1
|
Okamoto T, Mizuta R, Demachi-Okamura A, Muraoka D, Sasaki E, Masago K, Yamaguchi R, Teramukai S, Otani Y, Date I, Tanaka S, Takahashi Y, Hashimoto N, Matsushita H. Immune prognostic model for glioblastoma based on the ssGSEA enrichment score. Cancer Genet 2025; 294-295:32-41. [PMID: 40121844 DOI: 10.1016/j.cancergen.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 03/16/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE Few effective immune prognostic models based on the tumor immune microenvironment (TIME) for glioblastoma have been reported. Therefore, this study aimed to construct an immune prognostic model for glioblastoma by analyzing enriched biological processes and pathways in tumors. METHODS A comprehensive single-sample gene set enrichment analysis (ssGSEA) of gene sets from the Molecular Signatures Database was performed using TCGA RNA sequencing data (141 glioblastoma cases). After evaluating gene sets associated with prognosis using univariable Cox regression, gene sets related to biological processes and tumor immunity in gliomas were extracted. Finally, the least absolute shrinkage and selection operator Cox regression refined the gene sets and a nomogram was constructed. The model was validated using CGGA (183 cases) and Aichi Cancer Center (42 cases) datasets. RESULTS The immune prognostic model consisted of three gene sets related to biological processes (sphingolipids, steroid hormones, and intermediate filaments) and one related to tumor immunity (immunosuppressive chemokine pathways involving tumor-associated microglia and macrophages). Kaplan-Meier curves for the training (TCGA) and validation (CGGA) cohorts showed significantly worse overall survival in the high-risk group compared to the low-risk group (p < 0.001 and p = 0.04, respectively). Furthermore, in silico cytometry revealed a significant increase in macrophages with immunosuppressive properties and T cells with effector functions in the high-risk group (p < 0.01) across all cohorts. CONCLUSION Construction of an immune prognostic model based on the TIME assessment using ssGSEA could potentially provide valuable insights into the prognosis and immune profiles of patients with glioblastoma and guide treatment strategies.
Collapse
Affiliation(s)
- Takanari Okamoto
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan; Department of Neurosurgery, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan.
| | - Ryo Mizuta
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan; Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Ayako Demachi-Okamura
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Daisuke Muraoka
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Eiichi Sasaki
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Katsuhiro Masago
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center, Nagoya, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Satoshi Teramukai
- Department of Biostatistics, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Yoshihiro Otani
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Shota Tanaka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshinobu Takahashi
- Department of Neurosurgery, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Naoya Hashimoto
- Department of Neurosurgery, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| |
Collapse
|
2
|
Zhou H, Zheng Z, Fan C, Zhou Z. Mechanisms and strategies of immunosenescence effects on non-small cell lung cancer (NSCLC) treatment: A comprehensive analysis and future directions. Semin Cancer Biol 2025; 109:44-66. [PMID: 39793777 DOI: 10.1016/j.semcancer.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Non-small cell lung cancer (NSCLC), the most prevalent form of lung cancer, remains a leading cause of cancer-related mortality worldwide, particularly among elderly individuals. The phenomenon of immunosenescence, characterized by the progressive decline in immune cell functionality with aging, plays a pivotal role in NSCLC progression and contributes to the diminished efficacy of therapeutic interventions in older patients. Immunosenescence manifests through impaired immune surveillance, reduced cytotoxic responses, and increased chronic inflammation, collectively fostering a pro-tumorigenic microenvironment. This review provides a comprehensive analysis of the molecular, cellular, and genetic mechanisms of immunosenescence and its impact on immune surveillance and the tumor microenvironment (TME) in NSCLC. We explore how aging affects various immune cells, including T cells, B cells, NK cells, and macrophages, and how these changes compromise the immune system's ability to detect and eliminate tumor cells. Furthermore, we address the challenges posed by immunosenescence to current therapeutic strategies, particularly immunotherapy, which faces significant hurdles in elderly patients due to immune dysfunction. The review highlights emerging technologies, such as single-cell sequencing and CRISPR-Cas9, which offer new insights into immunosenescence and its potential as a therapeutic target. Finally, we outline future research directions, including strategies for rejuvenating the aging immune system and optimizing immunotherapy for older NSCLC patients, with the goal of improving treatment efficacy and survival outcomes. These efforts hold promise for the development of more effective, personalized therapies for elderly patients with NSCLC.
Collapse
Affiliation(s)
- Huatao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| |
Collapse
|
3
|
Huang J, Wang B, Wu Q, Wang L, Guan C. LncRNA EGFR‑AS1 promotes lung cancer cell proliferation, invasion and metastasis via regulation of miR‑449a/HDAC1. Exp Ther Med 2025; 29:27. [PMID: 39720671 PMCID: PMC11667440 DOI: 10.3892/etm.2024.12777] [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: 02/06/2024] [Accepted: 05/10/2024] [Indexed: 12/26/2024] Open
Abstract
There is increasing evidence that long non-coding (lnc)RNA EGFR-AS1 is involved in the development of numerous types of cancer, including non-small-cell lung cancer (NSCLC). The Cancer Genome Atlas (TCGA) demonstrates that EGFR-AS1 is highly expressed in NSCLC. Downregulation of EGFR-AS1 in A549 and PC9 NSCLC cells demonstrates inhibition of NSCLC proliferation, invasion and metastasis. The present study demonstrated that lncRNA EGFR-AS1 was essential for the development of NSCLC through its function as a competitive endogenous RNA binding to miR-449a and upregulating histone deacetylase 1. In brief, it identified a novel signaling pathway that mediated the invasion and metastasis of NSCLC and may therefore provide a new treatment target for NSCLC.
Collapse
Affiliation(s)
- Jie Huang
- Department of Respiratory Medicine, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Xuhui, Shanghai 200031, P.R. China
| | - Baoqing Wang
- Department of Respiratory Medicine, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Xuhui, Shanghai 200031, P.R. China
- Department of Respiratory Medicine, Zhongshan Hospital, Fudan University, Xuhui, Shanghai 200031, P.R. China
| | - Qin Wu
- Department of Respiratory Medicine, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Xuhui, Shanghai 200031, P.R. China
| | - Liming Wang
- Department of Respiratory Medicine, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Xuhui, Shanghai 200031, P.R. China
| | - Chao Guan
- Department of Respiratory Medicine, Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Fudan University, Xuhui, Shanghai 200031, P.R. China
| |
Collapse
|
4
|
Zhang Y, Zhou J, Jin Y, Liu C, Zhou H, Sun Y, Jiang H, Gan J, Zhang C, Lu Q, Chang Y, Zhang Y, Li X, Ning S. Single-Cell and Bulk Transcriptomics Reveal the Immunosenescence Signature for Prognosis and Immunotherapy in Lung Cancer. Cancers (Basel) 2024; 17:85. [PMID: 39796714 PMCID: PMC11720133 DOI: 10.3390/cancers17010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Immunosenescence is the aging of the immune system, which is closely related to the development and prognosis of lung cancer. Targeting immunosenescence is considered a promising therapeutic approach. METHODS We defined an immunosenescence gene set (ISGS) and examined it across 33 TCGA tumor types and 29 GTEx normal tissues. We explored the 46,993 single cells of two lung cancer datasets. The immunosenescence risk model (ISRM) was constructed in TCGA LUAD by network analysis, immune infiltration analysis, and lasso regression and validated by survival analysis, cox regression, and nomogram in four lung cancer cohorts. The predictive ability of ISRM for drug response and immunotherapy was detected by the oncopredict algorithm and XGBoost model. RESULTS We found that senescent lung tissues were significantly enriched in ISGS and revealed the heterogeneity of immunosenescence in pan-cancer. Single-cell and bulk transcriptomics characterized the distinct immune microenvironment between old and young lung cancer. The ISGS network revealed the crucial function modules and transcription factors. Multiplatform analysis revealed specific associations between immunosenescence and the tumor progression of lung cancer. The ISRM consisted of five risk genes (CD40LG, IL7, CX3CR1, TLR3, and TLR2), which improved the prognostic stratification of lung cancer across multiple datasets. The ISRM showed robustness in immunotherapy and anti-tumor therapy. We found that lung cancer patients with a high-risk score showed worse survival and lower expression of immune checkpoints, which were resistant to immunotherapy. CONCLUSIONS Our study performed a comprehensive framework for assessing immunosenescence levels and provided insights into the role of immunosenescence in cancer prognosis and biomarker discovery.
Collapse
Affiliation(s)
- Yakun Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Jiajun Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Yitong Jin
- The Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China;
| | - Chenyu Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Hanxiao Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Yue Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Han Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Jing Gan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Caiyu Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Qianyi Lu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Yetong Chang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (Y.Z.); (J.Z.); (C.L.); (H.Z.); (Y.S.); (H.J.); (J.G.); (C.Z.); (Q.L.); (Y.C.); (Y.Z.)
| |
Collapse
|
5
|
Bonis A, Lunardi F, Pagliarini G, Verzeletti V, Lione L, Busetto A, Cannone G, Comacchio GM, Mammana M, Faccioli E, Rebusso A, Schiavon M, Nicotra S, Dell’Amore A, Rea F. Peripheral Circulating Blood Cells Deviation Based on Tumor Inflammatory Microenvironment Activity in Resected Upstaged Lung Adenocarcinomas. J Clin Med 2024; 13:7597. [PMID: 39768520 PMCID: PMC11676159 DOI: 10.3390/jcm13247597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The tumour inflammatory microenvironment (TIME) reflects a selective activation of the central immune system (IS), particularly T-cells expansion, which leads to immune cells migrating to the target, such as lung cancer, via the bloodstream and lymphatic vessels. In this study, the aim is to investigate whether the distribution of peripheral blood cells varies based on the immune status of patients with lung adenocarcinoma. Methods: This is a single-center retrospective study conducted in the Thoracic Surgery Unit of the University of Padua (Italy) between 1 January 2016 and 1 April 2024. It included patients (>18 years old) with lung adenocarcinoma deemed resectable (cT2bN0M0 or lower) who experienced pathological upstaging (IIB or higher). Patients were classified as TIME-active (with tumour-infiltrating lymphocytes-TILs and/or PD-L1 expression) or TIME-silent (without TILs or PD-L1). According to the TIME status, peripheral blood cell counts with clinical and pathological data were compared between groups using the Fisher's, Pearson's or Wilcoxon's test when appropriate. A Kaplan-Meier estimator investigated overall survival (OS) and recurrence-free survival (RFS) adopting the log-rank test. Results: Preoperatively, the TIME-a group demonstrated a significantly higher lymphocyte count (p = 0.02) and a lower absolute neutrophil rate (p = 0.01) than TIME-s. These differences persisted after resection (p = 0.06 and p = 0.02) while they became similar one month after surgery (p = 1 and p = 0.32). The neutrophil-to-lymphocyte ratio-NLR showed similar trends (p = 0.01 and p = 1). Better OS and RFS were shown in the TIME-a group (p = 0.02 and 0.03, respectively). Conclusions: Resected upstaged lung adenocarcinomas show distinct peripheral blood cell profiles based on immune status. TIME-active patients had a significantly lower NLR, which normalized post-surgery. Surgical resection may help restore native immune surveillance.
Collapse
Affiliation(s)
- Alessandro Bonis
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Francesca Lunardi
- Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Giulia Pagliarini
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Vincenzo Verzeletti
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Luigi Lione
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Alberto Busetto
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Giorgio Cannone
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Giovanni Maria Comacchio
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Marco Mammana
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Eleonora Faccioli
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Alessandro Rebusso
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Marco Schiavon
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Samuele Nicotra
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Andrea Dell’Amore
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| | - Federico Rea
- Thoracic Surgery Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health—DSCTV, University of Padova, 35128 Padova, Italy
| |
Collapse
|
6
|
Fay M, Clavijo PE, Allen CT. Heterogeneous characterization of neutrophilic cells in head and neck cancers. Head Neck 2024; 46:2591-2599. [PMID: 38622975 PMCID: PMC11473716 DOI: 10.1002/hed.27774] [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: 12/08/2023] [Revised: 03/14/2024] [Accepted: 04/07/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Neutrophilic cells are among the most abundant immune populations within the head and neck tumor microenvironment (TME) and harbor multiple mechanisms of immunosuppression. Despite these important features, neutrophilic cells may be underrepresented in contemporary studies that aim to comprehensively characterize the immune landscape of the TME due to discrepancies in tissue processing and analysis techniques. Here, we review the role of pathologically activated neutrophilic cells within the TME and pitfalls of various approaches used to study their frequency and function in clinical samples. METHODS The literature was identified by searching PubMed for "immune landscape" and "tumor immune microenvironment" in combination with keywords describing solid tumor malignancies. Key publications that assessed the immune composition of solid tumors derived from human specimens were included. The tumor and blood processing methodologies in each study were reviewed in depth and correlated with the reported abundance of neutrophilic cells. RESULTS Neutrophilic cells do not survive cryopreservation, and many studies fail to identify and study neutrophilic cell populations due to cryopreservation of clinical samples for practical reasons. Additional single-cell transcriptomic studies filter out neutrophilic cells due to low transcriptional counts. CONCLUSIONS This report can help readers critically interpret studies aiming to comprehensively study the immune TME that fail to identify and characterize neutrophilic cells.
Collapse
Affiliation(s)
- Magdalena Fay
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Paul E. Clavijo
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Clint T. Allen
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Papadaki MA, Papadaki E, Chatziavraam S, Aggouraki D, Michaelidou K, Fotsitzoudis C, Vassilakopoulou M, Mavroudis D, Agelaki S. Prognostic Value of Fas/Fas Ligand Expression on Circulating Tumor Cells (CTCs) and Immune Cells in the Peripheral Blood of Patients with Metastatic Breast Cancer. Cancers (Basel) 2024; 16:2927. [PMID: 39272785 PMCID: PMC11393959 DOI: 10.3390/cancers16172927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
The Fas/Fas ligand (FasL) system is a major apoptosis-regulating pathway with a key role in tumor immune surveillance and metastasis. The expression of Fas/FasL on mammary tumor tissues holds prognostic value for breast cancer (BC) patients. We herein assessed Fas/FasL expression on circulating tumor cells (CTCs) and matched peripheral blood mononuclear cells (PBMCs) from 98 patients with metastatic BC receiving first-line treatment. Fas+, FasL+, and Fas+/FasL+ CTCs were identified in 88.5%, 92.3%, and 84.6% of CTC-positive patients, respectively. In addition, Fas+/FasL+, Fas-/FasL+, and Fas-/FasL- PBMCs were identified in 70.3%, 24.2%, and 5.5% of patients, respectively. A reduced progression-free survival (PFS) was revealed among CTC-positive patients (median PFS: 9.5 versus 13.4 months; p = 0.004), and specifically among those harboring Fas+/FasL+ CTCs (median PFS: 9.5 vs. 13.4 months; p = 0.009). On the other hand, an increased overall survival (OS) was demonstrated among patients with Fas+/FasL+ PBMCs rather than those with Fas-/FasL+ and Fas-/FasL- PBMCs (median OS: 35.7 vs. 25.9 vs. 14.4 months, respectively; p = 0.008). These data provide for the first time evidence on Fas/FasL expression on CTCs and PBMCs with significant prognostic value for patients with metastatic BC, thus highlighting the role of the Fas/FasL system in the peripheral immune response and metastatic progression of BC.
Collapse
Affiliation(s)
- Maria A Papadaki
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Eleni Papadaki
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Sofia Chatziavraam
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Despoina Aggouraki
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Kleita Michaelidou
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Charalampos Fotsitzoudis
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| | - Maria Vassilakopoulou
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
| | - Dimitrios Mavroudis
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| | - Sofia Agelaki
- Laboratory of Translational Oncology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| |
Collapse
|
8
|
Wang K, Chen X, Lin P, Wu J, Huang Q, Chen Z, Tian J, Wang H, Tian Y, Shi M, Qian M, Hui B, Zhu Y, Li L, Yao R, Bian H, Zhu P, Chen R, Chen L. CD147-K148me2-Driven Tumor Cell-Macrophage Crosstalk Provokes NSCLC Immunosuppression via the CCL5/CCR5 Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400611. [PMID: 38873823 PMCID: PMC11304266 DOI: 10.1002/advs.202400611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/15/2024] [Indexed: 06/15/2024]
Abstract
Immunosuppression is a major hallmark of tumor progression in non-small cell lung cancer (NSCLC). Cluster of differentiation 147 (CD147), an important pro-tumorigenic factor, is closely linked to NSCLC immunosuppression. However, the role of CD147 di-methylation in the immunosuppressive tumor microenvironment (TME) remains unclear. Here, di-methylation of CD147 at Lys148 (CD147-K148me2) is identified as a common post-translational modification (PTM) in NSCLC that is significantly associated with unsatisfying survival outcomes among NSCLC sufferers, especially those in the advanced stages of the disease. The methyltransferase NSD2 catalyzes CD147 to generate CD147-K148me2. Further analysis demonstrates that CD147-K148me2 reestablishes the immunosuppressive TME and promotes NSCLC progression. Mechanistically, this modification promotes the interaction between cyclophilin A (CyPA) and CD147, and in turn, increases CCL5 gene transcription by activating p38-ZBTB32 signaling, leading to increased NSCLC cell-derived CCL5 secretion. Subsequently, CD147-K148me2-mediated CCL5 upregulation facilitates M2-like tumor-associated macrophage (TAM) infiltration in NSCLC tissues via CCL5/CCR5 axis-dependent intercellular crosstalk between tumor cells and macrophages, which is inhibited by blocking CD147-K148me2 with the targeted antibody 12C8. Overall, this study reveals the role of CD147-K148me2-driven intercellular crosstalk in the development of NSCLC immunosuppression, and provides a potential interventional strategy for PTM-targeted NSCLC therapy.
Collapse
Affiliation(s)
- Ke Wang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Xiaohong Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Peng Lin
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Jiao Wu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Qiang Huang
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
- School of MedicineShanghai UniversityShanghai200444China
| | - Zhi‐Nan Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Jiale Tian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Hao Wang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ye Tian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Mingyan Shi
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Meirui Qian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Bengang Hui
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
- Department of Thoracic Surgery of Tangdu HospitalFourth Military Medical UniversityXi'an710038China
| | - Yumeng Zhu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ling Li
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Rui Yao
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Huijie Bian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ping Zhu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Ruo Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing HospitalFourth Military Medical UniversityXi'an710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
| | - Liang Chen
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesChina
- School of MedicineShanghai UniversityShanghai200444China
| |
Collapse
|
9
|
Sun Y, Qiu X, Zhou D, Ricciardi S, Shinohara S, Ma J. miR-16-5p specifically inhibits IFN-γ-regulated memory T helper cell differentiation in malignant pleural effusion of non-small cell lung cancer. Transl Lung Cancer Res 2024; 13:1727-1741. [PMID: 39118892 PMCID: PMC11304156 DOI: 10.21037/tlcr-24-505] [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: 06/12/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024]
Abstract
Background The mechanism for memory T helper (Th) cell differentiation in malignant pleural effusion (MPE) of non-small cell lung cancer (NSCLC) is poorly understood. MicroRNAs (miRNAs), as small non-coding RNA that regulate gene expression, play a crucial role in the regulation of memory Th cell differentiation. However, whether miRNAs can inhibit the differentiation of memory Th cells in MPE of NSCLC has not been reported. This study aimed to explore miR-16-5p specifically inhibits interferon-gamma (IFN-γ)-regulated memory Th cell differentiation in MPE of NSCLC. Methods A total of 30 patients with NSCLC and 30 age- and sex-matched patients, who were clinically diagnosed as benign pleural effusion (BPE) of lung disease and had not received any intervention, were collected. The expression of nucleic acids, miRNAs, and cytokines was detected by polymerase chain reaction (PCR), miRNA microarray, enzyme-linked immunosorbent assay (ELISA), flow cytometry, and western blotting. Results The expression of CD4+CD69+ T cells in NSCLC with MPE was lower than that in lung disease BPE. CD4+CD69+ T cells highly express CD45RO+ and mainly secrete anti-tumor cytokines IFN-γ, interleukin-2 (IL-2), and tumor necrosis factor-α (TNF-α). The expression of miR-16-5p in CD4+CD69+ CD45RO+ T cells in MPE was higher than that in BPE. Moreover, miR-16-5p can bind to both IFN-γ promoter and its 5'untranslated region (5'-UTR), suggesting that IFN-γ may be the target gene directly affected by miR-16-5p. IFN-γ also affects the differentiation of memory CD4+ T cells by regulating T-bet. Conclusions We believe that miR-16-5p may regulate the decrease of differentiation of naïve CD4+ T cells into memory CD4+CD69+ T cells through its target gene IFN-γ in MPE, thus reducing the number of cytokines that produce anti-tumor effects. It may be the main reason for the low response rate of lung cancer with MPE immunotherapy.
Collapse
Affiliation(s)
- Yi Sun
- Pediatric, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaofen Qiu
- State Key Laboratory of Oncology in South China, Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Dalei Zhou
- State Key Laboratory of Oncology in South China, Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Sara Ricciardi
- Unit of Thoracic Surgery, San Camillo Forlanini Hospital, Rome, Italy
- PhD Program University of Bologna, Bologna, Italy
| | - Shuichi Shinohara
- Department of Thoracic Surgery, Anjo Kosei Hospital, Anjo, Aichi, Japan
| | - Jiangjun Ma
- State Key Laboratory of Oncology in South China, Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| |
Collapse
|
10
|
Zhang F, Zou M, Bai C, Zhu M. Prognostic signature based on S100 calcium-binding protein family members for lung adenocarcinoma and its clinical significance. Comput Methods Biomech Biomed Engin 2024:1-17. [PMID: 39012268 DOI: 10.1080/10255842.2024.2376668] [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: 02/06/2024] [Accepted: 07/01/2024] [Indexed: 07/17/2024]
Abstract
The S100 family proteins (S100s) participate in multiple stages of tumorigenesis and are considered to have potential value as biomarkers for detecting and predicting various cancers. But the role of S100s in lung adenocarcinoma (LUAD) prognosis is elusive. Transcriptional data of LUAD patients were retrieved from TCGA, and relevant literature was extensively reviewed to collect S100 genes. Differential gene expression analysis was performed on the LUAD data, followed by intersection analysis between the differentially expressed genes (DEGs) and S100 genes. Unsupervised consensus clustering analysis identified two clusters. Significant variations in overall survival between the two clusters were shown by Kaplan-Meier analysis. DEGs between the two clusters were analyzed using Lasso regression and univariate/multivariate Cox regression analysis, leading to construction of an 11-gene prognostic signature. The signature exhibited stable and accurate predictive capability in TCGA and GEO datasets. Subsequently, we observed distinct immune cell infiltration, immunotherapy response, and tumor mutation characteristics in high and low-risk groups. Finally, small molecular compounds targeting prognostic genes were screened using CellMiner database, and molecular docking confirmed the binding of AMG-176, Estramustine, and TAK-632 with prognostic genes. In conclusion, we generated a prognostic signature with robust and reliable predictive ability, which may provide guidance for prognosis and treatment of LUAD.
Collapse
Affiliation(s)
- Fengshun Zhang
- Department of Pathology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Mi Zou
- Respiratory Department, The First Branch of The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunsheng Bai
- Academician Expert Workstation of Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, China
- Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, China
| | - Mengjiao Zhu
- Academician Expert Workstation of Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, China
- Zhejiang Luoxi Medical Technology Co., Ltd., Hangzhou, China
| |
Collapse
|
11
|
Yang Z, Liu X, Xu H, Teschendorff AE, Xu L, Li J, Fu M, Liu J, Zhou H, Wang Y, Zhang L, He Y, Lv K, Yang H. Integrative analysis of genomic and epigenomic regulation reveals miRNA mediated tumor heterogeneity and immune evasion in lower grade glioma. Commun Biol 2024; 7:824. [PMID: 38971948 PMCID: PMC11227553 DOI: 10.1038/s42003-024-06488-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/21/2024] [Indexed: 07/08/2024] Open
Abstract
The expression dysregulation of microRNAs (miRNA) has been widely reported during cancer development, however, the underling mechanism remains largely unanswered. In the present work, we performed a systematic integrative study for genome-wide DNA methylation, copy number variation and miRNA expression data to identify mechanisms underlying miRNA dysregulation in lower grade glioma. We identify 719 miRNAs whose expression was associated with alterations of copy number variation or promoter methylation. Integrative multi-omics analysis revealed four subtypes with differing prognoses. These glioma subtypes exhibited distinct immune-related characteristics as well as clinical and genetic features. By construction of a miRNA regulatory network, we identified candidate miRNAs associated with immune evasion and response to immunotherapy. Finally, eight prognosis related miRNAs were validated to promote cell migration, invasion and proliferation through in vitro experiments. Our study reveals the crosstalk among DNA methylation, copy number variation and miRNA expression for immune regulation in glioma, and could have important implications for patient stratification and development of biomarkers for immunotherapy approaches.
Collapse
Affiliation(s)
- Zhen Yang
- Center for Medical Research and Innovation of Pudong Hospital, and Intelligent Medicine Institute, Shanghai Medical College, Fudan University, 131 Dongan Road, Shanghai, 200032, China.
| | - Xiaocen Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, 241001, Anhui, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Lingjie Xu
- Emergency Department, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jingyi Li
- Department of Medical Cosmetology, Beijing Tiantan Hospital, Capital Medical University, 100070, Beijing, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
| | - Jun Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
| | - Hanyu Zhou
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, 241001, Anhui, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241001, Anhui, China
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
| | - Yingying Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
| | - Licheng Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
| | - Yungang He
- Shanghai Fifth People's Hospital, and Intelligent Medicine Institute, Shanghai Medical College, Fudan University, 131 Dongan Road, Shanghai, 200032, China
| | - Kun Lv
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, 241001, Anhui, China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241001, Anhui, China.
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China.
| | - Hui Yang
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, 241001, Anhui, China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241001, Anhui, China.
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China.
| |
Collapse
|
12
|
Sugita Y, Muraoka D, Demachi-Okamura A, Komuro H, Masago K, Sasaki E, Fukushima Y, Matsui T, Shinohara S, Takahashi Y, Nishida R, Takashima C, Yamaguchi T, Horio Y, Hashimoto K, Tanaka I, Hamana H, Kishi H, Miura D, Tanaka Y, Onoue K, Onoguchi K, Yamashita Y, Stratford R, Clancy T, Yamaguchi R, Kuroda H, Ishibashi H, Okubo K, Matsushita H. Candidate tumor-specific CD8 + T cell subsets identified in the malignant pleural effusion of advanced lung cancer patients by single-cell analysis. Oncoimmunology 2024; 13:2371556. [PMID: 38952674 PMCID: PMC11216099 DOI: 10.1080/2162402x.2024.2371556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 06/19/2024] [Indexed: 07/03/2024] Open
Abstract
Isolation of tumor-specific T cells and their antigen receptors (TCRs) from malignant pleural effusions (MPE) may facilitate the development of TCR-transduced adoptive cellular immunotherapy products for advanced lung cancer patients. However, the characteristics and markers of tumor-specific T-cells in MPE are largely undefined. To this end, to establish the phenotypes and antigen specificities of CD8+ T cells, we performed single-cell RNA and TCR sequencing of samples from three advanced lung cancer patients. Dimensionality reduction on a total of 4,983 CD8+ T cells revealed 10 clusters including naïve, memory, and exhausted phenotypes. We focused particularly on exhausted T cell clusters and tested their TCR reactivity against neoantigens predicted from autologous cancer cell lines. Four different TCRs specific for the same neoantigen and one orphan TCR specific for the autologous cell line were identified from one of the patients. Differential gene expression analysis in tumor-specific T cells relative to the other T cells identified CXCL13, as a candidate gene expressed by tumor-specific T cells. In addition to expressing CXCL13, tumor-specific T cells were present in a higher proportion of T cells co-expressing PDCD1(PD-1)/TNFRSF9(4-1BB). Furthermore, flow cytometric analyses in advanced lung cancer patients with MPE documented that those with high PD-1/4-1BB expression have a better prognosis in the subset of 57 adenocarcinoma patients (p = .039). These data suggest that PD-1/4-1BB co-expression might identify tumor-specific CD8+ T cells in MPE, which are associated with patients' prognosis. (233 words).
Collapse
Affiliation(s)
- Yusuke Sugita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Thoracic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Daisuke Muraoka
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Ayako Demachi-Okamura
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hiroyasu Komuro
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Katsuhiro Masago
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Eiichi Sasaki
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yasunori Fukushima
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Takuya Matsui
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Shuichi Shinohara
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yusuke Takahashi
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Reina Nishida
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Chieko Takashima
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Teppei Yamaguchi
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yoshitsugu Horio
- Department of Thoracic Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Kana Hashimoto
- Department of Respiratory Internal Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Ichidai Tanaka
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Hamana
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Hiroyuki Kishi
- Department of Immunology, Faculty of Medicine, Academic Assembly, University of Toyama, Toyama, Japan
| | - Daiki Miura
- AI Drug Development Division, NEC Corporation, Tokyo, Japan
| | - Yuki Tanaka
- AI Drug Development Division, NEC Corporation, Tokyo, Japan
| | - Kousuke Onoue
- AI Drug Development Division, NEC Corporation, Tokyo, Japan
| | | | | | | | - Trevor Clancy
- NEC OncoImmunity AS, Oslo Cancer Cluster, Oslo, Norway
| | - Rui Yamaguchi
- Division of Cancer System Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kuroda
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hironori Ishibashi
- Department of Thoracic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenichi Okubo
- Department of Thoracic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Immunogenomics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
13
|
Moerland JA, Liby KT. The Triterpenoid CDDO-Methyl Ester Reduces Tumor Burden, Reprograms the Immune Microenvironment, and Protects from Chemotherapy-Induced Toxicity in a Preclinical Mouse Model of Established Lung Cancer. Antioxidants (Basel) 2024; 13:621. [PMID: 38929060 PMCID: PMC11201246 DOI: 10.3390/antiox13060621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/17/2024] [Accepted: 05/18/2024] [Indexed: 06/28/2024] Open
Abstract
NRF2 activation protects epithelial cells from malignancy, but cancer cells can upregulate the pathway to promote survival. NRF2 activators including CDDO-Methyl ester (CDDO-Me) inhibit cancer in preclinical models, suggesting NRF2 activation in other cell types may promote anti-tumor activity. However, the immunomodulatory effects of NRF2 activation remain poorly understood in the context of cancer. To test CDDO-Me in a murine model of established lung cancer, tumor-bearing wildtype (WT) and Nrf2 knockout (KO) mice were treated with 50-100 mg CDDO-Me/kg diet, alone or combined with carboplatin/paclitaxel (C/P) for 8-12 weeks. CDDO-Me decreased tumor burden in an Nrf2-dependent manner. The combination of CDDO-Me plus C/P was significantly (p < 0.05) more effective than either drug alone, reducing tumor burden by 84% in WT mice. CDDO-Me reduced the histopathological grade of WT tumors, with a significantly (p < 0.05) higher proportion of low-grade tumors and a lower proportion of high-grade tumors. These changes were augmented by combination with C/P. CDDO-Me also protected WT mice from C/P-induced toxicity and improved macrophage and T cell phenotypes in WT mice, reducing the expression of CD206 and PD-L1 on macrophages, decreasing immunosuppressive FoxP3+ CD4+ T cells, and increasing activation of CD8+ T cells in a Nrf2-dependent manner.
Collapse
Affiliation(s)
- Jessica A. Moerland
- Department of Pharmacology and Toxicology, Michigan State University, 1355 Bogue Street, East Lansing, MI 48824, USA;
| | - Karen T. Liby
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA
| |
Collapse
|
14
|
Sui P, Liu X, Zhong C, Sha Z. Integrated single-cell and bulk RNA-Seq analysis enhances prognostic accuracy of PD-1/PD-L1 immunotherapy response in lung adenocarcinoma through necroptotic anoikis gene signatures. Sci Rep 2024; 14:10873. [PMID: 38740918 DOI: 10.1038/s41598-024-61629-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
In addition to presenting significant diagnostic and treatment challenges, lung adenocarcinoma (LUAD) is the most common form of lung cancer. Using scRNA-Seq and bulk RNA-Seq data, we identify three genes referred to as HMR, FAM83A, and KRT6A these genes are related to necroptotic anoikis-related gene expression. Initial validation, conducted on the GSE50081 dataset, demonstrated the model's ability to categorize LUAD patients into high-risk and low-risk groups with significant survival differences. This model was further applied to predict responses to PD-1/PD-L1 blockade therapies, utilizing the IMvigor210 and GSE78220 cohorts, and showed strong correlation with patient outcomes, highlighting its potential in personalized immunotherapy. Further, LUAD cell lines were analyzed using quantitative PCR (qPCR) and Western blot analysis to confirm their expression levels, further corroborating the model's relevance in LUAD pathophysiology. The mutation landscape of these genes was also explored, revealing their broad implication in various cancer types through a pan-cancer analysis. The study also delved into molecular subclustering, revealing distinct expression profiles and associations with different survival outcomes, emphasizing the model's utility in precision oncology. Moreover, the diversity of immune cell infiltration, analyzed in relation to the necroptotic anoikis signature, suggested significant implications for immune evasion mechanisms in LUAD. While the findings present a promising stride towards personalized LUAD treatment, especially in immunotherapy, limitations such as the retrospective nature of the datasets and the need for larger sample sizes are acknowledged. Prospective clinical trials and further experimental research are essential to validate these findings and enhance the clinical applicability of our prognostic model.
Collapse
Affiliation(s)
- Ping Sui
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
- Institute of Transfusion Medicine and Immunology, Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Xueping Liu
- Department of Pulmonary and Critical Care Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, Shandong, China
| | - Cheng Zhong
- Jiangmen Hospital of Traditional Chinese Medicine Affiliated to Jinan University, Jiangmen, 52900, China.
| | - Zhanming Sha
- Department of Anesthesiology, Shandong Provincial Third Hospital, Jinan, 250031, China.
| |
Collapse
|
15
|
Song L, Gong Y, Wang E, Huang J, Li Y. Unraveling the tumor immune microenvironment of lung adenocarcinoma using single-cell RNA sequencing. Ther Adv Med Oncol 2024; 16:17588359231210274. [PMID: 38606165 PMCID: PMC11008351 DOI: 10.1177/17588359231210274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 10/09/2023] [Indexed: 04/13/2024] Open
Abstract
Tumor immune microenvironment (TIME) and its indications for lung cancer patient prognosis and therapeutic response have become new hotspots in cancer research in recent years. Tumor cells, immune cells, various regulatory factors, and their interactions in the TIME have been suggested to commonly influence lung cancer development and therapeutic outcome. The heterogeneity of TIME is composed of dynamic immune-related components, including various cancer cells, immune cells, cytokine/chemokine environments, cytotoxic activity, or immunosuppressive factors. The specific composition of cell subtypes may facilitate or hamper the response to immunotherapy and influence patient prognosis. Various markers have been found to stratify the patient prognosis or predict the therapeutic outcome. In this article, we systematically reviewed the recent advancement of TIME studies in lung adenocarcinoma (LUAD) using single-cell RNA sequencing (scRNA-seq) techniques, with specific focuses on the roles of TIME in LUAD development, TIME heterogeneity, indications of TIME in patient prognosis and therapeutic response during immunotherapy and drug resistance. The main findings in TIME heterogeneity and relevant markers or models for prognosis stratification and response prediction have been summarized. We hope that this review provides an overview of TIME status in LUAD and an inspiration for future development of strategies and biomarkers in LUAD treatment.
Collapse
Affiliation(s)
- Lele Song
- Department of Oncology, Chinese PLA General Hospital, Beijing, P.R. China
| | - Yuan Gong
- Department of Gastroenterology, The Second Medical Center of the Chinese PLA General Hospital, Beijing, P.R. China
| | - Erpeng Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong province, P.R. China
| | - Jianchun Huang
- Department of Thoracic Surgery, The First Affiliated Hospital of Kunming Medical University. No. 295, Xichang Road, Wuhua District, Kunming, Yunnan Province 650032, P.R. China
| | - Yuemin Li
- Department of Oncology, Chinese PLA General Hospital. No.8, Dongdajie, Fengtai District, Beijing 100071, P.R. China
| |
Collapse
|
16
|
Rigamonti A, Viatore M, Polidori R, Rahal D, Erreni M, Fumagalli MR, Zanini D, Doni A, Putignano AR, Bossi P, Voulaz E, Alloisio M, Rossi S, Zucali PA, Santoro A, Balzano V, Nisticò P, Feuerhake F, Mantovani A, Locati M, Marchesi F. Integrating AI-Powered Digital Pathology and Imaging Mass Cytometry Identifies Key Classifiers of Tumor Cells, Stroma, and Immune Cells in Non-Small Cell Lung Cancer. Cancer Res 2024; 84:1165-1177. [PMID: 38315789 PMCID: PMC10982643 DOI: 10.1158/0008-5472.can-23-1698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/13/2023] [Accepted: 02/01/2024] [Indexed: 02/07/2024]
Abstract
Artificial intelligence (AI)-powered approaches are becoming increasingly used as histopathologic tools to extract subvisual features and improve diagnostic workflows. On the other hand, hi-plex approaches are widely adopted to analyze the immune ecosystem in tumor specimens. Here, we aimed at combining AI-aided histopathology and imaging mass cytometry (IMC) to analyze the ecosystem of non-small cell lung cancer (NSCLC). An AI-based approach was used on hematoxylin and eosin (H&E) sections from 158 NSCLC specimens to accurately identify tumor cells, both adenocarcinoma and squamous carcinoma cells, and to generate a classifier of tumor cell spatial clustering. Consecutive tissue sections were stained with metal-labeled antibodies and processed through the IMC workflow, allowing quantitative detection of 24 markers related to tumor cells, tissue architecture, CD45+ myeloid and lymphoid cells, and immune activation. IMC identified 11 macrophage clusters that mainly localized in the stroma, except for S100A8+ cells, which infiltrated tumor nests. T cells were preferentially localized in peritumor areas or in tumor nests, the latter being associated with better prognosis, and they were more abundant in highly clustered tumors. Integrated tumor and immune classifiers were validated as prognostic on whole slides. In conclusion, integration of AI-powered H&E and multiparametric IMC allows investigation of spatial patterns and reveals tissue relevant features with clinical relevance. SIGNIFICANCE Leveraging artificial intelligence-powered H&E analysis integrated with hi-plex imaging mass cytometry provides insights into the tumor ecosystem and can translate tumor features into classifiers to predict prognosis, genotype, and therapy response.
Collapse
Affiliation(s)
- Alessandra Rigamonti
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| | - Marika Viatore
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| | - Rebecca Polidori
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| | - Daoud Rahal
- Department of Pathology, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
| | - Marco Erreni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Maria Rita Fumagalli
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Damiano Zanini
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Andrea Doni
- Unit of Advanced Optical Microscopy, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Anna Rita Putignano
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
| | - Paola Bossi
- Department of Pathology, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
| | - Emanuele Voulaz
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
- Division of Thoracic Surgery, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Marco Alloisio
- Division of Thoracic Surgery, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Sabrina Rossi
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Paolo Andrea Zucali
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Armando Santoro
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
- Medical Oncology and Hematology Unit, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Vittoria Balzano
- Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Nisticò
- Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Alberto Mantovani
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Biomedical Science, Humanitas University, Pieve Emanuele, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Massimo Locati
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| | - Federica Marchesi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital; Rozzano (Milan), Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan; Milan, Italy
| |
Collapse
|
17
|
Occhiuto CJ, Liby KT. KEAP1-Mutant Lung Cancers Weaken Anti-Tumor Immunity and Promote an M2-like Macrophage Phenotype. Int J Mol Sci 2024; 25:3510. [PMID: 38542481 PMCID: PMC10970780 DOI: 10.3390/ijms25063510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 04/04/2024] Open
Abstract
Considerable advances have been made in lung cancer therapies, but there is still an unmet clinical need to improve survival for lung cancer patients. Immunotherapies have improved survival, although only 20-30% of patients respond to these treatments. Interestingly, cancers with mutations in Kelch-like ECH-associated protein 1 (KEAP1), the negative regulator of the nuclear factor erythroid 2-related factor 2 (NRF2) transcription factor, are resistant to immune checkpoint inhibition and correlate with decreased lymphoid cell infiltration. NRF2 is known for promoting an anti-inflammatory phenotype when activated in immune cells, but the study of NRF2 activation in cancer cells has not been adequately assessed. The objective of this study was to determine how lung cancer cells with constitutive NRF2 activity interact with the immune microenvironment to promote cancer progression. To assess, we generated CRISPR-edited mouse lung cancer cell lines by knocking out the KEAP1 or NFE2L2 genes and utilized a publicly available single-cell dataset through the Gene Expression Omnibus to investigate tumor/immune cell interactions. We show here that KEAP1-mutant cancers promote immunosuppression of the tumor microenvironment. Our data suggest KEAP1 deletion is sufficient to alter the secretion of cytokines, increase expression of immune checkpoint markers on cancer cells, and alter recruitment and differential polarization of immunosuppressive macrophages that ultimately lead to T-cell suppression.
Collapse
Affiliation(s)
- Christopher J. Occhiuto
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA;
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Karen T. Liby
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
18
|
Shi S, Chu Y, Liu H, Yu L, Sun D, Yang J, Tian G, Ji L, Zhang C, Lu X. Predictable regulation of survival by intratumoral microbe-immune crosstalk in patients with lung adenocarcinoma. MICROBIAL CELL (GRAZ, AUSTRIA) 2024; 11:29-40. [PMID: 38375207 PMCID: PMC10876218 DOI: 10.15698/mic2024.02.813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 02/21/2024]
Abstract
Intratumoral microbiota can regulate the tumor immune microenvironment (TIME) and mediate tumor prognosis by promoting inflammatory response or inhibiting anti-tumor effects. Recent studies have elucidated the potential role of local tumor microbiota in the development and progression of lung adenocarcinoma (LUAD). However, whether intratumoral microbes are involved in the TIME that mediates the prognosis of LUAD remains unknown. Here, we obtained the matched tumor microbiome and host transcriptome and survival data of 478 patients with LUAD in The Cancer Genome Atlas (TCGA). Machine learning models based on immune cell marker genes can predict 1- to 5-year survival with relative accuracy. Patients were stratified into high- and low-survival-risk groups based on immune cell marker genes, with significant differences in intratumoral microbial communities. Specifically, patients in the high-risk group had significantly higher alpha diversity (p < 0.05) and were characterized by an enrichment of lung cancer-related genera such as Streptococcus. However, network analysis highlighted a more active pattern of dominant bacteria and immune cell crosstalk in TIME in the low-risk group compared to the high-risk group. Our study demonstrated that intratumoral microbiota-immune crosstalk was strongly associated with prognosis in LUAD patients, which would provide new targets for the development of precise therapeutic strategies.
Collapse
Affiliation(s)
- Shuo Shi
- The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Yuwen Chu
- Geneis Beijing Co., Ltd., Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, Shandong, China
| | - Haiyan Liu
- College of Information Engineering, Changsha Medical University, Changsha 410219, Hunan, China
- Academician Workstation, Changsha Medical University, Changsha 410219, Hunan, China
| | - Lan Yu
- Clinical Medical Research Center, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Hohhot, Inner Mongolia, China
- Inner Mongolia Key Laboratory of Gene Regulation of The Metabolic Disease, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Hohhot, Inner Mongolia, China
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Hohhot, Inner Mongolia, China
| | - Dejun Sun
- Inner Mongolia Academy of Medical Sciences, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Hohhot, Inner Mongolia, China
- Pulmonary and Critical Care Medicine, Inner Mongolian People's Hospital, No. 20, Zhaowuda Road, Saihan District, Hohhot, Inner Mongolia, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd., Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, Shandong, China
- Academician Workstation, Changsha Medical University, Changsha 410219, Hunan, China
| | - Geng Tian
- Geneis Beijing Co., Ltd., Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, Shandong, China
| | - Lei Ji
- Geneis Beijing Co., Ltd., Beijing 100102, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, Shandong, China
| | - Cong Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine/No. 39, 12th Bridge Road, Jinniu District, Chengdu City, Sichuan Province, 610072, China
| | - Xinxin Lu
- Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research
| |
Collapse
|
19
|
Liu K, He S, Sun S, Zhang X, He Y, Quan F, Pang B, Xiao Y. Computational Quantification of Cancer Immunoediting. Cancer Immunol Res 2023; 11:1159-1167. [PMID: 37540180 DOI: 10.1158/2326-6066.cir-22-0926] [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: 11/21/2022] [Revised: 03/31/2023] [Accepted: 07/10/2023] [Indexed: 08/05/2023]
Abstract
The remarkable success of cancer immunotherapy has revolutionized cancer treatment, emphasizing the importance of tumor-immune interactions in cancer evolution and treatment. Cancer immunoediting describes the dual effect of tumor-immune interactions: inhibiting tumor growth by destroying tumor cells and facilitating tumor escape by shaping tumor immunogenicity. To better understand tumor-immune interactions, it is critical to develop computational methods to measure the extent of cancer immunoediting. In this review, we provide a comprehensive overview of the computational methods for quantifying cancer immunoediting. We focus on describing the basic ideas, computational processes, advantages, limitations, and influential factors. We also summarize recent advances in quantifying cancer immunoediting studies and highlight future research directions. As the methods for quantifying cancer immunoediting are continuously improved, future research will further help define the role of immunity in tumorigenesis and hopefully provide a basis for the design of new personalized cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Kun Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shengyuan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shangqin Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanzhen He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Fei Quan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Bo Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
20
|
Kudling TV, Clubb JH, Pakola S, Quixabeira DC, Lähdeniemi IA, Heiniö C, Arias V, Havunen R, Cervera-Carrascon V, Santos JM, Sutinen E, Räsänen J, Borenius K, Mäyränpää MI, Aaltonen E, Sorsa S, Hemminki O, Kanerva A, Verschuren EW, Ilonen I, Hemminki A. Effective intravenous delivery of adenovirus armed with TNFα and IL-2 improves anti-PD-1 checkpoint blockade in non-small cell lung cancer. Oncoimmunology 2023; 12:2241710. [PMID: 37546696 PMCID: PMC10399490 DOI: 10.1080/2162402x.2023.2241710] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
Lung cancer remains among the most difficult-to-treat malignancies and is the leading cause of cancer-related deaths worldwide. The introduction of targeted therapies and checkpoint inhibitors has improved treatment outcomes; however, most patients with advanced-stage non-small cell lung cancer (NSCLC) eventually fail these therapies. Therefore, there is a major unmet clinical need for checkpoint refractory/resistant NSCLC. Here, we tested the combination of aPD-1 and adenovirus armed with TNFα and IL-2 (Ad5-CMV-mTNFα/mIL-2) in an immunocompetent murine NSCLC model. Moreover, although local delivery has been standard for virotherapy, treatment was administered intravenously to facilitate clinical translation and putative routine use. We showed that treatment of tumor-bearing animals with aPD-1 in combination with intravenously injected armed adenovirus significantly decreased cancer growth, even in the presence of neutralizing antibodies. We observed an increased frequency of cytotoxic tumor-infiltrating lymphocytes, including tumor-specific cells. Combination treatment led to a decreased percentage of immunosuppressive tumor-associated macrophages and an improvement in dendritic cell maturation. Moreover, we observed expansion of the tumor-specific memory T cell compartment in secondary lymphoid organs in the group that received aPD-1 with the virus. However, although the non-replicative Ad5-CMV-mTNFα/mIL-2 virus allows high transgene expression in the murine model, it does not fully reflect the clinical outcome in humans. Thus, we complemented our findings using NSCLC ex vivo models fully permissive for the TNFα and IL-2- armed oncolytic adenovirus TILT-123. Overall, our data demonstrate the ability of systemically administered adenovirus armed with TNFα and IL-2 to potentiate the anti-tumor efficacy of aPD-1 and warrant further investigation in clinical trials.
Collapse
Affiliation(s)
- Tatiana V. Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - James H.A. Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dafne C.A. Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Iris A.K. Lähdeniemi
- Translational Lung Cancer Research Group, Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Joao M. Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Eva Sutinen
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pulmonary Medicine, Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Jari Räsänen
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristian Borenius
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko I. Mäyränpää
- Pathology, University of Helsinki and Helsinki University Hospital (HUSLAB), Helsinki, Finland
| | - Eero Aaltonen
- Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Otto Hemminki
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Gynecology and Obstetrics, Helsinki University Hospital, Helsinki, Finland
| | - Emmy W. Verschuren
- Translational Lung Cancer Research Group, Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Ilkka Ilonen
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
| |
Collapse
|
21
|
Komuro H, Shinohara S, Fukushima Y, Demachi-Okamura A, Muraoka D, Masago K, Matsui T, Sugita Y, Takahashi Y, Nishida R, Takashima C, Ohki T, Shigematsu Y, Watanabe F, Adachi K, Fukuyama T, Hamana H, Kishi H, Miura D, Tanaka Y, Onoue K, Onoguchi K, Yamashita Y, Stratford R, Clancy T, Yamaguchi R, Kuroda H, Doi K, Iwata H, Matsushita H. Single-cell sequencing on CD8 + TILs revealed the nature of exhausted T cells recognizing neoantigen and cancer/testis antigen in non-small cell lung cancer. J Immunother Cancer 2023; 11:e007180. [PMID: 37544663 PMCID: PMC10407349 DOI: 10.1136/jitc-2023-007180] [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: 07/10/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND CD8+tumor infiltrating lymphocytes (TILs) are often observed in non-small cell lung cancers (NSCLC). However, the characteristics of CD8+ TILs, especially T-cell populations specific for tumor antigens, remain poorly understood. METHODS High throughput single-cell RNA sequencing and single-cell T-cell receptor (TCR) sequencing were performed on CD8+ TILs from three surgically-resected lung cancer specimens. Dimensional reduction for clustering was performed using Uniform Manifold Approximation and Projection. CD8+ TIL TCR specific for the cancer/testis antigen KK-LC-1 and for predicted neoantigens were investigated. Differentially-expressed gene analysis, Gene Set Enrichment Analysis (GSEA) and single sample GSEA was performed to characterize antigen-specific T cells. RESULTS A total of 6998 CD8+ T cells was analyzed, divided into 10 clusters according to their gene expression profile. An exhausted T-cell (exhausted T (Tex)) cluster characterized by the expression of ENTPD1 (CD39), TOX, PDCD1 (PD1), HAVCR2 (TIM3) and other genes, and by T-cell oligoclonality, was identified. The Tex TCR repertoire (Tex-TCRs) contained nine different TCR clonotypes recognizing five tumor antigens including a KK-LC-1 antigen and four neoantigens. By re-clustering the tumor antigen-specific T cells (n=140), it could be seen that the individual T-cell clonotypes were present on cells at different stages of differentiation and functional states even within the same Tex cluster. Stimulating these T cells with predicted cognate peptide indicated that TCR signal strength and subsequent T-cell proliferation and cytokine production was variable but always higher for neoantigens than KK-LC-1. CONCLUSIONS Our approach focusing on T cells with an exhausted phenotype among CD8+ TILs may facilitate the identification of tumor antigens and clarify the nature of the antigen-specific T cells to specify the promising immunotherapeutic targets in patients with NSCLC.
Collapse
Affiliation(s)
- Hiroyasu Komuro
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of General Thoracic Surgery, Gifu University School of Medicine Graduate School of Medicine, Gifu, Japan
| | - Shuichi Shinohara
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yasunori Fukushima
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of General Thoracic Surgery, Gifu University School of Medicine Graduate School of Medicine, Gifu, Japan
| | - Ayako Demachi-Okamura
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Daisuke Muraoka
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Katsuhiro Masago
- Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Takuya Matsui
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yusuke Sugita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yusuke Takahashi
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Reina Nishida
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Chieko Takashima
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Takashi Ohki
- Department of Respiratory Surgery, Ichinomiya Nishi Hospital, Ichinomiya, Japan
| | - Yoshiki Shigematsu
- Department of Respiratory Surgery, Ichinomiya Nishi Hospital, Ichinomiya, Japan
| | - Fumiaki Watanabe
- Department of Thoracic Surgery, Mie Chuo Medical Center, Tsu, Japan
| | | | - Takashi Fukuyama
- Division of Biomedical Research, Kitasato University Medical Center, Kitamoto, Japan
| | - Hiroshi Hamana
- Department of Immunology, University of Toyama, Toyama, Japan
| | - Hiroyuki Kishi
- Department of Immunology, University of Toyama, Toyama, Japan
| | - Daiki Miura
- Drug Development Division, NEC Corporation, Minato-ku, Japan
| | - Yuki Tanaka
- Drug Development Division, NEC Corporation, Minato-ku, Japan
| | - Kousuke Onoue
- Drug Development Division, NEC Corporation, Minato-ku, Japan
| | | | | | | | - Trevor Clancy
- NEC OncoImmunity AS, Oslo Cancer Cluster, Oslo, Norway
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kuroda
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Kiyoshi Doi
- Department of General Thoracic Surgery, Gifu University School of Medicine Graduate School of Medicine, Gifu, Japan
| | - Hisashi Iwata
- Department of General Thoracic Surgery, Gifu University School of Medicine Graduate School of Medicine, Gifu, Japan
| | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cancer Immunogenomics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
22
|
Lim JU, Lee E, Lee SY, Cho HJ, Ahn DH, Hwang Y, Choi JY, Yeo CD, Park CK, Kim SJ. Current literature review on the tumor immune micro-environment, its heterogeneity and future perspectives in treatment of advanced non-small cell lung cancer. Transl Lung Cancer Res 2023; 12:857-876. [PMID: 37197639 PMCID: PMC10183402 DOI: 10.21037/tlcr-22-633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 02/07/2023] [Indexed: 03/21/2023]
Abstract
Background and Objective Immune checkpoint inhibitors (ICI) were a major clinical advancement that provided an opportunity to improve the prognosis of patients with non-small cell lung cancer (NSCLC). However, programmed death-ligand-1 (PD-L1) expression does not sufficiently predict ICI efficacy in NSCLC patients. In recent studies, the tumor immune microenvironment (TIME) was shown to have a central role in lung cancer progression and to affect clinical outcome of patients diagnosed with lung cancer. As development of new therapeutic targets to overcome ICI resistance is a priority, understanding the TIME is important. Recently, a series of studies was conducted to target each component of TIME to improve efficacy of cancer treatment. In this review, important features related to TIME, its heterogeneity and current trends in treatment targeting the component of TIME are discussed. Methods PubMed and PMC were searched from January 1st, 2012 to August 16th, 2022 using the following key words: "NSCLC", "Tumor microenvironment", "Immune", "Metastasis" and "Heterogeneity". Key Content and Findings Heterogeneity in the TIME can be either spatial or temporal. Subsequent to heterogeneous changes in the TIME, treatment of lung cancer can be more challenging because drug resistance is more likely to occur. In terms of the TIME, the main concept for increasing the chance of successful NSCLC treatment is to activate immune responses against tumor cells and inhibit immunosuppressive activities. In addition, relevant research is focused on normalizing an otherwise aberrant TIME in NSCLC patients. Potential therapeutic targets include immune cells, cytokine interactions, and non-immune cells such as fibroblasts or vessels. Conclusions In management of lung cancer, understanding TIME and its heterogeneity is significant to treatment outcomes. Ongoing trials including various treatment modalities such as radiotherapy, cytotoxic chemotherapy, and anti-angiogenic treatment and regimens inhibiting other immunoinhibitory molecules are promising.
Collapse
Affiliation(s)
- Jeong Uk Lim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eunyoung Lee
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang-Yun Lee
- Department of Biomedical Engineering, Gachon University, Seongnam, Republic of Korea
| | - Hyeong Jun Cho
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dong Hyuck Ahn
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yongki Hwang
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joon Young Choi
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Incheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chan Kwon Park
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Joon Kim
- Division of Pulmonology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Postech-Catholic Biomedical Engineering Institute, Songeui Multiplex Hall, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
23
|
Miller AR. Checkpoint inhibitors are a basic science-based, transformative new treatment for lung cancer. Respirology 2023; 28:101-106. [PMID: 36535890 PMCID: PMC10107528 DOI: 10.1111/resp.14437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/04/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Alistair R Miller
- Department of Respiratory and Sleep Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia.,Department of Internal Medicine, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia.,Department of Medicine (RMH), Faculty of Medicine Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|