1
|
Wang X, Tian Y, Wu X, Zhu Y, Chen H, Wang Z, Liu Z, Tan J, Pan Z, Cao J, Li Z, Zhang X, Shi Z, Wang J, Liu T. Targeting PERP promotes anti-tumor immunity in HNSCC by regulating tumor immune microenvironment and metabolic homeostasis. Mol Cancer 2025; 24:168. [PMID: 40483487 PMCID: PMC12144827 DOI: 10.1186/s12943-025-02360-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 05/20/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND PERP may have the potential to function as an oncogene. However, the precise function, prognostic value, and predictive significance remain shrouded in ambiguity. METHODS We conducted an in-depth analysis using pan-cancer RNA sequencing data and various online web tools to investigate the correlation between PERP and crucial clinical outcomes such as prognosis, tumor microenvironment, and tumor metabolism. In addition, we explored the tumor-promoting role of PERP and its potential mechanisms through models such as immunofluorescence staining, flow cytometry, cell proliferation assays, wound healing assays, cell migration assays, mass spectrometry analysis and isotope tracing. Further in vivo models confirmed the functional consistency of PERP across pan-cancer. Finally, we analyzed the potential of PERP as a predictive factor for immunotherapy sensitivity in a clinical cohort. RESULTS PERP exhibits elevated expression in the majority of cancer types and impedes immune cell infiltration as well as immune checkpoint reactivity in pan-cancer. We confirmed that PERP can promote tumor progression by tumor cell proliferation, scratch and transwell experiments. Meanwhile, the absence of PERP restricts the flux of 13C6-glucose into glycolysis and the tricarboxylic acid (TCA) cycle. Importantly, the deficiency of PERP enhances the in vivo anti-tumor efficacy of PD1 monoclonal antibodies. In addition, low PERP expression is highly correlated with the response of head and neck squamous cell carcinoma (HNSCC) patients to immunotherapy. CONCLUSIONS PERP represents a promising predictive/diagnostic biomarker and therapeutic target for HNSCC patients.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150000, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Yuxi Tian
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
- Department of Geriatrics, Respiratory Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, Hunan, 410008, People's Republic of China
- National Medical Metabolomics International Collaborative Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Xiaohong Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China
| | - Yewen Zhu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Huihong Chen
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Zeyao Wang
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150000, China
| | - Zihan Liu
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150000, China
| | - Jiaqi Tan
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Zhaoyu Pan
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Jiaoyan Cao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Zhenjiang Li
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Xin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China
| | - Zhongjie Shi
- Department of Pediatrics, Wayne State University, Detroit, MI, 48201, USA
| | - Juncheng Wang
- Department of Otolaryngology-Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P.R. China.
- National Medical Metabolomics International Collaborative Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China.
| | - Tong Liu
- Department of Surgical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, 150000, China.
- NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, 150001, China.
| |
Collapse
|
2
|
Liu S, Zhang W, Liu W, Ding Z, Zhang R, Han Y, Niu Z, Zhang M, Li H, Li D, Wang Z, Peng J, Wu Y, Han Y, Xie Z, Wu J, Qin L, Hu Z, Chen X, Hu Y, Liu Y, Ma S, Zha X. PRMT1-Mediated SWI/SNF Complex Recruitment via SMARCC1 Drives IGF2BP2 Transcription to Enhance Carboplatin Resistance in Head and Neck Squamous Cell Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417460. [PMID: 40270464 PMCID: PMC12165065 DOI: 10.1002/advs.202417460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/27/2025] [Indexed: 04/25/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a malignancy with poor prognosis and chemotherapy resistance. Here, protein arginine methyltransferase 1 (PRMT1) is identified as a key driver of carboplatin (CBP) resistance in HNSCC. Analyses of clinical samples, cell lines, patient-derived organoids, and xenograft models reveal that PRMT1 promotes tumor growth and CBP resistance through a novel, methyltransferase-independent mechanism. Conditional PRMT1 knockout suppresses tumorigenesis and enhances CBP sensitivity in vivo, highlighting its essential role in HNSCC progression. Mechanistically, PRMT1 recruits the SWI/SNF chromatin remodeling complex via direct interaction with SMARCC1, leading to the transcriptional activation of insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2), which enhances CBP resistance and tumor growth. Notably, this function is independent of PRMT1's enzymatic activity, distinguishing it from its well-established roles in arginine methylation. Furthermore, pre-B-cell leukemia homeobox 2 (PBX2) is identified as an upstream transcriptional activator that binds the PRMT1 promoter, driving its overexpression and reinforcing this oncogenic network. Clinically, high PBX2, PRMT1, SMARCC1, and IGF2BP2 expression correlates with malignant progression and poor prognosis in HNSCC patients. This study uncovers a previously unrecognized non-catalytic function of PRMT1 and highlights the PBX2-PRMT1-SWI/SNF-IGF2BP2 axis as a potential therapeutic target for overcoming CBP resistance in HNSCC.
Collapse
Affiliation(s)
- Shixian Liu
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Bengbu Medical UniversityBengbu233004China
- Department of Biochemistry & Molecular BiologySchool of Basic MedicineAnhui Medical UniversityHefei230032China
| | - Wentao Zhang
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
- Department of Biochemistry & Molecular BiologySchool of Basic MedicineAnhui Medical UniversityHefei230032China
| | - Weiwei Liu
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Bengbu Medical UniversityBengbu233004China
| | - Zhao Ding
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
- Department of Biochemistry & Molecular BiologySchool of Basic MedicineAnhui Medical UniversityHefei230032China
| | - Ruijing Zhang
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Bengbu Medical UniversityBengbu233004China
| | - Yuefeng Han
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Bengbu Medical UniversityBengbu233004China
| | - Zihao Niu
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Mengdie Zhang
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Hui Li
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Bengbu Medical UniversityBengbu233004China
| | - Dapeng Li
- Department of OtolaryngologyHead and Neck Surgeryand Scientific Research and Experiment CenterThe Affiliated Bozhou Hospital of Anhui Medical UniversityBozhou236800China
| | - Zixi Wang
- Department of Biochemistry & Molecular BiologySchool of Basic MedicineAnhui Medical UniversityHefei230032China
- Institutes of Biomedical SciencesChildren's Hospital of Fudan UniversityNational Children's Medical CenterFudan UniversityShanghai200032China
| | - Jie Peng
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Bengbu Medical UniversityBengbu233004China
| | - Yu Wu
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Yanxun Han
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
- Department of Biochemistry & Molecular BiologySchool of Basic MedicineAnhui Medical UniversityHefei230032China
| | - Zihui Xie
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
- Department of Biochemistry & Molecular BiologySchool of Basic MedicineAnhui Medical UniversityHefei230032China
| | - Jing Wu
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Liang Qin
- Department of UrologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese MedicineBeijing Research Institute of Chinese MedicineBeijing University of Chinese MedicineBeijing100029China
| | - Xu Chen
- Department of Biochemistry & Molecular BiologySchool of Basic MedicineAnhui Medical UniversityHefei230032China
| | - Yunlong Hu
- Department of Otolaryngology Head and Neck SurgeryAnhui NO.2 Provincial People's HospitalHefei230041China
| | - Yehai Liu
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Shiyin Ma
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Bengbu Medical UniversityBengbu233004China
| | - Xiaojun Zha
- Department of OtolaryngologyHead & Neck SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
- Department of Biochemistry & Molecular BiologySchool of Basic MedicineAnhui Medical UniversityHefei230032China
- Department of OtolaryngologyHead and Neck Surgeryand Scientific Research and Experiment CenterThe Affiliated Bozhou Hospital of Anhui Medical UniversityBozhou236800China
| |
Collapse
|
3
|
Kavaliauskas P, Acevedo W, Mickevičiūtė E, Grigalevičiūtė R, Grybaitė B, Sapijanskaitė-Banevič B, Pranaitytė G, Petraitis V, Petraitienė R, Mickevičius V. 3,3'-((3-Hydroxyphenyl)azanediyl)dipropionic Acid Derivatives as a Promising Scaffold Against Drug-Resistant Pathogens and Chemotherapy-Resistant Cancer. Pathogens 2025; 14:484. [PMID: 40430804 PMCID: PMC12115217 DOI: 10.3390/pathogens14050484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
The synthesis and antimicrobial and anticancer activity of 3,3'-((3-hydroxyphenyl)azanediyl)dipropionic acid derivatives (2-25) against drug-resistant bacterial pathogens and FaDu head and neck cancer cells were investigated. The derivatives were synthesized through various methods, including esterification, hydrazinolysis, and condensation reactions. The compounds demonstrated structure-dependent antimicrobial activity, predominantly targeting Gram-positive pathogens. Compounds containing 4-nitrophenyl, 1-naphthyl, and 5-nitro-2-thienyl groups exhibited enhanced activity against S. aureus and E. faecalis. Additionally, compounds 5, 6, and 25 showed antiproliferative activity in cisplatin-resistant FaDu cells at low micromolar concentrations. The in silico modeling revealed that compound 25 interacts with the HER-2 and c-MET proteins. These compounds also induced significant oxidative stress in FaDu cells and demonstrated low cytotoxic activity in non-cancerous HEK293 cells. These results highlight the potential of N-aryl-substituted β-amino acid derivatives as promising scaffolds for the further development of novel amino acid-based antimicrobial and anticancer agents targeting drug-resistant pathogens and cancers.
Collapse
Affiliation(s)
- Povilas Kavaliauskas
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
- Biological Research Center, Lithuanian University of Health Sciences, Tilzes Street 18, LT-47181 Kaunas, Lithuania;
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 655 W. Baltimore Street, Baltimore, MD 21201, USA
- Institute of Infectious Diseases and Pathogenic Microbiology, Birstono Street 38A, LT-59116 Prienai, Lithuania
| | - Waldo Acevedo
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso 2373223, Chile;
| | - Eglė Mickevičiūtė
- Department of Information Systems, Kaunas University of Technology, LT-51368 Kaunas, Lithuania;
| | - Ramunė Grigalevičiūtė
- Biological Research Center, Lithuanian University of Health Sciences, Tilzes Street 18, LT-47181 Kaunas, Lithuania;
- Department of Animal Nutrition, Lithuanian University of Health Sciences, Tilzes Street 18, LT-47181 Kaunas, Lithuania
| | - Birutė Grybaitė
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
| | - Birutė Sapijanskaitė-Banevič
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
| | - Guoda Pranaitytė
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
| | - Vidmantas Petraitis
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; (V.P.); (R.P.)
| | - Rūta Petraitienė
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA; (V.P.); (R.P.)
| | - Vytautas Mickevičius
- Department of Organic Chemistry, Kaunas University of Technology, LT-50254 Kaunas, Lithuania or (P.K.); (B.G.); (B.S.-B.); (G.P.)
| |
Collapse
|
4
|
Liu X, Zhang J, Yi T, Li H, Tang X, Liu D, Wu D, Li Y. Decoding tumor angiogenesis: pathways, mechanisms, and future directions in anti-cancer strategies. Biomark Res 2025; 13:62. [PMID: 40251641 PMCID: PMC12007322 DOI: 10.1186/s40364-025-00779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 04/13/2025] [Indexed: 04/20/2025] Open
Abstract
Angiogenesis, a crucial process in tumor growth and metastasis, necessitates targeted therapeutic intervention. This review reviews the latest knowledge of anti-angiogenesis targets in tumors, with emphasis on the molecular mechanisms and signaling pathways that regulate this process. We emphasize the tumor microenvironment's role in angiogenesis, examine endothelial cell metabolic changes, and evaluated potential therapeutic strategies targeting the tumor vascular system. At the same time, we analyzed the signaling pathway and molecular mechanism of tumor angiogenesis in detail. In addition, this paper also looks at the development trend of tumor anti-angiogenesis drugs, including their future development direction and challenges, aiming to provide prospective insight into the development of this field. Despite their potential, anti-angiogenic therapies encounter challenges like drug resistance and side effects, necessitating ongoing research to enhance cancer treatment strategies and the efficacy of these therapies.
Collapse
Affiliation(s)
- Xueru Liu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Ting Yi
- Department of Trauma Center, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Dan Liu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Daichao Wu
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China.
| |
Collapse
|
5
|
Liu C, Liu N, Zhang T, Tu Y. Adoptive immune cell therapy for colorectal cancer. Front Immunol 2025; 16:1557906. [PMID: 40236691 PMCID: PMC11996668 DOI: 10.3389/fimmu.2025.1557906] [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: 01/09/2025] [Accepted: 02/28/2025] [Indexed: 04/17/2025] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related morbidity and mortality worldwide, with limited options for patients at advanced stages. Immunotherapy, particularly immune cell-based therapies, has gained significant attention as an innovative approach for targeting CRC. This review summarizes the progress in various immune cell therapies, including DC vaccine, CAR/TCR-T cells, CAR-NK cells et al, each engineered to recognize and attack cancer cells expressing specific antigens. CAR-T cell therapy, which has been successful in hematologic cancers, faces challenges in CRC due to the solid tumor microenvironment, which limits cell infiltration and persistence. CAR-NK cells, CAR-M and CAR-γδ T cells, however, offer alternative strategies due to their unique properties, such as the ability to target tumor cells without prior sensitization and a lower risk of inducing severe cytokine release syndrome. Recent advances in lentiviral transduction have enabled effective expression of CARs on NK and γδ T cells, providing promising preclinical results in CRC models. This review explores the mechanisms, tumor targets, preclinical studies, and early-phase clinical trials of these therapies, addressing key challenges such as enhancing specificity to tumor antigens and overcoming the immunosuppressive tumor microenvironment. The potential of combination therapies, including immune checkpoint inhibitors and cytokine therapy, is also discussed some as a means to improve the effectiveness of immune cell-based treatments for CRC. Continued research is essential to translate these promising approaches into clinical settings, offering new hope for CRC patients.
Collapse
Affiliation(s)
- Chenxiao Liu
- Guangdong Province Science and Technology Expert Workstation, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
| | - Nan Liu
- Guangdong Province Science and Technology Expert Workstation, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Tongcun Zhang
- Guangdong Province Science and Technology Expert Workstation, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan, China
| | - Yanyang Tu
- Science Research Center, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
- Huizhou Central People’s Hospital Academy of Medical Sciences, Huizhou Central People’s Hospital, Huizhou, Guangdong, China
| |
Collapse
|
6
|
Sasi N, Preetha D, Iyyappan S, Selvamurugan N. Circular RNAs: Emerging regulators of signaling pathways in epithelial-mesenchymal transition and angiogenesis during breast cancer progression. Semin Oncol 2025; 52:152340. [PMID: 40220369 DOI: 10.1016/j.seminoncol.2025.152340] [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: 01/23/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 04/14/2025]
Abstract
Circular RNAs (circRNAs) have emerged as important regulators of gene expression and cellular activities, and abnormalities in circRNAs in breast cancer have been linked to important biological processes like epithelial-mesenchymal transition (EMT) and angiogenesis, both essential for tumor metastasis. EMT facilitates the transition of epithelial cancer cells into a mesenchymal phenotype, enhancing their invasive and migratory capabilities, while angiogenesis promotes tumor progression by forming new blood vessels. CircRNAs also interact with microRNAs to regulate signaling pathways such as TGF-β, Wnt/-catenin, and VEGF. Besides EMT and angiogenesis, studies have identified that circRNAs affect metabolic reprogramming, chemoresistance, tumor microenvironment remodeling, and immunological evasion. Thus, circRNAs play a multifaceted role in the development of breast cancer. They hold potential as non-invasive biomarkers and therapeutic targets due to their high stability, resistance to exonuclease degradation, abundance in body fluids, and diverse expression patterns across different tissues. This review summarizes and critically assesses existing understanding of the functional roles and molecular processes of circRNAs in controlling EMT and angiogenesis during breast cancer progression.
Collapse
Affiliation(s)
- Nivruthi Sasi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Tamil Nadu, India
| | - Dilipkumar Preetha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Tamil Nadu, India
| | - Saranya Iyyappan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Tamil Nadu, India.
| |
Collapse
|
7
|
Abo-Zaid OAR, Moawed FSM, Eldin ES, Farrag MA, Ahmed ESA. Antitumor activity of gamma-irradiated Rosa canina L. against lung carcinoma in rat model: a proposed mechanism. BMC Complement Med Ther 2025; 25:86. [PMID: 40022036 PMCID: PMC11869437 DOI: 10.1186/s12906-025-04813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/05/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Lung cancer is one of the most prevalent malignancies globally and is the leading cause of cancer-related mortality. Although cisplatin is a widely utilized chemotherapeutic agent, its clinical efficacy is often hampered by significant toxicity and undesirable side effects. Rosa canina, a medicinal plant, has demonstrated a range of beneficial biological activities, including anti-inflammatory, anticancer, immunomodulatory, antioxidant, and genoprotective effects. METHODS This study aimed to investigate the potential of Rosa canina to enhance the anticancer efficacy of cisplatin in a dimethyl benz(a)anthracene-induced lung cancer model using female rats. The animals were administered Rosa canina, cisplatin, or a combination of both treatments. The expression levels of critical signaling molecules were evaluated, including phosphoinositide-3-kinase (PI3K), Akt, mammalian target of rapamycin (mTOR), cleaved poly (ADP-ribose) polymerase (PARP-1), myeloid differentiation factor 88 (MyD88), and tumor necrosis factor receptor-associated factor (TRAF), in addition to various autophagic markers. Furthermore, we assessed the levels of toll-like receptor 2 (TLR2), nuclear factor kappa B (NF-κB), and apoptotic markers in lung tissue, complemented by histopathological examinations. RESULTS The combined treatment of Rosa canina extract and cisplatin significantly inhibited lung cancer cell proliferation by downregulating PARP-1 and the TLR2/MyD88/TRAF6/NF-κB signaling pathway, as well as the PI3K/Akt/mTOR pathway. Moreover, this combination therapy promoted autophagy and apoptosis, evidenced by elevated levels of autophagic and apoptotic markers. CONCLUSION Overall, the findings of this study suggest that Rosa canina enhances the anticancer effects of cisplatin by inhibiting cancer cell proliferation while simultaneously inducing autophagy and apoptosis. Thus, Rosa can be used as adjuvant to cisplatin chemotherapy to overcome its limitations which may be considered a new approach during lung cancer treatment strategy.
Collapse
Affiliation(s)
- Omayma A R Abo-Zaid
- Biochemistry and Molecular Biology Department, Faculty of Vet. Med, Benha University, Benha, Egypt
| | - Fatma S M Moawed
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Eman S Eldin
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mostafa A Farrag
- Radiation Biology , National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Cairo, 11787, Egypt
| | - Esraa S A Ahmed
- Radiation Biology , National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Nasr City, Cairo, 11787, Egypt.
| |
Collapse
|
8
|
Chen PJ, Lu YT, Yang WE, Su CW, Chang LC, Yang SF, Lin CW, Chou YE. The Impact of MET Variants in Oral Cancer Progression and Clinicopathological Characteristics. J Cancer 2025; 16:1747-1753. [PMID: 39991569 PMCID: PMC11843229 DOI: 10.7150/jca.106426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/26/2025] [Indexed: 02/25/2025] Open
Abstract
Epigenetic, genetic predisposition and epidemiological risk factors were suggested to be involved in the carcinogenesis of oral cancer. In this study, we focused on the associations of MET single-nucleotide polymorphisms (SNPs) to oral cancer susceptibility and clinicopathological characteristics. The MET SNPs rs41736, rs41739, rs1621, and rs33917957 in 1198 controls and 1318 male patients with oral cancer were analyzed with real-time polymerase chain reaction. Our results revealed that the cigarette smokers among the oral cancer patients who carried the MET rs1621 polymorphic variant "G" were significantly associated with lower risk to develop oral cancer [OR (95% CI) = 0.463 (0.226-0.948)]. The male oral cancer patients who with the genotypic variant "G" of MET rs33917957 were associated with lower risk of cell differentiated grade (p = 0.041). In the TCGA database, the MET expressions were upregulated in oral cancer tissues compared to normal tissues, and were correlated with poor cell differentiated and poorer prognoses in smoker groups. In conclusion, these novel findings underscore the role of MET genetic variants in oral cancer susceptibility, particularly in smokers, and highlight the potential of these variants for prognosis and disease prediction.
Collapse
Affiliation(s)
- Ping-Ju Chen
- Department of Dentistry, Changhua Christian Hospital, Changhua, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yen-Ting Lu
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Department of Otolaryngology, St. Martin De Porres Hospital, Chiayi, Taiwan
| | - Wei-En Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Wen Su
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Lun-Ching Chang
- Department of Mathematics and Statistics, Florida Atlantic University, Boca Raton, Florida, USA
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ying-Erh Chou
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
9
|
Luo D, Liu Y, Lu Z, Huang L. Targeted therapy and immunotherapy for gastric cancer: rational strategies, novel advancements, challenges, and future perspectives. Mol Med 2025; 31:52. [PMID: 39923010 PMCID: PMC11806620 DOI: 10.1186/s10020-025-01075-y] [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: 01/10/2025] [Indexed: 02/10/2025] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors worldwide, and its treatment has been a focus of medical research. Herein we systematically review the current status of and advancements in targeted therapy and immunotherapy for GC, which have emerged as important treatment strategies in recent years with great potential, and summarize the efficacy and safety of such treatments. Targeted therapies against key targets in GC, including epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), and vascular endothelial growth factor (VEGF)/VEGF receptor (VEGFR), have shown remarkable therapeutic efficacies by inhibiting tumor progression and/or blood supply. In particular, markable breakthroughs have been made in HER2-targeting drugs for HER2-positive GC patients. To address intrinsic and acquired resistances to HER2-targeting drugs, novel therapeutic agents including bispecific antibodies and antibody-drug conjugates (ADC) targeting HER2 have been developed. Immunotherapy enhances the recognition and elimination of cancer cells by activating body anticancer immune system. Programmed cell death protein 1 (PD-1) and programmed cell death-ligand 1 (PD-L1) antibodies are the most commonly used immunotherapeutic agents and have been used with some success in GC treatment. Innovative immunotherapy modalities, including adoptive immune cell therapy, tumor vaccines, and non-specific immunomodulators therapy, and oncolytic viruses have shown promise in early-stage clinical trials for GC. Clinical trials have supported that targeted therapy and immunotherapy can significantly improve the survival and quality of life of GC patients. However, the effects of such therapies need to be further improved and more personalized, with advancement in researches on tumor immune microenvironment. Further studies remain needed to address the issues of drug resistance and adverse events pertaining to such therapies for GC. The combined application of such therapies and individualized treatment strategies should be further explored with novel drugs developed, to provide more effective treatments for GC patients.
Collapse
Affiliation(s)
- Dong Luo
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
- Center of Structural Heart Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunmei Liu
- School of Cultural Heritage and Information Management, Shanghai University, Shanghai, 200444, China.
| | - Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Lei Huang
- Department of Gastroenterology, National Clinical Research Center for Digestive Diseases, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
- National Key Laboratory of Immunity and Inflammation, Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University/Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
10
|
Okuyama K, Tsuchiya M, Debnath KC, Islam S, Yanamoto S. Desmoplastic reaction in the microenvironment of head and neck and other solid tumors: the therapeutic barrier. Ther Adv Med Oncol 2025; 17:17588359251317144. [PMID: 39926258 PMCID: PMC11806477 DOI: 10.1177/17588359251317144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 01/15/2025] [Indexed: 02/11/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains a challenge due to limited prognostic biomarkers and therapeutic options. The tumor microenvironment (TME), particularly the desmoplastic reaction (DR) characterized by stromal fibrosis, plays a crucial role in cancer progression and resistance to therapy. This review aims to summarize the biological significance of DR in HNSCC initiation, progression, and treatment resistance. Histologically, DR in HNSCC correlates with invasion patterns and clinical outcomes, affecting disease-free and overall survival. The interaction between cancer-associated fibroblasts (CAFs) and TME influences immune responses, including resistance to immunotherapy. Notably, human papillomavirus-driven HNSCC exhibits distinct DR characteristics that further influence the prognosis. DR promotes epithelial-mesenchymal transition and cancer cell invasion through CAF-mediated extracellular matrix remodeling and signaling pathways such as transforming growth factor-beta. DR also affects bone invasion and chemotherapy resistance by modulating stromal responses. Therapeutic strategies targeting DR and stromal components show promise in overcoming therapeutic resistance including resistance to immune checkpoint inhibitors. Understanding the role of DR in HNSCC biology and its impact on treatment response is critical to developing effective therapeutic interventions.
Collapse
Affiliation(s)
- Kohei Okuyama
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 123, Houston, TX 77030-4009, USA
| | - Maiko Tsuchiya
- Department of Pathology, Teikyo University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Kala Chand Debnath
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shajedul Islam
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Souichi Yanamoto
- Department of Oral Oncology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima-shi, Hiroshima, Japan
| |
Collapse
|
11
|
He C, Wang X, Chiou YS, Basappa B, Zhu T, Pandey V, Lobie PE. Inhibition of TFF3 synergizes with c-MET inhibitors to decrease the CSC-like phenotype and metastatic burden in ER+HER2+ mammary carcinoma. Cell Death Dis 2025; 16:76. [PMID: 39920140 PMCID: PMC11806102 DOI: 10.1038/s41419-025-07387-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 02/09/2025]
Abstract
The interaction between HER2 and ERα signaling pathways contributes to resistance to anti-estrogen and HER2-targeted therapies, presenting substantial treatment challenges in ER-positive (ER+) HER2-positive (HER2+) mammary carcinoma (MC). Trefoil Factor-3 (TFF3) has been reported to mediate resistance to both anti-estrogen and anti-HER2 targeted therapies in ER+ and ER+HER2+ MC, respectively. Herein, the function and mechanism of TFF3 in ER+HER2+ MC were delineated; and novel combinatorial therapeutic strategies were identified. Elevated expression of TFF3 promoted the oncogenicity of ER+HER2+ MC cells, including enhanced cell proliferation, survival, anchorage-independent growth, 3D growth, cancer stem cell-like (CSC-like) phenotype, migration, invasion, and xenograft growth. Targeting TFF3 with an interfering RNA plasmid or a small-molecule inhibitor (AMPC) inhibited these oncogenic characteristics, highlighting the therapeutic potential of targeting TFF3 in ER+HER2+ MC. Furthermore, a high-throughput combinatorial anti-cancer compound library screening revealed that AMPC preferentially synergized with receptor tyrosine kinase c-MET inhibitors (c-METis) to reduce cell survival and the CSC-like phenotype. The combination of AMPC and c-METis also synergistically suppressed the in vivo growth of ER+HER2+ MC cell-derived xenografts and abrogated lung metastasis. Mechanistically, TFF3 was observed to activate c-MET signaling through a positive-feedback loop to enhance the CSC-like phenotype of ER+HER2+ MC. Therefore, proof of concept is provided herein that antagonizing of TFF3 is a promising therapeutic strategy in combination with c-MET inhibition for the treatment of ER+HER2+ MC.
Collapse
Affiliation(s)
- Chuyu He
- Institute of Biopharmaceutical and Health Engineering and Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, PR China
| | - Xuejuan Wang
- Institute of Biopharmaceutical and Health Engineering and Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, PR China
| | - Yi-Shiou Chiou
- Institute of Biopharmaceutical and Health Engineering and Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, PR China
- Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore, India
| | - Tao Zhu
- Shenzhen Bay Laboratory, Shenzhen, Guangdong, PR China
- Department of Oncology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, PR China
- Hefei National Laboratory for Physical Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering and Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, PR China.
| | - Peter E Lobie
- Institute of Biopharmaceutical and Health Engineering and Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, PR China.
- Shenzhen Bay Laboratory, Shenzhen, Guangdong, PR China.
| |
Collapse
|
12
|
Fatima S, Kumar V, Kumar D. Molecular mechanism of genetic, epigenetic, and metabolic alteration in lung cancer. Med Oncol 2025; 42:61. [PMID: 39893601 DOI: 10.1007/s12032-025-02608-5] [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: 11/15/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025]
Abstract
Lung cancer, a leading cause of cancer-related deaths worldwide, is primarily linked to smoking, tobacco use, air pollution, and exposure to hazardous chemicals. Genetic alterations, particularly in oncogenes like RAS, EGFR, MYC, BRAF, HER, and P13K, can lead to metabolic changes in cancer cells. These cells often rely on glycolysis for energy production, even in the presence of oxygen, a phenomenon known as aerobic glycolysis. This metabolic shift, along with other alterations, contributes to cancer cell growth and survival. To develop effective therapies, it's crucial to understand the genetic and metabolic changes that drive lung cancer. This review aims to identify specific genes associated with these metabolic alterations and screen phytochemicals for their potential to target these genes. By targeting both genetic and metabolic pathways, we hope to develop innovative therapeutic approaches to combat lung cancer.
Collapse
Affiliation(s)
- Sheeri Fatima
- School of Health Science and Technology (SoHST), UPES, Dehradun, Uttarakhand, 248007, India
| | - Vineet Kumar
- Chemistry & Bioprospecting Division, Forest Research Institute, Dehradun, 248006, India
| | - Dhruv Kumar
- School of Health Science and Technology (SoHST), UPES, Dehradun, Uttarakhand, 248007, India.
| |
Collapse
|
13
|
Ragab EM, Gamal DME, El-Najjar FF, Elkomy HA, Ragab MA, Elantary MA, Basyouni OM, Moustafa SM, El-Naggar SA, Elsherbiny AS. New insights into Notch signaling as a crucial pathway of pancreatic cancer stem cell behavior by chrysin-polylactic acid-based nanocomposite. Discov Oncol 2025; 16:107. [PMID: 39891818 PMCID: PMC11787125 DOI: 10.1007/s12672-025-01846-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Pancreatic cancer is an extremely deadly illness for which there are few reliable treatments. Recent research indicates that malignant tumors are highly variable and consist of a tiny subset of unique cancer cells, known as cancer stem cells (CSCs), which are responsible for the beginning and spread of tumors. These cells are typically identified by the expression of specific cell surface markers. A population of pancreatic cancer stem cells with aberrantly active developmental signaling pathways has been identified in recent studies of human pancreatic tumors. Among these Notch signaling pathway has been identified as a key regulator of CSCs self-renewal, making it an attractive target for therapeutic intervention. Chrysin-loaded polylactic acid (PLA) as polymeric nanoparticles systems have been growing interest in using as platforms for improved drug delivery. This review aims to explore innovative strategies for targeted therapy and optimized drug delivery in pancreatic CSCs by manipulating the Notch pathway and leveraging PLA-based drug delivery systems. Furthermore, we will assess the capability of PLA nanoparticles to enhance the bioavailability and effectiveness of gemcitabine in pancreatic cancer cells. The insights gained from this review have the potential to contribute to the development of novel treatment approaches that combine targeted therapy with advanced drug delivery utilizing biodegradable polymeric nanoparticles.
Collapse
Affiliation(s)
- Eman M Ragab
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doaa M El Gamal
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Fares F El-Najjar
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Hager A Elkomy
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mahmoud A Ragab
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mariam A Elantary
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Omar M Basyouni
- Chemistry/Zoology Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Sherif M Moustafa
- Chemistry/Biochemistry Division, chemistry department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Shimaa A El-Naggar
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Abeer S Elsherbiny
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
14
|
Tolentino-Molina BX, Loaeza-Loaeza J, Ortega-Soto A, Castro-Coronel Y, Fernández-Tilapa G, Hernández-Sotelo D. Hsa_circ_0009910 knockdown in HeLa cells increases miR‑198 expression levels and decreases c‑Met expression levels and cell viability. Oncol Lett 2025; 29:74. [PMID: 39650233 PMCID: PMC11622005 DOI: 10.3892/ol.2024.14820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 09/09/2024] [Indexed: 12/11/2024] Open
Abstract
Cervical cancer (CC) is considered a public health problem. Circular RNAs (circRNAs) serve important roles in different types of cancer, including CC. However, the mechanisms used by circRNAs to facilitate CC progression are currently unclear. The present study analyzed the effects of hsa_circ_0009910 knockdown on microRNA (miRNA/miR)-198 and mesenchymal-epithelial transition factor (c-Met) expression levels and its impact on apoptosis and the viability of HeLa cells. Differentially expressed circRNAs in CC were identified using analysis of circRNA microarray data. Bioinformatics analysis was performed to predict circRNA-microRNA (miRNA) and miRNA-mRNA interactions. The knockdown of hsa_circ_0009910 in HeLa cells was performed using small interfering RNA and the expression levels of hsa_circ_0009910, miR-198 and c-Met were assessed using reverse transcription-quantitative PCR. The viability and apoptosis of HeLa cells were evaluated using MTT, neutral red uptake and ApoLive-Glo™ multiplex assays. Hsa_circ_0009910 was significantly upregulated in HeLa cells and the knockdown of hsa_circ_0009910 increased miRNA-198 expression levels, reduced c-Met expression levels and decreased cellular viability, but not apoptosis, in HeLa cells. Overall, these results indicated that hsa_circ_0009910 could act as a molecular sponge of miRNA-198 and contribute to the upregulation of c-Met expression levels. The hsa_circ_0009910/miRNA-198/c-Met interaction network affects the viability, but not apoptosis, of HeLa cells. Based on this mechanism, the present study suggests that hsa_circ_0009910 may be a promising biomarker for CC.
Collapse
Affiliation(s)
- Bernardo Xavier Tolentino-Molina
- Laboratory of Cancer Epigenetics, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero 39070, Mexico
| | - Jaqueline Loaeza-Loaeza
- Laboratory of Neurotoxicology, Department of Toxicology, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07300, Mexico
| | - Arturo Ortega-Soto
- Laboratory of Neurotoxicology, Department of Toxicology, Center for Research and Advanced Studies of the National Polytechnic Institute, Mexico City 07300, Mexico
| | - Yaneth Castro-Coronel
- Laboratory of Cytopathology and Histochemistry, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero 39070, Mexico
| | - Gloria Fernández-Tilapa
- Clinical Research Laboratory, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero 39070, Mexico
| | - Daniel Hernández-Sotelo
- Laboratory of Cancer Epigenetics, School of Chemical and Biological Sciences, Autonomous University of Guerrero, Chilpancingo, Guerrero 39070, Mexico
| |
Collapse
|
15
|
Xia Y, Huang C, Zhong M, Zhong H, Ruan R, Xiong J, Yao Y, Zhou J, Deng J. Targeting HGF/c-MET signaling to regulate the tumor microenvironment: Implications for counteracting tumor immune evasion. Cell Commun Signal 2025; 23:46. [PMID: 39856684 PMCID: PMC11762533 DOI: 10.1186/s12964-025-02033-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The hepatocyte growth factor (HGF) along with its receptor (c-MET) are crucial in preserving standard cellular physiological activities, and imbalances in the c-MET signaling pathway can lead to the development and advancement of tumors. It has been extensively demonstrated that immune checkpoint inhibitors (ICIs) can result in prolonged remission in certain patients. Nevertheless, numerous preclinical studies have shown that MET imbalance hinders the effectiveness of anti-PD-1/PD-L1 treatments through various mechanisms. Consequently, clarifying the link between the c-MET signaling pathway and the tumor microenvironment (TME), as well as uncovering the effects of anti-MET treatment on ICI therapy, is crucial for enhancing the outlook for tumor patients. In this review, we examine the impact of abnormal activation of the HGF/c-MET signaling pathway on the control of the TME and the processes governing PD-L1 expression in cancer cells. The review thoroughly examines both clinical and practical evidence regarding the use of c-MET inhibitors alongside PD-1/PD-L1 inhibitors, emphasizing that focusing on c-MET with immunotherapy enhances the effectiveness of treating MET tumors exhibiting elevated PD-L1 expression.
Collapse
Affiliation(s)
- Yang Xia
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Chunye Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Min Zhong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Hongguang Zhong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jing Zhou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
16
|
Haas M, Cherfa S, Nguyen L, Bourgoin M, Caron G, Dessauge E, Marchand T, Delpy L, Auberger P, Moreaux J, Jacquel A, Fest T. PIM2 inhibition promotes MCL1 dependency in plasma cells involving integrated stress response-driven NOXA expression. Nat Commun 2025; 16:256. [PMID: 39747141 PMCID: PMC11696207 DOI: 10.1038/s41467-024-55572-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
Our study explores the complex dynamics of the integrated stress response (ISR) axis, highlighting PIM2 kinase's critical role and its interaction with the BCL2 protein family, uncovering key mechanisms of cell survival and tumor progression. Elevated PIM2 expression, a marker of various cancers, often correlates with disease aggressiveness. Using a model of normal and malignant plasma cells, we show that inhibiting PIM2 kinase inhibits phosphorylated BAD production and activates ISR-mediated NOXA expression. This shift towards MCL1 dependence underscores the synergy achieved through combined PIM/MCL1 inhibition, driven largely by ISR-mediated NOXA expression. In mouse xenograft models, dual targeting of PIM2 and MCL1 effectively controls tumor growth-a response reversed by ISR-specific inhibition and upregulation of genes linked to tumor cell dissemination. This work elucidates the molecular intricacies of PIM2 inhibition and its implications for cancer therapy, especially in tumors with elevated PIM2 expression.
Collapse
Affiliation(s)
- Marion Haas
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, F-35043, Rennes, France
- Laboratoire d'hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, F-35033, Rennes, France
| | - Sabrina Cherfa
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, F-35043, Rennes, France
| | - Léa Nguyen
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, F-35043, Rennes, France
- Laboratoire d'hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, F-35033, Rennes, France
| | | | - Gersende Caron
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, F-35043, Rennes, France
- Laboratoire d'hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, F-35033, Rennes, France
| | - Elise Dessauge
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, F-35043, Rennes, France
| | - Tony Marchand
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, F-35043, Rennes, France
- Service d'hématologie clinique, Centre Hospitalier Universitaire, F-35033, Rennes, France
| | - Laurent Delpy
- Université de Limoges, UMR CNRS 7276, INSERM U1262, F-87025, Limoges, France
| | | | - Jérôme Moreaux
- Institut de Génétique Humaine, UMR 9002 CNRS-UM; Pôle de biologie, Centre Hospitalier Universitaire, F-34000, Montpellier, France
| | | | - Thierry Fest
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, UMR_S1236, F-35043, Rennes, France.
- Laboratoire d'hématologie, Pôle de Biologie, Centre Hospitalier Universitaire, F-35033, Rennes, France.
| |
Collapse
|
17
|
Singh S, Nigam V, Kasana S, Kurmi BD, Gupta GD, Patel P. Targeting c-Met in Cancer Therapy: Unravelling Structure-activity Relationships and Docking Insights for Enhanced Anticancer Drug Design. Curr Top Med Chem 2025; 25:409-433. [PMID: 39484763 DOI: 10.2174/0115680266331025241015084546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/02/2024] [Accepted: 09/28/2024] [Indexed: 11/03/2024]
Abstract
The c-Met receptor, a pivotal player in oncogenesis and tumor progression, has become a compelling target for anticancer drug development. This review explores the intricate landscape of Structure-Activity Relationship (SAR) studies and molecular binding analyses performed on c-Met inhibitors. Through a comprehensive examination of various chemical scaffolds and modifications, SAR investigations have elucidated critical molecular features essential for the potent inhibition of c-Met activity. Additionally, molecular docking studies have provided invaluable insights into how c-Met inhibitors interact with their target receptor, facilitating the rational design of novel compounds with enhanced efficacy and selectivity. This review highlights key findings from recent SAR and docking studies, particularly focusing on the structural determinants that govern inhibition potency and selectivity. Furthermore, the integration of computational methodologies with experimental approaches has accelerated the discovery and optimization of c-Met inhibitors, fostering the advancement of promising candidates for clinical applications. Overall, this review underscores the pivotal role of SAR and molecular docking studies in advancing our understanding of c-Met inhibition and guiding the rational design of next-generation anticancer agents targeting this pathway.
Collapse
Affiliation(s)
- Surbhi Singh
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Vaibhav Nigam
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Shivani Kasana
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga-142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry and Analysis, ISF College of Pharmacy, Moga-142001, Punjab, India
| |
Collapse
|
18
|
Zheng S, Li Y, Wang L, Wei Q, Wei M, Yu T, Zhao L. Extrachromosomal circular DNA and their roles in cancer progression. Genes Dis 2025; 12:101202. [PMID: 39534571 PMCID: PMC11554924 DOI: 10.1016/j.gendis.2023.101202] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/26/2023] [Accepted: 11/19/2023] [Indexed: 11/16/2024] Open
Abstract
Extrachromosomal circular DNA (eccDNA), a chromosome-independent circular DNA, has garnered significant attention due to its widespread distribution and intricate biogenesis in carcinoma. Existing research findings propose that multiple eccDNAs contribute to drug resistance in cancer treatments through complex and interrelated regulatory mechanisms. The unique structure and genetic properties of eccDNA increase tumor heterogeneity. This increased diversity is a result of eccDNA's ability to stimulate oncogene remodeling and participate in anomalous splicing processes through chimeric cyclization and the reintegration of loop DNA back into the linear genome. Such actions promote oncogene amplification and silencing. eccDNA orchestrates protein interactions and modulates protein degradation by acting as a regulatory messenger. Moreover, it plays a pivotal role in modeling the tumor microenvironment and intensifying the stemness characteristics of tumor cells. This review presented detailed information about the biogenesis, distinguishing features, and functions of eccDNA, emphasized the role and mechanisms of eccDNA during cancer treatment, and further proposed the great potential of eccDNA in inspiring novel strategies for precision cancer therapy and facilitating the discovery of prognostic biomarkers for cancer.
Collapse
Affiliation(s)
- Siqi Zheng
- Department of Pharmacology, School of Pharmacy, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Yunong Li
- Department of Pharmacology, School of Pharmacy, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Lin Wang
- Department of Pharmacology, School of Pharmacy, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning 110042, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China
| |
Collapse
|
19
|
Kumar M, Kumar A, Srivastav A, Ghosh A, Kumar D. Genomic and molecular landscape of gallbladder cancer elucidating pathogenic mechanisms novel therapeutic targets and clinical implications. Mutat Res 2025; 830:111896. [PMID: 39754821 DOI: 10.1016/j.mrfmmm.2024.111896] [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: 11/04/2024] [Revised: 12/19/2024] [Accepted: 12/19/2024] [Indexed: 01/06/2025]
Abstract
Gallbladder cancer (GBC) is an aggressive malignancy with a poor prognosis, often diagnosed at advanced stages due to subtle early symptoms. Recent studies have provided a comprehensive view of GBC's genetic and mutational landscape, uncovering crucial pathways involved in its pathogenesis. Environmental exposures, particularly to heavy metals, have been linked to elevated GBC risk. Established signaling pathways, including hormonal, apoptotic, metabolic, inflammatory, and DNA damage repair pathways, are integral to GBC progression, and evidence points to the involvement of specific germline and somatic mutations in its development. Key mutations in genes such as KRAS, TP53, IDH1/2, ERBB, PIK3CA, MET, MYC, BRAF, MGMT, CDKN2A and p16 have been identified as contributors to tumorigenesis, with additional alterations including chromosomal aberrations and epigenetic modifications. These molecular insights reveal several potential therapeutic targets that could address the limited treatment options for GBC. Promising therapeutic avenues under investigation include immune checkpoint inhibitors, tyrosine kinase inhibitors, tumor necrosis factor-related apoptosis-inducing ligands (TRAIL), and phytochemicals. Numerous clinical trials are assessing the efficacy of these targeted therapies. This review provides a detailed examination of GBC's genetic and mutational underpinnings, highlighting critical pathways and emerging therapeutic strategies. We discuss the implications of germline and somatic mutations for early detection and individualized treatment, aiming to bridge current knowledge gaps. By advancing our understanding of GBC's molecular profile, we hope to enhance diagnostic accuracy and improve treatment outcomes, ultimately paving the way for precision medicine approaches in managing GBC.
Collapse
Affiliation(s)
- Manishankar Kumar
- School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand 248007, India
| | - Arun Kumar
- Mahavir Cancer Institute and Research Centre, Phulwarisharif, Patna, Bihar 801505, India
| | - Abhinav Srivastav
- Mahavir Cancer Institute and Research Centre, Phulwarisharif, Patna, Bihar 801505, India
| | - Ashok Ghosh
- Mahavir Cancer Institute and Research Centre, Phulwarisharif, Patna, Bihar 801505, India
| | - Dhruv Kumar
- School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand 248007, India.
| |
Collapse
|
20
|
Zhao L, Qian C, Ma X, Wang X. Bioactive Products Targeting C-Met As Potential Antitumour Drugs. Anticancer Agents Med Chem 2025; 25:688-696. [PMID: 39812063 DOI: 10.2174/0118715206346207241217064022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025]
Abstract
Mesenchymal‒epithelial transition factor (c-Met), a receptortyrosine kinase (RTK), plays a vital role in cell proliferation, migration and invasion, and tumour metastasis. OBJECTIVE With increasing duration of treatment, many tumours gradually develop drug resistance. Therefore, novel antitumour drugs need to be developed to treat patients with tumours. Targeting c-met inhibitors may be an effective treatment strategy. METHODS Scientific databases such as ScienceDirect, PubMed, the Wiley Online Library, and Social Sciences Citation Index were used to collect information. All the relevant literature was reviewed, and the available literature was screened. The upstream and downstream pathways of c-Met and their relevance to antitumour effects were searched based on the articles' title, abstract, and full text. The c-Met-targeting drugs with antitumour effects are summarized below. A "citation within a citation" or snowballing approach was used in this screening process to identify additional papers that may have been missed in the initial literature screening process. High-quality studies published in peer-reviewed journals were summarized and prioritized for citation in the review. RESULTS In recent years, research on small-molecule targeted drugs has developed rapidly. Many results have also been achieved in the synthesis and isolation of c-Met inhibitors from natural compounds and traditional Chinese medicines. CONCLUSION This article summarizes the developments in anti-c-Met drugs, which are synthesized and isolated from natural compounds and traditional Chinese medicine (TCM). This study provides primary resources for the development of c-Met inhibitors.
Collapse
Affiliation(s)
- Liying Zhao
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chunmei Qian
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Xiaoqi Ma
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoyu Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
21
|
Al Mashud MA, Devnath R, Anzuman M, Sumona MI, Hossain MS, Kumer A, Talukder MEK, Rahman MM, Imon RR, Akash S, El Moussaoui A, Salamatullah AM, Bourhia M. New Approach as Inhibitor Against Head-Neck Cancer by In silico, DFT, FMOs, Docking, Molecular Dynamic, and ADMET of Euphorbia tirucalli (Pencil Cactus). Med Chem 2025; 21:122-143. [PMID: 40007184 DOI: 10.2174/0115734064315601240628115330] [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: 03/10/2024] [Revised: 05/25/2024] [Accepted: 05/31/2024] [Indexed: 02/27/2025]
Abstract
BACKGROUND Head and neck cancer (HNC) is on the rise worldwide, endangering lives and straining healthcare systems in both developing and developed nations. Despite the availability of a number of therapy options, the success rate for treating and controlling head and neck cancer remains dismal. To combat the aggressiveness and drug resistance of Epstein-Barr virus (EBV)-positive Head-Neck cancer cells, this study looks into the potential of Euphorbia tirucalli (pencil cactus) leaf extract. OBJECTIVES The goal of this study is to identify prospective therapeutic candidates from the extract of Euphorbia tirucalli (pencil cactus) leaves, which have the ability to inhibit Epstein-Barr virus (EBV)-positive Head- Neck cancer cells. MATERIALS AND METHODS The thirteen most important chemical components found in Euphorbia tirucalli (pencil cactus) leaves were analyzed by means of molecular modeling techniques such as Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET), Quantum Mechanics (QM) calculation, molecular docking, and molecular dynamics (MD) simulations. Using the Prediction of Activity Spectra for Substances (PASS) model, we assess the potency of these compounds. Important molecular properties such as chemical potential, electronegativity, hardness, and softness can be determined with the use of quantum chemical calculations employing HOMO-LUMO analysis. These drugs' safety and toxicological characteristics are better understood to assessments of their pharmacokinetics and ADMET. Finally, molecular dynamics simulations are employed to verify binding interactions and assess the stability of docked complexes. RESULTS The molecular docking analysis identifies ligands (01), (02), and (10) as strong competitors, with strong binding affinity for the Epstein-Barr virus (EBV)-positive Head-Neck cancer cell line. Not only do the ligands (01), (02), and (10) match the criteria for a potential new inhibitor of head-neck cancer, but they also outperform the present FDA-approved treatment. CONCLUSION Taraxerol, euphol, and ephorginol, three phytochemicals isolated from the leaves of the Euphorbia tirucalli (pencil cactus), have been identified as effective anti-cancer agents with the potential to serve as a foundation for novel head-neck cancer therapies, particularly those targeting the Epstein-Barr virus (EBV)-overexpressing subtype of this disease. An effective, individualized treatment plan for head-neck cancer is a long way off, but this study is a major step forward that could change the lives of patients and reduce the global burden of this disease.
Collapse
Affiliation(s)
- Md Abdullah Al Mashud
- Biophysics and Biomedicine Research Lab, Department of Electrical and Electronic Engineering, Islamic University, Kushtia-7003, Bangladesh
- Computational Bio-info Lab, Research and Development Center for Sustainability, Scientific Foundation for Cancer Research, Kushtia-7000, Bangladesh
| | - Ramprosad Devnath
- Nanotechnology & Catalysis Research Centre (NANOCAT), Institute for Advanced Studies (IAS), Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Masuma Anzuman
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia-7003, Bangladesh
| | - Mahbuba Iasmin Sumona
- Biophysics and Biomedicine Research Lab, Department of Electrical and Electronic Engineering, Islamic University, Kushtia-7003, Bangladesh
| | - Md Shamim Hossain
- Department of Computer Science and Technology, University of Science and Technology of China, Hefei, Anhui, 230026, P.R. China
| | - Ajoy Kumer
- Laboratory of Computational Research for Drug Design and Material Science, Department of Chemistry, College of Arts and Sciences, IUBAT-International University of Business Agriculture and Technology, 4 Embankment Drive Road, Sector 10, Uttara Model Town, Dhaka 1230, Bangladesh
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Md Enamul Kabir Talukder
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore-7408, Bangladesh
| | - Md Mashiar Rahman
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore-7408, Bangladesh
| | - Raihan Rahman Imon
- Molecular and Cellular Biology Laboratory, Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore-7408, Bangladesh
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Birulia 1216 Ashulia, Dhaka, Bangladesh
| | - Abdelfattah El Moussaoui
- Plant Biotechnology Team, Faculty of Sciences, Abdelmalek Essaadi University, Tetouan 93002, Morocco
| | - Ahmad Mohammad Salamatullah
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, 11 P.O. Box 2460, Riyadh 11451, Saudi Arabia
| | - Mohammed Bourhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, 80060, Agadir, Morocco
| |
Collapse
|
22
|
Pei Y, Mou Z, Jiang L, Yang J, Gu Y, Min J, Sunzhang L, Xiong N, Xu X, Chi H, Xu K, Liu S, Luo H. Aging and head and neck cancer insights from single cell and spatial transcriptomic analyses. Discov Oncol 2024; 15:801. [PMID: 39692961 PMCID: PMC11655923 DOI: 10.1007/s12672-024-01672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma(HNSCC) is the sixth most common malignancy worldwide, with more than 890,000 new cases and 450,000 deaths annually. Its major risk factors include smoking, alcohol abuse, aging, and poor oral hygiene. Due to the lack of early and effective detection and screening methods, many patients are diagnosed at advanced stages with a five-year survival rate of less than 50%. In this study, we deeply explored the expression of Aging-related genes(ARGs) in HNSCC and analyzed their prognostic significance using single-cell sequencing and spatial transcriptomics analysis. This research aims to provide new theoretical support and directions for personalized treatment. Annually, more than 890,000 new cases of head and neck squamous cell carcinoma (HNSCC) are diagnosed globally, leading to 450,000 deaths, making it the sixth most common malignancy worldwide. The primary risk factors for HNSCC include smoking, alcohol abuse, aging, and poor oral hygiene. Many patients are diagnosed at advanced stages due to the absence of early and effective detection and screening methods, resulting in a five-year survival rate of less than 50%. In this research, single cell sequencing and spatial transcriptome analysis were used to investigate the expression of Aging-related genes (ARGs) in HNSCC and to analyse their prognostic significance. This research aims to provide new theoretical support and directions for personalized treatment. METHODS In this study, we investigated the association between HNSCC and AGRs by utilizing the GSE139324 series in the GEO database alongside the TCGA database, combined with single-cell sequencing and spatial transcriptomics analysis. The data were analyzed using Seurat and tSNE tools to reveal intercellular communication networks. For the spatial transcriptome data, SCTransform and RunPCA were applied to examine the metabolic activities of the cells. Gene expression differences were determined through spacerxr and RCTD tools, while the limma package was employed to identify differentially expressed genes and to predict recurrence rates using Cox regression analysis and column line plots. These findings underscore the potential importance of molecular classification, prognostic assessment, and personalized treatment of HNSCC. RESULTS This study utilized HNSCC single-cell sequencing data to highlight the significance of ARGs in the onset and prognosis of HNSCC. It revealed that the proportion of monocytes and macrophages increased, while the proportion of B cells decreased. Notably, high expression of the APOE gene in monocytes was closely associated with patient prognosis. Additionally, a Cox regression model was developed based on GSTP1 and age to provide personalized prediction tools for clinical use in predicting patient survival. CONCLUSIONS We utilized single-cell sequencing and spatial transcriptomics to explore the cellular characteristics of HNSCC and its interaction with the tumor microenvironment. Our findings reveal that HNSCC tissues show increased mononuclear cells and demonstrate enhanced activity in ARGs, thereby advancing our understanding of HNSCC development mechanisms.
Collapse
Affiliation(s)
- Yi Pei
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Zhuying Mou
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Jinyan Yang
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Yuheng Gu
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Jie Min
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Lingyi Sunzhang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Nan Xiong
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Xiang Xu
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China.
| | - Sinian Liu
- Department of Pathology, Xichong People's Hospital, Nanchong, 637200, China.
| | - Huiyan Luo
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China.
| |
Collapse
|
23
|
Li L, Liu Y, Li X, He Y, Tian Z. Effect of group management on disease cognition and fear of disease progression, nutritional status, and quality of life in patients with head and neck tumors. Am J Transl Res 2024; 16:7937-7947. [PMID: 39822501 PMCID: PMC11733339 DOI: 10.62347/cbkg3767] [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: 08/08/2024] [Accepted: 12/02/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVE To investigate the effect of group guided training management on disease cognition and fear of disease progression, nutritional status, and quality of life in patients undergoing chemotherapy for head and neck tumors. METHODS A total of 88 patients diagnosed with malignant head and neck tumors who were admitted to Beijing Tongren Hospital, Capital Medical University from January 2020 to February 2021 were included as the subjects of this study. Patients receiving standard care were set as the control group (n=43), and patients undergoing group education were set as the study group (n=45). The fear of disease progression, level of hope, nutritional status, knowledge, attitude, behaviors, quality of life, self-management efficacy, and adverse reactions were compared between the two groups. Factors affecting patient's prognosis were also analyzed. RESULTS Group guided management for chemotherapy patients with head and neck tumors significantly reduced the fear of disease progression in the study group compared to the control group (P=0.010). Additionally, the study group showed significantly higher levels of hope (P=0.006), nutritional status (P=0.019), nutritional knowledge (P=0.006), positive attitude (P=0.007), and health behavior (P=0.032) than those in the control group. The incidence of malnutrition at 1 month and 3 months after intervention (P=0.005, P=0.009) and adverse reactions (P=0.001) in the study group were significantly lower than those in control group. Furthermore, the quality of life (P=0.011) and self-management efficacy (P=0.008, P=0.019) in the study group were significantly higher than those in the control group. Nursing interventions and self-efficacy were identified as independent risk factors for fear of disease progression and hope level (all P < 0.05). CONCLUSION A group management model, through specialized health education and guidance, can significantly reduce patient's fear of disease progression, alleviate negative emotions, enhance their self-management ability, and improve quality of life. This approach fosters a proactive attitude toward treatment and contributes to better therapeutic outcomes.
Collapse
Affiliation(s)
- Li Li
- Department of ENT, Beijing Tongren Hospital, Capital Medical UniversityBeijing 100730, China
| | - Yongling Liu
- Department of ENT, Beijing Tongren Hospital, Capital Medical UniversityBeijing 100730, China
| | - Xiuya Li
- Department of ENT, Beijing Tongren Hospital, Capital Medical UniversityBeijing 100730, China
| | - Yiran He
- Department of ENT, Beijing Tongren Hospital, Capital Medical UniversityBeijing 100730, China
| | - Zirong Tian
- Department of Nursing, Beijing Tongren Hospital, Capital Medical UniversityBeijing 100730, China
| |
Collapse
|
24
|
Park R, Chung CH. Advanced Human Papillomavirus-Negative Head and Neck Squamous Cell Carcinoma: Unmet Need and Emerging Therapies. Mol Cancer Ther 2024; 23:1717-1730. [PMID: 39301607 PMCID: PMC11612620 DOI: 10.1158/1535-7163.mct-24-0281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/22/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024]
Abstract
Despite notable progress in the treatment of advanced head and neck squamous cell carcinoma (HNSCC), survival remains poor in patients with recurrent and/or metastatic (R/M) human papillomavirus (HPV)-negative HNSCC. Worse outcomes in patients who are HPV-negative may be partly related to loss of cell-cycle regulators and tumor suppressors as well as a noninflamed and hypoxic tumor microenvironment, both of which contribute to treatment resistance and disease progression. Anti-programmed cell death protein 1-based regimens as current standard-of-care treatment for R/M HNSCC are associated with durable responses in a limited number of patients. The anti-EGFR mAb, cetuximab, has antitumor activity in this treatment setting, but responses are short-lived and inevitably curtailed due to treatment resistance. Crosstalk between the EGFR and hepatocyte growth factor-dependent mesenchymal-epithelial transition (c-MET) receptor tyrosine kinase pathway is a known mechanism of resistance to cetuximab. Dual targeting of EGFR and c-MET pathways may overcome resistance to cetuximab in patients with HPV-negative HNSCC. Here, we review clinical data of treatments evaluated in patients with R/M HPV-negative HNSCC and highlight the potential role of combining hepatocyte growth factor/c-MET and EGFR pathway inhibitors to overcome cetuximab resistance in this population.
Collapse
Affiliation(s)
- Robin Park
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Christine H. Chung
- Department of Head and Neck-Endocrine Oncology, Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
25
|
Hao G, Gu F, Hu M, Ding W, Shi W, Dai J, Hou L. Prevalence and assessment of sleep-disordered breathing in head and neck cancer patients: a systematic review. Sleep Breath 2024; 28:2335-2343. [PMID: 39390313 DOI: 10.1007/s11325-024-03075-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 10/12/2024]
Abstract
STUDY OBJECTIVES Sleep-disordered breathing (SDB) is a very common and underdiagnosed condition in head and neck cancers (HNC) patients. If untreated, SDB can lead to negative health consequences. The identification of SDB in HNC patients is crucial to ensure appropriate treatment and to improve outcomes. The purpose of the study was to investigate the incidence of coexisting SDB in HNC patients and to evaluate methods of assessing SDB in the population. METHODS A systematic search of PubMed, Embase, CINAHL, Cochrane Database, the Web of Science, and Scopus was performed for studies related to SDB in HNC patients. In total, 1713 articles were identified. 19 articles were selected for qualitative synthesis. The studies involved 584 subjects. RESULTS The prevalence of SDB ranged from 57 to 90% before cancer treatment and from 12 to 96% after. When using an apnea-hypopnea index (AHI) cut-off ≥ 5/h to diagnosis SDB, the prevalence of SDB was 57-90% before cancer treatment and 12-94% after treatment. Sleep studies using polysomnography are the most commonly used assessment tools, but thresholds for diagnosis have been inconsistent. CONCLUSIONS There is a high prevalence of SDB in HNC patients. However, the diagnostic and thresholds methods used for detecting SDB vary widely. To determine the accurate prevalence of SDB, prospective, systematic studies of SDB in unselected cohorts of HNC participants are required.
Collapse
Affiliation(s)
- Guihua Hao
- Department of Nursing, Shanghai Ninth People's Hospital, School of Medicine, Shanghai jiaotong University, 639 S Zhizaoju Rd, Shanghai, 200011, China
- Shanghai JiaoTong University School of Nursing, No.227 South Chongqing Road, Shanghai, China
| | - Fen Gu
- Department of Nursing, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Min Hu
- Department of Nursing, Shanghai Ninth People's Hospital, School of Medicine, Shanghai jiaotong University, 639 S Zhizaoju Rd, Shanghai, 200011, China
| | - Wenjing Ding
- Shanghai Jiaotong University School of Medicine Library, Shanghai, China
| | - Wentao Shi
- Clinical research unit, Shanghai Ninth People's Hospital, School of Medicine, Shanghai jiaotong University, 639 S Zhizaoju Rd, Shanghai, 200011, China.
| | - Jingjing Dai
- Department of Nursing, Shanghai Ninth People's Hospital, School of Medicine, Shanghai jiaotong University, 639 S Zhizaoju Rd, Shanghai, 200011, China.
| | - Lili Hou
- Department of Nursing, Shanghai Ninth People's Hospital, School of Medicine, Shanghai jiaotong University, 639 S Zhizaoju Rd, Shanghai, 200011, China.
| |
Collapse
|
26
|
Wang J, Xu Q, Yu J, Xu A, Yu L, Chen Z, Cao Y, Yuan R, Yu Z. SCGB1A1 as a novel biomarker and promising therapeutic target for the management of HNSCC. Oncol Lett 2024; 28:527. [PMID: 39268163 PMCID: PMC11391500 DOI: 10.3892/ol.2024.14660] [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: 02/22/2024] [Accepted: 07/18/2024] [Indexed: 09/15/2024] Open
Abstract
Head and neck cancer (HNC) is the sixth most common type of cancer worldwide, and head and neck squamous cell carcinoma (HNSCC) accounts for 90% of HNC cases. Furthermore, HNSCC accounts for 400,000 cancer-associated deaths worldwide each year. However, at present there is an absence of a versatile biomarker that can be used for diagnosis, prognosis evaluation and as a therapeutic target for HNSCC. In the present study, bioinformatics analysis was used to assess the relationship between hub genes and the clinical features of patients with HNSCC. The findings from the bioinformatics analysis were then verified using clinical samples and in vitro experiments. A total of 51 overlapping genes were identified from the intersection of differentially expressed genes and co-expressed genes. The top 10 hub genes were obtained from a protein-protein interaction network of overlapping genes. Among the hub genes, only secretoglobin family 1A member 1 (SCGB1A1) was significantly associated with both overall and disease-free survival. Specifically, upregulated SCGB1A1 expression levels were associated with prolonged overall and disease-free survival. Moreover, the SCGB1A1 expression levels were negatively correlated with drug sensitivity. Notably, it was demonstrated that SCGB1A1 was involved in tumor immunoreaction by affecting the infiltration of cells and checkpoint regulation of immune cells. Additionally, it was shown that SCGB1A1 regulated multiple key cancer-related signaling pathways, including extracellular matrix receptor interaction, transforming growth factor-β and tumor metabolism signaling pathways. Based on the results of the present study, SCGB1A1 may serve as a novel biomarker for predicting the diagnosis, prognosis and therapeutic effectiveness of certain drugs in patients with HNSCC. Moreover, SCGB1A1 may serve as a potential therapeutic target for the management of HNSCC.
Collapse
Affiliation(s)
- Jing Wang
- Center of Oral Medicine, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
- R&D, Shandong Yinfeng Life Science Research Institute, Jinan, Shandong 250000, P.R. China
| | - Qianqian Xu
- Qingdao Cancer Institute, School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Jiangbo Yu
- Center of Oral Medicine, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Aotian Xu
- R&D, Qingdao Sino-cell Biomedicine Co., Ltd., Qingdao, Shandong 266000, P.R. China
| | - Lizheng Yu
- Department of Vascular Surgery, Qingdao Medical College, Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Zhenggang Chen
- Center of Oral Medicine, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Yang Cao
- Center of Oral Medicine, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Rongtao Yuan
- Center of Oral Medicine, Qingdao Municipal Hospital, Qingdao, Shandong 266000, P.R. China
| | - Zhongjie Yu
- R&D, Qingdao Sino-cell Biomedicine Co., Ltd., Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
27
|
Ma Y, Wang R, Liao J, Guo P, Wang Q, Li W. Xanthohumol overcomes osimertinib resistance via governing ubiquitination-modulated Ets-1 turnover. Cell Death Discov 2024; 10:454. [PMID: 39468027 PMCID: PMC11519634 DOI: 10.1038/s41420-024-02220-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is a prevalent and fatal malignancy with a significant global impact. Recent advancements have introduced targeted therapies like tyrosine kinase inhibitors (TKIs) such as osimertinib, which have improved patient outcomes, particularly in those with EGFR mutations. Despite these advancements, acquired resistance to TKIs remains a significant challenge. Hence, one of the current research priorities is understanding the resistance mechanisms and identifying new therapeutic targets to improve therapeutic efficacy. Herein, we identified high expression of c-Met in osimertinib-resistant NSCLC cells, and depletion of c-Met significantly inhibited the proliferation of osimertinib-resistant cells and prolonged survival in mice, suggesting c-Met as an attractive therapeutic target. To identify effective anti-tumor agents targeting c-Met, we screened a compound library containing 641 natural products and found that only xanthohumol exhibited potent inhibitory effects against osimertinib-resistant NSCLC cells. Moreover, combination treatment with xanthohumol and osimertinib sensitized osimertinib-resistant NSCLC cells to osimertinib both in vitro and in vivo. Mechanistically, xanthohumol disrupted the interaction between USP9X and Ets-1, and inhibited the phosphorylation of Ets-1 at Thr38, promoting its degradation, thereby targeting the Ets-1/c-Met signaling axis and inducing intrinsic apoptosis in osimertinib-resistant NSCLC cells. Overall, the research highlights the critical role of targeting c-Met to address osimertinib resistance in NSCLC. By demonstrating the efficacy of xanthohumol in overcoming resistance and enhancing therapeutic outcomes, this study provides valuable insights and potential new strategies for improving the clinical management of NSCLC.
Collapse
Affiliation(s)
- Ying Ma
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- NHC key laboratory of translantional research on transplantation medicine, Department of Transplant Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Ruirui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Pengfei Guo
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Qiang Wang
- NHC key laboratory of translantional research on transplantation medicine, Department of Transplant Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
28
|
Li C, Fang Y, Xu S, Zhao J, Dong D, Li S. Nanomedicine in HNSCC therapy-a challenge to conventional therapy. Front Pharmacol 2024; 15:1434994. [PMID: 39469621 PMCID: PMC11513379 DOI: 10.3389/fphar.2024.1434994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/01/2024] [Indexed: 10/30/2024] Open
Abstract
Squamous cell carcinoma of the head and neck (HNSCC) is a difficult-to-treat cancer and treatment is challenging due to recurrence or metastasis. Therefore, there is an urgent need to explore more effective targeted therapies to improve the clinical outcomes and survival of HNSCC patients. The nanomedicine is emerging as a promising strategy to achieve maximal anti-tumor effect in cancer therapy. In this review, we summarize some important signaling pathways and present the current and potential roles of various nanomaterial drug-delivery formulations in HNSCC treatment, aiming to understand the pathogenesis of HNSCC and further improve the therapeutic efficacy of nanomaterial HNSCC. This article seeks to highlight the exciting potential of novel nanomaterials for targeted cancer therapy in HNSCC and thus provide motivation for further research in this field.
Collapse
Affiliation(s)
- Chenyu Li
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- School of Pharmacy, Dalian Medical University, Dalian, China
| | - Yuan Fang
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Sanchun Xu
- School of Pharmacy, Dalian Medical University, Dalian, China
| | - Jingyuan Zhao
- Clinical Laboratory Center, Central Hospital of Dalian University of Technology, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shuai Li
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
29
|
Naguib BH, Elsebaie HA, Nafie MS, Mohamady S, Albujuq NR, Samir Ayed A, Nada D, Khalil AF, Hefny SM, Tawfik HO, Shaldam MA. Fragment-based design and synthesis of coumarin-based thiazoles as dual c-MET/STAT-3 inhibitors for potential antitumor agents. Bioorg Chem 2024; 151:107682. [PMID: 39137597 DOI: 10.1016/j.bioorg.2024.107682] [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: 06/30/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 08/15/2024]
Abstract
c-MET and STAT-3 are significant targets for cancer treatments. Here, we describe a class of very effective dual STAT-3 and c-MET inhibitors with coumarin-based thiazoles (3a-o) as its scaffold. Spectroscopic evidence (NMR, HRMS, and HPLC) validated the structural discoveries of the new compounds. The cytotoxic activity of these compounds was also tested against a panel of cancer cells in accordance with US-NCI guidelines. Compound 3g proved to be active at 10 µM, thus it was automatically scheduled to be tested at five doses. Towards SNB-75 (CNS cancer cell line), compound 3g showed notable in vitro anti-cancer activity with GI50 = 1.43 μM. For the molecular targets, compound 3g displayed potent activity towards STAT-3 and c-MET having IC50 of 4.7 µM and 12.67, respectively, compared to Cabozantinib (IC50 = 15 nM of c-MET) and STAT-3-IN-3 (IC50 = 2.1 µM of STAT-3). Moreover, compound 3g significantly induced apoptosis in SNB-75 cells, causing a 3.04-fold increase in apoptotic cell death (treated cells exhibited 11.53 % overall apoptosis, against 3.04 % in reference cells) and a 3.58-fold increase in necrosis. Moreover, it arrests cells at the G2 phase. Dual inhibition of c-MET and STAT-3 protein kinase was further validated using RT-PCR. The target compound's binding mechanism was determined by the application of molecular docking.
Collapse
Affiliation(s)
- Bassem H Naguib
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
| | - Heba A Elsebaie
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Samy Mohamady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
| | - Nader R Albujuq
- Department of Chemistry, School of Science, The University of Jordan, Amman 11942, Jordan
| | - Aya Samir Ayed
- Zoology Department, Faculty of Science, Suez Canal University, P.O. 41522, Ismailia, Egypt
| | - Dina Nada
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University, Egypt
| | - Ahmed F Khalil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Salma M Hefny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, P.O. Box 33516, Kafrelsheikh, Egypt
| |
Collapse
|
30
|
Shen Y, You Z, Li L, Tang X, Shan X. The interaction of PRDX1 with Cofilin promotes oral squamous cell carcinoma metastasis. Int J Cancer 2024; 155:1290-1302. [PMID: 38738971 DOI: 10.1002/ijc.34999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024]
Abstract
Peroxiredoxin 1 (PRDX1) is an important member of the peroxiredoxin family (PRDX) and is upregulated in a variety of tumors. Previous studies have found that high PRDX1 expression is closely related to the metastasis of oral squamous cell carcinoma (OSCC), but the specific molecular mechanism is elusive. To elucidate the role of PRDX1 in the metastasis process of OSCC, we evaluated the expression of PRDX1 in OSCC clinical specimens and its impact on the prognosis of OSCC patients. Then, the effect of PRDX1 on OSCC metastasis and cytoskeletal reconstruction was explored in vitro and in nude mouse tongue cancer models, and the molecular mechanisms were also investigated. PRDX1 can directly interact with the actin-binding protein Cofilin, inhibiting the phosphorylation of its Ser3 site, accelerating the depolymerization and turnover of actin, promoting OSCC cell movement, and aggravating the invasion and metastasis of OSCC. In clinical samples and mouse tongue cancer models, PRDX1 also increased lymph node metastasis of OSCC and was negatively correlated with the phosphorylation of Cofilin; PRDX1 also reduced the overall survival rate of OSCC patients. In summary, our study identified that PRDX1 may be a potential therapeutic target to inhibit OSCC metastasis.
Collapse
Affiliation(s)
- Yajun Shen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Zixuan You
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Lingyu Li
- Department of Oral Pathology, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Xiaofei Tang
- Division of Oral Pathology, Beijing Institute of Dental Research, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, China
| | - Xiaofeng Shan
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices& Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|
31
|
Wang L, Chen J, Li Q, Liu A, Lei Z, Li M, Yasin P, Yang S, Ren J, Hu Y, Ren Y, Cheng S, Liu Z. Cigarette smoke extract induces malignant transformation and DNA damage via c-MET phosphorylation in human bronchial epithelial cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 283:116985. [PMID: 39217894 DOI: 10.1016/j.ecoenv.2024.116985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Cigarette smoke, a complex mixture produced by tobacco combustion, contains a variety of carcinogens and can trigger DNA damage. Overactivation of c-MET, a receptor tyrosine kinase, may cause cancer and cellular DNA damage, but the underlying mechanisms are unknown. In this work, we investigated the mechanisms of cigarette smoke extract (CSE) induced malignant transformation and DNA damage in human bronchial epithelial cells (BEAS-2B). The results demonstrated that CSE treatment led to up-regulated mRNA expression of genes associated with the c-MET signaling pathway, increased expression of the DNA damage sensor protein γ-H2AX, and uncontrolled proliferation in BEAS-2B cells. ATR, ATR, and CHK2, which are involved in DNA damage repair, as well as the phosphorylation of c-MET and a group of kinases (ATM, ATR, CHK1, CHK2) involved in the DNA damage response were all activated by CSE. In addition, CSE activation promotes the phosphorylation modification of ATR, CHK1 proteins associated with DNA damage repair. The addition of PHA665752, a specific inhibitor of c-MET, or knock-down with c-MET both attenuated DNA damage, while overexpression of c-MET exacerbated DNA damage. Thus, c-MET phosphorylation may be involved in CSE-induced DNA damage, providing a potential target for intervention in the prevention and treatment of smoking-induced lung diseases.
Collapse
Affiliation(s)
- Li Wang
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China; School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Jin Chen
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Qianhui Li
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China; School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Anfei Liu
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Zhenhan Lei
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Meixin Li
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Pazilat Yasin
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Shuo Yang
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Jing Ren
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Yijie Hu
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Yihui Ren
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China
| | - Suizhi Cheng
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China; School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Zhenzhong Liu
- School of Public Health, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
32
|
Shao C, Chen J, Qiang B, Ye J, Yan F, Zhu Y. The role of cGAS-STING signaling in the development and therapy of head and neck squamous cell carcinoma. Front Immunol 2024; 15:1451305. [PMID: 39295867 PMCID: PMC11408205 DOI: 10.3389/fimmu.2024.1451305] [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: 06/19/2024] [Accepted: 08/19/2024] [Indexed: 09/21/2024] Open
Abstract
The cGAS-STING signaling pathway plays a critical role in innate immunity and defense against viral infections by orchestrating intracellular and adaptive immune responses to DNA. In the context of head and neck squamous cell carcinoma (HNSCC), this pathway has garnered significant attention due to its potential relevance in disease development and progression. HNSCC is strongly associated with risk factors such as smoking, heavy alcohol consumption, and human papillomavirus (HPV) infection. The presence or absence of HPV in HNSCC patients has been shown to have a profound impact on patient survival and prognosis, possibly due to the distinct biological characteristics of HPV-associated tumors. This review aims to provide a comprehensive overview of the current therapeutic approaches and challenges in HNSCC management, as well as the involvement of cGAS-STING signaling and its potential in the therapy of HNSCC. In addition, by advancing the present understanding of the mechanisms underlying this pathway, Activation of cGAS-STING-dependent inflammatory signaling downstream of chromosomal instability can exert both anti-tumoral and pro-tumoral effects in a cell-intrinsic manner, suggesting individualized therapy is of great importance. However, further exploration of the cGAS-STING signaling pathway is imperative for the effective management of HNSCC.
Collapse
Affiliation(s)
- Chengze Shao
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Jiawen Chen
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Bi Qiang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Junmei Ye
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fangrong Yan
- Research Center of Biostatistics and Computational Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yongbo Zhu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
33
|
Lin Y, Huang Y, Yang B, Zhang Y, Ji N, Li J, Zhou Y, Shen YQ, Chen Q. Precision therapy targeting CAMK2 to overcome resistance to EGFR inhibitors in FAT1-mutated oral squamous cell carcinoma. Chin Med J (Engl) 2024:00029330-990000000-01204. [PMID: 39227322 DOI: 10.1097/cm9.0000000000003217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a prevalent type of cancer with a high mortality rate in its late stages. One of the major challenges in OSCC treatment is the resistance to epidermal growth factor receptor (EGFR) inhibitors. Therefore, it is imperative to elucidate the mechanism underlying drug resistance and develop appropriate precision therapy strategies to enhance clinical efficacy. METHODS To evaluate the efficacy of the combination of the Ca2+/calmodulin-dependent protein kinase II (CAMK2) inhibitor KN93 and EGFR inhibitors, we performed in vitro and in vivo experiments using two FAT atypical cadherin 1 (FAT1)-deficient (SCC9 and SCC25) and two FAT1 wild-type (SCC47 and HN12) OSCC cell lines. We assessed the effects of EGFR inhibitors (afatinib or cetuximab), KN93, or their combination on the malignant phenotype of OSCC in vivo and in vitro. The alterations in protein expression levels of members of the EGFR signaling pathway and SRY-box transcription factor 2 (SOX2) were analyzed. Changes in the yes-associated protein 1 (YAP1) protein were characterized. Moreover, we analyzed mitochondrial dysfunction. Besides, the effects of combination therapy on mitochondrial dynamics were also evaluated. RESULTS OSCC with FAT1 mutations exhibited resistance to EGFR inhibitors treatment. The combination of KN93 and EGFR inhibitors significantly inhibited the proliferation, survival, and migration of FAT1-mutated OSCC cells and suppressed tumor growth in vivo. Mechanistically, combination therapy enhanced the therapeutic sensitivity of FAT1-mutated OSCC cells to EGFR inhibitors by modulating the EGFR pathway and downregulated tumor stemness-related proteins. Furthermore, combination therapy induced reactive oxygen species (ROS)-mediated mitochondrial dysfunction and disrupted mitochondrial dynamics, ultimately resulting in tumor suppression. CONCLUSION Combination therapy with EGFR inhibitors and KN93 could be a novel precision therapeutic strategy and a potential clinical solution for EGFR-resistant OSCC patients with FAT1 mutations.
Collapse
Affiliation(s)
- Yumeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences, Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Liu Y, Zhang N, Wen Y, Wen J. Head and neck cancer: pathogenesis and targeted therapy. MedComm (Beijing) 2024; 5:e702. [PMID: 39170944 PMCID: PMC11338281 DOI: 10.1002/mco2.702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
Head and neck cancer (HNC) is a highly aggressive type of tumor characterized by delayed diagnosis, recurrence, metastasis, relapse, and drug resistance. The occurrence of HNC were associated with smoking, alcohol abuse (or both), human papillomavirus infection, and complex genetic and epigenetic predisposition. Currently, surgery and radiotherapy are the standard treatments for most patients with early-stage HNC. For recurrent or metastatic (R/M) HNC, the first-line treatment is platinum-based chemotherapy combined with the antiepidermal growth factor receptor drug cetuximab, when resurgery and radiation therapy are not an option. However, curing HNC remains challenging, especially in cases with metastasis. In this review, we summarize the pathogenesis of HNC, including genetic and epigenetic changes, abnormal signaling pathways, and immune regulation mechanisms, along with all potential therapeutic strategies such as molecular targeted therapy, immunotherapy, gene therapy, epigenetic modifications, and combination therapies. Recent preclinical and clinical studies that may offer therapeutic strategies for future research on HNC are also discussed. Additionally, new targets and treatment methods, including antibody-drug conjugates, photodynamic therapy, radionuclide therapy, and mRNA vaccines, have shown promising results in clinical trials, offering new prospects for the treatment of HNC.
Collapse
Affiliation(s)
- Yan Liu
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengduChina
- National Facility for Translational Medicine (Sichuan)West China Hospital of Sichuan UniversityChengduChina
| | - Nannan Zhang
- National Center for Birth Defect MonitoringKey Laboratory of Birth Defects and Related Diseases of Women and ChildrenMinistry of EducationWest China Second University HospitalSichuan UniversityChengduChina
| | - Yi Wen
- State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Jiaolin Wen
- Frontiers Medical CenterTianfu Jincheng LaboratoryChengduChina
| |
Collapse
|
35
|
Gowda SV, Kim NY, Harsha KB, Gowda D, Suresh RN, Deivasigamani A, Mohan CD, Hui KM, Sethi G, Ahn KS, Rangappa KS. A new 1,2,3-triazole-indirubin hybrid suppresses tumor growth and pulmonary metastasis by mitigating the HGF/c-MET axis in hepatocellular carcinoma. J Adv Res 2024:S2090-1232(24)00377-1. [PMID: 39216686 DOI: 10.1016/j.jare.2024.08.033] [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: 05/01/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is a fatal cancer that is often diagnosed at the advanced stages which limits the available therapeutic options. The interaction of HGF with c-MET (a receptor tyrosine kinase) results in the activation of c-MET which subsequently triggers the PI3K/Akt/mTOR axis. Overexpression of c-MET in HCC tissues has been demonstrated to contribute to tumor progression and metastasis. OBJECTIVES We aimed to synthesize triazole-indirubin conjugates, examine their growth suppressor efficacy in cell-based assays, and investigate the antitumor as well as antimetastatic activity of lead cytotoxic agent in the orthotopic mice model. METHODS A series of triazole-indirubin hybrids were synthesized and cytotoxicity, apoptogenic, and antimigratory effect of the lead compound (CRI9) was evaluated using MTT assay, cell cycle analysis, annexin-V/PI assay, TUNEL assay, and wound healing assay. The effect of CRI9 on the operation of the HGF/c-MET/PI3K/Akt/mTOR axis was examined using western blotting and transfection experiments. Acute toxicity, antitumor, and antimetastatic activity of CRI9 were examined in NCr nude mice. The expression of c-MET/PI3K/Akt/mTOR, CD31, and Ki-67 was examined using immunohistochemistry and western blotting. RESULTS Among the new compounds, CRI9 consistently displayed potent cytotoxicity against HGF-induced HCC cells. CRI9 induced apoptosis as evidenced by increased sub G1 cells, annexin-V+/PI+ cells, TUNEL+ cells, and cleavage of procaspase-3 and PARP. CRI9 inhibited HGF-induced phosphorylation of c-METY1234/1235 and subsequently suppressed the PI3K/Akt/mTOR axis. Also, depletion of c-MET or inhibition of c-MET by CRI9 resulted in suppression of the PI3K/Akt/mTOR axis. CRI9 showed no toxic effects in NCr nude mice and displayed a potent antitumor and antimetastatic effect in the orthotopic HCC mice model. CRI9 also reduced the levels of phospho-c-MET, CD31, and Ki-67 and suppressed the activation of the PI3K/Akt/mTOR axis in tumor tissues. CONCLUSION CRI9 has been identified as a new inhibitor of the c-MET/PI3K/Akt/mTOR axis in HCC preclinical models.
Collapse
Affiliation(s)
- Shalini V Gowda
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - Na Young Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Kachigere B Harsha
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - Darshini Gowda
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - Rajaghatta N Suresh
- Department of Studies in Chemistry, University of Mysore, Manasagangotri, Mysore 570006, Karnataka, India
| | - Amudha Deivasigamani
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Chakrabhavi Dhananjaya Mohan
- Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226 001, Uttar Pradesh, India
| | - Kam Man Hui
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore 169610, Singapore.
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | | |
Collapse
|
36
|
Yao S, Liu X, Feng Y, Li Y, Xiao X, Han Y, Xia S. Unveiling the Role of HGF/c-Met Signaling in Non-Small Cell Lung Cancer Tumor Microenvironment. Int J Mol Sci 2024; 25:9101. [PMID: 39201787 PMCID: PMC11354629 DOI: 10.3390/ijms25169101] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is characterized by several molecular alterations that contribute to its development and progression. These alterations include the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), human epidermal growth factor receptor 2 (HER2), and mesenchymal-epithelial transition factor (c-MET). Among these, the hepatocyte growth factor (HGF)/c-MET signaling pathway plays a crucial role in NSCLC. In spite of this, the involvement of the HGF/c-MET signaling axis in remodeling the tumor microenvironment (TME) remains relatively unexplored. This review explores the biological functions of the HGF/c-MET signaling pathway in both normal and cancerous cells, examining its multifaceted roles in the NSCLC tumor microenvironment, including tumor cell proliferation, migration and invasion, angiogenesis, and immune evasion. Furthermore, we summarize the current progress and clinical applications of MET-targeted therapies in NSCLC and discuss future research directions, such as the development of novel MET inhibitors and the potential of combination immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shu Xia
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (S.Y.); (X.L.); (Y.F.); (Y.L.); (X.X.); (Y.H.)
| |
Collapse
|
37
|
Otálora-Otálora BA, Payán-Gómez C, López-Rivera JJ, Pedroza-Aconcha NB, Aristizábal-Guzmán C, Isaza-Ruget MA, Álvarez-Moreno CA. Global transcriptomic network analysis of the crosstalk between microbiota and cancer-related cells in the oral-gut-lung axis. Front Cell Infect Microbiol 2024; 14:1425388. [PMID: 39228892 PMCID: PMC11368877 DOI: 10.3389/fcimb.2024.1425388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/15/2024] [Indexed: 09/05/2024] Open
Abstract
Background The diagnosis and treatment of lung, colon, and gastric cancer through the histologic characteristics and genomic biomarkers have not had a strong impact on the mortality rates of the top three global causes of death by cancer. Methods Twenty-five transcriptomic analyses (10 lung cancer, 10 gastric cancer, and 5 colon cancer datasets) followed our own bioinformatic pipeline based on the utilization of specialized libraries from the R language and DAVID´s gene enrichment analyses to identify a regulatory metafirm network of transcription factors and target genes common in every type of cancer, with experimental evidence that supports its relationship with the unlocking of cell phenotypic plasticity for the acquisition of the hallmarks of cancer during the tumoral process. The network's regulatory functional and signaling pathways might depend on the constant crosstalk with the microbiome network established in the oral-gut-lung axis. Results The global transcriptomic network analysis highlighted the impact of transcription factors (SOX4, TCF3, TEAD4, ETV4, and FOXM1) that might be related to stem cell programming and cancer progression through the regulation of the expression of genes, such as cancer-cell membrane receptors, that interact with several microorganisms, including human T-cell leukemia virus 1 (HTLV-1), the human papilloma virus (HPV), the Epstein-Barr virus (EBV), and SARS-CoV-2. These interactions can trigger the MAPK, non-canonical WNT, and IFN signaling pathways, which regulate key transcription factor overexpression during the establishment and progression of lung, colon, and gastric cancer, respectively, along with the formation of the microbiome network. Conclusion The global transcriptomic network analysis highlights the important interaction between key transcription factors in lung, colon, and gastric cancer, which regulates the expression of cancer-cell membrane receptors for the interaction with the microbiome network during the tumorigenic process.
Collapse
Affiliation(s)
| | - César Payán-Gómez
- Dirección Académica, Universidad Nacional de Colombia, Sede de La Paz, La Paz, Colombia
| | - Juan Javier López-Rivera
- Grupo de Investigación INPAC, Specialized Laboratory, Clinica Universitaria Colombia, Clínica Colsanitas S.A., Bogotá, Colombia
| | | | - Claudia Aristizábal-Guzmán
- Grupo de Investigación INPAC, Unidad de Investigación, Fundación Universitaria Sanitas, Bogotá, Colombia
| | - Mario Arturo Isaza-Ruget
- Keralty, Sanitas International Organization, Grupo de Investigación INPAC, Fundación Universitaria Sanitas, Bogotá, Colombia
| | | |
Collapse
|
38
|
Le Maout C, Fahy L, Renou L, Devanand C, Duwat C, Barroca V, Le Gall M, Ballerini P, Petit A, Calvo J, Uzan B, Pflumio F, Petit V. T-cell acute lymphoblastic leukemia progression is supported by inflammatory molecules including hepatocyte growth factor. Biomed Pharmacother 2024; 177:117039. [PMID: 38955085 DOI: 10.1016/j.biopha.2024.117039] [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: 04/30/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is a malignant hematological disorder characterized by an increased proliferation of immature T lymphocytes precursors. T-ALL treatment includes chemotherapy with strong side effects, and patients that undergo relapse display poor prognosis. Although cell-intrinsic oncogenic pathways are well-studied, the tumor microenvironment, like inflammatory cellular and molecular components is less explored in T-ALL. We sought to determine the composition of the inflammatory microenvironment induced by T-ALL, and its role in T-ALL progression. We show in two mouse T-ALL cell models that T-ALLs enhance blood neutrophils and resident monocytes, accompanied with a plasmatic acute secretion of inflammatory molecules. Depleting neutrophils using anti-Ly6G treatment or resident monocytes by clodronate liposomes treatment does not modulate plasmatic inflammatory molecule secretion and mice survival. However, inhibiting the secretion of inflammatory molecules by microenvironment with NECA, an agonist of adenosine receptors, diminishes T-ALL progression enhancing mouse survival. We uncovered Hepatocyte Growth Factor (HGF), T-ALL-driven and the most decreased molecule with NECA, as a potential therapeutic target in T-ALL. Altogether, we identified a signature of inflammatory molecules that can potentially be involved in T-ALL evolution and uncovered HGF/cMET pathway as important to target for limiting T-ALL progression.
Collapse
Affiliation(s)
- Charly Le Maout
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France
| | - Lucine Fahy
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France
| | - Laurent Renou
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France
| | - Caroline Devanand
- CEA, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Plateforme d'expérimentation animale, Fontenay-aux-Roses, France
| | - Charlotte Duwat
- CEA, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Plateforme d'expérimentation animale, Fontenay-aux-Roses, France
| | - Vilma Barroca
- CEA, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Plateforme d'expérimentation animale, Fontenay-aux-Roses, France
| | - Morgane Le Gall
- Proteom'IC facility, Université Paris Cité, CNRS, Inserm, Institut Cochin, Paris F-75014, France
| | - Paola Ballerini
- Service D'hématologie Pédiatrique, Assistance Publique - Hôpitaux de Paris, Hôpital A. Trousseau, Paris, France
| | - Arnaud Petit
- Service D'hématologie Pédiatrique, Assistance Publique - Hôpitaux de Paris, Hôpital A. Trousseau, Paris, France
| | - Julien Calvo
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France; Institut Carnot OPALE, Hôpital Saint Louis, Paris F-75020, France
| | - Benjamin Uzan
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France; Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, Paris F-75013, France
| | - Françoise Pflumio
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire des cellules Souches Hématopoïétiques et des Leucémies (LSHL), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Fontenay-aux-Roses F-92260, France; CEA, Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), Plateforme d'expérimentation animale, Fontenay-aux-Roses, France; Institut Carnot OPALE, Hôpital Saint Louis, Paris F-75020, France.
| | - Vanessa Petit
- Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, iRCM/IBFJ, Fontenay-aux-Roses F-92260, France; Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, Laboratoire Réparation et Transcription dans les cellules Souches (LRTS), Institut de Radiobiologie Cellulaire et Moléculaire (iRCM), Institut de Biologie François Jacob (IBFJ), France.
| |
Collapse
|
39
|
Danishuddin, Haque MA, Malik MZ, Arya R, Singh P, Lee JS, Kim JJ, Lee KW, Jung TS. Unveiling the Mechanisms Underlying the Immunotherapeutic Potential of Gene-miRNA and Drugs in Head and Neck Cancer. Pharmaceuticals (Basel) 2024; 17:921. [PMID: 39065771 PMCID: PMC11280033 DOI: 10.3390/ph17070921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Head and neck cancer ranks as the sixth-most common malignancy worldwide, characterized by high mortality and recurrence rates. Research studies indicate that molecular diagnostics play a crucial role in the early detection and prognostic evaluation of these diseases. This study aimed to identify potential biomarkers for head and neck cancer and elucidate their interactions with miRNAs and possible therapeutic drugs. Four drivers, namely, FN1, IL1A, COL1A1, and MMP9, were identified using network biology and machine learning approaches. Gene set variation analysis (GSVA) showed that these genes were significantly involved in different biological processes and pathways, including coagulation, UV-response-down, apoptosis, NOTCH signaling, Wnt-beta catenin, and other signal pathways. The diagnostic value of these hub genes was validated using receiver operating characteristic (ROC) curves. The top interactive miRNAs, including miR-128-3p, miR-218-5p, miR-214-3p, miR-124-3p, miR-129-2-3p, and miR-1-3p, targeted the key genes. Furthermore, the interaction between the key genes and drugs was also identified. In summary, the key genes and miRNAs or drugs reported in this study might provide valuable information for potential biomarkers to increase the prognosis and diagnosis of head and neck cancer.
Collapse
Affiliation(s)
- Danishuddin
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (D.); (M.A.H.); (R.A.)
| | - Md Azizul Haque
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (D.); (M.A.H.); (R.A.)
| | - Md. Zubbair Malik
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman 15462, Kuwait;
| | - Rakesh Arya
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (D.); (M.A.H.); (R.A.)
| | - Pooja Singh
- Division of Applied Life Science (BK21 Four), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Gyeongsang National University (GNU), Jinju 52828, Republic of Korea;
| | - Jeong-Sang Lee
- GSCRO, Research Spin-Off Company, Innopolis Jeonbuk, Jeonju 55069, Republic of Korea;
- Department of Food and Nutrition, College of Medical Science, Jeonju University, Jeonju 55069, Republic of Korea
| | - Jong-Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea; (D.); (M.A.H.); (R.A.)
| | - Keun-Woo Lee
- Korea Quantum Computing (KQC), Busan 48058, Republic of Korea
- Angel i-Drug Design (AiDD), Jinju 52650, Republic of Korea
| | - Tae-Sung Jung
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University (GNU), Jinju 52828, Republic of Korea
| |
Collapse
|
40
|
Desilets A, Pfister DG, Stein S, Wong W, Sherman EJ, Fetten J, Hung TKW, Kriplani A, Dunn LA, Ho AL, Michel LS. A phase 1 study of concurrent cabozantinib and cetuximab in recurrent or metastatic head and neck squamous cell cancer. Oral Oncol 2024; 154:106861. [PMID: 38795600 PMCID: PMC11235871 DOI: 10.1016/j.oraloncology.2024.106861] [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: 09/12/2023] [Revised: 05/14/2024] [Accepted: 05/18/2024] [Indexed: 05/28/2024]
Abstract
OBJECTIVES Epidermal growth factor receptor (EGFR) inhibition with cetuximab is a standard treatment for head and neck squamous cell carcinoma (HNSCC). Activation of the receptor tyrosine kinases AXL, MET and VEGFR can mediate resistance to cetuximab. Cabozantinib, a multikinase inhibitor (MKI) targeting AXL/MET/VEGFR, has demonstrated antitumor activity in preclinical models of HNSCC. This investigator- initiated phase I trial evaluated the safety and efficacy of cetuximab plus cabozantinib in patients with recurrent/metastatic (R/M) HNSCC. MATERIALS AND METHODS Patients received cetuximab concurrently with cabozantinib daily on a 28-day cycle. Using a 3 + 3 dose-escalation design, the primary endpoint was to determine the maximally tolerated dose (MTD) of cabozantinib. Secondary endpoints included overall response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS) RESULTS: Among the 20 patients enrolled, most had prior disease progression on immune checkpoint inhibitors (95 %), platinum-based chemotherapy (95 %), and cetuximab (80 %). No dose-limiting toxicities were recorded and the MTD for cabozantinib was established to be 60 mg. Grade ≥ 3 adverse events occurred in 65 % of patients (n = 13). ORR was 20 %, with 4 partial responses (PRs). Two PRs were observed in cetuximab-naïve patients (n = 4), with an ORR of 50 % in this subgroup. In the overall population, DCR was 75 %, median PFS was 3.4 months and median OS was 8.1 months. CONCLUSION Cetuximab plus cabozantinib demonstrated a manageable toxicity profile and preliminary efficacy in patients with heavily treated R/M HNSCC. The combination of cetuximab with MKIs targeting the AXL/MET/VEGFR axis warrants further investigation, including in cetuximab-naïve patients.
Collapse
Affiliation(s)
- Antoine Desilets
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States.
| | - David G Pfister
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, United States
| | - Sarah Stein
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States
| | - Winston Wong
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, United States
| | - Eric J Sherman
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, United States
| | - James Fetten
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, United States
| | - Tony K W Hung
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, United States
| | - Anuja Kriplani
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, United States
| | - Lara A Dunn
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, United States
| | - Alan L Ho
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, United States
| | - Loren S Michel
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, United States; Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, United States
| |
Collapse
|
41
|
El Herch I, Tornaas S, Dongre HN, Costea DE. Heterogeneity of cancer-associated fibroblasts and tumor-promoting roles in head and neck squamous cell carcinoma. Front Mol Biosci 2024; 11:1340024. [PMID: 38966131 PMCID: PMC11222324 DOI: 10.3389/fmolb.2024.1340024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 05/23/2024] [Indexed: 07/06/2024] Open
Abstract
Tumor microenvironment (TME) in head and neck squamous cell carcinoma (HNSCC) has a major influence on disease progression and therapy response. One of the predominant stromal cell types in the TME of HNSCC is cancer-associated fibroblasts (CAF). CAF constitute a diverse cell population and we are only at the beginning of characterizing and understanding the functions of various CAF subsets. CAF have been shown to interact with tumor cells and other components of the TME to shape mainly a favourable microenvironment for HNSCC progression, although some studies report existence of tumor-restraining CAF subtypes. The numerous pathways used by CAF to promote tumorigenesis may represent potential therapeutic targets. This review summarizes current knowledge on the origins, subtypes and mechanisms employed by CAF in HNSCC. The aim is to contribute to the understanding on how CAF actively influence the TME and modulate different immune cell types, as well as cancer cells, to establish a conducive setting for cancer growth. Although CAF are currently a promising therapeutic target for the treatment of other types of cancer, there is no significant therapeutic advancement in HNSCC.
Collapse
Affiliation(s)
- Imane El Herch
- University of Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- Gade Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Stian Tornaas
- Gade Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Harsh Nitin Dongre
- Gade Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| | - Daniela Elena Costea
- Gade Laboratory for Pathology, Department of Clinical Medicine, Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Faculty of Medicine and Dentistry, University of Bergen, Bergen, Norway
| |
Collapse
|
42
|
Mohan CD, Shanmugam MK, Gowda SGS, Chinnathambi A, Rangappa KS, Sethi G. c-MET pathway in human malignancies and its targeting by natural compounds for cancer therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155379. [PMID: 38503157 DOI: 10.1016/j.phymed.2024.155379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 03/21/2024]
Abstract
BACKGROUND c-MET is a receptor tyrosine kinase which is classically activated by HGF to activate its downstream signaling cascades such as MAPK, PI3K/Akt/mTOR, and STAT3. The c-MET modulates cell proliferation, epithelial-mesenchymal transition (EMT), immune response, morphogenesis, apoptosis, and angiogenesis. The c-MET has been shown to serve a prominent role in embryogenesis and early development. The c-MET pathway is deregulated in a broad range of malignancies, due to overexpression of ligands or receptors, genomic amplification, and MET mutations. The link between the deregulation of c-MET signaling and tumor progression has been well-documented. Overexpression or overactivation of c-MET is associated with dismal clinical outcomes and acquired resistance to targeted therapies. Since c-MET activation results in the triggering of oncogenic pathways, abrogating the c-MET pathway is considered to be a pivotal strategy in cancer therapeutics. Herein, an analysis of role of the c-MET pathway in human cancers and its relevance in bone metastasis and therapeutic resistance has been undertaken. Also, an attempt has been made to summarize the inhibitory activity of selected natural compounds towards c-MET signaling in cancers. METHODS The publications related to c-MET pathway in malignancies and its natural compound modulators were obtained from databases such as PubMed, Scopus, and Google Scholar and summarized based on PRISMA guidelines. Some of the keywords used for extracting relevant literature are c-MET, natural compound inhibitors of c-MET, c-MET in liver cancer, c-MET in breast cancer, c-MET in lung cancer, c-MET in pancreatic cancer, c-MET in head and neck cancer, c-MET in bone metastasis, c-MET in therapeutic resistance, and combination of c-MET inhibitors and chemotherapeutic agents. The chemical structure of natural compounds was verified in PubChem database. RESULTS The search yielded 3935 publications, of which 195 reference publications were used for our analysis. Clinical trials were referenced using ClinicalTrials.gov identifier. The c-MET pathway has been recognized as a prominent target to combat the growth, metastasis, and chemotherapeutic resistance in cancers. The key role of the c-MET in bone metastasis as well as therapeutic resistance has been elaborated. Also, suppressive effect of selected natural compounds on the c-MET pathway in clinical/preclinical studies has been discussed.
Collapse
Affiliation(s)
- Chakrabhavi Dhananjaya Mohan
- FEST Division, CSIR-Indian Institute of Toxicology Research, Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226 001, India
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | | | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Kanchugarakoppal S Rangappa
- Institution of Excellence, Vijnana Bhavan, University of Mysore, Manasagangotri, Mysore, Karnataka 570006, India.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| |
Collapse
|
43
|
Jing N, Du X, Liang Y, Tao Z, Bao S, Xiao H, Dong B, Gao WQ, Fang YX. PAX6 promotes neuroendocrine phenotypes of prostate cancer via enhancing MET/STAT5A-mediated chromatin accessibility. J Exp Clin Cancer Res 2024; 43:144. [PMID: 38745318 PMCID: PMC11094950 DOI: 10.1186/s13046-024-03064-1] [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: 01/23/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Neuroendocrine prostate cancer (NEPC) is a lethal subset of prostate cancer which is characterized by neuroendocrine differentiation and loss of androgen receptor (AR) signaling. Growing evidence reveals that cell lineage plasticity is crucial in the failure of NEPC therapies. Although studies suggest the involvement of the neural transcription factor PAX6 in drug resistance, its specific role in NEPC remains unclear. METHODS The expression of PAX6 in NEPC was identified via bioinformatics and immunohistochemistry. CCK8 assay, colony formation assay, tumorsphere formation assay and apoptosis assay were used to illustrate the key role of PAX6 in the progression of in vitro. ChIP and Dual-luciferase reporter assays were conducted to confirm the binding sequences of AR in the promoter region of PAX6, as well as the binding sequences of PAX6 in the promoter regions of STAT5A and MET. For in vivo validation, the xenograft model representing NEPC subtype underwent pathological analysis to verify the significant role of PAX6 in disease progression. Complementary diagnoses were established through public clinical datasets and transcriptome sequencing of specific cell lines. ATAC-seq was used to detect the chromatin accessibility of specific cell lines. RESULTS PAX6 expression was significantly elevated in NEPC and negatively regulated by AR signaling. Activation of PAX6 in non-NEPC cells led to NE trans-differentiation, while knock-down of PAX6 in NEPC cells inhibited the development and progression of NEPC. Importantly, loss of AR resulted in an enhanced expression of PAX6, which reprogramed the lineage plasticity of prostate cancer cells to develop NE phenotypes through the MET/STAT5A signaling pathway. Through ATAC-seq, we found that a high expression level of PAX6 elicited enhanced chromatin accessibility, mainly through attenuation of H4K20me3, which typically causes chromatin silence in cancer cells. CONCLUSION This study reveals a novel neural transcription factor PAX6 could drive NEPC progression and suggest that it might serve as a potential therapeutic target for the management of NEPC.
Collapse
Affiliation(s)
- Nan Jing
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
- Med-X Research Institutes, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yu Liang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - ZhenKeke Tao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Shijia Bao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huixiang Xiao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Baijun Dong
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei-Qiang Gao
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China.
- Med-X Research Institutes, Shanghai Jiao Tong University, Shanghai, 200030, China.
| | - Yu-Xiang Fang
- State Key Laboratory of Systems Medicine for Cancer, Renji-Med-X Stem Cell Research Center, Ren Ji Hospital, School of Medicine, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
44
|
Jing W, Wang G, Cui Z, Li X, Zeng S, Jiang X, Li W, Han B, Xing N, Zhao Y, Chen S, Shi B. Tumor-neutrophil cross talk orchestrates the tumor microenvironment to determine the bladder cancer progression. Proc Natl Acad Sci U S A 2024; 121:e2312855121. [PMID: 38713626 PMCID: PMC11098120 DOI: 10.1073/pnas.2312855121] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 03/13/2024] [Indexed: 05/09/2024] Open
Abstract
The immune landscape of bladder cancer progression is not fully understood, and effective therapies are lacking in advanced bladder cancer. Here, we visualized that bladder cancer cells recruited neutrophils by secreting interleukin-8 (IL-8); in turn, neutrophils played dual functions in bladder cancer, including hepatocyte growth factor (HGF) release and CCL3highPD-L1high super-immunosuppressive subset formation. Mechanistically, c-Fos was identified as the mediator of HGF up-regulating IL-8 transcription in bladder cancer cells, which was central to the positive feedback of neutrophil recruitment. Clinically, compared with serum IL-8, urine IL-8 was a better biomarker for bladder cancer prognosis and clinical benefit of immune checkpoint blockade (ICB). Additionally, targeting neutrophils or hepatocyte growth factor receptor (MET) signaling combined with ICB inhibited bladder cancer progression and boosted the antitumor effect of CD8+ T cells in mice. These findings reveal the mechanism by which tumor-neutrophil cross talk orchestrates the bladder cancer microenvironment and provide combination strategies, which may have broad impacts on patients suffering from malignancies enriched with neutrophils.
Collapse
Affiliation(s)
- Weiqiang Jing
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Ganyu Wang
- Department of Pediatric Surgery, Qilu Hospital Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Zhiwei Cui
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Xinyuan Li
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Shuyan Zeng
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Xin Jiang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Wushan Li
- Department of Obstetrics, Jinan Maternity and Child Care Hospital Shandong First Medical University, Jinan, Shandong Province250000, China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Nianzeng Xing
- Department of Urology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing10021, China
| | - Yunxue Zhao
- Department of Immunology, Shandong Provincial Key Laboratory of Infection & Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Shouzhen Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province250012, China
| |
Collapse
|
45
|
Feng X, He S, Chen Y, Zhang L. Deubiquitinase BRCC3 promotes the migration, invasion and EMT progression of colon adenocarcinoma by stabilizing MET expression. Genes Genomics 2024; 46:637-646. [PMID: 38470543 DOI: 10.1007/s13258-024-01508-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 02/24/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND Breast cancer type 1 susceptibility protein/breast cancer type 2 susceptibility protein-containing complex subunit 3 (BRCC3), a deubiquitinase (DUBs), is overexpressed in various cancers. However, the underlying biological roles of BRCC3 in adenocarcinoma colon (COAD) have yet to be decrypted. OBJECTIVE In this work, we explored the potential biological function of BRCC3 in the natural process of COAD cells. METHODS The expression levels of BRCC3 in COAD tissues and cell lines were investigated via quantitative real time polymerase chain reaction and western blotting analyses. Meanwhile, short hairpin RNAs targeting BRCC3 (sh-BRCC3) or mesenchymal-epithelial transition factor (MET) (sh-MET) were used to investigate the biological function, including proliferation, apoptosis, migration, invasion, and epithelial-mesenchymal transition (EMT) progression in COAD cells. Furthermore, the expression levels of EMT-related biomarkers were detected with western blotting analysis. Furthermore, we also performed Co-IP assay to identify the correlation between BRCC3 and MET. RESULTS BRCC3 expression was increased in COAD tissues and cell lines. ShRNA-mediated downmodulation of BRCC3 in COAD cell lines induced EMT progression. BRCC3 knockdown resulted in decreased migration as well as invasion and increased apoptosis of SW480 and Lovo cells. Besides, MET was regulated by BRCC3 and involved in the migration, invasion, and EMT in SW480 and Lovo cells. Finally, we uncovered that the overexpressed MET reversed the effects of BRCC3 knockdown in COAD cell development. CONCLUSIONS BRCC3 acted as a critical factor in the development of COAD by deubiquitinating and stabilizing MET, which might provide an emerging biomarker for the therapeutic and diagnosis strategy of COAD.
Collapse
Affiliation(s)
- Xiu Feng
- Department of Oncology, Nantong First People's Hospital and Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Chongchuan District, Nantong, 226000, Jiangsu, China
| | - Shengnan He
- Department of Oncology, Nantong First People's Hospital and Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Chongchuan District, Nantong, 226000, Jiangsu, China
| | - Ying Chen
- Department of Oncology, Nantong First People's Hospital and Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Chongchuan District, Nantong, 226000, Jiangsu, China.
| | - Liang Zhang
- Department of Oncology, Nantong First People's Hospital and Affiliated Hospital 2 of Nantong University, No.666 Shengli Road, Chongchuan District, Nantong, 226000, Jiangsu, China.
| |
Collapse
|
46
|
Rezaei A, Moqadami A, Khalaj-Kondori M. Minocycline as a prospective therapeutic agent for cancer and non-cancer diseases: a scoping review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2835-2848. [PMID: 37991540 DOI: 10.1007/s00210-023-02839-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 11/05/2023] [Indexed: 11/23/2023]
Abstract
Minocycline is an FDA-approved secondary-generation tetracycline antibiotic. It is a synthetic antibiotic having many biological effects, such as antioxidant, anti-inflammatory, anti-cancer, and neuroprotective functions. This study discusses the pharmacological mechanisms of preventive and therapeutic effects of minocycline. Specifically, it provides a comprehensive overview of the molecular pathways by which minocycline acts on the different cancers, including ovarian, breast, glioma, colorectal, liver, pancreatic, lung, prostate, melanoma, head and neck, leukemia, and non-cancer diseases such as Alzheimer's disease, Parkinson, schizophrenia, multiple sclerosis, Huntington, polycystic ovary syndrome, and coronavirus disease 19. Minocycline may be a potential medication for these disorders due to its strong blood-brain barrier penetrance. It is also widely accepted as a specific medication, has a well-known side-effect characteristic, is reasonably priced, making it appropriate for continuous use in managing diseases, and has been demonstrated as an oral approach because it is effectively absorbed and accomplished almost all of the body's parts.
Collapse
Affiliation(s)
- Abedeh Rezaei
- Department of Animal Biology¸ Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Amin Moqadami
- Department of Animal Biology¸ Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology¸ Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
47
|
Mastronikolis NS, Delides A, Kyrodimos E, Piperigkou Z, Spyropoulou D, Giotakis E, Tsiambas E, Karamanos NK. Insights into metastatic roadmap of head and neck cancer squamous cell carcinoma based on clinical, histopathological and molecular profiles. Mol Biol Rep 2024; 51:597. [PMID: 38683372 PMCID: PMC11058607 DOI: 10.1007/s11033-024-09476-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/25/2024] [Indexed: 05/01/2024]
Abstract
The incidence of head and neck cancer (HNC), constituting approximately one in ten cancer cases worldwide, affects approximately 644,000 individuals annually. Managing this complex disease involves various treatment modalities such as systemic therapy, radiation, and surgery, particularly for patients with locally advanced disease. HNC treatment necessitates a multidisciplinary approach due to alterations in patients' genomes affecting their functionality. Predominantly, squamous cell carcinomas (SCCs), the majority of HNCs, arise from the upper aerodigestive tract epithelium. The epidemiology, staging, diagnosis, and management techniques of head and neck squamous cell carcinoma (HNSCC), encompassing clinical, image-based, histopathological and molecular profiling, have been extensively reviewed. Lymph node metastasis (LNM) is a well-known predictive factor for HNSCC that initiates metastasis and significantly impacts HNSCC prognosis. Distant metastasis (DM) in HNSCC has been correlated to aberrant expression of cancer cell-derived cytokines and growth factors triggering abnormal activation of several signaling pathways that boost cancer cell aggressiveness. Recent advances in genetic profiling, understanding tumor microenvironment, oligometastatic disease, and immunotherapy have revolutionized treatment strategies and disease control. Future research may leverage genomics and proteomics to identify biomarkers aiding individualized HNSCC treatment. Understanding the molecular basis, genetic landscape, atypical signaling pathways, and tumor microenvironment have enhanced the comprehension of HNSCC molecular etiology. This critical review sheds light on regional and distant metastases in HNSCC, presenting major clinical and laboratory features, predictive biomarkers, and available therapeutic approaches.
Collapse
Affiliation(s)
- Nicholas S Mastronikolis
- Department of Otorhinolaryngology - Head and Neck Surgery, School of Medicine, University of Patras, Patras, 26504, Greece.
| | - Alexander Delides
- 2nd Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Attikon' University Hospital, Rimini 1, Athens, 12462, Greece
| | - Efthymios Kyrodimos
- 1st Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Ippokrateion' General Hospital, Athens, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, 26504, Greece
| | - Despoina Spyropoulou
- Department of Radiation Oncology, Medical School, University of Patras, Patras, 26504, Greece
| | - Evangelos Giotakis
- 1st Otolaryngology Department, School of Medicine, National & Kapodistrian University of Athens, 'Ippokrateion' General Hospital, Athens, Greece
| | | | - Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, 26504, Greece
| |
Collapse
|
48
|
Li Y, Tao L, Xin J, Dai Y, Chen X, Zou J, Wang R, Wang B, Liu Z. Development and experimental verification of a prognosis model for disulfidptosis-associated genes in HNSCC. Medicine (Baltimore) 2024; 103:e37308. [PMID: 38518012 PMCID: PMC10957022 DOI: 10.1097/md.0000000000037308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/29/2024] [Indexed: 03/24/2024] Open
Abstract
Disulfidptosis is a newly discovered cell death pattern that has been less studied in head and neck squamous carcinoma (HNSCC). Exploring the molecular features of different subtypes of HNSCC based on disulfidptosis-associated genes (DAGs) is important for HNSCC. In addition, immunotherapy plays a pivotal role in the treatment of HNSCC. Exploring the sensitivity of immunotherapies and developing predictive models is essential for HNSCC. We analyzed the expression and mutational status of DAGs in 790 HNSCC patients and correlated the dates with clinical prognosis. HNSCC patients were divided into 2 groups based on their DAG expression. The relationship between DAGs, risk genes, and the immune microenvironment was analyzed using the CIBERSORT algorithm. A disulfidptosis risk model was constructed based on 5 risk genes using the LASSO COX method. To facilitate the clinical applicability of the proposed risk model, we constructed column line plots and performed stem cell correlation analysis and antitumor drug sensitivity analysis. Two different disulfidptosis-associated clusters were identified using consistent unsupervised clustering analysis. Correlations between multilayer DAG alterations and clinical characteristics and prognosis were observed. Then, a well-performing disulfidptosis-associated risk model (DAG score) was developed to predict the prognosis of HNSCC patients. We divided patients into high-risk and low-risk groups based on the DAG score and found that patients in the low-risk group were more likely to survive than those in the high-risk group (P < .05). A high DAG score implies higher immune cell infiltration and increased mutational burden. Also, univariate and multivariate Cox regression analyses revealed that the DAG score was an independent prognostic predictor for patients with HNSCC. Subsequently, a highly accurate predictive model was developed to facilitate the clinical application of DAG scores, showing good predictive and calibration power. Overall, we present a comprehensive overview of the DAG profile in HNSCC and develop a new risk model for the therapeutic status and prognosis of patients with HNSCC. Our findings highlight the potential clinical significance of DAG and suggest that disulfidptosis may be a potential therapeutic target for patients with HNSCC.
Collapse
Affiliation(s)
- Yushen Li
- Hospital of Stomatology, Jilin University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, People’s Republic of China
| | - Lu Tao
- Hospital of Stomatology, Jilin University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, People’s Republic of China
| | - Jiajun Xin
- Hospital of Stomatology, Jilin University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, People’s Republic of China
| | - Yifei Dai
- Hospital of Stomatology, Jilin University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, People’s Republic of China
| | - Xiantao Chen
- Hospital of Stomatology, Jilin University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, People’s Republic of China
| | - Jiatong Zou
- Hospital of Stomatology, Jilin University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, People’s Republic of China
| | - Rui Wang
- Hospital of Stomatology, Jilin University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, People’s Republic of China
| | - Bowei Wang
- The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Zhihui Liu
- Hospital of Stomatology, Jilin University, Changchun, People’s Republic of China
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun, People’s Republic of China
| |
Collapse
|
49
|
Duan XP, Qin BD, Jiao XD, Liu K, Wang Z, Zang YS. New clinical trial design in precision medicine: discovery, development and direction. Signal Transduct Target Ther 2024; 9:57. [PMID: 38438349 PMCID: PMC10912713 DOI: 10.1038/s41392-024-01760-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
In the era of precision medicine, it has been increasingly recognized that individuals with a certain disease are complex and different from each other. Due to the underestimation of the significant heterogeneity across participants in traditional "one-size-fits-all" trials, patient-centered trials that could provide optimal therapy customization to individuals with specific biomarkers were developed including the basket, umbrella, and platform trial designs under the master protocol framework. In recent years, the successive FDA approval of indications based on biomarker-guided master protocol designs has demonstrated that these new clinical trials are ushering in tremendous opportunities. Despite the rapid increase in the number of basket, umbrella, and platform trials, the current clinical and research understanding of these new trial designs, as compared with traditional trial designs, remains limited. The majority of the research focuses on methodologies, and there is a lack of in-depth insight concerning the underlying biological logic of these new clinical trial designs. Therefore, we provide this comprehensive review of the discovery and development of basket, umbrella, and platform trials and their underlying logic from the perspective of precision medicine. Meanwhile, we discuss future directions on the potential development of these new clinical design in view of the "Precision Pro", "Dynamic Precision", and "Intelligent Precision". This review would assist trial-related researchers to enhance the innovation and feasibility of clinical trial designs by expounding the underlying logic, which be essential to accelerate the progression of precision medicine.
Collapse
Affiliation(s)
- Xiao-Peng Duan
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhan Wang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
50
|
Cheng B, Li L, Luo T, Wang Q, Luo Y, Bai S, Li K, Lai Y, Huang H. Single-cell deconvolution algorithms analysis unveils autocrine IL11-mediated resistance to docetaxel in prostate cancer via activation of the JAK1/STAT4 pathway. J Exp Clin Cancer Res 2024; 43:67. [PMID: 38429845 PMCID: PMC10905933 DOI: 10.1186/s13046-024-02962-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/19/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Docetaxel resistance represents a significant obstacle in the treatment of prostate cancer. The intricate interplay between cytokine signalling pathways and transcriptional control mechanisms in cancer cells contributes to chemotherapeutic resistance, yet the underlying molecular determinants remain only partially understood. This study elucidated a novel resistance mechanism mediated by the autocrine interaction of interleukin-11 (IL-11) and its receptor interleukin-11 receptor subunit alpha(IL-11RA), culminating in activation of the JAK1/STAT4 signalling axis and subsequent transcriptional upregulation of the oncogene c-MYC. METHODS Single-cell secretion profiling of prostate cancer organoid was analyzed to determine cytokine production profiles associated with docetaxel resistance.Analysis of the expression pattern of downstream receptor IL-11RA and enrichment of signal pathway to clarify the potential autocrine mechanism of IL-11.Next, chromatin immunoprecipitation coupled with high-throughput sequencing (ChIP-seq) was performed to detect the nuclear localization and DNA-binding patterns of phosphorylated STAT4 (pSTAT4). Coimmunoprecipitation and reporter assays were utilized to assess interaction between pSTAT4 and the cotranscription factor CREB-binding protein (CBP) as well as their role in c-MYC transcriptional activity. RESULTS Autocrine secretion of IL-11 was markedly increased in docetaxel-resistant prostate cancer cells. IL-11 stimulation resulted in robust activation of JAK1/STAT4 signalling. Upon activation, pSTAT4 translocated to the nucleus and associated with CBP at the c-MYC promoter region, amplifying its transcriptional activity. Inhibition of the IL-11/IL-11RA interaction or disruption of the JAK1/STAT4 pathway significantly reduced pSTAT4 nuclear entry and its binding to CBP, leading to downregulation of c-MYC expression and restoration of docetaxel sensitivity. CONCLUSION Our findings identify an autocrine loop of IL-11/IL-11RA that confers docetaxel resistance through the JAK1/STAT4 pathway. The pSTAT4-CBP interaction serves as a critical enhancer of c-MYC transcriptional activity in prostate cancer cells. Targeting this signalling axis presents a potential therapeutic strategy to overcome docetaxel resistance in advanced prostate cancer.
Collapse
Affiliation(s)
- Bisheng Cheng
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lingfeng Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Tianlong Luo
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Qiong Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 511430, China
| | - Yong Luo
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shoumin Bai
- Department of Radiation Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Kaiwen Li
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Yiming Lai
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Hai Huang
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China.
| |
Collapse
|