1
|
Baldari S, Antonini A, Di Rocco G, Toietta G. Expression pattern and prognostic significance of aldehyde dehydrogenase 2 in lung adenocarcinoma as a potential predictor of immunotherapy efficacy. CANCER INNOVATION 2025; 4:e149. [PMID: 39640071 PMCID: PMC11620833 DOI: 10.1002/cai2.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/30/2024] [Accepted: 05/23/2024] [Indexed: 12/07/2024]
Abstract
Background The incidence of alcohol-associated cancers is higher within Asian populations having an increased prevalence of an inactivating mutation in aldehyde dehydrogenase 2 (ALDH2), a mitochondrial enzyme required for the clearance of acetaldehyde, a cytotoxic metabolite of ethanol. The role of alcohol consumption in promoting lung cancer is controversial, and little attention has been paid to the association between alcohol drinking and pulmonary ALDH2 expression. Methods We performed a comprehensive bioinformatic analysis of multi-omics data available in public databases to elucidate the role of ALDH2 in lung adenocarcinoma (LUAD). Results Transcriptional and proteomic data indicate a substantial pulmonary expression of ALDH2, which is functional for the metabolism of alcohol diffused from the bronchial circulation. ALDH2 expression is higher in healthy lung tissue than in LUAD and inhibits cell cycle, apoptosis, and epithelial-mesenchymal transition pathways. Moreover, low ALDH2 mRNA levels predict poor prognosis and low overall survival in LUAD patients. Interestingly, ALDH2 expression correlates with immune infiltration in LUAD. Conclusions A better understanding of the role of ALDH2 in lung tumor progression and immune infiltration might support its potential use as a prognostic marker and therapeutic target for improving immunotherapeutic response.
Collapse
Affiliation(s)
- Silvia Baldari
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Annalisa Antonini
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Giuliana Di Rocco
- Unit of Cellular Networks and Molecular Therapeutic TargetsIRCCS Regina Elena National Cancer InstituteRomeItaly
| | - Gabriele Toietta
- Tumor Immunology and Immunotherapy UnitIRCCS Regina Elena National Cancer InstituteRomeItaly
| |
Collapse
|
2
|
Dai Y, Tian X, Ye X, Gong Y, Xu L, Jiao L. Role of the TME in immune checkpoint blockade resistance of non-small cell lung cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:52. [PMID: 39802954 PMCID: PMC11724356 DOI: 10.20517/cdr.2024.166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 01/16/2025]
Abstract
Primary and secondary resistance to immune checkpoint blockade (ICB) reduces its efficacy. The mechanisms underlying immunotherapy resistance are highly complex. In non-small cell lung cancer (NSCLC), these mechanisms are primarily associated with the loss of programmed cell death-ligand 1 (PD-L1) expression, genetic mutations, circular RNA axis and transcription factor regulation, antigen presentation disorders, and dysregulation of signaling pathways. Additionally, alterations in the tumor microenvironment (TME) play a pivotal role in driving immunotherapy resistance. Primary resistance is mainly attributed to TME alterations, including mutations and co-mutations, modulation of T cell infiltration, enrichment of M2 tumor-associated macrophages (M2-TAMs) and mucosal-associated invariant T (MAIT) cells, vascular endothelial growth factor (VEGF), and pulmonary fibrosis. Acquired resistance mainly stems from changes in cellular infiltration patterns leading to "cold" or "hot" tumors, altered interferon (IFN) signaling pathway expression, involvement of extracellular vesicles (EVs), and oxidative stress responses, as well as post-treatment gene mutations and circadian rhythm disruption (CRD). This review presents an overview of various mechanisms underlying resistance to ICB, elucidates the alterations in the TME during primary, adaptive, and acquired resistance, and discusses existing strategies for overcoming ICB resistance.
Collapse
Affiliation(s)
- Yuening Dai
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xueqi Tian
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Xuanting Ye
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Yabin Gong
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Ling Xu
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Translational Cancer Research for Integrated Chinese and Western Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Lijing Jiao
- Department of Oncology I, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
- Institute of Translational Cancer Research for Integrated Chinese and Western Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| |
Collapse
|
3
|
Yang C, You J, Wang Y, Chen S, Tang Y, Chen H, Zhong H, Song R, Long H, Xiang T, Zhao ZR, Xia J. TLS and immune cell profiling: immunomodulatory effects of immunochemotherapy on tumor microenvironment in resectable stage III NSCLC. Front Immunol 2024; 15:1499731. [PMID: 39726591 PMCID: PMC11670196 DOI: 10.3389/fimmu.2024.1499731] [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: 09/21/2024] [Accepted: 11/21/2024] [Indexed: 12/28/2024] Open
Abstract
Background The use of programmed death-1 (PD-1) inhibitors in the neoadjuvant setting for patients with resectable stage III NSCLC has revolutionized this field in recent years. However, there is still 40%-60% of patients do not benefit from this approach. The complex interactions between immune cell subtypes and tertiary lymphoid structures (TLSs) within the tumor microenvironment (TME) may influence prognosis and the response to immunochemotherapy. This study aims to assess the relationship between immune cells subtypes and TLSs to better understand their impact on immunotherapy response. Methods This study initially compared the tertiary lymphoid structures (TLSs) density among patients who underwent immunochemotherapy, chemotherapy and upfront surgery using 123 tumor samples from stage-matched patients. Multiplex immunohistochemistry (mIHC) was employed to analyze the spatial distribution of PD-L1+CD11c+ cells and PD1+CD8+ T cells within TLSs. Cytometry by time-of-flight (CyTOF) was used to assess immune cell dynamics in paired biopsy and resection specimens from six patients who underwent immunochemotherapy. Key immune cells were validated in newly collected samples using flow cytometry, mIHC, and in vitro CAR-T cells model. Results Patients who underwent neoadjuvant chemotherapy or immunochemotherapy exhibited increased TLSs compared to those who opted for upfront surgery. The TLS area-to-tumor area ratio distinguished pCR+MPR and NR patients in the immunochemotherapy group. Spatial analysis revealed variations in the distance between PD-L1+CD11c+ cells and PD1+CD8+ T cells within TLSs in the immunochemotherapy group. CyTOF analysis revealed an increase in the frequency of key immune cells (CCR7+CD127+CD69+CD4+ and CD38+CD8+ cells) following combined therapy. Treatment responders exhibited an increase in CCR7+CD4+ T cells, whereas CD38+CD8+ T cells were associated with compromised treatment effectiveness. Conclusions Immunochemotherapy and chemotherapy increase TLSs and granzyme B+ CD8+ T cells in tumors. The TLS area-to-tumor ratio distinguishes responders from non-responders, with PD-L1+ dendritic cells near CD8+PD-1+ T cells linked to efficacy, suggesting that PD-1 inhibitors disrupt harmful interactions. Post-immunochemotherapy, CD8+ T cells increase, but CD38+CD8+ T cells show reduced functionality. These findings highlight the complex immune dynamics and their implications for NSCLC treatment.
Collapse
Affiliation(s)
- Chaopin Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jinqi You
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yizhi Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Si Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Yan Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Hao Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Haoran Zhong
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ruyue Song
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Hao Long
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Tong Xiang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Ze-Rui Zhao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Jianchuan Xia
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
- Department of Biotherapy, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
López-Rodríguez JC, Barral P. Mucosal associated invariant T cells: Powerhouses of the lung. Immunol Lett 2024; 269:106910. [PMID: 39128630 DOI: 10.1016/j.imlet.2024.106910] [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: 07/09/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The lungs face constant environmental challenges from harmless molecules, airborne pathogens and harmful agents that can damage the tissue. The lungs' immune system includes numerous tissue-resident lymphocytes that contribute to maintain tissue homeostasis and to the early initiation of immune responses. Amongst tissue-resident lymphocytes, Mucosal Associated Invariant T (MAIT) cells are present in human and murine lungs and emerging evidence supports their contribution to immune responses during infections, chronic inflammatory disorders and cancer. This review explores the mechanisms underpinning MAIT cell functions in the airways, their impact on lung immunity and the potential for targeting pulmonary MAIT cells in a therapeutic context.
Collapse
Affiliation(s)
- J C López-Rodríguez
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK.
| | - P Barral
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK.
| |
Collapse
|
5
|
Guo L, Li J, Wang J, Chen X, Cai C, Zhou F, Xiong A. Prognostic role of dynamic changes in inflammatory indicators in patients with non-small cell lung cancer treated with immune checkpoint inhibitors-a retrospective cohort study. Transl Lung Cancer Res 2024; 13:1975-1987. [PMID: 39263031 PMCID: PMC11384502 DOI: 10.21037/tlcr-24-637] [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: 07/21/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024]
Abstract
Background Immune checkpoint inhibitors (ICIs) have become one of the standard treatments for non-small cell lung cancer (NSCLC) patients without driver mutations. However, a considerable proportion of patients suffer from severe immune side effects and fail to respond to ICIs. As effective biomarkers, programmed cell death ligand 1 (PD-L1) expression, microsatellite instability (MSI), the tumor mutation burden (TMB) and tumor-infiltrating lymphocytes (TILs) require invasive procedures that place heavy physical and psychological burdens on patients. This study aims to identify simple and effective markers to optimize patient selection through therapeutic decisions and outcome prediction. Methods This retrospective study comprised 95 patients with metastatic NSCLC who were treated with ICIs either as the standard of care or in a clinical trial. The following data were extracted from the medical records. The baseline and dynamic neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) were calculated in the present study. Responses were assessed by computed tomography (CT) imaging and classified according to the Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 every 6-12 weeks during treatment. Results In total, 95 patients were included in the present study. The median age of patients was 61 years, 83.2% (79/95) patients were male, 62.1% (59/95) were former or current smokers, 66.3% (63/95) had adenocarcinoma, 93.7% (89/95) had stage IV disease, and 87.4% were without molecular alterations. A higher overall response rate (ORR) and prolonged median progression-free survival (PFS) was observed in patients with a lower cycle 3 (C3) NLR [7.7 vs. 5.5 months, hazard ratio (HR): 1.70, 95% confidence interval (CI): 0.90-3.22; P=0.12] and derived NLR (dNLR) (8.2 vs. 5.6 months, HR: 1.67, 95% CI: 0.94-2.97; P=0.08). After two cycles of ICI treatment, patients who had an increased NLR, dNLR, and PLR had a lower ORR and an inferior median PFS than those with a decreased NLR (5.5 vs. 8.5 months, HR: 1.87, 95% CI: 1.09-3.21; P=0.02), dNLR (5.6 vs. 8.4 months, HR: 1.49, 95% CI: 0.87-2.57; P=0.15), and PLR (11.8 vs. 5.5 months, HR: 2.28, 95% CI: 1.32-3.94; P=0.003). Moreover, patients with both an increased NLR and PLR had a worse ORR and median PFS than those with either an increased NLR or PLR, or both an increased NLR and PLR (11.8 vs. 5.5 vs. 5.6 months, P=0.003). In addition, the dynamic changes in the PLR could serve as an independent predictive factor of PFS in NSCLC patients treated with ICIs. Conclusions Elevated dynamic changes in the NLR and PLR were associated with lower response rates and shorter PFS in the patients with NSCLC treated with ICIs. Our results also highlight the role of dynamic changes in the PLR in identifying patients with NSCLC who could benefit from ICIs.
Collapse
Affiliation(s)
- Liang Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juanjuan Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Jing Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Xinru Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Chenlei Cai
- Department of Medical Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Fei Zhou
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Anwen Xiong
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Qu J, Wu B, Chen L, Wen Z, Fang L, Zheng J, Shen Q, Heng J, Zhou J, Zhou J. CXCR6-positive circulating mucosal-associated invariant T cells can identify patients with non-small cell lung cancer responding to anti-PD-1 immunotherapy. J Exp Clin Cancer Res 2024; 43:134. [PMID: 38698468 PMCID: PMC11067263 DOI: 10.1186/s13046-024-03046-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 04/13/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Mucosal-associated invariant T (MAIT) cells have been reported to regulate tumor immunity. However, the immune characteristics of MAIT cells in non-small cell lung cancer (NSCLC) and their correlation with the treatment efficacy of immune checkpoint inhibitors (ICIs) remain unclear. PATIENTS AND METHODS In this study, we performed single-cell RNA sequencing (scRNA-seq), flow cytometry, and multiplex immunofluorescence assays to determine the proportion and characteristics of CD8+MAIT cells in patients with metastatic NSCLC who did and did not respond to anti-PD-1 therapy. Survival analyses were employed to determine the effects of MAIT proportion and C-X-C chemokine receptor 6 (CXCR6) expression on the prognosis of patients with advanced NSCLC. RESULTS The proportion of activated and proliferating CD8+MAIT cells were significantly higher in responders-derived peripheral blood mononuclear cells (PBMCs) and lung tissues before anti-PD-1 therapy, with enhanced expression of cytotoxicity-related genes including CCL4, KLRG1, PRF1, NCR3, NKG7, GZMB, and KLRK1. The responders' peripheral and tumor-infiltrating CD8+MAIT cells showed an upregulated CXCR6 expression. Similarly, CXCR6+CD8+MAIT cells from responders showed higher expression of cytotoxicity-related genes, such as CST7, GNLY, KLRG1, NKG7, and PRF1. Patients with ≥15.1% CD8+MAIT cells to CD8+T cells ratio and ≥35.9% CXCR6+CD8+MAIT cells to CD8+MAIT cells ratio in peripheral blood showed better progression-free survival (PFS) after immunotherapy. The role of CD8+MAIT cells in lung cancer immunotherapy was potentially mediated by classical/non-classical monocytes through the CXCL16-CXCR6 axis. CONCLUSION CD8+MAIT cells are a potential predictive biomarker for patients with NSCLC responding to anti-PD-1 therapy. The correlation between CD8+MAIT cells and immunotherapy sensitivity may be ascribed to high CXCR6 expression.
Collapse
Affiliation(s)
- Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Binggen Wu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Zuoshi Wen
- Department of Cardiology, The First Affiliated Hospital, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
| | - Liangjie Fang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Jing Zheng
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Qian Shen
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Jianfu Heng
- Department of Clinical Pharmaceutical Research Institution, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, P. R. China.
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China.
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China.
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| |
Collapse
|
7
|
Guo H, Li X, Mao D, Wang H, Wei L, Qu D, Qin X, Li X, Liu Y, Chen Y. Homologous-magnetic dual-targeted metal-organic framework to improve the Anti-hepatocellular carcinoma efficacy of PD-1 inhibitor. J Nanobiotechnology 2024; 22:206. [PMID: 38658950 PMCID: PMC11044376 DOI: 10.1186/s12951-024-02469-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024] Open
Abstract
The insufficient abundance and weak activity of tumour-infiltrating lymphocytes (TILs) are two important reasons for the poor efficacy of PD-1 inhibitors in hepatocellular carcinoma (HCC) treatment. The combined administration of tanshinone IIA (TSA) and astragaloside IV (As) can up-regulate the abundance and activity of TILs by normalising tumour blood vessels and reducing the levels of immunosuppressive factors respectively. For enhancing the efficacy of PD-1 antibody, a magnetic metal-organic framework (MOF) with a homologous tumour cell membrane (Hm) coating (Hm@TSA/As-MOF) is established to co-deliver TSA&As into the HCC microenvironment. Hm@TSA/As-MOF is a spherical nanoparticle and has a high total drug-loading capacity of 16.13 wt%. The Hm coating and magnetic responsiveness of Hm@TSA/As-MOF provide a homologous-magnetic dual-targeting, which enable Hm@TSA/As-MOF to counteract the interference posed by ascites tumour cells and enhance the precision of targeting solid tumours. Hm coating also enable Hm@TSA/As-MOF to evade immune clearance by macrophages. The release of TSA&As from Hm@TSA/As-MOF can be accelerated by HCC microenvironment, thereby up-regulating the abundance and activity of TILs to synergistic PD-1 antibody against HCC. This study presents a nanoplatform to improve the efficacy of PD-1 inhibitors in HCC, providing a novel approach for anti-tumour immunotherapy in clinical practice.
Collapse
Affiliation(s)
- Hong Guo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Rd, Qixia Qu, Nanjing, Jiangsu, 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Probince Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Xia Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Rd, Qixia Qu, Nanjing, Jiangsu, 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Probince Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Dengxuan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Rd, Qixia Qu, Nanjing, Jiangsu, 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Probince Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Hong Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Rd, Qixia Qu, Nanjing, Jiangsu, 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Probince Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Liangyin Wei
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Rd, Qixia Qu, Nanjing, Jiangsu, 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Probince Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Ding Qu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Rd, Qixia Qu, Nanjing, Jiangsu, 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Probince Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Xiaoying Qin
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Rd, Qixia Qu, Nanjing, Jiangsu, 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Probince Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Xiaoqi Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Rd, Qixia Qu, Nanjing, Jiangsu, 210028, China
- Multi-component of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Probince Academy of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China
| | - Yuping Liu
- Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China.
| | - Yan Chen
- Jiangsu Clinical Innovation Center of Digestive Cancer of Traditional Chinese Medicine, Nanjing, Jiangsu, 210028, China.
| |
Collapse
|
8
|
Hou J, Yang X, Xie S, Zhu B, Zha H. Circulating T cells: a promising biomarker of anti-PD-(L)1 therapy. Front Immunol 2024; 15:1371559. [PMID: 38576625 PMCID: PMC10991692 DOI: 10.3389/fimmu.2024.1371559] [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: 01/16/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Anti-PD-(L)1 therapy has shown great efficacy in some patients with cancer. However, a significant proportion of patients with cancer do not respond to it. Another unmet clinical need for anti-PD-(L)1 therapy is the dynamic monitoring of treatment effects. Therefore, identifying biomarkers that can stratify potential responders before PD-(L)1 treatment and timely monitoring of the efficacy of PD-(L)1 treatment are crucial in the clinical setting. The identification of biomarkers by liquid biopsy has attracted considerable attention. Among the identified biomarkers, circulating T cells are one of the most promising because of their indispensable contribution to anti-PD-(L)1 therapy. The present review aimed to thoroughly explore the potential of circulating T cells as biomarkers of anti-PD-(L)1 therapy and its advantages and limitations.
Collapse
Affiliation(s)
- Junlei Hou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xuezhi Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shuanglong Xie
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Haoran Zha
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
9
|
Yigit M, Basoglu OF, Unutmaz D. Mucosal-associated invariant T cells in cancer: dual roles, complex interactions and therapeutic potential. Front Immunol 2024; 15:1369236. [PMID: 38545100 PMCID: PMC10965779 DOI: 10.3389/fimmu.2024.1369236] [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: 01/11/2024] [Accepted: 02/26/2024] [Indexed: 04/17/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells play diverse roles in cancer, infectious diseases, and immunotherapy. This review explores their intricate involvement in cancer, from early detection to their dual functions in promoting inflammation and mediating anti-tumor responses. Within the solid tumor microenvironment (TME), MAIT cells can acquire an 'exhausted' state and secrete tumor-promoting cytokines. On the other hand, MAIT cells are highly cytotoxic, and there is evidence that they may have an anti-tumor immune response. The frequency of MAIT cells and their subsets has also been shown to have prognostic value in several cancer types. Recent innovative approaches, such as programming MAIT cells with chimeric antigen receptors (CARs), provide a novel and exciting approach to utilizing these cells in cell-based cancer immunotherapy. Because MAIT cells have a restricted T cell receptor (TCR) and recognize a common antigen, this also mitigates potential graft-versus-host disease (GVHD) and opens the possibility of using allogeneic MAIT cells as off-the-shelf cell therapies in cancer. Additionally, we outline the interactions of MAIT cells with the microbiome and their critical role in infectious diseases and how this may impact the tumor responses of these cells. Understanding these complex roles can lead to novel therapeutic strategies harnessing the targeting capabilities of MAIT cells.
Collapse
Affiliation(s)
- Mesut Yigit
- Human Immunology Laboratory, Acibadem University School of Medicine, Istanbul, Türkiye
| | - Omer Faruk Basoglu
- Human Immunology Laboratory, Acibadem University School of Medicine, Istanbul, Türkiye
| | - Derya Unutmaz
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| |
Collapse
|
10
|
Yin Y, Zeng A, Abuduwayiti A, Xu Z, Chen K, Wang C, Fang X, Wang J, Jiang G, Dai J. MAIT cells are associated with responsiveness to neoadjuvant immunotherapy in COPD-associated NSCLC. Cancer Med 2024; 13:e7112. [PMID: 38509769 PMCID: PMC10955227 DOI: 10.1002/cam4.7112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/29/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Patients with non-small cell lung cancer (NSCLC) and chronic obstructive pulmonary disease (COPD) experience worse clinical outcomes but respond better to immunotherapy than patients with NSCLC without COPD. Mucosal-associated invariant T (MAIT) cells, a versatile population of innate immune T lymphocytes, have a crucial function in the response to infection and tumors. This study investigated the distribution of MAIT cells in COPD-associated NSCLC and their involvement in the immune response. METHODS Flow cytometry, immunohistochemistry, and immunofluorescence were performed on tissue samples of patients with NSCLC, with or without COPD, treated with or without anti-programmed death 1 (PD1) immunotherapy. MAIT cells were stimulated with 5-OP-RU using a mouse subcutaneous tumor model. RESULTS Tumors contained significantly more MAIT cells than paraneoplastic tissues, and CD8+ MAIT cells accounted for more than 90% of these cells. Patients with NSCLC and COPD had higher CD8+ MAIT cell counts than those with NSCLC without COPD. Additionally, patients with NSCLC and COPD displayed reduced expression of the activation marker, CD69, and functional markers, granzyme B (GZMB) and interferon γ (IFNγ), and higher expression of the immune exhaustion marker, PD1. Among patients who received immunotherapy, the proportion with a complete or partial response was higher in those with COPD than in those without COPD. In patients with NSCLC and COPD, the major pathologic response (MPR) group had higher MAIT levels than those in the no major pathologic response (NPR) group. In the mouse subcutaneous tumor model stimulation of MAIT cells using 5-OP-RU enhanced the antitumor effects of anti-PD1. CONCLUSIONS In patients with NSCLC and COPD, response to immunotherapy is associated with accumulation of CD8+ MAIT cells showing immune exhaustion. These findings may contribute to innovative approaches for immunotherapy targeting CD8+ MAIT cells.
Collapse
Affiliation(s)
- Yanze Yin
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Ao Zeng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Abudumijiti Abuduwayiti
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Zhilong Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Keyi Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Chao Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xinyun Fang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Jiarui Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Gening Jiang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Jie Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of MedicineTongji UniversityShanghaiChina
| |
Collapse
|
11
|
Sundström P, Dutta N, Rodin W, Hallqvist A, Raghavan S, Quiding Järbrink M. Immune checkpoint blockade improves the activation and function of circulating mucosal-associated invariant T (MAIT) cells in patients with non-small cell lung cancer. Oncoimmunology 2024; 13:2312631. [PMID: 38343750 PMCID: PMC10854269 DOI: 10.1080/2162402x.2024.2312631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/28/2024] [Indexed: 02/15/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells constitute one of the most numerous unconventional T cell subsets, and are characterized by rapid release of Th1- and Th17-associated cytokines and increased cytotoxic functions following activation. MAIT cells accumulate in tumor tissue but show an exhausted phenotype. Here, we investigated if immune checkpoint blockade (ICB) with antibodies to PD-1 or PD-L1 affects the function of circulating MAIT cells from non-small cell lung cancer patients. ICB increased the proliferation and co-expression of the activation markers HLA-DR and CD38 on MAIT cells in most patients after the first treatment cycle, irrespective of treatment outcome. Furthermore, production of cytokines, especially TNF and IL-2, also increased after treatment, as did MAIT cell polyfunctionality. These results indicate that MAIT cells respond to ICB, and that MAIT cell reinvigoration may contribute to tumor regression in patients undergoing immune checkpoint therapy.
Collapse
Affiliation(s)
- Patrik Sundström
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Nikita Dutta
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - William Rodin
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Andreas Hallqvist
- Department of Oncology, Institute of Clinical Sciences, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Sukanya Raghavan
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Marianne Quiding Järbrink
- Department of Microbiology and Immunology, Institute of Biomedicine, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
12
|
Katsnelson EN, Spengler A, Domenico J, Couts KL, Loh L, Gapin L, McCarter MD, Tobin RP. Dysfunctional states of unconventional T-cell subsets in cancer. J Leukoc Biol 2024; 115:36-46. [PMID: 37837379 PMCID: PMC10843843 DOI: 10.1093/jleuko/qiad129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/16/2023] Open
Abstract
Unconventional T cells represent a promising therapeutic agent to overcome the current limitations of immunotherapies due to their universal T-cell receptors, ability to respond directly to cytokine stimulation, and capacity to recruit and modulate conventional immune cells in the tumor microenvironment. Like conventional T cells, unconventional T cells can enter a dysfunctional state, and the functional differences associated with this state may provide insight into the discrepancies observed in their role in antitumor immunity in various cancers. The exhaustive signature of unconventional T cells differs from conventional αβ T cells, and understanding the differences in the mechanisms underlying exhaustive differentiation in these cell types may aid in the discovery of new treatments to improve sustained antitumor responses. Ongoing clinical trials investigating therapies that leverage unconventional T-cell populations have shown success in treating hematologic malignancies and reducing the immunosuppressive tumor environment. However, several hurdles remain to extend these promising results into solid tumors. Here we discuss the current knowledge on unconventional T-cell function/dysfunction and consider how the incorporation of therapies that modulate unconventional T-cell exhaustion may aid in overcoming the current limitations of immunotherapy. Additionally, we discuss how components of the tumor microenvironment alter the functions of unconventional T cells and how these changes can affect tumor infiltration by lymphocytes and alter conventional T-cell responses.
Collapse
Affiliation(s)
- Elizabeth N Katsnelson
- Department of Surgery, Division of Surgical Oncology, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, United States
| | - Andrea Spengler
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, United States
| | - Joanne Domenico
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, United States
| | - Kasey L Couts
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, United States
| | - Liyen Loh
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, United States
| | - Laurent Gapin
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, United States
| | - Martin D McCarter
- Department of Surgery, Division of Surgical Oncology, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, United States
| | - Richard P Tobin
- Department of Surgery, Division of Surgical Oncology, University of Colorado Anschutz Medical Campus, 12800 E 19th Ave, Aurora, CO 80045, United States
| |
Collapse
|
13
|
Guo H, Liu Y, Li X, Wang H, Mao D, Wei L, Ye X, Qu D, Huo J, Chen Y. Magnetic Metal-Organic Framework-Based Nanoplatform with Platelet Membrane Coating as a Synergistic Programmed Cell Death Protein 1 Inhibitor against Hepatocellular Carcinoma. ACS NANO 2023; 17:23829-23849. [PMID: 37991391 PMCID: PMC10722610 DOI: 10.1021/acsnano.3c07885] [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: 08/22/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/23/2023]
Abstract
Programmed cell death protein 1 (PD-1) inhibitors are the most common immune-checkpoint inhibitors and considered promising drugs for hepatocellular carcinoma (HCC). However, in clinical settings, they have a low objective response rate (15%-20%) for patients with HCC; this is because of the insufficient level and activity of tumor-infiltrating T lymphocytes (TILs). The combined administration of oxymatrine (Om) and astragaloside IV (As) can increase the levels of TILs by inhibiting the activation of cancer-associated fibroblasts (CAFs) and improve the activity of TILs by enhancing their mitochondrial function. In the present study, we constructed a magnetic metal-organic framework (MOF)-based nanoplatform with platelet membrane (Pm) coating (PmMN@Om&As) to simultaneously deliver Om and As into the HCC microenvironment. We observed that PmMN@Om&As exhibited a high total drug-loading capacity (33.77 wt %) and good immune escape. Furthermore, it can target HCC tissues in a magnetic field and exert long-lasting effects. The HCC microenvironment accelerated the disintegration of PmMN@Om&As and the release of Om&As, thereby increasing the level and activity of TILs by regulating CAFs and the mitochondrial function of TILs. In addition, the carrier could synergize with Om&As by enhancing the oxygen consumption rate and proton efflux rate of TILs, thereby upregulating the mitochondrial function of TILs. Combination therapy with PmMN@Om&As and α-PD-1 resulted in a tumor suppression rate of 84.15% and prolonged the survival time of mice. Our study provides a promising approach to improving the antitumor effect of immunotherapy in HCC.
Collapse
Affiliation(s)
- Hong Guo
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component
of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Yuping Liu
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component
of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
- Jiangsu
Clinical Innovation Center of Digestive Cancer of Traditional Chinese
Medicine, Nanjing 210028, China
| | - Xia Li
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component
of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Hong Wang
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component
of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Dengxuan Mao
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component
of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Liangyin Wei
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component
of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Xietao Ye
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component
of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Ding Qu
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Multi-component
of Traditional Chinese Medicine and Microecology Researh Center, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210028, China
| | - Jiege Huo
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu
Clinical Innovation Center of Digestive Cancer of Traditional Chinese
Medicine, Nanjing 210028, China
| | - Yan Chen
- Affiliated
Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210028, China
- Jiangsu
Clinical Innovation Center of Digestive Cancer of Traditional Chinese
Medicine, Nanjing 210028, China
| |
Collapse
|
14
|
Peng Q, Huang R, Wang H, Xiao H, Wang Y, Zhai Z, Wang Z. Immune characteristics and prognostic implications of mucosal-associated invariant T cells in acute myeloid leukemia. Cancer Immunol Immunother 2023; 72:4399-4414. [PMID: 37932426 PMCID: PMC10991463 DOI: 10.1007/s00262-023-03574-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
Increasing evidence suggests that mucosal-associated invariant T cells (MAITs) play a crucial role in anti-tumor responses against various cancers. In this study, we investigated the immune characteristics of MAIT cells in patients with acute myeloid leukemia (AML). Using multi-parameter flow cytometry, we performed phenotypic and functional analysis of MAITs in peripheral blood or bone marrow samples collected from 131 patients with AML including 99 newly diagnosed, 18 remission, and 14 relapsed cases, as well as 69 healthy controls. We found that MAITs exhibit signs of aging and exhaustion, particularly in CD8+ MAITs subset, at newly diagnosis. MAITs exhibit an effector memory or terminally differentiated phenotype. Frequency and number of MAITs reflect AML cell genetic features, tumor burden, disease status, and treatment responsiveness. Moreover, MAITs exhibit a highly activated or even exhausted state, as indicated by upregulation of PD-1. Furthermore, impaired production of Th1-type cytokines and increased secretion of Th17-type cytokines, granzyme B, and perforin were observed in MAITs from AML patients. Additionally, MAITs shifted toward producing cytokines that promote tumor progression, such as IL-8. Lower frequency of MAITs was associated with poorer overall survival (OS), and multivariate analysis revealed that MAITs frequency < 2.12% was an independent prognostic factor affecting OS. Collectively, our findings suggest that MAITs may play a role in immune deficiency in AML, emphasizing their potential importance in AML pathogenesis and treatment. These discoveries provide a theoretical basis for the development of novel immunotherapeutic strategies in AML.
Collapse
Affiliation(s)
- Qian Peng
- Hematology Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China
- Hematologic Diseases Research Center of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China
| | - Renhua Huang
- Hematology Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China
- Hematologic Diseases Research Center of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China
| | - Huiping Wang
- Hematology Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China
- Hematologic Diseases Research Center of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China
| | - Hao Xiao
- Hematology Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China
- Hematologic Diseases Research Center of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China
| | - Yiping Wang
- Centre for Transplantation and Renal Research, Westmead Millennium Institute, The University of Sydney, Sydney, Australia
| | - Zhimin Zhai
- Hematology Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China.
- Hematologic Diseases Research Center of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China.
| | - Zhitao Wang
- Hematology Department, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China.
- Hematologic Diseases Research Center of Anhui Medical University, Hefei, 230601, Anhui Province, People's Republic of China.
| |
Collapse
|