1
|
Wu Z, Shan Q, Jiang Y, Huang W, Wang Z, Zhuang Y, Liu J, Li T, Yang Z, Li C, Wei T, Wen C, Cui W, Qiu Z, Liu X, Wang Z. Irreversible electroporation combined with PD-L1/IL-6 dual blockade promotes anti-tumor immunity via cDC2/CD4 +T cell axis in MHC-I deficient pancreatic cancer. Cancer Lett 2025; 617:217620. [PMID: 40068706 DOI: 10.1016/j.canlet.2025.217620] [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: 12/13/2024] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/15/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a "cold" solid tumor with frequent Major Histocompatibility Complex I (MHC-I) deficiency, thereby making it resistant to type-1-conventional dendritic cell (cDC1)-CD8+T cell mediated anti-tumor immunity. Current studies have demonstrated the emerging compensatory role of MHC-II-mediated antigen presentation and CD4+T cell activation in anti-tumor immunity against MHC-I-deficient tumors. However, the underlying mechanism of the compensatory immune response by CD4+T cells in cancer ablation therapy remains to be elucidate. In clinical samples and murine models, we observed that irreversible electroporation (IRE) ablation therapy promoted immune infiltration and the conversion of CD4+T cells into anti-tumor IFN-γ+Th1 cells and Th17 cells in MHC-I low-expressed PDAC using scRNA-seq and flow-cytometry analyses. Furthermore, we found that PD-L1 blockade predominantly enhanced the activation of CD11b+CD103-type-2 conventional dendritic cells (cDC2s) and their antigen presentation to CD4+T cells after ablation, stimulating the anti-tumor immune response through the tumor antigen-specific IFN-γ+Th1-NK cell axis. Elevated plasma levels of IL-6 in pancreatic cancer patients receiving ablation therapy are significant indicators for impaired prognosis. IL-6 and PD-L1 dual blockade could significantly augment the ratio of IFN-γ+Th1 in CD4+T cells to boost the anti-tumor immunity of NK cells, leading to prolonged survival of mouse bearing pancreatic cancer. Collectively, we have elucidated that PD-L1 blockade activates the cDC2-CD4+T cell axis after IRE therapy, thereby playing a pivotal compensatory anti-tumor role in MHC-I low-expressed pancreatic cancer. Moreover, a combination strategy involving dual-target blockade of PD-L1/IL-6 along with ablation therapy could emerge as a novel therapeutic approach for MHC-I deficient tumors.
Collapse
Affiliation(s)
- Zhuozhuo Wu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Qungang Shan
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yuyue Jiang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Wei Huang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Ziyin Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Yaping Zhuang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jingjing Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Tiankuan Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Ziyu Yang
- Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Chaojie Li
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Department of Radiology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.149, South Chongqing Road, Shanghai, 200025, China
| | - Tao Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, China
| | - Chenlei Wen
- Department of Pancreatic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Zilong Qiu
- Songjiang Research Institute, Institute of Autism & MOE-Shanghai Key Laboratory for Children's Environmental Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China, No.748, Middle Zhongshan Road, Shanghai, 200025, China
| | - Xiaoyu Liu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China
| | - Zhongmin Wang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Faculty of Medical Imaging Technology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, No.197, Ruijin 2nd Road, Shanghai, 200025, China; Department of Radiology, Ruijin Hospital Luwan Branch, Shanghai Jiao Tong University School of Medicine, No.149, South Chongqing Road, Shanghai, 200025, China
| |
Collapse
|
2
|
Sui Y, Shen Z, Pan R, Ma R, Si R, Feng J, Zhou G. AHSA1-HSP90AA1 complex stabilized IFI6 and TGFB1 promotes mitochondrial stability and EMT in EGFR-mutated lung adenocarcinoma under Osimertinib pressure. Cell Death Dis 2025; 16:298. [PMID: 40234395 PMCID: PMC12000569 DOI: 10.1038/s41419-025-07650-9] [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: 08/12/2024] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 04/17/2025]
Abstract
Tyrosine kinase inhibitors (TKIs) have substantially improved the management of lung adenocarcinoma harboring epidermal growth factor receptor (EGFR) mutations, however, not all patients can derive benefit from it. We found that the overexpression of IFI6 under the influence of the AHSA1-HSP90AA1 complex significantly enhances Osimertinib resistance in EGFR-mutated lung adenocarcinoma cells. This effect is achieved by stabilizing mitochondrial function, reducing apoptosis, and promoting cell survival pathways via increased Akt phosphorylation. Additionally, we revealed that TGFB1 further promotes epithelial-mesenchymal transition (EMT) and enhances the invasive and migratory capabilities of these cells, thereby intensifying resistance. Regarding mechanisms, the AHSA1-HSP90AA1 complex stabilizes IFI6 and TGFB1 to enhance cell survival and Osimertinib resistance in EGFR mutant lung adenocarcinoma. IFI6 not only aids in cellular survival under drug stress but also promotes aggressive tumor phenotypes, suggesting its viability as a novel biomarker and therapeutic target for overcoming primary TKI resistance.
Collapse
Affiliation(s)
- Ying Sui
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Ziyang Shen
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Rongtian Pan
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Rong Ma
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Rujia Si
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China
| | - Jifeng Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China.
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China.
| | - Guoren Zhou
- The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, Jiangsu, China.
- Jiangsu Cancer Hospital, and Jiangsu Institute of Cancer Research, Nanjing, Jiangsu, China.
| |
Collapse
|
3
|
Chang K, Zhu LF, Wu TT, Zhang SQ, Yu ZC. Network Pharmacology and in vitro Experimental Verification on Intervention of Oridonin on Non-Small Cell Lung Cancer. Chin J Integr Med 2025; 31:347-356. [PMID: 39331210 DOI: 10.1007/s11655-024-4116-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2024] [Indexed: 09/28/2024]
Abstract
OBJECTIVE To explore the key target molecules and potential mechanisms of oridonin against non-small cell lung cancer (NSCLC). METHODS The target molecules of oridonin were retrieved from SEA, STITCH, SuperPred and TargetPred databases; target genes associated with the treatment of NSCLC were retrieved from GeneCards, DisGeNET and TTD databases. Then, the overlapping target molecules between the drug and the disease were identified. The protein-protein interaction (PPI) was constructed using the STRING database according to overlapping targets, and Cytoscape was used to screen for key targets. Molecular docking verification were performed using AutoDockTools and PyMOL software. Using the DAVID database, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis were conducted. The impact of oridonin on the proliferation and apoptosis of NSCLC cells was assessed using cell counting kit-8, cell proliferation EdU image kit, and Annexin V-FITC/PI apoptosis kit respectively. Moreover, real-time quantitative PCR and Western blot were used to verify the potential mechanisms. RESULTS Fifty-six target molecules and 12 key target molecules of oridonin involved in NSCLC treatment were identified, including tumor protein 53 (TP53), Caspase-3, signal transducer and activator of transcription 3 (STAT3), mitogen-activated protein kinase kinase 8 (MAPK8), and mammalian target of rapamycin (mTOR). Molecular docking showed that oridonin and its key target molecules bind spontaneously. GO and KEGG enrichment analyses revealed cancer, apoptosis, phosphoinositide-3 kinase/protein kinase B (PI3K/Akt), and other signaling pathways. In vitro experiments showed that oridonin inhibited the proliferation, induced apoptosis, downregulated the expression of Bcl-2 and Akt, and upregulated the expression of Caspase-3. CONCLUSION Oridonin can act on multiple targets and pathways to exert its inhibitory effects on NSCLC, and its mechanism may be related to upregulating the expression of Caspase-3 and downregulating the expressions of Akt and Bcl-2.
Collapse
Affiliation(s)
- Ke Chang
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
- Department of Pharmacy, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200065, China
| | - Li-Fei Zhu
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
| | - Ting-Ting Wu
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
| | - Si-Qi Zhang
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China
| | - Zi-Cheng Yu
- Department of Pharmacy, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, 200090, China.
| |
Collapse
|
4
|
Yokota H, Sato K, Sakamoto S, Okuda Y, Takeda M, Akamine Y, Nakayama K, Miura M. Influence of interleukin-6 on the pharmacokinetics and pharmacodynamics of osimertinib in patients with non-small cell lung cancer. Cancer Chemother Pharmacol 2025; 95:49. [PMID: 40156608 PMCID: PMC11954710 DOI: 10.1007/s00280-025-04772-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE The inflammatory cytokine interleukin (IL)-6 reduces the activity of drug metabolic enzymes and promotes tumor progression. We investigated the effect of IL-6 on the pharmacokinetics of osimertinib and the association between an IL-6 polymorphism and clinical outcomes in 30 patients with non-small cell lung cancer (NSCLC). METHODS Osimertinib and IL-6 plasma concentrations were measured on day 15 after therapy initiation. The genotype of IL-6 1800796G > C was identified using polymerase chain reaction-restriction fragment length polymorphism. Risk factors affecting overall survival (OS) were assessed by Cox proportional hazard regression analysis. RESULTS The IL-6 concentration was significantly correlated with the osimertinib trough plasma concentration (r = 0.423, P = 0.020) and area under the plasma concentration-time curve (r = 0.420, P = 0.021). The IL-6 concentration was significantly higher in patients with the IL-6 rs1800796G allele versus C/C genotype (P = 0.024). OS was significantly shorter in patients with the IL-6 rs1800796G allele versus C/C genotype (median: 15.1 vs. 48.9 months, P = 0.005). Univariate and multivariate analyses indicated that the IL-6 rs1800796G allele is an independent risk factor for OS (crude hazard ratio = 7.07; P = 0.014; adjusted hazard ratio = 6.38; P = 0.021). CONCLUSION A higher IL-6 concentration was associated with reduced metabolic activity of osimertinib, leading to increased osimertinib exposure. As the IL-6 concentration was higher in NSCLC patients with the IL-6 rs1800796G allele, it might be an independent prognostic factor for patients treated with osimertinib.
Collapse
Affiliation(s)
- Hayato Yokota
- Department of Pharmacy, Akita University Hospital, Akita, Japan
| | - Kazuhiro Sato
- Department of Internal Medicine Division of Respiratory Medicine, Akita University School of Medicine, Akita, Japan
| | - Sho Sakamoto
- Department of Internal Medicine Division of Respiratory Medicine, Akita University School of Medicine, Akita, Japan
| | - Yuji Okuda
- Department of Internal Medicine Division of Respiratory Medicine, Akita University School of Medicine, Akita, Japan
| | - Masahide Takeda
- Department of Internal Medicine Division of Respiratory Medicine, Akita University School of Medicine, Akita, Japan
| | - Yumiko Akamine
- Department of Pharmacy, Akita University Hospital, Akita, Japan
| | - Katsutoshi Nakayama
- Department of Internal Medicine Division of Respiratory Medicine, Akita University School of Medicine, Akita, Japan
| | - Masatomo Miura
- Department of Pharmacy, Akita University Hospital, Akita, Japan.
- Department of Pharmacokinetics, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan.
| |
Collapse
|
5
|
Meng Z, Li J, Wang H, Cao Z, Lu W, Niu X, Yang Y, Li Z, Wang Y, Lu S. NLRP4 unlocks an NK/macrophages-centered ecosystem to suppress non-small cell lung cancer. Biomark Res 2025; 13:44. [PMID: 40087771 PMCID: PMC11909883 DOI: 10.1186/s40364-025-00756-4] [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: 10/04/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Tumor immune evasion extends beyond T cells, affecting innate immune elements like natural killer cells (NK) and macrophages within the tumor-immune microenvironment (TIME). Nevertheless, translational strategies to trigger collaboration of NK cells and macrophages to initiate sufficient anti-tumor cytoxicity remain scarce and are urgently needed. METHODS In this study, TCGA datasets was used to confirm the prognosis value of the expression level of NLR family pyrin domain containing 4 (NLRP4) in NSCLC and the tumor tissues microarray was used to further check its clinical-relevance at protein-level. Subsequently, a tumor cell line with stable NLRP4 overexpression was established and subcutaneous tumor models in C57BL/6J mice were used to validate the anti-tumor characteristics of NLRP4. After analyzing the tumor microenvironment using flow cytometry and multiplex immunofluorescence, we further validated our findings through co-culture transwell assays and TCGA analysis. Utilizing bulk-RNA sequencing, proteomics, and mass spectrometry of mouse tumor tissues, we innovatively identified the downstream pathways of NLRP4 and verified them through co-immunoprecipitation (co-IP) and Western blot (WB) experiments. RESULTS NLRP4 could trigger a distinct anti-tumor ecosystem organized by TIGIT+TNFA+ NK and iNOS+ M1 in lung cancer, discovered in TCGA analysis and verified in murine model. NLRP4-eco exerted tumor-suppression capacity through chemokine reprogramming including CCL5 and CXCL2. Meanwhile, the cytoxicity of NK could be facilitated by iNOS+M1. Mechanistically, NLRP4 stimulated PI3K/Akt-NF-kB axis through suppression of the activity of PP2A. Besides, knockdown of CCL5 and blockade of CXCL2-CXCR2 axis abolished chemotaxis of TIGIT+TNFA+ NK and iNOS+ M1 respectively, as well as for LB-100, a PP2A inhibitor. CONCLUSION Altogether, we delineated NLRP4's unexplored facets and discovered an NLRP4-driven anti-tumor ecosystem composed of TIGIT+TNFA+ NK and iNOS+ M1. Finally, targeting PP2A by its inhibitor successfully mimicked the anti-tumor capacity of the overexpression of NLRP4.
Collapse
Affiliation(s)
- Zhouwenli Meng
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Jian Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Hui Wang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Zhengqi Cao
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Wenqing Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Xiaomin Niu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Yi Yang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
| | - Ying Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China.
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China.
| |
Collapse
|
6
|
Lim JU, Jung J, Kim YW, Kim CY, Lee SH, Park DW, Choi SI, Ji W, Yeo CD, Lee SH. Targeting the Tumor Microenvironment in EGFR-Mutant Lung Cancer: Opportunities and Challenges. Biomedicines 2025; 13:470. [PMID: 40002883 PMCID: PMC11852785 DOI: 10.3390/biomedicines13020470] [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: 01/10/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have transformed the treatment of epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer. However, treatment resistance remains a major challenge in clinical practice. The tumor microenvironment (TME) is a complex system composed of tumor cells, immune and non-immune cells, and non-cellular components. Evidence indicates that dynamic changes in TME during TKI treatment are associated with the development of resistance. Research has focused on identifying how each component of the TME interacts with tumors and TKIs to understand therapeutic targets that could address TKI resistance. In this review, we describe how TME components, such as immune cells, fibroblasts, blood vessels, immune checkpoint proteins, and cytokines, interact with EGFR-mutant tumors and how they can promote resistance to TKIs. Furthermore, we discuss potential strategies targeting TME as a novel therapeutic approach.
Collapse
Affiliation(s)
- Jeong Uk Lim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yeon Wook Kim
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Chi Young Kim
- Division of Pulmonology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Sang Hoon Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Institute of Chest Diseases, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Won Park
- Division of Pulmonary Medicine and Allergy, Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, Republic of Korea;
| | - Sue In Choi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Wonjun Ji
- Division of Pulmonology and Critical Care Medicine, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 44610, Republic of Korea
| | - Chang Dong Yeo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 03083, Republic of Korea
| | - Seung Hyeun Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Precision Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
7
|
Wu L, Lin L, Yu M, Li H, Dang Y, Liang H, Chen G, Muhetaer H, Zheng G, Li J, Jia X, Wu B, Li C. Antitumor Activity of USP7 Inhibitor GNE-6776 in Non-Small Cell Lung Cancer Involves Regulation of Epithelial-Mesenchymal Transition, Cell Cycle, Wnt/β-Catenin, and PI3K/AKT/mTOR Pathways. Pharmaceuticals (Basel) 2025; 18:245. [PMID: 40006058 PMCID: PMC11858873 DOI: 10.3390/ph18020245] [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: 01/25/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Objective: Non-small cell lung cancer (NSCLC) is a major cause of cancer-related deaths worldwide. This study investigated the effects and mechanisms of the USP7 inhibitor GNE-6776 on human NSCLC A549 and H1299 cells, providing insights for anti-NSCLC drug development. Methods: USP7 expression was analyzed in lung cancer tissue using data from public databases. RNA sequencing and functional enrichment analyses were conducted to explore differentially expressed genes (DEGs) and potentially related pathways. A549 and H1299 cells were treated with GNE-6776 at different concentrations, and its effects on cell proliferation, migration, invasion, apoptosis, mitochondrial membrane potential, and cell cycle were evaluated. Changes in protein expression following GNE-6776 treatment were assessed by Western blot. A xenograft tumor model in nude mice was used to evaluate the in vivo effects of GNE-6776. Results: GNE-6776 inhibited the proliferation, migration, and invasion of A549 and H1299 cells, induced apoptosis, and caused cells to arrest in the G1 phase in a concentration-dependent manner. GNE-6776 decreased the mitochondrial membrane potential, suppressed epithelial-mesenchymal transition (EMT) markers, and downregulated the PI3K/AKT/mTOR and Wnt/β-catenin signaling pathways. GNE-6776 significantly inhibited tumor growth without affecting body weight, reduced expression of CDK6, C-myc, and N-cadherin, and increased GSK3β expression in tumor tissue. Conclusions: In summary, GNE-6776 demonstrated potent anti-tumor activity in NSCLC both in vitro and in vivo. GNE-6776 suppresses NSCLC cell proliferation, invasion, and migration while promoting apoptosis by inhibiting the EMT and modulating the PI3K/AKT/mTOR and Wnt/β-catenin pathways. These findings support its potential as a therapeutic agent for treating NSCLC.
Collapse
Affiliation(s)
- Lipeng Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Long Lin
- Phase I Clinical Trial Center, Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou 510440, China
| | - Meng Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Huajian Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yuanye Dang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Huosheng Liang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Guangyang Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Halimulati Muhetaer
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Guodong Zheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong SAR 999077, China
| | - Xuejing Jia
- College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China
| | - Bo Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Chuwen Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
8
|
Zhou F, Guo H, Xia Y, Le X, Tan DSW, Ramalingam SS, Zhou C. The changing treatment landscape of EGFR-mutant non-small-cell lung cancer. Nat Rev Clin Oncol 2025; 22:95-116. [PMID: 39614090 DOI: 10.1038/s41571-024-00971-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/01/2024]
Abstract
The discovery of the association between EGFR mutations and the efficacy of EGFR tyrosine-kinase inhibitors (TKIs) has revolutionized the treatment paradigm for patients with non-small-cell lung cancer (NSCLC). Currently, third-generation EGFR TKIs, which are often characterized by potent central nervous system penetrance, are the standard-of-care first-line treatment for advanced-stage EGFR-mutant NSCLC. Rational combinations of third-generation EGFR TKIs with anti-angiogenic drugs, chemotherapy, the EGFR-MET bispecific antibody amivantamab or local tumour ablation are being investigated as strategies to delay drug resistance and increase clinical benefit. Furthermore, EGFR TKIs are being evaluated in patients with early stage or locally advanced EGFR-mutant NSCLC, with the ambitious aim of achieving cancer cure. Despite the inevitable challenge of acquired resistance, emerging treatments such as new TKIs, antibody-drug conjugates, new immunotherapeutic approaches and targeted protein degraders have shown considerable promise in patients with progression of EGFR-mutant NSCLC on or after treatment with EGFR TKIs. In this Review, we describe the current first-line treatment options for EGFR-mutant NSCLC, provide an overview of the mechanisms of acquired resistance to third-generation EGFR TKIs and explore novel promising treatment strategies. We also highlight potential avenues for future research that are aimed at improving the survival outcomes of patients with this disease.
Collapse
Affiliation(s)
- Fei Zhou
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haoyue Guo
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yang Xia
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuning Le
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Duke-NUS Medical School, Singapore, Singapore
| | - Suresh S Ramalingam
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Winship Cancer Institute, Atlanta, GA, USA
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Xie YQ, Yan FN, Yu LH, Yan HW, Kong YX, Yang ZY. Mechanism of Shashen-Maidong herb pair in treating hepatocellular carcinoma using network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118954. [PMID: 39419302 DOI: 10.1016/j.jep.2024.118954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 10/14/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hepatocellular carcinoma (HCC) is among the most prevalent malignant tumors globally and represents a significant public health issue worldwide. Immune cell dysfunction is the crucial factor for the formation of immunosuppression microenvironment of HCC. Glehnia littoralis (A.Gray) F.Schmidt ex Miq. (Shashen) and Ophiopogon japonicus (Thunb.) Ker Gawl. (Maidong) are classic herb pair in traditional Chinese medicine (TCM) of nourishing Yin, and is widely applied in the treatment of HCC and possesses multiple immunomodulatory functions. However, the role of the Shashen-Maidong herb pair (SS-MD) for the management of HCC and the potential mechanisms has not been explicated. AIM OF THE STUDY The purpose of the research is to investigate the potential mechanism of the SS-MD herb pair for the management of HCC. MATERIALS AND METHODS The known components of the SS-MD herb pair were preliminarily identified using UPLC-Q-Orbitrap-MS/MS. The active ingredients of SS-MD herb pair in treating HCC were screened by constructing herb-component-target network, and the key therapeutic targets were explored by constructing a protein-protein interaction (PPI) network. The binding affinity of the key targets and components were validated through molecular docking and molecular dynamics simulations. GO biological function and KEGG pathway analyses were operated to elucidate the potential mechanisms of the SS-MD herb pair for the management of HCC. And the mechanism was verified in the tumor bearing mice model and cell co-culture experiments. RESULTS Network pharmacology prediction revealed 39 active components and 138 targets of the SS-MD herb pair for the treatment of HCC. KEGG analysis mainly focused on Notch signaling pathway and Apoptosis signaling pathway. The targets were enriched in biological functions of lymphocyte effector function and lymphocyte apoptosis. In vivo and in vitro experiments proved that the SS-MD herb pair can improve the proportion of CD8+T cells in the HCC immune microenvironment, regulate its subgroup distribution. SS-MD herb pair promoted CD8+T cells to secrete IL-2, TNF-α, IFN-γ, Granzyme B and Perforin, and inhibited apoptosis by regulating Notch signaling pathway. CONCLUSIONS This study identified the key components, targets, and signaling pathways of the SS-MD herb pair, confirm that SS-MD herb pair play an immunomodulatory role in treating HCC, provides theoretical support for the collaborative treatment of HCC with TCM.
Collapse
Affiliation(s)
- Yu-Qing Xie
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| | - Feng-Na Yan
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| | - Li-Hua Yu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| | - Hui-Wen Yan
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| | - Ya-Xian Kong
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| | - Zhi-Yun Yang
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| |
Collapse
|
10
|
Thuya WL, Cao Y, Ho PCL, Wong ALA, Wang L, Zhou J, Nicot C, Goh BC. Insights into IL-6/JAK/STAT3 signaling in the tumor microenvironment: Implications for cancer therapy. Cytokine Growth Factor Rev 2025:S1359-6101(25)00003-6. [PMID: 39893129 DOI: 10.1016/j.cytogfr.2025.01.003] [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: 01/07/2025] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
The IL-6/JAK/STAT3 signaling pathway is a key regulator of tumor progression, immune evasion, and therapy resistance in various cancers. Frequently dysregulated in malignancies, this pathway drives cancer cell growth, survival, angiogenesis, and metastasis by altering the tumor microenvironment (TME). IL-6 activates JAK kinases and STAT3 through its receptor complex, leading to the transcription of oncogenic genes and fostering an immunosuppressive TME. This environment recruits tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and regulatory T cells (Tregs), collectively supporting immune evasion and tumor growth. IL-6/JAK/STAT3 axis also contributes to metabolic reprogramming, such as enhanced glycolysis and glutathione metabolism, helping cancer cells adapt to environmental stresses. Therapeutic targeting of this pathway has gained significant interest. Strategies include monoclonal antibodies against IL-6 or its receptor (e.g., Tocilizumab, Siltuximab), JAK inhibitors (e.g., Ruxolitinib), and STAT3-specific inhibitors (e.g., Napabucasin), which have exhibited promise in preclinical and initial clinical studies. These inhibitors can suppress tumor growth, reverse immune suppression, and enhance the efficacy of immunotherapies like immune checkpoint inhibitors. Combination therapies that integrate IL-6 pathway inhibitors with conventional treatments are particularly promising, addressing resistance mechanisms and improving patient outcomes. Advances in biomarker-driven patient selection, RNA-based therapies, and isoform-specific inhibitors pave the way for more precise interventions. This review delves into the diverse roles of IL-6/JAK/STAT3 signaling in cancer progression, therapeutic strategies targeting this pathway, and the potential for integrating these approaches into personalized medicine to enhance treatment outcomes.
Collapse
Affiliation(s)
- Win Lwin Thuya
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Yang Cao
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Laboratory Medicine, Lequn Brance, The First Hospital of Jilin University, Changchun, Jilin 130031, China
| | - Paul Chi-Lui Ho
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Jalan Lagoon, Selangor Darul Ehsan 47500, Malaysia
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, USA
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore.
| | - Christophe Nicot
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, USA
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; Department of Haematology-Oncology, National University Cancer Institute, National University Health System, 119074, Singapore
| |
Collapse
|
11
|
Foffano L, Bertoli E, Bortolot M, Torresan S, De Carlo E, Stanzione B, Del Conte A, Puglisi F, Spina M, Bearz A. Immunotherapy in Oncogene-Addicted NSCLC: Evidence and Therapeutic Approaches. Int J Mol Sci 2025; 26:583. [PMID: 39859299 PMCID: PMC11765476 DOI: 10.3390/ijms26020583] [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: 12/20/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a leading cause of cancer-related mortality worldwide. The discovery of specific driver mutations has revolutionized the treatment landscape of oncogene-addicted NSCLC through targeted therapies, significantly improving patient outcomes. However, immune checkpoint inhibitors (ICIs) have demonstrated limited effectiveness in this context. Emerging evidence, though, reveals significant heterogeneity among different driver mutation subgroups, suggesting that certain patient subsets may benefit from ICIs, particularly when combined with other therapeutic modalities. In this review, we comprehensively examine the current evidence on the efficacy of immunotherapy in oncogene-addicted NSCLC. By analyzing recent clinical trials and preclinical studies, along with an overview of mechanisms that may reduce immunotherapy efficacy, we explored potential strategies to address these challenges, to provide insights that could optimize immunotherapy approaches and integrate them effectively into the treatment algorithm for oncogene-addicted NSCLC.
Collapse
Affiliation(s)
- Lorenzo Foffano
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Elisa Bertoli
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Martina Bortolot
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Sara Torresan
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Elisa De Carlo
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Brigida Stanzione
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Alessandro Del Conte
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Fabio Puglisi
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Michele Spina
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| | - Alessandra Bearz
- Department of Medical Oncology, CRO di Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy; (E.B.); (M.B.); (E.D.C.); (B.S.); (A.D.C.); (F.P.); (M.S.); (A.B.)
| |
Collapse
|
12
|
Wang J, He X, Jia Z, Yan A, Xiao K, Liu S, Hou M, Long Y, Ding X. Shenqi Fuzheng injection restores the sensitivity to gefitinib in non-small cell lung cancer by inhibiting the IL-22/STAT3/AKT pathway. PHARMACEUTICAL BIOLOGY 2024; 62:33-41. [PMID: 38100532 PMCID: PMC10732196 DOI: 10.1080/13880209.2023.2292266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
CONTEXT Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. Gefitinib is a first-line treatment for NSCLC. However, its effectiveness is hindered by the development of drug resistance. At present, Shenqi Fuzheng injection (SFI) is widely accepted as an adjuvant therapy in NSCLC. OBJECTIVE This study investigates the molecular mechanism of SFI when combined with gefitinib in regulating cell progression among EGFR-TKI-resistant NSCLC. MATERIALS AND METHODS We established gefitinib-resistant PC9-GR cells by exposing gefitinib escalation from 10 nM with the indicated concentrations of SFI in PC9 cells (1, 4, and 8 mg/mL). Quantitative real-time polymerase chain reaction was performed to assess gene expression. PC9/GR and H1975 cells were treated with 50 ng/mL of interleukin (IL)-22 alone or in combination with 10 mg/mL of SFI. STAT3, p-STAT3, AKT, and p-AKT expression were evaluated using Western blot. The effects on cell proliferation, clonogenicity, and apoptosis in NSCLC cells were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), colony formation and flow cytometry assays. RESULTS SFI treatment alleviated the development of gefitinib resistance in NSCLC. PC9/GR and H1975 cells treated with SFI significantly exhibited a reduction in IL-22 protein and mRNA overexpression levels. SFI effectively counteracted the activation of the STAT3/AKT signaling pathway induced by adding exogenous IL-22 to PC9/GR and H1975 cells. Moreover, IL-22 combined with gefitinib markedly increased cell viability while reducing apoptosis. In contrast, combining SFI with gefitinib and the concurrent treatment of SFI with gefitinib and IL-22 demonstrated the opposite effect. DISCUSSION AND CONCLUSION SFI can be a valuable therapeutic option to address gefitinib resistance in NSCLC by suppressing the IL-22/STAT3/AKT pathway.
Collapse
Affiliation(s)
- Jiali Wang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xianhai He
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhirong Jia
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Aiwen Yan
- Jiangsu Food & Pharmaceutical Science College, Jiangsu Food Science College, Huanan, China
| | - Kang Xiao
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shuo Liu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mengjun Hou
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yaling Long
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xuansheng Ding
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
- Precision Medicine Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
13
|
Dong Y, Khan L, Yao Y. Immunological features of EGFR-mutant non-small cell lung cancer and clinical practice: a narrative review. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:289-298. [PMID: 39735443 PMCID: PMC11674437 DOI: 10.1016/j.jncc.2024.06.004] [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: 12/02/2023] [Revised: 05/02/2024] [Accepted: 06/15/2024] [Indexed: 12/31/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have significantly improved outcomes for patients with advanced driver-negative non-small cell lung cancer (NSCLC). However, targeted therapy remains the preferred treatment for advanced driver-positive NSCLC, including cases with epidermal growth factor receptor (EGFR) mutations. Considering the variability in EGFR-mutant NSCLC, including expression levels of programmed cell death ligand 1 (PD-L1), tumor mutation burden (TMB), and other immunological features, the application of immunotherapy in this group is still a subject of investigation. Therefore, we have summarized and analyzed the immunological characteristics and regulatory mechanisms of different EGFR mutations in NSCLC, as well as the current clinical application of immunotherapy in the EGFR-mutant population, to provide a reference for future research.
Collapse
Affiliation(s)
- Yi Dong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Liaqat Khan
- Research Center, Benazir Bhutto Hospital of Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
14
|
Bui VNV, Daugaard TF, Sorensen BS, Nielsen AL. Expression of the non-coding RNA nc886 facilitates the development of tyrosine kinase inhibitor resistance in EGFR-mutated non-small-cell lung cancer cells. Biochem Biophys Res Commun 2024; 731:150395. [PMID: 39024976 DOI: 10.1016/j.bbrc.2024.150395] [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: 05/27/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 07/20/2024]
Abstract
Treatment of non-small-cell lung cancer (NSCLC) patients possessing EGFR-activating mutations with tyrosine kinase inhibitors (TKIs) can confer an initial promising response. However, TKI resistance inevitably arises. Numerous TKI resistance mechanisms are identified including EGFR secondary mutations, bypass receptor tyrosine kinase (RTK) signaling, and cellular transition e.g. epithelial-mesenchymal transition (EMT). To increase the knowledge of TKI resistance we performed an epigenetic screen to identify small non-coding (nc) genes with DNA methylation alterations in HCC827 NSCLC EGFR-mutated cells with acquired TKI resistance. We analyzed Infinium Methylation EPIC 850K Array data for DNA methylation changes present in both TKI-resistant HCC827 cells with EMT and MET-amplification. Hereby, we identified that the polymorphic maternal imprinted gene nc886 (vtRNA2-1) has a decrease in promoter DNA methylation in TKI-resistant cells. This epigenetic change was associated with an increase in the expression of nc886. The induction of EMT did not affect nc886 expression. CRISPR/Cas9-mediated distortion of the nc886 sequence increased the sensitivity of HCC827 cells towards TKI. Finally, nc886 sequence distortion hindered MET RTK activation and instead was EMT the endpoint TKI resistance mechanism. In conclusion, the expression of nc886 contributes to TKI resistance in the HCC827 NSCLC cell line by supporting cell survival and selection of the endpoint TKI resistance mechanism. We propose DNA methylation and expression changes for nc886 to constitute a novel TKI resistance contributing mechanism in NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Cell Line, Tumor
- DNA Methylation
- Drug Resistance, Neoplasm/genetics
- Epigenesis, Genetic/drug effects
- Epithelial-Mesenchymal Transition/genetics
- Epithelial-Mesenchymal Transition/drug effects
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Lung Neoplasms/genetics
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Mutation
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Tyrosine Kinase Inhibitors/pharmacology
- Tyrosine Kinase Inhibitors/therapeutic use
Collapse
Affiliation(s)
- Vivian N V Bui
- Department of Biomedicine, Aarhus University, 8000, Aarhus, Denmark.
| | - Tina F Daugaard
- Department of Biomedicine, Aarhus University, 8000, Aarhus, Denmark.
| | - Boe S Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, 8200, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, 8200, Aarhus, Denmark.
| | - Anders L Nielsen
- Department of Biomedicine, Aarhus University, 8000, Aarhus, Denmark.
| |
Collapse
|
15
|
Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, Luo P, Cheng Q, Xu H, Han X. Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduct Target Ther 2024; 9:270. [PMID: 39389953 PMCID: PMC11467208 DOI: 10.1038/s41392-024-01955-5] [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: 02/24/2024] [Revised: 07/18/2024] [Accepted: 08/24/2024] [Indexed: 10/12/2024] Open
Abstract
The cascade of metastasis in tumor cells, exhibiting organ-specific tendencies, may occur at numerous phases of the disease and progress under intense evolutionary pressures. Organ-specific metastasis relies on the formation of pre-metastatic niche (PMN), with diverse cell types and complex cell interactions contributing to this concept, adding a new dimension to the traditional metastasis cascade. Prior to metastatic dissemination, as orchestrators of PMN formation, primary tumor-derived extracellular vesicles prepare a fertile microenvironment for the settlement and colonization of circulating tumor cells at distant secondary sites, significantly impacting cancer progression and outcomes. Obviously, solely intervening in cancer metastatic sites passively after macrometastasis is often insufficient. Early prediction of metastasis and holistic, macro-level control represent the future directions in cancer therapy. This review emphasizes the dynamic and intricate systematic alterations that occur as cancer progresses, illustrates the immunological landscape of organ-specific PMN creation, and deepens understanding of treatment modalities pertinent to metastasis, thereby identifying some prognostic and predictive biomarkers favorable to early predict the occurrence of metastasis and design appropriate treatment combinations.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingqi Chen
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|
16
|
Fu ZY, Huang Y, Lian LS, Huang HT, Zhan SF, Cai Y, Li JX, Liu XH. Potential of semen coicis in enhancing the anti-tumor effects of PD-1 inhibitor on A549 cell lines by blocking the PI3K-AKT-mTOR pathway. Clin Transl Oncol 2024; 26:2250-2261. [PMID: 38554191 DOI: 10.1007/s12094-024-03444-1] [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: 02/01/2024] [Accepted: 03/04/2024] [Indexed: 04/01/2024]
Abstract
BACKGROUND The objective of this research was to investigate how the combination of semen coicis extract and PD-1 inhibitors can potentially work together to enhance the anti-tumor effects, with a focus on understanding the underlying mechanism. METHODS We obtained the active components and specific targets of semen coicis in the treatment of NSCLC from various databases, namely TCMSP, GeneCard, and OMIM. By utilizing the STRING database and Cytoscape software, we established a protein interaction network (PPI) for the active ingredient of semen coicis and the target genes related to NSCLC. To explore the potential pathways involved, we conducted gene ontology (GO) and biological pathway (KEGG) enrichment analyses, which were further supported by molecular docking technology. Additionally, we conducted cyto-inhibition experiments to verify the inhibitory effects of semen coicis alone or in combination with a PD-1 inhibitor on A549 cells, along with examining the associated pathways. Furthermore, we investigated the synergistic mechanism of these two drugs through cytokine release experiments and the PD-L1 expression study on A549 cells. RESULTS Semen coicis contains two main active components, Omaine and (S)-4-Nonanolide. Its primary targets include PIK3R1, PIK3CD, PIK3CA, AKT2, and mTOR. Molecular docking experiments confirmed that these ingredients and targets form stable bonds. In vitro experiments showed that semen coicis demonstrates inhibitory effects against A549 cells, and this effect was further enhanced when combined with PD-1 inhibitors. PCR and WB analysis confirmed that the inhibition of the PI3K-AKT-mTOR pathway may contribute to this effect. Additionally, semen coicis was observed to decrease the levels of IFN-γ, IL-6, and TNF-α, promoting the recovery of the human anti-tumor immune response. And semen coicis could inhibit the induced expression of PD‑L1 of A549 cells stimulated by IFN‑γ as well. CONCLUSION Semen coicis not only has the ability to kill tumor cells directly but also alleviates the immunosuppression found in the tumor microenvironment. Additionally, it collaboratively enhances the effectiveness of PD-1 inhibitors against tumors by blocking the activation of PI3K-AKT-mTOR.
Collapse
Affiliation(s)
- Zi-Yi Fu
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Ying Huang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Le-Shen Lian
- Department of Pulmonary and Critical Care Medicine, Dongguan Hospital Affiliated to Guangzhou University of Chinese Medicine, Dongguan, 523000, China
| | - Hui-Ting Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Shao-Feng Zhan
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Yan Cai
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Jun-Xiong Li
- Department of Pulmonary and Critical Care Medicine, Guangdong Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Foshan, 528000, China
| | - Xiao-Hong Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510000, China.
| |
Collapse
|
17
|
Ou X, Gao G, Habaz IA, Wang Y. Mechanisms of resistance to tyrosine kinase inhibitor-targeted therapy and overcoming strategies. MedComm (Beijing) 2024; 5:e694. [PMID: 39184861 PMCID: PMC11344283 DOI: 10.1002/mco2.694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/27/2024] Open
Abstract
Tyrosine kinase inhibitor (TKI)-targeted therapy has revolutionized cancer treatment by selectively blocking specific signaling pathways crucial for tumor growth, offering improved outcomes with fewer side effects compared with conventional chemotherapy. However, despite their initial effectiveness, resistance to TKIs remains a significant challenge in clinical practice. Understanding the mechanisms underlying TKI resistance is paramount for improving patient outcomes and developing more effective treatment strategies. In this review, we explored various mechanisms contributing to TKI resistance, including on-target mechanisms and off-target mechanisms, as well as changes in the tumor histology and tumor microenvironment (intrinsic mechanisms). Additionally, we summarized current therapeutic approaches aiming at circumventing TKI resistance, including the development of next-generation TKIs and combination therapies. We also discussed emerging strategies such as the use of dual-targeted antibodies and PROteolysis Targeting Chimeras. Furthermore, we explored future directions in TKI-targeted therapy, including the methods for detecting and monitoring drug resistance during treatment, identification of novel targets, exploration of dual-acting kinase inhibitors, application of nanotechnologies in targeted therapy, and so on. Overall, this review provides a comprehensive overview of the challenges and opportunities in TKI-targeted therapy, aiming to advance our understanding of resistance mechanisms and guide the development of more effective therapeutic approaches in cancer treatment.
Collapse
Affiliation(s)
- Xuejin Ou
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| | - Ge Gao
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
- Clinical Trial Center, National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drugs, West China HospitalSichuan UniversityChengduChina
| | - Inbar A. Habaz
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonOntarioCanada
| | - Yongsheng Wang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
18
|
Hu Q, Remsing Rix LL, Desai B, Miroshnychenko D, Li X, Welsh EA, Fang B, Wright GM, Chaudhary N, Kroeger JL, Doebele RC, Koomen JM, Haura EB, Marusyk A, Rix U. Cancer-associated fibroblasts confer ALK inhibitor resistance in EML4-ALK -driven lung cancer via concurrent integrin and MET signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609975. [PMID: 39253447 PMCID: PMC11383036 DOI: 10.1101/2024.08.27.609975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are associated with tumor progression and modulate drug sensitivity of cancer cells. However, the underlying mechanisms are often incompletely understood and crosstalk between tumor cells and CAFs involves soluble secreted as well as adhesion proteins. Interrogating a panel of non-small cell lung cancer (NSCLC) cell lines driven by EML4-ALK fusions, we observed substantial CAF-mediated drug resistance to clinical ALK tyrosine kinase inhibitors (TKIs). Array-based cytokine profiling of fibroblast-derived conditioned- media identified HGF-MET signaling as a major contributor to CAF-mediated paracrine resistance that can be overcome by MET TKIs. However, 'Cell Type specific labeling using Amino acid Precursors' (CTAP)-based expression and phosphoproteomics in direct coculture also highlighted a critical role for the fibronectin-integrin pathway. Flow cytometry analysis confirmed activation of integrin β1 (ITGB1) in lung cancer cells by CAF coculture. Treatment with pharmacological inhibitors, cancer cell-specific silencing or CRISPR-Cas9-mediated knockout of ITGB1 overcame adhesion protein-mediated resistance. Concurrent targeting of MET and integrin signaling effectively abrogated CAF-mediated resistance of EML4-ALK -driven NSCLC cells to ALK TKIs in vitro . Consistently, combination of the ALK TKI alectinib with the MET TKI capmatinib and/or the integrin inhibitor cilengitide was significantly more efficacious than single agent treatment in suppressing tumor growth using an in vivo EML4-ALK -dependent allograft mouse model of NSCLC. In summary, these findings emphasize the complexity of resistance-associated crosstalk between CAFs and cancer cells, which can involve multiple concurrent signaling pathways, and illustrate how comprehensive elucidation of paracrine and juxtacrine resistance mechanisms can inform on more effective therapeutic approaches.
Collapse
|
19
|
Li D, Shao F, Yu Q, Wu R, Tuo Z, Wang J, Ye L, Guo Y, Yoo KH, Ke M, Okoli UA, Premkamon C, Yang Y, Wei W, Heavey S, Cho WC, Feng D. The complex interplay of tumor-infiltrating cells in driving therapeutic resistance pathways. Cell Commun Signal 2024; 22:405. [PMID: 39160622 PMCID: PMC11331645 DOI: 10.1186/s12964-024-01776-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024] Open
Abstract
Drug resistance remains a significant challenge in cancer treatment. Recently, the interactions among various cell types within the tumor microenvironment (TME) have deepened our understanding of the mechanisms behind treatment resistance. Therefore, this review aims to synthesize current research focusing on infiltrating cells and drug resistance suggesting that targeting the TME could be a viable strategy to combat this issue. Numerous factors, including inflammation, metabolism, senescence, hypoxia, and angiogenesis, contribute to drug resistance could be a viable strategy to combat this issue. Overexpression of STAT3 is commonly associated with drug-resistant cancer cells or stromal cells. Current research often generalizes the impact of stromal cells on resistance, lacking specificity and statistical robustness. Thus, future research should take notice of this issue and aim to provide high-quality evidence. Despite the existing limitations, targeting the TME to overcome therapy resistance hold promising and valuable potential.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fanglin Shao
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, 315211, China
- Department of Pathology, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhouting Tuo
- Department of Urology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, Seoul, Republic of Korea
| | - Mang Ke
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, China
| | - Uzoamaka Adaobi Okoli
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK
- Basic and Translational Cancer Research Group, Department of Pharmacology and Therapeutics, College of Medicine, University of Nigeria, Eastern part of Nigeria, Nsukka, Enugu State, Nigeria
| | - Chaipanichkul Premkamon
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK
| | - Yubo Yang
- Department of Urology, Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, 404000, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Susan Heavey
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Birmingham, Hong Kong SAR, China.
| | - Dechao Feng
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| |
Collapse
|
20
|
Tan J, Zhu L, Shi J, Zhang J, Kuang J, Guo Q, Zhu X, Chen Y, Zhou C, Gao X. Evaluation of drug resistance for EGFR-TKIs in lung cancer via multicellular lung-on-a-chip. Eur J Pharm Sci 2024; 199:106805. [PMID: 38763450 DOI: 10.1016/j.ejps.2024.106805] [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: 02/19/2024] [Revised: 04/10/2024] [Accepted: 05/17/2024] [Indexed: 05/21/2024]
Abstract
Drug resistance to irreversible epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) is a primary factor affecting their therapeutic efficacy in human non-small cell lung cancer (NSCLC). NSCLC cells can undergo epithelial-mesenchymal transition (EMT) induced by many factors in the tumour microenvironment (TME), which plays a crucial role in tumour drug resistance. In this study, a multicellular lung-on-a-chip that can realise the cell co-culture of the human non-small cell lung cancer cell line HCC827, human foetal lung fibroblasts (HFL-1), and human umbilical vein endothelial cells (HUVECs) is prepared. The TME was simulated on the chip combined with perfusion and other factors, and the drug evaluation of osimertinib was performed to explore the drug resistance mechanism of EGFR-TKIs. In the early stages, a two-dimensional static cell co-culture was achieved by microchip, and the results showed that HFL-1 cells could be transformed into cancer-associated fibroblasts (CAFs), and HCC827 cells could undergo EMT, both of which were mediated by Interleukin-6 (IL-6). Vimentin (VIM) and Alpha Skeletal Muscle Actin (a-SMA) expression of HFL-1 was upregulated, whereas E-cadherin (E-cad) expression of HCC827 was down-regulated. Further, N-cadherin (N-cad) expression of HCC827 was upregulated. In both the static cell co-culture and multicellular lung-on-a-chip, HCC827 cells with CAFs co-culture or IL-6 treatment developed resistance to osimertinib. Further use of the IL-6 antibody inhibitor tocilizumab could reverse EGFR-TKI resistance to a certain extent. Combination therapy with tocilizumab and EGFR-TKIs may provide a novel therapeutic strategy for overcoming EGFR-TKI resistance caused by EMT in NSCLC. Furthermore, the lung-on-a-chip can simulate complex TME and can be used for evaluating tumour resistance and exploring mechanisms, with the potential to become an important tool for personalised diagnosis, treatment, and biomedical research.
Collapse
Affiliation(s)
- Jianfeng Tan
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510030, China
| | - Leqing Zhu
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China; Shenzhen Clinical Medical College, Southern Medical University, Shenzhen,518101, China
| | - Jingyan Shi
- Materials Genome Institute, Shanghai University, Shanghai 200444, China
| | - Jianhua Zhang
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Jun Kuang
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Quanwei Guo
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Xiaojia Zhu
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Yuliang Chen
- Department of Thoracic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, China
| | - Chengbin Zhou
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510030, China; Department of Cardiovascular Surgery, Guangdong Provincial Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510030, China.
| | - Xinghua Gao
- Materials Genome Institute, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
21
|
Huang H, Zhu X, Yu Y, Li Z, Yang Y, Xia L, Lu S. EGFR mutations induce the suppression of CD8 + T cell and anti-PD-1 resistance via ERK1/2-p90RSK-TGF-β axis in non-small cell lung cancer. J Transl Med 2024; 22:653. [PMID: 39004699 PMCID: PMC11246587 DOI: 10.1186/s12967-024-05456-5] [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: 04/16/2024] [Accepted: 07/01/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) patients with EGFR mutations exhibit an unfavorable response to immune checkpoint inhibitor (ICI) monotherapy, and their tumor microenvironment (TME) is usually immunosuppressed. TGF-β plays an important role in immunosuppression; however, the effects of TGF-β on the TME and the efficacy of anti-PD-1 immunotherapy against EGFR-mutated tumors remain unclear. METHODS Corresponding in vitro studies used the TCGA database, clinical specimens, and self-constructed mouse cell lines with EGFR mutations. We utilized C57BL/6N and humanized M-NSG mouse models bearing EGFR-mutated NSCLC to investigate the effects of TGF-β on the TME and the combined efficacy of TGF-β blockade and anti-PD-1 therapy. The changes in immune cells were monitored by flow cytometry. The correlation between TGF-β and immunotherapy outcomes of EGFR-mutated NSCLC was verified by clinical samples. RESULTS We identified that TGF-β was upregulated in EGFR-mutated NSCLC by EGFR activation and subsequent ERK1/2-p90RSK phosphorylation. TGF-β directly inhibited CD8+ T cell infiltration, proliferation, and cytotoxicity both in vitro and in vivo, but blocking TGF-β did not suppress the growth of EGFR-mutated tumors in vivo. Anti-TGF-β antibody combined with anti-PD-1 antibody significantly inhibited the proliferation of recombinant EGFR-mutated tumors in C57BL/6N mice, which was superior to their monotherapy. Mechanistically, the combination of anti-TGF-β and anti-PD-1 antibodies significantly increased the infiltration of CD8+ T cells and enhanced the anti-tumor function of CD8+ T cells. Moreover, we found that the expression of TGF-β1 in EGFR-TKI resistant cell lines was significantly higher than that in parental cell lines. The combination of anti-TGF-β and nivolumab significantly inhibited the proliferation of EGFR-TKI resistant tumors in humanized M-NSG mice and prolonged their survival. CONCLUSIONS Our results reveal that TGF-β expression is upregulated in NSCLC with EGFR mutations through the EGFR-ERK1/2-p90RSK signaling pathway. High TGF-β expression inhibits the infiltration and anti-tumor function of CD8+ T cells, contributing to the "cold" TME of EGFR-mutated tumors. Blocking TGF-β can reshape the TME and enhance the therapeutic efficacy of anti-PD-1 in EGFR-mutated tumors, which provides a potential combination immunotherapy strategy for advanced NSCLC patients with EGFR mutations.
Collapse
Affiliation(s)
- Huayan Huang
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, Shanghai, 200030, China
| | - Xiaokuan Zhu
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, Shanghai, 200030, China
| | - Yongfeng Yu
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, Shanghai, 200030, China
| | - Ziming Li
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, Shanghai, 200030, China
| | - Yi Yang
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, Shanghai, 200030, China
| | - Liliang Xia
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, Shanghai, 200030, China.
| | - Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, West Huaihai Road 241, Shanghai, 200030, China.
| |
Collapse
|
22
|
Zhong Y, Liu J. Emerging roles of CAR-NK cell therapies in tumor immunotherapy: current status and future directions. Cell Death Discov 2024; 10:318. [PMID: 38987565 PMCID: PMC11236993 DOI: 10.1038/s41420-024-02077-1] [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: 03/29/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024] Open
Abstract
Cancer immunotherapy harnesses the body's immune system to combat malignancies, building upon an understanding of tumor immunosurveillance and immune evasion mechanisms. This therapeutic approach reactivates anti-tumor immune responses and can be categorized into active, passive, and combined immunization strategies. Active immunotherapy engages the immune system to recognize and attack tumor cells by leveraging host immunity with cytokine supplementation or vaccination. Conversely, passive immunotherapy employs exogenous agents, such as monoclonal antibodies (anti-CTLA4, anti-PD1, anti-PD-L1) or adoptive cell transfers (ACT) with genetically engineered chimeric antigen receptor (CAR) T or NK cells, to exert anti-tumor effects. Over the past decades, CAR-T cell therapies have gained significant traction in oncological treatment, offering hope through their targeted approach. However, the potential adverse effects associated with CAR-T cells, including cytokine release syndrome (CRS), off-tumor toxicity, and neurotoxicity, warrant careful consideration. Recently, CAR-NK cell therapy has emerged as a promising alternative in the landscape of tumor immunotherapy, distinguished by its innate advantages over CAR-T cell modalities. In this review, we will synthesize the latest research and clinical advancements in CAR-NK cell therapies. We will elucidate the therapeutic benefits of employing CAR-NK cells in oncology and critically examine the developmental bottlenecks impeding their broader application. Our discussion aims to provide a comprehensive overview of the current status and future potential of CAR-NK cells in cancer immunotherapy.
Collapse
Affiliation(s)
- Yan Zhong
- Department of Pathology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Jingfeng Liu
- Shenzhen Key Laboratory of Immunity and Inflammatory Diseases, Peking University Shenzhen Hospital, Shenzhen, China.
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China.
| |
Collapse
|
23
|
Liu HM, Yu ZL, Xia HF, Zhang LZ, Fu QY, Wang Y, Gong HY, Chen G. EGFR Mutation and TKI Treatment Promote Secretion of Small Extracellular Vesicle PD-L1 and Contribute to Immunosuppression in NSCLC. Biomolecules 2024; 14:820. [PMID: 39062533 PMCID: PMC11274907 DOI: 10.3390/biom14070820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/21/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
In Asian populations with non-small-cell lung cancer (NSCLC), EGFR mutations are highly prevalent, occurring in roughly half of these patients. Studies have revealed that individuals with EGFR mutation typically fare worse with immunotherapy. In patients who received EGFR tyrosine kinase inhibitor (TKI) treatment followed by anti-PD-1 therapy, poor results were observed. The underlying mechanism remains unclear. We used high-resolution flow cytometry and ELISA to detect the circulating level of small extracellular vesicle (sEV) PD-L1 in NSCLC individuals with EGFR mutations before and after receiving TKIs. The secretion amount of sEV PD-L1 of lung cancer cell lines with EGFR mutations under TKI treatment or not were detected using high-resolution flow cytometry and Western blotting. The results revealed that patients harboring EGFR mutations exhibit increased levels of sEV PD-L1 in circulation, which inversely correlated with the presence of CD8+ T cells in tumor tissues. Furthermore, tumor cells carrying EGFR mutations secrete a higher quantity of PD-L1-positive sEVs. TKI treatment appeared to amplify the levels of PD-L1-positive sEVs in the bloodstream. Mutation-induced and TKI-induced sEVs substantially impaired the functionality of CD8+ T cells. Importantly, our findings indicated that EGFR mutations and TKI therapies promote secretion of PD-L1-positive sEVs via distinct molecular mechanisms, namely the HRS and ALIX pathways, respectively. In conclusion, the increased secretion of PD-L1-positive sEVs, prompted by genetic alterations and TKI administration, may contribute to the limited efficacy of immunotherapy observed in EGFR-mutant patients and patients who have received TKI treatment.
Collapse
Affiliation(s)
- Hai-Ming Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Hou-Fu Xia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lin-Zhou Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qiu-Yun Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yi Wang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hong-Yun Gong
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430071, China
| |
Collapse
|
24
|
Wu Y, Yu G, Jin K, Qian J. Advancing non-small cell lung cancer treatment: the power of combination immunotherapies. Front Immunol 2024; 15:1349502. [PMID: 39015563 PMCID: PMC11250065 DOI: 10.3389/fimmu.2024.1349502] [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: 12/04/2023] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) remains an unsolved challenge in oncology, signifying a substantial global health burden. While considerable progress has been made in recent years through the emergence of immunotherapy modalities, such as immune checkpoint inhibitors (ICIs), monotherapies often yield limited clinical outcomes. The rationale behind combining various immunotherapeutic or other anticancer agents, the mechanistic underpinnings, and the clinical evidence supporting their utilization is crucial in NSCLC therapy. Regarding the synergistic potential of combination immunotherapies, this study aims to provide insights to help the landscape of NSCLC treatment and improve clinical outcomes. In addition, this review article discusses the challenges and considerations of combination regimens, including toxicity management and patient selection.
Collapse
Affiliation(s)
- Yuanlin Wu
- Department of Thoracic Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Guangmao Yu
- Department of Thoracic Surgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Ketao Jin
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People’s Hospital, Affiliated Xinchang Hospital, Wenzhou Medical University, Xinchang, Zhejiang, China
| |
Collapse
|
25
|
Lu C, Gao Z, Wu D, Zheng J, Hu C, Huang D, He C, Liu Y, Lin C, Peng T, Dou Y, Zhang Y, Sun F, Jiang W, Yin G, Han R, He Y. Understanding the dynamics of TKI-induced changes in the tumor immune microenvironment for improved therapeutic effect. J Immunother Cancer 2024; 12:e009165. [PMID: 38908857 PMCID: PMC11328648 DOI: 10.1136/jitc-2024-009165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND The dynamic interplay between tyrosine kinase inhibitors (TKIs) and the tumor immune microenvironment (TME) plays a crucial role in the therapeutic trajectory of non-small cell lung cancer (NSCLC). Understanding the functional dynamics and resistance mechanisms of TKIs is essential for advancing the treatment of NSCLC. METHODS This study assessed the effects of short-term and long-term TKI treatments on the TME in NSCLC, particularly targeting epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) mutations. We analyzed changes in immune cell composition, cytokine profiles, and key proteins involved in immune evasion, such as laminin subunit γ-2 (LAMC2). We also explored the use of aspirin as an adjunct therapy to modulate the TME and counteract TKI resistance. RESULTS Short-term TKI treatment enhanced T cell-mediated tumor clearance, reduced immunosuppressive M2 macrophage infiltration, and downregulated LAMC2 expression. Conversely, long-term TKI treatment fostered an immunosuppressive TME, contributing to drug resistance and promoting immune escape. Differential responses were observed among various oncogenic mutations, with ALK-targeted therapies eliciting a stronger antitumor immune response compared with EGFR-targeted therapies. Notably, we found that aspirin has potential in overcoming TKI resistance by modulating the TME and enhancing T cell-mediated tumor clearance. CONCLUSIONS These findings offer new insights into the dynamics of TKI-induced changes in the TME, improving our understanding of NSCLC challenges. The study underscores the critical role of the TME in TKI resistance and suggests that adjunct therapies, like aspirin, may provide new strategies to enhance TKI efficacy and overcome resistance.
Collapse
Affiliation(s)
- Conghua Lu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Ziyuan Gao
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Di Wu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Jie Zheng
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Chen Hu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Daijuan Huang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| | - Chao He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Yihui Liu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Caiyu Lin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Tao Peng
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Yuanyao Dou
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Yimin Zhang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Fenfen Sun
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Weiling Jiang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Guoqing Yin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Rui Han
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
- Chongqing Key Laboratory of Precision Medicine and Prevention of Major Respiratory Diseases, Chongqing, China
- School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
26
|
Lu K, Zhao Y, Li Y, Fu Z, Chen Y, Kong Y, Li G. IFI16 promotes the progression of clear cell renal cell carcinoma through the IL6/PI3K/AKT axis. J Transl Med 2024; 22:533. [PMID: 38831470 PMCID: PMC11149187 DOI: 10.1186/s12967-024-05354-w] [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: 03/11/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is a common disease in the urinary system, with a high incidence and poor prognosis in advanced stages. Although γ-interferon-inducible protein 16 (IFI16) has been reported to play a role in various tumors, its involvement in ccRCC remains poorly documented, and the molecular mechanisms are not yet clear. METHODS We conducted bioinformatics analysis to study the expression of IFI16 in ccRCC using public databases. Additionally, we analyzed and validated clinical specimens that we collected. Subsequently, we explored the impact of IFI16 on ccRCC cell proliferation, migration, and invasion through in vitro and in vivo experiments. Furthermore, we predicted downstream molecules and pathways using transcriptome analysis and confirmed them through follow-up experimental validation. RESULTS IFI16 was significantly upregulated in ccRCC tissue and correlated with poor patient prognosis. In vitro, IFI16 promoted ccRCC cell proliferation, migration, and invasion, while in vivo, it facilitated subcutaneous tumor growth and the formation of lung metastatic foci. Knocking down IFI16 suppressed its oncogenic function. At the molecular level, IFI16 promoted the transcription and translation of IL6, subsequently activating the PI3K/AKT signaling pathway and inducing epithelial-mesenchymal transition (EMT). CONCLUSION IFI16 induced EMT through the IL6/PI3K/AKT axis, promoting the progression of ccRCC.
Collapse
Affiliation(s)
- Ke Lu
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China
- Department of Urology, Affiliated Changshu Hospital of Nantong University, Changshu, 215500, Jiangsu, China
| | - Yan Zhao
- Department of Urology, Xuzhou Cancer Hospital, Affiliated Hospital of Jiangsu University, Xuzhou, 221000, Jiangsu, China
| | - Yu Li
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China
| | - Zhenyu Fu
- Department of Urology, Affiliated Changshu Hospital of Nantong University, Changshu, 215500, Jiangsu, China
| | - Yongchang Chen
- Department of Urology, Affiliated Changshu Hospital of Nantong University, Changshu, 215500, Jiangsu, China.
| | - Ying Kong
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China.
| | - Gang Li
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu, China.
| |
Collapse
|
27
|
Konen JM, Wu H, Gibbons DL. Immune checkpoint blockade resistance in lung cancer: emerging mechanisms and therapeutic opportunities. Trends Pharmacol Sci 2024; 45:520-536. [PMID: 38744552 PMCID: PMC11189143 DOI: 10.1016/j.tips.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Immune checkpoint blockade (ICB) therapy works by inhibiting suppressive checkpoints that become upregulated after T cell activation, like PD-1/PD-L1 and CTLA-4. While the initial FDA approvals of ICB have revolutionized cancer therapies and fueled a burgeoning immuno-oncology field, more recent clinical development of new agents has been slow. Here, focusing on lung cancer, we review the latest research uncovering tumor cell intrinsic and extrinsic ICB resistance mechanisms as major hurdles to treatment efficacy and clinical progress. These include genomic and non-genomic tumor cell alterations, along with host and microenvironmental factors like the microbiome, metabolite accumulation, and hypoxia. Together, these factors can cooperate to promote immunosuppression and ICB resistance. Opportunities to prevent resistance are constantly evolving in this rapidly expanding field, with the goal of moving toward personalized immunotherapeutic regimens.
Collapse
Affiliation(s)
- Jessica M Konen
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
| | - Haoyi Wu
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
28
|
Yin Y, Wang Y, Wang C, Zhang Y, Qi A, Song J, Xu L, Yang W, Jiao L. Predicting the mechanism of action of YQYYJD prescription in the treatment of non-small cell lung cancer using transcriptomics analysis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117984. [PMID: 38428661 DOI: 10.1016/j.jep.2024.117984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/16/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The efficacy of the herbal formula Yiqi Yangyin Jiedu (YQYYJD) in the treatment of advanced lung cancer has been reported in clinical trials. However, the key anti-lung cancer herbs and molecular mechanisms underlying its inhibition of lung cancer are not well-understood. AIM OF THE STUDY To identify the key anti-lung cancer herbs in the YQYYJD formula and investigate their therapeutic effect and potential mechanism of action in non-small cell lung cancer (NSCLC) using transcriptomics and bioinformatics techniques. MATERIALS AND METHODS A mouse Lewis lung carcinoma (LLC) subcutaneous inhibitory tumor model was established with 6 mice in each group. Mice were treated with the YQYYJD split formula: Yiqi Formula (YQ), Yangyin Formula (YY), and Ruanjian Jiedu Formula (RJJD) for 14 days. The tumor volume and mouse weight were recorded, and the status of tumor occurrence was further observed by taking photos. The tumor was stained with hematoxylin-eosin to observe its histopathological changes. Immunohistochemistry was used to detect the expression of the proliferation marker Ki67 and the apoptotic marker Caspase-3 in tumor tissues. Flow cytometry was used to detect the number of CD4+ and CD8+ T cells and cytokines interleukin-2 (IL-2) and interferon-gamma (IFN-γ) in the spleen and tumor tissues. The differential genes of key drugs against tumors were obtained by transcriptome sequencing of tumors. Gene Ontology (GO) and Kyoto Encyclopedia of Gene and Genomes (KEGG) enrichment analyses were performed on differential genes to obtain pathways and biological processes where targets were aggregated. TIMER2.0 and TISIDB databases were used to evaluate the impact of drugs on immune cell infiltration and immune-related genes. The binding activity of the key targets and compounds was verified by molecular docking. RESULTS YQ, YY, and RJJD inhibited the growth of subcutaneous transplanted tumors in LLC mice to varying degrees and achieved antitumor effects by inhibiting the expression of tumor cell proliferation, apoptosis, and metastasis-related proteins. Among the three disassembled prescriptions, YQ better inhibited the growth of subcutaneous transplanted tumors in LLC mice, significantly promoted tumor necrosis, significantly increased the expression of Caspase-3 protein in tumor tissue, and significantly decreased the expression of Ki-67 (P < 0.05), thereby increasing the infiltration of CD8+ T cells. YQ significantly increased the expression of CD4+ and CD8+ T cells in tumor and splenic tissues of tumor-bearing mice and up-regulated the expression of IL-2 and IFN-γ. Transcriptome sequencing and bioinformatics results showed that after YQ intervention, differentially expressed genes were enriched in more than one tumor-related pathway and multiple immune regulation-related biological functions. There were 12 key immune-related target genes. CONCLUSION YQ was the key disassembled prescription of YQYYJD, exerting significant antitumor effects and immune regulation effects on NSCLC. It may have relieved T cell exhaustion and regulated the immune microenvironment to exert antitumor effects by changing lung cancer-related targets, pathways, and biological processes.
Collapse
Affiliation(s)
- Yinan Yin
- Department of Oncology, Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yichao Wang
- Department of Oncology, Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chengyan Wang
- Department of Oncology, Jing'an Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Yilu Zhang
- Department of Oncology, Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ao Qi
- Department of Oncology, Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiajun Song
- Department of Oncology, Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Xu
- Department of Oncology, Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Institute of Translational Cancer Research for Integrated Chinese and Western Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenxiao Yang
- Department of Oncology, Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Lijing Jiao
- Department of Oncology, Yue Yang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Institute of Translational Cancer Research for Integrated Chinese and Western Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
29
|
Li Y, Yang W, Liu C, Zhou S, Liu X, Zhang T, Wu L, Li X, Zhang J, Chang E. SFXN1-mediated immune cell infiltration and tumorigenesis in lung adenocarcinoma: A potential therapeutic target. Int Immunopharmacol 2024; 132:111918. [PMID: 38537539 DOI: 10.1016/j.intimp.2024.111918] [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: 01/29/2024] [Revised: 03/17/2024] [Accepted: 03/20/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Sideroflexin 1 (SFXN1), a mitochondrial serine transporter implicated in one-carbon metabolism, is a prognostic biomarker in lung adenocarcinoma (LUAD). However, its role in LUAD progression remains elusive. This study aimed to investigate the functional significance of SFXN1 in LUAD and evaluate its potential as a therapeutic target. METHODS We analyzed SFXN1 expression and its diagnostic and prognostic value in LUAD using the Pan-cancer TCGA dataset. In vitro assays (CCK-8, cell cycle, EDU, wound-healing, and transwell) were employed to assess the role of SFXN1, complemented by in vivo experiments. RNA sequencing elucidated SFXN1-mediated cellular functions and potential mechanisms. Bulk RNA-seq and scRNA-seq data from TCGA and GEO were used to investigate the correlation between SFXN1 and the tumor immune microenvironment. RT-qPCR, Western blot, and IHC assays validated SFXN1 expression and its impact on the immune microenvironment in LUAD. RESULTS SFXN1 was upregulated in LUAD tissues and associated with poor prognosis. RNA-seq and scRNA-seq analyses revealed increased SFXN1 expression in tumor cells, accompanied by decreased infiltration of NK and cytotoxic T cells. SFXN1 knockdown significantly reduced cell proliferation and migration, and the inhibition of ERK phosphorylation and CCL20 expression may be the molecular mechanism involved. In vivo, targeting SFXN1 decreased Tregs infiltration and inhibited tumor growth. CONCLUSIONS Our findings suggest that SFXN1 may be a potential therapeutic target for LUAD treatment.
Collapse
Affiliation(s)
- Yanjun Li
- Department of Anaesthesiology and Perioperative Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Wenke Yang
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Chaojun Liu
- Department of Breast Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Shengli Zhou
- Department of Pathology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Xiaozhuan Liu
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Tingting Zhang
- Center for Clinical Single-Cell Biomedicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, China
| | - Lingzhi Wu
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faulty of Medicine, Imperial College London, Chelsea and Westminster Hospital, UK
| | - Xinyi Li
- Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faulty of Medicine, Imperial College London, Chelsea and Westminster Hospital, UK
| | - Jiaqiang Zhang
- Department of Anaesthesiology and Perioperative Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, China.
| | - Enqiang Chang
- Department of Anaesthesiology and Perioperative Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan 450003, China; Division of Anaesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faulty of Medicine, Imperial College London, Chelsea and Westminster Hospital, UK.
| |
Collapse
|
30
|
Miao X, Jiang P, Zhang X, Li X, Wu Z, Jiang Y, Liu H, Xie W, Li X, Shi B, Cai J, Gong W. Lactobacillus rhamnosus HN001 facilitates the efficacy of dual PI3K/mTOR inhibition prolonging cardiac transplant survival and enhancing antitumor effect. Microbiol Spectr 2024; 12:e0183923. [PMID: 38564670 PMCID: PMC11064485 DOI: 10.1128/spectrum.01839-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 11/24/2023] [Indexed: 04/04/2024] Open
Abstract
Solid organ transplantation is a crucial treatment for patients who have reached the end stage of heart, lung, kidney, or liver failure. However, the likelihood of developing cancer post-transplantation increases. Additionally, primary malignant tumors remain a major obstacle to the long-term survival of transplanted organs. Therefore, it is essential to investigate effective therapies that can boost the immune system's ability to combat cancer and prevent allograft rejection. We established a mouse orthotopic liver tumor model and conducted allogeneic heterotopic heart transplantation. Various treatments were administered, and survival curves were generated using the Kaplan-Meier method. We also collected graft samples and measured inflammatory cytokine levels in the serum using an inflammatory array. The specificity of the histochemical techniques was tested by staining sections. We administered a combination therapy of phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) dual inhibitor BEZ235 and Lactobacillus rhamnosus HN001 to primary liver cancer model mice with cardiac allografts. Consistent with our prior findings, L. rhamnosus HN001 alleviated the intestinal flora imbalance caused by BEZ235. Our previous research confirmed that the combination of BEZ235 and L. rhamnosus HN001 significantly prolonged cardiac transplant survival. IMPORTANCE We observed that the combination of phosphoinositide 3-kinase/mammalian target of rapamycin (PI3K/mTOR) dual inhibitor BEZ235 and Lactobacillus rhamnosus HN001 notably prolonged cardiac transplant survival while also inhibiting the progression of primary liver cancer. The combination therapy was efficacious in treating antitumor immunity and allograft rejection, as demonstrated by the efficacy results. We also found that this phenomenon was accompanied by the regulation of inflammatory IL-6 expression. Our study presents a novel and effective therapeutic approach to address antitumor immunity and prevent allograft rejection.
Collapse
Affiliation(s)
- Xiaolong Miao
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Peng Jiang
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaotong Zhang
- Medical department, Qingdao Eighth People’s Hospital, Qingdao, China
| | - Xinqiang Li
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zelai Wu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuancong Jiang
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Han Liu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Weixun Xie
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Xinwei Li
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bingfeng Shi
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jinzhen Cai
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weihua Gong
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| |
Collapse
|
31
|
Wang Y, Chen K, Liu G, Du C, Cheng Z, Wei D, Li F, Li C, Yang Y, Zhao Y, Nie G. Disruption of Super-Enhancers in Activated Pancreatic Stellate Cells Facilitates Chemotherapy and Immunotherapy in Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308637. [PMID: 38417121 PMCID: PMC11040371 DOI: 10.1002/advs.202308637] [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: 11/11/2023] [Revised: 01/27/2024] [Indexed: 03/01/2024]
Abstract
One major obstacle in the drug treatment of pancreatic ductal adenocarcinoma (PDAC) is its highly fibrotic tumor microenvironment, which is replete with activated pancreatic stellate cells (a-PSCs). These a-PSCs generate abundant extracellular matrix and secrete various cytokines to form biophysical and biochemical barriers, impeding drug access to tumor tissues. Therefore, it is imperative to develop a strategy for reversing PSC activation and thereby removing the barriers to facilitate PDAC drug treatment. Herein, by integrating chromatin immunoprecipitation (ChIP)-seq, Assays for Transposase-Accessible Chromatin (ATAC)-seq, and RNA-seq techniques, this work reveals that super-enhancers (SEs) promote the expression of various genes involved in PSC activation. Disruption of SE-associated transcription with JQ1 reverses the activated phenotype of a-PSCs and decreases stromal fibrosis in both orthotopic and patient-derived xenograft (PDX) models. More importantly, disruption of SEs by JQ1 treatments promotes vascularization, facilitates drug delivery, and alters the immune landscape in PDAC, thereby improving the efficacies of both chemotherapy (with gemcitabine) and immunotherapy (with IL-12). In summary, this study not only elucidates the contribution of SEs of a-PSCs in shaping the PDAC tumor microenvironment but also highlights that targeting SEs in a-PSCs may become a gate-opening strategy that benefits PDAC drug therapy by removing stromal barriers.
Collapse
Affiliation(s)
- Yazhou Wang
- Pancreas CenterThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210000China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Kai Chen
- Department of General SurgeryPeking University First HospitalBeijing100034China
| | - Gang Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of EducationNortheast Normal UniversityChangchun130024China
| | - Chong Du
- Department of OncologyThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'an710061China
| | - Zhaoxia Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Dan Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Fenfen Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Chen Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Yinmo Yang
- Department of General SurgeryPeking University First HospitalBeijing100034China
| | - Ying Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & NanosafetyCAS Center of Excellence in NanoscienceNational Center for Nanoscience and TechnologyBeijing100190China
| |
Collapse
|
32
|
Xie A, Wang J, Liu Y, Li G, Yang N. Impacts of β-1, 3-N-acetylglucosaminyltransferases (B3GNTs) in human diseases. Mol Biol Rep 2024; 51:476. [PMID: 38553573 DOI: 10.1007/s11033-024-09405-9] [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: 12/04/2023] [Accepted: 02/29/2024] [Indexed: 04/02/2024]
Abstract
Glycosylation modification of proteins is a common post-translational modification that exists in various organisms and has rich biological functions. It is usually catalyzed by multiple glycosyltransferases located in the Golgi apparatus. β-1,3-N-acetylglucosaminyltransferases (B3GNTs) are members of the glycosyltransferases and have been found to be involved in the occurrence and development of a variety of diseases including autoimmunity diseases, cancers, neurodevelopment, musculoskeletal system, and metabolic diseases. The functions of B3GNTs represent the glycosylation of proteins is a crucial and frequently life-threatening step in progression of most diseases. In this review, we give an overview about the roles of B3GNTs in tumor, nervous system, musculoskeletal and metabolic diseases, describing the recent results about B3GNTs, in order to provide a research direction and exploration value for the prevention, diagnosis and treatment of these diseases.
Collapse
Affiliation(s)
- Anna Xie
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jingjing Wang
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yi Liu
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Guoqing Li
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Nanyang Yang
- The Hengyang Key Laboratory of Cellular Stress Biology, Institute of Cytology and Genetics, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
33
|
Zhao Y, Jia Y, Wang J, Chen X, Han J, Zhen S, Yin S, Lv W, Yu F, Wang J, Xu F, Zhao X, Liu L. circNOX4 activates an inflammatory fibroblast niche to promote tumor growth and metastasis in NSCLC via FAP/IL-6 axis. Mol Cancer 2024; 23:47. [PMID: 38459511 PMCID: PMC10921747 DOI: 10.1186/s12943-024-01957-5] [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: 11/08/2023] [Accepted: 02/10/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) orchestrate a supportive niche that fuels cancer metastatic development in non-small cell lung cancer (NSCLC). Due to the heterogeneity and plasticity of CAFs, manipulating the activated phenotype of fibroblasts is a promising strategy for cancer therapy. However, the underlying mechanisms of fibroblast activation and phenotype switching that drive metastasis remain elusive. METHODS The clinical implications of fibroblast activation protein (FAP)-positive CAFs (FAP+CAFs) were evaluated based on tumor specimens from NSCLC patients and bioinformatic analysis of online databases. CAF-specific circular RNAs (circRNAs) were screened by circRNA microarrays of primary human CAFs and matched normal fibroblasts (NFs). Survival analyses were performed to assess the prognostic value of circNOX4 in NSCLC clinical samples. The biological effects of circNOX4 were investigated by gain- and loss-of-function experiments in vitro and in vivo. Fluorescence in situ hybridization, luciferase reporter assays, RNA immunoprecipitation, and miRNA rescue experiments were conducted to elucidate the underlying mechanisms of fibroblast activation. Cytokine antibody array, transwell coculture system, and enzyme-linked immunosorbent assay (ELISA) were performed to investigate the downstream effectors that promote cancer metastasis. RESULTS FAP+CAFs were significantly enriched in metastatic cancer samples, and their higher abundance was correlated with the worse overall survival in NSCLC patients. A novel CAF-specific circRNA, circNOX4 (hsa_circ_0023988), evoked the phenotypic transition from NFs into CAFs and promoted the migration and invasion of NSCLC in vitro and in vivo. Clinically, circNOX4 correlated with the poor prognosis of advanced NSCLC patients. Mechanistically, circNOX4 upregulated FAP by sponging miR-329-5p, which led to fibroblast activation. Furthermore, the circNOX4/miR-329-5p/FAP axis activated an inflammatory fibroblast niche by preferentially inducing interleukin-6 (IL-6) and eventually promoting NSCLC progression. Disruption of the intercellular circNOX4/IL-6 axis significantly suppressed tumor growth and metastatic colonization in vivo. CONCLUSIONS Our study reveals a role of the circRNA-induced fibroblast niche in tumor metastasis and highlights that targeting the circNOX4/FAP/IL-6 axis is a promising strategy for the intervention of NSCLC metastasis.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, China
- Department of Oncology, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050011, China
| | - Yunlong Jia
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, China
| | - Jiali Wang
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, China
| | - Xiaolin Chen
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050011, China
| | - Jingya Han
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050011, China
| | - Shuman Zhen
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, China
| | - Shuxian Yin
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, China
| | - Wei Lv
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, China
| | - Fan Yu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050011, China
| | - Jiaqi Wang
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, China
| | - Fan Xu
- Departments of Oncology, The Affiliated Hospital of Chengde Medical University, Chengde, 067000, China
| | - Xinming Zhao
- Department of Nuclear Medicine, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050011, China.
- Hebei Provincial Key Laboratory of Tumor Microenvironment and Drug Resistance, Shijiazhuang, 050011, China.
| | - Lihua Liu
- Department of Tumor Immunotherapy, The Fourth Hospital of Hebei Medical University and Hebei Provincial Tumor Hospital, Shijiazhuang, 050035, China.
- Cancer Research Institute of Hebei Province, Shijiazhuang, 050011, China.
- International Cooperation Laboratory of Stem Cell Research, Hebei Medical University, Shijiazhuang, 050011, China.
| |
Collapse
|
34
|
Xie Y, Wu H, He Y, Liu L, Huang IB, Zhou L, Lin CY, Leung RWH, Loh JJ, Lee TKW, Ding J, Man K, Ma S, Tong M. Targeting AXL induces tumor-intrinsic immunogenic response in tyrosine kinase inhibitor-resistant liver cancer. Cell Death Dis 2024; 15:110. [PMID: 38310091 PMCID: PMC10838288 DOI: 10.1038/s41419-024-06493-0] [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: 07/28/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/05/2024]
Abstract
Hepatocellular carcinoma (HCC) is an aggressive malignancy without effective therapeutic approaches. Here, we evaluate the tumor-intrinsic mechanisms that attenuate the efficacy of immune checkpoint inhibitor (ICI) that is observed in patients with advanced HCC who progress on first-line tyrosine kinase inhibitor (TKI) therapy. Upregulation of AXL observed in sorafenib- and lenvatinib-resistant HCCs is correlated with poor response towards TKI and ICI treatments. AXL upregulation protects sorafenib-resistant HCC cells from oxidative stress, mitochondrial damage, and accompanying immunogenic cell death through suppressed tumor necrosis factor-α (TNF-α) and STING-type I interferon pathways. Pharmacological inhibition of AXL abrogates the protective effect and re-sensitizes TKI-resistant HCC tumors to anti-PD-1 treatment. We suggest that targeting AXL in combination with anti-PD-1 may provide an alternative treatment scheme for HCC patients who progress on TKI treatment.
Collapse
Affiliation(s)
- Yunong Xie
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Haofeng Wu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yimiao He
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Linglin Liu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ianto Bosheng Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lei Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Cheuk-Yin Lin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Rainbow Wing-Hei Leung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jia-Jian Loh
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Terence Kin-Wah Lee
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jin Ding
- Clinical Cancer Institute, Center for Translational Medicine, Naval Medical University, Shanghai, China
| | - Kwan Man
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China
| | - Stephanie Ma
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong, China.
- Hong Kong University-Shenzhen Hospital, Shenzhen, China.
| | - Man Tong
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
35
|
Lyu C, Sun H, Sun Z, Liu Y, Wang Q. Roles of exosomes in immunotherapy for solid cancers. Cell Death Dis 2024; 15:106. [PMID: 38302430 PMCID: PMC10834551 DOI: 10.1038/s41419-024-06494-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
Although immunotherapy has made breakthrough progress, its efficacy in solid tumours remains unsatisfactory. Exosomes are the main type of extracellular vesicles that can deliver various intracellular molecules to adjacent or distant cells and organs, mediating various biological functions. Studies have found that exosomes can both activate the immune system and inhibit the immune system. The antigen and major histocompatibility complex (MHC) carried in exosomes make it possible to develop them as anticancer vaccines. Exosomes derived from blood, urine, saliva and cerebrospinal fluid can be used as ideal biomarkers in cancer diagnosis and prognosis. In recent years, exosome-based therapy has made great progress in the fields of drug transportation and immunotherapy. Here, we review the composition and sources of exosomes in the solid cancer immune microenvironment and further elaborate on the potential mechanisms and pathways by which exosomes influence immunotherapy for solid cancers. Moreover, we summarize the potential clinical application prospects of engineered exosomes and exosome vaccines in immunotherapy for solid cancers. Eventually, these findings may open up avenues for determining the potential of exosomes for diagnosis, treatment, and prognosis in solid cancer immunotherapy.
Collapse
Affiliation(s)
- Cong Lyu
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Haifeng Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Yang Liu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
36
|
Ozawa Y, Koh Y, Hase T, Chibana K, Kaira K, Okishio K, Ichihara E, Murakami S, Shimokawa M, Yamamoto N. Prospective observational study to explore genes and proteins predicting efficacy and safety of brigatinib for ALK-gene rearranged non-small-cell lung cancer: study protocol for ABRAID study (WJOG11919L). Ther Adv Med Oncol 2024; 16:17588359231225046. [PMID: 38282663 PMCID: PMC10822087 DOI: 10.1177/17588359231225046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 12/11/2023] [Indexed: 01/30/2024] Open
Abstract
Background ALK-tyrosine kinase inhibitors (ALK-TKIs) are effective for treating non-small-cell lung cancer with ALK gene rearrangement; however, resistance is inevitable. Brigatinib is a unique ALK-TKI that is effective against many resistance mutations. However, data on factors associated with its efficacy and resistance mechanisms are limited. Objectives This study will evaluate the efficacy and safety of brigatinib in the real world and explore factors related to its efficacy, safety, and resistance mechanisms. Design Prospective observational study. Ethics This study is approved by the Ethics Committee of Wakayama Medical University. Written informed consent will be obtained from all patients before study-related procedures. Methods and analysis This study comprises three cohorts. Cohorts A, B, and 0 will enroll patients receiving alectinib as the first ALK-TKI, receiving alectinib as the first ALK-TKI and subsequently cytotoxic agents and/or lorlatinib after alectinib, and without a history of ALK-TKI, respectively. Overall, 100, 30, and 50 patients will be enrolled in Cohorts A, B, and 0, respectively. Circulating tumor DNA before starting brigatinib and at disease progression will be analyzed in all cohorts using a hypersensitive next-generation sequencing (NGS) PGDx Elio plasma resolve panel. Serum protein levels will be analyzed using the Milliplex xMAP assay system with a Luminex 200 (Luminex, Austin, USA). The enrollment period is 31 months and the patients will be observed for 2 years after enrollment. Archived tissues will be collected for NGS analysis, gene expression analysis, and immunohistochemistry staining 1 year after completion of registration. Quality of life and safety evaluation using electronic patient-reported outcomes will be investigated. Discussion This study will elucidate predictors of ALK-TKI efficacy and resistance mechanisms and evaluate the efficacy and safety of brigatinib in a real-world setting. The results will provide crucial information for establishing treatment strategies, discovering novel biomarkers, and developing new therapeutic agents. Trial registration UMIN000042439.
Collapse
Affiliation(s)
- Yuichi Ozawa
- Department of Respiratory Medicine, Hamamatsu Medical Center, 328 Tomitsuka-cho, Naka-ku, Hamamatsu, Shizuoka 432-8580, Japan
- Internal Medicine III, Wakayama Medical University, Wakayama City, Wakayama 641-0012, Japan
| | - Yasuhiro Koh
- Internal Medicine III, Wakayama Medical University, Wakayama City, Wakayama, Japan
- Center for Biomedical Sciences, Wakayama Medical University, Wakayama City, Wakayama, Japan
| | - Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kenji Chibana
- Department of Respiratory Medicine, National Hospital Organization Okinawa National Hospital, Ginowan, Okinawa, Japan
| | - Kyoichi Kaira
- Department of Respiratory Medicine, Comprehensive Cancer Center, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Kyoichi Okishio
- Department of Internal Medicine, National Hospital Organization Kinki-chuo Chest Medical Center, Sakai City, Osaka, Japan
| | - Eiki Ichihara
- Center for Clinical Oncology, Okayama University Hospital, Okayama, Okayama, Japan
| | - Shuji Murakami
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Nobuyuki Yamamoto
- Internal Medicine III, Wakayama Medical University, Wakayama City, Wakayama, Japan
| |
Collapse
|
37
|
Ma J, Liu D, Mao X, Huang L, Ren Y, Xu X, Huang X, Deng C, Shi F, Sun P. Enhanced Diagnostic Efficiency of Endometrial Carcinogenesis and Progression in Women with Abnormal Uterine Bleeding through Peripheral Blood Cytokine Testing: A Multicenter Retrospective Cohort Study. Int J Med Sci 2024; 21:601-611. [PMID: 38464838 PMCID: PMC10920852 DOI: 10.7150/ijms.91506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 01/09/2024] [Indexed: 03/12/2024] Open
Abstract
Objective: This study aimed to evaluate the role of plasma cytokine detection in endometrial cancer screening and tumor progression assessment in patients with abnormal uterine bleeding. Methods: In this multicenter retrospective cohort study of 287 patients with abnormal uterine bleeding, comprehensive clinical information and laboratory assessments, including cytokines, routine blood tests, and tumor markers, were performed. Associations between the clinical indicators and endometrial carcinogenesis/progression were evaluated. The independent risk factors for endometrial cancer and endometrial cancer with deep myometrial invasion were analyzed using multivariate binary logistic regression. Additionally, a diagnostic model was used to evaluate the predictive efficacy of these identified risk factors. Results: In patients with abnormal uterine bleeding, low IL-4 and high IL-8 levels were independent risk factors for endometrial cancer (p < 0.05). Combining IL-4, IL-8, CA125, and menopausal status improved the accuracy of assessing endometrial cancer risk. The area under curve of the model is 0.816. High IL-6 and IL-8 levels were independent risk factors for deep myometrial invasion in patients with endometrial cancer (p < 0.05). Similarly, combining IL-6, IL-8, and Monocyte counts enhanced the accuracy of assessing endometrial cancer risk with deep myometrial invasion. The area under curve of the model is 0.753. Conclusions: Cytokines such as IL-4, IL-8, and IL-6 can serve as markers for monitoring endometrial cancer and its progression in women with abnormal uterine bleeding.
Collapse
Affiliation(s)
- Jincheng Ma
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Dabin Liu
- Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
| | - Xiaodan Mao
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Leyi Huang
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Yuan Ren
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Xiaozhen Xu
- Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Xiaoli Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, Fujian, China
- Department of Obstetrics and Gynecology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, Fujian, China
| | - Caiping Deng
- The Second Hospital of Nanping City, Nanping 354200, Fujian, China
| | - Feifeng Shi
- Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, Fujian, China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
- Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China
| |
Collapse
|
38
|
Meng X, Xu H, Liang Y, Liang M, Song W, Zhou B, Shi J, Du M, Gao Y. Enhanced CT-based radiomics model to predict natural killer cell infiltration and clinical prognosis in non-small cell lung cancer. Front Immunol 2024; 14:1334886. [PMID: 38283362 PMCID: PMC10811188 DOI: 10.3389/fimmu.2023.1334886] [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: 11/08/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Background Natural killer (NK) cells are crucial for tumor prognosis; however, their role in non-small-cell lung cancer (NSCLC) remains unclear. The current detection methods for NSCLC are inefficient and costly. Therefore, radiomics represent a promising alternative. Methods We analyzed the radiogenomics datasets to extract clinical, radiological, and transcriptome data. The effect of NK cells on the prognosis of NSCLC was assessed. Tumors were delineated using a 3D Slicer, and features were extracted using pyradiomics. A radiomics model was developed and validated using five-fold cross-validation. A nomogram model was constructed using the selected clinical variables and a radiomic score (RS). The CIBERSORTx database and gene set enrichment analysis were used to explore the correlations of NK cell infiltration and molecular mechanisms. Results Higher infiltration of NK cells was correlated with better overall survival (OS) (P = 0.002). The radiomic model showed an area under the curve of 0.731, with 0.726 post-validation. The RS differed significantly between high and low infiltration of NK cells (P < 0.01). The nomogram, using RS and clinical variables, effectively predicted 3-year OS. NK cell infiltration was correlated with the ICOS and BTLA genes (P < 0.001) and macrophage M0/M2 levels. The key pathways included TNF-α signaling via NF-κB and Wnt/β-catenin signaling. Conclusions Our radiomic model accurately predicted NK cell infiltration in NSCLC. Combined with clinical characteristics, it can predict the prognosis of patients with NSCLC. Bioinformatic analysis revealed the gene expression and pathways underlying NK cell infiltration in NSCLC.
Collapse
Affiliation(s)
- Xiangzhi Meng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haijun Xu
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Liang
- Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weijian Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Boxuan Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Shi
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjun Du
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
39
|
Olivera I, Luri-Rey C, Teijeira A, Eguren-Santamaria I, Gomis G, Palencia B, Berraondo P, Melero I. Facts and Hopes on Neutralization of Protumor Inflammatory Mediators in Cancer Immunotherapy. Clin Cancer Res 2023; 29:4711-4727. [PMID: 37522874 DOI: 10.1158/1078-0432.ccr-22-3653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/26/2023] [Accepted: 07/14/2023] [Indexed: 08/01/2023]
Abstract
In cancer pathogenesis, soluble mediators are responsible for a type of inflammation that favors the progression of tumors. The mechanisms chiefly involve changes in the cellular composition of the tumor tissue stroma and in the functional modulation of myeloid and lymphoid leukocytes. Active immunosuppression, proangiogenesis, changes in leukocyte traffic, extracellular matrix remodeling, and alterations in tumor-antigen presentation are the main mechanisms linked to the inflammation that fosters tumor growth and metastasis. Soluble inflammatory mediators and their receptors are amenable to various types of inhibitors that can be combined with other immunotherapy approaches. The main proinflammatory targets which can be interfered with at present and which are under preclinical and clinical development are IL1β, IL6, the CXCR1/2 chemokine axis, TNFα, VEGF, leukemia inhibitory factor, CCL2, IL35, and prostaglandins. In many instances, the corresponding neutralizing agents are already clinically available and can be repurposed as a result of their use in other areas of medicine such as autoimmune diseases and chronic inflammatory conditions.
Collapse
Affiliation(s)
- Irene Olivera
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Alvaro Teijeira
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Iñaki Eguren-Santamaria
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Gabriel Gomis
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Belen Palencia
- Department of Immunology and Immunotherapy, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
40
|
Rosell R, Jain A, Codony-Servat J, Jantus-Lewintre E, Morrison B, Ginesta JB, González-Cao M. Biological insights in non-small cell lung cancer. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0108. [PMID: 37381723 PMCID: PMC10466437 DOI: 10.20892/j.issn.2095-3941.2023.0108] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/05/2023] [Indexed: 06/30/2023] Open
Abstract
Lung oncogenesis relies on intracellular cysteine to overcome oxidative stress. Several tumor types, including non-small cell lung cancer (NSCLC), upregulate the system xc- cystine/glutamate antiporter (xCT) through overexpression of the cystine transporter SLC7A11, thus sustaining intracellular cysteine levels to support glutathione synthesis. Nuclear factor erythroid 2-related factor 2 (NRF2) serves as a master regulator of oxidative stress resistance by regulating SLC7A11, whereas Kelch-like ECH-associated protein (KEAP1) acts as a cytoplasmic repressor of the oxidative responsive transcription factor NRF2. Mutations in KEAP1/NRF2 and p53 induce SLC7A11 activation in NSCLC. Extracellular cystine is crucial in supplying the intracellular cysteine levels necessary to combat oxidative stress. Disruptions in cystine availability lead to iron-dependent lipid peroxidation, thus resulting in a type of cell death called ferroptosis. Pharmacologic inhibitors of xCT (either SLC7A11 or GPX4) induce ferroptosis of NSCLC cells and other tumor types. When cystine uptake is impaired, the intracellular cysteine pool can be sustained by the transsulfuration pathway, which is catalyzed by cystathionine-B-synthase (CBS) and cystathionine g-lyase (CSE). The involvement of exogenous cysteine/cystine and the transsulfuration pathway in the cysteine pool and downstream metabolites results in compromised CD8+ T cell function and evasion of immunotherapy, diminishing immune response and potentially reducing the effectiveness of immunotherapeutic interventions. Pyroptosis is a previously unrecognized form of regulated cell death. In NSCLCs driven by EGFR, ALK, or KRAS, selective inhibitors induce pyroptotic cell death as well as apoptosis. After targeted therapy, the mitochondrial intrinsic apoptotic pathway is activated, thus leading to the cleavage and activation of caspase-3. Consequently, gasdermin E is activated, thus leading to permeabilization of the cytoplasmic membrane and cell-lytic pyroptosis (indicated by characteristic cell membrane ballooning). Breakthroughs in KRAS G12C allele-specific inhibitors and potential mechanisms of resistance are also discussed herein.
Collapse
Affiliation(s)
- Rafael Rosell
- Germans Trias i Pujol Research Institute, Badalona 08028, Spain
- IOR, Hospital Quiron-Dexeus, Barcelona 08028, Spain
| | - Anisha Jain
- Department of Microbiology, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | | | - Eloisa Jantus-Lewintre
- Department of Biotechnology, Universitat Politècnica de Valencia; Mixed Unit TRIAL (General University Hospital of Valencia Research Foundation and Príncipe Felipe Research Center), CIBERONC, Valencia 46014, Spain
| | - Blake Morrison
- Sumitomo Pharma Oncology, Inc., Cambridge, MA and Lehi, UT 84043, USA
| | | | | |
Collapse
|
41
|
Yan R, Huang X, Liu H, Xiao Z, Liu J, An G, Ge Y. DCLK1 Drives EGFR-TKI-Acquired Resistance in Lung Adenocarcinoma by Remodeling the Epithelial-Mesenchymal Transition Status. Biomedicines 2023; 11:biomedicines11051490. [PMID: 37239162 DOI: 10.3390/biomedicines11051490] [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: 02/06/2023] [Revised: 05/11/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
OBJECTIVE Epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) is a first-line treatment for lung adenocarcinoma with EGFR-sensitive mutations, but acquired resistance to EGFR-TKIs remains a problem in clinical practice. The development of epithelial-mesenchymal transition (EMT) is a critical mechanism that induces acquired resistance to TKIs. Reversing acquired resistance to EGFR-TKIs through targeting the key molecules driving EMT provides an alternative choice for patients. We, therefore, aimed to explore the role of doublecortin-like kinase 1 (DCLK1) as an EMT driver gene in the acquired resistance of lung adenocarcinoma to EGFR-TKIs. METHODS The IC50 of Gefitinib or Osimertinib in PC9/HCC827 cells was measured using a cell counting kit-8 (CCK8) assay. The expression levels of EMT-related genes in PC9 and HCC827 cells were detected using RT-PCR and Western blot. Cell migration and invasion abilities were assessed via a transwell assay. For the in vivo experiments, PC9 cells were subcutaneously injected into BALB/c nude mice to form tumors. Upon harvesting, tumor tissues were retained for RT-PCR, Western blot, and polychromatic fluorescence staining to detect biomarker changes in the EMT process. RESULTS Gefitinib-resistant PC9 (PC9/GR) and Osimertinib-resistant HCC827 (HCC827/OR) cells showed remarkable activation of EMT and enhanced migration and invasion abilities compared to TKI-sensitive cells. In addition, DCLK1 expression was markedly increased in EGFR-TKI-resistant lung adenocarcinoma cells. The targeted knockout of DCLK1 effectively reversed the EMT phenotype in TKI-resistant cells and improved EGFR-TKI sensitivity, which was further validated by the in vivo experiments. CONCLUSIONS DCLK1 facilitates acquired resistance to EGFR-TKI in lung adenocarcinoma by inducting EMT and accelerating the migration and invasion abilities of TKI-resistant cells.
Collapse
Affiliation(s)
- Rui Yan
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, 8 Gongren Tiyuchang Nanlu Road, Chaoyang Dist., Beijing 100020, China
| | - Xuying Huang
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, 8 Gongren Tiyuchang Nanlu Road, Chaoyang Dist., Beijing 100020, China
| | - Heshu Liu
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, 8 Gongren Tiyuchang Nanlu Road, Chaoyang Dist., Beijing 100020, China
| | - Zeru Xiao
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, 8 Gongren Tiyuchang Nanlu Road, Chaoyang Dist., Beijing 100020, China
| | - Jian Liu
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, 8 Gongren Tiyuchang Nanlu Road, Chaoyang Dist., Beijing 100020, China
| | - Guangyu An
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, 8 Gongren Tiyuchang Nanlu Road, Chaoyang Dist., Beijing 100020, China
| | - Yang Ge
- Beijing Chao-Yang Hospital Department of Oncology, Capital Medical University, 8 Gongren Tiyuchang Nanlu Road, Chaoyang Dist., Beijing 100020, China
| |
Collapse
|
42
|
Kwok HH, Yang J, Lam DCL. Breaking the Invisible Barriers: Unleashing the Full Potential of Immune Checkpoint Inhibitors in Oncogene-Driven Lung Adenocarcinoma. Cancers (Basel) 2023; 15:2749. [PMID: 37345086 DOI: 10.3390/cancers15102749] [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: 04/11/2023] [Revised: 05/11/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
The rapid development of targeted therapy paved the way toward personalized medicine for advanced non-small cell lung cancer (NSCLC). Lung adenocarcinoma (ADC) harboring actionable genetic alternations including epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), Kirsten rat sarcoma virus (ALK) and c-ros oncogene 1 (ROS1) treated with tyrosine kinase inhibitors (TKIs) incurred lesser treatment toxicity but better therapeutic responses compared with systemic chemotherapy. Angiogenesis inhibitors targeting vascular endothelial growth factor (VEGF) have also shown an increase in overall survival (OS) for NSCLC patients. However, acquired resistance to these targeted therapies remains a major obstacle to long-term maintenance treatment for lung ADC patients. The emergence of immune checkpoint inhibitors (ICIs) against programmed cell death protein 1 (PD-1) or programmed cell death-ligand 1 (PD-L1) has changed the treatment paradigm for NSCLC tumors without actionable genetic alternations. Clinical studies have suggested, however, that there are no survival benefits with the combination of targeted therapy and ICIs. In this review, we will summarize and discuss the current knowledge on the tumor immune microenvironment and the dynamics of immune phenotypes, which could be crucial in extending the applicability of ICIs for this subpopulation of lung ADC patients.
Collapse
Affiliation(s)
- Hoi-Hin Kwok
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jiashuang Yang
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David Chi-Leung Lam
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
43
|
Rosell R, Aguilar-Hernández A, González-Cao M. Insights into EGFR Mutations and Oncogenic KRAS Mutations in Non-Small-Cell Lung Cancer. Cancers (Basel) 2023; 15:cancers15092519. [PMID: 37173989 PMCID: PMC10177428 DOI: 10.3390/cancers15092519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 04/23/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
Genetic mutations can activate different sets of proto-oncogenes and tumor suppressors genes [...].
Collapse
Affiliation(s)
- Rafael Rosell
- Germans Trias i Pujol Research Institute, 08916 Badalona, Spain
- IOR, Hospital Quiron-Dexeus, 08028 Barcelona, Spain
| | | | | |
Collapse
|
44
|
Rosell R, González-Cao M, Ito M, Santarpia M, Aguilar A, Codony-Servat J. The role of biomarkers in stage III non-small cell lung cancer. Expert Rev Respir Med 2023; 17:469-480. [PMID: 37317885 DOI: 10.1080/17476348.2023.2223985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
INTRODUCTION Stage III non-small cell lung cancer (NSCLC) is a composite of the regional spread of lung cancer with different levels of potential lymph node involvement and tumor size that often deem the stage at time of diagnosis to be unresectable and suitable for chemoradiation plus consolidation immunotherapy with durvalumab for 12 months. Chemoradiation plus durvalumab consolidation yielded a landmark 49.2% 5-year overall survival in unresectable NSCLC. AREAS COVERED Sub-optimal results lead us to focus on the mechanisms of resistance responsible for intractability in a significant proportion of cases that fail with chemoradiation and immunotherapy. In stage III NSCLC it is opportune to explore the accumulated evidence on ferroptosis resistance that can lead to cancer progression and metastasis. Strong data shows that three anti-ferroptosis pathways are principally involved in resistance to chemotherapy, radiation, and immunotherapy. EXPERT OPINION Because a large part of stage III NSCLCs is resistant to chemoradiation and durvalumab consolidation, a ferroptosis-based therapeutic approach, combined with standard-of-care therapy, can lead to improved clinical outcomes in patients diagnosed with stage III and possibly stage IV NSCLCs.
Collapse
Affiliation(s)
- Rafael Rosell
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital, Barcelona, Spain
- Laboratory of Molecular Biology of Cancer, Germans Trias I Pujol Health Sciences Institute and Hospital (IGTP), Badalona, Spain
- Medical Oncology Department, Catalan Institute of Oncology, Badalona, Spain
| | - María González-Cao
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital, Barcelona, Spain
| | - Masaoki Ito
- Department of Surgical Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy
| | - Andrés Aguilar
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Dexeus University Hospital, Barcelona, Spain
| | - Jordi Codony-Servat
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| |
Collapse
|