1
|
Jamal A, Aldreiwish AD, Banawas SS, Alqurashi YE, Kamal MA, Ahmad F. The paths toward immunotherapy of esophageal cancer: An overview of clinical trials. Int Immunopharmacol 2025; 151:114261. [PMID: 40015204 DOI: 10.1016/j.intimp.2025.114261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
As the seventh-leading contributor to global cancer-related deaths, esophageal cancer (EC) is one of the most challenging types of cancer. Despite advancements in conventional therapies, including surgery, chemotherapy, and radiotherapy, the five-year survival rate remains low, underscoring the need for the development of more efficacious treatment approaches. Immunotherapy has emerged as a promising treatment approach, offering new hope for EC patients. This review provides an in-depth examination of the latest immunotherapeutic strategies for EC, focusing on immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and oncolytic virotherapy. We critically analyze the current clinical data to highlight the progress and pitfalls of each immunotherapeutic approach for EC. Additionally, we explore the potential for combination therapies, which could overcome the resistance often seen with monotherapies. Finally, we discuss the limitations of current treatments and outline key areas for future research to improve patient outcomes and survival.
Collapse
Affiliation(s)
- Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Allolo D Aldreiwish
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Saeed S Banawas
- Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Yaser E Alqurashi
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh 13713, Saudi Arabia
| |
Collapse
|
2
|
Suzuki N, Shindo Y, Nakajima M, Tsunedomi R, Nagano H. Current status of vaccine immunotherapy for gastrointestinal cancers. Surg Today 2024; 54:1279-1291. [PMID: 38043066 DOI: 10.1007/s00595-023-02773-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/14/2023] [Indexed: 12/05/2023]
Abstract
Recent advances in tumor immunology and molecular drug development have ushered in a new era of cancer immunotherapy. Immunotherapy has shown promising results for several types of tumors, such as advanced melanoma, non-small cell lung cancer, renal cell carcinoma, bladder cancers, and refractory Hodgkin's lymphoma. Similarly, efforts have been made to develop immunotherapies such as adoptive T-cell transplantation, peptide vaccines, and dendritic cell vaccines, specifically for gastrointestinal tumors. However, before the advent of immune checkpoint inhibitors, immunotherapy did not work as well as expected. In this article, we review immunotherapy, focusing on cancer vaccines for gastrointestinal tumors, which generally target eliciting tumor-specific CD8 + cytotoxic T lymphocytes (CTLs). We also review various vaccine therapies and describe the relationship between vaccines and adjuvants. Finally, we discuss prospects for the combination of immunotherapy with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Nobuaki Suzuki
- Department of Gastroenterological, Breast, and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yoshitaro Shindo
- Department of Gastroenterological, Breast, and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Masao Nakajima
- Department of Gastroenterological, Breast, and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Ryouichi Tsunedomi
- Department of Gastroenterological, Breast, and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast, and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|
3
|
Makino T, Miyata H, Yasuda T, Kitagawa Y, Muro K, Park JH, Hikichi T, Hasegawa T, Igarashi K, Iguchi M, Masaoka Y, Yano M, Doki Y. A phase 3, randomized, double-blind, multicenter, placebo-controlled study of S-588410, a five-peptide cancer vaccine as an adjuvant therapy after curative resection in patients with esophageal squamous cell carcinoma. Esophagus 2024; 21:447-455. [PMID: 38990441 PMCID: PMC11405444 DOI: 10.1007/s10388-024-01072-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND S-588410, a cancer peptide vaccine (CPV), comprises five HLA-A*24:02-restricted peptides from five cancer-testis antigens. In a phase 2 study, S-588410 was well-tolerated and exhibited antitumor efficacy in patients with urothelial cancer. Therefore, we aimed to evaluate the efficacy, immune response, and safety of S-588410 in patients with completely resected esophageal squamous cell carcinoma (ESCC). METHODS This phase 3 study involved patients with HLA-A*24:02-positive and lymph node metastasis-positive ESCC who received neoadjuvant therapy followed by curative resection. After randomization, patients were administered S-588410 and placebo (both emulsified with Montanide™ ISA 51VG) subcutaneously. The primary endpoint was relapse-free survival (RFS). The secondary endpoints were overall survival (OS), cytotoxic T-lymphocyte (CTL) induction, and safety. Statistical significance was tested using the one-sided weighted log-rank test with the Fleming-Harrington class of weights. RESULTS A total of 276 patients were randomized (N = 138/group). The median RFS was 84.3 and 84.1 weeks in the S-588410 and placebo groups, respectively (P = 0.8156), whereas the median OS was 236.3 weeks and not reached, respectively (P = 0.6533). CTL induction was observed in 132/134 (98.5%) patients who received S-588410 within 12 weeks. Injection site reactions (137/140 patients [97.9%]) were the most frequent treatment-emergent adverse events in the S-588410 group. Prolonged survival was observed in S-588410-treated patients with upper thoracic ESCC, grade 3 injection-site reactions, or high CTL intensity. CONCLUSIONS S-588410 induced immune response and had acceptable safety but failed to reach the primary endpoint. A high CTL induction rate and intensity may be critical for prolonging survival during future CPV development.
Collapse
Affiliation(s)
- Tomoki Makino
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroshi Miyata
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
| | - Takushi Yasuda
- Department of Surgery, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, 589-8511, Japan.
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Jae-Hyun Park
- OncoTherapy Science, Inc., Kawasaki, Kanagawa, Japan
| | - Tetsuro Hikichi
- Laboratory Department, Cancer Precision Medicine, Inc., Kawasaki, Kanagawa, Japan
| | | | | | - Motofumi Iguchi
- Medical Affairs Department, Shionogi & Co., Ltd, Osaka, Japan
| | | | - Masahiko Yano
- Department of Gastroenterological Surgery, Osaka International Cancer Institute, Osaka, Japan
- Kyowakai Hospital, Osaka, Japan
| | - Yuichiro Doki
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
4
|
Abou Kors T, Meier M, Mühlenbruch L, Betzler AC, Oliveri F, Bens M, Thomas J, Kraus JM, Doescher J, von Witzleben A, Hofmann L, Ezic J, Huber D, Benckendorff J, Barth TFE, Greve J, Schuler PJ, Brunner C, Blackburn JM, Hoffmann TK, Ottensmeier C, Kestler HA, Rammensee HG, Walz JS, Laban S. Multi-omics analysis of overexpressed tumor-associated proteins: gene expression, immunopeptide presentation, and antibody response in oropharyngeal squamous cell carcinoma, with a focus on cancer-testis antigens. Front Immunol 2024; 15:1408173. [PMID: 39136024 PMCID: PMC11317303 DOI: 10.3389/fimmu.2024.1408173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Introduction The human leukocyte antigen complex (HLA) is essential for inducing specific immune responses to cancer by presenting tumor-associated peptides (TAP) to T cells. Overexpressed tumor associated antigens, mainly cancer-testis antigens (CTA), are outlined as essential targets for immunotherapy in oropharyngeal squamous cell carcinoma (OPSCC). This study assessed the degree to which presentation, gene expression, and antibody response (AR) of TAP, mainly CTA, are correlated in OPSCC patients to evaluate their potential as immunotherapy targets. Materials and methods Snap-frozen tumor (NLigand/RNA=40), healthy mucosa (NRNA=6), and healthy tonsils (NLigand=5) samples were obtained. RNA-Seq was performed using Illumina HiSeq 2500/NovaSeq 6000 and whole exome sequencing (WES) utilizing NextSeq500. HLA ligands were isolated from tumor tissue using immunoaffinity purification, UHPLC, and analyzed by tandem MS. Antibodies were measured in serum (NAb=27) utilizing the KREX™ CT262 protein array. Data analysis focused on 312 proteins (KREX™ CT262 panel + overexpressed self-proteins). Results 183 and 94 of HLA class I and II TAP were identified by comparative profiling with healthy tonsils. Genes from 26 TAP were overexpressed in tumors compared to healthy mucosa (LFC>1; FDR<0.05). Low concordance (r=0.25; p<0.0001) was found between upregulated mRNA and class I TAP. The specific mode of correlation of TAP was found to be dependent on clinical parameters. A lack of correlation was observed both between mRNA and class II TAP, as well as between class II tumor-unique TAP (TAP-U) presentation and antibody response (AR) levels. Discussion This study demonstrates that focusing exclusively on gene transcript levels fails to capture the full extent of TAP presentation in OPSCC. Furthermore, our findings reveal that although CTA are presented at relatively low levels, a few CTA TAP-U show potential as targets for immunotherapy.
Collapse
Affiliation(s)
- Tsima Abou Kors
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Matthias Meier
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Lena Mühlenbruch
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Eberhard Karls University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany
| | - Annika C. Betzler
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Core Facility Immune Monitoring, Ulm University Medical Center, Ulm, Germany
| | - Franziska Oliveri
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Martin Bens
- Core Facility Next Generation Sequencing, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Jaya Thomas
- Cancer Sciences Unit, University of Southampton, Faculty of Medicine, Southampton, United Kingdom
| | - Johann M. Kraus
- Institute of Medical Systems Biology, Faculty of Medicine, Ulm University, Ulm, Germany
| | - Johannes Doescher
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Department of Otolaryngology, Augsburg University Hospital, Augsburg, Germany
| | - Adrian von Witzleben
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Linda Hofmann
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Jasmin Ezic
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Diana Huber
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | | | | | - Jens Greve
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
| | - Patrick J. Schuler
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| | - Cornelia Brunner
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Core Facility Immune Monitoring, Ulm University Medical Center, Ulm, Germany
| | - Jonathan M. Blackburn
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Thomas K. Hoffmann
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| | - Christian Ottensmeier
- Institute of Systems, Molecular and Integrative Biology, Liverpool Head and Neck Center, University of Liverpool, Faculty of Medicine, Liverpool, United Kingdom
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Faculty of Medicine, Ulm University, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| | - Hans-Georg Rammensee
- Department of Immunology, Institute for Cell Biology, Eberhard Karls University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany
| | - Juliane S. Walz
- Department of Peptide-based Immunotherapy, Eberhard Karls University and University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK), Partner Site Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Simon Laban
- Department of Otorhinolaryngology and Head and Neck Surgery, Ulm University Medical Center, Ulm, Germany
- Surgical Oncology Ulm, i2SOUL Consortium, Ulm, Germany
| |
Collapse
|
5
|
Tamura R, Yamanobe Y, Fujioka M, Morimoto Y, Fukumura M, Nakaya M, Oishi Y, Sato M, Ueda R, Fujiwara H, Hikichi T, Noji S, Oishi N, Ozawa H, Ogawa K, Kawakami Y, Ohira T, Yoshida K, Toda M. Phase I/II Study of a Vascular Endothelial Growth Factor Receptor Vaccine in Patients With NF2-Related Schwannomatosis. J Clin Oncol 2024; 42:2578-2587. [PMID: 38776485 DOI: 10.1200/jco.23.02376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/15/2024] [Accepted: 03/21/2024] [Indexed: 05/25/2024] Open
Abstract
PURPOSE The humanized antivascular endothelial growth factor (VEGF) antibody bevacizumab (Bev) is efficacious for the treatment of NF2-related schwannomatosis (NF2), previously known as neurofibromatosis type 2. This study evaluated the safety and efficacy of a VEGF receptor (VEGFR) vaccine containing VEGFR1 and VEGFR2 peptides in patients with NF2 with progressive schwannomas (jRCTs031180184). MATERIALS AND METHODS VEGFR1 and VEGFR2 peptides were injected subcutaneously into infra-axillary and inguinal regions, once a week for 4 weeks and then once a month for 4 months. The primary end point was safety. Secondary end points included tolerability, hearing response, imaging response, and immunologic response. RESULTS Sixteen patients with NF2 with progressive schwannomas completed treatment and were assessed. No severe vaccine-related adverse events occurred. Among the 13 patients with assessable hearing, word recognition score improved in five patients at 6 months and two at 12 months. Progression of average hearing level of pure tone was 0.168 dB/mo during the year of treatment period, whereas long-term progression was 0.364 dB/mo. Among all 16 patients, a partial response was observed in more than one schwannoma in four (including one in which Bev had not been effective), minor response in 5, and stable disease in 4. Both VEGFR1-specific and VEGFR2-specific cytotoxic T lymphocytes (CTLs) were induced in 11 patients. Two years after vaccination, a radiologic response was achieved in nine of 20 assessable schwannomas. CONCLUSION This study demonstrated the safety and preliminary efficacy of VEGFR peptide vaccination in patients with NF2. Memory-induced CTLs after VEGFR vaccination may persistently suppress tumor progression.
Collapse
Affiliation(s)
- Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Yoshiharu Yamanobe
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masato Fujioka
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
- Clinical and Translational Research Center, Keio University School of Medicine, Tokyo, Japan
- Department of Molecular Genetics, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yukina Morimoto
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Mariko Fukumura
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Masato Nakaya
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Yumiko Oishi
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Mizuto Sato
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Ueda
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Hirokazu Fujiwara
- Department of Radiology, Keio University School of Medicine, Tokyo, Japan
| | | | - Shinobu Noji
- Division of Cellular Signaling Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, Tokyo, Japan
| | - Naoki Oishi
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroyuki Ozawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kaoru Ogawa
- Department of Otorhinolaryngology, Head and Neck Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kawakami
- Division of Cellular Signaling Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Department of Immunology, International University of Health and Welfare School of Medicine, Narita, Japan
| | - Takayuki Ohira
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
Zhang W, Sun S, Zhu W, Meng D, Hu W, Yang S, Gao M, Yao P, Wang Y, Wang Q, Ji J. Birinapant Reshapes the Tumor Immunopeptidome and Enhances Antigen Presentation. Int J Mol Sci 2024; 25:3660. [PMID: 38612472 PMCID: PMC11011986 DOI: 10.3390/ijms25073660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Birinapant, an antagonist of the inhibitor of apoptosis proteins, upregulates MHCs in tumor cells and displays a better tumoricidal effect when used in combination with immune checkpoint inhibitors, indicating that Birinapant may affect the antigen presentation pathway; however, the mechanism remains elusive. Based on high-resolution mass spectrometry and in vitro and in vivo models, we adopted integrated genomics, proteomics, and immunopeptidomics strategies to study the mechanism underlying the regulation of tumor immunity by Birinapant from the perspective of antigen presentation. Firstly, in HT29 and MCF7 cells, Birinapant increased the number and abundance of immunopeptides and source proteins. Secondly, a greater number of cancer/testis antigen peptides with increased abundance and more neoantigens were identified following Birinapant treatment. Moreover, we demonstrate the existence and immunogenicity of a neoantigen derived from insertion/deletion mutation. Thirdly, in HT29 cell-derived xenograft models, Birinapant administration also reshaped the immunopeptidome, and the tumor exhibited better immunogenicity. These data suggest that Birinapant can reshape the tumor immunopeptidome with respect to quality and quantity, which improves the presentation of CTA peptides and neoantigens, thus enhancing the immunogenicity of tumor cells. Such changes may be vital to the effectiveness of combination therapy, which can be further transferred to the clinic or aid in the development of new immunotherapeutic strategies to improve the anti-tumor immune response.
Collapse
Affiliation(s)
- Weiyan Zhang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Shenghuan Sun
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA 94143, USA;
| | - Wenyuan Zhu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Delan Meng
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Weiyi Hu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Siqi Yang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Mingjie Gao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Pengju Yao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Yuhao Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Qingsong Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| | - Jianguo Ji
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing 100871, China; (W.Z.)
| |
Collapse
|
7
|
Carter JA, Matta B, Battaglia J, Somerville C, Harris BD, LaPan M, Atwal GS, Barnes BJ. Identification of pan-cancer/testis genes and validation of therapeutic targeting in triple-negative breast cancer: Lin28a-based and Siglece-based vaccination induces antitumor immunity and inhibits metastasis. J Immunother Cancer 2023; 11:e007935. [PMID: 38135347 DOI: 10.1136/jitc-2023-007935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
BACKGROUND Cancer-testis (CT) genes are targets for tumor antigen-specific immunotherapy given that their expression is normally restricted to the immune-privileged testis in healthy individuals with aberrant expression in tumor tissues. While they represent targetable germ tissue antigens and play important functional roles in tumorigenesis, there is currently no standardized approach for identifying clinically relevant CT genes. Optimized algorithms and validated methods for accurate prediction of reliable CT antigens (CTAs) with high immunogenicity are also lacking. METHODS Sequencing data from the Genotype-Tissue Expression (GTEx) and The Genomic Data Commons (GDC) databases was used for the development of a bioinformatic pipeline to identify CT exclusive genes. A CT germness score was calculated based on the number of CT genes expressed within a tumor type and their degree of expression. The impact of tumor germness on clinical outcome was evaluated using healthy GTEx and GDC tumor samples. We then used a triple-negative breast cancer mouse model to develop and test an algorithm that predicts epitope immunogenicity based on the identification of germline sequences with strong major histocompatibility complex class I (MHCI) and MHCII binding affinities. Germline sequences for CT genes were synthesized as long synthetic peptide vaccines and tested in the 4T1 triple-negative model of invasive breast cancer with Poly(I:C) adjuvant. Vaccine immunogenicity was determined by flow cytometric analysis of in vitro and in vivo T-cell responses. Primary tumor growth and lung metastasis was evaluated by histopathology, flow cytometry and colony formation assay. RESULTS We developed a new bioinformatic pipeline to reliably identify CT exclusive genes as immunogenic targets for immunotherapy. We identified CT genes that are exclusively expressed within the testis, lack detectable thymic expression, and are significantly expressed in multiple tumor types. High tumor germness correlated with tumor progression but not with tumor mutation burden, supporting CTAs as appealing targets in low mutation burden tumors. Importantly, tumor germness also correlated with markers of antitumor immunity. Vaccination of 4T1 tumor-bearing mice with Siglece and Lin28a antigens resulted in increased T-cell antitumor immunity and reduced primary tumor growth and lung metastases. CONCLUSION Our results present a novel strategy for the identification of highly immunogenic CTAs for the development of targeted vaccines that induce antitumor immunity and inhibit metastasis.
Collapse
Affiliation(s)
- Jason A Carter
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
- Stony Brook University, Stony Brook, New York, USA
- Department of Surgery, University of Washington, Seattle, WA, USA
| | - Bharati Matta
- Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Jenna Battaglia
- Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Carter Somerville
- Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Benjamin D Harris
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
- Lyell Immunopharma, South San Francisco, CA, USA
| | - Margaret LaPan
- Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Gurinder S Atwal
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
- Regeneron Pharmaceuticals Inc, Tarrytown, NY, USA
| | - Betsy J Barnes
- Northwell Health Feinstein Institutes for Medical Research, Manhasset, New York, USA
- Departments of Pediatrics and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
8
|
Klein M, Wefers M, Hallermann C, Fischer HJ, Hölzle F, Wermker K. IMP3 Expression as a Potential Tumour Marker in High-Risk Localisations of Cutaneous Squamous Cell Carcinoma: IMP3 in Metastatic cSCC. Cancers (Basel) 2023; 15:4087. [PMID: 37627115 PMCID: PMC10452512 DOI: 10.3390/cancers15164087] [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: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND High IMP3 expression is correlated with a worse outcome. Until now, there have been no data about IMP3 expression and clinical outcome for high-risk localisation of squamous cell carcinoma of the skin (cSCC). METHODS One-hundred twenty-two patients with cSCC of the lip and ear were included, and IMP3 expression in the tumours was immunohistochemically assessed in different evaluation approaches. Subsequently, subgroups were analysed in a matched pair approach and correlated with clinical pathologic parameters. In the following, different IMP3 analysis methods were tested for clinical suitability. RESULTS We found a significant correlation between IMP3 expression and risk for lymph node metastasis, local relapse, and progression-free survival. CONCLUSIONS On basis of our data, we suggest a prognostic benefit cutoff value for high (>50%) and low (<50%) IMP3 expression. Thus, IMP3 expression has a high scientific potential for further studies and could potentially be used as a prognostic marker in diagnostic and therapeutic decision-making.
Collapse
Affiliation(s)
- Maurice Klein
- Department of Oral, Maxillofacial and Facial Plastic Surgery, School of Medicine, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany;
| | - Merle Wefers
- Orthodontics Meyer, Kurze Straße 6, 48151 Muenster, Germany;
| | - Christian Hallermann
- Laboratory for Dermatopathology and Pathology Hamburg-Niendorf, Tibarg 7, 22459 Hamburg, Germany;
- Department of Dermatology and Histopathology, Fachklinik Hornheide, Dorbaumstrasse 300, 48157 Muenster, Germany
| | - Henrike J. Fischer
- Department of Immunology, School of Medicine, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074 Aachen, Germany;
| | - Frank Hölzle
- Department of Oral, Maxillofacial and Facial Plastic Surgery, School of Medicine, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074 Aachen, Germany;
| | - Kai Wermker
- Department of Oral and Cranio-Maxillofacial Surgery, Klinikum Osnabrueck GmbH, Am Finkenhuegel 1, 49076 Osnabrueck, Germany;
| |
Collapse
|
9
|
Ren S, Zhang Z, Li M, Wang D, Guo R, Fang X, Chen F. Cancer testis antigen subfamilies: Attractive targets for therapeutic vaccine (Review). Int J Oncol 2023; 62:71. [PMID: 37144487 PMCID: PMC10198712 DOI: 10.3892/ijo.2023.5519] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Cancer‑testis antigen (CTA) is a well‑accepted optimal target library for cancer diagnosis and treatment. Most CTAs are located on the X chromosome and aggregate into large gene families, such as the melanoma antigen, synovial sarcoma X and G antigen families. Members of the CTA subfamily are usually co‑expressed in tumor tissues and share similar structural characteristics and biological functions. As cancer vaccines are recommended to induce specific antitumor responses, CTAs, particularly CTA subfamilies, are widely used in the design of cancer vaccines. To date, DNA, mRNA and peptide vaccines have been commonly used to generate tumor‑specific CTAs in vivo and induce anticancer effects. Despite promising results in preclinical studies, the antitumor efficacy of CTA‑based vaccines is limited in clinical trials, which may be partially attributed to weak immunogenicity, low efficacy of antigen delivery and presentation processes, as well as a suppressive immune microenvironment. Recently, the development of nanomaterials has enhanced the cancer vaccination cascade, improved the antitumor performance and reduced off‑target effects. The present study provided an in‑depth review of the structural characteristics and biofunctions of the CTA subfamilies, summarised the design and utilisation of CTA‑based vaccine platforms and provided recommendations for developing nanomaterial‑derived CTA‑targeted vaccines.
Collapse
Affiliation(s)
- Shengnan Ren
- Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Zhanyi Zhang
- Bethune Third Clinical Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Mengyuan Li
- Traditional Chinese Medicine College, Jilin Agricultural University, Changchun, Jilin 130118, P.R. China
| | - Daren Wang
- Bethune Third Clinical Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ruijie Guo
- Bethune Third Clinical Medical College, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xuedong Fang
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Fangfang Chen
- Key Laboratory of Pathobiology, Ministry of Education, Nanomedicine and Translational Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
10
|
Liu X, Chen J, Chen W, Xu Y, Shen Y, Xu X. Targeting IGF2BP3 in Cancer. Int J Mol Sci 2023; 24:ijms24119423. [PMID: 37298373 DOI: 10.3390/ijms24119423] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/20/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
RNA-binding proteins (RBPs) can regulate multiple pathways by binding to RNAs, playing a variety of functions, such as localization, stability, and immunity. In recent years, with the development of technology, researchers have discovered that RBPs play a key role in the N6-methyladenosine (m6A) modification process. M6A methylation is the most abundant form of RNA modification in eukaryotes, which is defined as methylation on the sixth N atom of adenine in RNA. Insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) is one of the components of m6A binding proteins, which plays an important role in decoding m6A marks and performing various biological functions. IGF2BP3 is abnormally expressed in many human cancers, often associated with poor prognosis. Here, we summarize the physiological role of IGF2BP3 in organisms and describe its role and mechanism in tumors. These data suggest that IGF2BP3 may be a valuable therapeutic target and prognostic marker in the future.
Collapse
Affiliation(s)
- Xin Liu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiayu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenliang Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yangtao Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yang Shen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ximing Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
11
|
Carter JA, Matta B, Battaglia J, Somerville C, Harris BD, LaPan M, Atwal GS, Barnes BJ. Identification of pan-cancer/testis genes and validation of therapeutic targeting in triple-negative breast cancer: Lin28a- and Siglece-based vaccination induces anti-tumor immunity and inhibits metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.09.539617. [PMID: 37214884 PMCID: PMC10197572 DOI: 10.1101/2023.05.09.539617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Background Cancer-testis (CT) genes are targets for tumor antigen-specific immunotherapy given that their expression is normally restricted to the immune-privileged testis in healthy individuals with aberrant expression in tumor tissues. While they represent targetable germ-tissue antigens and play important functional roles in tumorigenesis, there is currently no standardized approach for identifying clinically relevant CT genes. Optimized algorithms and validated methods for accurate prediction of reliable CT antigens with high immunogenicity are also lacking. Methods Sequencing data from the Genotype-Tissue Expression (GTEx) and The Genomic Data Commons (GDC) databases was utilized for the development of a bioinformatic pipeline to identify CT exclusive genes. A CT germness score was calculated based on the number of CT genes expressed within a tumor type and their degree of expression. The impact of tumor germness with clinical outcome was evaluated using healthy GTEx and GDC tumor samples. We then used a triple-negative breast cancer mouse model to develop and test an algorithm that predicts epitope immunogenicity based on the identification of germline sequences with strong MHCI and MHCII binding affinities. Germline sequences for CT genes were synthesized as long synthetic peptide vaccines and tested in the 4T1 triple-negative model of invasive breast cancer with Poly(I:C) adjuvant. Vaccine immunogenicity was determined by flow cytometric analysis of in vitro and in vivo T cell responses. Primary tumor growth and lung metastasis was evaluated by histopathology, flow cytometry and colony formation assay. Results We developed a new bioinformatic pipeline to reliably identify CT exclusive genes as immunogenic targets for immunotherapy. We identified CT genes that are exclusively expressed within the testis, lack detectable thymic expression, and are significantly expressed in multiple tumor types. High tumor germness correlated with tumor progression but not with tumor mutation burden, supporting CT antigens as appealing targets in low mutation burden tumors. Importantly, tumor germness also correlated with markers of anti-tumor immunity. Vaccination of 4T1 tumor bearing mice with Siglece and Lin28a antigens resulted in increased T cell anti-tumor immunity and reduced primary tumor growth and lung metastases. Conclusion Our results present a novel strategy for the identification of highly immunogenic CT antigens for the development of targeted vaccines that induce anti-tumor immunity and inhibit metastasis.
Collapse
|
12
|
Kajiwara Y, Takahashi A, Ueno H, Kakeji Y, Hasegawa H, Eguchi S, Goi T, Saiura A, Sasaki A, Takiguchi S, Takeuchi H, Tanaka C, Hashimoto M, Hiki N, Horiguchi A, Matsuda S, Mizushima T, Marubashi S, Gotoh M, Konno H, Yamamoto H, Miyata H, Seto Y, Kitagawa Y, The National Clinical Database. Annual report on National Clinical Database 2020 for gastroenterological surgery in Japan. Ann Gastroenterol Surg 2023; 7:367-406. [PMID: 37152776 PMCID: PMC10154850 DOI: 10.1002/ags3.12662] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 01/16/2023] [Accepted: 01/23/2023] [Indexed: 04/04/2023] Open
Abstract
Aim The National Clinical Database (NCD) of Japan is a nationwide data entry system for surgery, and it marked its 10th anniversary in 2020. The aim was to present the 2020 annual report of gastroenterological surgery of the NCD. Methods The data of the surgical procedures stipulated by the training curriculum for board-certified surgeons of the Japanese Society of Gastroenterological Surgery in the NCD from 2011 to 2020 were summarized. Results In total, 5 622 845 cases, including 593 088 cases in 2020, were extracted from the NCD. The total number of gastroenterological surgeries increased gradually in these 10 years, except for the year 2020 due to the COVID-19 pandemic. The annual number of surgeries of each organ, except the pancreas and liver, decreased by 0.4%-13.1% in 2020 compared to 2019. The surgical patients were consistently aging, with more than 20% of all gastroenterological surgeries in 2020 involving patients aged 80 years or older. The participation of board-certified surgeons increased for each organ (75.9%-95.7% in 2020). The rates of endoscopic surgery also increased constantly. Although the incidences of postoperative complications of each organ increased by 0.7%-7.9% in these 10 years, postoperative mortality rates decreased by 0.2%-1.5%. Conclusions We present here the short-term outcomes of each gastroenterological operative procedure in 2020. This review of the 10-years of NCD data of gastroenterological surgery revealed a consistent increase of the number of surgeries (except for in 2020), especially endoscopic procedures, and aging of the Japanese population. The good safety of Japanese gastroenterological surgeries was also indicated.
Collapse
Affiliation(s)
| | - Arata Takahashi
- Department of Health Policy and Management, School of MedicineKeio UniversityTokyoJapan
- Department of Healthcare Quality Assessment Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hideki Ueno
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | | | | | - Susumu Eguchi
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | - Takanori Goi
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | - Akio Saiura
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | - Akira Sasaki
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | - Shuji Takiguchi
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | - Hiroya Takeuchi
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | - Chie Tanaka
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | | | - Naoki Hiki
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | | | - Satoru Matsuda
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | | | | | - Mitsukazu Gotoh
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | - Hiroyuki Konno
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | - Hiroyuki Yamamoto
- Department of Health Policy and Management, School of MedicineKeio UniversityTokyoJapan
- Department of Healthcare Quality Assessment Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Hiroaki Miyata
- Department of Health Policy and Management, School of MedicineKeio UniversityTokyoJapan
- Department of Healthcare Quality Assessment Graduate School of MedicineThe University of TokyoTokyoJapan
| | - Yasuyuki Seto
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | - Yuko Kitagawa
- The Japanese Society of Gastroenterological SurgeryTokyoJapan
| | | |
Collapse
|
13
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
14
|
Shimizu N, Hussain SA, Obara W, Yamasaki T, Takashima S, Hasegawa T, Iguchi M, Igarashi K, Ogawa O, Fujioka T. A Phase 2 Study of S-588410 Maintenance Monotherapy for Platinum-Treated Advanced or Metastatic Urothelial Carcinoma. Bladder Cancer 2022; 8:179-192. [PMID: 38993370 PMCID: PMC11181746 DOI: 10.3233/blc-211592] [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: 08/03/2021] [Accepted: 04/06/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Effective maintenance therapy for urothelial carcinoma (UC) is needed to delay progression after first-line chemotherapy. OBJECTIVE To evaluate S-588410, a cancer peptide vaccine containing five human leukocyte antigen (HLA)-A*24:02-restricted epitope peptides derived from five cancer-testis antigens (DEPDC1, MPHOSPH1, URLC10, CDCA1, and KOC1) in chemotherapy-treated, clinically stable patients with advanced or metastatic UC. MATERIALS AND METHODS This open-label, international, phase 2 trial enrolled patients with UC who had completed≥4 cycles of first-line platinum-containing chemotherapy without disease progression. Forty-five HLA-A*24:02-positive patients received subcutaneous injections of S-588410 (Montanide ISA 51 VG with 1 mg/mL of each peptide) weekly for 12 weeks then once every 2 weeks thereafter for up to 24 months. Thirty-six HLA-A*24:02-negative patients did not receive S-588410 (observation group). The primary endpoint was the rate of cytotoxic T-lymphocyte (CTL) induction against≥1 of the peptides at 12 weeks. RESULTS The CTL induction rate in the S-588410 group was 93.3% (p < 0.0001, one-sided binomial test with a rate of≤50% as the null hypothesis). The antitumor response rate was 8.9% in the S-588410 group and 0% in the observation group; median progression-free survival was 18.1 versus 12.5 weeks and median overall survival was 71.0 versus 99.0 weeks, respectively. The most frequent treatment-emergent adverse event was injection-site reactions (47 events, grades 1-3) reported in 93.3% (n = 42/45) of participants. CONCLUSIONS S-588410 demonstrated a high CTL induction rate, acceptable safety profile, and modest clinical response, as maintenance therapy in participants with advanced or metastatic UC who had received first-line platinum-based chemotherapy (EudraCT 2013-005274-22).
Collapse
Affiliation(s)
- Nobuaki Shimizu
- Department of Urology, Gunma Prefectural Cancer Center, Ota, Gunma, Japan
| | | | - Wataru Obara
- Department of Urology, Iwate Medical University, Morioka, Iwate, Japan
| | - Toshinari Yamasaki
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | - Osamu Ogawa
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoaki Fujioka
- Department of Urology, Iwate Medical University, Morioka, Iwate, Japan
| |
Collapse
|
15
|
Identification of shared neoantigens in esophageal carcinoma by the combination of comprehensive analysis of genomic data and in silico neoantigen prediction. Cell Immunol 2022; 377:104537. [DOI: 10.1016/j.cellimm.2022.104537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 01/10/2023]
|
16
|
Yasuda T, Nishiki K, Hiraki Y, Kato H, Iwama M, Shiraishi O, Yasuda A, Shinkai M, Kimura Y, Sukegawa Y, Chiba Y, Imano M, Takeda K, Satou T, Shiozaki H, Nakamura Y. Phase II Adjuvant Cancer-specific Vaccine Therapy for Esophageal Cancer Patients Curatively Resected After Preoperative Therapy With Pathologically Positive Nodes; Possible Significance of Tumor Immune Microenvironment in its Clinical Effects. Ann Surg 2022; 275:e155-e162. [PMID: 33055588 DOI: 10.1097/sla.0000000000003880] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To elucidate the efficacy of adjuvant vaccine monotherapy using 3 Human Leukocyte Antigen (HLA)-A∗24-restricted tumor-specific peptide antigens for ESCC, upregulated lung cancer 10, cell division cycle associated 1, and KH domain-containing protein overexpressed in cancer 1. SUMMARY OF BACKGROUND DATA ESCC patients with pathologically positive nodes (pN(+)) have a high risk for postoperative recurrence, despite curative resection after preoperative therapy. Subclinical micrometastases are an appropriate target for cancer vaccine. METHODS This is a non-randomized prospective phase II clinical trial (UMIN000003557). ESCC patients curatively resected after preoperative therapy with pN(+) were allocated into the control and vaccine groups (CG and VG) according to the HLA-A status. One mg each of three epitope peptides was postoperatively injected 10 times weekly followed by 10 times biweekly to the VG. The primary and secondary endpoints were relapse-free survival (RFS) and esophageal cancer-specific survival (ECSS), respectively. RESULTS Thirty were in the CG and 33 in the VG. No significant difference was observed in RFS between the CG and VG (5-year RFS: 32.5% vs 45.3%), but the recurrence rate significantly decreased with the number of peptides which induced antigen-specific cytotoxic T lymphocytes. The VG showed a significantly higher 5-year ECSS than the CG (60.0% vs 32.4%, P = 0.045) and this difference was more prominent in patients with CD8+ and programmed death-ligand 1 double negative tumor (68.0% vs 17.7%, P = 0.010). CONCLUSIONS Our cancer peptide vaccine might improve the survival of ESCC patients, which is warranted to be verified in the phase III randomized controlled study.
Collapse
Affiliation(s)
- Takushi Yasuda
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kohei Nishiki
- Department of Surgery, Ohita Nakamura Hospital, Ohita, Japan
| | - Yoko Hiraki
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Hiroaki Kato
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Mitsuru Iwama
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Osamu Shiraishi
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Atsushi Yasuda
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Masayuki Shinkai
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yutaka Kimura
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yasushi Sukegawa
- Life Science Research Institute, Kindai University, Osaka, Japan
| | - Yasutaka Chiba
- Clinical Research Center, Kindai University Hospital, Osaka, Japan
| | - Motohiro Imano
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kazuyoshi Takeda
- Division of Cell Biology, Biomedical Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takao Satou
- Division of Hospital Pathology, Kindai University Hospital, Osaka, Japan
| | - Hitoshi Shiozaki
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | | |
Collapse
|
17
|
Wu SC, Münger K. Role and Clinical Utility of Cancer/Testis Antigens in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13225690. [PMID: 34830845 PMCID: PMC8616139 DOI: 10.3390/cancers13225690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/03/2021] [Accepted: 11/11/2021] [Indexed: 12/15/2022] Open
Abstract
Cancer/testis (CT) antigens exhibit selective expression predominantly in immunoprivileged tissues in non-pathological contexts but are aberrantly expressed in diverse cancers. Due to their expression pattern, they have historically been attractive targets for immunotherapies. A growing number of studies implicate CT antigens in almost all hallmarks of cancer, suggesting that they may act as cancer drivers. CT antigens are expressed in head and neck squamous cell carcinomas. However, their role in the pathogenesis of these cancers remains poorly studied. Given that CT antigens hold intriguing potential as therapeutic targets and as biomarkers for prognosis and that they can provide novel insights into oncogenic mechanisms, their further study in the context of head and squamous cell carcinoma is warranted.
Collapse
Affiliation(s)
- Sharon Changshan Wu
- Molecular Microbiology Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA 02111, USA;
| | - Karl Münger
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA 02111, USA
- Correspondence:
| |
Collapse
|
18
|
He S, Xu J, Liu X, Zhen Y. Advances and challenges in the treatment of esophageal cancer. Acta Pharm Sin B 2021; 11:3379-3392. [PMID: 34900524 PMCID: PMC8642427 DOI: 10.1016/j.apsb.2021.03.008] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/24/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Esophageal cancer (EC) is one of the most common cancers with high morbidity and mortality rates. EC includes two histological subtypes, namely esophageal squamous cell carcinoma (ESCC) and esophageal adenocarcinoma (EAC). ESCC primarily occurs in East Asia, whereas EAC occurs in Western countries. The currently available treatment strategies for EC include surgery, chemotherapy, radiation therapy, molecular targeted therapy, and combinations thereof. However, the prognosis remains poor, and the overall five-year survival rate is very low. Therefore, achieving the goal of effective treatment remains challenging. In this review, we discuss the latest developments in chemotherapy and molecular targeted therapy for EC, and comprehensively analyze the application prospects and existing problems of immunotherapy. Collectively, this review aims to provide a better understanding of the currently available drugs through in-depth analysis, promote the development of new therapeutic agents, and eventually improve the treatment outcomes of patients with EC.
Collapse
Affiliation(s)
- Shiming He
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Jian Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Yongsu Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
19
|
Wang L, Han H, Wang Z, Shi L, Yang M, Qin Y. Targeting the Microenvironment in Esophageal Cancer. Front Cell Dev Biol 2021; 9:684966. [PMID: 34513829 PMCID: PMC8427432 DOI: 10.3389/fcell.2021.684966] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Esophageal cancer (EC) is the eighth most common type of cancer and the sixth leading cause of cancer-related deaths worldwide. At present, the clinical treatment for EC is based mainly on radical surgery, chemotherapy, and radiotherapy. However, due to the limited efficacy of conventional treatments and the serious adverse reactions, the outcome is still unsatisfactory (the 5-year survival rate for patients is less than 25%). Thus, it is extremely important and urgent to identify new therapeutic targets. The concept of tumor microenvironment (TME) has attracted increased attention since it was proposed. Recent studies have shown that TME is an important therapeutic target for EC. Microenvironment-targeting therapies such as immunotherapy and antiangiogenic therapy have played an indispensable role in prolonging survival and improving the prognosis of patients with EC. In addition, many new drugs and therapies that have been developed to target microenvironment may become treatment options in the future. We summarize the microenvironment of EC and the latest advances in microenvironment-targeting therapies in this review.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Huiqiong Han
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Zehua Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Litong Shi
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Mei Yang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
20
|
Lampis A, Ratti M, Ghidini M, Mirchev MB, Okuducu AF, Valeri N, Hahne JC. Challenges and perspectives for immunotherapy in oesophageal cancer: A look to the future (Review). Int J Mol Med 2021; 47:97. [PMID: 33846775 PMCID: PMC8041478 DOI: 10.3892/ijmm.2021.4930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Oesophageal cancer is one of the most aggressive malignancies with limited treatment options, thus resulting in a high morbidity and mortality. With 5‑year survival rates of only 5‑10%, oesophageal cancer holds a dismal prognosis for patients. In order to improve overall survival, the early diagnosis and tools for patient stratification for personalized treatment are urgent needs. A minority of oesophageal cancers belong to the spectrum of Lynch syndrome‑associated cancers and are characterized by microsatellite instability (MSI). Microsatellite instability is a consequence of defective mismatch repair protein functions and it has been well characterized in other gastrointestinal tumours, such as colorectal and gastric cancer. In the latter, high levels of MSI are associated with a better prognosis and with an increased benefit to immune‑based therapies. Therefore, similar therapeutic approaches could offer an opportunity of treatment for oesophageal cancer patients with MSI. Apart from immune checkpoint inhibitors, other immunotherapies such as adoptive T‑cell transfer, peptide vaccine and oncolytic viruses are under investigation in oesophageal cancer patients. In the present review, the rationale and current knowledge about immunotherapies in oesophageal cancer are summarised.
Collapse
Affiliation(s)
- Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
| | - Margherita Ratti
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
- Medical Department, Division of Oncology, Hospital Trust of Cremona, I-26100 Cremona, Italy
| | - Michele Ghidini
- Division of Medical Oncology, Hospital Policlinic 'Fondazione IRCCS Ca' Granda Ospedale Maggiore', I-20122 Milan, Italy
| | - Milko B. Mirchev
- Clinic of Gastroenterology, Medical University, 9002 Varna, Bulgaria
| | | | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
- Department of Medicine, The Royal Marsden NHS Foundation Trust, Sutton SM25NG, UK
| | - Jens Claus Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton SM25NG, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, Sutton SM25NG, UK
| |
Collapse
|
21
|
Bevanda Glibo D, Bevanda D, Vukojević K, Tomić S. IMP3 protein is an independent prognostic factor of clinical stage II rectal cancer. Sci Rep 2021; 11:10844. [PMID: 34035433 PMCID: PMC8149387 DOI: 10.1038/s41598-021-90513-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/12/2021] [Indexed: 12/24/2022] Open
Abstract
Immunohistochemical level of IMP3-protein in patients with rectal cancer in clinical stage II (141), were correlated with sociodemographic, pathohistological and clinical indicators and duration of overall-survival and progression-free-survival. Vascular invasion was associated with IMP3-positive immunostaining (p < 0.001). Vascular invasion ratio in the group of poorly-differentiated-tumors was 21 times higher than in the group of well-differentiated-tumors. IMP3-positive patients lived 2.2 times shorter than negative (p < 0.001). Patients with well-differentiated-tumors lived 1.7 times longer than the subjects with poorly-differentiated-tumors (p < 0.001). Patients without vascular invasion lived 2.7 times longer than the subjects with vascular invasion (p < 0.001). The risk of mortality was 2.3 times higher for IMP3 positive patients (p = 0.027) and 10.4 higher for the patients with vascular invasion (p < 0.001). IMP3-negative participants had 2.3 times longer free interval without disease (p < 0.001). The free interval without disease was 3.6 times longer in the group without vascular invasion (p < 0.001). The risk of disease relapse in the IMP3 positive group was 5.3 times higher (p < 0.001) and with vascular invasion was 8 times longer (p < 0.001). The risk of disease relapse was 6.8 times higher in the group with vascular invasion (p < 0.001). Patients with rectal cancer and high IMP3-protein level will have a shorter overall survival relative to patients without or with low levels of IMP3. The analysis of IMP3 expression by immunohistochemistry pointed IMP3 as an independent prognostic factor of clinical stage II rectal cancer.
Collapse
Affiliation(s)
- Daniela Bevanda Glibo
- Department of Internal Medicine, University Hospital Center Mostar, 88000, Mostar, Bosnia and Herzegovina
| | - Danijel Bevanda
- Department of Internal Medicine, University Hospital Center Mostar, 88000, Mostar, Bosnia and Herzegovina
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21000, Split, Croatia. .,Department of Medical Genetics, School of Medicine, University of Mostar, 88000, Mostar, Bosnia and Herzegovina.
| | - Snježana Tomić
- Department of Pathology, Citology and Forensic Medicine, University Hospital Center Split, 21000, Split, Croatia
| |
Collapse
|
22
|
Kumar S, Buon L, Talluri S, Roncador M, Liao C, Zhao J, Shi J, Chakraborty C, Gonzalez G, Tai YT, Prabhala R, Samur MK, Munshi NC, Shammas MA. Integrated genomics and comprehensive validation reveal drivers of genomic evolution in esophageal adenocarcinoma. Commun Biol 2021; 4:617. [PMID: 34031527 PMCID: PMC8144613 DOI: 10.1038/s42003-021-02125-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 04/16/2021] [Indexed: 12/13/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is associated with a marked genomic instability, which underlies disease progression and development of resistance to treatment. In this study, we used an integrated genomics approach to identify a genomic instability signature. Here we show that elevated expression of this signature correlates with poor survival in EAC as well as three other cancers. Knockout and overexpression screens establish the relevance of these genes to genomic instability. Indepth evaluation of three genes (TTK, TPX2 and RAD54B) confirms their role in genomic instability and tumor growth. Mutational signatures identified by whole genome sequencing and functional studies demonstrate that DNA damage and homologous recombination are common mechanisms of genomic instability induced by these genes. Our data suggest that the inhibitors of TTK and possibly other genes identified in this study have potential to inhibit/reduce growth and spontaneous as well as chemotherapy-induced genomic instability in EAC and possibly other cancers. Subodh Kumar et al. identify a gene signature correlated with genomic instability and poor survival in esophageal adenocarcinoma (EAC), using a combination of integrative genomic analysis of patient data and laboratory validation in cell line models and mice. They find that inhibitors of some of the identified proteins, including TTK, could be used to reduce genomic evolution as well as inhibit growth of EAC cells.
Collapse
Affiliation(s)
- Subodh Kumar
- Dana Farber Cancer Institute, Boston, MA, USA.,Veterans Administration Healthcare System, Boston, MA, USA
| | - Leutz Buon
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Srikanth Talluri
- Dana Farber Cancer Institute, Boston, MA, USA.,Veterans Administration Healthcare System, Boston, MA, USA
| | | | - Chengcheng Liao
- Dana Farber Cancer Institute, Boston, MA, USA.,Veterans Administration Healthcare System, Boston, MA, USA
| | - Jiangning Zhao
- Dana Farber Cancer Institute, Boston, MA, USA.,Veterans Administration Healthcare System, Boston, MA, USA
| | - Jialan Shi
- Dana Farber Cancer Institute, Boston, MA, USA.,Veterans Administration Healthcare System, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | - Gabriel Gonzalez
- Veterans Administration Healthcare System, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Yu-Tzu Tai
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Rao Prabhala
- Dana Farber Cancer Institute, Boston, MA, USA.,Veterans Administration Healthcare System, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | | | - Nikhil C Munshi
- Dana Farber Cancer Institute, Boston, MA, USA.,Veterans Administration Healthcare System, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Masood A Shammas
- Dana Farber Cancer Institute, Boston, MA, USA. .,Veterans Administration Healthcare System, Boston, MA, USA.
| |
Collapse
|
23
|
Lonie JM, Barbour AP, Dolcetti R. Understanding the immuno-biology of oesophageal adenocarcinoma: Towards improved therapeutic approaches. Cancer Treat Rev 2021; 98:102219. [PMID: 33993033 DOI: 10.1016/j.ctrv.2021.102219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022]
Abstract
With an incidence that is constantly rising, oesophageal adenocarcinoma (OAC) is becoming an increasing health burden worldwide. Although significant advances in treatment regimens have improved patient outcomes, survival rates for this deadly cancer remain unsatisfactory. This highlights the need to improve current therapeutic approaches and develop novel therapeutic strategies for treating OAC patients. The advent of immunotherapy has revolutionised treatment across a range of malignancies, however outcomes in OAC show modest results. The inherent resistance of OAC to treatment reflects the complex genomic landscape of this cancer, which displays a lack of ubiquitous driver mutations and large-scale genomic alterations along with high tumour and immune heterogeneity. Research into the immune landscape of OAC is limited, and elucidation of the mechanisms surrounding the immune responses to this complex cancer will result in improved therapeutic approaches. This review explores what is known about the immuno-biology of OAC and explores promising therapeutic avenues that may improve responses to immunotherapeutic regimens.
Collapse
Affiliation(s)
- James M Lonie
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia.
| | - Andrew P Barbour
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia; Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Riccardo Dolcetti
- The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia; Sir Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
24
|
Liu W, Tang H, Li L, Wang X, Yu Z, Li J. Peptide-based therapeutic cancer vaccine: Current trends in clinical application. Cell Prolif 2021; 54:e13025. [PMID: 33754407 PMCID: PMC8088465 DOI: 10.1111/cpr.13025] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/21/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023] Open
Abstract
The peptide‐based therapeutic cancer vaccines have attracted enormous attention in recent years as one of the effective treatments of tumour immunotherapy. Most of peptide‐based vaccines are based on epitope peptides stimulating CD8+ T cells or CD4+ T helper cells to target tumour‐associated antigens (TAAs) or tumour‐specific antigens (TSAs). Some adjuvants and nanomaterials have been exploited to optimize the efficiency of immune response of the epitope peptide to improve its clinical application. At present, numerous peptide‐based therapeutic cancer vaccines have been developed and achieved significant clinical benefits. Similarly, the combination of peptide‐based vaccines and other therapies has demonstrated a superior efficacy in improving anti‐cancer activity. We delve deeper into the choices of targets, design and screening of epitope peptides, clinical efficacy and adverse events of peptide‐based vaccines, and strategies combination of peptide‐based therapeutic cancer vaccines and other therapies. The review will provide a detailed overview and basis for future clinical application of peptide‐based therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Wensi Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center, Shenyang, China
| | - Haichao Tang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center, Shenyang, China
| | - Luanfeng Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center, Shenyang, China
| | - Xiangyi Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center, Shenyang, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, Liaoning Cancer immune peptide drug Engineering Technology Research Center, Shenyang, China
| | - Jianping Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Transfusion Medicine Institute, Liaoning Blood Center, Shenyang, China.,Transfusion Medicine Institute, Harbin Blood Center, Harbin, China
| |
Collapse
|
25
|
Oguma J, Ozawa S, Sakakibara T, Kajiwara H, Nakamura N, Makuuchi H. Prognostic impact of LY6K and CDCA1 expression for patients with esophageal squamous cell carcinoma. Ann Gastroenterol Surg 2021; 5:194-203. [PMID: 33860139 PMCID: PMC8034699 DOI: 10.1002/ags3.12415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 11/06/2022] Open
Abstract
AIM In the present study, we investigated the relationship between the expressions of two cancer testis antigens (CTA), LY6K (lymphocyte antigen 6 complex locus K) and CDCA1 (cell division cycle associated 1), in esophageal squamous cell carcinoma (ESCC) tumors and the long-term outcomes of patients with ESCC to clarify the clinical significance of LY6K and CDCA1 expression in ESCC tumors. METHODS A total of 175 patients with thoracic ESCC who had undergone a thoracic esophagectomy with three-field lymphadenectomy without neoadjuvant therapy were retrospectively reviewed in this study. LY6K and CDCA1 expressions were evaluated in tumor tissues using immunohistochemical (IH) staining. RESULTS Median patient age was 63 years; 159 patients (90.9%) were men. Ninety-four patients (55.3%) were LY6K-positive, and 85 patients (48.6%) were CDCA1-positive. The LY6K-positive group had a significantly worse overall survival (OS) than the LY6K-negative group (P = 0.012), and the CDCA1-positive group had a significantly worse OS than the CDCA1-negative group (P = 0.010). A multivariate analysis suggested that pathological N stage, venous invasion, LK6Y-positive and CDCA1-positive were independent prognostic factors. The patients were classified into four groups according to the staining pattern combinations of the two CTA. The LY6K-positive and CDCA1-positive group was found to have a significantly poorer outcome than the other groups. CONCLUSION ESCC patients with a combination of LY6K and CDCA1 expression in their tumor tissues had a worse prognosis than all the other ESCC patients and it was an independent factor associated with prognosis for patients with ESCC.
Collapse
Affiliation(s)
- Junya Oguma
- Department of Gastroenterological SurgeryTokai University School of MedicineIseharaJapan
| | - Soji Ozawa
- Department of Gastroenterological SurgeryTokai University School of MedicineIseharaJapan
| | - Terue Sakakibara
- Department of Gastroenterological SurgeryTokai University School of MedicineIseharaJapan
| | - Hiroshi Kajiwara
- Department of PathologyTokai University School of MedicineIseharaJapan
| | - Naoya Nakamura
- Department of PathologyTokai University School of MedicineIseharaJapan
| | - Hiroyasu Makuuchi
- Department of Gastroenterological SurgeryTokai University School of MedicineIseharaJapan
| |
Collapse
|
26
|
Ikeda M, Okusaka T, Ohno I, Mitsunaga S, Kondo S, Ueno H, Morizane C, Gemmoto K, Suna H, Ushida Y, Furuse J. Phase I studies of peptide vaccine cocktails derived from GPC3, WDRPUH and NEIL3 for advanced hepatocellular carcinoma. Immunotherapy 2021; 13:371-385. [PMID: 33525928 DOI: 10.2217/imt-2020-0278] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aim: Two peptide cocktail vaccines using glypican-3, WD-repeat-containing protein up-regulated in hepatocellular carcinoma (HCC) and nei endonuclease VIII-like three epitopes were evaluated in advanced HCC in two Phase I studies. Patients & methods: Study 1 evaluated dose-limiting toxicities (DLTs) of peptides 1-3 (HLA-A24-restricted) and study 2 evaluated DLTs of peptides 1-6 (HLA-A24 or A02-restricted). Results: Overall, 18 and 14 patients were enrolled in studies 1 and 2, respectively. No DLTs were observed up to 7.1 mg of the vaccine cocktail. No complete response/partial response was observed. Stable disease was reported in nine and five patients with a disease control rate of 52.9% and 35.7% in studies 1 and 2, respectively. Conclusion: Both vaccines showed good tolerability and potential usefulness against HCC. Clinical trial registration: JapicCTI-121933; JapicCTI-142477.
Collapse
Affiliation(s)
- Masafumi Ikeda
- Department of Hepatobiliary & Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary & Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Izumi Ohno
- Department of Hepatobiliary & Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shuichi Mitsunaga
- Department of Hepatobiliary & Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shunsuke Kondo
- Department of Hepatobiliary & Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hideki Ueno
- Department of Hepatobiliary & Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary & Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kazuto Gemmoto
- Oncology Clinical Development Unit, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Hideaki Suna
- Oncology Clinical Development Unit, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Yasunori Ushida
- Clinical Development, Data Science, Statistical Analysis, Ono Pharmaceutical Co., Ltd, Osaka, Japan
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| |
Collapse
|
27
|
Vacchelli E, Martins I, Eggermont A, Fridman WH, Galon J, Sautès-Fridman C, Tartour E, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide vaccines in cancer therapy. Oncoimmunology 2021; 1:1557-1576. [PMID: 23264902 PMCID: PMC3525611 DOI: 10.4161/onci.22428] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Prophylactic vaccination constitutes one of the most prominent medical achievements of history. This concept was first demonstrated by the pioneer work of Edward Jenner, dating back to the late 1790s, after which an array of preparations that confer life-long protective immunity against several infectious agents has been developed. The ensuing implementation of nation-wide vaccination programs has de facto abated the incidence of dreadful diseases including rabies, typhoid, cholera and many others. Among all, the most impressive result of vaccination campaigns is surely represented by the eradication of natural smallpox infection, which was definitively certified by the WHO in 1980. The idea of employing vaccines as anticancer interventions was first theorized in the 1890s by Paul Ehrlich and William Coley. However, it soon became clear that while vaccination could be efficiently employed as a preventive measure against infectious agents, anticancer vaccines would have to (1) operate as therapeutic, rather than preventive, interventions (at least in the vast majority of settings), and (2) circumvent the fact that tumor cells often fail to elicit immune responses. During the past 30 y, along with the recognition that the immune system is not irresponsive to tumors (as it was initially thought) and that malignant cells express tumor-associated antigens whereby they can be discriminated from normal cells, considerable efforts have been dedicated to the development of anticancer vaccines. Some of these approaches, encompassing cell-based, DNA-based and purified component-based preparations, have already been shown to exert conspicuous anticancer effects in cohorts of patients affected by both hematological and solid malignancies. In this Trial Watch, we will summarize the results of recent clinical trials that have evaluated/are evaluating purified peptides or full-length proteins as therapeutic interventions against cancer.
Collapse
Affiliation(s)
- Erika Vacchelli
- Institut Gustave Roussy; Villejuif, France ; Université Paris-Sud/Paris XI; Le Kremlin-Bicêtre, France ; INSERM, U848; Villejuif, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Combined inhibition of PD-1/PD-L1, Lag-3, and Tim-3 axes augments antitumor immunity in gastric cancer-T cell coculture models. Gastric Cancer 2021; 24:611-623. [PMID: 33611641 PMCID: PMC8065004 DOI: 10.1007/s10120-020-01151-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Immunotherapy targeting PD-1 provides a limited survival benefit in patients with unresectable advanced or recurrent gastric cancer (GC). Beside PD-L1, the expression of inhibitory ligands such as CEACAM-1 and LSECtin on GC cells account for this limitation. Here we assessed their expression and immune suppressive effect in GC patients. METHODS Using multiplexed immunohistochemistry staining, we evaluated the distribution of different inhibitory ligands, including PD-L1, CEACAM-1, LSECtin, and MHC class II, in 365 GC patients. We analyzed their correlations and overall survival (OS) based on the expression of each inhibitory ligand and the independent prognostic factors that affect OS. Subsequently, we evaluated the additive effect of anti-PD-1 mAb or anti-PD-L1 mAb with/without anti-Lag-3 mAb with/without anti-Tim-3 mAb in cytotoxic assay using tumor-antigen specific CTL clones against GC cell lines. RESULTS Co-expression of the inhibitory ligands for PD-1, Tim-3, and Lag-3 was observed in the largest proportion (34.7%). CEACAM-1, LSECtin, and MHC class II expression showed significant correlation with PD-L1 expression and OS. Multivariable analysis demonstrated that CEACAM-1 low is an independent prognostic factor. Furthermore, combining dual and triple ICIs yielded additive effect on cytotoxicity of CTL clones against each immune inhibitory ligand positive GC cell lines. CONCLUSIONS Our findings suggested that the expression of inhibitory ligands for Tim-3 and Lag-3 on GC cells serve as potential biomarkers to predict the response to anti-PD-1 therapy and the combinatorial immunotherapy with ICIs targeting for PD-1, Tim-3, and Lag-3 has a therapeutic potential for GC patients.
Collapse
|
29
|
Chang M, Hou Z, Wang M, Li C, Lin J. Recent Advances in Hyperthermia Therapy-Based Synergistic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2004788. [PMID: 33289219 DOI: 10.1002/adma.202004788] [Citation(s) in RCA: 243] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/17/2020] [Indexed: 06/12/2023]
Abstract
The past decades have witnessed hyperthermia therapy (HTT) as an emerging strategy against malignant tumors. Nanomaterial-based photothermal therapy (PTT) and magnetic hyperthermia (MHT), as highly effective and noninvasive treatment models, offer advantages over other strategies in the treatment of different types of tumors. However, both PTT and MHT cannot completely cure cancer due to recurrence and distal metastasis. In recent years, cancer immunotherapy has attracted widespread attention owing to its capability to activate the body's own natural defense to identify, attack, and eradicate cancer cells. Significant efforts have been devoted to studying the activated immune responses caused by hyperthermia-ablated tumors. In this article, the synergistic mechanism of HTT in immunotherapy, including immunogenic cell death and reversal of the immunosuppressive tumor microenvironment is discussed. The reports of the combination of HTT or HTT-based multimodal therapy with immunotherapy, including immunoadjuvant exploitation, immune checkpoint blockade therapy, and adoptive cellular immunotherapy are summarized. As highlighted, these strategies could achieve synergistically enhanced therapeutic outcomes against both primary tumors and metastatic lesions, prevent cancer recurrence, and prolong the survival period. Finally, current challenges and prospective developments in HTT-synergized immunotherapy are also reviewed.
Collapse
Affiliation(s)
- Mengyu Chang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| | - Zhiyao Hou
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangdong, 511436, P. R. China
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P. R. China
| | - Man Wang
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
30
|
Peng C, Cohen DJ. Advances in the pharmacotherapeutic management of esophageal squamous cell carcinoma. Expert Opin Pharmacother 2020; 22:93-107. [PMID: 33034212 DOI: 10.1080/14656566.2020.1813278] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Esophageal squamous cancer remains an important cause of mortality worldwide with two new immunotherapy drugs recently approved for metastatic disease. AREAS COVERED The authors review the epidemiology and genomics of esophageal squamous cell carcinoma. They also examine prior trials involving targeted agents under investigation as well immunotherapies that have been approved and novel combinations. EXPERT OPINION Great advances have been made in characterizing the molecular changes in esophageal carcinoma. However, relatively few drugs have shown benefit in this disease. Targeted therapies have not shown to improve survival although many of these trials did not explore potential biomarkers. Pembrolizumab and nivolumab are now approved for esophageal squamous carcinoma but much more data are needed to understand how these agents may be used in non-metastatic settings. Novel treatments are still required as overall prognosis remains poor.
Collapse
Affiliation(s)
| | - Deirdre J Cohen
- Department of Hematology and Medical Oncology, Tisch Cancer Institute, Mount Sinai Health , New York, NY, USA
| |
Collapse
|
31
|
Tamura R, Morimoto Y, Sato M, Hikichi T, Yoshida K, Toda M. A Pilot Study of the Adverse Events Caused by the Combined Use of Bevacizumab and Vascular Endothelial Growth Factor Receptor-Targeted Vaccination for Patients with a Malignant Glioma. Vaccines (Basel) 2020; 8:vaccines8030498. [PMID: 32887369 PMCID: PMC7564945 DOI: 10.3390/vaccines8030498] [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: 07/02/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 11/28/2022] Open
Abstract
Anti-angiogenic therapy, targeting vascular endothelial growth factor (VEGF)-A/VEGF receptors (VEGFRs), is beneficial for tumor growth prevention in a malignant glioma. A simultaneous blockade using both bevacizumab (Bev), which targets circulating VEGF-A, and a multi-kinase inhibitor on VEGFRs was more effective for advanced solid cancers, including melanoma and renal cell carcinoma. However, previous clinical trials demonstrated a high adverse event rate. Additionally, no studies previously assessed treatment efficacy and safety using both VEGF-A and VEGFR-targeted agents for malignant gliomas. We had conducted clinical trials investigating VEGFRs peptide vaccination in patients with malignant gliomas, in which the treatment exhibited safety and yielded therapeutic effects in some patients. The combined use of Bev and VEGFRs vaccination may enhance the anti-tumor effect in malignant gliomas. In this pilot study, the adverse event profile in patients treated with Bev after the vaccination was investigated to establish this treatment strategy, in comparison to those treated with Bev collected from the published data or treated with the vaccination alone. In our previous clinical studies on patients with malignant gliomas, Bev was administered to 13 patients after VEGFRs vaccinations. One patient had a Grade 4 pulmonary embolism. Two patients had Grade 2 cerebral infarctions. There were no significant differences in the adverse event rates among patients treated with Bev, with the vaccination, or with Bev after the vaccination. Although careful observation is imperative for patients after this combination treatment strategy, VEGFRs-targeted vaccination may coexist with Bev for malignant gliomas.
Collapse
Affiliation(s)
- Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan; (R.T.); (Y.M.); (M.S.); (K.Y.)
| | - Yukina Morimoto
- Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan; (R.T.); (Y.M.); (M.S.); (K.Y.)
| | - Mizuto Sato
- Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan; (R.T.); (Y.M.); (M.S.); (K.Y.)
| | - Tetsuro Hikichi
- OncoTherapy Science Inc., 3-2-1, Sakado, Takatsu-ku, Kawasaki City, Kanagawa 213-0012, Japan;
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan; (R.T.); (Y.M.); (M.S.); (K.Y.)
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, Tokyo 160-8582, Japan; (R.T.); (Y.M.); (M.S.); (K.Y.)
- Correspondence:
| |
Collapse
|
32
|
Ogasawara M, Miyashita M, Yamagishi Y, Ota S. Immunotherapy employing dendritic cell vaccination for patients with advanced or relapsed esophageal cancer. Ther Apher Dial 2020; 24:482-491. [PMID: 32524770 DOI: 10.1111/1744-9987.13542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The prognosis of patients with advanced esophageal cancer is poor despite the recent introduction of immune checkpoint inhibitors. In the present pilot study, we have evaluated the safety and the feasibility of Wilms' tumor 1 (WT1) peptide-pulsed dendritic cell (DC) vaccination in combination with OK-432 in patients with advanced or relapsed esophageal cancer. Fifteen eligible patients were enrolled. No severe adverse events related to the vaccinations were observed. Objective response rate and disease control rate were 20% and 40%, respectively. Median progression free survival and overall survival was 4.1 months and 7.0 months, respectively. WT1 peptide-pulsed DC vaccinations augmented WT1specific immunity, which might be related to clinical outcome. These results indicate that DC-based immunotherapy combined with a conventional chemotherapy is safe and feasible for patients in advanced stage of esophageal cancer.
Collapse
Affiliation(s)
- Masahiro Ogasawara
- Department of Internal Medicine, Sapporo Hokuyu Hospital, Sapporo, Japan.,Institute for Artificial Organ, Transplantation and Cell Therapy, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Mamiko Miyashita
- Institute for Artificial Organ, Transplantation and Cell Therapy, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Yuka Yamagishi
- Cell Processing Center, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Shuichi Ota
- Department of Internal Medicine, Sapporo Hokuyu Hospital, Sapporo, Japan
| |
Collapse
|
33
|
Demir Y, Türkeş C, Beydemir Ş. Molecular Docking Studies and Inhibition Properties of Some Antineoplastic Agents against Paraoxonase-I. Anticancer Agents Med Chem 2020; 20:887-896. [DOI: 10.2174/1871520620666200218110645] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/15/2019] [Accepted: 01/27/2020] [Indexed: 01/28/2023]
Abstract
Background:
Currently, most of the drugs used in clinical applications show their pharmacological
influences by inhibiting or activating enzymes. Therefore, enzyme inhibitors have an essential place in the drug
design for many diseases.
Objective:
The current study aimed to contribute to this growing drug design field (i.e., medicine discovery and
development) by analyzing enzyme-drug interactions.
Methods:
For this reason, Paraoxonase-I (PON1) enzyme was purified from fresh human serum by using rapid
chromatographic techniques. Additionally, the inhibition effects of some antineoplastic agents were researched
on the PON1.
Results:
The enzyme was obtained with a specific activity of 2603.57 EU/mg protein. IC50 values for pemetrexed
disodium, irinotecan hydrochloride, dacarbazine, and azacitidine were determined to be 9.63μM,
30.13μM, 53.31μM, and 21.00mM, respectively. These agents found to strongly inhibit PON1, with Ki constants
ranging from 8.29±1.47μM to 23.34±2.71mM. Dacarbazine and azacitidine showed non-competitive inhibition,
while other drugs showed competitive inhibition. Furthermore, molecular docking was performed using maestro
for these agents. Among these, irinotecan hydrochloride and pemetrexed disodium possess the binding energy of
-5.46 and -8.43 kcal/mol, respectively.
Conclusion:
The interaction studies indicated that these agents with the PON1 possess binding affinity.
Collapse
Affiliation(s)
- Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Gole Vocational High School, Ardahan University, 75700, Ardahan, Turkey
| | - Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yildirim University, 24100, Erzincan, Turkey
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, 26470, Eskisehir, Turkey
| |
Collapse
|
34
|
Daiko H, Marafioti T, Fujiwara T, Shirakawa Y, Nakatsura T, Kato K, Puccio I, Hikichi T, Yoshimura S, Nakagawa T, Furukawa M, Stoeber K, Nagira M, Ide N, Kojima T. Exploratory open-label clinical study to determine the S-588410 cancer peptide vaccine-induced tumor-infiltrating lymphocytes and changes in the tumor microenvironment in esophageal cancer patients. Cancer Immunol Immunother 2020; 69:2247-2257. [PMID: 32500232 PMCID: PMC7568713 DOI: 10.1007/s00262-020-02619-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/20/2020] [Indexed: 12/24/2022]
Abstract
Cancer vaccines induce cancer-specific T-cells capable of eradicating cancer cells. The impact of cancer peptide vaccines (CPV) on the tumor microenvironment (TME) remains unclear. S-588410 is a CPV comprising five human leukocyte antigen (HLA)-A*24:02-restricted peptides derived from five cancer testis antigens, DEPDC1, MPHOSPH1, URLC10, CDCA1 and KOC1, which are overexpressed in esophageal cancer. This exploratory study investigated the immunologic mechanism of action of subcutaneous S-588410 emulsified with MONTANIDE ISA51VG adjuvant (median: 5 doses) by analyzing the expression of immune-related molecules, cytotoxic T-lymphocyte (CTL) response and T-lymphocytes bearing peptide-specific T-cell receptor (TCR) sequencing in tumor tissue or blood samples from 15 participants with HLA-A*24:02-positive esophageal cancer. Densities of CD8+, CD8+ Granzyme B+, CD8+ programmed death-1-positive (PD-1+) and programmed death-ligand 1-positive (PD-L1+) cells were higher in post- versus pre-vaccination tumor tissue. CTL response was induced in all patients for at least one of five peptides. The same sequences of peptide-specific TCRs were identified in post-vaccination T-lymphocytes derived from both tumor tissue and blood, suggesting that functional peptide-specific CTLs infiltrate tumor tissue after vaccination. Twelve (80%) participants had treatment-related adverse events (AEs). Injection site reaction was the most frequently reported AE (grade 1, n = 1; grade 2, n = 11). In conclusion, S-588410 induces a tumor immune response in esophageal cancer. Induction of CD8+ PD-1+ tumor-infiltrating lymphocytes and PD-L1 expression in the TME by vaccination suggests S-588410 in combination with anti-PD-(L)1 antibodies may offer a clinically useful therapy.Trial registration UMIN-CTR registration identifier: UMIN000023324.
Collapse
Affiliation(s)
- H Daiko
- Esophageal Surgery Division, National Cancer Center Hospital, 5-1-1, Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
| | - T Marafioti
- Department of Cellular Pathology, University College London Hospital, London, UK
| | - T Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Y Shirakawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - T Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - K Kato
- Gastrointestinal Medical Oncology Division, National Cancer Center Hospital, Tokyo, Japan
| | - I Puccio
- Department of Cellular Pathology, University College London Hospital, London, UK
| | - T Hikichi
- R&D Department, Cancer Precision Medicine, Inc., Kawasaki, Japan
| | - S Yoshimura
- R&D Department, Cancer Precision Medicine, Inc., Kawasaki, Japan
| | - T Nakagawa
- Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Toyonaka, Japan
| | - M Furukawa
- Biostatistics Department, Shionogi & Co., Ltd., Osaka, Japan
| | - K Stoeber
- Business Development, Shionogi & Co., Ltd., London, UK
| | - M Nagira
- Drug Discovery and Disease Research Laboratory, Shionogi & Co., Ltd., Toyonaka, Japan
| | - N Ide
- Project Management Department, Shionogi & Co., Ltd., Osaka, Japan
| | - T Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
35
|
Immunotherapy in gastrointestinal cancer: The current scenario and future perspectives. Cancer Treat Rev 2020; 88:102030. [PMID: 32505807 DOI: 10.1016/j.ctrv.2020.102030] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/07/2020] [Accepted: 05/09/2020] [Indexed: 02/06/2023]
Abstract
Gastrointestinal cancers include colorectal, gastric, oesophageal, pancreatic and liver cancers. They continue to be a significant cause of mortality and morbidity worldwide. Current treatment strategies include chemotherapy, surgery, radiotherapy and targeted therapies. Immunotherapy has recently been incorporated in treatment regimens for some gastrointestinal malignancies and research into different immune modifying treatments is being carried out in this context. Approaches to immune modulation such as vaccination, adoptive cell therapy and checkpoint inhibition have shown varying clinical benefit, with most of the benefit seen in checkpoint inhibition. This review summarises recent advances and future direction of immunotherapy in patients with gastrointestinal malignancies.
Collapse
|
36
|
Nakajima M, Hazama S, Tamada K, Udaka K, Kouki Y, Uematsu T, Arima H, Saito A, Doi S, Matsui H, Shindo Y, Matsukuma S, Kanekiyo S, Tokumitsu Y, Tomochika S, Iida M, Yoshida S, Nakagami Y, Suzuki N, Takeda S, Yamamoto S, Yoshino S, Ueno T, Nagano H. A phase I study of multi-HLA-binding peptides derived from heat shock protein 70/glypican-3 and a novel combination adjuvant of hLAG-3Ig and Poly-ICLC for patients with metastatic gastrointestinal cancers: YNP01 trial. Cancer Immunol Immunother 2020; 69:1651-1662. [PMID: 32219501 PMCID: PMC7347520 DOI: 10.1007/s00262-020-02518-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/12/2020] [Indexed: 12/14/2022]
Abstract
Background This phase I study aimed to evaluate the safety, peptide-specific immune responses, and anti-tumor effects of a novel vaccination therapy comprising multi-HLA-binding heat shock protein (HSP) 70/glypican-3 (GPC3) peptides and a novel adjuvant combination of hLAG-3Ig and Poly-ICLC against metastatic gastrointestinal cancers. Methods HSP70/GPC3 peptides with high binding affinities for three HLA types (A*24:02, A*02:01, and A*02:06) were identified with our peptide prediction system. The peptides were intradermally administered with combined adjuvants on a weekly basis. This study was a phase I dose escalation clinical trial, which was carried out in a three patients’ cohort; in total, 11 patients were enrolled for the recommended dose. Results Seventeen patients received this vaccination therapy without dose-limiting toxicity. All treatment-related adverse events were of grades 1 to 2. Peptide-specific CTL induction by HSP70 and GPC3 proteins was observed in 11 (64.7%) and 13 (76.5%) cases, respectively, regardless of the HLA type. Serum tumor marker levels were decreased in 10 cases (58.8%). Immunological analysis using PBMCs indicated that patients receiving dose level 3 presented with significantly reduced T cell immunoglobulin and mucin-domain containing-3 (TIM3)-expressing CD4 + T cells after one course of treatment. PD-1 or TIM3-expressing CD4 + T cells and T cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT)-expressing CD8 + T cells in PBMCs before vaccination were negative predictive factors for survival. Conclusions This novel peptide vaccination therapy was safe for patients with metastatic gastrointestinal cancers. Electronic supplementary material The online version of this article (10.1007/s00262-020-02518-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Masao Nakajima
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shoichi Hazama
- Department of Translational Research and Developmental Therapeutics Against Cancer, Yamaguchi University School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Koji Tamada
- Department of Immunology, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Keiko Udaka
- Department of Immunology, Kochi Medical School, Nankoku, Kochi, 783-8505, Japan
| | - Yasunobu Kouki
- Department of Pharmacy, Yamaguchi University Hospital, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Toshinari Uematsu
- Department of Pharmacy, Yamaguchi University Hospital, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Hideki Arima
- Department of Pharmacy, Yamaguchi University Hospital, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Akira Saito
- Department of AI Applied Quantitative Clinical Science, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku, Tokyo, 160-8402, Japan
| | - Shun Doi
- CYTLIMIC Inc, Shinagawa, Tokyo, 141-0021, Japan
| | - Hiroto Matsui
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yoshitaro Shindo
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Satoshi Matsukuma
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shinsuke Kanekiyo
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yukio Tokumitsu
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shinobu Tomochika
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Michihisa Iida
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shin Yoshida
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Yuki Nakagami
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shigeru Takeda
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shigeru Yamamoto
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Shigefumi Yoshino
- Oncology Center, Yamaguchi University Hospital, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Tomio Ueno
- Department of Digestive Surgery, Kawasaki University School of Medicine, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, Yamaguchi, 755-8505, Japan.
| |
Collapse
|
37
|
Abstract
Esophageal cancer (EC) seriously threatens human health, and a promising new avenue for EC treatment involves cancer immunotherapy. To improve the efficacy of EC immunotherapy and to develop novel strategies for EC prognosis prediction or clinical treatment, understanding the immune landscapes in EC is required. EC cells harbor abundant tumor antigens, including tumor-associated antigens and neoantigens, which have the ability to initiate dendritic cell-mediated tumor-killing cytotoxic T lymphocytes in the early stage of cancer development. As EC cells battle the immune system, they obtain an ability to suppress antitumor immunity through immune checkpoints, secreted factors, and negative regulatory immune cells. Cancer-associated fibroblasts also contribute to the immune evasion of EC cells. Some factors of the immune landscape in EC tumor microenvironment are associated with cancer development, patient survival, or treatment response. Based on the immune landscape, peptide vaccines, adoptive T cell therapy, and immune checkpoint blockade can be used for EC immunotherapy. Combined strategies are required for better clinical outcome in EC. This review provides directions to design novel and effective strategies for prognosis prediction and immunotherapy in EC.
Collapse
Affiliation(s)
- Tu-Xiong Huang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathology and Shenzhen International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, P. R. China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathology and Shenzhen International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, Guangdong, P. R. China.
| |
Collapse
|
38
|
Lin W, Liu J, Chen J, Li J, Qiu S, Ma J, Lin X, Zhang L, Wu J. Peptides of tetraspanin oncoprotein CD151 trigger active immunity against primary tumour and experimental lung metastasis. EBioMedicine 2019; 49:133-144. [PMID: 31668880 PMCID: PMC6945203 DOI: 10.1016/j.ebiom.2019.10.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Active immunotherapy is an effective, long-lasting, cheap, and safe approach to suppress cancer progression; however, the key issue is to develop appropriate tumour vaccines. Oncoproteins are up-regulated under various stress conditions and promote cell survival. Oncoproteins and their immunogenic domains could serve well as tumour vaccines and prime the hosts' active anti-tumour immunity. METHODS Proteomic and bioinformatic analyses were performed to identify potential tumour associated antigens (TAAs). Then, peptides derived from CD151 were designed and synthesized according to the major histocompatibility complex (MHC) I binding and immunogenicity. Cytotoxicity assay, flow cytometry, immunohistochemistry, and in vivo bioluminescence imaging were performed to assess the active anti-tumour immunity triggered by CD151 peptides in H22 primary hepatoma and experimental 4T1 breast cancer lung metastasis models. FINDINGS CD151 was identified as an ideal TAA based on proteomic and bioinformatic analyses. CD151 peptides as tumour vaccines triggered active anti-tumour immunity against H22 hepatoma and the lung metastasis of 4T1 breast cancer in two mouse models through the activation of CD8+IFNγ+ lymphocytes and the subsequent targeted cytotoxicity. Further, the peptides suppressed the negative regulators, myeloid-derived suppressor cells. Survival was prolonged for mice with lung metastases from CD151 peptide-immunised groups. INTERPRETATION The up-regulated oncoproteins in 8 Gy-irradiated tumour cells are good candidates for designing immunogenic peptides as tumour vaccines. Anti-tumour active immunity primed by peptides from CD151 may be an effective and safe approach to suppress cancer progression.
Collapse
Affiliation(s)
- Wanzun Lin
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, 420 Fuma Rd, Fuzhou 350014, China
| | - Jun Liu
- Department of Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, 420 Fuma Rd, Fuzhou 350014, China
| | - Juhui Chen
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, 420 Fuma Rd, Fuzhou 350014, China
| | - Jiancheng Li
- Department of Chest Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, 420 Fuma Rd, Fuzhou 350014, China
| | - Sufang Qiu
- Department of Head & Neck Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, 420 Fuma Rd, Fuzhou 350014, China
| | - Jiayu Ma
- Department of Chest Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, 420 Fuma Rd, Fuzhou 350014, China
| | - Xiandong Lin
- Laboratory of Radiobiology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, 420 Fuma Rd, Fuzhou 350014, China
| | - Lurong Zhang
- Laboratory of Radiobiology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, 420 Fuma Rd, Fuzhou 350014, China.
| | - Junxin Wu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, 420 Fuma Rd, Fuzhou 350014, China.
| |
Collapse
|
39
|
Sakakibara T, Ozawa S, Oguma J, Nakui M, Yamamoto S, Makuuchi H, Kajiwara H, Nakamura N. Prognostic significance of IMP-3 expression pattern in esophageal squamous cell carcinoma. J Thorac Dis 2019; 11:3776-3784. [PMID: 31656650 DOI: 10.21037/jtd.2019.09.25] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Esophageal cancer is one of the most malignant gastroenterological cancers. To improve the treatment outcomes of patients with esophageal squamous cell carcinoma (ESCC), a biomarker capable of predicting the malignant potential of the cancer cells is needed. The aim of the present study was to investigate the relationship between the expression pattern of insulin-like growth factor II m-RNA-binding protein 3 (IMP3), a promising cancer testis antigen for peptide vaccine therapy, in ESCC tumors and the outcomes of patients with ESCC. Methods One hundred and seventy patients with ESCC who underwent a radical transthoracic esophagectomy between 2003 and 2005 at Tokai University Hospital were investigated. IMP3 expression was immunohistochemically analyzed using sections from surgically resected tumor specimens and metastatic lymph nodes. Results Of the 170 patients, 160 patients (94%) exhibited IMP3 positivity in the cytoplasm of their cancer cells (IMP3-positive group), while 10 patients (6%) were IMP3-negative (IMP3-negative group). No significant difference in the overall survival curves were observed between the IMP3-positive and IMP3-negative groups. When the survival analysis was confined to the 160 IMP3-positive patients, however, an invasive front-type IMP3 expression pattern (IF-type) was seen in 46 patients (29%) and a diffuse-type pattern (D-type) was seen in 114 patients (71%). A multivariate analysis also showed that an IF-type was a prognostic factor (HR =1.618, P=0.049). The overall survival curve for patients with an IF-type was significantly worse than that of D-type patients (P=0.001). Conclusions An IF-type pattern of IMP3 expression might predict a poor outcome in patients with ESCC.
Collapse
Affiliation(s)
- Terue Sakakibara
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Soji Ozawa
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Junya Oguma
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Minoru Nakui
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Soichiro Yamamoto
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Hiroyasu Makuuchi
- Department of Gastroenterological Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | - Hiroshi Kajiwara
- Department of Pathology, Tokai University School of Medicine, Kanagawa, Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, Kanagawa, Japan
| |
Collapse
|
40
|
Zhao Q, Yu J, Meng X. A good start of immunotherapy in esophageal cancer. Cancer Med 2019; 8:4519-4526. [PMID: 31231980 PMCID: PMC6712478 DOI: 10.1002/cam4.2336] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/25/2019] [Indexed: 12/11/2022] Open
Abstract
Considering the benefits of immunotherapy in advanced melanoma, non–small cell lung cancer, renal cell carcinoma, bladder cancers, and refractory Hodgkin lymphoma, we begin to consider whether immunotherapy is effective for esophageal cancer, which is extremely malignant and has a poor prognosis. There are a large number of clinical trials to study the application of immunotherapy such as immune checkpoint inhibitors, peptide vaccine, adoptive T cell transfer and oncolytic virus in esophageal cancer. Some already have preliminary results and show the advantages of immunotherapy in esophageal cancer, while others are still in progress. This review aims to introduce the feasibility and current status of immunotherapy in esophageal cancer.
Collapse
Affiliation(s)
- Qian Zhao
- Cheeloo College of MedicineShanDong UniversityJinanChina
| | - Jinming Yu
- Department of Radiation OncologyShandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical SciencesJinanChina
| | - Xue Meng
- Cheeloo College of MedicineShanDong UniversityJinanChina
- Department of Radiation OncologyShandong Cancer Hospital affiliated to Shandong University, Shandong Academy of Medical SciencesJinanChina
| |
Collapse
|
41
|
Nemec PS, Kapatos A, Holmes JC, Stowe DM, Hess PR. Cancer-testis antigens in canine histiocytic sarcoma and other malignancies. Vet Comp Oncol 2019; 17:317-328. [PMID: 30854786 DOI: 10.1111/vco.12475] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/20/2022]
Abstract
Cancer-testis antigens (CTAs) are a category of self proteins aberrantly expressed in diverse malignancies, mostly solid tumours, due to epigenetic de-repression. Normally expressed only in fetal or gametogenic tissues, CTAs are tantalizing immunotherapy targets, since autoimmunity risks appear minimal. Few prevalent CTAs have been identified in human hematologic cancers, and just two in their veterinary counterparts. We sought to discover new CTAs in canine hematologic cancers such as histiocytic sarcoma (HS) and lymphoma to foster immunotherapy development. To accomplish this, the ligandome binding the dog leukocyte antigen (DLA)-88*508:01 class I allele overexpressed in an HS line was searched by mass spectrometry to identify possible CTA-derived peptides, which could serve as CD8+ T-cell epitopes. Twenty-two peptides mapped to 5 human CTAs and 12 additional proteins with CTA characteristics. Expression of five promising candidates was then evaluated in tumour and normal tissue by quantitative and end-point RT-PCR. The ortholog of an established CTA, IGF2BP3, had unexpectedly high expression in peripheral blood mononuclear cells (PBMCs). Four other testis-enhanced proteins were also assessed. AKR1E2, SPECC1 and TPX2 were expressed variably in HS and T-cell lymphoma biopsies, but also at high levels in critical tissues, including kidney, brain and marrow, diminishing their utility. A more tissue-restricted candidate, NT5C1B, was detected in T-cell lymphomas, but also at low levels in some normal dog tissues. These results illustrate the feasibility of discovering canine CTAs by a reverse approach, proceeding from identification of MHC class I-presented peptides to a comparative RNA expression survey of tumours and normal tissues.
Collapse
Affiliation(s)
- Paige S Nemec
- Department of Clinical Sciences, North Carolina State University, College of Veterinary Medicine, Raleigh, North Carolina
| | - Alexander Kapatos
- Department of Clinical Sciences, North Carolina State University, College of Veterinary Medicine, Raleigh, North Carolina
| | - Jennifer C Holmes
- Department of Clinical Sciences, North Carolina State University, College of Veterinary Medicine, Raleigh, North Carolina
| | - Devorah M Stowe
- Department of Population, Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina
| | - Paul R Hess
- Department of Clinical Sciences, North Carolina State University, College of Veterinary Medicine, Raleigh, North Carolina
| |
Collapse
|
42
|
Abstract
With the spotlight on cancer immunotherapy and the expanding use of immune checkpoint inhibitors, strategies to improve the response rate and duration of current cancer immunotherapeutics are highly sought. In that sense, investigators around the globe have been putting spurs on the development of effective cancer vaccines in humans after decades of efforts that led to limited clinical success. In more than three decades of research in pursuit of targeted and personalized immunotherapy, several platforms have been incorporated into the list of cancer vaccines from live viral or bacterial agents harboring antigens to synthetic peptides with the hope of stronger and durable immune responses that will tackle cancers better. Unlike adoptive cell therapy, cancer vaccines can take advantage of using a patient's entire immune system that can include more than engineered receptors or ligands in developing antigen-specific responses. Advances in molecular technology also secured the use of genetically modified genes or proteins of interest to enhance the chance of stronger immune responses. The formulation of vaccines to increase chances of immune recognition such as nanoparticles for peptide delivery is another area of great interest. Studies indicate that cancer vaccines alone may elicit tumor-specific cellular or humoral responses in immunologic assays and even regression or shrinkage of the cancer in select trials, but novel strategies, especially in combination with other cancer therapies, are under study and are likely to be critical to achieve and optimize reliable objective responses and survival benefit. In this review, cancer vaccine platforms with different approaches to deliver tumor antigens and boost immunity are discussed with the intention of summarizing what we know and what we need to improve in the clinical trial setting.
Collapse
Affiliation(s)
- Hoyoung M. Maeng
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jay A. Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
43
|
Wei X, Chen F, Xin K, Wang Q, Yu L, Liu B, Liu Q. Cancer-Testis Antigen Peptide Vaccine for Cancer Immunotherapy: Progress and Prospects. Transl Oncol 2019; 12:733-738. [PMID: 30877975 PMCID: PMC6423365 DOI: 10.1016/j.tranon.2019.02.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 02/19/2019] [Accepted: 02/19/2019] [Indexed: 12/31/2022] Open
Abstract
Cancer vaccines, including peptide-based vaccines, have been considered a key tool of effective and protective cancer immunotherapy because of their capacity to provide long-term clinical benefit for tumors. Among a large number of explorations of peptide antigen-based vaccines, cancer-testis antigens (CTAs), which are activated in cancers but silenced in normal tissues (except testis tissue), are considered as ideal targets. Currently, personalized treatment for cancer has become a trend due to its superior clinical efficacy. Thus, we envisage rational selection of CTA peptides to design "personalized" CTA peptide vaccines. This review summarizes the advances in CTA peptide vaccine research and discusses the feasibility of establishing "personalized" CTA peptide vaccines.
Collapse
Affiliation(s)
- Xiao Wei
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing Medical University
| | - Fangjun Chen
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University
| | - Kai Xin
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University
| | - Qin Wang
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University
| | - Lixia Yu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Nanjing Medical University; The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University
| | - Qin Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University.
| |
Collapse
|
44
|
Kikuchi R, Ueda R, Saito K, Shibao S, Nagashima H, Tamura R, Morimoto Y, Sasaki H, Noji S, Kawakami Y, Yoshida K, Toda M. A Pilot Study of Vaccine Therapy with Multiple Glioma Oncoantigen/Glioma Angiogenesis-Associated Antigen Peptides for Patients with Recurrent/Progressive High-Grade Glioma. J Clin Med 2019; 8:jcm8020263. [PMID: 30791546 PMCID: PMC6406695 DOI: 10.3390/jcm8020263] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 01/01/2023] Open
Abstract
High-grade gliomas (HGGs) carry a dismal prognosis despite current treatments. We previously confirmed the safety and immunogenicity of a vaccine treatment targeting tumor angiogenesis with synthetic peptides, for vascular endothelial growth factor receptor (VEGFR) epitopes in recurrent HGG patients. In this study, we evaluated a novel vaccine therapy targeting not only tumor vasculature but also tumor cells, using multiple glioma oncoantigen (GOA)/glioma angiogenesis-associated antigen (GAAA) peptides in HLA-A2402+ recurrent/progressive HGG patients. The vaccine included peptide epitopes from four GOAs (LY6K, DEPDC1, KIF20A, and FOXM1) and two GAAAs (VEGFR1 and VEGFR2). Ten patients received subcutaneous vaccinations. The primary endpoint was the safety of the treatment. T-lymphocyte responses against GOA/GAAA epitopes and treatment response were evaluated secondarily. The treatment was well tolerated without any severe systemic adverse events. The vaccinations induced immunoreactivity to at least three vaccine-targeted GOA/GAAA in all six evaluable patients. The median overall survival time in all patients was 9.2 months. Five achieved progression-free status lasting at least six months. Two recurrent glioblastoma patients demonstrated stable disease. One patient with anaplastic oligoastrocytoma achieved complete response nine months after the vaccination. Taken together, this regimen was well tolerated and induced robust GOA/GAAA-specific T-lymphocyte responses in recurrent/progressive HGG patients.
Collapse
Affiliation(s)
- Ryogo Kikuchi
- Department of Neurosurgery, Hiratsuka City Hospital, Hiratsuka, Kanagawa 254-0019, Japan.
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Ryo Ueda
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
- Department of Neurosurgery, Kawasaki Municipal Hospital, Kawasaki, Kanagawa 210-0013, Japan.
| | - Katsuya Saito
- Department of Neurosurgery, Ashikaga Red Cross Hospital, Ashikaga, Tochigi 326-0843, Japan.
| | - Shunsuke Shibao
- Department of Neurosurgery, Ashikaga Red Cross Hospital, Ashikaga, Tochigi 326-0843, Japan.
| | - Hideaki Nagashima
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Yukina Morimoto
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Hikaru Sasaki
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Shinobu Noji
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjku, Tokyo 160-8587, Japan.
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, Keio University School of Medicine, Shinjku, Tokyo 160-8587, Japan.
| | - Kazunari Yoshida
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, Shinjuku, Tokyo 160-8587, Japan.
| |
Collapse
|
45
|
Zhang Y, Zhang Y, Zhang L. Expression of cancer-testis antigens in esophageal cancer and their progress in immunotherapy. J Cancer Res Clin Oncol 2019; 145:281-291. [PMID: 30656409 PMCID: PMC6373256 DOI: 10.1007/s00432-019-02840-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/03/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE Esophageal cancer is a common disease in China with low survival rate due to no obvious early symptoms and lack of effective screening strategies. Traditional treatments usually do not produce desirable results in patients with advanced esophageal cancer, so immunotherapy which relies on tumor-related antigens is needed to combat low survival rates effectively. Cancer-testis antigens (CTA), a large family of tumor-related antigens, have a strong in vivo immunogenicity and tumor-restricted expressing patterns in normal adult tissues. These two characteristics are ideal features of anticancer immunotherapy targets and, therefore, promoted the development of some studies of CTA-based therapy. To provide ideas for the role of the cancer-testis antigens MAGE-A, NY-ESO-1, LAGE-1, and TTK in esophageal cancer, we summarized their expression, prognostic value, and development in immunotherapy. METHODS The relevant literature from PubMed is reviewed in this study. RESULTS In esophageal cancer, although the relationship between expression of MAGE-A, NY-ESO-1, LAGE-1, and TTK and prognosis value is still in a controversial situation, MAGE-A, NY-ESO-1, LAGE-1, and TTK are highly expressed and can induce specific CTL cells to produce particular killing effect on tumor cells, and some clinical trials have demonstrated that immunotherapy for esophageal cancer patients is effective and safe, which provides a new therapeutic strategy for the treatment of esophageal cancer in the future. CONCLUSION In this review, we summarize expression and prognostic value of MAGE-A, NY-ESO-1, LAGE-1, and TTK in esophageal cancer and point out recent advances in immunotherapy about them.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Yuxin Zhang
- Hepatic Surgery Center, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China
| | - Li Zhang
- Department of Oncology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.
| |
Collapse
|
46
|
Chai SJ, Fong SCY, Gan CP, Pua KC, Lim PVH, Lau SH, Zain RB, Abraham T, Ismail SM, Abdul Rahman ZA, Ponniah S, Patel V, Cheong SC, Lim KP. In vitro evaluation of dual-antigenic PV1 peptide vaccine in head and neck cancer patients. Hum Vaccin Immunother 2018; 15:167-178. [PMID: 30193086 DOI: 10.1080/21645515.2018.1520584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Peptide vaccines derived from tumour-associated antigens have been used as an immunotherapeutic approach to induce specific cytotoxic immune response against tumour. We previously identified that MAGED4B and FJX1 proteins are overexpressed in HNSCC patients; and further demonstrated that two HLA-A2-restricted 9-11 amino acid peptides derived from these proteins were able to induce anti-tumour immune responses in vitro independently using PBMCs isolated from these patients. In this study, we evaluated the immunogenicity and efficacy of a dual-antigenic peptide vaccine (PV1), comprised of MAGED4B and FJX1 peptides in HNSCC patients. We first demonstrated that 94.8% of HNSCC patients expressed MAGED4B and/or FJX1 by immunohistochemistry, suggesting that PV1 could benefit the majority of HNSCC patients. The presence of pre-existing MAGED4B and FJX1-specific T-cells was detected using a HLA-A2 dimer assay and efficacy of PV1 to induce T-cell to secrete cytotoxic cytokine was evaluated using ELISPOT assay. Pre-existing PV1-specific T-cells were detected in all patients. Notably, we demonstrated that patients' T-cells were able to secrete cytotoxic cytokines upon exposure to target cells expressing the respective antigen post PV1 stimulation. Furthermore, patients with high expression of MAGED4B and FJX1 in their tumours were more responsive to PV1 stimulation, demonstrating the specificity of the PV1 peptide vaccine. Additionally, we also demonstrated the expression of MAGED4B and FJX1 in breast, lung, colon, prostate and rectal cancer suggesting the potential use of PV1 in these cancers. In summary, PV1 could be a good vaccine candidate for the treatment of HNSCC patients and other cancers expressing these antigens.
Collapse
Affiliation(s)
- San Jiun Chai
- a Cancer Research Malaysia , Subang Jaya , Selangor , Malaysia
| | | | - Chai Phei Gan
- a Cancer Research Malaysia , Subang Jaya , Selangor , Malaysia
| | - Kin Choo Pua
- b Department of Otorhinolaryngology , Hospital Pulau Pinang , Penang , Malaysia
| | - Paul Vey Hong Lim
- c Ear, Nose and Throat Department , Tung Shin Hospital , Kuala Lumpur , Malaysia
| | - Shin Hin Lau
- d Stomatology Unit , Cancer Research Centre, Institute for Medical Research , Kuala Lumpur , Malaysia
| | - Rosnah Binti Zain
- e Faculty of Dentistry , MAHSA University , Selangor , Malaysia.,f Oral Cancer Research and Coordinating Centre, Faculty of Dentistry , University of Malaya , Kuala Lumpur , Malaysia
| | - Thomas Abraham
- g Department of Oral & Maxillofacial Surgery , Tengku Ampuan Rahimah Hospital , Klang , Malaysia
| | - Siti Mazlipah Ismail
- h Department of Oro-Maxillofacial Surgery and Medical Sciences, Faculty of Dentistry , University of Malaya , Kuala Lumpur , Malaysia
| | - Zainal Ariff Abdul Rahman
- h Department of Oro-Maxillofacial Surgery and Medical Sciences, Faculty of Dentistry , University of Malaya , Kuala Lumpur , Malaysia
| | - Sathibalan Ponniah
- i Cancer Vaccine Development Program, Department of Surgery , Uniformed Services University of the Health Sciences , Bethesda , MD , USA
| | - Vyomesh Patel
- a Cancer Research Malaysia , Subang Jaya , Selangor , Malaysia
| | - Sok Ching Cheong
- a Cancer Research Malaysia , Subang Jaya , Selangor , Malaysia.,h Department of Oro-Maxillofacial Surgery and Medical Sciences, Faculty of Dentistry , University of Malaya , Kuala Lumpur , Malaysia
| | - Kue Peng Lim
- a Cancer Research Malaysia , Subang Jaya , Selangor , Malaysia
| |
Collapse
|
47
|
He W, Chen L, Yuan K, Zhou Q, Peng L, Han Y. Gene set enrichment analysis and meta-analysis to identify six key genes regulating and controlling the prognosis of esophageal squamous cell carcinoma. J Thorac Dis 2018; 10:5714-5726. [PMID: 30505479 DOI: 10.21037/jtd.2018.09.55] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is a common malignancy with high mortality. Because of the lack of clarity in the relevant genes and mechanisms involved, and the current difficulty for oncotherapy in providing therapeutic solutions, there is an urgent need to study this matter. While gene probe studies have been used to select the most virulent genes and pathways, paucity of case controls during gene screening and lack of conclusive results to expound the etiology and pathogenesis of the disease, have reduced study reliability. Methods We chose six datasets from independent studies in the Gene Expression Omnibus (GEO) database and used gene set enrichment analysis and meta-analysis to select key genes and pathways. Results We found four down-regulated and four up-regulated pathways through gene set enrichment analysis, and 406 differential genes through meta-analysis. Based on The Cancer Genome Atlas (TCGA), 995 differentially expressed genes were screened out. Comparing the 406 gene set with the 995 gene set, we found 19 common genes, of which 6 had a common pathway and were screened out as key genes regulating and controlling the prognosis of ESCC. Conclusions Among the 19 genes, we found three genes that affect the chemotherapy of ESCC: BUB1B, BUB1, and TTK. Another three genes NDC1, NUP107, and NUP155 on the RNA transport pathway were also found. Altogether, these six genes are not only crucial in the development of ESCC, but also determine the prognosis of patients. The key genes and pathways identified in the present study will be used for the next stage in our study, which will involve gene elimination and other experimentation methods.
Collapse
Affiliation(s)
- Wenwu He
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Research Institute, Chengdu 610041, China
| | - Linxin Chen
- Department of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, China
| | - Kun Yuan
- Department of Anesthesiology, North Sichuan Medical College, Nanchong 637000, China
| | - Qiuxi Zhou
- Department of Respiratory Medicine, Nanchong Central Hospital, Nanchong 637000, China
| | - Lin Peng
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Research Institute, Chengdu 610041, China
| | - Yongtao Han
- Department of Thoracic Surgery, Sichuan Cancer Hospital and Research Institute, Chengdu 610041, China
| |
Collapse
|
48
|
Obara W, Hara I, Kato Y, Kato R, Inoue K, Sato F, Mimata H, Nakamura Y, Fujioka T. Immunotherapy with cancer peptides in combination with intravesical bacillus Calmette-Guerin for patients with non-muscle invasive bladder cancer. Cancer Immunol Immunother 2018; 67:1371-1380. [PMID: 29971464 PMCID: PMC11028097 DOI: 10.1007/s00262-018-2197-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 06/29/2018] [Indexed: 10/28/2022]
Abstract
PURPOSE A phase I study using two peptide vaccines derived from M phase phosphoprotein 1 (MPHOSPH1) and DEP domain containing 1 (DEPDC1) demonstrated promising results for the treatment of advanced bladder cancer. Therefore, we further tested the ability of these peptides to prevent recurrence after transurethral resection of the bladder tumor in patients with non-muscle invasive bladder cancer (NMIBC). MATERIALS AND METHODS 127 patients were enrolled in a multicenter, non-randomized phase II clinical trial. The primary endpoint was recurrence-free survival (RFS) rate, and secondary endpoints were safety and immunological response. HLA-A24-restricted peptides were subcutaneously administered in addition to intravesical BCG therapy. The exploratory endpoint evaluated differences of RFS rate between HLA-A*2402-positive (A24(+)) and -negative (A24(-)) groups. RESULTS A 2-year RFS rate in all patients was 74.0%. The RFS rate in the A24(+) group (n = 75) and in the A24(-) group (n = 52) were 76.0 and 71.2%, respectively. This vaccine therapy was well-tolerated and feasible. MPHOSPH1 and DEPDC1 peptide-specific cytotoxic T lymphocyte responses were observed in 75.8 and 77.5% of the A24(+) group, respectively. Patients having both peptide-specific CTL responses showed significantly better RFS than patients without CTL response (P = 0.014). In the A24(+) group, patients who had positive reaction at the injection sites (RAI) had significantly lower rates of recurrence than RAI-negative patients (P = 0.0019). CONCLUSIONS Cancer peptide vaccines in combination with intravesical BCG therapy demonstrated good immunogenicity and safety, and may provide benefit for preventing recurrence of NMIBC.
Collapse
Affiliation(s)
- Wataru Obara
- Department of Urology, Iwate Medical University School of Medicine, 19-1 Uchimaru, Morioka, 020-8505, Japan.
| | - Isao Hara
- Department of Urology, Wakayama Medical University, Wakayama, Japan
| | - Yoichiro Kato
- Department of Urology, Iwate Medical University School of Medicine, 19-1 Uchimaru, Morioka, 020-8505, Japan
| | - Renpei Kato
- Department of Urology, Iwate Medical University School of Medicine, 19-1 Uchimaru, Morioka, 020-8505, Japan
| | - Keiji Inoue
- Department of Urology, Kochi Medical School, Nankoku, Japan
| | - Fuminori Sato
- Department of Urology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Hiromitsu Mimata
- Department of Urology, Faculty of Medicine, Oita University, Yufu, Japan
| | - Yusuke Nakamura
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - Tomoaki Fujioka
- Department of Urology, Iwate Medical University School of Medicine, 19-1 Uchimaru, Morioka, 020-8505, Japan
| |
Collapse
|
49
|
Mimura K, Yamada L, Ujiie D, Hayase S, Tada T, Hanayama H, Thar Min AK, Shibata M, Momma T, Saze Z, Ohki S, Kono K. Immunotherapy for esophageal squamous cell carcinoma: a review. Fukushima J Med Sci 2018; 64:46-53. [PMID: 30058598 DOI: 10.5387/fms.2018-09] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cancer vaccines and immune checkpoint inhibitors (ICI) have recently been employed as immunotherapies for esophageal squamous cell carcinoma (ESCC). Cancer vaccines for ESCC have yielded several promising results from investigator-initiated phase I and II clinical trials. Furthermore, a Randomized Controlled Trial as an adjuvant setting after curative surgery is in progress in Japan. On the other hand, ICI, anti-CTLA-4 mAb and anti-PD-1 mAb, have demonstrated tumor shrinkage and improved overall survival in patients with multiple cancer types. For ESCC, several clinical trials using anti-PD-1/anti-PD-L1 mAb are underway with several recent promising results. In this review, cancer vaccines and ICI are discussed as novel therapeutic strategies for ESCC.
Collapse
Affiliation(s)
- Kosaku Mimura
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University.,Department of Advanced Cancer Immunotherapy, Fukushima Medical University.,Department of Progressive DOHaD Research, Fukushima Medical University
| | - Leo Yamada
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Daisuke Ujiie
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Suguru Hayase
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Takeshi Tada
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Hiroyuki Hanayama
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Aung Kyi Thar Min
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Masahiko Shibata
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University.,Department of Advanced Cancer Immunotherapy, Fukushima Medical University
| | - Tomoyuki Momma
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Zenichiro Saze
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Shinji Ohki
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University
| |
Collapse
|
50
|
Takeda K, Kitaura K, Suzuki R, Owada Y, Muto S, Okabe N, Hasegawa T, Osugi J, Hoshino M, Tsunoda T, Okumura K, Suzuki H. Quantitative T-cell repertoire analysis of peripheral blood mononuclear cells from lung cancer patients following long-term cancer peptide vaccination. Cancer Immunol Immunother 2018; 67:949-964. [PMID: 29568993 PMCID: PMC11028142 DOI: 10.1007/s00262-018-2152-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 03/13/2018] [Indexed: 12/14/2022]
Abstract
Therapeutic cancer peptide vaccination is an immunotherapy designed to elicit cytotoxic T-lymphocyte (CTL) responses in patients. A number of therapeutic vaccination trials have been performed, nevertheless there are only a few reports that have analyzed the T-cell receptors (TCRs) expressed on tumor antigen-specific CTLs. Here, we use next-generation sequencing (NGS) to analyze TCRs of vaccine-induced CTL clones and the TCR repertoire of bulk T cells in peripheral blood mononuclear cells (PBMCs) from two lung cancer patients over the course of long-term vaccine therapy. In both patients, vaccination with two epitope peptides derived from cancer/testis antigens (upregulated lung cancer 10 (URLC10) and cell division associated 1 (CDCA1)) induced specific CTLs expressing various TCRs. All URLC10-specific CTL clones tested showed Ca2+ influx, IFN-γ production, and cytotoxicity when co-cultured with URLC10-pulsed tumor cells. Moreover, in CTL clones that were not stained with the URLC10/MHC-multimer, the CD3 ζ chain was not phosphorylated. NGS of the TCR repertoire of bulk PBMCs demonstrated that the frequency of vaccine peptide-specific CTL clones was near the minimum detectable threshold level. These results demonstrate that vaccination induces antigen-specific CTLs expressing various TCRs at different time points in cancer patients, and that some CTL clones are maintained in PBMCs during long-term treatment, including some with TCRs that do not bind peptide/MHC-multimer.
Collapse
Affiliation(s)
- Kazuyoshi Takeda
- Division of Cell Biology, Biomedical Research Center, Graduate School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan.
- Department of Biofunctional Micribiota, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Kazutaka Kitaura
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Sagamihara, Kanagawa, 252-0392, Japan
| | - Ryuji Suzuki
- Department of Rheumatology and Clinical Immunology, Clinical Research Center for Allergy and Rheumatology, Sagamihara National Hospital, National Hospital Organization, Sagamihara, Kanagawa, 252-0392, Japan
| | - Yuki Owada
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Satoshi Muto
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Naoyuki Okabe
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Takeo Hasegawa
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Jun Osugi
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Mika Hoshino
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| | - Takuya Tsunoda
- Department of Clinical Immuno-oncology, Showa University, Setagaya-ku, Tokyo, 157-8577, Japan
| | - Ko Okumura
- Department of Biofunctional Micribiota, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, 113-8421, Japan
- Atopy (Allergy) Research Center, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroyuki Suzuki
- Department of Chest Surgery, School of Medicine, Fukushima Medical University, Fukushima, 960-1295, Japan
| |
Collapse
|