1
|
Zou Q, Liao K, Li G, Huang X, Zheng Y, Yang G, Luo M, Xue EY, Lan C, Wang S, Shen Y, Luo D, Ng DKP, Liu Q. Photo-metallo-immunotherapy: Fabricating Chromium-Based Nanocomposites to Enhance CAR-T Cell Infiltration and Cytotoxicity against Solid Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2407425. [PMID: 38899741 PMCID: PMC11733712 DOI: 10.1002/adma.202407425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Indexed: 06/21/2024]
Abstract
The infiltration and cytotoxicity of chimeric antigen receptor (CAR)-T cells are crucial for effective elimination of solid tumors. While metallo-immunotherapy is a promising strategy that can activate the antitumor immunity, its role in promoting CAR-T cell therapy remains elusive. The first single-element nanomaterial based on chromium nanoparticles (Cr NPs) for cancer photo-metallo-immunotherapy has been reported previously. Herein, an extended study using biodegradable polydopamine as a versatile carrier for these nanoparticles, enabling synergistic CAR-T cell therapy, is reported. The results show that these nanocomposites with or without further encapsulation of the anticancer drug alpelisib can promote the CAR-T cell migration and antitumor effect. Upon irradiation with near-infrared light, they caused mild hyperthermia that can "warm" the "cold" tumor microenvironment (TME). The administration of B7-H3 CAR-T cells to NOD severe combined immunodeficiency gamma mice bearing a human hepatoma or PIK3CA-mutated breast tumor can significantly inhibit the tumor growth after the induction of tumor hyperthermia by the nanocomposites and promote the secretion of serum cytokines, including IL-2, IFN-γ, and TNF-α. The trivalent Cr3+ ions, which are the major degradation product of these nanocomposites, can increase the CXCL13 and CCL3 chemokine expressions to generate tertiary lymphoid structures (TLSs) in the tumor tissues, facilitating the CAR-T cell infiltration.
Collapse
Affiliation(s)
- Qingshuang Zou
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Ke Liao
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
- Institute of Pharmacy and PharmacologySchool of Pharmaceutical ScienceHengyang Medical SchoolUniversity of South ChinaHengyangHunan421001China
| | - Guangchao Li
- Department of HematologyThe Affiliated Guangdong Second Provincial General Hospital of Jinan UniversityGuangzhou510317China
| | - Xin Huang
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Yongwei Zheng
- Research and Development DepartmentGuangzhou Bio‐Gene Technology Co. Ltd.Guangzhou510530China
| | - Gun Yang
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Min Luo
- Research and Development DepartmentGuangzhou Bio‐Gene Technology Co. Ltd.Guangzhou510530China
| | - Evelyn Y. Xue
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Chuanqing Lan
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Shuai Wang
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Yao Shen
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Dixian Luo
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| | - Dennis K. P. Ng
- Department of ChemistryThe Chinese University of Hong KongShatin, N.T.Hong Kong999077China
| | - Quan Liu
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)Shenzhen UniversityShenzhen518052China
| |
Collapse
|
2
|
Zhou X, Man M, Cui M, Zhou X, Hu Y, Liu Q, Deng Y. Relationship between EZH2 expression and prognosis of patients with hepatocellular carcinoma using a pathomics predictive model. Heliyon 2024; 10:e38562. [PMID: 39640777 PMCID: PMC11619983 DOI: 10.1016/j.heliyon.2024.e38562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/04/2024] [Accepted: 09/26/2024] [Indexed: 12/07/2024] Open
Abstract
Background Enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) is overexpressed in hepatocellular carcinoma, promoting tumorigenesis and correlating with poor prognosis. Traditional histopathological examinations are insufficient to accurately predict hepatocellular carcinoma (HCC) survival; however, pathomics models can predict EZH2 expression and HCC prognosis. This study aimed to investigate the relationship between pathomics features and EZH2 expression for predicting overall survival of patients with HCC. Methods We analyzed 267 patients with HCC from the Cancer Genome Atlas database, with available pathological images and gene expression data. RNA sequencing data were divided into high and low EZH2 expression groups for prognosis and survival analysis. Pathological image features were screened using mRMR_RFE. A pathological model was constructed using a gradient boosting machine (GBM) algorithm, and efficiency evaluation and survival analysis of the model were performed. The R package "survminer" took the pathomics score (PS) cutoff value of 0.4628 to divide the patients into two groups: high and low PS expression. Survival analyses included Kaplan-Meier curve analysis, univariate and multivariate Cox regression analyses, and interaction tests. Potential pathomechanisms were explored through enrichment, differential, immune cell infiltration abundance, and gene mutation analyses. Result EZH2 was highly expressed in tumor samples but poorly expressed in normal tissue samples. Univariate and multivariate Cox regression analyses revealed that EZH2 was an independent risk factor for HCC (hazard ratio [HR], 2.792 and 3.042, respectively). Seven imaging features were selected to construct a pathomics model to predict EZH2. Decision curve analysis showed that the model had high clinical utility. Multivariate Cox regression analysis showed that high PS expression was an independent risk factor for HCC prognosis (HR, 2.446). The Kaplan-Meier curve showed that high PS expression was a risk factor for overall survival. Conclusion EZH2 expression can affect the prognosis of patients with liver cancer. Our pathological model could predict EZH2 expression and prognosis of patients with HCC with high accuracy and robustness, making it a new and potentially valuable tool.
Collapse
Affiliation(s)
- Xulin Zhou
- Department of Oncology, Hefei BOE Hospital, Hefei, PR China
| | - Muran Man
- Department of Oncology, People's Hospital of Shizhong District, Zaozhuang City, Shandong Province, PR China
| | - Min Cui
- Affiliated Hospital Of Jining Medical University (Shanxian Central Hospital), Heze City, Shandong Province, PR China
| | - Xiang Zhou
- People's Hospital of Xinjiang Uygur Autonomous Region Urumqi, Xinjiang, CN, PR China
| | - Yan Hu
- Department of Oncology, Hefei BOE Hospital, Hefei, PR China
| | - Qinghua Liu
- Department of Oncology, Deyang People's Hospital, Deyang, Sichuan, CN, PR China
| | - Youxing Deng
- Department of Oncology, Hefei BOE Hospital, Hefei, PR China
| |
Collapse
|
3
|
Khan MN, Mao B, Hu J, Shi M, Wang S, Rehman AU, Li X. Tumor-associated macrophages and CD8+ T cells: dual players in the pathogenesis of HBV-related HCC. Front Immunol 2024; 15:1472430. [PMID: 39450177 PMCID: PMC11499146 DOI: 10.3389/fimmu.2024.1472430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
HBV infection is a key risk factor for the development and progression of hepatocellular carcinoma (HCC), a highly invasive tumor, and is characterized by its persistent immunosuppressive microenvironment. This review provides an in-depth analysis of HBV-related HCC and explores the interactions between neutrophils, natural killer cells, and dendritic cells, examining their roles in regulating tumor-associated macrophages and CD8+ T cells and shaping the tumor microenvironment. Two critical players in the immunosuppressive milieu of HBV-related HCC are CD8+ T cells and tumor-associated macrophages (TAMs). The study explores how TAMs, initially recruited to combat infection, transform, adopting a tumor-promoting phenotype, turning against the body, promoting tumor cell proliferation, suppressing anti-tumor immunity, and assisting in the spread of cancer. Meanwhile, CD8+ T cells, crucial for controlling HBV infection, become dysfunctional and exhausted in response to persistent chronic viral inflammation. The review then dissects how TAMs manipulate this immune response, further depleting CD8+ T cell functions through mechanisms like arginine deprivation and creating hypoxic environments that lead to exhaustion. Finally, it explores the challenges and promising therapeutic avenues that target TAMs and CD8+ T cells, either separately or in combination with antiviral therapy and personalized medicine approaches, offering hope for improved outcomes in HBV-related HCC.
Collapse
Affiliation(s)
- Muhammad Naveed Khan
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Western (Chongqing) Collaborative Innovation Center for Intelligent Diagnostics and Digital Medicine, Chongqing, China
| | - Binli Mao
- Department of Blood Transfusion, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Juan Hu
- Department of Clinical Laboratory Medicine, Suining Central Hospital, Suining, Sichuan, China
| | - Mengjia Shi
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shunyao Wang
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Adeel Ur Rehman
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaosong Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Western (Chongqing) Collaborative Innovation Center for Intelligent Diagnostics and Digital Medicine, Chongqing, China
| |
Collapse
|
4
|
Park R, Yu J, Shahzad M, Lee S, Ji JD. The immune regulatory function of B7-H3 in malignancy: spotlight on the IFN-STAT1 axis and regulation of tumor-associated macrophages. Immunol Res 2024; 72:526-537. [PMID: 38265550 DOI: 10.1007/s12026-024-09458-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/17/2024] [Indexed: 01/25/2024]
Abstract
B7-H3 is a member of the B7 superfamily and a putative inhibitory immune checkpoint molecule. Several early-phase clinical trials have reported promising anti-tumor activity and safety of anti-cancer drugs targeting B7-H3, suggesting that it may be a promising target for a potential next-generation immune checkpoint inhibitor. Despite ongoing clinical studies, most B7-H3-targeted drugs being currently investigated rely on direct cytotoxicity as their mechanisms of action rather than modulating its function as an immune checkpoint, at least in part due to its incompletely understood immune regulatory function. Recent studies have begun to elucidate the role of B7-H3 in regulating the tumor microenvironment (TME). Emerging evidence suggests that B7-H3 may regulate the interferon-STAT1 axis in the TME and promote immune suppression. Similarly, increasing evidence shows B7-H3 may be implicated in promoting M1 to M2 polarization of tumor-associated macrophages (TAMs). There is also accumulating evidence suggesting that B7-H3 may play a role in the heterotypic fusion of cancer stem cells and macrophages, thereby promoting tumor invasion and metastasis. Here, we review the recent advances in the understanding of B7-H3 cancer immunobiology with a focus on highlighting its potential role in the interferon priming of TAMs and the heterotypic fusion of TAMs with cancer stem cells and suggest future direction in elucidating its immune checkpoint function.
Collapse
Affiliation(s)
- Robin Park
- Department of Hematology/Oncology, Moffitt Cancer Center/University of South Florida, Tampa, FL, USA
| | - James Yu
- Department of Hematology/Oncology, Moffitt Cancer Center/University of South Florida, Tampa, FL, USA
| | - Moazzam Shahzad
- Department of Hematology/Oncology, Moffitt Cancer Center/University of South Florida, Tampa, FL, USA
| | - Sunggon Lee
- Department of Internal Medicine, Korea University, Seoul, South Korea
| | - Jong Dae Ji
- Department of Rheumatology, College of Medicine, Korea University, Seoul, South Korea.
| |
Collapse
|
5
|
Epperly R, Gottschalk S, DeRenzo C. CAR T cells redirected to B7-H3 for pediatric solid tumors: Current status and future perspectives. EJC PAEDIATRIC ONCOLOGY 2024; 3:100160. [PMID: 38957786 PMCID: PMC11218663 DOI: 10.1016/j.ejcped.2024.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Despite intensive therapies, pediatric patients with relapsed or refractory solid tumors have poor outcomes and need novel treatments. Immune therapies offer an alternative to conventional treatment options but require the identification of differentially expressed antigens to direct antitumor activity to sites of disease. B7-H3 (CD276) is an immune regulatory protein that is expressed in a range of malignancies and has limited expression in normal tissues. B7-H3 is highly expressed in pediatric solid tumors including osteosarcoma, rhabdomyosarcoma, Ewing sarcoma, Wilms tumor, neuroblastoma, and many rare tumors. In this article we review B7-H3-targeted chimeric antigen receptor (B7-H3-CAR) T cell therapies for pediatric solid tumors, reporting preclinical development strategies and outlining the landscape of active pediatric clinical trials. We identify challenges to the success of CAR T cell therapy for solid tumors including localizing to and penetrating solid tumor sites, evading the hostile tumor microenvironment, supporting T cell expansion and persistence, and avoiding intrinsic tumor resistance. We highlight strategies to overcome these challenges and enhance the effect of B7-H3-CAR T cells, including advanced CAR T cell design and incorporation of combination therapies.
Collapse
Affiliation(s)
- Rebecca Epperly
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Christopher DeRenzo
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
6
|
Sun H, Gao F, Liu Y, Shao J. Survival and clinicopathological significance of B7-H3 in bladder cancer: a systematic review and meta-analysis. BMC Urol 2024; 24:57. [PMID: 38468228 DOI: 10.1186/s12894-024-01446-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 03/01/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND B7-H3 has been implicated in clinical pathological features and prognosis across various cancer types, suggesting its potential as a cancer biomarker. Nevertheless, consensus remains elusive regarding its clinical-pathological and prognostic significance in bladder cancer. To address this gap, we conducted a systematic review and meta-analysis. METHODS We systematically searched PubMed, Embase, Web of Science, Cochrane, and CNKI databases from their inception up to October 6, 2022. We evaluated the literature's quality using the Newcastle-Ottawa Scale. We performed meta-analysis using Review Manager 5.3 and STATA 12.0, synthesizing data and calculating odds ratios (ORs) or hazard ratios (HRs) with corresponding 95% confidence intervals (CIs). RESULTS After applying eligibility criteria and conducting assessments, we included data from 8 studies, encompassing 1622 bladder cancer patients. Bladder tumor tissues exhibited significantly elevated B7-H3 protein expression compared to normal bladder tissues. Elevated B7-H3 expression was notably associated with patient age, tumor infiltration, and recurrence in bladder cancer. However, no significant correlations were observed with other clinical characteristics. Our pooled HR analysis indicated no significant association between B7-H3 expression and overall survival in bladder cancer patients. CONCLUSION Our meta-analysis unveils the complex role of B7-H3 in bladder cancer progression. It appears to be directly involved in tumor infiltration and recurrence but cannot definitively serve as a prognostic biomarker for bladder cancer. To validate these findings, further well-designed studies, encompassing larger sample sizes and diverse racial backgrounds, are warranted. PROSPERO REGISTRATION No. CRD42022364688.
Collapse
Affiliation(s)
- Haohao Sun
- Department of Urology, Wuxi No.2 People's Hospital (Jiangnan University Medical Center), Wuxi, 214002, China
- Department of Urology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Fei Gao
- Department of Urology, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Yuan Liu
- Department of General Surgery, Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214023, China
| | - Jianfeng Shao
- Department of Urology, Wuxi No.2 People's Hospital (Jiangnan University Medical Center), Wuxi, 214002, China.
| |
Collapse
|
7
|
Zhang D, Huang H, Gao X, Yu G, Zhang X, Jin H, Xu R, Wang Z, Zhang G. High expression of B7-H3 on monocyte/macrophages in tumor microenvironment promotes lung cancer progression by inhibiting apoptosis. Transl Oncol 2024; 41:101874. [PMID: 38262113 PMCID: PMC10832491 DOI: 10.1016/j.tranon.2023.101874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/03/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024] Open
Abstract
Monocyte/macrophages constitute a significant population of tumor-infiltrating immune cells and play a crucial role in tumor growth, invasion, and metastasis. B7-H3, has immune regulatory functions, however, it is unclear whether B7-H3 expressed on monocyte/macrophages plays a significance role in tumor progression. We found B7-H3 was high-expressed on monocyte/macrophages in tumor microenvironment compared with adjacent tissues in lung cancer, and its expression level was positively correlated with the number of monocyte/macrophages. Furthermore, the expression of B7-H3 was related to clinical stage and lymph node metastasis. Moreover, miR-29a-3p negatively regulated B7-H3, and the expression of B7-H3 on THP-1-derived macrophages was regulated by secreting exosomes containing miR-29a-3p. In addition, knockdown of B7-H3 promoted macrophage apoptosis under hypoxia. Mechanistically, B7-H3 enhanced the antiapoptotic ability of macrophage by up-regulating HIF-1ɑ via activating NF-κB. Taken together, these results imply that B7-H3 as a therapeutic target could hold promise for enhancing anti-tumor immune responses in individuals diagnosed with lung cancer.
Collapse
Affiliation(s)
- Dongze Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China
| | - Haitao Huang
- Department of Thoracic surgery, The First Affiliated Hospital of Soochow University, China
| | - Xin Gao
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, China
| | - Gehua Yu
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China
| | - Haiyan Jin
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China
| | - Ruyan Xu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China
| | - Zhenxin Wang
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, China.
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China.
| |
Collapse
|
8
|
Koumprentziotis IA, Theocharopoulos C, Foteinou D, Angeli E, Anastasopoulou A, Gogas H, Ziogas DC. New Emerging Targets in Cancer Immunotherapy: The Role of B7-H3. Vaccines (Basel) 2024; 12:54. [PMID: 38250867 PMCID: PMC10820813 DOI: 10.3390/vaccines12010054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Immune checkpoints (ICs) are molecules implicated in the fine-tuning of immune response via co-inhibitory or co-stimulatory signals, and serve to secure minimized host damage. Targeting ICs with various therapeutic modalities, including checkpoint inhibitors/monoclonal antibodies (mAbs), antibody-drug conjugates (ADCs), and CAR-T cells has produced remarkable results, especially in immunogenic tumors, setting a paradigm shift in cancer therapeutics through the incorporation of these IC-targeted treatments. However, the large proportion of subjects who experience primary or secondary resistance to available IC-targeted options necessitates further advancements that render immunotherapy beneficial for a larger patient pool with longer duration of response. B7-H3 (B7 Homolog 3 Protein, CD276) is a member of the B7 family of IC proteins that exerts pleiotropic immunomodulatory effects both in physiologic and pathologic contexts. Mounting evidence has demonstrated an aberrant expression of B7-H3 in various solid malignancies, including tumors less sensitive to current immunotherapeutic options, and has associated its expression with advanced disease, worse patient survival and impaired response to IC-based regimens. Anti-B7-H3 agents, including novel mAbs, bispecific antibodies, ADCs, CAR-T cells, and radioimmunotherapy agents, have exhibited encouraging antitumor activity in preclinical models and have recently entered clinical testing for several cancer types. In the present review, we concisely present the functional implications of B7-H3 and discuss the latest evidence regarding its prognostic significance and therapeutic potential in solid malignancies, with emphasis on anti-B7-H3 modalities that are currently evaluated in clinical trial settings. Better understanding of B7-H3 intricate interactions in the tumor microenvironment will expand the oncological utility of anti-B7-H3 agents and further shape their role in cancer therapeutics.
Collapse
|
9
|
Zhang X, Yu C, Zhao S, Wang M, Shang L, Zhou J, Ma Y. The role of tumor-associated macrophages in hepatocellular carcinoma progression: A narrative review. Cancer Med 2023; 12:22109-22129. [PMID: 38098217 PMCID: PMC10757104 DOI: 10.1002/cam4.6717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 11/04/2023] [Accepted: 11/07/2023] [Indexed: 12/31/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors in the world, with complex etiology and mechanism, and a high mortality rate. Tumor-associated macrophages (TAMs) are an important part of the HCC tumor microenvironment. Studies in recent years have shown that TAMs are involved in multiple stages of HCC and are related to treatment and prognosis in HCC. The specific mechanisms between TAMs and HCC are gradually being revealed. This paper reviews recent advances in the mechanisms associated with TAMs in HCC, concentrating on an overview of effects of TAMs on drug resistance in HCC and the signaling pathways linked with HCC, providing clues for the treatment and prognosis determination of HCC.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Chao Yu
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Siqi Zhao
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Min Wang
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Longcheng Shang
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Jin Zhou
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| | - Yong Ma
- Department of General Surgery, Nanjing First HospitalNanjing Medical UniversityNanjingChina
| |
Collapse
|
10
|
Chen S, Zhan S, Ding S, Zhang Q, Xuan H, Zhang X, Cao L. B7-H3 and CD47 co-expression in gastric cancer is a predictor of poor prognosis and potential targets for future dual-targeting immunotherapy. J Cancer Res Clin Oncol 2023; 149:16609-16621. [PMID: 37715830 DOI: 10.1007/s00432-023-05408-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND Gastric cancer (GC) is one of the most prevalent types of cancer worldwide. B7-H3, an immune checkpoint molecule with promising potential, has been found to be overexpressed in various cancers. CD47 is an anti-phagocytic molecule that interacts with the signal regulatory protein alpha (SIRPα) to affect phagocytes. The relationship between the expression of B7-H3 and CD47, two potential therapeutic targets found in tumor cells, remains unknown. In this study, our objective is to investigate the clinical significance of co-expression of B7-H3 and CD47, as well as the potential therapeutic value of combination therapy in GC. METHODS We utilized immunohistochemistry (IHC) to assess the expression of B7-H3, CD47, CD68, CD86 and CD163 in tissue microarrays obtained from 268 GC patients who underwent surgeries. Western blotting was employed to assess the protein level of B7-H3 and CD47 in GC tissues. The co-localization of B7-H3/CD47 and CD68 in GC tissues was determined using multiplex immunohistochemistry (m-IHC). We further verified the relationship between B7-H3/CD47 and macrophage infiltration via flow cytometry. To estimate the clinical outcomes of patients from different subgroups, we employed the Kaplan-Meier curve and the Cox model. RESULTS Among the 268 GC cases, a total of 180 cases exhibited positive expression of B7-H3, while 122 cases showed positive expression of CD47. In fresh GC clinical tissues, B7-H3 and CD47 protein level was also higher in tumor tissue than in adjacent normal tissue. Remarkably, 91 cases demonstrated co-expression of B7-H3 and CD47. We observed a significant correlation between B7-H3 expression and tumor stage (P = 0.001), differentiation (P = 0.045), and depth (P = 0.003). Additionally, there was a significant association between B7-H3 and CD47 expression (P = 0.018). The percentage of B7-H3 and CD47 double positive cells in fresh GC tumor tissues were elevated compared with control adjacent tissues regardless of CD45- or CD45+ cells (P = 0.0029, P = 0.0012). Patients with high B7-H3 or CD47 expression had significantly lower overall survival (OS) rates compared to those with low expression levels (P = 0.0176 or P = 0.0042). Surprisingly, patients with combined high expression of B7-H3 and CD47 exhibited a considerably worse prognosis than others (P = 0.0007). Univariate analysis revealed that cases with high expression of B7-H3, CD47, or both had significantly higher hazard ratios (HR) than cases with low expression of these markers. Furthermore, the results of multivariate analysis indicated that B7-H3/CD47 co-expression and CD47 expression alone are independent prognostic factors for overall survival. Moreover, significant correlations were observed between B7-H3 and CD68 expression, CD47 and CD68 expression, as well as B7-H3/CD47 co-expression and CD68 expression in GC patients (P < 0.001, P = 0.003, and P < 0.001). Flow cytometry test showed that the percentage of CD68-positive cells but not CD86-positive cells among B7-H3-positive or CD47-positive immune cells in GC tumor tissue was elevated significantly compared with adjacent tissue. CONCLUSION Our findings demonstrated a correlation between B7-H3 expression and CD47 expression in GC patient tissues. Co-expression of B7-H3 and CD47 can serve as an indicator of poor prognosis in GC patients. In GC tumor tissue, but not adjacent tissue, B7-H3 and CD47 expression was accompanied with macrophage infiltration.
Collapse
Affiliation(s)
- Siji Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China
| | - Shenghua Zhan
- Department of Pathology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, China
| | - Sisi Ding
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China
| | - Qiange Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China
| | - Hanqin Xuan
- Department of Pathology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, 215006, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China
| | - Lei Cao
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China.
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, 178 Ganjiang East Road, Suzhou, 215006, China.
| |
Collapse
|
11
|
Yuan Y, Wu D, Li J, Huang D, Zhao Y, Gao T, Zhuang Z, Cui Y, Zheng DY, Tang Y. Mechanisms of tumor-associated macrophages affecting the progression of hepatocellular carcinoma. Front Pharmacol 2023; 14:1217400. [PMID: 37663266 PMCID: PMC10470150 DOI: 10.3389/fphar.2023.1217400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 06/23/2023] [Indexed: 09/05/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are essential components of the immune cell stroma of hepatocellular carcinoma. TAMs originate from monocytic myeloid-derived suppressor cells, peripheral blood monocytes, and kupffer cells. The recruitment of monocytes to the HCC tumor microenvironment is facilitated by various factors, leading to their differentiation into TAMs with unique phenotypes. TAMs can directly activate or inhibit the nuclear factor-κB, interleukin-6/signal transducer and signal transducer and activator of transcription 3, Wnt/β-catenin, transforming growth factor-β1/bone morphogenetic protein, and extracellular signal-regulated kinase 1/2 signaling pathways in tumor cells and interact with other immune cells via producing cytokines and extracellular vesicles, thus affecting carcinoma cell proliferation, invasive and migratory, angiogenesis, liver fibrosis progression, and other processes to participate in different stages of tumor progression. In recent years, TAMs have received much attention as a prospective treatment target for HCC. This review describes the origin and characteristics of TAMs and their mechanism of action in the occurrence and development of HCC to offer a theoretical foundation for further clinical research of TAMs.
Collapse
Affiliation(s)
- Yi Yuan
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dailin Wu
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jing Li
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dan Huang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yan Zhao
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Tianqi Gao
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhenjie Zhuang
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ying Cui
- Department of Psychiatry, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Da-Yong Zheng
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Hepatology, TCM-Integrated Hospital of Southern Medical University, Guangzhou, Guangdong, China
- Department of Hepatopancreatobiliary, Cancer Center, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Tang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Mielcarska S, Dawidowicz M, Kula A, Kiczmer P, Skiba H, Krygier M, Chrabańska M, Piecuch J, Szrot M, Ochman B, Robotycka J, Strzałkowska B, Czuba Z, Waniczek D, Świętochowska E. B7H3 Role in Reshaping Immunosuppressive Landscape in MSI and MSS Colorectal Cancer Tumours. Cancers (Basel) 2023; 15:3136. [PMID: 37370746 DOI: 10.3390/cancers15123136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The study aimed to assess the expression of B7H3 concerning clinicopathological and histological parameters, including MSI/MSS status, CD-8 cells, tumour-infiltrating lymphocytes (TILs), budding, TNM scale and grading. Moreover, we analyzed the B7H3-related pathways using available online datasets and the immunological context of B7H3 expression, through the 48-cytokine screening panel of cancer tissues homogenates, immunogenic features and immune composition. The study included 158 patients diagnosed with CRC. To assess B7H3 levels, we performed an immunohistochemistry method (IHC) and enzyme-linked immunosorbent assay (ELISA). To elucidate the immune composition of colorectal cancer, we performed the Bio-Plex Pro Human 48-cytokine panel. To study biological characteristics of B7H3, we used online databases. Expression of B7H3 was upregulated in CRC tumour tissues in comparison to adjacent noncancerous margin tissues. The concentrations of B7H3 in tumours were positively associated with T parameter of patients and negatively with tumour-infiltrating lymphocytes score. Additionally, Principal Component Analysis showed that B7H3 expression in tumours correlated positively with cytokines associated with M2-macrophages and protumour growth factors. The expression of B7H3 in tumours was independent of MSI/MSS status. These findings will improve our understanding of B7H3 role in colorectal cancer immunity. Our study suggests that B7-H3 is a promising potential target for cancer therapy. Further studies must clarify the mechanisms of B7H3 overexpression and its therapeutic importance in colorectal cancer.
Collapse
Affiliation(s)
- Sylwia Mielcarska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland
| | - Miriam Dawidowicz
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland
| | - Agnieszka Kula
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland
| | - Paweł Kiczmer
- Department and Chair of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 13-15 3 Maja, 41-800 Zabrze, Poland
| | - Hanna Skiba
- Department and Chair of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 13-15 3 Maja, 41-800 Zabrze, Poland
| | - Małgorzata Krygier
- Department and Chair of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 13-15 3 Maja, 41-800 Zabrze, Poland
| | - Magdalena Chrabańska
- Department and Chair of Pathomorphology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 13-15 3 Maja, 41-800 Zabrze, Poland
| | - Jerzy Piecuch
- Department of General and Bariatric Surgery and Emergency Medicine in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10 Marii Curie-Skłodowskiej, 41-800 Zabrze, Poland
| | - Monika Szrot
- Department of General and Bariatric Surgery and Emergency Medicine in Zabrze, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 10 Marii Curie-Skłodowskiej, 41-800 Zabrze, Poland
| | - Błażej Ochman
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland
| | - Julia Robotycka
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland
| | - Bogumiła Strzałkowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland
| | - Zenon Czuba
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-808 Zabrze, Poland
| |
Collapse
|
13
|
Rasic P, Jeremic M, Jeremic R, Dusanovic Pjevic M, Rasic M, Djuricic SM, Milickovic M, Vukadin M, Mijovic T, Savic D. Targeting B7-H3-A Novel Strategy for the Design of Anticancer Agents for Extracranial Pediatric Solid Tumors Treatment. Molecules 2023; 28:molecules28083356. [PMID: 37110590 PMCID: PMC10145344 DOI: 10.3390/molecules28083356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/05/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Recent scientific data recognize the B7-H3 checkpoint molecule as a potential target for immunotherapy of pediatric solid tumors (PSTs). B7-H3 is highly expressed in extracranial PSTs such as neuroblastoma, rhabdomyosarcoma, nephroblastoma, osteosarcoma, and Ewing sarcoma, whereas its expression is absent or very low in normal tissues and organs. The influence of B7-H3 on the biological behavior of malignant solid neoplasms of childhood is expressed through different molecular mechanisms, including stimulation of immune evasion and tumor invasion, and cell-cycle disruption. It has been shown that B7-H3 knockdown decreased tumor cell proliferation and migration, suppressed tumor growth, and enhanced anti-tumor immune response in some pediatric solid cancers. Antibody-drug conjugates targeting B7-H3 exhibited profound anti-tumor effects against preclinical models of pediatric solid malignancies. Moreover, B7-H3-targeting chimeric antigen receptor (CAR)-T cells demonstrated significant in vivo activity against different xenograft models of neuroblastoma, Ewing sarcoma, and osteosarcoma. Finally, clinical studies demonstrated the potent anti-tumor activity of B7-H3-targeting antibody-radioimmunoconjugates in metastatic neuroblastoma. This review summarizes the established data from various PST-related studies, including in vitro, in vivo, and clinical research, and explains all the benefits and potential obstacles of targeting B7-H3 by novel immunotherapeutic agents designed to treat malignant extracranial solid tumors of childhood.
Collapse
Affiliation(s)
- Petar Rasic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
| | - Marija Jeremic
- Institute of Medical and Clinical Biochemistry, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Rada Jeremic
- Institute of Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Marija Dusanovic Pjevic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Milica Rasic
- Institute of Human Genetics, Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Slavisa M Djuricic
- Department of Clinical Pathology, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
- Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
| | - Maja Milickovic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Miroslav Vukadin
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
| | - Tanja Mijovic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
| | - Djordje Savic
- Department of Abdominal Surgery, Mother and Child Health Care Institute of Serbia "Dr. Vukan Cupic", 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| |
Collapse
|
14
|
Sanchez-Moral L, Paul T, Martori C, Font-Díaz J, Sanjurjo L, Aran G, Téllez É, Blanco J, Carrillo J, Ito M, Tuttolomondo M, Ditzel HJ, Fumagalli C, Tapia G, Sidorova J, Masnou H, Fernández-Sanmartín MA, Lozano JJ, Vilaplana C, Rodriguez-Cortés A, Armengol C, Valledor AF, Kremer L, Sarrias MR. Macrophage CD5L is a target for cancer immunotherapy. EBioMedicine 2023; 91:104555. [PMID: 37054630 PMCID: PMC10139961 DOI: 10.1016/j.ebiom.2023.104555] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND Reprogramming of immunosuppressive tumor-associated macrophages (TAMs) presents an attractive therapeutic strategy in cancer. The aim of this study was to explore the role of macrophage CD5L protein in TAM activity and assess its potential as a therapeutic target. METHODS Monoclonal antibodies (mAbs) against recombinant CD5L were raised by subcutaneous immunization of BALB/c mice. Peripheral blood monocytes were isolated from healthy donors and stimulated with IFN/LPS, IL4, IL10, and conditioned medium (CM) from different cancer cell lines in the presence of anti-CD5L mAb or controls. Subsequently, phenotypic markers, including CD5L, were quantified by flow cytometry, IF and RT-qPCR. Macrophage CD5L protein expression was studied in 55 human papillary lung adenocarcinoma (PAC) samples by IHC and IF. Anti-CD5L mAb and isotype control were administered intraperitoneally into a syngeneic Lewis Lung Carcinoma mouse model and tumor growth was measured. Tumor microenvironment (TME) changes were determined by flow cytometry, IHC, IF, Luminex, RNAseq and RT-qPCR. FINDINGS Cancer cell lines CM induced an immunosuppressive phenotype (increase in CD163, CD206, MERTK, VEGF and CD5L) in cultured macrophages. Accordingly, high TAM expression of CD5L in PAC was associated with poor patient outcome (Log-rank (Mantel-Cox) test p = 0.02). We raised a new anti-CD5L mAb that blocked the immunosuppressive phenotype of macrophages in vitro. Its administration in vivo inhibited tumor progression of lung cancer by altering the intratumoral myeloid cell population profile and CD4+ T-cell exhaustion phenotype, thereby significantly modifying the TME and increasing the inflammatory milieu. INTERPRETATION CD5L protein plays a key function in modulating the activity of macrophages and their interactions within the TME, which supports its role as a therapeutic target in cancer immunotherapy. FUNDING For a full list of funding bodies, please see the Acknowledgements.
Collapse
Affiliation(s)
- Lidia Sanchez-Moral
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Tony Paul
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Clara Martori
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain; Departament de Farmacologia, Terapèutica i Toxicologia, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Joan Font-Díaz
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona (IBUB), 08028 Barcelona, Spain
| | - Lucía Sanjurjo
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Gemma Aran
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Érica Téllez
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain
| | - Julià Blanco
- Virology and Cellular Immunology (VIC), IrsiCaixa, 08916 Badalona, Spain
| | - Jorge Carrillo
- Virology and Cellular Immunology (VIC), IrsiCaixa, 08916 Badalona, Spain
| | - Masaoki Ito
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 739-8527 Hiroshima, Japan
| | - Martina Tuttolomondo
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark
| | - Henrik J Ditzel
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; Department of Oncology, Odense University Hospital, 5220 Odense, Denmark
| | - Caterina Fumagalli
- Servicio de Anatomía Patológica, Hospital de la Santa Creu i Sant Pau, 08025, Barcelona, Spain
| | - Gustavo Tapia
- Pathology Department, Germans Trias i Pujol University Hospital (HUGTiP), 08916 Badalona, Spain
| | - Julia Sidorova
- Bioinformatics Platform, CIBERehd, 08036 Barcelona, Spain
| | - Helena Masnou
- Gastroenterology Department, Germans Trias i Pujol University Hospital (HUGTiP), 08916 Badalona, Spain; Network for Biomedical Research in Hepatic and Digestive Diseases (CIBERehd), 28029 Madrid, Spain
| | | | | | - Cristina Vilaplana
- Experimental Tuberculosis Unit, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain; Department of Genetics and Microbiology, Autonomous University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain; Microbiology Department, Laboratori Clínic Metropolitana Nord, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain
| | - Alhelí Rodriguez-Cortés
- Departament de Farmacologia, Terapèutica i Toxicologia, Facultat de Veterinària, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Carolina Armengol
- Network for Biomedical Research in Hepatic and Digestive Diseases (CIBERehd), 28029 Madrid, Spain; Childhood Liver Oncology Group, Program of Predictive and Personalized Medicine of Cancer (PMPCC), IGTP, 08916 Badalona, Spain
| | - Annabel F Valledor
- Department of Cell Biology, Physiology and Immunology, School of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona (IBUB), 08028 Barcelona, Spain
| | - Leonor Kremer
- Protein Tools Unit and Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain
| | - Maria-Rosa Sarrias
- Innate Immunity Group, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Spain; Network for Biomedical Research in Hepatic and Digestive Diseases (CIBERehd), 28029 Madrid, Spain.
| |
Collapse
|
15
|
Zhou Z, Yu X, Chen Y, Tan X, Liu W, Hua W, Chen L, Zhang W. Inhibition of the B7-H3 immune checkpoint limits hepatocellular carcinoma progression by enhancing T lymphocyte-mediated immune cytotoxicity in vitro and in vivo. Clin Transl Oncol 2023; 25:1067-1079. [PMID: 36512305 DOI: 10.1007/s12094-022-03013-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 11/08/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE The interaction between tumor cells and immune system in hepatocellular carcinoma (HCC) remains unclear. Great clinical achievements have progressed in HCC patients treated with immune checkpoint inhibitors (ICIs) for programmed death-1 and its ligands. However, response efficacy for these therapies is limited, thereby requiring alternative ICI candidates for HCC treatment. B7 homolog 3 protein (B7-H3), an immunoregulatory protein, plays a significant role in tumor immunity and disease progression. In this study, we evaluated the correlation between B7-H3 expression and prognosis of HCC patients, and investigated the therapeutic potential of B7-H3 targeting in HCC. METHODS B7-H3 expression was analyzed immunohistochemically in HCC patients, and its relationship with tumor-infiltrating lymphocyte infiltration was assessed. The anti-tumor efficacy of anti-B7-H3 antibody therapy was determined using an in vitro co-culture system and a subcutaneous HCC-bearing murine model. RESULTS We found that B7-H3 overexpressed in tumor cells and positively correlated with poor prognosis in HCC patients. B7-H3 inhibited the infiltration of CD8+ T cells in tumors. Furthermore, co-culture experiment indicated that inhibiting B7-H3 in tumor cells significantly increased T cells-mediated immune activities and tumor cell killing. Consistently, anti-B7-H3 antibody-treated HCC murine model showed decreased tumor size and enhanced anti-tumor immunity mediated by CD8+ T cells. CONCLUSION Altogether, our findings suggest that B7-H3 inhibition in tumor cells restores the immune cytotoxicity of T cells, which in turn promotes apoptosis of target cells. Therefore, B7-H3 serves as a key negative regulator in tumor immunity and the promising clinical utility of B7-H3-based immunotherapies for HCC treatment could be developed.
Collapse
Affiliation(s)
- Zixiong Zhou
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China
| | - Xijie Yu
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China
- Department of Pathology, Longyan First Hospital Affiliated to Fujian Medical University, Longyan, 364000, Fujian, China
| | - Yuqing Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China
- Diagnostic Pathology Center, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China
| | - Xiaodan Tan
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China
| | - Wenwen Liu
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China
- Diagnostic Pathology Center, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China
| | - Wenxi Hua
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China
- Diagnostic Pathology Center, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China
| | - Lihong Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China.
- Diagnostic Pathology Center, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China.
| | - Wenmin Zhang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China.
- Diagnostic Pathology Center, Fujian Medical University, 1 Xue Yuan Road, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
16
|
Muench MO, Fomin ME, Gutierrez AG, López-Terrada D, Gilfanova R, Nosworthy C, Beyer AI, Ostolaza G, Kats D, Matlock KL, Cairo S, Keller C. CD203c is expressed by human fetal hepatoblasts and distinguishes subsets of hepatoblastoma. Front Oncol 2023; 13:927852. [PMID: 36845728 PMCID: PMC9947649 DOI: 10.3389/fonc.2023.927852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 01/19/2023] [Indexed: 02/11/2023] Open
Abstract
Background & Aims Hepatocytic cells found during prenatal development have unique features compared to their adult counterparts, and are believed to be the precursors of pediatric hepatoblastoma. The cell-surface phenotype of hepatoblasts and hepatoblastoma cell lines was evaluated to discover new markers of these cells and gain insight into the development of hepatocytic cells and the phenotypes and origins of hepatoblastoma. Methods Human midgestation livers and four pediatric hepatoblastoma cell lines were screened using flow cytometry. Expression of over 300 antigens was evaluated on hepatoblasts defined by their expression of CD326 (EpCAM) and CD14. Also analyzed were hematopoietic cells, expressing CD45, and liver sinusoidal-endothelial cells (LSECs), expressing CD14 but lacking CD45 expression. Select antigens were further examined by fluorescence immunomicroscopy of fetal liver sections. Antigen expression was also confirmed on cultured cells by both methods. Gene expression analysis by liver cells, 6 hepatoblastoma cell lines, and hepatoblastoma cells was performed. Immunohistochemistry was used to evaluate CD203c, CD326, and cytokeratin-19 expression on three hepatoblastoma tumors. Results Antibody screening identified many cell surface markers commonly or divergently expressed by hematopoietic cells, LSECs, and hepatoblasts. Thirteen novel markers expressed on fetal hepatoblasts were identified including ectonucleotide pyrophosphatase/phosphodiesterase family member 3 (ENPP-3/CD203c), which was found to be expressed by hepatoblasts with widespread expression in the parenchyma of the fetal liver. In culture CD203c+CD326++ cells resembled hepatocytic cells with coexpression of albumin and cytokeratin-19 confirming a hepatoblast phenotype. CD203c expression declined rapidly in culture whereas the loss of CD326 was not as pronounced. CD203c and CD326 were co-expressed on a subset of hepatoblastoma cell lines and hepatoblastomas with an embryonal pattern. Conclusions CD203c is expressed on hepatoblasts and may play a role in purinergic signaling in the developing liver. Hepatoblastoma cell lines were found to consist of two broad phenotypes consisting of a cholangiocyte-like phenotype that expressed CD203c and CD326 and a hepatocyte-like phenotype with diminished expression of these markers. CD203c was expressed by some hepatoblastoma tumors and may represent a marker of a less differentiated embryonal component.
Collapse
Affiliation(s)
- Marcus O. Muench
- Vitalant Research Institute, San Francisco, CA, United States,Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States,*Correspondence: Marcus O. Muench,
| | - Marina E. Fomin
- Vitalant Research Institute, San Francisco, CA, United States
| | | | - Dolores López-Terrada
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States,Texas Children’s Cancer Center, Texas Children’s Hospital, Houston, TX, United States
| | | | | | - Ashley I. Beyer
- Vitalant Research Institute, San Francisco, CA, United States
| | | | - Dina Kats
- Pediatric Cancer Biology, Children’s Cancer Therapy Development Institute, Beaverton, OR, United States
| | | | - Stefano Cairo
- Research and Development Unit, XenTech, Evry, France
| | - Charles Keller
- Pediatric Cancer Biology, Children’s Cancer Therapy Development Institute, Beaverton, OR, United States
| |
Collapse
|
17
|
Qu W, Qiao S, Liu L, Chen Y, Peng C, Hou Y, Xu Z, Lv M, Wang T. Dectin3 protects against hepatocellular carcinoma by regulating glycolysis of macrophages. Int Immunopharmacol 2022; 113:109384. [PMID: 36461581 DOI: 10.1016/j.intimp.2022.109384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/10/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
|
18
|
Zhu S, Wang Y, Tang J, Cao M. Radiotherapy induced immunogenic cell death by remodeling tumor immune microenvironment. Front Immunol 2022; 13:1074477. [PMID: 36532071 PMCID: PMC9753984 DOI: 10.3389/fimmu.2022.1074477] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Emerging evidence indicates that the induction of radiotherapy(RT) on the immunogenic cell death (ICD) is not only dependent on its direct cytotoxic effect, changes in the tumor immune microenvironment also play an important role in it. Tumor immune microenvironment (TIME) refers to the immune microenvironment that tumor cells exist, including tumor cells, inflammatory cells, immune cells, various signaling molecules and extracellular matrix. TIME has a barrier effect on the anti-tumor function of immune cells, which can inhibit all stages of anti-tumor immune response. The remodeling of TIME caused by RT may affect the degree of immunogenicity, and make it change from immunosuppressive phenotype to immunostimulatory phenotype. It is of great significance to reveal the causes of immune escape of tumor cells, especially for the treatment of drug-resistant tumor. In this review, we focus on the effect of RT on the TIME, the mechanism of RT in reversing the TIME to suppress intrinsic immunity, and the sensitization effect of the remodeling of TIME caused by RT on the effectiveness of immunotherapy.
Collapse
|
19
|
Wang LT, Zeng QL, Jiang SL, Chen ZY, Wang XL, Li L, Li X. Ficolin-2: A potential immune-related therapeutic target with low expression in liver cancer. Front Oncol 2022; 12:987481. [PMID: 36425563 PMCID: PMC9679423 DOI: 10.3389/fonc.2022.987481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/04/2022] [Indexed: 11/09/2022] Open
Abstract
Objective This study aimed to investigate the role of ficolin-2 (FCN2) in the development and course of hepatocellular carcinoma (HCC) and to contribute to the evolution of innovative HCC therapeutics. Methods Oncomine, GEPIA (Gene Expression Profiling Interactive Analysis), TISIDB (Tumor Immune System Interactions and Drug Bank database), UALCAN (University of Alabama at Birmingham Cancer data analysis portal), UCSC (University of California, Santa Cruz), R package, the Kaplan–Meier technique, Cox regression analysis, LinkedOmics, Pearson’s correlation, and a nomogram were used to investigate the prognostic value of FCN2 in HCC. Co-expressed genes were screened. A protein–protein interaction network was created using the STRING database. Finally, immunohistochemistry was performed to establish the expression of FCN2 in HCC tissues. A pan-cancer study centered on HCC-related molecular analysis was also conducted to look for a link between FCN2 and immune infiltration, immune modulators, and chemokine receptors. Results In HCC tissues, the expression of FCN2 was observed to be lower than that in normal tissues. This was connected to the HCC marker alpha-fetoprotein, showing that FCN2 is involved in the development and progression of cancer. FCN2 may act through Staphylococcus aureus infection, lectins, and other pathways. Furthermore, at the immune level, the expression of FCN2 in HCC was associated with some immune cell infiltration, immunomodulators, and chemokine receptors. Conclusion FCN2 may be an immune checkpoint inhibitor for HCC, creating a breakthrough in the treatment of HCC.
Collapse
Affiliation(s)
- Li-ting Wang
- The First Clinical College of Guangxi Medical University, Nanning, China
| | - Qiu-ling Zeng
- The First Clinical College of Guangxi Medical University, Nanning, China
| | - Shao-lan Jiang
- The First Clinical College of Guangxi Medical University, Nanning, China
| | - Zhen-yu Chen
- The First Clinical College of Guangxi Medical University, Nanning, China
| | - Xiao-ling Wang
- The First Clinical College of Guangxi Medical University, Nanning, China
| | - Ling Li
- Department of Pathology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- *Correspondence: Ling Li, ; Xiaolong Li,
| | - Xiaolong Li
- Department of Cell Biology and Genetics, Key Laboratory of Longevity and Agingrelated Diseases of Chinese Ministry of Education, School of Pre-Clinical Medicine, Guangxi Medical University, Nanning, China
- *Correspondence: Ling Li, ; Xiaolong Li,
| |
Collapse
|
20
|
Zhao B, Li H, Xia Y, Wang Y, Wang Y, Shi Y, Xing H, Qu T, Wang Y, Ma W. Immune checkpoint of B7-H3 in cancer: from immunology to clinical immunotherapy. J Hematol Oncol 2022; 15:153. [PMID: 36284349 PMCID: PMC9597993 DOI: 10.1186/s13045-022-01364-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/30/2022] [Indexed: 11/28/2022] Open
Abstract
Immunotherapy for cancer is a rapidly developing treatment that modifies the immune system and enhances the antitumor immune response. B7-H3 (CD276), a member of the B7 family that plays an immunoregulatory role in the T cell response, has been highlighted as a novel potential target for cancer immunotherapy. B7-H3 has been shown to play an inhibitory role in T cell activation and proliferation, participate in tumor immune evasion and influence both the immune response and tumor behavior through different signaling pathways. B7-H3 expression has been found to be aberrantly upregulated in many different cancer types, and an association between B7-H3 expression and poor prognosis has been established. Immunotherapy targeting B7-H3 through different approaches has been developing rapidly, and many ongoing clinical trials are exploring the safety and efficacy profiles of these therapies in cancer. In this review, we summarize the emerging research on the function and underlying pathways of B7-H3, the expression and roles of B7-H3 in different cancer types, and the advances in B7-H3-targeted therapy. Considering different tumor microenvironment characteristics and results from preclinical models to clinical practice, the research indicates that B7-H3 is a promising target for future immunotherapy, which might eventually contribute to an improvement in cancer immunotherapy that will benefit patients.
Collapse
Affiliation(s)
- Binghao Zhao
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Huanzhang Li
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yu Xia
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yaning Wang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yuekun Wang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yixin Shi
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Hao Xing
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Tian Qu
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yu Wang
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| | - Wenbin Ma
- grid.506261.60000 0001 0706 7839Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730 People’s Republic of China ,grid.506261.60000 0001 0706 7839State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
21
|
Sung PS, Park DJ, Roh PR, Mun KD, Cho SW, Lee GW, Jung ES, Lee SH, Jang JW, Bae SH, Choi JY, Choi J, Ahn J, Yoon SK. Intrahepatic inflammatory IgA +PD-L1 high monocytes in hepatocellular carcinoma development and immunotherapy. J Immunother Cancer 2022; 10:e003618. [PMID: 35577505 PMCID: PMC9114848 DOI: 10.1136/jitc-2021-003618] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND IgA neutralizes pathogens to prevent infection at mucosal sites. However, emerging evidence shows that IgA contributes to aggravating inflammation or dismantling antitumor immunity in human diseased liver. The aim of this study was to elucidate the roles of inflammation-induced intrahepatic inflammatory IgA+ monocytes in the development of hepatocellular carcinoma (HCC). METHODS Patient cohorts including steatohepatitis cohort (n=61) and HCC cohort (n=271) were established. Patients' surgical and biopsy specimens were analyzed using immunohistochemistry. Multicolor flow cytometry was performed with a subset of patient samples. Single-cell RNA-Seq analysis was performed using Gene Expression Omnibus (GEO) datasets. Additionally, we performed in vitro differentiation of macrophages, stimulation with coated IgA, and RNA sequencing. Hepa1-6 cells and C57BL/6N mice were used to obtain HCC syngeneic mouse models. RESULTS Serum IgA levels were associated (p<0.001) with fibrosis progression and HCC development in patients with chronic liver diseases. Additionally, immunohistochemical staining of inflamed livers or HCC revealed IgA positivity in monocytes, with a correlation between IgA+ cell frequency and IgA serum levels. Compared with IgA- monocytes, intrahepatic IgA+ monocytes expressed higher levels of programmed death-ligand 1 (PD-L1) in inflamed livers and in HCC tumor microenvironment. Single-cell RNA sequencing using NCBI GEO database indicated an upregulation in inflammation-associated genes in the monocytes of patients whose plasma cell IGHA1 expression was greater than or equal to the median value. Bulk RNA sequencing demonstrated that in vitro stimulation of M2-polarized macrophages using coated IgA complex induced PD-L1 upregulation via YAP-mediated signaling. In vivo blockade of IgA signaling decreased the number of tumor-infiltrating IgA+PD-L1high macrophages and increased the number of CD69+CD8+ T cells to enhance antitumor effects in HCC mice models. CONCLUSIONS Overall, the findings of this study showed that serum IgA levels was correlated with intrahepatic and intratumoral infiltration of inflammatory IgA+PD-L1high monocytes in chronic liver diseases and HCC, providing potential therapeutic targets.
Collapse
Affiliation(s)
- Pil Soo Sung
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Dong Jun Park
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Pu Reun Roh
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Kyoung Do Mun
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Sung Woo Cho
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Gil Won Lee
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Eun Sun Jung
- Department of Hospital Pathology, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Jeong Won Jang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Si Hyun Bae
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Jong Young Choi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Jonghwan Choi
- Department of Computer Science, Yonsei University, Seoul, South Korea
| | - Jaegyoon Ahn
- Department of Computer Science & Engineering, Incheon National University, Incheon, South Korea
| | - Seung Kew Yoon
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
22
|
Microbiota-dependent activation of the myeloid calcineurin-NFAT pathway inhibits B7H3- and B7H4-dependent anti-tumor immunity in colorectal cancer. Immunity 2022; 55:701-717.e7. [PMID: 35364006 DOI: 10.1016/j.immuni.2022.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/07/2023]
Abstract
Bacterial sensing by intestinal tumor cells contributes to tumor growth through cell-intrinsic activation of the calcineurin-NFAT axis, but the role of this pathway in other intestinal cells remains unclear. Here, we found that myeloid-specific deletion of calcineurin in mice activated protective CD8+ T cell responses and inhibited colorectal cancer (CRC) growth. Microbial sensing by myeloid cells promoted calcineurin- and NFAT-dependent interleukin 6 (IL-6) release, expression of the co-inhibitory molecules B7H3 and B7H4 by tumor cells, and inhibition of CD8+ T cell-dependent anti-tumor immunity. Accordingly, targeting members of this pathway activated protective CD8+ T cell responses and inhibited primary and metastatic CRC growth. B7H3 and B7H4 were expressed by the majority of human primary CRCs and metastases, which was associated with low numbers of tumor-infiltrating CD8+ T cells and poor survival. Therefore, a microbiota-, calcineurin-, and B7H3/B7H4-dependent pathway controls anti-tumor immunity, revealing additional targets for immune checkpoint inhibition in microsatellite-stable CRC.
Collapse
|
23
|
Glennon EKK, Tongogara T, Primavera VI, Reeder SM, Wei L, Kaushansky A. Elucidating Spatially-Resolved Changes in Host Signaling During Plasmodium Liver-Stage Infection. Front Cell Infect Microbiol 2022; 11:804186. [PMID: 35111697 PMCID: PMC8801743 DOI: 10.3389/fcimb.2021.804186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022] Open
Abstract
Upon transmission to the human host, Plasmodium sporozoites exit the skin, are taken up by the blood stream, and then travel to the liver where they infect and significantly modify a single hepatocyte. Low infection rates within the liver have made proteomic studies of infected hepatocytes challenging, particularly in vivo, and existing studies have been largely unable to consider how protein and phosphoprotein differences are altered at different spatial locations within the heterogeneous liver. Using digital spatial profiling, we characterized changes in host signaling during Plasmodium yoelii infection in vivo without disrupting the liver tissue. Moreover, we measured alterations in protein expression around infected hepatocytes and identified a subset of CD163+ Kupffer cells that migrate towards infected cells during infection. These data offer the first insight into the heterogeneous microenvironment that surrounds the infected hepatocyte and provide insights into how the parasite may alter its milieu to influence its survival and modulate immunity.
Collapse
Affiliation(s)
- Elizabeth K. K. Glennon
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Tinotenda Tongogara
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Grinnell College, Grinnell, IA, United States
| | - Veronica I. Primavera
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Sophia M. Reeder
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Ling Wei
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
| | - Alexis Kaushansky
- Seattle Children’s Research Institute, Center for Global Infectious Disease Research, Seattle, WA, United States
- Department of Global Health, University of Washington, Seattle, WA, United States
- Department of Pediatrics, University of Washington, Seattle, WA, United States
- Brotman Baty Institute for Precision Medicine, Seattle, WA, United States
- *Correspondence: Alexis Kaushansky,
| |
Collapse
|
24
|
Immune suppressive checkpoint interactions in the tumour microenvironment of primary liver cancers. Br J Cancer 2022; 126:10-23. [PMID: 34400801 PMCID: PMC8727557 DOI: 10.1038/s41416-021-01453-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 05/05/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022] Open
Abstract
Liver cancer is one of the most prevalent cancers, and the third most common cause of cancer-related mortality worldwide. The therapeutic options for the main types of primary liver cancer-hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA)-are very limited. HCC and CCA are immunogenic cancers, but effective immune-mediated tumour control is prevented by their immunosuppressive tumour microenvironment. Despite the critical involvement of key co-inhibitory immune checkpoint interactions in immunosuppression in liver cancer, only a minority of patients with HCC respond to monotherapy using approved checkpoint inhibitor antibodies. To develop effective (combinatorial) therapeutic immune checkpoint strategies for liver cancer, in-depth knowledge of the different mechanisms that contribute to intratumoral immunosuppression is needed. Here, we review the co-inhibitory pathways that are known to suppress intratumoral T cells in HCC and CCA. We provide a detailed description of insights from preclinical studies in cellular crosstalk within the tumour microenvironment that results in interactions between co-inhibitory receptors on different T-cell subsets and their ligands on other cell types, including tumour cells. We suggest alternative immune checkpoints as promising targets, and draw attention to the possibility of combined targeting of co-inhibitory and co-stimulatory pathways to abrogate immunosuppression.
Collapse
|
25
|
Miyamoto T, Murakami R, Hamanishi J, Tanigaki K, Hosoe Y, Mise N, Takamatsu S, Mise Y, Ukita M, Taki M, Yamanoi K, Horikawa N, Abiko K, Yamaguchi K, Baba T, Matsumura N, Mandai M. B7-H3 Suppresses Antitumor Immunity via the CCL2-CCR2-M2 Macrophage Axis and Contributes to Ovarian Cancer Progression. Cancer Immunol Res 2022; 10:56-69. [PMID: 34799346 PMCID: PMC9414298 DOI: 10.1158/2326-6066.cir-21-0407] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/03/2021] [Accepted: 11/18/2021] [Indexed: 01/07/2023]
Abstract
New approaches beyond PD-1/PD-L1 inhibition are required to target the immunologically diverse tumor microenvironment (TME) in high-grade serous ovarian cancer (HGSOC). In this study, we explored the immunosuppressive effect of B7-H3 (CD276) via the CCL2-CCR2-M2 macrophage axis and its potential as a therapeutic target. Transcriptome analysis revealed that B7-H3 is highly expressed in PD-L1-low, nonimmunoreactive HGSOC tumors, and its expression negatively correlated with an IFNγ signature, which reflects the tumor immune reactivity. In syngeneic mouse models, B7-H3 (Cd276) knockout (KO) in tumor cells, but not in stromal cells, suppressed tumor progression, with a reduced number of M2 macrophages and an increased number of IFNγ+CD8+ T cells. CCL2 expression was downregulated in the B7-H3 KO tumor cell lines. Inhibition of the CCL2-CCR2 axis partly negated the effects of B7-H3 suppression on M2 macrophage migration and differentiation, and tumor progression. In patients with HGSOC, B7-H3 expression positively correlated with CCL2 expression and M2 macrophage abundance, and patients with B7-H3-high tumors had fewer tumoral IFNγ+CD8+ T cells and poorer prognosis than patients with B7-H3-low tumors. Thus, B7-H3 expression in tumor cells contributes to CCL2-CCR2-M2 macrophage axis-mediated immunosuppression and tumor progression. These findings provide new insights into the immunologic TME and could aid the development of new therapeutic approaches against the unfavorable HGSOC phenotype.
Collapse
Affiliation(s)
- Taito Miyamoto
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryusuke Murakami
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Gynecology, Shiga General Hospital, Moriyama, Japan.,Corresponding Author: Ryusuke Murakami, Department of Gynecology, Shiga General Hospital, 5-4-30, Moriyama, Moriyama City, Shiga 524-8524, Japan. Phone: 817-7582-5031; Fax: 817-7582-5931; E-mail:
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Yuko Hosoe
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nathan Mise
- Department of Environmental and Preventive Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Shiro Takamatsu
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuka Mise
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayo Ukita
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mana Taki
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koji Yamanoi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Naoki Horikawa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Obstetrics and Gynecology, Shizuoka General Hospital, Shizuoka, Japan
| | - Kaoru Abiko
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan.,Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Obstetrics and Gynecology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University School of Medicine, Higashiosaka, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
26
|
Feng R, Chen Y, Liu Y, Zhou Q, Zhang W. The role of B7-H3 in tumors and its potential in clinical application. Int Immunopharmacol 2021; 101:108153. [PMID: 34678689 DOI: 10.1016/j.intimp.2021.108153] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
B7-H3 (CD276 molecule) is an immune checkpoint from the B7 family of molecules that acts more as a co-inhibitory molecule to promote tumor progression. It is abnormally expressed on tumor cells and can be induced to express on antigen-presenting cells (APCs) including dendritic cells (DCs) and macrophages. In the tumor microenvironment (TME), B7-H3 promotes tumor progression by impairing T cell response, promoting the polarization of tumor-associated macrophages (TAMs) to M2, inhibiting the function of DCs, and promoting the migration and invasion of cancer-associated fibroblasts (CAFs). In addition, through non-immunological functions, B7-H3 promotes tumor cell proliferation, invasion, metastasis, resistance, angiogenesis, and metabolism, or in the form of exosomes to promote tumor progression. In this process, microRNAs can regulate the expression of B7-H3. B7-H3 may serve as a potential biomarker for tumor diagnosis and a marker of poor prognosis. Immunotherapy targeting B7-H3 and the combination of B7-H3 and other immune checkpoints have shown certain efficacy. In this review, we summarized the basic characteristics of B7-H3 and its mechanism to promote tumor progression by inducing immunosuppression and non-immunological functions, as well as the potential clinical applications of B7-H3 and immunotherapy based on B7-H3.
Collapse
Affiliation(s)
- Ranran Feng
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Department of Andrology, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Yong Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ying Liu
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qing Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wenling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
27
|
Lavoie RR, Gargollo PC, Ahmed ME, Kim Y, Baer E, Phelps DA, Charlesworth CM, Madden BJ, Wang L, Houghton PJ, Cheville J, Dong H, Granberg CF, Lucien F. Surfaceome Profiling of Rhabdomyosarcoma Reveals B7-H3 as a Mediator of Immune Evasion. Cancers (Basel) 2021; 13:cancers13184528. [PMID: 34572755 PMCID: PMC8466404 DOI: 10.3390/cancers13184528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/10/2021] [Accepted: 08/17/2021] [Indexed: 12/18/2022] Open
Abstract
Novel therapeutic strategies are needed for the treatment of rhabdomyosarcoma (RMS), the most common soft-tissue sarcoma in children. By using a combination of cell surface proteomics and transcriptomic profiling of RMS and normal muscle, we generated a catalog of targetable cell surface proteins enriched in RMS tumors. Among the top candidates, we identified B7-H3 as the major immunoregulatory molecule expressed by RMS tumors. By using a large cohort of tissue specimens, we demonstrated that B7-H3 is expressed in a majority of RMS tumors while not detected in normal human tissues. Through a deconvolution analysis of the RMS tumor RNA-seq data, we showed that B7-H3-rich tumors are enriched in macrophages M1, NK cells, and depleted in CD8+-T cells. Furthermore, in vitro functional assays showed that B7-H3 knockout in RMS tumor cells increases T-cell mediated cytotoxicity. Altogether, our study uncovers new potential targets for the treatment of RMS and provides the first biological insights into the role of B7-H3 in RMS biology, paving the way for the development of next-generation immunotherapies.
Collapse
Affiliation(s)
- Roxane R. Lavoie
- Department of Urology, Mayo Clinic, Rochester, MN 55902, USA; (R.R.L.); (P.C.G.); (M.E.A.); (Y.K.); (E.B.); (H.D.); (C.F.G.)
| | - Patricio C. Gargollo
- Department of Urology, Mayo Clinic, Rochester, MN 55902, USA; (R.R.L.); (P.C.G.); (M.E.A.); (Y.K.); (E.B.); (H.D.); (C.F.G.)
| | - Mohamed E. Ahmed
- Department of Urology, Mayo Clinic, Rochester, MN 55902, USA; (R.R.L.); (P.C.G.); (M.E.A.); (Y.K.); (E.B.); (H.D.); (C.F.G.)
| | - Yohan Kim
- Department of Urology, Mayo Clinic, Rochester, MN 55902, USA; (R.R.L.); (P.C.G.); (M.E.A.); (Y.K.); (E.B.); (H.D.); (C.F.G.)
| | - Emily Baer
- Department of Urology, Mayo Clinic, Rochester, MN 55902, USA; (R.R.L.); (P.C.G.); (M.E.A.); (Y.K.); (E.B.); (H.D.); (C.F.G.)
| | - Doris A. Phelps
- Greehey Children’s Cancer Research Institute, San Antonio, TX 78229, USA; (D.A.P.); (P.J.H.)
| | | | - Benjamin J. Madden
- Proteomic Core, Mayo Clinic, Rochester, MN 55902, USA; (C.M.C.); (B.J.M.)
| | - Liguo Wang
- Division of Computational Biology, Mayo Clinic, Rochester, MN 55902, USA;
| | - Peter J. Houghton
- Greehey Children’s Cancer Research Institute, San Antonio, TX 78229, USA; (D.A.P.); (P.J.H.)
| | - John Cheville
- Department of Anatomic Pathology, Mayo Clinic, Rochester, MN 55902, USA;
| | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN 55902, USA; (R.R.L.); (P.C.G.); (M.E.A.); (Y.K.); (E.B.); (H.D.); (C.F.G.)
- Department of Immunology, Mayo Clinic, Rochester, MN 55902, USA
| | - Candace F. Granberg
- Department of Urology, Mayo Clinic, Rochester, MN 55902, USA; (R.R.L.); (P.C.G.); (M.E.A.); (Y.K.); (E.B.); (H.D.); (C.F.G.)
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, MN 55902, USA; (R.R.L.); (P.C.G.); (M.E.A.); (Y.K.); (E.B.); (H.D.); (C.F.G.)
- Correspondence:
| |
Collapse
|
28
|
Wang J, Chen X, Xie C, Sun M, Hu C, Zhang Z, Luan L, Zhou J, Zhou J, Zhu X, Ouyang J, Dong X, Li D, Zhang J, Zhao X. MicroRNA miR-29a Inhibits Colon Cancer Progression by Downregulating B7-H3 Expression: Potential Molecular Targets for Colon Cancer Therapy. Mol Biotechnol 2021; 63:849-861. [PMID: 34100183 DOI: 10.1007/s12033-021-00348-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/01/2021] [Indexed: 12/11/2022]
Abstract
MiR-29a belongs to one of the subtypes of miRNAs known as non-coding single-stranded RNAs and is preferentially expressed in normal tissues. B7-H3, a member of the B7/CD28 immunoglobulin superfamily, was shown to be overexpressed in several solid malignant tumors, including colon cancer. In addition, it is associated with tumor progression and poor prognosis. We used immunohistochemical and Western blotting to assess B7-H3 protein expression levels in colon cancer and adjacent normal tissues and then compared their relationships with clinicopathological factors. Quantitative real-time reverse-transcription PCR was used to assess B7-H3 and miRNA-29a mRNA expression levels, and then their relationship and clinical significance were evaluated. In addition, colon cancer Caco-2 cells, which constitutively overexpress B7-H3, were transfected with lentivirus particles for miR-29a upregulation. Invasion and migration assays were carried out in vitro along with the establishment of a subcutaneous xenograft model in vivo to determine the role of miRNA-29a in colon cancer progression. The B7-H3 protein showed elevated expression in colon carcinoma and was relevant to TNM staging, lymph node metastasis, and reduced survival. Meanwhile, miR-29a was preferentially expressed in normal colon tissues, while B7-H3 transcript levels had no marked differences between tumor and normal tissue specimens. In vitro, miR-29a upregulation resulted in reduced B7-H3 expression. Furthermore, miR-29a upregulation reduced the invasive and migratory abilities of colon carcinoma cells. In animal models, upregulation of miR-29a slowed down the growth of subcutaneous xenotransplanted tumors and resulted in prolonged survival time. MiR-29a downregulates B7-H3 expression and accordingly inhibits colon cancer progression, invasion, and migration, indicating miR-29a and B7-H3 might represent novel molecular targets for advanced immunotherapy in colon cancer.
Collapse
Affiliation(s)
- Jin Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of General Surgery, Suzhou Dushu Lake Hospital (Dushu Lake Hospital Affiliated to Soochow University), Suzhou, China
- Jiangsu Institute of Clinical Immunology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaojuan Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chen Xie
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Mingbing Sun
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chenrui Hu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhe Zhang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Lipeng Luan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinguo Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Ouyang
- Department of Urology Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaoqiang Dong
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dechun Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianglei Zhang
- Department of Urology Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Xin Zhao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Jiangsu Institute of Clinical Immunology, Soochow University, Suzhou, China.
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
29
|
CD276 is an important player in macrophage recruitment into the tumor and an upstream regulator for PAI-1. Sci Rep 2021; 11:14849. [PMID: 34290311 PMCID: PMC8295264 DOI: 10.1038/s41598-021-94360-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/07/2021] [Indexed: 12/16/2022] Open
Abstract
More than 70% of colorectal, prostate, ovarian, pancreatic and breast cancer specimens show expression of CD276 (B7–H3), a potential immune checkpoint family member. Several studies have shown that high CD276 expression in cancer cells correlates with a poor clinical prognosis. This has been associated with the presence of lower tumor infiltrating leukocytes. Among those, tumor-associated macrophages can comprise up to 50% of the tumor mass and are thought to support tumor growth through various mechanisms. However, a lack of information on CD276 function and interaction partner(s) impedes rigorous evaluation of CD276 as a therapeutic target in oncology. Therefore, we aimed to understand the relevance of CD276 in tumor-macrophage interaction by employing a 3D spheroid coculture system with human cells. Our data show a role for tumor-expressed CD276 on the macrophage recruitment into the tumor spheroid, and also in regulation of the extracellular matrix modulator PAI-1. Furthermore, our experiments focusing on macrophage-expressed CD276 suggest that the antibody-dependent CD276 engagement triggers predominantly inhibitory signaling networks in human macrophages.
Collapse
|
30
|
Granito A, Marinelli S, Forgione A, Renzulli M, Benevento F, Piscaglia F, Tovoli F. Regorafenib Combined with Other Systemic Therapies: Exploring Promising Therapeutic Combinations in HCC. J Hepatocell Carcinoma 2021; 8:477-492. [PMID: 34079777 PMCID: PMC8165211 DOI: 10.2147/jhc.s251729] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/25/2021] [Indexed: 02/05/2023] Open
Abstract
Regorafenib was the first drug to demonstrate a survival benefit as a second-line agent after sorafenib failure in patients with unresectable hepatocellular carcinoma (HCC). Recent studies have shown that its mechanism of action is not only limited to its very broad spectrum of inhibition of angiogenesis, tumor proliferation, spread, and metastasis, but also to its immunomodulatory properties that have favorable effects on the very intricate role that the tumor microenvironment plays in carcinogenesis and tumor growth. In this review, we discuss rationale and evidence supporting regorafenib efficacy in HCC and that led to its approval as a second-line treatment, after sorafenib failure. We also discuss the evidence from clinical practice studies that confirm the results previously achieved in clinical trials. Finally, we analyze the potential role of regorafenib in emerging combined treatment approach with immunotherapy strategies using immune checkpoint blockade and its potential extension to patient categories not included in the registrative study.
Collapse
Affiliation(s)
- Alessandro Granito
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Sara Marinelli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Antonella Forgione
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Matteo Renzulli
- Radiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Benevento
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Fabio Piscaglia
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesco Tovoli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
31
|
Park DJ, Sung PS, Lee GW, Cho SW, Kim SM, Kang BY, Hur WH, Yang H, Lee SK, Lee SH, Jung ES, Seo CH, Ahn J, Choi HJ, You YK, Jang JW, Bae SH, Choi JY, Yoon SK. Preferential Expression of Programmed Death Ligand 1 Protein in Tumor-Associated Macrophages and Its Potential Role in Immunotherapy for Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:4710. [PMID: 33946835 PMCID: PMC8124544 DOI: 10.3390/ijms22094710] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/23/2021] [Accepted: 04/25/2021] [Indexed: 02/07/2023] Open
Abstract
A predictive biomarker of immune checkpoint inhibitor (ICI)-based treatments in hepatocellular carcinoma (HCC) has not been clearly demonstrated. In this study, we focused on the infiltration and programmed death ligand 1 (PD-L1) expression of tumor-associated macrophages (TAMs) in the tumor microenvironment of HCC. Immunohistochemistry demonstrated that PD-L1 was preferentially expressed on CD68+ macrophages in the tumor microenvironment of HCC, suggestive of its expression in TAMs rather than in T cells or tumor cells (P < 0.05). A co-culture experiment using activated T cells and M2 macrophages confirmed a significant increase in T cell functionality after the pretreatment of M2 macrophages with anti-PD-L1. Syngeneic mouse model experiments demonstrated that TAMs expressed PD-L1 and tumors treated with anti-PD-L1 showed smaller diameters than those treated with IgG. In these mice, anti-PD-L1 treatment increased activation markers in intratumoral CD8+ T cells and reduced the size of the TAM population. Regarding nivolumab-treated patients, three of eight patients responded to the anti-PD-1 treatment. The percentage of Ki-67-positive CD4+ and CD8+ T cells was higher in responders than non-responders after nivolumab. Overall, PD-L1 expression on TAMs may be targeted by immune-based HCC treatment, and ICI treatment results in the reinvigoration of exhausted CD8+ T cells in HCC.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/biosynthesis
- B7-H1 Antigen/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Coculture Techniques
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Immunotherapy/methods
- Ki-67 Antigen/biosynthesis
- Ki-67 Antigen/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/therapy
- Liver Neoplasms, Experimental/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice
- Mice, Inbred C57BL
- Molecular Targeted Therapy/methods
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Nivolumab/pharmacology
- Nivolumab/therapeutic use
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Tumor Cells, Cultured
- Tumor Microenvironment/immunology
- Tumor-Associated Macrophages/drug effects
- Tumor-Associated Macrophages/metabolism
Collapse
Affiliation(s)
- Dong-Jun Park
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
| | - Pil-Soo Sung
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
- Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea
| | - Gil-Won Lee
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
| | - Sung-Woo Cho
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
| | - Sung-Min Kim
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
| | - Byung-Yoon Kang
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
| | - Won-Hee Hur
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
| | - Hyun Yang
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
- Department of Internal Medicine, College of Medicine, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul 03383, Korea
| | - Soon-Kyu Lee
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
- Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea
| | - Sung-Hak Lee
- Department of Clinical Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Eun-Sun Jung
- Department of Hospital Pathology, College of Medicine, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul 03383, Korea;
| | - Chang-Ho Seo
- Department of Surgery, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea; (C.-H.S.); (J.A.); (H.-J.C.); (Y.-K.Y.)
| | - Joseph Ahn
- Department of Surgery, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea; (C.-H.S.); (J.A.); (H.-J.C.); (Y.-K.Y.)
| | - Ho-Joong Choi
- Department of Surgery, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea; (C.-H.S.); (J.A.); (H.-J.C.); (Y.-K.Y.)
| | - Young-Kyoung You
- Department of Surgery, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea; (C.-H.S.); (J.A.); (H.-J.C.); (Y.-K.Y.)
| | - Jeong-Won Jang
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
- Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea
| | - Si-Hyun Bae
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
- Department of Internal Medicine, College of Medicine, Eunpyeong St. Mary’s Hospital, The Catholic University of Korea, Seoul 03383, Korea
| | - Jong-Young Choi
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
- Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea
| | - Seung-Kew Yoon
- The Catholic University Liver Research Center, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; (D.-J.P.); (P.-S.S.); (G.-W.L.); (S.-W.C.); (S.-M.K.); (B.-Y.K.); (W.-H.H.); (H.Y.); (S.-K.L.); (J.-W.J.); (S.-H.B.); (J.-Y.C.)
- Department of Internal Medicine, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
32
|
Sakr MA, Mohamed KAH, Hussein AM, Fouad MH, Allam AS, Safwat E. Diagnostic and prognostic value of serum soluble CD163 in cirrhotic patients with hepatitis C virus-related hepatocellular carcinoma before and after locoregional therapy. EGYPTIAN LIVER JOURNAL 2021. [DOI: 10.1186/s43066-021-00090-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Abstract
Background
Tumor-associated macrophages (TAMs), inflammatory cells in tumor microenvironment, are crucial for the tumor occurrence and progression which in turn increase the expression of soluble CD163 (sCD163). Nevertheless, not much has been established regarding sCD163 and its connection to HCC diagnosis and prognosis. This study was conducted to evaluate the diagnostic and prognostic role of sCD163 in patients with HCC on top of HCV-related liver cirrhosis. Forty adult patients with HCV-related liver cirrhosis and HCC (HCC group) were randomly selected and subjected to locoregional therapies, either transarterial chemoembolization (TACE) or radiofrequency ablation (RFA). Four patients were excluded because of portal vein invasion. Another group of 20 patients with liver cirrhosis only served as controls (LC group). Routine laboratory studies and abdominal ultrasound were done for all. Alpha-fetoprotein (AFP) and sCD163 were measured twice, at baseline and 1-month post-intervention, using a commercially available enzyme-linked immunosorbent assay kit.
Results
At baseline, sCD163 showed an insignificant higher value in HCC group (p > 0.05). The best cutoff value for sCD163 and AFP was 6.2 mg/L and 195 ng/mL, respectively. AFP had a larger area under the curve (0.88 vs. 0.767). An overall significant decline was seen in sCD163 after treatment (6.5±1.5 to 3.1±2.5 mg/L; p < 0.001), while AFP showed an insignificant decrease (p > 0.05). Also, sCD163 decreased significantly in the eradicated cases (6.1±1.4 mg/L before intervention vs. 2.3±1.4 mg/L after intervention, p < 0.01), while there was a significant increase in the recurrent cases (8.4±0.4 mg/L before intervention vs. 10.3±1.6 after intervention; p < 0.05). Moreover, sCD163 showed a significant difference in its pre-intervention and post-intervention values between recurrent and eradicated HCC cases (p < 0.01).
Conclusions
It is concluded that sCD163 has a minor role as a diagnostic marker for HCC, yet it could be used as a good prognostic marker in predicting the tumor response to locoregional therapies.
Collapse
|
33
|
Mattsson J, Ekdahl L, Junghus F, Ajore R, Erlandsson E, Niroula A, Pertesi M, Frendéus B, Teige I, Nilsson B. Accelerating target deconvolution for therapeutic antibody candidates using highly parallelized genome editing. Nat Commun 2021; 12:1277. [PMID: 33627649 PMCID: PMC7904777 DOI: 10.1038/s41467-021-21518-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/26/2021] [Indexed: 12/26/2022] Open
Abstract
Therapeutic antibodies are transforming the treatment of cancer and autoimmune diseases. Today, a key challenge is finding antibodies against new targets. Phenotypic discovery promises to achieve this by enabling discovery of antibodies with therapeutic potential without specifying the molecular target a priori. Yet, deconvoluting the targets of phenotypically discovered antibodies remains a bottleneck; efficient deconvolution methods are needed for phenotypic discovery to reach its full potential. Here, we report a comprehensive investigation of a target deconvolution approach based on pooled CRISPR/Cas9. Applying this approach within three real-world phenotypic discovery programs, we rapidly deconvolute the targets of 38 of 39 test antibodies (97%), a success rate far higher than with existing approaches. Moreover, the approach scales well, requires much less work, and robustly identifies antibodies against the major histocompatibility complex. Our data establish CRISPR/Cas9 as a highly efficient target deconvolution approach, with immediate implications for the development of antibody-based drugs. Efficient deconvolution of antibody targets is needed for phenotype-based discovery. Here, the authors investigate a deconvolution approach based on pooled CRISPR Cas9 to achieve 97% deconvolution success rate.
Collapse
Affiliation(s)
- Jenny Mattsson
- Department of Laboratory Medicine, Hematology and Transfusion Medicine, Lund, Sweden.,BioInvent International AB, Ideongatan 1, Lund, Sweden
| | - Ludvig Ekdahl
- Department of Laboratory Medicine, Hematology and Transfusion Medicine, Lund, Sweden
| | - Fredrik Junghus
- Department of Laboratory Medicine, Hematology and Transfusion Medicine, Lund, Sweden
| | - Ram Ajore
- Department of Laboratory Medicine, Hematology and Transfusion Medicine, Lund, Sweden
| | - Eva Erlandsson
- Department of Laboratory Medicine, Hematology and Transfusion Medicine, Lund, Sweden
| | - Abhishek Niroula
- Department of Laboratory Medicine, Hematology and Transfusion Medicine, Lund, Sweden.,Broad Institute, 415 Main Street, Cambridge, MA, USA
| | - Maroulio Pertesi
- Department of Laboratory Medicine, Hematology and Transfusion Medicine, Lund, Sweden
| | | | - Ingrid Teige
- BioInvent International AB, Ideongatan 1, Lund, Sweden
| | - Björn Nilsson
- Department of Laboratory Medicine, Hematology and Transfusion Medicine, Lund, Sweden. .,Broad Institute, 415 Main Street, Cambridge, MA, USA.
| |
Collapse
|
34
|
Wei J, Chen P, Gupta P, Ott M, Zamler D, Kassab C, Bhat KP, Curran MA, de Groot JF, Heimberger AB. Immune biology of glioma-associated macrophages and microglia: functional and therapeutic implications. Neuro Oncol 2021; 22:180-194. [PMID: 31679017 DOI: 10.1093/neuonc/noz212] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
CNS immune defenses are marshaled and dominated by brain resident macrophages and microglia, which are the innate immune sentinels and frontline host immune barriers against various pathogenic insults. These myeloid lineage cells are the predominant immune population in gliomas and can constitute up to 30-50% of the total cellular composition. Parenchymal microglial cells and recruited monocyte-derived macrophages from the periphery exhibit disease-specific phenotypic characteristics with spatial and temporal distinctions and are heterogeneous subpopulations based on their molecular signatures. A preponderance of myeloid over lymphoid lineage cells during CNS inflammation, including gliomas, is a contrasting feature of brain immunity relative to peripheral immunity. Herein we discuss glioma-associated macrophage and microglia immune biology in the context of their identity, molecular drivers of recruitment, nomenclature and functional paradoxes, therapeutic reprogramming and polarization strategies, relevant challenges, and our perspectives on therapeutic modulation.
Collapse
Affiliation(s)
- Jun Wei
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Peiwen Chen
- Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Pravesh Gupta
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Martina Ott
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Zamler
- Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cynthia Kassab
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Krishna P Bhat
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael A Curran
- Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - John F de Groot
- Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amy B Heimberger
- Departments of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
35
|
Jiang Y, Han Q, Zhao H, Zhang J. Promotion of epithelial-mesenchymal transformation by hepatocellular carcinoma-educated macrophages through Wnt2b/β-catenin/c-Myc signaling and reprogramming glycolysis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:13. [PMID: 33407720 PMCID: PMC7788901 DOI: 10.1186/s13046-020-01808-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/09/2020] [Indexed: 12/28/2022]
Abstract
Background Tumour-associated macrophages (TAMs) in the tumour microenvironment (TME) can promote the progression of hepatocellular carcinoma (HCC). Some tumours can be suppressed by targeting Wnt2b in tumour cells. However, the role of Wnt2b in HCC is still unknown. In particular, the role of Wnt2b-mediated signal activation in macrophage polarization in the HCC microenvironment, and the regulatory effect between Wnt and glycolysis in TAMs has not been described. Methods The expression of Wnt2b in TAMs was detected by qPCR and immunofluorescence. Wnt2b/β-catenin interference in HCC-TAMs was performed by lentivirus carrying targeted shRNA or TLR9 agonist. Markers related to macrophage polarization and the changes of key glycolytic enzymes expression were detected by flow cytometry and qPCR. ECAR was analysed by Seahorse analyser. MTT assay, wound healing assay, western blotting were used to evaluate the promoting effect of different HCC-TAMs on the proliferation, migration and EMT of HCC in vitro. Tumour cells and different HCC-TAMs were injected via subcutaneously into immunodeficient mice to assess the effects of CpG ODN, Wnt2b, or β-catenin on HCC-TAMs in tumour growth in vivo. Results Polarization-promoting factors derived from HCC cells upregulated the expression of Wnt2b in macrophages, which promoted the polarization of TAMs to M2-like macrophages by activating Wnt2b/β-catenin/c-Myc signalling. Furthermore, this process was associated with the activation of glycolysis in HCC-TAMs. These HCC-TAMs could promote the development of EMT, proliferation, and migration of HCC. In addition to silencing Wnt2b or β-catenin expression, TLR9 agonist CpG ODN downregulated the level of glycolysis and inhibited the M2 polarization of HCC-TAMs, reversing the tumour-promoting effects of TAMs in vitro and vivo. Conclusions As a potential target for HCC therapy, Wnt2b may play an important regulatory role for the functions of TAMs in the TME. Moreover, the TLR9 agonist CpG ODN might act as a Wnt2b signal inhibitor and can potentially be employed for HCC therapy by disturbing Wnt2b/β-catenin/c-Myc and inhibiting glycolysis in HCC-TAMs.
Collapse
Affiliation(s)
- Yu Jiang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, Shandong Province, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, Shandong Province, China
| | - Huajun Zhao
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, Shandong Province, China
| | - Jian Zhang
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
36
|
Granito A, Forgione A, Marinelli S, Renzulli M, Ielasi L, Sansone V, Benevento F, Piscaglia F, Tovoli F. Experience with regorafenib in the treatment of hepatocellular carcinoma. Therap Adv Gastroenterol 2021; 14:17562848211016959. [PMID: 34104211 PMCID: PMC8165525 DOI: 10.1177/17562848211016959] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Regorafenib is a diphenylurea oral multikinase inhibitor, structurally comparable to sorafenib, which targets a variety of kinases implicated in angiogenic and tumor growth-promoting pathways. Regorafenib was the first agent to positively show significant survival advantage as a second-line therapy in patients with unresectable hepatocellular carcinoma (HCC) who had previously failed first-line treatment with sorafenib. Recent evidence has shown that its antitumor efficacy is due to a comprehensive spectrum of tumor neo-angiogenesis and proliferation inhibition and immunomodulatory effects on the tumor microenvironment, which plays a crucial role in tumor development. This review addresses the rationale and supporting evidence for regorafenib's efficacy in HCC that led to regorafenib's approval as a second-line therapy. In addition, we review proof from clinical practice studies that validate the RESORCE trial results. We discuss regorafenib's potential role in the newly emerging therapeutic strategy based on combination with immune checkpoint blockade and its possible extensibility to patient categories not enrolled in the registrative study.
Collapse
Affiliation(s)
| | - Antonella Forgione
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italia
| | - Sara Marinelli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Matteo Renzulli
- Radiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luca Ielasi
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italia
| | - Vito Sansone
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italia
| | - Francesca Benevento
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italia
| | - Fabio Piscaglia
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italia
| | - Francesco Tovoli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italia
| |
Collapse
|
37
|
Hong GQ, Cai D, Gong JP, Lai X. Innate immune cells and their interaction with T cells in hepatocellular carcinoma. Oncol Lett 2021; 21:57. [PMID: 33281968 PMCID: PMC7709558 DOI: 10.3892/ol.2020.12319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/08/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor and is associated with necroinflammation driven by various immune cells, such as dendritic cells, macrophages and natural killer cells. Innate immune cells can directly affect HCC or regulate the T-cell responses that mediate HCC. In addition, innate immune cells and T cells are not isolated, which means the interaction between them is important in the HCC microenvironment. Considering the current unsatisfactory efficacy of immunotherapy in patients with HCC, understanding the relationship between innate immune cells and T cells is necessary. In the present review the roles and clinical value of innate immune cells that have been widely reported to be involved in HCC, including dendritic cells, macrophages (including kupffer cells), neutrophils, eosinophils, basophils and innate lymphoid cells and the crosstalk between the innate and adaptive immune responses in the antitumor process have been discussed. The present review will facilitate researchers in understanding the importance of innate immune cells in HCC and lead to innovative immunotherapy approaches for the treatment of HCC.
Collapse
Affiliation(s)
- Guo-Qing Hong
- Department of Hepatobiliary and Thyroid Breast Surgery, Tongnan District People's Hospital, Chongqing 402660, P.R. China
| | - Dong Cai
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jian-Ping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xing Lai
- Department of Hepatobiliary and Thyroid Breast Surgery, Tongnan District People's Hospital, Chongqing 402660, P.R. China
- Correspondence to: Dr Xing Lai, Department of Hepatobiliary and Thyroid Breast Surgery, Tongnan District People's Hospital, 271 Datong, Tongnan, Chongqing 402660, P.R. China, E-mail:
| |
Collapse
|
38
|
Ge Z, Ding S. The Crosstalk Between Tumor-Associated Macrophages (TAMs) and Tumor Cells and the Corresponding Targeted Therapy. Front Oncol 2020; 10:590941. [PMID: 33224886 PMCID: PMC7670061 DOI: 10.3389/fonc.2020.590941] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Tumor microenvironment (TME) is composed of tumor cells and surrounding non-tumor stromal cells, mainly including tumor associated macrophages (TAMs), endothelial cells, and carcinoma-associated fibroblasts (CAFs). The TAMs are the major components of non-tumor stromal cells, and play an important role in promoting the occurrence and development of tumors. Macrophages originate from bone marrow hematopoietic stem cells and embryonic yolk sacs. There is close crosstalk between TAMs and tumor cells. With the occurrence of tumors, tumor cells secrete various chemokines to recruit monocytes to infiltrate tumor tissues and further promote their M2-type polarization. Importantly, M2-like TAMs can in turn accelerate tumor growth, promote tumor cell invasion and metastasis, and inhibit immune killing to promote tumor progression. Therefore, targeting TAMs in tumor tissues has become one of the principal strategies in current tumor immunotherapy. Current treatment strategies focus on reducing macrophage infiltration in tumor tissues and reprogramming TAMs to M1-like to kill tumors. Although these treatments have had some success, their effects are still limited. This paper mainly summarized the recruitment and polarization of macrophages by tumors, the support of TAMs for the growth of tumors, and the research progress of TAMs targeting tumors, to provide new treatment strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Zhe Ge
- School of Physical Education & Health Care, East China Normal University, Shanghai, China.,Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Shuzhe Ding
- School of Physical Education & Health Care, East China Normal University, Shanghai, China.,Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| |
Collapse
|
39
|
Flem-Karlsen K, Fodstad Ø, Nunes-Xavier CE. B7-H3 Immune Checkpoint Protein in Human Cancer. Curr Med Chem 2020; 27:4062-4086. [PMID: 31099317 DOI: 10.2174/0929867326666190517115515] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/29/2019] [Accepted: 05/04/2019] [Indexed: 02/07/2023]
Abstract
B7-H3 belongs to the B7 family of immune checkpoint proteins, which are important regulators of the adaptive immune response and emerging key players in human cancer. B7-H3 is a transmembrane protein expressed on the surface of tumor cells, antigen presenting cells, natural killer cells, tumor endothelial cells, but can also be present in intra- and extracellular vesicles. Additionally, B7-H3 may be present as a circulating soluble isoform in serum and other body fluids. B7-H3 is overexpressed in a variety of tumor types, in correlation with poor prognosis. B7-H3 is a promising new immunotherapy target for anti-cancer immune response, as well as a potential biomarker. Besides its immunoregulatory role, B7-H3 has intrinsic pro-tumorigenic activities related to enhanced cell proliferation, migration, invasion, angiogenesis, metastatic capacity and anti-cancer drug resistance. B7-H3 has also been found to regulate key metabolic enzymes, promoting the high glycolytic capacity of cancer cells. B7-H3 receptors are still not identified, and little is known about the molecular mechanisms underlying B7-H3 functions. Here, we review the current knowledge on the involvement of B7-H3 in human cancer.
Collapse
Affiliation(s)
- Karine Flem-Karlsen
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Øystein Fodstad
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway.,Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Caroline E Nunes-Xavier
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, Oslo, Norway
| |
Collapse
|
40
|
Gao Y, Fang P, Li WJ, Zhang J, Wang GP, Jiang DF, Chen FP. LncRNA NEAT1 sponges miR-214 to regulate M2 macrophage polarization by regulation of B7-H3 in multiple myeloma. Mol Immunol 2019; 117:20-28. [PMID: 31731055 DOI: 10.1016/j.molimm.2019.10.026] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/30/2019] [Accepted: 10/30/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND LncRNA NEAT1 was associated with the tumorigenesis of multiple myeloma (MM). However, the mechanisms of M2 macrophage polarization involved with NEAT1 in MM are still unknown. METHODS Bone marrow samples, multiple myeloma cells RPMI 8226 and monocyte cell line THP-1 were used in this study. The expression of NEAT1 and miR-214 was modified by transfection with the shNEAT1 or miR-214 inhibitor. The expression of NEAT1, miR-214 and B7-H3 in MM patient tissues and cells was analyzed by RT-qPCR. ELISA assay was used to determine the release of B7-H3 in the supernatant of cell culture. The patient survival curve was analyzed using Kaplan-Meier method. The macrophage polarization markers were examined by RT-qPCR and western blotting. The interaction between NEAT1, miR-214 and B7-H3 was analyzed by Dual-Luciferase reporter and RIP assays. AG490 was used to block the JAK2/STAT3 signaling. Co-culture of THP-1 and RPMI 8226 cells was used for macrophage polarization. RESULTS NEAT1 and B7-H3 were up-regulated, but miR-214 was obviously down-regulated in MM patients. B7-H3, NEAT1 and miR-214 were associated with overall survival time of MM patients. NEAT1 silencing induced miR-214 and inhibited the expression and release of B7-H3 and then suppressed M2 macrophage polarization via inhibiting the JAK2/STAT3 signaling. NEAT1 directly targeted miR-214, and miR-214 directly bound to B7-H3. MiR-214 inhibitor reversed the down-regulation and release of B7-H3 and M2 macrophage polarization caused by shNEAT1. The specific JAK2/STAT3 signaling inhibitor AG490 abrogated M2 macrophage polarization. CONCLUSION NEAT1 promoted M2 macrophage polarization by sponging miR-214 and then regulating B7-H3, thus accelerating MM progression via the JAK2/STAT3 signaling pathway. Our study revealed novel mechanisms of M2 macrophage polarization and provided new potential clinical therapeutic targets for MM.
Collapse
Affiliation(s)
- Yin Gao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Peng Fang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Wen-Jin Li
- Department of Hematology, JiangXi PingXiang People's Hospital, Pingxiang 337000, Jiangxi Province, P.R. China
| | - Jian Zhang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410003, Hunan Province, P.R. China
| | - Guang-Ping Wang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China
| | - Duan-Feng Jiang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410003, Hunan Province, P.R. China
| | - Fang-Ping Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, P.R. China; Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410003, Hunan Province, P.R. China.
| |
Collapse
|
41
|
Zhang J, Shi Z, Xu X, Yu Z, Mi J. The influence of microenvironment on tumor immunotherapy. FEBS J 2019; 286:4160-4175. [PMID: 31365790 PMCID: PMC6899673 DOI: 10.1111/febs.15028] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/24/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022]
Abstract
Tumor immunotherapy has achieved remarkable efficacy, with immune-checkpoint inhibitors as especially promising candidates for cancer therapy. However, some issues caused by immunotherapy have raised attention, such as limited efficacy for some patients, narrow antineoplastic spectrum, and adverse reactions, suggesting that using regulators of tumor immune response may prove to be more complicated than anticipated. Current evidence indicates that different factors collectively constituting the unique tumor microenvironment promote immune tolerance, and these include the expression of co-inhibitory molecules, the secretion of lactate, and competition for nutrients between tumor cells and immune cells. Furthermore, cancer-associated fibroblasts, the main cellular components of solid tumors, promote immunosuppression through inhibition of T cell function and extracellular matrix remodeling. Here, we summarize the research advances in tumor immunotherapy and the latest insights into the influence of microenvironment on tumor immunotherapy.
Collapse
Affiliation(s)
- Jieying Zhang
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
- Research Center for Translational MedicineEast HospitalTongJi University School of MedicineShanghaiChina
| | - Zhaopeng Shi
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
| | - Xiang Xu
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
| | - Zuoren Yu
- Research Center for Translational MedicineEast HospitalTongJi University School of MedicineShanghaiChina
| | - Jun Mi
- Department of Biochemistry and Molecular Cell BiologyKey Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of EducationShanghai Jiao Tong University School of MedicineChina
- Hongqiao International Institute of MedicineTongren HospitalShanghai Jiao Tong University School of MedicineChina
| |
Collapse
|
42
|
Zhang J, Shi Z, Xu X, Yu Z, Mi J. The influence of microenvironment on tumor immunotherapy. THE FEBS JOURNAL 2019. [PMID: 31365790 DOI: 10.1111/febs.15028.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
Tumor immunotherapy has achieved remarkable efficacy, with immune-checkpoint inhibitors as especially promising candidates for cancer therapy. However, some issues caused by immunotherapy have raised attention, such as limited efficacy for some patients, narrow antineoplastic spectrum, and adverse reactions, suggesting that using regulators of tumor immune response may prove to be more complicated than anticipated. Current evidence indicates that different factors collectively constituting the unique tumor microenvironment promote immune tolerance, and these include the expression of co-inhibitory molecules, the secretion of lactate, and competition for nutrients between tumor cells and immune cells. Furthermore, cancer-associated fibroblasts, the main cellular components of solid tumors, promote immunosuppression through inhibition of T cell function and extracellular matrix remodeling. Here, we summarize the research advances in tumor immunotherapy and the latest insights into the influence of microenvironment on tumor immunotherapy.
Collapse
Affiliation(s)
- Jieying Zhang
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China.,Research Center for Translational Medicine, East Hospital, TongJi University School of Medicine, Shanghai, China
| | - Zhaopeng Shi
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China
| | - Xiang Xu
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China
| | - Zuoren Yu
- Research Center for Translational Medicine, East Hospital, TongJi University School of Medicine, Shanghai, China
| | - Jun Mi
- Department of Biochemistry and Molecular Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, China.,Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
43
|
Zhang S, Zhou C, Zhang D, Huang Z, Zhang G. The anti-apoptotic effect on cancer-associated fibroblasts of B7-H3 molecule enhancing the cell invasion and metastasis in renal cancer. Onco Targets Ther 2019; 12:4119-4127. [PMID: 31213832 PMCID: PMC6538013 DOI: 10.2147/ott.s201121] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/05/2019] [Indexed: 01/26/2023] Open
Abstract
Background: Renal cancer is one of the most common malignancies. However, the mechanisms underlying its development are still ambiguous. B7-H3 has been described as an important tumor antigen in various human tumors. An abnormal high expression of B7-H3 molecules is often observed in tumor cells and tumor stromal cells in the tumor microenvironment. On the basis of the above findings, we hypothesized that cancer-associated fibroblasts (CAFs) clustered in the renal cell microenvironment can survive for a long time with the anti-apoptotic effect of B7-H3, and then secrete cytokines to enhance the malignant behavior of renal cancer cells. Methods: The expression of B7-H3 protein in CAFs was detected in renal cancer tissues. Then, the CAFs cells were stably transfected with shRNA and their expression was silenced to determine the role of B7-H3 in CAFs. Western blot was used to detect the expression of apoptosis-related proteins, hepatocyte growth factor (HGF) protein and stromal cell-derived factor-1 (CXCL12) protein. CAF-NC cells and CAFs-shRNA cells were co-cultured with A498 cells to assess the biological function changes of A498. Results: A group of CAFs were found with B7-H3 expression in renal cancer. B7-H3 can stimulate CAFs to secrete HGF and Cxcl-12, and has strong anti-apoptotic effect on CAFs. We also found that CAFs-NC promotes the proliferation, invasion and migration of A498 cells in vitro and promotes the tumor formation of A498 in vivo. Conclusion: B7-H3+ CAFs promote the invasion and metastasis in renal cancer.
Collapse
Affiliation(s)
- Shuai Zhang
- Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University
- Jiangsu Institute of Jiangsu key Laboratory of Clinical Immunology, Soochow University
| | - Chenchao Zhou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou216007, People’s Republic of China
| | - Dongze Zhang
- Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University
- Jiangsu Institute of Jiangsu key Laboratory of Clinical Immunology, Soochow University
| | - Ziyi Huang
- Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University
- Jiangsu Institute of Jiangsu key Laboratory of Clinical Immunology, Soochow University
| | - Guangbo Zhang
- Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University
- Jiangsu Institute of Jiangsu key Laboratory of Clinical Immunology, Soochow University
| |
Collapse
|
44
|
Son Y, Kwon SM, Cho JY. CD276 (B7-H3) Maintains Proliferation and Regulates Differentiation in Angiogenic Function in Late Endothelial Progenitor Cells. Stem Cells 2018; 37:382-394. [PMID: 30379377 DOI: 10.1002/stem.2944] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/22/2018] [Accepted: 10/16/2018] [Indexed: 12/24/2022]
Abstract
Endothelial progenitor cells (EPCs) provide an important source of recovery from blood vessel dysfunction. Late EPCs (LEPCs) are circulating blood cells that are capable of promoting vascular repair. Using transcriptome analysis, we identified distinctive LEPC profiles and found that CD276 (B7-H3) mRNA is strongly expressed in LEPCs. CD276 protein is present abundantly on the cell surface of LEPC when analyzed by fluorescence-activated cell sorter and immunocytochemistry. CD276, a B7 family member, is a type I transmembrane glycoprotein. The role of CD276 in LEPCs remains unknown. CD276 knockdown by lentivirus transduction in LEPCs significantly decreased proliferation and increased apoptosis of LEPCs in vitro. After CD276 silencing, the cell cycle of LEPCs was prone to remain at the G0/G1 phase, and the cell migration rates as well as transwell and wound-healing migration were decreased. CD276 knockdown in LEPCs increased the G1 phase regulators cyclin D2/D3/E1-cyclin-dependent kinases (CDK2/4/6), but decreased the S-G2-M phase regulators cyclin A/B-CDK1. However, LEPCs with CD276 knockdown resulted in increased tube formation in vitro and angiogenesis in a Matrigel plug assay in vivo. FoxC1/C2, an upstream signal of Notch in arterial cell proliferation, and Hey1/2, which is known to promote arterial differentiation in the vasculature, were upregulated in CD276 knockdown LEPCs. In LEPCS, CD276 has a positive effect on proliferation and migration of endothelial cells, but negative effects on angiogenesis, particularly endothelial cell differentiation. Our data indicate, for therapeutic purpose, that CD276 can be used to acquire and maintain cell populations of LEPCs and blocking CD276 will promote angiogenetic differentiation. We found that CD276 (B7-H3) is enriched on the cell membrane of LEPCs. CD276 knockdown reduced proliferation and migration of LEPCs by increasing cell cycle inhibitors such as p21cip1 and pRb and decreasing pErk1/2 and pAkt but promoted angiogenesis and endothelial cell differentiation by elevating vascular endothelial growth factor-vascular endothelial growth factor receptor 1 and p-p38. Stem Cells 2019;37:382-394.
Collapse
Affiliation(s)
- YeonSung Son
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine & Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul, Korea
| |
Collapse
|
45
|
Yu D, Cheng J, Xue K, Zhao X, Wen L, Xu C. Expression of Programmed Death-Ligand 1 in Laryngeal Carcinoma and its Effects on Immune Cell Subgroup Infiltration. Pathol Oncol Res 2018; 25:1437-1443. [PMID: 30361911 DOI: 10.1007/s12253-018-0501-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 10/15/2018] [Indexed: 12/11/2022]
Abstract
To study the expression of programmed death-ligand 1 (PD-L1), and its effects on CD8+ tumor infiltrating lymphocytes (TILs) and tumor associated macrophages (TAMs) in human laryngeal squamous cell carcinoma. Sixty-nine patients with laryngeal carcinoma and 10 with vocal cord leukoplakia received tumor resection at Neck Surgery Department in the Second Affiliation Hospital of Jilin University (Changchun, Jilin) from Jan. 2010 to Dec. 2015. The expressions of PD-L1, CD8, CD16 and CD206 in laryngeal carcinoma, paracancerous and vocal cord leukoplakia tissues were detected with immunohistochemistry. The associations between PD-L1 expression and clinicopathologic features, expression of TAMs and CD8+ T cell infiltration were analyzed. Expression of PD-L1 is significantly higher in laryngeal carcinoma than in paracancerous or leukoplakia tissue. The expression of PD-L1 is closely associated with stage of laryngeal cancer, histological differentiation and neck lymphatic metastasis. PD-L1 expression is negatively correlated with the number of CD8+ TILs and CD16+ cells (M1 type TAMs), while is positively associated with CD206+ (M2 type TAMs). PD-L1 is highly expressed in the laryngeal cancer with the tumor microenvironment immunosuppression.
Collapse
Affiliation(s)
- Dan Yu
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China
| | - Jinzhang Cheng
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China
| | - Kai Xue
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China
| | - Xue Zhao
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China
| | - Lianji Wen
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China
| | - Chengbi Xu
- Department of Otolaryngology Head and Neck Surgery, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun, 130041, China.
| |
Collapse
|
46
|
Wang Z, Wang Z, Zhang C, Liu X, Li G, Liu S, Sun L, Liang J, Hu H, Liu Y, Zhang W, Jiang T. Genetic and clinical characterization of B7-H3 (CD276) expression and epigenetic regulation in diffuse brain glioma. Cancer Sci 2018; 109:2697-2705. [PMID: 30027617 PMCID: PMC6125452 DOI: 10.1111/cas.13744] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 06/25/2018] [Accepted: 07/03/2018] [Indexed: 12/21/2022] Open
Abstract
Gliomas are the most common malignant tumors of the brain. Immune checkpoints have been increasingly emphasized as targets for treating malignant tumors. B7-H3 has been identified as an immune checkpoint that shows potential value for targeting therapies. We set out to characterize the expression pattern and biological function of B7-H3 in brain gliomas using high-throughput data obtained from the Chinese Glioma Genome Atlas (CGGA) and the Cancer Genome Atlas (TCGA) projects. B7-H3 was upregulated more in higher-grade gliomas than that in lower-grade gliomas in both CGGA and TCGA datasets. Isocitrate dehydrogenase (IDH) mutation seemed to exert significant influence on B7-H3 expression in gliomas but led to quite different results between grade II gliomas and higher-grade gliomas. In addition to IDH, methylation of B7-H3 promoter and microRNA-29 family also showed a potential regulatory effect on B7-H3 expression. Gene ontology analysis revealed that B7-H3 was associated with mitotic cell cycle, cell proliferation and immune response. Further investigation suggested that B7-H3 was mostly involved in the Toll-like receptor signaling pathway. Survival analysis indicated that B7-H3 was an independent unfavorable prognosticator for glioma patients in both CGGA and TCGA datasets. B7-H3 expression is regulated by multiple mechanisms and is potentially involved in the T-cell receptor signaling pathway. Higher B7-H3 expression indicates a worse prognosis for glioma patients, which warrants further research into the development of inhibitors for targeting this immune checkpoint, but we still need to be cautious about immune checkpoint inhibition for central nervous system tumors.
Collapse
Affiliation(s)
- Zhiliang Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Zheng Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Chuanbao Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Xing Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Guanzhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Shuai Liu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Lihua Sun
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Jingshan Liang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Huimin Hu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Yanwei Liu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Wei Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Glioma Genome Atlas network, Beijing, China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
47
|
Xu Z, Wang L, Tian J, Man H, Li P, Shan B. High expression of B7-H3 and CD163 in cancer tissues indicates malignant clinicopathological status and poor prognosis of patients with urothelial cell carcinoma of the bladder. Oncol Lett 2018; 15:6519-6526. [PMID: 29725402 DOI: 10.3892/ol.2018.8173] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 11/29/2017] [Indexed: 11/05/2022] Open
Abstract
The objective of the present study was to investigate the association of B7-H3 expression and cluster of differentiation (CD)163+ tumor-associated macrophage (TAM) infiltration with clinicopathological parameters in urothelial cell carcinoma of the bladder (UCB), and to investigate their potential conjoint effects on progression of UCB. B7-H3 expression and CD163+ TAM infiltration in tumor specimens from 134 consecutive patients that underwent radical cystectomy for UCB were tested using immunohistochemistry, followed by statistical analysis. In these 134 patients, B7-H3 expression and CD163+ TAM infiltration in the bladder carcinoma tissues were significantly associated with an increased ratio of vascular invasion (P=0.009; P=0.012) and distant metastasis (P=0.015; P=0.038); however, they were not associated with gender, age, pathologic grade, tumor stage, recurrence or lymphatic metastasis. The results of χ2 test analysis indicated that CD163+ TAM infiltration and B7-H3 expression were positively correlated (χ2=20.714; P<0.001). Overall survival (OS) and progression-free survival (PFS) rates were significantly worsened by high B7-H3 expression (P=0.002; P=0.020). However, CD163+ TAM infiltration was not associated with OS or PFS rate. Notably, the OS and PFS rates in patients with high B7-H3 expression or high CD163+ TAM infiltration were significantly poorer than the patients with low B7-H3 expression (P<0.001; P<0.001) or low CD163+ TAM infiltration (P=0.022; P=0.017) in the subgroup of 115 patients with muscle-invasive bladder cancer. The results of the present study indicate that B7-H3 expression level could be used as an independent prognostic indicator following radical cystectomy for UCB and patients with high B7-H3 expression and high CD163+ TAM infiltration experience a poorer prognosis.
Collapse
Affiliation(s)
- Zhili Xu
- Department of Immunology, Cancer Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Ling Wang
- Department of Immunology, Cancer Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Jianhua Tian
- Department of Urology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Hongwei Man
- Department of Immunology, Cancer Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Pengfei Li
- Department of Immunology, Cancer Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Baoen Shan
- Department of Immunology, Cancer Research Institute, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
48
|
Fujimura T, Kakizaki A, Kambayashi Y, Sato Y, Tanita K, Lyu C, Furudate S, Aiba S. Cytotoxic antimelanoma drugs suppress the activation of M2 macrophages. Exp Dermatol 2018; 27:64-70. [PMID: 28833504 DOI: 10.1111/exd.13417] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2017] [Indexed: 12/18/2022]
Abstract
Together with regulatory T cells (Tregs), tumor-associated macrophages (TAMs) play roles in maintaining the tumor microenvironment. Although cytotoxic antimelanoma drugs such as dacarbazine (DTIC), nimustine hydrochloride (ACNU) and vincristine (VCR) have been used for the treatment of malignant melanoma as adjuvant therapy in Japan, the detailed mechanisms of their immunomodulatory effects are not fully understood. As the majority of TAMs are alternatively activated M2 macrophages that favour tumor development, the aim of this study was to elucidate the immunomodulatory effects of these reagents on human monocyte-derived M2 macrophages. First, mRNA expressions and protein production of immune checkpoint molecules, PD-L1 and chemokines by CD163+ CD206+ M2 macrophages derived from peripheral blood mononuclear cells were investigated to determine the immunomodulatory effects of DTIC, ACNU, and VCR. DTIC and VCR significantly decreased PD-L1 mRNA expression, which was confirmed by flow cytometry. Moreover, the mRNA expression and production of CCL22 were significantly decreased by DTIC, which suggested that DTIC might suppress the recruitment of Tregs in the tumor site. Furthermore, the decreased expression of PD-L1 and production of CCL22 were validated in vivo, using the B16F10 mouse melanoma model, leading to abrogation of the suppressive function of T-cell proliferation. The present report suggests one of the possible antimelanoma mechanisms of DAV combination chemotherapy for melanoma patients.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antineoplastic Agents/pharmacology
- B7-H1 Antigen/metabolism
- Dacarbazine/pharmacology
- Female
- Japan
- Lectins, C-Type/metabolism
- Leukocytes, Mononuclear/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Mannose Receptor
- Mannose-Binding Lectins/metabolism
- Melanoma/drug therapy
- Melanoma, Experimental
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Monocytes/metabolism
- Nimustine/pharmacology
- RNA, Messenger/metabolism
- Receptors, Cell Surface/metabolism
- Skin Neoplasms/drug therapy
- Vincristine/pharmacology
- Melanoma, Cutaneous Malignant
Collapse
Affiliation(s)
- Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Aya Kakizaki
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yota Sato
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kayo Tanita
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chunbing Lyu
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sadanori Furudate
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
49
|
Zhang X, Ji J, Zhang G, Fang C, Jiang F, Ma S, Hou J. Expression and significance of B7-H3 and Tie-2 in the tumor vasculature of clear cell renal carcinoma. Onco Targets Ther 2017; 10:5417-5424. [PMID: 29180874 PMCID: PMC5692197 DOI: 10.2147/ott.s147041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tumor angiogenesis is required for tumor growth and metastasis, and the Ang/Tie-2 axis plays a pivotal role in angiogenesis. B7-H3, a new member of the B7 family of costimulatory molecules, has a critical function in the T-cell-mediated antitumor immune response, and abnormal tumor B7-H3 expression is frequently associated with a poor prognosis. However, the relationship between B7-H3 and angiogenesis in clear cell renal carcinoma (ccRCC) remains unclear. In this study, we used immunohistochemical methods to detect tumor vascular expression of B7-H3 and Tie-2 in tissue microarrays of 82 ccRCC patient samples. According to the results, B7-H3 is highly expressed in the tumor vascular endothelium of ccRCC and is associated with the ccRCC grade and tumor-node-metastasis (TNM) stage. Although vascular Tie-2 expression was also correlated with T stage and lymph node metastasis, it was not related to ccRCC grade or distant metastasis. The microvessel density (MVD) labeled by CD34 was correlated with tumor grade and TNM stage. Expression of B7-H3 and Tie-2 was positively correlated, and the levels were positively associated with the MVD. Additionally, immunofluorescence staining revealed coexpression of B7-H3 and Tie-2 in the vascular endothelia of ccRCC. Collectively, our findings suggest that expression of B7-H3 and Tie-2 in ccRCC tumor vasculature is closely related to the progression and prognosis of the disease. Furthermore, B7-H3 possibly promotes ccRCC angiogenesis through the Tie-2 pathway. Thus, B7-H3 might serve as an effective endothelial marker for ccRCC prognosis and become a promising target for ccRCC anti-angiogenic-targeted therapy.
Collapse
Affiliation(s)
- Xianyun Zhang
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou.,Department of Urology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an
| | - Jindong Ji
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou
| | - Guangbo Zhang
- Clinical Immunology Laboratory, First Affiliated Hospital of Soochow University, Suzhou
| | - Chuntao Fang
- Department of Gynaecology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, Jiangsu, China
| | - Fujin Jiang
- Department of Urology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an
| | - Song Ma
- Department of Urology, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an
| | - Jianquan Hou
- Department of Urology, First Affiliated Hospital of Soochow University, Suzhou
| |
Collapse
|
50
|
Cai XJ, Wang Z, Cao JW, Ni JJ, Xu YY, Yao J, Xu H, Liu F, Yang GY. Anti-angiogenic and anti-tumor effects of metronomic use of novel liposomal zoledronic acid depletes tumor-associated macrophages in triple negative breast cancer. Oncotarget 2017; 8:84248-84257. [PMID: 29137420 PMCID: PMC5663592 DOI: 10.18632/oncotarget.20539] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 07/30/2017] [Indexed: 12/26/2022] Open
Abstract
Zoledronic acid (ZOL) has been used as an adjuvant therapy for breast cancer. It is suggested that ZOL might be associated with inhibition of macrophages, which in turn reduces tumor growth, metastasis and tumor angiogenesis. Moreover, metronomic therapy can inhibit tumor angiogenesis and tumor immune cells. Previously we developed ZOL based cationic liposomes that allowed a higher intratumor delivery of drug compared with free ZOL in vivo. Therefore, in this study, Asn-Gly-Arg (NGR) and PEG2000 were used as ligands to modify the surface of liposomes (NGR-PEG-LP-ZOL) in metronomic therapy to clear the tumor-associated macrophages (TAMs) and inhibit the formation of tumor angiogenesis, achieving the purpose of anti-tumor growth. Our data showed that NGR-PEG-LP-ZOL metronomic therapy has the strongest inhibitory effect on tumor growth. Further, NGR-PEG-LP-ZOL metronomic therapy could significantly impair TAMs by inhibiting the expression of CD206 antibody in tumor tissues, decreasing the expression of cytokine related gene expression of TAMs, as well as reducing the percentage of TAMs in tumor tissues. In addition, NGR-PEG-LP-ZOL metronomic therapy could significantly inhibit the expression of tumor neovascular specific antibody CD31 and reduce the microvessel density. In conclusion, our study demonstrated that NGR-PEG-LP-ZOL metronomic therapy could impair TAMs by inhibiting tumor angiogenesis and enhance the antitumor effect of ZOL.
Collapse
Affiliation(s)
- Xin-Jun Cai
- Department of Pharmacy, Zhe Jiang Chinese Medicine and Western Medicine Integrated Hospital, Hangzhou 310003, People's Republic of China
| | - Zeng Wang
- Department of Pharmacy, Zhejiang Cancer Hospital, Hangzhou 310022, People's Republic of China
| | - Jia-Wei Cao
- Department of Pharmacy, Zhe Jiang Chinese Medicine and Western Medicine Integrated Hospital, Hangzhou 310003, People's Republic of China
| | - Jian-Jun Ni
- Department of Pharmacy, Zhe Jiang Chinese Medicine and Western Medicine Integrated Hospital, Hangzhou 310003, People's Republic of China
| | - Ying-Ying Xu
- Department of Pharmacy, Zhe Jiang Chinese Medicine and Western Medicine Integrated Hospital, Hangzhou 310003, People's Republic of China
| | - Jun Yao
- Department of Pharmacy, Zhe Jiang Chinese Medicine and Western Medicine Integrated Hospital, Hangzhou 310003, People's Republic of China
| | - Hong Xu
- Department of Gastroenterology and Hepatology, Integrated Chinese and Western Medicine Hospital of Zhejiang Province, Hangzhou 310003, People's Republic of China
| | - Fang Liu
- Department of Acupuncture, Integrated Chinese and Western Medicine Hospital of Zhejiang Province, Hangzhou 310003, People's Republic of China
| | - Gao-Yi Yang
- Department of Ultrasound, Integrated Chinese and Western Medicine Hospital of Zhejiang Province, Hangzhou 310003, People's Republic of China
| |
Collapse
|