1
|
Wang X, Wang Z, Liu Z, Huang F, Pan Z, Zhang Z, Liu T. Nutritional strategies in oncology: The role of dietary patterns in modulating tumor progression and treatment response. Biochim Biophys Acta Rev Cancer 2025; 1880:189322. [PMID: 40228747 DOI: 10.1016/j.bbcan.2025.189322] [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: 02/10/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Dietary interventions can influence tumor growth by restricting tumor-specific nutritional requirements, altering the nutrient availability in the tumor microenvironment, or enhancing the cytotoxicity of anticancer drugs. Metabolic reprogramming of tumor cells, as a significant hallmark of tumor progression, has a profound impact on immune regulation, severely hindering tumor eradication. Dietary interventions can modify tumor metabolic processes to some extent, thereby further improving the efficacy of tumor treatment. In this review, we emphasize the impact of dietary patterns on tumor progression. By exploring the metabolic differences of nutrients in normal cells versus cancer cells, we further clarify how dietary patterns influence cancer treatment. We also discuss the effects of dietary patterns on traditional treatments such as immunotherapy, chemotherapy, radiotherapy, and the gut microbiome, thereby underscoring the importance of precision nutrition.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zeyao Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zihan Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Fanxuan Huang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zhaoyu Pan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Hunan, China
| | - Zhiren Zhang
- Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China; Departments of Cardiology and Pharmacy and Breast Cancer surgery, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, China.
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China; Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.
| |
Collapse
|
2
|
Saeed Issa B, Adhab AH, Salih Mahdi M, Kyada A, Ganesan S, Bhanot D, Naidu KS, Kaur S, Mansoor AS, Radi UK, Saadoun Abd N, Kariem M. Decoding the complex web: cellular and molecular interactions in the lung tumour microenvironment. J Drug Target 2025; 33:666-690. [PMID: 39707828 DOI: 10.1080/1061186x.2024.2445772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
The lung tumour microenvironment (TME) or stroma is a dynamic space of numerous cells and their released molecules. This complicated web regulates tumour progression and resistance to different modalities. Lung cancer cells in conjunction with their stroma liberate a wide range of factors that dampen antitumor attacks by innate immunity cells like natural killer (NK) cells and also adaptive responses by effector T cells. These factors include numerous growth factors, exosomes and epigenetic regulators, and also anti-inflammatory cytokines. Understanding the intricate interactions between tumour cells and various elements within the lung TME, such as immune and stromal cells can help provide novel strategies for better management and treatment of lung malignancies. The current article discusses the complex network of cells and signalling molecules, which mediate communications in lung TME. By elucidating these multifaceted interactions, we aim to provide insights into potential therapeutic targets and strategies for lung cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Deepak Bhanot
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sharnjeet Kaur
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | | | - Usama Kadem Radi
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Nasr Saadoun Abd
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
3
|
Xu W, Fang H, Cao X, Xu MZ, Yan Y, Shen M, Yang Y, Jiang K. NADH:ubiquinone oxidoreductase core subunit S8 expression and functional significance in non-small cell lung cancer. Cell Death Dis 2025; 16:321. [PMID: 40258810 PMCID: PMC12012183 DOI: 10.1038/s41419-025-07638-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/23/2025]
Abstract
Hyperfunctional mitochondria provide a growth advantage by supporting the energy-intensive processes essential for non-small cell lung cancer (NSCLC). NADH:ubiquinone oxidoreductase core subunit S8 (NDUFS8) is a key subunit of mitochondrial complex I involved in oxidative phosphorylation (OXPHOS) and cellular energy production. Bioinformatics and local tissue examinations show that NDUFS8 expression is elevated in NSCLC compared to normal lung tissue. Both immortalized and primary human NSCLC cells exhibit higher NDUFS8 levels. Single-cell RNA sequencing confirmed NDUFS8 upregulation in cancerous cells of NSCLC tumor. Silencing NDUFS8 via shRNA or Cas9/sgRNA-mediated knockout (KO) disrupted mitochondrial functions, leading to decreased complex I activity, ATP depletion, mitochondrial depolarization, increased reactive oxygen species (ROS) production, and heightened lipid peroxidation. Furthermore, NDUFS8 silencing/KO triggered apoptosis and significantly reduced Akt-mTOR activation, cell viability, proliferation, and motility in various NSCLC cells. In contrast, ectopic overexpression of NDUFS8 boosted mitochondrial complex I activity and ATP levels, promoting Akt-mTOR activation, and enhancing NSCLC cell proliferation and motility. NDUFS8 also contributes to radioresistance in NSCLC; silencing or KO enhanced ionizing radiation (IR)-induced cytotoxicity, while overexpression mitigated it. Intratumoral injection of NDUFS8 shRNA-expressing adeno-associated virus significantly inhibited growth of primary NSCLC xenografts in nude mice, with observed NDUFS8 silencing, ATP reduction, oxidative damage, proliferation inhibition, Akt-mTOR inactivation and apoptosis in treated tissues. These findings highlight the pivotal pro-tumorigenic role of NDUFS8 in NSCLC.
Collapse
Affiliation(s)
- Weihua Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongpeng Fang
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Xianbao Cao
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Min-Zhao Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yubo Yan
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Mingjing Shen
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yi Yang
- Department of Nuclear Medicine, the Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, China.
| | - Kanqiu Jiang
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
4
|
Yang L, Wei W, Yuan X, Guo E, Peng P, Wang J, Sun W. Targeting DNA Damage Repair to Enhance Antitumor Immunity in Radiotherapy: Mechanisms and Opportunities. Int J Mol Sci 2025; 26:3743. [PMID: 40332379 PMCID: PMC12027993 DOI: 10.3390/ijms26083743] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025] Open
Abstract
Radiotherapy is a standard cancer treatment that involves the induction of DNA damage. DNA damage repair (DDR) pathways maintain genomic integrity and make tumors resistant to radiotherapy and certain chemotherapies. In turn, DDR dysfunction results in cumulative DNA damage, leading to increased sensitivity for antitumor treatment. Moreover, radiotherapy has been shown to trigger antitumor immunity. Currently, immunotherapy has become a new and widely used standard strategy for treating a broad spectrum of tumor types. Notably, recent studies have demonstrated that DDR pathways play important roles in driving the response to immunotherapy. Herein, we review and discuss how DDR affects antitumor immunity induced by radiotherapy. Furthermore, we summarize the development of strategies for combining DDR inhibitors with radiotherapy and/or immunotherapy to enhance their efficacy against cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (L.Y.); (W.W.); (X.Y.); (E.G.); (P.P.); (J.W.)
| |
Collapse
|
5
|
Tao Y, Ji H, Hu W, Jiang G, Yang F, Peng X, Zhang X, Yin Y, Yuan Z, Chen D. SMARCC1 promotes M2 macrophage polarization and reduces ferroptosis in lung cancer by activating FLOT1 transcription. J Mol Med (Berl) 2025; 103:453-467. [PMID: 40108025 DOI: 10.1007/s00109-025-02531-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Grounded on the bioinformatics insights, this study explores the role of flotillin 1 (FLOT1) in modulating macrophage phenotype and immune evasion in lung cancer cells. The bioinformatics analyses revealed positive correlations between FLOT1 expression and infiltration of M2 macrophages, neutrophils, dendritic cells, and CD4 memory T cells. Furthermore, elevated FLOT1 expression was associated with a poor prognosis in lung cancer patients. Analysis of tumor and adjacent non-tumor tissues from 53 lung cancer patients revealed significantly higher immunohistochemical staining of FLOT1 in tumor tissues, showing positive correlation with the staining intensity of PD-L1. Additionally, staining intensities for markers of M2 macrophages (Arg1), CD4 memory T cells (CD4), dendritic cells (CD83), and neutrophils (CD177) were significantly higher in tumor tissues with high FLOT1 levels. Silencing of FLOT1 was induced in two lung cancer cell lines. Co-culturing in conditioned media of the FLOT1-silenced cancer cells led to reduced chemotactic migration and M2 skewing of macrophages in vitro. Using xenograft models, we observed that FLOT1 silencing weakened tumorigenic activity of A549 cells in mice and reduced M2 macrophage infiltration in tumors. SWI/SNF related BAF chromatin remodeling complex subunit C1 (SMARCC1) was identified as a transcription factor that activated FLOT1 transcription by binding to its promoter. Knockdown of SMARCC1 in lung cancer cells similarly reduced the migration and M2 polarization of macrophages as well as weakened tumorigenesis in mice. However, these effects were counteracted by FLOT1 overexpression. Further analysis of the downstream effectors of the SMARCC1/FLOT1 cascade revealed the enrichment of these factors in ferroptosis-related pathways. Mechanistically, SMARCC1 knockdown led to a decreased GSH:GSSG ratio and increased lipid peroxidation in macrophages, while FLOT1 overexpression restored these changes. Transmission electron microscopic observation revealed typical features of ferroptosis-resistant mitochondria following SMARCC1 knockdown, including fragmented or reduced cristae and increased outer membrane integrity. These mitochondrial changes were mitigated by FLOT1 overexpression. In conclusion, SMARCC1 promotes immune evasion in lung cancer by activating FLOT1 transcription. This activation enhances recruitment and M2 polarization of macrophages, and increases PD-L1 expression, reduces ferroptosis. These findings provide valuable insights into the molecular mechanisms of immune evasion and suggest potential therapeutic targets for lung cancer treatment. KEY MESSAGES: • FLOT1 is associated with poor prognosis in lung cancer patients. • Association between FLOT1 and immune cell infiltration in lung cancer. • Silencing FLOT1 inhibits the recruitment of macrophages by lung cancer cells. • SMARCC1 is highly expressed in lung cancer and promotes the transcription of FLOT1. • FLOT1 overexpression rescues the inhibitory effect of SMARCC1 knockdown on M2 macrophage infiltration and activation of Ferroptosis.
Collapse
Affiliation(s)
- Youliang Tao
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Huafeng Ji
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Wensheng Hu
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Guojun Jiang
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Fangding Yang
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Xu Peng
- Department of Orthopedics, The First People's Hospital of Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Xu Zhang
- Department of General Surgery, The First People's Hospital of Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Yuqin Yin
- Department of Nephrology, The First People's Hospital of Lin'an District, Hangzhou, 311300, China
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China
| | - Zhize Yuan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Rd, Shanghai, 200433, China.
| | - Dukai Chen
- Department of Thoracic Surgery, The First People's Hospital of Hangzhou Lin'an District, Hangzhou Medical College, No. 360, Yikang Street, Lin'an District, Hangzhou, 311300, China.
- Lin'an Peoples' Hospital Affiliated to Hangzhou Medical College, Hangzhou, 311300, China.
- Lin'an District of Zhejiang Provincial People's Hospital, Hangzhou, 311300, China.
| |
Collapse
|
6
|
Gao Y, Xie J, Yang Z, Li M, Yuan H, Li R. Functional tumor-derived exosomes in NSCLC progression and clinical implications. Front Pharmacol 2025; 16:1485661. [PMID: 40176898 PMCID: PMC11962733 DOI: 10.3389/fphar.2025.1485661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer cases and remains one of the leading causes of cancer-related mortality worldwide. The high mortality rate is primarily driven by delayed diagnosis, rapid metastasis, and frequent recurrence. Tumor-derived exosomes (TEXs) have emerged as critical mediators in NSCLC progression, offering valuable insights into the tumor microenvironment. Exosomes are small membrane vesicles that facilitate intercellular communication and transport bioactive molecules, including proteins, RNAs, and DNAs, thereby reflecting the genetic complexity of tumors. These exosomes play a key role in promoting tumor metastasis, epithelial-mesenchymal transition (EMT), neovascularization, drug resistance, and immune evasion, all of which are pivotal in the development of NSCLC. This review explores the diverse roles of TEXs in NSCLC progression, focusing on their involvement in pre-metastatic niche formation, tissue metastasis, and immune modulation. Specifically, we discuss the roles of exosome-associated RNAs and proteins in NSCLC, and their contribute to tumor growth and metastasis. Furthermore, we explore the potential of TEXs as biomarkers for NSCLC, emphasizing their application in diagnosis, prognosis, and prediction of resistance to targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Yuxin Gao
- Department of Abdominal Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Chengdu, China
- Information Technology Center, West China Sanya Hospital of Sichuan University, Sanya, China
| | - Zhenya Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Mengxi Li
- College of pharmacy, Chengdu Medical College, Chengdu, China
| | - Hongfan Yuan
- Department of Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
7
|
Kostecki KL, Harmon RL, Iida M, Harris MA, Crossman BE, Bruce JY, Salgia R, Wheeler DL. Axl Regulation of NK Cell Activity Creates an Immunosuppressive Tumor Immune Microenvironment in Head and Neck Cancer. Cancers (Basel) 2025; 17:994. [PMID: 40149328 PMCID: PMC11940164 DOI: 10.3390/cancers17060994] [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: 01/27/2025] [Revised: 03/07/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Head and neck cancer (HNC) evades immune responses by manipulating the tumor immune microenvironment (TIME). Tumor-bound Axl has been implicated in promoting an immunosuppressive TIME in HNC, though its precise role remains unclear. Understanding Axl's contribution to immune evasion in HNC could lead to the identification of new therapeutic targets; therapies directed at these targets could be combined with and thereby enhance immunotherapies. Results: Using Axl knockout (Axl KO) cell lines derived from the immunologically "cold" MOC2 mouse model, we found that Axl loss delayed tumor growth in immunocompetent mice. This was accompanied by reduced immunosuppressive cells, including MDSCs, Tregs, B cells, and neutrophils, and increased infiltration of cytotoxic CD8 T cells and NK cells. To identify the immune population(s) responsible for these changes, Axl KO tumors were implanted in immune-deficient mice. Axl KO tumor growth in athymic nude mice (which lack T cells) was unchanged, whereas tumor growth in NCG mice (which lack NK cells) was rescued, suggesting that NK cells mediate the Axl KO tumor growth delay. Further, Axl loss enhanced NK cell cytotoxicity in vitro and in vivo, and NK cell depletion reversed delayed Axl KO tumor growth. Mechanistically, Axl KO tumors showed decreased expression of CD73 and CCL2, which inhibit NK cells, and increased expression of CCL5 and CXCL10, which promote NK cell recruitment and activation. Conclusions: These novel findings suggest that tumor-bound Axl fosters an immunosuppressive TIME by inhibiting NK cell recruitment and function, thereby promoting tumor growth. Targeting Axl may enhance NK cell-mediated tumor killing and improve immunotherapy efficacy in HNC.
Collapse
Affiliation(s)
- Kourtney L. Kostecki
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA; (K.L.K.); (R.L.H.); (M.I.); (M.A.H.); (B.E.C.)
| | - Regan L. Harmon
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA; (K.L.K.); (R.L.H.); (M.I.); (M.A.H.); (B.E.C.)
| | - Mari Iida
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA; (K.L.K.); (R.L.H.); (M.I.); (M.A.H.); (B.E.C.)
| | - Madelyn A. Harris
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA; (K.L.K.); (R.L.H.); (M.I.); (M.A.H.); (B.E.C.)
| | - Bridget E. Crossman
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA; (K.L.K.); (R.L.H.); (M.I.); (M.A.H.); (B.E.C.)
| | - Justine Yang Bruce
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA;
- Carbone Cancer Center, University of Wisconsin, Madison, WI 43792, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA;
| | - Deric L. Wheeler
- Department of Human Oncology, School of Medicine and Public Health, University of Wisconsin, Madison, WI 53705, USA; (K.L.K.); (R.L.H.); (M.I.); (M.A.H.); (B.E.C.)
- Carbone Cancer Center, University of Wisconsin, Madison, WI 43792, USA
| |
Collapse
|
8
|
Zha J, Li J, Yin H, Shen M, Xia Y. TIMM23 overexpression drives NSCLC cell growth and survival by enhancing mitochondrial function. Cell Death Dis 2025; 16:174. [PMID: 40082395 PMCID: PMC11906786 DOI: 10.1038/s41419-025-07505-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 02/16/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Mitochondrial hyperfunction is implicated in promoting non-small cell lung cancer (NSCLC) cell growth. TIMM23 (translocase of inner mitochondrial membrane 23) is a core component of the mitochondrial import machinery, facilitating the translocation of proteins across the inner mitochondrial membrane into the matrix. Its expression and potential functions in NSCLC were tested. Comprehensive bioinformatic analysis revealed a strong correlation between TIMM23 overexpression and adverse clinical outcomes in NSCLC patients. Single-cell RNA sequencing data further corroborated these findings, demonstrating elevated TIMM23 expression within the cancer cells of NSCLC mass. Subsequent experimental validation confirmed significantly increased TIMM23 mRNA and protein levels in locally-treated NSCLC tissues compared to matched normal lung tissues. Moreover, TIMM23 expression was consistently elevated across multiple primary/established NSCLC cells. Silencing or ablation of TIMM23 via shRNA or CRISPR/Cas9 in NSCLC cells resulted in impaired mitochondrial function, characterized by reduced complex I activity, ATP depletion, mitochondrial membrane potential dissipation, oxidative stress, and lipid peroxidation. These mitochondrial perturbations coincided with attenuated cell viability, proliferation, and migratory capacity, and concomitant induction of apoptosis. Conversely, ectopic overexpression of TIMM23 significantly enhanced mitochondrial complex I activity and ATP production, promoting NSCLC cell proliferation and motility. In vivo, intratumoral delivery of a TIMM23 shRNA-expressing adeno-associated virus significantly suppressed the growth of subcutaneous NSCLC xenografts in nude mice. Subsequent analysis of tumor tissues revealed depleted TIMM23 expression, ATP reduction, oxidative damage, proliferative arrest, and apoptotic induction. Collectively, these findings establish TIMM23 as a critical pro-tumorigenic factor in NSCLC, highlighting its potential as a prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Jianhua Zha
- Department of Thoracic Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine Nanchang, Nanchang, China
| | - Jiaxin Li
- Department of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Hui Yin
- Department of Thoracic Surgery, The First Affiliated Hospital of Shaoyang University, Shaoyang, China.
| | - Mingjing Shen
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Yingchen Xia
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
9
|
He S, Yang F, Tang L, Yang J, Chen S, Chen Z, Wang L, Zhang Z, Ma L, Luo X. A questionnaire-based cross-sectional survey of cutaneous adverse events in cancer patients treated with molecular targeted therapy and immunotherapies. Support Care Cancer 2025; 33:257. [PMID: 40056220 DOI: 10.1007/s00520-025-09308-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 02/25/2025] [Indexed: 03/10/2025]
Abstract
BACKGROUND Advances in anticancer treatments have significantly improved disease control and progression-free survival. These therapies are associated with various adverse events (AEs), especially cutaneous toxicities. However, there is a paucity of patient-reported outcomes on cutaneous AEs (CAEs) and associated alternation of quality of life (QoL) in cancer patients. OBJECTIVE To evaluate the impact of CAEs on QoL and related social factors in cancer patients. METHODS A cross-sectional questionnaire-based study was conducted in Huashan Hospital affiliated with Fudan University, Fudan University Shanghai Cancer Center, and Zhongshan Hospital affiliated with Fudan University. This study utilized a patient-based questionnaire including social factors, skincare routines, incidence of CAEs, and the impact on QoL. RESULTS Of the 1004 survey participants, 599 reported suffering from CAEs after targeted therapies. The majority of participants were female. Breast cancer was the most frequently reported cancer type. Itching was the most frequently reported problem, followed by eczema, hair loss, hand-foot reaction syndrome, and dry skin. Female participants had significantly lower DLQI scores than males. The need for multidisciplinary approaches in managing skin adverse events was emphasized by 78.30% of participants. CONCLUSION Patient self-report is critical to early detection and proper management for CAEs in cancer patients receiving novel anticancer therapies, thus improving patients' prognosis and QoL. Furthermore, our study emphasized the importance of closer collaboration between dermatologists and oncologists.
Collapse
Affiliation(s)
- Shan He
- Department of Allergy & Immunology, Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Research Center of Allergy and Disease, Fudan University, Shanghai, China
| | - Fanping Yang
- Department of Allergy & Immunology, Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Research Center of Allergy and Disease, Fudan University, Shanghai, China
| | - Lichen Tang
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jin Yang
- Department of Allergy & Immunology, Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Research Center of Allergy and Disease, Fudan University, Shanghai, China
| | - Shengan Chen
- Department of Allergy & Immunology, Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Research Center of Allergy and Disease, Fudan University, Shanghai, China
| | - Zihua Chen
- Department of Allergy & Immunology, Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Research Center of Allergy and Disease, Fudan University, Shanghai, China
| | - Lanting Wang
- Department of Allergy & Immunology, Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Research Center of Allergy and Disease, Fudan University, Shanghai, China
| | - Zhen Zhang
- Department of Allergy & Immunology, Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Research Center of Allergy and Disease, Fudan University, Shanghai, China
| | - Li Ma
- Department of Allergy & Immunology, Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Research Center of Allergy and Disease, Fudan University, Shanghai, China
| | - Xiaoqun Luo
- Department of Allergy & Immunology, Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| |
Collapse
|
10
|
Chen Y, Huang C, Wei W. Establishment of liquid-liquid phase separation-related prognostic model in lung adenocarcinoma and systematic analysis of its clinical significance. Int J Biol Markers 2025; 40:12-23. [PMID: 39791348 DOI: 10.1177/03936155241310887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
PurposeTo detect the prognostic importance of liquid-liquid phase separation (LLPS) in lung adenocarcinoma.MethodsThe gene expression files, copy number variation data, and clinical data were downloaded from The Cancer Genome Atlas cohort. LLPS-related genes were acquired from the DrLLPS website. The prognostic model based on LLPS was constructed by the Cox regression and LASSO regression analyses after the identification of LLPS-related differentially expressed genes (DEGs). Gene Ontology functional and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed. The LLPS-related prognostic risk score was validated by GSE31210 and GSE72094. The overall survival of lung adenocarcinoma patients was predicted by plotting a nomogram. The biological features of the high-risk lung adenocarcinoma were evaluated by the CIBERSORT, ESTIMATE, Gene Set Variation Analysis, and Genomics of Drug Sensitivity in Cancer. Reverse transcription-quantitative polymerase chain reaction detected hub gene expression.ResultsA total of 91 DEGs were screened out in LLPS, among which 9 genes were discovered as prognostic biomarkers of lung adenocarcinoma. GRIA1, CRTAC1, MAGEA4, and MAPK4 were identified as hub genes by the LASSO Cox regression analysis. High-risk and low-risk groups were divided according to the risk index, with the high-risk group displaying a markedly worse outcome. CRTAC1 expression was significantly decreased, MAGEA4 and MAPK4 expressions were increased, while GRIA1 expression was altered in lung adenocarcinoma cells. Tumor microenvironment, signaling pathway enrichment, and drug sensitivity significantly differed between different risk groups.ConclusionsThis work proposed a prognostic tool based on the LLPS-related gene signature to offer prospective and effective biomarkers for lung adenocarcinoma prognosis.
Collapse
Affiliation(s)
- Yan Chen
- Department of Respiratory and Critical Care Medicine, Anyue County People's Hospital, Anyue, China
| | - Cheng Huang
- Department of Respiratory and Critical Care Medicine, Anyue County People's Hospital, Anyue, China
| | - Wei Wei
- Department of Respiratory and Critical Care Medicine, Anyue County People's Hospital, Anyue, China
| |
Collapse
|
11
|
Yoon YC, Lee D, Park JH, Kim OH, Choi HJ, Kim SJ. Enhancing Pancreatic Cancer Therapy With Targeted CD133-Exosome Delivery of PD-L1 siRNA: A Preclinical Investigation. Pancreas 2025; 54:e210-e220. [PMID: 39590886 PMCID: PMC11882183 DOI: 10.1097/mpa.0000000000002419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 08/04/2024] [Indexed: 11/28/2024]
Abstract
OBJECTIVES This study assessed the anticancer potential of genetically modified exosomes engineered to express CD133-binding peptides on their surface and carry PD-L1 siRNA for the treatment of murine model of metastatic pancreatic cancer. MATERIALS AND METHODS CD133-targeting exosomes (tEx) were generated by harvesting conditioned media from adipose-derived stem cells (ASCs) that had undergone transformation using pDisplay vectors encoding CD133-binding peptide sequences. Subsequently, siPD-L1-loaded CD133-targeting Exosomes, referred to as tEx(s), were created by incorporating PD-L1 siRNA into the tEx using Exofect kit. RESULTS tEx(s) demonstrated superior targetability compared to other materials, including Ex, Ex(p), and tEx. This was substantiated by higher total radiant efficiency (TRE) observed in metastatic liver and pancreatic tissues following intravenous administration of tEx(s) ( P < 0.05). Furthermore, the intravenous delivery of tEx(s) resulted in the most pronounced upregulation of proapoptotic markers (BIM and c-caspase 3) and the least downregulation of the antiapoptotic markers (Mcl-1 and Bcl-xL), which has been demonstrated in various methods, including real-time polymerase chain reaction, western blot analysis, and immunohistochemistry in the metastatic lesions in the livers ( P < 0.05). CONCLUSIONS PD-L1 siRNA-loaded CD133-tEx demonstrated remarkable anticancer efficacy, characterized by specific binding to CD133-positive pancreatic cancer cells and suppression of PD-L1 expression within these cells.
Collapse
Affiliation(s)
- Young Chul Yoon
- From the Department of Surgery, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dosang Lee
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jung Hyun Park
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Surgery, Eunpyeong St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ok-Hee Kim
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Translational Research Team, Surginex Co., Ltd., Seoul, Republic of Korea
| | - Ho Joong Choi
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Say-June Kim
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
12
|
Ma F, Qi Z, Liao G, Zhao L, Su X, Dong C, Lu F, Sun Y. Study on PD-1 inhibitor combined with recombinant human endostatin and chemotherapy followed by IMRT in the treatment of advanced NSCLC. Medicine (Baltimore) 2025; 104:e41306. [PMID: 39928822 PMCID: PMC11812992 DOI: 10.1097/md.0000000000041306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 09/18/2024] [Accepted: 10/10/2024] [Indexed: 02/12/2025] Open
Abstract
Although chemotherapy, targeted therapy, and antiangiogenic drugs have become the cornerstones of treatment for advanced non-small-cell carcinoma (NSCLC) in clinical practice, the emergence of immune checkpoint inhibitors (such as PD-1 inhibitors) in recent years has also provided new options for the treatment of NSCLC. To explore whether PD-1 inhibitors combined with recombinant human endostatin and chemotherapy followed by IMRT have a certain curative effect in the treatment of advanced NSCLC. We retrospectively analyzed 47 patients with stage IIIB, IIIC, and IV NSCLC admitted to our hospital from August 2022 to June 2023. According to the treatment method, the patients were divided into an observation group (24 cases) and a control group (23 cases). The control group only received recombinant human endostatin 105 g and chemotherapy followed by IMRT; the observation group received domestic PD-1 inhibitors, 200 mg, and chemotherapy followed by IMRT on the basis of the treatment plan of the control group. After treatment, the objective response rate (ORR), disease control rate, overall survival, progression-free survival (PFS), duration of response, and incidence of adverse reactions were compared. After treatment, the ORR and disease control rate of the observation group were higher than those of the control group; compared with the control group, the PFS, overall survival, and duration of response period of the observation group were longer (P < .05); the incidence of adverse reactions in the observation group was significantly lower than that in the control group (P < .05). After chemotherapy, the CD3+ and CD4+ index of the observation group was significantly increased, the CD3+ CD8+ was slightly lower than that of the control group, without statistical significance, and the ratio of CD4/CD8 was higher than that of the control group, and the interleukin-2 index was significantly better than that of the routine group, P < .05. PD-1 inhibitor combined with recombinant human endostatin and chemotherapy followed by IMRT in the treatment of advanced NSCLC can significantly improve the ORR and prolong the PFS of patients, and the adverse reactions are controllable. The results of our study may provide help for the treatment strategies of patients with refractory advanced NSCLC, and are worthy of promotion and use.
Collapse
Affiliation(s)
- FaQiang Ma
- Department of Oncology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, China
| | - ZhengJun Qi
- Department of Oncology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, China
| | - GuangHui Liao
- Department of Oncology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, China
| | - LiLi Zhao
- Department of Oncology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, China
| | - XiaLu Su
- Department of Oncology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, China
| | - ChangFen Dong
- Department of Oncology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, China
| | - FangYang Lu
- Department of Oncology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou, China
| | - Yi Sun
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, Guizhou, China
| |
Collapse
|
13
|
Ren J, Wang J, Wang Y, Yang D, Sheng J, Zhu S, Liu Y, Li X, Liu W, Zhang B. Efficacy and safety of PD-1/PD-L1 inhibitors in advanced or recurrent endometrial cancer: a meta-analysis with trial sequential analysis of randomized controlled trials. Front Immunol 2025; 16:1521362. [PMID: 39958346 PMCID: PMC11825832 DOI: 10.3389/fimmu.2025.1521362] [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: 11/01/2024] [Accepted: 01/07/2025] [Indexed: 02/18/2025] Open
Abstract
Background The combination of programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors with chemotherapy (CT) is currently under evaluation as a first-line treatment for advanced or recurrent endometrial cancer (EC). This study sought to assess the efficacy and safety of this therapeutic combination in patients with advanced or recurrent EC. Methods We performed an exhaustive review of randomized controlled trials (RCTs) up to September 25, 2024, examining the efficacy and safety of combining PD-1/PD-L1 inhibitors with CT versus CT alone (or plus placebo) in advanced or recurrent EC. Efficacy was measured by progression-free survival (PFS) and overall survival (OS), while safety was assessed by the incidence of any grade or grade ≥ 3 adverse events (AEs). We calculated hazard ratios (HRs) for PFS and OS, as well as risk ratios (RRs) for AEs, each accompanied by 95% confidence intervals (CIs). To evaluate heterogeneity, we employed Cochran's Q test, I2 statistics, and 95% prediction intervals (PIs). Trial sequential analysis (TSA) was conducted using R Version 4.3.1, STATA Version 12.0, and TSA Version 0.9.5.10 Beta software. Results Our analysis incorporated 6 studies, encompassing a total of 2,954 patients. The combination of PD-1/PD-L1 inhibitors with CT significantly improved PFS (HR = 0.617, 95% CI: 0.506-0.752; 95% PI: 0.334-1.140) and OS (HR = 0.774, 95% CI: 0.664-0.902; 95% PI: 0.553-1.083) compared to CT alone (or plus placebo) in the overall population. Subgroup analysis based on mismatch repair (MMR) status revealed pronounced benefits in PFS and OS for patients with deficient MMR (dMMR) (PFS: HR = 0.344, 95% CI: 0.269-0.438; 95% PI: 0.231-0.510; OS: HR = 0.371, 95% CI: 0.245-0.562; 95% PI: 0.025-5.461) compared to those with proficient MMR (pMMR) (PFS: HR = 0.772, 95% CI: 0.627-0.950; 95% PI: 0.394-1.512; OS: HR = 0.996, 95% CI: 0.692-1.435; 95% PI: 0.021-47.662). Although there was no observed difference in the incidence of any grades AEs (RR = 0.994, 95% CI: 0.982-1.006; 95% PI: 0.978-1.009), the risk of grade ≥ 3 AEs was elevated in the group receiving PD-1/PD-L1 inhibitors in combination with CT (RR = 1.132, 95% CI: 1.023-1.252; 95% PI: 0.836-1.532). Conclusion The combination of PD-1/PD-L1 inhibitors with CT significantly improved PFS and OS in advanced or recurrent EC patients, with particularly pronounced benefits observed in those with dMMR. Clinicians can tailor treatment strategies according to individual patient characteristics to optimize therapeutic outcomes, while remaining alert to the possibility of AEs in clinical practice. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42024595455.
Collapse
Affiliation(s)
- Ji Ren
- Department of Medicine and Health, Dezhou University, Dezhou, China
| | - Jinghe Wang
- Department of Medicine and Health, Dezhou University, Dezhou, China
| | - Yanan Wang
- Department of Medicine and Health, Dezhou University, Dezhou, China
| | - Dongying Yang
- Department of Medicine and Health, Dezhou University, Dezhou, China
| | - Jianming Sheng
- Department of Medicine and Health, Dezhou University, Dezhou, China
| | - Shili Zhu
- Department of Medicine and Health, Dezhou University, Dezhou, China
| | - Yunli Liu
- Department of Medicine and Health, Dezhou University, Dezhou, China
| | - Xiaoqi Li
- Department of Medicine and Health, Dezhou University, Dezhou, China
| | - Wei Liu
- Department of Medicine and Health, Dezhou University, Dezhou, China
| | - Binbin Zhang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
14
|
Zhao L, Gong J, Liao S, Huang W, Zhao J, Xing Y. Preclinical evaluation and preliminary clinical study of 68Ga-NODAGA-NM-01 for PET imaging of PD-L1 expression. Cancer Imaging 2025; 25:6. [PMID: 39871394 PMCID: PMC11771120 DOI: 10.1186/s40644-025-00826-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/16/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Programmed cell death 1/programmed death ligand-1 (PD-L1)-based immune checkpoint blockade is an effective treatment approach for non-small-cell lung cancer (NSCLC). However, immunohistochemistry does not accurately or dynamically reflect PD-L1 expression owing to its spatiotemporal heterogeneity. Herein, we assessed the feasibility of using a 68Ga-labeled anti-PD-L1 nanobody, 68Ga-NODAGA-NM-01, for PET imaging of PD-L1. METHODS Micro-PET/CT and biodistribution studies were performed on PD-L1-positive and -negative tumor-bearing mice. Additionally, a preliminary clinical study was performed on two patients with NSCLC. NM-01 was radiolabeled with 68Ga without further purification under mild conditions. RESULTS 68Ga-NODAGA-NM-01 exhibited radiochemical purity (> 98%), high stability in vitro, and rapid blood clearance in vivo. Specific accumulation of 68Ga-NODAGA-NM-01 was observed in PD-L1-positive tumor-bearing mice, with a good tumor-to-background ratio 0.5h post-injection. Furthermore, 68Ga-NODAGA-NM-01 PET/CT imaging was found to be safe with no adverse events and distinct uptake in primary and metastatic lesions of the PD-L1-positive patient, with a higher maximal standardized uptake value than that in lesions of the PD-L1-negative patient 1h post-injection. CONCLUSIONS 68Ga-NODAGA-NM-01 can be prepared using a simple method under mild conditions and reflect PD-L1 expression in primary and metastatic lesions. However, our findings need to be confirmed in a large cohort. TRIAL REGISTRATION NCT02978196. Registered February 15, 2018.
Collapse
Affiliation(s)
- Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou District, No. 100, Haining Road, Shanghai, 200080, China
| | - Jiali Gong
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou District, No. 100, Haining Road, Shanghai, 200080, China
| | - Sisi Liao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou District, No. 100, Haining Road, Shanghai, 200080, China
| | - Wenhua Huang
- Nanomab Technology Limited, No. 333, North Chengdu Road, Jingan District, Shanghai, 200041, China
| | - Jinhua Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou District, No. 100, Haining Road, Shanghai, 200080, China.
| | - Yan Xing
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Hongkou District, No. 100, Haining Road, Shanghai, 200080, China.
| |
Collapse
|
15
|
Chen W, Lin Q, Wang D, Xie W, Huang C, Fan W, Wu S, Fan X, Li C. Establishment and clinical value of a circulating tumor cell system based on a multi-site immune lipid magnetic sphere technique in laryngopharyngeal head and neck tumors. Am J Cancer Res 2025; 15:19-31. [PMID: 39949932 PMCID: PMC11815362 DOI: 10.62347/mvrg3697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/03/2025] [Indexed: 02/16/2025] Open
Abstract
This study aimed to construct multi-site magnetic nanospheres to capture circulating tumor cells (CTCs) from peripheral blood specimens of laryngopharyngeal head and neck tumors. Separated CTCs were used to measure downstream molecular markers and to detect and analyze the disease status. A stable CTC multisite nano-enrichment system was used to determine changes in CTCs levels in Programmed Death-Ligand-1 (PD-L1)-positive patients and to assess the extent of real-time changes in CTCs over the course of the disease in correlation with clinicopathological indicators. The results demonstrated that the constructed immunomagnetic spheres could effectively capture CTCs and that the constructed lipid nanoparticles exhibited high capture efficiency and low cytotoxicity. The results of the concordance or complementarity analyses of PD-L1 expression at the CTC and tissue levels indicated good concordance between the two at up to 70%. The analysis of PD-L1 expression and the changes in CTCs in PD-L1-positive cells plays an auxiliary role in clinical diagnosis and can be used as a dynamic detection index for the course of head, neck, and throat tumor treatment and as a predictor of recurrence risk.
Collapse
Affiliation(s)
- Wei Chen
- Department of Otolaryngology, Fujian Medical University Union HospitalFuzhou 350001, Fujian, China
| | - Qin Lin
- Department of Otolaryngology, Fujian Medical University Union HospitalFuzhou 350001, Fujian, China
| | - Desheng Wang
- Department of Otolaryngology, Fujian Medical University Union HospitalFuzhou 350001, Fujian, China
| | - Wenting Xie
- Department of Otolaryngology, Fujian Medical University Union HospitalFuzhou 350001, Fujian, China
| | - Chunyan Huang
- Department of Otolaryngology, Fujian Medical University Union HospitalFuzhou 350001, Fujian, China
| | - Wenjing Fan
- Department of Otolaryngology, Fujian Medical University Union HospitalFuzhou 350001, Fujian, China
| | - Shipu Wu
- Department of ENT, Zhenghe County HospitalNanping 353605, Fujian, China
| | - Xiaomei Fan
- Department of ENT, Zhenghe County HospitalNanping 353605, Fujian, China
| | - Chen Li
- Department of Otolaryngology, Fujian Medical University Union HospitalFuzhou 350001, Fujian, China
| |
Collapse
|
16
|
Zhang B, Liu Q, Li L, Ye Y, Guo X, Xu W, Chen L, Mo X, Nian S, Yuan Q. Therapeutic effect of fully human anti-Nrp-1 antibody on non-small cell lung cancer in vivo and in vitro. Cancer Immunol Immunother 2025; 74:50. [PMID: 39751948 PMCID: PMC11699024 DOI: 10.1007/s00262-024-03893-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 11/11/2024] [Indexed: 01/04/2025]
Abstract
Although immune checkpoint inhibitors have changed the treatment paradigm for non-small cell lung cancer (NSCLC), not all patients benefit from them. Therefore, there is an urgent need to explore novel immune checkpoint inhibitors. Neuropilin-1 (Nrp-1) is a unique immune checkpoint capable of exerting antitumor effects through CD8+ T cells. It is also a T-cell memory checkpoint that regulates long-term antitumor immunity. However, its role in NSCLC remains unclear. The aim of this study was to develop a fully human anti-Nrp-1 antibody with therapeutic effects against NSCLC in vitro and in vivo. We screened and constructed of a high-affinity anti-Nrp-1 IgG antibody from a constructed high-capacity fully human single-chain fragment variable (scFv) phage library. This novel anti-Nrp-1 IgG antibody partially restored the killing function of exhausted CD8+ T cells in malignant pleural fluid in vitro. Co-culture of peripheral blood mononuclear cells (PBMC) with A549 and the addition of anti-Nrp1-IgG enhanced the killing of A549 target cells, leading to an increase in late-stage apoptosis of target cells. Importantly, anti-Nrp1-IgG treatment significantly reduced tumor volume in a mouse model of lung cancer with humanized immune system. These findings suggest that 53-IgG has a promising application as a potent Nrp-1-targeting agent in NSCLC immunotherapy.
Collapse
Grants
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022NSFSC0699, 2023NSFSC0727 Scienceand Technology Department of Sichuan Province
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022YFS0636-B3, 2022YFS0608-B1, 2022YFS0630-B3 the Sichuan Science and Technology program
- 2022CXY06 the project of Southwest Medical University
- 2022CXY06 the project of Southwest Medical University
- 2022CXY06 the project of Southwest Medical University
- 2022CXY06 the project of Southwest Medical University
- 2022CXY06 the project of Southwest Medical University
- 2022CXY06 the project of Southwest Medical University
- 2022CXY06 the project of Southwest Medical University
- 2022CXY06 the project of Southwest Medical University
- 2022CXY06 the project of Southwest Medical University
Collapse
Affiliation(s)
- Bo Zhang
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
- Clinical Laboratory, Female and Child Health Care and Family Planning Service Center, Binhai New Area, Tianjin, 300450, China
| | - Qin Liu
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Lin Li
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Yingchun Ye
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Xiyuan Guo
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Wenfeng Xu
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Neurosurgery, Luzhou, 646000, Sichuan Province, China
| | - Xianming Mo
- Department of Gastrointestinal Surgery, Laboratory of Stem Cell Biology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Siji Nian
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
| | - Qing Yuan
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, Sichuan Province, China.
- Institute of Nuclear Medicine, Southwest Medical University, Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China.
| |
Collapse
|
17
|
Zeng F, Zhang Y, Luo T, Wang C, Fu D, Wang X. Daidzein Inhibits Non-small Cell Lung Cancer Growth by Pyroptosis. Curr Pharm Des 2025; 31:884-924. [PMID: 39623715 DOI: 10.2174/0113816128330530240918073721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/08/2024] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) represents the leading cause of cancer deaths in the world. We previously found that daidzein, one of the key bioactivators in soy isoflavone, can inhibit NSCLC cell proliferation and migration, while the molecular mechanisms of daidzein in NSCLC remain unclear. METHODS We developed an NSCLC nude mouse model using H1299 cells and treated the mice with daidzein (30 mg/kg/day). Mass spectrometry analysis of tumor tissues from daidzein-treated mice identified 601 differentially expressed proteins (DEPs) compared to the vehicle-treated group. Gene enrichment analysis revealed that these DEPs were primarily associated with immune regulatory functions, including B cell receptor and chemokine pathways, as well as natural killer cell-mediated cytotoxicity. Notably, the NOD-like receptor signaling pathway, which is closely linked to pyroptosis, was significantly enriched. RESULTS Further analysis of key pyroptosis-related molecules, such as ASC, CASP1, GSDMD, and IL-1β, revealed differential expression in NSCLC versus normal tissues. High levels of ASC and CASP1 were associated with a favorable prognosis in NSCLC, suggesting that they may be critical effectors of daidzein's action. In NSCLC-bearing mice treated with daidzein, RT-qPCR and Western blot analyses showed elevated mRNA and protein levels of ASC, CASP1, and IL-1β but not GSDMD, which was consistent with the proteomic data. CONCLUSION In summary, this study demonstrated that daidzein inhibits NSCLC growth by inducing pyroptosis. Key pathway modulators ASC, CASP1, and IL-1β were identified as primary targets of daidzein. These findings offer insights into the molecular mechanisms underlying the anti-NSCLC effects of daidzein and could offer dietary recommendations for managing NSCLC.
Collapse
Affiliation(s)
- Fanfan Zeng
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yu Zhang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Ting Luo
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Department of Infection Control, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Chengman Wang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Denggang Fu
- College of Medicine, Medical University of South Carolina, Columbia, Charleston, SC 29425, United States
| | - Xin Wang
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, 332005, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
18
|
Lu J, Zhu J, Jiang G. Association of CirAEs and Efficacy in NSCLC Patients Treated with PD-1/PD-L1: A Meta-analysis of Cohort Studies. Comb Chem High Throughput Screen 2025; 28:435-446. [PMID: 38265378 DOI: 10.2174/0113862073267261231106093845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/14/2023] [Accepted: 10/09/2023] [Indexed: 01/25/2024]
Abstract
PURPOSE To systematically evaluate the relationship between cutaneous immunerelated adverse events (cirAEs) and the efficacy of PD-1/PD-L1 in the treatment of non-small cell lung cancer (NSCLC) and to provide an evidence-based reference for the clinical application of PD-1/PD-L1 and safety evaluation. METHODS Electronic databases (PubMed, Embase, Medline, Web of Science, and the Cochrane Library) were screened systematically to collect prospective or retrospective cohort studies on the correlation between cirAEs and efficacy of PD-1/PD-L1 in the treatment of NSCLC. RESULTS A total of 3514 participants were included in 13 cohort studies (enclosing an ambidirectional cohort study). Outcomes revealed that compared with those patients with non cirAEs, patients suffering cirAEs were associated with significantly higher objective response rate (ORR) [risk ratio (RR): 1.74, 95% confidence interval (CI): 1.42-2.14, P<0.00001], longer progressionfree survival (PFS) [RR: 0.52, 95% CI: 0.45-0.60, P<0.00001], and longer overall survival (OS) [RR:0.46, 95% CI: 0.38-0.56]. Sensitivity analyses through the exclusion of one study at a time did not significantly influence the outcomes, indicating that the meta-analysis results were relatively robust. Furthermore, subgroup analyses revealed consistent results in the study design (prospective or retrospective cohort studies), as well as in the endpoint results (PFS and OS) of Kaplan-Meier curves or Cox proportional hazards regression for evaluable patients. CONCLUSION Currently, evidence reveals that cirAEs development may be associated with a good prognosis and can be an early predictor of the efficacy of PD-1/PD-L1 in the treatment of NSCLC patients.
Collapse
Affiliation(s)
- Junru Lu
- Department of Dermatology, Xuzhou Medical University, Xuzhou 221004, China
| | - Jiaming Zhu
- Department of Dermatology, Xuzhou Medical University, Xuzhou 221004, China
| | - Guan Jiang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| |
Collapse
|
19
|
Su Z, Guan M, Zhang L, Lian X. Factors associated with immune‑related severe adverse events (Review). Mol Clin Oncol 2025; 22:3. [PMID: 39563998 PMCID: PMC11574706 DOI: 10.3892/mco.2024.2798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are frequently used in cancer treatment. Despite their clinical benefits, they can also cause a wide range of immune-related adverse events (ir-AEs). The overall incidence of irAEs in cancer patients treated with immunotherapy ranges from 70-90%, while that of immune-related severe adverse events (ir-SAEs) is 10-43%. ir-SAEs pose a significant risk to patient safety as they are extremely frequent and lethal. Due to non-specific manifestations, rapid progression and significant morbidity, it is essential to identify factors associated with ir-SAEs early to predict high-risk groups for treatment safety. However, less information is available on the factors causing ir-SAEs, and further research is needed. The present study reviews the factors associated with ir-SAEs in terms of demographic characteristics, disease-related information and laboratory examinations to provide a clinical reference. In terms of demographic characteristics, age, body mass index, smoking, ethnicity and cancer family history may influence the incidence of ir-SAEs. Regarding disease-related information, the risks factors associated with ir-SAEs may include disease history, treatment regimen and cancer type. For laboratory examinations, risk factors associated with ir-SAEs include the laboratory examination parameters of peripheral blood cells, immunocytes, cytokines/chemokines, genetics, gut microbia, proteins and brain injury markers. All of these risk factors can stimulate the body's inflammatory response, leading to over proliferation of T cells and other inflammatory factors. In addition, the use of ICIs may disrupt gut microbial homeostasis and dysregulate the pre-existing intestinal ecology, which may therefore trigger inflammatory signaling pathways, affect overall immune function and increase the occurrence of ir-SAEs. In response to the aforementioned risk factors, it is recommended that medical professionals incorporate their analysis into routine patient testing for early identification of patient ir-SAEs and to create early individualized interventions to improve the safety for immunotherapy patients.
Collapse
Affiliation(s)
- Zhenzhen Su
- School of Nursing, Peking University, Beijing 100191, P.R. China
| | - Miaomiao Guan
- School of Nursing, Peking University, Beijing 100191, P.R. China
| | - Liyan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xuemin Lian
- School of Nursing, Peking University, Beijing 100191, P.R. China
- Department of Health and Medical, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
20
|
Li G, Li Q, Tong Y, Zeng J, Dang T, Yang N, Zhou Y, Ma L, Ge Q, Zhao Z. The anticancer mechanisms of Toxoplasma gondii rhoptry protein 16 on lung adenocarcinoma cells. Cancer Biol Ther 2024; 25:2392902. [PMID: 39174877 PMCID: PMC11346528 DOI: 10.1080/15384047.2024.2392902] [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: 09/06/2023] [Revised: 04/29/2024] [Accepted: 08/11/2024] [Indexed: 08/24/2024] Open
Abstract
Lung adenocarcinoma is the most prevalent subtype of lung cancer, which is the leading cause of cancer-related mortality worldwide. Toxoplasma gondii (T.gondii) Rhoptry protein 16 (ROP16) has been shown to quickly enter the nucleus, and through activate host cell signaling pathways by phosphorylation STAT3 and may affect the survival of tumor cells. This study constructed recombinant lentiviral expression vector of T. gondii ROP16 I/II/III and stably transfected them into A549 cells, and the effects of ROP16 on cell proliferation, cell cycle, apoptosis, invasion, and migration of A549 cells were explored by utilizing CCK-8, flow cytometry, qPCR, Western blotting, TUNEL, Transwell assay, and cell scratch assay, and these effects were confirmed in the primary human lung adenocarcinoma cells from postoperative cancer tissues of patients. The type I and III ROP16 activate STAT3 and inhibited A549 cell proliferation, regulated the expression of p21, CDK6, CyclinD1, and induced cell cycle arrest at the G1 phase. ROP16 also regulated the Bax, Bcl-2, p53, cleaved-Caspase3, and Caspase9, inducing cell apoptosis, and reduced the invasion and migration of A549 cells, while type II ROP16 protein had no such effect. Furthermore, in the regulation of ROP16 on primary lung adenocarcinoma cells, type I and III ROP16 showed the same anticancer potential. These findings confirmed the anti-lung adenocarcinoma effect of type I and III ROP16, offering fresh perspectives on the possible application of ROP16 as a target with adjuvant therapy for lung adenocarcinoma and propelling the field of precision therapy research toward parasite treatment of tumors.
Collapse
Affiliation(s)
- Guangqi Li
- Medical Laboratory Center, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical Pathogenic Microorganisms, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Qinhui Li
- College of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Yongqing Tong
- Department of Clinical laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jin Zeng
- College of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Tiantian Dang
- Medical Laboratory Center, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical Pathogenic Microorganisms, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Ningai Yang
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yuning Zhou
- The First Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Lei Ma
- College of Life Sciences, Ningxia University, Yinchuan, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western, Ningxia University, Yinchuan, China
| | - Qirui Ge
- The First Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Zhijun Zhao
- Medical Laboratory Center, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Clinical Pathogenic Microorganisms, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Clinical Research Center of Medical Laboratory, General Hospital of Ningxia Medical University, Yinchuan, China
| |
Collapse
|
21
|
Huang Q, Tao Y, Zhang Y, Chen Y, Tan F, Ou Y. Pyropheophorbide-α methyl ester-mediated photodynamic therapy triggers pyroptosis in osteosarcoma cells via the ROS/caspase-3/GSDME pathway. Photodiagnosis Photodyn Ther 2024; 50:104427. [PMID: 39615558 DOI: 10.1016/j.pdpdt.2024.104427] [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: 09/05/2024] [Revised: 11/10/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024]
Abstract
BACKGROUND Pyropheophorbide-α methyl ester-mediated photodynamic therapy(MPPa-PDT) is a candidate treatment for solid tumors, including osteosarcoma. Pyroptosis has garnered significant attention in cancer research due to its pro-inflammatory and immunomodulatory nature. This study investigated the mechanism and role of MPPa-PDT-induced pyroptosis in osteosarcoma cells. METHODS We treated human osteosarcoma 143b and HOS cells with MPPa at concentrations of 0.5 μM and 0.25 μM, respectively, then irradiated the cells with LED light at 630 nm wavelength with an energy density of 4.8 J/cm2. Cell viability and apoptosis ratio were detected using CCK-8 and Annexin V-Propidium Iodide staining, respectively. Intracellular reactive oxygen species (ROS) levels and mitochondrial membrane potential (MtΔψ) were assessed using 2',7'-Dichlorofluorescin diacetate, and JC-1 staining kits, respectively. Scanning Electron Microscopy (SEM) was utilized to examine cell ultrastructure. The morphological changes of the cells were observed by an inverted microscope. Western blotting analysis was conducted to measure protein levels. To elucidate the mechanism and role, we re-evaluated relevant parameters after pretreating with NAC,Si caspase-3, and Si GSDME. RESULTS MPPa-PDT inhibited the activity of osteosarcoma 143b and HOS cells and induced pyroptosis with mitochondrial damage, ROS aggregation, and activation of Caspase-3 and GSDME. The effects of MPPa-PDT on the activity and apoptosis of osteosarcoma cells were partially reversed after pretreating with Si GSDME. After NAC pretreatment, the activation of pyroptosis and Caspase-3 induced by MPPa-PDT was partially reversed. After Si Caspase-3 pretreatment, the pyroptosis induced by MPPa-PDT was partially reversed. CONCLUSION MPPa-PDT can induce pyroptosis in osteosarcoma cells, which has the effect of enhancing apoptotic processes. Mitochondrial damage and ROS/caspase-3/GSDME pathway are the possible mechanisms of pyroptosis induced by MPPa-PDT.
Collapse
Affiliation(s)
- Qiu Huang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Yuzhong, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Yuzhong, Chongqing, 400016, China; Department of Orthopaedics, People's Hospital of Leshan, Leshan, Sichuan, 614000, China
| | - Yong Tao
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Yuzhong, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Yuzhong, Chongqing, 400016, China
| | - Ye Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Yuzhong, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Yuzhong, Chongqing, 400016, China
| | - Yuxing Chen
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Yuzhong, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Yuzhong, Chongqing, 400016, China
| | - Fuqiang Tan
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Yuzhong, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Yuzhong, Chongqing, 400016, China
| | - Yunsheng Ou
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing, 400016, China; Chongqing Municipal Health Commission Key Laboratory of Musculoskeletal Regeneration and Translational Medicine, Yuzhong, Chongqing, 400016, China; Orthopaedic Research Laboratory of Chongqing Medical University, Yuzhong, Chongqing, 400016, China.
| |
Collapse
|
22
|
Kang DH, Lee J, Im S, Chung C. Navigating the Complexity of Resistance in Lung Cancer Therapy: Mechanisms, Organoid Models, and Strategies for Overcoming Treatment Failure. Cancers (Basel) 2024; 16:3996. [PMID: 39682183 DOI: 10.3390/cancers16233996] [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: 09/24/2024] [Revised: 11/17/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Background: The persistence of chemotherapy-resistant and dormant cancer cells remains a critical challenge in the treatment of lung cancer. Objectives: This review focuses on non-small cell lung cancer and small cell lung cancer, examining the complex mechanisms that drive treatment resistance. Methods: This review analyzed current studies on chemotherapy resistance in NSCLC and SCLC, focusing on tumor microenvironment, genetic mutations, cancer cell heterogeneity, and emerging therapies. Results: Conventional chemotherapy and targeted therapies, such as tyrosine kinase inhibitors, often fail due to factors including the tumor microenvironment, genetic mutations, and cancer cell heterogeneity. Dormant cancer cells, which can remain undetected in a quiescent state for extended periods, pose a significant risk of recurrence upon reactivation. These cells, along with intrinsic resistance mechanisms, greatly complicate treatment efforts. Understanding these pathways is crucial for the development of more effective therapies. Emerging strategies, including combination therapies that target multiple pathways, are under investigation to improve treatment outcomes. Innovative approaches, such as antibody-drug conjugates and targeted protein degradation, offer promising solutions by directly delivering cytotoxic agents to cancer cells or degrading proteins that are essential for cancer survival. The lung cancer organoid model shows substantial promise to advance both research and clinical applications in this field, enhancing the ability to study resistance mechanisms and develop personalized treatments. The integration of current research underscores the need for continuous innovation in treatment modalities. Conclusions: Personalized strategies that combine novel therapies with an in-depth understanding of tumor biology are essential to overcome the challenges posed by treatment-resistant and dormant cancer cells in lung cancer. A multifaceted approach has the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Da Hyun Kang
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jisoo Lee
- College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Subin Im
- College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Chaeuk Chung
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
23
|
Wu S, Luo T, Lei X, Yang X. Emerging role of competing endogenous RNA in lung cancer drug resistance. J Chemother 2024; 36:546-565. [PMID: 38124356 DOI: 10.1080/1120009x.2023.2294582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
Lung cancer remains one of the most common malignant cancers worldwide, and its survival rate is extremely low. Chemotherapy, the mainstay of lung cancer treatment, is not as effective as it could be due to the development of cellular resistance. The molecular mechanisms of drug resistance in lung cancer remain to be elucidated. Accumulating evidence suggests that ceRNAs are involved in various carcinogenesis and development. CeRNA is a transcript that regulates each other through competition with miRNA. However, the relationship between ceRNAs and chemoresistance in lung cancer remains unclear. In this narrative review, we provided a summary of treatment approaches that focus on ceRNA networks to overcome drug resistance.
Collapse
Affiliation(s)
- Shijie Wu
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, People's Republic of China
| | - Ting Luo
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, People's Republic of China
| | - Xiaoyong Lei
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, People's Republic of China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, People's Republic of China
| | - Xiaoyan Yang
- School of Pharmaceutical Science, Hengyang Medical College, University of South China, Hengyang, People's Republic of China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, University of South China, Hengyang, People's Republic of China
| |
Collapse
|
24
|
Yu Z, Sun J, Fang K, Xu J, Yang J, Chunlei D, Gong Y, Ma H. SLC2A1 boosts the resistance of non-small cell lung cancer to taxanes by stimulating the formation of EPCAM+ cancer stem-like cells via glycolysis. Transl Oncol 2024; 49:102082. [DOI: doi.org/10.1016/j.tranon.2024.102082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025] Open
|
25
|
Martínez-Lira JL, Hernández-Gallegos E, DE Guadalupe Chávez-López M, Villalobos-Valencia R, Camacho J. The Effects of Nebivolol-Gefitinib-Loratadine Against Lung Cancer Cell Lines. In Vivo 2024; 38:2688-2695. [PMID: 39477390 PMCID: PMC11535926 DOI: 10.21873/invivo.13746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND/AIM Non-small-cell lung cancer (NSCLC) is the most frequently diagnosed malignancy and the first cause of cancer-related death. Thus, finding alternative therapeutic options is crucial. Drug repurposing offers therapeutic options in a simplified and affordable manner, especially to cancer patients in developing countries. Several drugs including antihistamines and beta-adrenergic receptor blockers (beta-blockers) display antiproliferative properties on cancer cells. Interestingly, NSCLC patients who had used either antihistamines or beta-blockers showed improved response to chemotherapy or reduced mortality in comparison to non-users of any of these drugs. However, combination therapy is gaining substantial interest in many cancers including non-EGFR mutated NSCLC. Here, we investigated the antineoplastic effect of the combination of the antihistamine loratadine, the beta-blocker nebivolol, and the tyrosine-kinase inhibitor gefitinib on NSCLC cell lines. MATERIALS AND METHODS A-549 and NCI-H1975 cell lines were used. The effect of nebivolol, gefitinib, and loratadine on the metabolic activity was studied using the MTT assay. The inhibitory concentrations (IC20 and IC50) were calculated and used in the drug-combination experiments. Apoptosis was investigated using flow cytometry; and cell survival using the colony formation assay. RESULTS The combination nebivolol-loratadine-gefitinib produced a significant synergistic effect on inhibiting the metabolic activity and colony formation, as well as on promoting apoptosis in both cell lines. Noteworthy, the effect on the cell line carrying the EGFR mutation (NCI-H1975) was very similar to the cell line that does not exhibit such mutation (A-549 cells). CONCLUSION The nebivolol-gefitinib-loratadine combination may be a promising alternative for lung cancer treatment.
Collapse
Affiliation(s)
- José Luis Martínez-Lira
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
- UMAE, Hospital de Especialidades Nο. 25, Instituto Mexicano del Seguro Social, Monterrey Nuevo León, México
| | - Elisabeth Hernández-Gallegos
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - María DE Guadalupe Chávez-López
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - Ricardo Villalobos-Valencia
- Departamento de Oncología Médica, Hospital de Oncología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de México, México
| | - Javier Camacho
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México;
| |
Collapse
|
26
|
Zhang Y, Zhang F, Liu Z, Li M, Wu G, Li H. P2RX1-Negative neutrophils promote the immunosuppressive microenvironment in Non-Small cell lung cancer by Upregulating PD-L1 expression. Hum Immunol 2024; 85:111105. [PMID: 39317128 DOI: 10.1016/j.humimm.2024.111105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/16/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND The most abundant innate immune cells, neutrophils, contribute significantly to cancer development by stimulating immunosuppression. However, it remains unclear about its function and molecular mechanisms in the immunosuppressive microenvironment of non-small cell lung cancer (NSCLC). METHODS Blood samples were collected from NSCLC patients and healthy volunteers to detect the expression of P2RX1 and PD-L1 in neutrophils using qRT-PCR, western blot (WB), and flow cytometry. Neutrophils were sorted into P2RX1-positive (P2RX1+)/P2RX1-negative (P2RX1-) groups and co-cultured with CD8+ T cells. Changes in the proliferative and cytotoxic capabilities of CD8+ T cells were then detected using flow cytometry and enzyme-linked immunosorbent assay. The content of granzyme B was determined by enzyme-linked immunosorbent assay. The effects of P2RX1-deficient neutrophils on fatty acids, triglycerides, lipid droplet content and FASN expression were detected using kits, Nile red staining and WB, respectively. RESULTS This study revealed a deficiency in P2RX1 expression in peripheral blood neutrophils of NSCLC patients, which was negatively correlated with the expression of PD-L1. P2RX1-neutrophils inhibited T cell proliferation and granzyme B expression and promoted T cell exhaustion. Furthermore, in P2RX1-deficient neutrophils, there was a notable increase in the levels of fatty acids, triglycerides, and lipid droplet accumulation, as well as an upregulation of FASN protein expression. Mechanistically, P2RX1-neutrophils upregulated PD-L1 expression by inducing fatty acid metabolism to improve immunosuppression in NSCLC. CONCLUSION The mechanism by which P2RX1-deficient neutrophils contributed to immunosuppressive effects in NSCLC was clarified by our findings, indicating that P2RX1 could be one potential target for counteracting the immunosuppressive effects of neutrophils.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Medical Oncology, Ma'anshan People's Hospital, Ma'anshan, 243000, Anhui Province, China.
| | - Fenglin Zhang
- Department of Medical Oncology, Ma'anshan People's Hospital, Ma'anshan, 243000, Anhui Province, China
| | - Zhi Liu
- Department of Pathology, Ma'anshan People's Hospital, Ma'anshan, 243000, Anhui Province, China
| | - Min Li
- Department of Medical Oncology, Ma'anshan People's Hospital, Ma'anshan, 243000, Anhui Province, China
| | - Ge Wu
- Department of Medical Oncology, Ma'anshan People's Hospital, Ma'anshan, 243000, Anhui Province, China
| | - Hui Li
- Department of Medical Oncology, Ma'anshan People's Hospital, Ma'anshan, 243000, Anhui Province, China
| |
Collapse
|
27
|
Yu Z, Sun J, Fang K, Xu J, Yang J, Chunlei D, Gong Y, Ma H. SLC2A1 boosts the resistance of non-small cell lung cancer to taxanes by stimulating the formation of EPCAM + cancer stem-like cells via glycolysis. Transl Oncol 2024; 49:102082. [PMID: 39126936 PMCID: PMC11364050 DOI: 10.1016/j.tranon.2024.102082] [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: 04/06/2024] [Revised: 07/01/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The mechanisms by which SLC2A1 enhances chemo-resistance of taxanes to non-small cell lung cancer (NSCLC) remains enigmatic. METHODS An investigation into the SLC2A1 expression pattern and prognosis across diverse datasets, as well as our internally collected samples, was undertaken. Additionally, the biological function of SLC2A1 was further delved into through in vitro experiments. The study also examined the chemo-resistance of NSCLC to taxanes using CCK-8, Annexin-V, and caspase-3 assays. Furthermore, the impact of taxanes on SLC2A1 expression was determined via western blot analysis. The effects of SLC2A1 on the formation of CSCs was examined via flow cytometry and metabolomics techniques. Finally, the impact of SLC2A1 on the tumor microenvironment was analyzed using single-cell sequencing and cellchat. RESULTS In the present investigation, it was observed that there was an elevated expression of SLC2A1 in NSCLC tumor tissues, which exhibited a significant association with a poorer prognosis. SLC2A1 overexpression in vitro promoted NSCLC cell proliferation, invasion, migration, chemo-resistance, and the formation of CD90+ and EpCAM+ CSCs. NSCLC cells were categorized based on SLC2A1 and EpCAM expression. SLC2A1highEpCAM+ CSCs were more chemo-resistance to taxanes. NSCLC patients with high SLC2A1 and EpCAM expression had poorer prognosis. Mechanically, SLC2A1 promoted the formation of CD90+ and EpCAM+ CSCs via activating glycolysis. Finally, SLC2A1low tumor cells promoted CD8+T cell function via HLA-A, B, C, and suppressed NK cell function via HLA-E. CONCLUSION Together, SLC2A1 plays an important role in enhancing chemo-resistance of taxanes to NSCLC.
Collapse
Affiliation(s)
- Zhe Yu
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jian Sun
- Center for Reproduction and Genetics, Suzhou Municipal Hospital &The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Kai Fang
- Department of Oncological Surgery, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu province, China
| | - Jingwei Xu
- Department of Thoracic Surgery, Suzhou Municipal Hospital & The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jian Yang
- Department of Thoracic Surgery, Suzhou Municipal Hospital & The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Dai Chunlei
- Department of Thoracic Surgery, Suzhou Municipal Hospital & The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yongsheng Gong
- Department of Thoracic Surgery, Suzhou Municipal Hospital & The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Haitao Ma
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
28
|
Jiang J, Ye P, Sun N, Zhu W, Yang M, Yu M, Yu J, Zhang H, Gao Z, Zhang N, Guo S, Ji Y, Li S, Zhang C, Miao S, Chai M, Liu W, An Y, Hong J, Wei W, Zhang S, Qiu H. Yap methylation-induced FGL1 expression suppresses anti-tumor immunity and promotes tumor progression in KRAS-driven lung adenocarcinoma. Cancer Commun (Lond) 2024; 44:1350-1373. [PMID: 39340215 PMCID: PMC12015977 DOI: 10.1002/cac2.12609] [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: 12/15/2023] [Revised: 09/04/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Despite significant strides in lung cancer immunotherapy, the response rates for Kirsten rat sarcoma viral oncogene homolog (KRAS)-driven lung adenocarcinoma (LUAD) patients remain limited. Fibrinogen-like protein 1 (FGL1) is a newly identified immune checkpoint target, and the study of related resistance mechanisms is crucial for improving the treatment outcomes of LUAD patients. This study aimed to elucidate the potential mechanism by which FGL1 regulates the tumor microenvironment in KRAS-mutated cancer. METHODS The expression levels of FGL1 and SET1 histone methyltransferase (SET1A) in lung cancer were assessed using public databases and clinical samples. Lentiviruses were constructed for transduction to overexpress or silence FGL1 in lung cancer cells and mouse models. The effects of FGL1 and Yes-associated protein (Yap) on the immunoreactivity of cytotoxic T cells in tumor tissues were evaluated using immunofluorescence staining and flow cytometry. Chromatin immunoprecipitation and dual luciferase reporter assays were used to study the SET1A-directed transcriptional program. RESULTS Upregulation of FGL1 expression in KRAS-mutated cancer was inversely correlated with the infiltration of CD8+ T cells. Mechanistically, KRAS activated extracellular signal-regulated kinase 1/2 (ERK1/2), which subsequently phosphorylated SET1A and increased its stability and nuclear localization. SET1A-mediated methylation of Yap led to Yap sequestration in the nucleus, thereby promoting Yap-induced transcription of FGL1 and immune evasion in KRAS-driven LUAD. Notably, dual blockade of programmed cell death-1 (PD-1) and FGL1 further increased the therapeutic efficacy of anti-PD-1 immunotherapy in LUAD patients. CONCLUSION FGL1 could be used as a diagnostic biomarker of KRAS-mutated lung cancer, and targeting the Yap-FGL1 axis could increase the efficacy of anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Ji Jiang
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Pengfei Ye
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Ningning Sun
- School of Nursing, Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Weihua Zhu
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Mei Yang
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Manman Yu
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Jingjing Yu
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Hui Zhang
- School of Nursing, Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Zijie Gao
- School of Nursing, Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Ningjie Zhang
- School of Nursing, Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Shijie Guo
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Yuru Ji
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Siqi Li
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Cuncun Zhang
- School of Nursing, Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Sainan Miao
- School of Nursing, Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Mengqi Chai
- School of Nursing, Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Wenmin Liu
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Yue An
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Jian Hong
- Department of HematologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiP. R. China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Shihao Zhang
- Institute of Clinical Pharmacology, Anhui Medical University; Key Laboratory of Anti‐Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Centre of Anti‐Inflammatory and Immune MedicineHefeiAnhuiP. R. China
| | - Huan Qiu
- School of Nursing, Anhui Medical UniversityHefeiAnhuiP. R. China
| |
Collapse
|
29
|
Lin Y, Chen M, Huang S, Chen Y, Ho JH, Lin F, Tan X, Chiang H, Huang C, Tu C, Cho D, Chiu S. Targeting Dual Immune Checkpoints PD-L1 and HLA-G by Trispecific T Cell Engager for Treating Heterogeneous Lung Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309697. [PMID: 39234811 PMCID: PMC11538689 DOI: 10.1002/advs.202309697] [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: 12/11/2023] [Revised: 06/04/2024] [Indexed: 09/06/2024]
Abstract
Immunotherapy targeting immune checkpoints (ICPs), such as programmed death-ligand-1 (PD-L1), is used as a treatment option for advanced or metastatic non-small cell lung cancer (NSCLC). However, overall response rate to anti-PD-L1 treatment is limited due to antigen heterogeneity and the immune-suppressive tumor microenvironment. Human leukocyte antigen-G (HLA-G), an ICP as well as a neoexpressed tumor-associated antigen, is previously demonstrated to be a beneficial target in combination with anti-PD-L1. In this study, a nanobody-based trispecific T cell engager (Nb-TriTE) is developed, capable of simultaneously binding to T cells, macrophages, and cancer cells while redirecting T cells toward tumor cells expressing PD-L1- and/or HLA-G. Nb-TriTE shows broad spectrum anti-tumor effects in vitro by augmenting cytotoxicity mediated by human peripheral blood mononuclear cells (PBMCs). In a humanized immunodeficient murine NSCLC model, Nb-TriTE exhibits superior anti-cancer potency compared to monoclonal antibodies and bispecific T cell engagers. Nb-TriTE, at the dose with pharmacoactivity, does not induce additional enhancement of circulating cytokines secretion from PMBCs. Nb-TriTE effectively prolongs the survival of mice without obvious adverse events. In conclusion, this study introduces an innovative therapeutic approach to address the challenges of immunotherapy and the tumor microenvironment in NSCLC through utilizing the dual ICP-targeting Nb-TriTE.
Collapse
Affiliation(s)
- Yu‐Chuan Lin
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
| | - Mei‐Chih Chen
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
| | - Shi‐Wei Huang
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichung City404Taiwan
- Institute of Biomedical SciencesNational Chung Hsing UniversityTaichung City402Taiwan
| | - Yeh Chen
- Department of Food Science and BiotechnologyNational Chung Hsing UniversityTaichung City402Taiwan
| | - Jennifer Hui‐Chun Ho
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- Center for Translational Genomics and Regenerative Medicine ResearchChina Medical University HospitalTaichung City404Taiwan
- Department of OphthalmologyChina Medical University HospitalChina Medical UniversityTaichung City404Taiwan
- Department of Medical ResearchEye CenterChina Medical University HospitalTaichung City404Taiwan
| | - Fang‐Yu Lin
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
| | - Xiao‐Tong Tan
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
| | - Hung‐Che Chiang
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- College of MedicineChina Medical UniversityTaichung City404Taiwan
| | - Chiu‐Ching Huang
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- Division of Nephrology and the Kidney InstituteDepartment of Internal MedicineChina Medical University HospitalTaichung City404Taiwan
| | - Chih‑Yen Tu
- Division of Pulmonary and Critical CareDepartment of Internal MedicineChina Medical University HospitalTaichung City404Taiwan
- School of MedicineCollege of MedicineChina Medical UniversityTaichung City404Taiwan
| | - Der‐Yang Cho
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichung City404Taiwan
- Drug Development CenterChina Medical UniversityTaichung City404Taiwan
- Department of NeurosurgeryChina Medical University HospitalTaichung City404Taiwan
| | - Shao‐Chih Chiu
- Translational Cell Therapy CenterChina Medical University HospitalNo. 2, Yude Rd., North Dist.Taichung City404Taiwan
- Shine‐On BioMedical Co. Ltd.Rm. B, 10F., No. 573, Sec. 2, Taiwan Blvd., West Dist.Taichung City403Taiwan
- Institute of New Drug DevelopmentChina Medical UniversityTaichung City404Taiwan
- Drug Development CenterChina Medical UniversityTaichung City404Taiwan
| |
Collapse
|
30
|
Feng X, Li Z, Liu Y, Chen D, Zhou Z. CRISPR/Cas9 technology for advancements in cancer immunotherapy: from uncovering regulatory mechanisms to therapeutic applications. Exp Hematol Oncol 2024; 13:102. [PMID: 39427211 PMCID: PMC11490091 DOI: 10.1186/s40164-024-00570-y] [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: 07/25/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
In recent years, immunotherapy has developed rapidly as a new field of tumour therapy. However, the efficacy of tumour immunotherapy is not satisfactory due to the immune evasion mechanism of tumour cells, induction of immunosuppressive tumour microenvironment (TME), and reduction of antigen delivery, etc. CRISPR/Cas9 gene editing technology can accurately modify immune and tumour cells in tumours, and improve the efficacy of immunotherapy by targeting immune checkpoint molecules and immune regulatory genes, which has led to the great development and application. In current clinical trials, there are still many obstacles to the application of CRISPR/Cas9 in tumour immunotherapy, such as ensuring the accuracy and safety of gene editing, overcoming overreactive immune responses, and solving the challenges of in vivo drug delivery. Here we provide a systematic review on the application of CRISPR/Cas9 in tumour therapy to address the above existing problems. We focus on CRISPR/Cas9 screening and identification of immunomodulatory genes, targeting of immune checkpoint molecules, manipulation of immunomodulators, enhancement of tumour-specific antigen presentation and modulation of immune cell function. Second, we also highlight preclinical studies of CRISPR/Cas9 in animal models and various delivery systems, and evaluate the efficacy and safety of CRISPR/Cas9 technology in tumour immunotherapy. Finally, potential synergistic approaches for combining CRISPR/Cas9 knockdown with other immunotherapies are presented. This study underscores the transformative potential of CRISPR/Cas9 to reshape the landscape of tumour immunotherapy and provide insights into novel therapeutic strategies for cancer patients.
Collapse
Affiliation(s)
- Xiaohang Feng
- Department of Colorectal Surgery, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhengxing Li
- Department of Colorectal Surgery, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuping Liu
- Department of Colorectal Surgery, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Di Chen
- Biomedical Sciences, College of Medicine and Veterinary Medicine, Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK
- Center for Reproductive Medicine of The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhuolong Zhou
- Department of Colorectal Surgery, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
- Biomedical Sciences, College of Medicine and Veterinary Medicine, Edinburgh Medical School, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
31
|
Ou Y, Liang S, Gao Q, Shang Y, Liang J, Zhang W, Liu S. Prognostic value of inflammatory markers NLR, PLR, LMR, dNLR, ANC in melanoma patients treated with immune checkpoint inhibitors: a meta-analysis and systematic review. Front Immunol 2024; 15:1482746. [PMID: 39493767 PMCID: PMC11527641 DOI: 10.3389/fimmu.2024.1482746] [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: 08/18/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) are an emerging tumor treatment pathway after traditional surgery, chemoradiotherapy, and targeted therapy. They have proven to be effective in a variety of cancers, but may not respond to non-target populations. Inflammatory markers such as neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), lymphocyte to monocyte ratio (LMR), derived neutrophil lymphocyte ratio (dNLR), and neutrophil count (ANC) have been shown to be strongly associated with tumor prognosis, but their prognostic significance remains controversial. We therefore performed a meta-analysis to explore the association between NLR, PLR, LMR, dNLR, ANC and prognostic and clinicopathological factors in melanoma patients treated with ICIs. Methods A comprehensive search was conducted in Pubmed, Embase, Web Of Science and Cochrane databases, and the last search time was July 2024. To estimate the prognostic value of NLR, PLR, LMR, dNLR, ANC for PFS and OS, hazard ratio (HR) and corresponding 95% confidence interval (CI) estimates were used. Results This meta-analysis ultimately included 22 cohort studies involving 3235 melanoma patients. Meta-analysis results showed that high levels of NLR in melanoma patients receiving ICIs were associated with poorer OS and PFS, Merging the HR respectively OS [HR = 2.21, 95% CI (1.62, 3.02), P < 0.001], PFS [HR = 1.80, 95% CI (1.40, 2.30), P < 0.001]; High levels of PLR were associated with poor OS and PFS, and the combined HR was OS[HR=2.15,95%CI(1.66,2.80),P < 0.001] and PFS[HR=1.67,95%CI(1.31,2.12),P < 0.001]. High levels of dNLR were associated with poor OS and PFS, with combined HR being OS[HR=2.34,95%CI(1.96,2.79),P < 0.001] and PFS[HR=2.05,95%CI(1.73,2.42),P < 0.001], respectively. High ANC was associated with poor OS and PFS, and combined HR was OS[HR=1.95,95%CI(1.16,3.27),P < 0.001] and PFS[HR=1.63,95%CI(1.04,2.54),P=0.032], respectively. Increased LMR was associated with prolonged OS and PFS, with combined HR being OS[HR=0.36, 95%CI(0.19,0.70),P < 0.001] and PFS[HR=0.56,95%CI(0.40,0.79),P=0.034], respectively. Conclusion In melanoma patients treated with ICIs, elevated levels of NLR, PLR, dNLR, and ANC were associated with poorer overall survival OS and PFS. Conversely, a high LMR correlated with improved OS and PFS. Subgroup analyses indicated that dNLR may be linked to a worse prognosis in melanoma patients. In summary, inflammatory markers such as NLR, PLR, LMR, dNLR, and ANC serve as effective biomarkers for the prognostic assessment of melanoma patients following ICI treatment. These markers provide valuable insights for treatment decision-making in the realm of melanoma immunotherapy, and we anticipate further high-quality prospective studies to validate our findings in the future. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#recordDetails, identifier CRD42024573406.
Collapse
Affiliation(s)
- Yan Ou
- Department of Plastic and Aesthetic Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Shufang Liang
- Department of Burns and Plastic Surgery, 969th Hospital of PLA Joint Logistic Support Force, Inner Mongolia, China
| | - Qiangqiang Gao
- Department of Proctology, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Yongran Shang
- The First Clinical Medical College of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Junfang Liang
- Department of Plastic and Aesthetic Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Weitao Zhang
- Department of Plastic and Aesthetic Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| | - Sha Liu
- Department of Plastic and Aesthetic Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Shaanxi, China
| |
Collapse
|
32
|
Groeger S, Meyle J. The role of programmed death receptor (PD-)1/PD-ligand (L)1 in periodontitis and cancer. Periodontol 2000 2024; 96:150-169. [PMID: 38351432 PMCID: PMC11579837 DOI: 10.1111/prd.12548] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/02/2023] [Accepted: 12/14/2023] [Indexed: 11/22/2024]
Abstract
The programmed-death-ligand-1 (PD-L1) is an immune-modulating molecule that is constitutively expressed on various immune cells, different epithelial cells and a multitude of cancer cells. It is a costimulatory molecule that may impair T-cell mediated immune response. Ligation to the programmed-death-receptor (PD)-1, on activated T-cells and further triggering of the related signaling pathways can induce T-cells apoptosis or anergy. The upregulation of PD-L1 in various cancer types, including oral squamous cell carcinomas, was demonstrated and has been linked to immune escape of tumors and poor prognosis. A bidirectional relationship exists between the increased PD-L1 expression and periodontitis as well as the epithelial-mesenchymal transition (EMT), a process of interconversion of epithelial cells to mesenchymal cells that may induce immune escape of tumors. Interaction between exosomal PD-L1 and PD-1 on T-cells may cause immunosuppression by blocking the activation and proliferation of T-cells. The efficacy and importance of treatment with PD-1/PD-L1 checkpoint inhibitors and their prognostic influence on human cancers was demonstrated. Regarding PD-1/PD-L1 checkpoint inhibitors, resistances exist or may develop, basing on various factors. Further investigations of the underlying mechanisms will help to overcome the therapeutic limitations that result from resistances and to develop new strategies for the treatment of cancer.
Collapse
Affiliation(s)
- Sabine Groeger
- Department of Periodontology, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
- Department of Orthodontics, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
| | - Joerg Meyle
- Department of Periodontology, Dental SchoolJustus‐Liebig‐University of GiessenGiessenGermany
| |
Collapse
|
33
|
Li H, Lei Y, Chen N, Guo G, Xiang X, Huang Y. circRNA-CPA4 Regulates Cell Proliferation and Apoptosis of Non-small Cell Lung Cancer via the miR-1183/PDPK1 Axis. Biochem Genet 2024; 62:4087-4102. [PMID: 38273153 DOI: 10.1007/s10528-023-10641-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/12/2023] [Indexed: 01/27/2024]
Abstract
Non-small-cell lung cancer (NSCLC) stands as a prevalent subtype of lung cancer, with circular RNAs emerging as key players in cancer development. This study elucidates the role of circRNA-CPA4 in NSCLC. Elevated circRNA-CPA4 expression in NSCLC lines was confirmed through qRT-PCR. Silencing circRNA-CPA4 with shRNA revealed, through CCK-8, colony formation, and flow cytometry assays, a notable constraint on proliferation and promotion of apoptosis in NSCLC cells. Subcellular localization analysis, RNA immunoprecipitation, and expression level assessments were employed to decipher the intricate interplay among miR-1183, circRNA-CPA4, and PDPK1. Results demonstrated heightened circRNA-CPA4 levels in NSCLC, and its knockdown curtailed NSCLC growth in vivo. Acting as a molecular sponge for miR-1183, circRNA-CPA4 regulated PDPK1 expression. Conversely, inhibiting miR-1183 counteracted the impact of circRNA-CPA4 silencing, reinstating NSCLC cell proliferation, and impeding apoptosis. Overall, this study unveils a novel mechanism: circRNA-CPA4 promotes PDPK1 expression by sequestering miR-1183, fostering NSCLC cell proliferation, and hindering apoptosis.
Collapse
Affiliation(s)
- Heng Li
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China
| | - Yujie Lei
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunzhou Road 519, Kunming, 650118, China
| | - Nan Chen
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China
| | - Gang Guo
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China
| | - Xudong Xiang
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China
| | - Yunchao Huang
- Department of Thoracic Surgery II, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, 650118, China.
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunzhou Road 519, Kunming, 650118, China.
| |
Collapse
|
34
|
Liao CY, Chen YM, Wu YT, Chao HS, Chiu HY, Wang TW, Chen JR, Shiao TH, Lu CF. Personalized prediction of immunotherapy response in lung cancer patients using advanced radiomics and deep learning. Cancer Imaging 2024; 24:129. [PMID: 39350284 PMCID: PMC11440728 DOI: 10.1186/s40644-024-00779-4] [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: 05/10/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Lung cancer (LC) is a leading cause of cancer-related mortality, and immunotherapy (IO) has shown promise in treating advanced-stage LC. However, identifying patients likely to benefit from IO and monitoring treatment response remains challenging. This study aims to develop a predictive model for progression-free survival (PFS) in LC patients with IO based on clinical features and advanced imaging biomarkers. MATERIALS AND METHODS A retrospective analysis was conducted on a cohort of 206 LC patients receiving IO treatment. Pre-treatment computed tomography images were used to extract advanced imaging biomarkers, including intratumoral and peritumoral-vasculature radiomics. Clinical features, including age, gene status, hematology, and staging, were also collected. Key radiomic and clinical features for predicting IO outcomes were identified using a two-step feature selection process, including univariate Cox regression and chi-squared test, followed by sequential forward selection. The DeepSurv model was constructed to predict PFS based on clinical and radiomic features. Model performance was evaluated using the area under the time-dependent receiver operating characteristic curve (AUC) and concordance index (C-index). RESULTS Combining radiomics of intratumoral heterogeneity and peritumoral-vasculature with clinical features demonstrated a significant enhancement (p < 0.001) in predicting IO response. The proposed DeepSurv model exhibited a prediction performance with AUCs ranging from 0.76 to 0.80 and a C-index of 0.83. Furthermore, the predicted personalized PFS curves revealed a significant difference (p < 0.05) between patients with favorable and unfavorable prognoses. CONCLUSIONS Integrating intratumoral and peritumoral-vasculature radiomics with clinical features enabled the development of a predictive model for PFS in LC patients with IO. The proposed model's capability to estimate individualized PFS probability and differentiate the prognosis status held promise to facilitate personalized medicine and improve patient outcomes in LC.
Collapse
Affiliation(s)
- Chien-Yi Liao
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Beitou District, Taipei, 112, Taiwan
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Te Wu
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Heng-Sheng Chao
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hwa-Yen Chiu
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Wei Wang
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Jyun-Ru Chen
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Beitou District, Taipei, 112, Taiwan
| | - Tsu-Hui Shiao
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chia-Feng Lu
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Linong Street, Beitou District, Taipei, 112, Taiwan.
- Institute of Biophotonics, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
35
|
Medina-Ceballos E, Machado I, Giner F, Blázquez-Bujeda Á, Espino M, Navarro S, Llombart-Bosch A. Immunological Tumor Microenvironment of Solitary Fibrous Tumors-Associating Immune Infiltrate with Variables of Prognostic Significance. Cancers (Basel) 2024; 16:3222. [PMID: 39335193 PMCID: PMC11430690 DOI: 10.3390/cancers16183222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/14/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Solitary fibrous tumors (SFTs) are morphologically heterogeneous tumors characterized by the NAB2::STAT6 gene fusion. Clinical outcomes may vary widely, and while most cases have favorable outcomes, some can progress to aggressive disease, manifesting as recurrence and metastasis, and ultimately resulting in patient death. Herein, we analyze the immunological tumor microenvironment (ITME) of SFTs, aiming to determine its prognostic value and correlation with established risk stratification systems (RSSs). METHODS A retrospective observational multicenter study of 52 fusion-confirmed SFTs with clinical follow-up data. Immunohistochemical analysis including CD163, CD68, CD3, CD8, CD20, PDL-1, PD-1, and LAG1 were evaluated in tissue microarrays, using an analog scale with scores ranging from 0 to 3 (0 = ≤9, 1 = 10-49, 2 = 50-99, and 3 = >100 positive cells per 10 high-power fields). The expression of these markers was correlated with clinical outcomes, morphological characteristics previously evaluated in whole slide tissue sections (hypercellularity/hypocellularity, round-oval or spindle dominant constituent cell (DCC) morphology, and necrosis), Ki67, overall survival, and RSS. RESULTS Only one of the fifty-two cases studied showed progression. In the multivariate analysis, neither the presence nor absence of immune cells (B-lymphocytes, T-lymphocytes, and macrophages) showed any association with the assessed RSSs (Demicco, Sugita, G-score, and Huang). Interestingly, the case that showed progression had high immune infiltrate with expression of CD68, CD163, CD8, and CD20 markers (score of 3). Round-oval cell morphology was associated with the presence of higher levels of CD163 macrophages. Lastly, the scant presence of CD20+ lymphocytes correlated with less necrosis, and cases with higher PDL-1 expression correlated with increased Ki67 values. All cases were negative for LAG-1 and PD-1. CONCLUSIONS SFT ITME components correlated with independent variables with prognostic significance. Nevertheless, ITME did not correlate with RSS scores.
Collapse
Affiliation(s)
| | - Isidro Machado
- Pathology Department, Instituto Valenciano de Oncología, 46009 Valencia, Spain
- Patologika Laboratory, Quirón-Salud, 46010 Valencia, Spain
- Cancer CIBER (CIBERONC), 28029 Madrid, Spain
| | - Francisco Giner
- Pathology Department, University Hospital La Fe, 46010 Valencia, Spain
- Pathology Department, University of Valencia, 46010 Valencia, Spain
| | | | - Mónica Espino
- Pathology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
| | - Samuel Navarro
- Pathology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain
- Cancer CIBER (CIBERONC), 28029 Madrid, Spain
- Pathology Department, University of Valencia, 46010 Valencia, Spain
| | | |
Collapse
|
36
|
Si Q, Bai M, Wang X, Wang T, Qin Y. Photonanozyme-Kras-ribosome combination treatment of non-small cell lung cancer after COVID-19. Front Immunol 2024; 15:1420463. [PMID: 39308869 PMCID: PMC11412844 DOI: 10.3389/fimmu.2024.1420463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 08/14/2024] [Indexed: 09/25/2024] Open
Abstract
With the outbreak of the coronavirus disease 2019 (COVID-19), reductions in T-cell function and exhaustion have been observed in patients post-infection of COVID-19. T cells are key mediators of anti-infection and antitumor, and their exhaustion increases the risk of compromised immune function and elevated susceptibility to cancer. Non-small cell lung cancer (NSCLC) is the most common subtype of lung cancer with high incidence and mortality. Although the survival rate after standard treatment such as surgical treatment and chemotherapy has improved, the therapeutic effect is still limited due to drug resistance, side effects, and recurrence. Recent advances in molecular biology and immunology enable the development of highly targeted therapy and immunotherapy for cancer, which has driven cancer therapies into individualized treatments and gradually entered clinicians' views for treating NSCLC. Currently, with the development of photosensitizer materials, phototherapy has been gradually applied to the treatment of NSCLC. This review provides an overview of recent advancements and limitations in different treatment strategies for NSCLC under the background of COVID-19. We discuss the latest advances in phototherapy as a promising treatment method for NSCLC. After critically examining the successes, challenges, and prospects associated with these treatment modalities, their profound prospects were portrayed.
Collapse
Affiliation(s)
- Qiaoyan Si
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
- School of Biomedical Engineering, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Mingjian Bai
- School of Biomedical Engineering, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Xiaolong Wang
- School of Biomedical Engineering, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Tianyu Wang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, China
| | - Yan Qin
- School of Biomedical Engineering, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
37
|
Cánovas-Cervera I, Nacher-Sendra E, Suay G, Lahoz A, García-Giménez JL, Mena-Mollá S. Role of miRNAs as epigenetic regulators of immune checkpoints in lung cancer immunity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 390:109-139. [PMID: 39864893 DOI: 10.1016/bs.ircmb.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The advent of immunotherapy in cancer has provided new avenues in the treatment of many malignancies at various stages. Specifically, immune checkpoint inhibitors (ICIs) have transformed the field of lung cancer treatment. However, since some tumors can evade the immune system, not all patients respond properly. Recent research has provided evidence showing how microRNAs (miRNAs) are involved in regulating many immune checkpoints. MiRNAs have demonstrated their ability to modulate immune evasion of tumor cells. Currently, reliable markers are being sought to predict the efficacy of immunotherapy in these types of cancers. Therefore, the association of serum miRNAs and the response of ICIs in lung cancer is under study. Many miRNA molecules and their corresponding target genes have been identified in the regulation of chemoresistance. Therefore, elucidating how these miRNAs control the function of immune checkpoints, as well as the effectiveness of therapies based on ICIs set the basis for the development of new biomarkers to predict treatment response to ICIs. This chapter delves into the molecular role of miRNAs interacting with ICs, such as PD-1 and PD-L1, and the clinical utility of miRNAs, such as miR-16, miR-146a, and miR-335, in predicting treatment response to ICI-based therapy in lung cancer. The aim is to provide a deep insight of the current landscape, serving as a cornerstone for further research.
Collapse
Affiliation(s)
- Irene Cánovas-Cervera
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Elena Nacher-Sendra
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain
| | - Guillermo Suay
- Medical Oncology Department, Hospital Universitari i Politècnic La Fe, Valencia, Spain
| | - Agustin Lahoz
- Biomarkers and Precision Medicine Unit, Health Research Institute-Hospital La Fe, Valencia, Spain; Analytical Unit, Health Research Institute-Hospital La Fe, Valencia, Spain
| | - José Luis García-Giménez
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia, Spain; Consortium Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, Valencia, Spain.
| | - Salvador Mena-Mollá
- INCLIVA Health Research Institute, INCLIVA, Valencia, Spain; Department of Physiology, Faculty of Pharmacy, University of Valencia, Burjassot, Spain
| |
Collapse
|
38
|
Zhao H, Zhang W, Lu Y, Dong Y, He Z, Zhen H, Li Q. Inosine enhances the efficacy of immune-checkpoint inhibitors in advanced solid tumors: A randomized, controlled, Phase 2 study. Cancer Med 2024; 13:e70143. [PMID: 39267574 PMCID: PMC11393481 DOI: 10.1002/cam4.70143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND This study aimed to evaluate whether inosine enhances the efficacy of immune-checkpoint inhibitors in human malignant solid tumors. METHODS This single-center, prospective, randomized, open-label study was conducted, from January 2021 to December 2022, in Beijing Friendship Hospital, Capital Medical University, and participants were randomly assigned (1:1) to either the inosine (trial) or non-inosine (control) group that received inosine (dosage: 0.2 g, three times/day) + PD-1/PD-L1 inhibitor or only PD-1/PD-L1 inhibitor ± targeted ± chemotherapy, respectively. Efficacy was assessed every 6 weeks (i.e., after every two-three treatment cycles). The primary endpoint was the objective response rate (ORR); the secondary endpoints were disease control rate, overall survival (OS), and progression-free survival (PFS). The trial was registered at ClinicalTrials.gov (NCT05809336). RESULTS Among the 172 participants with advanced malignant solid tumors, 86 each were assigned to the inosine and non-inosine groups, wherein the median PFS (95% CI) was 7.00 (5.31-8.69) and 4.40 (3.10-5.70) months, respectively (hazard ratio [HR] 0.63; 95% CI 0.44-0.90, p = 0.011), and the ORR was 26.7% and 15.1%, respectively (p = 0.061). In the inosine and non-inosine groups, the median OS was not reached and was 29.67 (95% CI 17.40-41.94) months, respectively (HR 1.05 [95% CI 0.59-1.84], p = 0.874). Compared with the non-inosine group, the median PFS and ORR of the inosine group were significantly prolonged and improved in the multiple exploratory subgroup analyses. The safety analysis showed that Grades 3 and 4 adverse reactions occurred in 25 (29%) and 31 (36%) patients in the inosine and non-inosine groups, respectively, and tended to decrease in the inosine group compared with the non-inosine group. CONCLUSION Inosine had a tendency to enhance the efficacy of immune-checkpoint inhibitors and reduced immunotherapy-related adverse reactions.
Collapse
Affiliation(s)
- Haiqing Zhao
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
- Internal Medicine DepartmentPeople's Hospital of Shen chi CountyShanxiPeople's Republic of China
| | - Wei Zhang
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Yuting Lu
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Yin Dong
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Zhihao He
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Hongchao Zhen
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Qin Li
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| |
Collapse
|
39
|
Iwata K, Suzawa K, Hashimoto K, Tanaka S, Shien K, Miyoshi K, Yamamoto H, Okazaki M, Sugimoto S, Toyooka S. Utility of neutrophil-to-lymphocyte ratio as an indicator of tumor immune status in non-small cell lung cancer. Jpn J Clin Oncol 2024; 54:895-902. [PMID: 38704243 PMCID: PMC11322889 DOI: 10.1093/jjco/hyae058] [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: 01/21/2024] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Neutrophil-to-lymphocyte ratio (NLR) has been reported as a prognostic biomarker in non-small cell lung cancer (NSCLC); however, the underlying biological rationale remains unclear. The present study aimed to explore the potential utility of NLR as a surrogate biomarker for immune response to cancer and to elucidate the underlying mechanism. METHODS This retrospective study included the medical records of 120 patients with NSCLC who underwent surgery at the study institution in 2012. NLR in peripheral blood was determined from blood test within 30 days before surgery. Tumor immune status was evaluated using immunohistochemical staining to identify CD3+, CD8+ and FOXP3+ tumor-infiltrating lymphocytes (TILs), and the relationship of NLR, with clinicopathologic characteristics including 5-year overall survival (OS), and the tumor immune status was investigated. The median values of NLR and TIL count were used as cutoff points. RESULTS The 5-year OS was significantly better in patients with low NLR (<2.2) than in those with high NLR (≥2.2) (70.1% vs. 56.8%, P = 0.042) and in patients with high CD3+ TIL count (≥242) than in those with low CD3+ TIL count (<242) (70% vs. 56.8%, P = 0.019). Additionally, the CD3+ TIL count was negatively correlated with preoperative NLR (P = 0.005). CONCLUSION NLR might potentially reflect the immune status of tumor microenvironment, explaining its impact on prognosis of patients with NSCLC.
Collapse
Affiliation(s)
- Kazuma Iwata
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Ken Suzawa
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
- Department of Thoracic Surgery, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kohei Hashimoto
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
- Department of Thoracic Surgery, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Shin Tanaka
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
- Department of Thoracic Surgery, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kazuhiko Shien
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
- Department of Thoracic Surgery, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Kentaroh Miyoshi
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
- Department of Thoracic Surgery, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Hiromasa Yamamoto
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
- Department of Thoracic Surgery, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Mikio Okazaki
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
- Department of Thoracic Surgery, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Seiichiro Sugimoto
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
- Department of Thoracic Surgery, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Brest and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
- Department of Thoracic Surgery, Okayama University Hospital, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| |
Collapse
|
40
|
Chen H, Wei J, Zhu Z, Hou Y. Multifaceted roles of PD-1 in tumorigenesis: From immune checkpoint to tumor cell-intrinsic function. Mol Carcinog 2024; 63:1436-1448. [PMID: 38751009 DOI: 10.1002/mc.23740] [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: 12/21/2023] [Revised: 02/27/2024] [Accepted: 05/04/2024] [Indexed: 07/10/2024]
Abstract
Programmed cell death 1 (PD-1), a key immune checkpoint receptor, has been extensively studied for its role in regulating immune responses in cancer. However, recent research has unveiled a complex and dual role for PD-1 in tumorigenesis. While PD-1 is traditionally associated with immune cells, this article explores its expression in various cancer cells and its impact on cancer progression. PD-1's functions extend beyond immune regulation, as it has been found to both promote and suppress tumor growth, depending on the cancer type. These findings have significant implications for the future of cancer treatment and our understanding of the immune response in the context of cancer. This article calls for further research into the multifaceted roles of PD-1 to optimize its therapeutic potential and improve patient outcomes in the fight against cancer.
Collapse
Affiliation(s)
- Huiqing Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Jiayu Wei
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| | - Zhen Zhu
- Zhenjiang Stomatological Hospital, Zhenjiang, China
| | - Yongzhong Hou
- School of Life Sciences, Jiangsu University, Zhenjiang, China
| |
Collapse
|
41
|
Chen M, Li H, Qu B, Huang X. The Roles of T cells in Immune Checkpoint Inhibitor-Induced Arthritis. Aging Dis 2024:AD.2024.0546. [PMID: 39122457 DOI: 10.14336/ad.2024.0546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy, a novel anti-tumor strategy, can specifically eliminate tumors by activating the immune system and inhibiting tumor immune escape. However, ICI therapy can lead to notable negative outcomes known as immune-related adverse events (irAEs). ICI-induced arthritis, also known as ICI arthritis, stands as the prevailing form of irAEs. The purpose of this review is to highlight the crucial functions of T cells in the progression of ICI arthritis. Under the influence of different signaling molecules, T cells could gather in large numbers within the synovial membrane of joints, releasing inflammatory substances and enzymes that harm healthy tissues, ultimately causing ICI arthritis. Moreover, considering the functions of T cells in triggering ICI arthritis, this review suggests several treatments to prevent ICI arthritis, including inhibiting the overstimulation of T cells at the synovial sac of joints, enhancing the precision of ICI medications, and directing ICI drugs specifically towards tumor tissues instead of joints. Collectively, T lymphocytes play a vital role in the onset of ICI arthritis, offering a hopeful perspective on treating ICI arthritis through the specific targeting of T cells within the affected joints.
Collapse
Affiliation(s)
- Maike Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Huili Li
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Baicheng Qu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
42
|
Sun D, Tan L, Chen Y, Yuan Q, Jiang K, Liu Y, Xue Y, Zhang J, Cao X, Xu M, Luo Y, Xu Z, Xu Z, Xu W, Shen M. CXCL5 impedes CD8 + T cell immunity by upregulating PD-L1 expression in lung cancer via PXN/AKT signaling phosphorylation and neutrophil chemotaxis. J Exp Clin Cancer Res 2024; 43:202. [PMID: 39034411 PMCID: PMC11264977 DOI: 10.1186/s13046-024-03122-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Lung cancer remains one of the most prevalent cancer types worldwide, with a high mortality rate. Upregulation of programmed cell death protein 1 (PD-1) and its ligand (PD-L1) may represent a key mechanism for evading immune surveillance. Immune checkpoint blockade (ICB) antibodies against PD-1 or PD-L1 are therefore widely used to treat patients with lung cancer. However, the mechanisms by which lung cancer and neutrophils in the microenvironment sustain PD-L1 expression and impart stronger inhibition of CD8+ T cell function remain unclear. METHODS We investigated the role and underlying mechanism by which PD-L1+ lung cancer and PD-L1+ neutrophils impede the function of CD8+ T cells through magnetic bead cell sorting, quantitative real-time polymerase chain reaction (RT-PCR), western blotting, enzyme-linked immunosorbent assays, confocal immunofluorescence, gene silencing, flow cytometry, etc. In vivo efficacy and safety studies were conducted using (Non-obeseDiabetes/severe combined immune deficiency) SCID/NOD mice. Additionally, we collected clinical and prognostic data from 208 patients who underwent curative lung cancer resection between 2017 and 2018. RESULTS We demonstrated that C-X-C motif chemokine ligand 5 (CXCL5) is markedly overexpressed in lung cancer cells and is positively correlated with a poor prognosis in patients with lung cancer. Mechanistically, CXCL5 activates the phosphorylation of the Paxillin/AKT signaling cascade, leading to upregulation of PD-L1 expression and the formation of a positive feedback loop. Moreover, CXCL5 attracts neutrophils, compromising CD8+ T cell-dependent antitumor immunity. These PD-L1+ neutrophils aggravate CD8+ T cell exhaustion following lung cancer domestication. Combined treatment with anti-CXCL5 and anti-PD-L1 antibodies significantly inhibits tumor growth in vivo. CONCLUSIONS Our findings collectively demonstrate that CXCL5 promotes immune escape through PD-L1 upregulation in lung cancer and neutrophils chemotaxis through autocrine and paracrine mechanisms. CXCL5 may serve as a potential therapeutic target in synergy with ICBs in lung cancer immunotherapy.
Collapse
Affiliation(s)
- Dantong Sun
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Lipin Tan
- Department of nursing administration, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yongbing Chen
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Qiang Yuan
- Department of interventional medicine, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Kanqiu Jiang
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yangyang Liu
- Department of Vascular Surgery, Hospital of Zhangjiagang, Suzhou, 215600, China
| | - Yuhang Xue
- Department of Thoracic Surgery, Hospital of Yancheng, Yancheng, 224000, China
| | - Jinzhi Zhang
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Xianbao Cao
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Minzhao Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yang Luo
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Zhonghua Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Zhonghen Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Weihua Xu
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Mingjing Shen
- Department of Thoracic and Cardiac Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China.
| |
Collapse
|
43
|
Ma S, Li F, Wang L. The Construction of a Nomogram Using the Pan-Immune-Inflammation Value Combined with a PILE Score for Immunotherapy Prediction Prognosis in Advanced NSCLC. Cancer Manag Res 2024; 16:741-751. [PMID: 38974092 PMCID: PMC11227331 DOI: 10.2147/cmar.s461964] [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: 01/30/2024] [Accepted: 06/18/2024] [Indexed: 07/09/2024] Open
Abstract
Purpose The purpose of this study was to investigate the predictive value of Pan-Immune-Inflammation Value (PIV) combined with the PILE score for immunotherapy in patients with advanced non-small cell lung cancer (NSCLC) and to construct a nomogram prediction model to provide reference for clinical work. Patients and Methods Patients with advanced NSCLC who received ICIs treatment in Qingdao Municipal Hospital from January 2019 to December 2021 were selected as the study subjects. The chi-square test, Kaplan-Meier survival analysis, and Cox proportional risk regression analysis were used to evaluate the prognosis. The results were visualized by a nomogram, and the performance of the model was judged by indicators such as the area under the subject operating characteristic curve (AUC) and C-index. The patients were divided into high- and low-risk groups by PILE score, and the prognosis of patients in different risk groups was evaluated. Results Multivariate Cox regression analysis showed that immune-related adverse events (irAEs) were prognostic factors for overall survival (OS) improvement, and ECOG PS score ≥2, bone metastases before treatment, and high PIV expression were independent risk factors for OS. The C index of OS predicted by the nomogram model is 0.750 (95% CI: 0.677-0.823), and the Calibration and ROC curves show that the model has good prediction performance. Compared with the low-risk group, patients in the high-risk group of PILE were associated with a higher inflammatory state and poorer physical condition, which often resulted in a poorer prognosis. Conclusion PIV can be used as a prognostic indicator for patients with advanced NSCLC treated with ICIs, and a nomogram prediction model can be constructed to evaluate the survival prediction of patients, thus contributing to better clinical decision-making and prognosis assessment.
Collapse
Affiliation(s)
- Shixin Ma
- Graduate School, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
- Department of Thoracic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266071, People’s Republic of China
| | - Fei Li
- Department of Thoracic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266071, People’s Republic of China
| | - Lunqing Wang
- Department of Thoracic Surgery, Qingdao Municipal Hospital, Qingdao, Shandong, 266071, People’s Republic of China
| |
Collapse
|
44
|
Zhang C, Shao J, Tang X, Wu J, Li P, Li W, Wang C. The real-world treatment characteristic and efficacy of immune checkpoint inhibitors in non-small cell lung cancer: Data from a retrospective cohort study. Int Immunopharmacol 2024; 134:112152. [PMID: 38761777 DOI: 10.1016/j.intimp.2024.112152] [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: 12/24/2023] [Revised: 04/21/2024] [Accepted: 04/22/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND The efficacy and prognosis of immune checkpoint inhibitors (ICIs) remain unresolved issues. Here, we assessed the treatment characteristics and efficacy of ICIs in non-small cell lung cancer (NSCLC) using real-world data and evaluated the predictive value of factors, including programmed death-ligand 1 (PD-L1) expression, for the clinical outcome of ICIs in NSCLC. METHODS Analyzed data was collected from hospitalized patients in the West China Hospital of Sichuan University between January 2017 and March 2023. The Kaplan-Meier method was utilized for analyzing real-world progression-free survival (rwPFS), while Cox regression models was employed to access the correlation between the efficacy of immunotherapy and sociodemographic characteristics, disease information, and characteristics of ICI treatment. RESULTS A total of 545 patients were included in the retrospective study and characteristics of immunotherapy varied significantly among PD-L1 expression groups. The median rwPFS for the entire population was 9.76 months. Subgroup analyses revealed that patients with high PD-L1 expression, early TNM stage, first-line immunotherapy, EGFR wild-type and those who have not received radiotherapy and targeted therapy previously were more likely to have better rwPFS. Furthermore, multivariate Cox regression analyses identified PD-L1 expression, EGFR mutation status and previous radiotherapy as the most influential predictors of the response to ICI treatment. CONCLUSIONS This study presents the real-world experience of Chinese NSCLC patients undergoing ICI treatment, offering guidance for clinical decision-making based on various patient conditions, preferences, and indications for ICIs, through the evaluation of immunotherapy efficacy and predictors in NSCLC patients.
Collapse
Affiliation(s)
- Chenyang Zhang
- Institute of Hospital Management, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Jun Shao
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Targeted Tracer Research and Development Laboratory, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Xiaolong Tang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiayang Wu
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Targeted Tracer Research and Development Laboratory, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Peiyi Li
- Department of Anesthesiology, Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Centre of Translational Medicine of Anesthesiology, The Research Units of West China (2018RU012)-Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu, China.
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Targeted Tracer Research and Development Laboratory, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China.
| | - Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, Targeted Tracer Research and Development Laboratory, Med-X Center for Manufacturing, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China.
| |
Collapse
|
45
|
Iyer K, Ivanov J, Tenchov R, Ralhan K, Rodriguez Y, Sasso JM, Scott S, Zhou QA. Emerging Targets and Therapeutics in Immuno-Oncology: Insights from Landscape Analysis. J Med Chem 2024; 67:8519-8544. [PMID: 38787632 PMCID: PMC11181335 DOI: 10.1021/acs.jmedchem.4c00568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/03/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Abstract
In the ever-evolving landscape of cancer research, immuno-oncology stands as a beacon of hope, offering novel avenues for treatment. This study capitalizes on the vast repository of immuno-oncology-related scientific documents within the CAS Content Collection, totaling over 350,000, encompassing journals and patents. Through a pioneering approach melding natural language processing with the CAS indexing system, we unveil over 300 emerging concepts, depicted in a comprehensive "Trend Landscape Map". These concepts, spanning therapeutic targets, biomarkers, and types of cancers among others, are hierarchically organized into eight major categories. Delving deeper, our analysis furnishes detailed quantitative metrics showcasing growth trends over the past three years. Our findings not only provide valuable insights for guiding future research endeavors but also underscore the merit of tapping the vast and unparalleled breadth of existing scientific information to derive profound insights.
Collapse
Affiliation(s)
| | - Julian Ivanov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Rumiana Tenchov
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | | - Yacidzohara Rodriguez
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Janet M. Sasso
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | - Sabina Scott
- CAS,
A Division of the American Chemical Society, Columbus, Ohio 43210, United States
| | | |
Collapse
|
46
|
Brägelmann J, Becker S, Stenzel D, Budahn A. [Unexplained reduction in the general condition of a female lung cancer patient]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2024; 65:612-616. [PMID: 38240815 PMCID: PMC11136821 DOI: 10.1007/s00108-023-01652-5] [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] [Accepted: 12/14/2023] [Indexed: 05/31/2024]
Abstract
A 63-year-old female patient with lung cancer presented to our emergency room for the first time with a sudden reduction in general condition, vomiting and severe weakness. She stated that she was receiving chemotherapy for the lung cancer and reported that she had no other relevant previous illnesses. Our initial suspected diagnosis was cytostatic-induced nausea and vomiting. Contrary to this suspected diagnosis, diagnostics carried out in the emergency room revealed the findings of ketoacidosis on the basis of an initial manifestation of diabetes mellitus with hyperglycemic decompensation as well as severe, manifest hypothyroidism. After obtaining the preliminary findings, it became evident that the patient was not receiving chemotherapy, but rather immune checkpoint therapy using durvalumab. The initial manifestations described were therefore to be viewed as immune reactions associated with durvalumab. After initiating diabetic recompensation therapy and substitution with L‑thyroxine, a rapid improvement in the patient's general condition was achieved.
Collapse
Affiliation(s)
- J Brägelmann
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsmedizin Essen, Hufelandstraße 55, 45147, Essen, Deutschland.
| | - S Becker
- Marienhaus Klinikum Mainz, Mainz, Deutschland
| | - D Stenzel
- Marienhaus Klinikum Mainz, Mainz, Deutschland
| | - A Budahn
- Marienhaus Klinikum Mainz, Mainz, Deutschland
| |
Collapse
|
47
|
Fang Z, Wu X, Xiao L, Wang C, Zhao Y, Zhang Q, Jablonska PA, La Rosa A, Dempke WCM, Furqan M, Fan H. Somatic KMT2D loss-of-function mutations in lung squamous cell carcinoma: a single-center cohort study. J Thorac Dis 2024; 16:3338-3349. [PMID: 38883659 PMCID: PMC11170359 DOI: 10.21037/jtd-24-134] [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: 01/23/2024] [Accepted: 05/08/2024] [Indexed: 06/18/2024]
Abstract
Background The significant progress has been made in targeted therapy for lung adenocarcinoma (LUAD) in the past decade. Only few targeted therapeutics have yet been approved for the treatment of lung squamous cell carcinoma (LUSC). Several higher frequency of gene alterations are identified as potentially actionable in LUSC. Our work aimed to explore the complex interplay of multiple genetic alterations and pathways contributing to the pathogenesis of LUSC, with a very low frequency of a single driver molecular alterations to develop more effective therapeutic strategies in the future. Methods We retrospectively analyzed the targeted next-generation sequencing (NGS) data (approximately 600 genes) of 335 patients initially diagnosed with non-small cell lung cancer (NSCLC) at our institution between January 2019 and March 2023 and explored the somatic genome alteration difference between LUSC and LUAD. Results We analyzed that the presence of loss-of-function (LoF) mutations (nonsense, frameshift, and splice-site variants) in histone-lysine N-methyltransferase 2D (KMT2D) was much more prevalent in LUSC (11/53, 20.8%) than in LUAD (6/282, 2.1%). Moreover, our data indicated TP53 co-mutated with KMT2D LoF in 90.9% (10/11) LUSC and 33.3% (2/6) LUAD. Notably, the mutation allele fraction (MAF) of KMT2D was very similar to that of TP53 in the co-mutated cases. Genomic profiling of driver gene mutations of NSCLC showed that 81.8% (9/11) of the patients with LUSC with KMT2D LoF mutations had PIK3CA amplification and/or FGFR1 amplification. Conclusions Our results prompted that somatic LoF mutations of KMT2D occur frequently in LUSC, but are less frequent in LUAD and therefore may potentially contribute to the pathogenesis of LUSC. Concurrent TP53 mutations, FGFR1 amplification, and PIK3CA amplification are very common in LUSC cases with KMT2D LoF mutations. It needs more deeper investigation on the interplay of the genes and pathways and uses larger cohorts in the future.
Collapse
Affiliation(s)
- Zekui Fang
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiping Wu
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Li Xiao
- Department of Organ Transplantation, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chunli Wang
- Mygene Diagnostics Co., Ltd., Guangzhou, China
- Guangdong Engineering Technology Research Center of Multiplex PCR & Tumor Diagnostics, Guangzhou, China
| | - Yanyan Zhao
- Mygene Diagnostics Co., Ltd., Guangzhou, China
- Guangdong Engineering Technology Research Center of Multiplex PCR & Tumor Diagnostics, Guangzhou, China
| | - Qingchao Zhang
- Mygene Diagnostics Co., Ltd., Guangzhou, China
- Guangdong Engineering Technology Research Center of Multiplex PCR & Tumor Diagnostics, Guangzhou, China
| | - Paola Anna Jablonska
- Radiation Oncology Department, Hospital Universitario de Navarra, Pamplona, Spain
| | - Alonso La Rosa
- Department of Radiation Oncology, Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
| | - Wolfram C M Dempke
- Department of Haematology and Oncology, University of Munich, Munich, Germany
| | - Muhammad Furqan
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, Iowa, USA
| | - Huizhen Fan
- Department of Pulmonary and Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
48
|
Li F, Tian C, Wang Y, Wu H, Jin M, Du X, Yan J, Yang X, Yu H. Prognostic significance of peripheral and tumor-infiltrating lymphocytes in newly diagnosed stage III/IV non-small-cell lung cancer. Front Med (Lausanne) 2024; 11:1349178. [PMID: 38841570 PMCID: PMC11150824 DOI: 10.3389/fmed.2024.1349178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 05/01/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND AND AIM Lymphocytes are effector cells that fight cancer by killing tumor cells. Here, we aim to explore the prognostic significance of both peripheral and tumor-infiltrating lymphocytes (TILs) in newly diagnosed stage III/IV non-small-cell lung cancer (NSCLC). MATERIALS AND METHODS In total, 105 cases of newly diagnosed stage III/IV NSCLC from July 2017 to October 2022 at the Tianjin Beichen Hospital were retrospectively investigated. Peripheral blood samples at the time of diagnosis and tumor tissue slices from these patients were collected. General peripheral blood cell composition and TILs were measured and analyzed via an automatic blood analyzer and immunofluorescence staining analysis. The overall survival (OS) time of all patients was also obtained and analyzed. RESULTS The median overall survival (mOS) of all patients is 12 months. The 1-, 2-, and 3-year overall survival rates were 60.5, 28.4, and 18.6%, respectively. Peripheral lymphocyte and neutrophil percentages, serum C-reactive protein (CRP) expression, tumor size, and tumor pathology are the prognostic factors of OS for newly diagnosed stage III/IV NSCLC patients. Moreover, patients with high tumor CD4+ and CD8+ T cell infiltration survived significantly longer compared to patients with low tumor CD4+ and CD8+ T cell infiltration (p < 0.0001 and p = 0.011, respectively). Compared to low tumor CD33+ cell infiltration, high tumor CD33+ cell infiltration was associated with worse OS (p = 0.018). High tumor CD8+ T cell infiltration was associated with lower peripheral lymphocyte number, lower serum CRP expression, smaller tumor size, and better tumor pathology (p = 0.012, p = 0.040, p = 0.012, and p = 0.029, respectively). CONCLUSION Increased numbers of peripheral lymphocytes, CD33+ cells, CD4+ TILs, and CD8+ TILs were significantly associated with OS in newly diagnosed stage III/IV NSCLC patients, which were positively associated with several basic clinical factors.
Collapse
Affiliation(s)
- Fenge Li
- Department of Oncology, Tianjin Beichen Hospital, Tianjin, China
- Core Laboratory, Tianjin Beichen Hospital, Tianjin, China
| | - Chong Tian
- Department of Interventional Therapy, Tianjin Medical University Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin Medical University Institute and Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Institute and Hospital, Tianjin, China
| | - Yupeng Wang
- Department of Oncology, Tianjin Beichen Hospital, Tianjin, China
| | - Huancheng Wu
- Department of Neurosurgery, Tianjin Beichen Hospital, Tianjin, China
| | - Mengli Jin
- Core Laboratory, Tianjin Beichen Hospital, Tianjin, China
| | - Xueming Du
- Department of Oncology, Tianjin Beichen Hospital, Tianjin, China
| | - Jin Yan
- Department of Oncology, Tianjin Beichen Hospital, Tianjin, China
| | - Xueling Yang
- Department of Interventional Therapy, Tianjin Medical University Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Haipeng Yu
- Department of Interventional Therapy, Tianjin Medical University Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
49
|
Lee E, Jang JY, Yang J. Uncommon Adverse Events of Immune Checkpoint Inhibitors in Small Cell Lung Cancer: A Systematic Review of Case Reports. Cancers (Basel) 2024; 16:1896. [PMID: 38791974 PMCID: PMC11119772 DOI: 10.3390/cancers16101896] [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: 04/08/2024] [Revised: 05/03/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
BACKGROUND This study aimed to systematically review case reports documenting rare adverse events in patients with small cell lung cancer (SCLC) following the administration of immune checkpoint inhibitors (ICIs). METHODS A systematic literature review was conducted to identify case reports detailing previously unreported adverse drug reactions to ICIs in patients with SCLC. The scope of the literature reviewed was restricted to case studies on SCLC published up to 31 December 2023. RESULTS We analyzed twenty-four studies on ICI use for patients with SCLC. There were six reports on atezolizumab, four on durvalumab, and three on adverse events from monotherapy with nivolumab. Reports involving combination treatments were the most frequent, with a total of six, predominantly involving using nivolumab in combination with ipilimumab. Additionally, there was one report each on using pembrolizumab, nofazinilimab, sintilimab, tislelizumab, and toripalimab. We collected detailed information on the clinical course, including patient and disease characteristics, symptoms, treatment for each adverse event, and recovery status. Among the patients included in the case reports, 21 out of 24 (87.5%) had extensive-stage SCLC when initiating ICI therapy, with only 1 patient diagnosed with limited-stage SCLC. Respiratory system adverse events were most common, with seven cases, followed by neurological, endocrinological, and gastroenterological events. Three case reports documented adverse events across multiple systems in a single patient. In most cases, patients showed symptom improvement; however, four studies reported cases where patients either expired without symptom improvement or experienced sequelae. CONCLUSIONS Efforts to develop reliable biomarkers for predicting irAEs continue, with ongoing research to enhance predictive precision. Immunotherapy presents diverse and unpredictable adverse events, underscoring the need for advanced diagnostic tools and a multidisciplinary approach to improve patient management.
Collapse
Affiliation(s)
- Eunso Lee
- Division of Allergy and Pulmonology, Department of Internal Medicine, Chungnam National University Sejong Hospital, Sejong 30099, Republic of Korea
| | - Jeong Yun Jang
- Department of Radiation Oncology, Konkuk University Medical Center, Konkuk University School of Medicine, 120-1, Neungdong-ro, Gwangjin-gu, Seoul 05030, Republic of Korea
| | - Jinho Yang
- Department of Occupational Health and Safety, Semyung University, 65 Semyung-ro, Jecheon 27136, Republic of Korea
| |
Collapse
|
50
|
Weishan H, Donglin Z, Guangmei D, Wenya L, Fasheng W, Jibing C. Immunoradiotherapy for NSCLC: mechanisms, clinical outcomes, and future directions. Clin Transl Oncol 2024; 26:1063-1076. [PMID: 37921958 PMCID: PMC11026276 DOI: 10.1007/s12094-023-03337-9] [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: 09/04/2023] [Accepted: 10/10/2023] [Indexed: 11/05/2023]
Abstract
Non-small-cell lung cancer (NSCLC) has an extremely low 5-year survival rate, with the only effective treatment being immunoradiotherapy (iRT). Here, we review the progress of clinical research on iRT for non-small-cell lung cancer (NSCLC) over 2018-2023, as well as the future directions. We first discuss the synergistic mechanisms of iRT, reflected in three aspects: immune regulation of RT, RT-activated immune-related pathways, and RT-related immune sensitization. iRT may include either external-beam or stereotactic-body RT combined with either immune checkpoint inhibitors (e.g., immunoglobulins against immune programmed cell death (PD) 1/PD ligand 1 or CD8+ T lymphocyte antigen 4) or traditional Chinese medicine drugs. Regarding clinical effectiveness and safety, iRT increases overall and progression-free survival and tumor control rate among patients with NSCLC but without a considerable increase in toxicity risk. We finally discuss iRT challenges and future directions reported over 2018-2023.
Collapse
Affiliation(s)
- He Weishan
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zheng Donglin
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Deng Guangmei
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Liu Wenya
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wu Fasheng
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| | - Chen Jibing
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| |
Collapse
|