1
|
Nojima I, Eikawa S, Tomonobu N, Hada Y, Kajitani N, Teshigawara S, Miyamoto S, Tone A, Uchida HA, Nakatsuka A, Eguchi J, Shikata K, Udono H, Wada J. Dysfunction of CD8 + PD-1 + T cells in type 2 diabetes caused by the impairment of metabolism-immune axis. Sci Rep 2020; 10:14928. [PMID: 32913271 PMCID: PMC7484782 DOI: 10.1038/s41598-020-71946-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 08/24/2020] [Indexed: 11/10/2022] Open
Abstract
The metabolic changes and dysfunction in CD8 + T cells may be involved in tumor progression and susceptibility to virus infection in type 2 diabetes (T2D). In C57BL/6JJcl mice fed with high fat-high sucrose chow (HFS), multifunctionality of CD8 + splenic and tumor-infiltrating lymphocytes (TILs) was impaired and associated with enhanced tumor growth, which were inhibited by metformin. In CD8 + splenic T cells from the HFS mice, glycolysis/basal respiration ratio was significantly reduced and reversed by metformin. In the patients with T2D (DM), multifunctionality of circulating CD8 + PD-1 + T cells stimulated with PMA/ionomycin as well as with HLA-A*24:02 CMV peptide was dampened, while metformin recovered multifunctionality. Both glycolysis and basal respiration were reduced in DM, and glycolysis was increased by metformin. The disturbance of the link between metabolism and immune function in CD8 + PD-1 + T cells in T2D was proved by recovery of antigen-specific and non-specific cytokine production via metformin-mediated increase in glycolytic activity.
Collapse
Affiliation(s)
- Ichiro Nojima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kitaku, Okayama City, 700-8558, Japan
| | - Shingo Eikawa
- Department of Hematology/Oncology, Hess Cancer Institute, Icahn School of Medicine At Mount Sinai, New York, USA.,Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Nahoko Tomonobu
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshiko Hada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kitaku, Okayama City, 700-8558, Japan
| | - Nobuo Kajitani
- Department of Internal Medicine, Diabetes Center, Okayama City Hospital, Okayama, Japan
| | | | - Satoshi Miyamoto
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Atsuhito Tone
- Diabetes Center, Okayama Saiseikai General Hospital, Okayama, Japan
| | - Haruhito A Uchida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Atsuko Nakatsuka
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kitaku, Okayama City, 700-8558, Japan
| | - Jun Eguchi
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kitaku, Okayama City, 700-8558, Japan
| | - Kenichi Shikata
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Heiichiro Udono
- Department of Immunology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kitaku, Okayama City, 700-8558, Japan.
| |
Collapse
|
2
|
Identification of a tumor-specific allo-HLA-restricted γδTCR. Blood Adv 2020; 3:2870-2882. [PMID: 31585951 DOI: 10.1182/bloodadvances.2019032409] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/28/2019] [Indexed: 12/25/2022] Open
Abstract
γδT cells are key players in cancer immune surveillance because of their ability to recognize malignant transformed cells, which makes them promising therapeutic tools in the treatment of cancer. However, the biological mechanisms of how γδT-cell receptors (TCRs) interact with their ligands are poorly understood. Within this context, we describe the novel allo-HLA-restricted and CD8α-dependent Vγ5Vδ1TCR. In contrast to the previous assumption of the general allo-HLA reactivity of a minor fraction of γδTCRs, we show that classic anti-HLA-directed, γδTCR-mediated reactivity can selectively act on hematological and solid tumor cells, while not harming healthy tissues in vitro and in vivo. We identified the molecular interface with proximity to the peptide-binding groove of HLA-A*24:02 as the essential determinant for recognition and describe the critical role of CD8 as a coreceptor. We conclude that alloreactive γδT-cell repertoires provide therapeutic opportunities, either within the context of haplotransplantation or as individual γδTCRs for genetic engineering of tumor-reactive T cells.
Collapse
|
3
|
Zhang J, He J, Li J, Zhou Q, Chen H, Zheng Z, Chen Q, Chen D, Chen J. The immunogenicity and protective immunity of multi-epitopes DNA prime-protein boost vaccines encoding Amastin-Kmp-11, Kmp11-Gp63 and Amastin-Gp63 against visceral leishmaniasis. PLoS One 2020; 15:e0230381. [PMID: 32176727 PMCID: PMC7075555 DOI: 10.1371/journal.pone.0230381] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 02/28/2020] [Indexed: 11/27/2022] Open
Abstract
Visceral leishmaniasis (VL) is the most fatal form of leishmaniasis if left untreated and 50,000 to 90,000 new cases of VL occur worldwide each year. Although various vaccines had been studied in animal models, none of them was eligible to prevent human from infections. In this study, according to the silico analysis of Leishmania Amastin, Kmp-11 and Gp63 protein, dominant epitope sequences of these proteins were selected and linked to construct dominant multi-epitopes DNA and protein vaccines (Amastin-Kmp-11, Amastin-Gp63 and Kmp-11-Gp63) against VL. BALB/c mice were immunized with a DNA prime-protein boost immunization strategy and challenged with a new Leishmania parasite strain isolated from a VL patient. After immunization, the results including specific antibody titers, IL-4 and TNF-α levels, and CD4 and CD8 T cell proportion suggested the potent immunogenicity of the three vaccines. After infection, the results of spleen parasite burdens in the three vaccine groups were significantly lower than those of control groups, and the parasite reduction rates of Amastin-Kmp-11, Amastin-Gp63 and Kmp-11-Gp63 groups were 89.38%, 91.01% and 88.42%, respectively. Spleen smear observation and liver histopathological changes showed that all vaccine groups could produce significant immunoprotection against VL and Amastin-Gp63 vaccine was the best. In conclusion, our work demonstrated that the three dominant multi-epitopes Amastin-Kmp-11, Amastin-Gp63 and Kmp-11-Gp63 DNA prime-protein boost vaccines might be new vaccine candidates for VL, and the Amastin-Gp63 vaccine have best efficacy.
Collapse
Affiliation(s)
- Jianhui Zhang
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jinlei He
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jiao Li
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Qi Zhou
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Han Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Zhiwan Zheng
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Qiwei Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Dali Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jianping Chen
- Department of Pathogenic Biology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China.,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Shimizu Y, Yoshikawa T, Kojima T, Shoda K, Nosaka K, Mizuno S, Wada S, Fujimoto Y, Sasada T, Kohashi K, Bando H, Endo I, Nakatsura T. Heat shock protein 105 peptide vaccine could induce antitumor immune reactions in a phase I clinical trial. Cancer Sci 2019; 110:3049-3060. [PMID: 31390678 PMCID: PMC6778658 DOI: 10.1111/cas.14165] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/02/2019] [Accepted: 08/05/2019] [Indexed: 01/06/2023] Open
Abstract
Heat shock protein 105 (HSP105) is overexpressed in many cancers, including colorectal cancer (CRC) and esophageal cancer (EC). We carried out a phase I clinical trial of HLA-A24- and HLA-A2-restricted HSP105 peptide vaccines in patients with CRC or EC. In this additional study of the trial, we examined the immunological efficacy of the novel vaccine. Thirty patients with advanced CRC or EC underwent HSP105 peptide vaccination. Immunological responses were evaluated by ex vivo and in vitro γ-interferon enzyme-linked immunospot assays and their correlation with patients' prognosis was analyzed. The HSP105 peptide vaccines induced peptide-specific CTLs in 15 of 30 patients. Among HLA-A24 patients (n = 15), 7 showed induction of CTLs only ex vivo, whereas among HLA-A2 patients (n = 15), 4 showed the induction ex vivo and 6 in vitro. Heat shock protein 105-specific CTL induction correlated with suppression of cancer progression and was revealed as a potential predictive biomarker for progression-free survival (P = .008; hazard ratio = 3.03; 95% confidence interval, 1.34-6.85) and overall survival (P = .025; hazard ratio = 2.72; 95% confidence interval, 1.13-6.52). Production of cytokines by HSP105 peptide-specific CTLs was observed at the injection sites (skin) and tumor tissues, suggesting that HSP105-specific CTLs not only accumulated at vaccination sites but also infiltrated tumors. Furthermore, we established 2 HSP105 peptide-specific CTL clones, which showed HSP105-specific cytokine secretion and cytotoxicity. Our results suggest that the HSP105 peptide vaccine could induce immunological effects in cancer patients and improve their prognosis.
Collapse
Affiliation(s)
- Yasuhiro Shimizu
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
- Department of Gastroenterological SurgeryYokohama City University Graduate School of MedicineYokohamaJapan
| | - Toshiaki Yoshikawa
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| | - Takashi Kojima
- Department of GastroenterologyNational Cancer Center Hospital EastKashiwaJapan
| | - Kayoko Shoda
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| | - Kazuto Nosaka
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| | - Shoichi Mizuno
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| | - Satoshi Wada
- Division of Cancer ImmunotherapyKanagawa Cancer Center Research InstituteYokohamaJapan
| | - Yuki Fujimoto
- Division of Cancer ImmunotherapyKanagawa Cancer Center Research InstituteYokohamaJapan
| | - Tetsuro Sasada
- Division of Cancer ImmunotherapyKanagawa Cancer Center Research InstituteYokohamaJapan
| | - Kenichi Kohashi
- Department of Anatomic PathologyPathological ScienceKyusyu University Graduate School of MedicineFukuokaJapan
| | - Hideaki Bando
- Department of GastroenterologyNational Cancer Center Hospital EastKashiwaJapan
| | - Itaru Endo
- Department of Gastroenterological SurgeryYokohama City University Graduate School of MedicineYokohamaJapan
| | - Tetsuya Nakatsura
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial CenterNational Cancer CenterKashiwaJapan
| |
Collapse
|
5
|
Battaglin F, Naseem M, Lenz HJ, Salem ME. Microsatellite instability in colorectal cancer: overview of its clinical significance and novel perspectives. CLINICAL ADVANCES IN HEMATOLOGY & ONCOLOGY : H&O 2018; 16:735-745. [PMID: 30543589 PMCID: PMC7493692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Microsatellite instability (MSI) is a key biomarker in colorectal cancer (CRC), with crucial diagnostic, prognostic, and predictive implications. Testing for mismatch repair deficiency (MMR-D)/MSI is recommended during screening for Lynch syndrome, an autosomal-dominant hereditary disease that is characterized by germline mutations in the MMR genes and associated with an increased risk for several types of cancer. Additionally, MSI-high (MSI-H) status is associated with a better prognosis in early-stage CRC and a lack of benefit from adjuvant treatment with 5-fluorouracil in stage II disease. More recently, MSI has emerged as a predictor of sensitivity to immunotherapy-based treatments. The groundbreaking success of checkpoint inhibitors in MMR-D metastatic CRC has opened a new therapeutic scenario for patients with these tumors. MSI-H CRC, in both the sporadic and hereditary settings, is characterized by distinctive molecular and clinicopathologic features and represents a unique subset of CRC that is the object of growing interest and fervent research efforts. This article, an overview of the expanding role of MSI in CRC, covers its clinical significance, the available data on molecular profiling, novel perspectives on MSI testing, biomarkers in MSI-H CRC, immunotherapy resistance, and novel immunotherapy strategies.
Collapse
Affiliation(s)
- Francesca Battaglin
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California and Veneto Oncologic Institute IOV-IRCCS, Padua, Italy
| | - Madiha Naseem
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Heinz-Josef Lenz
- USC Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Mohamed E Salem
- Levine Cancer Institute of the Carolinas HealthCare System, Charlotte, North Carolina
| |
Collapse
|
6
|
He J, Huang F, Zhang J, Chen H, Chen Q, Zhang J, Li J, Zheng Z, Chen D, Chen J. DNA prime-protein boost vaccine encoding HLA-A2, HLA-A24 and HLA-DR1 restricted epitopes of CaNA2 against visceral leishmaniasis. Immunology 2018; 156:94-108. [PMID: 30285279 DOI: 10.1111/imm.13007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 09/09/2018] [Accepted: 09/26/2018] [Indexed: 12/21/2022] Open
Abstract
Visceral leishmaniasis is a tropical and neglected disease with an estimated 200 000-400 000 cases and 60 000 deaths worldwide each year. Currently, no clinically valid vaccine is available for this disease. In this study, we formulated DNA and protein vaccines encoding HLA-A2, HLA-A24 and HLA-DR1 restricted epitopes of CaNA2 against visceral leishmaniasis. We predicted the secondary and tertiary structures, surface properties, subcellular localization, potential binding sites and HLA-A2, HLA-A24 and HLA-DR1 restricted epitopes of CaNA2. The best candidate CpG ODN (2395, M362, D-SL03 or 685) was screened out as a DNA vaccine adjuvant. We also prepared Kmp-11 and Kmp-11/CaNA2 DNA and protein vaccines, respectively, for comparison. BALB/c mice were immunized with a DNA prime-protein boost immunization strategy and challenged with a newly isolated Leishmania strain from an individual with visceral leishmaniasis. The IgG antibody titers showed that our vaccine had strong immunogenicity with a long duration, especially cellular immunity. The spleen parasite burden of each group demonstrated that the CaNA2 vaccine had a certain immune protective effect on visceral leishmaniasis in BALB/c mice, and the amastigote reduction rate reached 76%. Preliminary safety tests confirmed the safety of the vaccine. Our work demonstrates that the HLA-A2, HLA-A24 and HLA-DR1 restricted epitope CaNA2 DNA prime-protein boost vaccine may be a safe and effective epitope vaccine candidate against visceral leishmaniasis.
Collapse
Affiliation(s)
- Jinlei He
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Fan Huang
- Surgical Department, Chengdu Shuangliu Hospital of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jianhui Zhang
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Han Chen
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Qiwei Chen
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Junrong Zhang
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jiao Li
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Zhiwan Zheng
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Dali Chen
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jianping Chen
- Department of Parasitology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China.,Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Oh HJ, Kim JH, Lee TH, Park HE, Bae JM, Lee HS, Kang GH. Dominant high expression of wild-type HSP110 defines a poor prognostic subgroup of colorectal carcinomas with microsatellite instability: a whole-section immunohistochemical analysis. APMIS 2017; 125:1076-1083. [PMID: 28971530 DOI: 10.1111/apm.12770] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/10/2017] [Indexed: 01/03/2023]
Abstract
The aim of this study is to establish heat shock protein 110 (HSP110) as a prognostic biomarker of colorectal carcinomas (CRCs) with microsatellite instability-high (MSI-H) by considering the intratumoral heterogeneity of HSP110 expression. We performed whole-section immunohistochemistry (IHC) for wild-type HSP110 (HSP110wt) in 164 MSI-H CRCs. The intensity of the HSP110wt expression in tumor cells was semiquantitatively scored (0/1/2/3), and the HSP110wt expression status of each tumor was classified as low or high using the following four scoring criteria: H-score, dominant intensity score, lowest intensity score, and highest intensity score. Among the four criteria, only the dominant intensity score-based dichotomous classification of HSP110wt expression was significantly associated with a difference in disease-free survival (log-rank p = 0.035) in 164 MSI-H CRCs. The HSP110wt-low MSI-H CRCs were significantly correlated with larger deletions in the HSP110 T17 mononucleotide repeat (≥4 bp; p < 0.001). In conclusion, the dominant intensity score-based assessment of HSP110wt IHC can be a simple and useful method for the prognostic stratification of MSI-H CRCs. It is expected that HSP110wt IHC may be used to identify a subgroup of MSI-H CRCs with poor prognosis and/or candidates for further treatment, such as immunotherapy using immune checkpoint inhibitors in MSI-H CRCs.
Collapse
Affiliation(s)
- Hyeon Jeong Oh
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Ho Kim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Tae Hun Lee
- Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hye Eun Park
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Mo Bae
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Gyeong Hoon Kang
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea.,Laboratory of Epigenetics, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
8
|
Huang X, Dong H, Ting Z. Cloning, expression of a truncated HSP110 protein that augments the activities of tumor antigen-specific cytotoxic and apoptosis via tHSP110-peptide complex vaccines. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:10304-10314. [PMID: 31966365 PMCID: PMC6965763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/09/2017] [Indexed: 06/10/2023]
Abstract
The present study used a genetic engineering method to express a truncated heat shock protein 110 (tHSP110) isoform in Escherichia coli and verified its ability to bind to and present macromolecular antigens. Polymerase chain reaction (PCR) was used to obtain the truncated HSP110 gene, which was expressed in E. coli. The tHSP110 protein was non-covalently coupled to the intracellular domain (ICD) of human epidermal growth factor receptor 2 (HER2/Neu) in vitro to construct the antigen peptide complex tHSP110-ICD, which was identified by a co-immunoprecipitation assay. BALB/c mice were immunized 14-day interval for three times with the HSP110, tHSP110, HSP110-ICD, tHSP110-ICD, HSP110-P851-859 (a complex formed by full-length HSP110 with a cytotoxic T lymphocyte (CTL) epitope peptide of the Her2/neu ICD) and tHSP110-P851-859 complexes. Fourteen days after the last immunization, D2F2 cells were inoculated into BALB/c mice. The in vivo tumor volume of each group was measured every three days after cell inoculation to evaluate the immunization efficacy of the vaccine in each group. The level of the IFN-γ secreted by activated lymphocytes, the specific CTLs activity was detected. Immunohistochemical staining of bcl-2 and bax were measured on the tumor tissues of each group. Sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) showed that the expressed tHSP110 protein was 66 kDa in size. The non-covalent coupling of tHSP110 with ICD and peptide were confirmed by a co-immunoprecipitation assay. The in vivo tumor experiment results indicated no differences in the tumor volumes of the tHSP110-ICD and HSP110-ICD groups. In contrast, the tumor volume of the tHSP110-ICD group was significantly different compared with the tumor volume of the tHSP110-P851-859 group. After the mice immunized with tHSP110-ICD, tHSP110-P851-859 complexes, the complexes have potential immunogenicity, and can induce specific CTLs activity and apoptosis in BALB/c mice. As a tumor vaccine to inhibit in vivo tumor growth, the tHSP110 has the same ability to bind macromolecular antigens and activate tumor immune responses as full-length HSP110.
Collapse
Affiliation(s)
- Xu Huang
- Jiaxing University Medical CollegeJiaxing, Zhejiang, China
| | - Han Dong
- Jiaxing University Medical CollegeJiaxing, Zhejiang, China
| | - Zhang Ting
- College of Medical Technology, Zhejiang Chinese Medical UniversityHangzhou, China
| |
Collapse
|
9
|
Das K, Eisel D, Lenkl C, Goyal A, Diederichs S, Dickes E, Osen W, Eichmüller SB. Generation of murine tumor cell lines deficient in MHC molecule surface expression using the CRISPR/Cas9 system. PLoS One 2017; 12:e0174077. [PMID: 28301575 PMCID: PMC5354463 DOI: 10.1371/journal.pone.0174077] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/02/2017] [Indexed: 01/05/2023] Open
Abstract
In this study, the CRISPR/Cas9 technology was used to establish murine tumor cell lines, devoid of MHC I or MHC II surface expression, respectively. The melanoma cell line B16F10 and the murine breast cancer cell line EO-771, the latter stably expressing the tumor antigen NY-BR-1 (EO-NY), were transfected with an expression plasmid encoding a β2m-specific single guide (sg)RNA and Cas9. The resulting MHC I negative cells were sorted by flow cytometry to obtain single cell clones, and loss of susceptibility of peptide pulsed MHC I negative clones to peptide-specific CTL recognition was determined by IFNγ ELISpot assay. The β2m knockout (KO) clones did not give rise to tumors in syngeneic mice (C57BL/6N), unless NK cells were depleted, suggesting that outgrowth of the β2m KO cell lines was controlled by NK cells. Using sgRNAs targeting the β-chain encoding locus of the IAb molecule we also generated several B16F10 MHC II KO clones. Peptide loaded B16F10 MHC II KO cells were insusceptible to recognition by OT-II cells and tumor growth was unaltered compared to parental B16F10 cells. Thus, in our hands the CRISPR/Cas9 system has proven to be an efficient straight forward strategy for the generation of MHC knockout cell lines. Such cell lines could serve as parental cells for co-transfection of compatible HLA alleles together with human tumor antigens of interest, thereby facilitating the generation of HLA matched transplantable tumor models, e.g. in HLAtg mouse strains of the newer generation, lacking cell surface expression of endogenous H2 molecules. In addition, our tumor cell lines established might offer a useful tool to investigate tumor reactive T cell responses that function independently from MHC molecule surface expression by the tumor.
Collapse
Affiliation(s)
- Krishna Das
- GMP & T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Eisel
- GMP & T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Clarissa Lenkl
- GMP & T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ashish Goyal
- Division of RNA Biology and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sven Diederichs
- Division of RNA Biology and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Cancer Research, Dept. of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg & German Cancer Consortium (DKTK), Freiburg, Germany
| | - Elke Dickes
- GMP & T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wolfram Osen
- GMP & T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B. Eichmüller
- GMP & T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
- * E-mail:
| |
Collapse
|
10
|
Sun X, Suo J, Yan J. Immunotherapy in human colorectal cancer: Challenges and prospective. World J Gastroenterol 2016; 22:6362-6372. [PMID: 27605872 PMCID: PMC4968118 DOI: 10.3748/wjg.v22.i28.6362] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/02/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
Human colorectal cancer (CRC) is the third most commonly diagnosed malignancies and the prognosis for patients with recurrent or metastatic disease is extremely poor. Although new chemotherapeutic regimen improves survival rates, therapy with better efficacy and less adverse effects is drastically needed. Immunotherapy has been investigated in human CRC for decades with limited success. However, recent developments of immunotherapy, particularly immune checkpoint inhibitor therapy, have achieved promising clinical benefits in many types of cancer and revived the hope for utilizing such therapy in human CRC. In this review, we will discuss important immunological landscape within the CRC microenvironment and introduce immunoscore system to better describe immunophenotyping in CRC. We will also discuss different immunotherapeutic approaches currently utilized in different phases of clinical trials. Some of those completed or ongoing trials are summarized. Finally, we provide a brief prospective on the future human CRC immunotherapy.
Collapse
|
11
|
Fujinami N, Yoshikawa T, Sawada Y, Shimomura M, Iwama T, Sugai S, Kitano S, Uemura Y, Nakatsura T. Enhancement of antitumor effect by peptide vaccine therapy in combination with anti-CD4 antibody: Study in a murine model. Biochem Biophys Rep 2016; 5:482-491. [PMID: 28955856 PMCID: PMC5600353 DOI: 10.1016/j.bbrep.2016.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 01/30/2023] Open
Abstract
Purpose The clinical efficacy of cancer peptide vaccine therapy is insufficient. To enhance the anti-tumor effect of peptide vaccine therapy, we combined this therapy with an anti-CD4 mAb (GK1.5), which is known to deplete CD4+ cells, including regulatory T cells (Tregs). Methods To determine the treatment schedule, the number of lymphocyte subsets in the peripheral blood of mice was traced by flow cytometry after administration of anti-CD4 mAb. The ovalbumin (OVA)257–264 peptide vaccine was injected intradermally and anti-CD4 mAb was administered intraperitoneally into C57BL/6 mice at different schedules. We evaluated the enhancement of OVA peptide-specific cytotoxic T lymphocyte (CTL) induction in the combination therapy using the ELISPOT assay, CD107a assay, and cytokine assay. We then examined the in vivo metastasis inhibitory effect by OVA peptide vaccine therapy in combination with anti-CD4 mAb against OVA-expressing thymoma (EG7) in a murine liver metastatic model. Results We showed that peptide-specific CTL induction was enhanced by the peptide vaccine in combination with anti-CD4 mAb and that the optimized treatment schedule had the strongest induction effect of peptide-specific CTLs using an IFN-γ ELISPOT assay. We also confirmed that the CD107a+ cells secreted perforin and granzyme B and the amount of IL-2 and TNF produced by these CTLs increased when the peptide vaccine was combined with anti-CD4 mAb. Furthermore, metastasis was inhibited by peptide vaccines in combination with anti-CD4 mAb compared to peptide vaccine alone in a murine liver metastatic model. Conclusion The use of anti-CD4 mAb in combination with the OVA peptide vaccine therapy increased the number of peptide-specific CTLs and showed a higher therapeutic effect against OVA-expressing tumors. The combination with anti-CD4 mAb may provide a new cancer vaccine strategy. Peptide-specific CTL induction and function were enhanced by depletion of CD4+ cells. Anti-tumor effect by the peptide vaccine was enhanced by the depletion of CD4+ cells. Metastasis was inhibited by vaccine with depletion of CD4+ cells in a murine model. Combination with the depletion of CD4+ cells could be a new cancer vaccine strategy.
Collapse
Key Words
- 7-AAD, 7-amino-actinomycin D
- Anti-CD4 antibody
- CTL, cytotoxic T lymphocyte
- Cancer
- DC, dendritic cell
- ELISPOT assay, enzyme-linked immunospot assay
- FITC, fluorescein isothiocyanate
- FOXP3, forkhead box P3
- GPC3, glypican-3
- HCC, hepatocellular carcinoma
- IFN-γ, interferon-γ
- IL-2, interleukine-2
- Immunotherapy
- MHC, major histocompatibility complex
- Murine liver metastatic model
- OVA, ovalbumin
- PD-1, programmed death-1
- PE, phycoerythrin
- Peptide vaccine
- QOL, quality of life
- TGF-β, transforming growth factor-βl
- TNF, tumor necrosis factor
- Treg, regulatory T cell
- mAb, monoclonal antibody
Collapse
Affiliation(s)
- Norihiro Fujinami
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Toshiaki Yoshikawa
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Yu Sawada
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Manami Shimomura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Tatsuaki Iwama
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Shiori Sugai
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| | - Shigehisa Kitano
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Department of Experimental Therapeutics, National Cancer Center Hospital, Tsukiji, Tokyo, Japan
| | - Yasushi Uemura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research & Clinical Trial Center National Cancer Center, Kashiwa, Chiba, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Japan
| |
Collapse
|
12
|
Wurz GT, Kao CJ, DeGregorio MW. Novel cancer antigens for personalized immunotherapies: latest evidence and clinical potential. Ther Adv Med Oncol 2016; 8:4-31. [PMID: 26753003 DOI: 10.1177/1758834015615514] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The clinical success of monoclonal antibody immune checkpoint modulators such as ipilimumab, which targets cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), and the recently approved agents nivolumab and pembrolizumab, which target programmed cell death receptor 1 (PD-1), has stimulated renewed enthusiasm for anticancer immunotherapy, which was heralded by Science as 'Breakthrough of the Year' in 2013. As the potential of cancer immunotherapy has been recognized since the 1890s when William Coley showed that bacterial products could be beneficial in cancer patients, leveraging the immune system in the treatment of cancer is certainly not a new concept; however, earlier attempts to develop effective therapeutic vaccines and antibodies against solid tumors, for example, melanoma, frequently met with failure due in part to self-tolerance and the development of an immunosuppressive tumor microenvironment. Increased knowledge of the mechanisms through which cancer evades the immune system and the identification of tumor-associated antigens (TAAs) and negative immune checkpoint regulators have led to the development of vaccines and monoclonal antibodies targeting specific tumor antigens and immune checkpoints such as CTLA-4 and PD-1. This review first discusses the established targets of currently approved cancer immunotherapies and then focuses on investigational cancer antigens and their clinical potential. Because of the highly heterogeneous nature of tumors, effective anticancer immunotherapy-based treatment regimens will likely require a personalized combination of therapeutic vaccines, antibodies and chemotherapy that fit the specific biology of a patient's disease.
Collapse
Affiliation(s)
- Gregory T Wurz
- Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, Sacramento, CA, USA
| | - Chiao-Jung Kao
- Department of Obstetrics and Gynecology, University of California, Davis Sacramento, CA, USA
| | - Michael W DeGregorio
- Department of Internal Medicine, Division of Hematology and Oncology, University of California, Davis, 4501 X Street Suite 3016, Sacramento, CA 95817, USA
| |
Collapse
|
13
|
Sun W, Wei X, Niu A, Ma X, Li JJ, Gao D. Enhanced anti-colon cancer immune responses with modified eEF2-derived peptides. Cancer Lett 2015; 369:112-23. [PMID: 26304717 DOI: 10.1016/j.canlet.2015.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/22/2022]
Abstract
Eukaryotic elongation factor-2 (eEF2) is overexpressed in many human cancers and is an attractive target for cancer immunotherapy. The eEF2 derived polypeptides have been shown to be able to induce cytotoxic T lymphocytes from healthy donor. Here, we demonstrate the evidence indicating that modification of a segment of peptides from wild type eEF2-derived immunogenic peptides is able to further enhance its capacity of inducing antigen-specific cytotoxic T lymphocytes (CTLs) against colon cancer cells. Using peptide-MHC binding algorithms, potential HLA-A2.1-restricted epitopes capable of inducing specific CD8(+) CTLs were identified. By analyzing HLA-A2.1 affinity and immunogenicity, we further identified one novel immunogenic peptide, P739-747 (RLMEPIYLV), that elicited specific CTL responses in HLA-A2.1/K(b) transgenic mice and culture with peripheral blood lymphocytes from colon cancer patients. Furthermore, replacing certain amino acids (at positions 1, 3, 7) within the P739-747 sequence improved the immunogenicity against eEF2. Several analogs containing the auxiliary HLA-A*0201 anchor residues were able to stably bind to HLA-A*0201 and enhance CTL responses compared with the native sequence; two of them showed increased anti-tumor effects during the adoptive immunotherapy in vivo. Thus, these results support that modified immunogenic analogs are promising candidates for peptide-based cancer vaccination and immunotherapy.
Collapse
Affiliation(s)
- Weihong Sun
- Biotherapy Center, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China.
| | - Xiaofang Wei
- Biotherapy Center, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| | - Airong Niu
- Department of Clinical Laboratory, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| | - Xuezhen Ma
- Department of Oncology, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| | - Jian Jian Li
- Department of Radiation Oncology, NCI-designated Compressive Cancer Center, University of California Davis, Sacramento, CA 95817, USA.
| | - Daiqing Gao
- Biotherapy Center, The Second Affiliated Hospital, Qingdao University Medical College, Qingdao 266042, China
| |
Collapse
|