101
|
Okoye I, Xu L, Motamedi M, Parashar P, Walker JW, Elahi S. Galectin-9 expression defines exhausted T cells and impaired cytotoxic NK cells in patients with virus-associated solid tumors. J Immunother Cancer 2020; 8:e001849. [PMID: 33310773 PMCID: PMC7735134 DOI: 10.1136/jitc-2020-001849] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND We have previously reported that the upregulation of galectin-9 (Gal-9) on CD4+ and CD8+ T cells in HIV patients was associated with impaired T cell effector functions. Gal-9 is a ligand for T cell immunoglobulin and mucin domain-3, and its expression on T cells in cancer has not been investigated. Therefore, we aimed to investigate the expression level and effects of Gal-9 on T cell functions in patients with virus-associated solid tumors (VASTs). METHODS 40 patients with VASTs through a non-randomized and biomarker-driven phase II LATENT trial were investigated. Peripheral blood mononuclear cells and tumor biopsies were obtained and subjected to immunophenotyping. In this trial, the effects of oral valproate and avelumab (anti-PD-L1) was investigated in regards to the expression of Gal-9 on T cells. RESULTS We report the upregulation of Gal-9 expression by peripheral and tumor-infiltrating CD4+ and CD8+ T lymphocytes in patients with VASTs. Our results indicate that Gal-9 expression is associated with dysfunctional T cell effector functions in the periphery and tumor microenvironment (TME). Coexpression of Gal-9 with PD-1 or T cell immunoglobulin and ITIM domain (TIGIT) exhibited a synergistic inhibitory effect and enhanced an exhausted T cell phenotype. Besides, responding patients to treatment had lower Gal-9 mRNA expression in the TME. Translocation of Gal-9 from the cytosol to the cell membrane of T cells following stimulation suggests persistent T cell receptor (TCR) stimulation as a potential contributing factor in Gal-9 upregulation in patients with VASTs. Moreover, partial colocalization of Gal-9 with CD3 on T cells likely impacts the initiation of signal transduction via TCR as shown by the upregulation of ZAP70 in Gal-9+ T cells. Also, we found an expansion of Gal-9+ but not TIGIT+ NK cells in patients with VASTs; however, dichotomous to TIGIT+ NK cells, Gal-9+ NK cells exhibited impaired cytotoxic molecules but higher Interferon gamma (IFN-γ) expression. CONCLUSION Our data indicate that higher Gal-9-expressing CD8+ T cells were associated with poor prognosis following immunotherapy with anti-Programmed death-ligand 1 (PD-L1) (avelumab) in our patients' cohort. Therefore, for the very first time to our knowledge, we report Gal-9 as a novel marker of T cell exhaustion and the potential target of immunotherapy in patients with VASTs.
Collapse
Affiliation(s)
- Isobel Okoye
- School of Dentistry, Faculty of Medicine and Dentistrty, University of Alberta, Edmonton, AB, Canada
| | - Lai Xu
- School of Dentistry, Faculty of Medicine and Dentistrty, University of Alberta, Edmonton, AB, Canada
| | - Melika Motamedi
- Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Pallavi Parashar
- School of Dentistry, Faculty of Medicine and Dentistrty, University of Alberta, Edmonton, AB, Canada
| | - John W Walker
- Medical Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- School of Dentistry, Faculty of Medicine and Dentistrty, University of Alberta, Edmonton, AB, Canada
- Medical Microbiology and Immunology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Medical Oncology, Cross Cancer Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
102
|
Efficacy of Acupuncture and Moxibustion as a Subsequent Treatment after Second-Line Chemotherapy in Advanced Gastric Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:8274021. [PMID: 33144870 PMCID: PMC7599393 DOI: 10.1155/2020/8274021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/18/2020] [Indexed: 02/06/2023]
Abstract
Objective To explore whether acupuncture and moxibustion can prevent disease progression of advanced gastric cancer patients completing second-line chemotherapy and, if so, the related mechanism. Method Progression-free survival (PFS) and overall survival (OS) were main outcome measures. The real-time quantitative PCR was used to detect the expression of genes including T-bet, IFN-γ, GATA3, and IL-4 in peripheral blood mononuclear cells (PBMCs). IL-4, IL-6, Ca199, CRP, and IFN-γ in plasma levels were checked. Results 170 patients were randomly assigned in a 3 : 2 ratio to receive either acupuncture and moxibustion or sham acupuncture until progression. 135 patients were included in the primary analysis. Both PFS and OS in treatment group were proven to be better than control group. Acupuncture and moxibustion promoted typical Th1 cells drifting, as confirmed by increased T-bet/IFN-γ and decreased GATA3/IL-4 in mRNA levels from PBMCs, as well as upregulating IFN-γ and downregulating IL-4 in plasma levels. IL-6, Ca199, and CRP in plasma levels were also reduced by acupuncture and moxibustion. Conclusions Acupuncture and moxibustion can prolong PFS and OS of advanced gastric cancer patients completing second-line chemotherapy by reversing Th1/Th2 shift and attenuating inflammatory responses.
Collapse
|
103
|
Xu Y, Ma S, Si X, Zhao J, Yu H, Ma L, Song W, Tang Z. Polyethyleneimine-CpG Nanocomplex as an In Situ Vaccine for Boosting Anticancer Immunity in Melanoma. Macromol Biosci 2020; 21:e2000207. [PMID: 33107202 DOI: 10.1002/mabi.202000207] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/06/2020] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy is redefining the field of cancer therapy. However, current cancer immunotherapies are limited by insufficient immune activation, which results in low response rate. Herein, polyethyleneimine-CpG nanocomplex (CpG@PEI) is reported as an in situ vaccine for boosting anticancer immunity in melanoma. CpG, a Toll-like receptor (TLR) 9 agonist, can activate antigen-presenting cells and increase the expression of costimulatory molecules, while PEI can help to enhance the stability and cellular internalization of CpG. It is proved that PEI loading can significantly enhance the cellular internalization and immune stimulation ability of CpG, and the CpG@PEI nanocomplex can effectively inhibit murine B16F10 melanoma growth after intratumoral injection. Further analysis reveals that this CpG@PEI nanocomplex therapy elicits both innate and adaptive immunity, with much increased natural killer (NK) cells and T cells infiltration in the tumor, as well as CD80 expression on the dendritic cells (DCs). This study will inspire more attempts in directly using single nanoparticle-loaded pattern recognition receptor (PRR) agonists for cancer immunotherapy.
Collapse
Affiliation(s)
- Yudi Xu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,University of Chinese Academy of Sciences, Beijing, 100039, China
| | - Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Jiayu Zhao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| | - Haiyang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Lili Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China.,School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
104
|
Th17-inducing autologous dendritic cell vaccination promotes antigen-specific cellular and humoral immunity in ovarian cancer patients. Nat Commun 2020; 11:5173. [PMID: 33057068 PMCID: PMC7560895 DOI: 10.1038/s41467-020-18962-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 09/21/2020] [Indexed: 01/13/2023] Open
Abstract
In ovarian cancer (OC), IL-17-producing T cells (Th17s) predict improved survival, whereas regulatory T cells predict poorer survival. We previously developed a vaccine whereby patient-derived dendritic cells (DCs) are programmed to induce Th17 responses to the OC antigen folate receptor alpha (FRα). Here we report the results of a single-arm open-label phase I clinical trial designed to determine vaccine safety and tolerability (primary outcomes) and recurrence-free survival (secondary outcome). Immunogenicity is also evaluated. Recruitment is complete with a total of 19 Stage IIIC-IV OC patients in first remission after conventional therapy. DCs are generated using our Th17-inducing protocol and are pulsed with HLA class II epitopes from FRα. Mature antigen-loaded DCs are injected intradermally. All patients have completed study-related interventions. No grade 3 or higher adverse events are seen. Vaccination results in the development of Th1, Th17, and antibody responses to FRα in the majority of patients. Th1 and antibody responses are associated with prolonged recurrence-free survival. Antibody-dependent cell-mediated cytotoxic activity against FRα is also associated with prolonged RFS. Of 18 patients evaluable for efficacy, 39% (7/18) remain recurrence-free at the time of data censoring, with a median follow-up of 49.2 months. Thus, vaccination with Th17-inducing FRα-loaded DCs is safe, induces antigen-specific immunity, and is associated with prolonged remission.
Collapse
|
105
|
Li T, Wu B, Yang T, Zhang L, Jin K. The outstanding antitumor capacity of CD4 + T helper lymphocytes. Biochim Biophys Acta Rev Cancer 2020; 1874:188439. [PMID: 32980465 DOI: 10.1016/j.bbcan.2020.188439] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/10/2020] [Accepted: 09/21/2020] [Indexed: 02/05/2023]
Abstract
Over the past decades, tumor-resident immune cells have been extensively studied to dissect their biological functions and clinical roles. Tumor-infiltrating CD8+ T cells, because of their cytotoxic and killing ability, have been under the spotlight for a long time, whereas CD4+ T cells are considered just a supporting actor in the field of cancer immunotherapy. Until recently, accumulating evidence has demonstrated the ability of CD4+ T cells in eradicating solid tumors, and their functions in mediating antitumor immunity have been investigated in various orientations. In this review, we highlight the pivotal role of CD4+ T cells in eliciting vigorous antitumor immune responses, summarize key signaling axes and molecular networks behind these antitumor functions, and also propose possible targets and promising strategies which might translate into more efficient immunotherapies against human cancers.
Collapse
Affiliation(s)
- Tong Li
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China; State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bowen Wu
- School of Medicine, Stanford University, Stanford, CA 94304, USA
| | - Tao Yang
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Ke Jin
- Laboratory of Human Diseases and Immunotherapies, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
106
|
Kan S, Bito T, Shimabuku M, Taguchi J, Ohkusa T, Shimodaira S, Sugiyama H, Koido S. Impact of mature dendritic cells pulsed with a novel WT1 helper peptide on the induction of HLA‑A2‑restricted WT1‑reactive CD8+ T cells. Int J Oncol 2020; 57:1047-1056. [PMID: 32945369 DOI: 10.3892/ijo.2020.5110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 07/31/2020] [Indexed: 11/06/2022] Open
Abstract
The proliferation and activation of CD4+ T helper 1 (Th1) cells and CD8+ cytotoxic T lymphocytes (CTLs) that produce interferon‑γ (IFN‑γ) is an essential action of effective cancer vaccines. Recently, a novel Wilms' tumor 1 (WT1) helper peptide (WT1 HP34‑51; amino acid sequence, WAPVLDFAPPGASAYGSL) applicable for various human leukocyte antigen (HLA) subtypes (HLA‑DR, HLA‑DP and HLA‑DQ) was reported to increase peptide immunogenicity; however, the function of WT1 HP34‑51 remains unclear. In the present study, mature dendritic cells (mDCs) pulsed with WT1 HP34‑51 (mDC/WT1 HP34‑51) activated not only WT1‑specific CD4+ T cells but also CD8+ T cells that produced IFN‑γ following stimulation with immature dendritic cells (imDCs) pulsed with WT1 killer peptide (imDC/WT1 KP37‑45) in an HLA‑A*02:01‑ or HLA‑A*02:06‑restricted manner. Furthermore, the activated WT1‑reactive CD4+ Th1 cells were predominantly effector memory (EM) T cells. In 5 of 12 (41.7%) patients with cancer carrying the HLA‑A*02:01 or HLA‑A*02:06 allele, WT1‑reactive CD8+ T cells stimulated with mDC/WT1 HP34‑51 enhanced their levels of WT1 KP37‑45‑specific IFN‑γ production, with an increase >10%. Simultaneous activation of CD4+ and CD8+ T cells occurred more often when stimulation with mDC/WT1 HP34‑51 was combined with imDC/WT1 KP37‑45 restimulation. These results indicated that the novel mDC/WT1 HP34‑51 combination induced responses by WT1‑specific EM CD4+ Th1 cells and HLA‑A*02:01‑ or HLA‑A*02:06‑restricted CD8+ CTLs, suggesting its potential as a WT1‑targeting cancer vaccine.
Collapse
Affiliation(s)
- Shin Kan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277‑8567, Japan
| | - Tsuuse Bito
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277‑8567, Japan
| | - Masamori Shimabuku
- Tokyo Midtown Center for Advanced Medical Science and Technology, Tokyo 107‑6206, Japan
| | - Junichi Taguchi
- Tokyo Midtown Center for Advanced Medical Science and Technology, Tokyo 107‑6206, Japan
| | - Toshifumi Ohkusa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277‑8567, Japan
| | - Shigetaka Shimodaira
- Department of Regenerative Medicine, Kanazawa Medical University, Ishikawa 920‑0293, Japan
| | - Haruo Sugiyama
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Osaka 565‑0871, Japan
| | - Shigeo Koido
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, The Jikei University School of Medicine, Kashiwa Hospital, Kashiwa, Chiba 277‑8567, Japan
| |
Collapse
|
107
|
Xia Y, Li W, Li Y, Liu Y, Ye S, Liu A, Yu J, Jia Y, Liu X, Chen H, Guo Y. The clinical value of the changes of peripheral lymphocyte subsets absolute counts in patients with non-small cell lung cancer. Transl Oncol 2020; 13:100849. [PMID: 32866935 PMCID: PMC7475266 DOI: 10.1016/j.tranon.2020.100849] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/13/2020] [Accepted: 08/06/2020] [Indexed: 12/24/2022] Open
Abstract
Introduction Immune function strongly influences the outcome of patients with non-small cell lung cancer (NSCLC). It's vital to understand the immune state of patients through detecting the percentage and number of lymphocyte subsets accurately, and helpful to evaluate conditions of prognosis and adjust treatment for patients. Methods We conducted a retrospective cohort study in First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China. The absolute counts and percentages of CD3+, CD3 + CD4+, CD3 + CD8+, B and NK cells were determined by single platform technologies. 172 patients received treatment including surgery or chemotherapy after surgery. The factors affecting disease progression were analyzed by Binary Logistic regression. Progression free survival (PFS) calculating survivals were with the method of Kaplan-Meier. The log-rank test and cox's proportional hazard regression (enter method) were used for univariable and multivariable analyses respectively. Results Relative to normal controls, patients with NSCLC at different stages showed decreased absolute lymphocyte count obviously, rather than lymphocyte percentages. Different treatments had unlike influence on the homeostasis of lymphocytes and the effects last for a long time. Logistic regression showed CD3 + CD4+ and CD3 + CD8+ could contribute to favorable prognosis. Multivariate analysis of prognostic factors of PFS showed CD3 + CD4+ cell was independent factor for predicting PFS. Conclusions The absolute count of CD3+, CD3 + CD4+, CD3 + CD8+, B and NK cells were better indication of the patient's immune state than percentages of each lymphocyte subsets. Immune function was impaired in patients with non-small cell lung. The high level of baseline absolute CD3 + CD4+ cells count contributed to longer progression free survival. Chinese Clinic Trial Registry number: ChiCTR-IOR-17014139; Registry date: 2017/12/25.
Collapse
Affiliation(s)
- Ying Xia
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Oncology Department, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Graduate School, Tianjin, China
| | - Wentao Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Oncology Department, Tianjin, China
| | - Yongmin Li
- Hebei University of Chinese Medicine, Hebei, China
| | - Yunhe Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Oncology Department, Tianjin, China
| | - Songshan Ye
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Oncology Department, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Graduate School, Tianjin, China
| | - Aqing Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Oncology Department, Tianjin, China; Tianjin University of Traditional Chinese Medicine, Graduate School, Tianjin, China
| | - Jianchun Yu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Oncology Department, Tianjin, China.
| | - Yingjie Jia
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Oncology Department, Tianjin, China
| | - Xu Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Clinic Laboratory, Tianjin, China
| | - Huayu Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Clinic Laboratory, Tianjin, China
| | - Yongtie Guo
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Clinic Laboratory, Tianjin, China
| |
Collapse
|
108
|
Bi F, Chen Y, Yang Q. Significance of tumor mutation burden combined with immune infiltrates in the progression and prognosis of ovarian cancer. Cancer Cell Int 2020; 20:373. [PMID: 32774167 PMCID: PMC7405355 DOI: 10.1186/s12935-020-01472-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
Background Ovarian cancer (OC) is the most malignant tumor in the female reproductive system. About 75% of OC in complete remission of clinical symptoms still develop a recurrence. Therefore, searching for new treatment methods plays an important role in improving the prognosis of OC. Methods We downloaded the MAF files, RNA-seq data and clinical information from the TCGA database. The “maftools” package in R software was used to visualize the OC mutation data. We calculated the tumor mutation burden (TMB) of OC and analyzed its correlation with clinicopathological parameters and prognostic value. Tumor mutation burden related signature model was constructed to predict the overall survival (OS) of OC. Results The results revealed that there was a statistical correlation between TMB and FIGO stage, grade and tumor residual size of ovarian cancer patients. The Kaplan–Meier curve indicated that a high TMB is associated with better clinical outcomes of OC. The difference analysis indicated 24 upregulated genes and 619 downregulated genes in the high-TMB group compared with the low-TMB group. Besides, the TMBRS model based on five hub genes (RBMS3, PLA2G5, CDH2, AMHR2 and ADAMTS8) was constructed to predict the OS of OC. The ROC curve and validation data sets all revealed that the TMBRS model was reliable in predicting recurrence risk. Immune microenvironment analysis indicated the correlations between TMB and infiltrating immune cells. Conclusions Our results suggest that TMB plays an important role in the prognosis and guiding immunotherapy of OC. By detecting the TMB of OC, clinicians can more accurately treat patients with immunotherapy, thereby improving their survival rate.
Collapse
Affiliation(s)
- Fangfang Bi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, NO. 36 Sanhao Road, Shenyang, 110000 China
| | - Ying Chen
- Department of Ultrasound, Jiangnan Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, NO. 36 Sanhao Road, Shenyang, 110000 China
| |
Collapse
|
109
|
Kang N, Eccleston M, Clermont PL, Latarani M, Male DK, Wang Y, Crea F. EZH2 inhibition: a promising strategy to prevent cancer immune editing. Epigenomics 2020; 12:1457-1476. [PMID: 32938196 PMCID: PMC7607396 DOI: 10.2217/epi-2020-0186] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapies are revolutionizing the clinical management of a wide range of cancers. However, intrinsic or acquired unresponsiveness to immunotherapies does occur due to the dynamic cancer immunoediting which ultimately leads to immune escape. The evolutionarily conserved histone modifier enhancer of zeste 2 (EZH2) is aberrantly overexpressed in a number of human cancers. Accumulating studies indicate that EZH2 is a main driver of cancer cells' immunoediting and mediate immune escape through downregulating immune recognition and activation, upregulating immune checkpoints and creating an immunosuppressive tumor microenvironment. In this review, we overviewed the roles of EZH2 in cancer immunoediting, the preclinical and clinical studies of current pharmacologic EZH2 inhibitors and the prospects for EZH2 inhibitor and immunotherapy combination for cancer treatment.
Collapse
Affiliation(s)
- Ning Kang
- Department of Experimental Therapeutics, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
| | - Mark Eccleston
- Belgian Volition SPRL, Parc Scientifique Créalys, Rue Phocas Lejeune 22, BE-5032 Isnes, Belgium
| | - Pier-Luc Clermont
- Faculty of Medicine, Université Laval, 1050, avenue de la Médecine, Québec, QC, G1V 0A6, Canada
| | - Maryam Latarani
- Cancer Research Group, School of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - David Kingsley Male
- Cancer Research Group, School of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| | - Yuzhuo Wang
- Department of Experimental Therapeutics, BC Cancer Research Centre, 675 West 10th Avenue, Vancouver, BC, V5Z 1L3, Canada
- Department of Urologic Sciences, The Vancouver Prostate Centre, The University of British Columbia, 2660 Oak St, Vancouver, BC, V6H 3Z6, Canada
| | - Francesco Crea
- Cancer Research Group, School of Life Health & Chemical Sciences, The Open University, Walton Hall, Milton Keynes, MK7 6AA, UK
| |
Collapse
|
110
|
Jacoberger-Foissac C, Saliba H, Wantz M, Seguin C, Flacher V, Frisch B, Heurtault B, Fournel S. Liposomes as tunable platform to decipher the antitumor immune response triggered by TLR and NLR agonists. Eur J Pharm Biopharm 2020; 152:348-357. [PMID: 32479782 DOI: 10.1016/j.ejpb.2020.05.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 05/04/2020] [Accepted: 05/25/2020] [Indexed: 01/15/2023]
Abstract
Liposomes are powerful tools for the optimization of peptides and adjuvant composition in cancer vaccines. Here, we take advantage of a liposomal platform versatility to develop three vaccine candidates associating a peptide from HA influenza virus protein as CD4 epitope, a peptide from HPV16 E7 oncoprotein as CD8 epitope and TLR4, TLR2/6 or NOD1 agonists as adjuvant. Liposomal vaccine containing MPLA (TLR4 liposomes), are the most effective treatment against the HPV-transformed orthotopic lung tumor mouse model, TC-1. This vaccine induces a potent Th1-oriented antitumor immunity, which leads to a significant reduction in tumor growth and a prolonged survival of mice, even when injected after tumor appearance. This efficacy is dependent on CD8+ T cells. Subcutaneous injection of this treatment induces the migration of skin DCs to draining lymph nodes. Interestingly, TLR2/6 liposomes trigger a weaker Th1-immune response which is not sufficient for the induction of a prolonged antitumor activity. Although NOD1 liposome treatment results in the control of early tumor growth, it does not extend mice survival. Surprisingly, the antitumor activity of NOD1 vaccine is not associated with a specific adaptive immune response. This study shows that our modulable platform can be used for the strategical development of vaccines.
Collapse
Affiliation(s)
- Célia Jacoberger-Foissac
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Hanadi Saliba
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - May Wantz
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Cendrine Seguin
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Vincent Flacher
- Laboratory I(2)CT - Immunology, Immunopathology and Therapeutic Chemistry, CNRS UPR 3572, Institut de Biologie Moléculaire et Cellulaire, 15 Rue René Descartes, 67084 Strasbourg Cedex, France
| | - Benoît Frisch
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France
| | - Béatrice Heurtault
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France.
| | - Sylvie Fournel
- Université de Strasbourg, CNRS, 3Bio team, Laboratoire de Conception et Application de Molécules Bioactives, UMR 7199, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch Cedex, France.
| |
Collapse
|
111
|
Wang X, Gao Y, Zhang X, Wang X, Wang B, Meng X, Yoshikai Y, Wang Y, Sun X. CD30L/CD30 signaling regulates the formation of the tumor immune microenvironment and inhibits intestinal tumor development of colitis-associated colon cancer in mice. Int Immunopharmacol 2020; 84:106531. [PMID: 32353687 DOI: 10.1016/j.intimp.2020.106531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 04/19/2020] [Accepted: 04/19/2020] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease is one of the major causes of colitis-associated colon cancer (CAC). Therefore, it is necessary to explore new therapies to prevent colon cancer (CRC) in view of the relationship between chronic inflammation and tumor development. Previous studies on the correlation between CD30L/CD30 and cancer were mostly limited to lymphoid or homogenous tumors, while there have been only a few reports on the role of CD30L/CD30 signal transduction in the pathogenesis of CAC. In this study, we established an AOM/DSS-induced CAC model with CD30LKO mice to explore the effect of CD30L/CD30 signal transduction on the formation of the intestinal tumor immune microenvironment (TIME) during the development of intestinal tumors. Our results revealed that CD30L deficiency promoted the accumulation of myeloid derived suppressor cells (MDSCs), increased the expression of PD-L1 on MDSCs and tumor associated macrophages (TAMs), and enhanced the secretion of various inflammatory and immunosuppressive factors in the intestinal mucosa of CAC mice. Furthermore, CD30L gene deletion could selectively promote the upregulation of PD-1 expression on CD4+ and CD8+ T cells and inhibit their activation, differentiation and secretion of effector cytokines, which led to an attenuation of antitumor immune responses mediated by TEM (CD44+CD62L-) cells. Thus, our data suggest that CD30L/CD30 signaling might be a potential candidate target for immunological therapy in CAC.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Immunology, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Yaxian Gao
- Department of Immunology, Chengde Medical College, Chengde, Hebei 067000, PR China; Department of Immunology, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Xiaoqing Zhang
- Department of Immunology, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Xiaonan Wang
- Department of Immunology, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China
| | - Biao Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning Province, PR China
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning Province, PR China
| | - Yasunobu Yoshikai
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuanyuan Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning Province, PR China.
| | - Xun Sun
- Department of Immunology, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, PR China.
| |
Collapse
|
112
|
Zhang Y, Ma JA, Zhang HX, Jiang YN, Luo WH. Cancer vaccines: Targeting KRAS-driven cancers. Expert Rev Vaccines 2020; 19:163-173. [PMID: 32174221 DOI: 10.1080/14760584.2020.1733420] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Mutant KRAS is a genetic driver of multiple cancers that has challenged clinical anti-cancer therapeutics in the last 3 decades. Neo-antigens encoded by KRAS mutations have been identified as tumor-specific with high immunogenicity and can be used to deliver precision cancer vaccines to promote anti-tumor immune responses. KRAS mutation-based cancer vaccines have produced encouraging preclinical and clinical results. Cancer vaccines represent a promising approach to treat KRAS-driven cancers.Areas covered: In this review, we summarize the development and progress of vaccines targeting KRAS and evaluate their potential benefits and obstacles in the current landscape of therapy for KRAS-driven cancers.Expert opinion: KRAS mutation-based cancer vaccines can induce immunogenicity in patients with KRAS-driven cancers. However, the mechanisms of tumor suppression including cellular and molecular factors within the tumor microenvironment may limit vaccine efficacy. Combining KRAS-driven therapeutic cancer vaccines with other methods and adjuvants can circumvent immunosuppression and promote therapeutic successes.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin-An Ma
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hai-Xia Zhang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Na Jiang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Hao Luo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
113
|
Wang M, Li Z, Peng Y, Fang J, Fang T, Wu J, Zhou J. Identification of immune cells and mRNA associated with prognosis of gastric cancer. BMC Cancer 2020; 20:206. [PMID: 32164594 PMCID: PMC7068972 DOI: 10.1186/s12885-020-6702-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background The clinical success demonstrates the enormous potential of immunotherapy in cancer treatment. Methods This article presented research linking gastric cancer to immune cells, based on RNA-seq data of Stomach adenocarcinoma (STAD) and gene expression profile of GSE84437, 24 kinds of tumor-infiltrating immune cells were quantified by single-sample gene set enrichment analysis. Results Th2 cells, T helper cells, and Mast cells were identified as prognostic immune cells in both TCGA and GEO groups. Then SUPV3L1 and SLC22A17 were identified as hub genes which may affect immune cell infiltration by correlation analysis. Survival analysis further proved that hub genes and prognostic immune cells are associated with the prognosis of gastric cancer. In gastrointestinal tumors, hub genes and prognostic immune cells also found differences in non-tumor and tumor tissues. Conclusions We found that three immune cells infiltration are associated with the prognosis of gastric cancer and further identify two hub genes. These two key genes may affect immune cell infiltration, result in the different prognosis of patients.
Collapse
Affiliation(s)
- Mingming Wang
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Zedong Li
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yu Peng
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jianyu Fang
- Department of Nursing, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tao Fang
- Department of General Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiajia Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jun Zhou
- Department of Minimally Invasive Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
114
|
Abstract
Peptides, as a large group of molecules, are composed of amino acid residues and can be divided into linear or cyclic peptides according to the structure. Over 13,000 molecules of natural peptides have been found and many of them have been well studied. In artificial peptide libraries, the number of peptide diversity could be up to 1 × 1013. Peptides have more complex structures and higher affinity to target proteins comparing with small molecular compounds. Recently, the development of targeting cancer immune checkpoint (CIP) inhibitors is having a very important role in tumor therapy. Peptides targeting ligands or receptors in CIP have been designed based on three-dimensional structures of target proteins or directly selected by random peptide libraries in biological display systems. Most of these targeting peptides work as inhibitors of protein-protein interaction and improve CD8+ cytotoxic T-lymphocyte (CTL) activation in the tumor microenvironment, for example, PKHB1, Ar5Y4 and TPP1. Peptides could be designed to regulate CIP protein degradation in vivo, such as PD-LYSO and PD-PALM. Besides its use in developing therapeutic drugs for targeting CIP, targeting peptides could be used in drug's targeted delivery and diagnosis in tumor immune therapy.
Collapse
|
115
|
Knutson KL, Block MS, Norton N, Erskine CL, Hobday TJ, Dietz AB, Padley D, Gustafson MP, Puglisi-Knutson D, Mangskau TK, Chumsri S, Dueck AC, Karyampudi L, Wilson G, Degnim AC. Rapid Generation of Sustainable HER2-specific T-cell Immunity in Patients with HER2 Breast Cancer using a Degenerate HLA Class II Epitope Vaccine. Clin Cancer Res 2019; 26:1045-1053. [PMID: 31757875 DOI: 10.1158/1078-0432.ccr-19-2123] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 10/14/2019] [Accepted: 11/18/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE Patients with HER2+ breast cancer benefit from trastuzumab-containing regimens with improved survival. Adaptive immunity, including cytotoxic T-cell and antibody immunity, is critical to clinical efficacy of trastuzumab. Because Th cells are central to the activation of these antitumor effectors, we reason that HER2 patients treated with trastuzumab may benefit by administering vaccines that are designed to stimulate Th-cell immunity. PATIENTS AND METHODS We developed a degenerate HER2 epitope-based vaccine consisting of four HLA class II-restricted epitopes mixed with GM-CSF that should immunize most (≥84%) patients. The vaccine was tested in a phase I trial. Eligible women had resectable HER2+ breast cancer and had completed standard treatment prior to enrollment and were disease free. Patients were vaccinated monthly for six doses and monitored for safety and immunogenicity. RESULTS Twenty-two subjects were enrolled and 20 completed all six vaccines. The vaccine was well tolerated. All patients were alive at analysis with a median follow-up of 2.3 years and only two experienced disease recurrence. The percent of patients that responded with augmented T-cell immunity was high for each peptide ranging from 68% to 88%, which led to 90% of the patients generating T cells that recognized naturally processed HER2 antigen. The vaccine also augmented HER2-specific antibody. Immunity was sustained in patients with little sign of diminishing at 2 years following the vaccination. CONCLUSIONS Degenerate HLA-DR-based HER2 vaccines induce sustainable HER2-specific T cells and antibodies. Future studies, could evaluate whether vaccination during adjuvant treatment with trastuzumab-containing regimens improves patient outcomes.
Collapse
Affiliation(s)
- Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, Florida.
| | | | - Nadine Norton
- Department of Cancer Biology, Mayo Clinic, Jacksonville, Florida
| | | | | | - Allan B Dietz
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Douglas Padley
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Michael P Gustafson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | | | - Toni Kay Mangskau
- Mayo Clinic Cancer Education Program, Mayo Clinic, Rochester, Minnesota
| | | | - Amylou C Dueck
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, Arizona
| | | | | | - Amy C Degnim
- Department of Surgery, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
116
|
Zhang Y, Lin Z, Wan Y, Cai H, Deng L, Li R. The Immunogenicity and Anti-tumor Efficacy of a Rationally Designed Neoantigen Vaccine for B16F10 Mouse Melanoma. Front Immunol 2019; 10:2472. [PMID: 31749795 PMCID: PMC6848027 DOI: 10.3389/fimmu.2019.02472] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/03/2019] [Indexed: 12/30/2022] Open
Abstract
Tumor neoantigens are ideal targets for cancer immunotherapy as they are recognized by host immune system as foreigners and can elicit tumor-specific immune responses. However, existing strategies utilizing RNA or long peptides for the neoantigen vaccines render limited immune responses since only 20–30% of neoantigens predicted in silico to bind MHC I molecules are capable of eliciting immune responses with the majority of responding T cells are CD4+. Therefore, it warrants further exploration to enhance neoantigen-specific CD8+ T cells responses. Since neoantigens are naturally weak antigens, we asked whether foreign T help epitopes could enhance their immunogenicity. In present study, we chose 4 weak B16F10 neoantigens as vaccine targets, and fused them to the transmembrane domain of diphtheria toxin, namely DTT-neoAg. Strikingly, the vaccine elicited anti-tumor CD8+ T cells responses and enhanced tumor infiltration of both T cells and NK cells. Impressively, DTT-neoAg vaccine significantly deterred tumor growth with the inhibition rate reached 88% in the preventive model and 100% in the therapeutic model at low dose of tumor challenge. Furthermore, after second challenge with higher dose of tumor cells, 33.3% of the immunized mice remained tumor-free for 6 months in the therapeutic model. Because DTT is a non-toxic domain of diphtheria toxin, it may be not of great concern in terms of safety as a Th epitope provider. Thus, the fusion strategy employed by this study may become a feasible and powerful approach for development of personalized cancer vaccines.
Collapse
Affiliation(s)
- Yan Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhibing Lin
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yuhua Wan
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Huaman Cai
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Li Deng
- Shanghai HyCharm Inc., Shanghai, China
| | - Rongxiu Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.,Shanghai HyCharm Inc., Shanghai, China.,Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, Shanghai, China
| |
Collapse
|
117
|
Comoli P, Chabannon C, Koehl U, Lanza F, Urbano-Ispizua A, Hudecek M, Ruggeri A, Secondino S, Bonini C, Pedrazzoli P. Development of adaptive immune effector therapies in solid tumors. Ann Oncol 2019; 30:1740-1750. [PMID: 31435646 DOI: 10.1093/annonc/mdz285] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
State-of-the-art treatment strategies have drastically ameliorated the outcome of patients affected by cancer. However, resistant and recurrent solid tumors are generally nonresponsive to conventional therapies. A central factor in the sequence of events that lead to cancer is an alteration in antitumor immune surveillance, which results in failure to recognize and eliminate the transformed tumor cell. A greater understanding of the dysregulation and evasion of the immune system in the evolution and progression of cancer provides the basis for improved therapies. Targeted strategies, such as T-cell therapy, not only generally spare normal tissues, but also use alternative antineoplastic mechanisms that synergize with other therapeutics. Despite encouraging success in hematologic malignancies, adaptive cellular therapies for solid tumors face unique challenges because of the immunosuppressive tumor microenvironment, and the hurdle of T-cell trafficking within scarcely accessible tumor sites. This review provides a brief overview of current cellular therapeutic strategies for solid tumors, research carried out to increase efficacy and safety, and results from ongoing clinical trials.
Collapse
Affiliation(s)
- P Comoli
- Cell Factory and Pediatric Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - C Chabannon
- Institut Paoli-Calmettes, Aix-Marseille University, INSERM CBT 1409, Centre for Clinical Investigation in Biotherapy, Marseille, France
| | - U Koehl
- Institute of Clinical Immunology, University of Leipzig and Fraunhofer Institute for Cell Therapy and Immunology, Leipzig; Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - F Lanza
- Hematology and Stem Cell Transplant, Romagna Transplant Network, Ravenna, Italy
| | - A Urbano-Ispizua
- Department of Hematology, IDIBAPS, Institute of Research Josep Carreras, Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - M Hudecek
- Department of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - A Ruggeri
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Rome
| | - S Secondino
- Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia
| | - C Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, University Vita-Salute San Raffaele and Ospedale San Raffaele Scientific Institute, Milan, Italy
| | - P Pedrazzoli
- Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia.
| |
Collapse
|
118
|
Kristensen LK, Fröhlich C, Christensen C, Melander MC, Poulsen TT, Galler GR, Lantto J, Horak ID, Kragh M, Nielsen CH, Kjaer A. CD4 + and CD8a + PET imaging predicts response to novel PD-1 checkpoint inhibitor: studies of Sym021 in syngeneic mouse cancer models. Theranostics 2019; 9:8221-8238. [PMID: 31754392 PMCID: PMC6857046 DOI: 10.7150/thno.37513] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022] Open
Abstract
Predicting the outcome of immunotherapy is essential for efficient treatment. The recent clinical success of immunotherapy is increasingly changing the paradigm of cancer treatment. Accordingly, the development of immune-based agents is accelerating and the number of agents in the global immuno-oncology pipeline has grown 60-70% over the past year. However, despite remarkable clinical efficacy in some patients, only few achieve a lasting clinical response. Treatment failure can be attributed to poorly immunogenic tumors that do not attract tumor infiltrating lymphocytes (TILs). Therefore, we developed positron emission tomography (PET) radiotracers for non-invasive detection of CD4+ and CD8a+ TILs in syngeneic mouse tumor models for preclinical studies. Methods: Seven syngeneic mouse tumor models (B16F10, P815, CT26, MC38, Renca, 4T1, Sa1N) were quantified for CD4+ and CD8a+ TILs using flow cytometry and immunohistochemistry (IHC), as well as for tumor growth response to Sym021, a humanized PD-1 antibody cross-reactive with mouse PD-1. Radiotracers were generated from F(ab)'2 fragments of rat-anti-mouse CD4 and CD8a antibodies conjugated to the p-SCN-Bn-Desferrioxamine (SCN-Bn-DFO) chelator and radiolabeled with Zirconium-89 (89Zr-DFO-CD4/89Zr-DFO-CD8a). Tracers were optimized for in vivo PET/CT imaging in CT26 tumor-bearing mice and specificity was evaluated by depletion studies and isotype control imaging. 89Zr-DFO-CD4 and 89Zr-DFO-CD8a PET/CT imaging was conducted in the panel of syngeneic mouse models prior to immunotherapy with Sym021. Results: Syngeneic tumor models were characterized as "hot" or "cold" according to number of TILs determined by flow cytometry and IHC. 89Zr-DFO-CD4 and 89Zr-DFO-CD8a were successfully generated with a radiochemical purity >99% and immunoreactivity >85%. The optimal imaging time-point was 24 hours post-injection of ~1 MBq tracer with 30 µg non-labeled co-dose. Reduced tumor and spleen uptake of 89Zr-DFO-CD8a was observed in CD8a+ depleted mice and the uptake was comparable with that of isotype control (89Zr-DFO-IgG2b) confirming specificity. PET imaging in syngeneic tumor models revealed a varying maximum tumor-to-heart ratio of 89Zr-DFO-CD4 and 89Zr-DFO-CD8a across tumor types and in-between subjects that correlated with individual response to Sym021 at day 10 relative to start of therapy (p=0.0002 and p=0.0354, respectively). The maximum 89Zr-DFO-CD4 tumor-to-heart ratio could be used to stratify mice according to Sym021 therapy response and overall survival was improved in mice with a 89Zr-DFO-CD4 ratio >9 (p=0.0018). Conclusion: We developed 89Zr-DFO-CD4 and 89Zr-DFO-CD8a PET radiotracers for specific detection and whole-body assessment of CD4+ and CD8a+ status. These radiotracers can be used to phenotype preclinical syngeneic mouse tumor models and to predict response to an immune checkpoint inhibitor. We foresee development of such non-invasive in vivo biomarkers for prediction and evaluation of clinical efficacy of immunotherapeutic agents, such as Sym021.
Collapse
Affiliation(s)
- Lotte K. Kristensen
- Minerva Imaging, Copenhagen, Denmark
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Dept. of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Denmark
| | | | - Camilla Christensen
- Minerva Imaging, Copenhagen, Denmark
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Dept. of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Denmark
| | | | | | | | | | | | | | - Carsten H. Nielsen
- Minerva Imaging, Copenhagen, Denmark
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Dept. of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Denmark
| | - Andreas Kjaer
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Dept. of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Denmark
| |
Collapse
|
119
|
Milcent B, Josseaume N, Riller Q, Giglioli I, Rabia E, Deligne C, Latouche JB, Hamieh M, Couture A, Toutirais O, Lone YC, Jeger-Madiot R, Graff-Dubois S, Amorim S, Loiseau P, Toubert A, Brice P, Thieblemont C, Teillaud JL, Sibéril S. Presence of T cells directed against CD20-derived peptides in healthy individuals and lymphoma patients. Cancer Immunol Immunother 2019; 68:1561-1572. [PMID: 31494742 PMCID: PMC6805815 DOI: 10.1007/s00262-019-02389-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023]
Abstract
Preclinical and clinical studies have suggested that cancer treatment with antitumor antibodies induces a specific adaptive T cell response. A central role in this process has been attributed to CD4+ T cells, but the relevant T cell epitopes, mostly derived from non-mutated self-antigens, are largely unknown. In this study, we have characterized human CD20-derived epitopes restricted by HLA-DR1, HLA-DR3, HLA-DR4, and HLA-DR7, and investigated whether T cell responses directed against CD20-derived peptides can be elicited in human HLA-DR-transgenic mice and human samples. Based on in vitro binding assays to recombinant human MHC II molecules and on in vivo immunization assays in H-2 KO/HLA-A2+-DR1+ transgenic mice, we have identified 21 MHC II-restricted long peptides derived from intracellular, membrane, or extracellular domains of the human non-mutated CD20 protein that trigger in vitro IFN-γ production by PBMCs and splenocytes from healthy individuals and by PBMCs from follicular lymphoma patients. These CD20-derived MHC II-restricted peptides could serve as a therapeutic tool for improving and/or monitoring anti-CD20 T cell activity in patients treated with rituximab or other anti-CD20 antibodies.
Collapse
Affiliation(s)
- Benoit Milcent
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Nathalie Josseaume
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Quentin Riller
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Ilenia Giglioli
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Emilia Rabia
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Claire Deligne
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France
| | - Jean-Baptiste Latouche
- Inserm U1245, Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen University Hospital, Rouen, France
| | - Mohamad Hamieh
- Inserm U1245, Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen University Hospital, Rouen, France
| | - Alexandre Couture
- Inserm U1245, Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen University Hospital, Rouen, France
| | - Olivier Toutirais
- Unicaen, Inserm 1237, Physiopathology and Imaging of Neurological Disorders, Normandie University, Caen, France.,French Blood Service (Etablissement Français du Sang, EFS), Caen, France
| | - Yu-Chun Lone
- Inserm U1014, Hôpital Paul Brousse, Villejuif, France
| | - Raphaël Jeger-Madiot
- Inserm U1135, CNRS ERL8255, Center for Immunology and Microbial Infection, Paris, France
| | - Stéphanie Graff-Dubois
- Inserm U1135, CNRS ERL8255, Center for Immunology and Microbial Infection, Paris, France
| | - Sandy Amorim
- APHP, Saint-Louis Hospital, Hemato-oncology, Diderot University, Sorbonne Paris Cité, Paris, France
| | - Pascale Loiseau
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Paris, France.,Inserm UMR-S 1160, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot-Paris 7, Paris, France
| | - Antoine Toubert
- Laboratoire d'Immunologie et Histocompatibilité, Hôpital Saint-Louis, Paris, France.,Inserm UMR-S 1160, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot-Paris 7, Paris, France
| | - Pauline Brice
- APHP, Saint-Louis Hospital, Hemato-oncology, Diderot University, Sorbonne Paris Cité, Paris, France
| | - Catherine Thieblemont
- APHP, Saint-Louis Hospital, Hemato-oncology, Diderot University, Sorbonne Paris Cité, Paris, France.,EA7324 Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jean-Luc Teillaud
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France.,Laboratory "Immune Microenvironment and Immunotherapy", Sorbonne Université UMRS 1135, INSERM U.1135, Centre d'Immunologie et des Maladies Infectieuses (CIMI), Paris, France
| | - Sophie Sibéril
- Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Inserm UMRS 1138, "Cancer, Immune Control and Escape" Laboratory, Centre de Recherche des Cordeliers, Paris, France. .,Cordeliers Research Center-Inserm UMR-S 1138, "Cancer, Immune Control and Escape" Laboratory, 15 rue de l'Ecole de Médecine, 75006, Paris, France.
| |
Collapse
|
120
|
Abstract
Cancer immunotherapy aims to promote the activity of cytotoxic T lymphocytes (CTLs) within a tumour, assist the priming of tumour-specific CTLs in lymphoid organs and establish efficient and durable antitumour immunity. During priming, help signals are relayed from CD4+ T cells to CD8+ T cells by specific dendritic cells to optimize the magnitude and quality of the CTL response. In this Review, we highlight the cellular dynamics and membrane receptors that mediate CD4+ T cell help and the molecular mechanisms of the enhanced antitumour activity of CTLs. We outline how deficient CD4+ T cell help reduces the response of CTLs and how maximizing CD4+ T cell help can improve outcomes in cancer immunotherapy strategies.
Collapse
|
121
|
Negahdaripour M, Owji H, Eslami M, Zamani M, Vakili B, Sabetian S, Nezafat N, Ghasemi Y. Selected application of peptide molecules as pharmaceutical agents and in cosmeceuticals. Expert Opin Biol Ther 2019; 19:1275-1287. [PMID: 31382850 DOI: 10.1080/14712598.2019.1652592] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Peptide molecules are being vastly investigated as an emerging class of therapeutic molecules in recent years. Currently, 60 peptides have been approved by the US Food and Drug Administration (FDA), and more would enter the market in near future. Peptides have already opened their ways into cosmeceutical and food industries as well.Areas covered: Antibodies, vaccines, and antimicrobial agents are the major classes of therapeutic peptides. Additionally, peptides may be employed in drug development to support cell penetration or targeting. The interest in antimicrobial peptides is surging due to the increasing risk of antibiotic-resistant pathogens. Peptide vaccines with their significant advantages compared with traditional vaccines, are expected to find their place in coming years, especially for cancer, microbial and allergen-specific immunotherapy. The usage of peptides in cosmeceuticals is also growing rapidly.Expert opinion: Peptide synthesis has become accessible, and advances in peptide engineering, sequencing technologies, and structural bioinformatics have resulted in the rational designing of novel peptides. All these advancements would lead to the more prominent roles of peptides in the mentioned areas. In this review, we discuss applications of peptides in different fields including pharmaceuticals, cosmeceuticals, besides the critical factors in designing efficient peptide molecules.
Collapse
Affiliation(s)
- Manica Negahdaripour
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Hajar Owji
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Mahboobeh Eslami
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Mozhdeh Zamani
- Colorectal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bahareh Vakili
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran.,Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soudabeh Sabetian
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Navid Nezafat
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
| | - Younes Ghasemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Pharmaceutical Sciences Research Center, Shiraz University of Medical Science, Shiraz, Iran
| |
Collapse
|
122
|
Batham J, Lim PS, Rao S. SETDB-1: A Potential Epigenetic Regulator in Breast Cancer Metastasis. Cancers (Basel) 2019; 11:cancers11081143. [PMID: 31405032 PMCID: PMC6721492 DOI: 10.3390/cancers11081143] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023] Open
Abstract
The full epigenetic repertoire governing breast cancer metastasis is not completely understood. Here, we discuss the histone methyltransferase SET Domain Bifurcated Histone Lysine Methyltransferase 1 (SETDB1) and its role in breast cancer metastasis. SETDB1 serves as an exemplar of the difficulties faced when developing therapies that not only specifically target cancer cells but also the more elusive and aggressive stem cells that contribute to metastasis via epithelial-to-mesenchymal transition and confer resistance to therapies.
Collapse
Affiliation(s)
- Jacob Batham
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia
| | - Pek Siew Lim
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| | - Sudha Rao
- Melanie Swan Memorial Translational Centre, Faculty of Sci-Tech, University of Canberra, Bruce ACT 2617, Australia.
| |
Collapse
|
123
|
Mohammadi MR, Corbo C, Molinaro R, Lakey JRT. Biohybrid Nanoparticles to Negotiate with Biological Barriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1902333. [PMID: 31250985 DOI: 10.1002/smll.201902333] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 05/30/2019] [Indexed: 06/09/2023]
Abstract
Incapability of effective cross-talk with biological environments has partly impaired the in vivo functionality of nanoparticles (NPs). Homing, biodistribution, and function of NPs could be engineered through regulating their interactions with in vivo niches. Inspired by communications in biological systems, endowing a "biological identity" to synthetic NPs is one approach to control their biodistribution, and immunonegotiation profiles. This synthetic-biological combination is referred to as biohybrid NPs, which comprise both i) engineerable, readily producible, and trackable synthetic NPs as well as ii) biological moieties with the capability to cross-talk with immunological barriers. Here, the latest understanding on the in vivo interactions of NPs, biological barriers they face, and emerging methods for quantitative measurements of NPs' biodistribution are reviewed. Some key biomolecules that have emerged as negotiators with the immune system in the context of cancer and autoimmunity, and their inspirations on biohybrid NPs are introduced. Critical design considerations for efficient cross-talk between NPs and innate and adaptive immunity followed by hybridization methods are also discussed. Finally, clinical translation challenges and future perspectives regarding biohybrid NPs are discussed.
Collapse
Affiliation(s)
- M Rezaa Mohammadi
- Department of Chemical Engineering and Materials Science, University of California, Irvine, Sue and Bill Gross Stem Cell Research Center, Irvine, CA, 92697, USA
- Department of Surgery and Biomedical Engineering, University of California, Irvine, CA, 92697, USA
| | - Claudia Corbo
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, MI, 20126, Italy
| | - Roberto Molinaro
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, PU, 61029, Italy
- Department of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jonathan R T Lakey
- Department of Surgery and Biomedical Engineering, University of California, Irvine, CA, 92697, USA
| |
Collapse
|
124
|
Distinct prognostic value of circulating anti-telomerase CD4 + Th1 immunity and exhausted PD-1 +/TIM-3 + T cells in lung cancer. Br J Cancer 2019; 121:405-416. [PMID: 31358938 PMCID: PMC6738094 DOI: 10.1038/s41416-019-0531-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 06/25/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Despite the critical roles of Th1-polarised CD4+ T cells in cancer immunosurveillance, the translation of their potential to clinical use remains challenging. Here, we investigate the clinical relevance of circulating antitumor Th1 immunity in non-small cell lung cancer (NSCLC). METHODS The circulating antitumor Th1 response was assessed by the ELISpot assay in 170 NSCLC patients using a mixture of HLA class II-restricted peptides from telomerase (TERT). Phenotyping of blood immune cells was performed by flow cytometry. RESULTS TERT-reactive CD4 T-cell response was detected in 35% of NSCLC patients before any treatment. Functional analysis showed that these cells were effector memory and Th1 polarised capable to produce effector cytokines, such as IFN-γ, TNF-α and IL-2. The presence of anti-TERT Th1 response was inversely correlated with the level of exhausted PD-1+/TIM-3+CD4 T cells. The level of these two immune parameters differentially affected the survival, so that increased level of anti-TERT Th1 response and low rate of exhausted PD-1+TIM-3+CD4+ T cells were associated with a better prognosis. CONCLUSIONS Systemic anti-TERT Th1 response plays a strong antitumor protective role in NSCLC. This study underlines the potential interest of monitoring circulating antitumor Th1 response for patients' stratification and therapy decision.
Collapse
|
125
|
Melssen MM, Petroni GR, Chianese-Bullock KA, Wages NA, Grosh WW, Varhegyi N, Smolkin ME, Smith KT, Galeassi NV, Deacon DH, Gaughan EM, Slingluff CL. A multipeptide vaccine plus toll-like receptor agonists LPS or polyICLC in combination with incomplete Freund's adjuvant in melanoma patients. J Immunother Cancer 2019; 7:163. [PMID: 31248461 PMCID: PMC6598303 DOI: 10.1186/s40425-019-0625-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cancer vaccines require adjuvants to induce effective immune responses; however, there is no consensus on optimal adjuvants. We hypothesized that toll-like receptor (TLR)3 agonist polyICLC or TLR4 agonist lipopolysaccharide (LPS), combined with CD4 T cell activation, would support strong and durable CD8+ T cell responses, whereas addition of an incomplete Freund's adjuvant (IFA) would reduce magnitude and persistence of immune responses. PATIENTS AND METHODS Participants with resected stage IIB-IV melanoma received a vaccine comprised of 12 melanoma peptides restricted by Class I MHC (12MP), plus a tetanus helper peptide (Tet). Participants were randomly assigned 2:1 to cohort 1 (LPS dose-escalation) or cohort 2 (polyICLC). Each cohort included 3 subgroups (a-c), receiving 12MP + Tet + TLR agonist without IFA (0), or with IFA in vaccine one (V1), or all six vaccines (V6). Toxicities were recorded (CTCAE v4). T cell responses were measured with IFNγ ELIspot assay ex vivo or after one in vitro stimulation (IVS). RESULTS Fifty-three eligible patients were enrolled, of which fifty-one were treated. Treatment-related dose-limiting toxicities (DLTs) were observed in 0/33 patients in cohort 1 and in 2/18 patients in cohort 2 (11%). CD8 T cell responses to 12MP were detected ex vivo in cohort 1 (42%) and in cohort 2 (56%) and in 18, 50, and 72% for subgroups V0, V1, and V6, respectively. T cell responses to melanoma peptides were more durable and of highest magnitude for IFA V6. CONCLUSIONS LPS and polyICLC are safe and effective vaccine adjuvants when combined with IFA. Contrary to the central hypothesis, IFA enhanced T cell responses to peptide vaccines when added to TLR agonists. Future studies will aim to understand mechanisms underlying the favorable effects with IFA. TRIAL REGISTRATION The clinical trial Mel58 was performed with IRB (#15781) and FDA approval and is registered with Clinicaltrials.gov on April 25, 2012 (NCT01585350). Patients provided written informed consent to participate. Enrollment started on June 24, 2012.
Collapse
Affiliation(s)
- Marit M Melssen
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, 1352 Pinn Hall, P.O. Box 801457, Charlottesville, VA, 22908, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Gina R Petroni
- Department of Public Health Sciences/Division of Translational Research & Applied Statistics, University of Virginia, Charlottesville, VA, USA
| | - Kimberly A Chianese-Bullock
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, 1352 Pinn Hall, P.O. Box 801457, Charlottesville, VA, 22908, USA
| | - Nolan A Wages
- Department of Public Health Sciences/Division of Translational Research & Applied Statistics, University of Virginia, Charlottesville, VA, USA
| | - William W Grosh
- Department of Medicine/Division of Hematology/Oncology, University of Virginia, Charlottesville, VA, USA
| | - Nikole Varhegyi
- Department of Public Health Sciences/Division of Translational Research & Applied Statistics, University of Virginia, Charlottesville, VA, USA
| | - Mark E Smolkin
- Department of Public Health Sciences/Division of Translational Research & Applied Statistics, University of Virginia, Charlottesville, VA, USA
| | - Kelly T Smith
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, 1352 Pinn Hall, P.O. Box 801457, Charlottesville, VA, 22908, USA
| | - Nadejda V Galeassi
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, 1352 Pinn Hall, P.O. Box 801457, Charlottesville, VA, 22908, USA
| | - Donna H Deacon
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, 1352 Pinn Hall, P.O. Box 801457, Charlottesville, VA, 22908, USA
| | - Elizabeth M Gaughan
- Department of Medicine/Division of Hematology/Oncology, University of Virginia, Charlottesville, VA, USA
| | - Craig L Slingluff
- Department of Surgery/Division of Surgical Oncology and the Human Immune Therapy Center, Cancer Center, University of Virginia, 1352 Pinn Hall, P.O. Box 801457, Charlottesville, VA, 22908, USA.
| |
Collapse
|
126
|
Khan IA, Hwang S, Moretto M. Toxoplasma gondii: CD8 T Cells Cry for CD4 Help. Front Cell Infect Microbiol 2019; 9:136. [PMID: 31119107 PMCID: PMC6504686 DOI: 10.3389/fcimb.2019.00136] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/15/2019] [Indexed: 11/13/2022] Open
Abstract
Toxoplasma gondii, an apicomplexan parasite, is a pathogenic protozoan that can infect the central nervous system. In pregnant women, infection can result in congenital problems of the fetus, while in immunocompromised individual it can lead to severe neurological consequences. Although CD8 T cells play an important effector role in controlling the chronic infection, their maintenance is dependent on the critical help provided by CD4 T cells. In a recent study, we demonstrated that reactivation of the infection in chronically infected host is a consequence of CD8 T dysfunction caused by CD4 T cell exhaustion. Furthermore, treatment of chronically infected host with antigen-specific non-exhausted CD4 T cells can restore CD8 T cell functionality and prevent reactivation of the latent infection. The exhaustion status of CD4 T cells is mediated by the increased expression of the transcription factor BLIMP-1, and deletion of this molecule led to the restoration of CD4 T cell function, reversal of CD8 exhaustion and prevention of reactivation of the latent infection. In a recent study from our laboratory, we also observed an increased expression of miR146a levels by CD4 T cells from the chronically infected animals. Recent reports have demonstrated that microRNAs (especially miR146a) has a strong impact on the immune system of T. gondii infected host. Whether these molecules have any role in the BLIMP-1 up-regulation and dysfunctionality of these cells needs to be investigated.
Collapse
Affiliation(s)
- Imtiaz A. Khan
- Department Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC, United States
| | | | | |
Collapse
|
127
|
Shukla S. A Viral Nanoparticle Cancer Vaccine Delays Tumor Progression and Prolongs Survival in a HER2 + Tumor Mouse Model. ADVANCED THERAPEUTICS 2019; 2:1800139. [PMID: 33855164 PMCID: PMC8043622 DOI: 10.1002/adtp.201800139] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Indexed: 12/17/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) overexpression is associated with aggressive tumors with increased incidence of metastasis and recurrence. Therapeutic antibodies such as Trastuzumab inhibit tumor growth through blockade of HER2 receptors. However, the short lifespan of such therapeutic antibodies necessitates repeat administrations with ensuing cardiac toxicity and development of resistance, while offering no protection against relapse. Cancer vaccines targeting HER2 can overcome these shortcomings of passive immunotherapy by instigating an endogenous and sustained immune response and memory against the cancer antigen. The efficacy of a viral nanoparticle (VNP)-based cancer vaccine is demonstrated here in activating a potent anti-HER2 immune response that delays progression of primary tumors as well as metastases and prolongs survival in mice. The results illustrate that the VNP-based vaccine instigates HER2-specific antibodies as well as effector and memory T cells, which contributes to the effectiveness of the vaccine. Given the highly aggressive course of HER2+ cancers, inhibition of disease progression by such cancer vaccines could provide a critical window for interventions with other adjuvant therapies. Moreover, the immune memory generated by this viral nanoparticle-based cancer vaccine could mitigate relapse of the disease.
Collapse
|
128
|
Kroon P, Frijlink E, Iglesias-Guimarais V, Volkov A, van Buuren MM, Schumacher TN, Verheij M, Borst J, Verbrugge I. Radiotherapy and Cisplatin Increase Immunotherapy Efficacy by Enabling Local and Systemic Intratumoral T-cell Activity. Cancer Immunol Res 2019; 7:670-682. [PMID: 30782666 DOI: 10.1158/2326-6066.cir-18-0654] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/18/2018] [Accepted: 02/11/2019] [Indexed: 11/16/2022]
Abstract
To increase cancer immunotherapy success, PD-1 blockade must be combined with rationally selected treatments. Here, we examined, in a poorly immunogenic mouse breast cancer model, the potential of antibody-based immunomodulation and conventional anticancer treatments to collaborate with anti-PD-1 treatment. One requirement to improve anti-PD-1-mediated tumor control was to promote tumor-specific cytotoxic T-cell (CTL) priming, which was achieved by stimulating the CD137 costimulatory receptor. A second requirement was to overrule PD-1-unrelated mechanisms of CTL suppression in the tumor microenvironment (TME). This was achieved by radiotherapy and cisplatin treatment. In the context of CD137/PD-1-targeting immunotherapy, radiotherapy allowed for tumor elimination by altering the TME, rather than intrinsic CTL functionality. Combining this radioimmunotherapy regimen with low-dose cisplatin improved CTL-dependent regression of a contralateral tumor outside the radiation field. Thus, systemic tumor control may be achieved by combining immunotherapy protocols that promote T-cell priming with (chemo)radiation protocols that permit CTL activity in both the irradiated tumor and (occult) metastases.
Collapse
Affiliation(s)
- Paula Kroon
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands
| | - Elselien Frijlink
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands
| | - Victoria Iglesias-Guimarais
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands
| | - Andriy Volkov
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands
| | - Marit M van Buuren
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands
| | - Ton N Schumacher
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands.,Division of Molecular Oncology and Immunology, Oncode Institute, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands
| | - Marcel Verheij
- Department of Radiation Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands
| | - Jannie Borst
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands
| | - Inge Verbrugge
- Division of Tumor Biology and Immunology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
129
|
Khan IA, Ouellette C, Chen K, Moretto M. Toxoplasma: Immunity and Pathogenesis. CURRENT CLINICAL MICROBIOLOGY REPORTS 2019; 6:44-50. [PMID: 31179204 DOI: 10.1007/s40588-019-0114-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Toxoplasma gondii infection induces a strong immunity in the host. Although the response is manifested by innate response during early infection, adaptive immunity is critical for long-term protection. Amongst the adaptive immune response CD4 T cells play an important helper role for CD8 T cells which are the primary effector cells responsible for controlling the infection. Notwithstanding the induction of robust CD8 T immunity during acute infection, the parasite is not eradicated. One of the reasons for this is the functional exhaustion of CD8 T cells during latent infection. Recent studies from our laboratory have reported that primary cause of CD8 T cell exhaustion is compromised CD4 T cell help during latent toxoplasmosis. CD8 T cell dysfunctionality is preceded by CD4 exhaustion and effector immunity is severely compromised.
Collapse
Affiliation(s)
- Imtiaz A Khan
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC 20037
| | - Charlotte Ouellette
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC 20037
| | - Keer Chen
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC 20037
| | - Magali Moretto
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University, Washington DC 20037
| |
Collapse
|
130
|
Enfield KSS, Martin SD, Marshall EA, Kung SHY, Gallagher P, Milne K, Chen Z, Nelson BH, Lam S, English JC, MacAulay CE, Lam WL, Guillaud M. Hyperspectral cell sociology reveals spatial tumor-immune cell interactions associated with lung cancer recurrence. J Immunother Cancer 2019; 7:13. [PMID: 30651131 PMCID: PMC6335759 DOI: 10.1186/s40425-018-0488-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/19/2018] [Indexed: 01/06/2023] Open
Abstract
Background The tumor microenvironment (TME) is a complex mixture of tumor epithelium, stroma and immune cells, and the immune component of the TME is highly prognostic for tumor progression and patient outcome. In lung cancer, anti-PD-1 therapy significantly improves patient survival through activation of T cell cytotoxicity against tumor cells. Direct contact between CD8+ T cells and target cells is necessary for CD8+ T cell activity, indicating that spatial organization of immune cells within the TME reflects a critical process in anti-tumor immunity. Current immunohistochemistry (IHC) imaging techniques identify immune cell numbers and densities, but lack assessment of cell–cell spatial relationships (or “cell sociology”). Immune functionality, however, is often dictated by cell-to-cell contact and cannot be resolved by simple metrics of cell density (for example, number of cells per mm2). To address this issue, we developed a Hyperspectral Cell Sociology technology platform for the analysis of cell–cell interactions in multi-channel IHC-stained tissue. Methods Tissue sections of primary tumors from lung adenocarcinoma patients with known clinical outcome were stained using multiplex IHC for CD3, CD8, and CD79a, and hyperspectral image analysis determined the phenotype of all cells. A Voronoi diagram for each cell was used to approximate cell boundaries, and the cell type of all neighboring cells was identified and quantified. Monte Carlo analysis was used to assess whether cell sociology patterns were likely due to random distributions of the cells. Results High density of intra-tumoral CD8+ T cells was significantly associated with non-recurrence of tumors. A cell sociology pattern of CD8+ T cells surrounded by tumor cells was more significantly associated with non-recurrence compared to CD8+ T cell density alone. CD3+ CD8- T cells surrounded by tumor cells was also associated with non-recurrence, but at a similar significance as cell density alone. Cell sociology metrics improved recurrence classifications of 12 patients. Monte Carlo re-sampling analysis determined that these cell sociology patterns were non-random. Conclusion Hyperspectral Cell Sociology expands our understanding of the complex interplay between tumor cells and immune infiltrate. This technology could improve predictions of responses to immunotherapy and lead to a deeper understanding of anti-tumor immunity. Electronic supplementary material The online version of this article (10.1186/s40425-018-0488-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katey S S Enfield
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.
| | - Spencer D Martin
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Erin A Marshall
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| | - Sonia H Y Kung
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| | - Paul Gallagher
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| | - Katy Milne
- Deeley Research Centre, Victoria, BC, Canada
| | - Zhaoyang Chen
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| | - Brad H Nelson
- Deeley Research Centre, Victoria, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Stephen Lam
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John C English
- Pathology and Laboratory Medicine, Vancouver Coastal Health, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Calum E MacAulay
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wan L Lam
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Martial Guillaud
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| |
Collapse
|
131
|
Nakata J, Nakajima H, Hayashibara H, Imafuku K, Morimoto S, Fujiki F, Motooka D, Okuzaki D, Hasegawa K, Hosen N, Tsuboi A, Oka Y, Kumanogoh A, Oji Y, Sugiyama H. Extremely strong infiltration of WT1-specific CTLs into mouse tumor by the combination vaccine with WT1-specific CTL and helper peptides. Oncotarget 2018; 9:36029-36038. [PMID: 30542516 PMCID: PMC6267595 DOI: 10.18632/oncotarget.26338] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/01/2018] [Indexed: 11/29/2022] Open
Abstract
In immunotherapy by cancer antigen-derived peptide vaccine, vaccination of cytotoxic T lymphocyte (CTL) peptide alone is common, while it remains unclear whether the addition of helper peptide vaccine to the CTL peptide vaccine is of great advantage for the enhancement of tumor immunity. In the present study, combination vaccine of Wilms’ tumor gene 1(WT1) protein-derived CTL and helper peptides induced the strong infiltration of WT1-specific CD8+ T cells into mouse tumor at frequencies of 8.8%, resulting in the formation of multiple microscopic necrotic lesions in the tumor, whereas the frequencies of WT1-specific CD8+ T cell infiltration into the tumor in the vaccination of the CTL peptide alone were only 0.32%. The majority of the infiltrated WT1-specific CD8+ T cells was effector phenotype T cells, but importantly, WT1-specific CD8+CD44+CD62L+CD103+ resident memory T cells, which could differentiate into a lot of effector phenotype T cells, existed in the tumor of mice vaccinated with the both WT1 peptides. Furthermore, T-cell receptor repertoire analysis showed the oligoclonality of these tumor infiltrating WT1 tetramer+ CD8+ T cells, and 3 clones occupied about half of them. These results indicated that WT1-specific CD4+ T cells played an essential role not only in the priming and activation of WT1-specific CD8+ T cells, but also in trafficking and infiltration of the CD8+ T cells into tumors. These results should provide us with the concept that in the clinical setting, combination vaccine of WT1-specific CTL and helper peptides would be more advantageous than the CTL peptide vaccine alone.
Collapse
Affiliation(s)
- Jun Nakata
- Department of Biomedical Informatics, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Hiroko Nakajima
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Hiromu Hayashibara
- Department of Biomedical Informatics, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Kanako Imafuku
- Department of Biomedical Informatics, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Soyoko Morimoto
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Kana Hasegawa
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Naoki Hosen
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Yoshihiro Oka
- Department of Cancer Stem Cell Biology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan.,Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan.,Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan.,Department of Immunopathology, WP1 Immunology Frontier Research Center, Osaka University, Suita City, Osaka 565-0871, Japan
| | - Yusuke Oji
- Department of Functional Diagnostic Science, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| | - Haruo Sugiyama
- Department of Cancer Immunology, Osaka University Graduate School of Medicine, Suita City, Osaka 565-0871, Japan
| |
Collapse
|
132
|
Benonisson H, Altıntaş I, Sluijter M, Verploegen S, Labrijn AF, Schuurhuis DH, Houtkamp MA, Verbeek JS, Schuurman J, van Hall T. CD3-Bispecific Antibody Therapy Turns Solid Tumors into Inflammatory Sites but Does Not Install Protective Memory. Mol Cancer Ther 2018; 18:312-322. [PMID: 30381448 DOI: 10.1158/1535-7163.mct-18-0679] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/01/2018] [Accepted: 10/24/2018] [Indexed: 11/16/2022]
Abstract
Immunotherapy of cancer with CD3-targeting bispecific antibodies (CD3 bsAb) is a fast developing field, and multiple tumor-associated antigens (TAA) are evaluated for hematologic and solid malignancies. The efficacy of these CD3 bsAb is usually examined in xenograft mouse tumor models with human T cells or in genetically engineered mouse models, where human TAA are introduced. These models often fail to fully recapitulate the natural tumor environment, especially for solid cancers, because of interspecies differences. Here, we investigated the systemic and intratumoral effects of a mouse CD3 bsAb in a fully immune-competent mouse melanoma model. Systemic administration of 0.5 mg/kg antibody induced a brief overall T-cell activation that was selectively sustained in the tumor microenvironment for several days. A fast subsequent influx of inflammatory macrophages into the tumor microenvironment was observed, followed by an increase in the number of CD4+ and CD8+ T cells. Although the capacity to directly kill melanoma cells in vitro was very modest, optimal tumor elimination was observed in vivo, even in the absence of CD8+ T cells, implying a redundancy in T-cell subsets for therapeutic efficacy. Finally, we took advantage of the full immune competence of our mouse model and tested immune memory induction. Despite a strong initial immunity against melanoma, treatment with the CD3 bsAb did not install protective memory responses. The observed mechanisms of action revealed in this immune-competent mouse model might form a rational basis for combinatorial approaches.
Collapse
Affiliation(s)
- Hreinn Benonisson
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
133
|
ICOSL-augmented adenoviral-based vaccination induces a bipolar Th17/Th1 T cell response against unglycosylated MUC1 antigen. Vaccine 2018; 36:6262-6269. [PMID: 30219366 DOI: 10.1016/j.vaccine.2018.09.010] [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: 03/07/2018] [Revised: 07/18/2018] [Accepted: 09/07/2018] [Indexed: 01/14/2023]
Abstract
Cellular immunity established via immunotherapy holds the potential to eliminate solid tumors. Yet, cancer vaccines have failed to induce tumor-reactive T cells of sufficient quality to control disease. The inducible T cell costimulator (ICOS) pathway has been implicated in both the selective induction of immunity over tolerance as well as licensing of IL-17-polarized cellular immunity. Herein, we evaluated the ability of ICOS ligand (ICOSL) to augment the immunogenicity of adenoviral-based vaccination targeting the unglycosylated MUC1 peptide antigen. Vaccination disrupted immunotolerance in a transgenic mouse model recognizing human MUC1 as a self-antigen, inducing robust MUC1-specific immunity. Augmenting vaccination with ICOSL induced a bipolar Th17/Th1 effector profile, marked by increased MUC1-specific IL-17A production and RORγt expression in CD4+ but not CD8+ T cells which predominantly expressed IFNγ/IL-2 and T-bet. The polarization and maintenance of Th17 cells established following ICOSL augmented vaccination was highly durable, with elevated IL-17A and RORγt levels detected in CD4+ T cells up to 10 months after initial immunization. Furthermore, provision of ICOSL significantly enhanced MUC1-specific IgG antibody in response to immunization. ICOSL signaling dramatically influenced CD4+ T cell phenotype, altering gene expression of transcription factors and regulators of effector function following immunization. Interestingly, ICOSL augmentation failed to alter the transcriptional profile of CD8+ T cells following immunization, affecting the magnitude, but not distribution, of gene expression. Collectively, ICOSL supports the induction of durable, antigen-specific Th17/Th1-mediated immunity in vivo, establishing a vaccination platform to enhance CD4+ T cell-mediated antitumor immunity and providing a crucial component of an effective cancer vaccine.
Collapse
|
134
|
Bezu L, Kepp O, Cerrato G, Pol J, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Peptide-based vaccines in anticancer therapy. Oncoimmunology 2018; 7:e1511506. [PMID: 30524907 PMCID: PMC6279318 DOI: 10.1080/2162402x.2018.1511506] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
Peptide-based anticancer vaccination aims at stimulating an immune response against one or multiple tumor-associated antigens (TAAs) following immunization with purified, recombinant or synthetically engineered epitopes. Despite high expectations, the peptide-based vaccines that have been explored in the clinic so far had limited therapeutic activity, largely due to cancer cell-intrinsic alterations that minimize antigenicity and/or changes in the tumor microenvironment that foster immunosuppression. Several strategies have been developed to overcome such limitations, including the use of immunostimulatory adjuvants, the co-treatment with cytotoxic anticancer therapies that enable the coordinated release of damage-associated molecular patterns, and the concomitant blockade of immune checkpoints. Personalized peptide-based vaccines are also being explored for therapeutic activity in the clinic. Here, we review recent preclinical and clinical progress in the use of peptide-based vaccines as anticancer therapeutics.Abbreviations: CMP: carbohydrate-mimetic peptide; CMV: cytomegalovirus; DC: dendritic cell; FDA: Food and Drug Administration; HPV: human papillomavirus; MDS: myelodysplastic syndrome; MHP: melanoma helper vaccine; NSCLC: non-small cell lung carcinoma; ODD: orphan drug designation; PPV: personalized peptide vaccination; SLP: synthetic long peptide; TAA: tumor-associated antigen; TNA: tumor neoantigen
Collapse
Affiliation(s)
- Lucillia Bezu
- Faculty of Medicine, University of Paris Sud/Paris XI, Le Kremlin-Bicêtre, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Giulia Cerrato
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Jonathan Pol
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio, Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Faculty of Medicine, University of Paris Sud/Paris XI, Le Kremlin-Bicêtre, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,INSERM, U1015, Gustave Roussy Cancer Campus, Villejuif, France
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France.,Equipe 11 labellisée Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers,Paris, France.,U1138, INSERM, Paris, France.,Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, Paris, France.,Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
135
|
Tan S, Wang K, Sun F, Li Y, Gao Y. CXCL9 promotes prostate cancer progression through inhibition of cytokines from T cells. Mol Med Rep 2018; 18:1305-1310. [PMID: 29901197 PMCID: PMC6072144 DOI: 10.3892/mmr.2018.9152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 03/27/2018] [Indexed: 01/29/2023] Open
Abstract
Chemokines have been demonstrated to serve an important role in a variety of diseases, particularly in tumor progression. There have been numerous studies that have reported that T cells serve major roles in tumor progression. However, the function of CXC motif chemokine ligand 9 (CXCL9) in prostate cancer remains unknown. The present study aimed to investigate the role of CXCL9 in prostate cancer. A prostate cancer mouse model was generated by treating C57/BL‑6 and B6.Cg‑Selplgtm1Fur/J mice with 3,2'‑dimethyl 4‑aminobiphenyl (DMAB). Hematoxylin and eosin staining detected the histopathological alterations of mouse prostate tissues. Immunohistochemistry (IHC) staining determined cell proliferation of the mice. Flow cytometry was used to detect the alterations of T cells in C57+DMAB or CXCL9+DMAB mice. Immunofluorescence revealed that there was positive expression of interleukin‑6 (IL‑6) and transforming growth factor (TGF)‑β in the mouse tissues. The survival rates of C57+DMAB and CXCL9+DMAB mice was analyzed. The association of CXCL9 expression and clinical stages was also evaluated. Results revealed that prostate cancer pathology and cell proliferation in CXCL9+DMAB mice were significantly greater compared with the C57+DMAB mice. Compared with C57+DMAB mice, the number of T cells in peripheral blood and spleen of CXCL9+DMAB mice was significantly reduced. IHC demonstrated that the expression of IL‑6 and TGF‑β was significantly downregulated in the CXCL9+DMAB mice. The survival rate of CXCL9+DMAB mice was significantly decreased compared with the C57+DMAB mice. In addition, reverse transcription‑quantitative polymerase chain reaction analysis demonstrated that CXCL9 mRNA expression in clinical samples was positively associated with clinical pathological stages of prostate cancer. In conclusion, CXCL9 may promote prostate cancer progression via inhibition of cytokines from T cells.
Collapse
Affiliation(s)
- Shanfeng Tan
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Kai Wang
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Fuguang Sun
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Yang Li
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| | - Yisheng Gao
- Department of Urology, Linyi People's Hospital, Linyi, Shandong 276000, P.R. China
| |
Collapse
|
136
|
Tsukamoto H, Fujieda K, Miyashita A, Fukushima S, Ikeda T, Kubo Y, Senju S, Ihn H, Nishimura Y, Oshiumi H. Combined Blockade of IL6 and PD-1/PD-L1 Signaling Abrogates Mutual Regulation of Their Immunosuppressive Effects in the Tumor Microenvironment. Cancer Res 2018; 78:5011-5022. [PMID: 29967259 DOI: 10.1158/0008-5472.can-18-0118] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/24/2018] [Accepted: 06/27/2018] [Indexed: 11/16/2022]
Abstract
Recently emerging cancer immunotherapies combine the applications of therapeutics to disrupt the immunosuppressive conditions in tumor-bearing hosts. In this study, we found that targeting the proinflammatory cytokine IL6 enhances tumor-specific Th1 responses and subsequent antitumor effects in tumor-bearing mice. IL6 blockade upregulated expression of the immune checkpoint molecule programmed death-ligand 1 (PD-L1) on melanoma cells. This PD-L1 induction was canceled in IFNγ-deficient mice or CD4+ T cell-depleted mice, suggesting that CD4+ T cell-derived IFNγ is important for PD-L1 induction in tumor-bearing hosts. In some patients with melanoma, however, treatment with the anti-PD-1 antibody nivolumab increased systemic levels of IL6, which was associated with poor clinical responses. This PD-L1 blockade-evoked induction of IL6 was reproducible in melanoma-bearing mice. We found that PD-1/PD-L1 blockade prompted PD-1+ macrophages to produce IL6 in the tumor microenvironment. Depletion of macrophages in melanoma-bearing mice reduced the levels of IL6 during PD-L1 blockade, suggesting macrophages are responsible for the IL6-mediated defective CD4+ Th1 response. Combined blockade of the mutually regulated immunosuppressive activities of IL6 and PD-1/PD-L1 signals enhanced expression of T cell-attracting chemokines and promoted infiltration of IFNγ-producing CD4+ T cells in tumor tissues, exerting a synergistic antitumor effect, whereas PD-L1 blockade alone did not promote Th1 response. Collectively, these findings suggest that IL6 is a rational immunosuppressive target for overcoming the narrow therapeutic window of anti-PD-1/PD-L1 therapy.Significance: These findings advance our understanding of IL6-PD1/PD-L1 cross-talk in the tumor microenvironment and provide clues for targeted interventional therapy that may prove more effective against cancer. Cancer Res; 78(17); 5011-22. ©2018 AACR.
Collapse
Affiliation(s)
| | - Koji Fujieda
- Department of Immunogenetics, Kumamoto University, Kumamoto, Japan
| | - Azusa Miyashita
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tokunori Ikeda
- Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yosuke Kubo
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Kumamoto University, Kumamoto, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Kumamoto University, Kumamoto, Japan.,Nishimura Project Laboratory, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
137
|
Dalle Vedove E, Costabile G, Merkel OM. Mannose and Mannose-6-Phosphate Receptor-Targeted Drug Delivery Systems and Their Application in Cancer Therapy. Adv Healthc Mater 2018; 7:e1701398. [PMID: 29719138 PMCID: PMC6108418 DOI: 10.1002/adhm.201701398] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/16/2018] [Indexed: 12/21/2022]
Abstract
In order to overcome the main disadvantages of conventional cancer therapies, which prove to be inadequate because of their lack of selectivity, the development of targeted delivery systems is one of the main focuses in anticancer research. It is repeatedly shown that decorating the surface of nanocarriers with high-affinity targeting ligands, such as peptides or small molecules, is an effective way to selectively deliver therapeutics by enhancing their specific cellular uptake via the binding between a specific receptor and the nanosystems. Nowadays, the need of finding new potential biological targets with a high endocytic efficiency as well as a low tendency to mutate is urgent and, in this context, mannose and mannose-6-phosphate receptors appear promising to target anticancer drugs to cells where their expression is upregulated. Moreover, they open the path to encouraging applications in immune-based and gene therapies as well as in theragnostic purposes. In this work, the potential of mannose- and mannose-6-phosphate-targeted delivery systems in cancer therapy is discussed, emphasizing their broad application both in direct treatments against cancer cells with conventional chemotherapeutics or by gene therapy and also their encouraging capabilities in immunotherapy and diagnostics purposes.
Collapse
Affiliation(s)
- Elena Dalle Vedove
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, 81337 Munich, Germany
| | - Gabriella Costabile
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, 81337 Munich, Germany
| | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, 81337 Munich, Germany
| |
Collapse
|
138
|
Ueno T, Tsuchikawa T, Hatanaka KC, Hatanaka Y, Mitsuhashi T, Nakanishi Y, Noji T, Nakamura T, Okamura K, Matsuno Y, Hirano S. Prognostic impact of programmed cell death ligand 1 (PD-L1) expression and its association with epithelial-mesenchymal transition in extrahepatic cholangiocarcinoma. Oncotarget 2018; 9:20034-20047. [PMID: 29732001 PMCID: PMC5929444 DOI: 10.18632/oncotarget.25050] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 03/21/2018] [Indexed: 12/31/2022] Open
Abstract
Extrahepatic cholangiocarcinoma (eCCA) has a poor prognosis. Although the possibility of immunotherapy has been studied, immune checkpoint molecules such as programmed death ligand 1 (PD-L1) in eCCA are not well understood. Epithelial-mesenchymal transition (EMT) has recently been shown to regulate PD-L1 expression. Our aims were to assess the clinicopathological significance of tumor-infiltrating lymphocytes (TILs) and tumor PD-L1 expression in eCCA and to compare these immune responses with EMT marker expression. In this retrospective study, we conducted immunohistochemical analyses for 117 patients with eCCA. We stained for CD4, CD8, Foxp3, and PD-L1 as markers reflecting local immune responses, and for E-cadherin, N-cadherin, vimentin, ZEB1, ZEB2, SNAIL, and TWIST as markers associated with EMT. High numbers of CD4+ and CD8+ TILs correlated with node-negative (P = 0.009 and P = 0.046, respectively) and low SNAIL expression (P = 0.016 and P = 0.022, respectively). High PD-L1 expression was associated with poor histopathological classification (P = 0.034), and low E-cadherin (P = 0.001), high N-cadherin (P = 0.044), high vimentin (P < 0.001) and high ZEB1 (P = 0.036) expression. Multivariate analysis showed that CD4+ TILs, PD-L1 expression and N-cadherin expression were independent prognostic factors (hazard ratio (HR) = 0.61; 95% confidence interval (CI) = 0.38-1.00; HR=4.27; 95% CI = 1.82-9.39; HR = 2.20; 95% CI = 1.18-3.92, respectively). These findings could help to identify potential biomarkers for predicting not only the prognosis, but also the therapeutic response to immunotherapy for eCCA.
Collapse
Affiliation(s)
- Takashi Ueno
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Takahiro Tsuchikawa
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Kanako C. Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Yutaka Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Tomoko Mitsuhashi
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Yoshitsugu Nakanishi
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takehiro Noji
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Toru Nakamura
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Keisuke Okamura
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Satoshi Hirano
- Department of Gastroenterological Surgery II, Division of Surgery, Faculty of Medicine, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
139
|
Hakim MS, Ikram A, Zhou J, Wang W, Peppelenbosch MP, Pan Q. Immunity against hepatitis E virus infection: Implications for therapy and vaccine development. Rev Med Virol 2017; 28. [PMID: 29272060 DOI: 10.1002/rmv.1964] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/10/2017] [Accepted: 11/14/2017] [Indexed: 12/20/2022]
Abstract
Hepatitis E virus (HEV) is the leading cause of acute viral hepatitis worldwide and an emerging cause of chronic infection in immunocompromised patients. As with viral infections in general, immune responses are critical to determine the outcome of HEV infection. Accumulating studies in cell culture, animal models and patients have improved our understanding of HEV immunopathogenesis and informed the development of new antiviral therapies and effective vaccines. In this review, we discuss the recent progress on innate and adaptive immunity in HEV infection, and the implications for the devolopment of effective vaccines and immune-based therapies.
Collapse
Affiliation(s)
- Mohamad S Hakim
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center and Postgraduate School Molecular Medicine, Rotterdam, The Netherlands.,Department of Microbiology, Faculty of Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Aqsa Ikram
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center and Postgraduate School Molecular Medicine, Rotterdam, The Netherlands.,Atta-Ur-Rahman School of Applied Biosciences, National University of Science and Technology, Islamabad, Pakistan
| | - Jianhua Zhou
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center and Postgraduate School Molecular Medicine, Rotterdam, The Netherlands.,State Key Laboratory of Veterinary Etiological Biology, National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, PR China
| | - Wenshi Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center and Postgraduate School Molecular Medicine, Rotterdam, The Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center and Postgraduate School Molecular Medicine, Rotterdam, The Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center and Postgraduate School Molecular Medicine, Rotterdam, The Netherlands
| |
Collapse
|
140
|
Tsukamoto H, Fujieda K, Senju S, Ikeda T, Oshiumi H, Nishimura Y. Immune-suppressive effects of interleukin-6 on T-cell-mediated anti-tumor immunity. Cancer Sci 2017; 109:523-530. [PMID: 29090850 PMCID: PMC5834784 DOI: 10.1111/cas.13433] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/12/2022] Open
Abstract
Accompanied by the growing clinical applications of immunotherapy in the treatment of cancer patients, development of novel therapeutic approaches to reverse the immune-suppressive environment in cancer patients is eagerly anticipated, because the success of cancer immunotherapy is currently limited by immune-suppressive effects in tumor-bearing hosts. Interleukin (IL)-6, a pleotropic proinflammatory cytokine, participates in tumor cell-autonomous processes that are required for their survival and growth, and is therefore known as a poor prognostic factor in cancer patients. In addition, an emerging role of IL-6 in modulating multiple functions of immune cells including T cells, dendritic cells, and macrophages is responsible for the dysfunction of innate and adaptive immunity against tumors. Therefore, the IL-6-targeting approach is of value as a promising strategy for desensitization and prevention of immune-suppressive effects, and should be an effective treatment when combined with current immunotherapies. The aim of the present review is to discuss the immune-suppressive aspects of IL-6, notably with modification of T-cell functions in cancer patients, and their relationship to anti-tumor immune responses and cancer immunotherapy.
Collapse
Affiliation(s)
- Hirotake Tsukamoto
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koji Fujieda
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tokunori Ikeda
- Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Nishimura Project Laboratory, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
141
|
Tsukamoto H, Fujieda K, Senju S, Ikeda T, Oshiumi H, Nishimura Y. Immune-suppressive effects of interleukin-6 on T-cell-mediated anti-tumor immunity. Cancer Sci 2017. [PMID: 29090850 DOI: 10.1111/cas.13433.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Accompanied by the growing clinical applications of immunotherapy in the treatment of cancer patients, development of novel therapeutic approaches to reverse the immune-suppressive environment in cancer patients is eagerly anticipated, because the success of cancer immunotherapy is currently limited by immune-suppressive effects in tumor-bearing hosts. Interleukin (IL)-6, a pleotropic proinflammatory cytokine, participates in tumor cell-autonomous processes that are required for their survival and growth, and is therefore known as a poor prognostic factor in cancer patients. In addition, an emerging role of IL-6 in modulating multiple functions of immune cells including T cells, dendritic cells, and macrophages is responsible for the dysfunction of innate and adaptive immunity against tumors. Therefore, the IL-6-targeting approach is of value as a promising strategy for desensitization and prevention of immune-suppressive effects, and should be an effective treatment when combined with current immunotherapies. The aim of the present review is to discuss the immune-suppressive aspects of IL-6, notably with modification of T-cell functions in cancer patients, and their relationship to anti-tumor immune responses and cancer immunotherapy.
Collapse
Affiliation(s)
- Hirotake Tsukamoto
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koji Fujieda
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoru Senju
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tokunori Ikeda
- Department of Clinical Investigation, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasuharu Nishimura
- Department of Immunogenetics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.,Nishimura Project Laboratory, Institute of Resource Development and Analysis, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
142
|
Linette GP, Carreno BM. Neoantigen Vaccines Pass the Immunogenicity Test. Trends Mol Med 2017; 23:869-871. [PMID: 28867556 DOI: 10.1016/j.molmed.2017.08.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 08/16/2017] [Indexed: 12/30/2022]
Abstract
Neoantigens arising from tumor-specific genomic alterations constitute authentic non-self antigens and represent a new class of targets for cancer immunotherapy. Recent reports on various vaccine platforms targeting neoantigens suggest a basis for precision therapies customized to each patient's tumor mutational profile.
Collapse
Affiliation(s)
- Gerald P Linette
- Department of Hematology/Oncology, Parker Institute for Cancer Immunotherapy and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5157, USA.
| | - Beatriz M Carreno
- Department of Pathology and Laboratory Medicine, Parker Institute for Cancer Immunotherapy and Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-5157, USA
| |
Collapse
|