1
|
Chen P, Chen Z, Sui W, Han W. Recent advances in the mechanisms of PD-L1 expression in gastric cancer: a review. Biol Res 2025; 58:16. [PMID: 40091086 PMCID: PMC11912799 DOI: 10.1186/s40659-025-00597-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 03/07/2025] [Indexed: 03/19/2025] Open
Abstract
In the progression of gastric cancer (GC), various cell types in the tumor microenvironment (TME) exhibit upregulated expression of programmed death ligand 1 (PD-L1), leading to impaired T-cell function and evasion of immune surveillance. Infection with H. pylori and EBV leads to increased PD-L1 expression in various cell types within TME, resulting in immune suppression and facilitating immune escape of GC cells. In the TME, mesenchymal stem cells (MSCs), M1-like tumor-associated macrophages (MI-like TAM), and myeloid-derived suppressor cells (MDSCs) contribute to the upregulation of PD-L1 expression in GC cells. Conversely, mast cells, M2-like tumor-associated macrophages (M2-like TAM), and tumor-associated neutrophils (TANs) exhibit elevated levels of PD-L1 expression in response to the influence of GC cells. Together, these factors collectively contribute to the upregulation of PD-L1 expression in GC. This review aims to provide a comprehensive summary of the cellular expression patterns of PD-L1 in GC and the underlying molecular mechanisms. Understanding the complex regulatory pathways governing PD-L1 expression may offer novel insights for the development of effective immunotherapeutic interventions.
Collapse
Affiliation(s)
- Peifeng Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China
| | - Zhangming Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China
| | - Wannian Sui
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China
| | - Wenxiu Han
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Avenue, Shushan District, Hefei, Anhui Province, 230022, China.
| |
Collapse
|
2
|
Yang Y, Sun H, Yu H, Wang L, Gao C, Mei H, Jiang X, Ji M. Tumor-associated-fibrosis and active collagen-CD44 axis characterize a poor-prognosis subtype of gastric cancer and contribute to tumor immunosuppression. J Transl Med 2025; 23:123. [PMID: 39871345 PMCID: PMC11773867 DOI: 10.1186/s12967-025-06070-9] [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: 08/28/2024] [Accepted: 01/04/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Tumor-associated fibrosis modifies the tumor microenvironment (TME), hinders the infiltration and activity of cytotoxic immune cells, and is a critical pathological process leading to the ineffectiveness of tumor immunotherapy in gastric cancer (GC). However, the specific mechanisms and interventions are yet to be fully explored. METHODS Our study included 375 gastric cancer samples from TCGA, 1 single-cell RNA sequencing (scRNA-seq) dataset comprising of 15 gastric cancer samples from GEO, 19 cohorts of immunotherapy and 2 GWAS datasets. Consensus clustering identified a gastric cancer subtype characterized primarily by fibrosis, and various methods such as pseudotime analysis, CellChat analysis and Colocalization analysis were used to explore its mechanisms. RESULTS A subtype of gastric cancer was identified with poor prognosis, characterized by higher malignancy, drug resistance, and poor immune infiltration, associated with elevated expression of genes related with Extracellular matrix (ECM). Single-cell transcriptome analysis showed active Collagen-CD44 signaling axis between cancer-associated fibroblasts (CAFs) and immune cells in gastric cancer, with ECM-related genes upregulated during tumor progression. The expression of CD44 was significantly elevated in the subtype, associated with poor prognosis and tumor immune suppression in gastric cancer, potentially involved in the recruitment of immunosuppressive cells such as M2 macrophages and regulatory T cells (Tregs) and the upregulation of multiple immune checkpoints including PD-1/PD-L1. CONCLUSION Our study identified a new subtype of gastric cancer, revealing that fibrosis is a critical mechanism driving immune suppression in gastric cancer and emphasizing the central role of the Collagen-CD44 signaling axis. The Collagen-CD44 signaling axis has the potential to serve as a novel therapeutic target for gastric cancer by enhancing immune cell-mediated tumor suppression. By combining it with immune checkpoint inhibitors (ICIs), it may improve the efficacy of immunotherapy for gastric cancer and offer new hope for treatment.
Collapse
Affiliation(s)
- Yingqi Yang
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Haohan Sun
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Hongkai Yu
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Luyao Wang
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Chang Gao
- The Second School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Haokun Mei
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Xiaomeng Jiang
- Department of Gastroenterology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, China.
| | - Minghui Ji
- School of Nursing, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
3
|
Yang M, Lin W, Huang J, Mannucci A, Luo H. Novel immunotherapeutic approaches in gastric cancer. PRECISION CLINICAL MEDICINE 2024; 7:pbae020. [PMID: 39397869 PMCID: PMC11467695 DOI: 10.1093/pcmedi/pbae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/08/2024] [Accepted: 09/08/2024] [Indexed: 10/15/2024] Open
Abstract
Gastric cancer is a malignant tumor that ranks third in cancer-related deaths worldwide. Early-stage gastric cancer can often be effectively managed through surgical resection. However, the majority of cases are diagnosed in advanced stages, where outcomes with conventional radiotherapy and chemotherapy remain unsatisfactory. Immunotherapy offers a novel approach to treating molecularly heterogeneous gastric cancer by modifying the immunosuppressive tumor microenvironment. Immune checkpoint inhibitors and adoptive cell therapy are regarded as promising modalities in cancer immunotherapy. Food and Drug Administration-approved programmed death-receptor inhibitors, such as pembrolizumab, in combination with chemotherapy, have significantly extended overall survival in gastric cancer patients and is recommended as a first-line treatment. Despite challenges in solid tumor applications, adoptive cell therapy has demonstrated efficacy against various targets in gastric cancer treatment. Among these approaches, chimeric antigen receptor-T cell therapy research is the most widely explored and chimeric antigen receptor-T cell therapy targeting claudin18.2 has shown acceptable safety and robust anti-tumor capabilities. However, these advancements primarily remain in preclinical stages and further investigation should be made to promote their clinical application. This review summarizes the latest research on immune checkpoint inhibitors and adoptive cell therapy and their limitations, as well as the role of nanoparticles in enhancing immunotherapy.
Collapse
Affiliation(s)
- Meng Yang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Wuhao Lin
- Department of Molecular Diagnostics, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jiaqian Huang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| | - Alessandro Mannucci
- Gastroenterology and Gastrointestinal Emndoscopy Unit, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan 20132, Italy
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope; Monrovia, CA 91016, USA
| | - Huiyan Luo
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
- Research Unit of Precision Diagnosis and Treatment for Gastrointestinal Cancer, Chinese Academy of Medical Sciences, Guangzhou 510060, China
| |
Collapse
|
4
|
Groeger S, Meyle J. The role of programmed death receptor (PD-)1/PD-ligand (L)1 in periodontitis and cancer. Periodontol 2000 2024; 96:150-169. [PMID: 38351432 PMCID: PMC11579837 DOI: 10.1111/prd.12548] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/02/2023] [Accepted: 12/14/2023] [Indexed: 11/22/2024]
Abstract
The programmed-death-ligand-1 (PD-L1) is an immune-modulating molecule that is constitutively expressed on various immune cells, different epithelial cells and a multitude of cancer cells. It is a costimulatory molecule that may impair T-cell mediated immune response. Ligation to the programmed-death-receptor (PD)-1, on activated T-cells and further triggering of the related signaling pathways can induce T-cells apoptosis or anergy. The upregulation of PD-L1 in various cancer types, including oral squamous cell carcinomas, was demonstrated and has been linked to immune escape of tumors and poor prognosis. A bidirectional relationship exists between the increased PD-L1 expression and periodontitis as well as the epithelial-mesenchymal transition (EMT), a process of interconversion of epithelial cells to mesenchymal cells that may induce immune escape of tumors. Interaction between exosomal PD-L1 and PD-1 on T-cells may cause immunosuppression by blocking the activation and proliferation of T-cells. The efficacy and importance of treatment with PD-1/PD-L1 checkpoint inhibitors and their prognostic influence on human cancers was demonstrated. Regarding PD-1/PD-L1 checkpoint inhibitors, resistances exist or may develop, basing on various factors. Further investigations of the underlying mechanisms will help to overcome the therapeutic limitations that result from resistances and to develop new strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Sabine Groeger
- Department of Periodontology, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
- Department of Orthodontics, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
| | - Joerg Meyle
- Department of Periodontology, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
| |
Collapse
|
5
|
Winter L, Ries J, Vogl C, Trumet L, Geppert CI, Lutz R, Kesting M, Weber M. Comparative Analysis of Inhibitory and Activating Immune Checkpoints PD-1, PD-L1, CD28, and CD86 in Non-Melanoma Skin Cancer. Cells 2024; 13:1569. [PMID: 39329753 PMCID: PMC11430031 DOI: 10.3390/cells13181569] [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: 06/05/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
The establishment of immunotherapy applying immune checkpoint inhibitors (ICI) has provided an important new option for the treatment of solid malignant diseases. However, different tumor entities show dramatically different responses to this therapy. BCC responds worse to anti-PD-1 ICIs as compared to cSCC. Differential immune checkpoint expression could explain this discrepancy and, therefore, the aim of this study was to analyze activating and inhibitory immune checkpoints in cSCC and BCC tissues. Tissue microarrays of the invasive front as well as the tumor core of BCC and cSCC samples were used to evaluate PD-1, PD-L1, CD28, and CD86 expression and their topographic distribution profiles by chromogenic immunohistochemistry. QuPath was used to determine the labeling index. The expression of PD-1, PD-L1, and CD28 was significantly higher in both the tumor core and the invasive front of cSCC samples as compared to BCC (p < 0.001). In addition, the ratios of PD-L1/CD86 (p < 0.001) and CD28/CD86 (p < 0.001) were significantly higher in cSCC. The invasive front of both tumor entities showed higher expression levels of all immune markers compared to the tumor core in both tumor entities. The significantly higher expression of PD-1, PD-L1, and CD28 in cSCC, along with the predominance of the inhibitory ligand PD-L1 as compared to the activating CD86 in cSCC, provide a potential explanation for the better objective response rates to anti-PD-1 immunotherapy as compared to BCC. Furthermore, the predominant site of interaction between the immune system and the tumor was within the invasive front in both tumor types.
Collapse
Affiliation(s)
- Linus Winter
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Jutta Ries
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Christoph Vogl
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Leah Trumet
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
- Department of Operative Dentistry and Periodontology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Carol Immanuel Geppert
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Rainer Lutz
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Marco Kesting
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| | - Manuel Weber
- Department of Oral and Cranio-Maxillofacial Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.W.); (C.V.)
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
- Comprehensive Cancer Center Alliance WERA (CCC WERA), 91052 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91052 Erlangen, Germany
| |
Collapse
|
6
|
Sun D, Altalbawy FMA, Yumashev A, Hjazi A, Menon SV, Kaur M, Deorari M, Abdulwahid AS, Shakir MN, Gabal BC. Shedding Light on the Role of Exosomal PD-L1 (ExoPD-L1) in Cancer Progression: an Update. Cell Biochem Biophys 2024; 82:1709-1720. [PMID: 38907940 DOI: 10.1007/s12013-024-01340-7] [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] [Accepted: 05/25/2024] [Indexed: 06/24/2024]
Abstract
Exosomes are the primary category of extracellular vesicles (EVs), which are lipid-bilayer vesicles with biological activity spontaneously secreted from either normal or tansformed cells. They serve a crucial role for intercellular communication and affect extracellular environment and the immune system. Tumor-derived exosomes (TEXs) enclose high levels of immunosuppressive proteins, including programmed death-ligand 1 (PD-L1). PD-L1 and its receptor PD-1 act as crucial immune checkpoint molecules, thus facilitating tumor advancement by inhibiting immune responses. PDL-1 is abundantly present on tumor cells and interacts with PD-1 on activated T cells, resulting in T cell suppression and allowing immune evasion of cancer cells. Various FDA-approved monoclonal antibodies inhibiting the PD-1/PD-L1 interaction are commonly used to treat a diverse range of tumors. Although the achieved results are significant, some individuals have a poor reaction to PD-1/PD-L1 blocking. PD-L1-enriched TEXs may mimic the impact of cell-surface PD-L1, consequently potentiating tumor resistance to PD1/PD-L1 based therapy. In light of this, a strong correlation between circulating exosomal PD-L1 levels and response rate to anti-PD-1/PD-L1 antibody treatment has been evinced. This article inspects the function of exosomal PDL-1 in developing resistance to anti-PD-1/PD-L1 therapy for opening new avenues for overcoming tumor resistance to such modalities and development of more favored combination therapy.
Collapse
Affiliation(s)
- Dongmei Sun
- Siping City Central People's Hospital, Siping, Jilin, 136000, P. R. China
| | - Farag M A Altalbawy
- Department of Biochemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
| | - Alexey Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Alzahraa S Abdulwahid
- Department of Medical Laboratories Technology, Al-Hadi University College, Baghdad, 10011, Iraq
| | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Baneen Chasib Gabal
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
7
|
Cheng W, Kang K, Zhao A, Wu Y. Dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 in lung cancer. J Hematol Oncol 2024; 17:54. [PMID: 39068460 PMCID: PMC11283714 DOI: 10.1186/s13045-024-01581-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Cancer immunotherapies, represented by immune checkpoint inhibitors (ICIs), have reshaped the treatment paradigm for both advanced non-small cell lung cancer and small cell lung cancer. Programmed death receptor-1/programmed death receptor ligand-1 (PD-1/PD-L1) and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) are some of the most common and promising targets in ICIs. Compared to ICI monotherapy, which occasionally demonstrates treatment resistance and limited efficacy, the dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 operates at different stages of T cell activation with synergistically enhancing immune responses against cancer cells. This emerging dual therapy heralds a new direction for cancer immunotherapy, which, however, may increase the risk of drug-related adverse reactions while improving efficacy. Previous clinical trials have explored combination therapy strategy of anti-PD-1/PD-L1 and anti-CTLA-4 agents in lung cancer, yet its efficacy remains to be unclear with the inevitable incidence of immune-related adverse events. The recent advent of bispecific antibodies has made this sort of dual targeting more feasible, aiming to alleviate toxicity without compromising efficacy. Thus, this review highlights the role of dual blockade immunotherapy targeting PD-1/PD-L1 and CTLA-4 in treating lung cancer, and further elucidates its pre-clinical mechanisms and current advancements in clinical trials. Besides, we also provide novel insights into the potential combinations of dual blockade therapies with other strategies to optimize the future treatment mode for lung cancer.
Collapse
Affiliation(s)
- Weishi Cheng
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Kai Kang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Yijun Wu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
8
|
Guo F, Kong W, Li D, Zhao G, Anwar M, Xia F, Zhang Y, Ma C, Ma X. M2-type tumor-associated macrophages upregulated PD-L1 expression in cervical cancer via the PI3K/AKT pathway. Eur J Med Res 2024; 29:357. [PMID: 38970071 PMCID: PMC11225336 DOI: 10.1186/s40001-024-01897-2] [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: 12/23/2023] [Accepted: 05/21/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND AND PURPOSE PD-1/PD-L1 inhibitors have become a promising therapy. However, the response rate is lower than 30% in patients with cervical cancer (CC), which is related to immunosuppressive components in tumor microenvironment (TME). Tumor-associated macrophages (TAMs), as one of the most important immune cells, are involved in the formation of tumor suppressive microenvironment. Therefore, it will provide a theoretical basis for curative effect improvement about the regulatory mechanism of TAMs on PD-L1 expression. METHODS The clinical data and pathological tissues of CC patients were collected, and the expressions of PD-L1, CD68 and CD163 were detected by immunohistochemistry. Bioinformatics was used to analyze the macrophage subtypes involved in PD-L1 regulation. A co-culture model was established to observe the effects of TAMs on the morphology, migration and invasion function of CC cells, and the regulatory mechanism of TAMs on PD-L1. RESULTS PD-L1 expression on tumor cells could predict the poor prognosis of patients. And there was a strong correlation between PD-L1 expression with CD163+TAMs infiltration. Similarly, PD-L1 expression was associated with M1/M2-type TAMs infiltration in bioinformatics analysis. The results of cell co-culture showed that M1/M2-type TAMs could upregulate PD-L1 expression, especially M2-type TAMs may elevate the PD-L1 expression via PI3K/AKT pathway. Meanwhile, M1/M2-type TAMs can affect the morphological changes, and enhance migration and invasion abilities of CC cells. CONCLUSIONS PD-L1 expression in tumor cells can be used as a prognostic factor and is closely related to CD163+TAMs infiltration. In addition, M2-type TAMs can upregulate PD-L1 expression in CC cells through PI3K/AKT pathway, enhance the migration and invasion capabilities, and affect the tumor progression.
Collapse
Affiliation(s)
- Fan Guo
- Department of Medical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, No 789 Suzhou Road, Urumqi, 830011, Xinjiang, China
- Postdoctoral Research Workstation of Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Weina Kong
- Department of Medical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, No 789 Suzhou Road, Urumqi, 830011, Xinjiang, China
| | - Dewei Li
- Center of Respiratory and Critical Care Medicine, The People's Hospital of the Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Gang Zhao
- Department of Blood Transfusion, Affiliated Traditional Chinese Medicine Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Miyessar Anwar
- Department of Medical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, No 789 Suzhou Road, Urumqi, 830011, Xinjiang, China
| | - Feifei Xia
- Department of Medical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, No 789 Suzhou Road, Urumqi, 830011, Xinjiang, China
| | - Yuanming Zhang
- Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Cailing Ma
- Department of Gynecology, The First Affiliated Hospital of Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, 137 Li Yu Shan South Road, Urumqi, 830054, Xinjiang, China.
| | - Xiumin Ma
- Department of Medical Laboratory Center, Tumor Hospital Affiliated to Xinjiang Medical University, State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, No 789 Suzhou Road, Urumqi, 830011, Xinjiang, China.
| |
Collapse
|
9
|
Ouyang P, Wang L, Wu J, Tian Y, Chen C, Li D, Yao Z, Chen R, Xiang G, Gong J, Bao Z. Overcoming cold tumors: a combination strategy of immune checkpoint inhibitors. Front Immunol 2024; 15:1344272. [PMID: 38545114 PMCID: PMC10965539 DOI: 10.3389/fimmu.2024.1344272] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 02/26/2024] [Indexed: 04/12/2024] Open
Abstract
Immune Checkpoint Inhibitors (ICIs) therapy has advanced significantly in treating malignant tumors, though most 'cold' tumors show no response. This resistance mainly arises from the varied immune evasion mechanisms. Hence, understanding the transformation from 'cold' to 'hot' tumors is essential in developing effective cancer treatments. Furthermore, tumor immune profiling is critical, requiring a range of diagnostic techniques and biomarkers for evaluation. The success of immunotherapy relies on T cells' ability to recognize and eliminate tumor cells. In 'cold' tumors, the absence of T cell infiltration leads to the ineffectiveness of ICI therapy. Addressing these challenges, especially the impairment in T cell activation and homing, is crucial to enhance ICI therapy's efficacy. Concurrently, strategies to convert 'cold' tumors into 'hot' ones, including boosting T cell infiltration and adoptive therapies such as T cell-recruiting bispecific antibodies and Chimeric Antigen Receptor (CAR) T cells, are under extensive exploration. Thus, identifying key factors that impact tumor T cell infiltration is vital for creating effective treatments targeting 'cold' tumors.
Collapse
Affiliation(s)
- Peng Ouyang
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Lijuan Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jianlong Wu
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yao Tian
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Caiyun Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Dengsheng Li
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zengxi Yao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Ruichang Chen
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Guoan Xiang
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Jin Gong
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhen Bao
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
Cheng S, Wang H, Kang X, Zhang H. Immunotherapy Innovations in the Fight against Osteosarcoma: Emerging Strategies and Promising Progress. Pharmaceutics 2024; 16:251. [PMID: 38399305 PMCID: PMC10892906 DOI: 10.3390/pharmaceutics16020251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Immunosuppressive elements within the tumor microenvironment are the primary drivers of tumorigenesis and malignant advancement. The presence, as well as the crosstalk between myeloid-derived suppressor cells (MDSCs), osteosarcoma-associated macrophages (OS-Ms), regulatory T cells (Tregs), and endothelial cells (ECs) with osteosarcoma cells cause the poor prognosis of OS. In addition, the consequent immunosuppressive factors favor the loss of treatment potential. Nanoparticles offer a means to dynamically and locally manipulate immuno-nanoparticles, which present a promising strategy for transforming OS-TME. Additionally, chimeric antigen receptor (CAR) technology is effective in combating OS. This review summarizes the essential mechanisms of immunosuppressive cells in the OS-TME and the current immune-associated strategies. The last part highlights the limitations of existing therapies and offers insights into future research directions.
Collapse
Affiliation(s)
- Shigao Cheng
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Orthopedics, Hunan Loudi Central Hospital, Loudi 417000, China
| | - Huiyuan Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuejia Kang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Hui Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
11
|
Wang J, Wu W, Yuan T, Wang L, Zang L, Liu Q, Wang L, Huo X, Huo B, Tang Y, Wang H, Zhao Z. Tumor-associated macrophages and PD-L1 in prostate cancer: a possible key to unlocking immunotherapy efficacy. Aging (Albany NY) 2024; 16:445-465. [PMID: 38189834 PMCID: PMC10817380 DOI: 10.18632/aging.205378] [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: 07/12/2023] [Accepted: 11/15/2023] [Indexed: 01/09/2024]
Abstract
PURPOSE Prostate cancer (PCa) is often considered as a "cold" tumor with low responsiveness to immunotherapy. Recent evidence suggests the activation of specific immune cells, such as tumor-associated macrophages (TAMs), could potentially influence the efficacy of immunotherapy in PCa. However, the relationship between TAMs and PD-L1, a significant regulator in immunotherapy, within PCa remains unexplored. METHODS In this study, we assessed TAM infiltration and PD-L1 expression levels in a local cohort of 95 PCa tissue samples and two publicly available PCa datasets. We employed a combination of bioinformatics and experimental techniques, including gene set enrichment analysis, CIBERSORTx, tissue microarray, immunohistochemistry staining, and analysis of single-cell sequencing datasets, to provide a comprehensive understanding of the association between PD-L1 and TAMs in the PCa microenvironment. RESULTS The study showed that CD68+ TAMs and CD163+ TAMs (M2-TAMs) were more abundant in the tumor microenvironment than in non-cancerous surrounding tissues. The infiltration of CD163+ TAMs was significantly associated with the Gleason score and risk stratification of PCa. Importantly, elevated PD-L1 expression correlated significantly with high infiltration of CD163+ TAMs. Furthermore, patients displaying high levels of CD163+ TAMs and PD-L1 expression exhibited shorter times to biochemical recurrence-free survival. CONCLUSION Our study suggests that CD163+ TAMs are closely associated with PD-L1 expression and can act as a valuable prognostic indicator for PCa. The high infiltration of M2-TAMs, coupled with the overexpression of PD-L1, may contribute to immune escape mechanisms in PCa, thereby influencing disease prognosis.
Collapse
Affiliation(s)
- Jinhuan Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Wenqi Wu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Tian Yuan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Lili Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Li Zang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Qing Liu
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Lei Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xiaodong Huo
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Bin Huo
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yong Tang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Haitao Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhigang Zhao
- Department of Medical Oncology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China
| |
Collapse
|
12
|
Han Z, Wang N, Qiao Q, He X, Wang N. Association of PD-L1 Expression with Clinicopathologic Characters in Gastric Cancer: A Comprehensive Meta-analysis. Curr Med Chem 2024; 31:3198-3216. [PMID: 37921182 DOI: 10.2174/0109298673263784230922060257] [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: 05/24/2023] [Revised: 07/18/2023] [Accepted: 08/16/2023] [Indexed: 11/04/2023]
Abstract
PURPOSE The expression level of programmed death ligand-1(PD-L1) in patients with gastric cancer is the key to determining the use of immune drugs. The relationship between PD-L1 expression level and clinical characteristics is worth exploring. METHODS By setting the search terms correlated to PD-L1 and gastric cancer, a nearly comprehensive search was carried out in four major databases, and the deadline for searching was September 1, 2022. The retrieved documents were further screened by strict inclusion and exclusion criteria after removing the duplication. Next, the quality of the included studies was evaluated with the Newcastle-Ottawa Scale (NOS) scale. Finally, the STATA15.1 software was used to process data and draw plots, and the odds ratios (ORs) were adopted to assess the pooled effect size. RESULTS A total of 85 works of literature were included in this study through screening strictly, and detailed data were extracted after evaluating the quality of the literature. The process of analysis was conducted in the whole population, Asia-Africa population, European and American population, and Asian population with CPS≥1, amd all found that the expression of PD-L1 in gastric cancer was correlated with age, tumor size, EBV infection, Her-2 expression and microsatellite status. However, the subgroup of the region also found some differences in Asian and Western regions, which was interesting and worth studying further. The included research of this study did not have significant publish bias. CONCLUSION After careful analysis, this study found that age (>60 years), tumor size (>5cm), EBV infection (+), Her-2 expression (+), microsatellite status (MSI), and mismatch repair status (dMMR) were risk factors for positive expression of PD-L1 in gastric cancer.
Collapse
Affiliation(s)
- Zhuo Han
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| | - Nan Wang
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| | - Qing Qiao
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| | - Xianli He
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| | - Nan Wang
- Department of General Surgery, Tangdu Hospital, The Air Force Medical University, Xi'an, 710038, China
| |
Collapse
|
13
|
Sharma N, Panigrahi R, Pradhan P, Parida S, Sahoo SR. Expression of CD68+ Tumor associated macrophages in relation to β-catenin in carcinoma stomach. INDIAN J PATHOL MICR 2024; 67:15-20. [PMID: 38358183 DOI: 10.4103/ijpm.ijpm_535_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2024] Open
Abstract
Background With no unified system for tumor associated macrophages (TAMs) density assessment, limited information is available on their relationship with β-catenin expression. Aim To evaluate the density of CD68+ TAMs in gastric adenocarcinoma samples by immunohistochemistry and correlate it with grade, stage, invasion, and beta-catenin. Designs and Settings Formalin fixed paraffin embedded (FFPE) blocks from gastrectomy specimens of proven gastric adenocarcinoma were prospectively and retrospectively were studied over a period of two years. Materials and Methods Immunohistochemistry with CD68 and β-catenin was performed. TAM density was qualitatively compared in "tumor" versus "stroma" and "tumor" versus "non-tumor" regions. Quantitative CD68+ TAM density was assessed using different methods and compared. Cases were classified as high and low TAM based on the median value and correlated with histologic type, location, grade, stage and β-catenin expression pattern. Statistical Analysis Spearman's rank correlation test was used to compare the different methods of TAM density evaluation. The categorical variables were studied using Pearson's Chi-square or Fisher's exact test. CD68+ TAM density and β-catenin expression were correlated by analysis of variance. A P value ≤ 0.05 was taken as statistically significant. Results The CD68+ TAMs in the "tumor" versus "non-tumor" area (p = 0.34) and "tumor" versus "stroma distribution" (p = 0.81) did not show any statistical significance. All methods of TAM density were found to be comparable. High TAM group is significantly associated with lymphovascular invasion, tumor depth, lymph node metastasis, and abnormal β-catenin expression. Conclusion TAMs density plays an important role in the tumor stage. Macrophages may possibly induce gastric cancer invasiveness by activating β-catenin pathway.
Collapse
Affiliation(s)
- Nikhil Sharma
- Department of Pathology, Kalinga Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Ranjita Panigrahi
- Department of Pathology, Kalinga Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Prita Pradhan
- Department of Pathology, Kalinga Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Sabyasachi Parida
- Department of Surgical Oncology, Kalinga Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Saroj R Sahoo
- Department of Surgical Oncology, Kalinga Institute of Medical Sciences, Bhubaneswar, Odisha, India
| |
Collapse
|
14
|
Khan SU, Khan SU, Suleman M, Khan MU, Khan MS, Arbi FM, Hussain T, Mohammed Alsuhaibani A, S Refat M. Natural Allies for Heart Health: Nrf2 Activation and Cardiovascular Disease Management. Curr Probl Cardiol 2024; 49:102084. [PMID: 37714318 DOI: 10.1016/j.cpcardiol.2023.102084] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/17/2023]
Abstract
The term "cardiovascular diseases" (CVD) refers to various ailments that affect the heart and blood vessels, including myocardial ischemia, congenital heart defects, heart failure, rheumatic heart disease, hypertension, peripheral artery disease, atherosclerosis, and cardiomyopathies. Despite significant breakthroughs in preventative measures and treatment choices, CVDs significantly contribute to morbidity and mortality, imposing a considerable financial burden. Oxidative stress (OS) is a fundamental contributor to the development and progression of CVDs, resulting from an inherent disparity in generating reactive oxygen species. The disparity above significantly contributes to the aberrant operation of the cardiovascular system. To tackle this issue, therapeutic intervention primarily emphasizes the nuclear erythroid 2-related factor 2 (Nrf2), a transcription factor crucial in regulating endogenous antioxidant defense systems against OS. The Nrf2 exhibits potential as a promising target for effectively managing CVDs. Significantly, an emerging field of study is around the utilization of natural substances to stimulate the activation of Nrf2, hence facilitating the promotion of cardioprotection. This technique introduces a new pathway for treating CVD. The substances above elicit their advantageous effects by mitigating the impact of OS via initiating Nrf2 signaling. The primary objective of our study is to provide significant insights that can contribute to advancing treatment methods, including natural products. These strategies aim to tackle the obstacles associated with CVDs.
Collapse
Affiliation(s)
- Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and South west University, College of Agronomy and Biotechnology, Southwest University, Chongqing, China; Department of Biochemistry, Women Medical and Dental College, Khyber Medical University, Abbottabad, Khyber Pakhtunkhwa, Pakistan.
| | - Muhammad Suleman
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan; Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar
| | - Munir Ullah Khan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | | | | | - Talib Hussain
- Women Dental College Abbottabad, Khyber Pakhtunkhwa, Pakistan
| | - Amnah Mohammed Alsuhaibani
- Department of Physical Sport Science, College of Education, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Moamen S Refat
- Department of Chemistry, College of Science, Taif University, Taif, Saudi Arabia
| |
Collapse
|
15
|
Yerolatsite M, Torounidou N, Gogadis A, Kapoulitsa F, Ntellas P, Lampri E, Tolia M, Batistatou A, Katsanos K, Mauri D. TAMs and PD-1 Networking in Gastric Cancer: A Review of the Literature. Cancers (Basel) 2023; 16:196. [PMID: 38201623 PMCID: PMC10778110 DOI: 10.3390/cancers16010196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/11/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common and aggressive types of cancer. Immune checkpoint inhibitors (ICIs) have proven effective in treating various types of cancer. The use of ICIs in GC patients is currently an area of ongoing research. The tumor microenvironment (TME) also seems to play a crucial role in cancer progression. Tumor-associated macrophages (TAMs) are the most abundant population in the TME. TAMs are capable of displaying programmed cell death protein 1 (PD-1) on their surface and can form a ligand with programmed death ligand 1 (PD-L1), which is found on the surface of cancer cells. Therefore, it is expected that TAMs may significantly influence the immune response related to immune checkpoint inhibitors (ICIs). AIM OF THE STUDY Understanding the role of TAMs and PD-1/PD-L1 networking in GC. METHODS A systematic review of published data was performed using MEDLINE (PubMed), Embase, and Cochrane databases. We retrieved articles investigating the co-existence of TAMs and PD-1 in GC and the prognosis of patients expressing high levels of PD-1+ TAMs. RESULTS Ten articles with a total of 2277 patients were included in the systematic review. The examined data suggest that the expression of PD-L1 has a positive correlation with the infiltration of TAMs and that patients who express high levels of PD-1+ TAMs may have a worse prognosis than those who express low levels of PD-1+ TAMs. CONCLUSIONS TAMs play a pivotal role in the regulation of PD-1/PD-L1 networking and the progression of GC cells. Nevertheless, additional studies are needed to better define the role of TAMs and PD-1/PD-L1 networking in GC.
Collapse
Affiliation(s)
- Melina Yerolatsite
- Department of Medical Oncology, University of Ioannina, 45500 Ioannina, Greece; (N.T.); (A.G.); (F.K.); (P.N.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Nanteznta Torounidou
- Department of Medical Oncology, University of Ioannina, 45500 Ioannina, Greece; (N.T.); (A.G.); (F.K.); (P.N.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Aristeidis Gogadis
- Department of Medical Oncology, University of Ioannina, 45500 Ioannina, Greece; (N.T.); (A.G.); (F.K.); (P.N.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Fani Kapoulitsa
- Department of Medical Oncology, University of Ioannina, 45500 Ioannina, Greece; (N.T.); (A.G.); (F.K.); (P.N.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| | - Panagiotis Ntellas
- Department of Medical Oncology, University of Ioannina, 45500 Ioannina, Greece; (N.T.); (A.G.); (F.K.); (P.N.); (D.M.)
| | - Evangeli Lampri
- Department of Pathology, University of Ioannina, 45500 Ioannina, Greece; (E.L.); (A.B.)
| | - Maria Tolia
- Department of Radiotherapy, University of Crete, 71003 Heraklion, Greece;
| | - Anna Batistatou
- Department of Pathology, University of Ioannina, 45500 Ioannina, Greece; (E.L.); (A.B.)
| | | | - Davide Mauri
- Department of Medical Oncology, University of Ioannina, 45500 Ioannina, Greece; (N.T.); (A.G.); (F.K.); (P.N.); (D.M.)
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45445 Ioannina, Greece
| |
Collapse
|
16
|
Zhang J, Hu C, Zhang R, Xu J, Zhang Y, Yuan L, Zhang S, Pan S, Cao M, Qin J, Cheng X, Xu Z. The role of macrophages in gastric cancer. Front Immunol 2023; 14:1282176. [PMID: 38143746 PMCID: PMC10746385 DOI: 10.3389/fimmu.2023.1282176] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023] Open
Abstract
As one of the deadliest cancers of the gastrointestinal tract, there has been limited improvement in long-term survival rates for gastric cancer (GC) in recent decades. The poor prognosis is attributed to difficulties in early detection, minimal opportunity for radical resection and resistance to chemotherapy and radiation. Macrophages are among the most abundant infiltrating immune cells in the GC stroma. These cells engage in crosstalk with cancer cells, adipocytes and other stromal cells to regulate metabolic, inflammatory and immune status, generating an immunosuppressive tumour microenvironment (TME) and ultimately promoting tumour initiation and progression. In this review, we summarise recent advances in our understanding of the origin of macrophages and their types and polarisation in cancer and provide an overview of the role of macrophages in GC carcinogenesis and development and their interaction with the GC immune microenvironment and flora. In addition, we explore the role of macrophages in preclinical and clinical trials on drug resistance and in treatment of GC to assess their potential therapeutic value in this disease.
Collapse
Affiliation(s)
- Jiaqing Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Can Hu
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ruolan Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Jingli Xu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Li Yuan
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shengjie Zhang
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Siwei Pan
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Mengxuan Cao
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiangjiang Qin
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
17
|
Liu K, Yuan S, Wang C, Zhu H. Resistance to immune checkpoint inhibitors in gastric cancer. Front Pharmacol 2023; 14:1285343. [PMID: 38026944 PMCID: PMC10679741 DOI: 10.3389/fphar.2023.1285343] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023] Open
Abstract
Gastric cancer (GC) is one of the most common gastrointestinal malignancies worldwide. In the past decade, with the development of early diagnostic techniques, a clear decline in GC incidence has been observed, but its mortality remains high. The emergence of new immunotherapies such as immune checkpoint inhibitors (ICIs) has changed the treatment of GC patients to some extent. However, only a small number of patients with advanced GC have a durable response to ICI treatment, and the efficacy of ICIs is very limited. Existing studies have shown that the failure of immunotherapy is mainly related to the development of ICI resistance in patients, but the understanding of the resistance mechanism is still insufficient. Therefore, clarifying the mechanism of GC immune resistance is critical to improve its treatment and clinical benefit. In this review, we focus on summarizing the mechanisms of primary or acquired resistance to ICI immunotherapy in GC from both internal and external aspects of the tumor. At the same time, we also briefly discuss some other possible resistance mechanisms in light of current studies.
Collapse
Affiliation(s)
- Kai Liu
- The Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Shiman Yuan
- The Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Chenyu Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Hong Zhu
- Cancer Center, Department of Medical Oncology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Yi Z, Jia Q, Wang Y, Zhang Y, Xie T, Ling J. Elian granules alleviate precancerous lesions of gastric cancer in rats by suppressing M2-type polarization of tumor-associated macrophages through NF-κB signaling pathway. BMC Complement Med Ther 2023; 23:188. [PMID: 37291549 DOI: 10.1186/s12906-023-04015-7] [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: 12/23/2022] [Accepted: 05/28/2023] [Indexed: 06/10/2023] Open
Abstract
BACKGROUND Precancerous lesions of gastric cancer (PLGC) refer to a kind of histopathological changes in the gastric mucosa that can progress to gastric cancer. Elian granules (ELG), a Chinese medicinal prescription, have achieved satisfactory results in the treatment of PLGC. However, the exact mechanism underlying the therapeutic effect of ELG remains unclear. Here, this study aims to explore the mechanism of ELG alleviating PLGC in rats. METHODS The chemical ingredients of ELG were analyzed using ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS). Specific Pathogen Free SD rats were randomly assigned to 3 groups: the control, model, and ELG groups. The 1-Methyl-3-nitro-1-nitrosoguanidine (MNNG) integrated modeling method was adopted to construct the PLGC rat model in groups except for the control group. Meanwhile, normal saline was used as an intervention for the control and model groups, and ELG aqueous solution for the ELG group, lasting 40 weeks. Subsequently, the stomach of rats was harvested for further analysis. Hematoxylin-eosin staining of the gastric tissue was conducted to assess the pathological changes. Immunofluorescence was carried out for the expression of CD68, and CD206 proteins. Real-time quantitative PCR combined with Western blot was conducted to analyze the expression of arginase-1(Arg-1), inducible nitric oxide synthase (iNOS), p65, p-p65, nuclear factor inhibitor protein-α (IκBα), and p-IκBα in gastric antrum tissue. RESULTS Five chemical ingredients including Curcumol, Curzerenone, Berberine, Ferulic Acid, and 2-Hydroxy-3-Methylanthraquine were identified in ELG. The gastric mucosal glands of rats treated with ELG were orderly arranged, with no intestinal metaplasia and no dysplasia. Furthermore, ELG decreased the percentage of M2-type TAMs marked with CD68 and CD206 proteins, and the ratio of Arg-1 to iNOS in the gastric antrum tissue of rats with PLGC. In addition, ELG could also down-regulate the protein and mRNA expression of p-p65, p65, and p-IκBα, but up-regulate the expression of IκBα mRNA in rats with PLGC. CONCLUSIONS The results showed that ELG attenuates PLGC in rats by suppressing the M2-type polarization of tumor-associated macrophages (TAMs) through NF-κB signaling pathway.
Collapse
Affiliation(s)
- Zhirong Yi
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
| | - Qingling Jia
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, China
| | - Yujiao Wang
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, China
| | - Yuqin Zhang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
| | - Tianyi Xie
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, China
| | - Jianghong Ling
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200021, China.
| |
Collapse
|
19
|
Ornella MSC, Badrinath N, Kim KA, Kim JH, Cho E, Hwang TH, Kim JJ. Immunotherapy for Peritoneal Carcinomatosis: Challenges and Prospective Outcomes. Cancers (Basel) 2023; 15:cancers15082383. [PMID: 37190310 DOI: 10.3390/cancers15082383] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Peritoneal metastasis, also known as peritoneal carcinomatosis (PC), is a refractory cancer that is typically resistant to conventional therapies. The typical treatment for PC is a combination of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). Recently, research in this area has seen significant advances, particularly in immunotherapy as an alternative therapy for PC, which is very encouraging. Catumaxomab is a trifunctional antibody intraperitoneal (IP) immunotherapy authorized in Europe that can be used to diminish malignant ascites by targeting EpCAM. Intraperitoneal (IP) immunotherapy breaks immunological tolerance to treat peritoneal illness. Increasing T-cell responses and vaccination against tumor-associated antigens are two methods of treatment. CAR-T cells, vaccine-based therapeutics, dendritic cells (DCs) in combination with pro-inflammatory cytokines and NKs, adoptive cell transfer, and immune checkpoint inhibitors are promising treatments for PC. Carcinoembryonic antigen-expressing tumors are suppressed by IP administration of CAR-T cells. This reaction was strengthened by anti-PD-L1 or anti-Gr1. When paired with CD137 co-stimulatory signaling, CAR-T cells for folate receptor cancers made it easier for T-cell tumors to find their way to and stay alive in the body.
Collapse
Affiliation(s)
- Mefotse Saha Cyrelle Ornella
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Narayanasamy Badrinath
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Kyeong-Ae Kim
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Jung Hee Kim
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Euna Cho
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Tae-Ho Hwang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
- Bionoxx Inc., Parkview Tower #1905, 248 Jeongjail-ro, Bundang-gu, Seongnam 13554, Republic of Korea
| | - Jae-Joon Kim
- Division of Hematology & Oncology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
20
|
Ni JJ, Zhang ZZ, Ge MJ, Chen JY, Zhuo W. Immune-based combination therapy to convert immunologically cold tumors into hot tumors: an update and new insights. Acta Pharmacol Sin 2023; 44:288-307. [PMID: 35927312 PMCID: PMC9889774 DOI: 10.1038/s41401-022-00953-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/03/2022] [Indexed: 02/04/2023]
Abstract
As a breakthrough strategy for cancer treatment, immunotherapy mainly consists of immune checkpoint inhibitors (ICIs) and other immunomodulatory drugs that provide a durable protective antitumor response by stimulating the immune system to fight cancer. However, due to the low response rate and unique toxicity profiles of immunotherapy, the strategies of combining immunotherapy with other therapies have attracted enormous attention. These combinations are designed to exert potent antitumor effects by regulating different processes in the cancer-immunity cycle. To date, immune-based combination therapy has achieved encouraging results in numerous clinical trials and has received Food and Drug Administration (FDA) approval for certain cancers with more studies underway. This review summarizes the emerging strategies of immune-based combination therapy, including combinations with another immunotherapeutic strategy, radiotherapy, chemotherapy, anti-angiogenic therapy, targeted therapy, bacterial therapy, and stroma-targeted therapy. Here, we highlight the rationale of immune-based combination therapy, the biomarkers and the clinical progress for these immune-based combination therapies.
Collapse
Affiliation(s)
- Jiao-Jiao Ni
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
- Department of Gastroenterology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310016, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Zi-Zhen Zhang
- Department of Gastroenterology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310016, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Ming-Jie Ge
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Hangzhou, 310006, China
| | - Jing-Yu Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310016, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
| | - Wei Zhuo
- Department of Cell Biology and Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
- Institution of Gastroenterology, Zhejiang University, Hangzhou, 310016, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
21
|
Wang SJ, Liu BR, Zhang F, Li YP, Su XR, Yang CT, Cong B, Zhang ZH. Abnormal fatty acid metabolism and ceramide expression may discriminate myocardial infarction from strangulation death: A pilot study. Tissue Cell 2023; 80:101984. [PMID: 36434828 DOI: 10.1016/j.tice.2022.101984] [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: 09/13/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022]
Abstract
Determining myocardial infarction (MI) and mechanical asphyxia (MA) was one of the most challenging tasks in forensic practice. The present study aimed to investigate the potential of fatty acid (FAs) metabolism, and lipid alterations in determining MI and MA. MA and MI mouse models were constructed, and metabolic profiles were obtained by LC-MS-based untargeted metabolomics. The metabolic alterations were explored using the PCA, OPLS-DA, the Wilcoxon test, and fold change analysis. The contents of lipid droplets (LDs) were detected by the transmission scanning electron microscope and Oil red O staining. The immunohistochemical assay was performed to detect CD36 and dysferlin. The ceramide was assessed by LC-MS. PCA showed considerable differences in the metabolite profiles, and the well-fitting OPLS-DA model was developed to screen differential metabolites. Thereinto, 9 metabolites in the MA were reduced, while metabolites were up- and down-regulated in MI. The increased CD36 suggested that MI and MA could enhance the intake of FAs and disturb energy metabolism. The increased LDs, decreased dysferlin, and increased ceramide (C18:0, C22:0, and C24:0) were observed in MI groups, confirming the lipid deposition. The present study indicated significant differences in myocardial FAs metabolism and lipid alterations between MI and MA, suggesting that FAs metabolism and related proteins, certain ceramide may harbor the potential as biomarkers for discrimination of MI and MA.
Collapse
Affiliation(s)
- Song-Jun Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, China.
| | - Bing-Rui Liu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, China.
| | - Fu Zhang
- Forensic Pathology Lab, Guangdong Public Security Department, China.
| | - Ya-Ping Li
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, China.
| | - Xiao-Rui Su
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, China.
| | - Chen-Teng Yang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, China.
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, China.
| | - Zhi-Hua Zhang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, China; HeBei Chest Hospital, China.
| |
Collapse
|
22
|
Saito M. Novel Roles of Nanog in Cancer Cells and Their Extracellular Vesicles. Cells 2022; 11:cells11233881. [PMID: 36497144 PMCID: PMC9736053 DOI: 10.3390/cells11233881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
The use of extracellular vesicle (EV)-based vaccines is a strategically promising way to prevent cancer metastasis. The effective roles of immune cell-derived EVs have been well understood in the literature. In the present paper, we focus on cancer cell-derived EVs to enforce, more thoroughly, the use of EV-based vaccines against unexpected malignant cells that might appear in poor prognostic patients. As a model of such a cancer cell with high malignancy, Nanog-overexpressing melanoma cell lines were developed. As expected, Nanog overexpression enhanced the metastatic potential of melanomas. Against our expectations, a fantastic finding was obtained that determined that EVs derived from Nanog-overexpressing melanomas exhibited a metastasis-suppressive effect. This is considered to be a novel role for Nanog in regulating the property of cancer cell-derived EVs. Stimulated by this result, the review of Nanog's roles in various cancer cells and their EVs has been updated once again. Although there was no other case presenting a similar contribution by Nanog, only one case suggested that NANOG and SOX might be better prognosis markers in head and neck squamous cell carcinomas. This review clarifies the varieties of Nanog-dependent phenomena and the relevant signaling factors. The information summarized in this study is, thus, suggestive enough to generate novel ideas for the construction of an EV-based versatile vaccine platform against cancer metastasis.
Collapse
Affiliation(s)
- Mikako Saito
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan
| |
Collapse
|
23
|
Kovaleva OV, Podlesnaya PA, Chang VL, Ognerubov NA, Gratchev AN, Kozlov NA, Stilidi IS, Kushlinskii NE. Comprehensive Analysis of Stromal and Serum Markers in Gastric Cancer. Acta Naturae 2022; 14:75-83. [PMID: 36694901 PMCID: PMC9844092 DOI: 10.32607/actanaturae.11753] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 10/20/2022] [Indexed: 01/22/2023] Open
Abstract
A comprehensive analysis of the cell phenotype of the inflammatory infiltrate of the tumor stroma represents a promising area of molecular oncology. The study of not only soluble forms of various immunoregulatory molecules, but also their membrane-bound forms is also considered highly relevant. We performed a comprehensive analysis of tissue and circulating forms of the PD-1 and PD-L1 proteins, as well as macrophage and B-cell markers in the tumor stroma of gastric cancer, to assess their clinical and prognostic significance. The tumor and blood plasma samples from 63 gastric cancer patients were studied using ELISA and immunohistochemistry. Malignant gastric tumors were shown to be strongly infiltrated by B-cells, and their number was comparable to that of macrophages. For PU.1 expression, an association with tumor size was observed; i.e., larger tumors were characterized by fewer PU.1+ infiltrating cells (p = 0.005). No clinical significance was found for CD20 and CD163, but their numbers were higher at earlier stages of the disease and in the absence of metastases. It was also demonstrated that the PD-L1 content in tumor cells was not associated with the clinical and morphological characteristics of GC. At the same time, PD-L1 expression in tumor stromal cells was associated with the presence of distant metastases. The analysis of the prognostic significance of all the markers studied demonstrated that CD163 was statistically significantly associated with a poor prognosis for the disease (p = 0.019). In addition, PD-L1 expression in tumor cells tended to indicate a favorable prognosis (p = 0.122). The results obtained in this work indicate that the study of soluble and tissue markers of tumor stroma is promising in prognosticating the course of GC. The search for combinations of markers seems to be highly promising, with their comprehensive analysis capable of helping personalize advanced antitumor therapy.
Collapse
Affiliation(s)
- O. V. Kovaleva
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russian Federation, Moscow, 115552 Russia
| | - P. A. Podlesnaya
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russian Federation, Moscow, 115552 Russia
| | - V. L. Chang
- Medical Institute of G.P. Derzhavin Tambov State University, Tambov, 392000 Russia
| | - N. A. Ognerubov
- Medical Institute of G.P. Derzhavin Tambov State University, Tambov, 392000 Russia
| | - A. N. Gratchev
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russian Federation, Moscow, 115552 Russia
| | - N. A. Kozlov
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russian Federation, Moscow, 115552 Russia
| | - I. S. Stilidi
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russian Federation, Moscow, 115552 Russia
| | - N. E. Kushlinskii
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of Russian Federation, Moscow, 115552 Russia
| |
Collapse
|
24
|
Agnarelli A, Vella V, Samuels M, Papanastasopoulos P, Giamas G. Incorporating Immunotherapy in the Management of Gastric Cancer: Molecular and Clinical Implications. Cancers (Basel) 2022; 14:cancers14184378. [PMID: 36139540 PMCID: PMC9496849 DOI: 10.3390/cancers14184378] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/30/2022] [Accepted: 09/05/2022] [Indexed: 01/30/2023] Open
Abstract
Simple Summary Gastric cancer is one of the most common malignant tumours worldwide, with the fifth and third highest morbidity and mortality, respectively, of all cancers. Survival is limited, as most of the patients are diagnosed at an advanced stage, and are not suitable for surgery with a curative intent. Chemotherapy has only modestly improved patients’ outcomes and is mainly given with a palliative intent. Immunotherapy has improved overall survival of patients with gastric cancer, and has thus become a new standard of care in clinic. In this review we discuss the strong molecular rationale for the administration of immunotherapy in this disease and analyse the clinical data supporting its use. Abstract Gastric cancer has a median survival of 11 months, and this poor prognosis has not improved over the last 30 years. Recent pre-clinical data suggest that there is high tumour-related neoantigen expression in gastric cancer cells, suggesting that a clinical strategy that enhances the host’s immune system against cancer cells may be a successful approach to improve clinical outcomes. Additionally, there has been an increasing amount of translational evidence highlighting the relevance of PD-L1 expression in gastric cancer cells, indicating that PD-1/PD-L1 inhibitors may be useful. Several molecular subgroups of gastric cancer have been identified to respond with excellent outcomes to immunotherapy, including microsatellite instable tumours, tumours bearing a high tumour mutational burden, and tumours related to a chronic EBV infection. In gastric cancer, immunotherapy has produced durable responses in chemo-refractory patients; however, most recently there has been a lot of enthusiasm as several large-scale clinical trials highlight the improved survival noted from the incorporation of immunotherapy in the first line setting for advanced gastric cancer. Our review aims to discuss current pre-clinical and clinical data supporting the innovative role of immunotherapy in gastric cancer.
Collapse
|
25
|
Chen TW, Hung WZ, Chiang SF, Chen WTL, Ke TW, Liang JA, Huang CY, Yang PC, Huang KCY, Chao KSC. Dual inhibition of TGFβ signaling and CSF1/CSF1R reprograms tumor-infiltrating macrophages and improves response to chemotherapy via suppressing PD-L1. Cancer Lett 2022; 543:215795. [PMID: 35718267 DOI: 10.1016/j.canlet.2022.215795] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/02/2022]
Abstract
TGFβ contributes to chemoresistance in advanced colorectal cancer (CRC) via diverse immune-microenvironment mechanisms. Here, we found that cancer cell autonomous TGFβ directly triggered tumor programmed cell death 1 ligand 1 (PD-L1) upregulation, resulting in resistance to chemotherapy. Inhibition of tumor PD-L1 expression sensitized cancer cells to chemotherapy, reduced lung metastasis and increased the influx of CD8+ T cells. However, chemorefractory cancer cell-derived CSF1 recruited TAMs for TGFβ-mediated PD-L1 upregulation via a vicious cycle. High infiltration of macrophages was clinically correlated with the status of tumor PD-L1 after chemotherapy treatment in CRC patients. We found that depletion of immunosuppressive CSF1R+ TAM infiltration and blockade of the TGFβ receptor resulted in an increased influx of cytotoxic CD8+ T and effector memory CD8+ cells, a reduction in regulatory T cells, and a synergistic inhibition of tumor growth when combined with chemotherapy. These findings show that CSF1R+ TAMs and TGFβ are the dominant components that regulate PD-L1 expression within the immunosuppressive tumor microenvironment, providing a therapeutic strategy for advanced CRC patients.
Collapse
Affiliation(s)
- Tsung-Wei Chen
- Graduate Institute of Biomedical Science, China Medical University, Taichung, 40402, Taiwan; Department of Pathology, Asia University Hospital, Asia University, Taichung, 41354, Taiwan
| | - Wei-Ze Hung
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan
| | - Shu-Fen Chiang
- Lab of Precision Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Taichung, 42055, Taiwan
| | - William Tzu-Liang Chen
- Department of Colorectal Surgery, China Medical University HsinChu Hospital, China Medical University, HsinChu, 302, Taiwan; Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan; Department of Surgery, School of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Tao-Wei Ke
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan; School of Chinese Medicine & Graduate Institute of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Ji-An Liang
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan; Department of Radiotherapy, School of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Science, China Medical University, Taichung, 40402, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan; Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, 970, Taiwan; Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan
| | - Pei-Chen Yang
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan
| | - Kevin Chih-Yang Huang
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 40402, Taiwan; Translation Research Core, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan.
| | - K S Clifford Chao
- Graduate Institute of Biomedical Science, China Medical University, Taichung, 40402, Taiwan; Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan; Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| |
Collapse
|
26
|
Kang D, Kim IH. Molecular Mechanisms and Potential Rationale of Immunotherapy in Peritoneal Metastasis of Advanced Gastric Cancer. Biomedicines 2022; 10:biomedicines10061376. [PMID: 35740397 PMCID: PMC9220323 DOI: 10.3390/biomedicines10061376] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 11/16/2022] Open
Abstract
Peritoneal metastasis (PM) is one of the most frequent metastasis patterns of gastric cancer (GC), and the prognosis of patients with PM is very dismal. According to Paget’s theory, disseminated free cancer cells are seeded and survive in the abdominal cavity, adhere to the peritoneum, invade the subperitoneal tissue, and proliferate through angiogenesis. In these sequential processes, several key molecules are involved. From a therapeutic point of view, immunotherapy with chemotherapy combination has become the standard of care for advanced GC. Several clinical trials of newer immunotherapy agents are ongoing. Understanding of the molecular process of PM and the potential rationale of immunotherapy for PM treatment is necessary. Beyond understanding of the molecular aspect of PM, many studies have been conducted on the modality of treatment of PM. Notably, intraperitoneal approaches, including chemotherapy or immunotherapy, have been conducted, because systemic treatment of PM has limitations. In this study, we reviewed the molecular mechanisms and immunologic aspects of PM, and intraperitoneal approaches under investigation for treating PM.
Collapse
Affiliation(s)
- Donghoon Kang
- Division of Gastroenterology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea;
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence:
| |
Collapse
|
27
|
Wei Z, Zhang Y. Immune Cells in Hyperprogressive Disease under Immune Checkpoint-Based Immunotherapy. Cells 2022; 11:cells11111758. [PMID: 35681453 PMCID: PMC9179330 DOI: 10.3390/cells11111758] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 01/27/2023] Open
Abstract
Immunotherapy, an antitumor therapy designed to activate antitumor immune responses to eliminate tumor cells, has been deeply studied and widely applied in recent years. Immune checkpoint inhibitors (ICIs) are capable of preventing the immune responses from being turned off before tumor cells are eliminated. ICIs have been demonstrated to be one of the most effective and promising tumor treatments and significantly improve the survival of patients with multiple tumor types. However, low effective rates and frequent atypical responses observed in clinical practice limit their clinical applications. Hyperprogressive disease (HPD) is an unexpected phenomenon observed in immune checkpoint-based immunotherapy and is a challenge facing clinicians and patients alike. Patients who experience HPD not only cannot benefit from immunotherapy, but also experience rapid tumor progression. However, the mechanisms of HPD remain unclear and controversial. This review summarized current findings from cell experiments, animal studies, retrospective studies, and case reports, focusing on the relationships between various immune cells and HPD and providing important insights for understanding the pathogenesis of HPD.
Collapse
Affiliation(s)
- Zhanqi Wei
- School of Medicine, Tsinghua University, Haidian District, Beijing 100084, China;
- Hepatopancreatbiliary Center, Tsinghua University Affiliated Beijing Tsinghua Changgung Hospital, Changping District, Beijing 102218, China
| | - Yuewei Zhang
- Hepatopancreatbiliary Center, Tsinghua University Affiliated Beijing Tsinghua Changgung Hospital, Changping District, Beijing 102218, China
- Correspondence:
| |
Collapse
|
28
|
Frohwitter G, Kerta M, Vogl C, Geppert CI, Werry JE, Ries J, Kesting M, Weber M. Macrophage and T-Cell Infiltration and Topographic Immune Cell Distribution in Non-Melanoma Skin Cancer of the Head and Neck. Front Oncol 2022; 12:809687. [PMID: 35463364 PMCID: PMC9022069 DOI: 10.3389/fonc.2022.809687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Non-melanoma skin cancer (NMSC) is a heterogeneous tumor entity that is vastly determined by age and UV-light exposure leading to a great mutational burden in cancer cells. However, the success of immune checkpoint blockade in advanced NMSC and the incidence and disease control rates of NMSC in organ transplant recipients compared to immunologically uncompromised patients point toward the emerging importance of the immunologic activity of NMSC. To gain first insight into the role of T-cell and macrophage infiltration in NMSC of the head and neck and capture their different immunogenic profiles, which appear to be highly relevant for the response to immunotherapy, we conducted a whole slide analysis of 107 basal cell carcinoma (BCC) samples and 117 cutaneous squamous cell carcinoma (cSCC) samples. The CD8+ and CD68+ immune cell expression in both cancer types was evaluated by immunohistochemistry and a topographic distribution profile, and the proportion of both cell populations within the two tumor entities was assessed. The results show highly significant differences in terms of CD8+ T-cell and CD68+ macrophage infiltration in BCC and cSCC and indicate cSCC as a highly immunogenic tumor. Yet, BCC presents less immune cell infiltration; the relation between the immune cells compared to cSCC does not show any significant difference. These findings help explain disparities in local aggressiveness, distant metastasis, and eligibility for immune checkpoint blockade in both tumor entities and encourage further research.
Collapse
Affiliation(s)
- Gesche Frohwitter
- Department for Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Bavaria, Germany
| | - Marie Kerta
- Department for Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Bavaria, Germany
| | - Christoph Vogl
- Department for Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Bavaria, Germany
| | - Carol Immanuel Geppert
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Bavaria, Germany
| | - Jan-Erik Werry
- Department for Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Bavaria, Germany
| | - Jutta Ries
- Department for Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Bavaria, Germany
| | - Marco Kesting
- Department for Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Bavaria, Germany
| | - Manuel Weber
- Department for Oral and Maxillofacial Surgery, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Bavaria, Germany
| |
Collapse
|
29
|
Bazhin AV, von Ahn K, Fritz J, Bunge H, Maier C, Isayev O, Neff F, Siveke JT, Karakhanova S. Pivotal antitumor role of the immune checkpoint molecule B7-H1 in pancreatic cancer. Oncoimmunology 2022; 11:2043037. [PMID: 35251770 PMCID: PMC8890402 DOI: 10.1080/2162402x.2022.2043037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Immune checkpoint molecule B7-H1 plays a decisive immune regulatory role in different pathologies including cancer, and manipulation of B7-H1 expression became an attractive approach in cancer immunotherapy. Pancreatic cancer (PDAC) is characterized by pronounced immunosuppressive environment and B7-H1 expression correlates with PDAC prognosis. However, the first attempts to diminish B7-H1 expression in patients were not so successful. This points the complicity of PDAC immunosuppressive network and requires further examinations. We investigated the effect of B7-H1 deficiency in PDAC. Our results clearly show that partial or complete B7-H1 inhibition in vivo let to reduced tumor volume and improved survival of PDAC-bearing mice. This oncological benefit is due to the abrogation of immunosuppression provided by MDSC, macrophages, DC and Treg, which resulted in simultaneous restoration of anti-tumor immune response, namely improved accumulation and functionality of effector-memory CD4 and CD8 T cells. Our results underline the potential of B7-H1 molecule to control immunosuppressive network in PDAC and provide new issues for further clinical investigations.
Collapse
Affiliation(s)
- Alexandr V. Bazhin
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Katharina von Ahn
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Jasmin Fritz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Henriette Bunge
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Caroline Maier
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Orkhan Isayev
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku, Azerbaijan
| | - Florian Neff
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site University Hospital Essen, Heidelberg, Germany
| | - Jens T. Siveke
- Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site University Hospital Essen, Heidelberg, Germany
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Svetlana Karakhanova
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
30
|
Ruiz Hispán E, Pedregal M, Cristobal I, García-Foncillas J, Caramés C. Immunotherapy for Peritoneal Metastases from Gastric Cancer: Rationale, Current Practice and Ongoing Trials. J Clin Med 2021; 10:4649. [PMID: 34682772 PMCID: PMC8539177 DOI: 10.3390/jcm10204649] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 02/07/2023] Open
Abstract
Peritoneal metastases from gastric cancer play a key role in the fatal prognosis of the disease. The lack of efficacy of actual therapeutic approaches together with the outcomes achieved with checkpoint inhibitors in gastric cancer compel us to address the current state-of-the-art immunotherapy treatment of peritoneal dissemination. The immunogenicity of the peritoneum has been described to be particularly active at omentum and peritoneal lymph nodes. Also, both innate and acquired immunity seems to be involved at different molecular levels. Recent works show PDL1 expression being less present at the peritoneal level; however, some clinical trials have begun to yield results. For example, the ATTRACTION-2 trial has demonstrated the activity of Nivolumab in heavily pretreated patients even though peritoneal metastases were diagnosed in a 30% of them. Despite positive results in the metastatic setting, peritoneal responses to systemic checkpoint inhibitors remains unclear, therefore, new strategies for intraperitoneal immunotherapy are being proposed for different ongoing clinical trials.
Collapse
Affiliation(s)
- Eva Ruiz Hispán
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| | - Manuel Pedregal
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| | - Ion Cristobal
- Cancer Unit for Research on Novel Therapeutic Targets, Oncohealth Institute, IIS-Fundación Jiménez Díaz-UAM Madrid, 28040 Madrid, Spain;
| | - Jesús García-Foncillas
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| | - Cristina Caramés
- Department of Oncology, Fundación Jiménez Díaz University Hospital, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain; (E.R.H.); (M.P.)
| |
Collapse
|
31
|
Zhang SX, Liu W, Ai B, Sun LL, Chen ZS, Lin LZ. Current Advances and Outlook in Gastric Cancer Chemoresistance: A Review. Recent Pat Anticancer Drug Discov 2021; 17:26-41. [PMID: 34587888 DOI: 10.2174/1574892816666210929165729] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 08/19/2021] [Accepted: 09/20/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Surgical resection of the lesion is the standard primary treatment of gastric cancer. Unfortunately, most patients are already in the advanced stage of the disease when they are diagnosed with gastric cancer. Alternative therapies, such as radiation therapy and chemotherapy, can achieve only very limited benefits. The emergence of cancer drug resistance has always been the major obstacle to the cure of tumors. The main goal of modern cancer pharmacology is to determine the underlying mechanism of anticancer drugs. OBJECTIVE Here, we mainly review the latest research results related to the mechanism of chemotherapy resistance in gastric cancer, the application of natural products in overcoming the chemotherapy resistance of gastric cancer, and the new strategies currently being developed to treat tumors based on immunotherapy and gene therapy. CONCLUSION The emergence of cancer drug resistance is the main obstacle in achieving alleviation and final cure for gastric cancer. Mixed therapies are considered to be a possible way to overcome chemoresistance. Natural products are the main resource for discovering new drugs specific for treating chemoresistance, and further research is needed to clarify the mechanism of natural product activity in patients. .
Collapse
Affiliation(s)
- Sheng-Xiong Zhang
- Guangdong Province Work Injury Rehabilitation Hospital, Guangzhou, 510440. China
| | - Wei Liu
- College of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006. China
| | - Bo Ai
- Huazhong University of Science and Technology, Wuhan, 430030. China
| | - Ling-Ling Sun
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405. China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, New York. United States
| | - Li-Zhu Lin
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405. China
| |
Collapse
|
32
|
Yang Y, Yang Y, Chen M, Chen J, Wang J, Ma Y, Qian H. Injectable shear-thinning polylysine hydrogels for localized immunotherapy of gastric cancer through repolarization of tumor-associated macrophages. Biomater Sci 2021; 9:6597-6608. [PMID: 34582523 DOI: 10.1039/d1bm01053k] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Immunotherapy has emerged as one of the most promising treatments for cancer in recent years. However, it works only for a small proportion of patients, which can in part be attributed to the immunosuppressive tumor microenvironment (TME). Tumor associated macrophages (TAMs) are the critical components of tumors and play an important role in the development of the immunosuppressive TME. The transition of TAMs from the pro-tumor (M2) phenotype to anti-tumor (M1) phenotype is crucial for the immunotherapy of gastric cancer. Herein, we developed a shear-thinning, injectable hydrogel co-loaded with polyphyllin II (PP2) and resiquimod (R848) (PR-Gel) for potentiating localized immunotherapy of gastric cancer through the repolarization of TAMs. In this work, we evaluate the effects of PR-Gel on TAM repolarization and explored its therapeutic effect for localized immunotherapy. The hydrogels were synthesized through the Schiff base reactions between aldehyde-functionalized polyethylene glycol and the amino group of polylysine. A M2-to-M1 repolarization of TAMs and increased production of TNF-α and IL-6 were observed after treatment with PR-Gel in vitro. The anti-tumor efficacy of PR-Gel in a subcutaneous xenograft model of gastric cancer showed that the hydrogels possess good tumor growth suppression properties after a single injection. Furthermore, an increased iNOS/CD206 ratio in TAMs and enhanced CD8+ T cell infiltration were also observed within the TME after the treatment with PR-Gel. Hence, the biocompatible, shear-thinning, injectable hydrogels are a promising noninvasive drug-delivery platform for the regulation of the immunosuppressive TME and have great potential in localized immunotherapy against gastric cancer.
Collapse
Affiliation(s)
- Yan Yang
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| | - Yang Yang
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, Jiangsu, China
| | - Meili Chen
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| | - Jianquan Chen
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| | - Jinyan Wang
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| | - Yajun Ma
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| | - Hanqing Qian
- Central Laboratory, Translational Medicine Research Center, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing 211100, Jiangsu, China.
| |
Collapse
|
33
|
Hang Q, Ying H, Cheng G, Yang S, Jin J, Chen Y, Chen Q, Jiang Y, Zhao Q, Fang M, Chen M, Lai X. [Prognostic Analysis of NSCLC Based on the Tumor-associated Macrophages, Tumor Neo-vessels and PD-L1 Expression in Tumor Microenvironment]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 23:837-844. [PMID: 33070512 PMCID: PMC7583870 DOI: 10.3779/j.issn.1009-3419.2020.103.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
背景与目的 肿瘤微环境是肿瘤细胞赖以生存的复杂环境。其中肿瘤相关巨噬细胞(tumor-associated macrophages, TAMs)、肿瘤新生血管及程序性死亡受体1/程序性死亡受体-配体1(programmed cell death protein 1/programmed cell death ligand 1, PD-1/PD-L1)作为关键部分,在肿瘤发生、发展过程中起重要作用,影响患者预后。本研究旨在阐明TAMs、肿瘤新生血管和PD-L1的表达与非小细胞肺癌(non-small cell lung cancer, NSCLC)临床病理特征的相关性,并探讨它们对NSCLC预后的影响。 方法 收集92例NSCLC患者的临床病理资料及手术标本,采用免疫组化法检测癌组织和癌旁组织中TAMs、肿瘤新生血管和PD-L1的表达,采用配备有Olympus-DP72图像采集系统的Olympus-BX51正置显微镜进行拍照并用Image-pro Plus 6.0软件进行半定量分析。 结果 癌组织与癌旁组织中TAMs、肿瘤新生血管和PD-L1的表达差异无统计学意义(P > 0.05)。根据肿瘤微环境中各组分的定量表达,可将其分为低、中、高表达组。癌组织中TAMs的低、中和高密度组的中位总生存(overall survival, OS)分别是36个月(95%CI: 25.3-46.7)、26个月(95%CI: 12.2-39.8)和16个月(95%CI: 9.4-22.6),差异具有统计学意义(P=0.016);肿瘤新生血管的低、中和高密度组的中位OS分别为30个月(95%CI: 22.5-37.5)、28个月(95%CI: 18.1-37.9)和25个月(95%CI: 14.6-35.4),差异无统计学意义(P=0.626);PD-L1的低、中和高表达组的中位OS分别为35个月(95%CI: 29.4-40.6),28个月(95%CI: 13.6-42.4)和17个月(95%CI: 10.5-23.5),差异具有统计学意义(P=0.002)。联合低、中和高表达组的中位OS分别为36个月(95%CI: 30.6-41.4)、26个月(95%CI: 19.2-32.8)和9个月(95%CI: 4.4-13.6),差异具有统计学意义(P=0.001)。Cox回归分析结果显示,病理分型、TAMs和PD-L1均为肺癌患者的独立预后因素。 结论 肿瘤微环境关键成分PD-L1及TAMs的表达与NSCLC患者的预后密切相关。
Collapse
Affiliation(s)
- Qingqing Hang
- The Second Clinical Medical College Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Hangjie Ying
- Zhejiang Key Laboratory of Radiation Oncology, Department of Thoracic Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Guoping Cheng
- Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Shifeng Yang
- Department of Pathology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Jianan Jin
- The Second Clinical Medical College Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yamei Chen
- Zhejiang Key Laboratory of Radiation Oncology, Department of Thoracic Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Qixun Chen
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Youhua Jiang
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Qiang Zhao
- Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Min Fang
- Zhejiang Key Laboratory of Radiation Oncology, Department of Thoracic Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Ming Chen
- Zhejiang Key Laboratory of Radiation Oncology, Department of Thoracic Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Xiaojing Lai
- Zhejiang Key Laboratory of Radiation Oncology, Department of Thoracic Radiotherapy, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China
| |
Collapse
|
34
|
Jiang Q, Sun J, Chen H, Ding C, Tang Z, Ruan Y, Liu F, Sun Y. Establishment of an Immune Cell Infiltration Score to Help Predict the Prognosis and Chemotherapy Responsiveness of Gastric Cancer Patients. Front Oncol 2021; 11:650673. [PMID: 34307129 PMCID: PMC8299334 DOI: 10.3389/fonc.2021.650673] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/22/2021] [Indexed: 12/24/2022] Open
Abstract
The immune microenvironment plays a critical role in tumor biology. The molecular profiles of immune components and related genes are of tremendous value for the study of primary resistance to immune checkpoint blockers (ICBs) for gastric cancer (GC) and serve as prognostic biomarkers to predict GC survival. Recent studies have revealed that tumor immune cell infiltration (ICI) is an indicator of the survival and responsiveness to chemotherapy in GC patients. Here, we describe the immune cell landscape based on the ESTIMATE and CIBERSORT algorithms to help separate GC into 3 ICI clusters using the unsupervised clustering method. Further in-depth analyses, such as differential expression gene (DEG) analysis and principal component analysis (PCA), help to establish an ICI scoring system. A low ICI score is characterized by an increased tumor mutation burden (TMB). The combination of the ICI score and TMB score better predicts the survival of GC patients. Analyses based on public and our own database revealed that the ICI scoring system could also help predict the survival and chemotherapy responsiveness of GC patients. The present study demonstrated that the ICI score may be an effective prognostic biomarker and predictive indicator for chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Quan Jiang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Human Phenome Institute, Fudan University, Shanghai, China
| | - Jie Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Ding
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhaoqing Tang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuanyuan Ruan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Fenglin Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Gao L, Wang X, Yang X, Gu R, Zhu G, Gao X. A clinicopathologic analysis of microscopic extension in small cell lung cancer and lung adenocarcinoma: Determination of clinical target volume with precise radiotherapy. Thorac Cancer 2021; 12:1973-1982. [PMID: 34028192 PMCID: PMC8258354 DOI: 10.1111/1759-7714.14000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/14/2022] Open
Abstract
PURPOSE The identification of the clinical target volume (CTV) is particularly important in the precise radiotherapy of lung cancer. The purpose of this study was to determine the extension margin from gross tumor volume (GTV) to CTV in primary small cell lung cancer (SCLC) and lung adenocarcinoma (ADC) by microscopic extension (ME). MATERIAL AND METHODS The data of 25 cases of SCLC and 29 cases of ADC from August 2015 to August 2020 were analyzed. The measurement of tumor size between preoperative thoracic computed tomography (CT) and postoperative macroscopic specimens was compared, and the ME range of tumor cells was measured under a microscope to determine its correlation with clinical features and pathological manifestations. RESULTS A total of 217 slides were examined, corresponding to 103 slides for SCLC and 114 slides for ADC. The radiologic sizes of the tumors in SCLC and ADC were 12.8 and 7.9 mm, respectively (p = 0.09), and the macroscopic sizes were 12.5 and 8.5 mm, respectively (p = 0.07). There was a significant correlation between the radiologic and macroscopic size of the same tumor sample (r = 0.886). Compared with ADC, more SCLC tumor cells infiltrated through vascular or lymphatic dissemination (16% vs. 9%, p = 0.047). The mean ME value was 2.81 mm for SCLC and 2.02 mm for ADC (p = 0.012). To take into account 95% of the ME, a margin of 8 and 7.7 mm must be expanded for SCLC and ADC, respectively. The ME value of the tumor was related to the presence of atelectasis, the location of the tumor, and the Ki-67 cell proliferation index. CONCLUSION The GTV of the tumor was contoured according to CT images, which was basically consistent with the actual tumor size. The GTVs of SCLC and ADC should be expanded by 8 and 7.7 mm, respectively, to fully cover the subclinical lesions in 95% of cases.
Collapse
Affiliation(s)
- Liwei Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| | - Xiuhong Wang
- Department of Pathology, China-Japan Friendship Hospital, Beijing, China
| | - Xiongtao Yang
- Department of Radiation Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Runchuan Gu
- Department of Radiation Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Guangying Zhu
- Department of Radiation Oncology, China-Japan Friendship Hospital, Beijing, China
| | - Xianshu Gao
- Department of Radiation Oncology, Peking University First Hospital, Beijing, China
| |
Collapse
|
36
|
Yin T, Xin H, Yu J, Teng F. The role of exosomes in tumour immunity under radiotherapy: eliciting abscopal effects? Biomark Res 2021; 9:22. [PMID: 33789758 PMCID: PMC8011088 DOI: 10.1186/s40364-021-00277-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/19/2021] [Indexed: 12/11/2022] Open
Abstract
As a curative treatment of localized tumours or as palliative control, radiotherapy (RT) has long been known to kill tumour cells and trigger the release of proinflammatory factors and immune cells to elicit an immunological response to cancer. As a crucial part of the tumour microenvironment (TME), exosomes, which are double-layered nanometre-sized vesicles, can convey molecules, present antigens, and mediate cell signalling to regulate tumour immunity via their contents. Different contents result in different effects of exosomes. The abscopal effect is a systemic antitumour effect that occurs outside of the irradiated field and is associated with tumour regression. This effect is mediated through the immune system, mainly via cell-mediated immunity, and results from a combination of inflammatory cytokine cascades and immune effector cell activation. Although the abscopal effect has been observed in various malignancies for many years, it is still a rarely identified clinical event. Researchers have indicated that exosomes can potentiate abscopal effects to enhance the effects of radiation, but the specific mechanisms are still unclear. In addition, radiation can affect exosome release and composition, and irradiated cells release exosomes with specific contents that change the cellular immune status. Hence, fully understanding how radiation affects tumour immunity and the interaction between specific exosomal contents and radiation may be a potential strategy to maximize the efficacy of cancer therapy. The optimal application of exosomes as novel immune stimulators is under active investigation and is described in this review.
Collapse
Affiliation(s)
- Tianwen Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Huixian Xin
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
37
|
Wei Y, Liang M, Xiong L, Su N, Gao X, Jiang Z. PD-L1 induces macrophage polarization toward the M2 phenotype via Erk/Akt/mTOR. Exp Cell Res 2021; 402:112575. [PMID: 33771483 DOI: 10.1016/j.yexcr.2021.112575] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/04/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023]
Abstract
PD-L1 (programmed death-ligand 1) is the ligand of PD-1 (programmed cell death protein 1) and regulates inhibitory immune responses. It is well known that PD-L1 suppresses T cell function via binding to PD-1. However, little is known about the role of the PD-1/PD-L1 axis in macrophage polarization. According to previous studies, the function of the PD-1/PD-L1 axis in macrophage polarization is controversial, and the underlying mechanism has not been fully elucidated. Thus, we treated THP-1-derived macrophages with human PD-L1 Fc to determine the role of the PD-1/PD-L1 axis in macrophage polarization. To further explore the mechanism, we performed RNA sequencing and used specific inhibitors to identify the implicated signalling pathways. In this study, we found that PD-L1 induces the upregulation of CD206 expression, which is inhibited by nivolumab, LY294002, U0126, and rapamycin. Evaluation of differentially expressed genes (DEGs) and bioinformatics analysis indicated that PD-L1 also induces the upregulation of the expression of genes that maintain mitochondrial function and mediate metabolic switching. In addition, we did not detect PD-L1-induced CD86 alterations, indicating that PD-L1 treatment has no significant influence on M1 polarization. Taken together, these results suggest that PD-L1 binds to PD-1 and promotes M2 polarization accompanied by mitochondrial function enhancement and metabolic reprogramming via Erk/Akt/mTOR. This study elucidates the role of PD-L1 in macrophage polarization and verifies the underlying mechanisms for the first time. Considering that aberrantly upregulated PD-L1 expression contributes to a wide variety of diseases, targeting PD-L1-mediated macrophage polarization is a prospective therapeutic strategy for both neoplastic and nonneoplastic diseases.
Collapse
Affiliation(s)
- Yi Wei
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mengjun Liang
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liping Xiong
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ning Su
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiang Gao
- Department of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zongpei Jiang
- Department of Nephrology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
38
|
Petty AJ, Dai R, Lapalombella R, Baiocchi RA, Benson DM, Li Z, Huang X, Yang Y. Hedgehog-induced PD-L1 on tumor-associated macrophages is critical for suppression of tumor-infiltrating CD8+ T cell function. JCI Insight 2021; 6:146707. [PMID: 33749663 PMCID: PMC8026184 DOI: 10.1172/jci.insight.146707] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The programmed death-1 (PD-1) and the PD ligand 1 (PD-L1) interaction represents a key immune checkpoint within the tumor microenvironment (TME), and PD-1 blockade has led to exciting therapeutic advances in clinical oncology. Although IFN-γ–dependent PD-L1 induction on tumor cells was initially thought to mediate the suppression on effector cells, recent studies have shown that PD-L1 is also expressed at high level on tumor-associated macrophages (TAMs) in certain types of tumors. However, the precise role of PD-L1 expression on TAMs in suppressing antitumor immunity within the TME remains to be defined. Using a myeloid-specific Pdl1-knockout mouse model, here we showed definitive evidence that PD-L1 expression on TAMs is critical for suppression of intratumor CD8+ T cell function. We further demonstrated that tumor-derived Sonic hedgehog (Shh) drives PD-L1 expression in TAMs to suppress tumor-infiltrating CD8+ T cell function, leading to tumor progression. Mechanistically, Shh-dependent upregulation of PD-L1 in TAMs is mediated by signal transducer and activator of transcription 3, a cascade that has not been previously reported to our knowledge. Last, single-cell RNA sequencing analysis of human hepatocellular carcinoma revealed that PD-L1 is mainly expressed on M2 TAMs, supporting the clinical relevance of our findings. Collectively, our data revealed an essential role for Shh-dependent PD-L1 upregulation in TAMs in suppressing antitumor immunity within the TME, which could lead to the development of new immunotherapeutic strategies for treating cancer.
Collapse
Affiliation(s)
- Amy J Petty
- Department of Medicine and.,Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA.,Division of Hematology
| | - Rui Dai
- Department of Medicine and.,Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA.,Division of Hematology
| | | | | | | | - Zihai Li
- Division of Medical Oncology, and.,Pelotonia Institute for Immuno-Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | | | - Yiping Yang
- Department of Medicine and.,Division of Hematology.,Pelotonia Institute for Immuno-Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
39
|
Zhang Y, Yang H, Zhao J, Wan P, Hu Y, Lv K, Hu Y, Yang X, Ma M. Activation of MAT2A-RIP1 signaling axis reprograms monocytes in gastric cancer. J Immunother Cancer 2021; 9:e001364. [PMID: 33593829 PMCID: PMC7888314 DOI: 10.1136/jitc-2020-001364] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The activation of tumor-associated macrophages (TAMs) facilitates the progression of gastric cancer (GC). Cell metabolism reprogramming has been shown to play a vital role in the polarization of TAMs. However, the role of methionine metabolism in function of TAMs remains to be explored. METHODS Monocytes/macrophages were isolated from peripheral blood, tumor tissues or normal tissues from healthy donors or patients with GC. The role of methionine metabolism in the activation of TAMs was evaluated with both in vivo analyses and in vitro experiments. Pharmacological inhibition of the methionine cycle and modulation of key metabolic genes was employed, where molecular and biological analyses were performed. RESULTS TAMs have increased methionine cycle activity that are mainly attributed to elevated methionine adenosyltransferase II alpha (MAT2A) levels. MAT2A modulates the activation and maintenance of the phenotype of TAMs and mediates the upregulation of RIP1 by increasing the histone H3K4 methylation (H3K4me3) at its promoter regions. CONCLUSIONS Our data cast light on a novel mechanism by which methionine metabolism regulates the anti-inflammatory functions of monocytes in GC. MAT2A might be a potential therapeutic target for cancer cells as well as TAMs in GC.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, China
- Department of Gastroenterology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Hui Yang
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Jun Zhao
- Department of General Surgery, Yijishan Hospital, The First Aflliated Hospital of Wannan Medical College, Wuhu, China
| | - Ping Wan
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Ye Hu
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Lv
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - YiRen Hu
- Department of General Surgery, Wenzhou No. 3 Clinical Institute of Wenzhou Medical University,Wenzhou People's Hospital, Wenzhou, China
| | - Xi Yang
- Shanghai Institute of Head Trauma, Shanghai, China
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Mingzhe Ma
- Key Laboratory of Non-coding RNA Transformation Research of Anhui Higher Education Institution (Wannan Medical College), Wuhu, China
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Immune Cell Landscape in Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2021; 2021:1930706. [PMID: 33575321 PMCID: PMC7857889 DOI: 10.1155/2021/1930706] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/28/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023]
Abstract
Background The tumor-infiltrating immune cells are closely associated with the prognosis of gastric cancer (GC). This article is aimed at determining the composition change of immune cells and immune regulatory factors in GC and normal tissues, depicting their prognosis value in GC, and revealing the relationship between them and GC clinical parameters. Methods We used CIBERSORT to calculate the proportion of 22 immune cells in the GC or normal tissues; a t-test was applied to assess the expression difference of immune cells and immune regulatory factors in normal and GC tissues. The relationship of the immune cells, immune regulatory factors, and GC patients' clinical characteristics was assessed by univariate analysis. Results In this study, we found that the proportion of macrophages increased, while plasma cells and monocytes decreased in GC tissues. In these immune fractions, Tregs and naïve B cells were found to be correlated with GC patients' prognosis. Interestingly, the expression of immune regulatory factors was ambiguous with their classical function in GC tissues. For example, TIM-3, FOXP3, and CMTM6 were overexpressed, while CD27 and PD-1 were underexpressed in GC tissues. We also found that IDO1, PD-1, TIGIT, and TIM-3 were highly expressed in high-grade GC tissues, the HERC2 expression level was related to patients' gender, and the TIGIT expression level was sensitive to targeted therapy. Furthermore, our results suggested that the infiltration of Tregs and naive B cells was strongly correlated with the T stage, radiation therapy, targeted molecular therapy, and the expression levels of TIM-3 and FOXP3 in GC. Conclusion The expression pattern of tumor-infiltrating immune cells and immune regulatory factors was systematically depicted in the GC tumor microenvironment, indicating that individualized treatment based on the tumor-infiltrating immune cells and immune regulatory factors may be beneficial to GC patients.
Collapse
|
41
|
Zhu S, Luo Z, Li X, Han X, Shi S, Zhang T. Tumor-associated macrophages: role in tumorigenesis and immunotherapy implications. J Cancer 2021; 12:54-64. [PMID: 33391402 PMCID: PMC7738842 DOI: 10.7150/jca.49692] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor-associated macrophages (TAMs) occupy an important position in the tumor microenvironment (TME), they are a highly plastic heterogeneous population with complex effects on tumorigenesis and development. TAMs secrete a variety of cytokines, chemokines, and proteases, which promote the remodeling of extracellular matrix, tumor cell growth and metastasis, tumor vessel and lymphangiogenesis, and immunosuppression. TAMs with different phenotypes have different effects on tumor proliferation and metastasis. TAMs act a pivotal part in occurrence and development of tumors, and are very attractive target to inhibit tumor growth and metastasis in tumor immunotherapy. This article reviews the interrelationship between TAMs and tumor microenvironment and its related applications in tumor therapy.
Collapse
Affiliation(s)
- Shunyao Zhu
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ziyi Luo
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xixi Li
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xi Han
- Xiaoshan Hosptital of Traditional Chinese Medicine, Hangzhou 311201, China
| | - Senlin Shi
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ting Zhang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| |
Collapse
|
42
|
Zhao R, Wan Q, Wang Y, Wu Y, Xiao S, Li Q, Shen X, Zhuang W, Zhou Y, Xia L, Song Y, Chen Y, Yang H, Wu X. M1-like TAMs are required for the efficacy of PD-L1/PD-1 blockades in gastric cancer. Oncoimmunology 2020; 10:1862520. [PMID: 33457080 PMCID: PMC7781754 DOI: 10.1080/2162402x.2020.1862520] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The efficacy of PD-1/PD-L1 blockades is heterogeneous in different molecular subtypes of gastric cancer (GC). In this study, we analyzed relevant clinical trials to identify the molecular subtypes associated with the efficacy of PD-1/PD-L1 blockades, and public datasets, patient samples, and GC cell lines were used for investigating potential mechanisms. We found that GC with EBV-positive, MSI-H/dMMR, TMB-H or PIK3CA mutant subtype had enhanced efficacy of PD-L1/PD-1 blockades. Also, differentially expressed genes of these molecular subtypes shared the same gene signature and functional annotations related to immunity. Meanwhile, CIBERSORT identified that the overlapping landscapes of tumor-infiltrating immune cells in the four molecular subtypes were mainly M1-like macrophages (M1). The relationships between M1 and clinical characteristics, M1, and gene signatures associated with PD-1/PD-L1 blockades also revealed that M1 was associated with improved prognosis and required for the efficacy of PD-L1/PD-1 blockades in GC. We identified that tumor-infiltrating CD68+CD163− macrophages could represent M1 calculated by CIBERSORT in clinical application, and CXCL9, 10, 11/CXCR3 axis was involved in the mechanism of CD68+CD163− macrophages in the enhanced efficacy of PD-L1/PD-1 blockades. In conclusion, CD68+CD163− macrophages are required for the efficacy of PD-L1/PD-1 blockades and expand the applicable candidates in GC patients without the molecular subtypes.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qianyi Wan
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yutao Wu
- West China College of Stomatology, West China Dental Hospital, Sichuan University, Chengdu, China
| | - Shuomeng Xiao
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiqi Li
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Xiaoding Shen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Zhuang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Zhou
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Xia
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yinghan Song
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Chen
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Xiaoting Wu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Wang JB, Li P, Liu XL, Zheng QL, Ma YB, Zhao YJ, Xie JW, Lin JX, Lu J, Chen QY, Cao LL, Lin M, Liu LC, Lian NZ, Yang YH, Huang CM, Zheng CH. An immune checkpoint score system for prognostic evaluation and adjuvant chemotherapy selection in gastric cancer. Nat Commun 2020; 11:6352. [PMID: 33311518 PMCID: PMC7732987 DOI: 10.1038/s41467-020-20260-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 11/18/2020] [Indexed: 12/24/2022] Open
Abstract
Immunosuppressive molecules are extremely valuable prognostic biomarkers across different cancer types. However, the diversity of different immunosuppressive molecules makes it very difficult to accurately predict clinical outcomes based only on a single immunosuppressive molecule. Here, we establish a comprehensive immune scoring system (ISSGC) based on 6 immunosuppressive ligands (NECTIN2, CEACAM1, HMGB1, SIGLEC6, CD44, and CD155) using the LASSO method to improve prognostic accuracy and provide an additional selection strategy for adjuvant chemotherapy of gastric cancer (GC). The results show that ISSGC is an independent prognostic factor and a supplement of TNM stage for GC patients, and it can improve their prognosis prediction accuracy; in addition, it can distinguish GC patients with better prognosis from those with high prognostic nutritional index score; furthermore, ISSGC can also be used as a tool to select GC patients who would benefit from adjuvant chemotherapy independent of their TNM stages, MSI status and EBV status.
Collapse
Affiliation(s)
- Jia-Bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Xiao-Long Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Fuzhou, China
| | - Qiao-Ling Zheng
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yu-Bin Ma
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qinghai University, Xining, China
| | - Ya-Jun Zhao
- Division of Life Sciences and Medicine, Department of Gastrointestinal Surgery, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Long-Long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Li-Chao Liu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ning-Zi Lian
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China
| | - Ying-Hong Yang
- Department of Pathology, Fujian Medical University Union Hospital, Fuzhou, China.
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, China.
- Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
44
|
Antonangeli F, Natalini A, Garassino MC, Sica A, Santoni A, Di Rosa F. Regulation of PD-L1 Expression by NF-κB in Cancer. Front Immunol 2020; 11:584626. [PMID: 33324403 PMCID: PMC7724774 DOI: 10.3389/fimmu.2020.584626] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 08/25/2020] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoints are inhibitory receptor/ligand pairs regulating immunity that are exploited as key targets of anti-cancer therapy. Although the PD-1/PD-L1 pair is one of the most studied immune checkpoints, several aspects of its biology remain to be clarified. It has been established that PD-1 is an inhibitory receptor up-regulated by activated T, B, and NK lymphocytes and that its ligand PD-L1 mediates a negative feedback of lymphocyte activation, contributing to the restoration of the steady state condition after acute immune responses. This loop might become detrimental in the presence of either a chronic infection or a growing tumor. PD-L1 expression in tumors is currently used as a biomarker to orient therapeutic decisions; nevertheless, our knowledge about the regulation of PD-L1 expression is limited. The present review discusses how NF-κB, a master transcription factor of inflammation and immunity, is emerging as a key positive regulator of PD-L1 expression in cancer. NF-κB directly induces PD-L1 gene transcription by binding to its promoter, and it can also regulate PD-L1 post-transcriptionally through indirect pathways. These processes, which under conditions of cellular stress and acute inflammation drive tissue homeostasis and promote tissue healing, are largely dysregulated in tumors. Up-regulation of PD-L1 in cancer cells is controlled via NF-κB downstream of several signals, including oncogene- and stress-induced pathways, inflammatory cytokines, and chemotherapeutic drugs. Notably, a shared signaling pathway in epithelial cancers induces both PD-L1 expression and epithelial–mesenchymal transition, suggesting that PD-L1 is part of the tissue remodeling program. Furthermore, PD-L1 expression by tumor infiltrating myeloid cells can contribute to the immune suppressive features of the tumor environment. A better understanding of the interplay between NF-κB signaling and PD-L1 expression is highly relevant to cancer biology and therapy.
Collapse
Affiliation(s)
- Fabrizio Antonangeli
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
| | - Ambra Natalini
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
| | - Marina Chiara Garassino
- Medical Oncology Department, Istituto Nazionale dei Tumori, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Antonio Sica
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, A. Avogadro, Novara, Italy.,Humanitas Clinical and Research Center, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Laboratory Affiliated to Istituto Pasteur Italia, Sapienza University of Rome, Rome, Italy
| | - Francesca Di Rosa
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Rome, Italy
| |
Collapse
|
45
|
Taghiloo S, Asgarian-Omran H. Immune evasion mechanisms in acute myeloid leukemia: A focus on immune checkpoint pathways. Crit Rev Oncol Hematol 2020; 157:103164. [PMID: 33271388 DOI: 10.1016/j.critrevonc.2020.103164] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/09/2020] [Accepted: 11/05/2020] [Indexed: 12/14/2022] Open
Abstract
Immune surveillance mechanisms comprising of adaptive and innate immune systems are naturally designed to eliminate AML development. However, leukemic cells apply various immune evasion mechanisms to deviate host immune responses resulting tumor progression. One of the recently well-known immune escape mechanisms is over-expression of immune checkpoint receptors and their ligands. Introduction of blocking antibodies targeting co-inhibitory molecules achieved invaluable success in tumor targeted therapy. Moreover, several new co-inhibitory pathways are currently studying for their potential impacts on improving anti-tumor immune responses. Although immunotherapeutic strategies based on the blockade of immune checkpoint molecules have shown promising results in a number of hematological malignances, their effectiveness in AML patients showed less remarkable success. This review discusses current knowledge about the involvement of co-inhibitory signaling pathways in immune evasion mechanisms of AML and potential application of immune checkpoint inhibitors for targeted immunotherapy of this malignancy.
Collapse
Affiliation(s)
- Saeid Taghiloo
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Asgarian-Omran
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Gastrointestinal Cancer Research Center, Non-Communicable Diseases Institute, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
46
|
Deng R, Lu J, Liu X, Peng XH, Wang J, Li XP. PD-L1 Expression is Highly Associated with Tumor-Associated Macrophage Infiltration in Nasopharyngeal Carcinoma. Cancer Manag Res 2020; 12:11585-11596. [PMID: 33209062 PMCID: PMC7669506 DOI: 10.2147/cmar.s274913] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/12/2020] [Indexed: 01/06/2023] Open
Abstract
Purpose Tumour-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment and provide a barrier against the cytotoxic effector functions of T cells and natural killer (NK) cells. Recently, TAMs have become increasingly recognised as an attractive target in combination therapy with PD-1/PD-L1 immuno-checkpoint blockades (ICBs). However, the relationship between PD-L1 expression and TAMs remains unknown in nasopharyngeal carcinoma (NPC). Patients and Methods A total of 212 NPC patients from Nanfang hospital were collected in this study. We evaluated the expression of PD-L1 in tumor cells, CD68 (pan-macrophages), and CD163 (M2-like macrophage) in NPC tissues using immunohistochemical (IHC) staining. Results The positivity of PD-L1 on tumor cells was 61.3% (130/212). The infiltration densities of CD68+ cells and CD163+ cells in PD-L1-positive NPC tissues were significantly higher than those in PD-L1-negative NPC tissues (P=0.0012 for CD68; P<0.0001 for CD163). Logistic regression analysis showed that high densities of CD68+ macrophages and CD163+ TAMs were significantly associated with increased PD-L1 expression. Subgroup analyses demonstrated that a positive PD-L1 expression on tumor cells in combination with lower CD163+ TAMs density was significantly associated with favorable prognosis, whereas negative PD-L1 expression on tumor cells with higher CD163+ TAMs density was associated with worse prognosis. Conclusion The PD-L1 expression in tumor cells was positively correlated with TAMs density in tumor microenvironment of NPC, suggesting TAMs as a new target for combination therapy to improve the response rate of ICBs in NPC treatment.
Collapse
Affiliation(s)
- Rui Deng
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Juan Lu
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiong Liu
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiao-Hong Peng
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jie Wang
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Xiang-Ping Li
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
47
|
Ubukata Y, Ogata K, Sohda M, Yokobori T, Shimoda Y, Handa T, Nakazawa N, Kimura A, Kogure N, Sano A, Sakai M, Ogawa H, Kuwano H, Shirabe K, Oyama T, Saeki H. Role of PD-L1 Expression during the Progression of Submucosal Gastric Cancer. Oncology 2020; 99:15-22. [PMID: 33113541 DOI: 10.1159/000509033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 05/28/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Programmed death-ligand 1 (PD-L1) expression is a prognostic marker for gastric cancer that correlates with tumor diameter and depth of penetration. But the role of PD-L1 and mechanism(s) employed in the initial phase of invasion in early gastric cancer is yet to be understood. OBJECTIVE This study aims to elucidate the role of PD-L1 during the progression of gastric cancer, specifically invading the submucosa beyond the lamina muscularis mucosa. METHODS Using 107 patients with pathological submucosal gastric cancer, we determined the expression of PD-L1 based on the staining of the cell membrane or cytoplasm of tumor cells in the central and invasive front of the tumor. Samples were categorized into 3 groups based on the intensity of PD-L1 expression. CD8+ lymphocytes expressing PD-1 and CD163+ macrophages were used to determine the number of cell nuclei at the invasive front, similar to PD-L1. CMTM6 levels were determined and used to stratify samples into 3 groups. RESULTS PD-L1 expression was higher in the invasive front (26.2%) than in the central portion of the tumors (7.4%; p < 0.001). Moreover, lymphatic and vascular invasion were more frequently observed in samples with high levels of PD-L1 (lymphatic invasion: 60.7 vs. 35.4%, p = 0.0026, and vascular invasion: 39.3 vs. 16.5%, p = 0.0018). There was no correlation between PD-L1 expression and the levels of PD-1, CD8, CD163, and CMTM6. CONCLUSIONS PD-L1-expressing cancer cells at the invasive front of gastric cancer influence the initial stages of tumor invasion and lymphovascular permeation in early-stage gastric cancers. Immune checkpoint signaling may be the driving force in the invasive front during the invasion of the submucosa beyond the lamina muscularis mucosa.
Collapse
Affiliation(s)
- Yasunari Ubukata
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Kyoichi Ogata
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Makoto Sohda
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan,
| | - Takehiko Yokobori
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Yuki Shimoda
- Department of Pathology, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Tadashi Handa
- Department of Pathology, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Nobuhiro Nakazawa
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Akiharu Kimura
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Norimichi Kogure
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Akihiko Sano
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Makoto Sakai
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Hiroomi Ogawa
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Hiroyuki Kuwano
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Ken Shirabe
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Tetsunari Oyama
- Department of Pathology, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| | - Hiroshi Saeki
- Department of General Surgery, Graduate School of Medical Sciences, Gunma University, Maebashi, Japan
| |
Collapse
|
48
|
Yao X, Ajani JA, Song S. Molecular biology and immunology of gastric cancer peritoneal metastasis. Transl Gastroenterol Hepatol 2020; 5:57. [PMID: 33073052 DOI: 10.21037/tgh.2020.02.08] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/03/2020] [Indexed: 12/24/2022] Open
Abstract
Peritoneal metastases occur in 55-60% of patients with gastric cancer (GC) and are associated with a 2% 5-year overall survival rate. There are limited treatment options for these patients, and no targeted therapy or immunotherapy is available. Rational therapeutic targets remain to be found. In this review, we present the published literature and our own recent experience in molecular biology to identify important molecules and signaling pathways as well as cellular immunity involved in the peritoneal metastasis of GC. We also suggest potential novel strategies for improving the outcomes of GC patients with peritoneal metastasis.
Collapse
Affiliation(s)
- Xiaodan Yao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
49
|
Ju X, Zhang H, Zhou Z, Chen M, Wang Q. Tumor-associated macrophages induce PD-L1 expression in gastric cancer cells through IL-6 and TNF-ɑ signaling. Exp Cell Res 2020; 396:112315. [PMID: 33031808 DOI: 10.1016/j.yexcr.2020.112315] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/08/2020] [Accepted: 10/01/2020] [Indexed: 12/21/2022]
Abstract
PD-1/PD-L1 immune checkpoint blockade therapy has been widely used for the clinical treatment of cancer. However, recent clinical trials have shown that only a small proportion of cancer patients respond to PD1/PD-L1 immunotherapy. The tumor immune microenvironment plays an important regulatory role in PD1/PD-L1 immunotherapy. Macrophages are one of the most important immune cells in the tumor immune microenvironment. In this study, we found a high correlation between macrophage infiltration and PD-L1 expression in gastric cancer (GC) specimens. Further study revealed that infiltrated macrophages released the proinflammatory cytokines TNF-ɑ and IL-6, which induced PD-L1 expression in tumor cells. The release of TNF-ɑ and IL-6 activated the NF-kB and STAT3 signaling pathway to regulate PD-L1 expression. TNF-α, p-65 and STAT3 expression in cancer patients has prognostic value in stomach adenocarcinoma. Furthermore, infiltrated macrophages can also promote GC cell proliferation by inducing PD-L1 expression in GC cells. Taken together, our results suggest that macrophages play a dual role in regulating the expression of PD-L1 in tumor cells. On the one hand, macrophages induce PD-L1 expression in tumor cells, helping tumor cells escape cytotoxic T cell killing; on the other hand, they can promote the proliferation of tumor cells by regulating the expression of PD-L1.
Collapse
Affiliation(s)
- Xiaoli Ju
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Heng Zhang
- Department of General Surgery, Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, Jiangsu, 211200, China
| | - Zidi Zhou
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Miao Chen
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China; Department of Pathology, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Qiang Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
50
|
Han X, Alu A, Xiao Y, Wei Y, Wei X. Hyperprogression: A novel response pattern under immunotherapy. Clin Transl Med 2020; 10:e167. [PMID: 32997401 PMCID: PMC7510779 DOI: 10.1002/ctm2.167] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 02/05/2023] Open
Abstract
Checkpoint blockade therapy has shown significant therapeutic benefits and resulted in durable responses in patients with various tumors. However, accumulating evidence has demonstrated that 4-29% of all patients with cancers with various histologies may suffer from tumor flare following such therapy. This novel tumor response pattern, termed hyperprogression, is a potentially deleterious side effect of checkpoint blockade therapy that accelerates disease progression in a subset of patients. In this review, we describe possible immune checkpoint blockade biomarkers and the epidemiology, different definitions, and predictors of hyperprogression based on the research findings and further present the available evidence supporting pathophysiological hypotheses that might explain hyperprogression during checkpoint blockade therapy. We also compare hyperprogression and pseudoprogression. Finally, we discuss areas requiring further study.
Collapse
Affiliation(s)
- Xue‐jiao Han
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Aqu Alu
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Yi‐nan Xiao
- West China School of MedicineWest China HospitalSichuan UniversityChengduChina
| | - Yu‐quan Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| | - Xia‐wei Wei
- Laboratory of Aging Research and Cancer Drug TargetState Key Laboratory of BiotherapyNational Clinical Research Center for GeriatricsWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|