future immunotherapies in PCa.
Collapse
Affiliation(s)
- Adeline N. Boettcher
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Ahmed Usman
- Department of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alicia Morgans
- Department of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - David J. VanderWeele
- Department of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jeffrey Sosman
- Department of Hematology and Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jennifer D. Wu
- Department of Urology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
20
|
Nakahara Y, Kouro T, Igarashi Y, Kawahara M, Sasada T. Prospects for a personalized peptide vaccine against lung cancer. Expert Rev Vaccines 2019; 18:703-709. [DOI: 10.1080/14760584.2019.1635461] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yoshiro Nakahara
- Department of Respiratory Medicine, Kanagawa Cancer Center, Yokohama, Japan
| | - Taku Kouro
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yuka Igarashi
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Mamoru Kawahara
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Tetsuro Sasada
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine Center, Kanagawa Cancer Center, Yokohama, Japan
| |
Collapse
|
21
|
Chen F, Zou Z, Du J, Su S, Shao J, Meng F, Yang J, Xu Q, Ding N, Yang Y, Liu Q, Wang Q, Sun Z, Zhou S, Du S, Wei J, Liu B. Neoantigen identification strategies enable personalized immunotherapy in refractory solid tumors. J Clin Invest 2019; 129:2056-2070. [PMID: 30835255 DOI: 10.1172/jci99538] [Citation(s) in RCA: 164] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Recent genomic and bioinformatic technological advances have made it possible to dissect the immune response to personalized neoantigens encoded by tumor-specific mutations. However, timely and efficient identification of neoantigens is still one of the major obstacles to using personalized neoantigen-based cancer immunotherapy. METHODS Two different pipelines of neoantigens identification were established in this study: (1) Clinical grade targeted sequencing was performed in patients with refractory solid tumor, and mutant peptides with high variant allele frequency and predicted high HLA-binding affinity were de novo synthesized. (2) An inventory-shared neoantigen peptide library of common solid tumors was constructed, and patients' hotspot mutations were matched to the neoantigen peptide library. The candidate neoepitopes were identified by recalling memory T-cell responses in vitro. Subsequently, neoantigen-loaded dendritic cell vaccines and neoantigen-reactive T cells were generated for personalized immunotherapy in six patients. RESULTS Immunogenic neo-epitopes were recognized by autologous T cells in 3 of 4 patients who utilized the de novo synthesis mode and in 6 of 13 patients who performed shared neoantigen peptide library, respectively. A metastatic thymoma patient achieved a complete and durable response beyond 29 months after treatment. Immune-related partial response was observed in another patient with metastatic pancreatic cancer. The remaining four patients achieved the prolonged stabilization of disease with a median PFS of 8.6 months. CONCLUSIONS The current study provided feasible pipelines for neoantigen identification. Implementing these strategies to individually tailor neoantigens could facilitate the neoantigen-based translational immunotherapy research.TRIAL REGSITRATION. ChiCTR.org ChiCTR-OIC-16010092, ChiCTR-OIC-17011275, ChiCTR-OIC-17011913; ClinicalTrials.gov NCT03171220. FUNDING This work was funded by grants from the National Key Research and Development Program of China (Grant No. 2017YFC1308900), the National Major Projects for "Major New Drugs Innovation and Development" (Grant No.2018ZX09301048-003), the National Natural Science Foundation of China (Grant No. 81672367, 81572329, 81572601), and the Key Research and Development Program of Jiangsu Province (No. BE2017607).
Collapse
|
22
|
Noguchi M, Koga N, Moriya F, Suekane S, Yutani S, Yamada A, Shichijo S, Kakuma T, Itoh K. Survival analysis of multiple peptide vaccination for the selection of correlated peptides in urological cancers. Cancer Sci 2018; 109:2660-2669. [PMID: 29938870 PMCID: PMC6277968 DOI: 10.1111/cas.13709] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/16/2018] [Indexed: 12/24/2022] Open
Abstract
Peptide‐based cancer vaccines are able to induce strong immune responses, but their clinical results are unsatisfactory. To determine clinically correlated peptides, we analyzed survival data from urological cancer patients treated by personalized peptide vaccination (PPV), in which different multiple peptides were used for individual patients based on human leukocyte antigen (HLA) type and pre‐existing immunity. Survival data were obtained from a database of 265 urological cancer patients treated in 5 clinical PPV trials comprising 154 patients with castration‐resistant prostate cancer (CRPC) and 111 patients with advanced urothelial cancer (UC). Expression of tumor‐associated antigens (TAA) was evaluated in 10 prostate cancer tissues, 4 metastatic lymph nodes from prostate cancer, and 10 UC tissues using immunohistochemical staining. Clinical efficacy of individual peptides for overall survival was evaluated by the Cox proportional hazards regression model. All TAA coding candidate peptides used in PPV treatment were expressed in tumor cells from prostate cancer and UC samples except for p56Lck in both, and prostate‐specific antigen (PSA), prostatic acid phosphatase (PAP) and prostate‐specific membrane antigen (PSMA) in the UC samples. Patients with the following peptides had a significantly longer survival than patients without the peptides (hazard ratio <1.0, 95% confidence intervals <1.0 and P < .05): SART3‐109, PTHrP‐102, HNPRL‐140, SART3‐302 and Lck‐90 in CRPC patients, and EGF‐R‐800, Lck‐486, PSMA‐624, CypB‐129 and SART3‐734 in advanced UC patients, respectively. Correlated peptides selected using both survival data and pre‐existing immunity for PPV treatment may enhance the clinical benefits for urological cancer patients.
Collapse
Affiliation(s)
- Masanori Noguchi
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Noriko Koga
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Shigeru Yutani
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Akira Yamada
- Division of Cancer Vaccines in Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Shigeki Shichijo
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Tatuyuki Kakuma
- Bio-statistics Center, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
23
|
Guan X, Chen J, Hu Y, Lin L, Sun P, Tian H, Chen X. Highly enhanced cancer immunotherapy by combining nanovaccine with hyaluronidase. Biomaterials 2018; 171:198-206. [PMID: 29698869 DOI: 10.1016/j.biomaterials.2018.04.039] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/14/2018] [Accepted: 04/18/2018] [Indexed: 01/04/2023]
Abstract
Tumor vaccine has been one of the research hotspots for cancer immunotherapy in recent years. By introducing tumor antigens into the body, the patient's own immune system will be specifically activated to induce effective immune responses for controlling or eliminating the malignant tumor cells. In this study, a simple nanovaccine was developed to induce antigen-specific anti-tumor immune responses. Polycationic polyethylenimine (PEI) was utilized to co-deliver the antigen ovalbumin (OVA) and the adjuvant unmethylated cytosine-phosphate-guanine (CpG) by electrostatic binding. The positively charged PEI could be beneficial to augment the PEI/CpG/OVA nanovaccine uptake in dendritic cells (DCs) and facilitate the endosomal escape of the nanovaccine for antigen delivering into the cytoplasm. The nanovaccine showed significant stimulation on DCs' maturation in vitro, and it was further applied for in vivo anti-tumor immunotherapy. To enhance the tumor infiltration of the nanovaccine-generated tumor-specific T cells, hyaluronidase (HAase) was employed to increase the permeability of the tumor tissues by breaking down the hyaluronan (HA) in the extracellular matrix (ECM) of tumors. Highly enhanced in vivo anti-tumor therapeutic efficiency was achieved by combining the PEI/CpG/OVA nanovaccine with HAase, which was attributed to the increased quantity of OVA-specific T cells in tumor tissues. The combination of nanovaccine with HAase has offered a simple and efficient strategy for inducing powerful anti-tumor effect in cancer immunotherapy.
Collapse
Affiliation(s)
- Xiuwen Guan
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Jie Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Yingying Hu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Lin Lin
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Pingjie Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Huayu Tian
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| |
Collapse
|
24
|
Abstract
OPINION STATEMENT Biliary tract cancers (BTCs) are rare aggressive neoplasms with a poor prognosis and a median survival of less than 1 year in the locally advanced or metastatic setting. Among the few patients who undergo curative resection the recurrence rates are high. About 90% of patients are detected at advanced stages, and systemic chemotherapy is the mainstay of their treatment. The treatment options for these patients are limited and multiple modalities of therapy from targeted therapy to immunotherapy and combination therapies (immunotherapy, targeted therapy, and chemotherapy) have been tested in this disease. Targeted therapies have failed to show a survival benefit. The deregulation of the immune system plays a significant role in the pathogenesis of BTCs. Therefore, immunotherapy, especially, immune checkpoint inhibitors hold great promise for this group of cancers. Numerous trials of immunotherapy in BTC are currently ongoing. In this review, we will discuss the available data and evidence for immunotherapy in BTC.
Collapse
Affiliation(s)
- Urvi A Shah
- Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, 2nd Floor, Bronx, NY, 10461, USA
| | - Amara G Nandikolla
- Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, 2nd Floor, Bronx, NY, 10461, USA
| | - Lakshmi Rajdev
- Department of Medical Oncology, Montefiore Medical Center, 1695 Eastchester Road, 2nd Floor, Bronx, NY, 10461, USA.
| |
Collapse
|
25
|
Wada S, Yada E, Ohtake J, Sasada T. Personalized peptide vaccines for cancer therapy: current progress and state of the art. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1403286] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Satoshi Wada
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| | - Erica Yada
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| | - Junya Ohtake
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| | - Tetsuro Sasada
- Cancer Immunotherapy, Kanagawa Cancer Center, Asahi-ku, Yokohama, Japan
| |
Collapse
|
26
|
Suekane S, Ueda K, Nishihara K, Sasada T, Yamashita T, Koga N, Yutani S, Shichijo S, Itoh K, Igawa T, Noguchi M. Personalized peptide vaccination as second-line treatment for metastatic upper tract urothelial carcinoma. Cancer Sci 2017; 108:2430-2437. [PMID: 28940789 PMCID: PMC5715265 DOI: 10.1111/cas.13404] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/06/2017] [Accepted: 09/15/2017] [Indexed: 01/02/2023] Open
Abstract
This study investigated the applicability of personalized peptide vaccination (PPV) for patients with metastatic upper tract urothelial cancer (mUTUC) after failure of platinum-based chemotherapy. In this single arm, open-label, phase II clinical trial, patients with mUTUC received PPV at a single institution. Personalized peptide vaccination treatment used a maximum of four peptides chosen from 27 candidate peptides according to human leukocyte antigen types and peptide-reactive IgG titers, for six s.c. injections weekly as one cycle. The safety of PPV, as well as its influence on host immunity and effect on overall survival were assessed. Forty-eight patients were enrolled in this study. Personalized peptide vaccinations were well tolerated without severe adverse events. Median survival time was 7.3 months (95% confidence interval [CI], 5.3-13.1) with 13.0 months for patients receiving combined salvage chemotherapy (95% CI, 5.7-17.5) and 4.5 months for patients receiving PPV alone (95% CI, 1.7-10.1) (P = 0.080). Patients with positive CTL responses showed a significantly longer survival than patients with negative CTL responses (hazard ratio, 0.37; 95% CI, 0.16-0.85; P = 0.019). Multivariate Cox regression analysis showed that lower numbers of Bellmunt risk factors and lower levels of B-cell activating factor were significantly associated with favorable overall survival for patients under PPV treatment. This study indicated that PPV for patients with mUTUC after failure of platinum-based chemotherapy induced substantial peptide-specific CTL responses without severe adverse events and has the potential to prolong survival when combined with salvage chemotherapy. UMIN Clinical Trials Registry ID: 000001854.
Collapse
Affiliation(s)
- Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume, Japan.,Kurume University Cancer Vaccine Center, Kurume, Japan
| | - Kousuke Ueda
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Kiyoaki Nishihara
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | | | - Takuto Yamashita
- Biostatics Center, Kurume University School of Medicine, Kurume, Japan
| | - Noriko Koga
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | | | | | - Kyogo Itoh
- Kurume University Cancer Vaccine Center, Kurume, Japan
| | - Tsukasa Igawa
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Masanori Noguchi
- Department of Urology, Kurume University School of Medicine, Kurume, Japan.,Kurume University Cancer Vaccine Center, Kurume, Japan.,Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
27
|
Koga N, Moriya F, Waki K, Yamada A, Itoh K, Noguchi M. Immunological efficacy of herbal medicines in prostate cancer patients treated by personalized peptide vaccine. Cancer Sci 2017; 108:2326-2332. [PMID: 28898532 PMCID: PMC5715291 DOI: 10.1111/cas.13397] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/04/2017] [Accepted: 09/07/2017] [Indexed: 12/20/2022] Open
Abstract
This randomized phase II study investigated the immunological efficacy of herbal medicines (HM) using Hochu‐ekki‐to and Keishi‐bukuryo‐gan in combination with personalized peptide vaccination (PPV) for castration‐resistant prostate cancer (CRPC). Seventy patients with CRPC were assigned to two arms; PPV plus HM or PPV alone. Two to four peptides were chosen from 31 peptides derived from cancer antigens for a s.c. injection of PPV given eight times according to the patient's human leukocyte antigen type and levels of antigen‐specific IgG titer before PPV treatment. Peptide‐specific CTL, IgG, regulatory T cells (Treg), monocytic myeloid‐derived suppressor cells (Mo‐MDSC), and interleukin‐6 (IL‐6) responses were measured before and at the eighth vaccination. Clinical outcomes were also analyzed. Combination therapy of PPV with HM was well tolerated without severe adverse events. There was no significant change in antigen‐specific IgG, CTL, Treg or clinical outcomes. Combination therapy of PPV with HM stabilized the frequency of Mo‐MDSC (1.91%–1.92%, P = 0.96) and serum levels of IL‐6 (19.2 pg/mL to 16.1 pg/mL, P = 0.63) during the treatment. In contrast, the frequency of Mo‐MDSC and levels of IL‐6 in the PPV‐alone group were significantly increased (0.91%–1.49% for Mo‐MDSC and 9.2 pg/mL to 19.4 pg/mL for IL‐6, respectively). These results suggest that the combined use of HM has the potential to prevent the immunosuppression induced by Mo‐MDSC or IL‐6 during immunotherapy. More research is needed to validate the findings of the present study.
Collapse
Affiliation(s)
- Noriko Koga
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Kayoko Waki
- Division of Cancer Vaccines, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Akira Yamada
- Division of Cancer Vaccines, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Masanori Noguchi
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan.,Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan.,Department of Urology, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
28
|
Xie J, Yang C, Liu Q, Li J, Liang R, Shen C, Zhang Y, Wang K, Liu L, Shezad K, Sullivan M, Xu Y, Shen G, Tao J, Zhu J, Zhang Z. Encapsulation of Hydrophilic and Hydrophobic Peptides into Hollow Mesoporous Silica Nanoparticles for Enhancement of Antitumor Immune Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2017; 13:1701741. [PMID: 28861951 DOI: 10.1002/smll.201701741] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/18/2017] [Indexed: 06/07/2023]
Abstract
Codelivery of combinational antigenic peptides and adjuvant to antigen presenting cells is expected to amplify tumor specific T lymphocytes immune responses while minimizing the possibility of tumor escaping and reducing immune tolerance to single antigenic peptide. However, the varied hydrophobicities of these multivariant derived short antigenic peptides limit their codelivery efficiency in conventional delivery systems. Here, a facile yet effective route is presented to generate monodisperse and stable hollow mesoporous silica nanoparticles (HMSNs) for codelivering of HGP10025-33 and TRP2180-188 , two melanoma-derived peptides with varied hydrophobicities. The HMSNs with large pore size can improve the encapsulation efficiency of both HGP100 and TRP2 after NH2 modification on the inner hollow core and COOH modification in the porous channels. HGP100 and TRP2 loaded HMSNs (HT@HMSNs) are further enveloped within monophosphoryl lipid A adjuvant entrapped lipid bilayer (HTM@HMLBs), for improved stability/biocompatibility and codelivery efficiency of multiple peptides, adjuvant, and enhanced antitumor immune responses. HTM@HMLBs increase uptake by dendritic cells (DCs) and stimulate DCs maturation efficiently, which further induce the activation of both tumor specific CD8+ and CD4+ T lymphocytes. Moreover, HTM@HMLBs can significantly inhibit tumor growth and lung metastasis in murine melanoma models with good safety profiles. HMSNs enveloped with lipid bilayers (HMLBs) are believed to be a promising platform for codelivery of multiple peptides, adjuvant, and enhancement of antitumor efficacy of conventional vaccinations.
Collapse
Affiliation(s)
- Jun Xie
- Tongji School of Pharmacy and National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology (HUST), Wuhan, 430030, China
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Chaohua Yang
- Tongji School of Pharmacy and National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology (HUST), Wuhan, 430030, China
| | - Qianqian Liu
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Jun Li
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Ruijing Liang
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Chen Shen
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Yi Zhang
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Ke Wang
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Liping Liu
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Khurram Shezad
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Martin Sullivan
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Yong Xu
- Department of Immunology, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Guanxin Shen
- Department of Immunology, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Juan Tao
- Department of Dermatology, Affiliated Union Hospital, Tongji Medical College, HUST, Wuhan, 430022, China
| | - Jintao Zhu
- School of Chemistry and Chemical Engineering, National Engineering Center for Nanomedicine, HUST, Wuhan, 430074, China
| | - Zhiping Zhang
- Tongji School of Pharmacy and National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology (HUST), Wuhan, 430030, China
| |
Collapse
|
29
|
Shen J, Wang LF, Zou ZY, Kong WW, Yan J, Meng FY, Chen FJ, Du J, Shao J, Xu QP, Ren HZ, Li RT, Wei J, Qian XP, Liu BR. Phase I clinical study of personalized peptide vaccination combined with radiotherapy for advanced hepatocellular carcinoma. World J Gastroenterol 2017; 23:5395-5404. [PMID: 28839440 PMCID: PMC5550789 DOI: 10.3748/wjg.v23.i29.5395] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/25/2017] [Accepted: 03/04/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the efficacy and safety of a new treatment modality, cellular immune therapy based on personalized peptide vaccination (PPV-DC-CTL) combined with radiotherapy, for treating advanced hepatocellular carcinoma (HCC). METHODS A total of nine patients with advanced HCC were enrolled. Multidisciplinary consultation confirmed that all the patients definitely had no opportunity of surgery, because four patients had multiple liver metastases (the number of liver lesions > 3), one patient had liver metastases and portal vein tumor thrombosis, one patient had lung and bone metastases, two patients had liver and lung metastases and one patient had liver metastasis and peritoneal metastasis. Patients with metastasis were treated with precise radiotherapy combined with PPV-DC-CTL. RESULTS Following radiotherapy and one to three cycles of PPV-DC-CTL treatment, AFP levels were significantly decreased in six patients and imaging assessment of the lesions showed a partial response (PR) in three patients and stable disease in the other three patients. The response rate was 33% and disease control rate was 66%. This regimen was found to be safe and well tolerated. None of the patients developed liver or kidney side effects. Only one patient developed grade II bone marrow suppression and the remaining patients had no significant hematological side effects. CONCLUSION Radiotherapy combined with PPV-DC-CTL provides a new therapeutic strategy for patients with advanced HCC, which is well tolerated, safe, feasible and effective.
Collapse
|
30
|
Further insight into AE37 peptide vaccination in prostate cancer. Future Sci OA 2017; 3:FSO192. [PMID: 28883993 PMCID: PMC5583690 DOI: 10.4155/fsoa-2017-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/24/2017] [Indexed: 11/17/2022] Open
|
31
|
Yutani S, Shirahama T, Muroya D, Matsueda S, Yamaguchi R, Morita M, Shichijo S, Yamada A, Sasada T, Itoh K. Feasibility study of personalized peptide vaccination for hepatocellular carcinoma patients refractory to locoregional therapies. Cancer Sci 2017. [PMID: 28622427 PMCID: PMC5581512 DOI: 10.1111/cas.13301] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Overall survival of patients with hepatocellular carcinoma (HCC) refractory to locoregional therapy is dismal, even following treatment with sorafenib, a multikinase inhibitor. To develop a more efficacious treatment, we undertook a feasibility study of personalized peptide vaccination (PPV) for HCC, in which the peptides were selected from 31 peptide candidates based on the pre‐existing immunity. Twenty‐six HCC patients refractory to locoregional therapies (cohort 1) and 30 patients refractory to both locoregional and systemic therapies (cohort 2) were entered into the study. There were no severe adverse events related to PPV except for one injection site reaction. At the end of the first cycle of six vaccinations, successful CTL or IgG boosting was observed in 57% or 46% of patients in cohort 1 and in 54% or 52% of patients in cohort 2, respectively. Successful IgG boosting at the end of the second cycle was observed in the majority of patients tested. Median overall survival was 18.7 months (95% confidence interval, 12.2–22.5 months) in cohort 1, and 8.5 months (95% confidence interval, 5.9–12.2 months) in cohort 2. Based on the higher rates of immune boosting and the safety profile of PPV, further clinical studies of PPV would be warranted for patients with HCC refractory to not only locoregional therapy but also both locoregional and systemic therapies. The protocol of this study was registered with the UMIN Clinical Trials Registry (UMIN000001882 and UMIN000003590).
Collapse
Affiliation(s)
| | - Takahisa Shirahama
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Daisuke Muroya
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | | | - Rin Yamaguchi
- Division of Pathology, Medical Center of Kurume University, Kurume, Japan
| | - Michi Morita
- Division of Pathology, Medical Center of Kurume University, Kurume, Japan
| | | | - Akira Yamada
- Cancer Vaccine Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Tetsuro Sasada
- Cancer Vaccine Center, Kanagawa Cancer Center, Yokohama, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Japan
| |
Collapse
|
32
|
Effects of Viral Peptide Presentation on CD4+ T Cell Responses to MHC Class II-Restricted Tumor Peptides. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2017. [DOI: 10.5812/ijcm.8174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
33
|
Abstract
Prostate cancer is the most common cancer in men, and the second leading cause of cancer-related death in Western countries. Prostate cancer-related death occurs in patients with metastatic castration-resistant prostate cancer. Although several new drugs for castration-resistant prostate cancer have been approved, each of these has prolonged survival by just a few months. Consequently, new therapies are sorely needed. Recently, it has been recognized that immunotherapy is an effective treatment for prostate cancer patients. Several strategies, such as cancer vaccines and immune checkpoint inhibitors, have been investigated in clinical studies for prostate cancer patients. In the present review, the results of the most recent clinical studies investigating immunotherapy in prostate cancer patients are reported, and the future clinical development of immunotherapy for prostate cancer is discussed.
Collapse
Affiliation(s)
- Masanori Noguchi
- Clinical Research Division, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan.,Department of Urology, Kurume University School of Medicine, Kurume, Japan.,Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Noriko Koga
- Clinical Research Division, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Tsukasa Igawa
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
34
|
Yu DP, Cheng X, Liu ZD, Xu SF. Comparative beneficiary effects of immunotherapy against chemotherapy in patients with advanced NSCLC: Meta-analysis and systematic review. Oncol Lett 2017; 14:1568-1580. [PMID: 28789381 PMCID: PMC5529907 DOI: 10.3892/ol.2017.6274] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/04/2017] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is the most commonly diagnosed cancer among men and it is the third ranked in women. There are two major types of lung cancer, namely, small cell lung cancer (SCLC), which accounts for ~20% of the cases, and non-small cell lung cancer (NSCLC), which is the most common. Chemotherapy and chemoradiotherapy have been used as the first-line therapies but suffer from lack of efficacy and also of several toxic adverse effects. Immunotherapeutic approaches including tumor antigen vaccination, monoclonal antibodies targeting checkpoint pathways and also activated immune cells are being developed and have been shown to be effective in treating NSCLC. Despite their promise, efficacy of several immunotherapies has not been consistent. We undertook this meta-analysis study to analyze results from clinical trials that compared efficacy and safety of immunotherapies with placebo or chemotherapy/radiotherapy in improving overall survival (OS) and progression-free survival (PFS) of NSCLC patients. Various databases were searched to identify randomized clinical studies examining the efficacy and safety of antibody- and vaccine-based immunotherapies in NSCLC patients in comparison to chemotherapy or chemoradiotherapy or placebo. Effects on OS and PFS and also adverse events have been compared. In accordance with the selection criteria, a total of 13 studies with 3,513 patients in immunotherapy and 3,072 patients in chemotherapy/placebo, were selected. PFS (odds ratio 1.81, 95% CI 1.36, 2.42; P<0.0001) and OS (P<0.0001) are found to be greatly improved by immunotherapies. Immunotherapy of NSCLC patients was also found to prevent several adverse effects and to improve daily living ability of the patients. The present meta-analysis strongly suggests that immunotherapy improves OS and PFS of patients with NSCLC.
Collapse
Affiliation(s)
- Da-Ping Yu
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Xu Cheng
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Zhi-Dong Liu
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| | - Shao-Fa Xu
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University, Beijing 101149, P.R. China
| |
Collapse
|
35
|
Sakamoto S, Matsueda S, Takamori S, Toh U, Noguchi M, Yutani S, Yamada A, Shichijo S, Yamada T, Suekane S, Kawano K, Naitou M, Sasada T, Hattori N, Kohno N, Itoh K. Immunological evaluation of peptide vaccination for cancer patients with the HLA -A11 + or -A33 + allele. Cancer Sci 2017; 108:598-603. [PMID: 28178396 PMCID: PMC5406587 DOI: 10.1111/cas.13189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 01/25/2017] [Accepted: 02/04/2017] [Indexed: 01/16/2023] Open
Abstract
The HLA‐A11 or ‐A33 allele is found in approximately 18% or 10% of the Asian population, respectively, but each of which is a minor allele worldwide, and therefore no clinical trials were previously conducted. To develop a therapeutic peptide vaccine for each of them, we investigated immunological responses of advanced cancer patients with the HLA‐A11+/A11+ (n = 18) or ‐A33+/A33+ (n = 13) allele to personalized peptide vaccine (PPV) regimens. The primary sites of HLA‐A11+/A11+ or ‐A33+/A33+ patients were the colon (n = 4 or 2), stomach (2 or 3), breast (3 or 2), lung and pancreas (2 or 2), and so on. For PPV, a maximum of four peptides were selected from nine different peptides capable of binding to HLA‐A11 and ‐A33 molecules based on the pre‐existing peptide‐specific IgG responses. There were no severe adverse events related to PPV. At the end of the first cycle, peptide‐specific CTL responses were augmented in 4/12 or 2/9 of HLA‐A11+/A11+ or ‐A33+/A33+ patients, while peptide‐specific IgG responses were augmented in 6/14 or 4/10 patients, respectively. Clinical responses consisted of four stable diseases and 14 progressive diseases in HLA‐A11+/A11+patients, versus seven and six in ‐A33+/A33+patients, respectively. Further clinical study of PPV could be recommended because of the safety and positive immunological responses.
Collapse
Affiliation(s)
- Shinjiro Sakamoto
- Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan.,Cancer Vaccine Center, Kurume University, Kurume, Japan.,Department of Molecular and Internal Medicine School of Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | - Shinzo Takamori
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Uhi Toh
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Masanori Noguchi
- Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | | | - Akira Yamada
- Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | | | - Teppei Yamada
- Department of Gastroenterological Surgery, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Kouichiro Kawano
- Department of Obstetrics and Gynecology, Kurume University School of Medicine, Kurume, Japan
| | | | - Tetsuro Sasada
- Cancer Vaccine Center, Kurume University, Kurume, Japan.,Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine School of Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Japan
| |
Collapse
|
36
|
Sakamoto S, Yamada T, Terazaki Y, Yoshiyama K, Sugawara S, Takamori S, Matsueda S, Shichijo S, Yamada A, Noguchi M, Itoh K, Hattori N, Kohno N, Sasada T. Feasibility Study of Personalized Peptide Vaccination for Advanced Small Cell Lung Cancer. Clin Lung Cancer 2017; 18:e385-e394. [PMID: 28416261 DOI: 10.1016/j.cllc.2017.03.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/15/2017] [Accepted: 03/15/2017] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The prognosis of patients with small cell lung cancer (SCLC) remains very poor. Therefore, the development of new therapeutic approaches, including immunotherapies, is desirable. PATIENTS AND METHODS We conducted a phase II study of personalized peptide vaccination (PPV), in which a maximum of 4 human leukocyte antigen-matched peptides were selected from 31 pooled peptides according to the pre-existing peptide-specific IgG responses before vaccination. The PPV was subcutaneously administered. RESULTS Forty-six patients were enrolled (median age, 63 years; 40 patients were men). Grade 1 (n = 13), 2 (n = 10), or 3 (n = 1) skin reactions at the injection sites were observed; however, no other severe adverse events related to the PPV were observed. The median survival time was 466, 397, 401, and 107 days in the subgroups with 0 (n = 5), 1 (n = 15), 2 (n = 12), and ≥ 3 (n = 14) previous chemotherapy regimens, respectively. Peptide-specific IgG responses to the vaccinated peptides were augmented in 70% and 95% of patients after 1 and 2 vaccination cycles, respectively. The overall survival (OS) of patients with augmented IgG responses to a greater number of nonvaccinated peptides after the second cycle of vaccination was significantly longer (median survival time, 1237 days vs. 382 days; P = .010). In addition, augmentation of IgG responses specific to 6 peptides, including Lck-derived peptides, was significantly related to better OS (P < .05, in each peptide). CONCLUSION These results suggest the feasibility of PPV for SCLC patients from the viewpoints of safety, immune boosting, and possible prolongation of OS. Therefore, further evaluation of PPV for advanced SCLC in prospective randomized trials is warranted.
Collapse
Affiliation(s)
- Shinjiro Sakamoto
- Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan; Cancer Vaccine Center, Kurume University, Kurume, Japan; Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Teppei Yamada
- Cancer Vaccine Center, Kurume University, Kurume, Japan; Department of Gastroenterological Surgery, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Yasuhiro Terazaki
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Koichi Yoshiyama
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Shinzo Takamori
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | | | | | - Akira Yamada
- Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Masanori Noguchi
- Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | - Tetsuro Sasada
- Cancer Vaccine Center, Kurume University, Kurume, Japan; Kanagawa Cancer Center Research Institute, Yokohama, Japan
| |
Collapse
|
37
|
Loi M, Desideri I, Greto D, Mangoni M, Sottili M, Meattini I, Becherini C, Terziani F, Delli Paoli C, Olmetto E, Bonomo P, Livi L. Radiotherapy in the age of cancer immunology: Current concepts and future developments. Crit Rev Oncol Hematol 2017; 112:1-10. [PMID: 28325250 DOI: 10.1016/j.critrevonc.2017.02.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/24/2016] [Accepted: 02/06/2017] [Indexed: 02/06/2023] Open
Abstract
Major advances in the knowledge of cancer biology and its interactions with tumor immune environment led to the emergence, in the last five years of new immunotherapy-based treatment strategies in cancer patients. At the same time, improvement in radiation technique and progress in radiobiology allowed in the last decade to expand the applications of radiotherapy in a growing number of settings. At present, there are strong theoretical basis to propose immune-enhanced radiation therapy that may represent in the future a new paradigm of treatment, combining the intrinsic power of radiotherapy to elicit a specific, systemic, tumor-directed immune response with modern highly conformal and precise dose delivery, in order to maximize response at the major site of disease and obtain durable disease control. The aim of this review is to describe the principal mechanisms of immune modulation of response to radiation and investigational strategies to harness the potential of radiation-inducible immune response: radiation therapy is expected to be not just a local treatment but the cornerstone of a multimodal strategy that might achieve long-lasting tumor remission at the primary site and systemic efficacy metastatic lesions.
Collapse
Affiliation(s)
- Mauro Loi
- Department of Radiation Oncology, University of Florence, Florence, Italy.
| | - Isacco Desideri
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Daniela Greto
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Monica Mangoni
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Mariangela Sottili
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Icro Meattini
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Carlotta Becherini
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Francesca Terziani
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | | | - Emanuela Olmetto
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Pierluigi Bonomo
- Department of Radiation Oncology, University of Florence, Florence, Italy
| | - Lorenzo Livi
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
38
|
Minami T, Matsumura N, Sugimoto K, Shimizu N, De Velasco M, Nozawa M, Yoshimura K, Harashima N, Harada M, Uemura H. Hypoxia-inducing factor (HIF)-1α-derived peptide capable of inducing cancer-reactive cytotoxic T lymphocytes from HLA-A24 + patients with renal cell carcinoma. Int Immunopharmacol 2017; 44:197-202. [PMID: 28110220 DOI: 10.1016/j.intimp.2017.01.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 12/12/2016] [Accepted: 01/10/2017] [Indexed: 10/20/2022]
Abstract
Hypoxic tumor microenvironment makes cancer cells to be therapy-resistant and hypoxia-inducing factors (HIFs) play a central role in hypoxic adaptation. Especially, renal cell carcinoma (RCC) is often associated with von Hippel-Lindau (VHL) gene mutations, leading to up-regulation of HIFs. However, from a different point of view, this suggests the possibility that HIFs could be promising targets in anti-cancer therapy. In this study, we searched for HIF-1α-derived peptides that are able to induce RCC-reactive cytotoxic T lymphocytes (CTLs) from HLA-A24+ RCC patients. Among five peptides derived from HIF-1α, which were prepared based on the binding motif to the HLA-A24 allele, a HIF-1α278-287 peptide induced peptide-specific CTLs from peripheral blood mononuclear cells of HLA-A24+ RCC patients most effectively. In immunoblot assays, the expression of HIF-1α was lowly detected in whole and nuclear lysates of RCC cell lines even under normoxia (20% O2), and their expression in whole lysates was increased under hypoxia (1% O2). Additionally, HIF-1α278-287 peptide-stimulated T cells showed a higher cytotoxicity against HLA-A24+ HIF-1α-expressing RCC cells than against HLA-A24- HIF-1α-expressing RCC cells. The cytotoxicity was inhibited by the addition of HIF-1α278-287 peptide-pulsed cold target cells. Altogether, these results indicate that the HIF-1α278-287 peptide could be a candidate for peptide-based anti-cancer vaccines for HLA-A24+ RCC patients.
Collapse
Affiliation(s)
- Takafumi Minami
- Department of Urology, Kindai University School of Medicine, Osaka, Japan.
| | - Naoki Matsumura
- Department of Urology, Kindai University School of Medicine, Osaka, Japan
| | - Koichi Sugimoto
- Department of Urology, Kindai University School of Medicine, Osaka, Japan
| | - Nobutaka Shimizu
- Department of Urology, Kindai University School of Medicine, Osaka, Japan
| | - Marco De Velasco
- Department of Urology, Kindai University School of Medicine, Osaka, Japan
| | - Masahiro Nozawa
- Department of Urology, Kindai University School of Medicine, Osaka, Japan
| | - Kazuhiro Yoshimura
- Department of Urology, Kindai University School of Medicine, Osaka, Japan
| | - Nanae Harashima
- Department of Immunology, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Mamoru Harada
- Department of Immunology, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kindai University School of Medicine, Osaka, Japan.
| |
Collapse
|
39
|
Voutsas IF, Anastasopoulou EA, Tzonis P, Papamichail M, Perez SA, Baxevanis CN. Unraveling the role of preexisting immunity in prostate cancer patients vaccinated with a HER-2/neu hybrid peptide. J Immunother Cancer 2016; 4:75. [PMID: 27891225 PMCID: PMC5109671 DOI: 10.1186/s40425-016-0183-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 10/27/2016] [Indexed: 01/09/2023] Open
Abstract
Background Cancer vaccines aim at eliciting not only an immune response against specific tumor antigens, but also at enhancing a preexisting immunity against the tumor. In this context, we recently reported on the levels of preexisting immunity in prostate cancer patients vaccinated with the HER-2 hybrid peptide (AE37), during a phase I clinical trial. The purpose of the current study was to correlate between preexisting immunity to the native HER-2 peptide, AE36, and expression of HLA-A2 and -A24 molecules with the clinical outcome. Additionally, we investigated the ability of the AE37 vaccine to induce an antitumor immune response against other tumor associated antigens, not integrated in the vaccine formulation, with respect to the clinical response. Methods We analyzed prostate cancer patients who were vaccinated with the AE37 vaccine [Ii-Key-HER-2/neu(776–790) hybrid peptide vaccine (AE37), which is a MHC class II long peptide vaccine encompassing MHC class I epitopes, during a phase I clinical trial. Preexisting immunity to the native HER-2/neu(776–790) (AE36) peptide was assessed by IFNγ response or dermal reaction at the inoculation site. Antigen specificity against other tumor antigens was defined using multimer analysis. Progression free survival (PFS) was considered as the patients’ clinical outcome. Two-tailed Wilcoxon signed rank test at 95 % confidence interval was used for statistical evaluation at different time points and Kaplan–Meier curves with log-rank (Mantel-Cox) test were used for the evaluation of PFS. Results Preexisting immunity to AE36, irrespectively of HLA expression, was correlated with longer PFS. Specific CD8+ T cell immunity against E75 and PSA146–151 (HLA-A2 restricted), as well as, PSA153–161 (HLA-A24 restricted) was detected at relatively high frequencies which were further enhanced during vaccinations. Specific immunity against PSA153–161 correlated with longer PFS in HLA-A24+ patients. However, HLA-A2+ patients with high preexisting or vaccine-induced immunity to E75, showed a trend for shorter PFS. Conclusions Our data cast doubt on whether preexisting immunity or epitope spreading specific for HLA-class I-restricted peptides can actually predict a favorable clinical outcome. They also impose that preexisting immunity to long vaccine peptides, encompassing both HLA class II and I epitopes should be considered as an important prerequisite for the improvement of future immunotherapeutic protocols. Protocol ID Code: Generex-06-07 National Organization for Medicines (EOF) ID Code: IS-107-01-06 NEC Study Code: EED107/1/06 EudraCT Number: 2006-003299-37 Date of registration: 07/06/2006 Date of enrolment of the first participant to the trial: Nov 1st, 2007 Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0183-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ioannis F Voutsas
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | | | - Panagiotis Tzonis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | - Michael Papamichail
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | - Sonia A Perez
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece
| | | |
Collapse
|
40
|
Sakamoto S, Yutani S, Shichijo S, Morita M, Yamada A, Itoh K, Noguchi M. Immunological evaluation of personalized peptide vaccination for patients with histologically unfavorable carcinoma of unknown primary site. Cancer Immunol Immunother 2016; 65:1223-31. [PMID: 27549314 PMCID: PMC11029246 DOI: 10.1007/s00262-016-1887-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 08/11/2016] [Indexed: 12/22/2022]
Abstract
The immunological characteristics of carcinoma of unknown primary site (CUP) are not well established due to inclusion of heterogeneous types of metastatic tumors with the absence of any detectable primary site. We evaluated the immune responses in patients with histologically unfavorable CUP during personalized peptide vaccination (PPV). Ten patients with histologically unfavorable CUP who had been treated by PPV after chemotherapy failure were analyzed. In PPV treatment, up to four human leukocyte antigen-matched peptides of a total 31 peptides were selected according to preexisting host immunity before vaccination and administered subcutaneously. Peptides derived from the Lck antigen were most often chosen for use among all patients. CTL responses were increased in 8 of the 10 and 5 of the five patients tested at the end of the first and second PPV cycles, respectively. Increases in humoral responses after vaccination, including IgG, IgG1, IgG3, IgA, and IgM, were observed against not only the vaccinated peptides but also the non-vaccinated peptides. Severe adverse events due to PPV were not observed. Median overall survival was 13.9 months (95 % CI 4.0-22.5 months). PPV activated both cellular and humoral immune responses to short peptides derived from CTL epitopes in the majority of CUP patients. PPV with Lck-derived peptides may be a feasible, new treatment modality for histologically unfavorable CUP patients due to its safety and strong ability to boost immune responses, although its clinical efficacy remains to be investigated in larger-scale trials.
Collapse
MESH Headings
- Adenocarcinoma/mortality
- Adenocarcinoma/pathology
- Adenocarcinoma/therapy
- Adult
- Aged
- Cancer Vaccines/immunology
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/therapy
- Cytotoxicity, Immunologic
- Drug Resistance, Neoplasm
- Epitopes, T-Lymphocyte/metabolism
- Female
- HLA Antigens/metabolism
- Humans
- Immunity, Humoral
- Immunotherapy/methods
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism
- Male
- Middle Aged
- Neoplasm Metastasis
- Neoplasms, Unknown Primary/mortality
- Neoplasms, Unknown Primary/pathology
- Neoplasms, Unknown Primary/therapy
- Peptide Fragments/metabolism
- Precision Medicine
- Survival Analysis
- T-Lymphocytes, Cytotoxic/immunology
- Vaccination
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Shinjiro Sakamoto
- Cancer Vaccine Center, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
- Clinical Research Division, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan
- Department of Molecular and Internal Medicine, Hiroshima University School of Medicine, Hiroshima, Japan
| | - Shigeru Yutani
- Cancer Vaccine Center, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Shigeki Shichijo
- Cancer Vaccine Center, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Michi Morita
- Department of Surgery, Nagasaki University Graduate Biomedical Science, Nagasaki, Japan
| | - Akira Yamada
- Cancer Vaccine Division, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Masanori Noguchi
- Cancer Vaccine Center, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan.
- Clinical Research Division, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, 830-0011, Japan.
| |
Collapse
|
41
|
Dammeijer F, Lievense LA, Veerman GDM, Hoogsteden HC, Hegmans JP, Arends LR, Aerts JG. Efficacy of Tumor Vaccines and Cellular Immunotherapies in Non-Small-Cell Lung Cancer: A Systematic Review and Meta-Analysis. J Clin Oncol 2016; 34:3204-12. [PMID: 27432922 DOI: 10.1200/jco.2015.66.3955] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Programmed cell death protein-1- checkpoint blockers have recently been approved as second-line treatment for advanced non-small-cell lung cancer (NSCLC). Unfortunately, only a subgroup of patients responds and shows long-term survival to these therapies. Tumor vaccines and cellular immunotherapies could synergize with checkpoint blockade, but which of these treatments is most efficacious is unknown. In this meta-analysis, we assessed the efficacy of tumor vaccination and cellular immunotherapy in NSCLC. METHODS We searched for randomized controlled trials (RCTs) investigating cellular immunotherapy or vaccines in NSCLC. We used random effects models to analyze overall survival (OS) and progression-free survival (PFS), expressed as hazard ratios (HRs), and differences in time (months). The effect of immunotherapy type, disease stage, tumor histology, and concurrent chemotherapy was assessed using subgroup analysis and meta-regression. All procedures were performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS We identified 18 RCTs that matched our selection criteria; these included a total of 6,756 patients. Immunotherapy extended NSCLC survival and PFS, expressed as HR (OS: HR, 0.81, 95% CI, 0.70 to 0.94, P = .01; PFS: HR, 0.83, 95% CI, 0.72 to 0.95, P = .006) and month difference (OS: difference, 5.43 months, 95% CI, 3.20 to 7.65, P < .005; PFS: difference, 3.24 months, 95% CI, 1.61 to 4.88, P < .005). Cellular therapies outperformed tumor vaccines (OS as HR: P = .005, month difference: P < .001; PFS as HR: P = .001, month difference: P = .004). There was a benefit of immunotherapy in low-stage compared with high-stage NSCLC and with concurrent administration of chemotherapy only in one of four outcome measures evaluated (PFS in months: P = .01 and PFS as HR: P = .031, respectively). There was no significant effect of tumor histology on survival or PFS. CONCLUSION Tumor vaccines and cellular immunotherapies enhanced OS and PFS in NSCLC. Cellular immunotherapy was found to be more effective than tumor vaccination. These findings have implications for future studies investigating combination immunotherapy in NSCLC.
Collapse
Affiliation(s)
- Floris Dammeijer
- Floris Dammeijer, Lysanne A. Lievense, G.D. Marijn Veerman, Henk C. Hoogsteden, Joost P. Hegmans, Joachim G. Aerts, and Lidia R. Arends, Erasmus Medical Center, Rotterdam, The Netherlands; and Joachim G. Aerts, Amphia Hospital, Breda, The Netherlands
| | - Lysanne A Lievense
- Floris Dammeijer, Lysanne A. Lievense, G.D. Marijn Veerman, Henk C. Hoogsteden, Joost P. Hegmans, Joachim G. Aerts, and Lidia R. Arends, Erasmus Medical Center, Rotterdam, The Netherlands; and Joachim G. Aerts, Amphia Hospital, Breda, The Netherlands
| | - G D Marijn Veerman
- Floris Dammeijer, Lysanne A. Lievense, G.D. Marijn Veerman, Henk C. Hoogsteden, Joost P. Hegmans, Joachim G. Aerts, and Lidia R. Arends, Erasmus Medical Center, Rotterdam, The Netherlands; and Joachim G. Aerts, Amphia Hospital, Breda, The Netherlands
| | - Henk C Hoogsteden
- Floris Dammeijer, Lysanne A. Lievense, G.D. Marijn Veerman, Henk C. Hoogsteden, Joost P. Hegmans, Joachim G. Aerts, and Lidia R. Arends, Erasmus Medical Center, Rotterdam, The Netherlands; and Joachim G. Aerts, Amphia Hospital, Breda, The Netherlands
| | - Joost P Hegmans
- Floris Dammeijer, Lysanne A. Lievense, G.D. Marijn Veerman, Henk C. Hoogsteden, Joost P. Hegmans, Joachim G. Aerts, and Lidia R. Arends, Erasmus Medical Center, Rotterdam, The Netherlands; and Joachim G. Aerts, Amphia Hospital, Breda, The Netherlands
| | - Lidia R Arends
- Floris Dammeijer, Lysanne A. Lievense, G.D. Marijn Veerman, Henk C. Hoogsteden, Joost P. Hegmans, Joachim G. Aerts, and Lidia R. Arends, Erasmus Medical Center, Rotterdam, The Netherlands; and Joachim G. Aerts, Amphia Hospital, Breda, The Netherlands
| | - Joachim G Aerts
- Floris Dammeijer, Lysanne A. Lievense, G.D. Marijn Veerman, Henk C. Hoogsteden, Joost P. Hegmans, Joachim G. Aerts, and Lidia R. Arends, Erasmus Medical Center, Rotterdam, The Netherlands; and Joachim G. Aerts, Amphia Hospital, Breda, The Netherlands.
| |
Collapse
|
42
|
Iwasa S, Yamada Y, Heike Y, Shoji H, Honma Y, Komatsu N, Matsueda S, Yamada A, Morita M, Yamaguchi R, Tanaka N, Kawahara A, Kage M, Shichijo S, Sasada T, Itoh K. Phase I study of a new cancer vaccine of ten mixed peptides for advanced cancer patients. Cancer Sci 2016; 107:590-600. [PMID: 26920496 PMCID: PMC4970826 DOI: 10.1111/cas.12919] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 02/05/2016] [Accepted: 02/21/2016] [Indexed: 12/22/2022] Open
Abstract
A phase I study of a new cancer vaccine (KRM‐10), consisting of a mixture of 10 different short peptides, was conducted for patients with advanced gastrointestinal cancers. Primary or secondary endpoints included the dose‐limiting toxicity (DLT), or safety and immune responses, respectively. Peptide‐specific cytotoxic T lymphocytes (CTL) and immunoglobulin G (IgG), together with soluble inflammatory factors, were measured before and after vaccination. Twenty‐one patients were vaccinated with KRM‐10 at dose levels of 10 (n = 6), 20 (n = 8) or 30 mg (n = 7) of peptides every week for 6 weeks. No DLT were observed in the dose range evaluated. Common treatment‐related adverse events were a grade 1 injection site reaction in 15 patients, and fever in three patients (grade 1 in two patients and grade 2 in one patient). CTL activity to at least one peptide at the time of the third and sixth vaccination increased in 2 and 3 of 6 (10 mg), 2 of 8 and 4 of 6 (20 mg), or 2 and 1 of 6 (30 mg) patients, respectively. IgG levels, at the third and sixth vaccination, were also increased in 1 and 1 of 6 (10 mg), 2 of 8 and 4 of 6 (20 mg), or 1 and 3 of 6 (30 mg) patients, respectively. The KRM‐10 vaccine consisting of 20 mg of peptides was determined as the optimal dose for a coming phase II trial because of its safety, and also for demonstrating the most potent activity for augmenting the immune response of the three doses tested. This trial was registered at the UMIN Clinical Trials Registry as UMIN000008820.
Collapse
Affiliation(s)
- Satoru Iwasa
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yasuhide Yamada
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yuji Heike
- Immunotherapy and Cell Therapy Department, St. Lucas International Hospital, Tokyo, Japan
| | - Hirokazu Shoji
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshitaka Honma
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - Nobukazu Komatsu
- Department of Immunology, Kurume University School of Medicine, Kurume, Japan
| | | | - Akira Yamada
- Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Michi Morita
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.,Division of Pathology, Medical Center of Kurume University, Kurume, Japan
| | - Rin Yamaguchi
- Division of Pathology, Medical Center of Kurume University, Kurume, Japan
| | - Natsuki Tanaka
- Department of Surgery, Kurume University School of Medicine, Kurume, Japan
| | - Akihiko Kawahara
- Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Japan
| | - Masayoshi Kage
- Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan.,Department of Diagnostic Pathology, Kurume University Hospital, Kurume, Japan
| | | | - Tetsuro Sasada
- Cancer Vaccine Center, Kanagawa Cancer Center Institute, Yokohama, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Japan
| |
Collapse
|
43
|
Noguchi M, Moriya F, Koga N, Matsueda S, Sasada T, Yamada A, Kakuma T, Itoh K. A randomized phase II clinical trial of personalized peptide vaccination with metronomic low-dose cyclophosphamide in patients with metastatic castration-resistant prostate cancer. Cancer Immunol Immunother 2016; 65:151-60. [PMID: 26728480 PMCID: PMC11028889 DOI: 10.1007/s00262-015-1781-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/11/2015] [Indexed: 01/21/2023]
Abstract
This study investigated the effect of metronomic cyclophosphamide (CPA) in combination with personalized peptide vaccination (PPV) on regulatory T cells (Treg) and myeloid-derived suppressor cells (MDSC), and whether it could improve the antitumor effect of PPV. Seventy patients with metastatic castration-resistant prostate cancer were randomly assigned (1:1) to receive PPV plus oral low-dose CPA (50 mg/day), or PPV alone. PPV treatment used a maximum of four peptides chosen from 31 pooled peptides according to human leukocyte antigen types and antigen-specific humoral immune responses before PPV, for 8 subcutaneous weekly injections. Peptide-specific cytotoxic T lymphocyte (CTL) and immunoglobulin G responses were measured before and after PPV. The incidence of grade 3 or 4 hematologic adverse events was higher in the PPV plus CPA arm than in the PPV alone arm. Decrease in Treg and increase in MDSC were more pronounced in PPV plus CPA treatment than in PPV alone (p = 0.036 and p = 0.048, respectively). There was no correlation between the changes in Treg or MDSC and CTL response. There was no difference in positive immune responses between the two arms, although overall survival in patients with positive immune responses was longer than in those with negative immune responses (p = 0.001). Significant differences in neither progression-free survival nor overall survival were observed between the two arms. Low-dose CPA showed no change in the antitumor effect of PPV, possibly due to the simultaneous decrease in Treg and increase in MDSC, in patients under PPV.
Collapse
MESH Headings
- Administration, Metronomic
- Aged
- Aged, 80 and over
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Cancer Vaccines/immunology
- Combined Modality Therapy
- Cyclophosphamide/administration & dosage
- Humans
- Male
- Middle Aged
- Neoplasm Grading
- Neoplasm Metastasis
- Peptides/administration & dosage
- Peptides/chemistry
- Peptides/immunology
- Precision Medicine/methods
- Prostatic Neoplasms, Castration-Resistant/immunology
- Prostatic Neoplasms, Castration-Resistant/mortality
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/therapy
- Survival Analysis
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Treatment Outcome
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/adverse effects
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Masanori Noguchi
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan.
- Department of Urology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan.
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan.
| | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Noriko Koga
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Satoko Matsueda
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Tetsuro Sasada
- Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
| | - Akira Yamada
- Division of Cancer Vaccines, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuyuki Kakuma
- Bio-statistics Center, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
44
|
Abstract
Biliary tract cancers (BTCs), which encompass intra- and extrahepatic cholangiocarcinomas as well as gallbladder carcinomas, are one of the most aggressive malignancies. Although the development of systemic chemotherapy approaches has made progress, the prognosis of BTC remains poor. Chronic inflammation plays an important role in the carcinogenesis of BTC, highlighting the immune etiology of this disease. Immunotherapy has emerged as a promising new modality of treatment for BTC. Here, we summarize the relevant tumor immunology of BTC and recently completed and ongoing clinical trials of immunotherapy for BTC.
Collapse
Affiliation(s)
- Yi Chai
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
45
|
Noguchi M, Koga N, Moriya F, Itoh K. Immunotherapy in prostate cancer: challenges and opportunities. Immunotherapy 2016; 8:69-77. [DOI: 10.2217/imt.15.101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Although treatment options for castration-resistant prostate cancer (CRPC) have increased over the last decade, there remains a need for strategies that can provide durable disease control and long-term benefit. Recently, immunotherapy has emerged as a viable and attractive strategy for the treatment of CRPC. To date, there are multiple strategies to target the immune system, and several approaches including therapeutic cancer vaccines and immune checkpoint inhibitors have been most successful in clinical trials. With regard to this, we report the results of the most recent clinical trials investigating immunotherapy in CRPC and discuss the future development of immunotherapy for CRPC, as well as the potential importance of biomarkers in the future progress of this field.
Collapse
Affiliation(s)
- Masanori Noguchi
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | - Noriko Koga
- Division of Clinical Research, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011, Japan
| | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
46
|
Araki H, Pang X, Komatsu N, Soejima M, Miyata N, Takaki M, Muta S, Sasada T, Noguchi M, Koda Y, Itoh K, Kuhara S, Tashiro K. Haptoglobin promoter polymorphism rs5472 as a prognostic biomarker for peptide vaccine efficacy in castration-resistant prostate cancer patients. Cancer Immunol Immunother 2015; 64:1565-73. [PMID: 26428930 PMCID: PMC11028849 DOI: 10.1007/s00262-015-1756-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 09/07/2015] [Indexed: 01/16/2023]
Abstract
Personalized peptide vaccination (PPV) is an attractive approach to cancer immunotherapy with strong immune-boosting effects conferring significant clinical benefit. However, as with most therapeutic agents, there is a difference in clinical efficacy among patients receiving PPV. Therefore, a useful biomarker is urgently needed for prognosticating clinical outcomes to preselect patients who would benefit the most from PPV. In this retrospective study, to detect a molecular prognosticator of clinical outcomes for PPV, we analyzed whole-genome gene expression profiles of peripheral blood mononuclear cells (PBMCs) in castration-resistant prostate cancer (CRPC) patients before administration of PPV. Cox regression analysis revealed that mRNA expression of myeloperoxidase, haptoglobin, and neutrophil elastase was significantly associated with overall survival (OS) among vaccinated CRPC patients (adjusted P < 0.01). By promoter sequence analysis of these three genes, we found that rs5472 of haptoglobin (HP), an acute-phase plasma glycoprotein, was strongly correlated to OS of vaccinated CRPC patients (P = 0.0047, hazard ratio 0.47; 95 % confidence interval 0.28-0.80). Furthermore, both HP mRNA expression in PBMCs and protein level in plasma of CRPC patients before administration of PPV exhibited rs5472 dependence (P < 0.001 for mRNA expression and P < 0.05 for protein level). Our findings suggest that rs5472 may play an important role in the immune response to PPV via regulation of HP. Thus, we concluded that rs5472 is a potential prognostic biomarker for PPV.
Collapse
Affiliation(s)
- Hiromitsu Araki
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 6-10-1 Hakozaki Higashi-Ku, Fukuoka, 812-8581, Japan
| | - Xiaoliang Pang
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 6-10-1 Hakozaki Higashi-Ku, Fukuoka, 812-8581, Japan
| | - Nobukazu Komatsu
- Department of Immunology and Immunotherapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Mikiko Soejima
- Department of Forensic Medicine and Human Genetics, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Nawoe Miyata
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 6-10-1 Hakozaki Higashi-Ku, Fukuoka, 812-8581, Japan
| | - Mari Takaki
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 6-10-1 Hakozaki Higashi-Ku, Fukuoka, 812-8581, Japan
| | - Shigeru Muta
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 6-10-1 Hakozaki Higashi-Ku, Fukuoka, 812-8581, Japan
| | - Tetsuro Sasada
- Department of Immunology and Immunotherapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Masanori Noguchi
- Clinical Research Division of Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Yoshiro Koda
- Department of Forensic Medicine and Human Genetics, Kurume University School of Medicine, 67 Asahi-machi, Kurume, 830-0011, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Japan
| | - Satoru Kuhara
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 6-10-1 Hakozaki Higashi-Ku, Fukuoka, 812-8581, Japan
| | - Kosuke Tashiro
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, 6-10-1 Hakozaki Higashi-Ku, Fukuoka, 812-8581, Japan.
| |
Collapse
|
47
|
Noguchi M, Matsumoto K, Uemura H, Arai G, Eto M, Naito S, Ohyama C, Nasu Y, Tanaka M, Moriya F, Suekane S, Matsueda S, Komatsu N, Sasada T, Yamada A, Kakuma T, Itoh K. An Open-Label, Randomized Phase II Trial of Personalized Peptide Vaccination in Patients with Bladder Cancer that Progressed after Platinum-Based Chemotherapy. Clin Cancer Res 2015; 22:54-60. [PMID: 26581246 DOI: 10.1158/1078-0432.ccr-15-1265] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/18/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE The prognosis of platinum-based chemotherapy-resistant metastatic urothelial cancer of the bladder remains poor. Personalized selection of the right peptides for each patient could be a novel approach for a cancer vaccine to boost anticancer immunity. EXPERIMENTAL DESIGN In this randomized, open-label, phase II study, patients ages ≥18 years with progressive bladder cancer after first-line platinum-based chemotherapy were randomly assigned (1:1) to receive personalized peptide vaccination (PPV) plus best supportive care (BSC) or BSC. PPV treatment used a maximum of four peptides chosen from 31 candidate peptides according to human leukocyte antigen types and peptide-reactive IgG titers, for 12 s.c. injections (8 injections, weekly; 4 injections, bi-weekly). The primary endpoint was progression-free survival (PFS). Secondary endpoints were overall survival (OS), immune response, and toxicity. RESULTS Eighty patients were randomly assigned to receive either PPV plus BSC (n = 39) or BSC (n = 41). No significant improvement in PFS was noted [HR, 0.7; 95% confidence interval (CI), 0.4-1.2, P = 0.17]. For the secondary endpoints, PPV plus BSC significantly prolonged OS compared with BSC (HR, 0.58; 95% CI, 0.34-0.99, P = 0.049), with median OS of 7.9 months (95% CI, 3.5-12.0) in the PPV plus BSC and 4.1 months (95% CI, 2.8-6.9) in the BSC. PPV treatment was well tolerated, without serious adverse drug reactions. CONCLUSIONS PPV could not prolong PFS, but OS appeared to be improved with low toxicity and immune responses. Further large-scale, randomized trials are needed to confirm these results.
Collapse
Affiliation(s)
- Masanori Noguchi
- Divisions of Clinical Research, Kurume University School of Medicine, Kurume, Japan. Department of Urology, Kurume University School of Medicine, Kurume, Japan.
| | - Kazumasa Matsumoto
- Department of Urology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kinki University Faculty of Medicine, Osaka, Japan
| | - Gaku Arai
- Department of Urology, Dokkyo Medical University Koshigaya Hospital, Koshigaya, Japan
| | - Masatoshi Eto
- Department of Urology, Kumamoto University, Kumamoto, Japan
| | - Seiji Naito
- Department of Urology, Graduate School of Medical Sciences, University of Kyushu, Fukuoka, Japan
| | - Chikara Ohyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yasutomo Nasu
- Department of Urology, Okayama University Graduate School of Medicine, Okayama, Japan
| | | | - Fukuko Moriya
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Shigetaka Suekane
- Department of Urology, Kurume University School of Medicine, Kurume, Japan
| | - Satoko Matsueda
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| | | | - Tetsuro Sasada
- Department of Immunology, Kurume University School of Medicine, Kurume, Japan
| | - Akira Yamada
- Cancer Vaccines, Research Center for Innovative Cancer Therapy, Kurume University School of Medicine, Kurume, Japan
| | - Tatsuyuki Kakuma
- Bio-Statistics Center, Kurume University School of Medicine, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
48
|
Marks EI, Yee NS. Immunotherapeutic approaches in biliary tract carcinoma: Current status and emerging strategies. World J Gastrointest Oncol 2015; 7:338-346. [PMID: 26600933 PMCID: PMC4644856 DOI: 10.4251/wjgo.v7.i11.338] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/17/2015] [Accepted: 09/16/2015] [Indexed: 02/05/2023] Open
Abstract
For biliary tract carcinoma (BTC), complete surgical resection of tumor is only feasible in a minority of patients, and the treatment options for patients with unresectable or metastatic disease are limited. Advances in cancer immunology have led to identification of tumor-infiltrating immune cells as indicators of prognosis and response to treatment in BTC. This has also facilitated development of immunotherapy that focuses on enhancing the immune system against biliary tumors. This includes peptide- and dendritic cell-based vaccines that stimulate in-vivo immune responses against tumor-specific antigens. Adoptive immunotherapy, which entails the ex-vivo expansion of tumor-infiltrating immune cells for subsequent reintroduction, and cytokine-based therapies have been developed in BTC. Clinical studies indicate that this type of therapy is generally well tolerated. Combination therapy with dendritic cell-based vaccines and adoptive immunotherapy has shown particularly good potential. Emerging strategies through discovery of novel antigen targets and by reversal of tumor-associated immunosuppression are expected to improve the efficacy of immunotherapy in BTC. Collaborative efforts by integration of targeted immunotherapeutics with molecular profiling of biliary tumor will hopefully make a positive impact on advancing towards the goal of developing precision treatment of patients with this highly lethal disease.
Collapse
|
49
|
Strategies and Advancements in Harnessing the Immune System for Gastric Cancer Immunotherapy. J Immunol Res 2015; 2015:308574. [PMID: 26579545 PMCID: PMC4633567 DOI: 10.1155/2015/308574] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/05/2015] [Indexed: 12/12/2022] Open
Abstract
In cancer biology, cells and molecules that form the fundamental components of the tumor microenvironment play a major role in tumor initiation, and progression as well as responses to therapy. Therapeutic approaches that would enable and harness the immune system to target tumor cells mark the future of anticancer therapy as it could induce an immunological memory specific to the tumor type and further enhance tumor regression and relapse-free survival in cancer patients. Gastric cancer is one of the leading causes of cancer-related mortalities that has a modest survival benefit from existing treatment options. The advent of immunotherapy presents us with new approaches in gastric cancer treatment where adaptive cell therapies, cancer vaccines, and antibody therapies have all been used with promising outcomes. In this paper, we review the current advances and prospects in the gastric cancer immunotherapy. Special focus is laid on new strategies and clinical trials that attempt to enhance the efficacy of various immunotherapeutic modalities in gastric cancer.
Collapse
|
50
|
Matsueda S, Shichijo S, Nagata S, Seki C, Yamada A, Noguchi M, Itoh K. Identification of novel Lck-derived T helper epitope long peptides applicable for HLA-A2(+) cancer patients as cancer vaccine. Cancer Sci 2015; 106:1493-8. [PMID: 26331453 PMCID: PMC4714684 DOI: 10.1111/cas.12805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/20/2015] [Accepted: 08/27/2015] [Indexed: 01/04/2023] Open
Abstract
The present study attempted to identify T helper epitope long peptides capable of inducing cytotoxic T lymphocytes (CTL) from Lck antigen (p56(Lck) ), the src family tyrosine kinase, which is known to be aberrantly expressed in metastatic cancers cells, in order to develop a long peptide-based cancer vaccine for HLA-A2(+) cancer patients. Based on the biding motif to the HLA-DR and HLA-A2 alleles, 94 peptides were prepared from the Lck antigen. These peptides were screened for their reactivity to immunoglobulin G (IgG) from plasma of cancer patients, followed by testing of their ability to induce both CD4(+) and CD8(+) T lymphocytes showing not only peptide-specific IFN-γ production but cytotoxicity against HLA-A2(+) cancer cells from peripheral blood mononuclear cells (PBMC) of HLA-A2(+) cancer patients. Among 94 peptides tested, the three T helper epitope long peptides and their inner CTL epitope short peptides with HLA-A2 binding motifs were frequently recognized by IgG of cancer patients, and efficiently induced both CD4(+) IFN-γ(+) and CD8(+) IFN-γ(+) T lymphocytes. Patients' PBMC stimulated with these long peptides showed cytotoxicity against HLA-A2(+) Lck(+) cancer cells in HLA-class I and HLA-class II dependent manners. These three peptides might be useful for long peptide-based vaccines for HLA-A2(+) cancer patients with Lck(+) tumor cells.
Collapse
Affiliation(s)
| | | | - Sayaka Nagata
- Cancer Vaccine Center, Kurume University, Kurume, Japan
| | - Chieko Seki
- Cancer Vaccine Center, Kurume University, Kurume, Japan
| | - Akira Yamada
- Cancer Vaccine Development Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Masanori Noguchi
- Cancer Vaccine Center, Kurume University, Kurume, Japan.,Clinical Research Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Kyogo Itoh
- Cancer Vaccine Center, Kurume University, Kurume, Japan
| |
Collapse
|