1
|
Liao KL, Watt KD. Adaptive Immunity Determines the Cancer Treatment Outcome of Oncolytic Virus and Anti-PD-1. Bull Math Biol 2025; 87:36. [PMID: 39878909 DOI: 10.1007/s11538-025-01413-5] [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: 08/27/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
The immune checkpoint inhibitor, anti-programmed death protein-1 (anti-PD-1), enhances adaptive immunity to kill tumor cells, and the oncolytic virus (OV) triggers innate immunity to clear the infected tumor cells. We create a mathematical model to investigate how the interaction between adaptive and innate immunities under OV and anti-PD-1 affects tumor reduction. For different immunity strength, we create the corresponding virtual baseline patients and cohort patients to decipher the major factors determining the treatment outcome. Global sensitivity analysis indicates that adaptive immunity has more control on the treatment outcome than innate immunity, and whether anti-PD-1 cancels out the OV treatment efficacy depends on the OV dosage and the balance between clearance of infected tumor cells and OV by T cells. The optimal OV infection rate and dosage suggest that OV treatment is more sensitive to adaptive immunity than innate immunity. Our model prediction also indicates that tumor reduction is more sensitive to anti-PD-1 efficacy as adaptive immunity becomes stronger, and anti-PD-1 trends to cancel out the OV treatment efficacy as innate immunity becomes stronger. Based on these results, the recommended treatment protocol for patients with different immunity strength can be determined.
Collapse
Affiliation(s)
- Kang-Ling Liao
- Department of Mathematics, University of Manitoba, 340 UMSU University Centre, Winnipeg, MB, R3T 2N2, Canada.
| | - Kenton D Watt
- Department of Mathematics, University of Manitoba, 340 UMSU University Centre, Winnipeg, MB, R3T 2N2, Canada
| |
Collapse
|
2
|
Mathiesen H, Juul-Madsen K, Tramm T, Vorup-Jensen T, Møller HJ, Etzerodt A, Andersen MN. Prognostic value of CD163 + macrophages in solid tumor malignancies: A scoping review. Immunol Lett 2025; 272:106970. [PMID: 39778658 DOI: 10.1016/j.imlet.2025.106970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025]
Abstract
Tumor-associated macrophages (TAMs) play crucial roles in development and progression of malignant diseases. Notably, CD163+ TAMs likely perform specific pro-tumorigenic functions, suggesting that this subset may serve as both prognostic biomarkers and targets for future anti-cancer therapy. We conducted a scoping review to map the current knowledge on the prognostic role of CD163+ TAMs in the five most lethal cancers worldwide: Lung, colorectal, gastric, liver, and breast cancer. For all cancer types, most studies showed that high tumoral presence of CD163+ cells was associated with poor patient outcome, and this association was more frequently observed when CD163+ cells were measured at the tumor periphery compared to more central parts of the tumor. These results support that CD163+ TAMs represent a biomarker of poor patient outcome across a variety of solid tumors, and highlight the relevance of further investigations of CD163+ TAMs as targets of future immunotherapies.
Collapse
Affiliation(s)
- Henriette Mathiesen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Kristian Juul-Madsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Trine Tramm
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Holger Jon Møller
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Etzerodt
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Morten Nørgaard Andersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Department of Hematology, Aarhus University Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark.
| |
Collapse
|
3
|
Zhao Y, Cheng HE, Wang J, Zang Y, Liu Z, Sun Y, Sun Y. Disulfidptosis-related lncRNA signature to assess the immune microenvironment and drug sensitivity in acute myeloid leukemia. Sci Rep 2024; 14:32015. [PMID: 39738722 DOI: 10.1038/s41598-024-83560-8] [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: 07/29/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025] Open
Abstract
Acute myeloid leukemia (AML) represents a hematological malignancy that arises from the abnormal proliferation of progenitor cells or myeloid hematopoietic stem. The current standard treatments for AML include chemotherapy and hematopoietic stem cell transplantation. However, chemotherapy suffers from high toxicity and a shortage of hematopoietic stem cell donors, which significantly shortens patient survival. A new type of cell death, disulfidptosis, has shown potential in medicine. However, its specific biological mechanism of action in AML is currently unclear. This research developed a prognostic model of disulfidptosis-related long non-coding RNAs (DRLs) based on 132 AML patients with GDC TCGA Acute myeloid leukemia (LAML). In this model, eight DRLs: AL049835.1, EXOC3-AS1, AC009237.14, LINC00944, AP002761.4, LINC00926, AC010247.2, and AC099811.5 were included. Patients with high-risk AML evaluated based on the model had shorter survival, significant infiltration of monocytes and M2 macrophages, and elevated transcriptional levels of immune checkpoint genes. In addition, AML was classified into three subtypes according to the model, and patients in different subtypes showed different overall survival (OS) and drug sensitivity. Overall, we formulated a pioneering prognostic model utilizing DRLs, achieving precise AML outcome predictions. The correlations between the DRL prognostic models and the AML immune microenvironment, drug sensitivity, and tumor subtype were explored. In addition, further studies on the molecular mechanisms of key biomarkers, such as LINC00944 and LINC00926, will greatly contribute to our understanding of AML pathogenesis and drug resistance mechanisms in the future.
Collapse
Affiliation(s)
- Yuying Zhao
- Department of Laboratory Medicine, School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China
| | - Hai-En Cheng
- Department of Laboratory Medicine, School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China
| | - Jingfei Wang
- Department of Laboratory Medicine, School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China
| | - Yunke Zang
- Department of Laboratory Medicine, School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China
| | - Zhijun Liu
- School of Basic Medicine, Shandong Second Medical University, Weifang, 261053, People's Republic of China.
| | - Yanhua Sun
- Department of Hematology, Weifang People's Hospital, Weifang, 261000, People's Republic of China.
| | - Yanli Sun
- Department of Laboratory Medicine, School of Medical Laboratory, Shandong Second Medical University, Weifang, 261053, People's Republic of China.
| |
Collapse
|
4
|
Wang M, Li S, Li R, Ning F, Tian L. Efficacy and Mechanism of Combining Radiotherapy and Immunotherapy in Stage IV Non-Small Cell Lung Cancer. Curr Treat Options Oncol 2024; 25:1605-1614. [PMID: 39625619 PMCID: PMC11638397 DOI: 10.1007/s11864-024-01260-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] [Accepted: 08/26/2024] [Indexed: 12/13/2024]
Abstract
OPINION STATEMENT Lung cancer is the leading cause of cancer-related deaths worldwide, with about 85% of patients being diagnosed as non-small cell lung cancer (NSCLC); and most presenting with stage IV disease initially. With the continuous advancement of treatment strategies of oncology, immunotherapy with/without chemo-immunotherapy has become the first-line treatment for patients with stage IV NSCLC. However, a proportion of patients still develop resistance to the treatment regimen and experience local progression, and primary lung lesion progression is the main progression pattern of stage IV NSCLC. Preclinical and clinical studies have demonstrated the potential of radiotherapy in anti-tumor treatment and suggest that administering local radiotherapy prior to cancer progression can prolong survival. Therefore, we consider whether adding local radiotherapy before the progression of a pulmonary lesion in stage IV NSCLC patients receiving chemo-immunotherapy would be beneficial. The present review aims to explore the efficacy and safety of combining radiotherapy with immunotherapy in the treatment of stage IV NSCLC, delving into the intricacies of their underlying mechanism.
Collapse
Affiliation(s)
- Mingyue Wang
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China
| | - Shuo Li
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China
| | - Runyu Li
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China
| | - Fangling Ning
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China
| | - Lijun Tian
- The Department of Oncology, Binzhou Medical University Hospital, Binzhou City, Shandong Province, China.
| |
Collapse
|
5
|
Choi JY, Seok HJ, Lee DH, Kwon J, Shin US, Shin I, Bae IH. miR-1226-5p is involved in radioresistance of colorectal cancer by activating M2 macrophages through suppressing IRF1. J Transl Med 2024; 22:980. [PMID: 39472937 PMCID: PMC11523791 DOI: 10.1186/s12967-024-05797-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Although the representative treatment for colorectal cancer (CRC) is radiotherapy, cancer cells survive due to inherent radioresistance or resistance acquired after radiation treatment, accelerating tumor malignancy and causing local recurrence and metastasis. However, the detailed mechanisms of malignancy induced after radiotherapy are not well understood. To develop more effective and improved radiotherapy and diagnostic methods, it is necessary to clearly identify the mechanisms of radioresistance and discover related biomarkers. METHODS To analyze the expression pattern of miRNAs in radioresistant CRC, sequence analysis was performed in radioresistant HCT116 cells using Gene Expression Omnibus, and then miR-1226-5p, which had the highest expression in resistant cells compared to parental cells, was selected. To confirm the effect of miR-1226-5 on tumorigenicity, Western blot, qRT-PCR, transwell migration, and invasion assays were performed to confirm the expression of EMT factors, cell mobility and invasiveness. Additionally, the tumorigenic ability of miR-1226-5p was confirmed in organoids derived from colorectal cancer patients. In CRC cells, IRF1, a target gene of miR-1226-5p, and circSLC43A1, which acts as a sponge for miR-1226-5p, were discovered and the mechanism was analyzed by confirming the tumorigenic phenotype. To analyze the effect of tumor-derived miR-1226-5p on macrophages, the expression of M2 marker in co-cultured cells and CRC patient tissues were confirmed by qRT-PCR and immunohistochemical (IHC) staining analyses. RESULTS This study found that overexpressed miR-1226-5p in radioresistant CRC dramatically promoted epithelial-mesenchymal transition (EMT), migration, invasion, and tumor growth by suppressing the expression of its target gene, IRF1. Additionally, we discovered circSLC43A1, a factor that acts as a sponge for miR-1226-5p and suppresses its expression, and verified that EMT, migration, invasion, and tumor growth are suppressed by circSLC43A1 in radioresistant CRC cells. Resistant CRC cells-derived miR-1226-5p was transferred to macrophages and contributed to tumorigenicity by inducing M2 polarization and secretion of TGF-β. CONCLUSIONS This study showed that the circSLC43A1/miR-1226-5p/IRF1 axis is involved in radioresistance and cancer aggressiveness in CRC. It was suggested that the discovered signaling factors could be used as potential biomarkers for diagnosis and treatment of radioresistant CRC.
Collapse
Affiliation(s)
- Jae Yeon Choi
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Ro, Nowon-Gu, Seoul, 01812, Republic of Korea
- Department of Life Science, Hanyang University, Seoul, Republic of Korea
| | - Hyun Jeong Seok
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Ro, Nowon-Gu, Seoul, 01812, Republic of Korea
| | - Dong Hyeon Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Ro, Nowon-Gu, Seoul, 01812, Republic of Korea
| | - Junhye Kwon
- Medical Sciences Substantiation Center, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Ui Sup Shin
- Department of Surgery, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Republic of Korea
| | - Incheol Shin
- Department of Life Science, Hanyang University, Seoul, Republic of Korea
| | - In Hwa Bae
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-Ro, Nowon-Gu, Seoul, 01812, Republic of Korea.
| |
Collapse
|
6
|
Peng J, Hu F, Mao X, Niu Y, Ma M, Jiang L. Evaluation of the prognostic value of the new 9th edition Tumor-Node-Metastases (TNM) staging system for epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma patients with bone metastases. BMC Pulm Med 2024; 24:508. [PMID: 39394157 PMCID: PMC11468161 DOI: 10.1186/s12890-024-03331-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 10/07/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND There are some changes in the new 9th edition Tumor-Node-Metastases (TNM) staging system for lung cancer, including subdividing M1c into M1c1 and M1c2 stage. The aim of this study was to assess the prognostic performance of the updated classification system and try to provide some real-world application data among advanced lung adenocarcinoma patients with bone metastases. METHODS Advanced lung adenocarcinoma patients in M1c stage with bone metastases who receiving first-line first-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) and T790M-guided osimertinib as the second-line therapy were retrospectively screened from December 2016 to December 2021. A total of 126 patients were enrolled in this study. 62 patients and 64 patients were subdivided into M1c1 and M1c2 groups according to the 9th edition of TNM staging system.The first-line real-world progression-free survival (1LrwPFS), the second-line real-world progression-free survival (2LrwPFS), post-progression survival (PPS) and real-world overall survival (rwOS) were analyzed. RESULTS The overall median rwOS was 40.1 months (95% CI 35.996-44.204). 1LrwPFS was 13.9 months (95% CI 12.653-15.147) and 2LrwPFS was 14.5 months (95% CI 11.665-17.335) for all patients.Patients in M1c2 stage was inferior to M1c1 stage patients in rwOS (35.2 months vs. 42.9 months, HR = 0.512, P = 0.005). 2LrwPFS was moderately correlated with rwOS (r = 0.621, R2 = 0.568, P = 0.000). Multivariate analysis showed performance status (PS) score ≥ 2 and TP53 alteration positive were independent prognostic factors of worse rwOS. CONCLUSIONS More refined stratification of M1c according to the 9th edition of TNM staging system is conducive to the judgment of prognosis and the implementation of precision medicine for patients.
Collapse
Affiliation(s)
- Jin Peng
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 Huaihai West Road, Xuhui District, Shanghai, 200030, PR China
| | - Fang Hu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 Huaihai West Road, Xuhui District, Shanghai, 200030, PR China
| | - Xiaowei Mao
- Department of Respiratory and Critical Care Medicine, Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Yanjie Niu
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 Huaihai West Road, Xuhui District, Shanghai, 200030, PR China
| | - Meili Ma
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 Huaihai West Road, Xuhui District, Shanghai, 200030, PR China
| | - Liyan Jiang
- Department of Respiratory and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, 241 Huaihai West Road, Xuhui District, Shanghai, 200030, PR China.
| |
Collapse
|
7
|
Coullomb A, Monsarrat P, Pancaldi V. mosna reveals different types of cellular interactions predictive of response to immunotherapies and survival in cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.16.532947. [PMID: 36993595 PMCID: PMC10055099 DOI: 10.1101/2023.03.16.532947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Spatially resolved omics enable the discovery of tissue organization of biological or clinical importance. Despite the existence of several methods, performing a rational analysis including multiple algorithms while integrating different conditions such as clinical data is still not trivial. To make such investigations more accessible, we developed mosna, a Python package to analyze spatial omics data with respect to clinical or biological data and to gain insight on cell interaction patterns or tissue architecture of biological relevance. mosna is compatible with all spatial omics methods, it leverages tysserand to build accurate spatial networks, and is compatible with Squidpy. It proposes an analysis pipeline, in which increasingly complex features computed at each step can be explored in integration with clinical data, either with easy-to-use descriptive statistics and data visualization, or by seamlessly training machine learning models and identifying variables with the most predictive power. mosna can take as input any dataset produced by spatial omics methods, including sub-cellular resolved transcriptomics (MERFISH, seqFISH) and proteomics (CODEX, MIBI-TOF, low-plex immuno-fluorescence), as well as spot-based spatial transcriptomics (10x Visium). Integration with experimental metadata or clinical data is adapted to binary conditions, such as biological treatments or response status of patients, and to survival data. We demonstrate the proposed analysis pipeline on two spatially resolved proteomic datasets containing either binary response to immunotherapy or survival data. mosna identifies features describing cellular composition and spatial distribution that can provide biological insight regarding factors that affect response to immunotherapies or survival.
Collapse
Affiliation(s)
- Alexis Coullomb
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVT, Toulouse, France
| | - Paul Monsarrat
- RESTORE Research Center, Université de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVT, Toulouse, France
- Oral Medicine Department and Hospital of Toulouse - Toulouse Institute of Oral Medicine and Science, CHU de Toulouse, Toulouse, France
- Artificial and Natural Intelligence Toulouse Institute ANITI, Toulouse, France
| | - Vera Pancaldi
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| |
Collapse
|
8
|
Aktar T, Modak S, Majumder D, Maiti D. A detailed insight into macrophages' role in shaping lung carcinogenesis. Life Sci 2024; 352:122896. [PMID: 38972632 DOI: 10.1016/j.lfs.2024.122896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
Despite significant advancements in cancer treatment in recent decades, the high mortality rate associated with lung cancer remains a significant concern. The development and proper execution of new targeted therapies needs more deep knowledge regarding the lung cancer associated tumour microenvironment. One of the key component of that tumour microenvironment is the lung resident macrophages. Although in normal physiological condition the lung resident macrophages are believed to maintain lung homeostasis, but they may also initiate a vicious inflammatory response in abnormal conditions which is linked to lung cancer development. Depending on the activation pathway, the lung resident macrophages are either of M1 or M2 sub-type. The M1 and M2 sub-types differ significantly in various prospectuses, from phenotypic markers to metabolic pathways. In addition to this generalized classification, the recent advancement of the multiomics technology is able to identify some other sub-types of lung resident macrophages. Researchers have also observed that these different sub-types can manipulate the pathogenesis of lung carcinogenesis in a context dependent manner and can either promote or inhibit the development of lung carcinogenesis upon receiving proper activation. As proper knowledge about the role played by the lung resident macrophages' in shaping the lung carcinogenesis is limited, so the main purpose of this review is to bring all the available information under the same roof. We also elaborated the different mechanisms involved in maintenance of the plasticity of M1/M2 sub-type, as this plasticity can be a good target for lung cancer treatment.
Collapse
Affiliation(s)
- Tamanna Aktar
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Snehashish Modak
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Debabrata Majumder
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India; Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Debasish Maiti
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India.
| |
Collapse
|
9
|
Ding Y, Yuan X, Wang Y, Yan J. CASQ2 alleviates lung cancer by inhibiting M2 tumor-associated macrophage polarization and JAK/STAT pathway. J Biochem Mol Toxicol 2024; 38:e23801. [PMID: 39132772 DOI: 10.1002/jbt.23801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Lung cancer (LC) is a major inducer of cancer-related death. We aim to reveal the effect of Calsequestrin2 (CASQ2) on macrophage polarization and Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway in LC. Hub genes were determined from protein-protein interaction networks based on GSE21933 and GSE1987 data sets using bioinformatic analysis. Expression of hub genes was verified by real-time quantitative polymerase chain reaction (RT-qPCR). Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine, wound-healing, colony formation, and transwell assays were performed to assess the impact of CASQ2 on LC cells. A xenograft mouse model was evaluated using hematoxylin-eosin, immunohistochemistry, and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling staining to investigate the effect of CASQ2 on LC. The role of CASQ2 in regulating macrophage polarization and JAK/STAT pathway was evaluated by western blot andRT-qPCR. We screened out 155 common differentially expressed genes in GSE21933 and GSE1987 data sets. Myomesin-2, tyrosine kinase, sex determining region Y-box 2, platelet and endothelial cell adhesion molecule 1, matrix metallopeptidase 9, claudin-5, caveolin-1, CASQ2, recombinant ATPase, Ca++ transporting, cardiac muscle, slow twitch 2 (ATP2A2), and ankyrin repeat domain 1 were identified as the hub genes with high prediction value. CASQ2 was selected as a pivotal regulator of LC. In vitro experiments and xenograft models revealed that CASQ2 overexpression suppressed proliferation, colony formation, migration, invasion of LC cells, and tumor growth in vivo. Additionally, overexpression of CASQ2 promoted the expression of M1 macrophage markers (cluster of differentiation 80 [CD80], interleukin [IL]-12, inducible nitric oxide synthase [iNOS]), while decreasing the expression of M2 macrophage markers (CD163, IL-10, Arg1) in tumor-associated macrophages and xenograft tissues. Finally, we found that overexpression of CASQ2 inhibited JAK/STAT pathway. CASQ2 is a novel biomarker, which can alleviate LC via inhibiting M2 tumor-associated macrophage polarization and JAK/STAT pathway.
Collapse
Affiliation(s)
- Yan Ding
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Xiaoliang Yuan
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Ying Wang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| | - Jun Yan
- Department of Oncology, First Affiliated Hospital of Gannan Medical University, Ganzhou City, Jiangxi Province, China
| |
Collapse
|
10
|
Sun X, He W, Lin B, Huang W, Ye D. Defining three ferroptosis-based molecular subtypes and developing a prognostic risk model for high-grade serous ovarian cancer. Aging (Albany NY) 2024; 16:9106-9126. [PMID: 38795391 PMCID: PMC11164503 DOI: 10.18632/aging.205857] [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: 10/27/2023] [Accepted: 04/08/2024] [Indexed: 05/27/2024]
Abstract
BACKGROUND As a newly defined regulated cell death, ferroptosis is a potential biomarker in ovarian cancer (OV). However, its underlying mechanism in tumor microenvironment (TME) and clinical prediction significance in OV remained to be elucidated. METHODS The transcriptome data of high-grade serous OV from The Cancer Genome Atlas (TCGA) database were downloaded. Molecular subtypes were classified based on ferroptosis-correlated genes from the FerrDb database by performing consensus clustering analysis. The associations between the subtypes and clinicopathologic characteristics, mutation, regulatory pathways and immune landscape were assessed. A ferroptosis-related prognostic model was constructed and verified using International Cancer Genome Consortium (ICGC) cohort and GSE70769. RESULTS Three molecular subtypes of OV were defined. Patients in subtype C3 tended to have the most favorable prognosis, while subtype C1 showing more mesenchymal cells, increased immune infiltration of Macrophages_M2, lower tumor purity, and epithelial-to-mesenchymal transition (EMT) features had the poorest prognosis. A ferroptosis-related risk model was constructed using 8 genes (PDP1, FCGBP, EPHA4, GAS1, SLC7A11, BLOC1S1, SPOCK2, and CXCL9) and manifested a strong prediction performance. High-risk patients had enriched EMT pathways, more Macrophages_M2, less plasma cells and CD8 cell infiltration, greater tendency of immune escape and worse prognosis. The risk score has negatively correlated relation with LAG3, TIGIT, CTLA4, IDO1, CD27, ICOS, and IL2RB but positively correlated with PVR, CD276, and CD28. Moreover, low-risk patients were more sensitive to Cisplatin and Gefitinib, Gemcitabine. CONCLUSIONS Our results could improve the understanding of ferroptosis in OV, providing promising insights for the clinical targeted therapy for the cancer.
Collapse
Affiliation(s)
- Xiang Sun
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Wenbin He
- Department of Otolaryngology, Lanzhou University Second Hospital, Gansu 730030, China
| | - Baohua Lin
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Weiming Huang
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| | - Danping Ye
- Department of Obstetrics and Gynecology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, China
| |
Collapse
|
11
|
Pukhalskaia TV, Yurakova TR, Bogdanova DA, Demidov ON. Tumor-Associated Senescent Macrophages, Their Markers, and Their Role in Tumor Microenvironment. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:839-852. [PMID: 38880645 DOI: 10.1134/s0006297924050055] [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: 12/21/2023] [Revised: 04/27/2024] [Accepted: 04/27/2024] [Indexed: 06/18/2024]
Abstract
Tumor-associated macrophages (TAMs) are an important component of the tumor microenvironment (TME) and the most abundant population of immune cells infiltrating a tumor. TAMs can largely determine direction of anti-tumor immune response by promoting it or, conversely, contribute to formation of an immunosuppressive TME that allows tumors to evade immune control. Through interactions with tumor cells or other cells in the microenvironment and, as a result of action of anti-cancer therapy, macrophages can enter senescence. In this review, we have attempted to summarize information available in the literature on the role of senescent macrophages in tumors. With the recent development of senolytic therapeutic strategies aimed at removing senescent cells from an organism, it seems important to discuss functions of the senescent macrophages and potential role of the senolytic drugs in reprogramming TAMs to enhance anti-tumor immune response and improve efficacy of cancer treatment.
Collapse
Affiliation(s)
- Tamara V Pukhalskaia
- Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Taisiya R Yurakova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Daria A Bogdanova
- Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
| | - Oleg N Demidov
- Sirius University of Science and Technology, Federal Territory Sirius, 354340, Russia.
- Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russia
- INSERM UMR1231, Université de Bourgogne, Dijon, 21000, France
| |
Collapse
|
12
|
Ni Y, Li R, Shen X, Yi D, Ren Y, Wang F, Geng Y, You Q. Diaphorobacter nitroreducens synergize with oxaliplatin to reduce tumor burden in mice with lung adenocarcinoma. mSystems 2024; 9:e0132323. [PMID: 38483163 PMCID: PMC11019951 DOI: 10.1128/msystems.01323-23] [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: 12/07/2023] [Accepted: 02/29/2024] [Indexed: 04/17/2024] Open
Abstract
Lung adenocarcinoma (LADC) is the most common lung cancer and the leading cause of cancer-related deaths globally. Accumulating evidence suggests that the gut microbiota regulates the host response to chemotherapeutic drugs and can be targeted to reduce the toxicity of current chemotherapeutic agents. However, the effect of Diaphorobacter nitroreducens synergized with oxaliplatin on the gut microbiota and their impact on LADC have never been explored. This study aimed to evaluate the anti-cancer effects of D. nitroreducens, oxaliplatin, and their combined treatment on tumor growth in tumor-bearing mice. The composition of gut microbiota and the immune infiltration of tumors were evaluated by using 16S rRNA gene high-throughput sequencing and immunofluorescence, respectively. The inhibitory effect of the combination treatment with D. nitroreducens and oxaliplatin was significantly stronger than that of oxaliplatin alone in tumor-bearing mice. Furthermore, we observed that the combination treatment significantly increased the relative abundance of Lactobacillus and Akkermansia in the gut microbiota. Meanwhile, the combination treatment significantly increased the proportions of macrophage but decreased the proportion of regulatory T cells in the LADC tumor tissues of mice. These findings underscored the relationship between D. nitroreducens and the gut microbiota-immune cell-LADC axis, highlighting potential therapeutic avenues for LADC treatment. IMPORTANCE Oxaliplatin is widely used as an effective chemotherapeutic agent in cancer treatment, but its side effects and response rate still need to be improved. Conventional probiotics potentially benefit cancer chemotherapy by regulating gut microbiota and tumor immune infiltration. This study was novel in reporting a more significant inhibitory effect of Diaphorobacter nitroreducens on lung adenocarcinoma (LADC) cells compared with common traditional probiotics and validating its potential as an adjuvant therapy for LADC chemotherapy in mice. This study investigated the impact of D. nitroreducens combined with oxaliplatin on the gut microbiota and immune infiltration of tumors as a potential mechanism to improve anticancer effects.
Collapse
Affiliation(s)
- Yalan Ni
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Rui Li
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiaoyu Shen
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Deli Yi
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yilin Ren
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Fudong Wang
- Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Yan Geng
- School of Life Science and Health Engineering, Jiangnan University, Wuxi, China
| | - Qingjun You
- Department of Oncology, Affiliated Children’s Hospital of Jiangnan University, Wuxi, Jiangsu, China
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
13
|
Yu Z, Zou J, Xu F. Tumor-associated macrophages affect the treatment of lung cancer. Heliyon 2024; 10:e29332. [PMID: 38623256 PMCID: PMC11016713 DOI: 10.1016/j.heliyon.2024.e29332] [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: 12/26/2023] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/17/2024] Open
Abstract
As one of the most common malignant tumors in the world, lung cancer has limited benefits for patients despite its diverse treatment methods due to factors such as personalized medicine targeting histological type, immune checkpoint expression, and driver gene mutations. The high mortality rate of lung cancer is partly due to the immune-suppressive which limits the effectiveness of anti-cancer drugs and induces tumor cell resistance. The currently widely recognized TAM phenotypes include the anti-tumor M1 and pro-tumor M2 phenotypes. M2 macrophages promote the formation of an immune-suppressive microenvironment and hinder immune cell infiltration, thereby inhibiting activation of the anti-tumor immune system and aiding tumor cells in resisting treatment. Analyzing the relationship between different treatment methods and macrophages in the TME can help us better understand the impact of TAMs on lung cancer and confirm the feasibility of targeted TAM therapy. Targeting TAMs to reduce the M2/M1 ratio and reverse the immune-suppressive microenvironment can improve the clinical efficacy of conventional treatment methods and potentially open up more efficient combination treatment strategies, maximizing the benefit for lung cancer patients.
Collapse
Affiliation(s)
- Zhuchen Yu
- Clinical Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Juntao Zou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| | - Fei Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, China
| |
Collapse
|
14
|
ZHOU Y, REN D, BI H, YI B, ZHANG C, WANG H, SUN J. [Tumor-associated Macrophage:
Emerging Targets for Modulating the Tumor Microenvironment]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:231-240. [PMID: 38590197 PMCID: PMC11002190 DOI: 10.3779/j.issn.1009-3419.2024.102.13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Indexed: 04/10/2024]
Abstract
Tumor-associated macrophage (TAM) play a crucial role in the immune microenvironment of lung cancer. Through changes in their phenotype and phagocytic functions, TAM contribute to the initiation and progression of lung cancer. By promoting the formation of an immune-suppressive microenvironment and accelerating the growth of abnormal tumor vasculature, TAM facilitate the invasion and metastasis of lung cancer. Macrophages can polarize into different subtypes with distinct functions and characteristics in response to various stimuli, categorized as anti-tumor M1 and pro-tumor M2 types. In tumor tissues, TAM typically polarize into the alternatively activated M2 phenotype, exhibiting inhibitory effects on tumor immunity. This article reviews the role of anti-angiogenic drugs in modulating TAM phenotypes, highlighting their potential to reprogram M2-type TAM into an anti-tumor M1 phenotype. Additionally, the functional alterations of TAM play a significant role in anti-angiogenic therapy and immunotherapy strategies. In summary, the regulation of TAM polarization and function opens up new avenues for lung cancer treatment and may serve as a novel target for modulating the immune microenvironment of tumors.
.
Collapse
|
15
|
Zhang W, Zhou D, Song S, Hong X, Xu Y, Wu Y, Li S, Zeng S, Huang Y, Chen X, Liang Y, Guo S, Pan H, Li H. Prediction and verification of the prognostic biomarker SLC2A2 and its association with immune infiltration in gastric cancer. Oncol Lett 2024; 27:70. [PMID: 38192676 PMCID: PMC10773219 DOI: 10.3892/ol.2023.14203] [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: 07/01/2023] [Accepted: 11/15/2023] [Indexed: 01/10/2024] Open
Abstract
Gastric cancer (GC) is the fifth most common cause of cancer-associated deaths; however, its treatment options are limited. Despite clinical improvements, chemotherapy resistance and metastasis are major challenges in improving the prognosis and quality of life of patients with GC. Therefore, effective prognostic biomarkers and targets associated with immunological interventions need to be identified. Solute carrier family 2 member 2 (SLC2A2) may serve a role in tumor development and invasion. The present study aimed to evaluate SLC2A2 as a prospective prognostic marker and chemotherapeutic target for GC. SLC2A2 expression in several types of cancer and GC was analyzed using online databases, and the effects of SLC2A2 expression on survival prognosis in GC were investigated. Clinicopathological parameters were examined to explore the association between SLC2A2 expression and overall survival (OS). Associations between SLC2A2 expression and immune infiltration, immune checkpoints and IC50 were estimated using quantification of the tumor immune contexture from human RNA-seq data, the Tumor Immune Estimation Resource 2.0 database and the Genomics of Drug Sensitivity in Cancer database. Differential SLC2A2 expression and the predictive value were validated using the Human Protein Atlas, Gene Expression Omnibus, immunohistochemistry and reverse transcription-quantitative PCR. SLC2A2 expression was downregulated in most types of tumor but upregulated in GC. Functional enrichment analysis revealed an association between SLC2A2 expression and lipid metabolism and the tumor immune microenvironment. According to Gene Ontology term functional enrichment analysis, SLC2A2-related differentially expressed genes were enriched predominantly in 'chylomicron assembly', 'plasma lipoprotein particle assembly', 'high-density lipoprotein particle', 'chylomicron', 'triglyceride-rich plasma lipoprotein particle', 'very-low-density lipoprotein particle'. 'intermembrane lipid transfer activity', 'lipoprotein particle receptor binding', 'cholesterol transporter activity' and 'intermembrane cholesterol transfer activity'. In addition, 'cholesterol metabolism', and 'fat digestion and absorption' were significantly enriched in the Kyoto Encyclopedia of Genes and Genomes pathway analysis. Patients with GC with high SLC2A2 expression had higher levels of neutrophil and M2 macrophage infiltration and a significant inverse correlation was observed between SLC2A2 expression and MYC targets, tumor mutation burden, microsatellite instability and immune checkpoints. Furthermore, patients with high SLC2A2 expression had worse prognosis, including OS, disease-specific survival and progression-free interval. Multivariate regression analysis demonstrated that SLC2A2 could independently prognosticate GC and the nomogram model showed favorable performance for survival prediction. SLC2A2 may be a prospective prognostic marker for GC. The prediction model may improve the prognosis of patients with GC in clinical practice, and SLC2A2 may serve as a novel therapeutic target to provide immunotherapy plans for GC.
Collapse
Affiliation(s)
- Weijian Zhang
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Dishu Zhou
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Shuya Song
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Xinxin Hong
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Yifei Xu
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Yuqi Wu
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Shiting Li
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Sihui Zeng
- The Fourth Clinical Medical College, Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Yanzi Huang
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Xinbo Chen
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Yizhong Liang
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Shaoju Guo
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| | - Huafeng Pan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, P.R. China
| | - Haiwen Li
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong 518033, P.R. China
| |
Collapse
|
16
|
Takam Kamga P, Mayenga M, Sebane L, Costantini A, Julie C, Capron C, Parent F, Seferian A, Guettier C, Emile JF, Giroux Leprieur E. Colony stimulating factor-1 (CSF-1) signalling is predictive of response to immune checkpoint inhibitors in advanced non-small cell lung cancer. Lung Cancer 2024; 188:107447. [PMID: 38176297 DOI: 10.1016/j.lungcan.2023.107447] [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: 08/23/2023] [Revised: 11/13/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024]
Abstract
The identification of biomarkers related to treatment in patients with non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitors (ICIs) represents a significant challenge. The aim of this study was to determine the predictive value of macrophage-related markers assessed in plasma and tissue samples of patients with NSCLC undergoing ICI treatment. This bicentric study included a prospective cohort of 88 patients with advanced NSCLC who received first-line therapy with ICI (either as monotherapy or in combination with chemotherapy) or chemotherapy alone (CT). Samples were collected from the patients at baseline and during follow-up. Plasma levels of CSF-1 and IL-34 were measured using ELISA, while expression levels of the macrophage receptors CD163 and CSF-1-R were evaluated using immunohistochemistry on lung biopsies. At baseline, the median plasma CSF-1 expression was higher in patients who did not respond to immunotherapy compared to those who responded (8898 pg/mL vs. 14031 pg/mL, p = 0.0005). Importantly, high CSF-1 levels at the initial assessment were associated with disease progression regardless of the treatment received. Furthermore, high CSF-1 levels were associated with shorter progression-free survival (PFS) and overall survival (OS) in patients receiving ICI therapy, but not in those treated with chemotherapy. There was no correlation between IL-34, CSF-1R, CD163 and therapeutic response. We observed in vitro that the activation of lymphocytes mediated by pembrolizumab was hindered by the treatment of PBMC with recombinant CSF-1, suggesting that CSF-1 creates a systemic immunosuppressive state that interferes with ICI treatment. In conclusion, baseline CSF-1 levels represent a potential predictive marker to ICI treatment in NSCLC.
Collapse
Affiliation(s)
- Paul Takam Kamga
- Université Paris-Saclay, UVSQ, EA4340 BECCOH, Boulogne-Billancourt, France
| | - Marie Mayenga
- Université Paris-Saclay, UVSQ, EA4340 BECCOH, Boulogne-Billancourt, France
| | - Louise Sebane
- Université Paris-Saclay, UVSQ, EA4340 BECCOH, Boulogne-Billancourt, France
| | - Adrien Costantini
- Université Paris-Saclay, UVSQ, EA4340 BECCOH, Boulogne-Billancourt, France; APHP - Hôpital Ambroise Paré, Department of Respiratory Diseases and Thoracic Oncology, Boulogne-Billancourt, France
| | - Catherine Julie
- Université Paris-Saclay, UVSQ, EA4340 BECCOH, Boulogne-Billancourt, France; APHP - Hôpital Ambroise Paré, Department of Pathology, Boulogne-Billancourt, France
| | - Claude Capron
- Université Paris-Saclay, UVSQ, EA4340 BECCOH, Boulogne-Billancourt, France; APHP - Hôpital Ambroise Paré, Department of Hematology Immunology, Boulogne-Billancourt, France
| | - Florence Parent
- APHP - Hôpital Bicêtre, Department of Pulmonology, Kremlin-Bicêtre, France
| | - Andrei Seferian
- APHP - Hôpital Bicêtre, Department of Pulmonology, Kremlin-Bicêtre, France
| | - Catherine Guettier
- APHP - Hôpital Bicêtre, Department of Pathology, Kremlin-Bicêtre, France
| | - Jean-François Emile
- Université Paris-Saclay, UVSQ, EA4340 BECCOH, Boulogne-Billancourt, France; APHP - Hôpital Ambroise Paré, Department of Pathology, Boulogne-Billancourt, France
| | - Etienne Giroux Leprieur
- Université Paris-Saclay, UVSQ, EA4340 BECCOH, Boulogne-Billancourt, France; APHP - Hôpital Ambroise Paré, Department of Respiratory Diseases and Thoracic Oncology, Boulogne-Billancourt, France.
| |
Collapse
|
17
|
Xu X, Zhang X, Kou R, Liu Y, Chen S, Li Z, Jian Z, Wang Z. Prognosis and immunotherapy response prediction based on M2 macrophage-related genes in colon cancer. J Cancer Res Clin Oncol 2024; 150:31. [PMID: 38270646 PMCID: PMC10811099 DOI: 10.1007/s00432-023-05573-6] [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: 08/04/2023] [Accepted: 12/12/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND M2 macrophage were revealed to play a crucial role in immune evasion and immunotherapies. This study aims to explore the potential significance of M2 macrophage-related genes in colon adenocarcinoma (COAD) by analysizing the transcriptome data in a comprehensive way. METHODS We collected RNA-sequencing (RNA-seq) data of COAD from The Cancer Genome Atlas (TCGA) and Gene Expression Ominibus (GEO) databases. We calculated the immune infiltration scores of every sample using CIBERSORT algorithm. Through weighted gene co-expression network analysis (WGCNA), we picked out M2 macrophage-related genes. With these genes we screened out prognosis related genes which were utilized to construct a signature to assess the prognosis of patients. To extend the potential application of the signature, we also calculated the correlations with immune infiltration. Finally, we applied techniques such as quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunoblotting (Western Blotting) to validate the RNF32 gene in cellular in vitro assays. RESULTS Seven M2 macrophage-related genes signature was constructed, which was an excellent prognostic predictor in two independent groups. The high-risk group showed lower immune infiltration and poorer response to immunotherapies than those of the low-risk group. The cell vitro experiments showed that the expression level of RNF32 was upregulated in colon cancer cell lines compared with normal cell lines. Moreover, we found that RNF32 may promote the proliferation, migration and invasion of cancer cells in vitro by inhibiting apoptosis. CONCLUSION A novel M2 macrophage-related gene signature affects the prognosis and immune characteristics of colon cancer.
Collapse
Affiliation(s)
- Xiaochen Xu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, Guilin, 541001, China
| | - Xinwen Zhang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, Guilin, 541001, China
| | - Ruilong Kou
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, Guilin, 541001, China
| | - Yihao Liu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, Guilin, 541001, China
| | - Siqi Chen
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, Guilin, 541001, China
| | - Zuguo Li
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, Guilin, 541001, China
| | - Zhiyuan Jian
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, Guilin, 541001, China.
| | - Zhenran Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guilin Medical University, Guangxi Zhuang Autonomous Region, Guilin, 541001, China.
| |
Collapse
|
18
|
Wang J, Wu W, Yuan T, Wang L, Zang L, Liu Q, Wang L, Huo X, Huo B, Tang Y, Wang H, Zhao Z. Tumor-associated macrophages and PD-L1 in prostate cancer: a possible key to unlocking immunotherapy efficacy. Aging (Albany NY) 2024; 16:445-465. [PMID: 38189834 PMCID: PMC10817380 DOI: 10.18632/aging.205378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/15/2023] [Indexed: 01/09/2024]
Abstract
PURPOSE Prostate cancer (PCa) is often considered as a "cold" tumor with low responsiveness to immunotherapy. Recent evidence suggests the activation of specific immune cells, such as tumor-associated macrophages (TAMs), could potentially influence the efficacy of immunotherapy in PCa. However, the relationship between TAMs and PD-L1, a significant regulator in immunotherapy, within PCa remains unexplored. METHODS In this study, we assessed TAM infiltration and PD-L1 expression levels in a local cohort of 95 PCa tissue samples and two publicly available PCa datasets. We employed a combination of bioinformatics and experimental techniques, including gene set enrichment analysis, CIBERSORTx, tissue microarray, immunohistochemistry staining, and analysis of single-cell sequencing datasets, to provide a comprehensive understanding of the association between PD-L1 and TAMs in the PCa microenvironment. RESULTS The study showed that CD68+ TAMs and CD163+ TAMs (M2-TAMs) were more abundant in the tumor microenvironment than in non-cancerous surrounding tissues. The infiltration of CD163+ TAMs was significantly associated with the Gleason score and risk stratification of PCa. Importantly, elevated PD-L1 expression correlated significantly with high infiltration of CD163+ TAMs. Furthermore, patients displaying high levels of CD163+ TAMs and PD-L1 expression exhibited shorter times to biochemical recurrence-free survival. CONCLUSION Our study suggests that CD163+ TAMs are closely associated with PD-L1 expression and can act as a valuable prognostic indicator for PCa. The high infiltration of M2-TAMs, coupled with the overexpression of PD-L1, may contribute to immune escape mechanisms in PCa, thereby influencing disease prognosis.
Collapse
Affiliation(s)
- Jinhuan Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Wenqi Wu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Tian Yuan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Lili Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Li Zang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Qing Liu
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Lei Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xiaodong Huo
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Bin Huo
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Yong Tang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Haitao Wang
- Department of Oncology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Zhigang Zhao
- Department of Medical Oncology, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China
| |
Collapse
|
19
|
Esteban V, Javaloyes J, Martínez-López S, Sancho-Chust JN, Gálvez B, Chiner E, Ferrer C, Colom MF. Alveolar Macrophage Expression Differs According to Lung Cancer Subtype. Arch Bronconeumol 2024; 60:59-61. [PMID: 37993306 DOI: 10.1016/j.arbres.2023.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Affiliation(s)
- Violeta Esteban
- Department of Respiratory Medicine, San Juan de Alicante University Hospital, Alicante, Spain.
| | - Javier Javaloyes
- Department of Plant Production and Microbiology, Miguel Hernández University, San Juan de Alicante, Alicante, Spain
| | - Sebastián Martínez-López
- Department of Clinical Medicine, Miguel Hernández University, San Juan de Alicante, Alicante, Spain
| | | | - Beatriz Gálvez
- Department of Respiratory Medicine, Vinalopó University Hospital, Alicante, Spain
| | - Eusebi Chiner
- Department of Respiratory Medicine, San Juan de Alicante University Hospital, Alicante, Spain
| | - Consuelo Ferrer
- Department of Plant Production and Microbiology, Miguel Hernández University, San Juan de Alicante, Alicante, Spain; Institute for Sanitary and Biomedical Research of Alicante (ISABIAL), Alicante, Spain
| | - María Francisca Colom
- Department of Plant Production and Microbiology, Miguel Hernández University, San Juan de Alicante, Alicante, Spain; Institute for Sanitary and Biomedical Research of Alicante (ISABIAL), Alicante, Spain
| |
Collapse
|
20
|
Liu Y, Lu M, Liu F, Xu G, Feng C, Chen Y, Cai D, Sun H, Zeng Y, Xie J, Ma W, Gao X. Extracellular Vesicles Obtained From Lung Adenocarcinoma Cells Cultured Under Intermittent Hypoxia Induce M2 Macrophage Polarization via miR-20a-5p Delivery. Technol Cancer Res Treat 2024; 23:15330338231219415. [PMID: 38327167 PMCID: PMC10851739 DOI: 10.1177/15330338231219415] [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: 08/09/2023] [Revised: 11/07/2023] [Accepted: 10/10/2023] [Indexed: 02/09/2024] Open
Abstract
Conclusion: These findings indicate that EVs obtained from lung adenocarcinoma cells cultured under IH deliver miR-20a-5p to promote M2 macrophage polarization by targeting PTEN.
Collapse
Affiliation(s)
- Yuanling Liu
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Minzhen Lu
- Department of Pulmonary and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, China
| | - Feng Liu
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Gang Xu
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Congrui Feng
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yuluo Chen
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Danyan Cai
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Huake Sun
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yanjun Zeng
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Jian Xie
- Department of Pharmacy, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Wei Ma
- Department of Geriatric Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Xinglin Gao
- Department of Geriatric Respiratory Medicine, Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Khaled ML, Ren Y, Kundalia R, Alhaddad H, Chen Z, Wallace GC, Evernden B, Ospina OE, Hall M, Liu M, Darville LN, Izumi V, Chen YA, Pilon-Thomas S, Stewart PA, Koomen JM, Corallo SA, Jain MD, Robinson TJ, Locke FL, Forsyth PA, Smalley I. Branched-chain keto acids promote an immune-suppressive and neurodegenerative microenvironment in leptomeningeal disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.18.572239. [PMID: 38187773 PMCID: PMC10769272 DOI: 10.1101/2023.12.18.572239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Leptomeningeal disease (LMD) occurs when tumors seed into the leptomeningeal space and cerebrospinal fluid (CSF), leading to severe neurological deterioration and poor survival outcomes. We utilized comprehensive multi-omics analyses of CSF from patients with lymphoma LMD to demonstrate an immunosuppressive cellular microenvironment and identified dysregulations in proteins and lipids indicating neurodegenerative processes. Strikingly, we found a significant accumulation of toxic branched-chain keto acids (BCKA) in the CSF of patients with LMD. The BCKA accumulation was found to be a pan-cancer occurrence, evident in lymphoma, breast cancer, and melanoma LMD patients. Functionally, BCKA disrupted the viability and function of endogenous T lymphocytes, chimeric antigen receptor (CAR) T cells, neurons, and meningeal cells. Treatment of LMD mice with BCKA-reducing sodium phenylbutyrate significantly improved neurological function, survival outcomes, and efficacy of anti-CD19 CAR T cell therapy. This is the first report of BCKA accumulation in LMD and provides preclinical evidence that targeting these toxic metabolites improves outcomes.
Collapse
Affiliation(s)
- Mariam Lotfy Khaled
- The Department of Metabolism and Physiology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Egypt
| | - Yuan Ren
- The Department of Metabolism and Physiology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Ronak Kundalia
- The Department of Metabolism and Physiology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Hasan Alhaddad
- The Department of Metabolism and Physiology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Zhihua Chen
- Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Gerald C. Wallace
- Department of Hematology/Oncology, Georgia Cancer Center at Medical College of Georgia, Augusta, GA, USA
| | - Brittany Evernden
- Department of Neuro Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Oscar E. Ospina
- Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - MacLean Hall
- Department of Immunology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Min Liu
- The Proteomics and Metabolomics Core, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Lancia N.F. Darville
- The Proteomics and Metabolomics Core, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Victoria Izumi
- The Proteomics and Metabolomics Core, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Y. Ann Chen
- Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Shari Pilon-Thomas
- Department of Immunology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Paul A. Stewart
- Department of Biostatistics and Bioinformatics, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - John M. Koomen
- The Proteomics and Metabolomics Core, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
- Department of Molecular Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Salvatore A. Corallo
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Michael D. Jain
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Timothy J. Robinson
- Therapeutic Radiology, Smilow Cancer Hospital at Yale New Haven, 35 Park Street, New Haven, CT, USA
| | - Fredrick L. Locke
- Department of Blood and Marrow Transplant and Cellular Immunotherapy, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Peter A. Forsyth
- Department of Neuro Oncology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
- The Department of Tumor Biology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| | - Inna Smalley
- The Department of Metabolism and Physiology, The Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, USA
| |
Collapse
|
22
|
Su J, Tan S, Gong H, Luo Y, Cheng T, Yang H, Wen X, Jiang Z, Li Y, Zhang L. The Evaluation of Prognostic Value and Immune Characteristics of Ferroptosis-Related Genes in Lung Squamous Cell Carcinoma. Glob Med Genet 2023; 10:285-300. [PMID: 37915460 PMCID: PMC10615648 DOI: 10.1055/s-0043-1776386] [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] [Indexed: 11/03/2023] Open
Abstract
Background The purpose of our study was to construct a prognostic model based on ferroptosis-related gene signature to improve the prognosis prediction of lung squamous carcinoma (LUSC). Methods The mRNA expression profiles and clinical data of LUSC patients were downloaded. LUSC-related essential differentially expressed genes were integrated for further analysis. Prognostic gene signatures were identified through random forest regression and univariate Cox regression analyses for constructing a prognostic model. Finally, in a preliminary experiment, we used the reverse transcription-quantitative polymerase chain reaction assay to verify the relationship between the expression of three prognostic gene features and ferroptosis. Results Fifty-six ferroptosis-related essential genes were identified by using integrated analysis. Among these, three prognostic gene signatures (HELLS, POLR2H, and POLE2) were identified, which were positively affected by LUSC prognosis but negatively affected by immune cell infiltration. Significant overexpression of immune checkpoint genes occurred in the high-risk group. In preliminary experiments, we confirmed that the occurrence of ferroptosis can reduce three prognostic gene signature expression. Conclusions The three ferroptosis-related genes could predict the LUSC prognostic risk of antitumor immunity.
Collapse
Affiliation(s)
- Jialin Su
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Changsha, Hunan Province, People's Republic of China
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, People's Republic of China
| | - Shuhua Tan
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, People's Republic of China
| | - Houwu Gong
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, People's Republic of China
| | - Yongzhong Luo
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Changsha, Hunan Province, People's Republic of China
| | - Tianli Cheng
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Changsha, Hunan Province, People's Republic of China
| | - Hua Yang
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Changsha, Hunan Province, People's Republic of China
| | - Xiaoping Wen
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Changsha, Hunan Province, People's Republic of China
| | - Zhou Jiang
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Changsha, Hunan Province, People's Republic of China
| | - Yuning Li
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, People's Republic of China
| | - Lemeng Zhang
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Changsha, Hunan Province, People's Republic of China
| |
Collapse
|
23
|
Liao KL, Watt KD, Protin T. Different mechanisms of CD200-CD200R induce diverse outcomes in cancer treatment. Math Biosci 2023; 365:109072. [PMID: 37734537 DOI: 10.1016/j.mbs.2023.109072] [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: 06/20/2023] [Revised: 08/09/2023] [Accepted: 08/26/2023] [Indexed: 09/23/2023]
Abstract
The CD200 is a cell membrane protein expressed by tumor cells, and its receptor CD200 receptor (CD200R) is expressed by immune cells including macrophages and dendritic cells. The formation of CD200-CD200R inhibits the cellular functions of the targeted immune cells, so CD200 is one type of the immune checkpoint and blockade CD200-CD200R formation is a potential cancer treatment. However, the CD200 blockade has opposite treatment outcomes in different types of cancers. For instance, the CD200R deficient mice have a higher tumor load than the wild type (WT) mice in melanoma suggesting that CD200-CD200R inhibits melanoma. On the other hand, the antibody anti-CD200 treatment in pancreatic ductal adenocarcinoma (PDAC) and head and neck squamous cell carcinoma (HNSCC) significantly reduces the tumor load indicating that CD200-CD200R promotes PDAC and HNSCC. In this work, we hypothesize that different mechanisms of CD200-CD200R in tumor microenvironment could be one of the reasons for the diverse treatment outcomes of CD200 blockade in different types of cancers. We create one Ordinary Differential Equations (ODEs) model for melanoma including the inhibition of CCL8 and regulatory T cells and the switching from M2 to M1 macrophages by CD200-CD200R to capture the tumor inhibition by CD200-CD200R. We also create another ODEs model for PDAC and HNSCC including the promotion of the polarization and suppressive activities of M2 macrophages by CD200-CD200R to generate the tumor promotion by CD200-CD200R. Furthermore, we use these two models to investigate the treatment efficacy of the combination treatment between the CD200-CD200R blockade and the other immune checkpoint inhibitor, anti-PD-1. Our result shows that different mechanisms of CD200-CD200R can induce different treatment outcomes in combination treatments, namely, only the CD200-CD200R blockade reduces tumor load in melanoma and only the anti-PD-1 and CD200 knockout decrease tumor load in PDAC and HNSCC. Moreover, in melanoma, the CD200-CD200R mainly utilizes the inhibitions on M1 macrophages and dendritic cells to inhibit tumor growth, instead of M2 macrophages.
Collapse
Affiliation(s)
- Kang-Ling Liao
- Department of Mathematics, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada.
| | - Kenton D Watt
- Department of Mathematics, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada
| | - Tom Protin
- Department of Applied Mathematics, INSA Rennes, France
| |
Collapse
|
24
|
Wu D, Zhang H, Huang H, Li X, Liu X, Liu H, Chen J. Establishment of a prognostic signature for patients with advanced lung squamous cell carcinoma based on tumor-infiltrating immune cells. Transl Cancer Res 2023; 12:2706-2716. [PMID: 37969402 PMCID: PMC10643955 DOI: 10.21037/tcr-23-545] [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: 03/29/2023] [Accepted: 09/01/2023] [Indexed: 11/17/2023]
Abstract
Background With the advancements in the fields of science, technology, and medical therapy, there is an increasing awareness among the general public regarding tumor-infiltrating immune cells. These immune cells have a close association with the prognosis of clinical patients with lung cancer. Methods The research used a comprehensive analysis and assessed tumor-infiltrating immune cells in advanced lung squamous cell carcinoma (LUSC) using The Cancer Genome Atlas (TCGA) database and the CIBERSORT algorithm. The research examined 22 types of tumor-infiltrating immune cells and observed notable differences in the infiltration patterns of immune cells between normal tissue and advanced LUSC. Results Univariate Cox regression analyses revealed a positive correlation between macrophages M2 and patient prognoses, as well as potential influences on patient prognosis by natural killer (NK) cells resting, monocytes, and activated mast cells. Multivariate Cox regression models were developed, incorporating three types of immune cells. The efficacy of the model was evaluated using a receiver operating characteristic (ROC) curve. Furthermore, the research constructed a nomogram model to individually predict the mortality risk in patients with advanced LUSC. This prediction model serves as a valuable tool for clinicians, enabling them to provide effective guidance based on tumor-infiltrating immune cells for advanced LUSC patients. Conclusions The research comprehensively analyzed and evaluated 22 types of tumor-infiltrating immune cells from advanced LUSC, revealing the correlation between immune cell infiltration and overall survival (OS) in clinical patients. Based on the nomogram of NK cells resting, monocytes, and macrophages M2, it can make specific prognostic predictions for advanced LUSC patients.
Collapse
Affiliation(s)
- Di Wu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongbing Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hua Huang
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuanguang Li
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xingyu Liu
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyu Liu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jun Chen
- Department of Lung Cancer Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenvironment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
25
|
Lonie JM, Brosda S, Bonazzi VF, Aoude LG, Patel K, Brown I, Sharma S, Lampe G, Addala V, Koufariotis LT, Wood S, Waddell N, Dolcetti R, Barbour AP. The oesophageal adenocarcinoma tumour immune microenvironment dictates outcomes with different modalities of neoadjuvant therapy - results from the AGITG DOCTOR trial and the cancer evolution biobank. Front Immunol 2023; 14:1220129. [PMID: 37965317 PMCID: PMC10642165 DOI: 10.3389/fimmu.2023.1220129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/14/2023] [Indexed: 11/16/2023] Open
Abstract
A plateau in treatment effect can be seen for the current 'one-size-fits-all' approach to oesophageal adenocarcinoma (OAC) management using neoadjuvant chemoradiotherapy (nCRT) or chemotherapy (nCT). In OAC, the tumour microenvironment (TME) is largely immunosuppressed, however a subgroup of patients with an immune-inflamed TME exist and show improved outcomes. We aimed to understand the overall immune-based mechanisms underlying treatment responses and patient outcomes in OAC, and in relation to neoadjuvant therapy modality. This study included 107 patients; 68 patients were enrolled in the Australian Gastro-Intestinal Trials Group sponsored DOCTOR Trial, and 38 patients were included from the Cancer Evolution Biobank. Matched pre-treatment and post-treatment tumour biopsies were used to perform multi-modality analysis of the OAC TME including NanoString mRNA expression analysis, multiplex and single colour immunohistochemistry (IHC), and peripheral blood mononuclear cell analysis of tumour-antigen specific T cell responses. Patients with the best clinicopathological outcomes and survival had an immune-inflamed TME enriched with anti-tumour immune cells and pathways. Those with the worst survival showed a myeloid T regulatory cell enriched TME, with decreased CD8+ cell infiltration and increased pro-tumour immune cells. Multiplex IHC analysis identified that high intra-tumoural infiltration of CD8+ cells, and low infiltration with CD163+ cells was associated with improved survival. High tumour core CD8+ T cell infiltration, and a low tumour margin infiltration of CD163+ cells was also associated with improved survival. nCRT showed improved survival compared with nCT for patients with low CD8+, or high CD163+ cell infiltration. Poly-functional T cell responses were seen with tumour-antigen specific T cells. Overall, our study supports the development of personalised therapeutic approaches based on the immune microenvironment in OAC. Patients with an immune-inflamed TME show favourable outcomes regardless of treatment modality. However, in those with an immunosuppressed TME with CD163+ cell infiltration, treatment with nCRT can improve outcomes. Our findings support previous studies into the TME of OAC and with more research, immune based biomarker selection of treatment modality may lead in improved outcomes in this deadly disease.
Collapse
Affiliation(s)
- James M. Lonie
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Sandra Brosda
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Vanessa F. Bonazzi
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Lauren G. Aoude
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Kalpana Patel
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Ian Brown
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Envoi Specialist Pathologists, Brisbane, QLD, Australia
- Department of Pathology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Sowmya Sharma
- Medlab Pathology, Sydney, NSW, Australia
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Guy Lampe
- Department of Anatomical Pathology, Central Laboratory Pathology Queensland, Brisbane, QLD, Australia
| | - Venkateswar Addala
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | - Scott Wood
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Nicola Waddell
- Medical Genomics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Riccardo Dolcetti
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
- Translational and Clinical Immunotherapy, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, VIC, Australia
| | - Andrew P. Barbour
- Surgical Oncology Group, Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
- Department of Surgery, Princess Alexandra Hospital, Brisbane, QLD, Australia
| |
Collapse
|
26
|
Cao Y, Wu Y, Tu H, Gu Z, Yu F, Huang W, Shen L, Wang L, Li Y. (-)-Guaiol inhibit epithelial-mesenchymal transition in lung cancer via suppressing M2 macrophages mediated STAT3 signaling pathway. Heliyon 2023; 9:e19817. [PMID: 37809930 PMCID: PMC10559221 DOI: 10.1016/j.heliyon.2023.e19817] [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: 06/26/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 10/10/2023] Open
Abstract
In the context of cancer expansion, epithelial-mesenchymal transition (EMT) plays an essential role in driving invasion and metastasis potential of cancer cells. Tumor-associated macrophages (TAMs)-derived factors involved in the initiation and progression of EMT. We assess the role of M2 macrophage in suppressing lung tumors of a natural compound (-)-Guaiol by using macrophage depleted model. Bone marrow-derived monocytes (BMDMs) were extracted and induced to M2-like phenotype in vitro. The co-culture of M2 macrophage and lung cancer cells was established to observe that inhibition of lung tumor growth by (-)-Guaiol requires presence of macrophages. This suppressed effect of (-)-Guaiol was alleviated when mice macrophage was depleted. The expression of M2-like macrophages was strongly reduced by (-)-Guaiol treated mice, but not the changes of M1-like macrophages. In vitro studies, we demonstrated that (-)-Guaiol suppressed M2 polarization of BMDMs, as well as migration, invasion, and EMT of lung cancer cells in co-culture. M2 macrophage-derived interleukin 10 (IL-10) was investigated as a critical signaling molecule between M2 macrophage and lung cancer cells. We have also verified that the mechanism of (-)-Guaiol inhibiting the EMT process of lung cancer is related to the activation of IL-10-mediated signal transducer and activator of transcription 3 (STAT3). These results suggested that the suppressive effect role of (-)-Guaiol in M2 macrophage promoting EMT of lung cancer, which was associated with inhibition of IL-10 mediated STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yajuan Cao
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Yangpu District, Shanghai 200433, China
| | - Yonghui Wu
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Yangpu District, Shanghai 200433, China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongbin Tu
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Yangpu District, Shanghai 200433, China
| | - Zhan Gu
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Yangpu District, Shanghai 200433, China
| | - Fengzhi Yu
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Yangpu District, Shanghai 200433, China
| | - Weiling Huang
- Shanghai Jing 'an District Hospital of Traditional Chinese Medicine, Shanghai 200072, China
| | - Liping Shen
- LongHua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lixin Wang
- Department of Integrated Traditional Chinese and Western Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Yangpu District, Shanghai 200433, China
| | - Yan Li
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| |
Collapse
|
27
|
Wang X, Luo X, Wang Z, Wang Y, Zhao J, Bian L. Identification of cancer stemness and M2 macrophage-associated biomarkers in lung adenocarcinoma. Heliyon 2023; 9:e19114. [PMID: 37662825 PMCID: PMC10472008 DOI: 10.1016/j.heliyon.2023.e19114] [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: 02/07/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 09/05/2023] Open
Abstract
Objective Cancer stemness and M2 macrophages are intimately linked to the prognosis of lung adenocarcinoma (LUAD). For this reason, this investigation sought to identify the key genes relevant to cancer stemness and M2 macrophages, explore the relationship between these genes and clinical characteristics, and determine the potential mechanism. Methods LUAD transcriptomic data was analyzed from The Cancer Genome Atlas (TCGA) as well as the Gene Expression Omnibus databases. Differential expression analysis was performed to discern abnormally expressed genes between LUAD and control samples in TCGA cohort. The Cell type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT) algorithm was applied to determine varyingly infiltrated immune cells in LUAD compared with the control samples in TCGA cohort. Weighted correlation network analysis (WGCNA) was performed to identify genes associated with mRNA expression-based stemness index (mRNAsi) and M2 macrophages. Least absolute shrinkage and selection operator (LASSO), RandomForest (RF) and support vector machine-recursive feature elimination (SVM-RFE) machine learning methods were conducted to detect gene signatures. Global survival evaluation (Kaplan-Meier curve) was applied to investigate the relationship between gene signatures and the survival time of LUAD patients. Receiver operating characteristic (ROC) curves were produced to define biomarkers relevant to diagnosis. Gene Set Enrichment Analysis (GSEA) was performed to probe the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways related to diagnostic biomarkers. The public single-cell dataset of LUAD (GSE123902) was used to investigate the expression differences of diagnostic biomarkers in various cell types in LUAD. Real-time quantitative PCR (qRT-PCR) was performed to confirm key genes in lung adenocarcinoma cells. Results A total of 5,410 differentialy expressed genes (DEGs) as well as 15 differentially infiltrated immune cells were identified between LUAD and control sepcimens in TCGA cohort. Thirty-seven DEGs were associated with both M2 macrophages and mRNAsi according to the WGCNA analysis. Sixteen common gene signatures were obtained using three diverse algorithms. CBFA2T3, DENND3 and FCAMR were correlated to overall and disease-free survival of LUAD patients. ROC curves revealed that CBFA2T3 and DENND3 expression accurately classified LUAD and control samples. The results of single cell related analysis showed that two diagnostic biomarkers expressions were differed between the different tissue sources in M2-like macrophages. QRT-PCR demonstrated the mRNA expressions of CBFA2T3 and DENND3 were upregulated in lung adenocarcinoma cells A549 and H2122. Conclusion Our study identified CBFA2T3 and DENND3 as key genes associated with mRNAsi and M2 macrophages in LUAD and investigated the potential molecular mechanisms underlying this relationship.
Collapse
Affiliation(s)
| | | | - ZhiYuan Wang
- The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - YangHao Wang
- The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Juan Zhao
- The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Bian
- The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
28
|
Zhu YT, Wu SY, Yang S, Ying J, Tian L, Xu HL, Zhang HP, Yao H, Zhang WY, Jin QQ, Yang YT, Jiang XY, Zhang N, Yao S, Zhou SG, Chen G. Identification and validation of a novel anoikis-related signature for predicting prognosis and immune landscape in ovarian serous cystadenocarcinoma. Heliyon 2023; 9:e18708. [PMID: 37554782 PMCID: PMC10404752 DOI: 10.1016/j.heliyon.2023.e18708] [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: 03/08/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Ovarian serous cystadenocarcinoma (OSC) is the most prevalent histological subtype of ovarian cancer (OV) and presents a serious threat to women's health. Anoikis is an essential component of metastasis, and tumor cells can get beyond it to become viable. The impact of anoikis on OSC, however, has only been the topic of a few studies. METHODS The mRNA sequencing and clinical information of OSC came from The Cancer Genome Atlas Target Genotype-Tissue Expression (TCGA TARGET GTEx) dataset. Anoikis-related genes (ARGs) were collected by Harmonizome and GeneCards websites. Centered on these ARGs, we used unsupervised consensus clustering to explore potential tumor typing and filtered hub ARGs to create a model of predictive signature for OSC patients. Furthermore, we presented clinical specialists with a novel nomogram based on ARGs, revealing the underlying clinical relevance of this signature. Finally, we explored the immune microenvironment among various risk groups. RESULTS We identified 24 ARGs associated with the prognosis of OSC and classified OSC patients into three subtypes, and the subtype with the best prognosis was more enriched in immune-related pathways. Seven ARGs (ARHGEF7, NOTCH4, CASP2, SKP2, PAK4, LCK, CCDC80) were chosen to establish a risk model and a nomogram that can provide practical clinical decision support. Risk scores were found to be an independent and significant prognostic factor in OSC patients. The CIBERSORTx result revealed an inflammatory microenvironment is different for risk groups, and the proportion of immune infiltrates of Macrophages M1 is negatively correlated with risk score (rs = -0.21, P < 0.05). Ultimately, quantitative reverse transcription polymerase chain reaction (RT-PCR) was utilized to validate the expression of the seven pivotal ARGs. CONCLUSION In this study, based on seven ARGs, a risk model and nomogram established can be used for risk stratification and prediction of survival outcomes in patients with OSC, providing a reliable reference for individualized therapy of OSC patients.
Collapse
Affiliation(s)
- Yu-Ting Zhu
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Shuang-Yue Wu
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Song Yang
- Department of Pain Treatment, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, China
| | - Jie Ying
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Lu Tian
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Hong-Liang Xu
- Department of Pathology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - He-Ping Zhang
- Department of Pathology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Hui Yao
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Wei-Yu Zhang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Qin-Qin Jin
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Yin-Ting Yang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Xi-Ya Jiang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Nan Zhang
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Shun Yao
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Shu-Guang Zhou
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| | - Guo Chen
- Department of Gynecology, Maternal and Child Health Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230001, China
- Department of Gynecology, Anhui Province Maternity and Child Healthcare Hospital, Hefei, Anhui 230001, China
| |
Collapse
|
29
|
SUN DONGJIE, ZHANG CHI. Immunogenic cell death-related long noncoding RNA influences immunotherapy against lung adenocarcinoma. Oncol Res 2023; 31:753-767. [PMID: 37547766 PMCID: PMC10398397 DOI: 10.32604/or.2023.029287] [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: 02/10/2023] [Accepted: 05/06/2023] [Indexed: 08/08/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is the leading cause of cancer-related deaths, accounting for over a million deaths worldwide annually. Immunogenic cell death (ICD) elicits an adaptive immune response. However, the role of ICD-related long noncoding RNAs (lncRNAs) in LUAD is unknown. In this study, we investigated the characteristics of the tumor microenvironment in LUAD, the prognostic significance of ICD-related lncRNAs, and the half-maximal inhibitory concentration (IC50) of possible chemotherapeutic drugs. We sorted prognostic lncRNAs using univariate Cox regression and constructed a risk signature based on them. We then confirmed the model's accuracy and generated a nomogram. Additionally, we performed immune microenvironment analysis, somatic mutation calculation, Tumor Immune Dysfunction and Exclusion (TIDE) analysis, and anticancer pharmaceutical IC50 prediction. Least absolute shrinkage and selection operator Cox regression identified 27 prognostic lncRNAs related to ICD, and a unique risk signature using 10 ICD-related lncRNAs was constructed. The risk score was confirmed to be a reliable predictor of survival, with the highest c-index score. The signature had a remarkable predictive performance with clinical applicability and could accurately predict the overall survival in LUAD. Furthermore, the lncRNA signature was closely associated with immunocyte invasion. We also analyzed the correlation between the risk score, tumor-infiltrating immune cells, and prognosis and identified high immune and ESTIMATE scores in low-risk patients. Moreover, we observed elevated checkpoint gene expression and low TIDE scores in high-risk patients, indicating a good immunotherapy response. Finally, high-risk patients were shown to be susceptible to anticancer medications. Therefore, our unique risk signature comprising 10 ICD-related lncRNAs was demonstrated to indicate the characteristics of the tumor-immune microenvironment in LUAD, predict patients' overall survival, and guide individualized treatment.
Collapse
Affiliation(s)
- DONGJIE SUN
- Department of Translational Medicine, The First Hospital of Jilin University, Changchun, China
- College of Basic Medical Sciences, Jilin University, Changchun, China
| | - CHI ZHANG
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Haston S, Gonzalez-Gualda E, Morsli S, Ge J, Reen V, Calderwood A, Moutsopoulos I, Panousopoulos L, Deletic P, Carreno G, Guiho R, Manshaei S, Gonzalez-Meljem JM, Lim HY, Simpson DJ, Birch J, Pallikonda HA, Chandra T, Macias D, Doherty GJ, Rassl DM, Rintoul RC, Signore M, Mohorianu I, Akbar AN, Gil J, Muñoz-Espín D, Martinez-Barbera JP. Clearance of senescent macrophages ameliorates tumorigenesis in KRAS-driven lung cancer. Cancer Cell 2023; 41:1242-1260.e6. [PMID: 37267953 DOI: 10.1016/j.ccell.2023.05.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 03/07/2023] [Accepted: 05/05/2023] [Indexed: 06/04/2023]
Abstract
The accumulation of senescent cells in the tumor microenvironment can drive tumorigenesis in a paracrine manner through the senescence-associated secretory phenotype (SASP). Using a new p16-FDR mouse line, we show that macrophages and endothelial cells are the predominant senescent cell types in murine KRAS-driven lung tumors. Through single cell transcriptomics, we identify a population of tumor-associated macrophages that express a unique array of pro-tumorigenic SASP factors and surface proteins and are also present in normal aged lungs. Genetic or senolytic ablation of senescent cells, or macrophage depletion, result in a significant decrease in tumor burden and increased survival in KRAS-driven lung cancer models. Moreover, we reveal the presence of macrophages with senescent features in human lung pre-malignant lesions, but not in adenocarcinomas. Taken together, our results have uncovered the important role of senescent macrophages in the initiation and progression of lung cancer, highlighting potential therapeutic avenues and cancer preventative strategies.
Collapse
Affiliation(s)
- Scott Haston
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London WC1N 1EH, UK.
| | | | - Samir Morsli
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Jianfeng Ge
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Virinder Reen
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Alexander Calderwood
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Ilias Moutsopoulos
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Leonidas Panousopoulos
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Polina Deletic
- Division of Medicine, University College London, London, UK
| | - Gabriela Carreno
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Romain Guiho
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Saba Manshaei
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London WC1N 1EH, UK
| | | | - Hui Yuan Lim
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London WC1N 1EH, UK
| | | | - Jodie Birch
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Husayn A Pallikonda
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Tamir Chandra
- MRC Human Generics Unit, University of Edinburgh, Edinburgh, UK
| | - David Macias
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Gary J Doherty
- Cambridge University Hospitals NHS Foundation Trust, Department of Oncology, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Doris M Rassl
- Royal Papworth Hospital NHS Foundation Trust. Cambridge Biomedical Campus, Cambridge CB2 0AY, UK
| | - Robert C Rintoul
- Royal Papworth Hospital NHS Foundation Trust. Cambridge Biomedical Campus, Cambridge CB2 0AY, UK; Department of Oncology, University of Cambridge, Cambridge, UK; CRUK Cambridge Centre Thoracic Cancer Programme, Cambridge, UK
| | - Massimo Signore
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Irina Mohorianu
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Arne N Akbar
- Division of Medicine, University College London, London, UK
| | - Jesús Gil
- MRC London Institute of Medical Sciences (LMS), Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Daniel Muñoz-Espín
- Early Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, UK; CRUK Cambridge Centre Thoracic Cancer Programme, Cambridge, UK.
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, UCL Institute of Child Health, London WC1N 1EH, UK.
| |
Collapse
|
31
|
Zhang Q, Feng J, Liu K, Yang X, Huang Y, Tang B. STK11 mutation impacts CD1E expression to regulate the differentiation of macrophages in lung adenocarcinoma. Immun Inflamm Dis 2023; 11:e958. [PMID: 37506141 PMCID: PMC10373563 DOI: 10.1002/iid3.958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND The deficiency of serine/threonine protein kinase 11 (STK11), one of the most common tumor suppressor genes in non-small-cell lung cancer, is a crucial player in tumor immune microenvironment regulation. This study attempted to unveil how mutated STK11 impact the differentiation of macrophages in lung adenocarcinoma (LUAD). METHODS STK11 and CD1E expression levels in different cell models were assessed by quantitative reverse transcription polymerase chain reaction. Western blot was utilized to detect the protein expression levels of STK11, CD1E, apoptosis markers, and AMPK signaling pathway markers after transfection treatment. Cell viability and macrophage differentiation were detected by CCK-8 and flow cytometry. Immunohistochemistry and immunofluorescence were employed to detect the expression of related genes and macrophage markers, respectively. RESULTS This study found that STK11 mutations promoted the proliferation of LUAD cells and inhibited the differentiation of M1 macrophages, apoptosis, and the AMPK signaling pathway. Mutated STK11 led to CD1E downregulation, which curbed the differentiation of M1 macrophages and hence promoted LUAD progression. It was further validated by the in vivo experimental results that STK11 mutation significantly decreased the immune infiltration of M1 macrophages and promoted LUAD progression. CONCLUSION It was revealed that STK11 mutation affected CD1E expression to regulate macrophage differentiation in LUAD and then promote tumor progression. In this way, CD1E could be a potential biological target for the therapeutic interventions of STK11-mutant LUAD patients. These findings also threw new light on a new therapeutic strategy for STK11-mutant tumor patients that assisted the macrophage polarization pathway.
Collapse
Affiliation(s)
- Qingfeng Zhang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| | - Juan Feng
- Department of Operating Room, Zigong Fourth People's Hospital, Zigong, China
| | - Kui Liu
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| | - Xiaoyan Yang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| | - Yun Huang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| | - Bo Tang
- Department of Cardio-Thoracic Surgery, Zigong Fourth People's Hospital, Zigong, China
| |
Collapse
|
32
|
Yokota S, Kaji K, Yonezawa T, Momoi Y, Maeda S. CD204⁺ tumor-associated macrophages are associated with clinical outcome in canine pulmonary adenocarcinoma and transitional cell carcinoma. Vet J 2023; 296-297:105992. [PMID: 37164121 DOI: 10.1016/j.tvjl.2023.105992] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/12/2023]
Abstract
Tumor-associated macrophages are abundant infiltrating cells in the tumor microenvironment (TME). Macrophages can be classified into several types of subsets based on their immune responses. Among those subsets, M2 macrophages contribute to anti-inflammatory responses and create an immunosuppressive environment that promotes tumor cell proliferation. In a previous study, human cancer patients with high M2 macrophages showed a worse prognosis for many types of tumors. However, studies examining the relationship between M2 macrophages and clinical outcomes in canine tumors are limited. In the previous human and canine studies, CD204 has been used as the marker for detecting M2 macrophages. Then we evaluated CD204+ and total macrophages infiltration and its association with clinical outcomes in canine solid tumors. In this study, we examined dogs with oral malignant melanoma (OMM), pulmonary adenocarcinoma (PA), hepatocellular carcinoma (HCC), and transitional cell carcinoma (TCC). Compared to healthy tissues, CD204+ and total macrophages were increased in OMM, PA, and TCC, but not in HCC. High CD204+ macrophage levels were significantly associated with lung metastasis in TCC (P = 0.030). Kaplan-Meier analysis revealed that high CD204+ macrophage levels were associated with shorter overall survival (OS) in canine patients with PA (P = 0.012) and TCC (P = 0.0053). These results suggest that CD204+ macrophages contribute to tumor progression and could be a prognostic factor in dogs with PA and TCC.
Collapse
Affiliation(s)
- S Yokota
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - K Kaji
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - T Yonezawa
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Y Momoi
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - S Maeda
- Department of Veterinary Clinical Pathobiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| |
Collapse
|
33
|
Liao Y, Cheng W, Mou R, Li X, Jia Y. RGN as a prognostic biomarker with immune infiltration and ceRNA in lung squamous cell carcinoma. Sci Rep 2023; 13:7553. [PMID: 37161020 PMCID: PMC10170118 DOI: 10.1038/s41598-023-32217-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/24/2023] [Indexed: 05/11/2023] Open
Abstract
Regucalcin (RGN) is a potent inhibitory protein of calcium signaling and expresses in various tissues. However, the role of RGN in the tumor immunological microenvironment in lung squamous cell carcinoma (LUSC) remains unclear. This study identified the expression of RGN from public databases and immunohistochemistry with clinical specimen. The association between RGN and the tumor immune microenvironment (TIME) was investigated in LUSC by ESTIMATE and CIBERSORT algorithms. Similarly, the Tumor IMmune Estimation Resource (TIMER) database was used to identify the correlation between RGN and immune cells. The ceRNA network was established based on the data obtained from public databases. Finally, prediction of drug response to chemotherapy and immunotherapy was performed to evaluate clinical significance. This study found that RGN expression was significantly downregulated in tumor tissues and closely related to clinical factors and prognosis of LUSC patients. Differentially expressed genes (DEGs) grouped by the expression of RGN were mostly involved in immunobiological processes such as humoral immune response and leukocyte mediated immunity. RGN and its related miRNA (has-miR-203a-3p) and lncRNAs (ZNF876P and PSMG3-AS1) constructed the novel prognosis-related ceRNA network. Plasma cells, T cells CD4 memory resting, Macrophages M0, Macrophages M1, Mast cells resting, Mast cells activated and Neutrophils showed significantly different levels of infiltration between high and low RGN expression groups. The TIMER database showed that RGN expression was positively correlated with certain immune infiltrating cells. High RGN expression group showed a higher TIDE score, a higher dysfunction score and a lower MSI score, presenting a possible lower efficacy after accepting the immunotherapy than low RGN expression group. RGN expression was closely associated with prognosis of LUSC patients and played an important role in tumor microenvironment. This suggests that RGN could be a promising biomarker for assessing immunotherapy efficacy and prognosis.
Collapse
Affiliation(s)
- Yang Liao
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300008, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300008, China
| | - Wen Cheng
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300008, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300008, China
| | - Ruiyu Mou
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300008, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300008, China
| | - Xiaojiang Li
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300008, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300008, China.
| | - Yingjie Jia
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300008, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300008, China.
| |
Collapse
|
34
|
Yalamarty SSK, Filipczak N, Khan MM, Torchilin VP. Role of circular RNA and its delivery strategies to cancer - An overview. J Control Release 2023; 356:306-315. [PMID: 36878321 DOI: 10.1016/j.jconrel.2023.02.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023]
Abstract
With the passage of years and the progress of research on ribonucleic acids, the range of forms in which these molecules have been observed grows. One of them, discovered relatively recently, is circular RNA - covalently closed circles (circRNA). In recent years, there has been a huge increase in the interest of researchers in this group of molecules. It entailed a significant increase in the state of knowledge about them, which in turn caused a dramatic change in their perception. Rather than seeing circular RNAs as curiosities that represent a minor information noise in a cell or a result of RNA misprocessing, they came to be regarded as a common, essential, and potentially extremely useful group of molecules. Nevertheless, the current state of the art of circRNA is full of white cards. A lot of valuable information has been obtained from high-throughput methods to study whole transcriptomes, but many issues related to circular RNAs still need to be clarified. Presumably, each answer obtained will raise several new questions. However, circRNAs have a wealth of potential applications, including therapeutic applications.
Collapse
Affiliation(s)
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | | | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA; Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
35
|
Niblock A, Rajendran S, Laverty C, Logue P, Alexander HD. Assessing the frequency of CD163+ tumor-associated macrophages and CD3+ T lymphocytes between MGUS and plasma cell myeloma. Exp Hematol 2023; 119-120:3-7. [PMID: 36516929 DOI: 10.1016/j.exphem.2022.12.001] [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: 01/17/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Plasma cell dyscrasias (PCDs) are a heterogeneous group of diseases, and the most common is monoclonal gammopathy of undetermined significance (MGUS). This premalignant PCD consistently precedes multiple myeloma (MM), with a 1% risk of progression per year. Evading and suppressing the host immune system is an important step in the progression of MGUS to MM. This pilot study was designed to assess whether MGUS and MM have a distinct microenvironment, characterized by a unique distribution of immune cells, including tumor-associated macrophages. Evaluation of bone marrow (BM) tumor microenvironment was performed using immunohistochemical quantification of T cells (CD3), macrophages (CD68), and a macrophage subtype (CD163). The findings were compared between MGUS and MM to determine whether differences existed. The results suggest that there is a significantly lower percentage of CD3-positive, CD68-positive and CD163-positive immune effector cells in BM trephine biopsy samples from patients with MGUS than in those from patients with untreated MM (p < 0.001). Interestingly, in a patient treated for MM, the percentages of CD3+ and CD68+ cells were the same as those in other patients with untreated MM; however, the percentage of CD163+ cells reduced and correlated with low plasma cell count. Future studies are required to investigate whether the percentage of CD163+ cells is correlated with disease burden in patients with MM. If this is the case, then the level of soluble CD163 in plasma could be a potential biomarker of disease burden in patients with nonsecretory myelomas, in whom measurements of levels of paraprotein and free light chains are inconclusive.
Collapse
Affiliation(s)
- Aaron Niblock
- Queen Mary University, London, United Kingdom; School of Medicine, Ulster University, Northern Ireland.
| | | | | | - Philip Logue
- Antrim Laboratory, Antrim Area Hospital, Northern Ireland
| | - H Denis Alexander
- Personalised Medicine Centre, School of Medicine, Ulster University, Northern Ireland
| |
Collapse
|
36
|
Yi B, Cheng Y, Chang R, Zhou W, Tang H, Gao Y, Zhang C. Prognostic significance of tumor-associated macrophages polarization markers in lung cancer: a pooled analysis of 5105 patients. Biosci Rep 2023; 43:BSR20221659. [PMID: 36633963 PMCID: PMC9902841 DOI: 10.1042/bsr20221659] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/01/2022] [Accepted: 01/11/2023] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The prognostic significance of tumor-associated macrophages (TAMs) in patients with lung cancer (LCa) remains controversial. We therefore conducted the present study to systematically evaluate the role of different TAMs markers and histologic locations on the prognosis of LCa. METHODS Searches of Web of Science, PubMed, and EMBASE databases were performed up to 28 February 2022. The pooled analysis was conducted in random-effect or fixed-effects model with hazard risk (HR) and 95% confidence interval (CI) for survival data including overall survival (OS), and disease-free survival (DFS) from raw or adjusted measures, according to different TAMs markers and histologic locations. RESULTS Including a total of 5105 patients from 30 eligible studies, the results indicated that the total count of CD68+ TAMs was negatively associated with OS and DFS, which was also observed in the relationship of CD68+ or CD204+ TAMs in tumor stroma (TS) with OS and DFS (all P<0.05). Conversely, higher CD68+ TAMs density in tumor nest (TN) or TN/TS ratio of CD68+ TAMs predicted better OS (all P<0.05). Similarly, higher HLA-DR+ TAMs density was correlated with better OS in TN and TS (all P<0.05). Besides, neither nest CD163+ TAM density nor stromal CD163+ TAM density was a prognostic factor in LCa patients (all P>0.05). CONCLUSION Our study indicated that different TAMs markers and histologic locations could bring about different prognostic effects in LCa patients. Great understanding of the infiltration modes of TAMs may contribute to improve outcomes of LCa patients.
Collapse
Affiliation(s)
- Bin Yi
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008 Hunan, P. R. China
| | - Yuanda Cheng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008 Hunan, P. R. China
| | - Ruimin Chang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008 Hunan, P. R. China
| | - Wolong Zhou
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008 Hunan, P. R. China
| | - Huili Tang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008 Hunan, P. R. China
| | - Yang Gao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008 Hunan, P. R. China
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008 Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008 Hunan, P. R. China
| |
Collapse
|
37
|
Tumor Suppressor miR-613 Alleviates Non-Small Cell Lung Cancer Cell via Repressing M2 Macrophage Polarization. JOURNAL OF ONCOLOGY 2023; 2023:2311231. [PMID: 36844868 PMCID: PMC9950322 DOI: 10.1155/2023/2311231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/10/2022] [Accepted: 11/24/2022] [Indexed: 02/18/2023]
Abstract
Background Non-small cell lung cancer (NSCLC) is a crucial crux of cancer-related death, and M2 macrophage polarization facilitates NSCLC development. MicroRNA-613 (miR-613) is a tumor suppressor. This research aimed to clarify the miR-613 function in NSCLC and its impact on M2 macrophage polarization. Methods. miR-613 expressions in NSCLC tissues and cells were evaluated using quantitative real-time PCR. For miR-613 function in NSCLC, cell proliferation analysis, cell counting kit-8, flow cytometry, western blot, transwell, and wound-healing were conducted. Meanwhile, the miR-613 impact on M2 macrophage polarization was assessed by the NSCLC models. Results. miR-613 was lessened in NSCLC cells and tissues. It was corroborated that miR-613 overexpression retrained NSCLC cell proliferation, invasion, and migration but facilitated cell apoptosis. Moreover, miR-613 overexpression restrained NSCLC development by repressing M2 macrophage polarization. Conclusion Tumor suppressor miR-613 ameliorated NSCLC by restraining M2 macrophage polarization.
Collapse
|
38
|
Wang GC, Zhou M, Zhang Y, Cai HM, Chiang ST, Chen Q, Han TZ, Li RX. Screening and identifying a novel M-MDSCs-related gene signature for predicting prognostic risk and immunotherapeutic responses in patients with lung adenocarcinoma. Front Genet 2023; 13:989141. [PMID: 36699465 PMCID: PMC9869425 DOI: 10.3389/fgene.2022.989141] [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: 07/08/2022] [Accepted: 11/30/2022] [Indexed: 01/05/2023] Open
Abstract
Background: Lung adenocarcinoma (LUAD) shows intratumoral heterogeneity, a highly complex phenomenon that known to be a challenge during cancer therapy. Considering the key role of monocytic myeloid-derived suppressor cells (M-MDSCs) in the tumor microenvironment (TME), we aimed to build a prognostic risk model using M-MDSCs-related genes. Methods: M-MDSCs-related genes were extracted from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Utilized univariate survival analysis and random forest algorithm to screen candidate genes. A least absolute shrinkage and selection operator (LASSO) Cox regression analysis was selected to build the risk model. Patients were scored and classified into high- and low-risk groups based on the median risk scores. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis along with R packages "estimate" and "ssGSEA" were performed to reveal the mechanism of risk difference. Prognostic biomarkers and tumor mutation burden (TMB) were combined to predict the prognosis. Nomogram was carried out to predict the survival probability of patients in 1, 3, and 5 years. Results: 8 genes (VPREB3, TPBG, LRFN4, CD83, GIMAP6, PRMT8, WASF1, and F12) were identified as prognostic biomarkers. The GEO validation dataset demonstrated the risk model had good generalization effect. Significantly enrichment level of cell cycle-related pathway and lower content of CD8+ T cells infiltration in the high-risk group when compared to low-risk group. Morever, the patients were from the intersection of high-TMB and low-risk groups showed the best prognosis. The nomogram demonstrated good consistency with practical outcomes in predicting the survival rate over 1, 3, and 5 years. Conclusion: The risk model demonstrate good prognostic predictive ability. The patients from the intersection of low-risk and high-TMB groups are not only more sensitive response to but also more likely to benefit from immune-checkpoint-inhibitors (ICIs) treatment.
Collapse
Affiliation(s)
- Geng-Chong Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Mi Zhou
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China,Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yan Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Hua-Man Cai
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Seok-Theng Chiang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Qi Chen
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Tian-Zhen Han
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | | |
Collapse
|
39
|
Moerland JA, Leal AS, Lockwood B, Demireva EY, Xie H, Krieger-Burke T, Liby KT. The Triterpenoid CDDO-Methyl Ester Redirects Macrophage Polarization and Reduces Lung Tumor Burden in a Nrf2-Dependent Manner. Antioxidants (Basel) 2023; 12:116. [PMID: 36670978 PMCID: PMC9854457 DOI: 10.3390/antiox12010116] [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: 12/12/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
The NRF2/KEAP1 pathway protects healthy cells from malignant transformation and maintains cellular homeostasis. Up to 30% of human lung tumors gain constitutive NRF2 activity which contributes to cancer cell survival and chemoresistance, but the effects of NRF2 activation in immune cells within the tumor microenvironment are underexplored. Macrophages can promote cancer progression or regression depending on context, and NRF2 activation affects macrophage activity. The NRF2 activator CDDO-Methyl ester (CDDO-Me or bardoxolone methyl) reprogrammed Nrf2 wild-type (WT) tumor-educated bone marrow-derived macrophages (TE-BMDMs) from a tumor-promoting to a tumor-inhibiting phenotype, marked by an increase in M1 markers TNFα, IL-6, and MHC-II and a decrease in the tumor-promoting factors VEGF, CCL2, and CD206. No changes were observed in Nrf2 knockout (KO) TE-BMDMs. CDDO-Me decreased tumor burden (p < 0.001) and improved pathological grade (p < 0.05) in WT but not Nrf2 KO A/J mice. Tumor burden in Nrf2 KO mice was 4.6-fold higher (p < 0.001) than in WT mice, irrespective of treatment. CDDO-Me increased the number of lung-infiltrating macrophages in WT mice but lowered CD206 expression in these cells (p < 0.0001). In summary, Nrf2 KO exacerbates lung tumorigenesis in A/J mice, and CDDO-Me promotes an Nrf2-dependent, anti-cancer macrophage phenotype.
Collapse
Affiliation(s)
- Jessica A. Moerland
- Department of Pharmacology & Toxicology, College of Osteopathic Medicine, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI 48824, USA
| | - Ana S. Leal
- Department of Pharmacology & Toxicology, College of Osteopathic Medicine, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI 48824, USA
| | - Beth Lockwood
- Department of Pharmacology & Toxicology, College of Osteopathic Medicine, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI 48824, USA
| | - Elena Y. Demireva
- Transgenic and Genome Editing Facility, Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Huirong Xie
- Transgenic and Genome Editing Facility, Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, MI 48824, USA
| | | | - Karen T. Liby
- Department of Pharmacology & Toxicology, College of Osteopathic Medicine, Michigan State University, B430 Life Science Building, 1355 Bogue Street, East Lansing, MI 48824, USA
| |
Collapse
|
40
|
Balážová K, Clevers H, Dost AFM. The role of macrophages in non-small cell lung cancer and advancements in 3D co-cultures. eLife 2023; 12:82998. [PMID: 36809334 PMCID: PMC9943070 DOI: 10.7554/elife.82998] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 02/09/2023] [Indexed: 02/23/2023] Open
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths worldwide. Traditional therapeutic approaches such as chemotherapy or radiotherapy have provided only a marginal improvement in the treatment of lung carcinomas. Inhibitors targeting specific genetic aberrations present in non-small cell lung cancer (NSCLC), the most common subtype (85%), have improved the prognostic outlook, but due to the complexity of the LC mutational spectrum, only a fraction of patients benefit from these targeted molecular therapies. More recently, the realization that the immune infiltrate surrounding solid tumors can foster tumor-promoting inflammation has led to the development and implementation of anticancer immunotherapies in the clinic. In NSCLC, one of the most abundant leukocyte infiltrates is macrophages. These highly plastic phagocytes, which are part of the cellular repertoire of the innate immunity, can have a pivotal role in early NSCLC establishment, malignant progression, and tumor invasion. Emerging macrophage-targeting therapies have been focused on the re-differentiation of the macrophages toward an antitumorigenic phenotype, depletion of tumor-promoting macrophage subtypes, or combination therapies combining traditional cytotoxic treatments with immunotherapeutic agents. The most extensively used models employed for the exploration of NSCLC biology and therapy have been 2D cell lines and murine models. However, studying cancer immunology requires appropriately complex models. 3D platforms, including organoid models, are quickly advancing powerful tools to study immune cell-epithelial cell interactions within the tumor microenvironment. Co-cultures of immune cells along with NSCLC organoids allow for an in vitro observation of the tumor microenvironment dynamics closely resembling in vivo settings. Ultimately, the implementation of 3D organoid technology into tumor microenvironment-modeling platforms might facilitate the exploration of macrophage-targeted therapies in NSCLC immunotherapeutic research, thus establishing a new frontier in NSCLC treatment.
Collapse
Affiliation(s)
- Katarína Balážová
- Hubrecht Institute for Developmental Biology and Stem Cell Research-KNAW & University Medical Centre UtrechtUtrechtNetherlands,Oncode Institute, Hubrecht Institute-KNAWUtrechtNetherlands
| | - Hans Clevers
- Roche Pharma Research and early DevelopmentBaselSwitzerland
| | - Antonella FM Dost
- Hubrecht Institute for Developmental Biology and Stem Cell Research-KNAW & University Medical Centre UtrechtUtrechtNetherlands,Oncode Institute, Hubrecht Institute-KNAWUtrechtNetherlands
| |
Collapse
|
41
|
Li Z, Wang YJ, Zhou J, Umakoshi M, Goto A. The prognostic role of M2 tumor-associated macrophages in non-small-cell lung cancer. Histol Histopathol 2022; 37:1167-1175. [PMID: 35638244 DOI: 10.14670/hh-18-474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lung cancer is a high-risk tumor and is a main cause of death worldwide. The tumor aggressiveness and degree of malignancy depend not only on the tumor itself, but also on the microenvironment. The inflammatory microenvironment is one of the key factors in promoting the progression of lung cancer. It has been found that macrophages are the most abundant immune cells in the tumor microenvironment, with strong plasticity and heterogeneity. Tumor-Associated Macrophages (TAMs) are important components of the tumor immune microenvironment. TAMs are thought to be polarized into two main phenotypes: inflammatory or classically activated (M1) and antiinflammatory or alternatively activated (M2) macrophages. Their phenotype and function change according to environment and the appearance of tumor cells. M2 macrophages have been reported to be protumorigenic, because they can promote the formation of blood vessels in the tumor microenvironment, helping tumor cells escape the body's immune defense and promote their growth, by releasing a variety of cytokines, including chemokines, inflammatory factors and growth factor. However, the prognostic impact of TAMs and their phenotypes in non-small-cell lung cancer (NSCLC) remains to be fully elucidated. Some reports of the association between the characteristics of macrophages in lung tumor and patients' survival outcomes show contradicting results. In order to explore the prognostic role of TAMs in NSCLS, the association between the phenotype, density and distribution of macrophages and the prognosis of human NSCLC, as well as the potential mechanisms of M2 macrophages leading to poor prognosis in NSCLC, are reviewed in this study.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China.,Department of Cellular and Organ Pathology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Yun-Jie Wang
- Department of Laboratory Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Jian Zhou
- Department of Laboratory Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, China
| | - Michinobu Umakoshi
- Department of Cellular and Organ Pathology, Graduate School of Medicine, Akita University, Akita, Japan
| | - Akiteru Goto
- Department of Cellular and Organ Pathology, Graduate School of Medicine, Akita University, Akita, Japan.
| |
Collapse
|
42
|
Tang Y, Hu X, Wu K, Li X. Immune landscape and immunotherapy for penile cancer. Front Immunol 2022; 13:1055235. [PMID: 36524123 PMCID: PMC9745054 DOI: 10.3389/fimmu.2022.1055235] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022] Open
Abstract
Penile cancer is a rare malignancy and usually refers to penile squamous cell carcinoma (PSCC), which accounts for more than 95% of all penile malignancies. Although organ-sparing surgery is an effective treatment for early-stage PSCC, surgical intervention alone is often not curative for advanced PSCC with metastases to the inguinal and/or pelvic lymph nodes; thus, systemic therapy is required (usually platinum-based chemotherapy and surgery combined). However, chemotherapy for PSCC has proven to be of limited efficacy and is often accompanied by high toxicity, and patients with advanced PSCC usually have poor prognosis. The limited treatment options and poor prognosis indicate the unmet need for advanced PSCC. Immune-based therapies have been approved for a variety of genitourinary and squamous cell carcinomas but are rarely reported in PSCC. To date, several studies have reported high expression of PDL1 in PSCC, supporting the potential application of immune checkpoint inhibitors in PSCC. In addition, human papillomavirus (HPV) infection is highly prevalent in PSCC and plays a key role in the carcinogenesis of HPV-positive PSCC, suggesting that therapeutic HPV vaccine may also be a potential treatment modality. Moreover, adoptive T cell therapy (ATC) has also shown efficacy in treating advanced penile cancer in some early clinical trials. The development of new therapeutics relies on understanding the underlying biological mechanisms and processes of tumor initiation, progression and metastasis. Therefore, based on the interest, we reviewed the tumor immune microenvironment and the emerging immunotherapy for penile cancer.
Collapse
Affiliation(s)
| | | | | | - Xiang Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Van Hiep N, Sun WL, Feng PH, Lin CW, Chen KY, Luo CS, Dung LN, Van Quyet H, Wu SM, Lee KY. Heparin binding epidermal growth factor-like growth factor is a prognostic marker correlated with levels of macrophages infiltrated in lung adenocarcinoma. Front Oncol 2022; 12:963896. [PMID: 36439487 PMCID: PMC9686304 DOI: 10.3389/fonc.2022.963896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Background The interactions between tumor cells and the host immune system play a crucial role in lung cancer progression and resistance to treatment. The alterations of EGFR signaling have the potential to produce an ineffective tumor-associated immune microenvironment by upregulating a series of immune suppressors, including inhibitory immune checkpoints, immunosuppressive cells, and cytokines. Elevated Heparin-binding EGF-like growth factor (HB-EGF) expression, one EGFR ligand correlated with higher histology grading, worse patient prognosis, and lower overall survival rate, acts as a chemotactic factor. However, the role of heparin-binding epidermal growth factor-like growth factor (HB-EGF) in the accumulation of immune cells in the tumor microenvironment remains unclear. Methods The clinical association of HB-EGF expression in lung cancer was examined using the Gene Expression Omnibus (GEO) repository. HB-EGF expression in different cell types was determined using single-cell RNA sequencing (scRNA-seq) dataset. The correlation between HB-EGF expression and cancer-immune infiltrated cells was investigated by performing TIMER and ClueGo pathways analysis from TCGA database. The chemotaxis of HB-EGF and macrophage infiltration was investigated using migration and immunohistochemical staining. Results The high HB-EGF expression was significantly correlated with poor overall survival in patients with lung adenocarcinoma (LUAD) but not lung squamous cell carcinoma (LUSC). Moreover, HB-EGF expression was correlated with the infiltration of monocytes, macrophages, neutrophils, and dendritic cells in LUAD but not in LUSC. Analysis of scRNA-seq data revealed high HB-EGF expression in lung cancer cells and myeloid cells. Results from the pathway analysis and cell-based experiment indicated that elevated HB-EGF expression was associated with the presence of macrophage and lung cancer cell migration. HB-EGF was highly expressed in tumors and correlated with M2 macrophage infiltration in LUAD. Conclusions HB-EGF is a potential prognostic marker and therapeutic target for lung cancer progression, particularly in LUAD.
Collapse
Affiliation(s)
- Nguyen Van Hiep
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,Oncology Center, Bai Chay Hospital, Quang Ninh, Ha Long, Vietnam,Department of Thoracic and Neurological Surgery, Bai Chay Hospital, Quang Ninh, Ha Long, Vietnam
| | - Wei-Lun Sun
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Hao Feng
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Wei Lin
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei, Taiwan,Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Yuan Chen
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei, Taiwan,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ching-Shan Luo
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan,TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei, Taiwan
| | - Le Ngoc Dung
- Department of Thoracic and Neurological Surgery, Bai Chay Hospital, Quang Ninh, Ha Long, Vietnam
| | - Hoang Van Quyet
- Department of Thoracic and Neurological Surgery, Bai Chay Hospital, Quang Ninh, Ha Long, Vietnam
| | - Sheng-Ming Wu
- Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei, Taiwan,*Correspondence: Kang-Yun Lee, ; Sheng-Ming Wu,
| | - Kang-Yun Lee
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan,Division of Pulmonary Medicine, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,TMU Research Center for Thoracic Medicine, Taipei Medical University, Taipei, Taiwan,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan,*Correspondence: Kang-Yun Lee, ; Sheng-Ming Wu,
| |
Collapse
|
44
|
[Consensus on Postoperative Recurrence Prediction of Non-small Cell Lung Cancer
Based on Molecular Markers]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:701-714. [PMID: 36285390 PMCID: PMC9619343 DOI: 10.3779/j.issn.1009-3419.2022.102.44] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Significant progress has been made in lung cancer screening, surgery, chemoradiation, targeted therapy, and immunotherapy recently. Surgical resection is the most important treatment for localized non-small cell lung cancer (NSCLC) so far, but there are still many patients who develop local recurrence or distant metastases within 5 years of surgery. Currently, the risk factors of recurrence in patients with NSCLC are mainly based on clinical and pathological features, which hardly identify patients at high risk of recurrence accurately. With the development of new detection technologies, a number of molecular markers that may have a predictive risk of recurrence in NSCLC have been discovered over the years. In order to summarize the molecular markers related to postoperative recurrence in NSCLC patients, we have formulated a consensus on the prediction of postoperative recurrence of NSCLC based on molecular markers. This consensus mainly focuses on the early stage NSCLC patients, discusses and summarizes the risk factors of disease recurrence from the molecular level. It is hoped that more and more valuable information can be provided for the management of patients, so as to provide more guidance for the perioperative management of the patients with early stage NSCLC in the future.
.
Collapse
|
45
|
Park JV, Chandra R, Cai L, Ganguly D, Li H, Toombs JE, Girard L, Brekken RA, Minna JD. Tumor Cells Modulate Macrophage Phenotype in a Novel In Vitro Co-Culture Model of the NSCLC Tumor Microenvironment. J Thorac Oncol 2022; 17:1178-1191. [PMID: 35798240 PMCID: PMC9529910 DOI: 10.1016/j.jtho.2022.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Macrophage phenotype in the tumor microenvironment correlates with prognosis in NSCLC. Immunosuppressive macrophages promote tumor progression, whereas proinflammatory macrophages may drive an antitumor immune response. How individual NSCLCs affect macrophage phenotype is a major knowledge gap. METHODS To systematically study the impact of lung cancer cells on macrophage phenotypes, we developed an in vitro co-culture model that consisted of molecularly and clinically annotated patient-derived NSCLC lines, human cancer-associated fibroblasts, and murine macrophages. Induced macrophage phenotype was studied through quantitative real-time polymerase chain reaction and validated in vivo using NSCLC xenografts through quantitative immunohistochemistry and clinically with The Cancer Genome Atlas (TCGA)-"matched" patient tumors. RESULTS A total of 72 NSCLC cell lines were studied. The most frequent highly induced macrophage-related gene was Arginase-1, reflecting an immunosuppressive M2-like phenotype. This was independent of multiple clinicopathologic factors, which also did not affect M2:M1 ratios in matched TCGA samples. In vivo, xenograft tumors established from high Arginase-1-inducing lines (Arghi) had a significantly elevated density of Arg1+ macrophages. Matched TCGA clinical samples to Arghi NSCLC lines had a significantly higher ratio of M2:M1 macrophages (p = 0.0361). CONCLUSIONS In our in vitro co-culture model, a large panel of patient-derived NSCLC lines most frequently induced high-expression Arginase-1 in co-cultured mouse macrophages, independent of major clinicopathologic and oncogenotype-related factors. Arghi cluster-matched TCGA tumors contained a higher ratio of M2:M1 macrophages. Thus, this in vitro model reproducibly characterizes how individual NSCLC modulates macrophage phenotype, correlates with macrophage polarization in clinical samples, and can serve as an accessible platform for further investigation of macrophage-specific therapeutic strategies.
Collapse
Affiliation(s)
- Josiah Voth Park
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Raghav Chandra
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ling Cai
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas; Quantitative Biomedical Research Center, Department of Population and Data Science, University of Texas Southwestern Medical Center, Dallas, Texas; Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Debolina Ganguly
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas; Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Huiyu Li
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jason E Toombs
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Luc Girard
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas; Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas; Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, Texas; Cancer Biology Graduate Program, University of Texas Southwestern Medical Center, Dallas, Texas; Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
46
|
Clinical relevance and therapeutic aspects of professional antigen-presenting cells in lung cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:237. [PMID: 36175603 DOI: 10.1007/s12032-022-01841-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/01/2022] [Indexed: 10/14/2022]
Abstract
Lung cancer stays the preeminent cause of death worldwide. Despite recent advancements in chemotherapy, radiotherapy, and immunotherapy, the survival rate for people with advanced stages of the disease is still appalling. Moreover, there is a severe lack of reliable prognoses and indicators for classification in newly developed immunotherapies. A better understanding of immune cells is necessary to harness immune response mechanisms for therapeutic effects. Professional antigen-presenting cells are responsible for determining the fate of the immune response through the antigen processing and presentation pathway (APP). The most professional antigen-presenting cells (APC) include the dendritic cells (DC), macrophages, and B cells, which present antigens to the T-helper cells. Dendritic cells are significantly explored as a tool for immunotherapy owing to their precise ability to provoke and alter T-cell responses. Moreover, the role of tumor-associated macrophages (TAMs), an abundant leukocyte in lung cancer, is also a potential target for adjuvant anti-cancer therapies. In this review, we summarize the recent advances in our understanding of the various types of immunotherapy mapped out via professional antigen-presenting cells in lung cancer.
Collapse
|
47
|
Shen T, Miao S, Zhou Y, Yi X, Xue S, Du B, Tang C, Qu L, Fu D, Jia R, He H. Exosomal AP000439.2 from clear cell renal cell carcinoma induces M2 macrophage polarization to promote tumor progression through activation of STAT3. Cell Commun Signal 2022; 20:152. [PMID: 36153596 PMCID: PMC9509597 DOI: 10.1186/s12964-022-00957-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 08/09/2022] [Indexed: 11/28/2022] Open
Abstract
Background Tumorigenic phenotype of M2 tumor-associated macrophages promote tumor progression in response to exosomes cues imposed by tumor cells. However, the effect and underlying mechanisms of clear cell renal cell carcinoma (ccRCC)-derived exosomes (ccRCC-exo) on instructing macrophages phenotype remains unclear. Methods Macrophages were cocultured with ccRCC-exo and then evaluate the polarization of macrophages and migration of ccRCC cells. The effect and mechanism of lncRNA AP000439.2 overexpressed or deleted exosomes on macrophages M2 polarization were examined. Xenograft tumor mice model was used for in vivo validation. Results The ccRCC-exo significantly activated macrophages to M2 phenotype presented by increased expression of transforming growth factor-beta (TGF-β) and interleukin 10 (IL-10) at mRNA and protein levels, and these M2 macrophages in turn facilitating the migration of ccRCC cells. LncRNA AP000439.2 was highly enriched in the ccRCC-exo. Overexpression of exosomal AP000439.2 promoted M2 macrophage polarization whereas AP000439.2-deficient exosome had the opposite effects. Nuclear-localized AP000439.2 directly interacted with signal transducer and activator of transcription 3 (STAT3) proteins and phosphorylated STAT3 in macrophages. RNA-Seq results showed overexpression of AP000439.2 activated NF-κB signaling pathway. Silencing of STAT3 suppressed overexpression of AP000439.2-induced up-regulation of TGF-β and IL-10 expression, and p65 phosphorylation. AP000439.2-deleted exosome inhibited tumor growth in vivo. Conclusion Exosomes from ccRCC deliver AP000439.2 to promote M2 macrophage polarization via STAT3, thus enhancing ccRCC progression, indicating exosomal AP000439.2 might be a novel therapeutic target in ccRCC. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00957-6.
Collapse
|
48
|
Chen Z, Yang J, Li Y, Zeng W, Bai Y, Ding C, Xu C, Li C, Chen J, Ju S, Tang L, Zhao J. Integration of single-cell and bulk RNA-seq to establish a predictive signature based on the differentiation trajectory of M2 macrophages in lung adenocarcinoma. Front Genet 2022; 13:1010440. [PMID: 36171876 PMCID: PMC9510778 DOI: 10.3389/fgene.2022.1010440] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Tumor-associated macrophages as important members of the tumor microenvironment, are highly plastic and heterogeneous. TAMs can be classified into two preliminary subtypes: M1 and M2 macrophages. M2 macrophages are significantly associated with the progression of lung adenocarcinoma. However, no study has investigated the heterogeneity among M2 macrophages and their differentiation-related genes at the single-cell level to guide the clinical treatment of lung adenocarcinoma. Methods: Using the available annotation information from the Tumor Immune Single-cell Hub database, we clustered and annotated 12 lung adenocarcinoma samples using the R package ‘Seurat’. Subsequently, we extracted M2 macrophages for secondary clustering analysis and performed cell trajectory analysis using the R package ‘monocle2’. Based on heterogeneous genes associated with the differentiation trajectory of M2 macrophages, we established a prognostic lung adenocarcinoma model using Lasso-Cox and multivariate stepwise regression. In addition, we also performed immunotherapy and chemotherapy predictions. Results: M2 macrophages exhibit heterogeneity among themselves. M2 macrophages in different differentiation states showed significant differences in pathway activation and immune cell communication. Prognostic signature based on heterogeneous genes can be used to classify the prognostic status and abundance of immune cell infiltration in lung adenocarcinoma patients. In addition, the calculation of the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and the validation of the GSE126044 database indicated that lung adenocarcinoma patients with high-risk scores had poorer treatment outcomes when receiving immune checkpoint inhibitors treatment. Conclusion: Based on scRNA-seq and Bulk-seq data, we identified M2 macrophage-associated prognostic signature with a potential clinical utility to improve precision therapy.
Collapse
Affiliation(s)
- Zhike Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yu Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weibiao Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yiling Bai
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Cheng Ding
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chun Xu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chang Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Sheng Ju
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Sheng Ju, ; Lijuan Tang, ; Jun Zhao,
| | - Lijuan Tang
- Dalian Medical University, Dalian, Liaoning, China
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- *Correspondence: Sheng Ju, ; Lijuan Tang, ; Jun Zhao,
| | - Jun Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- *Correspondence: Sheng Ju, ; Lijuan Tang, ; Jun Zhao,
| |
Collapse
|
49
|
Transcription Factor MAFB as a Prognostic Biomarker for the Lung Adenocarcinoma. Int J Mol Sci 2022; 23:ijms23179945. [PMID: 36077342 PMCID: PMC9456510 DOI: 10.3390/ijms23179945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/18/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
MAFB is a basic leucine zipper (bZIP) transcription factor specifically expressed in macrophages. We have previously identified MAFB as a candidate marker for tumor-associated macrophages (TAMs) in human and mouse models. Here, we analyzed single-cell sequencing data of patients with lung adenocarcinoma obtained from the GEO database (GSE131907). Analyzed data showed that general macrophage marker CD68 and macrophage scavenger receptor 1 (CD204) were expressed in TAM and lung tissue macrophage clusters, while transcription factor MAFB was expressed specifically in TAM clusters. Clinical records of 120 patients with lung adenocarcinoma stage I (n = 57), II (n = 21), and III (n = 42) were retrieved from Tsukuba Human Tissue Biobank Center (THB) in the University of Tsukuba Hospital, Japan. Tumor tissues from these patients were extracted and stained with anti-human MAFB antibody, and then MAFB-positive cells relative to the tissue area (MAFB+ cells/tissue area) were morphometrically quantified. Our results indicated that higher numbers of MAFB+ cells significantly correlated to increased local lymph node metastasis (nodal involvement), high recurrence rate, poor pathological stage, increased lymphatic permeation, higher vascular invasion, and pleural infiltration. Moreover, increased amounts of MAFB+ cells were related to poor overall survival and disease-free survival, especially in smokers. These data indicate that MAFB may be a suitable prognostic biomarker for smoker lung cancer patients.
Collapse
|
50
|
Wu J, Liu X, Wu J, Lou C, Zhang Q, Chen H, Yang Z, Long S, Wang Y, Shang Z, Hu Z, Zhang R, Zhang J, Zeng Z. CXCL12 derived from CD248-expressing cancer-associated fibroblasts mediates M2-polarized macrophages to promote nonsmall cell lung cancer progression. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166521. [PMID: 35985448 DOI: 10.1016/j.bbadis.2022.166521] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/09/2022] [Accepted: 08/11/2022] [Indexed: 12/23/2022]
Abstract
Nonsmall cell lung cancer (NSCLC) is among the most prevalent malignant tumours threatening human health. In the tumour microenvironment (TME), cancer-associated fibroblasts (CAFs) induce M2-polarized macrophages, which strongly regulate tumour progression. However, little is known about the association between CAFs and M2 macrophages. CD248 is a transmembrane glycoprotein found in several cancer cells, tumour stromal cells, and pericytes. Here, we isolated CAFs from tumour tissues of NSCLC patients to detect the relationship between CD248 expression and patient prognosis. We knocked down the expression of CD248 on CAFs to detect CXCL12 secretion and macrophage polarization. We then examined the effects of CD248-expressing CAF-induced M2 macrophage polarization to promote NSCLC progression in vitro and in vivo. We found that CD248 is expressed mainly in NSCLC-derived CAFs and that the expression of CD248 correlates with poor patient prognosis. Blocking CXCL12 receptor (CXCR4) drastically decreased M2 macrophage chemotaxis. CD248 promotes CAFs secreting CXCL12 to mediate M2-polarized macrophages to promote NSCLC progression both in vitro and in vivo. Collectively, our data suggest that CD248-positive CAFs induce NSCLC progression by mediating M2-polarized macrophages.
Collapse
Affiliation(s)
- Jieheng Wu
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China; Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, China.
| | - Xinlei Liu
- Guizhou Prenatal Diagnsis Center, The Affiliated Hospital of Guizhou Medical University, 550001 Guiyang, China
| | - Jiangwei Wu
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China
| | - Chunju Lou
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China
| | - Qiaoling Zhang
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China
| | - Huiping Chen
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China
| | - Zeyang Yang
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China
| | - Shiqi Long
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China; Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, China
| | - Yun Wang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, China
| | - Zhenling Shang
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China
| | - Zuquan Hu
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, China
| | - Rui Zhang
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China; Department of Immunology, The Fourth Military Medical University, Xi'an 710032, China
| | - Jian Zhang
- Department of Thoracic Surgery, The Affiliated Hospital of Guizhou Medical University, 550001 Guiyang, China.
| | - Zhu Zeng
- Department of Immunology, Guizhou Medical University, 550025 Guiyang, China; Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang 550025, China; Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|