1
|
Zhang H, Cai W, Miao Y, Gu Y, Zhou X, Kaneda H, Wang L. Long Non-Coding RNA LINC01116 Promotes the Proliferation of Lung Adenocarcinoma by Targeting miR-9-5p/CCNE1 Axis. J Cell Mol Med 2024; 28:e70270. [PMID: 39648148 PMCID: PMC11625508 DOI: 10.1111/jcmm.70270] [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: 09/20/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/10/2024] Open
Abstract
Long non-coding RNA (lncRNA) LINC01116 is crucial in promoting cell proliferation, invasion and migration in solid tumours, including lung adenocarcinoma (LUAD). LINC01116 acts as a competing endogenous RNAs (ceRNA) that binds competitively to microRNAs and plays a critical role in tumour migration and invasion. However, other mechanisms of action besides the ceRNA theory have been rarely reported and remain to be elucidated further. The differences in RNA and protein levels in cells and tissues were assessed through real-time quantitative PCR and Western blot analysis. In vitro functional assays and in vivo xenograft models were used to analyse the function of LINC01116 in LUAD. Thus, the molecular correlation between miR-9-5p and CCNE1 was investigated through direct and indirect mechanism experiments. LINC01116, miR-9-5p and CCNE1 were upregulated in LUAD cell lines and tissues and were associated with a poor prognosis in patients. LINC01116 depletion inhibited proliferation but facilitated cell apoptosis. AGO2-RNA binding protein immunoprecipitation (AGO2-RIP) experiments confirmed that AGO2 binds to LINC01116 and miR-9-5p, indicating that LINC01116 interacts with miR-9-5p. The overexpression of miR-9-5p and CCNE1 effectively counteracts the biological effects of LINC01116 knockdown on reduced proliferation and cell cycle arrest in LUAD cells. The downregulation of miR-9-5p significantly reduces the CCNE1 level in A549 cells, and the upregulation of LINC01116 counteracts the downregulation of miR-9-5p effect, restoring the expression level of CCNE1. Our data demonstrated that LINC01116 regulates the expression of CCNE1 by positively regulating miR-9-5p, thereby affecting cell cycle, proliferation and participating in the development of LUAD.
Collapse
Affiliation(s)
- Hui Zhang
- The Jiangyin Clinical College of Xuzhou Medical UniversityXuzhouChina
| | - Wenwen Cai
- Sanmen County People's HospitalTaizhouChina
| | - Yiyan Miao
- The Jiangyin Clinical College of Xuzhou Medical UniversityXuzhouChina
| | - Yihang Gu
- Department of GeriatricsThe Jiangyin Clinical College of Xuzhou Medical UniversityJiangyinChina
| | - Xiaorong Zhou
- Department of Immunology, School of MedicineNantong UniversityNantongChina
| | - Hiroyasu Kaneda
- Department of Clinical Oncology, Graduate School of MedicineOsaka Metropolitan UniversityOsakaJapan
| | - Lan Wang
- Department of Respiratory and Critical Care MedicineThe Jiangyin Clinical College of Xuzhou Medical UniversityJiangyinChina
| |
Collapse
|
2
|
Xia T, Zhang Y, Peng H, Jia X, Yang D, Wei L, Li T, Yao W. EVA1B facilitates esophageal squamous carcinoma progression and recruitment of immunosuppressive myeloid-derived suppressor cells in the tumor microenvironment. Pharmacol Res 2024; 210:107521. [PMID: 39603573 DOI: 10.1016/j.phrs.2024.107521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024]
Abstract
Eva-1 Homolog B (EVA1B) has been preliminarily found to be associated with prognostic outcomes and immune microenvironment in several human cancer types, but the implications of EVA1B in ESCC remain unclear. Human ESCC and paracancerous tissues were gathered in this study, and EVA1B expression was measured via immunoblotting. EC109 and KYSE-180 ESCC cells were stably infected by sh-EVA1B lentivirus, and functional experiments were subsequently implemented. Syngeneic mouse models were built, and the expansion and recruitment of myeloid-derived suppressor cells (MDSCs) were then evaluated. The results showed that EVA1B presented the notable up-regulation in clinical ESCC tissues versus controls, and was connected to more advanced stages and the abundance of MDSCs. Silencing EVA1B notably attenuated proliferation of ESCC cells and tumor growth in syngeneic mouse models. Moreover, EVA1B suppression resulted in apoptosis and cell cycle arrest, and impaired ESCC cell aggressiveness. Among ESCC patients, EVA1B was strongly correlated to EMT pathway activity. Targeted suppression of EVA1B mitigated the expression of Wnt3a, β-catenin and LRP6 in ESCC cells and tumor xenografts. Additionally, inhibition of EVA1B attenuated the expansion and recruitment of MDSCs within the immune microenvironment based upon the reduction in the percentage of CD11b+Gr-1+ immunosuppressive MDSCs as well as the expression of MDSC expansion stimulators (S100A8, S100A9, Arg-1, and VEGF). Collectively, our findings unveiled the contribution of high expression of EVA1B to ESCC progression and MDSCs expansion and recruitment, indicating that targeted suppression of EVA1B may be a potential treatment choice for ESCC patients.
Collapse
Affiliation(s)
- Tian Xia
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| | - Yongkang Zhang
- Department of Thoracic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Haodong Peng
- Department of Thoracic Surgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, China.
| | - Xiangbo Jia
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| | - Dong Yang
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| | - Li Wei
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| | - Tian Li
- Tianjin Medical University, Tianjin 300102, China.
| | - Wenjian Yao
- Department of Thoracic Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan 450003, China.
| |
Collapse
|
3
|
Fan L, Lin Y, Fu Y, Wang J. Small cell lung cancer with liver metastases: from underlying mechanisms to treatment strategies. Cancer Metastasis Rev 2024; 44:5. [PMID: 39585433 DOI: 10.1007/s10555-024-10220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024]
Abstract
Small cell lung cancer (SCLC) represents an aggressive neuroendocrine (NE) tumor within the pulmonary region, characterized by very poor prognoses. Druggable targets for SCLC remain limited, thereby constraining treatment options available to patients. Immuno-chemotherapy has emerged as a pivotal therapeutic strategy for extensive-stage SCLC (ES-SCLC), yet it fails to confer significant efficacy in cases involving liver metastases (LMs) originating from SCLC. Therefore, our attention is directed towards the challenging subset of SCLC patients with LMs. Disease progression of LM-SCLC patients is affected by various factors in the tumor microenvironment (TME), including immune cells, blood vessels, inflammatory mediators, metabolites, and NE substances. Beyond standard immuno-chemotherapy, ongoing efforts to manage LMs in SCLC encompass anti-angiogenic therapy, radiotherapy, microwave ablation (MWA) / radiofrequency ablation (RFA), trans-arterial chemoembolization (TACE), and systemic therapies in conjunction with local interventions. Prospective experimental and clinical investigations into SCLC should prioritize precise and individualized approaches to enhance the prognosis across distinct patient cohorts.
Collapse
Affiliation(s)
- Linjie Fan
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yiwen Lin
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yunjie Fu
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
4
|
He Y, Qiu Y, Yang X, Lu G, Zhao SS. Remodeling of tumor microenvironment by cellular senescence and immunosenescence in cervical cancer. Semin Cancer Biol 2024; 108:17-32. [PMID: 39586414 DOI: 10.1016/j.semcancer.2024.11.002] [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: 09/06/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
Cellular senescence is a response to various stress signals, which is characterized by stable cell cycle arrest, alterations in cellular morphology, metabolic reprogramming and production of senescence-associated secretory phenotype (SASP). When it occurs in the immune system, it is called immunosenescence. Cervical cancer is a common gynecological malignancy, and cervical cancer screening is generally recommended before the age of 65. Elderly women (≥65 years) are more often diagnosed with advanced disease and have poorer prognosis compared to younger patients. Despite extensive research, the tumor microenvironment requires more in-depth exploration, particularly in elderly patients. In cervical cancer, senescent cells have a double-edged sword effect on tumor progression. Induction of preneoplastic cell senescence prevents tumor initiation, and several treatment approaches of cervical cancer act in part by inducing cancer cell senescence. However, senescent immune cell populations within the tumor microenvironment facilitate tumor development, recurrence, treatment resistance, etc. Amplification of beneficial effects and inhibition of aging-related pro-tumorigenic pathways contribute to improving antitumor effects. This review discusses senescent cancer and immune cells present in the tumor microenvironment of cervical cancer and how these senescent cells and their SASP remodel the tumor microenvironment, influence antitumor immunity and tumor initiation and development. Moreover, we discuss the significance of senotherapeutics that enable to eliminate senescent cells and prevent tumor progression and development through improving antitumor immunity and affecting the tumor microenvironment.
Collapse
Affiliation(s)
- Yijiang He
- Abdominal Radiation Oncology Ward II, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yue Qiu
- Department of Digestive Diseases 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Xiansong Yang
- Department of Day Chemotherapy Ward, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital), Qingdao, Shandong 266042, China
| | - Guimei Lu
- Department of Laboratory, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Shan-Shan Zhao
- Department of Gynecology Surgery 1, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| |
Collapse
|
5
|
Saqib M, Din ZS, Zafar S, Munawar N, Nawaz R, Ahmed S, Hamdard MH. Lung cancer, platinum analog-based frontline treatment and pharmacogenetic limitations. Per Med 2024; 21:385-400. [PMID: 39560009 DOI: 10.1080/17410541.2024.2391269] [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: 02/21/2024] [Accepted: 08/08/2024] [Indexed: 11/20/2024]
Abstract
Lung cancer has the highest mortality rate among all the highly prevalent neoplasia globally. The major concern with its frontline treatment-cisplatin, is the rapid progression of chemoresistance and multi-organ-based toxicities including hearing loss and tinnitus, nephrotoxicity, hepatotoxicity and myelosuppression including anemia and neutropenia. In this review, studies concluding the association of single nucleotide polymorphisms (SNP) in disparate genes with aforementioned toxicity points are summarized to observe the pharmacogenomic pattern. Especially, SNPs in ATP7B, ERCC-1, ERCC-2, MATE-1, OCT-2, ABCB-1, ABCC-1, ABCG-2, ABCC-2, SLC22A, ERCC-5, BRCA-1, GSTM-3, GSTM-4 and GSTM-5 genes appear to be associated with the therapeutic response and/or adverse effects of cisplatin. We recommend utilizing this information to minimize the risk of treatment failure due to chemoresistance and adverse effects on other organs.
Collapse
Affiliation(s)
- Maryam Saqib
- Shifa College of Medicine, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Zari Salahud Din
- Shifa College of Medicine, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Sehrish Zafar
- Combined Military Hospital College, Lahore, Pakistan
| | - Nayla Munawar
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| | - Rukhsana Nawaz
- Department of Clinical Psychology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain, 15551, United Arab Emirates
| | - Sagheer Ahmed
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | | |
Collapse
|
6
|
Jiang Y, Xie J, Cheng Q, Cai Z, Xu K, Lu W, Wang F, Wu X, Song Y, Lv T, Zhan P. Comprehensive genomic and spatial immune infiltration analysis of survival outliers in extensive-stage small cell lung cancer receiving first-line chemoimmunotherapy. Int Immunopharmacol 2024; 141:112901. [PMID: 39151386 DOI: 10.1016/j.intimp.2024.112901] [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: 04/26/2024] [Revised: 07/20/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND A minority of patients with extensive-stage small cell lung cancer (ES-SCLC) exhibit prolonged survival following first-line chemoimmunotherapy, which warrants the use of reliable biomarkers. Here, we investigated the disparities in genomics and immune cell spatial distribution between long- and short-term survival of patients with ES-SCLC. METHODS We retrospectively recruited 11 long-term (>2 years) and 13 short-term (<9 months) ES-SCLC survivors receiving first-line chemoimmunotherapy. The samples were processed using targeted next-generation sequencing (tNGS), programmed death ligand-1 staining, multiplex immunohistochemical staining for immune cells (mIHC), tumor mutation burden (TMB), and chromosomal instability score measurements. The expression of putative genes in SCLC at the bulk and single-cell RNA-sequencing levels, as well as the role of putative genes in pan-cancer immunotherapy cohorts, were analyzed. RESULTS At the genomic level, a greater proportion of the smoking signature and higher TMB (>3.1) were associated with favorable survival. At the single-gene and pathway levels, tNGS revealed that MCL1 and STMN1 amplification and alterations in the apoptosis pathway were more common in short-term survivors, whereas alterations in the DLL3, KMT2B, HGF, EPHA3, ADGRB3, lysine deprivation, and HGF-cMET pathways were observed more frequently in long-term survivors. mIHC analysis of immune cells with different spatial distributions revealed that long-term survivors presented increased numbers of M1-like macrophages in all locations and decreased numbers of CD8+ T cells in the tumor stroma. Bulk transcriptomic analysis demonstrated that high levels of STMN1 and DLL3 represented an immune-suppressive tumor immune microenvironment (TIME), whereas HGF indicated an immune-responsive TIME. The expression levels of our putative genes were comparative in both TP53/RB1 mutant-type and TP53/RB1 wild-type. At the single-cell level, STMN1, MCL1, and DLL3 were highly expressed among all molecular subtypes (SCLC-A, SCLC-N, and SCLC-P), with STMN1 being enriched in cell division and G2M checkpoint pathways. CONCLUSIONS For ES-SCLC patients receiving first-line chemoimmunotherapy, alterations in DLL3, KMT2B, HGF, EPHA3, and ADGRB3 and a greater proportion of M1-like macrophages infiltration in all locations were predictors of favorable survival, while MCL1 and STMN1 amplification, as well as a greater proportion of CD8+ T cells infiltrating the tumor stroma, predicted worse survival.
Collapse
Affiliation(s)
- Yuxin Jiang
- School of Medicine, Southeast University, Nanjing 210000, China
| | - Jingyuan Xie
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Qinpei Cheng
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Zijing Cai
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China
| | - Ke Xu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Wanjun Lu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Fufeng Wang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xiaoying Wu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Yong Song
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Tangfeng Lv
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Ping Zhan
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| |
Collapse
|
7
|
Helmy MW, Youssef MH, Yamari I, Amr A, Moussa FI, El Wakil A, Chtita S, El-Samad LM, Hassan MA. Repurposing of sericin combined with dactolisib or vitamin D to combat non-small lung cancer cells through computational and biological investigations. Sci Rep 2024; 14:27034. [PMID: 39505930 PMCID: PMC11541877 DOI: 10.1038/s41598-024-76947-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
This study aims to repurpose sericin in combating non-small lung cancer cells (A549 and H460) by combining it with dactolisib or vitamin D to reduce the dose of dactolisib and boost the anticancer effectiveness of dactolisib and vitamin D. Therefore, the binding affinities of individual and combined drugs were examined using in silico and protein-protein interaction studies, targeting NF-κB, Cyclin D1, p-AKT, and VEGF1 proteins. The findings manifested remarkable affinities for combinatorial drugs compared to individual compounds. To substantiate these findings, the combined IC50 for each combination (sericin + dactolisib and sericin + vitamin D) were determined, reporting 31.9 and 41.8 µg/ml, respectively, against A549 cells and 47.9 and 55.3 µg/ml, respectively, against H460 cells. Furthermore, combination indices were assessed to lower the doses of each drug. Interestingly, in vitro results exhibited marked diminutions in NF-κB, Cyclin D1, p-AKT, and VEGF1 after treatment with sericin + dactolisib and sericin + vitamin D compared to control lung cancer cells and those treated with a single drug. Moreover, A549 and H460 cells treated with both combinations demonstrated augmented caspase-3 levels, implying substantial apoptotic activity. Altogether, these results accentuated the prospective implementation of sericin in combination with dactolisib and vitamin D at low doses to preclude lung cancer cell proliferation.
Collapse
Affiliation(s)
- Maged W Helmy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Damanhour University, 22511, Damanhour, Egypt
| | - Mariam H Youssef
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Imane Yamari
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, P. O. Box 7955, Casablanca, Morocco
| | - Alaa Amr
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Farouzia I Moussa
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Abeer El Wakil
- Department of Biological and Geological Sciences, Faculty of Education, Alexandria University, Alexandria, Egypt
| | - Samir Chtita
- Laboratory of Analytical and Molecular Chemistry, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, P. O. Box 7955, Casablanca, Morocco
| | - Lamia M El-Samad
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt.
| | - Mohamed A Hassan
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, 21934, Alexandria, Egypt.
| |
Collapse
|
8
|
Miao S, Cheng Y, Li Y, Chen X, Chen F, Zha D, Xue T. Prediction of recurrence-free survival and risk factors of sinonasal inverted papilloma after surgery by machine learning models. Eur J Med Res 2024; 29:528. [PMID: 39497172 PMCID: PMC11533278 DOI: 10.1186/s40001-024-02099-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/08/2024] [Indexed: 11/06/2024] Open
Abstract
OBJECTIVES Our research aims to construct machine learning prediction models to identify patients proned to recurrence after inverted papilloma (IP) surgery and guide their follow-up treatment. METHODS This study collected 210 patients underwent IP resection surgery at a university hospital from January 2010 to December 2023. Six machine learning algorithms including ExtraSurvivalTrees (EST), GradientBoostingSurvivalAnalysis (GBSA), RandomSurvivalForest (RSF), SurvivalSVM, Coxnet and Coxph, were used to construct the prediction models. Shapley Additive Explanations (SHAP) values were used to explain the importance of various features in predicting IP recurrence. RESULTS We found that the recurrence rate of IP patients is 20.00%, with a median recurrence time of 35.5 months. Multivariate Cox regression analysis identified mild or moderate dysplasia as an independent risk factor for recurrence. The EST model performs the best in predicting postoperative recurrence of IP, with C-index of 0.968 and 0.878 in the training and testing sets. SHAP emphasizes five important predictive factors for recurrence, including bone defects, orbital involvement, smoking, no processing of tumor attachment sites and drinking. CONCLUSIONS To our knowledge, this is the first study to use multiple ML models to predict postoperative recurrence of IP. The EST model has the best predictive performance, with SHAP emphasizing several key predictive factors for IP recurrence. This study emphasizes the practicality of machine learning algorithms in predicting IP clinical outcomes, providing valuable insights into the potential for improving clinical decision-making.
Collapse
Affiliation(s)
- Siyu Miao
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, The Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
- The Outpatient Department, Lintong Rehabilitation and Convalescent Centre, Xi'an, 710600, China
| | - Yang Cheng
- Department of Endocrinology, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Yaqi Li
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, The Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Xiaodong Chen
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, The Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Fuquan Chen
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, The Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China
| | - Dingjun Zha
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, The Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China.
| | - Tao Xue
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, The Air Force Medical University, 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
9
|
Gu M, Liu Y, Zheng W, Jing Z, Li X, Guo W, Zhao Z, Yang X, Liu Z, Zhu X, Gao W. Combined targeting of senescent cells and senescent macrophages: A new idea for integrated treatment of lung cancer. Semin Cancer Biol 2024; 106-107:43-57. [PMID: 39214157 DOI: 10.1016/j.semcancer.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Lung cancer is one of the most common cancers worldwide and a leading cause of cancer-related deaths. Macrophages play a key role in the immune response and the tumour microenvironment. As an important member of the immune system, macrophages have multiple functions, including phagocytosis and clearance of pathogens, modulation of inflammatory responses, and participation in tissue repair and regeneration. In lung cancer, macrophages are considered to be the major cellular component of the tumor-associated inflammatory response and are closely associated with tumorigenesis, progression and metastasis. However, macrophages gradually undergo a senescence process with age and changes in pathological states. Macrophage senescence is an important change in the functional and metabolic state of macrophages and may have a significant impact on lung cancer development. In lung cancer, senescent macrophages interact with other cells in the tumor microenvironment (TME) by secreting senescence-associated secretory phenotype (SASP) factors, which can either promote the proliferation, invasion and metastasis of tumor cells or exert anti-tumor effects through reprogramming or clearance under specific conditions. Therefore, senescent macrophages are considered important potential targets for lung cancer therapy. In this paper, a systematic review of macrophages and their senescence process, and their role in tumors is presented. A variety of inhibitory strategies against senescent macrophages, including enhancing autophagy, inhibiting SASP, reducing DNA damage, and modulating metabolic pathways, were also explored. These strategies are expected to improve lung cancer treatment outcomes by restoring the anti-tumor function of macrophages.
Collapse
Affiliation(s)
- Ming Gu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yang Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Wenhui Zheng
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zuoqian Jing
- Department of Ophthalmology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xiang Li
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zimo Zhao
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Xu Yang
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zhe Liu
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Xinwang Zhu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Wei Gao
- Department of Gastrointestinal Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
10
|
Yin L, Sun P, Guo S, Shuai P, Zhang J. CAR-T cell therapy: Challenge and opportunity for effective treatment of small cell lung cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189228. [PMID: 39615863 DOI: 10.1016/j.bbcan.2024.189228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 12/08/2024]
Abstract
Small cell lung cancer (SCLC) is a devastating malignancy characterized by rapid metastasis, drug resistance, and frequent recurrence. Owing to the paucity of existing therapeutic options, the prognosis of SCLC remains poor. Recently, the combination of immune checkpoint inhibitors and chemotherapy has resulted in modest improvements in treatment responses. In this review, we characterize the biological signature of SCLC and outline the obstacles to current treatment, including impaired antigen presentation and T cell infiltration. These obstacles may potentially be overcome by chimeric antigen receptor (CAR)-T cell therapy. For the first time, we summarize the available data and discuss the future prospects of CAR-T cell therapy for the treatment of SCLC. Given the high heterogeneity and immunosuppressive tumor microenvironment of SCLC, structural modifications of CAR-T cells and combination therapy may be required to elicit a successful antitumor response. Further research, including clinical trials, is needed to determine the suitability of CAR-T cell therapy as a treatment for SCLC.
Collapse
Affiliation(s)
- Limei Yin
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ping Sun
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shujin Guo
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ping Shuai
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Junlin Zhang
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
11
|
Qi Y, Li T, Zhou Y, Hao Y, Zhang J. RNA modification regulators as promising biomarkers in gynecological cancers. Cell Biol Toxicol 2024; 40:92. [PMID: 39472384 PMCID: PMC11522084 DOI: 10.1007/s10565-024-09924-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/02/2024] [Indexed: 11/02/2024]
Abstract
This review explores the evolving landscape of gynecological oncology by focusing on emerging RNA modification signatures as promising biomarkers for assessing the risk and progression of ovarian, cervical, and uterine cancers. It provides a comprehensive overview of common RNA modifications, especially m6A, and their roles in cellular processes, emphasizing their implications in gynecological cancer development. The review meticulously examines specific m6A regulators including "writers", "readers", and "erasers" associated with three gynecological cancer types, discussing their involvement in initiation and progression. Methodologies for detecting RNA modifications are surveyed, highlighting advancements in high-throughput techniques with high sensitivity. A critical analysis of studies identifying m6A regulators as potential biomarkers is presented, addressing their diagnostic or prognostic significance. Mechanistic insights into RNA modification-mediated cancer progression are explored, shedding light on molecular pathways and potential therapeutic targets. Despite current challenges, the review discusses ongoing research efforts, future directions, and the transformative possibility of RNA modifications on early assessment and personalized therapy in gynecological oncology.
Collapse
Affiliation(s)
- Yue Qi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, Liaoning, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, Shanxi, China.
| | - Yang Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, Liaoning, China
| | - Yingying Hao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, Liaoning, China.
| | - Jin Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
12
|
Almarza C, Villalobos-Nova K, Toro MA, González M, Niechi I, Brown-Brown DA, López-Muñoz RA, Silva-Pavez E, Gaete-Ramírez B, Varas-Godoy M, Burzio VA, Jara L, Aguayo F, Tapia JC. Cisplatin-resistance and aggressiveness are enhanced by a highly stable endothelin-converting enzyme-1c in lung cancer cells. Biol Res 2024; 57:74. [PMID: 39443981 PMCID: PMC11515556 DOI: 10.1186/s40659-024-00551-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Lung cancer constitutes the leading cause of cancer mortality. High levels of endothelin-1 (ET-1), its cognate receptor ETAR and its activating enzyme, the endothelin-converting enzyme-1 (ECE-1), have been reported in several cancer types, including lung cancer. ECE-1 comprises four isoforms, which only differ in their cytoplasmic N-terminus. Protein kinase CK2 phosphorylates the N-terminus of isoform ECE-1c, increasing its stability and leading to enhanced invasiveness in glioblastoma and colorectal cancer cells, which is believed to be mediated by the amino acid residue Lys-6, a conserved putative ubiquitination site neighboring the CK2-phosphorylated residues Ser-18 and Ser-20. Whether Lys-6 is linked to the acquisition of a cancer stem cell (CSC)-like phenotype and aggressiveness in human non-small cell lung cancer (NSCLC) cells has not been studied. METHODS In order to establish the role of Lys-6 in the stability of ECE-1c and its involvement in lung cancer aggressiveness, we mutated this residue to a non-ubiquitinable arginine and constitutively expressed the wild-type (ECE-1cWT) and mutant (ECE-1cK6R) proteins in A549 and H1299 human NSCLC cells by lentiviral transduction. We determined the protein stability of these clones alone or in the presence of the CK2 inhibitor silmitasertib, compared to ECE-1cWT and mock-transduced cells. In addition, the concentration of secreted ET-1 in the growth media was determined by ELISA. Expression of stemness genes were determined by Western blot and RT-qPCR. Chemoresistance to cisplatin was studied by MTS viability assay. Migration and invasion were measured through transwell and Matrigel assays, respectively, and the side-population was determined using flow cytometry. RESULTS ECE-1cK6R displayed higher stability in NSCLC cells compared to ECE-1cWT-expressing cells, but ET-1 secreted levels showed no difference up to 48 h. Most importantly, ECE-1cK6R promoted expression of the stemness genes c-Myc, Sox-2, Oct-4, CD44 and CD133, which enhance cellular self-renewal capability. Also, the ECE-1cK6R-expressing cells showed higher cisplatin chemoresistance, correlating with an augmented side-population abundance due to the increased expression of the ABCG2 efflux pump. Finally, the ECE-1cK6R-expressing cells showed enhanced invasiveness, which correlated with the regulated expression of known EMT markers. CONCLUSIONS Our findings suggest an important role of ECE-1c in lung cancer. ECE-1c is key in a non-canonical ET-1-independent mechanism which triggers a CSC-like phenotype, leading to enhanced lung cancer aggressiveness. Underlying this mechanism, ECE-1c is stabilized upon phosphorylation by CK2, which is upregulated in many cancers. Thus, phospho-ECE-1c may be considered as a novel prognostic biomarker of recurrence, as well as the CK2 inhibitor silmitasertib as a potential therapy for lung cancer patients.
Collapse
Affiliation(s)
- Cristopher Almarza
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Karla Villalobos-Nova
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - María A Toro
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Manuel González
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ignacio Niechi
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - David A Brown-Brown
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Rodrigo A López-Muñoz
- Instituto de Farmacología y Morfofisiología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Eduardo Silva-Pavez
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago, Chile
| | - Belén Gaete-Ramírez
- Centro de Biología Celular y Biomedicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Manuel Varas-Godoy
- Centro de Biología Celular y Biomedicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Verónica A Burzio
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Facultad de Medicina, Universidad Andrés Bello, Santiago, Chile
| | - Lilian Jara
- Programa de Genética, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Francisco Aguayo
- Departamento de Biomedicina, Facultad de Medicina, Universidad de Tarapacá, Arica, Chile
| | - Julio C Tapia
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile.
- Laboratorio de Transformación Celular, Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Av. Independencia 1027, Santiago, 8380453, Chile.
| |
Collapse
|
13
|
Zhou R, Zhang K, Dai T, Guo Z, Li T, Hong X. Construction and validation of cell cycle-related prognostic genetic model for glioblastoma. Medicine (Baltimore) 2024; 103:e39205. [PMID: 39465756 PMCID: PMC11460857 DOI: 10.1097/md.0000000000039205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Indexed: 10/29/2024] Open
Abstract
Glioblastoma (GBM) is a common primary malignant brain tumor and the prognosis of these patients remains poor. Therefore, further understanding of cell cycle-related molecular mechanisms of GBM and identification of appropriate prognostic markers and therapeutic targets are key research imperatives. Based on RNA-seq expression datasets from The Cancer Genome Atlas database, prognosis-related biological processes in GBM were screened out. Gene Set Variation Analysis (GSVA), LASSO-COX, univariate and multivariate Cox regression analyses, Kaplan-Meier survival analysis, and Pearson correlation analysis were performed for constructing a predictive prognostic model. A total of 58 cell cycle-related genes were identified by GSVA and analysis of differential expression between GBM and control samples. By univariate Cox and LASSO regression analyses, 8 genes were identified as prognostic biomarkers in GBM. A nomogram with superior performance to predict the survival of GBM patients was established regarding risk score, cancer status, recurrence type, and mRNAsi. This study revealed the prognostic value of cell cycle-related genes in GBM. In addition, we constructed a reliable model for predicting the prognosis of GBM patients. Our findings reinforce the relationship between cell cycle and GBM and may help improve the prognostic assessment of patients with GBM. Our predictive prognostic model, based on independent prognostic factors, enables tailored treatment strategies for GBM patients. It is particularly useful for subgroups with uncertain prognosis or treatment challenges.
Collapse
Affiliation(s)
- Runpeng Zhou
- Department of Neurosurgery, Pu’er People’s Hospital, Pu’er, China
| | - Kai Zhang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Tingting Dai
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zeshang Guo
- Department of Neurosurgery, The First Bethune Hospital of Jilin University, Changchun, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Xinyu Hong
- Department of Neurosurgery, The First Bethune Hospital of Jilin University, Changchun, China
| |
Collapse
|
14
|
Shen S, Hong Y, Huang J, Qu X, Sooranna SR, Lu S, Li T, Niu B. Targeting PD-1/PD-L1 in tumor immunotherapy: Mechanisms and interactions with host growth regulatory pathways. Cytokine Growth Factor Rev 2024; 79:16-28. [PMID: 39179486 DOI: 10.1016/j.cytogfr.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024]
Abstract
Tumor immunotherapy has garnered considerable attention, emerging as a new standard of care in cancer treatment. The conventional targets, such as VEGF and EGFR, have been extended to others including BRAF and PD-1/PD-L1, which have shown significant potential in recent cancer treatments. This review aims to succinctly overview the impact and mechanisms of therapies that modulate PD-1/PD-L1 expression by targeting VEGF, EGFR, LAG-3, CTLA-4 and BRAF. We investigated how modulation of PD-1/PD-L1 expression impacts growth factor signaling, shedding light on the interplay between immunomodulatory pathways and growth factor networks within the tumor microenvironment. By elucidating these interactions, we aim to provide insights into novel potential synergistic therapeutic strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Songyu Shen
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Yihan Hong
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Jiajun Huang
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Xiaosheng Qu
- Guangxi Botanical Garden of Medicinal Plants, Nanning, Guangxi 530023, China
| | - Suren Rao Sooranna
- Department of Metabolism, Digestion and Reproduction, Imperial College London, 369 Fulham Road, London SW10 9NH, United Kingdom
| | - Sheng Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, 169 Changle West Rd, Xi'an 710032, China.
| | - Bing Niu
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China.
| |
Collapse
|
15
|
Yu W, Xing Y, Song X, Li T, Zhang M. EGFR-Tyrosine Kinase Inhibitor Combined with Radiotherapy in 105 Patients of Lung Adenocarcinoma with Brain Metastasis: A Retrospective Study of Prognostic Factor Analysis. Oncol Res Treat 2024; 47:531-548. [PMID: 39293411 DOI: 10.1159/000541494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 09/08/2024] [Indexed: 09/20/2024]
Abstract
INTRODUCTION This study aimed to retrospectively analyse the response and prognosis factors for patients with lung adenocarcinoma exhibiting brain metastasis and epidermal growth factor receptor (EGFR) mutations, who were treated with a combination of EGFR-tyrosine kinase inhibitor (TKI) and brain radiotherapy (RT). METHODS Clinicopathological data of patients with lung adenocarcinoma were collected from January 2021 to January 2024 at the First Affiliated Hospital of Hebei North University. Statistical analysis was performed using SPSS version 26.0, with significance set at p < 0.05. RESULTS A total of 105 patients were included. The overall survival (OS) rates at 1, 2, and 3 years were 82.9%, 61.2%, and 33.7%, respectively. The progression-free survival 1 (PFS1) rates at 1, 2, and 3 years were 62.7%, 36.6%, and 22.1%, respectively. The progression-free survival 2 (PFS2) rates at 1, 2, and 3 years were 80.8%, 54.6%, and 31.4%, respectively. The median OS, PFS1, and PFS2 were 29.8, 18.0, and 28.1 months, respectively. Cox multivariate analysis identified gene mutation status and brain radiation dose as independent prognostic factors for OS. For PFS1, gene mutation status, brain radiation dose, and initial treatment response were independent prognostic factors. Clinical stage, gene mutation status, brain radiation dose, and initial treatment response were independent prognostic factors for PFS2. CONCLUSION The combination of TKIs and brain RT is effective for patients with lung adenocarcinoma with EGFR mutations and brain metastases. Patients with exon 19 Del or exon 21 L858R mutations and brain radiation doses ≥40 Gy exhibit longer OS, PFS1, and PFS2. Additionally, complete remission + partial remission is associated with extended PFS1 and PFS2, while patients in stage IVA show longer PFS2.
Collapse
Affiliation(s)
- Wenjuan Yu
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yuan Xing
- Department of Pharmacy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Xiao Song
- Department of Radiotherapy, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Mi Zhang
- Department of Respiratory, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Liu Y, Yang H, Li T, Zhang N. Immunotherapy in liver cancer: overcoming the tolerogenic liver microenvironment. Front Immunol 2024; 15:1460282. [PMID: 39295859 PMCID: PMC11409253 DOI: 10.3389/fimmu.2024.1460282] [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: 07/05/2024] [Accepted: 08/21/2024] [Indexed: 09/21/2024] Open
Abstract
Liver cancer is a major global health concern, ranking among the top causes of cancer-related deaths worldwide. Despite advances in medical research, the prognosis for liver cancer remains poor, largely due to the inherent limitations of current therapies. Traditional treatments like surgery, radiation, and chemotherapy often fail to provide long-term remission and are associated with significant side effects. Immunotherapy has emerged as a promising avenue for cancer treatment, leveraging the body's immune system to target and destroy cancer cells. However, its application in liver cancer has been limited. One of the primary challenges is the liver's unique immune microenvironment, which can inhibit the effectiveness of immunotherapeutic agents. This immune microenvironment creates a barrier, leading to drug resistance and reducing the overall efficacy of treatment. Recent studies have focused on understanding the immunological landscape of liver cancer to develop strategies that can overcome these obstacles. By identifying the specific factors within the liver that contribute to immune suppression and drug resistance, researchers aim to enhance the effectiveness of immunotherapy. Prospective strategies include combining immunotherapy with other treatments, using targeted therapies to modulate the immune microenvironment, and developing new agents that can bypass or counteract the inhibitory mechanisms in the liver. These advancements hold promise for improving outcomes in liver cancer treatment.
Collapse
Affiliation(s)
- Yanju Liu
- Department of Infectious Diseases, Weifang People's Hospital, Weifang, Shandong, China
| | - Hongyuan Yang
- Department of Infectious Diseases, Weifang People's Hospital, Weifang, Shandong, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Na Zhang
- Department of Infectious Diseases, Weifang People's Hospital, Weifang, Shandong, China
| |
Collapse
|
17
|
Xu Y, He C, Xi Y, Zhang Y, Bai Y. Gut microbiota and immunosenescence in cancer. Semin Cancer Biol 2024; 104-105:32-45. [PMID: 39127266 DOI: 10.1016/j.semcancer.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
Cancer is generally defined as a disease of aging. With aging, the composition, diversity and functional characteristics of the gut microbiota occur changes, with a decline of beneficial commensal microbes triggered by intrinsic and extrinsic factors (e.g., diet, drugs and chronic health conditions). Nowadays, dysbiosis of the gut microbiota is recognized as a hallmark of cancer. At the same time, aging is accompanied by changes in innate and adaptive immunity, known as immunosenescence, as well as chronic low-grade inflammation, known as inflammaging. The elevated cancer incidence and mortality in the elderly are linked with aging-associated alterations in the gut microbiota that elicit systemic metabolic alterations, leading to immune dysregulation with potentially tumorigenic effects. The gut microbiota and immunosenescence might both affect the response to treatment in cancer patients. In-depth understanding of age-associated alterations in the gut microbiota and immunity will shed light on the risk of cancer development and progression in the elderly. Here, we describe the aging-associated changes of the gut microbiota in cancer, and review the evolving understanding of the gut microbiota-targeted intervention strategies. Furthermore, we summarize the knowledge on the cellular and molecular mechanisms of immunosenescence and its impact on cancer. Finally, we discuss the latest knowledge about the relationships between gut microbiota and immunosenescence, with implications for cancer therapy. Intervention strategies targeting the gut microbiota may attenuate inflammaging and rejuvenate immune function to provide antitumor benefits in elderly patients.
Collapse
Affiliation(s)
- Yaozheng Xu
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110136, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning 110136, China.
| | - Chuan He
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Ying Xi
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110136, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning 110136, China.
| | - Yue Zhang
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110136, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning 110136, China.
| | - Yibo Bai
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110136, China; Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning 110136, China.
| |
Collapse
|
18
|
Zhao J, Wang Z, Tian Y, Ning J, Ye H. T cell exhaustion and senescence for ovarian cancer immunotherapy. Semin Cancer Biol 2024; 104-105:1-15. [PMID: 39032717 DOI: 10.1016/j.semcancer.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/30/2024] [Accepted: 07/09/2024] [Indexed: 07/23/2024]
Abstract
Ovarian cancer is a common gynecological malignancy, and its treatment remains challenging. Although ovarian cancer may respond to immunotherapy because of endogenous immunity at the molecular or T cell level, immunotherapy has so far not had the desired effect. The functional status of preexisting T cells is an indispensable determinant of powerful antitumor immunity and immunotherapy. T cell exhaustion and senescence are two crucial states of T cell dysfunction, which share some overlapping phenotypic and functional features, but each status possesses unique molecular and developmental signatures. It has been widely accepted that exhaustion and senescence of T cells are important strategies for cancer cells to evade immunosurveillance and maintain the immunosuppressive microenvironment. Herein, this review summarizes the phenotypic and functional features of exhaust and senescent T cells, and describes the key drivers of the two T cell dysfunctional states in the tumor microenvironment and their functional roles in ovarian cancer. Furthermore, we present a summary of the molecular machinery and signaling pathways governing T cell exhaustion and senescence. Possible strategies that can prevent and/or reverse T cell dysfunction are also explored. An in-depth understanding of exhausted and senescent T cells will provide novel strategies to enhance immunotherapy of ovarian cancer through redirecting tumor-specific T cells away from a dysfunctional developmental trajectory.
Collapse
Affiliation(s)
- Jiao Zhao
- Department of Gynecology Surgery 3, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Zhongmiao Wang
- Department of Digestive Diseases 1, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Yingying Tian
- Department of Oncology Radiotherapy 2, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao, Shandong 266042, China
| | - Jing Ning
- Department of General Internal Medicine (VIP Ward), Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| | - Huinan Ye
- Department of Digestive Diseases 1, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China.
| |
Collapse
|
19
|
Li Q, Zhang K, Liu H, Zhai S, Jia Y, Li T, Pan Y. Clinical effects of neuroendoscopic infratentorial supracerebellar approach surgical technique for resecting pineal tumors: a retrospective study. Neurochirurgie 2024; 70:101576. [PMID: 38908132 DOI: 10.1016/j.neuchi.2024.101576] [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: 04/13/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/24/2024]
Abstract
OBJECTIVE Pineal tumors are relatively rare central nervous system lesions with a predilection for the pediatric population. This article aims to explore the clinical effects of neuroendoscopic infratentorial supracerebellar approach for resecting tumors in the pineal area. METHODS This is a retrospective study that included patients who underwent neuroendoscopic infratentorial supracerebellar approach to resect nine tumors in the pineal area at the Department of Neurosurgery of the Second Hospital of Lanzhou University from December 2017 to October 2023. RESULTS The results of postoperative MRI revealed that all tumors were resected. Five patients received postoperative radiotherapy, three patients received radiotherapy along with chemotherapy, and one patient received neither radiotherapy nor chemotherapy. The pathological results showed that four patients were diagnosed with germinoma, two patients with teratoma, two patients with mixed germ cell tumors, and one patient with central neurocytoma. After surgery, one patient developed psychiatric symptoms, two patients developed binocular upward vision and diplopia, and one patient developed unstable walking and diplopia. With a follow-up of 1.7-4.8 years, all nine patients lived normally. Furthermore, none of them had tumor recurrence or death. CONCLUSION The simple neuroendoscopic infratentorial supracerebellar approach has some safety and efficacy. It is suitable for tumors in the pineal region where the disease is mainly located below the Galen vein complex.
Collapse
Affiliation(s)
- Qiang Li
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Kai Zhang
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Hanruo Liu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Shijia Zhai
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou 730030, China.
| | - Yanfei Jia
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou 730030, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Yawen Pan
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
20
|
Ying Q, Fan R, Shen Y, Chen B, Zhang J, Li Q, Shi X. Small Cell Lung Cancer-An Update on Chemotherapy Resistance. Curr Treat Options Oncol 2024; 25:1112-1123. [PMID: 39066852 DOI: 10.1007/s11864-024-01245-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] [Accepted: 07/03/2024] [Indexed: 07/30/2024]
Abstract
OPINION STATEMENT Compared to other types of lung cancer, small cell lung cancer (SCLC) exhibits aggressive characteristics that promote drug resistance. Despite platinum-etoposide chemotherapy combined with immunotherapy being the current standard treatment, the rapid development of drug resistance has led to unsatisfactory clinical outcomes. This review focuses on the mechanisms contributing to the chemotherapy resistance phenotype in SCLC, such as increased intra-tumoral heterogeneity, alterations in the tumor microenvironment, changes in cellular metabolism, and dysregulation of apoptotic pathways. A comprehensive understanding of these drug resistance mechanisms in SCLC is imperative for ushering in a new era in cancer research, which will promise revolutionary advancements in cancer diagnosis and treatment methodologies.
Collapse
Affiliation(s)
- Qian Ying
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China
| | - Ruiyun Fan
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China
- Department of Respiratory Medicine, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China
| | - Yili Shen
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China
- Department of Respiratory Medicine, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China
| | - Boyi Chen
- Department of Respiratory Medicine, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, People's Republic of China
| | - Jianhui Zhang
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China
| | - Qiuhui Li
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China.
- Department of Respiratory Medicine, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China.
| | - Xuefei Shi
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, People's Republic of China.
- Department of Respiratory Medicine, Fifth School of Clinical Medicine of Zhejiang, Huzhou Central Hospital, Chinese Medical University, Huzhou, People's Republic of China.
| |
Collapse
|
21
|
Tian Y, Xin S, Wan Z, Dong H, Liu L, Fan Z, Li T, Peng F, Xiong Y, Han Y. TCF19 promotes cell proliferation and tumor formation in lung cancer by activating the Raf/MEK/ERK signaling pathway. Transl Oncol 2024; 45:101978. [PMID: 38701650 PMCID: PMC11088346 DOI: 10.1016/j.tranon.2024.101978] [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: 01/26/2024] [Revised: 04/09/2024] [Accepted: 04/27/2024] [Indexed: 05/05/2024] Open
Abstract
OBJECTIVE This study aimed to investigate TCF19's role in lung cancer development, specifically its involvement in the RAF/MEK/ERK signaling pathway. METHODS Lung cancer tissue analysis revealed significant TCF19 overexpression. In vitro experiments using A549 and Hop62 cells with TCF19 overexpression demonstrated enhanced cell growth. Transgenic mouse models confirmed TCF19's role in primary tumor development. Transcriptome sequencing identified altered gene expression profiles, linking TCF19 to RAF/MEK/ERK pathway activation. Functional assays elucidated underlying mechanisms, revealing increased phosphorylation of Raf1, MEK1/2, and ERK1/2. Inhibiting RAF1 or ERK through shRaf1 or ERK inhibitor reduced cell cycle-related proteins and inhibited TCF19-overexpressing cell growth. RESULTS TCF19 was identified as an oncogene in lung carcinoma, specifically impacting the RAF/MEK/ERK pathway. Elevated TCF19 levels in lung cancer suggest targeting TCF19 or its associated pathways as a promising strategy for disease management. CONCLUSION This study unveils TCF19's oncogenic role in lung cancer, emphasizing its modulation of the RAF/MEK/ERK pathway and presenting a potential therapeutic target for TCF19-overexpressing lung cancers.
Collapse
Affiliation(s)
- Yahui Tian
- Department of Thoracic Surgery, Air Force Medical Center, Air Force Medical University, 30 Fucheng Rd, Beijing 100142, China; School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Shaowei Xin
- Department of Thoracic Surgery, Air Force Medical Center, Air Force Medical University, 30 Fucheng Rd, Beijing 100142, China
| | - Zitong Wan
- College of life Science, Northwestern University, Xi'an, China
| | - Honghong Dong
- Department of Thoracic Surgery, Air Force Medical Center, Air Force Medical University, 30 Fucheng Rd, Beijing 100142, China
| | - Lu Liu
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhenzhen Fan
- CAS Key Laboratory of Genomics and Precision Medicine, Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Fujun Peng
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China.
| | - Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Yong Han
- Department of Thoracic Surgery, Air Force Medical Center, Air Force Medical University, 30 Fucheng Rd, Beijing 100142, China.
| |
Collapse
|
22
|
Wu X, Fan J, Zhang X, Li T, Song J. Global trends of single cell sequence associated in cancer from 2011 to 2024: A bibliometric analysis. Heliyon 2024; 10:e32847. [PMID: 38975217 PMCID: PMC11226897 DOI: 10.1016/j.heliyon.2024.e32847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
Objective Exploring the different molecular and clinicopathological features of nodal cancer based on single cell sequencing can reveal the intertumoral heterogeneity in cancer, and provide new ideas for early diagnosis, treatment and prognosis analysis of cancer. Methods The hotspots, the features of worldwide scientific output, and the frontiers concerning single cell sequence related to cancer from 2011 to 2024 were determined using our bibliometric analysis. Web of Science Core Collection (WOSCC) database was searched for publications on single cell sequence associated with cancer that were published between 2011 and 2024. According to the journals, keywords, number of records, affiliations, citations, and countries, we conducted a bibliometric analysis. With the use of the data gathered from the WOSCC, geographic distribution was visualized, keyword, affiliation, and author cluster analyses were conducted, and co-cited references were reviewed and a descriptive analysis was also performed. Results From the analysis, it was concluded that 6189 articles that were published between 2011 and 2024 in total were identified. Frontiers in immunology is the leading journal with the most publications in field of the research. The five clusters that were identified for hotspots included immunotherapy, single-cell RNA sequencing, hepatocellular carcinoma, proliferation, gene expression appeared the most frequently. Journals, nations, organizations, scholars with most contribution and most referenced publications globally were extracted. Studies have mostly concentrated on the spatial transcriptomics, pan-cancer analysis, hepatocellular carcinoma et al. Conclusion Single-cell sequencing plays a significant role in tumor diagnosis, treatment and prognosis.
Collapse
Affiliation(s)
- Xueliang Wu
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, 075000, China
- Institute of Cancer, The First Affiliated Hospital of Hebei North University, 075000, China
| | - Jianchun Fan
- Department of General Surgery, The First Affiliated Hospital of Hebei North University, 075000, China
| | - Xingmei Zhang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610000, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Jichao Song
- Department of Gynaecology, Xinchang Country People's Hospital/Xinchang County Maternal and Child Health Hospital, 117 Gushan Middle Road, Xinchang, 312500, Zhejiang Province, China
| |
Collapse
|
23
|
Wang Y, Zhou C, Li T, Luo J. Prognostic value of CDKN2A in head and neck squamous cell carcinoma via pathomics and machine learning. J Cell Mol Med 2024; 28:e18394. [PMID: 38751024 PMCID: PMC11096642 DOI: 10.1111/jcmm.18394] [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: 03/10/2024] [Revised: 04/11/2024] [Accepted: 04/27/2024] [Indexed: 05/19/2024] Open
Abstract
This study aims to enhance the prognosis prediction of Head and Neck Squamous Cell Carcinoma (HNSCC) by employing artificial intelligence (AI) to analyse CDKN2A gene expression from pathology images, directly correlating with patient outcomes. Our approach introduces a novel AI-driven pathomics framework, delineating a more precise relationship between CDKN2A expression and survival rates compared to previous studies. Utilizing 475 HNSCC cases from the TCGA database, we stratified patients into high-risk and low-risk groups based on CDKN2A expression thresholds. Through pathomics analysis of 271 cases with available slides, we extracted 465 distinctive features to construct a Gradient Boosting Machine (GBM) model. This model was then employed to compute Pathomics scores (PS), predicting CDKN2A expression levels with validation for accuracy and pathway association analysis. Our study demonstrates a significant correlation between higher CDKN2A expression and improved median overall survival (66.73 months for high expression vs. 42.97 months for low expression, p = 0.013), establishing CDKN2A's prognostic value. The pathomic model exhibited exceptional predictive accuracy (training AUC: 0.806; validation AUC: 0.710) and identified a strong link between higher Pathomics scores and cell cycle activation pathways. Validation through tissue microarray corroborated the predictive capacity of our model. Confirming CDKN2A as a crucial prognostic marker in HNSCC, this study advances the existing literature by implementing an AI-driven pathomics analysis for gene expression evaluation. This innovative methodology offers a cost-efficient and non-invasive alternative to traditional diagnostic procedures, potentially revolutionizing personalized medicine in oncology.
Collapse
Affiliation(s)
- Yandan Wang
- Department of Otolaryngology, Huaihe HospitalHenan UniversityKaifengChina
| | - Chaoqun Zhou
- Department of Pathology, Huaihe HospitalHenan UniversityKaifengChina
| | - Tian Li
- School of Basic MedicineFourth Military Medical UniversityXi'anChina
| | - Junpeng Luo
- Translational Medical Center of Huaihe HospitalHenan UniversityKaifengChina
| |
Collapse
|
24
|
Yuan F, Hu Y, Xu F, Feng X. A review of obstructive sleep apnea and lung cancer: epidemiology, pathogenesis, and therapeutic options. Front Immunol 2024; 15:1374236. [PMID: 38605948 PMCID: PMC11007033 DOI: 10.3389/fimmu.2024.1374236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
Despite undeniable advances in modern medicine, lung cancer still has high morbidity and mortality rates. Lung cancer is preventable and treatable, and it is important to identify new risk factors for lung cancer, especially those that can be treated or reversed. Obstructive sleep apnea (OSA) is a very common sleep-breathing disorder that is grossly underestimated in clinical practice. It can cause, exacerbate, and worsen adverse outcomes, including death and various diseases, but its relationship with lung cancer is unclear. A possible causal relationship between OSA and the onset and progression of lung cancer has been established biologically. The pathophysiological processes associated with OSA, such as sleep fragmentation, intermittent hypoxia, and increased sympathetic nervous excitation, may affect normal neuroendocrine regulation, impair immune function (especially innate and cellular immunity), and ultimately contribute to the occurrence of lung cancer, accelerate progression, and induce treatment resistance. OSA may be a contributor to but a preventable cause of the progression of lung cancer. However, whether this effect exists independently of other risk factors is unclear. Therefore, by reviewing the literature on the epidemiology, pathogenesis, and treatment of lung cancer and OSA, we hope to understand the relationships between the two and promote the interdisciplinary exchange of ideas between basic medicine, clinical medicine, respiratory medicine, sleep medicine, and oncology.
Collapse
Affiliation(s)
- Fang Yuan
- Department of Respiratory, The First Hospital of Jiujiang City, Jiujiang, China
| | - Yanxia Hu
- Department of Respiratory, The First Hospital of Jiujiang City, Jiujiang, China
| | - Fei Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xujun Feng
- Department of Respiratory, The First Hospital of Jiujiang City, Jiujiang, China
- Department of Respiratory and Critical Care Medicine, Sleep Medicine Center, Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal 2024; 22:109. [PMID: 38347575 PMCID: PMC10860306 DOI: 10.1186/s12964-023-01302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/30/2023] [Indexed: 02/15/2024] Open
Abstract
Cancer treatment faces many hurdles and resistance is one among them. Anti-cancer treatment strategies are evolving due to innate and acquired resistance capacity, governed by genetic, epigenetic, proteomic, metabolic, or microenvironmental cues that ultimately enable selected cancer cells to survive and progress under unfavorable conditions. Although the mechanism of drug resistance is being widely studied to generate new target-based drugs with better potency than existing ones. However, due to the broader flexibility in acquired drug resistance, advanced therapeutic options with better efficacy need to be explored. Combination therapy is an alternative with a better success rate though the risk of amplified side effects is commonplace. Moreover, recent groundbreaking precision immune therapy is one of the ways to overcome drug resistance and has revolutionized anticancer therapy to a greater extent with the only limitation of being individual-specific and needs further attention. This review will focus on the challenges and strategies opted by cancer cells to withstand the current therapies at the molecular level and also highlights the emerging therapeutic options -like immunological, and stem cell-based options that may prove to have better potential to challenge the existing problem of therapy resistance. Video Abstract.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Holcombe Blvd, Houston, TX, 77030, USA.
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
26
|
Zeng J, Cheng Y, Xie W, Lin X, Ding C, Xu H, Cui B, Chen Y, Gao S, Zhang S, Liu K, Lu Y, Zhou J, Shi Z, Sun Y. Calcium-sensing receptor and NF-κB pathways in TN breast cancer contribute to cancer-induced cardiomyocyte damage via activating neutrophil extracellular traps formation. Cell Mol Life Sci 2024; 81:19. [PMID: 38196005 PMCID: PMC11073098 DOI: 10.1007/s00018-023-05051-9] [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: 08/18/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 01/11/2024]
Abstract
Cardiovascular disorders are commonly prevalent in cancer patients, yet the mechanistic link between them remains poorly understood. Because neutrophil extracellular traps (NETs) have implications not just in cardiovascular diseases (CVD), but also in breast cancer (BC), it was hypothesized to contribute to CVD in the context of oncogenesis. We established a mouse model using nude mice to simulate liver metastasis of triple-negative BC (TNBC) through the injection of MDA-MB-231 cells. Multiple imaging and analysis techniques were employed to assess the cardiac function and structure, including echocardiography, HE staining, Masson staining, and transmission electron microscopy (TEM). MDA-MB-231 cells underwent treatment with a CaSR inhibitor, CaSR agonist, and NF-κB channel blocker. The phosphorylation of NF-κB channel protein p65 and the expression and secretion of IL-8 were assessed using qRT-PCR, Western Blot, and ELISA, respectively. In addition, MDA-MB-231 cells were co-cultured with polymorphonuclear neutrophils (PMN) under varying conditions. The co-localization of PMN extracellular myeloperoxidase (MPO) and DNA were observed by cellular immunofluorescence staining to identify the formation of NETs. Then, the cardiomyocytes were co-cultured with the above medium that contains NETs or not, respectively; the effects of NETs on cardiomyocytes apoptosis were perceived by flow cytometry. The ultrastructural changes of myocardial cells were perceived by TEM, and ELISA detected the levels of myocardial enzyme (LDH, MDA and SOD). Overall, according to our research, CaSR has been found to have a regulatory role in IL-8 secretion in MDA-MB-231 cells, as well as in the formation of NETs by PMN cells. These findings suggest CaSR-mediated stimulation in PMN can lead to increased NETs formation and subsequently to cytotoxicity in cardiomyocytes, which potentially via activation of the NF-κB signaling cascade of BC cell.
Collapse
Affiliation(s)
- Jingya Zeng
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Yangyang Cheng
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Wanlin Xie
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Xin Lin
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Chenglong Ding
- Department of Pathology, The First Affiliated Hospital of Jiamusi University, Jiamusi, 154003, Heilongjiang, China
| | - Huimin Xu
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Baohong Cui
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Yixin Chen
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Song Gao
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Siwen Zhang
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Kaiyue Liu
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Yue Lu
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Jialing Zhou
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Zhongxiang Shi
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China
| | - Yihua Sun
- Department of Clinical Laboratory, Harbin Medical University Cancer Hospital, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
27
|
Zhang YH, Chen Y, Shi L, Han X, Xie JC, Chen Y, Xiang M, Li BW, Li J, Xing HR, Wang JY. A novel lung cancer stem cell extracellular vesicles lncRNA ROLLCSC modulate non-stemness cancer cell plasticity through miR-5623-3p and miR-217-5p targeting lipid metabolism. Int J Biol Macromol 2024; 256:128412. [PMID: 38029909 DOI: 10.1016/j.ijbiomac.2023.128412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/19/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND The high mortality rate of lung cancer is largely attributed to metastasis. Lung cancer stem cells (CSC) are conducive to cancer heterogeneity. Long noncoding RNAs are known to participate in various biological processes regulating the development of lung cancer. However, characterization of the role and mechanisms of lncRNA in lung cancer metastasis remains a challenge. RESULTS We demonstrate that ROLLCSC, a highly expressed lncRNA in LLC-SDs, promotes the metastasis of the low metastatic LLCs both in vitro and in vivo. ROLLCSC can be transferred from LLC-SD to LLC through encapsulation in extracellular vesicles (EVs), ultimately leading to the enhancement of the metastatic phenotype of LLCs. Mechanistically, we demonstrate that the pro-metastatic activity of ROLLCSC is achieved through its function as a competing endogenous RNA (ceRNA) of miR-5623-3p and miR-217-5p to stimulate lipid metabolism. CONCLUSION In this study, we have characterized ROLLCSC, a novel lncRNA, as a pivotal regulator in the metastasis of lung cancer, highlighting its potential as a therapeutic target. Specifically, we show that ROLLCSC is encapsulated by the EVs of LLC-SDs and transmitted to the LLCs, where it acts as a ceRNA of miR-5623-3p and miR-217-5p to stimulate lipid metabolism and ultimately augments metastatic colonization of LLCs.
Collapse
Affiliation(s)
- Yu-Han Zhang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yan Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Lei Shi
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Xue Han
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Jia-Cheng Xie
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yuting Chen
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Meng Xiang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Bo-Wen Li
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Jie Li
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - H Rosie Xing
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400016, China; Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China.
| | - Jian-Yu Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
28
|
Tian W, Huang J, Zhang W, Wang Y, Jin R, Guo H, Tang Y, Wang Y, Lai H, Leung ELH. Harnessing natural product polysaccharides against lung cancer and revisit its novel mechanism. Pharmacol Res 2024; 199:107034. [PMID: 38070793 DOI: 10.1016/j.phrs.2023.107034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024]
Abstract
The incidence and mortality of lung cancer are on the rise worldwide. However, the benefit of clinical treatment in lung cancer is limited. Owning to important sources of drug development, natural products have received constant attention around the world. Main ingredient polysaccharides in natural products have been found to have various activities in pharmacological research. In recent years, more and more scientists are looking for the effects and mechanisms of different natural product polysaccharides on lung cancer. In this review, we focus on the following aspects: First, natural product polysaccharides have been discovered to directly suppress the growth of lung cancer cells, which can be effective in limiting tumor progression. Additionally, polysaccharides have been considered to enhance immune function, which can play a pivotal role in fighting lung cancer. Lastly, polysaccharides can improve the efficacy of drugs in lung cancer treatment by regulating the gut microbiota. Overall, the research of natural product polysaccharides in the treatment of lung cancer is a promising area that has the potential to lead to new clinical treatments. With better understanding, natural product polysaccharides have the potential to become important components of future lung cancer treatments.
Collapse
Affiliation(s)
- Wangqi Tian
- College of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-xianyang New Economic Zone, Shaanxi Province, China
| | - Jumin Huang
- Cancer Center, Faculty of Health Sciences, and MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau
| | - Weitong Zhang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-xianyang New Economic Zone, Shaanxi Province, China
| | - Yifan Wang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-xianyang New Economic Zone, Shaanxi Province, China
| | - Ruyi Jin
- College of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-xianyang New Economic Zone, Shaanxi Province, China
| | - Hui Guo
- College of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-xianyang New Economic Zone, Shaanxi Province, China
| | - Yuping Tang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-xianyang New Economic Zone, Shaanxi Province, China
| | - Yuwei Wang
- College of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-xianyang New Economic Zone, Shaanxi Province, China.
| | - Huanling Lai
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangdong Province, China; Guangzhou Laboratory, Guangzhou 510005, Guangdong Province, China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Sciences, and MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau; State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau.
| |
Collapse
|
29
|
Zheng X, Song X, Zhu G, Pan D, Li H, Hu J, Xiao K, Gong Q, Gu Z, Luo K, Li W. Nanomedicine Combats Drug Resistance in Lung Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308977. [PMID: 37968865 DOI: 10.1002/adma.202308977] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/03/2023] [Indexed: 11/17/2023]
Abstract
Lung cancer is the second most prevalent cancer and the leading cause of cancer-related death worldwide. Surgery, chemotherapy, molecular targeted therapy, immunotherapy, and radiotherapy are currently available as treatment methods. However, drug resistance is a significant factor in the failure of lung cancer treatments. Novel therapeutics have been exploited to address complicated resistance mechanisms of lung cancer and the advancement of nanomedicine is extremely promising in terms of overcoming drug resistance. Nanomedicine equipped with multifunctional and tunable physiochemical properties in alignment with tumor genetic profiles can achieve precise, safe, and effective treatment while minimizing or eradicating drug resistance in cancer. Here, this work reviews the discovered resistance mechanisms for lung cancer chemotherapy, molecular targeted therapy, immunotherapy, and radiotherapy, and outlines novel strategies for the development of nanomedicine against drug resistance. This work focuses on engineering design, customized delivery, current challenges, and clinical translation of nanomedicine in the application of resistant lung cancer.
Collapse
Affiliation(s)
- Xiuli Zheng
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Xiaohai Song
- Department of General Surgery, Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Guonian Zhu
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Dayi Pan
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Haonan Li
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Jiankun Hu
- Department of General Surgery, Gastric Cancer Center and Laboratory of Gastric Cancer, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Kai Xiao
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Qiyong Gong
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Precision Medicine Key Laboratory of Sichuan Province, Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen, Fujian, 361000, China
| | - Zhongwei Gu
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
| | - Kui Luo
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Precision Medicine Key Laboratory of Sichuan Province, Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Weimin Li
- Department of Radiology, Department of Respiratory, Huaxi MR Research Center (HMRRC) and Critical Care Medicine, Institute of Respiratory Health, Precision Medicine Center, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, China
- Precision Medicine Key Laboratory of Sichuan Province, Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
30
|
Alsaikhan F. Hyaluronic acid-empowered nanotheranostics in breast and lung cancers therapy. ENVIRONMENTAL RESEARCH 2023; 237:116951. [PMID: 37633628 DOI: 10.1016/j.envres.2023.116951] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 08/28/2023]
Abstract
Nanomedicine application in cancer therapy is an urgency because of inability of current biological therapies for complete removal of tumor cells. The development of smart and novel nanoplatforms for treatment of cancer can provide new insight in tumor suppression. Hyaluronic acid is a biopolymer that can be employed for synthesis of smart nanostructures capable of selective targeting CD44-overexpressing tumor cells. The breast and lung cancers are among the most malignant and common tumors in both females and males that environmental factors, lifestyle and genomic alterations are among the risk factors for their pathogenesis and development. Since etiology of breast and lung tumors is not certain and multiple factors participate in their development, preventative measures have not been completely successful and studies have focused on developing new treatment strategies for them. The aim of current review is to provide a comprehensive discussion about application of hyaluronic acid-based nanostructures for treatment of breast and lung cancers. The main reason of using hyaluronic acid-based nanoparticles is their ability in targeting breast and lung cancers in a selective way due to upregulation of CD44 receptor on their surface. Moreover, nanocarriers developed from hyaluronic acid or functionalized with hyaluronic acid have high biocompatibility and their safety is appreciated. The drugs and genes used for treatment of breast and lung cancers lack specific accumulation at cancer site and their cytotoxicity is low, but hyaluronic acid-based nanostructures provide their targeted delivery to tumor site and by increasing internalization of drugs and genes in breast and lung tumor cells, they improve their therapeutic index. Furthermore, hyaluronic acid-based nanostructures can be used for phototherapy-mediated breast and lung cancers ablation. The stimuli-responsive and smart kinds of hyaluronic acid-based nanostructures such as pH- and light-responsive can increase selective targeting of breast and lung cancers.
Collapse
Affiliation(s)
- Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
| |
Collapse
|
31
|
Li T, Giaccone G. Advances in biology and novel treatments of SCLC. Semin Cancer Biol 2023; 96:1-2. [PMID: 37611726 DOI: 10.1016/j.semcancer.2023.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Affiliation(s)
- Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Giuseppe Giaccone
- Department of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, United States.
| |
Collapse
|
32
|
Mai Y, Su J, Yang C, Xia C, Fu L. The strategies to cure cancer patients by eradicating cancer stem-like cells. Mol Cancer 2023; 22:171. [PMID: 37853413 PMCID: PMC10583358 DOI: 10.1186/s12943-023-01867-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023] Open
Abstract
Cancer stem-like cells (CSCs), a subpopulation of cancer cells, possess remarkable capability in proliferation, self-renewal, and differentiation. Their presence is recognized as a crucial factor contributing to tumor progression and metastasis. CSCs have garnered significant attention as a therapeutic focus and an etiologic root of treatment-resistant cells. Increasing evidence indicated that specific biomarkers, aberrant activated pathways, immunosuppressive tumor microenvironment (TME), and immunoevasion are considered the culprits in the occurrence of CSCs and the maintenance of CSCs properties including multi-directional differentiation. Targeting CSC biomarkers, stemness-associated pathways, TME, immunoevasion and inducing CSCs differentiation improve CSCs eradication and, therefore, cancer treatment. This review comprehensively summarized these targeted therapies, along with their current status in clinical trials. By exploring and implementing strategies aimed at eradicating CSCs, researchers aim to improve cancer treatment outcomes and overcome the challenges posed by CSC-mediated therapy resistance.
Collapse
Affiliation(s)
- Yansui Mai
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jiyan Su
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, China; School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| | - Liwu Fu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Guangdong Esophageal Cancer Institute; Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
33
|
Deng H, Chen Y, Wang L, Zhang Y, Hang Q, Li P, Zhang P, Ji J, Song H, Chen M, Jin Y. PI3K/mTOR inhibitors promote G6PD autophagic degradation and exacerbate oxidative stress damage to radiosensitize small cell lung cancer. Cell Death Dis 2023; 14:652. [PMID: 37802999 PMCID: PMC10558571 DOI: 10.1038/s41419-023-06171-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 10/08/2023]
Abstract
Our previous study revealed that PI3K/AKT/mTOR signaling was associated with SCLC radioresistance. SBC2 cells were used as primary radioresistance models, while H446 cells were continuously exposed to ionizing radiation (IR) to develop acquired radioresistance. Cell viability and apoptosis assays were used to investigate synergistic effects of BEZ235/GSK2126458 and IR in vitro, while immunoblotting, metabolite quantitative analysis and bioinformatic analyses were utilized to explore the underlying mechanism. Both genetically engineered mouse models (GEMM) and subcutaneous tumor models were used to confirm the synergistic effect in vivo. Key molecules of PI3K/AKT/mTOR signaling were upregulated after IR, which was correlated with primary radioresistance, and they were more expressed in acquired radioresistant cells. BEZ235/GSK2126458 effectively enhanced the cytotoxic effects of IR. BEZ235/GSK2126458 plus IR elevated γ-H2AX and p-Nrf2 expression, suggesting DNA and oxidative stress damage were intensified. Mechanistically, BEZ235/GSK2126458 plus IR significantly reduced the expression of G6PD protein, the rate-limiting enzyme of the pentose phosphate pathway (PPP). In detail, PI3K/mTOR inhibitors reinforced interaction between G6PD and HSPA8/HSC70, and G6PD was degraded by chaperone-mediated autophagy processes. Their metabolites (NADPH and R-5P) were decreased, and ROS levels were indirectly elevated, both of which exacerbated cell death. PI3K/AKT/mTOR signaling activator, insulin, enhanced SCLC radioresistance, while the synergistic effect of BEZ235/GSK2126458 and IR can be attenuated by N-acetylcysteine, and enhanced by 6-amino niacinamide. GEMM and allograft transplantation assays further confirmed their synergistic effect in vivo. This study provided insights into the connection between PI3K/AKT/mTOR signaling and the PPP underlying radioresistance and provided evidence of mechanisms supporting PI3K/mTOR inhibitors as possible therapeutic strategies to abrogate SCLC radioresistance.
Collapse
Affiliation(s)
- Huan Deng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yamei Chen
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200240, China
| | - Li Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yibi Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 331800, China
| | - Qingqing Hang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Peijing Li
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Peng Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Jing Ji
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Hai Song
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Ming Chen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co., Ltd, Guangzhou, China.
| | - Ying Jin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, 310022, China.
| |
Collapse
|
34
|
Zeng L, Liang L, Fang X, Xiang S, Dai C, Zheng T, Li T, Feng Z. Glycolysis induces Th2 cell infiltration and significantly affects prognosis and immunotherapy response to lung adenocarcinoma. Funct Integr Genomics 2023; 23:221. [PMID: 37400733 DOI: 10.1007/s10142-023-01155-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/22/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023]
Abstract
Glycolysis has a major role in cancer progression and can affect the tumor immune microenvironment, while its specific role in lung adenocarcinoma (LUAD) remains poorly studied. We obtained publicly available data from The Cancer Genome Atlas and Gene Expression Omnibus databases and used R software to analyze the specific role of glycolysis in LUAD. The Single Sample Gene Set Enrichment Analysis (ssGSEA) indicated a correlation between glycolysis and unfavorable clinical outcome, as well as a repression effect on the immunotherapy response of LUAD patients. Pathway enrichment analysis revealed a significant enrichment of MYC targets, epithelial-mesenchymal transition (EMT), hypoxia, G2M checkpoint, and mTORC1 signaling pathways in patients with higher activity of glycolysis. Immune infiltration analysis showed a higher infiltration of M0 and M1 macrophages in patients with elevated activity of glycolysis. Moreover, we developed a prognosis model based on six glycolysis-related genes, including DLGAP5, TOP2A, KIF20A, OIP5, HJURP, and ANLN. Both the training and validation cohorts demonstrated the high efficiency of prognostic prediction in this model, which identified that patients with high risk may have a poorer prognosis and lower sensitivity to immunotherapy. Additionally, we also found that Th2 cell infiltration may predict poorer survival and resistance to immunotherapy. The study indicated that glycolysis is significantly associated with poor prognosis in patients with LUAD and immunotherapy resistance, which might be partly dependent on the Th2 cell infiltration. Additionally, the signature comprised of six genes related to glycolysis showed promising predictive value for LUAD prognosis.
Collapse
Affiliation(s)
- Liping Zeng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- College of Basic Medicine, Hunan University of Medicine, 492 Jinxi South Rd, Huaihua, 418000, China
| | - Lu Liang
- Department of Pathology, The First Affiliated Hospital of Hunan University of Medicine, Yushi RD, Huaihua, 418000, China
| | - Xianlei Fang
- College of Basic Medicine, Hunan University of Medicine, 492 Jinxi South Rd, Huaihua, 418000, China
| | - Sha Xiang
- College of Basic Medicine, Hunan University of Medicine, 492 Jinxi South Rd, Huaihua, 418000, China
| | - Chenglong Dai
- Department of Physical Diagnosis, The First Affiliated Hospital of Hunan University of Medicine, 383 Yushi RD, Huaihua, 418000, China
| | - Tao Zheng
- Department of Radiotherapy Oncology, The No. 2 People's Hospital of Huaihua, Huaihua, 418000, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Zhenbo Feng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Rd, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
35
|
Zhang X, Yu S, Li X, Wen X, Liu S, Zu R, Ren H, Li T, Yang C, Luo H. Research progress on the interaction between oxidative stress and platelets: Another avenue for cancer? Pharmacol Res 2023; 191:106777. [PMID: 37080257 DOI: 10.1016/j.phrs.2023.106777] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 04/06/2023] [Accepted: 04/18/2023] [Indexed: 04/22/2023]
Abstract
Oxidative stress (OS) is a chemical imbalance between an oxidant and an antioxidant, causing damage to redox signaling and control or causing molecular damage. Unbalanced oxidative metabolism can produce excessive reactive oxygen species (ROS). These excess ROS can cause drastic changes in platelet metabolism and further affect platelet function. It will also lead to an increase in platelet procoagulant phenotype and cell apoptosis, which will increase the risk of thrombosis. The creation of ROS and subsequent platelet activation, adhesion, and recruitment are then further encouraged in an auto-amplifying loop by ROS produced from platelets. Meanwhile, cancer cells produce a higher concentration of ROS due to their fast metabolism and high proliferation rate. However, excessive ROS can result in damage to and modification of cellular macromolecules. The formation of cancer and its progression is strongly associated with oxidative stress and the resulting oxidative damage. In addition, platelets are an important part of the tumor microenvironment, and there is a significant cross-communication between platelets and cancer cells. Cancer cells alter the activation status of platelets, their RNA spectrum, proteome, and other properties. The "cloaking" of cancer cells by platelets providing physical protection,avoiding destruction from shear stress and the attack of immune cells, promoting tumor cell invasion.We explored the vicious circle interaction between ROS, platelets, and cancer in this review, and we believe that ROS can play a stimulative role in tumor growth and metastasis through platelets.
Collapse
Affiliation(s)
- Xingmei Zhang
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Sisi Yu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China
| | - Xiaobo Li
- Molecular Diagnostic Laboratory of Department of Microbiology and Immunology, 3201 Hospital Affiliated to Medical College of Xi'an Jiaotong University, Hanzhong 723099, China
| | - Xiaoxia Wen
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Shan Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Ruiling Zu
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China
| | - Hanxiao Ren
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Chaoguo Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610042, China.
| | - Huaichao Luo
- Department of Clinical Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041 China.
| |
Collapse
|