151
|
Pellegrino M, Del Bufalo F, De Angelis B, Quintarelli C, Caruana I, de Billy E. Manipulating the Metabolism to Improve the Efficacy of CAR T-Cell Immunotherapy. Cells 2020; 10:cells10010014. [PMID: 33374128 PMCID: PMC7824126 DOI: 10.3390/cells10010014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 12/23/2020] [Indexed: 12/19/2022] Open
Abstract
The adoptive transfer of the chimeric antigen receptor (CAR) expressing T-cells has produced unprecedented successful results in the treatment of B-cell malignancies. However, the use of this technology in other malignancies remains less effective. In the setting of solid neoplasms, CAR T-cell metabolic fitness needs to be optimal to reach the tumor and execute their cytolytic function in an environment often hostile. It is now well established that both tumor and T cell metabolisms play critical roles in controlling the immune response by conditioning the tumor microenvironment and the fate and activity of the T cells. In this review, after a brief description of the tumoral and T cell metabolic reprogramming, we summarize the latest advances and new strategies that have been developed to improve the metabolic fitness and efficacy of CAR T-cell products.
Collapse
Affiliation(s)
- Marsha Pellegrino
- Department of Onco-hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital–IRCCS, 00146 Rome, Italy; (M.P.); (F.D.B.); (B.D.A.); (C.Q.); (I.C.)
| | - Francesca Del Bufalo
- Department of Onco-hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital–IRCCS, 00146 Rome, Italy; (M.P.); (F.D.B.); (B.D.A.); (C.Q.); (I.C.)
| | - Biagio De Angelis
- Department of Onco-hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital–IRCCS, 00146 Rome, Italy; (M.P.); (F.D.B.); (B.D.A.); (C.Q.); (I.C.)
| | - Concetta Quintarelli
- Department of Onco-hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital–IRCCS, 00146 Rome, Italy; (M.P.); (F.D.B.); (B.D.A.); (C.Q.); (I.C.)
- Department of Clinical Medicine and Surgery, Federico II University of Naples, 81100 Naples, Italy
| | - Ignazio Caruana
- Department of Onco-hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital–IRCCS, 00146 Rome, Italy; (M.P.); (F.D.B.); (B.D.A.); (C.Q.); (I.C.)
- Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University Children’s Hospital of Würzburg, 97080 Würzburg, Germany
| | - Emmanuel de Billy
- Department of Onco-hematology, Cell and Gene Therapy, Bambino Gesù Children’s Hospital–IRCCS, 00146 Rome, Italy; (M.P.); (F.D.B.); (B.D.A.); (C.Q.); (I.C.)
- Correspondence: ; Tel.: +39-06-6859-3516
| |
Collapse
|
152
|
Ai L, Wang H. Effects of propofol and sevoflurane on tumor killing activity of peripheral blood natural killer cells in patients with gastric cancer. J Int Med Res 2020; 48:300060520904861. [PMID: 32216484 PMCID: PMC7133410 DOI: 10.1177/0300060520904861] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective This study aimed to investigate the effects of propofol and sevoflurane on cytotoxicity of natural killer (NK) cells in patients with gastric cancer. Methods Patients with gastric cancer were anesthetized by propofol or sevoflurane. Peripheral blood NK cells were isolated and co-cultured with BGC-823 gastric cancer cell culture supernatant, and the rate of apoptosis and effector molecules were analyzed by flow cytometry. Effects of propofol and sevoflurane on NK cell function and SMAD4 protein expression were investigated. Results Cytotoxicity of NK cells in patients with gastric cancer was inhibited before surgery, but it was enhanced in patients who were anesthetized by propofol compared with those who had sevoflurane. In vitro co-culture with BGC-823 cells significantly inhibited the cytotoxicity of NK cells, which was abolished by treatment of propofol or transforming growth factor (TGF)-β1. SMAD4 protein expression in the NK cell nucleus was significantly downregulated by TGF-β1 treatment and BGC-823 supernatant co-culture, and this expression could be restored by propofol. Conclusions Cytotoxicity of NK cells in patients with gastric cancer is low, but it can be promoted by propofol. Propofol regulates cytotoxicity in NK cells by promoting SMAD4, thereby affecting cellular function.
Collapse
Affiliation(s)
- Lili Ai
- Department of Anesthesiology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China.,Department of Anesthesiology, the Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Hao Wang
- Department of Anesthesiology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
153
|
Liu X, Du Q, Tian C, Tang M, Jiang Y, Wang Y, Cao Y, Wang Z, Wang Z, Yang J, Li Y, Jiao X, Xie P. Discovery of CAPE derivatives as dual EGFR and CSK inhibitors with anticancer activity in a murine model of hepatocellular carcinoma. Bioorg Chem 2020; 107:104536. [PMID: 33342565 DOI: 10.1016/j.bioorg.2020.104536] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 12/17/2022]
Abstract
Caffeic acid phenethyl ester (CAPE), a bioactive component extracted from propolis of honeybee hives, can inhibit hepatocellular carcinoma (HCC). In order to explore more stable CAPE derivatives, 25 compounds were designed, synthesized, and pharmacologically assessed in vitro and in vivo as anti-tumor agents in HCC. Compounds 8d, 8f, 8l, 8j, and 8k showed favorable antiproliferative activity than other compounds including CAPE in the HCC cell lines. Based on the result of QTRP (Quantitative Thiol Reactivity Profiling), epidermal growth factor receptor (EGFR) and C-terminal Src kinase (CSK) were supposed to the targets of 8f, which was confirmed by binding mode analysis. Furthermore, compounds 8f, 8l, 8j, 8k, 8g, and 8h showed potent inhibitory effects against both CSK and EGFR than other derivatives in an ADP-Glo™ kinase assay. The representative compound, 8f, potently inhibited various tumor growth in murine model including murine hepatocellular carcinoma H22, meanwhile downregulating the EGFR/AKT pathway and enhancing T cell proliferation through inhibition of CSK. Metabolic stability in vitro suggested 8f and 8k were more stable in mouse plasma than CAPE and susceptible to metabolism in liver microsomes. The overall excellent profile of compound 8f makes it a potential candidate for further preclinical investigation.
Collapse
Affiliation(s)
- Xiaoyu Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qianqian Du
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Caiping Tian
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Beijing Institute of Lifeomics, Beijing 102206, China; School of Medicine, Tsinghua University, Beijing, China
| | - Mei Tang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yingjun Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yong Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yang Cao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhe Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhenwei Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jing Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences Beijing, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Yan Li
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Xiaozhen Jiao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ping Xie
- State Key Laboratory of Bioactive Substance and Function of Natural Medicine, Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
154
|
Murakami T, Takasawa A, Takasawa K, Akimoto T, Aoyama T, Magara K, Saito Y, Ota M, Kyuno D, Yamamoto S, Hasegawa T, Saito T, Osanai M. Aberrant expression of junctional adhesion molecule-A contributes to the malignancy of cervical adenocarcinoma by interaction with poliovirus receptor/CD155. Cancer Sci 2020; 112:906-917. [PMID: 33185939 PMCID: PMC7893988 DOI: 10.1111/cas.14734] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022] Open
Abstract
Recent studies have shown that aberrant expression of tight junction proteins (TJP) contributes to malignant potential of various cancers. In the present study, we investigated the expression of junctional adhesion molecule-A (JAM-A), one of the transmembrane TJP, in uterine cervical adenocarcinoma and the significance of its expression for malignancy. Immunohistochemistry on human surgical specimens showed that JAM-A was aberrantly expressed in neoplastic regions including adenocarcinoma in situ (AIS). Knockout of JAM-A significantly suppressed cell proliferation and colony-forming and migration abilities. We also showed that an antibody specific to an extracellular region of JAM-A reduced cell proliferation ability and that loss of JAM-A increased drug sensitivity of cervical adenocarcinoma cells. Based on a comprehensive proteome analysis, we found that poliovirus receptor (PVR/CD155) was regulated by JAM-A and formed a physical interaction with JAM-A. In human surgical specimens, PVR/CD155 expression was significantly correlated with some clinicopathological features and prognosis of cervical adenocarcinoma. Interestingly, most of the PVR/CD155-positive cases expressed a high level of JAM-A, and patients with the expression pattern of PVR/CD155 positive/JAM-A high had significantly shorter periods of relapse-free survival (P = .00964) and overall survival (P = .0204) than those for the other patients. Our observations suggest that aberrant expression of JAM-A promotes malignancy of uterine cervical adenocarcinoma by regulation of PVR/CD155, and JAM-A is therefore a potential therapeutic target for this malignancy.
Collapse
Affiliation(s)
- Taro Murakami
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Akira Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kumi Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Taishi Akimoto
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tomoyuki Aoyama
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kazufumi Magara
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuki Saito
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Misaki Ota
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Daisuke Kyuno
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Surgery, Surgical Oncology and Science, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Soh Yamamoto
- Department of Microbiology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tadashi Hasegawa
- Department of Surgical Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Makoto Osanai
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
155
|
Ding QQ, Chauvin JM, Zarour HM. Targeting novel inhibitory receptors in cancer immunotherapy. Semin Immunol 2020; 49:101436. [PMID: 33288379 DOI: 10.1016/j.smim.2020.101436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022]
Abstract
T cells play a critical role in promoting tumor regression in both experimental models and humans. Yet, T cells that are chronically exposed to tumor antigen during cancer progression can become dysfunctional/exhausted and fail to induce tumor destruction. Such tumor-induced T cell dysfunction may occur via multiple mechanisms. In particular, immune checkpoint inhibitory receptors that are upregulated by tumor-infiltrating lymphocytes in many cancers limit T cell survival and function. Overcoming this inhibitory receptor-mediated T cell dysfunction has been a central focus of recent developments in cancer immunotherapy. Immunotherapies targeting inhibitory receptor pathways such as programmed cell death 1 (PD-1)/programmed death ligand 1 and cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), alone or in combination, confer significant clinical benefits in multiple tumor types. However, many patients with cancer do not respond to immune checkpoint blockade, and dual PD-1/CTLA-4 blockade may cause serious adverse events, which limits its indications. Targeting novel non-redundant inhibitory receptor pathways contributing to tumor-induced T cell dysfunction in the tumor microenvironment may prove efficacious and non-toxic. This review presents preclinical and clinical findings supporting the roles of two key pathways-T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and T cell immunoreceptor with Ig and ITIM domain (TIGIT)/CD226/CD96/CD112R-in cancer immunotherapy.
Collapse
Affiliation(s)
- Quan-Quan Ding
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Joe-Marc Chauvin
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Hassane M Zarour
- Department of Medicine and Division of Hematology/Oncology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
156
|
Wang Y, Jin B, Zhou N, Sun Z, Li J, Chen Q, Wu X, Zhou Y, Shi Y, Lu X, Sang X, Mao Y, Du S, Wang W, Bai C. Identification of WDFY3 Neoantigens as Prognostic Markers in Longterm Survivors of Extrahepatic Cholangiocarcinoma. Curr Cancer Drug Targets 2020; 20:875-886. [PMID: 32957886 DOI: 10.2174/1568009620999200918121456] [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: 04/17/2020] [Revised: 07/18/2020] [Accepted: 07/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Neoantigens are newly formed antigens that have not been previously recognized by the immune system. They may arise from altered tumor proteins that form as a result of mutations. Although neoantigens have recently been linked to antitumor immunity in long-term survivors of cancers, such as melanoma and colorectal cancer, their prognostic and immune-modulatory role in many cancer types remains undefined. OBJECTIVE The purpose of this study is to identify prognostic markers for long-term extrahepatic cholangiocarcinoma (EHCC) survival. METHODS We investigated neoantigens in EHCC, a rare, aggressive cancer with a 5-year overall survival rate lower than 10%, using a combination of whole-exome sequencing (WES), RNA sequencing (RNA-seq), computational biophysics, and immunohistochemistry. RESULTS Our analysis revealed a decreased neutrophil infiltration-related trend of high-quality neoantigen load with IC50 <500 nM (r=-0.445, P=0.043). Among 24 EHCC patients examined, we identified four long-term survivors with WDFY3 neoantigens and none with WDFY3 neoantigens in the short-term survivors. The WDFY3 neoantigens are associated with a lower infiltration of neutrophils (p=0.013), lower expression of CCL5 (p=0.025), CXCL9 (p=0.036) and TIGIT (p=0.016), and less favorable prognosis (p=0.030). In contrast, the prognosis was not significantly associated with tumor mutation burden, neoantigen load, or immune cell infiltration. CONCLUSION We suggest that the WDFY3 neoantigens may affect prognosis by regulating antitumor immunity and that the WDFY3 neoantigens may be harnessed as potential targets for immunotherapy of EHCC.
Collapse
Affiliation(s)
- Yingyi Wang
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing, China
| | - Bao Jin
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Na Zhou
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing, China
| | - Zhao Sun
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing, China
| | - Jiayi Li
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Qiao Chen
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiangan Wu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yi Zhou
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yue Shi
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xin Lu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xinting Sang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yilei Mao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wenze Wang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences
and Peking Union Medical College, Beijing, China
| |
Collapse
|
157
|
Han HS, Jeong S, Kim H, Kim HD, Kim AR, Kwon M, Park SH, Woo CG, Kim HK, Lee KH, Seo SP, Kang HW, Kim WT, Kim WJ, Yun SJ, Shin EC. TOX-expressing terminally exhausted tumor-infiltrating CD8 + T cells are reinvigorated by co-blockade of PD-1 and TIGIT in bladder cancer. Cancer Lett 2020; 499:137-147. [PMID: 33249194 DOI: 10.1016/j.canlet.2020.11.035] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/28/2020] [Accepted: 11/23/2020] [Indexed: 12/25/2022]
Abstract
Exhausted T cells in the tumor microenvironment are major targets of immunotherapies. However, the exhaustion status of CD8+ tumor-infiltrating lymphocytes (TILs) in bladder cancer has not been comprehensively evaluated. Herein, we examined distinct exhaustion status of CD8+ TILs based on the level of programmed cell death-1 (PD-1) and thymocyte selection-associated high mobility group box protein (TOX) expression in urothelial bladder cancer. We also evaluated the reinvigoration of exhausted CD8+ TILs upon ex vivo treatment with inhibitory checkpoint blockers. TOX-expressing PD-1highCD8+ TILs had the highest expression of immune checkpoint receptors (ICRs), the most terminally exhausted features, and the highest tumor antigen reactivity among PD-1+CD8+ TILs. Bladder cancer patients with a high percentage of PD-1highTOX+CD8+ TILs had more progressed T-cell exhaustion features and higher programmed death-ligand 1 expression in tumor tissues. TIGIT was the most frequent co-expressed ICR on PD-1+CD8+ TILs, and TIGIT blockade enhanced the PD-1 blockade-mediated cytokine production by CD8+ TILs from bladder cancer patients. Our findings provide an improved understanding of the heterogeneous exhaustion status of CD8+ TILs and additional immunotherapy strategies to improve outcomes of bladder cancer patients.
Collapse
Affiliation(s)
- Hye Sook Han
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Republic of Korea; Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Seongju Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Hyunglae Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Hyung-Don Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - A Reum Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Minsuk Kwon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Chang Gok Woo
- Department of Pathology, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Hee Kyung Kim
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Ki Hyeong Lee
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju, Republic of Korea; Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Sung Pil Seo
- Department of Urology, Chungbuk National University Hospital, Cheongju, Republic of Korea
| | - Ho Won Kang
- Department of Urology, Chungbuk National University Hospital, Cheongju, Republic of Korea; Department of Urology, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Won Tae Kim
- Department of Urology, Chungbuk National University Hospital, Cheongju, Republic of Korea; Department of Urology, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Wun-Jae Kim
- Department of Urology, Chungbuk National University Hospital, Cheongju, Republic of Korea; Department of Urology, Chungbuk National University College of Medicine, Cheongju, Republic of Korea
| | - Seok Joong Yun
- Department of Urology, Chungbuk National University Hospital, Cheongju, Republic of Korea; Department of Urology, Chungbuk National University College of Medicine, Cheongju, Republic of Korea.
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.
| |
Collapse
|
158
|
Shi F, Su J, Wang J, Liu Z, Wang T. Activation of STING inhibits cervical cancer tumor growth through enhancing the anti-tumor immune response. Mol Cell Biochem 2020; 476:1015-1024. [PMID: 33141310 DOI: 10.1007/s11010-020-03967-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/27/2020] [Indexed: 01/07/2023]
Abstract
Cervical cancer remains the second leading cause of gynecologic cancer-related mortality among women worldwide. STING (stimulator of interferon genes) was reported to be involved in the immune surveillance of tumors. However, the specific role of STING in cervical cancer remains unclear. In this study, we found that the cGAS (Cyclic GMP-AMP synthase)/STING signal decreased in cervical cancer cells. Knockdown of STING by siRNA enhanced the cell viability and migration of cervical cancer cells, while activation of STING by ADU-S100 inhibited the cell viability of cervical cancer cells, with no effect on the migration and apoptosis. In addition, ADU-S100 promoted the secretion of IFNβ and IL-6, and the activation of TBK1 (TANK-binding kinase 1)/NF-κB (nuclear factor kappa-B) pathway. Meanwhile, knockdown of STING inhibited the production of IFNβ and IL-6 that were triggered by dsDNA and suppressed the TBK1/NF-κB signaling. ADU-S100 also suppressed tumor growth in vivo and increased the tumor-infiltrating CD8+ T cell and CD103+ dendritic cell numbers. The NF-κB signal inhibitor limited the increasing numbers of CD8+ T cell and CD103+ dendritic cells induced by ADU-S100, without influence on tumor growth. Hence, our study suggested that STING could serve as a potential novel immunotherapeutic target for cervical cancer.
Collapse
Affiliation(s)
- Fan Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, No.277 West yanta road, Xi'an, 710061, China
| | - Jin Su
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, No.277 West yanta road, Xi'an, 710061, China
| | - Juan Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, No.277 West yanta road, Xi'an, 710061, China
| | - Zi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, No.277 West yanta road, Xi'an, 710061, China
| | - Tao Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, No.277 West yanta road, Xi'an, 710061, China.
| |
Collapse
|
159
|
Abstract
PURPOSE OF REVIEW Immunotherapy has shown an unprecedented response in treatment of tumors. However, challenges such as lack of cytotoxic lymphocytes to mount an immune response or development of resistance to therapy can limit efficacy. Here, we discuss alternative checkpoints that can be targeted to improve cytotoxic lymphocyte function while harnessing other components of the immune system. RECENT FINDINGS Blockade of alternative checkpoints has improved anti-tumor immunity in mouse models and is being tested clinically with encouraging findings. In addition to modulating T cell function directly, alternative checkpoints can also regulate activity of myeloid cells and regulatory T cells to affect anti-tumor response. Combination of immune checkpoint inhibitors can improve treatment of tumors by activating multiple arms of the immune system.
Collapse
Affiliation(s)
- Ayush Pant
- Department of Neurosurgery, Neurosurgery Oncology, Radiation Oncology, Otolaryngology, and Institute of NanoBiotechnology, Brain Tumor Immunotherapy Program, Metastatic Brain Tumor Center, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Phipps 123, Baltimore, MD, 21287, USA
| | - Ravi Medikonda
- Department of Neurosurgery, Neurosurgery Oncology, Radiation Oncology, Otolaryngology, and Institute of NanoBiotechnology, Brain Tumor Immunotherapy Program, Metastatic Brain Tumor Center, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Phipps 123, Baltimore, MD, 21287, USA
| | - Michael Lim
- Department of Neurosurgery, Neurosurgery Oncology, Radiation Oncology, Otolaryngology, and Institute of NanoBiotechnology, Brain Tumor Immunotherapy Program, Metastatic Brain Tumor Center, The Johns Hopkins University School of Medicine, 600 N. Wolfe Street, Phipps 123, Baltimore, MD, 21287, USA.
| |
Collapse
|
160
|
Liu X, Li Q, Zhou Y, He X, Fang J, Lu H, Wang X, Wang D, Ma D, Cheng B, Liao G, Wang Z. Dysfunctional role of elevated TIGIT expression on T cells in oral squamous cell carcinoma patients. Oral Dis 2020; 27:1667-1677. [PMID: 33125794 DOI: 10.1111/odi.13703] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 10/06/2020] [Accepted: 10/24/2020] [Indexed: 01/13/2023]
Abstract
OBJECTIVE This study was aimed to analyze the role of T-cell immunoreceptor with immunoglobulin and tyrosine-based inhibitory motif domains (TIGIT) expression on T cells in patients with oral squamous cell carcinoma (OSCC). MATERIALS AND METHODS Peripheral blood mononuclear cells (PBMC) and tumor-infiltrating lymphocytes (TILs) were collected from OSCC patients. The correlation between TIGIT expression and clinicopathologic features was analyzed by chi-square test. Phenotypic and functional study of TIGIT+ T cells were performed by flow cytometry. RESULTS TIGIT was highly expressed on T cells from PBMC and TILs. High expression of TIGIT on CD4+ T cells (19.0%) and CD8+ T cells (35.9%) was also associated with higher T stage and nodal invasion. Moreover, TIGIT+ CD4+ and TIGIT+ CD8+ T cells sorted from OSCC patients showed a dysfunctional phenotype (low cell proliferation and low secretion of IL-2, TNF-α and IFN-γ), and TIGIT+ CD4+ T cells exhibited inhibitory function (high expression of Foxp3 and high amounts of IL-10). Importantly, TIGIT blockade can enhance the proliferation ability and effective cytokine production (IL-2, TNF-α, and IFN-γ) of CD4+ and CD8+ T cells from OSCC patients in vitro. CONCLUSIONS TIGIT-expressing T cells exhibit a lower effector cytokine-releasing phenotype in OSCC patients.
Collapse
Affiliation(s)
- Xiangqi Liu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Qunxing Li
- Department of Oral Medicine, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ying Zhou
- Department of Oral Medicine, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xinlin He
- Department of Oral Medicine, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Juan Fang
- Department of Oral Medicine, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Huanzi Lu
- Department of Oral Medicine, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xi Wang
- Department of Oral Medicine, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Dikan Wang
- Department of Oral Medicine, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Da Ma
- Department of Prosthodontics, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Bin Cheng
- Department of Oral Medicine, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Guiqing Liao
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhi Wang
- Department of Oral Medicine, Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
161
|
Cao Y, Wang X, Jin T, Tian Y, Dai C, Widarma C, Song R, Xu F. Immune checkpoint molecules in natural killer cells as potential targets for cancer immunotherapy. Signal Transduct Target Ther 2020; 5:250. [PMID: 33122640 PMCID: PMC7596531 DOI: 10.1038/s41392-020-00348-8] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/13/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
Recent studies have demonstrated the potential of natural killer (NK) cells in immunotherapy to treat multiple types of cancer. NK cells are innate lymphoid cells that play essential roles in tumor surveillance and control that efficiently kill the tumor and do not require the major histocompatibility complex. The discovery of the NK's potential as a promising therapeutic target for cancer is a relief to oncologists as they face the challenge of increased chemo-resistant cancers. NK cells show great potential against solid and hematologic tumors and have progressively shown promise as a therapeutic target for cancer immunotherapy. The effector role of these cells is reliant on the balance of inhibitory and activating signals. Understanding the role of various immune checkpoint molecules in the exhaustion and impairment of NK cells when their inhibitory receptors are excessively expressed is particularly important in cancer immunotherapy studies and clinical implementation. Emerging immune checkpoint receptors and molecules have been found to mediate NK cell dysfunction in the tumor microenvironment; this has brought up the need to explore further additional NK cell-related immune checkpoints that may be exploited to enhance the immune response to refractory cancers. Accordingly, this review will focus on the recent findings concerning the roles of immune checkpoint molecules and receptors in the regulation of NK cell function, as well as their potential application in tumor immunotherapy.
Collapse
Affiliation(s)
- Yuqing Cao
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China
| | - Xiaoyu Wang
- College of Life and Health Science, Northeastern University, 110819, Shenyang, China
| | - Tianqiang Jin
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China
| | - Chaoliu Dai
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China
| | - Crystal Widarma
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| | - Feng Xu
- Department of General Surgery, Shengjing Hospital of China Medical University, 110004, Shenyang, China.
| |
Collapse
|
162
|
Wu F, Cheng Y, Wu L, Zhang W, Zheng W, Wang Q, Cao H, Pan X, Tang W. Emerging Landscapes of Tumor Immunity and Metabolism. Front Oncol 2020; 10:575037. [PMID: 33117713 PMCID: PMC7575711 DOI: 10.3389/fonc.2020.575037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
The metabolic reprogramming of cancer tissue has higher metabolic activity than surrounding tissues. At the same time, the local infiltration of immunosuppressive cells is also significantly increased, resulting in a significant decrease in tumor immunity. During the progression of cancer cells, immunosuppressive tumor microenvironment is formed around the tumor due to their metabolic reprogramming. In addition, it is the changes in metabolic patterns that make tumor cells resistant to certain drugs, impeding cancer treatment. This article reviews the mechanisms of immune escape caused by metabolic reprogramming, and aims to provide new ideas for clinical tumor immunotherapy combined with metabolic intervention for tumor treatment.
Collapse
Affiliation(s)
- Fan Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ye Cheng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liangliang Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Wenling Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wubing Zheng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qian Wang
- Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Hongyong Cao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Xiongxiong Pan
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weiwei Tang
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
163
|
Immunological Gene Signature Associated With the Tumor Microenvironment of Pancreatic Cancer After Neoadjuvant Chemotherapy. Pancreas 2020; 49:1240-1245. [PMID: 32898010 DOI: 10.1097/mpa.0000000000001665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Neoadjuvant chemotherapy (NAC) has improved overall survival in patients with pancreatic ductal adenocarcinoma (PDAC), but its effects on immune gene signatures are unknown. Here, we examined the immune transcriptome after NAC for PDAC. METHODS Resected tumor specimens were obtained from 140 patients with PDAC who received surgery first (n = 93) or NAC (n = 47). Six patients were randomly selected from each group, and RNA was extracted from tumor tissues. We compared 770 immune-related genes among the 2 groups using nCounterPanCancer Immune Profiling (NanoString Technologies, Seattle, Wash). Gene clusters were classified into 14 immune function groups based on gene ontology argolism by nSolver 4.0 software (NanoString Technologies), and corresponding immune cell function scores were compared. RESULTS Eleven genes (LY86, SH2D1A, CD247, TIGIT, CR2, CD83, LAMP3, CXCR4, DUSP4, SELL, and IL2RA) were significantly downregulated in the NAC group. Gene expression analysis showed that the functions of regulatory T cells, B cells, and natural killer CD56 dim cells were significantly decreased in the NAC group. CONCLUSIONS Neoadjuvant chemotherapy may suppress regulatory T cells and B-cell function in the PDAC microenvironment. The 11 identified genes could be useful for predicting the efficacy of NAC and could be therapeutic targets for PDAC.
Collapse
|
164
|
The impaired anti-tumoral effect of immune surveillance cells in the immune microenvironment of gastric cancer. Clin Immunol 2020; 219:108551. [DOI: 10.1016/j.clim.2020.108551] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/07/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022]
|
165
|
Zhou X, Du J, Wang H, Chen C, Jiao L, Cheng X, Zhou X, Chen S, Gou S, Zhao W, Zhai W, Chen J, Gao Y. Repositioning liothyronine for cancer immunotherapy by blocking the interaction of immune checkpoint TIGIT/PVR. Cell Commun Signal 2020; 18:142. [PMID: 32894141 PMCID: PMC7487564 DOI: 10.1186/s12964-020-00638-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
Background Inhibitors targeting immune checkpoint were proved effective in cancer immunotherapy, such as PD-1/PD-L1 blockade. The novel immune checkpoint TIGIT/PVR plays critical roles in suppressing the anti-tumor effects of CD8+ T and NK cells, and dual blockade of TIGIT/PVR and PD-1/PD-L1 by antibody can elicit synergistic effects in tumor models and clinical trials. However, small molecules for TIGIT/PVR blockade have not been investigated. Methods The expression of PVR in tumors were analyzed by using TCGA, Oncomine and GEO database, and in cancer cell lines examined by flow cytometry. Natural product compounds were docked to PVR for virtual screening by using the software Molecular Operating Environment (MOE). Candidate compounds were further tested by biolayer interferometry-based binding assay, microscale thermophoresis assay and cell based blocking assay. The in vitro activity of the candidate compound was determined by MTT, peripheral blood mononuclear cells (PBMCs) activation assay and coculture assay. The anti-tumor effects and mechanism were also investigated by using MC38 tumor-bearing mice model and immune cell depletion tumor model. Results PVR was over-expressed in many tumor tissues and cancer cell lines, making it a promising therapeutic target. Through virtual screening, binding, and blocking assay, liothyronine was discovered to bind PVR and block the interaction of TIGIT/PVR. Liothyronine could enhance the function of CD4+ and CD8+ T cells in PBMCs. Besides, in the Jurkat-hTIGIT and CHOK1-hPVR coculture assay, liothyronine could reverse the IL-2 secretion inhibition resulted by TIGIT/PVR ligation. Although had no influence on the proliferation of tumor cells in vitro, liothyronine could significantly inhibit tumor growth when administrated in vivo, by enhancing CD8+ T cell infiltration and immune responses in the tumor bearing mice. The immune cell depletion model showed that the anti-tumor effects of liothyronine depends on CD4+ T cells, CD8+ T cells and NK cells. Conclusions A small molecule liothyronine was discovered to serve as a potential candidate for cancer immunotherapy by blocking the immune checkpoint TIGIT/PVR. Video abstract
Graphical abstract ![]()
Collapse
Affiliation(s)
- Xiuman Zhou
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518035, China
| | - Jiangfeng Du
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hongfei Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chunxia Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ling Jiao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiangrui Cheng
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaowen Zhou
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Shaomeng Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Shanshan Gou
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenshan Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wenjie Zhai
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Junhui Chen
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518035, China.
| | - Yanfeng Gao
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518035, China. .,School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
166
|
Abstract
Tumors evade immune-mediated recognition through multiple mechanisms of immune escape. On chronic tumor antigen exposure, T cells become dysfunctional/exhausted and upregulate various checkpoint inhibitory receptors (IRs) that limit T cells' survival and function. During the last decade, immunotherapies targeting IRs such as programmed cell death receptor 1 (PD-1) and anticytotoxic T lymphocyte-associated antigen 4 (CTLA-4) have provided ample evidence of clinical benefits in many solid tumors. Beyond CTLA-4 and PD-1, multiple other IRs are also targeted with immune checkpoint blockade in the clinic. Specifically, T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) is a promising new target for cancer immunotherapy. TIGIT is upregulated by immune cells, including activated T cells, natural killer cells, and regulatory T cells. TIGIT binds to two ligands, CD155 (PVR) and CD112 (PVRL2, nectin-2), that are expressed by tumor cells and antigen-presenting cells in the tumor microenvironment. There is now ample evidence that the TIGIT pathway regulates T cell-mediated and natural killer cell-mediated tumor recognition in vivo and in vitro. Dual PD-1/TIGIT blockade potently increases tumor antigen-specific CD8+ T cell expansion and function in vitro and promotes tumor rejection in mouse tumor models. These findings support development of ongoing clinical trials with dual PD-1/TIGIT blockade in patients with cancer.
Collapse
Affiliation(s)
- Joe-Marc Chauvin
- Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hassane M Zarour
- Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
167
|
Abstract
Through the successes of checkpoint blockade and adoptive cellular therapy, immunotherapy has become an established treatment modality for cancer. Cellular metabolism has emerged as a critical determinant of the viability and function of both cancer cells and immune cells. In order to sustain prodigious anabolic needs, tumours employ a specialized metabolism that differs from untransformed somatic cells. This metabolism leads to a tumour microenvironment that is commonly acidic, hypoxic and/or depleted of critical nutrients required by immune cells. In this context, tumour metabolism itself is a checkpoint that can limit immune-mediated tumour destruction. Because our understanding of immune cell metabolism and cancer metabolism has grown significantly in the past decade, we are on the cusp of being able to unravel the interaction of cancer cell metabolism and immune metabolism in therapeutically meaningful ways. Although there are metabolic processes that are seemingly fundamental to both cancer and responding immune cells, metabolic heterogeneity and plasticity may serve to distinguish the two. As such, understanding the differential metabolic requirements of the diverse cells that comprise an immune response to cancer offers an opportunity to selectively regulate immune cell function. Such a nuanced evaluation of cancer and immune metabolism can uncover metabolic vulnerabilities and therapeutic windows upon which to intervene for enhanced immunotherapy.
Collapse
Affiliation(s)
- Robert D Leone
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jonathan D Powell
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Research Center, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
168
|
Li Y, He X, Fan L, Zhang X, Xu Y, Xu X. Identification of a novel immune prognostic model in gastric cancer. Clin Transl Oncol 2020; 23:846-855. [PMID: 32857339 DOI: 10.1007/s12094-020-02478-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 08/10/2020] [Indexed: 12/24/2022]
Abstract
PURPOSE The tumor immune microenvironment (TIME) is now considered as an important factor during gastric cancer (GC) development. This study identified a novel immune-related risk model for predicting prognosis and assessing the immune status of GC patients. METHODS Transcriptomic data were obtained from the TCGA database. The differential expressed immune-related genes (IRGs) were identified through the ImmPort portal. Enrichment analysis was performed to explore the potential molecular mechanism of these IRGs. By the Cox regression analysis, we constructed the immune prognostic model. Then, the association between the model and the immune microenvironment was estimated. The model was validated in the GSE84433 dataset. RESULTS Totally, we identified 222 differentially expressed IRGs. These IRGs were closely correlated with immune response and immune signaling pathways. Through the Cox regression analysis, we developed the immune prognostic model based on the expression of seven IRGs (CXCL3, NOX4, PROC, FAM19A4, RNASE2, IGHD2-15, CGB5). Patients were stratified into two groups according to immune-related risk scores. Survival analysis indicated that the prognosis of high-risk patients was poorer than low-risk patients. Moreover, the immune-related risk score was an independent prognostic biomarker. More importantly, we found that the infiltration level of immunosuppressive cells and the expression of inhibitory immune checkpoints were higher in high-risk patients. The immune microenvironment tended to be a suppressive status in patients with high-risk scores. CONCLUSION This study demonstrated that our model had predictive value for prognosis and the TIME in GC. It might be a robust tool to improve personalized patient management.
Collapse
Affiliation(s)
- Y Li
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - X He
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - L Fan
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - X Zhang
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - Y Xu
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China
| | - X Xu
- Cancer Center, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
169
|
Hossain MA, Liu G, Dai B, Si Y, Yang Q, Wazir J, Birnbaumer L, Yang Y. Reinvigorating exhausted CD8 + cytotoxic T lymphocytes in the tumor microenvironment and current strategies in cancer immunotherapy. Med Res Rev 2020; 41:156-201. [PMID: 32844499 DOI: 10.1002/med.21727] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/26/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy has revolutionized the treatment of cancer in recent years and achieved overall success and long-term clinical benefit in patients with a wide variety of cancer types. However, there is still a large proportion of patients exhibiting limited or no responses to immunotherapeutic strategy, some of which were even observed with hyperprogressive disease. One major obstacle restricting the efficacy is that tumor-reactive CD8+ T cells, which are central for tumor control, undergo exhaustion, and lose their ability to eliminate cancer cells after infiltrating into the strongly immunosuppressive tumor microenvironment. Thus, as a potential therapeutic rationale in the development of cancer immunotherapy, targeting or reinvigorating exhausted CD8+ T cells has been attracting much interest. Hitherto, both intrinsic and extrinsic mechanisms that govern CD8+ T-cell exhaustion have been explored. Specifically, the transcriptional and epigenetic landscapes have been depicted utilizing single-cell RNA sequencing or mass cytometry (CyTOF). In addition, cellular metabolism dictating the tumor-infiltrating CD8+ T-cell fate is currently under investigation. A series of clinical trials are being carried out to further establish the current strategies targeting CD8+ T-cell exhaustion. Taken together, despite the proven benefit of immunotherapy in cancer patients, additional efforts are still needed to fully circumvent limitations of exhausted T cells in the treatment. In this review, we will focus on the current cellular and molecular understanding of metabolic changes, epigenetic remodeling, and transcriptional regulation in CD8+ T-cell exhaustion and describe hypothetical treatment approaches based on immunotherapy aiming at reinvigorating exhausted CD8+ T cells.
Collapse
Affiliation(s)
- Md Amir Hossain
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Guilai Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Beiying Dai
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yaxuan Si
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Qitao Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Junaid Wazir
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lutz Birnbaumer
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, Durham, North Carolina, USA.,Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina
| | - Yong Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.,Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
170
|
Zeng Z, Xie D, Gong J. Genome-wide identification of CpG island methylator phenotype related gene signature as a novel prognostic biomarker of gastric cancer. PeerJ 2020; 8:e9624. [PMID: 32821544 PMCID: PMC7396145 DOI: 10.7717/peerj.9624] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 07/07/2020] [Indexed: 12/24/2022] Open
Abstract
Background Gastric cancer (GC) is one of the most fatal cancers in the world. Results of previous studies on the association of the CpG island methylator phenotype (CIMP) with GC prognosis are conflicting and mainly based on selected CIMP markers. The current study attempted to comprehensively assess the association between CIMP status and GC survival and to develop a CIMP-related prognostic gene signature of GC. Methods We used a hierarchical clustering method based on 2,082 GC-related methylation sites to stratify GC patients from the cancer genome atlas into three different CIMP subgroups according to the CIMP status. Gene set enrichment analysis, tumor-infiltrating immune cells, and DNA somatic mutations analysis were conducted to reveal the genomic characteristics in different CIMP-related patients. Cox regression analysis and the least absolute shrinkage and selection operator were performed to develop a CIMP-related prognostic signature. Analyses involving a time-dependent receiver operating characteristic (ROC) curve and calibration plot were adopted to assess the performance of the prognostic signature. Results We found a positive relationship between CIMP and prognosis in GC. Gene set enrichment analysis indicated that cancer-progression-related pathways were enriched in the CIMP-L group. High abundances of CD8+ T cells and M1 macrophages were found in the CIMP-H group, meanwhile more plasma cells, regulatory T cells and CD4+ memory resting T cells were detected in the CIMP-L group. The CIMP-H group showed higher tumor mutation burden, more microsatellite instability-H, less lymph node metastasis, and more somatic mutations favoring survival. We then established a CIMP-related prognostic gene signature comprising six genes (CST6, SLC7A2, RAB3B, IGFBP1, VSTM2L and EVX2). The signature was capable of classifying patients into high‐and low‐risk groups with significant difference in overall survival (OS; p < 0.0001). To assess performance of the prognostic signature, the area under the ROC curve (AUC) for OS was calculated as 0.664 at 1 year, 0.704 at 3 years and 0.667 at 5 years. When compared with previously published gene-based signatures, our CIMP-related signature was comparable or better at predicting prognosis. A multivariate Cox regression analysis indicated the CIMP-related prognostic gene signature was an independent prognostic indicator of GC. In addition, Gene ontology analysis indicated that keratinocyte differentiation and epidermis development were enriched in the high-risk group. Conclusion Collectively, we described a positive association between CIMP status and prognosis in GC and proposed a CIMP-related gene signature as a promising prognostic biomarker for GC.
Collapse
Affiliation(s)
- Zhuo Zeng
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Daxing Xie
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianping Gong
- Molecular Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Department of GI Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
171
|
Liu S, Liu F, Zhou Y, Jin B, Sun Q, Guo S. Immunosuppressive Property of MSCs Mediated by Cell Surface Receptors. Front Immunol 2020; 11:1076. [PMID: 32849489 PMCID: PMC7399134 DOI: 10.3389/fimmu.2020.01076] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022] Open
Abstract
In the past decade, mesenchymal stem cells (MSCs) tend to exhibit inherent tropism for refractory inflammatory diseases and engineered MSCs have appeared on the market as therapeutic agents. Recently, engineered MSCs target to cell surface molecules on immune cells has been a new strategy to improve MSC applications. In this review, we discuss the roles of multiple receptors (ICAM-1, Gal-9, PD-L1, TIGIT, CD200, and CXCR4) in the process of MSCs' immunosuppressive properties. Furthermore, we discuss the principles and strategies for developing receptor-regulated MSCs and their mechanisms of action and the challenges of using MSCs as immunosuppressive therapies.
Collapse
Affiliation(s)
- Siyu Liu
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Fei Liu
- Department of Breast Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - You Zhou
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Baeku Jin
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qiang Sun
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
172
|
Lee BR, Chae S, Moon J, Kim MJ, Lee H, Ko HW, Cho BC, Shim HS, Hwang D, Kim HR, Ha SJ. Combination of PD-L1 and PVR determines sensitivity to PD-1 blockade. JCI Insight 2020; 5:128633. [PMID: 32554931 DOI: 10.1172/jci.insight.128633] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/04/2020] [Indexed: 12/13/2022] Open
Abstract
Expression of immune checkpoint ligands (ICLs) is necessary to trigger the inhibitory signal via immune checkpoint receptors (ICRs) in exhausted T cells under tumor immune microenvironment. Nevertheless,to our knowledge, ICL expression profile in cancer patients has not been investigated. Using previously reported RNA-seq data sets, we found that expression of ICLs was patient specific but their coexpression can be patterned in non-small-cell lung cancers (NSCLCs). Since the expression of PD-L1 and poliovirus receptor (PVR) among various ICLs was independently regulated, we could stratify the patients who were treated with anti-PD-1 later into 4 groups according to the expression level of PD-L1 and PVR. Of interest, high PVR and low PVR expressions in PD-L1-expressing patients enriched nonresponders and responders to PD-1 blockade, respectively, helping in further selection of responders. Using a genetically engineered cancer model, we also found that PVR-deficient and PD-L1-sufficient tumor-bearing mice were highly sensitive to anti-PD-1 therapy, whereas PVR-sufficient and PD-L1-deficient tumor-bearing mice were resistant to anti-PD-1 therapy. Taken together, our study provides a concept that combinatorial expression patterns of PVR and PD-L1 are key determinants for PD-1 blockade and furthermore suggest a better therapeutic usage of immune checkpoint blockades (ICBs).
Collapse
Affiliation(s)
- Bo Ryeong Lee
- Department of Biochemistry, College of Life Science & Biotechnology, and.,Brain Korea 21 (BK21) PLUS Program, Initiative for Biological Functions & Systems, Yonsei University, Seoul, Republic of Korea
| | - Sehyun Chae
- Korea Brain Bank, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Jihyun Moon
- Department of Biochemistry, College of Life Science & Biotechnology, and.,Brain Korea 21 (BK21) PLUS Program, Initiative for Biological Functions & Systems, Yonsei University, Seoul, Republic of Korea
| | - Myeong Joon Kim
- Department of Biochemistry, College of Life Science & Biotechnology, and.,Brain Korea 21 (BK21) PLUS Program, Initiative for Biological Functions & Systems, Yonsei University, Seoul, Republic of Korea
| | - Hankyu Lee
- Department of Biochemistry, College of Life Science & Biotechnology, and.,Brain Korea 21 (BK21) PLUS Program, Initiative for Biological Functions & Systems, Yonsei University, Seoul, Republic of Korea
| | - Hyuk Wan Ko
- Department of Biochemistry, College of Life Science & Biotechnology, and.,Brain Korea 21 (BK21) PLUS Program, Initiative for Biological Functions & Systems, Yonsei University, Seoul, Republic of Korea
| | | | - Hyo Sup Shim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Daehee Hwang
- Department of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hye Ryun Kim
- Yonsei Cancer Center, Division of Medical Oncology, and
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, and.,Brain Korea 21 (BK21) PLUS Program, Initiative for Biological Functions & Systems, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
173
|
Tian H, Zhu X, Lv Y, Jiao Y, Wang G. Glucometabolic Reprogramming in the Hepatocellular Carcinoma Microenvironment: Cause and Effect. Cancer Manag Res 2020; 12:5957-5974. [PMID: 32765096 PMCID: PMC7381782 DOI: 10.2147/cmar.s258196] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/30/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a tumor that exhibits glucometabolic reprogramming, with a high incidence and poor prognosis. Usually, HCC is not discovered until an advanced stage. Sorafenib is almost the only drug that is effective at treating advanced HCC, and promising metabolism-related therapeutic targets of HCC are urgently needed. The “Warburg effect” illustrates that tumor cells tend to choose aerobic glycolysis over oxidative phosphorylation (OXPHOS), which is closely related to the features of the tumor microenvironment (TME). The HCC microenvironment consists of hypoxia, acidosis and immune suppression, and contributes to tumor glycolysis. In turn, the glycolysis of the tumor aggravates hypoxia, acidosis and immune suppression, and leads to tumor proliferation, angiogenesis, epithelial–mesenchymal transition (EMT), invasion and metastasis. In 2017, a mechanism underlying the effects of gluconeogenesis on inhibiting glycolysis and blockading HCC progression was proposed. Treating HCC by increasing gluconeogenesis has attracted increasing attention from scientists, but few articles have summarized it. In this review, we discuss the mechanisms associated with the TME, glycolysis and gluconeogenesis and the current treatments for HCC. We believe that a treatment combination of sorafenib with TME improvement and/or anti-Warburg therapies will set the trend of advanced HCC therapy in the future.
Collapse
Affiliation(s)
- Huining Tian
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin, People's Republic of China
| | - Xiaoyu Zhu
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130021, Jilin, People's Republic of China
| | - You Lv
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin, People's Republic of China
| | - Yan Jiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, Jilin, People's Republic of China
| | - Guixia Wang
- Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun 130021, Jilin, People's Republic of China
| |
Collapse
|
174
|
Yang ZZ, Kim HJ, Wu H, Jalali S, Tang X, Krull JE, Ding W, Novak AJ, Ansell SM. TIGIT Expression Is Associated with T-cell Suppression and Exhaustion and Predicts Clinical Outcome and Anti-PD-1 Response in Follicular Lymphoma. Clin Cancer Res 2020; 26:5217-5231. [PMID: 32631956 DOI: 10.1158/1078-0432.ccr-20-0558] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/01/2020] [Accepted: 06/30/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE T-cell immunoglobulin and ITIM domain (TIGIT), a member of the immune checkpoint family, is important in normal T-cell biology. However, the phenotypical profile and clinical relevance of TIGIT in follicular lymphoma is largely unknown. EXPERIMENTAL DESIGN Biopsy specimens from a cohort of 82 patients with follicular lymphoma were analyzed using mass cytometry to explore the phenotype and biological and clinical significance of TIGIT+ T cells. RESULTS TIGIT is highly expressed on intratumoral T cells and its expression alters T-cell phenotype in follicular lymphoma. TIGIT is abundantly expressed on Treg cells, resulting in an enhanced suppressive property. TIGIT expression on non-Treg/TFH T cells defines a population that exhibits an exhausted phenotype. Clinically, increased numbers of TIGIT+ T cells are associated with inferior patient outcomes and poor survival. We observe that anti-PD-1 therapy with pembrolizumab alters the phenotype of TIGIT+ T subsets and identifies a role for CD28 expression on TIGIT+ T cells in treatment response. CONCLUSIONS The current study provides a comprehensive analysis of the phenotypic profile of intratumoral TIGIT+ T subsets and their prognostic relevance in follicular lymphoma. Inhibition of TIGIT signaling may be an additional mechanism to prevent T-cell suppression and exhaustion in B-cell lymphoma.
Collapse
Affiliation(s)
- Zhi-Zhang Yang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota.
| | - Hyo Jin Kim
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Hongyan Wu
- Department of Immunology, Medical College, China Three Gorges University, Yichang, Hubei, China
| | - Shahrzad Jalali
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Xinyi Tang
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jordan E Krull
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Wei Ding
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Anne J Novak
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Stephen M Ansell
- Division of Hematology and Internal Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
175
|
Mechanisms of immune escape in the cancer immune cycle. Int Immunopharmacol 2020; 86:106700. [PMID: 32590316 DOI: 10.1016/j.intimp.2020.106700] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 06/10/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022]
Abstract
Cancer is a critical issue globally with high incidence and mortality, imposing great burden on the society. Although great progress has been made in immunotherapy based on immune checkpoint, only a subset of patients responds to this treatment, suggesting that cancer immune evasion is still a major barrier in current immunotherapy. There are a series of factors contributing to immune evasion despite in an immunocompetent environment. Given that these factors are involved in different steps of the cancer immune cycle. In this review, we discuss the mechanisms of immune escape in each step of the cancer immune cycle and then present therapeutic strategies for overcoming immune escape, with the potential to better understand the determinants of immune escape and make anti-tumor immunity more effective.
Collapse
|
176
|
Meggyes M, Nagy DU, Szigeti B, Csiszar B, Sandor B, Tamas P, Szereday L. Investigation of mucosal-associated invariant T (MAIT) cells expressing immune checkpoint receptors (TIGIT and CD226) in early-onset preeclampsia. Eur J Obstet Gynecol Reprod Biol 2020; 252:373-381. [PMID: 32682212 DOI: 10.1016/j.ejogrb.2020.06.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/08/2020] [Accepted: 06/15/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE During our work, we examined the possible contribution of MAIT cells in the pathogenesis of the clinical phase of early-onset preeclampsia and how this could be influenced by TIGIT and CD226 immune checkpoint molecules. STUDY DESIGN 37 pregnant women diagnosed with early-onset preeclampsia and 36 healthy, age-matched control women were involved in this study. Peripheral blood mononuclear cells were isolated by density gradient and frozen. After thawing, cells were stained with monoclonal antibodies to characterize MAIT, MAIT-like, and non-MAIT cells. Flow cytometric analyses were used to measure TIGIT, CD226, intracellular perforin, and granzyme B expression. RESULTS MAIT (CD3+ CD8+ Vα7.2+ CD161++), MAIT-like (CD3+ CD8+ Vα7.2+ CD161+) and non-MAIT (CD3+ CD8+ Vα7.2+ CD161-) cell population were identified based on their CD161 receptor positivity. MAIT cells markedly differed in proportion, TIGIT expression, granzyme B, and perforin content compared to MAIT-like and non-MAIT cells. A significant difference was determined in TIGIT expression by non-MAIT cells and in CD8/CD226 positive relationship between the preeclamptic and healthy condition. CONCLUSIONS Considering that we did not detect a notable difference between early-onset preeclampsia and healthy pregnancy, we suppose that peripheral MAIT cells expressing TIGIT and CD226 immune checkpoint molecules have a marginal role in the pathogenesis of early-onset preeclampsia.
Collapse
Affiliation(s)
- Matyas Meggyes
- University of Pecs, Medical School, Department of Medical Microbiology and Immunology, 7624, Pecs, 12 Szigeti Street, Hungary; Janos Szentagothai Research Centre, 7624 Pecs, 20 Ifjusag Street, Hungary.
| | - David U Nagy
- University of Pécs, Department of Genetics and Molecular Biology, 7624 Pecs, 6 Ifjusag Street, Hungary.
| | - Brigitta Szigeti
- University of Pecs, Medical School, Department of Medical Microbiology and Immunology, 7624, Pecs, 12 Szigeti Street, Hungary.
| | - Beata Csiszar
- University of Pecs, Medical School, 1st Department of Medicine, 7624 Pecs, 13 Ifjusag Street, Hungary.
| | - Barbara Sandor
- University of Pecs, Medical School, 1st Department of Medicine, 7624 Pecs, 13 Ifjusag Street, Hungary.
| | - Peter Tamas
- University of Pecs, Medical School, Department of Obstetrics and Gynaecology, 7624 Pecs, 17 Edesanyak Street, Hungary.
| | - Laszlo Szereday
- University of Pecs, Medical School, Department of Medical Microbiology and Immunology, 7624, Pecs, 12 Szigeti Street, Hungary; Janos Szentagothai Research Centre, 7624 Pecs, 20 Ifjusag Street, Hungary.
| |
Collapse
|
177
|
Hoseini-Aghdam M, Sheikh V, Eftekharian MM, Rezaeepoor M, Behzad M. Enhanced expression of TIGIT but not neuropilin-1 in patients with type 2 diabetes mellitus. Immunol Lett 2020; 225:1-8. [PMID: 32540486 DOI: 10.1016/j.imlet.2020.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND The aggressive T helper cell responses and regulatory T (Treg) cells dysfunction exist in type 2 diabetes mellitus (T2DM). The co-inhibitory T cell immunoglobulin and ITIM-domain (TIGIT), neuropilin-1 (Nrp-1), and the co-stimulatory CD226 play a critical role in the inhibition or activation of immune responses. In this project, the expression of TIGIT, CD226, Nrp-1, and their ligands, CD155 and semaphorin 3A (Sema-3A) were investigated in T2DM. METHODS Peripheral blood mononuclear cells (PBMCs) were collected from 30 patients with T2DM, and 30 healthy controls (HCs). The frequencies of TIGIT and Nrp-1 on CD4+CD25hi Treg cells, CD4+CD25- responder T cells, total CD4+ T cells, and non-CD4+ cells were assessed using flow cytometry. The mRNA levels of TIGIT, CD226, Nrp-1, CD155, and Sema-3A were assessed by real-time PCR. RESULTS The percentage and MFI of TIGIT on CD4+CD25hi T cells, CD4+CD25- T cells, total CD4+ T cells, and non-CD4+ cells were higher in patients versus HCs (p < 0.05 for all). The mRNA level of TIGIT was increased in patients compared with HCs (p = 0.003). No differences were observed in the expression of CD226, CD155, Nrp-1, and Sema-3A between the groups. CONCLUSIONS The expression of TIGIT was enhanced in T2DM and the TIGIT axis could be considered as a new therapeutic purpose for the T2DM.
Collapse
Affiliation(s)
- Mirhamed Hoseini-Aghdam
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Vida Sheikh
- Department of Internal Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mahsa Rezaeepoor
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mahdi Behzad
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
178
|
Nie H, Mei J, Zhang Q, An F, Zhan Q. Systematic Characterization of the Expression and Prognostic Values of Formin-Like Gene Family in Gastric Cancer. DNA Cell Biol 2020; 39:1664-1677. [PMID: 32551946 DOI: 10.1089/dna.2020.5508] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Formin-like genes (FMNLs) are members of formins family and have been implicated to the development and progression of multiple cancers. This research aims to analyze the expression profiles, prognostic values, and immune infiltrating associations of FMNLs in gastric cancer (GC) using multiple online bioinformatics website, including Oncomine, UALCAN, Kaplan-Meier Plotter, TIMER, GeneMANIA, DAVID, and LinkedOmics databases. The mRNA levels of FMNL1/2/3 were higher in GC tissues than normal. Meanwhile, FMNLs expressions tend to be upregulated in advanced and poorly differentiated GC. Prognostic value analysis suggested that high transcription levels of FMNL1/3 were associated with poor overall survival in GC patients. Correlation analysis between FMNLs expressions and immune infiltrating GC revealed that the expressions of FMNLs were significantly associated with immune infiltrating. Protein-protein interaction network and enrichment analysis of FMNLs in GC showed that FMNLs coexpressed genes mainly participated in organizing actin cytoskeleton through affecting small G proteins activity. Moreover, Gene Set Enrichment Analysis (GSEA) analysis uncovered FMNLs and their coexpressed genes was tightly associated with immune-related cellular functions. These findings demonstrate that FMNLs might play significant immunomodulatory roles in tumor immunity and could be novel therapeutic targets and potential prognostic biomarkers in GC.
Collapse
Affiliation(s)
- He Nie
- Department of Gastroenterology and Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Jie Mei
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Qinglin Zhang
- Department of Gastroenterology and Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Fangmei An
- Department of Gastroenterology and Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Qiang Zhan
- Department of Gastroenterology and Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
179
|
Zhou X, Ding X, Li H, Yang C, Ma Z, Xu G, Yang S, Zhang D, Xie X, Xin L, Luo X. Upregulation of TIGIT and PD-1 in Colorectal Cancer with Mismatch-repair Deficiency. Immunol Invest 2020; 50:338-355. [PMID: 32397769 DOI: 10.1080/08820139.2020.1758130] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Xuebing Zhou
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
- Department of General Surgery Center, People's Hospital of Ningxia Hui Autonomous region, Yinchuan, China
| | - Xiaoling Ding
- Department of Gastroenterology, People's Hospital of Ningxia Hui Autonomous region, Yinchuan, China
| | - Hai Li
- Department of Anal-Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Chun Yang
- Department of Anal-Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Zhanbing Ma
- Department of Medical Genetics and Cell Biology, Ningxia Medical University, Yinchuan, China
| | - Guangxian Xu
- Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Shaoqi Yang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Dong Zhang
- Department of Anal-Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiaoliang Xie
- Department of Anal-Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Lei Xin
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Xiaoli Luo
- Department of Anal-Colorectal Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
180
|
Fenwick C, Joo V, Jacquier P, Noto A, Banga R, Perreau M, Pantaleo G. T-cell exhaustion in HIV infection. Immunol Rev 2020; 292:149-163. [PMID: 31883174 PMCID: PMC7003858 DOI: 10.1111/imr.12823] [Citation(s) in RCA: 217] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022]
Abstract
The T‐cell response is central in the adaptive immune‐mediated elimination of pathogen‐infected and/or cancer cells. This activated T‐cell response can inflict an overwhelming degree of damage to the targeted cells, which in most instances leads to the control and elimination of foreign invaders. However, in conditions of chronic infection, persistent exposure of T cells to high levels of antigen results in a severe T‐cell dysfunctional state called exhaustion. T‐cell exhaustion leads to a suboptimal immune‐mediated control of multiple viral infections including the human immunodeficiency virus (HIV). In this review, we will discuss the role of T‐cell exhaustion in HIV disease progression, the long‐term defect of T‐cell function even in aviremic patients on antiretroviral therapy (ART), the role of exhaustion‐specific markers in maintaining a reservoir of latently infected cells, and exploiting these markers in HIV cure strategies.
Collapse
Affiliation(s)
- Craig Fenwick
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Victor Joo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Patricia Jacquier
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alessandra Noto
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Riddhima Banga
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Matthieu Perreau
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Service of Immunology and Allergy, Department of Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Swiss Vaccine Research Institute, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
181
|
Therapeutic Development of Immune Checkpoint Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:619-649. [PMID: 32185726 DOI: 10.1007/978-981-15-3266-5_23] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immune checkpoint blockade (ICB) has been proven to be an effective strategy for enhancing the effector activity of anti-tumor T cells, and checkpoint blockers targeting CTLA-4, PD-1, and PD-L1 have displayed strong and durable clinical responses in certain cancer patients. The new hope brought by ICB therapy has led to the boost in therapeutic development of ICBs in recent years. Nonetheless, the therapeutic efficacy of ICBs varies substantially among cancer types and patients, and only a proportion of cancer patients could benefit from ICBs. The emerging targets and molecules for enhancing anticancer immunity may bring additional therapeutic opportunities for cancer patients. The current challenges in the ICB therapy have been discussed, aimed to provide further strategies for maximizing the efficacy of ICB therapy.
Collapse
|
182
|
You H, Zhang YZ, Lai HL, Li D, Liu YQ, Li RZ, Khan I, Hsiao WWL, Duan FG, Fan XX, Yao XJ, Cao YB, Wu QB, Leung ELH, Wang MF. Prognostic significance of tumor poliovirus receptor and CTLA4 expression in patients with surgically resected non-small-cell lung cancer. J Cancer Res Clin Oncol 2020; 146:1441-1450. [PMID: 32248302 DOI: 10.1007/s00432-020-03189-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/17/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Poliovirus receptor (PVR) is a tumor promoter and a regulatory checkpoint that enhances immunosuppression. We investigated PVR expression by applying immunohistochemistry (IHC) staining. A positive association existed between PVR expression and cytotoxic T lymphocyte-associated antigen 4 (CTLA4) expression in patients with surgically resected non-small-cell lung cancer (NSCLC). PVR expression is a prognosis predictor of lung adenocarcinoma. PURPOSE To investigate the prognostic significance of PVR expression and CTLA4 expression for surgically resected NSCLC. PATIENTS AND METHODS The medical records of 108 Chinese patients with primary NSCLC who underwent surgery were retrospectively reviewed. The expression of PVR and CTLA4 were measured through IHC. Clinical characteristics, the association between PVR and CTLA4, and the prognostic significance of PVR were analyzed. RESULTS A significant positive association was observed between PVR and CTLA4 expression in NSCLC (P = 0.016). PVR had a high positive rate among females, nonsmokers, and patients with adenocarcinoma and advanced lung cancer. The overall survival (OS) of patients with negative PVR expression was significantly longer than that of patients with positive PVR expression (P = 0.049), especially among females (P = 0.03) and nonsmokers (P = 0.025). Multivariate analysis results showed that advanced tumor stage and PVR expression were independent prognosis predictors of poor OS. CONCLUSION PVR can potentially serve as a prognostic predictor and biomarker for NSCLC and cancer anti-CTLA4 immunotherapy response.
Collapse
Affiliation(s)
- Hui You
- Department of Respiratory and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yi-Zhong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Huan-Ling Lai
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Dan Li
- Department of Pathology, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yu-Quan Liu
- Department of Respiratory and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Run-Ze Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Imran Khan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Wendy Wen-Lun Hsiao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Fu-Gang Duan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Xing-Xing Fan
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Xiao-Jun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Ya-Bing Cao
- Department of Oncology, Kiang Wu Hospital, Macau (SAR), China
| | - Qi-Biao Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China.
| | - Elaine Lai-Han Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China.
| | - Mei-Fang Wang
- Department of Respiratory and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Shiyan, China.
| |
Collapse
|
183
|
Chen F, Xu Y, Chen Y, Shan S. TIGIT enhances CD4 + regulatory T-cell response and mediates immune suppression in a murine ovarian cancer model. Cancer Med 2020; 9:3584-3591. [PMID: 32212317 PMCID: PMC7221438 DOI: 10.1002/cam4.2976] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/27/2022] Open
Abstract
Ovarian cancer (OC) is the fifth-leading cause of cancer-related death in women with a pathogenesis involving activation of regulatory T cells (Tregs). The T-cell immunoglobulin and ITIM domain (TIGIT) is a well-known immune checkpoint molecule that inhibits T-cell responses. However, the role of TIGIT in OC is not comprehensively understood. In this study, we revealed crucial functions of TIGIT in the development and progression of OC. ID8 cells were used to establish a murine OC model. TIGIT expression was increased in immune cells of OC mice, particularly in CD4+ Tregs. Anti-TIGIT monoclonal antibodies (mAb) were used to block the function of TIGIT in OC mice, and we found that the anti-TIGIT treatment reduced the proportion of CD4+ Tregs, but did not affect CD4+ and CD8+ T cells or natural killer cells. Splenic CD4+ Tregs from OC mice were isolated after the anti-TIGIT treatment, and their functioning was examined. Inhibition of TIGIT lowered the degree of immunosuppression induced by CD4+ Tregs. A survival curve suggested that anti-TIGIT treatment can improve the survival rate of OC in mice. We conclude that TIGIT enhanced CD4+ Tregs response and mediated immunosuppression in the OC model. Our data suggest that inhibition of TIGIT is a potential therapeutic target in OC patients.
Collapse
Affiliation(s)
- Fengzhen Chen
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yanying Xu
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yulong Chen
- Department of Lung Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Shu Shan
- Department of Gynecology and Obstetrics, Affiliated Tongji Hospital, Tongji University, Shanghai, China
| |
Collapse
|
184
|
Zhang X, Gao F, Li N, Zhang J, Dai L, Yang H. Peroxiredoxins and Immune Infiltrations in Colon Adenocarcinoma: Their Negative Correlations and Clinical Significances, an In Silico Analysis. J Cancer 2020; 11:3124-3143. [PMID: 32231717 PMCID: PMC7097948 DOI: 10.7150/jca.38057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 01/04/2020] [Indexed: 01/05/2023] Open
Abstract
Background: Peroxiredoxins (PRDXs) were reported to be associated with inflammation response in previous studies. In colon adenocarcinoma (COAD), however, their correlations and clinical significance were unclear. Methods: The RNA-seq data of 452 COAD patients with clinical information was downloaded from The Cancer Genome Atlas (TCGA) and transcripts per million (TPM) normalized. Comparisons of relative expressions of PRDXs between COAD tumor and normal controls were applied. PRDXs dy-regulations in COAD were validated via Oncomine, Human Protein Atlas (HPA) and Gene Expression Omnibus (GEO) repository. Through Tumor Immune Estimation Resource (TIMER), the immune estimation of TCGA-COAD patients was downloaded and the dy-regulated PRDXs were analyzed for their correlations with immune infiltrations in COAD. The TCGA-COAD patients were divided into younger group (age≤65 years) and older group (age>65 years) to investigate the prognostic roles of age, TNM stage, dy-regulated PRDXs and the immune infiltrations in different age groups through Kaplan-Meier survival and Cox regression analyses. Results: Three of the PRDX members showed their expressional differences both at protein and mRNA level. PRDX2 was consistently up-regulated while PRDX6 down-regulated in COAD. PRDX1 was overexpressed (mRNA) while nuclear absent (protein) in the tumor tissues. PRDX1 overexpression and PRDX6 under-expression were also shown in the stem-like colonospheres from colon cancer cells. Via TIMER, PRDX1, PRDX2, and PRDX6 were found to be negatively correlated with the immune infiltrations in COAD. Both in the younger and older patients, TNM stage had prognostic effects on their overall survival (OS) and recurrence-free survival (RFS). CD4+ T cell had independent unfavorable effects on OS of the younger patients while age had similar effects on RFS of the older ones. CD8+ T cell was independently prognostic for RFS in the two groups. Conclusions: Late diagnosis indicated poor prognosis in COAD and dy-regulated PRDXs w might be new markers for its early diagnosis. Age was prognostic and should be considered in the treatments of the older patients. Dy-regulated PRDXs were negatively correlated with immune infiltration levels. CD4+ T cell and CD8+ T cell infiltrations were prognostic in COAD and their potential as immune targets needed further investigation.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, Henan Province, China.,Medical Laboratory Center, Henan Medical College, Zhengzhou, Henan Province, China.,Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Fenglan Gao
- Department of Pathology, Henan Medical College, Zhengzhou, Henan Province, China
| | - Ningning Li
- Department of Pathology, Henan Medical College, Zhengzhou, Henan Province, China
| | - Jinzhong Zhang
- Medical Laboratory Center, Henan Medical College, Zhengzhou, Henan Province, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Hongmei Yang
- Department of Pathology, Henan Medical College, Zhengzhou, Henan Province, China.,Medical Laboratory Center, Henan Medical College, Zhengzhou, Henan Province, China
| |
Collapse
|
185
|
Khan M, Arooj S, Wang H. NK Cell-Based Immune Checkpoint Inhibition. Front Immunol 2020; 11:167. [PMID: 32117298 PMCID: PMC7031489 DOI: 10.3389/fimmu.2020.00167] [Citation(s) in RCA: 212] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/21/2020] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy, with an increasing number of therapeutic dimensions, is becoming an important mode of treatment for cancer patients. The inhibition of immune checkpoints, which are the source of immune escape for various cancers, is one such immunotherapeutic dimension. It has mainly been aimed at T cells in the past, but NK cells are a newly emerging target. Simultaneously, the number of checkpoints identified has been increasing in recent times. In addition to the classical NK cell receptors KIRs, LIRs, and NKG2A, several other immune checkpoints have also been shown to cause dysfunction of NK cells in various cancers and chronic infections. These checkpoints include the revolutionized CTLA-4, PD-1, and recently identified B7-H3, as well as LAG-3, TIGIT & CD96, TIM-3, and the most recently acknowledged checkpoint-members of the Siglecs family (Siglec-7/9), CD200 and CD47. An interesting dimension of immune checkpoints is their candidacy for dual-checkpoint inhibition, resulting in therapeutic synergism. Furthermore, the combination of immune checkpoint inhibition with other NK cell cytotoxicity restoration strategies could also strengthen its efficacy as an antitumor therapy. Here, we have undertaken a comprehensive review of the literature to date regarding NK cell-based immune checkpoints.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Oncology, The First Affiliated Hospital, Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Sumbal Arooj
- Department of Biochemistry, University of Sialkot, Sialkot, Pakistan
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital, Institute for Liver Diseases of Anhui Medical University, Hefei, China
| |
Collapse
|
186
|
Sayitoglu EC, Georgoudaki AM, Chrobok M, Ozkazanc D, Josey BJ, Arif M, Kusser K, Hartman M, Chinn TM, Potens R, Pamukcu C, Krueger R, Zhang C, Mardinoglu A, Alici E, Temple HT, Sutlu T, Duru AD. Boosting Natural Killer Cell-Mediated Targeting of Sarcoma Through DNAM-1 and NKG2D. Front Immunol 2020; 11:40. [PMID: 32082316 PMCID: PMC7001093 DOI: 10.3389/fimmu.2020.00040] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 01/08/2020] [Indexed: 12/16/2022] Open
Abstract
Sarcomas are malignancies of mesenchymal origin that occur in bone and soft tissues. Many are chemo- and radiotherapy resistant, thus conventional treatments fail to increase overall survival. Natural Killer (NK) cells exert anti-tumor activity upon detection of a complex array of tumor ligands, but this has not been thoroughly explored in the context of sarcoma immunotherapy. In this study, we investigated the NK cell receptor/ligand immune profile of primary human sarcoma explants. Analysis of tumors from 32 sarcoma patients identified the proliferative marker PCNA and DNAM-1 ligands CD112 and/or CD155 as commonly expressed antigens that could be efficiently targeted by genetically modified (GM) NK cells. Despite the strong expression of CD112 and CD155 on sarcoma cells, characterization of freshly dissociated sarcomas revealed a general decrease in tumor-infiltrating NK cells compared to the periphery, suggesting a defect in the endogenous NK cell response. We also applied a functional screening approach to identify relevant NK cell receptor/ligand interactions that induce efficient anti-tumor responses using a panel NK-92 cell lines GM to over-express 12 different activating receptors. Using GM NK-92 cells against primary sarcoma explants (n = 12) revealed that DNAM-1 over-expression on NK-92 cells led to efficient degranulation against all tested explants (n = 12). Additionally, NKG2D over-expression showed enhanced responses against 10 out of 12 explants. These results show that DNAM-1+ or NKG2D+ GM NK-92 cells may be an efficient approach in targeting sarcomas. The degranulation capacity of GM NK-92 cell lines was also tested against various established tumor cell lines, including neuroblastoma, Schwannoma, melanoma, myeloma, leukemia, prostate, pancreatic, colon, and lung cancer. Enhanced degranulation of DNAM-1+ or NKG2D+ GM NK-92 cells was observed against the majority of tumor cell lines tested. In conclusion, DNAM-1 or NKG2D over-expression elicited a dynamic increase in NK cell degranulation against all sarcoma explants and cancer cell lines tested, including those that failed to induce a notable response in WT NK-92 cells. These results support the broad therapeutic potential of DNAM-1+ or NKG2D+ GM NK-92 cells and GM human NK cells for the treatment of sarcomas and other malignancies.
Collapse
Affiliation(s)
- Ece Canan Sayitoglu
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States.,NSU Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Anna-Maria Georgoudaki
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States.,NSU Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL, United States.,Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Michael Chrobok
- Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Didem Ozkazanc
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Benjamin J Josey
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States.,NSU Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Muhammad Arif
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Kim Kusser
- Translational Research and Economic Development, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Michelle Hartman
- Translational Research and Economic Development, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Tamara M Chinn
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Renee Potens
- NSU Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Cevriye Pamukcu
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - Robin Krueger
- Translational Research and Economic Development, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Cheng Zhang
- Faculty of Dentistry, Oral & Craniofacial Sciences, Centre for Host-Microbiome Interactions, King's College London, London, United Kingdom
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden.,Faculty of Dentistry, Oral & Craniofacial Sciences, Centre for Host-Microbiome Interactions, King's College London, London, United Kingdom
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Harry Thomas Temple
- Department of Surgery, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Tolga Sutlu
- Department of Molecular Biology and Genetics, Bogaziçi University, Istanbul, Turkey
| | - Adil Doganay Duru
- Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, United States.,NSU Cell Therapy Institute, Nova Southeastern University, Fort Lauderdale, FL, United States.,Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
187
|
Sun Y, Luo J, Chen Y, Cui J, Lei Y, Cui Y, Jiang N, Jiang W, Chen L, Chen Y, Kuang Y, Tang K, Ke Z. Combined evaluation of the expression status of CD155 and TIGIT plays an important role in the prognosis of LUAD (lung adenocarcinoma). Int Immunopharmacol 2020; 80:106198. [PMID: 31954274 DOI: 10.1016/j.intimp.2020.106198] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/13/2019] [Accepted: 01/06/2020] [Indexed: 12/25/2022]
Abstract
The interaction between CD155 and its high-affinity ligand TIGIT is being increasingly investigated in various solid tumors. However, the prognostic significance of CD155 and TIGIT in lung adenocarcinoma (LUAD) remains unclear. In this study, immunohistochemistry was applied in 334 LUAD cases to evaluate the expression of CD155 and TIGIT. Western blotting was conducted in 5 paired primary LUAD and adjacent normal lung tissues. Our results reveal that CD155 and TIGIT are overexpressed in LUAD tissues and that aberrant overexpression is closely correlated with poor clinical outcomes (P < 0.01). The multivariate model also shows that CD155 expression is an independent risk factor for LUAD (RR, 1.34; P = 0.036). Moreover, patients expressing high CD155 and TIGIT simultaneously presented shorter overall survival (OS) (P < 0.01) and progression-free survival (PFS) (P < 0.01). These findings suggest that CD155 and TIGIT can make up a prognosticating tool to predict clinical outcomes, thereby contributing to personalized medical care in LUAD.
Collapse
Affiliation(s)
- Yu Sun
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Jiping Luo
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Yangshan Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Ji Cui
- Gastrointestinal Surgery Center, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Yiyan Lei
- Department of Thoracic Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Yongmei Cui
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Neng Jiang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Wenting Jiang
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Lili Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Yanyang Chen
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Yukun Kuang
- Department of Respiratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Kejing Tang
- Department of Respiratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, PR China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510080, PR China.
| |
Collapse
|
188
|
Overexpression of an Immune Checkpoint (CD155) in Breast Cancer Associated with Prognostic Significance and Exhausted Tumor-Infiltrating Lymphocytes: A Cohort Study. J Immunol Res 2020; 2020:3948928. [PMID: 32411795 PMCID: PMC7201814 DOI: 10.1155/2020/3948928] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/06/2019] [Accepted: 12/26/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose The immune checkpoint inhibitor is approved for breast cancer treatment, but the low expression of PD-L1 limits the immunotherapy. CD155 is another immune checkpoint protein in cancers and interacts with ligands to regulate immune microenvironment. This study is aimed at investigating the expression of CD155 and the association with prognosis and pathological features of breast cancer. Methods 126 patients were recruited this cohort study consecutively, and CD155 expression on tumor cells was detected by immunohistochemistry. The Kaplan-Meier survival curve and Cox hazard regression model were used to estimate the association. Results 38.1% patients had an overexpression of CD155, and the proportion of tumor cells with CD155 overexpression was 17%, 39%, 37%, and 62% among Luminal A, Luminal B, HER2-positive, and triple negative breast cancer cases, respectively (p < 0.05). Patients with CD155 overexpression had the Ki-67 index significantly higher than that of patients with low expression (42% vs. 26%). Though the number of tumor-infiltrating lymphocytes was higher among patients with CD155 overexpression (144/HPF vs. 95/HPF), the number of PD-1+ lymphocytes was significantly higher (52/HPF vs. 25/HPF, p < 0.05). Patients of CD155 overexpression had the disease-free and overall survival decreased by 13 months and 9 months, respectively (p < 0.05). CD155 overexpression was associated with an increased relapse (HR = 13.93, 95% CI 2.82, 68.91) and death risk for breast cancer patients (HR = 5.47, 1.42, 20.99). Conclusions Overexpression of CD155 was correlated with more proliferative cancer cells and a dysfunctional immune microenvironment. CD155 overexpression introduced a worse relapse-free and overall survival and might be a potential immunotherapy target for breast cancer.
Collapse
|
189
|
Sheppard AD, Lysaght J. Immunometabolism and Its Potential to Improve the Current Limitations of Immunotherapy. Methods Mol Biol 2020; 2184:233-263. [PMID: 32808230 DOI: 10.1007/978-1-0716-0802-9_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The last century of research in tumor immunology has culminated in the advent of immunotherapy, most notably immune checkpoint inhibitors. These drugs have shown encouraging results across a multitude of malignancies and have shifted the paradigm of cancer treatment. However, no more than 40% of patients treated with these immune checkpoint blockade inhibitors respond. Thus, resistance is a barrier to therapy that remains poorly understood. All cells require energy and biosynthetic precursors for survival, growth, and functioning, where multiple metabolic pathways allow for flexibility in how nutrients are utilized. A defining hallmark of many cancers is altered cellular metabolism, creating an imbalanced demand for nutrients within the tumor microenvironment. Immunometabolism is increasingly understood to be vital to the functions and phenotypes of a myriad of immune cell subsets. In tumors, the high demand for nutrients by the tumor drives competition between tumor cells and infiltrating immune cells, culminating in dysfunctional immune responses. This chapter discusses the recent successes in cancer immunotherapy and highlights challenges to therapy. We also outline the major metabolic processes involved in the generation of an immune response, how this can become dysregulated in the context of the tumor microenvironment, and how this contributes to resistance to immunotherapy. Finally, we explore the potential for targeting immunometabolic pathways to improve immunotherapy, and examine current trials targeting various aspects of metabolism in an attempt to improve the outcomes from immunotherapy.
Collapse
Affiliation(s)
- Andrew D Sheppard
- Cancer Immunology and Immunotherapy Group, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland
| | - Joanne Lysaght
- Cancer Immunology and Immunotherapy Group, Trinity Translational Medicine Institute, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
190
|
Hargadon KM. Tumor microenvironmental influences on dendritic cell and T cell function: A focus on clinically relevant immunologic and metabolic checkpoints. Clin Transl Med 2020; 10:374-411. [PMID: 32508018 PMCID: PMC7240858 DOI: 10.1002/ctm2.37] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/23/2020] [Accepted: 04/23/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer immunotherapy is fast becoming one of the most promising means of treating malignant disease. Cancer vaccines, adoptive cell transfer therapies, and immune checkpoint blockade have all shown varying levels of success in the clinical management of several cancer types in recent years. However, despite the clinical benefits often achieved by these regimens, an ongoing problem for many patients is the inherent or acquired resistance of their cancer to immunotherapy. It is now appreciated that dendritic cells and T lymphocytes both play key roles in antitumor immune responses and that the tumor microenvironment presents a number of barriers to the function of these cells that can ultimately limit the success of immunotherapy. In particular, the engagement of several immunologic and metabolic checkpoints within the hostile tumor microenvironment can severely compromise the antitumor functions of these important immune populations. This review highlights work from both preclinical and clinical studies that has shaped our understanding of the tumor microenvironment and its influence on dendritic cell and T cell function. It focuses on clinically relevant targeted and immunotherapeutic strategies that have emerged from these studies in an effort to prevent or overcome immune subversion within the tumor microenvironment. Emphasis is also placed on the potential of next-generation combinatorial regimens that target metabolic and immunologic impediments to dendritic cell and T lymphocyte function as strategies to improve antitumor immune reactivity and the clinical outcome of cancer immunotherapy going forward.
Collapse
Affiliation(s)
- Kristian M. Hargadon
- Hargadon LaboratoryDepartment of BiologyHampden‐Sydney CollegeHampden‐SydneyVirginiaUSA
| |
Collapse
|
191
|
Harjunpää H, Guillerey C. TIGIT as an emerging immune checkpoint. Clin Exp Immunol 2019; 200:108-119. [PMID: 31828774 DOI: 10.1111/cei.13407] [Citation(s) in RCA: 308] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2019] [Indexed: 12/15/2022] Open
Abstract
T cell immunoglobulin and ITIM domain (TIGIT) is an inhibitory receptor expressed on lymphocytes that was recently propelled under the spotlight as a major emerging target in cancer immunotherapy. TIGIT interacts with CD155 expressed on antigen-presenting cells or tumour cells to down-regulate T cell and natural killer (NK) cell functions. TIGIT has emerged as a key inhibitor of anti-tumour responses that can hinder multiple steps of the cancer immunity cycle. Pre-clinical studies indicated that TIGIT blockade may protect against various solid and haematological cancers. Several monoclonal antibodies (mAbs) that block the inhibitory activity of human TIGIT have been developed. Clinical trials are ongoing, investigating TIGIT blockade as a monotherapy or in combination with anti-PD1/PD-L1 mAbs for the treatment of patients with advanced solid malignancies. In this review, we cover our current knowledge on TIGIT, from its discovery in 2009 to its current status as a clinical target.
Collapse
Affiliation(s)
- H Harjunpää
- Molecular and Integrative Biosciences, Faculty of Biological and Environmental Sciences, The University of Helsinki, Helsinki, Finland
| | - C Guillerey
- Cancer Immunotherapies Laboratory, Mater Research Institute, The University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
192
|
Anestakis D, Petanidis S, Domvri K, Tsavlis D, Zarogoulidis P, Katopodi T. Carboplatin chemoresistance is associated with CD11b +/Ly6C + myeloid release and upregulation of TIGIT and LAG3/CD160 exhausted T cells. Mol Immunol 2019; 118:99-109. [PMID: 31862674 DOI: 10.1016/j.molimm.2019.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/29/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
Abstract
Immunosuppressive chemoresistance is a major barrier in lung cancer treatment. Tumor immunosuppressive environments mediated by myeloid-derived suppressor cells (MDSCs) play a key role in chemotherapy induced MDSC development and differentiation but their mechanistic role has not been elucidated. Here, we define a role for carboplatin based chemotherapy in potentiating an MDSC-dependent pathway that triggers the chemoresistance mechanism. Findings reveal MDSC differentiation and activation of IL-13/IL-33-mediated pathway through VCAM/RANTES following carboplatin treatment. Furthemore, secretion of T regulatory IL-10-producing CD4+Foxp3+ cells was increased followed by expression of co-inhibitory receptor TIGIT on T cells, leading to a dysfunctional T cell phenotype. These cells were characterized by an immunosuppressive phenotype with impaired activation, proliferation and cytokine production. Lung cancer tissues expressed CD155, which bound TIGIT receptors and inactivated CD8 T cells. This TIGIT expression on tumor-infiltrating T cells was found to be associated with tumor progression and was linked to functional exhaustion of T cells. In addition, the presence of plasmacytoid dendritic cells (pDCs) exposed to tumor-derived factors further enhanced tumor progression through IL-10 production and up-regulation of the inducible co-stimulatory ligand (ICOS-L). Deciphering these deranged immune mechanisms and how they are impacted by chemotherapy induction is essential for incorporation of novel immune-based strategies in order to restore immunity and inhibit the immunosuppressive phenotype of metastatic lung cancer.
Collapse
Affiliation(s)
- Doxakis Anestakis
- Department of Medicine, Laboratory of Forensic Medicine and Toxicology, Aristotle University of Thessaloniki, 54124, Greece
| | - Savvas Petanidis
- Department of Medicine, Laboratory of Medical Biology and Genetics, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece; Department of Pulmonology, I.M. Sechenov First Moscow State Medical University, Moscow, 119992, Russian Federation.
| | - Kalliopi Domvri
- Pulmonary Department-Oncology Unit, "G. Papanikolaou" General Hospital, Aristotle University of Thessaloniki, Thessaloniki, 57010, Greece
| | - Drosos Tsavlis
- Department of Medicine, Laboratory of Experimental Physiology, Aristotle University of Thessaloniki, 54124, Greece
| | - Paul Zarogoulidis
- Third Department of Surgery, "AHEPA" University Hospital, Aristotle University of Thessaloniki, 55236, Thessaloniki, Greece
| | - Theodora Katopodi
- Department of Medicine, Laboratory of Medical Biology and Genetics, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| |
Collapse
|
193
|
Song WM, Lin X, Liao X, Hu D, Lin J, Sarpel U, Ye Y, Feferman Y, Labow DM, Walsh MJ, Zheng X, Zhang B. Multiscale network analysis reveals molecular mechanisms and key regulators of the tumor microenvironment in gastric cancer. Int J Cancer 2019; 146:1268-1280. [PMID: 31463974 PMCID: PMC7004118 DOI: 10.1002/ijc.32643] [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: 04/02/2019] [Revised: 07/18/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is the third leading cause of cancer deaths and the fourth most prevalent malignancy worldwide. The high incidence and mortality rates of gastric cancer result from multiple factors such as ineffective screening, diagnosis, and limited treatment options. In our study, we sought to systematically identify predictive molecular networks and key regulators to elucidate complex interacting signaling pathways in GC. We performed an integrative network analysis of the transcriptomic data in The Cancer Genome Atlas (TCGA) gastric cancer cohort and then comprehensively characterized the predictive subnetworks and key regulators by the matched genetic and epigenetic data. We identified 221 gene subnetworks (modules) in GC. The most prognostic subnetworks captured multiple aspects of the tumor microenvironment in GC involving interactions among stromal, epithelial and immune cells. We revealed the genetic and epigenetic underpinnings of those subnetworks and their key transcriptional regulators. We computationally predicted and experimentally validated specific mechanisms of anticancer effects of GKN2 in gastric cancer proliferation and invasion in vitro. The network models and the key regulators of the tumor microenvironment in GC identified here pave a way for developing novel therapeutic strategies for GC. What's new? Gene signatures have been identified for diagnosis and classification of gastric cancer (GC) as well as prediction of therapeutic response. However, key molecular mechanisms underlying prognosis remain to be revealed. Our study systematically identifies and characterizes predictive molecular networks and key regulators. The most prognostic subnetworks capture multiple aspects of the tumor microenvironment in GC involving interactions among stromal, epithelial, and immune cells. The authors computationally predicted and experimentally validated specific mechanisms of anti‐cancer effects of GKN2 in GC proliferation and invasion in vitro. These network models and key regulators pave the way for developing novel therapeutic strategies for GC.
Collapse
Affiliation(s)
- Won-Min Song
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY.,Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Xiandong Lin
- Laboratory of Radiation Oncology and Radiobiolog, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China.,Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
| | - Xuehong Liao
- Department of Pathology, Zhongshan Hospital of Xiamen University, Xiamen, Fujian, China
| | - Dan Hu
- Department of Pathology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Jieqiong Lin
- Department of Pathology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Umut Sarpel
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yunbin Ye
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China.,Laboratory of Immuno-Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Yael Feferman
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Daniel M Labow
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Martin J Walsh
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY.,Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY.,The Mount Sinai Center for RNA Biology and Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Xiongwei Zheng
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China.,Department of Pathology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian, China
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY.,Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
194
|
Zhao L, Liu Y, Zhang J, Liu Y, Qi Q. LncRNA SNHG14/miR-5590-3p/ZEB1 positive feedback loop promoted diffuse large B cell lymphoma progression and immune evasion through regulating PD-1/PD-L1 checkpoint. Cell Death Dis 2019; 10:731. [PMID: 31570691 PMCID: PMC6769008 DOI: 10.1038/s41419-019-1886-5] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/12/2019] [Accepted: 08/20/2019] [Indexed: 12/22/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL) is the commonest disorder derived from the B-lymphocytes. Inhibiting the immune checkpoint through naturalizing programmed death-1 (PD-1) and programmed death ligand 1 (PD-L1) is proved to be a successful therapeutic regime for lymphoma. Long non-coding RNAs (lncRNAs) are unceasingly reported to be promising biological targets for the cancer therapies. This study planned to explore the regulation of small nucleolar RNA host gene 14 (SNHG14) on DLBCL. SNHG14 level in DLBCL samples and cell lines was analyzed by GEPIA bioinformatics tool and RT-qPCR. Biological functions of SNHG14 in DLBCL were detected by CCK-8, colony formation, and transwell invasion assays. Molecular interaction was determined by RNA immunoprecipitation (RIP) and luciferase reporter assays. MiR-5590-3p-related pathway was identified through KEGG pathway analysis applying DAVID6.8 online bioinformatics tool. Effect of SNHG14 on CD8+ T cells was detected by flow cytometry. Results depicted that SNHG14 was upregulated in DLBCL and its depletion retarded proliferation, migration and epithelial-to-mesenchymal transition (EMT). Mechanistically, SNHG14 sponged miR-5590-3p to upregulate Zinc finger E-box binding homeobox 1 (ZEB1), and ZEB1 transcriptionally activated SNHG14 and PD-L1 to promote the immune evasion of DLBCL cells. In conclusion, we firstly showed that SNHG14/miR-5590-3p/ZEB1 positive feedback loop promoted diffuse large B cell lymphoma progression and immune evasion through regulating PD-1/PD-L1 checkpoint, indicating that targeting SNHG14 was a potential approach to improve the efficacy of immunotherapy in DLBCL.
Collapse
Affiliation(s)
- Lina Zhao
- Department of Hematology, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, China.
| | - Ye Liu
- Department of Immunology Teaching and Research, Harbin Medical University, Harbin, China
| | - Jingbo Zhang
- Department of Hematology, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Yan Liu
- Department of Hematology, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| | - Qi Qi
- Department of Hematology, The Tumor Hospital Affiliated to Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
195
|
Gastric Cancer in the Era of Immune Checkpoint Blockade. JOURNAL OF ONCOLOGY 2019; 2019:1079710. [PMID: 31662748 PMCID: PMC6778883 DOI: 10.1155/2019/1079710] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/22/2019] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is one of the most important malignancies worldwide because of its high incidence and mortality. The very low survival rates are mainly related to late diagnosis and limited treatment options. GC is the final clinical outcome of a stepwise process that starts with a chronic and sustained inflammatory reaction mounted in response to Helicobacter pylori infection. The bacterium modulates innate and adaptive immunity presumably as part of the strategies to survive, which favors the creation of an immunosuppressive microenvironment that ultimately facilitates GC progression. T-cell exhaustion, which is characterized by elevated expression of immune checkpoint (IC) proteins, is one of the most salient manifestations of immunosuppressive microenvironments. It has been consistently demonstrated that the tumor-immune microenvironment(TIME)‐exhausted phenotype can be reverted by blocking ICs with monoclonal antibodies. Although these therapies are associated with long-lasting response rates, only a subset of patients derive clinical benefit, which varies according to tumor site. The search for biomarkers to predict the response to IC inhibition is a matter of intense investigation as this may contribute to maximize disease control, reduce side effects, and minimize cost. The approval of pembrolizumab for its use in GC has rocketed immuno-oncology research in this cancer type. In this review, we summarize the current knowledge centered around the immune contexture and recent findings in connection with IC inhibition in GC.
Collapse
|
196
|
Xue W, Xu X, Tan Y, Qian Y, Wang H, Wang Y, Xu Y, Zhu X, Jiang P, Ding W. Evaluating and validating the predictive ability of preoperative systemic inflammatory/immune cells in gastric cancer following R0 resection. Oncol Lett 2019; 18:5205-5214. [PMID: 31612031 PMCID: PMC6781767 DOI: 10.3892/ol.2019.10867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 08/29/2019] [Indexed: 01/17/2023] Open
Abstract
The present study aimed to compare the predictive abilities of preoperative systemic inflammatory/immune cell ratios in gastric cancer (GC) following curative R0 resection, and to screen the optimal parameter incorporated into nomograms to predict the postoperative overall survival (OS) and recurrence-free survival (RFS). A total of 679 patients with GC were included in the study, divided into a primary cohort (300 cases), an internal validation cohort (278 cases), and an external validation cohort (101 cases). In the primary cohort, the prognostic abilities of all systemic inflammatory/immune cell accounts or ratios were compared by receiver operating characteristic (ROC) curve analysis. The area under the ROC curve (AUC) of the neutrophil-monocyte-lymphocyte ratio (NMLR) was largest for the prediction of OS (AUC=0.728) and RFS (AUC=0.695). The independent predictive factors for OS or RFS, including NMLR, degree of differentiation (DD), T-stage and N-stage were used to establish the 2 nomograms. The comprehensive predictive power of nomograms was compared with that of the tumor-nodes-metastasis (TNM) staging system and validated by bootstrap resampling. The concordance indexes (C-indexes) of the nomograms for OS [C-index, 0.851; 95% confidence interval (CI), 0.817-0.883] and RFS (C-index, 0.860; 95% CI, 0.831-0.889), were increased compared with those for the DD, the NMLR and the TNM stage. The AUCs of the 2 nomograms (0.933 for OS and 0.944 for RFS) were largest among all predictive scoring systems. In the internal validation cohort, the C-indexes of the nomograms for OS and RFS were 0.840 and 0.916, respectively. In the external validation cohort, the C-indexes of the nomograms for OS and RFS nomograms were 0.827 and 0.891, respectively. The present study demonstrated that the NMLR was an independent prognostic factor for patients with GC. The proposed nomograms were demonstrated to have a good predictive ability with improved sensitivity and accuracy in survival and recurrence in patients with GC undergoing R0 resection.
Collapse
Affiliation(s)
- Wenbo Xue
- Department of General Surgery, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu 213002, P.R. China
| | - Xuezhong Xu
- Department of General Surgery, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu 213002, P.R. China
| | - Yulin Tan
- Department of General Surgery, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu 213002, P.R. China
| | - Yan Qian
- Department of Respiration, Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou, Jiangsu 213164, P.R. China
| | - Hao Wang
- Department of Medical Records, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu 213002, P.R. China
| | - Yibo Wang
- Department of General Surgery, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu 213002, P.R. China
| | - Yixin Xu
- Department of General Surgery, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu 213002, P.R. China
| | - Xiaojun Zhu
- Department of General Surgery, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu 213002, P.R. China
| | - Peng Jiang
- Department of General Surgery, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu 213002, P.R. China
| | - Wei Ding
- Department of General Surgery, Wujin Hospital Affiliated to Jiangsu University, Changzhou, Jiangsu 213002, P.R. China
| |
Collapse
|
197
|
Hoogi S, Eisenberg V, Mayer S, Shamul A, Barliya T, Cohen CJ. A TIGIT-based chimeric co-stimulatory switch receptor improves T-cell anti-tumor function. J Immunother Cancer 2019; 7:243. [PMID: 31500665 PMCID: PMC6734436 DOI: 10.1186/s40425-019-0721-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 08/28/2019] [Indexed: 12/16/2022] Open
Abstract
Background Tumors can employ different mechanisms to evade immune surveillance and function. Overexpression of co-inhibitory ligands that bind to checkpoint molecules on the surface of T-cells can greatly impair the function of latter. TIGIT (T cell immunoreceptor with Ig and ITIM domains) is such a co-inhibitory receptor expressed by T and NK cells which, upon binding to its ligand (e.g., CD155), can diminish cytokine production and effector function. Additionally, the absence of positive co-stimulation at the tumor site can further dampen T-cell response. Methods As T-cell genetic engineering has become clinically-relevant in the recent years, we devised herein a strategy aimed at enhancing T-cell anti-tumor function by diverting T-cell coinhibitory signals into positive ones using a chimeric costimulatory switch receptor (CSR) composed of the TIGIT exodomain fused to the signaling domain of CD28. Results After selecting an optimized TIGIT-28 CSR, we co-transduced it along with tumor-specific TCR or CAR into human T-cells. TIGIT-28-equipped T-cells exhibited enhanced cytokine secretion and upregulation of activation markers upon co-culture with tumor cells. TIGIT-28 enhancing capability was also demonstrated in an original in vitro model of T-cell of hypofunction induction upon repetitive antigen exposure. Finally, we tested the function of this molecule in the context of a xenograft model of established human melanoma tumors and showed that TIGIT-28-engineered human T-cells demonstrated superior anti-tumor function. Conclusion Overall, we propose that TIGIT-based CSR can substantially enhance T-cell function and thus contribute to the improvement of engineered T cell-based immunotherapy. Electronic supplementary material The online version of this article (10.1186/s40425-019-0721-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shiran Hoogi
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, 52900-02, Ramat Gan, Israel
| | - Vasyl Eisenberg
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, 52900-02, Ramat Gan, Israel
| | - Shimrit Mayer
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, 52900-02, Ramat Gan, Israel
| | - Astar Shamul
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, 52900-02, Ramat Gan, Israel
| | - Tilda Barliya
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, 52900-02, Ramat Gan, Israel
| | - Cyrille J Cohen
- The Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, 52900-02, Ramat Gan, Israel.
| |
Collapse
|
198
|
Duan X, Liu J, Cui J, Ma B, Zhou Q, Yang X, Lu Z, Du Y, Su C. Expression of TIGIT/CD155 and correlations with clinical pathological features in human hepatocellular carcinoma. Mol Med Rep 2019; 20:3773-3781. [PMID: 31485637 PMCID: PMC6755146 DOI: 10.3892/mmr.2019.10641] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 06/25/2019] [Indexed: 12/16/2022] Open
Abstract
T cell immunoglobulin and ITIM domain (TIGIT) is a recently identified T cell coinhibitory receptor. Studies have shown that TIGIT is expressed in colon adenocarcinoma, uterine corpus endometrioid carcinoma, breast carcinoma and kidney renal clear cell carcinoma. However, the role of the TIGIT/human poliovirus receptor (CD155) pathway in the pathogenesis of hepatocellular carcinoma (HCC) remains to be elucidated. In the present study, the expression of TIGIT and CD155 in HCC tissues and peripheral blood were determined, and correlations among TIGIT, CD155, TIGIT+ CD4+ T cells, TIGIT+ regulatory T (Treg) cells and α-fetoprotein (AFP) were investigated in order to identify a potential target for diagnosing and treating HCC. Immunohistochemistry, reverse transcription-quantitative PCR analysis and western blotting were used to examine the expression of TIGIT and CD155 in cancerous tissues and peripheral blood collected from patients with HCC. The frequency of TIGIT+ CD4+ T cells and TIGIT+ Treg cells and the concentration of inflammatory cytokines secreted by T cell subsets were analyzed by flow cytometry and a Merck Milliplex assay. Correlations between the frequency of TIGIT+ CD4+ T and TIGIT+ Treg cells and AFP were analyzed using Spearman's rank correlation test. With the degree of cancerous differentiation from high to low, the expression levels of TIGIT and CD155 were upregulated in the cancerous tissues from patients with HCC. TIGIT+ CD4+ T cell and TIGIT+ Treg cell frequencies were decreased in peripheral blood from postoperative patients with HCC. The increased expression of TIGIT was positively correlated with the level of AFP. These results indicate that co-inhibitory receptor TIGIT may be involved in the pathogenesis of HCC and represent a novel target for the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Xiangguo Duan
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Juanxi Liu
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Jianjian Cui
- Department of Clinical Laboratory, Ningxia Chinese Medicine Research Center, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Bin Ma
- Department of Oncology Surgery, The First People's Hospital of Yinchuan, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Qiunan Zhou
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Xiaojuan Yang
- Department of Laboratory Medicine, College of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Zhenhui Lu
- Department of Laboratory Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Yong Du
- Department of Laboratory Surgery, General Hospital of Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| | - Chunxia Su
- Department of Pathogen Biology and Immunology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750004, P.R. China
| |
Collapse
|
199
|
Zhang J, Zhu Y, Wang Q, Kong Y, Sheng H, Guo J, Xu J, Dai B. Poliovirus receptor CD155 is up-regulated in muscle-invasive bladder cancer and predicts poor prognosis. Urol Oncol 2019; 38:41.e11-41.e18. [PMID: 31383549 DOI: 10.1016/j.urolonc.2019.07.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/06/2019] [Accepted: 07/11/2019] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the expression pattern of CD155 and evaluate the prognostic value of CD155 in muscle-invasive bladder cancer (MIBC). PATIENTS AND METHODS Immunohistochemical staining of CD155 and survival analysis were conducted on 228 nonmetastatic MIBC patients underwent radical cystectomy in cohorts from Fudan University Shanghai Cancer Center and Zhongshan Hospital. Association of CD155 gene expression with tumor stage and survival were analyzed in TCGA and GSE13507 dataset. RESULTS CD155 was significantly up-regulated in MIBC compared to matched normal urothelium and majorly stained on the membrane of tumor cells. In Fudan MIBC cohort, CD155 high expression was significantly correlated with shorter recurrence-free survival (HR = 2.13, P < 0.001) and overall survival (HR = 2.49, P < 0.001). CD155 expression, T stage, and lymph node status were independent factors for predicting survival in multivariate analysis. In TCGA dataset, CD155 high expression was independently associated with shorter overall survival (HR = 1.74, P = 0.001) beyond age, T stage, and lymph node status. Further, explorative analysis in Fudan MIBC cohort showed that adjuvant chemotherapy was associated with longer recurrence-free survival and overall survival in stage III and IV disease with CD155-high tumors. CONCLUSIONS These findings suggest that CD155 is a robust prognostic factor and may help predict the benefit of adjuvant chemotherapy in MIBC.
Collapse
Affiliation(s)
- Junyu Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qifeng Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yunyi Kong
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Haoyue Sheng
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jianming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Bo Dai
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
200
|
CD155 expression and its prognostic value in postoperative patients with breast cancer. Biomed Pharmacother 2019; 115:108884. [DOI: 10.1016/j.biopha.2019.108884] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/06/2019] [Accepted: 04/15/2019] [Indexed: 12/24/2022] Open
|