1
|
Torre-Cea I, Berlana-Galán P, Guerra-Paes E, Cáceres-Calle D, Carrera-Aguado I, Marcos-Zazo L, Sánchez-Juanes F, Muñoz-Félix JM. Basement membranes in lung metastasis growth and progression. Matrix Biol 2025; 135:135-152. [PMID: 39719224 DOI: 10.1016/j.matbio.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 12/26/2024]
Abstract
The lung is a highly vascularized tissue that often harbors metastases from various extrathoracic malignancies. Lung parenchyma consists of a complex network of alveolar epithelial cells and microvessels, structured within an architecture defined by basement membranes. Consequently, understanding the role of the extracellular matrix (ECM) in the growth of lung metastases is essential to uncover the biology of this pathology and developing targeted therapies. These basement membranes play a critical role in the progression of lung metastases, influencing multiple stages of the metastatic cascade, from the acquisition of an aggressive phenotype to intravasation, extravasation and colonization of secondary sites. This review examines the biological composition of basement membranes, focusing on their core components-collagens, fibronectin, and laminin-and their specific roles in cancer progression. Additionally, we discuss the function of integrins as primary mediators of cell adhesion and signaling between tumor cells, basement membranes and the extracellular matrix, as well as their implications for metastatic growth in the lung. We also explore vascular co-option (VCO) as a form of tumor growth resistance linked to basement membranes and tumor vasculature. Finally, the review covers current clinical therapies targeting tumor adhesion, extracellular matrix remodeling, and vascular development, aiming to improve the precision and effectiveness of treatments against lung metastases.
Collapse
Affiliation(s)
- Irene Torre-Cea
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Patricia Berlana-Galán
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Elena Guerra-Paes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Daniel Cáceres-Calle
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Iván Carrera-Aguado
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Laura Marcos-Zazo
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain
| | - Fernando Sánchez-Juanes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain.
| | - José M Muñoz-Félix
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Spain.
| |
Collapse
|
2
|
Hu GS, Zheng ZZ, He YH, Wang DC, Nie R, Liu W. Integrated analysis of proteome and transcriptome profiling reveals pan-cancer-associated pathways and molecular biomarkers. Mol Cell Proteomics 2025:100919. [PMID: 39884577 DOI: 10.1016/j.mcpro.2025.100919] [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: 06/12/2024] [Revised: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
Understanding dysregulated genes and pathways in cancer is critical for precision oncology. Integrating mass spectrometry-based proteomic data with transcriptomic data presents unique opportunities for systematic analyses of dysregulated genes and pathways in pan-cancer. Here, we compiled a comprehensive set of datasets, encompassing proteomic data from 2,404 samples and transcriptomic data from 7,752 samples across 13 cancer types. Comparisons between normal or adjacent normal tissues (ANTs) and tumor tissues identified several dysregulated pathways including mRNA splicing, interferon pathway, fatty acid metabolism, and complement coagulation cascade in pan-cancer. Additionally, pan-cancer up- and down-regulated genes (PCUGs and PCDGs) were also identified. Notably, RRM2 and ADH1B, two genes belong to PCUGs and PCDGs, respectively, were identified as robust pan-cancer diagnostic biomarkers. TNM stage-based comparisons revealed dysregulated genes and biological pathways involved in cancer progression, among which the dysregulation of complement coagulation cascade and epithelial-mesenchymal transition are frequent in multiple types of cancers. A group of pan-cancer continuously up- and down-regulated proteins in different tumor stages (PCCUPs and PCCDPs) were identified. We further constructed prognostic risk stratification models for corresponding cancer types based on dysregulated genes, which effectively predict the prognosis for patients with these cancers. Drug prediction based on PCUPs and PCDPs as well as PCCUPs and PCCDPs revealed that small molecule inhibitors targeting CDK, HDAC, MEK, JAK, PI3K, and others might be effective treatments for pan-cancer, thereby supporting drug repurposing. We also developed web tools for cancer diagnosis, pathologic stage assessment, and risk evaluation. Overall, this study highlights the power of combining proteomic and transcriptomic data to identify valuable diagnostic and prognostic markers as well as drug targets and treatments for cancer.
Collapse
Affiliation(s)
- Guo-Sheng Hu
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, 350117, China; State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Zao-Zao Zheng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Yao-Hui He
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Du-Chuang Wang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Ruichao Nie
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China
| | - Wen Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiang'an South Road, Xiamen, Fujian 361102, China.
| |
Collapse
|
3
|
Chung SY, Yeh YC, Huang CJ, Chiang NJ, Hsu DSS, Chan MH, Lu ML, Hsu TS, Hung YP, Yeh CN, Hsiao M, Chang YC, Wang YC, Chen MH. Comparative impact of tertiary lymphoid structures and tumor-infiltrating lymphocytes in cholangiocarcinoma. J Immunother Cancer 2025; 13:e010173. [PMID: 39870490 PMCID: PMC11772930 DOI: 10.1136/jitc-2024-010173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/01/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Cholangiocarcinoma is a challenging malignancy with limited responses to conventional therapies, particularly immune checkpoint inhibitor therapy. Tumor-infiltrating lymphocytes (TILs) and tertiary lymphoid structures (TLSs) are key components of the tumor microenvironment (TME) and have been implicated in the immune response to cancer. However, the role and difference of TLSs and TILs in patients with cholangiocarcinoma remains unclear. This study elucidates their contributions to the TME. METHODS We examined 16 tumor samples from a single-arm, phase II trial of nivolumab plus modified gemcitabine and S-1 and various datasets. Immunohistochemistry and RNA sequencing were employed to assess TLSs and TILs presence and activity. Differential gene expression and signature of immune cell composition were examined by GeoMx Digital Spatial Profiler and Cancer Transcriptome Altas analysis. RESULTS TLS-positive (N=7) patients demonstrated significantly better immunotherapy outcomes compared with TLS-negative (N=9) patients, including higher objective response rates (71% vs 0%) and disease control rates (100% vs 67%). The presence of TLSs correlated with improved progression-free and overall survival (p=0.03). TLSs were associated with "inflamed" tumors characterized by substantial immune infiltration, particularly involving T and B cells. Gene expression analyses identified significant upregulation of B cell-related genes in TLSs. Additionally, TLSs exhibited higher properties of memory B cells and myeloid dendritic cells but lower levels of innate immune cells compared with TILs. T cells within TLSs showed elevated expression of precursor-exhausted-related genes and lower cytotoxicity signature. Furthermore, TILs in TLS-positive tumors had higher levels of exhaustion signatures compared with TILs in TLS-negative tumors. Clinical data corroborated these findings, with higher PD-L1 and LAG-3 expression in TLS-positive tumors. CONCLUSION Our findings revealed that TILs in TLS-positive tumors have more exhausted T cell signature and PD-1 and LAG-3 protein expression in CCA which support our clinical finding. TLSs can predict favorable immunotherapy responses in patients with cholangiocarcinoma, highlighting their potential as a biomarker and therapeutic target to enhance treatment efficacy.
Collapse
Affiliation(s)
- Shin-Yi Chung
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Chen Yeh
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chien-Jung Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Nai-Jung Chiang
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | | | - Ming-Hsien Chan
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Meng-Lun Lu
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Sheng Hsu
- Institute of Molecular and Cellular Biology, College of Life Sciences and Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ping Hung
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Nan Yeh
- General Surgery, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Chao Wang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Huang Chen
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
4
|
Niu N, Li K, Wang J, Funes V, Espinoza B, Li P, Wang J, Lyman M, He M, Herbst B, Wichroski M, Novosiadly R, Shoucair S, Mou Y, Zheng L. Chemotherapy in synergy with innate immune agonists enhances T cell priming for checkpoint inhibitor treatment in pancreatic cancer. Biomark Res 2025; 13:21. [PMID: 39871312 PMCID: PMC11773940 DOI: 10.1186/s40364-024-00721-7] [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: 11/28/2024] [Accepted: 12/27/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND The combination of conventional chemotherapy and immune checkpoint inhibitors (ICIs) has been unsuccessful for pancreatic ductal adenocarcinoma (PDAC). Administration of maximum tolerated dose of chemotherapy drugs may have immunosuppressive effects. METHODS We thus tested, by using the preclinical model of PDACs including the genetically engineered mouse KPC spontaneous pancreatic tumor model and the pancreatic KPC tumor orthotopic implant model, the combinations of synthetic innate immune agonists including STING and NLRP3 agonist, respectively, and ICIs with or without chemotherapy. RESULTS We found that innate agonists potentiate the role of chemotherapy in inducing effector T cells and subsequently to prime the tumor microenvironment (TME) better for ICI treatments. Triple combination of chemotherapy, innate agonists, and ICIs is superior to single modalities or double modalities in antitumor efficacies. Adding chemotherapy to innate agonists enhances the infiltration of overall CD8+ T cells and the memory cytotoxic subtype. NLRP3 agonist has a less effect than STING agonist on driving the T cell exhaustion. Adding chemotherapy to innate agonists enhances the infiltration of dendritic cells (DCs) in the tumors and CD86+ mature DCs in tumor draining lymph nodes. RNA sequencing analysis of the pancreatic tumors demonstrates the role of the combination of STING/NLRP3 agonist and chemotherapy, but not either treatment modality alone, in upregulating the T cell activation signaling. The NLRP3 agonist-mediated T cell activation is likely through regulating the nitrogen metabolism pathways. CONCLUSION This study supports the clinical testing of both STING and NLRP3 agonists, respectively, in combination with chemotherapy to sensitize PDAC patients for ICI treatments.
Collapse
Affiliation(s)
- Nan Niu
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310003, China
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Keyu Li
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Junke Wang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Division of BiliarySurgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Vanessa Funes
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Birginia Espinoza
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Multidisciplinary Gastrointestinal Cancer Laboratories Program, the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Pan Li
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The First-affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianxin Wang
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The First-affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Melissa Lyman
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Mengni He
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Brian Herbst
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Multidisciplinary Gastrointestinal Cancer Laboratories Program, the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | | | | | - Sami Shoucair
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Yiping Mou
- Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- The Multidisciplinary Gastrointestinal Cancer Laboratories Program, the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Mays Cancer Center, University of Texas Health San Antonio MD Anderson, San Antonio, USA.
| |
Collapse
|
5
|
Zou J, Han W, Hu Y, Zeng C, Li J, Lei W, Cao J, Fei Q, Shao M, Yi J, Cheng Z, Wang L, Wu F, Liu W. Gene mutation, clinical characteristics and pathology in resectable lung adenocarcinoma. World J Surg Oncol 2025; 23:16. [PMID: 39844176 PMCID: PMC11752792 DOI: 10.1186/s12957-025-03680-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/19/2025] [Indexed: 01/24/2025] Open
Abstract
OBJECTIVE With the wide use of CT scan in clinical practice, more lung cancer was diagnosed in resectable stage. Pathological examination and genetic testing have become a routine procedure for lung adenocarcinoma following radical resection. This study analyzed special pathological components and gene mutations to explore their relationship with clinical characteristics and overall survival. METHODS Clinical, pathological, and gene mutation data from 1,118 patients were collected. All patients underwent surgery at the Department of Thoracic Surgery, the Second Xiangya Hospital of Central South University. Patients were grouped based on pathological components and gene mutations. Differences in clinical features and overall survival were analyzed as well. RESULTS Patients with mucinous, neuroendocrine, and poor-differentiated components were presented with more prognostic risk factors, including pleural invasion, carcinothrombosis, STAS, and advanced stages, along with varying frequencies of gene mutations. These factors significantly shortened overall survival. ALK and KRAS mutations were also associated with risk factors such as solid nodules, pleural invasion, STAS, and later stages. However, a significant reduction in overall survival was observed only in patients with the KRAS mutation. Relationship between gene mutations and pathological components still requires further investigation. CONCLUSION Special pathological components (mucinous, neuroendocrine, and poor-differentiated) and gene mutations had an influence on biological behavior of tumors, resulting in different clinical characteristics and prognosis.
Collapse
Affiliation(s)
- Ji'an Zou
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wei Han
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yan Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chao Zeng
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jina Li
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Weixuan Lei
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jieming Cao
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Quanming Fei
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Mengqi Shao
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Junqi Yi
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zeyu Cheng
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Fang Wu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Cancer Mega-Data Intelligent Application and Engineering Research Centre, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Wenliang Liu
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Early Diagnosis and Precision Treatment of Lung Cancer, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
6
|
Tashireva LA, Larionova IV, Ermak NA, Maltseva AA, Livanos EI, Kalinchuk AY, Stakheyeva MN, Kolomiets LA. Predicting immunotherapy efficacy in endometrial cancer: focus on the tumor microenvironment. Front Immunol 2025; 15:1523518. [PMID: 39902047 PMCID: PMC11788352 DOI: 10.3389/fimmu.2024.1523518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/24/2024] [Indexed: 02/05/2025] Open
Abstract
Immunotherapy represents a groundbreaking therapeutic approach, based on the immune system's intrinsic capacity to interfere with tumor progression, that opens the horizons in the treatment of endometrial cancer. However, the clinical efficacy of immunotherapy is hampered by the development of resistance in patients. The resistance to immunotherapy is multifactorial mechanism, encompassed genetic and epigenetic alterations in tumor cells modulating immune checkpoint molecules, resulted in escaping immune surveillance. The tumor microenvironment can orchestrate an immunosuppressive milieu, attenuating the immune response and facilitating tumor progression. To overcome immunotherapeutic resistance in endometrial cancer we must bring to light the mechanisms of intricate interplay between neoplastic cells, the host immune system, and the tumor microenvironment. The identification of predictive biomarkers for immunotherapeutic response and the innovative agents capable of reversing resistance pathways must be developed. Our review summarizes accumulated data on the role of cells of the tumor microenvironment and their regulatory molecules in the mechanisms underlying therapeutic effects of immune checkpoint inhibitors, including resistance to therapy. Major question we raise here - which group of patients is the most favorable to achieve durable immunotherapy response in endometrial cancer?
Collapse
Affiliation(s)
- Liubov A. Tashireva
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Irina V. Larionova
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
- Laboratory of Translational Cellular and Molecular Biomedicine, National Research Tomsk State University, Tomsk, Russia
| | - Nikita A. Ermak
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Anastasia A. Maltseva
- Department of Gynecology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Ekaterina I. Livanos
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Anna Yu. Kalinchuk
- Laboratory of Molecular Therapy of Cancer, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Marina N. Stakheyeva
- Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| | - Larisa A. Kolomiets
- Department of Gynecology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, Russia
| |
Collapse
|
7
|
Lopez de Rodas M, Villalba-Esparza M, Sanmamed MF, Chen L, Rimm DL, Schalper KA. Biological and clinical significance of tumour-infiltrating lymphocytes in the era of immunotherapy: a multidimensional approach. Nat Rev Clin Oncol 2025:10.1038/s41571-024-00984-x. [PMID: 39820025 DOI: 10.1038/s41571-024-00984-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Immune-checkpoint inhibitors (ICIs) have improved clinical outcomes across several solid tumour types. Prominent efforts have focused on understanding the anticancer mechanisms of these agents, identifying biomarkers of response and uncovering resistance mechanisms to develop new immunotherapeutic approaches. This research has underscored the crucial roles of the tumour microenvironment and, particularly, tumour-infiltrating lymphocytes (TILs) in immune-mediated tumour elimination. Numerous studies have evaluated the prognostic and predictive value of TILs and the mechanisms that govern T cell dysfunction, fuelled by technical developments in single-cell transcriptomics, proteomics, high-dimensional spatial platforms and advanced computational models. However, questions remain regarding the definition of TILs, optimal strategies to study them, specific roles of different TIL subpopulations and their clinical implications in different treatment contexts. Additionally, most studies have focused on the abundance of major TIL subpopulations but have not developed standardized quantification strategies or analysed other crucial aspects such as their functional profile, spatial distribution and/or arrangement, tumour antigen-reactivity, clonal diversity and heterogeneity. In this Review, we discuss a conceptual framework for the systematic study of TILs and summarize the evidence regarding their biological properties and biomarker potential for ICI therapy. We also highlight opportunities, challenges and strategies to support future developments in this field.
Collapse
Affiliation(s)
- Miguel Lopez de Rodas
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Cancer Center Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Maria Villalba-Esparza
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Miguel F Sanmamed
- Department of Immunology and Immunotherapy, Centro de Investigación Médica Aplicada and Clínica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - David L Rimm
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kurt A Schalper
- Department of Pathology and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
8
|
Bernatowicz K, Amat R, Prior O, Frigola J, Ligero M, Grussu F, Zatse C, Serna G, Nuciforo P, Toledo R, Escobar M, Garralda E, Felip E, Perez-Lopez R. Radiomics signature for dynamic monitoring of tumor inflamed microenvironment and immunotherapy response prediction. J Immunother Cancer 2025; 13:e009140. [PMID: 39800381 PMCID: PMC11749429 DOI: 10.1136/jitc-2024-009140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/14/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND The efficacy of immune checkpoint inhibitors (ICIs) depends on the tumor immune microenvironment (TIME), with a preference for a T cell-inflamed TIME. However, challenges in tissue-based assessments via biopsies have triggered the exploration of non-invasive alternatives, such as radiomics, to comprehensively evaluate TIME across diverse cancers. To address these challenges, we develop an ICI response signature by integrating radiomics with T cell-inflamed gene-expression profiles. METHODS We conducted a pan-cancer investigation into the utility of radiomics for TIME assessment, including 1360 tumors from 428 patients. Leveraging contrast-enhanced CT images, we characterized TIME through RNA gene expression analysis, using the T cell-inflamed gene expression signature. Subsequently, a pan-cancer CT-radiomic signature predicting inflamed TIME (CT-TIME) was developed and externally validated. Machine learning was employed to select robust radiomic features and predict inflamed TIME. The study also integrated independent cohorts with longitudinal CT images, baseline biopsies, and comprehensive immunohistochemistry panel evaluation to assess the pan-cancer biological associations, spatiotemporal landscape and clinical utility of the CT-TIME. RESULTS The CT-TIME signature, comprising four radiomic features linked to a T-cell inflamed microenvironment, demonstrated robust performance with AUCs (95% CI) of 0.85 (0.73 to 0.96) (training) and 0.78 (0.65 to 0.92) (external validation). CT-TIME scores exhibited positive correlations with CD3, CD8, and CD163 expression. Intrapatient analysis revealed considerable heterogeneity in TIME between tumors, which could not be assessed using biopsies. Evaluation of aggregated per-patient CT-TIME scores highlighted its promising clinical utility for dynamically assessing the immune microenvironment and predicting immunotherapy response across diverse scenarios in advanced cancer. Despite demonstrating progression disease at the first follow-up, patients within the inflamed status group, identified by CT-TIME, exhibited significantly prolonged progression-free survival (PFS), with some surpassing 5 months, suggesting a potential phenomenon of pseudoprogression. Cox models using aggregated CT-TIME scores from baseline images revealed a statistically significant reduction in the risk of PFS in the pan-cancer cohort (HR 0.62, 95% CI 0.44 to 0.88, p=0.007), and Kaplan-Meier analysis further confirmed substantial differences in PFS between patients with inflamed and uninflamed status (log-rank test p=0.009). CONCLUSIONS The signature holds promise for impacting clinical decision-making, pan-cancer patient stratification, and treatment outcomes in immune checkpoint therapies.
Collapse
Affiliation(s)
| | - Ramon Amat
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Olivia Prior
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Joan Frigola
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Marta Ligero
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | | | - Garazi Serna
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | | | | | - Elena Garralda
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Vall d'Hebron University Hospital, Barcelona, Spain
| | - Enriqueta Felip
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Vall d'Hebron University Hospital, Barcelona, Spain
| | - Raquel Perez-Lopez
- Vall d'Hebron Institute of Oncology, Barcelona, Spain
- Vall d'Hebron University Hospital, Barcelona, Spain
| |
Collapse
|
9
|
Wei X, Xing X, Yao W, Wang C, Xiao Y, Du X. First-line combination therapy of immunotherapy plus anti-angiogenic drug for thoracic SMARCA4-deficient undifferentiated tumors in AIDS: a case report and review of the literature. Front Immunol 2025; 15:1473578. [PMID: 39850892 PMCID: PMC11754271 DOI: 10.3389/fimmu.2024.1473578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/12/2024] [Indexed: 01/25/2025] Open
Abstract
Background Thoracic SMARCA4-deficient undifferentiated tumors (SMARCA4-UT) exhibit a notably aggressive phenotype, which is associated with poor patient survival outcomes. These tumors are generally resistant to conventional cytotoxic chemotherapy, thereby limiting the availability of effective treatment options. Case presentation We describe a 69-year-old AIDS patient who initially presented with a fused, enlarged lymph node on the right clavicle and mild, unexplained pain under the right axilla that worsened with severe coughing episodes. An initial chest CT scan revealed multiple nodular and mass shadows in the mediastinum and multiple nodules in both lungs, as well as a small amount of pericardial effusion. Additionally, serum biomarkers of lung cancer were abnormal as follows: carcinoembryonic antigen (CEA) at 13.74 ng/mL, cytokeratin 19 fragment (CYFRA21-1) at 6.82 ng/mL, neuron-specific enolase (NSE) at 25.49 ng/mL, and progastrin-releasing peptide precursor (ProGRP) at 89.35 pg/mL. Subsequent pathology confirmed SMARCA4-deficient undifferentiated tumors. Considering that the weak immune status and intermediate PD-L1 level, the patient was treated with a first-line combination therapy of immunotherapy and anti-angiogenic drug instead of chemo-immunotherapy. The patient responded well to immunotherapy combining anti-angiogenic drugs and achieved an overall survival for more than 22 months. Conclusion Our study presented a rare case of thoracic SMARCA4-deficient undifferentiated tumors and AIDS, suggesting that first-line immunotherapy plus anti-angiogenic drugs as a potential therapeutic option for SMARCA4-UT patients under specific conditions.
Collapse
Affiliation(s)
- Xiaoling Wei
- Department of Respiratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiangju Xing
- Department of Respiratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Yao
- Department of Respiratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Changzheng Wang
- Department of Respiratory Medicine, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yajie Xiao
- Department of Translational Medicine, Shenzhen Yucebio Technology Co., Ltd., Shenzhen, China
| | - Xianzhi Du
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
10
|
Fournier C, Mercey-Ressejac M, Derangère V, Al Kadi A, Rageot D, Charrat C, Leroy A, Vollaire J, Josserand V, Escudé M, Escaich S, Ghiringhelli F, Decaens T, Navarro FP, Jouvin-Marche E, Marche PN. Nanostructured lipid carriers based mRNA vaccine leads to a T cell-inflamed tumour microenvironment favourable for improving PD-1/PD-L1 blocking therapy and long-term immunity in a cold tumour model. EBioMedicine 2025; 112:105543. [PMID: 39793480 PMCID: PMC11774803 DOI: 10.1016/j.ebiom.2024.105543] [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: 09/04/2024] [Revised: 11/29/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND mRNA-based cancer vaccines show promise in triggering antitumour immune responses. To combine them with existing immunotherapies, the intratumoral immune microenvironment needs to be deeply characterised. Here, we test nanostructured lipid carriers (NLCs), the so-called Lipidots®, for delivering unmodified mRNA encoding Ovalbumin (OVA) antigen to elicit specific antitumour responses. METHODS We evaluated whether NLC OVA mRNA complexes activate dendritic cells (DCs) in vitro and identified the involved signalling pathways using specific inhibitors. We tested the anti-tumoral impact of Ova mRNA vaccine in B16-OVA and E.G7-OVA cold tumour-bearing C57Bl6 female mice as well as its synergy effect with an anti-PD-1 therapy by following the tumour growth and performing immunophenotyping of innate and adaptive immune cells. The intratumoral vaccine-related gene signature was assessed by RNA-sequencing. The immune memory response was assessed by re-challenging surviving treated mice with tumour cells. FINDINGS Our vaccine activates DCs in vitro through the TLR4/8 and ROS signalling pathways and induces specific T cell activation while exhibits significant preventive and therapeutic antitumour efficacy in vivo. A profound intratumoral remodelling of the innate and adaptive immunity in association with an increase in the gene expression of chemokines (Cxcl10, Cxcl11, Cxcl9) involved in CD8+ T cell attraction were observed in immunised mice. The combination of vaccine and anti-PD-1 therapy improves the rates of complete responses and memory immune responses compared to monotherapies. INTERPRETATION Lipidots® are effective platform for the development of vaccines against cancer based on mRNA delivery. Their combination with immune checkpoint blockers could counter tumour resistance and promote long-term antitumour immunity. FUNDING This work was supported by Inserm Transfert, la Région Auvergne Rhône Alpes, FINOVI, and the French Ministry of Higher Education, research and innovation (LipiVAC, COROL project, funding reference N° 2102992411).
Collapse
Affiliation(s)
- Carole Fournier
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France; Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France.
| | - Marion Mercey-Ressejac
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Valentin Derangère
- INSERM U1231, Equipe TIRECS, Dijon, 21000, France; Université de Bourgogne, Dijon, 21000, France; Centre de Lutte contre le Cancer Georges François Leclerc, Plateforme de Transfert en Biologie du Cancer, Dijon, 21000, France
| | - Amal Al Kadi
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - David Rageot
- INSERM U1231, Equipe TIRECS, Dijon, 21000, France; Université de Bourgogne, Dijon, 21000, France; Centre de Lutte contre le Cancer Georges François Leclerc, Plateforme de Transfert en Biologie du Cancer, Dijon, 21000, France
| | - Christine Charrat
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Alexis Leroy
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Julien Vollaire
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Véronique Josserand
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Marie Escudé
- CEA, LETI, Technologies for Healthcare and Biology Division, Microtechnologies for Living Systems Interactions Research Unit, Univ. Grenoble Alpes, Grenoble, F-38000, France
| | - Séverine Escaich
- CEA, LETI, Technologies for Healthcare and Biology Division, Microtechnologies for Living Systems Interactions Research Unit, Univ. Grenoble Alpes, Grenoble, F-38000, France
| | - François Ghiringhelli
- INSERM U1231, Equipe TIRECS, Dijon, 21000, France; Université de Bourgogne, Dijon, 21000, France; Centre de Lutte contre le Cancer Georges François Leclerc, Plateforme de Transfert en Biologie du Cancer, Dijon, 21000, France
| | - Thomas Decaens
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France; Hepato-Gastroenterology and Digestive Oncology Department, CHU Grenoble Alpes, Grenoble, France
| | - Fabrice P Navarro
- CEA, LETI, Technologies for Healthcare and Biology Division, Microtechnologies for Living Systems Interactions Research Unit, Univ. Grenoble Alpes, Grenoble, F-38000, France
| | - Evelyne Jouvin-Marche
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| | - Patrice N Marche
- Univ. Grenoble Alpes, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR, Grenoble, 5309, France
| |
Collapse
|
11
|
Xu Q, Hu J, Wang Y, Wang Z. The role of tumor types in immune-related adverse events. Clin Transl Oncol 2024:10.1007/s12094-024-03798-6. [PMID: 39738878 DOI: 10.1007/s12094-024-03798-6] [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: 08/02/2024] [Accepted: 11/13/2024] [Indexed: 01/02/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that block inhibitors of T cell activation and function. With the widespread use of ICIs in cancer therapy, immune-related adverse events (irAEs) have gradually emerged as urgent clinical issues. Tumors not only exhibit high heterogeneity, and their response to ICIs varies, with "hot" tumors showing better anti-tumor effects but also a higher susceptibility to irAEs. The manifestation of irAEs displays a tumor-heterogeneous pattern, correlating with the tumor type in terms of the affected organs, incidence, median onset time, and severity. Understanding the mechanisms underlying the pathogenic patterns of irAEs can provide novel insights into the prevention and management of irAEs, guide the development of biomarkers, and contribute to a deeper understanding of the toxicological characteristics of ICIs. In this review, we explore the impact of tumor type on the therapeutic efficacy of ICIs and further elucidate how these tumor types influence the occurrence of irAEs. Finally, we assess key candidate biomarkers and their relevance to proposed irAE mechanisms. This paper also outlines management strategies for patients with various types of tumors, based on their disease patterns.
Collapse
Affiliation(s)
- Qian Xu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Jing Hu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Yan Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
| | - Zhaohui Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
| |
Collapse
|
12
|
Shi R, Ran L, Tian Y, Guo W, Zhao L, Jin S, Cheng J, Zhang Z, Ma Y. Prospects and challenges of neoantigen applications in oncology. Int Immunopharmacol 2024; 143:113329. [PMID: 39405926 DOI: 10.1016/j.intimp.2024.113329] [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: 07/23/2024] [Revised: 09/11/2024] [Accepted: 10/02/2024] [Indexed: 10/30/2024]
Abstract
Neoantigen, unique peptides resulting from tumor-specific mutations, represent a promising frontier in oncology for personalized cancer immunotherapy. Their unique features allow for the development of highly specific and effective cancer treatments, which can potentially overcome the limitations of conventional therapies. This paper explores the current prospects and challenges associated with the application of neoantigens in oncology. We examine the latest advances in neoantigen identification, vaccine development, and adoptive T cell therapy. Additionally, we discuss the obstacles related to neoantigen heterogeneity, immunogenicity prediction, and the tumor microenvironment. Through a comprehensive analysis of current research and clinical trials, this paper aims to provide a detailed overview of how neoantigens could revolutionize cancer treatment and the hurdles that must be overcome to realize their full potential.
Collapse
Affiliation(s)
- Ranran Shi
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Ling Ran
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Yuan Tian
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Wei Guo
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China
| | - Lifang Zhao
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Shaoju Jin
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Henan Province Engineering & Technology Research Center of Foods for Special Medical Purpose, Luohe Medical College, Luohe 462000, China
| | - Jiang Cheng
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China; Department of Neurology, General Hospital of Ningxia Medical University, Yinchuan 750000, China
| | - Zhe Zhang
- School of Sciences, Henan University of Technology, Zhengzhou 450001, China.
| | - Yongchao Ma
- Department of Basic Medical Sciences, Luohe Medical College, Luohe 462000, China.
| |
Collapse
|
13
|
Kong S, Zhang J, Ding B, He C, Zhang X. Nanoplatform-based synergistic cancer Immuno-Chemodynamic therapy. Int J Pharm 2024; 667:124956. [PMID: 39550012 DOI: 10.1016/j.ijpharm.2024.124956] [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: 07/17/2024] [Revised: 10/22/2024] [Accepted: 11/12/2024] [Indexed: 11/18/2024]
Abstract
Immunotherapy has made excellent breakthroughs in the field of cancer treatments, but faces challenges with low immunogenicity of tumor cells and an immunosuppressive tumor microenvironment (ITME). The emerging chemodynamic therapy (CDT) based on the Fenton/Fenton-like reaction can induce immunogenic cell death (ICD) to enhance tumor immunogenicity, facilitating the transition from immune-cold to immune-hot tumors. Synergistic CDT and immunotherapy based on advanced nanotechnology have shown immense promise for improving therapeutic efficacy while minimizing side effects in cancer treatment. This review summarizes and discusses recent advances in the field, with the goal of designing a high-quality nanoplatform to enhance synergistic CDT in combination with immunotherapy and lay the foundation for its future clinical translation.
Collapse
Affiliation(s)
- Shuaizhi Kong
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, 310014, PR China; Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, PR China
| | - Jie Zhang
- Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, PR China
| | - Baoyue Ding
- Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, PR China.
| | - Chuanchuan He
- Jiaxing Maternity and Child Health Care Hospital, Affiliated Women and Children Hospital, Jiaxing University, Jiaxing, PR China.
| | - Xiaojuan Zhang
- Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, 314001, PR China.
| |
Collapse
|
14
|
Qu F, Wu S, Yu W. Progress of Immune Checkpoint Inhibitors Therapy for pMMR/MSS Metastatic Colorectal Cancer. Onco Targets Ther 2024; 17:1223-1253. [PMID: 39735789 PMCID: PMC11681808 DOI: 10.2147/ott.s500281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 12/31/2024] Open
Abstract
Immunotherapy is one of the research hotspots in colorectal cancer field in recent years. The colorectal cancer patients with mismatch repair-deficient (dMMR) or high microsatellite instability (MSI-H) are the primary beneficiaries of immunotherapy. However, the vast majority of colorectal cancers are mismatch repair proficient (pMMR) or microsatellite stability (MSS), and their immune microenvironment is characterized by "cold tumors" that are generally insensitive to single immunotherapy based on immune checkpoint inhibitors (ICIs). Studies have shown that some pMMR/MSS colorectal cancer patients regulate the immune microenvironment by combining other treatments, such as multi-target tyrosine kinase inhibitors, anti-vascular endothelial growth factor (VEGF) monoclonal antibodies, chemotherapy, radiotherapy, anti-epithelial growth factor receptor (EGFR) monoclonal antibodies, and mitogen-activated protein kinase (MAPK) signaling pathway inhibitors and oncolytic viruses, etc. to transform "cold tumor" into "hot tumor", thereby improving the response to immunotherapy. In addition, screening for potential prognostic biomarkers can also enrich the population benefiting from immunotherapy for microsatellite stable colorectal cancer. Therefore, in pMMR or MSS metastatic colorectal cancer (mCRC), the optimization of immunotherapy regimens and the search for effective efficacy prediction biomarkers are currently important research directions. In this paper, we review the progress of efficacy of immunotherapy (mainly ICIs) in pMMR /MSS mCRC, challenges and potential markers, in order to provide research ideas for the development of immunotherapy for mCRC.
Collapse
Affiliation(s)
- Fanjie Qu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - Shuang Wu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| | - WeiWei Yu
- Department of Oncology, Affiliated Dalian Third People’s Hospital of Dalian Medical University, Dalian, Liaoning Province, 116033, People’s Republic of China
| |
Collapse
|
15
|
Rogado J, Pozo F, Troule K, Pacheco M, Adrados M, Sánchez-Torres JM, Al-Shahrour F, Aspa J, Alfranca A, Romero-Laorden N, Colomer R. The role of the CXCL12/CXCR4 axis in the immunotherapy of non-small cell lung cancer. Clin Transl Oncol 2024:10.1007/s12094-024-03828-3. [PMID: 39708256 DOI: 10.1007/s12094-024-03828-3] [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: 08/29/2024] [Accepted: 12/09/2024] [Indexed: 12/23/2024]
Abstract
INTRODUCTION Peripheral blood mononuclear cells (PBMCs) trafficking is regulated by chemokines, which modulate leukocyte migration toward tumors and may collaborate in the efficacy of immunotherapy. In our study, we investigated whether the CXCL12/CXCR4 axis plays a role in the efficacy of immunotherapy in non-small cell lung cancer (NSCLC) by analyzing CXCR4 expression for CXCR4 in peripheral blood (PB), and the expression of its ligand CXCL12 in tumor. METHODS We identified PBMCs expressing CXCR4 using flow cytometry in a prospective cohort of NSCLC patients before starting anti-PD-1 immunotherapy. As a control, we studied patients with advanced cancer before starting any non-immunotherapy treatment. The relative frequency of PBMCs was correlated with treatment outcomes. Uni- and multivariate survival analyses were performed. The expression of CXCL12 in tumor tissue was studied and correlated with the expression of its receptor (CXCR4) in PBMCs. RESULTS The experimental group included 39 patients and the control group included 40. Low expression of CXCR4-expressing CD8 + T lymphocytes was correlated with a greater benefit from immunotherapy: median OS NR vs. 22.0 months, HR 0.6, p < 0.01; and median PFS 14.2 vs. 5.0 months, HR 0.38, p = 0.05. These differences were not observed in controls. Low expression in PB of these lymphocytes was correlated with a higher expression of CXCL12 in tumor (trend toward significance: p = 0.14). CONCLUSION Patients diagnosed with advanced NSCLC with low percentage of cytotoxic T lymphocytes expressing CXCR4 in PB, show greater benefit from immunotherapy, probably due to increased tumor infiltration by lymphocytes in response to CXCL12 produced by the tumor.
Collapse
Affiliation(s)
- Jacobo Rogado
- Medical Oncology Department, Hospital Universitario Infanta Leonor, Gran Vía del Este, 80, 28031, Madrid, Spain.
- Instituto de Investigación Sanitaria Princesa, Madrid, Spain.
| | - Fernando Pozo
- Bioinformatics Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Kevin Troule
- Bioinformatics Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - María Pacheco
- Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Medical Oncology Department, Hospital Universitario de La Princesa, Madrid, Spain
| | - Magdalena Adrados
- Pathology Department, Hospital Universitario de La Princesa, Madrid, Spain
| | - José Miguel Sánchez-Torres
- Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Medical Oncology Department, Hospital Universitario de La Princesa, Madrid, Spain
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Centro Nacional de Investigaciones Oncológicas, Madrid, Spain
| | - Javier Aspa
- Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Pneumonology Department, , Hospital Universitario de La Princesa, Madrid, Spain
| | - Arantzazu Alfranca
- Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Immunology Department, Hospital Universitario de La Princesa, Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
- Cátedra UAM-Fundación Instituto Roche de Medicina Personalizada de Precisión, Madrid, Spain
| | - Nuria Romero-Laorden
- Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Medical Oncology Department, Hospital Universitario de La Princesa, Madrid, Spain
- Cátedra UAM-Fundación Instituto Roche de Medicina Personalizada de Precisión, Madrid, Spain
| | - Ramon Colomer
- Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Medical Oncology Department, Hospital Universitario de La Princesa, Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
- Cátedra UAM-Fundación Instituto Roche de Medicina Personalizada de Precisión, Madrid, Spain
| |
Collapse
|
16
|
Zhu CY, Zhai TT, Su M, Pan HC, Tang Q, Huang BH, Chi XR, Li N, Xie LH, Qiu SQ, Pan F, Huang GW. EZH2 elicits CD8 + T-cell desert in esophageal squamous cell carcinoma via suppressing CXCL9 and dendritic cells. Commun Biol 2024; 7:1645. [PMID: 39702756 DOI: 10.1038/s42003-024-07341-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024] Open
Abstract
CD8+ T cell spatial distribution in the context of tumor microenvironment (TME) dictates the immunophenotypes of tumors, comprised of immune-infiltrated, immune-excluded and immune-desert, discriminating "hot" from "cold" tumors. The infiltration of cytotoxic CD8+ T cells is associated with favorable therapeutic response. Hitherto, the immunophenotypes of esophageal squamous cell carcinoma (ESCC) have not yet been comprehensively delineated. Herein, we comprehensively characterized the immunophenotypes of ESCC and identified a subset of ESCC, which was defined as cold tumor and characterized with CD8+ T cell-desert TME. However, the mechanism underlying the defect of CD8+ T cells in TME is still pending. Herein, we uncovered that tumor cell-intrinsic EZH2 with high expression was associated with the immunophenotype of immune-desert tumors. Targeted tumor cell-intrinsic EZH2 rewired the transcriptional activation of CXCL9 mediated by NF-κB and concomitantly reinvigorated DC maturation differentiation via inducing the reduction of VEGFC secretion, thereby enhancing the infiltration of cytotoxic CD8+ T cells into TME and inhibiting tumor immune evasion. Our findings identify EZH2 as a potential therapeutic target and point to avenues for targeted therapy applied to patients with ESCC characterized by CD8+ T cell-desert tumors.
Collapse
Affiliation(s)
- Chun-Yan Zhu
- Department of Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Tian-Tian Zhai
- Department of radiotherapy, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China.
| | - Meng Su
- Department of Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Hong-Chao Pan
- The Breast Center, Surgical Oncology Session No. 1, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong, 515041, China
| | - Qian Tang
- Department of Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Bao-Hua Huang
- Department of Pathology, Shantou Central Hospital, Shantou, Guangdong, 515041, China
| | - Xin-Rui Chi
- Department of Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Nuo Li
- Department of Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Ling-Hui Xie
- Department of Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Si-Qi Qiu
- Clinical research center, Shantou Central Hospital, Shantou, Guangdong, 515041, China.
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, 515041, Shantou, China.
| | - Feng Pan
- Clinical research center, Shantou Central Hospital, Shantou, Guangdong, 515041, China.
| | - Guo-Wei Huang
- Department of Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
17
|
Li Y, Wu Y, Gao S, Sun T, Jiang C. PROTAC delivery in tumor immunotherapy: Where are we and where are we going? J Control Release 2024; 378:116-144. [PMID: 39637991 DOI: 10.1016/j.jconrel.2024.11.076] [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: 10/18/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Immunotherapy has emerged as a pioneering therapeutic modality, particularly within the realm of oncology, where Chimeric Antigen Receptor T-cell (CAR-T) therapy has manifested significant efficacy in the treatment of hematological malignancies. Nonetheless, the extension of immunotherapy to solid tumors poses a considerable challenge. This challenge is largely attributed to the intrinsic "cold" characteristics of certain tumors, which are defined by scant T-cell infiltration and a diminished immune response. Additionally, the impediment is exacerbated by the elusive nature of numerous targets within the tumor microenvironment, notably those deemed "undruggable" by small molecule inhibitors. This scenario underscores an acute necessity for the inception of innovative therapeutic strategies aimed at countering the resistance mechanisms underlying immune evasion in cold tumors, thereby amplifying the efficacy of cancer immunotherapy. Among the promising strategies is the deployment of Proteolysis Targeting Chimeras (PROTACs), which facilitate the targeted degradation of proteins. PROTACs present unique advantages and have become indispensable in oncology. However, they concurrently grapple with challenges such as solubility issues, permeability barriers, and the classical Hook effect. Notably, advanced delivery systems have been instrumental in surmounting these obstacles. This review commences with an analysis of the factors contributing to the suboptimal responses to immunotherapy in cold tumors. Subsequently, it delivers a thorough synthesis of immunotherapeutic concepts tailored for these tumors, clarifying the integral role of PROTACs in their management and delineating the trajectory of PROTAC technology from bench-side investigation to clinical utilization, facilitated by drug delivery systems. Ultimately, the review extrapolates the prospective future of this approach, aspiring to present novel insights that could catalyze progress in immunotherapy for the treatment of cold tumors.
Collapse
Affiliation(s)
- Yiyang Li
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yike Wu
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Sihan Gao
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tao Sun
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; Quzhou Fudan Institute, Quzhou 324003, China.
| | - Chen Jiang
- Key Laboratory of Smart Drug DeliveryMinistry of Education, Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China; Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
18
|
Shi W, Xu W, Song L, Zeng Q, Qi G, Qin Y, Li Z, Liu X, Jiao Z, Zhao Y, Liu N, Lu H. A tumor-conditional IL-15 safely synergizes with immunotherapy to enhance antitumor immune responses. Mol Ther 2024; 32:4482-4496. [PMID: 39489922 PMCID: PMC11638872 DOI: 10.1016/j.ymthe.2024.10.021] [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: 08/03/2024] [Revised: 09/20/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
It is a challenge to invigorate tumor-infiltrating lymphocytes without causing immune-related adverse events, which also stands as a primary factor contributing to resistance against cancer immunotherapies. Interleukin (IL)-15 can potently promote expansion and activation of T cells, but its clinical use has been limited by dose-limiting toxicities. In this study, we develop a tumor-conditional IL-15 (pro-IL-15), which masks IL-15 with steric hindrance caused by Fc fragment and IL-15Rα-sushi domain. Upon reaching the tumor site, it can be cleaved by tumor-associated proteases to release an IL-15 superagonist, resulting in potent antitumor activities. Systemic delivery of pro-IL-15 demonstrates significantly reduced toxicity but uncompromised antitumor efficacy. Pro-IL-15 can yield better effectors and vitalize terminally exhausted CD8+ T cells to overcome checkpoint blockade resistance. Moreover, pro-IL-15 promotes chemotaxis and activation of adoptive T cells, leading to eradication of advanced solid tumors and durable cures. Furthermore, pro-IL-15 shows promise for synergizing with other immunotherapies like IL-12 and oncolytic virus by improving the CD8/Treg ratio and interferon-γ levels, resulting in substantial regression of both local and metastatic cold tumors. Collectively, our results suggest that pro-IL-15 represents a compelling strategy for overcoming resistance to current immunotherapies while avoiding toxicities.
Collapse
Affiliation(s)
- Wenqiang Shi
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; Shanghai Frontiers Science Center for Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Wei Xu
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Luyao Song
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; Shanghai Frontiers Science Center for Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Qiongya Zeng
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; Shanghai Frontiers Science Center for Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Gen Qi
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; Shanghai Frontiers Science Center for Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Ying Qin
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; Shanghai Frontiers Science Center for Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Zhikun Li
- Department of Orthopedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai 200336, China
| | - Xianglei Liu
- National Key Laboratory of Lead Druggability Research, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, China
| | - Zheng Jiao
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 Huaihai West Road, Shanghai 200030, China
| | - Yonggang Zhao
- Suzhou HKeyBio Company Ltd, 218 Xinghu Street, Suzhou 215004, China
| | - Nan Liu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.
| | - Huili Lu
- Engineering Research Center of Cell and Therapeutic Antibody, Ministry of Education, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; Shanghai Frontiers Science Center for Drug Target Identification and Delivery, National Key Laboratory of Innovative Immunotherapy, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China; Chongqing Research Institute, Shanghai Jiao Tong University, Chongqing 401135, China.
| |
Collapse
|
19
|
Zhang Z, Lu Y, Liu W, Huang Y. Nanomaterial-assisted delivery of CpG oligodeoxynucleotides for boosting cancer immunotherapy. J Control Release 2024; 376:184-199. [PMID: 39368710 DOI: 10.1016/j.jconrel.2024.09.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 08/03/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
Cancer immunotherapy aims to improve immunity to not only eliminate the primary tumor but also inhibit metastasis and recurrence. It is considered an extremely promising therapeutic approach that breaks free from the traditional paradigm of oncological treatment. As the medical community learns more about the immune system's mechanisms that "turn off the brake" and "step on the throttle", there is increasingly successful research on immunomodulators. However, there are still more restrictions than countermeasures with immunotherapy related to immunomodulators, such as low responsiveness and immune-related adverse events that cause multiple adverse reactions. Therefore, medical experts and materials scientists attempted to the efficacy of immunomodulatory treatments through various methods, especially nanomaterial-assisted strategies. CpG oligodeoxynucleotides (CpG) not only act as an adjuvant to promote immune responses, but also induce autophagy. In this review, the enhancement of immunotherapy using nanomaterial-based CpG formulations is systematically elaborated, with a focus on the delivery, protection, synergistic promotion of CpG efficacy by nanomaterials, and selection of the timing of treatment. In addition, we also discuss and prospect the existing problems and future directions of research on nanomaterials in auxiliary CpG therapy.
Collapse
Affiliation(s)
- Zhiyu Zhang
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Yu Lu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| | - Wenjing Liu
- Department of Pharmacology, Beijing Chest Hospital, Capital Medical University/Beijing Key Laboratory of Drug Resistance Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
20
|
Xie MZ, Li YQ, Liang R, Huang SY, Qin SY, Hu BL. Synergistic Effects of Fruquintinib Combined with Immune Checkpoint Inhibitors on Metastatic Colorectal Cancer. J Gastrointest Cancer 2024; 55:1620-1627. [PMID: 39316296 DOI: 10.1007/s12029-024-01108-5] [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: 08/17/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Fruquintinib has received approval for the management of patients with chemotherapy-resistant metastatic colorectal cancer (mCRC). However, combination of fruquintinib with immune checkpoint inhibitors (ICIs) is yet to be extensively studied. This study aims to assess the clinical efficacy, safety, and prognostic indicators of treatment regimen combining fruquintinib with ICIs in mCRC patients. METHODS We analyzed data from mCRC patients who were administered fruquintinib either as a monotherapy or in conjunction with ICIs following conventional chemotherapy. Parameters such as the objective response rate (ORR), disease control rate (DCR), overall survival (OS), progression-free survival (PFS), and incidence of adverse events were meticulously evaluated. Furthermore, the relationship between blood markers and patient prognosis was examined. RESULTS A total of 72 mCRC patients were included in this study, with a median observation period of 48 months, 19 were treated with fruquintinib alone, while 53 received a combination therapy involving fruquintinib and ICIs. The combined therapy group exhibited superior ORR and DCR compared to the fruquintinib monotherapy group. Additionally, significant improvements in OS and PFS were observed in the combined treatment group. The occurrence of adverse events was generally manageable and well-tolerated across both groups, with no significant difference in incidence rates. Notably, albumin levels were identified as a prognostic marker for PFS and OS in the univariate Cox regression analysis. CONCLUSIONS The combination of fruquintinib with ICIs demonstrated enhanced clinical efficacy and improved survival outcomes compared to fruquintinib monotherapy in mCRC patients. The safety of the combination regimen was deemed manageable and acceptable.
Collapse
Affiliation(s)
- Ming-Zhi Xie
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Yong-Qiang Li
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Rong Liang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Shi-Ying Huang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, No. 71 Hedi Road, Nanning, 530021, Guangxi, China
| | - Shan-Yu Qin
- Department of Gastroenterology, First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| | - Bang-Li Hu
- Department of Research, Guangxi Medical University Cancer Hospital, No.71 Hedi Road, Nanning, 530021, Guangxi, China
| |
Collapse
|
21
|
Eto S, Kato D, Saeki K, Iguchi T, Shiyu Q, Kamoto S, Yoshitake R, Shinada M, Ikeda N, Tsuboi M, Chambers J, Uchida K, Nishimura R, Nakagawa T. Comprehensive Analysis of the Tumour Immune Microenvironment in Canine Urothelial Carcinoma Reveals Immunosuppressive Mechanisms Induced by the COX-Prostanoid Cascade. Vet Comp Oncol 2024; 22:500-512. [PMID: 39179510 DOI: 10.1111/vco.12999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/04/2024] [Accepted: 07/13/2024] [Indexed: 08/26/2024]
Abstract
A comprehensive understanding of the tumour immune microenvironment (TIME) is essential for advancing precision medicine and identifying potential therapeutic targets. This study focused on canine urothelial carcinoma (cUC) recognised for its high sensitivity to cyclooxygenase (COX) inhibitors. Using immunohistochemical techniques, we quantified the infiltration of seven immune cell populations within cUC tumour tissue to identify clinicopathological features that characterise the TIME in cUC. Our results revealed several notable factors, including the significantly higher levels of CD3+ T cells and CD8+ T cells within tumour cell nests in cases treated with preoperative COX inhibitors compared to untreated cases. Based on the immunohistochemistry data, we further performed a comparative analysis using publicly available RNA-seq data from untreated cUC tissues (n = 29) and normal bladder tissues (n = 4) to explore the link between COX-prostanoid pathways and the immune response to tumours. We observed increased expression of COX-2, microsomal prostaglandin E2 synthase-1 (mPGES-1) and mPGES-2 in cUC tissues. However, only mPGES-2 showed a negative correlation with the cytotoxic T-cell (CTL)-related genes CD8A and granzyme B (GZMB). In addition, a broader analysis of solid tumours using The Cancer Genome Atlas (TCGA) database revealed similar patterns in several human tumours, suggesting a common mechanism in dogs and humans. Our results suggest that the COX-2/mPGES-2 pathway may act as a cross-species tumour-intrinsic factor that weakens anti-tumour immunity, and that COX inhibitors may convert TIME from a 'cold tumour' to a 'hot tumour' state by counteracting COX/mPGES-2-mediated immunosuppression.
Collapse
Affiliation(s)
- Shotaro Eto
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Daiki Kato
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kohei Saeki
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
- Department of Veterinary Medicine, Okayama University of Science, Imabari, Ehime, Japan
| | - Takaaki Iguchi
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Qin Shiyu
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Satoshi Kamoto
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryohei Yoshitake
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Masahiro Shinada
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Namiko Ikeda
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Masaya Tsuboi
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - James Chambers
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki Uchida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryohei Nishimura
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takayuki Nakagawa
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
22
|
Hamza FN, Mohammad KS. Immunotherapy in the Battle Against Bone Metastases: Mechanisms and Emerging Treatments. Pharmaceuticals (Basel) 2024; 17:1591. [PMID: 39770433 PMCID: PMC11679356 DOI: 10.3390/ph17121591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/20/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025] Open
Abstract
Bone metastases are a prevalent complication in advanced cancers, particularly in breast, prostate, and lung cancers, and are associated with severe skeletal-related events (SREs), including fractures, spinal cord compression, and debilitating pain. Conventional bone-targeted treatments like bisphosphonates and RANKL inhibitors (denosumab) reduce osteoclast-mediated bone resorption but do not directly impact tumor progression within the bone. This review focuses on examining the growing potential of immunotherapy in targeting the unique challenges posed by bone metastases. Even though immune checkpoint inhibitors (ICIs) have significantly changed cancer treatment, their impact on bone metastases appears limited because of the bone microenvironment's immunosuppressive traits, which include high levels of transforming growth factor-beta (TGFβ) and the immune-suppressing cells, such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). This review underscores the investigation of combined therapeutic approaches that might ease these difficulties, such as the synergy of immune checkpoint inhibitors with agents aimed at bones (denosumab, bisphosphonates), chemotherapy, and radiotherapy, as well as the combination of immune checkpoint inhibitors with different immunotherapeutic methods, including CAR T-cell therapy. This review provides a comprehensive analysis of preclinical studies and clinical trials that show the synergistic potential of these combination approaches, which aim to both enhance immune responses and mitigate bone destruction. By offering an in-depth exploration of how these strategies can be tailored to the bone microenvironment, this review underscores the need for personalized treatment approaches. The findings emphasize the urgent need for further research into overcoming immune evasion in bone metastases, with the goal of improving patient survival and quality of life.
Collapse
Affiliation(s)
- Fatheia N. Hamza
- Department of Biochemistry, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Khalid Said Mohammad
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| |
Collapse
|
23
|
John M, Helal M, Duell J, Mattavelli G, Stanojkovska E, Afrin N, Leipold AM, Steinhardt MJ, Zhou X, Žihala D, Anilkumar Sithara A, Mersi J, Waldschmidt JM, Riedhammer C, Kadel SK, Truger M, Werner RA, Haferlach C, Einsele H, Kretzschmar K, Jelínek T, Rosenwald A, Kortüm KM, Riedel A, Rasche L. Spatial transcriptomics reveals profound subclonal heterogeneity and T-cell dysfunction in extramedullary myeloma. Blood 2024; 144:2121-2135. [PMID: 39172759 DOI: 10.1182/blood.2024024590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/24/2024] Open
Abstract
ABSTRACT Extramedullary disease (EMD) is a high-risk feature of multiple myeloma (MM) and remains a poor prognostic factor, even in the era of novel immunotherapies. Here, we applied spatial transcriptomics (RNA tomography for spatially resolved transcriptomics [tomo-seq] [n = 2] and 10x Visium [n = 12]) and single-cell RNA sequencing (n = 3) to a set of 14 EMD biopsies to dissect the 3-dimensional architecture of tumor cells and their microenvironment. Overall, infiltrating immune and stromal cells showed both intrapatient and interpatient variations, with no uniform distribution over the lesion. We observed substantial heterogeneity at the copy number level within plasma cells, including the emergence of new subclones in circumscribed areas of the tumor, which is consistent with genomic instability. We further identified the spatial expression differences between GPRC5D and TNFRSF17, 2 important antigens for bispecific antibody therapy. EMD masses were infiltrated by various immune cells, including T cells. Notably, exhausted TIM3+/PD-1+ T cells diffusely colocalized with MM cells, whereas functional and activated CD8+ T cells showed a focal infiltration pattern along with M1 macrophages in tumor-free regions. This segregation of fit and exhausted T cells was resolved in the case of response to T-cell-engaging bispecific antibodies. MM and microenvironment cells were embedded in a complex network that influenced immune activation and angiogenesis, and oxidative phosphorylation represented the major metabolic program within EMD lesions. In summary, spatial transcriptomics has revealed a multicellular ecosystem in EMD with checkpoint inhibition and dual targeting as potential new therapeutic avenues.
Collapse
Affiliation(s)
- Mara John
- Mildred Scheel Early Career Center, University Hospital Würzburg, Würzburg, Germany
| | - Moutaz Helal
- Mildred Scheel Early Career Center, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Duell
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Greta Mattavelli
- Mildred Scheel Early Career Center, University Hospital Würzburg, Würzburg, Germany
| | - Emilia Stanojkovska
- Mildred Scheel Early Career Center, University Hospital Würzburg, Würzburg, Germany
| | - Nazia Afrin
- Mildred Scheel Early Career Center, University Hospital Würzburg, Würzburg, Germany
| | - Alexander M Leipold
- Helmholtz Institute for RNA-based Infection Research, Helmholtz-Center for Infection Research, Würzburg, Germany
| | | | - Xiang Zhou
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - David Žihala
- Department of Hematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Anjana Anilkumar Sithara
- Department of Hematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Julia Mersi
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | - Christine Riedhammer
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Sofie-Katrin Kadel
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | | | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, University Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Hermann Einsele
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Kai Kretzschmar
- Mildred Scheel Early Career Center, University Hospital Würzburg, Würzburg, Germany
| | - Tomáš Jelínek
- Department of Hematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Hematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | | | - K Martin Kortüm
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Angela Riedel
- Mildred Scheel Early Career Center, University Hospital Würzburg, Würzburg, Germany
| | - Leo Rasche
- Mildred Scheel Early Career Center, University Hospital Würzburg, Würzburg, Germany
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
24
|
Xiong X, Liu Y, Pu D, Yang Z, Bi Z, Tian L, Li X. DeSide: A unified deep learning approach for cellular deconvolution of tumor microenvironment. Proc Natl Acad Sci U S A 2024; 121:e2407096121. [PMID: 39514318 PMCID: PMC11573681 DOI: 10.1073/pnas.2407096121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Cellular deconvolution via bulk RNA sequencing (RNA-seq) presents a cost-effective and efficient alternative to experimental methods such as flow cytometry and single-cell RNA-seq (scRNA-seq) for analyzing the complex cellular composition of tumor microenvironments. Despite challenges due to heterogeneity within and among tumors, our innovative deep learning-based approach, DeSide, shows exceptional accuracy in estimating the proportions of 16 distinct cell types and subtypes within solid tumors. DeSide integrates biological pathways and assesses noncancerous cell types first, effectively sidestepping the issue of highly variable gene expression profiles (GEPs) associated with cancer cells. By leveraging scRNA-seq data from six cancer types and 185 cancer cell lines across 22 cancer types as references, our method introduces distinctive sampling and filtering techniques to generate a high-quality training set that closely replicates real tumor GEPs, based on The Cancer Genome Atlas (TCGA) bulk RNA-seq data. With this model and high-quality training set, DeSide outperforms existing methods in estimating tumor purity and the proportions of noncancerous cells within solid tumors. Our model precisely predicts cellular compositions across 19 cancer types from TCGA and proves its effectiveness with multiple additional external datasets. Crucially, DeSide enables the identification and analysis of combinatorial cell type pairs, facilitating the stratification of cancer patients into prognostically significant groups. This approach not only provides deeper insights into the dynamics of tumor biology but also highlights potential therapeutic targets by underscoring the importance of specific cell type or subtype interactions.
Collapse
Affiliation(s)
- Xin Xiong
- Department of Physics, Hong Kong Baptist University, Hong Kong, China
| | - Yerong Liu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Dandan Pu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Zhu Yang
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Zedong Bi
- Lingang Laboratory, Shanghai 200031, China
| | - Liang Tian
- Department of Physics, Hong Kong Baptist University, Hong Kong, China
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
- Institute of Computational and Theoretical Studies, Hong Kong Baptist University, Hong Kong, China
- Institute of Systems Medicine and Health Sciences, Hong Kong Baptist University, Hong Kong, China
| | - Xuefei Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
25
|
Fan Y, Chiu A, Zhao F, George JT. Understanding the interplay between extracellular matrix topology and tumor-immune interactions: Challenges and opportunities. Oncotarget 2024; 15:768-781. [PMID: 39513932 PMCID: PMC11546212 DOI: 10.18632/oncotarget.28666] [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: 07/12/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024] Open
Abstract
Modern cancer management comprises a variety of treatment strategies. Immunotherapy, while successful at treating many cancer subtypes, is often hindered by tumor immune evasion and T cell exhaustion as a result of an immunosuppressive tumor microenvironment (TME). In solid malignancies, the extracellular matrix (ECM) embedded within the TME plays a central role in T cell recognition and cancer growth by providing structural support and regulating cell behavior. Relative to healthy tissues, tumor associated ECM signatures include increased fiber density and alignment. These and other differentiating features contributed to variation in clinically observed tumor-specific ECM configurations, collectively referred to as Tumor-Associated Collagen Signatures (TACS) 1-3. TACS is associated with disease progression and immune evasion. This review explores our current understanding of how ECM geometry influences the behaviors of both immune cells and tumor cells, which in turn impacts treatment efficacy and cancer evolutionary progression. We discuss the effects of ECM remodeling on cancer cells and T cell behavior and review recent in silico models of cancer-immune interactions.
Collapse
Affiliation(s)
- Yijia Fan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
- Translational Medical Sciences, Texas A&M University Health Science Center, Houston, TX 77030, USA
| | - Alvis Chiu
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Feng Zhao
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Jason T. George
- Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
- Translational Medical Sciences, Texas A&M University Health Science Center, Houston, TX 77030, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
- Department of Hematopoietic Biology and Malignancy, MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
26
|
Ping W, Zhang X, Zeng H, Zhu T, Zhang N, Yan Q. Ultrasound-Driven Nanomachine for Enhanced Sonodynamic Therapy of Non-Small-Cell Lung Cancer. ACS APPLIED MATERIALS & INTERFACES 2024; 16:59803-59813. [PMID: 39437325 DOI: 10.1021/acsami.4c11546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Non-small-cell lung cancer (NSCLC) is the most prevalent type of lung cancer, and there is an urgent need for developing novel therapies. Sonodynamic therapy exhibits exceptional tissue penetration and minimal harm to healthy tissue, making it extremely promising for cancer treatment. The efficacy of SDT is limited by the intricate immunological microenvironment and the resistance to tumor treatment. This study developed targeted nanoparticles that use ultrasound to concentrate on treating NSCLC. The hybrid targeted nanoparticles utilize gold nanoparticles as their fundamental component, with the outside modified with engineered macrophage exosomes and the aptamer S11e to specifically target NSCLC. Ultrasound could effectively eliminate tumors in NSCLC cells by destroying lysosomes via targeted nanoparticles. Simultaneously, fragmented tumor antigens could effectively activate dendritic cell cells to recruit T cells. This method has significant efficacy in suppressing the development of NSCLC and exhibits potential for therapeutic application.
Collapse
Affiliation(s)
- Wei Ping
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoxin Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Hao Zeng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Taomin Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Ni Zhang
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Yan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| |
Collapse
|
27
|
Liu YT, Chen L, Li SJ, Wang WY, Wang YY, Yang QC, Song A, Zhang MJ, Mo WT, Li H, Hu CY, Sun ZJ. Dysregulated Wnt/β-catenin signaling confers resistance to cuproptosis in cancer cells. Cell Death Differ 2024; 31:1452-1466. [PMID: 38987382 PMCID: PMC11520902 DOI: 10.1038/s41418-024-01341-2] [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: 11/21/2023] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024] Open
Abstract
Cuproptosis is characterized by the aggregation of lipoylated enzymes of the tricarboxylic acid cycle and subsequent loss of iron-sulfur cluster proteins as a unique copper-dependent form of regulated cell death. As dysregulation of copper homeostasis can induce cuproptosis, there is emerging interest in exploiting cuproptosis for cancer therapy. However, the molecular drivers of cancer cell evasion of cuproptosis were previously undefined. Here, we found that cuproptosis activates the Wnt/β-catenin pathway. Mechanistically, copper binds PDK1 and promotes its interaction with AKT, resulting in activation of the Wnt/β-catenin pathway and cancer stem cell (CSC) properties. Notably, aberrant activation of Wnt/β-catenin signaling conferred resistance of CSCs to cuproptosis. Further studies showed the β-catenin/TCF4 transcriptional complex directly binds the ATP7B promoter, inducing its expression. ATP7B effluxes copper ions, reducing intracellular copper and inhibiting cuproptosis. Knockdown of TCF4 or pharmacological Wnt/β-catenin blockade increased the sensitivity of CSCs to elesclomol-Cu-induced cuproptosis. These findings reveal a link between copper homeostasis regulated by the Wnt/β-catenin pathway and cuproptosis sensitivity, and suggest a precision medicine strategy for cancer treatment through selective cuproptosis induction.
Collapse
Affiliation(s)
- Yuan-Tong Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Lei Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Shu-Jin Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Wu-Yin Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Yuan-Yuan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Qi-Chao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - An Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Meng-Jie Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Wen-Tao Mo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Hao Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Chuan-Yu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
28
|
Bitar R, Salem R, Finn R, Greten TF, Goldberg SN, Chapiro J, Atzen S. Interventional Oncology Meets Immuno-oncology: Combination Therapies for Hepatocellular Carcinoma. Radiology 2024; 313:e232875. [PMID: 39560477 PMCID: PMC11605110 DOI: 10.1148/radiol.232875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 11/20/2024]
Abstract
The management of hepatocellular carcinoma (HCC) is undergoing transformational changes due to the emergence of various novel immunotherapies and their combination with image-guided locoregional therapies. In this setting, immunotherapy is expected to become one of the standards of care in both neoadjuvant and adjuvant settings across all disease stages of HCC. Currently, more than 50 ongoing prospective clinical trials are investigating various end points for the combination of immunotherapy with both percutaneous and catheter-directed therapies. This review will outline essential tumor microenvironment mechanisms responsible for disease evolution and therapy resistance, discuss the rationale for combining locoregional therapy with immunotherapy, summarize ongoing clinical trials, and report on developing imaging end points and novel biomarkers that are relevant to both diagnostic and interventional radiologists participating in the management of HCC.
Collapse
Affiliation(s)
- Ryan Bitar
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Riad Salem
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Richard Finn
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Tim F. Greten
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - S. Nahum Goldberg
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Julius Chapiro
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| | - Sarah Atzen
- From the Departments of Radiology (R.B., J.C.) and Digestive Diseases
(Hepatology) (J.C.), Yale University School of Medicine, New Haven, Conn;
Department of Radiology, Feinberg School of Medicine, Northwestern University,
Chicago, Ill (R.S.); Department of Medical Oncology, Geffen School of Medicine,
University of California Los Angeles, Los Angeles, Calif (R.F.); Center for
Cancer Research, National Institutes of Health, Bethesda, Md (T.F.G.);
Department of Radiology, Hadassah Hebrew University Medical Center, Hebrew
University, Jerusalem, Israel (S.N.G.); and Department of Biomedical
Engineering, Yale School of Engineering and Applied Sciences, 789 Howard Ave,
Clinic Bldg 363H, New Haven, CT 06520 (J.C.)
| |
Collapse
|
29
|
Zheng J, Zheng Z, Zhang T, Chen X, Pang Q, Wang P, Yan C, Zhang W. Optimization of radiation target volume for locally advanced esophageal cancer in the immunotherapy era. Expert Opin Biol Ther 2024; 24:1221-1232. [PMID: 39460561 DOI: 10.1080/14712598.2024.2423009] [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: 06/29/2024] [Revised: 10/19/2024] [Accepted: 10/25/2024] [Indexed: 10/28/2024]
Abstract
INTRODUCTION Locally advanced esophageal cancer (EC) has poor prognosis. Preliminary clinical studies have demonstrated the synergistic efficacy of radiotherapy combined with immunotherapy in EC. Adjusting the radiotherapy target volume to protect immune function favors immunotherapy. However, there is no clear consensus on the exact definition of the EC target volume. AREAS COVERED Preclinical studies have provided a wealth of information on immunotherapy combined with different radiotherapy modalities, and several clinical studies have evaluated the impact of immunotherapy combined with radiotherapy on locally advanced EC. Here, we illustrate the rational target volume delineation for radiotherapy in terms of patient prognosis, pattern of radiotherapy failure, treatment-related toxicities, tumor-draining lymph nodes, and systemic immunity and summarize the clinical trials of radiotherapy combined with immunotherapy in EC. EXPERT OPINION We recommend applying involved-field irradiation (IFI) instead of elective nodal irradiation (ENI) for irradiated fields when immunotherapy is combined with chemoradiotherapy (CRT) for locally advanced EC. We expect that this target design will be evaluated in clinical trials to further explore more precise diagnostic modalities, long-term toxic responses, and quality of survival, and stratification factors for personalized treatment, and to provide more treatment benefits for patients.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhunhao Zheng
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Tian Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xi Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Qingsong Pang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ping Wang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wencheng Zhang
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
30
|
Yan T, Zhou W, Li C. Discovery of a T cell proliferation-associated regulator signature correlates with prognosis risk and immunotherapy response in bladder cancer. Int Urol Nephrol 2024; 56:3447-3462. [PMID: 38789872 DOI: 10.1007/s11255-024-04086-6] [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: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND The efficacy of immunotherapy is heavily influenced by T cell activity. This study aimed to examine how T cell proliferation regulators can predict the prognosis and response to immunotherapy in patients with bladder cancer (BCa). METHODS T cell proliferation-related subtypes were determined by employing the non-negative matrix factorization (NMF) algorithm that analyzed the expression patterns of T cell proliferation regulators. Subtypes were assessed for variations in prognosis, immune infiltration, and functional behaviors. Subsequently, a risk model related to T cell proliferation was created through Cox and Lasso regression analyses in the TCGA cohort and then confirmed in two GEO cohorts and an immunotherapy cohort. RESULTS BCa patients were categorized into two subtypes (C1 and C2) according to the expression profiles of 31 T cell proliferation-related genes (TRGs) with distinct prognoses and immune landscapes. The C2 subtype had a shorter overall survival (OS), with higher levels of M2 macrophage infiltration, and the activation of cancer-related pathways than the C1 subtype. Following this, thirteen prognosis-related genes that were involved in T cell proliferation were utilized to create the prognostic signature. The model's predictive accuracy was confirmed by analyzing both internal and external datasets. Individuals in the high-risk category experienced a poorer prognosis, increased immunosuppressive factors in the tumor microenvironment, and diminished responses to immunotherapy. Additionally, the immunotherapeutic prediction efficacy of the model was further confirmed by an immunotherapy cohort (anti-PD-L1 in the IMvigor210 cohort). CONCLUSIONS Our study characterized two subtypes linked to T cell proliferation in BCa patients with distinct prognoses and tumor microenvironment (TME) patterns, providing new insights into the heterogeneity of T cell proliferation in BCa and its connection to the immune landscape. The signature has prospective clinical implications for predicting outcomes and may help physicians to select prospective responders who prioritize current immunotherapy.
Collapse
Affiliation(s)
- Ting Yan
- Department of Blood Purification Center, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, People's Republic of China
| | - Wei Zhou
- Department of Urology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, People's Republic of China
| | - Chun Li
- Department of Blood Purification Center, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, No.141, Tianjin Road, Huangshi, 435000, Hubei, People's Republic of China.
| |
Collapse
|
31
|
Fan H, Liang X, Tang Y. Neuroscience in peripheral cancers: tumors hijacking nerves and neuroimmune crosstalk. MedComm (Beijing) 2024; 5:e784. [PMID: 39492832 PMCID: PMC11527832 DOI: 10.1002/mco2.784] [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: 05/21/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Cancer neuroscience is an emerging field that investigates the intricate relationship between the nervous system and cancer, gaining increasing recognition for its importance. The central nervous system governs the development of the nervous system and directly affects brain tumors, and the peripheral nervous system (PNS) shapes the tumor microenvironment (TME) of peripheral tumors. Both systems are crucial in cancer initiation and progression, with recent studies revealing a more intricate role of the PNS within the TME. Tumors not only invade nerves but also persuade them through remodeling to further promote malignancy, creating a bidirectional interaction between nerves and cancers. Notably, immune cells also contribute to this communication, forming a triangular relationship that influences protumor inflammation and the effectiveness of immunotherapy. This review delves into the intricate mechanisms connecting the PNS and tumors, focusing on how various immune cell types influence nerve‒tumor interactions, emphasizing the clinical relevance of nerve‒tumor and nerve‒immune dynamics. By deepening our understanding of the interplay between nerves, cancer, and immune cells, this review has the potential to reshape tumor biology insights, inspire innovative therapies, and improve clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Hua‐Yang Fan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Xin‐Hua Liang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial SurgeryWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ya‐Ling Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Oral PathologyWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
32
|
Yin C, Xing Y, Zhao P, Yin Y, Yao H, Xue J, Gu W. Tetradecanol-wrapped, CpG-loaded porous Prussian blue nanoimmunomodulator for photothermal-responsive in situ anti-tumor vaccine-like immunotherapy. BIOMATERIALS ADVANCES 2024; 164:213996. [PMID: 39146604 DOI: 10.1016/j.bioadv.2024.213996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
Therapeutic vaccine becomes a promising strategy to fight cancer by enhancing and sustaining specific anti-tumor immune responses. However, its efficacy is often impeded by low immunogenicity, the immunosuppressive tumor microenvironment (TME), and immune-related adverse events. Herein, we introduce 1-tetradecanol (TD)-wrapped, CpG-loaded porous Prussian blue nanoparticles (pPBNPs-CpG@TD) as a nanoimmunomodulator to initiate photothermal-induced immunogenic cell death (ICD) and photothermal-responsive release of CpG for augmenting the ICD effect. It was revealed that the dual-photothermal action significantly potentiated the in situ anti-tumor vaccine-like immunotherapy in terms of enhanced immunogenicity, promoted dendritic cell maturation, and increased T lymphocyte infiltration, consequently eliciting a robust immune response for inhibiting both primary and rechallenge tumors on a subcutaneous 4T1 tumor-bearing mouse model. The development and use of photoactive nanoimmunomodulators represents a novel and effective strategy to boost immunogenicity and counteract immunosuppressive TME, marking a significant advancement in the realm of ICD-driven in situ anti-tumor vaccine-like immunotherapy.
Collapse
Affiliation(s)
- Chenlu Yin
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Yixin Xing
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Peng Zhao
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Yuying Yin
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Hanye Yao
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Jingqiang Xue
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China
| | - Wei Gu
- School of pharmaceutical sciences, Capital medical university, Beijing 100069, PR China.
| |
Collapse
|
33
|
Hu C, Jiang Y, Chen Y, Wang Y, Wu Z, Zhang Q, Wu M. Low-Intensity Focused Ultrasound-Responsive Phase-Transitional Liposomes Loaded with STING Agonist Enhances Immune Activation for Breast Cancer Immunotherapy. Cancers (Basel) 2024; 16:3657. [PMID: 39518096 PMCID: PMC11545222 DOI: 10.3390/cancers16213657] [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: 08/22/2024] [Revised: 10/26/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Pharmacologically targeting the STING pathway offers a novel approach to cancer immunotherapy. However, small-molecule STING agonists face challenges such as poor tumor accumulation, rapid clearance, and short-lived effects within the tumor microenvironment, thus limiting their therapeutic potential. To address the challenges of poor specificity and inadequate targeting of STING in breast cancer treatment, herein, we report the design and development of a targeted liposomal delivery system modified with the tumor-targeting peptide iRGD (iRGD-STING-PFP@liposomes). With LIFU irradiation, the liposomal system exploits acoustic cavitation, where gas nuclei form and collapse within the hydrophobic region of the liposome lipid bilayer (transient pore formation), which leads to significantly enhanced drug release. Methods: Transmission electron microscopy (TEM) was used to investigate the physicochemical properties of the targeted liposomes. Encapsulation efficiency and in vitro release were assessed using the dialysis bag method, while the effects of iRGD on liposome targeting were evaluated through laser confocal microscopy. The CCK-8 assay was used to investigate the toxicity and cell growth effects of this system on 4T1 breast cancer cells and HUVEC vascular endothelial cells. A subcutaneous breast cancer tumor model was established to evaluate the tumor-killing effects and therapeutic mechanism of the newly developed liposomes. Results: The liposome carrier exhibited a regular morphology, with a particle size of 232.16 ± 19.82 nm, as indicated by dynamic light scattering (DLS), and demonstrated low toxicity to both HUVEC and 4T1 cells. With an encapsulation efficiency of 41.82 ± 5.67%, the carrier exhibited a slow release pattern in vitro after STING loading. Targeting results indicated that iRGD modification enhanced the system's ability to target 4T1 cells. The iRGD-STING-PFP@liposomes group demonstrated significant tumor growth inhibition in the subcutaneous breast cancer mouse model with effective activation of the immune system, resulting in the highest populations of matured dendritic cells (71.2 ± 5.4%), increased presentation of tumor-related antigens, promoted CD8+ T cell infiltration at the tumor site, and enhanced NK cell activity. Conclusions: The iRGD-STING-PFP@liposomes targeted drug delivery system effectively targets breast cancer cells, providing a new strategy for breast cancer immunotherapy. These findings indicate that iRGD-STING-PFP@liposomes could successfully deliver STING agonists to tumor tissue, trigger the innate immune response, and may serve as a potential platform for targeted immunotherapy.
Collapse
Affiliation(s)
- Cong Hu
- Department of Radiation Oncology, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China;
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Yuancheng Jiang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Yixin Chen
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Ying Wang
- Teaching Office, Zhongnan Hospital of Wuhan University, Wuhan 430071, China;
| | - Ziling Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Qi Zhang
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| | - Meng Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; (Y.J.); (Y.C.); (Z.W.); (Q.Z.)
| |
Collapse
|
34
|
Zhang C, Wu Q, Yang H, Zhang H, Liu C, Yang B, Hu Q. Ferroptosis-related gene signature for predicting prognosis and identifying potential therapeutic drug in EGFR wild-type lung adenocarcinoma. Commun Biol 2024; 7:1416. [PMID: 39478024 PMCID: PMC11525656 DOI: 10.1038/s42003-024-07117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Epidermal growth factor receptor wild type lung adenocarcinoma (EGFRWT LUAD) still has limited treatment options and unsatisfactory clinical outcomes. Ferroptosis, as a form of cell death, has been reported to play a dual role in regulating tumor cell survival. In this study, we constructed a 3-ferroptosis-gene signature, FeSig, and verified its accuracy and efficacy in predicting EGFRWT LUAD prognosis at both the RNA and protein levels. Patients with higher FeSig scores were found to have worse clinical outcomes. Additionally, we explored the relationship between FeSig and tumor microenvironment, revealing that enhanced interactions between fibroblasts and tumor cells in FeSighigh patients causing tumor resistance to ferroptosis. To address this challenge, we screened potential drugs from NCI-60 (The US National Cancer Institute 60 human tumour cell line anticancer drug screen) and Connectivity map database, ultimately identifying 6-mercatopurine (6-MP) as a promising candidate. Both in vitro and in vivo experiments demonstrated its efficacy in treating FeSighigh EGFRWT LUAD tumor models. In summary, we develop a novel FeSig for predicting prognosis and guiding drug application.
Collapse
Affiliation(s)
- Chuankai Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qi Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hongwei Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hui Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Changqing Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bo Yang
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qingsong Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
| |
Collapse
|
35
|
Xu Z, Wu Y, Hu J, Mei Z, Zhao Y, Yang K, Shi Y, Xu X. Recent advances in nanoadjuvant-triggered STING activation for enhanced cancer immunotherapy. Heliyon 2024; 10:e38900. [PMID: 39640775 PMCID: PMC11620084 DOI: 10.1016/j.heliyon.2024.e38900] [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: 04/30/2024] [Revised: 07/22/2024] [Accepted: 10/01/2024] [Indexed: 12/07/2024] Open
Abstract
The development of effective cancer treatments is a popular in contemporary medical research. Immunotherapy, the fourth most common cancer treatment method, relies on activating autoimmune function to eradicate tumors and exhibits advantages such as a good curative effect and few side effects. In recent years, tumor vaccines that activate the stimulator of interferon genes (STING) pathway are being actively researched in the field of immunotherapy; however, their application is still limited because of the rapid clearance rate of tumor-related lymph nodes and low efficiency of antigen presentation. The rise of nanomedicine has provided new opportunities for solving these problems. By preparing materials with adjuvant effects nanoparticles, the small size of nanoparticles can be exploited to enable the entry of vaccines into tumor-related lymph nodes to accurately deliver STING agonists and activate the immune response. Based on this, this paper reviews various types of nano-adjuvants based on metals, platinum chemotherapy drugs, camptothecin derivatives, deoxyribonucleic acid, etc. and highlights the transformation prospects of these nano-adjuvants in tumor vaccines to provide a reference for promoting the development of nano-medicine and tumor vaccinology.
Collapse
Affiliation(s)
- Zicong Xu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yihong Wu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Junjie Hu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Zhaozhao Mei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yutong Zhao
- College of Pharmacy, Jiamusi University, Jiamusi, 154007, PR China
| | - Keda Yang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Yi Shi
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, PR China
| | - Xiaoling Xu
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| |
Collapse
|
36
|
Shang X, Zhang C, Lv Y, Zhang X, Guo K, Li H, Wang H. Patients with Extensive-Stage Small Cell Lung Cancer Harboring Less Than 4 Metastatic Sites May Benefit from Immune Checkpoint Inhibitor Rechallenge by Reshaping Tumor Microenvironment. Immunotargets Ther 2024; 13:571-583. [PMID: 39478941 PMCID: PMC11523948 DOI: 10.2147/itt.s483093] [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: 06/17/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) has prolonged survival in patients with extensive-stage small cell lung cancer (ES-SCLC) as first-line treatment. However, whether ICI rechallenge could bring survival benefit to patients with ES-SCLC following its failure as first-line treatment remains unknown. Therefore, we aim to address the issue and identify the cohort of patients that may derive such benefit. Methods Patients with ES-SCLC from both the IMpower133 study and Shandong Cancer Hospital and Institute (shanzhong cohort) who failed first-line ICI were included. Kaplan Meier analysis was performed to compare overall survival (OS). Both univariate and multivariate Cox regression analyses were conducted to identify factors affecting survival. Tumor immune cell infiltration was evaluated by the CIBERSORT algorithm and detected by multiplex immunofluorescence (mIF). Results A total of 125 ES-SCLC patients undergoing atezolizumab and 161 patients undergoing ICI as first-line treatment were recruited from IMpower133 and shanzhong cohort. Those receiving ICI rechallenge had a longer OS than those without in IMpower133 (P = 0.08) and shanzhong cohort (P = 0.013). In IMpower133 cohort, subgroup analyses found that patients with <4 metastatic sites derived more survival benefit from atezolizumab (P = 0.008). For patients with ES-SCLC harboring <4 metastatic sites, there was significant OS difference between atezolizumab versus non-atezolizumab as retreatment (P = 0.036). Moreover, for ES-SCLC patients with <4 metastatic sites, atezolizumab improved survival compared with non-atezolizumab (hazard ratio [HR]: 0.457; 95% CI: 0.256-0.817; P = 0.008). These findings were confirmed in shanzhong cohort. Those harboring <4 metastatic sites had fewer M2 macrophage and more CD4 naïve T cells infiltration, which was further confirmed by mIF of ES-SCLC samples from shanzhong cohort. Conclusion Our study provides rationale for ICI rechallenge among ES-SCLC patients with <4 metastatic sites, suggesting beneficial outcome by reshaping TME.
Collapse
Affiliation(s)
- Xiaoling Shang
- Shandong Cancer Hospital and Institute, Shandong University, Jinan, 250117, People’s Republic of China
| | - Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, People’s Republic of China
| | - Yuanyuan Lv
- Department of Clinical Drug Research, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Xiaoxiao Zhang
- Department of Clinical Drug Research, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Kaiyue Guo
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Shandong University, Jinan, 250117, People’s Republic of China
| | - Huijuan Li
- Department of Clinical Drug Research, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117, People’s Republic of China
| |
Collapse
|
37
|
Cao Z, Zhu J, Wang Z, Peng Y, Zeng L. Comprehensive pan-cancer analysis reveals ENC1 as a promising prognostic biomarker for tumor microenvironment and therapeutic responses. Sci Rep 2024; 14:25331. [PMID: 39455818 PMCID: PMC11512054 DOI: 10.1038/s41598-024-76798-9] [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: 06/24/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Accumulating research showed that ENC1 plays a critical role in maintaining the physiological functions. However, little is known about its role in predicting prognosis and immunotherapy response across cancers. In our results, compared to normal tissues, most cancer tissues exhibit increased ENC1 expression. We found that the most common type of genetic variation was gene mutation. In addition, a positive correlation was found between CNV and ENC1 expression. Moreover, the overexpression of ENC1 was positively correlated with poor clinical outcomes. The GSEA results showed that ENC1 is closely correlated with tumor-promoting biological functions in most cancers. ENC1 is also closely negatively associated with the infiltration levels of T cells, activated NK cells, and B cells. Most immunomodulators are positively associated with ENC1. Further, we verified that inhibition of ENC1 expression suppressed the proliferation and migration of breast cancer, pancreatic cancer and glioma cells. In conclusion, our study demonstrated that ENC1 plays a protumorigenic role in most cancers. Additionally, ENC1 is closely correlated with tumor microenvironment features and immune checkpoint inhibitors expression. Overall, ENC1 could serve as a promising potential prognostic biomarker in various tumors.
Collapse
Affiliation(s)
- Zhenyu Cao
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Jinfeng Zhu
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, Hunan Province, China
| | - Zicheng Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yuhuai Peng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Liyun Zeng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China.
| |
Collapse
|
38
|
Hansen SB, Unal B, Kuzu OF, Saatcioglu F. Immunological facets of prostate cancer and the potential of immune checkpoint inhibition in disease management. Theranostics 2024; 14:6913-6934. [PMID: 39629128 PMCID: PMC11610136 DOI: 10.7150/thno.100555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/27/2024] [Indexed: 12/06/2024] Open
Abstract
Prostate cancer (PCa) is the most common non-cutaneous cancer in men and a major cause of cancer-related deaths. Whereas localized PCa can be cured by surgery and radiotherapy, metastatic disease can be treated, but is not curable. Inhibition of androgen signaling remains the main therapeutic intervention for treatment of metastatic PCa, in addition to chemotherapy, radionuclide therapy and emerging targeted therapies. Although initial responses are favorable, resistance to these therapies invariably arise with development of castration resistant PCa (CRPC) and lethal phenotypes. Recent findings have implicated the crosstalk between PCa cells and the tumor microenvironment (TME) as a key factor for disease progression and metastasis, and the immune system is becoming an increasingly attractive target for therapy. Given the striking success of immune checkpoint inhibitors (ICIs) in various cancer types, preclinical and clinical studies have begun to explore their potential in PCa. It has become clear that the PCa TME is largely immunosuppressive, and ICI therapy does not have efficacy for PCa. Intense effort is therefore being made in the field to understand the mechanisms of suppression and to turn the immunosuppressive TME into an immune active one that would enable ICI efficacy. Herein we examine this recent body of knowledge and how the mutational landscape of PCa integrates with an immunosuppressive TME to circumvent ICI-mediated T-cell activity and tumor killing. We then review the emerging potential success of combinatorial ICI approaches, utility of careful patient selection, and potential novel strategies to improve the efficacy of ICI for PCa therapy.
Collapse
Affiliation(s)
| | - Bilal Unal
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Omer Faruk Kuzu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
39
|
Ricci AD, Rizzo A, Schirizzi A, D’Alessandro R, Frega G, Brandi G, Shahini E, Cozzolongo R, Lotesoriere C, Giannelli G. Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: Regulatory Mechanisms, Functions, and Therapeutic Implications. Cancers (Basel) 2024; 16:3542. [PMID: 39456636 PMCID: PMC11505966 DOI: 10.3390/cancers16203542] [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: 09/18/2024] [Revised: 10/06/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Treatment options for intrahepatic cholangiocarcinoma (iCCA), a highly malignant tumor with poor prognosis, are limited. Recent developments in immunotherapy and immune checkpoint inhibitors (ICIs) have offered new hope for treating iCCA. However, several issues remain, including the identification of reliable biomarkers of response to ICIs and immune-based combinations. Tumor immune microenvironment (TIME) of these hepatobiliary tumors has been evaluated and is under assessment in this setting in order to boost the efficacy of ICIs and to convert these immunologically "cold" tumors to "hot" tumors. Herein, the review TIME of ICCA and its critical function in immunotherapy. Moreover, this paper also discusses potential avenues for future research, including novel targets for immunotherapy and emerging treatment plans aimed to increase the effectiveness of immunotherapy and survival rates for iCCA patients.
Collapse
Affiliation(s)
- Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Alessandro Rizzo
- S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
| | - Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Rosalba D’Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Giorgio Frega
- Osteoncology, Soft Tissue and Bone Sarcomas, Innovative Therapy Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy
| | - Raffaele Cozzolongo
- Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| |
Collapse
|
40
|
Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther 2024; 9:274. [PMID: 39420203 PMCID: PMC11491057 DOI: 10.1038/s41392-024-01979-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the "hot" (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct "cold" (immune-desert) phenotype, differing from the features of "hot" tumors. Additionally, there is a more nuanced "excluded" immune phenotype, positioned between the "cold" and "hot" categories, known as the immune "excluded" type. Effective differentiation between "cold" and "hot" tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on "hot" tumors, with limited efficacy against "cold" or "altered" tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert "cold" or "altered" tumors into "hot" ones. Therefore, aligning with the traits of "cold" and "hot" tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on "cold" and "hot" tumors to assess clinical efficacy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Youth League Committee, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bowen Li
- Department of Pancreatic and Gastrointestinal Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Haoqi Wu
- Department of Gynaecology and Obstetrics, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Jiang
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
41
|
Su L, Wang Z, Cai M, Wang Q, Wang M, Yang W, Gong Y, Fang F, Xu L. Single-cell analysis of matrisome-related genes in breast invasive carcinoma: new avenues for molecular subtyping and risk estimation. Front Immunol 2024; 15:1466762. [PMID: 39493752 PMCID: PMC11530991 DOI: 10.3389/fimmu.2024.1466762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Background The incidence of breast cancer remains high and severely affects human health. However, given the heterogeneity of tumor cells, identifying additional characteristics of breast cancer cells is essential for accurate treatment. Purpose This study aimed to analyze the relevant characteristics of matrix genes in breast cancer through the multigroup data of a breast cancer multi-database. Methods The related characteristics of matrix genes in breast cancer were analyzed using multigroup data from the breast cancer multi database in the Cancer Genome Atlas, and the differential genes of breast cancer matrix genes were identified using the elastic net penalty logic regression method. The risk characteristics of matrix genes in breast cancer were determined, and matrix gene expression in different breast cancer cells was evaluated using real-time fluorescent quantitative polymerase chain reaction (PCR). A consensus clustering algorithm was used to identify the biological characteristics of the population based on the matrix molecular subtypes in breast cancer, followed by gene mutation, immune correlation, pathway, and ligand-receptor analyses. Results This study reveals the genetic characteristics of cell matrix related to breast cancer. It is found that 18.1% of stromal genes are related to the prognosis of breast cancer, and these genes are mostly concentrated in the biological processes related to metabolism and cytokines in protein. Five different matrix-related molecular subtypes were identified by using the algorithm, and it was found that the five molecular subtypes were obviously different in prognosis, immune infiltration, gene mutation and drug-making gene analysis. Conclusions This study involved analyzing the characteristics of cell-matrix genes in breast cancer, guiding the precise prevention and treatment of the disease.
Collapse
Affiliation(s)
- Lingzi Su
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhe Wang
- The First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Mengcheng Cai
- The First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Qin Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Man Wang
- The First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Wenxiao Yang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yabin Gong
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fanfu Fang
- The First Affiliated Hospital of Naval Military Medical University, Shanghai, China
| | - Ling Xu
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
42
|
Li S, Wang Y, Li C, Zhou B, Zeng X, Zhu H. Supramolecular nanomedicine in the intelligent cancer therapy: recent advances and future. Front Pharmacol 2024; 15:1490139. [PMID: 39464634 PMCID: PMC11502448 DOI: 10.3389/fphar.2024.1490139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/30/2024] [Indexed: 10/29/2024] Open
Abstract
In recent years, the incidence of cancer has been increasing year by year, and the burden of the disease and the economic burden caused by it has been worsening. Although chemotherapy, immunotherapy, targeted therapy and other therapeutic means continue to progress, they still inevitably have problems such as high toxicity and side effects, susceptibility to drug resistance, and high price. Photothermal therapy and photodynamic therapy have demonstrated considerable advantages in cancer imaging and treatment due to their minimally invasive and selective nature. However, their development has been constrained by challenges related to drug delivery. In recent times, drug delivery systems constructed based on supramolecular chemistry have been the subject of considerable interest, particularly in view of their compatibility with the high permeability and long retention effect of tumors. Furthermore, the advantage of dissociating the active ingredient under pH, light and other stimuli makes them unique in cancer therapy. This paper reviews the current status of supramolecular nanomedicines in cancer therapy, elucidating the challenges faced and providing a theoretical basis for the efficient and precise treatment of malignant tumors.
Collapse
Affiliation(s)
- Shuo Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yujiao Wang
- Division of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Binghao Zhou
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Xiaoxi Zeng
- Division of Nephrology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
43
|
Reste M, Ajazi K, Sayi-Yazgan A, Jankovic R, Bufan B, Brandau S, Bækkevold ES, Petitprez F, Lindstedt M, Adema GJ, Almeida CR. The role of dendritic cells in tertiary lymphoid structures: implications in cancer and autoimmune diseases. Front Immunol 2024; 15:1439413. [PMID: 39483484 PMCID: PMC11526390 DOI: 10.3389/fimmu.2024.1439413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 09/23/2024] [Indexed: 11/03/2024] Open
Abstract
Tertiary Lymphoid Structures (TLS) are organized aggregates of immune cells such as T cells, B cells, and Dendritic Cells (DCs), as well as fibroblasts, formed postnatally in response to signals from cytokines and chemokines. Central to the function of TLS are DCs, professional antigen-presenting cells (APCs) that coordinate the adaptive immune response, and which can be classified into different subsets, with specific functions, and markers. In this article, we review current data on the contribution of different DC subsets to TLS function in cancer and autoimmunity, two opposite sides of the immune response. Different DC subsets can be found in different tumor types, correlating with cancer prognosis. Moreover, DCs are also present in TLS found in autoimmune and inflammatory conditions, contributing to disease development. Broadly, the presence of DCs in TLS appears to be associated with favorable clinical outcomes in cancer while in autoimmune pathologies these cells are associated with unfavorable prognosis. Therefore, it is important to analyze the complex functions of DCs within TLS in order to enhance our fundamental understanding of immune regulation but also as a possible route to create innovative clinical interventions designed for the specific needs of patients with diverse pathological diseases.
Collapse
Affiliation(s)
- Mariana Reste
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Kristi Ajazi
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Ayca Sayi-Yazgan
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Istanbul Technical University, Istanbul, Türkiye
- Department of Life Sciences, Centre for Inflammation Research and Translational Medicine, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Radmila Jankovic
- Faculty of Medicine, Institute of Pathology, University of Belgrade, Belgrade, Serbia
| | - Biljana Bufan
- Department of Microbiology and Immunology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| | - Sven Brandau
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Espen S. Bækkevold
- Department of Pathology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Florent Petitprez
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Gosse J. Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Catarina R. Almeida
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
44
|
Wang B, Xiao L, Chen P, Zhang T, Zhang P, Cao L, Zhou Z, Cheng H, Zhang T, Li S. Uncovering the role of traditional Chinese medicine in immune-metabolic balance of gastritis from the perspective of Cold and Hot: Jin Hong Tablets as a case study. Chin Med 2024; 19:134. [PMID: 39367502 PMCID: PMC11451182 DOI: 10.1186/s13020-024-00998-8] [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: 06/23/2024] [Accepted: 09/05/2024] [Indexed: 10/06/2024] Open
Abstract
Chronic gastritis (CG) is a common inflammatory disease of chronic inflammatory lesion of gastric mucosa and in the diagnosis of gastritis in traditional Chinese medicine (TCM), CG can be classified into Cold ZHENG (syndrome in TCM) and Hot ZHENG. However, the molecular features of Cold/Hot ZHENG in CG and the mechanism of Cold/Hot herbs in formulae for CG remained unclear. In this study, we collected a transcriptomics data including 35 patients of Cold/Hot ZHENG CG and 3 scRNA-seq CG samples. And 25 formulae for CG and 89 herbs recorded in these formulae were also collected. We conduct a comprehensive analysis based on the combination of transcriptomics datasets and machine learning algorithms, to discover biomarkers for Cold/Hot ZHENG CG. Then the target profiles of the collected formulae and Cold/Hot herbs were predicted to uncover the features and biomarkers of them against Cold/Hot ZHENG CG. These biomarkers suggest that Hot ZHENG CG might be characterized by over-inflammation and exuberant metabolism, and Cold ZHENG CG showed a trend of suppression in immune regulation and energy metabolism. Biomarkers and specific pathways of Hot herbs tend to regulate immune responses and energy metabolism, while those of Cold herbs are more likely to participate in anti-inflammatory effects. Finally, the findings were verified based on public transcriptomics datasets, as well as transcriptomics and ELISA detection, taking Jin Hong tablets as a case study. Biomarkers like leptin and IL-6 together with proportions of immune cells showed significant changes after the intervention. These findings might reflect the mechanism and build a bridge between macro and micro views of Cold/Hot ZHENG as well as Cold/Hot herbs.
Collapse
Affiliation(s)
- Boyang Wang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Lihao Xiao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co.,Ltd., Lianyungang, 222047, Jiangshu, China
| | - Pan Chen
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing, 100084, China
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangshu, China
| | - Tingyu Zhang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Peng Zhang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Liang Cao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co.,Ltd., Lianyungang, 222047, Jiangshu, China
| | - Ziyi Zhou
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Haibo Cheng
- Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangshu, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Shao Li
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
45
|
Guo W, Tan J, Wang L, Egelston CA, Simons DL, Ochoa A, Lim MH, Wang L, Solomon S, Waisman J, Wei CH, Hoffmann C, Song J, Schmolze D, Lee PP. Tumor draining lymph nodes connected to cold triple-negative breast cancers are characterized by Th2-associated microenvironment. Nat Commun 2024; 15:8592. [PMID: 39366933 PMCID: PMC11452381 DOI: 10.1038/s41467-024-52577-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/10/2024] [Indexed: 10/06/2024] Open
Abstract
Tumor draining lymph nodes (TDLN) represent a key component of the tumor-immunity cycle. There are few studies describing how TDLNs impact lymphocyte infiltration into tumors. Here we directly compare tumor-free TDLNs draining "cold" and "hot" human triple negative breast cancers (TDLNCold and TDLNHot). Using machine-learning-based self-correlation analysis of immune gene expression, we find unbalanced intranodal regulations within TDLNCold. Two gene pairs (TBX21/GATA3-CXCR1) with opposite correlations suggest preferential priming of T helper 2 (Th2) cells by mature dendritic cells (DC) within TDLNCold. This is validated by multiplex immunofluorescent staining, identifying more mature-DC-Th2 spatial clusters within TDLNCold versus TDLNHot. Associated with this Th2 priming preference, more IL4 producing mast cells (MC) are found within sinus regions of TDLNCold. Downstream, Th2-associated fibrotic TME is found in paired cold tumors with increased Th2/T-helper-1-cell (Th1) ratio, upregulated fibrosis growth factors, and stromal enrichment of cancer associated fibroblasts. These findings are further confirmed in a validation cohort and public genomic data. Our results reveal a potential role of IL4+ MCs within TDLNs, associated with Th2 polarization and reduced immune infiltration into tumors.
Collapse
Affiliation(s)
- Weihua Guo
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Jiayi Tan
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Irell & Manella Graduate School of Biological Sciences, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Lei Wang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- International Cancer Center, Shenzhen University Medical School, 518060, Shenzhen, Guangdong, China
| | - Colt A Egelston
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Diana L Simons
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Aaron Ochoa
- Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Min Hui Lim
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Genomics Core, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Lu Wang
- Mork Family Department of Chemical Engineering & Material Science, University of Southern California, Los Angeles, CA, 90089, USA
| | - Shawn Solomon
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - James Waisman
- Department of Medical Oncology, City of Hope, Duarte, CA, 91010, USA
| | - Christina H Wei
- Department of Pathology, City of Hope, Duarte, CA, 91010, USA
- Pathology Laboratory Administration, Los Angeles General Medical Center, Los Angeles, CA, 90033, USA
| | - Caroline Hoffmann
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
- Owkin, Inc., New York, NY, 10003, USA
| | - Joo Song
- Department of Pathology, City of Hope, Duarte, CA, 91010, USA
| | - Daniel Schmolze
- Department of Pathology, City of Hope, Duarte, CA, 91010, USA
| | - Peter P Lee
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
46
|
Fischer A, Albert TK, Moreno N, Interlandi M, Mormann J, Glaser S, Patil P, de Faria FW, Richter M, Verma A, Balbach ST, Wagener R, Bens S, Dahlum S, Göbel C, Münter D, Inserte C, Graf M, Kremer E, Melcher V, Di Stefano G, Santi R, Chan A, Dogan A, Bush J, Hasselblatt M, Cheng S, Spetalen S, Fosså A, Hartmann W, Herbrüggen H, Robert S, Oyen F, Dugas M, Walter C, Sandmann S, Varghese J, Rossig C, Schüller U, Tzankov A, Pedersen MB, d'Amore FA, Mellgren K, Kontny U, Kancherla V, Veloza L, Missiaglia E, Fataccioli V, Gaulard P, Burkhardt B, Soehnlein O, Klapper W, de Leval L, Siebert R, Kerl K. Lack of SMARCB1 expression characterizes a subset of human and murine peripheral T-cell lymphomas. Nat Commun 2024; 15:8571. [PMID: 39362842 PMCID: PMC11452211 DOI: 10.1038/s41467-024-52826-0] [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: 05/04/2023] [Accepted: 09/23/2024] [Indexed: 10/05/2024] Open
Abstract
Peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS) is a heterogeneous group of malignancies with poor outcome. Here, we identify a subgroup, PTCL-NOSSMARCB1-, which is characterized by the lack of the SMARCB1 protein and occurs more frequently in young patients. Human and murine PTCL-NOSSMARCB1- show similar DNA methylation profiles, with hypermethylation of T-cell-related genes and hypomethylation of genes involved in myeloid development. Single-cell analyses of human and murine tumors revealed a rich and complex network of interactions between tumor cells and an immunosuppressive and exhausted tumor microenvironment (TME). In a drug screen, we identified histone deacetylase inhibitors (HDACi) as a class of drugs effective against PTCL-NOSSmarcb1-. In vivo treatment of mouse tumors with SAHA, a pan-HDACi, triggered remodeling of the TME, promoting replenishment of lymphoid compartments and reversal of the exhaustion phenotype. These results provide a rationale for further exploration of HDACi combination therapies targeting PTCL-NOSSMARCB1- within the TME.
Collapse
MESH Headings
- Animals
- SMARCB1 Protein/genetics
- SMARCB1 Protein/metabolism
- Humans
- Lymphoma, T-Cell, Peripheral/genetics
- Lymphoma, T-Cell, Peripheral/drug therapy
- Lymphoma, T-Cell, Peripheral/metabolism
- Lymphoma, T-Cell, Peripheral/pathology
- Mice
- Histone Deacetylase Inhibitors/pharmacology
- Tumor Microenvironment/genetics
- Tumor Microenvironment/drug effects
- DNA Methylation
- Gene Expression Regulation, Neoplastic
- Female
- Cell Line, Tumor
- Male
- Vorinostat/pharmacology
- Single-Cell Analysis
Collapse
Affiliation(s)
- Anja Fischer
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Thomas K Albert
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Natalia Moreno
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Marta Interlandi
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Jana Mormann
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Selina Glaser
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Paurnima Patil
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Flavia W de Faria
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Mathis Richter
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Archana Verma
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Sebastian T Balbach
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Rabea Wagener
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Susanne Bens
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Sonja Dahlum
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Carolin Göbel
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Eppendorf (UKE), 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Daniel Münter
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Clara Inserte
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Monika Graf
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Eva Kremer
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Viktoria Melcher
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Gioia Di Stefano
- Pathological Anatomy Section, Careggi University Hospital, Florence, Italy
| | - Raffaella Santi
- Pathological Anatomy Section, Careggi University Hospital, Florence, Italy
| | - Alexander Chan
- Department of Pathology, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Ahmet Dogan
- Department of Pathology, Hematopathology Service, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Jonathan Bush
- Division of Anatomical Pathology, British Columbia Children's Hospital and Women's Hospital and Health Center, Vancouver, BC, Canada
| | - Martin Hasselblatt
- Institute of Neuropathology, University Hospital Münster, 48149, Münster, Germany
| | - Sylvia Cheng
- Division of Pediatric Hematology/Oncology/BMT, Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Signe Spetalen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Alexander Fosså
- Department of Oncology, Oslo University Hospital-Norwegian Radium Hospital, Oslo, Norway
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard-Domagk-Institut für Pathologie, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude D17, 48149, Münster, Germany
| | - Heidi Herbrüggen
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Stella Robert
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Florian Oyen
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Eppendorf (UKE), 20251, Hamburg, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Sarah Sandmann
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Julian Varghese
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg, Eppendorf (UKE), 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf (UKE), 20251, Hamburg, Germany
| | - Alexandar Tzankov
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Martin B Pedersen
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Francesco A d'Amore
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karin Mellgren
- Department of Pediatric Oncology and Hematology, Sahlgrenska University Hospital, The Queen Silvia Children's Hospital, Gothenburg, Sweden
| | - Udo Kontny
- Section of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Department of Pediatric and Adolescent Medicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Venkatesh Kancherla
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Luis Veloza
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Edoardo Missiaglia
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Virginie Fataccioli
- INSERM U955, Université Paris-Est, Créteil, France
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, INSERM U955, Université Paris Est Créteil, Créteil, France
| | - Philippe Gaulard
- Département de Pathologie, Hôpitaux Universitaires Henri Mondor, AP-HP, INSERM U955, Université Paris Est Créteil, Créteil, France
| | - Birgit Burkhardt
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany
| | - Oliver Soehnlein
- Institute for Experimental Pathology, Center for Molecular Biology of Inflammation, University of Münster, Münster, Germany
| | - Wolfram Klapper
- Department of Pathology, Haematopathology Section and Lymph Node Registry, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital, Lausanne, Switzerland
| | - Reiner Siebert
- Institute of Human Genetics, Ulm University Medical Center, Ulm, Germany
| | - Kornelius Kerl
- Department of Pediatric Hematology and Oncology, University Children's Hospital Münster, Münster, Germany.
| |
Collapse
|
47
|
Lan J, Zeng R, Li Z, Yang X, Liu L, Chen L, Sun L, Shen Y, Zhang T, Ding Y. Biomimetic Nanomodulators With Synergism of Photothermal Therapy and Vessel Normalization for Boosting Potent Anticancer Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408511. [PMID: 39180264 DOI: 10.1002/adma.202408511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/09/2024] [Indexed: 08/26/2024]
Abstract
Combination therapy using photothermal therapy (PTT) and immunotherapy is one of the most promising approaches for eliciting host immune responses to ablate tumors. However, its therapeutic efficacy is limited due to inefficient immune cell infiltration and cellular immune responses. In this study, a biomimetic immunostimulatory nanomodulator, Tm@PDA-GA (4T1 membrane@polydopamine-gambogic acid), with homologous targeting is developed. The 4T1 membrane (Tm) coating reduced immunogenicity and facilitated uptake of Tm@PDA-GA by tumor cells. Polydopamine (PDA) as a drug carrier can induce PTT under near-infrared ray (NIR) irradiation and immunogenic cell death (ICD) to activate dendritic cells (DCs). Moreover, Tm@PDA-GA on-demand released gambogic acid (GA) in an acidic tumor microenvironment, inhibiting the expression of heat shock proteins (HSPs) for synergetic chemo-photothermal anti-tumor activity and increasing the ICD of 4T1 cells. More importantly, GA can normalize the vessels via HIF-1α and VEGF inhibition to enhance immune infiltration and alleviate hypoxia stress. Thus, Tm@PDA-GA induced ICD, activated DCs, stimulated cytotoxic T cells, and suppressed Tregs. Moreover, Tm@PDA-GA is combined with anti-PD-L1 to further augment the tumor immune response and effectively suppress tumor growth and lung metastasis. In conclusion, biomaterial-mediated PTT combined with vessel normalization is a promising strategy for effective immunotherapy of triple-negative breast cancer (TNBC).
Collapse
Affiliation(s)
- Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ruifeng Zeng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xuguang Yang
- Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lixia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Liyan Sun
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yi Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- National Innovation Platform for Medical Industry-Education Integration, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
48
|
Chen D, Zhang H, Zhao L, Liu X, Lou Y, Wu P, Xue S, Jiang H. RILPL2 as a potential biomarker for predicting enhanced T cell infiltration in non-small cell lung cancer. Immunol Res 2024; 72:1174-1184. [PMID: 39078518 PMCID: PMC11564405 DOI: 10.1007/s12026-024-09520-6] [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: 07/01/2024] [Accepted: 07/19/2024] [Indexed: 07/31/2024]
Abstract
Our previous bioinformatics analysis has revealed that Rab-interacting lysosomal protein-like 2 (RILPL2) is associated with tumor immune microenvironment in non-small cell lung cancer (NSCLC). In our study, we collected 140 patients with primary NSCLC to verify the RILPL2 expression and its prognostic value, the relationship between RILPL2 expression and CD4+, CD8+T cell infiltration. A total of 140 patients who had been diagnosed with primary NSCLC (including 66 lung adenocarcinomas and 74 lung squamous cell carcinomas) were enrolled in our study. Immunohistochemical (IHC) staining was performed to analyze the expression of RILPL2, CD4, and CD8 in these patients. Compared with peri-cancer tissues, the RILPL2 expression in NSCLC tissues was significantly lower (P < 0.0001). RILPL2 expression was significantly related to clinical stage (P = 0.019), and low RILPL2 expression indicated higher stage. Low RILPL2 expression predicted worse overall survival (OS) in NSCLC patients (P = 0.017). Correlational analyses revealed that RILPL2 expression was significantly positively correlated with CD4+T cell infiltration in NSCLC (R = 0.294, P < 0.001), LUAD subgroup (R = 0.256, P = 0.038), and LUSC subgroup (R = 0.333, P = 0.004); RILPL2 expression was also significantly positively correlated with CD8+ T cell infiltration in NSCLC (R = 0.263, P = 0.002), LUAD subgroup (R = 0.280, P = 0.023), and LUSC subgroup (R = 0.250, P = 0.031). In conclusion, RILPL2 expression was downregulated in NSCLC; low RILPL2 expression was significantly related to higher stage and worse prognosis; RILPL2 expression was significantly positively correlated with CD4+, CD8+T cell infiltration.
Collapse
Affiliation(s)
- Dongfang Chen
- Department of Respiratory and Critical Care Medicine, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Hongyan Zhang
- Department of Respiratory and Critical Care Medicine, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Lifang Zhao
- Department of Respiratory and Critical Care Medicine, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xueqing Liu
- Department of Respiratory and Critical Care Medicine, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Yueyan Lou
- Department of Respiratory and Critical Care Medicine, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Peiling Wu
- Department of Respiratory and Critical Care Medicine, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Shan Xue
- Department of Respiratory and Critical Care Medicine, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Handong Jiang
- Department of Respiratory and Critical Care Medicine, Renji Hospital Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
49
|
Li X, Li Y, Tuerxun H, Zhao Y, Liu X, Zhao Y. Firing up "cold" tumors: Ferroptosis causes immune activation by improving T cell infiltration. Biomed Pharmacother 2024; 179:117298. [PMID: 39151313 DOI: 10.1016/j.biopha.2024.117298] [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: 06/09/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
Immune checkpoint blocking (ICB), a tumor treatment based on the mechanism of T-cell activation, has shown high efficacy in clinical trials, but not all patients benefit from it. Immune checkpoint inhibitors (ICIs) do not respond to cold tumors that lack effective T-cell infiltration but respond well to hot tumors with sufficient T-cell infiltration. How to convert an unresponsive cold tumor into a responsive hot tumor is an important topic in cancer immunotherapy. Ferroptosis, a newly discovered immunogenic cell death (ICD) form, has great potential in cancer therapy. In the process of deeply understanding the mechanism of cold tumor formation, it was found that ferroptosis showed a powerful immune-activating effect by improving T-cell infiltration, and the combination of ICB therapy significantly enhanced the anti-tumor efficacy. This paper reviews the complex relationship between T cells and ferroptosis, as well as summarizes the various mechanisms by which ferroptosis enhances T cell infiltration: reactivation of T cells and reversal of immunosuppressive tumor microenvironment (TME), as well as recent advances of ICI in combination with targeted ferroptosis therapies, which provides guidance for better improving the ICB efficacy of cold tumors.
Collapse
Affiliation(s)
- Xinru Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yawen Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Halahati Tuerxun
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xingyu Liu
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
50
|
Bao X, Sun M, Meng L, Zhang H, Yi X, Zhang P. Applications of pyroptosis activators in tumor immunotherapy. Mater Today Bio 2024; 28:101191. [PMID: 39221221 PMCID: PMC11363858 DOI: 10.1016/j.mtbio.2024.101191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Contemporary progress in tumor immunotherapy has solidified its role as an effective approach in combating cancer. Nonetheless, the prevalent "immune cold" state within the tumor microenvironment poses a substantial barrier to its efficacy. Addressing this, pyroptosis-a gasdermin-mediated programmed cell death characterized by its inflammatory profile-emerges as a crucial mechanism. It catalyzes the release of vast quantities of pro-inflammatory cytokines and immunogens, potentially transforming immunosuppressive "cold" tumors into reactive "hot" ones. Herein, we will initially present an overview of pyroptosis as a distinct form of cell death, along with its molecular mechanisms. Subsequently, we will focus on introducing how pyroptosis activators are utilized in the field of tumor immunotherapy. Insights gained from applications of pyroptosis activators in tumor immunotherapy could lead to the development of safe and efficient pyroptosis activators, significantly enriching the arsenal for tumor immunotherapy.
Collapse
Affiliation(s)
- Xin Bao
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Mengmeng Sun
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Lingfei Meng
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Hong Zhang
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Xuan Yi
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| |
Collapse
|