1
|
Guo H, Wei J, Zhang Y, Wang L, Wan J, Wang W, Gao L, Li J, Sun T, Ma L. Protein ubiquitination in ovarian cancer immunotherapy: The progress and therapeutic strategy. Genes Dis 2024; 11:101158. [PMID: 39253578 PMCID: PMC11382211 DOI: 10.1016/j.gendis.2023.101158] [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: 06/04/2023] [Revised: 09/04/2023] [Accepted: 10/10/2023] [Indexed: 09/11/2024] Open
Abstract
Ovarian cancer is a common cancer for females, and the incidence and mortality rates are on the rise. Many treatment strategies have been developed for ovarian cancer, including chemotherapy and immunotherapy, but they are often ineffective and prone to drug resistance. Protein ubiquitination is an important class of post-translation modifications that have been found to be associated with various human diseases and cancer development. Recent studies have revealed that protein ubiquitination is involved in the progression of ovarian cancer and plays an important role in the tumor immune process. Moreover, the combination of ubiquitinase/deubiquitinase inhibitors and cancer immunotherapy approaches can effectively reduce treatment resistance and improve treatment efficacy, which provides new ideas for cancer treatment. Herein, we review the role of protein ubiquitination in relation to ovarian cancer immunotherapy and recent advances in the use of ubiquitinase/deubiquitinase inhibitors in combination with cancer immunotherapy.
Collapse
Affiliation(s)
- Huiling Guo
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, China
| | - Jianwei Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yuyan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Li Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ling Gao
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan 450052, China
| | - Jiajing Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ting Sun
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, China
| | - Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Key Clinical Laboratory of Henan Province, Zhengzhou, Henan 450052, China
| |
Collapse
|
2
|
Ma S, Zhang P, Ye J, Tian Y, Tian X, Jung J, Macauley MS, Zhang J, Wu P, Wen L. Enzyme-Sialylation-Controlled Chemical Sulfation of Glycan Epitopes for Decoding the Binding of Siglec Ligands. J Am Chem Soc 2024; 146:29469-29480. [PMID: 39417319 DOI: 10.1021/jacs.4c08817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Widely distributed in nature, sulfated glycan epitopes play important roles in diverse pathophysiological processes. However, due to their structural complexity, the preparation of glycan epitopes with structurally defined sulfation patterns is challenging, which significantly hampers the detailed elucidation of their biological functions at the molecular level. Here, we introduce a strategy for site-specific chemical sulfation of glycan epitopes, leveraging enzymatic sialylation and desialylation processes to precisely control the regio-specificity of sulfation of disaccharide or trisaccharide glycan backbones. Using this method, a sulfated glycan library covering the most common sialylated glycan epitopes was prepared in high yield and efficiency. By screening a microarray prepared with this glycan library, we systematically probed their binding specificity with human Siglecs (sialic acid-binding immunoglobulin-type lectins), many of which function as glyco-immune checkpoints to suppress immune system activation. Our investigation revealed that sulfation and sialylation patterns serve as important determinants of Siglec binding affinity and specificity. Thus, these findings offer new insights for the development of research tools and potential therapeutic agents targeting glyco-immune checkpoints by modulating the Siglec signaling pathway.
Collapse
Affiliation(s)
- Shengzhou Ma
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Pengfei Zhang
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jinfeng Ye
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Yinping Tian
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiao Tian
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, China
| | - Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Jiabin Zhang
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, Guangdong 528400, China
| | - Peng Wu
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California 92037, United States
| | - Liuqing Wen
- Carbohydrate-Based Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
3
|
Murray CE, Kornepati AVR, Ontiveros C, Liao Y, de la Peña Avalos B, Rogers CM, Liu Z, Deng Y, Bai H, Kari S, Padron AS, Boyd JT, Reyes R, Clark CA, Svatek RS, Li R, Hu Y, Wang M, Conejo-Garcia JR, Byers LA, Ramkumar K, Sood AK, Lee JM, Burd CE, Vadlamudi RK, Gupta HB, Zhao W, Dray E, Sung P, Curiel TJ. Tumour-intrinsic PDL1 signals regulate the Chk2 DNA damage response in cancer cells and mediate resistance to Chk1 inhibitors. Mol Cancer 2024; 23:242. [PMID: 39478560 PMCID: PMC11523829 DOI: 10.1186/s12943-024-02147-z] [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: 02/07/2024] [Accepted: 10/05/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Aside from the canonical role of PDL1 as a tumour surface-expressed immune checkpoint molecule, tumour-intrinsic PDL1 signals regulate non-canonical immunopathological pathways mediating treatment resistance whose significance, mechanisms, and therapeutic targeting remain incompletely understood. Recent reports implicate tumour-intrinsic PDL1 signals in the DNA damage response (DDR), including promoting homologous recombination DNA damage repair and mRNA stability of DDR proteins, but many mechanistic details remain undefined. METHODS We genetically depleted PDL1 from transplantable mouse and human cancer cell lines to understand consequences of tumour-intrinsic PDL1 signals in the DNA damage response. We complemented this work with studies of primary human tumours and inducible mouse tumours. We developed novel approaches to show tumour-intrinsic PDL1 signals in specific subcellular locations. We pharmacologically depleted tumour PDL1 in vivo in mouse models with repurposed FDA-approved drugs for proof-of-concept clinical translation studies. RESULTS We show that tumour-intrinsic PDL1 promotes the checkpoint kinase-2 (Chk2)-mediated DNA damage response. Intracellular but not surface-expressed PDL1 controlled Chk2 protein content post-translationally and independently of PD1 by antagonising PIRH2 E3 ligase-mediated Chk2 polyubiquitination and protein degradation. Genetic tumour PDL1 depletion specifically reduced tumour Chk2 content but not ATM, ATR, or Chk1 DDR proteins, enhanced Chk1 inhibitor (Chk1i) synthetic lethality in vitro in diverse human and murine tumour models, and improved Chk1i efficacy in vivo. Pharmacologic tumour PDL1 depletion with cefepime or ceftazidime replicated genetic tumour PDL1 depletion by reducing tumour Chk2, inducing Chk1i synthetic lethality in a tumour PDL1-dependent manner, and reducing in vivo tumour growth when combined with Chk1i. CONCLUSIONS Our data challenge the prevailing surface PDL1 paradigm, elucidate important and previously unappreciated roles for tumour-intrinsic PDL1 in regulating the ATM/Chk2 DNA damage response axis and E3 ligase-mediated protein degradation, suggest tumour PDL1 as a biomarker for Chk1i efficacy, and support the rapid clinical potential of pharmacologic tumour PDL1 depletion to treat selected cancers.
Collapse
Affiliation(s)
- Clare E Murray
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Anand V R Kornepati
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Present address: Department of Internal Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Carlos Ontiveros
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yiji Liao
- Dartmouth Cancer Center and Dartmouth Health, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Bárbara de la Peña Avalos
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Cody M Rogers
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Zexuan Liu
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Yilun Deng
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Haiyan Bai
- Dartmouth Cancer Center and Dartmouth Health, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Suresh Kari
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Alvaro S Padron
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Jacob T Boyd
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ryan Reyes
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Curtis A Clark
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Present address: Department of Radiation Oncology, School of Medicine, University of Alabama Birmingham, Birmingham, USA
| | - Robert S Svatek
- Department of Urology, University of Texas Health San Antonio, San Antonio, TX, USA
- UT Health Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Rong Li
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Present address: Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, DC, USA
| | - Yanfen Hu
- Department of Molecular Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- Present address: Department of Anatomy & Cell Biology, School of Medicine & Health Sciences, The George Washington University, Washington, DC, USA
| | - Meiling Wang
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | | | - Lauren A Byers
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kavya Ramkumar
- Department of Thoracic/Head & Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology & Reproductive Medicine, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Jung-Min Lee
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christin E Burd
- Departments of Molecular Genetics, Cancer Biology and Genetics, The Ohio State University, Columbus, OH, USA
| | - Ratna K Vadlamudi
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA
- UT Health Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Harshita B Gupta
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Weixing Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Eloïse Dray
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Patrick Sung
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- UT Health Mays Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Tyler J Curiel
- Graduate School of Biomedical Sciences and Long School of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA.
- Dartmouth Cancer Center and Dartmouth Health, Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA.
- UT Southwestern, Dallas, TX, USA.
- Dartmouth Health and Dartmouth Cancer Center, Lebanon, NH, USA.
- The Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Department of Immunology, Dartmouth College, Hanover, NH, USA.
- Department of Integrative Immunobiology, Duke University, Durham, NC, USA.
| |
Collapse
|
4
|
Zhu Q, Li J, Sun H, Fan Z, Hu J, Chai S, Lin B, Wu L, Qin W, Wang Y, Hsieh-Wilson LC, Yi W. O-GlcNAcylation of enolase 1 serves as a dual regulator of aerobic glycolysis and immune evasion in colorectal cancer. Proc Natl Acad Sci U S A 2024; 121:e2408354121. [PMID: 39446384 PMCID: PMC11536113 DOI: 10.1073/pnas.2408354121] [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: 04/29/2024] [Accepted: 09/26/2024] [Indexed: 10/27/2024] Open
Abstract
Aerobic glycolysis and immune evasion are two key hallmarks of cancer. However, how these two features are mechanistically linked to promote tumor growth is not well understood. Here, we show that the glycolytic enzyme enolase-1 (ENO1) is dynamically modified with an O-linked β-N-acetylglucosamine (O-GlcNAcylation), and simultaneously regulates aerobic glycolysis and immune evasion via differential glycosylation. Glycosylation of threonine 19 (T19) on ENO1 promotes its glycolytic activity via the formation of active dimers. On the other hand, glycosylation of serine 249 (S249) on ENO1 inhibits its interaction with PD-L1, decreases association of PD-L1 with the E3 ligase STUB1, resulting in stabilization of PD-L1. Consequently, blockade of T19 glycosylation on ENO1 inhibits glycolysis, and decreases cell proliferation and tumor growth. Blockade of S249 glycosylation on ENO1 reduces PD-L1 expression and enhances T cell-mediated immunity against tumor cells. Notably, elimination of glycosylation at both sites synergizes with PD-L1 monoclonal antibody therapy to promote antitumor immune response. Clinically, ENO1 glycosylation levels are up-regulated and show a positive correlation with PD-L1 levels in human colorectal cancers. Thus, our findings provide a mechanistic understanding of how O-GlcNAcylation bridges aerobic glycolysis and immune evasion to promote tumor growth, suggesting effective therapeutic opportunities.
Collapse
Affiliation(s)
- Qiang Zhu
- Department of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou310058, China
- Department of Biophysics, College of Life Sciences, Zhejiang University,Hangzhou310058, China
| | - Jingchao Li
- Department of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou310058, China
- Department of Biophysics, College of Life Sciences, Zhejiang University,Hangzhou310058, China
| | - Haofan Sun
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing100026, China
| | - Zhiya Fan
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing100026, China
| | - Jiating Hu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| | - Siyuan Chai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| | - Bingyi Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| | - Liming Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| | - Weijie Qin
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing100026, China
| | - Yong Wang
- Department of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou310058, China
- Department of Biophysics, College of Life Sciences, Zhejiang University,Hangzhou310058, China
| | - Linda C. Hsieh-Wilson
- Division of Chemistry & Chemical Engineering, California Institute of Technology, Pasadena, CA91125
| | - Wen Yi
- Department of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou310058, China
- Department of Biophysics, College of Life Sciences, Zhejiang University,Hangzhou310058, China
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang Provincial Key Laboratory of Pancreatic Disease, School of Medicine, Zhejiang University, Hangzhou310002, China
| |
Collapse
|
5
|
Dong T, Niu H, Chu Z, Zhou C, Gao Y, Jia M, Sun B, Zheng X, Zhang W, Zhang J, Luo Y, Sun Y, Wang C, Lu Q, Liu C, Shao G, Lou H, Yuan H. Targeting VPS18 hampers retromer trafficking of PD-L1 and augments immunotherapy. SCIENCE ADVANCES 2024; 10:eadp4917. [PMID: 39413192 PMCID: PMC11482321 DOI: 10.1126/sciadv.adp4917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/12/2024] [Indexed: 10/18/2024]
Abstract
Immune checkpoint inhibitors targeting programmed cell death 1 (PD-1) or programmed cell death-ligand 1 (PD-L1) have achieved impressive antitumor clinical outcomes. However, the limited response rates suggest the incomplete understanding of PD-L1 regulation. Here, we demonstrate that vacuole protein sorting 11 and 18 (VPS11/18), two key players in vesicular trafficking, positively regulate PD-L1 and confer resistance to immune checkpoint blockade therapy. VPS11/18 interact with PD-L1 in endosome recycling accompanied by promoting PD-L1 glycosylation and protein stability. VPS18 deficiency enhances antitumor immune response. Pharmacological inhibition by VPS18 inhibitor RDN impaired PD-L1 member trafficking and protein stability. Combination treatment of RDN and anti-cytotoxic T lymphocyte-associated antigen 4 synergistically enhances antitumor efficacy in aggressive and drug-resistant tumors. RDN exerted lung-preferred distribution and good bioavailability, suggesting a favorable drug efficacy. Together, our study links VPS18/11-mediated trans-Golgi network recycling of PD-L1 and points to a promising treatment strategy for the enhancement of antitumor immunity.
Collapse
Affiliation(s)
- Ting Dong
- Key Laboratory of Natural Products & Chemical Biology of Ministry of Education, Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Huanmin Niu
- Key Laboratory of Natural Products & Chemical Biology of Ministry of Education, Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Zhaojun Chu
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Chengjun Zhou
- Department of Pathology/Urinary Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Yinghui Gao
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Mengqi Jia
- Key Laboratory of Natural Products & Chemical Biology of Ministry of Education, Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Bin Sun
- National Glycoengineering Research Center, Shandong University, Jinan 250012, China
| | - Xiaoxue Zheng
- Key Laboratory of Natural Products & Chemical Biology of Ministry of Education, Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Wenru Zhang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Jiaozhen Zhang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Yanhai Luo
- Key Laboratory of Natural Products & Chemical Biology of Ministry of Education, Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Yong Sun
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Chan Wang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
| | - Qiqi Lu
- Key Laboratory of Natural Products & Chemical Biology of Ministry of Education, Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Changhong Liu
- Key Laboratory of Natural Products & Chemical Biology of Ministry of Education, Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| | - Guangfeng Shao
- Department of Pathology/Urinary Surgery, The Second Hospital of Shandong University, Jinan, China
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, Shandong Province, China
- Traditional Chinese Medicine Department, Shandong Provincial Maternal and Child Health Care Hospital, Jinan, Shandong Province, China
| | - Huiqing Yuan
- Key Laboratory of Natural Products & Chemical Biology of Ministry of Education, Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, China
| |
Collapse
|
6
|
Xiao K, Zhang S, Peng Q, Du Y, Yao X, Ng II, Tang H. PD-L1 protects tumor-associated dendritic cells from ferroptosis during immunogenic chemotherapy. Cell Rep 2024; 43:114868. [PMID: 39423128 DOI: 10.1016/j.celrep.2024.114868] [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: 01/23/2023] [Revised: 06/13/2024] [Accepted: 09/27/2024] [Indexed: 10/21/2024] Open
Abstract
Dendritic cells (DCs) express high levels of PD-L1 in the tumor microenvironment. However, the physiological functions of PD-L1 on DCs remain incompletely understood. Here, we explored the roles of PD-L1 signaling during immunogenic chemotherapy. We found that antitumor efficacy was dramatically reduced in the absence of PD-L1 on DCs. Chemotherapy reshaped the tumor immune microenvironment, particularly the DC compartment. In the absence of PD-L1, DCs were more susceptible to the cytotoxicity induced by chemotherapy. Mechanistically, loss of PD-L1 led to the downregulation of SLC7A11, resulting in increased lipid peroxidation that caused DCs to succumb to ferroptosis and dampened antitumor immune responses. Mice with Pdl1-deficient DCs were less efficient at priming T cells during chemotherapy. In cancer patients, a higher level of PD-L1 on DCs correlated with better prognosis after immunogenic chemotherapy. Collectively, these findings reveal an underappreciated role of PD-L1 in orchestrating DC survival, which is critical during chemoimmunotherapy.
Collapse
Affiliation(s)
- Kaimin Xiao
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Silin Zhang
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Qi Peng
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China; Joint Graduate Program of Peking-Tsinghua-NIBS, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yuxia Du
- Department of General Practice, The Second Affiliated Hospital of Fujian Medical University, Quanzhou City, Fujian Province 362000, China
| | - Xiyue Yao
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Ian-Ian Ng
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Haidong Tang
- State Key Laboratory of Molecular Oncology, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
7
|
Liao Y, Zhao C, Pan Y, Guo Y, Liu L, Wu J, Zhang Y, Rao L, Li Q. Genetically engineered cellular nanoparticles loaded with curcuminoids for cancer immunotherapy. Theranostics 2024; 14:6409-6425. [PMID: 39431008 PMCID: PMC11488111 DOI: 10.7150/thno.99033] [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: 05/29/2024] [Accepted: 09/21/2024] [Indexed: 10/22/2024] Open
Abstract
Background: Inducing immunogenic cell death (ICD) is a promising strategy to enhance immune responses for immune checkpoint blockade (ICB) therapy, but the lack of a simple and effective platform to integrate ICD and ICB therapy limits their clinical application. Methods: Here, we developed programmed cell death protein 1 (PD1)-overexpressing genetically engineered nanovesicles (NVs)-coated curcumin (Cur)-loaded poly (lactic-co-poly-polyglycolic acid) nanoparticles (PD1@Cur-PLGA) to integrate ICD and ICB therapy for enhancing tumor immunotherapy. Results: Genetically engineered NVs greatly enhanced the tumor targeting of nanoparticles, and the PD1 on NVs dramatically blocked the PD1/PDL1 signaling pathway and stimulated antitumor immune responses. Meanwhile, the delivered Cur successfully induced tumor cell apoptosis and activated ICD by inhibiting NF-κB phosphorylation and Bcl-2 protein expression and activating caspase and Bax apoptotic signaling. By synergizing the ICD effect of Cur and the PD1/PDL1 axis blocking function of genetically engineered NVs, the PD1@Cur-PLGA enhanced the intratumoral infiltration rate of mature dendritic cells and CD8+ T cells in tumor tissues, resulting in significantly inhibiting tumor growth in breast and prostate tumor-bearing mouse models. Conclusion: This synergistic ICD and ICB therapy based on genetically engineered NVs provides a low-cost, safe, and effective strategy to enhance cancer immunotherapy.
Collapse
Affiliation(s)
- Yifang Liao
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Chenchen Zhao
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yuanwei Pan
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Yiming Guo
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Lujie Liu
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Jicheng Wu
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yunjiao Zhang
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Lang Rao
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen 518132, China
| | - Qi Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
8
|
Wang M, Qin L, Thia K, Nguyen T, MacDonald S, Belobrov S, Kranz S, Goode D, Trapani JA, Wiesenfeld D, Neeson PJ. Cancer cell-specific PD-L1 expression is a predictor of poor outcome in patients with locally advanced oral cavity squamous cell carcinoma. J Immunother Cancer 2024; 12:e009617. [PMID: 39357980 PMCID: PMC11448134 DOI: 10.1136/jitc-2024-009617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Locally advanced oral cavity squamous cell carcinoma (OCSCC) presents a significant clinical challenge despite being partially responsive to standard treatment modalities. This study investigates the prognostic implications of programmed death-ligand 1 (PD-L1) expression in these tumors, focusing on its association with treatment outcomes and the immune microenvironment. METHODS We assessed tumor-infiltrating lymphocytes (TILs) in 132 patients with OCSCC to evaluate their impact on survival. Multiplex immunohistochemistry staining for CD3, CD68, CD11c, PD-L1, and P40 was used to explore correlations with clinical outcomes in patients with early-stage (n=22) and locally advanced (n=36) OCSCC. These initial findings were validated through differential gene expression analysis, gene set enrichment, and immune cell deconvolution in a The Cancer Genome Atlas cohort of 163 locally advanced OCSCC tumors. Additionally, single-cell RNA sequencing (scRNA-seq) on a smaller cohort (n=10) further characterized the PD-L1hi or PD-L1lo cancer cells in these tumors. RESULTS Elevated PD-L1 expression was associated with poor outcomes in patients with locally advanced OCSCC undergoing standard adjuvant therapy, irrespective of "hot" or "cold" classification based on TILs assessment. PD-L1hi tumors exhibited an active immune response phenotype, enriched with M1 macrophages, CD8+ T cells and T regulatory cells in the tumor microenvironment. Notably, the negative impact of PD-L1 expression on outcomes was primarily attributed to its expression by cancer cells, rather than immune cells. Furthermore, scRNA-seq revealed that immune interactions were not essential for PD-L1 upregulation in cancer cells, instead, complex regulatory networks were involved. Additionally, PD-L1lo locally advanced tumors exhibited more complex pathway enrichment and diverse T-cell populations compared with those in the early-stage. CONCLUSION Our findings underscore the prognostic significance of PD-L1 expression in locally advanced OCSCC, and unveil the complex interplay between PD-L1 expression, immune responses, and molecular pathways in the tumor microenvironment. This study provides insights that may inform future therapeutic strategies, including the possibility of tailored immunotherapeutic approaches for patients with PD-L1hi locally advanced OCSCC.
Collapse
Affiliation(s)
- Minyu Wang
- Cancer Immunology Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Cancer Immunotherapy, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Lei Qin
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kevin Thia
- Cancer Immunology Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Cancer Immunotherapy, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Thu Nguyen
- Cancer Immunology Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Sean MacDonald
- Cancer Immunology Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Cancer Immunotherapy, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Simone Belobrov
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
| | - Sevastjan Kranz
- Department of Pathology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - David Goode
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Computational Cancer Biology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Joseph A Trapani
- Cancer Immunology Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Cancer Immunotherapy, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - David Wiesenfeld
- Melbourne Dental School, The University of Melbourne, Melbourne, Victoria, Australia
- Oral and Maxillofacial Surgery Unit, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Paul Joseph Neeson
- Cancer Immunology Research, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Cancer Immunotherapy, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Huang C, Wang X, Wang L, Liu Y, Xia Z, Wang X, Chen J. Targeting tumor associated macrophages (TAMs) reprograms tumor immune microenvironment to promote solid tumor immunotherapy. Cell Oncol (Dordr) 2024; 47:2011-2014. [PMID: 39235585 DOI: 10.1007/s13402-024-00987-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Affiliation(s)
- Chunliu Huang
- Nasopharyngeal Carcinoma Center, The Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Zhuhai, China.
| | - Xiumei Wang
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lixiang Wang
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yujia Liu
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zijin Xia
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinyu Wang
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Jun Chen
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Guangdong Engineering and Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China.
- Jinfeng Laboratory, Chongqing, China.
| |
Collapse
|
10
|
Peng L, Sferruzza G, Yang L, Zhou L, Chen S. CAR-T and CAR-NK as cellular cancer immunotherapy for solid tumors. Cell Mol Immunol 2024; 21:1089-1108. [PMID: 39134804 PMCID: PMC11442786 DOI: 10.1038/s41423-024-01207-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/22/2024] [Indexed: 10/02/2024] Open
Abstract
In the past decade, chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising immunotherapeutic approach for combating cancers, demonstrating remarkable efficacy in relapsed/refractory hematological malignancies in both pediatric and adult patients. CAR-natural killer (CAR-NK) cell complements CAR-T cell therapy by offering several distinct advantages. CAR-NK cells do not require HLA compatibility and exhibit low safety concerns. Moreover, CAR-NK cells are conducive to "off-the-shelf" therapeutics, providing significant logistic advantages over CAR-T cells. Both CAR-T and CAR-NK cells have shown consistent and promising results in hematological malignancies. However, their efficacy against solid tumors remains limited due to various obstacles including limited tumor trafficking and infiltration, as well as an immuno-suppressive tumor microenvironment. In this review, we discuss the recent advances and current challenges of CAR-T and CAR-NK cell immunotherapies, with a specific focus on the obstacles to their application in solid tumors. We also analyze in depth the advantages and drawbacks of CAR-NK cells compared to CAR-T cells and highlight CAR-NK CAR optimization. Finally, we explore future perspectives of these adoptive immunotherapies, highlighting the increasing contribution of cutting-edge biotechnological tools in shaping the next generation of cellular immunotherapy.
Collapse
Affiliation(s)
- Lei Peng
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- System Biology Institute, Yale University, West Haven, CT, USA.
| | - Giacomo Sferruzza
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
| | - Luojia Yang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA
| | - Liqun Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- System Biology Institute, Yale University, West Haven, CT, USA
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA
- Immunobiology Program, Yale University, New Haven, CT, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- System Biology Institute, Yale University, West Haven, CT, USA.
- Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA.
- Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, CT, USA.
- Immunobiology Program, Yale University, New Haven, CT, USA.
- Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA.
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Liver Center, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Biomedical Data Science, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
11
|
Pei S, Piao HL. Exploring Protein S-Palmitoylation: Mechanisms, Detection, and Strategies for Inhibitor Discovery. ACS Chem Biol 2024; 19:1868-1882. [PMID: 39160165 DOI: 10.1021/acschembio.4c00110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
S-palmitoylation is a reversible and dynamic process that involves the addition of long-chain fatty acids to proteins. This protein modification regulates various aspects of protein function, including subcellular localization, stability, conformation, and biomolecular interactions. The zinc finger DHHC (ZDHHC) domain-containing protein family is the main group of enzymes responsible for catalyzing protein S-palmitoylation, and 23 members have been identified in mammalian cells. Many proteins that undergo S-palmitoylation have been linked to disease pathogenesis and progression, suggesting that the development of effective inhibitors is a promising therapeutic strategy. Reducing the protein S-palmitoylation level can target either the PATs directly or their substrates. However, there are rare clinically effective S-palmitoylation inhibitors. This review aims to provide an overview of the S-palmitoylation field, including the catalytic mechanism of ZDHHC, S-palmitoylation detection methods, and the functional impact of protein S-palmitoylation. Additionally, this review focuses on current strategies for expanding the chemical toolbox to develop novel and effective inhibitors that can reduce the level of S-palmitoylation of the target protein.
Collapse
Affiliation(s)
- Shaojun Pei
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
| | - Hai-Long Piao
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
- University of Chinese Academy of Sciences, 100049 Beijing, China
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, 110122 Shenyang, China
| |
Collapse
|
12
|
Liu J, Qin J, Liang L, Zhang X, Gao J, Hao Y, Zhao P. Novel insights into the regulation of exosomal PD-L1 in cancer: From generation to clinical application. Eur J Pharmacol 2024; 979:176831. [PMID: 39047964 DOI: 10.1016/j.ejphar.2024.176831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/28/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Programmed cell death ligand 1 (PD-L1) interacts with programmed cell death 1 (PD-1), leading to T cell exhaustion and promoting tumor cell survival, ultimately mediating immunosuppression. While FDA-approved monoclonal antibodies targeting the PD-1/PD-L1 interaction have shown success in cancer treatment, some patients experience limited and short-lived therapeutic outcomes. Recent studies have identified PD-L1 expression not only on tumor cell surfaces but also on exosomes, with secretion pathways including both conventional and unconventional endocytosis routes, presenting a unique therapeutic opportunity. Emerging evidence suggests that exosomal PD-L1 contributes to systemic immunosuppression, potentially counteracting the effects of anti-PD-1 checkpoint therapies. However, the significance of exosomal PD-L1 in clinical cancer patients unresponsive to anti-PD-1/PD-L1 immunotherapy, as well as the factors regulating its generation, remain unclear. Moreover, the mechanisms underlying PD-L1 expression on exosomes and its regulation in cancer are yet to be fully elucidated. This review primarily focuses on the mechanisms modulating exosomal PD-L1 generation in cancer, while also outlining its involvement in immunosuppression, tumor proliferation, and response to cancer immunotherapy. Additionally, we explore the potential of exosomal PD-L1 as a cancer biomarker and therapeutic target, aiming to provide a comprehensive overview of this emerging field and its implications for cancer treatment and diagnosis.
Collapse
Affiliation(s)
- Jie Liu
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China; Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052, Erlangen, Germany
| | - Junxia Qin
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Lili Liang
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Xinzhong Zhang
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Jie Gao
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Youwei Hao
- Department of Cardiology, Taiyuan People's Hospital, Taiyuan, 030000, China
| | - Peng Zhao
- The Dermatology Department of Shanxi Provincial People's Hospital, Five Hospital of Shanxi Medical University, Taiyuan, 030012, China.
| |
Collapse
|
13
|
Guo Y, Dai Y, Yin J, Song Y, Wang T, Zhang L, Lu YJ, Song D. Novel tumor gene expression signatures improve the overall survival prediction efficiency over tumor mutation burden and PD-L1 expression in bladder carcinoma with checkpoint blockade immunotherapy. Am J Cancer Res 2024; 14:4411-4428. [PMID: 39417183 PMCID: PMC11477819 DOI: 10.62347/timd7591] [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: 07/06/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024] Open
Abstract
Although immune checkpoint blockade therapy (ICBT) has revolutionized cancer treatment with good therapeutic response in a number of human cancers, including bladder cancer, many cancers still do not respond to ICBT. Analyzing genetic signatures helps the understanding of underlying biological mechanisms. Here, based on two cohorts of bladder cancer patients receiving ICBT, we identified three novel ICBT-associated signatures in the bladder cancer microenvironment, involving genomic stability, angiogenesis and RNA regulatory, which affect PD-L1 expression and patient response to ICBT. The combinations of these signatures with TMB or PD-L1 expression improved the overall survival prediction efficiency over TMB and PD-L1 expression alone for patients receiving ICBT. Moreover, we utilized two methods to search potential drugs or small-molecules that have an impact on ICBT-associated signatures. This study provides new molecular insight into ICBT response of bladder cancer and has the potential to improve the prediction accuracy for patients to benefit from ICBT.
Collapse
Affiliation(s)
- Yufeng Guo
- Department of Urology, The First Affiliated Hospital and Academy of Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, China
| | - Yuanheng Dai
- Department of Urology, The First Affiliated Hospital and Academy of Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, China
| | - Jianjian Yin
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, China
| | - Yanliang Song
- Department of Urology, The First Affiliated Hospital and Academy of Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, China
- College of Public Health, Zhengzhou UniversityZhengzhou, Henan, China
| | - Tao Wang
- Department of Urology, The First Affiliated Hospital and Academy of Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, China
| | - Lirong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, China
| | - Yong-Jie Lu
- Department of Urology, The First Affiliated Hospital and Academy of Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, China
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of LondonLondon, The United Kingdom
| | - Dongkui Song
- Department of Urology, The First Affiliated Hospital and Academy of Medical Sciences, Zhengzhou UniversityZhengzhou, Henan, China
| |
Collapse
|
14
|
Wang L, Lin Y, Yao Z, Babu N, Lin W, Chen C, Du L, Cai S, Pan Y, Xiong X, Ye Q, Ren H, Zhang D, Chen Y, Yeung SCJ, Bremer E, Zhang H. Targeting undruggable phosphatase overcomes trastuzumab resistance by inhibiting multi-oncogenic kinases. Drug Resist Updat 2024; 76:101118. [PMID: 39094301 DOI: 10.1016/j.drup.2024.101118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/12/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024]
Abstract
AIMS Resistance to targeted therapy is one of the critical obstacles in cancer management. Resistance to trastuzumab frequently develops in the treatment for HER2+ cancers. The role of protein tyrosine phosphatases (PTPs) in trastuzumab resistance is not well understood. In this study, we aim to identify pivotal PTPs affecting trastuzumab resistance and devise a novel counteracting strategy. METHODS Four public datasets were used to screen PTP candidates in relation to trastuzumab responsiveness in HER2+ breast cancer. Tyrosine kinase (TK) arrays were used to identify kinases that linked to protein tyrosine phosphate receptor type O (PTPRO)-enhanced trastuzumab sensitivity. The efficacy of small activating RNA (saRNA) in trastuzumab-conjugated silica nanoparticles was tested for PTPRO upregulation and resistance mitigation in cell models, a transgenic mouse model, and human cancer cell line-derived xenograft models. RESULTS PTPRO was identified as the key PTP which influences trastuzumab responsiveness and patient survival. PTPRO de-phosphorated several TKs, including the previously overlooked substrate ERBB3, thereby inhibiting multiple oncogenic pathways associated with drug resistance. Notably, PTPRO, previously deemed "undruggable," was effectively upregulated by saRNA-loaded nanoparticles. The upregulated PTPRO simultaneously inhibited ERBB3, ERBB2, and downstream SRC signaling pathways, thereby counteracting trastuzumab resistance. CONCLUSIONS Antibody-conjugated saRNA represents an innovative approach for targeting "undruggable" PTPs.
Collapse
Affiliation(s)
- Lu Wang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Yusheng Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China; Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; Department of Hematology, University of Groningen, University Medical Center Groningen, the Netherlands; Shantou University Medical College, Shantou, China
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China; Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Nipun Babu
- Shantou University Medical College, Shantou, China
| | - Wan Lin
- Shantou University Medical College, Shantou, China
| | | | - Liang Du
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Songwang Cai
- Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yunlong Pan
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao Xiong
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Qiantao Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Zhuhai Institute of Jinan University, Zhuhai, China
| | - Hongzheng Ren
- Department of Pathology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China; Department of Pathology, Heping Hospital, Changzhi Medical College, Changzhi, China
| | - Dianzheng Zhang
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Yexi Chen
- Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin Bremer
- Department of Hematology, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Hao Zhang
- Department of Pathology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou, China; Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, China; Department of Thyroid, Breast and Hernia Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, China.
| |
Collapse
|
15
|
Qu Y, Gao N, Zhang S, Gao L, He B, Wang C, Gong C, Shi Q, Li Z, Yang S, Xiao Y. Role of N6-methyladenosine RNA modification in cancer. MedComm (Beijing) 2024; 5:e715. [PMID: 39252821 PMCID: PMC11381670 DOI: 10.1002/mco2.715] [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: 03/17/2024] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
N6-methyladenosine (m6A) is the most abundant modification of RNA in eukaryotic cells. Previous studies have shown that m6A is pivotal in diverse diseases especially cancer. m6A corelates with the initiation, progression, resistance, invasion, and metastasis of cancer. However, despite these insights, a comprehensive understanding of its specific roles and mechanisms within the complex landscape of cancer is still elusive. This review begins by outlining the key regulatory proteins of m6A modification and their posttranslational modifications (PTMs), as well as the role in chromatin accessibility and transcriptional activity within cancer cells. Additionally, it highlights that m6A modifications impact cancer progression by modulating programmed cell death mechanisms and affecting the tumor microenvironment through various cancer-associated immune cells. Furthermore, the review discusses how microorganisms can induce enduring epigenetic changes and oncogenic effect in microorganism-associated cancers by altering m6A modifications. Last, it delves into the role of m6A modification in cancer immunotherapy, encompassing RNA therapy, immune checkpoint blockade, cytokine therapy, adoptive cell transfer therapy, and direct targeting of m6A regulators. Overall, this review clarifies the multifaceted role of m6A modification in cancer and explores targeted therapies aimed at manipulating m6A modification, aiming to advance cancer research and improve patient outcomes.
Collapse
Affiliation(s)
- Yi Qu
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Nannan Gao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Shengwei Zhang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Limin Gao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Bing He
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Chao Wang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Chunli Gong
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Qiuyue Shi
- Department of Gastroenterology the First Affiliated Hospital of Guangxi Medical University Nanning Guangxi China
| | - Zhibin Li
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Shiming Yang
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| | - Yufeng Xiao
- Department of Gastroenterology Xinqiao Hospital Army Medical University Chongqing China
| |
Collapse
|
16
|
Xi Q, Yang G, He X, Zhuang H, Li L, Lin B, Wang L, Wang X, Fang C, Chen Q, Yang Y, Yu Z, Zhang H, Cai W, Li Y, Shen H, Liu L, Zhang R. M 6A-mediated upregulation of lncRNA TUG1 in liver cancer cells regulates the antitumor response of CD8 + T cells and phagocytosis of macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400695. [PMID: 38981064 PMCID: PMC11425850 DOI: 10.1002/advs.202400695] [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: 01/19/2024] [Revised: 06/11/2024] [Indexed: 07/11/2024]
Abstract
Tumor immune evasion relies on the crosstalk between tumor cells and adaptive/innate immune cells. Immune checkpoints play critical roles in the crosstalk, and immune checkpoint inhibitors have achieved promising clinical effects. The long non-coding RNA taurine-upregulated gene 1 (TUG1) is upregulated in hepatocellular carcinoma (HCC). However, how TUG1 is upregulated and the effects on tumor immune evasion are incompletely understood. Here, METTL3-mediated m6A modification led to TUG1 upregulation is demonstrated. Knockdown of TUG1 inhibited tumor growth and metastasis, increased the infiltration of CD8+ T cells and M1-like macrophages in tumors, promoted the activation of CD8+ T cells through PD-L1, and improved the phagocytosis of macrophages through CD47. Mechanistically, TUG1 regulated PD-L1 and CD47 expressions by acting as a sponge of miR-141 and miR-340, respectively. Meanwhile, TUG1 interacted with YBX1 to facilitate the upregulation of PD-L1 and CD47 transcriptionally, which ultimately regulated tumor immune evasion. Clinically, TUG1 positively correlated with PD-L1 and CD47 in HCC tissues. Moreover, the combination of Tug1-siRNA therapy with a Pdl1 antibody effectively suppressed tumor growth. Therefore, the mechanism of TUG1 in regulating tumor immune evasion is revealed and can inform existing strategies targeting TUG1 for enhancing HCC immune therapy and drug development.
Collapse
Affiliation(s)
- Qing Xi
- Department of Gastroenterology and HepatologyThe First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhou510080China
- School of Biomedical Sciences and EngineeringSouth China University of TechnologyGuangzhou511442China
| | - Guangze Yang
- Laboratory of Immunology and InflammationDepartment of ImmunologyKey Laboratory of Immune Microenvironment and Diseases of Educational Ministry of ChinaTianjin Medical UniversityTianjin300070China
| | - Xue He
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Hao Zhuang
- Department of Hepatobiliopancreatic SurgeryThe Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhou450008China
| | - Li Li
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Bing Lin
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Lingling Wang
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Xianyang Wang
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Chunqiang Fang
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Qiurui Chen
- Department of BioscienceSchool of Life Sciences and BiopharmaceuticsGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Yongjie Yang
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Zhaoan Yu
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Hao Zhang
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Wenqian Cai
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Yan Li
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Han Shen
- Department of BioscienceSchool of Life Sciences and BiopharmaceuticsGuangdong Pharmaceutical UniversityGuangzhou51006China
| | - Li Liu
- Department of RadiologyThe University of Texas Southwestern Medical Center5323 Harry Hines Blvd.DallasTX75390USA
| | - Rongxin Zhang
- Laboratory of Immunology and InflammationDepartment of BiotechnologySchool of Life Sciences and BiopharmaceuticsGuangdong Provincial Key Laboratory of Advanced Drug DeliveryGuangdong Provincial Engineering Center of Topical Precise Drug Delivery SystemGuangdong Pharmaceutical UniversityGuangzhou51006China
| |
Collapse
|
17
|
Bernadett L, Alexandra K, Georgina F, Erika T, András S, Ilona P, Ferenc O, Orsolya D. No Correlation between PD-L1 and NIS Expression in Lymph Node Metastatic Papillary Thyroid Carcinoma. Diagnostics (Basel) 2024; 14:1858. [PMID: 39272644 PMCID: PMC11394040 DOI: 10.3390/diagnostics14171858] [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: 07/05/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Approximately 90% of thyroid cancers are differentiated thyroid cancers (DTCs), originating from follicular epithelial cells. Out of these, 90% are papillary thyroid cancer (PTC), and 10% are follicular thyroid cancer (FTC). The standard care procedure for PTC includes surgery, followed by radioiodine (RAI) ablation and thyroid-stimulating hormone (TSH) suppressive therapy. Globally, treating radioiodine-refractory DTC poses a challenge. During malignant transformation, thyroid epithelial cells often lose their ability to absorb radioiodine due to impaired membrane targeting or lack of NIS (sodium/iodide symporter) expression. Recent reports show an increase in PD-L1 (programmed death ligand 1) expression in thyroid cancer cells during dedifferentiation. However, no research exists wherein NIS and PD-L1 expression are analyzed together in thyroid cancer. Therefore, we aimed to investigate and correlate PD-L1 and NIS expression within primary tumor samples of lymph node metastatic PTC. We analyzed the expression of hNIS (human sodium/iodide symporter) and PD-L1 in primary tumor samples from metastatic PTC patients using immunohistochemistry. Immunohistochemistry analysis of PD-L1 and NIS was conducted in 89 and 86 PTC cases, respectively. Any subcellular NIS localization was counted as a positive result. PD-L1 expression was absent in 25 tumors, while 58 tumors displayed PD-L1 expression in 1-50% of their cells; in 6 tumors, over 50% of the cells tested positive for PD-L1. NIS immunohistochemistry was performed for 86 primary papillary carcinomas, with 51 out of 86 tumors showcasing NIS expression. Only in seven cases was NIS localized in the plasma membrane; in most tumors, NIS was primarily found in the intracytoplasmic membrane compartments. In the case of PD-L1 staining, cells showing linear membrane positivity of any intensity were counted as positive. The evaluation of NIS immunostaining was simpler: cells showing staining of any intensity of cytoplasmic or membranous fashion were counted as positive. The number of NIS positive cells can be further divided into cytoplasmic and membrane positive compartments. There was no observed correlation between PD-L1 and NIS expression. We can speculate that the manipulation of the PD-1/PD-L1 axis using anti-PD-L1 or anti-PD-1 antibodies could reinstate the functional expression of NIS. However, based on our study, the only conclusion that can be drawn is that there is no correlation between the percentage of NIS- or PD-L1-expressing tumor cells in the primary tumor of lymph node metastatic PTC.
Collapse
Affiliation(s)
- Lévay Bernadett
- National Institute of Oncology, Multidisciplinary Head and Neck Cancer Center, 1124 Budapest, Hungary
| | - Kiss Alexandra
- National Institute of Oncology, Multidisciplinary Head and Neck Cancer Center, 1124 Budapest, Hungary
| | - Fröhlich Georgina
- National Institute of Oncology, Budapest, Center of Radiotherapy, 1124 Budapest, Hungary
| | - Tóth Erika
- National Institute of Oncology, Department of Molecular Pathology and Surgical Pathology Center, 1124 Budapest, Hungary
| | - Slezák András
- National Institute of Oncology, Department of Molecular Pathology and Surgical Pathology Center, 1124 Budapest, Hungary
| | - Péter Ilona
- National Institute of Oncology, Department of Molecular Pathology and Surgical Pathology Center, 1124 Budapest, Hungary
| | - Oberna Ferenc
- National Institute of Oncology, Multidisciplinary Head and Neck Cancer Center, 1124 Budapest, Hungary
| | - Dohán Orsolya
- Department of Internal Medicine and Clinical Oncology, Semmelweis University, 1124 Budapest, Hungary
| |
Collapse
|
18
|
Liu D, Yan J, Ma F, Wang J, Yan S, He W. Reinvigoration of cytotoxic T lymphocytes in microsatellite instability-high colon adenocarcinoma through lysosomal degradation of PD-L1. Nat Commun 2024; 15:6922. [PMID: 39134545 PMCID: PMC11319731 DOI: 10.1038/s41467-024-51386-7] [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: 10/20/2023] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Compensation and intracellular storage of PD-L1 may compromise the efficacy of antibody drugs targeting the conformational blockade of PD1/PD-L1 on the cell surface. Alternative therapies aiming to reduce the overall cellular abundance of PD-L1 thus might overcome resistance to conventional immune checkpoint blockade. Here we show by bioinformatics analysis that colon adenocarcinoma (COAD) with high microsatellite instability (MSI-H) presents the most promising potential for this therapeutic intervention, and that overall PD-L1 abundance could be controlled via HSC70-mediated lysosomal degradation. Proteomic and metabolomic analyses of mice COAD with MSI-H in situ unveil a prominent acidic tumor microenvironment. To harness these properties, an artificial protein, IgP β, is engineered using pH-responsive peptidic foldamers. This features customized peptide patterns and designed molecular function to facilitate interaction between neoplastic PD-L1 and HSC70. IgP β effectively reduces neoplastic PD-L1 levels via HSC70-mediated lysosomal degradation, thereby persistently revitalizing the action of tumor-infiltrating CD8 + T cells. Notably, the anti-tumor effect of lysosomal-degradation-based therapy surpasses that of antibody-based immune checkpoint blockade for MSI-H COAD in multiple mouse models. The presented strategy expands the use of peptidic foldamers in discovering artificial protein drugs for targeted cancer immunotherapy.
Collapse
Affiliation(s)
- Dan Liu
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Jin Yan
- Department of infectious Diseases, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China.
- Department of Tumor and Immunology in precision medical institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China.
| | - Fang Ma
- Department of Tumor and Immunology in precision medical institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Jingmei Wang
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Siqi Yan
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wangxiao He
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China.
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China.
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
19
|
Cai S, Yang G, Hu M, Li C, Yang L, Zhang W, Sun J, Sun F, Xing L, Sun X. Spatial cell interplay networks of regulatory T cells predict recurrence in patients with operable non-small cell lung cancer. Cancer Immunol Immunother 2024; 73:189. [PMID: 39093404 PMCID: PMC11297009 DOI: 10.1007/s00262-024-03762-x] [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: 11/05/2023] [Accepted: 06/13/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND The interplay between regulatory T cells (Tregs) and neighboring cells, which is pivotal for anti-tumor immunity and closely linked to patient prognosis, remains to be fully elucidated. METHODS Tissue microarrays of 261 operable NSCLC patients were stained by multiplex immunofluorescence (mIF) assay, and the interaction between Tregs and neighboring cells in the tumor microenvironment (TME) was evaluated. Employing various machine learning algorithms, we developed a spatial immune signature to predict the prognosis of NSCLC patients. Additionally, we explored the interplay between programmed death-1/programmed death ligand-1 (PD-1/PD-L1) interactions and their relationship with Tregs. RESULTS Survival analysis indicated that the interplay between Tregs and neighboring cells in the invasive margin (IM) and tumor center was associated with recurrence in NSCLC patients. We integrated the intersection of the three algorithms to identify four crucial spatial immune features [P(CD8+Treg to CK) in IM, P(CD8+Treg to CD4) in IM, N(CD4+Treg to CK) in IM, N(CD4+Tcon to CK) in IM] and employed these characteristics to establish SIS, an independent prognosticator of recurrence in NSCLC patients [HR = 2.34, 95% CI (1.53, 3.58), P < 0.001]. Furthermore, analysis of cell interactions demonstrated that a higher number of Tregs contributed to higher PD-L1+ cells surrounded by PD-1+ cells (P < 0.001) with shorter distances (P = 0.004). CONCLUSION We dissected the cell interplay network within the TME, uncovering the spatial architecture and intricate interactions between Tregs and neighboring cells, along with their impact on the prognosis of NSCLC patients.
Collapse
Affiliation(s)
- Siqi Cai
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Guanqun Yang
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mengyu Hu
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chaozhuo Li
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Liying Yang
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wei Zhang
- Shandong Cancer Hospital and Institute and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Jujie Sun
- Department of Pathology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Fenghao Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440, Jiyan Road, Huaiyin District, Jinan, 250117, Shandong, China
| | - Ligang Xing
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaorong Sun
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China.
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, No.440, Jiyan Road, Huaiyin District, Jinan, 250117, Shandong, China.
| |
Collapse
|
20
|
Albarrán V, Guerrero P, de Quevedo CG, González C, Chamorro J, Rosero DI, Moreno J, Calvo JC, de Aguado PP, Alía V, Sotoca P, Barrill AM, Román MS, Álvarez-Ballesteros P, Serrano JJ, Soria A, Olmedo ME, Saavedra C, Cortés A, Gómez A, Lage Y, Ruiz Á, Ferreiro MR, Longo F, Garrido P, Gajate P. Negative association of steroids with immunotherapy efficacy in a multi-tumor cohort: time and dose-dependent. Cancer Immunol Immunother 2024; 73:186. [PMID: 39093378 PMCID: PMC11297225 DOI: 10.1007/s00262-024-03772-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024]
Abstract
Previous studies have suggested a negative impact of steroids on the efficacy of immune checkpoint inhibitors (ICI), but how this effect is modulated by the dosage and time of administration is yet to be clarified. We have performed a retrospective analysis of 475 patients with advanced solid tumors treated with ICI as monotherapy from 2015 to 2022. Data regarding immune-related adverse events (irAEs) and clinical outcomes were collected. For each patient, the daily steroid dose (in mg/kg of prednisone) was registered until disease progression or death. The impact of cumulative doses on response rates and survival outcomes was analyzed within different periods. The objective response rate (ORR) was significantly lower among patients exposed to steroids within 30 days before the first cycle of ICI (C1) (20.3% vs. 36.7%, p < 0.01) and within the first 90 days of treatment (25.7% vs. 37.7%, p = 0.01). This negative association was confirmed by multivariable analysis. Higher mean steroid doses were observed among non-responders, and cumulative doses were inversely correlated with the disease control rate (DCR) around ICI initiation. Remarkably, poorer outcomes were observed even in patients belonging to the lowest dose quartile compared to the steroid-naïve population. The exposure to steroids after 6 months of ICI was not associated with worse survival outcomes. Our results suggest that the potential impact of steroids on ICI efficacy may be time-dependent, prevailing around ICI initiation, and dose-dependent, with modulation of neutrophil-to-lymphocyte ratio as a possible underlying mechanism.
Collapse
Affiliation(s)
- Víctor Albarrán
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| | - Patricia Guerrero
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Coral García de Quevedo
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Carlos González
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Jesús Chamorro
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Diana Isabel Rosero
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Jaime Moreno
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Juan Carlos Calvo
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Patricia Pérez de Aguado
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Víctor Alía
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pilar Sotoca
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Ana María Barrill
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - María San Román
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pablo Álvarez-Ballesteros
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Juan José Serrano
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Ainara Soria
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - María Eugenia Olmedo
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Cristina Saavedra
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Alfonso Cortés
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Ana Gómez
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Yolanda Lage
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Álvaro Ruiz
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - María Reyes Ferreiro
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Federico Longo
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pilar Garrido
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pablo Gajate
- Department of Medical Oncology, Ramon y Cajal University Hospital (Madrid), Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| |
Collapse
|
21
|
Ortega MA, Boaru DL, De Leon-Oliva D, Fraile-Martinez O, García-Montero C, Rios L, Garrido-Gil MJ, Barrena-Blázquez S, Minaya-Bravo AM, Rios-Parra A, Álvarez-Mon M, Jiménez-Álvarez L, López-González L, Guijarro LG, Diaz R, Saez MA. PD-1/PD-L1 axis: implications in immune regulation, cancer progression, and translational applications. J Mol Med (Berl) 2024; 102:987-1000. [PMID: 38935130 DOI: 10.1007/s00109-024-02463-3] [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: 03/11/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024]
Abstract
The PD-1/PD-L1 axis is a complex signaling pathway that has an important role in the immune system cells. Programmed cell death protein 1 (PD-1) acts as an immune checkpoint on the T lymphocytes, B lymphocytes, natural killer (NK), macrophages, dendritic cells (DCs), monocytes, and myeloid cells. Its ligand, the programmed cell death 1 ligand (PD-L1), is expressed in the surface of the antigen-presenting cells (APCs). The binding of both promotes the downregulation of the T cell response to ensure the activation to prevent the onset of chronic immune inflammation. This axis in the tumor microenvironment (TME) performs a crucial role in the tumor progression and the escape of the tumor by neutralizing the immune system, the engagement of PD-L1 with PD-1 in the T cell causes dysfunctions, neutralization, and exhaustion, providing the tumor mass production. This review will provide a comprehensive overview of the functions of the PD-1/PD-L1 system in immune function, cancer, and the potential therapeutic implications of the PD-1/PD-L1 pathway for cancer management.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain.
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain.
- Cancer Registry and Pathology Department, Principe de, Asturias University Hospital, Alcala de Henares, Spain.
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Laura Rios
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Maria J Garrido-Gil
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Silvestra Barrena-Blázquez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of Nursing and Physiotherapy, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Ana M Minaya-Bravo
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Antonio Rios-Parra
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Cancer Registry and Pathology Department, Principe de, Asturias University Hospital, Alcala de Henares, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Immune System Diseases-Rheumatology Service, University Hospital Principe de Asturias, CIBEREHD, 28801, Alcala de Henares, Spain
| | - Laura Jiménez-Álvarez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Laura López-González
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain
| | - Luis G Guijarro
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
| | - Raul Diaz
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain.
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801, Alcala de Henares, Spain.
- Surgery Service, University Hospital Principe de Asturias, 28801, Alcala de Henares, Spain.
| | - Miguel A Saez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, CIBEREHD, University of Alcalá, 28801, Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034, Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-University of Alcalá (UAH) Madrid, Alcala de Henares, Spain
| |
Collapse
|
22
|
Yu L, Huang K, Liao Y, Wang L, Sethi G, Ma Z. Targeting novel regulated cell death: Ferroptosis, pyroptosis and necroptosis in anti-PD-1/PD-L1 cancer immunotherapy. Cell Prolif 2024; 57:e13644. [PMID: 38594879 PMCID: PMC11294428 DOI: 10.1111/cpr.13644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/02/2024] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
Chemotherapy, radiotherapy, and immunotherapy represent key tumour treatment strategies. Notably, immune checkpoint inhibitors (ICIs), particularly anti-programmed cell death 1 (PD1) and anti-programmed cell death ligand 1 (PD-L1), have shown clinical efficacy in clinical tumour immunotherapy. However, the limited effectiveness of ICIs is evident due to many cancers exhibiting poor responses to this treatment. An emerging avenue involves triggering non-apoptotic regulated cell death (RCD), a significant mechanism driving cancer cell death in diverse cancer treatments. Recent research demonstrates that combining RCD inducers with ICIs significantly enhances their antitumor efficacy across various cancer types. The use of anti-PD-1/PD-L1 immunotherapy activates CD8+ T cells, prompting the initiation of novel RCD forms, such as ferroptosis, pyroptosis, and necroptosis. However, the functions and mechanisms of non-apoptotic RCD in anti-PD1/PD-L1 therapy remain insufficiently explored. This review summarises the emerging roles of ferroptosis, pyroptosis, and necroptosis in anti-PD1/PD-L1 immunotherapy. It emphasises the synergy between nanomaterials and PD-1/PD-L1 inhibitors to induce non-apoptotic RCD in different cancer types. Furthermore, targeting cell death signalling pathways in combination with anti-PD1/PD-L1 therapies holds promise as a prospective immunotherapy strategy for tumour treatment.
Collapse
Affiliation(s)
- Li Yu
- Health Science CenterYangtze UniversityJingzhouHubeiChina
- Department of UrologyJingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze UniversityJingzhouHubeiChina
| | - Ke Huang
- Health Science CenterYangtze UniversityJingzhouHubeiChina
| | - Yixiang Liao
- Department of UrologyJingzhou Central Hospital, Jingzhou Hospital Affiliated to Yangtze UniversityJingzhouHubeiChina
| | - Lingzhi Wang
- Department of PharmacologyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- Cancer Science Institute of Singapore, National University of SingaporeSingaporeSingapore
- NUS Centre for Cancer Research (N2CR), National University of SingaporeSingaporeSingapore
| | - Gautam Sethi
- Department of PharmacologyYong Loo Lin School of Medicine, National University of SingaporeSingaporeSingapore
- NUS Centre for Cancer Research (N2CR), National University of SingaporeSingaporeSingapore
| | - Zhaowu Ma
- Health Science CenterYangtze UniversityJingzhouHubeiChina
| |
Collapse
|
23
|
Jin Y, Christenson ES, Zheng L, Li K. Neutrophils in pancreatic ductal adenocarcinoma: bridging preclinical insights to clinical prospects for improved therapeutic strategies. Expert Rev Clin Immunol 2024; 20:945-958. [PMID: 38690749 DOI: 10.1080/1744666x.2024.2348605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy characterized by a dismal five-year survival rate of less than 10%. Neutrophils are key components of the innate immune system, playing a pivotal role in the PDAC immune microenvironment. AREAS COVERED This review provides a comprehensive survey of the pivotal involvement of neutrophils in the tumorigenesis and progression of PDAC. Furthermore, it synthesizes preclinical and clinical explorations aimed at targeting neutrophils within the milieu of PDAC, subsequently proposing a conceptual framework to propel further inquiry focused on enhancing the therapeutic efficacy of PDAC through neutrophil-targeted strategies. PubMed and Web of Science databases were utilized for researching neutrophils in pancreatic cancer publications prior to 2024. EXPERT OPINION Neutrophils play roles in promoting tumor growth and metastasis in PDAC and are associated with poor prognosis. However, the heterogeneity and plasticity of neutrophils and their complex relationships with other immune cells and extracellular matrix also provide new insights for immunotherapy targeting neutrophils to achieve a better prognosis for PDAC.
Collapse
Affiliation(s)
- Yi Jin
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Eric S Christenson
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Cancer Convergence Institute at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Keyu Li
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Skip Viragh Pancreatic Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
24
|
Ye QW, Liu YJ, Li JQ, Han M, Bian ZR, Chen TY, Li JP, Liu SL, Zou X. GJA4 expressed on cancer associated fibroblasts (CAFs)-A 'promoter' of the mesenchymal phenotype. Transl Oncol 2024; 46:102009. [PMID: 38833783 PMCID: PMC11190749 DOI: 10.1016/j.tranon.2024.102009] [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: 11/23/2023] [Revised: 05/09/2024] [Accepted: 05/25/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer worldwide. Connexin is a transmembrane protein involved in gap junctions (GJs) formation. Our previous study found that connexin 37 (Cx37), encoded by gap junction protein alpha 4 (GJA4), expressed on fibroblasts acts as a promoter of CRC and is closely related to epithelial-mesenchymal transition (EMT) and tumor immune microenvironment. However, to date, the mechanism concerning the malignancy of GJA4 in tumor stroma has not been studied. METHODS Hematoxylin-eosin (HE) and immunohistochemical (IHC) staining were used to validate the expression and localization of GJA4. Using single-cell analysis, enrichment analysis, spatial transcriptomics, immunofluorescence staining (IF), Sirius red staining, wound healing and transwell assays, western blotting (WB), Cell Counting Kit-8 (CCK8) assay and in vivo experiments, we investigated the possible mechanisms of GJA4 in promoting CRC. RESULTS We discovered that in CRC, GJA4 on fibroblasts is involved in promoting fibroblast activation and promoting EMT through a fibroblast-dependent pathway. Furthermore, GJA4 may act synergistically with M2 macrophages to limit T cell infiltration by stimulating the formation of an immune-excluded desmoplasic barrier. Finally, we found a significantly correlation between GJA4 and pathological staging (P < 0.0001) or D2 dimer (R = 0.03, P < 0.05). CONCLUSION We have identified GJA4 expressed on fibroblasts is actually a promoter of the tumor mesenchymal phenotype. Our findings suggest that the interaction between GJA4+ fibroblasts and M2 macrophages may be an effective target for enhancing tumor immunotherapy.
Collapse
Affiliation(s)
- Qian-Wen Ye
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, PR China; No.1 Clinical Medicial College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Yuan-Jie Liu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, PR China; No.1 Clinical Medicial College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Jia-Qi Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, PR China; No.1 Clinical Medicial College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Mei Han
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Ze-Ren Bian
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, PR China; No.1 Clinical Medicial College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Tian-Yuan Chen
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, PR China; No.1 Clinical Medicial College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Jie-Pin Li
- Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Nanjing, Jiangsu, PR China
| | - Shen-Lin Liu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, PR China.
| | - Xi Zou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, Nanjing, Jiangsu, PR China.
| |
Collapse
|
25
|
Lv J, Wang Z, Wang B, Deng C, Wang W, Sun L. S100A9 Induces Macrophage M2 Polarization and Immunomodulatory Role in the Lesion Site After Spinal Cord Injury in Rats. Mol Neurobiol 2024; 61:5525-5540. [PMID: 38206470 DOI: 10.1007/s12035-024-03920-3] [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: 09/23/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024]
Abstract
Immune response is pivotal in the secondary injury of spinal cord injury (SCI). Polarization of macrophages (MΦ) influences the immune response in the secondary injury, which is regulated by several immune-related proteins. M2Φ plays the immunomodulatory role in the central nervous system. This study used bioinformatic analysis and machine algorithms to screen hub immune-related proteins after SCI and experimentally investigate the role of the target protein in the M2Φ polarization and immunomodulation in rats and in vitro after SCI. We downloaded GSE151371 and GSE45006, hub immune-related genes were screened using machine learning algorithms, and the expression of S100A9 was verified by datasets. Allen's weight-drop injury SCI model in Sprague-Dawley rat and bone marrow-derived rat MΦ with myelin debris model were used to study the effects of S100A9 on M2Φ polarization and immunomodulation at the lesion site and in vitro. Bioinformatic analysis showed that S100A9 acts as a hub immune-related gene in the SCI patients and rats. S100A9 increased at the lesion site in SCI rats, and its inhibition reduced CD206 and ARG-1 expression. Exogenous S100A9 promoted CD206 and ARG-1 expression in MΦ. S100A9 also increased the expression of PD-L1 and decreased MHC II at the lesion site in SCI rats and MΦ with myelin debris, and enhanced mitochondrial activity in rat MΦ with myelin debris. In conclusion, S100A9 is an indispensable factor in the immune process in secondary injury following SCI.
Collapse
Affiliation(s)
- Junqiao Lv
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Zhiqiang Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Beiyang Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Chen Deng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Wei Wang
- Department of Urology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Lin Sun
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China.
| |
Collapse
|
26
|
Chen Z, Zhang D, Huang H, Chen J, Li Z, Hu Y, Liu R. NIR Absorbing Organic Chromophores Combination with NSAIDs for Remodeling of the Inflammatory Microenvironment to Amplify Tumor Ferroptosis-Photothermal Synergistic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400361. [PMID: 38708879 DOI: 10.1002/smll.202400361] [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: 01/16/2024] [Revised: 02/29/2024] [Indexed: 05/07/2024]
Abstract
Photothermal therapy has emerged as a promising approach for cancer treatment, which can cause ferroptosis to enhance immunotherapeutic efficacy. However, excessively generated immunogenicity will induce serious inflammatory response syndrome, resulting in a discounted therapeutic effect. Herein, a kind of NIR absorption small organic chromophore nanoparticles (TTHM NPs) with high photothermal conversion efficiency (68.33%) is developed, which can induce mitochondria dysfunction, generate mitochondrial superoxide, and following ferroptosis. TTHM NPs-based photothermal therapy is combined with Sulfasalazine (SUZ), a kind of nonsteroidal anti-inflammatory drugs, to weaken inflammation and promote ferroptosis through suppressing glutamate/cystine (Glu/Cys) antiporter system Xc- (xCT). Additionally, the combination of SUZ with PTT can induce immunogenic cell death (ICD), followed by promoting the maturation of DCs and the attraction of CD8+ T cell, which will secrete IFN-γ and trigger self-amplified ferroptosis via inhibiting xCT and simulating Acyl-CoA synthetase long-chain family member 4 (ACSL4). Moreover, the in vivo results demonstrate that this combination therapy can suppress the expression of inflammatory factors, enhance dendritic cell activation, facilitate T-cell infiltration, and realize effective thermal elimination of primary tumors and distant tumors. In general, this work provides an excellent example of combined medication and stimulates new thinking about onco-therapy and inflammatory response.
Collapse
Affiliation(s)
- Zhian Chen
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Di Zhang
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Huilin Huang
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jian Chen
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Zhenhao Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Yanfeng Hu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Ruiyuan Liu
- Guangdong Provincial Key Laboratory of Medical Image Processing, School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| |
Collapse
|
27
|
Sun F, Ding Z, Shao F, Gao X, Tian H, Zhang X, Chen H, Wang C. Albumin-Based MUC13 Peptide Nanomedicine Suppresses Liver Cancer Stem Cells via JNK-ERK Signaling Pathway-Mediated Autophagy Inhibition. ACS APPLIED MATERIALS & INTERFACES 2024; 16:38968-38978. [PMID: 39024013 DOI: 10.1021/acsami.4c06034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Targeting liver cancer stem cells (LCSCs) is a promising strategy for hepatocellular carcinoma (HCC) therapy. Target selection and corresponding inhibitor screening are of vital importance for eliminating the stemness of LCSCs. Peptide-based agents are hopeful but have long been hindered for in vivo application. Herein, we selected a clinically significant target MUC13 and screened out a suitable peptide for preparation of an albumin-based MUC13 peptide nanomedicine, P3@HSA, which suppressed liver cancer stem cells via JNK-ERK signaling pathway-mediated autophagy inhibition. The selected target MUC13 was highly expressed in LCSCs and associated with the prognosis of liver cancer patients. Encouraged by this observation, we screened the corresponding peptide-based inhibitor P3 for further evaluation. P3 could interact with albumin through the intrinsic hydrophobic force and formed the nanomedicine P3@HSA. The prepared nanomedicine could inhibit LCSCs through JNK-ERK signaling pathway-mediated autophagy inhibition and exert potent antitumor effect both in vitro and in vivo. Together, this study provides a promising peptide-based nanomedicine for high-performance HCC treatment.
Collapse
Affiliation(s)
- Fen Sun
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zongyao Ding
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Fengying Shao
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiaoyang Gao
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Haina Tian
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Xiao Zhang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huaqing Chen
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Changlong Wang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an 271016, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| |
Collapse
|
28
|
Reddy SU, Sadia FZ, Vancura A, Vancurova I. IFNγ-Induced Bcl3, PD-L1 and IL-8 Signaling in Ovarian Cancer: Mechanisms and Clinical Significance. Cancers (Basel) 2024; 16:2676. [PMID: 39123403 PMCID: PMC11311860 DOI: 10.3390/cancers16152676] [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: 05/19/2024] [Revised: 07/18/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
IFNγ, a pleiotropic cytokine produced not only by activated lymphocytes but also in response to cancer immunotherapies, has both antitumor and tumor-promoting functions. In ovarian cancer (OC) cells, the tumor-promoting functions of IFNγ are mediated by IFNγ-induced expression of Bcl3, PD-L1 and IL-8/CXCL8, which have long been known to have critical cellular functions as a proto-oncogene, an immune checkpoint ligand and a chemoattractant, respectively. However, overwhelming evidence has demonstrated that these three genes have tumor-promoting roles far beyond their originally identified functions. These tumor-promoting mechanisms include increased cancer cell proliferation, invasion, angiogenesis, metastasis, resistance to chemotherapy and immune escape. Recent studies have shown that IFNγ-induced Bcl3, PD-L1 and IL-8 expression is regulated by the same JAK1/STAT1 signaling pathway: IFNγ induces the expression of Bcl3, which then promotes the expression of PD-L1 and IL-8 in OC cells, resulting in their increased proliferation and migration. In this review, we summarize the recent findings on how IFNγ affects the tumor microenvironment and promotes tumor progression, with a special focus on ovarian cancer and on Bcl3, PD-L1 and IL-8/CXCL8 signaling. We also discuss promising novel combinatorial strategies in clinical trials targeting Bcl3, PD-L1 and IL-8 to increase the effectiveness of cancer immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Ivana Vancurova
- Department of Biological Sciences, St. John’s University, New York, NY 11439, USA; (S.U.R.); (F.Z.S.); (A.V.)
| |
Collapse
|
29
|
Si H, Fu X, Hao Y, Wang Y, Lin G, Wang D, Xu C, Zhang Y, Song Z. The influence of PD-L1 expression levels on the efficacy of combination therapy in thymic epithelial tumors. Clin Transl Oncol 2024:10.1007/s12094-024-03618-x. [PMID: 39046681 DOI: 10.1007/s12094-024-03618-x] [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: 04/07/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024]
Abstract
BACKGROUND The significant expression of PD-L1 in thymic epithelial tumors (TETs) has been confirmed, and immunotherapy and its combination therapy have been effective in TETs. However, there is no present evidence that the expression levels of PD-L1 affects the efficacy of combination therapy. Our study aimed to shed light on this relationship. METHODS Patients with thymic epithelial tumors (TETs) from multicenter hospitals were retrospectively identified. Objective response rate (ORR), progression-free survival (PFS), overall survival (OS), and immune-related adverse events (irAEs) in 22 patients were included. We divided the patients the 22 patients with PD-L1 test into three levels (high expression, low expression and no expression) and analyzed the relationship between the levels of PD-L1 expression and the efficacy of combination therapy. RESULTS Combination therapy showed an effective benefit in 22 patients with TETs, the median PFS (mPFS) was 16 months (95% CI: 8.5-23.5) and the median OS (mOS) was 38 months (95% CI: 21.5-54.5). Cox-regressive analysis found whether PD-L1 expression affected the PFS of patients (p = 0.017). Among the patients with PD-L1 expression, the levels of expression were correlated with curative effect (Kruskal-Wallis test, PFS: P = 0.012; OS: P = 0.01), and high expression group was along with better efficacy than low expression (Wilcoxon test, P = 0.01). Moreover, in 17 patients treated with immunotherapy combined with chemotherapy, the expression of PD-L1 was also associated with efficacy (Kruskal-Wallis test, p = 0.021). CONCLUSIONS PD-L1 expression affects the PFS of patients. High expression of PD-L1 patients with TETs responded better to combination therapy, which could provide a therapeutic option in clinic. Besides, other targeted treatments should be considered.
Collapse
Affiliation(s)
- Han Si
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
| | - Xiaoshuang Fu
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
| | - Yue Hao
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China
| | - Yina Wang
- Department of Oncology, College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310000, China
| | - Gen Lin
- Department of Medical Oncology, Fujian Medical University Cancer Hospital & Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Dong Wang
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210000, China
| | - Chunwei Xu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Zhongbo Song
- Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, China.
- Department of Clinical Trial, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No.1 East Banshan Road, Gongshu District, Hangzhou, 310022, Zhejiang, China.
| |
Collapse
|
30
|
Feng YYF, Li YC, Liu HM, Xu R, Liu YT, Zhang W, Yang HY, Chen G. Synthetic lethal CRISPR screen identifies a cancer cell-intrinsic role of PD-L1 in regulation of vulnerability to ferroptosis. Cell Rep 2024; 43:114477. [PMID: 38985676 DOI: 10.1016/j.celrep.2024.114477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 05/20/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Despite the success of programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) inhibition in tumor therapy, many patients do not benefit. This failure may be attributed to the intrinsic functions of PD-L1. We perform a genome-wide CRISPR synthetic lethality screen to systematically explore the intrinsic functions of PD-L1 in head and neck squamous cell carcinoma (HNSCC) cells, identifying ferroptosis-related genes as essential for the viability of PD-L1-deficient cells. Genetic and pharmacological induction of ferroptosis accelerates cell death in PD-L1 knockout cells, which are also more susceptible to immunogenic ferroptosis. Mechanistically, nuclear PD-L1 transcriptionally activates SOD2 to maintain redox homeostasis. Lower reactive oxygen species (ROS) and ferroptosis are observed in patients with HNSCC who have higher PD-L1 expression. Our study illustrates that PD-L1 confers ferroptosis resistance in HNSCC cells by activating the SOD2-mediated antioxidant pathway, suggesting that targeting the intrinsic functions of PD-L1 could enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Yang-Ying-Fan Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yi-Cun Li
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Guangdong 518036, China; Guangdong Provincial High-level Clinical Key Specialty, Guangdong 518036, China; Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Guangdong 518036, China; The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong 518036, China
| | - Hai-Ming Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Rui Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yu-Tong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Wei Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Hong-Yu Yang
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital, Guangdong 518036, China; Guangdong Provincial High-level Clinical Key Specialty, Guangdong 518036, China; Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Guangdong 518036, China; The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong 518036, China.
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China; TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
31
|
Zhang Y, Chen X, Hu B, Zou B, Xu Y. Advancements in nanomedicine delivery systems: unraveling immune regulation strategies for tumor immunotherapy. Nanomedicine (Lond) 2024; 19:1821-1840. [PMID: 39011582 PMCID: PMC11418288 DOI: 10.1080/17435889.2024.2374230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/26/2024] [Indexed: 07/17/2024] Open
Abstract
This review highlights the significant role of nanodrug delivery systems (NDDS) in enhancing the efficacy of tumor immunotherapy. Focusing on the integration of NDDS with immune regulation strategies, it explores their transformative impacts on the tumor microenvironment and immune response dynamics. Key advancements include the optimization of drug delivery through NDDS, targeting mechanisms like immune checkpoint blockade and modulating the immunosuppressive tumor environment. Despite the progress, challenges such as limited clinical efficacy and complex manufacturing processes persist. The review emphasizes the need for further research to optimize these systems, potentially revolutionizing cancer treatment by improving delivery efficiency, reducing toxicity and overcoming immune resistance.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Xi Chen
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Binbin Hu
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Bingwen Zou
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| | - Yong Xu
- Department of Radiation Oncology, Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu610041, P.R. China
| |
Collapse
|
32
|
Luo S, Li S, Liu C, Yu D, Sun L, Zhang S, Zhao N, Zhang M, Nie J, Zhao Y, Li C, Zhang Y, Zhang Q, Meng H, Li X, Shi J, Zheng T. Stage-specificity of STING activation in intrahepatic cholangiocarcinoma determines the efficacy of its agonism. Cancer Lett 2024; 594:216992. [PMID: 38797231 DOI: 10.1016/j.canlet.2024.216992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/11/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Intrahepatic cholangiocarcinoma (iCCA) is an aggressive cancer with an extremely poor prognosis, and new treatment options are needed. Recently, immunotherapy has emerged as an efficient treatment against malignant tumors, but less effective in iCCA. Activation of stimulator of interferon genes (STING) signaling could reignite immunologically inert tumors, but the expression and role of STING in iCCA remains to be determined. Here, we show STING is expressed in iCCA, and patients with high expression of STING in early-stage iCCA have a longer overall survival than those have low expression. Increased immune cell infiltration in early-stage iCCA corresponds to elevated STING expression. In mice iCCA models, treatment with the STING agonist MSA-2 show stage-specific inhibitory effects on tumors, with beneficial effects in early-stage tumors but not with advanced-stage cancer. This discrepancy was associated with greater programmed cell death ligand 1 (PD-L1) expression in advanced-stage tumors. Combination therapy targeting PD-L1 and MSA-2 strikingly reduced tumor burden in such tumors compared to either monotherapy. Cumulatively, these data demonstrate that STING agonism monotherapy improves the immune landscape of the tumor microenvironment in early-stage iCCA, while combination therapy ameliorates advanced-stage iCCA.
Collapse
Affiliation(s)
- Shengnan Luo
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Shun Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Caiqi Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Dongyu Yu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Linlin Sun
- Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Shuyuan Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Na Zhao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Meng Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Jianhua Nie
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Ying Zhao
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Chunyue Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China
| | - Yan Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Qian Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, 150 Haping Road, Nangang District, Harbin, 150081, PR China
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, 157 Baojian Road, Nangang District, Harbin, 150081, PR China.
| | - Jiaqi Shi
- Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China; Department of Phase 1 Trials Center, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China.
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China; Heilongjiang Province Key Laboratory of Molecular Oncology, No. 150 Haping Road, Nangang District, Harbin, Heilongjiang Province, 150081, PR China; Department of Phase 1 Trials Center, Harbin Medical University Cancer Hospital, No. 150 Haping Road, Nangang District, Harbin, 150081, PR China.
| |
Collapse
|
33
|
Hodgins JJ, Abou-Hamad J, O’Dwyer CE, Hagerman A, Yakubovich E, Tanese de Souza C, Marotel M, Buchler A, Fadel S, Park MM, Fong-McMaster C, Crupi MF, Makinson OJ, Kurdieh R, Rezaei R, Dhillon HS, Ilkow CS, Bell JC, Harper ME, Rotstein BH, Auer RC, Vanderhyden BC, Sabourin LA, Bourgeois-Daigneault MC, Cook DP, Ardolino M. PD-L1 promotes oncolytic virus infection via a metabolic shift that inhibits the type I IFN pathway. J Exp Med 2024; 221:e20221721. [PMID: 38869480 PMCID: PMC11176258 DOI: 10.1084/jem.20221721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/04/2024] [Accepted: 03/14/2024] [Indexed: 06/14/2024] Open
Abstract
While conventional wisdom initially postulated that PD-L1 serves as the inert ligand for PD-1, an emerging body of literature suggests that PD-L1 has cell-intrinsic functions in immune and cancer cells. In line with these studies, here we show that engagement of PD-L1 via cellular ligands or agonistic antibodies, including those used in the clinic, potently inhibits the type I interferon pathway in cancer cells. Hampered type I interferon responses in PD-L1-expressing cancer cells resulted in enhanced efficacy of oncolytic viruses in vitro and in vivo. Consistently, PD-L1 expression marked tumor explants from cancer patients that were best infected by oncolytic viruses. Mechanistically, PD-L1 promoted a metabolic shift characterized by enhanced glycolysis rate that resulted in increased lactate production. In turn, lactate inhibited type I IFN responses. In addition to adding mechanistic insight into PD-L1 intrinsic function, our results will also help guide the numerous ongoing efforts to combine PD-L1 antibodies with oncolytic virotherapy in clinical trials.
Collapse
Affiliation(s)
- Jonathan J. Hodgins
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - John Abou-Hamad
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Colin Edward O’Dwyer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Ash Hagerman
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Edward Yakubovich
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | | | - Marie Marotel
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Ariel Buchler
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
- University of Ottawa Heart Institute, Ottawa, Canada
| | - Saleh Fadel
- The Ottawa Hospital, Ottawa, Canada
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Maria M. Park
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Claire Fong-McMaster
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Ottawa Institute for Systems Biology, Ottawa, Canada
| | - Mathieu F. Crupi
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Olivia Joan Makinson
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Reem Kurdieh
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Reza Rezaei
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Harkirat Singh Dhillon
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Carolina S. Ilkow
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - John C. Bell
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
- Ottawa Institute for Systems Biology, Ottawa, Canada
| | - Benjamin H. Rotstein
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
- University of Ottawa Heart Institute, Ottawa, Canada
| | - Rebecca C. Auer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Barbara C. Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Luc A. Sabourin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Department of Microbiology, Infectious Diseases, and Immunology, University of Montreal, Montreal, Canada
- Centre Hospitalier de l’Université de Montréal Research Centre, Cancer and Immunopathology axes, Montreal, Canada
| | - David P. Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Michele Ardolino
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| |
Collapse
|
34
|
Jeong H, Koh J, Kim S, Song SG, Lee SH, Jeon Y, Lee CH, Keam B, Lee SH, Chung DH, Jeon YK. Epithelial-mesenchymal transition induced by tumor cell-intrinsic PD-L1 signaling predicts a poor response to immune checkpoint inhibitors in PD-L1-high lung cancer. Br J Cancer 2024; 131:23-36. [PMID: 38729997 PMCID: PMC11231337 DOI: 10.1038/s41416-024-02698-4] [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: 08/01/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND We investigated the role of tumor cell-intrinsic PD-L1 signaling in the epithelial-mesenchymal transition (EMT) in non-small-cell lung cancer (NSCLC) and the role of EMT as a predictive biomarker for immune checkpoint inhibitor (ICI) therapy. METHODS PD-L1-overexpressing or PD-L1-knockdown NSCLC cells underwent RNA-seq and EMT phenotype assessment. Mouse lung cancer LLC cells were injected into nude mice. Two cohorts of patients with NSCLC undergoing ICI therapy were analyzed. RESULTS RNA-seq showed that EMT pathways were enriched in PD-L1-high NSCLC cells. EMT was enhanced by PD-L1 in NSCLC cells, which was mediated by transforming growth factor-β (TGFβ). PD-L1 promoted the activation of p38-MAPK by binding to and inhibiting the protein phosphatase PPM1B, thereby increasing the TGFβ production. Tumor growth and metastasis increased in nude mice injected with PD-L1-overexpressing LLC cells. In the ICI cohort, EMT signature was higher in patients with progressive disease than in those with responses, and EMT was significantly associated with poor survival in PD-L1-high NSCLC. In PD-L1-high NSCLC, EMT was associated with increased M2-macrophage and regulatory T-cell infiltrations and decreased cytotoxic T-cell infiltration. CONCLUSIONS Tumor cell-intrinsic PD-L1 function contributes to NSCLC progression by promoting EMT. EMT may predict an unfavorable outcome after ICI therapy in PD-L1-high NSCLC.
Collapse
Affiliation(s)
- Hyein Jeong
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Interdiscipilinary Program of Cancer Biology, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Jaemoon Koh
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sehui Kim
- Department of Pathology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seung Geun Song
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soo Hyun Lee
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Youngjoo Jeon
- Interdiscipilinary Program of Cancer Biology, Seoul National University Graduate School, Seoul, Republic of Korea
| | - Chul-Hwan Lee
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Bhumsuk Keam
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology, Sungkyunkwan University, Seoul, Republic of Korea
| | - Doo Hyun Chung
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yoon Kyung Jeon
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea.
- Interdiscipilinary Program of Cancer Biology, Seoul National University Graduate School, Seoul, Republic of Korea.
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
35
|
Zhou Z, Jiang X, Yi L, Li C, Wang H, Xiong W, Li Z, Shen J. Mitochondria Energy Metabolism Depression as Novel Adjuvant to Sensitize Radiotherapy and Inhibit Radiation Induced-Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401394. [PMID: 38715382 PMCID: PMC11234447 DOI: 10.1002/advs.202401394] [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: 02/07/2024] [Indexed: 07/11/2024]
Abstract
Currently, the typical combination therapy of programmed death ligand-1 (PD-L1) antibodies with radiotherapy (RT) still exhibits impaired immunogenic antitumor response in clinical due to lessened DNA damage and acquired immune tolerance via the upregulation of some other immune checkpoint inhibitors. Apart from this, such combination therapy may raise the occurrence rate of radiation-induced lung fibrosis (RIPF) due to enhanced systemic inflammation, leading to the ultimate death of cancer patients (average survival time of about 3 years). Therefore, it is newly revealed that mitochondria energy metabolism regulation can be used as a novel effective PD-L1 and transforming growth factor-β (TGF-β) dual-downregulation method. Following this, IR-TAM is prepared by conjugating mitochondria-targeted heptamethine cyanine dye IR-68 with oxidative phosphorylation (OXPHOS) inhibitor Tamoxifen (TAM), which then self-assembled with albumin (Alb) to form IR-TAM@Alb nanoparticles. By doing this, tumor-targeting IR-TAM@Alb nanoparticle effectively reversed tumor hypoxia and depressed PD-L1 and TGF-β expression to sensitize RT. Meanwhile, due to the capacity of heptamethine cyanine dye in targeting RIPF and the function of TAM in depressing TGF-β, IR-TAM@Alb also ameliorated fibrosis development induced by RT.
Collapse
Affiliation(s)
- Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xin Jiang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lei Yi
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Cheng Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Haoxiang Wang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wei Xiong
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, 410013, China
| | - Zhipeng Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|
36
|
Liu YJ, Li JP, Han M, Li JX, Ye QW, Lin ST, Zhou JY, Liu SL, Zou X. IFIT1 + neutrophil is a causative factor of immunosuppressive features of poorly cohesive carcinoma (PCC). J Transl Med 2024; 22:580. [PMID: 38898490 PMCID: PMC11188200 DOI: 10.1186/s12967-024-05389-z] [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: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024] Open
Abstract
The importance of the immune microenvironment in poorly cohesive carcinoma (PCC) has been highlighted due to its limited response rate to conventional therapy and emerging treatment resistance. A combination of clinical cohorts, bioinformatics analyses, and functional/molecular experiments revealed that high infiltration of Interferon Induced Protein with Tetratricopeptide Repeats 1 (IFIT1) + tumor-associated neutrophils (TANs) is a distinguishing feature of PCC patients. Upregulation of IFIT1 + TANs promote migration and invasion of gastric cancer (GC) cell lines (MKN45 and MKN74) and stimulates the growth of cell-derived xenograft models. Besides, by promoting macrophage secreted phosphoprotein 1 (SPP1) expression and facilitating cancer-associated fibroblast and endothelial cell recruitment and activation through TANs, IFIT1 promotes a mesenchymal phenotype, which is associated with a poor prognosis. Importantly, compared to non-PCC (NPCC), PCC tumors is more immunosuppressive. Mechanistically, IFIT1 can be stimulated by IFN-γ and contributes to the expression of Programmed Cell Death 1 Ligand (PDL1) in TANs. We demonstrated in mouse models that IFIT1 + PDL1 + TANs can induce acquired resistance to anti-PD-1 immunotherapy, which may be responsible for the difficulty of PCC patients to benefit from immunotherapy. This work highlights the role of IFIT1 + TANs in mediating the remodeling of the tumor immune microenvironment and immunotherapeutic resistance and introduces IFIT1 + TANs as a promising target for precision therapy of PCC.
Collapse
Affiliation(s)
- Yuan-Jie Liu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
- Key Laboratory of Tumor System Biology of Traditional Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jie-Pin Li
- Key Laboratory of Tumor System Biology of Traditional Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Mei Han
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Jing-Xiao Li
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Qian-Wen Ye
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Si-Tian Lin
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Jin-Yong Zhou
- Central Laboratory, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Shen-Lin Liu
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
| | - Xi Zou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, Jiangsu, China.
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of Tumor, Nanjing, 210029, Jiangsu, China.
- Key Laboratory of Tumor System Biology of Traditional Chinese Medicine, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
37
|
Liu Z, Li L, Liu L, Zhu Z, Yu Y, Zhan S, Luo Z, Li Y, Long H, Cai J, Huang K, Du X. PDK3 drives colorectal carcinogenesis and immune evasion and is a therapeutic target for boosting immunotherapy. Am J Cancer Res 2024; 14:3117-3129. [PMID: 39005672 PMCID: PMC11236793 DOI: 10.62347/qwkz3078] [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: 04/20/2024] [Accepted: 05/27/2024] [Indexed: 07/16/2024] Open
Abstract
Pyruvate Dehydrogenase Kinase 3 (PDK3) has emerged as a significant player in various cancer types, yet its specific impact on cancers including colon cancer remains ambiguous. Through pan-cancer analysis using TCGA data, we found that the expression of PDK3 and the composition of the immune microenvironment for different tumors were highly heterogeneous across tumors. PDK3 is highly expressed in colorectal cancer and may promote tumor proliferation by activating PI3K-AKT signaling. In addition, we found that PDK3 was able to inhibit tumor antigen presentation signals to suppress immune killing. High PDK3 expression predicts less CD8+ T cell infiltration and effector function. Moreover, inhibition of PDK3 expression bolstered CD8+ T cell-mediated cytotoxicity CD8+ T cell infiltration and activation in vivo. Notably, PDK3 was found to facilitate STAT1 activation and elevate programmed death-ligand 1 (PD-L1) expression in colon cancer cells. Importantly, PDK3 inhibition combination with PD-1 blockade significantly activates the infiltrated CD8+ T cells to suppress tumor growth and improves the survival benefit in several murine tumor models. In summary, these findings underscore PDK3's role in fueling colon cancer growth by orchestrating PI3K-AKT signaling and PD-L1 expression and dampening CD8+ T cell function.
Collapse
Affiliation(s)
- Zhiqiang Liu
- Department of Neurology, Jiangxi Provincial People's Hospital, The First Affiliated Hospital of Nanchang Medical College Nanchang 330006, Jiangxi, China
| | - Liang Li
- Department of Emergency, Jiangxi Maternal and Child Health Hospital Nanchang 330006, Jiangxi, China
| | - Lei Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Zhengming Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Yi Yu
- Department of Urology Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Shaochao Zhan
- Department of Urology Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Zepeng Luo
- Department of Urology Surgery, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Yuchen Li
- Department of Gastrointestinal Surgery, Jiangxi Provincial Cancer Hospital Nanchang 330029, Jiangxi, China
| | - He Long
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| | - Kai Huang
- Department of Gastrointestinal Surgery, Jiangxi Provincial Cancer Hospital Nanchang 330029, Jiangxi, China
| | - Xiaohong Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University Nanchang 330006, Jiangxi, China
| |
Collapse
|
38
|
Xia J, Zhou Y, Wang Y, Liu Y, Chen Q, Koh K, Hu X, Chen H. Ultrasensitive electrochemical sensor based on synergistic effect of Ag@MXene and antifouling cyclic multifunctional peptide for PD-L1 detection in serum. Mikrochim Acta 2024; 191:380. [PMID: 38858258 DOI: 10.1007/s00604-024-06470-6] [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: 02/29/2024] [Accepted: 05/26/2024] [Indexed: 06/12/2024]
Abstract
A sensing interface co-constructed from the two-dimensional conductive material (Ag@MXene) and an antifouling cyclic multifunctional peptide (CP) is described. While the large surface area of Ag@MXene loads more CP probes, CP binds to Ag@MXene to form a fouling barrier and ensure the structural rigidity of the targeting sequence. This strategy synergistically enhances the biosensor's sensitivity and resistance to contamination. The SPR results showed that the binding affinity of the CP to the target was 6.23 times higher than that of the antifouling straight-chain multifunctional peptide (SP) to the target. In the 10 mg/mL BSA electrochemical fouling test, the fouling resistance of Ag@MXene + CP (composite sensing interface of CP combined with Ag@MXene) was 30 times higher than that of the bare electrode. The designed electrochemical sensor exhibited good selectivity and wide dynamic response range at PD-L1 concentrations from 0.1 to 50 ng/mL. The lowest detection limit was 24.54 pg/mL (S/N = 3). Antifouling 2D materials with a substantial specific surface area, coupled with non-straight chain antifouling multifunctional peptides, offer a wide scope for investigating the sensitivity and antifouling properties of electrochemical sensors.
Collapse
Affiliation(s)
- Junjie Xia
- School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yangyang Zhou
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yindian Wang
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yawen Liu
- School of Medicine, Shanghai University, Shanghai, 200444, China
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Qiang Chen
- School of Medicine, Shanghai University, Shanghai, 200444, China
| | - Kwangnak Koh
- Institute of General Education, Pusan National University, Busan, 609-735, Republic of Korea
| | - Xiaojun Hu
- School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| | - Hongxia Chen
- School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
39
|
Salminen A. The role of the immunosuppressive PD-1/PD-L1 checkpoint pathway in the aging process and age-related diseases. J Mol Med (Berl) 2024; 102:733-750. [PMID: 38600305 PMCID: PMC11106179 DOI: 10.1007/s00109-024-02444-6] [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: 02/15/2024] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024]
Abstract
The accumulation of senescent cells within tissues is a hallmark of the aging process. Senescent cells are also commonly present in many age-related diseases and in the cancer microenvironment. The escape of abnormal cells from immune surveillance indicates that there is some defect in the function of cytotoxic immune cells, e.g., CD8+ T cells and natural killer (NK) cells. Recent studies have revealed that the expression of programmed death-ligand 1 (PD-L1) protein is abundantly increased in senescent cells. An increase in the amount of PD-L1 protein protects senescent cells from clearance by the PD-1 checkpoint receptor in cytotoxic immune cells. In fact, the activation of the PD-1 receptor suppresses the cytotoxic properties of CD8+ T and NK cells, promoting a state of immunosenescence. The inhibitory PD-1/PD-L1 checkpoint pathway acts in cooperation with immunosuppressive cells; for example, activation of PD-1 receptor can enhance the differentiation of regulatory T cells (Treg), myeloid-derived suppressor cells (MDSC), and M2 macrophages, whereas the cytokines secreted by immunosuppressive cells stimulate the expression of the immunosuppressive PD-L1 protein. Interestingly, many signaling pathways known to promote cellular senescence and the aging process are crucial stimulators of the expression of PD-L1 protein, e.g., epigenetic regulation, inflammatory mediators, mTOR-related signaling, cGAS-STING pathway, and AhR signaling. It seems that the inhibitory PD-1/PD-L1 immune checkpoint axis has a crucial role in the accumulation of senescent cells and thus it promotes the aging process in tissues. Thus, the blockade of the PD-1/PD-L1 checkpoint signaling might be a potential anti-aging senolytic therapy. KEY MESSAGES: Senescent cells accumulate within tissues during aging and age-related diseases. Senescent cells are able to escape immune surveillance by cytotoxic immune cells. Expression of programmed death-ligand 1 (PD-L1) markedly increases in senescent cells. Age-related signaling stimulates the expression of PD-L1 protein in senescent cells. Inhibitory PD-1/PD-L1 checkpoint pathway suppresses clearance of senescent cells.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
40
|
Wang Y, Zhang C, Yan M, Ma X, Song L, Wang B, Li P, Liu P. PD-L1 regulates tumor proliferation and T-cell function in NF2-associated meningiomas. CNS Neurosci Ther 2024; 30:e14784. [PMID: 38828669 PMCID: PMC11145367 DOI: 10.1111/cns.14784] [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: 07/07/2023] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 06/05/2024] Open
Abstract
INTRODUCTION Programmed death-ligand 1 (PD-L1) expression is an immune evasion mechanism that has been demonstrated in many tumors and is commonly associated with a poor prognosis. Over the years, anti-PD-L1 agents have gained attention as novel anticancer therapeutics that induce durable tumor regression in numerous malignancies. They may be a new treatment choice for neurofibromatosis type 2 (NF2) patients. AIMS The aims of this study were to detect the expression of PD-L1 in NF2-associated meningiomas, explore the effect of PD-L1 downregulation on tumor cell characteristics and T-cell functions, and investigate the possible pathways that regulate PD-L1 expression to further dissect the possible mechanism of immune suppression in NF2 tumors and to provide new treatment options for NF2 patients. RESULTS PD-L1 is heterogeneously expressed in NF2-associated meningiomas. After PD-L1 knockdown in NF2-associated meningioma cells, tumor cell proliferation was significantly inhibited, and the apoptosis rate was elevated. When T cells were cocultured with siPD-L1-transfected NF2-associated meningioma cells, the expression of CD69 on both CD4+ and CD8+ T cells was partly reversed, and the capacity of CD8+ T cells to kill siPD-L1-transfected tumor cells was partly restored. Results also showed that the PI3K-AKT-mTOR pathway regulates PD-L1 expression, and the mTOR inhibitor rapamycin rapidly and persistently suppresses PD-L1 expression. In vivo experimental results suggested that anti-PD-L1 antibody may have a synergetic effect with the mTOR inhibitor in reducing tumor cell proliferation and that reduced PD-L1 expression could contribute to antitumor efficacy. CONCLUSIONS Targeting PD-L1 could be helpful for restoring the function of tumor-infiltrating lymphocytes and inducing apoptosis to inhibit tumor proliferation in NF2-associated meningiomas. Dissecting the mechanisms of the PD-L1-driven tumorigenesis of NF2-associated meningioma will help to improve our understanding of the mechanisms underlying tumor progression and could facilitate further refinement of current therapies to improve the treatment of NF2 patients.
Collapse
Affiliation(s)
- Ying Wang
- Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Chao Zhang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Minjun Yan
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Xin Ma
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Lairong Song
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Bo Wang
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Peng Li
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Pinan Liu
- Beijing Neurosurgical InstituteCapital Medical UniversityBeijingChina
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
41
|
Fan J, Zhu J, Zhu H, Xu H. Potential therapeutic targets in myeloid cell therapy for overcoming chemoresistance and immune suppression in gastrointestinal tumors. Crit Rev Oncol Hematol 2024; 198:104362. [PMID: 38614267 DOI: 10.1016/j.critrevonc.2024.104362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/26/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024] Open
Abstract
In the tumor microenvironment (TME), myeloid cells play a pivotal role. Myeloid-derived immunosuppressive cells, including tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), are central components in shaping the immunosuppressive milieu of the tumor. Within the TME, a majority of TAMs assume an M2 phenotype, characterized by their pro-tumoral activity. These cells promote tumor cell growth, angiogenesis, invasion, and migration. In contrast, M1 macrophages, under appropriate activation conditions, exhibit cytotoxic capabilities against cancer cells. However, an excessive M1 response may lead to pro-tumoral inflammation. As a result, myeloid cells have emerged as crucial targets in cancer therapy. This review concentrates on gastrointestinal tumors, detailing methods for targeting macrophages to enhance tumor radiotherapy and immunotherapy sensitivity. We specifically delve into monocytes and tumor-associated macrophages' various functions, establishing an immunosuppressive microenvironment, promoting tumorigenic inflammation, and fostering neovascularization and stromal remodeling. Additionally, we examine combination therapeutic strategies.
Collapse
Affiliation(s)
- Jiawei Fan
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - Jianshu Zhu
- Department of Spine Surgery, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - He Zhu
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China
| | - Hong Xu
- Department of Gastroenterology, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130021, PR China.
| |
Collapse
|
42
|
Chen F, Sheng J, Li X, Gao Z, Zhao S, Hu L, Chen M, Fei J, Song Z. Unveiling the promise of PD1/PD-L1: A new dawn in immunotherapy for cholangiocarcinoma. Biomed Pharmacother 2024; 175:116659. [PMID: 38692063 DOI: 10.1016/j.biopha.2024.116659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
Cholangiocarcinoma (CCA), a rare yet notably aggressive cancer, has experienced a surge in incidence in recent years. Presently, surgical resection remains the most effective curative strategy for CCA. Nevertheless, a majority of patients with CCA are ineligible for surgical removal at the time of diagnosis. For advanced stages of CCA, the combination of gemcitabine and cisplatin is established as the standard chemotherapy regimen. Despite this, treatment efficacy is often hindered by the development of resistance. In recent times, immune checkpoint inhibitors, particularly those that block programmed death 1 and its ligand (PD1/PD-L1), have emerged as promising strategies against a variety of cancers and are being increasingly integrated into the therapeutic landscape of CCA. A growing body of research supports that the use of PD1/PD-L1 monoclonal antibodies in conjunction with chemotherapy may significantly improve patient outcomes. This article seeks to meticulously review the latest studies on PD1/PD-L1 involvement in CCA, delving into their expression profiles, prognostic significance, contribution to oncogenic processes, and their potential clinical utility.
Collapse
Affiliation(s)
- Fei Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Jian Sheng
- Department of Research and Teaching, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoping Li
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhaofeng Gao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Siqi Zhao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Lingyu Hu
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Minjie Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Jianguo Fei
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Zhengwei Song
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
43
|
Ren Z, Xu Z, Chang X, Liu J, Xiao W. STC1 competitively binding βPIX enhances melanoma progression via YAP nuclear translocation and M2 macrophage recruitment through the YAP/CCL2/VEGFA/AKT feedback loop. Pharmacol Res 2024; 204:107218. [PMID: 38768671 DOI: 10.1016/j.phrs.2024.107218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/22/2024]
Abstract
This study investigates the role of Stanniocalcin-1 (STC1) in melanoma progression, with a focus on its impact on metastasis, angiogenesis, and immune evasion. Systematic bioinformatics analysis revealed the potential influence of STC1 dysregulation on prognosis, immune cell infiltration, response to immune therapy, and cellular functions. In vitro assays were conducted to assess the proliferation, invasion, migration, and angiogenesis capabilities of A375 cells. In vivo experiments utilizing C57BL/6 J mice established a lung metastasis model using B16-F10 cells to evaluate macrophage infiltration and M2 polarization. A Transwell co-culture system was employed to explore the crosstalk between melanoma and macrophages. Molecular interactions among STC1, YAP, βPIX, and CCL2 are investigated using mass spectrometry, Co-Immunoprecipitation, Dual-Luciferase Reporter Assay, and Chromatin Immunoprecipitation experiments. STC1 was found to enhance lung metastasis by promoting the recruitment and polarization of M2 macrophages, thereby fostering an immunosuppressive microenvironment. Mechanistically, STC1 competes with YAP for binding to βPIX within the KER domain in melanoma cells, leading to YAP activation and subsequent CCL2 upregulation. CCL2-induced M2 macrophages secrete VEGFA, which enhances tumor vascularization and increases STC1 expression via the AKT signaling pathway in melanoma cells, establishing a pro-metastatic feedback loop. Notably, STC1-induced YAP activation increases PD-L1 expression, promoting immune evasion. Silencing STC1 enhances the efficacy of PD-1 immune checkpoint therapy in mice. This research elucidates STC1's role in melanoma metastasis and its complex interactions with tumor-associated macrophages, proposing STC1 as a potential therapeutic target for countering melanoma metastasis and augmenting the efficacy of PD-1 immunotherapy.
Collapse
Affiliation(s)
- Zhaozhou Ren
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Zhijie Xu
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Xiyue Chang
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Jie Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China
| | - Wan'an Xiao
- Department of Orthopedics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, China.
| |
Collapse
|
44
|
Jia D, Zhao S, Liu H, Zhan X, Zhou Z, Lv M, Tang X, Guo W, Li H, Sun L, Zhong Y, Tian B, Yuan D, Tang X, Fan Q. ICG-labeled PD-L1-antagonistic affibody dimer for tumor imaging and enhancement of tumor photothermal-immunotherapy. Int J Biol Macromol 2024; 269:132058. [PMID: 38704065 DOI: 10.1016/j.ijbiomac.2024.132058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/06/2024]
Abstract
In clinical practice, tumor-targeting diagnosis and immunotherapy against programmed death ligand 1 (PD-L1) have a significant impact. In this research, a PD-L1-antagonistic affibody dimer (ZPD-L1) was successfully prepared through Escherichia coli expression system, and conjugated with the photosensitizer of ICG via N-hydroxysuccinimide (NHS) ester to develop a novel tumor-targeting agent (ICG-ZPD-L1) for both tumor imaging diagnosis and photothermal-immunotherapy simultaneously. In vitro, ZPD-L1 could specifically bind to PD-L1-positive LLC and MC38 tumor cells, and ICG-ZPD-L1-mediated photothermal therapy (PTT) also showed excellent phototoxicity to these tumor cells. In vivo, ICG-ZPD-L1 selectively enriched into the PD-L1-positive MC38 tumor tissues, and the high-contrast optical imaging of tumors was obtained. ICG-ZPD-L1-mediated PTT exhibited a potent anti-tumor effect in vivo due to its remarkable photothermal properties. Furthermore, ICG-ZPD-L1-mediated PTT significantly induced the immunogenic cell death (ICD) of primary tumors, promoted maturation of dendritic cells (DCs), up-regulated anti-tumor immune response, enhanced immunotherapy, and superiorly inhibited the growth of metastatic tumors. In addition, ICG-ZPD-L1 showed favorable biosafety throughout the brief duration of treatment. In summary, these results suggest that ICG-ZPD-L1 is a multifunctional tumor-targeting drug integrating tumor imaging diagnosis and photothermal-immunotherapy, and has great guiding significance for the diagnosis and treatment of clinical PD-L1-positive tumor patients.
Collapse
Affiliation(s)
- Dianlong Jia
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, PR China
| | - Shiqi Zhao
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Huimin Liu
- The Second Hospital of Coal Mining Group, Xuzhou 221011, PR China
| | - Xinyu Zhan
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, PR China
| | - Zhongxia Zhou
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Mingjia Lv
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, PR China
| | - Xiufeng Tang
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Wen Guo
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252000, PR China
| | - Hui Li
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Lilan Sun
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Yidong Zhong
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Baoqing Tian
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Dandan Yuan
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Xiaohui Tang
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China.
| | - Qing Fan
- Department of Pharmacy (Shandong Provincinal Key Traditional Chinese Medical Discipline of Clinical Chinese Pharmacy), Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, PR China.
| |
Collapse
|
45
|
Choi Y, Seok SH, Yoon HY, Ryu JH, Kwon IC. Advancing cancer immunotherapy through siRNA-based gene silencing for immune checkpoint blockade. Adv Drug Deliv Rev 2024; 209:115306. [PMID: 38626859 DOI: 10.1016/j.addr.2024.115306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 05/23/2024]
Abstract
Cancer immunotherapy represents a revolutionary strategy, leveraging the patient's immune system to inhibit tumor growth and alleviate the immunosuppressive effects of the tumor microenvironment (TME). The recent emergence of immune checkpoint blockade (ICB) therapies, particularly following the first approval of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors like ipilimumab, has led to significant growth in cancer immunotherapy. The extensive explorations on diverse immune checkpoint antibodies have broadened the therapeutic scope for various malignancies. However, the clinical response to these antibody-based ICB therapies remains limited, with less than 15% responsiveness and notable adverse effects in some patients. This review introduces the emerging strategies to overcome current limitations of antibody-based ICB therapies, mainly focusing on the development of small interfering ribonucleic acid (siRNA)-based ICB therapies and innovative delivery systems. We firstly highlight the diverse target immune checkpoint genes for siRNA-based ICB therapies, incorporating silencing of multiple genes to boost anti-tumor immune responses. Subsequently, we discuss improvements in siRNA delivery systems, enhanced by various nanocarriers, aimed at overcoming siRNA's clinical challenges such as vulnerability to enzymatic degradation, inadequate pharmacokinetics, and possible unintended target interactions. Additionally, the review presents various combination therapies that integrate chemotherapy, phototherapy, stimulatory checkpoints, ICB antibodies, and cancer vaccines. The important point is that when used in combination with siRNA-based ICB therapy, the synergistic effect of traditional therapies is strengthened, improving host immune surveillance and therapeutic outcomes. Conclusively, we discuss the insights into innovative and effective cancer immunotherapeutic strategies based on RNA interference (RNAi) technology utilizing siRNA and nanocarriers as a novel approach in ICB cancer immunotherapy.
Collapse
Affiliation(s)
- Youngjin Choi
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Su Hyun Seok
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hong Yeol Yoon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ju Hee Ryu
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Ick Chan Kwon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
46
|
Sullivan MR, Finocchiaro M, Yang Y, Thomas J, Ali A, Kaplan I, Abdulhamid Y, Bobilev E, Sheffer M, Romee R, Konry T. An innovative single-cell approach for phenotyping and functional genotyping of CAR NK cells. J Immunother Cancer 2024; 12:e008912. [PMID: 38821719 PMCID: PMC11149162 DOI: 10.1136/jitc-2024-008912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2024] [Indexed: 06/02/2024] Open
Abstract
BACKGROUND To accelerate the translation of novel immunotherapeutic treatment approaches, the development of analytic methods to assess their efficacy at early in vitro stages is necessary. Using a droplet-based microfluidic platform, we have established a method for multiparameter quantifiable phenotypic and genomic observations of immunotherapies. Chimeric antigen receptor (CAR) natural killer (NK) cells are of increased interest in the current immunotherapy landscape and thus provide an optimal model for evaluating our novel methodology. METHODS For this approach, NK cells transduced with a CD19 CAR were compared with non-transduced NK cells in their ability to kill a lymphoma cell line. Using our microfluidic platform, we were able to quantify the increase in cytotoxicity and synaptic contact formation of CAR NK cells over non-transduced NK cells. We then optimized our droplet sorter and successfully used it to separate NK cells based on target cell killing to perform transcriptomic analyses. RESULTS Our data revealed expected improvement in cytotoxicity with the CD19 CAR but more importantly, provided unique insights into the factors involved in the cytotoxic mechanisms of CAR NK cells. This demonstrates a novel, improved system for accelerating the pre-clinical screening of future immunotherapy treatments. CONCLUSIONS This study provides a new potential approach for enhanced early screening of immunotherapies to improve their development, with a highly relevant cell model to demonstrate. Additionally, our validation studies provided some potential insights into transcriptomic determinants influencing CAR NK cytotoxicity.
Collapse
Affiliation(s)
- Matthew Ryan Sullivan
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Michael Finocchiaro
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Yichao Yang
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Judene Thomas
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Alaa Ali
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Isabel Kaplan
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Yasmin Abdulhamid
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Eden Bobilev
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Michal Sheffer
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Rizwan Romee
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Tania Konry
- Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Bao L, Li Y, Hu X, Gong Y, Chen J, Huang P, Tan Z, Ge M, Pan Z. Targeting SIGLEC15 as an emerging immunotherapy for anaplastic thyroid cancer. Int Immunopharmacol 2024; 133:112102. [PMID: 38652971 DOI: 10.1016/j.intimp.2024.112102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 04/07/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
Anaplastic thyroid carcinoma (ATC) is the most aggressive subtype of thyroid cancer with few effective therapies. Though immunotherapies such as targeting PD-1/PD-L1 axis have benefited patients with solid tumor, the druggable immune checkpoints are quite limited in ATC. In our study, we focused on the anti-tumor potential of sialic acid-binding Ig-like lectins (Siglecs) in ATC. Through screening by integrating microarray datasets including 216 thyroid-cancer tissues and single-cell RNA-sequencing, SIGLEC family members CD33, SIGLEC1, SIGLEC10 and SIGLEC15 were significantly overexpressed in ATC, among which SIGLEC15 increased highest and mainly expressed on cancer cells. SIGLEC15high ATC cells are characterized by high expression of serine protease PRSS23 and cancer stem cell marker CD44. Compared with SIGLEC15low cancer cells, SIGLEC15high ATC cells exhibited higher interaction frequency with tumor microenvironment cells. Further study showed that SIGLEC15high cancer cells mainly interacted with T cells by immunosuppressive signals such as MIF-TNFRSF14 and CXCL12-CXCR4. Notably, treatment of anti-SIGLEC15 antibody profoundly increased the cytotoxic ability of CD8+ T cells in a co-culture model and zebrafish-derived ATC xenografts. Consistently, administration of anti-SIGLEC15 antibody significantly inhibited tumor growth and prolonged mouse survival in an immunocompetent model of murine ATC, which was associated with increase of M1/M2, natural killer (NK) cells and CD8+ T cells, and decrease of myeloid-derived suppressor cells (MDSCs). SIGLEC15 inhibited T cell activation by reducing NFAT1, NFAT2, and NF-κB signals. Blocking SIGLEC15 increased the secretion of IFN-γ and IL-2 in vitro and in vivo. In conclusion, our finding demonstrates that SIGLEC15 is an emerging and promising target for immunotherapy in ATC.
Collapse
Affiliation(s)
- Lisha Bao
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, China
| | - Ying Li
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Xiaoping Hu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China
| | - Yingying Gong
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, China
| | - Jinming Chen
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Ping Huang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China
| | - Zhuo Tan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, 310014, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China.
| | - Zongfu Pan
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, 310014, Hangzhou, China; Clinical Research Center for Cancer of Zhejiang Province, 310014, Hangzhou, China.
| |
Collapse
|
48
|
He Y, Zhu M, Lai X, Zhang H, Jiang W. The roles of PD-L1 in the various stages of tumor metastasis. Cancer Metastasis Rev 2024:10.1007/s10555-024-10189-4. [PMID: 38733457 DOI: 10.1007/s10555-024-10189-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/08/2024] [Indexed: 05/13/2024]
Abstract
The interaction between tumor programmed death ligand 1 (PD-L1) and T-cell programmed cell death 1 (PD-1) has long been acknowledged as a mechanism for evading immune surveillance. Recent studies, however, have unveiled a more nuanced role of tumor-intrinsic PD-L1 in reprograming tumoral phenotypes. Preclinical models emphasize the synchronized effects of both intracellular and extracellular PD-L1 in promoting metastasis, with intricate interactions with the immune system. This review aims to summarize recent findings to elucidate the spatiotemporal heterogeneity of PD-L1 expression and the pro-metastatic roles of PD-L1 in the entire process of tumor metastasis. For example, PD-L1 regulates the epithelial-to-mesenchymal transition (EMT) process, facilitates the survival of circulating tumor cells, and induces the formation of immunosuppressive environments at pre-metastatic niches and metastatic sites. And the complexed and dynamic regulation process of PD-L1 for tumor metastasis is related to the spatiotemporal heterogeneity of PD-L1 expression and functions from tumor primary sites to various metastatic sites. This review extends the current understandings for the roles of PD-L1 in mediating tumor metastasis and provides new insights into therapeutic decisions in clinical practice.
Collapse
Affiliation(s)
- Yinjun He
- Department of Colorectal Surgery, First Affiliated Hospital, Zhejiang University Medical School, Hangzhou, 310009, China
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China
| | - Ming Zhu
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China
| | - Xuan Lai
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China
| | - Honghe Zhang
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China.
| | - Weiqin Jiang
- Department of Colorectal Surgery, First Affiliated Hospital, Zhejiang University Medical School, Hangzhou, 310009, China.
- Department of Pathology, Zhejiang University Medical School, Hangzhou, 310058, China.
| |
Collapse
|
49
|
Wang X, Miao Y, Shen J, Li D, Deng X, Yang C, Ji Y, Dai Z, Ma Y. Unlocking PD-1 antibody resistance: The MUC1 DNA vaccine augments CD8 + T cell infiltration and attenuates tumour suppression. Scand J Immunol 2024; 99:e13356. [PMID: 38605549 DOI: 10.1111/sji.13356] [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: 07/07/2023] [Revised: 10/11/2023] [Accepted: 01/05/2024] [Indexed: 04/13/2024]
Abstract
In light of increasing resistance to PD1 antibody therapy among certain patient populations, there is a critical need for in-depth research. Our study assesses the synergistic effects of a MUC1 DNA vaccine and PD1 antibody for surmounting PD1 resistance, employing a murine CT26/MUC1 colon carcinoma model for this purpose. When given as a standalone treatment, PD1 antibodies showed no impact on tumour growth. Additionally, there was no change observed in the intra-tumoural T-cell ratios or in the functionality of T-cells. In contrast, the sole administration of a MUC1 DNA vaccine markedly boosted the cytotoxicity of CD8+ T cells by elevating IFN-γ and granzyme B production. Our compelling evidence highlights that combination therapy more effectively inhibited tumour growth and prolonged survival compared to either monotherapy, thus mitigating the limitations intrinsic to single-agent therapies. This enhanced efficacy was driven by a significant alteration in the tumour microenvironment, skewing it towards pro-immunogenic conditions. This assertion is backed by a raised CD8+/CD4+ T-cell ratio and a decrease in immunosuppressive MDSC and Treg cell populations. On the mechanistic front, the synergistic therapy amplified expression levels of CXCL13 in tumours, subsequently facilitating T-cell ingress into the tumour setting. In summary, our findings advocate for integrated therapy as a potent mechanism for surmounting PD1 antibody resistance, capitalizing on improved T-cell functionality and infiltration. This investigation affords critical perspectives on enhancing anti-tumour immunity through the application of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoqin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
- The Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yinsha Miao
- Department of Clinical laboratory, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, China
| | | | - Dandan Li
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xinyue Deng
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chengcheng Yang
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanhong Ji
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - ZhiJun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yunfeng Ma
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
50
|
Sasabe E, Tomomura A, Yamamoto T. The involvement of epidermal growth factor receptor/protein kinase B signaling in the tumor intrinsic PD-L1-induced malignant potential of oral squamous cell carcinoma. J Oral Pathol Med 2024; 53:310-320. [PMID: 38693616 DOI: 10.1111/jop.13540] [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: 12/09/2023] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Various antigen-presenting cells and tumor cells-expressing PD-L1 inhibits antitumor immune responses in the tumor microenvironment. Recently, numerous studies have shown that tumor cell intrinsic PD-L1 also plays important roles in tumor growth and progression. On the other hand, oral squamous cell carcinoma (OSCC) cells overexpress epidermal growth factor receptor (EGFR) and EGFR signal pathway exacerbates tumor progression. Therefore, this study assessed whether tumor-intrinsic PD-L1 facilitates malignant potential of OSCC cells through regulation of EGFR signaling. METHODS Two OSCC cell lines, SAS and HSC-3, were transfected with PD-L1 and EGFR-specific small interfering RNA (siRNA). Influences of PD-L1 knockdown on malignant potentials of OSCC cells were examined by Cell Counting kit-8 assay, transwell assay, sphere formation assay, flow cytometry, and Western blot. Effects of PD-L1 and EGFR knockdown on each expression were examined by quantitative real-time PCR (qRT-PCR), Western blot, and flow cytometry. RESULTS Transfection of an PD-L1-siRNA into OSCC cells decreased the abilities of proliferation, stemness, and mobility of these cells significantly. PD-L1 knockdown also decreased EGFR expression through the promotion of proteasome- and lysosome-mediated degradation and following activation of the EGFR/protekin kinase B (AKT) signal pathway. Meanwhile, EGFR knockdown did not influence PD-L1 expression in SAS and HSC-3 cells, but treatment with a recombinant human EGF induced its expression. Treatment with erlotinib and cetuximab suppressed rhEGF-induced PD-L1 expression and localization in the cellular membrane of both OSCC cells. CONCLUSION OSCC cells-expressing PD-L1 induced by EGF stimulation may promote malignancy intrinsically via the activation of the EGFR/AKT signaling cascade.
Collapse
Affiliation(s)
- Eri Sasabe
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Ayumi Tomomura
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Nankoku, Japan
| | - Tetsuya Yamamoto
- Department of Oral and Maxillofacial Surgery, Kochi Medical School, Kochi University, Nankoku, Japan
| |
Collapse
|