1
|
Break MKB, Ansari SA, Katamesh AA, Albadari N, Alshammari MD, Alkahtani HM. Synthesis, in vitro and in silico studies of a novel chrysin-ferrocene Schiff base with potent anticancer activity via G1 arrest, caspase-dependent apoptosis and inhibition of topoisomerase II. J Enzyme Inhib Med Chem 2025; 40:2501377. [PMID: 40396612 PMCID: PMC12096666 DOI: 10.1080/14756366.2025.2501377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025] Open
Abstract
A novel chrysin-ferrocene Schiff base (CFSB) was synthesised as a potential anticancer agent. CFSB demonstrated high cytotoxicity against cancer cells with HepG2 (liver) being the most susceptible (IC50 = 3.11 µM). The compound was less toxic towards normal MRC5 cells and exhibited ∼5-fold selectivity towards most cancer cells. CFSB caused G1-phase arrest, induced caspase-dependent apoptosis by increasing Bax/Bcl2 ratio and reduced metastasis by decreasing MMP9 in HepG2. Furthermore, CFSB was inactive against CDK2, EGFR, TrkA and VEGFR, but it strongly inhibited topoisomerase II (IC50 = 20 µM) with potency comparable to etoposide (IC50 = 15 µM), while weak inhibition was observed against tubulin (IC50 = 76 µM). DFT calculations revealed that CFSB had desirable reactivity, while docking indicated high binding affinity with topoisomerase II. Molecular dynamics and MM-GBSA analyses showed that CFSB-topoisomerase II complex was stable with favourable binding energies, while in silico ADMET studies showed drug-like properties for CFSB.
Collapse
Affiliation(s)
- Mohammed Khaled Bin Break
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha’il, Ha’il, Saudi Arabia
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A. Katamesh
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
| | - Najah Albadari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
| | - Maali D. Alshammari
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
| | - Hamad M. Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Liu QG, Wu J, Wang ZY, Chen BB, Du YF, Niu JB, Song J, Zhang SY. ALK-based dual inhibitors: Focus on recent development for non-small cell lung cancer therapy. Eur J Med Chem 2025; 291:117646. [PMID: 40262298 DOI: 10.1016/j.ejmech.2025.117646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
As a prevalent oncogenic driver gene in non-small cell lung cancer (NSCLC), ALK represents a crucial and efficacious therapeutic target. To date, seven ALK inhibitors have been approved for ALK fusion-positive NSCLC, with several others undergoing clinical trials. These therapies demonstrate significant efficacy in ALK fusion-positive NSCLC patients. However, acquired resistance mechanisms, including ALK kinase domain mutations, ALK gene amplification, and bypass pathway activation, significantly compromise the efficacy of targeted therapy in ALK fusion-positive NSCLC. Therefore, the discovery of novel ALK inhibitors and the development of related treatment strategies remain critical. Compared to the combination therapy strategy based on ALK inhibitors, dual-target inhibitors (targeting two distinct pathways within a single molecule) may reduce systemic toxicity and mitigate resistance mechanisms in cancer treatment. Notably, recent years have witnessed remarkable progress in dual-target ALK inhibitor development for NSCLC. Consequently, this review aims to summarize the advancements achieved through dual ALK-based inhibitors in NSCLC therapy, analyze their rational design and structure-activity relationships, and provide perspectives for overcoming resistance through next-generation inhibitors and innovative therapeutic approaches.
Collapse
Affiliation(s)
- Qiu-Ge Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ji Wu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Zi-Yue Wang
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Bing-Bing Chen
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yi-Fei Du
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jin-Bo Niu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
3
|
Martínez-Ortega U, Aguayo-Ortiz R, Aguilar-Cazares D, Guerrero-Molina ED, Aguilar-Martínez V, Moreno-Rodríguez A, López-González JS, Vázquez-Ramos JM, Hernández-Luis F. Alchemical free energy-based optimization of quinazoline derivatives as potent EGFR inhibitors with cytotoxic activity. Bioorg Med Chem 2025; 124:118179. [PMID: 40199186 DOI: 10.1016/j.bmc.2025.118179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/27/2025] [Accepted: 03/30/2025] [Indexed: 04/10/2025]
Abstract
Gefitinib (GFB) is a well-established EGFR inhibitor used in the treatment of non-small cell lung cancer (NSCLC) that has shown resistance in certain cases of this cancer. In this work, we aimed to enhance GFB's inhibitory activity using alchemical free energy calculations, leading to the design of five new quinazoline derivatives. Among these, compound 8a was the most potent, inhibiting EGFR at 10 µM and showing significant antiproliferative effects at 25 µM. Further optimization identified two new compounds, NCU00 and NCU01, with improved EGFR inhibition and superior cytotoxicity in four NSCLC cell lines compared to GFB. Molecular dynamics simulations revealed crucial interactions that contribute to the enhanced inhibitory activity of NCU00 and NCU01. Toxicological assessments in mice showed no adverse effects on kidney or liver function, and NCU01 exhibited no developmental toxicity in zebrafish embryos. This study highlights the effectiveness of alchemical free energy methods in optimizing quinazoline-bearing EGFR inhibitors.
Collapse
Affiliation(s)
- U Martínez-Ortega
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - R Aguayo-Ortiz
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - D Aguilar-Cazares
- Departamento de Enfermedades Crónico-Degenerativas, Laboratorio de Cáncer Pulmonar, Instituto Nacional de Enfermedades Respiratorias, Ismael Cosio Villegas, Mexico City 14080, Mexico
| | - E D Guerrero-Molina
- Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - V Aguilar-Martínez
- Laboratorio de Estudios Epidemiológicos, Clínicos, Diseños Experimentales e Investigación, Facultad de Ciencias Químicas, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico
| | - A Moreno-Rodríguez
- Laboratorio de Estudios Epidemiológicos, Clínicos, Diseños Experimentales e Investigación, Facultad de Ciencias Químicas, Universidad Autónoma Benito Juárez de Oaxaca, Oaxaca 68120, Mexico
| | - J S López-González
- Departamento de Enfermedades Crónico-Degenerativas, Laboratorio de Cáncer Pulmonar, Instituto Nacional de Enfermedades Respiratorias, Ismael Cosio Villegas, Mexico City 14080, Mexico
| | - J M Vázquez-Ramos
- Departamento de Bioquímica, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - F Hernández-Luis
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico.
| |
Collapse
|
4
|
Zhao M, Wu D, Qiu W, Zhang H, Chen M, Zhou H, Ye X, Han Z, Shi X. A novel Quinazoline derivative exerts anti-tumor effects in non-small cell lung cancer through Wnt/β-catenin pathway inhibition. Bioorg Chem 2025; 160:108481. [PMID: 40253763 DOI: 10.1016/j.bioorg.2025.108481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/07/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
4-anilinoquinazoline scaffold, a key structure in the development of EGFR-TKI anticancer drugs such as Gefitinib and Erlotinib, continues to drive advancements in non-small cell lung cancer (NSCLC) therapy. In this study, we synthesized a series of novel derivatives (B3-B11). Structure-activity relationship (SAR) analysis revealed that electron-donating groups para-substituted at the 4-anilino position significantly enhanced antiproliferative activity. Notably, B10 incorporating an acrylamide group at the 4-anilino position demonstrated the most potent activity against NSCLC, with an IC50 of 1.28 μM in A549 cells, nearly six times more effective than Gefitinib (7.81 μM). Acute toxicity studies confirmed that B10 is safe at doses below 50 mg/kg. In vivo studies using an A549 xenograft model showed a 46 % tumor growth inhibition (TGI) with B10 (25 mg/kg), surpassing the 21 % inhibition of Gefitinib at the same dose. Mechanistic studies revealed that B10 inhibited the AKT/GSK-3β/β-catenin signaling pathway, downregulating Wnt target genes (c-Myc, c-Jun), and reduced COX2 expression and IL-8 levels, showcasing its dual anti-inflammatory and antitumor effects. These results position B10 as a promising NSCLC therapeutic candidate, combining potent efficacy with a favorable safety profile, and enhance our understanding of the SAR and mechanism of action of 4-anilinoquinazoline derivatives.
Collapse
Affiliation(s)
- Menglong Zhao
- Clinical Research Center, Shantou Central Hospital, Shantou 515041, PR China
| | - Danqing Wu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Weiqian Qiu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Hongchen Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Min Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Haixia Zhou
- The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325088, PR China
| | - Xiaoxia Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Zhong Han
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, PR China
| | - Xiangchao Shi
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China.
| |
Collapse
|
5
|
Yang M, Xu J, Zhao M, Zhou J, Hou Y, Zhao L, Liu F, Huang Y. Synthesis of quinazoline derivatives and their in vitro inhibition activity against MDA-MB-231 cells and A549 cells. Bioorg Med Chem Lett 2025:130308. [PMID: 40516763 DOI: 10.1016/j.bmcl.2025.130308] [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: 03/26/2025] [Revised: 05/23/2025] [Accepted: 06/11/2025] [Indexed: 06/16/2025]
Abstract
A series of novel 4-aniline quinazoline derivatives (Y1-Y26) were synthesised from 2-amino-6-nitrobenzoic acid based on the quinazoline parent nucleus via trifluoroacetylation, ring-closing and chlorination; The CCK-8 method was used to assess the in vitro inhibitory activities of the resulting compounds against two distinct cell lines: breast cancer cells (MDA-MB-231) and non-small cell lung cancer (A549). The results demonstrated that most of the compounds exhibited in vitro proliferation inhibitory activity against both MDA-MB-231 and A549 cells. Among these, compound Y22 exhibited the strongest inhibitory effect on MDA-MB-231 cells (IC50 = 4.53 μM); As the concentration of Y22 increased, the inhibition of cell proliferation was enhanced, and the cells gradually shrank and underwent morphological changes consistent with apoptosis; Transwell assay demonstrated that the compound Y22 exhibited a substantial inhibitory effect on cell migration; Flow cytometry revealed a substantial augmentation in apoptosis with elevated compound concentrations; Western blot analysis indicated that Y22 may exert in vitro antitumour activity by decreasing the expression of the anti-apoptotic protein Bcl-2 while increasing the expression of the pro-apoptotic protein Bax. The findings of these studies suggest that Y22 can potentially plays a significant role in the design and synthesis of antitumour drugs.
Collapse
Affiliation(s)
- Mengyuan Yang
- School of Pharmacy, Bengbu Medical University, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, PR China
| | - Jie Xu
- School of Pharmacy, Bengbu Medical University, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, PR China
| | - Mengmeng Zhao
- School of Pharmacy, Bengbu Medical University, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, PR China
| | - Jiaxin Zhou
- Department of Biological Sciences, Bengbu Medical University, Bengbu 233030, Anhui, PR China
| | - Yifan Hou
- School of Pharmacy, Bengbu Medical University, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, PR China
| | - Long Zhao
- School of Pharmacy, Bengbu Medical University, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, PR China
| | - Fang Liu
- School of Pharmacy, Bengbu Medical University, Anhui Engineering Technology Research Center of Biochemical Pharmaceuticals, Bengbu 233030, Anhui, PR China.
| | - Yinjiu Huang
- Department of Biological Sciences, Bengbu Medical University, Bengbu 233030, Anhui, PR China.
| |
Collapse
|
6
|
Meng X, Li X, Gao Y, Zhang S. Nuclear receptors as novel regulators that modulate cancer radiosensitivity and normal tissue radiotoxicity. Mol Cancer 2025; 24:155. [PMID: 40442680 PMCID: PMC12124050 DOI: 10.1186/s12943-025-02362-2] [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: 04/15/2025] [Accepted: 05/20/2025] [Indexed: 06/02/2025] Open
Abstract
Nuclear receptors (NRs) are a superfamily of transcription factors that are involved in various pathophysiological processes. The human genome contains 48 types of nuclear receptors, including steroid hormone receptors (e.g., estrogen receptor [ER] and vitamin D receptor [VDR]), nonsteroid hormone receptors (e.g. peroxisome proliferator-activated receptor [PPAR] and retinoic acid receptor [RAR]), and orphan nuclear receptors (e.g. neuron-derived clone 77 [Nur77] and testicular nuclear receptor 4 [TR4]) and certain nuclear receptors are specifically overexpressed in tumor cells or surrounding normal tissues. Radiotherapy is one of the main methods of tumor treatment, but radioresistance in tumors and radiotoxicity to normal tissues strongly affect radiotherapy efficacy. Accumulating evidence has indicated the critical role of nuclear receptor modulators (including agonists and antagonists) as promising radiosensitizers in radiotherapy through various mechanisms. In addition, several nuclear receptors and their agonists alleviate normal tissue toxicity during radiotherapy. Thus, nuclear receptors serve as novel targets for tumor radiosensitization and for protecting of normal tissues from radiation damage. This review summarizes the research progress of nuclear receptors and highlights a promising synergistic strategy in radiotherapy.
Collapse
Affiliation(s)
- Xiaochen Meng
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiaoqian Li
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yi Gao
- Department of Gastroenterology, Affiliated Jiangyin Hospital of Nantong University, Jiangyin, 214400, China.
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China.
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital), Mianyang, 621099, China.
- Medical College of Tibet University, Lasa, 850000, China.
| |
Collapse
|
7
|
Rudawska A, Szermer-Olearnik B, Szczygieł A, Mierzejewska J, Węgierek-Ciura K, Żeliszewska P, Kozień D, Chaszczewska-Markowska M, Adamczyk Z, Rusiniak P, Wątor K, Rapak A, Pędzich Z, Pajtasz-Piasecka E. Functionalized Boron Carbide Nanoparticles as Active Boron Delivery Agents Dedicated to Boron Neutron Capture Therapy. Int J Nanomedicine 2025; 20:6637-6657. [PMID: 40438187 PMCID: PMC12118811 DOI: 10.2147/ijn.s516534] [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: 01/31/2025] [Accepted: 04/30/2025] [Indexed: 06/01/2025] Open
Abstract
Introduction Boron neutron capture therapy (BNCT) is a promising targeted radiotherapy that enables the treatment of cancers at the cellular level. The crucial aspect of BNCT are boron carriers, which should selectively reach cancer cells by delivering high concentrations of boron. Therefore, we propose the use of boron carbide (B4C) nanoparticles functionalized with antibodies directed against receptors overexpressed in cancer cells, such as the low-density lipoprotein receptor (LDLR) and the epidermal growth factor receptor (EGFR). Methods Hydrodynamic diameter measurements confirmed the stability of functionalized B4C nanoparticles in culture media during biological tests lasting up to 72 hours. The toxicity of the nanoparticles was assessed using the MTT assay and BrdU cell cycle assay on three types of cancer cells (PC-3, T98G, and SCC-25) with different levels of LDLR and EGFR surface expression. The uptake of functionalized B4C nanoparticles by cancer cells was assessed based on flow cytometry, fluorescence microscopy, and holotomography. Boron concentrations in cancer cells were quantified via ICP-MS. Results Functionalized B4C nanoparticles showed even 2-fold higher interaction with SCC-25 cells characterized by the highest surface expression of both receptors than with PC-3 and T98G cells. Holotomographic imaging confirmed the greater intracellular uptake of functionalized B4C nanoparticles compared to unmodified B4C, providing further evidence for the selective targeting of boron to cancer cells. ICP-MS analyses showed that B4C anti-LDLR nanoparticles were the most effective in delivering a high boron concentration to cancer cells. Particularly in SCC-25 cells, the concentration was 9.58 ± 2.6 mg/L boron per million cells. The highest uptake by these cells was associated with a decrease in viability to 63% and a slight reduction in the percentage of cells in S phase after 24-hour exposure. Conclusion Stable complexes of antibody-functionalized B4C nanoparticles were successfully obtained, demonstrating increased tropism towards cancer cells overexpressing LDLR and EGFR.
Collapse
Affiliation(s)
- Anna Rudawska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Bożena Szermer-Olearnik
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Agnieszka Szczygieł
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Jagoda Mierzejewska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Katarzyna Węgierek-Ciura
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Paulina Żeliszewska
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Krakow, Poland
| | - Dawid Kozień
- Depatment of Materials Science and Ceramics, AGH University of Krakow, Krakow, Poland
| | | | - Zbigniew Adamczyk
- Jerzy Haber Institute of Catalysis and Surface Chemistry, Polish Academy of Sciences, Krakow, Poland
| | - Piotr Rusiniak
- Department of Geology, Geophysics and Environmental Protection, AGH University of Krakow, Krakow, Poland
| | - Katarzyna Wątor
- Department of Geology, Geophysics and Environmental Protection, AGH University of Krakow, Krakow, Poland
| | - Andrzej Rapak
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Zbigniew Pędzich
- Depatment of Materials Science and Ceramics, AGH University of Krakow, Krakow, Poland
| | - Elżbieta Pajtasz-Piasecka
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
8
|
Wu J, Gao Q, Xia Q, Wang Y, Zheng Z, He A, Liu Y, Yang Y, Miao Y, Han D. Highly Specific Cytokine Receptor-Targeting Chimeras for Targeted Membrane Protein Degradation and Sensitization of Osimertinib in EGFR-Mutated Non-Small-Cell Lung Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2504050. [PMID: 40401615 DOI: 10.1002/adma.202504050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 05/01/2025] [Indexed: 05/23/2025]
Abstract
The ability of cytokine receptors to mediate the internalization of targets in lysosomes positions them as specific and effective effectors for protein degradation strategies. However, challenges remain, including the potential unintended activation of cell-proliferation-related cytokine receptors, as well as limitations in programmability and structural flexibility of protein degradators. In this work, a CXCR7-targeting chimera (AP-CRTAC) that functions as a CXCR7 inducer by covalently linking a membrane protein-targeting aptamer with a mutant-CXCL12 mimic peptide is developed. This peptide selectively binds to CXCR7 without activating CXCR4. The AP-CRTAC, which incorporates various aptamer forms from DNA, RNA, or even bispecific aptamers, has shown significant efficacy in degrading one or more proteins or protein mutants on the cell surface. Moreover, the AP-CRTAC constructed with a 2' F-pyrimidine-modified RNA aptamer targeting EGFR effectively degrades various EGFR activating mutations. Notably, AP-CRTAC enhances the sensitivity of the L858R/T790M/C797S triple mutant lung cancer cells, which are resistant to current EGFR-targeted therapies, to the third-generation EGFR inhibitor osimertinib in both in vitro and in vivo settings. This research introduces an engineered CXCR7 inducer with high specificity and programmability for the targeted degradation of cell surface proteins, while minimizing unwanted side effects.
Collapse
Affiliation(s)
- Jiawei Wu
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Qianqian Gao
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Qing Xia
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Yaru Wang
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zixuan Zheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Axin He
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yu Liu
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Yang Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, P. R. China
| | - Yanyan Miao
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Da Han
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
9
|
Chen C, Xu L, Chen L, Zhai Z, Cheng M, Luo S, Wang H. DNAJC5 facilitates the proliferation and migration of lung adenocarcinoma cells by augmenting EGFR trafficking. Commun Biol 2025; 8:757. [PMID: 40374748 PMCID: PMC12081771 DOI: 10.1038/s42003-025-08191-9] [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: 06/27/2024] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is a highly prevalent and lethal malignant tumor, with the aberrantly activated EGFR signaling pathway playing a crucial role in its development. However, resistance to tyrosine-kinase inhibitors (TKIs) targeting EGFR significantly limits the efficacy of LUAD clinical therapy. Therefore, it is imperative to identify novel therapeutic targets and elucidate the regulatory mechanisms of EGFR for improving LUAD treatment outcomes. In this study, we discover that DNAJC5 functions as an oncogene in LUAD. We observe elevated protein levels of DNAJC5 in tissues from LUAD patients, which are strongly associated with poor prognosis among these individuals. Furthermore, overexpression of DNAJC5 promotes proliferation and migration of LUAD cells both in vitro and in vivo. Mechanistic investigations reveal that DNAJC5 interacts with the intracellular domain of EGFR and enhances its endocytosis and recycle, thereby augmenting EGFR activity and downstream signaling pathways. Additionally, we find that DNAJC5 binds to AP2A1 protein-a key player in EGFR endocytosis-and strengthens its interaction with EGFR. Knockdown experiments targeting AP2A1 attenuate the ability of DNAJC5 to promote proliferation and migration of LUAD cells. Collectively, our findings unveil a functional role for DNAJC5 in regulating EGFR trafficking and driving LUAD progression.
Collapse
Affiliation(s)
- Can Chen
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Center for Experimental Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Linlin Xu
- Department of Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Limin Chen
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhenyu Zhai
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Minzhang Cheng
- Jiangxi Institute of Respiratory Disease, the Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Shiwen Luo
- Center for Experimental Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Hailong Wang
- Medical Innovation Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
10
|
Sun L, Chen W, Yuan W, Huang Q, Yang H, Zhang W, Tang J, Hu P. Ginkgetin inhibits the proliferation and migration of lung cancer cells via FAK/STAT3/AKT pathway. Mol Biol Rep 2025; 52:458. [PMID: 40366441 DOI: 10.1007/s11033-025-10540-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/23/2025] [Indexed: 05/15/2025]
Abstract
PURPOSE Lung cancer has become a primary illness that severely endangers human life and health due to its extremely high morbidity and mortality rates. Ginkgetin has been proven to have toxic effects on various tumor cells. Nevertheless, the mechanism of Ginkgetin on lung cancer is uncertain. In the present study, the effect and possible mechanism of Ginkgetin on lung cancer were explored. METHODS The cell counting kit-8 assay and colony formation assay were performed to detect the effect of Ginkgetin on cell proliferation. The wound healing assay was performed to detect the effect of Ginkgetin on cell migration. Additionally, western blot and immunofluorescence assay were performed to detect the expression of proteins. RESULTS Our results demonstrated that Ginkgetin effectively inhibited the proliferation and migration of A549 and H1299 cells. Mechanistically, Ginkgetin downregulated the phosphorylated expression of focal adhesion kinase (FAK), signal transducer and activator of transcription 3 (STAT3), and protein kinase B (AKT) and blocked the FAK/STAT3/AKT phosphorylation induced by epidermal growth factor (EGF). Furthermore, Ginkgetin suppressed the proliferation and migration of A549 and H1299 cells induced by EGF. Notably, Ginkgetin decreased the Cyclin A2 and Cyclin D1 expression. CONCLUSION Collectively, these findings concluded that Ginkgetin may suppress the proliferation and migration of lung cancer cells via the FAK/STAT3/AKT pathway, suggesting that Ginkgetin has potential applications in lung cancer treatment.
Collapse
Affiliation(s)
- Longhua Sun
- The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Jiangxi Provincial Key Laboratory of Respirtory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang, Jiangxi, 330200, P.R. China
| | - Wen Chen
- The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Jiangxi Provincial Key Laboratory of Respirtory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Wenxin Yuan
- Department of Ultrasonography, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Qianwen Huang
- The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Hong Yang
- The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Wei Zhang
- The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Jiangxi Provincial Key Laboratory of Respirtory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Jianjun Tang
- The Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Jiangxi Provincial Key Laboratory of Respirtory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- China-Japan Friendship Jiangxi Hospital, National Regional Center for Respiratory Medicine, Nanchang, Jiangxi, 330200, P.R. China
| | - Ping Hu
- Jiangxi Provincial Key Laboratory of Respirtory Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P.R. China.
- Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330001, P.R. China.
| |
Collapse
|
11
|
Fu X, Wang Y, Zhang W, Yang Y, Zeng J, Li X, Feng C, Li B, Liu Y, Zhang Y, Zhang C, Ma S. Nano-Mediated Fluorescence Switching for Epidermal Growth Factor Receptor Detection. Cell Prolif 2025:e70063. [PMID: 40368641 DOI: 10.1111/cpr.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/19/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
Identification of the epidermal growth factor receptor (EGFR) in biological specimens is essential for cancer diagnostics, drug development and therapeutic monitoring. However, real-time techniques for accurate EGFR expression monitoring are currently limited. In this study, we report the development of a novel nano detector (Cy3-AptEGFR@BPNSs) with the capabilities of quenching and recovery to enable visual EGFR expression analysis. Cy3-AptEGFR is a Cy3-labelled single-stranded RNA (ssRNA) that exhibits specific binding to EGFR. Black phosphorus nanosheets (BPNSs) possess the ability to adsorb Cy3-AptEGFR via van der Waals forces, quenching its fluorescence when combined. The detection of EGFR receptors on cancer cell surfaces prompts the release of Cy3-AptEGFR from BPNSs, a consequence of the robust binding interaction between the receptor and aptamer, thereby leading to fluorescence reinstatement. The recovered fluorescence intensity of this detector is found to be directly correlated with EGFR expression levels in cancer cells, indicating its potential for guiding tumour diagnosis and treatment. The specificity of Cy3-AptEGFR@BPNSs further enhances its utility in detecting EGFR. More importantly, our research demonstrates that the reduction in EGFR expression levels within cancer cells corresponds to a proportional decline in fluorescence intensity, thereby facilitating precise tracking of EGFR dynamics.
Collapse
Affiliation(s)
- Xin Fu
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Inspection, Ningxia Medical University, Yinchuan, China
| | - Yuhao Wang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wenxin Zhang
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yuepeng Yang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jialin Zeng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaodie Li
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chengyu Feng
- Department of Pediatric Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Bin Li
- School of Inspection, Ningxia Medical University, Yinchuan, China
| | - Yingying Liu
- School of Chemistry and Chemical Engineering, Center for Transformative Molecules, Zhangjiang Institute for Advanced Study and National Center for Translational Medicine (Shanghai), Shanghai Jiao Tong University, Shanghai, China
| | - Yinan Zhang
- School of Chemical Science and Engineering, Tongji University, Shanghai, China
| | - Chao Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Inspection, Ningxia Medical University, Yinchuan, China
| | - Sicong Ma
- Department of Intensive Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Global Health Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
12
|
Li J, Gan J, Shi S, Huang J, Yang Y. The potential of targeting autophagy-related non-coding RNAs in the treatment of lung cancer. Front Pharmacol 2025; 16:1551258. [PMID: 40438586 PMCID: PMC12116551 DOI: 10.3389/fphar.2025.1551258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/30/2025] [Indexed: 06/01/2025] Open
Abstract
Lung cancer is the most prevalent malignant tumor worldwide and remains the leading cause of cancer-related mortality. Despite advances in treatment development, lung cancer patients often face poor quality of life and low survival rates. Increasing evidence highlights the significant roles of autophagy and non-coding RNAs (ncRNAs) in the initiation, progression, and therapeutic response of lung cancer. Autophagy and ncRNAs can function as both tumor-promoting and tumor-suppressing factors in lung cancer. Therefore, investigating the roles of autophagy and ncRNAs in lung cancer provides valuable insights into its pathophysiology. At the same time, non-coding RNA also plays an important role in regulating autophagy. This study reveals that autophagy affects the occurrence and development of lung cancer through multiple pathways. Then, we also studied that in lung cancer, ncRNAs (e.g., lncRNAs, miRNAs, circRNAs and piRNAs) can regulate autophagy to promote or inhibit tumorigenesis, metastasis and drug resistance in lung cancer. Finally, the problems and solutions of autophagy and ncRNAs in the treatment of lung cancer were explored. These findings suggest that autophagy and ncRNAs can be potential targets for the treatment of lung cancer.
Collapse
Affiliation(s)
- Juan Li
- College of Pharmacy, Southwest Medical University, Luzhou, China
- Department of Pharmacy, Chengdu Wenjiang District People’s Hospital, Chengdu, Sichuan
| | - Jimei Gan
- College of Pharmacy, Southwest Medical University, Luzhou, China
- Department of Pharmacy, Chengdu Wenjiang District People’s Hospital, Chengdu, Sichuan
| | - Shenggan Shi
- Department of Pharmacy, Chengdu Wenjiang District People’s Hospital, Chengdu, Sichuan
| | - Juying Huang
- Department of Pharmacy, Chengdu Wenjiang District People’s Hospital, Chengdu, Sichuan
| | - Yong Yang
- College of Pharmacy, Southwest Medical University, Luzhou, China
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Lu C, Cheng D, Xie Y, Shang M, Chen R, Zhu Y, Gong J, Zhou H. Effect of CYP3A4 inhibitor and induction on the pharmacokinetics and safety of FHND9041, a novel EGFR T790M inhibitor, in healthy Chinese. BMC Pharmacol Toxicol 2025; 26:97. [PMID: 40336126 PMCID: PMC12060349 DOI: 10.1186/s40360-025-00930-2] [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: 03/10/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Non-small cell carcinoma is the main pathologic type of lung cancer, and a large number of clinical trials have shown that epidermal growth factor receptor tyrosinase inhibitors exhibit superior clinical efficacy and lower toxicity compared with chemotherapy. FHND9041 is a new irreversible EGFR T790M mutation-selective small molecule kinase inhibitor, a third-generation EGFR inhibitor developed by Nanjing Chuangte Pharmaceutical Technology Co., Ltd. The aim of this study was to evaluate the effects of oral Itraconazole capsules and oral Rifampicin capsules on the pharmacokinetic profile and safety and tolerability of a single oral dose of FHND9041 capsules in healthy Chinese male subjects. PATIENTS AND METHODS This study employed a single-center, open-label, fixed-sequence design, comprising two parallel groups: Group 1 received FHND9041 40 mg in combination with Itraconazole, while Group 2 received Rifampicin in combination with FHND9041 80 mg. Each group enrolled 16 subjects for a two-period study, with the first period involving monotherapy and the second period involving co-administration. All subjects participating in this clinical trial were healthy adult Chinese males. RESULTS In healthy subjects, after a single oral administration of 40 mg FHND9041 capsules, the corrected geometric mean ratios (90% confidence intervals) of FHND9041 Cmax, AUC0 - last, and AUC0 - inf when co-administered with itraconazole capsules compared to the monotherapy phase were 111.46% (103.26 - 120.30%), 169.53% (156.21 - 183.99%), and 168.25% (156.26 - 181.15%), respectively. The 90% confidence interval for Cmax fell within the 80-125% range, while the 90% confidence intervals for both AUC0 - last and AUC0 - inf exceeded the 80-125% range. Following a single oral dose of 80 mg FHND9041 capsules, the adjusted geometric mean ratios (90% confidence intervals) of Cmax, AUC0 - last, and AUC0 - inf for FHND9041 during co-administration with Rifampicin compared to monotherapy were 52.12% (41.95 - 64.74%), 16.47% (13.34 - 20.31%), and 16.51% (13.56 - 20.09%), respectively. The 90% confidence intervals for Cmax, AUC0 - last, and AUC0 - inf all fell outside the 80 - 125% range. No serious adverse events occurred during the trial. CONCLUSIONS Co-administration with Rifampicin significantly reduced the exposure of FHND9041. Therefore, it is recommended to avoid co-administration of FHND9041 with Rifampicin and other potent CYP3A4 inducers. Conversely, co-administration with Itraconazole significantly increased the total exposure of FHND9041. Caution is advised when FHND9041 is co-administered with Itraconazole or other strong CYP3A4 inhibitors. Close monitoring of tolerability during co-administration is essential, and dose reduction may be necessary if required. FHND9041 capsules demonstrated good safety and tolerability when used alone or in combination with strong CYP3A4 inhibitors or inducers. TRIAL REGISTRATION Registered 03/27/2023 ( http://www.chinadrugtrials.org.cn/index.html , CTR202300931).
Collapse
Affiliation(s)
- Chang Lu
- Clinical Research Center of Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Research Group of Jian Gong on Pharmacoepidemiology and Clinical Drug Evaluation, Shenyang Pharmaceutical University, Shenyang, 110016, China
- Key Laboratory of Innovative Pharmaceutical Research and Clinical Evaluation Jointly Constructed by Anhui, Bengbu, Anhui Province, 233000, China
| | - Dongmei Cheng
- Clinical Research Center of Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Key Laboratory of Innovative Pharmaceutical Research and Clinical Evaluation Jointly Constructed by Anhui, Bengbu, Anhui Province, 233000, China
| | - Yunqiu Xie
- Clinical Research Center of Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
- Key Laboratory of Innovative Pharmaceutical Research and Clinical Evaluation Jointly Constructed by Anhui, Bengbu, Anhui Province, 233000, China
| | - Minghong Shang
- Jiangsu Chia Tai Feng Hai Pharmaceutical Co., Ltd, Nanjing, 210046, China
| | - Rongzhen Chen
- Jiangsu Chia Tai Feng Hai Pharmaceutical Co., Ltd, Nanjing, 210046, China
| | - Yongqiang Zhu
- Jiangsu Chia Tai Feng Hai Pharmaceutical Co., Ltd, Nanjing, 210046, China.
- School of Life Sciences, Nanjing Normal University, Nanjing, 210042, China.
| | - Jian Gong
- Research Group of Jian Gong on Pharmacoepidemiology and Clinical Drug Evaluation, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Huan Zhou
- Clinical Research Center of Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.
- Research Group of Jian Gong on Pharmacoepidemiology and Clinical Drug Evaluation, Shenyang Pharmaceutical University, Shenyang, 110016, China.
- Key Laboratory of Innovative Pharmaceutical Research and Clinical Evaluation Jointly Constructed by Anhui, Bengbu, Anhui Province, 233000, China.
| |
Collapse
|
14
|
Nosair AM, Abdelaziz AA, Abo-Kamer AM, Al-Madboly LA, Farghali MH. Nutritional optimization for bioprocess production of staphyloxanthin from Staphylococcus aureus with response surface methodology: promising anticancer scaffold targeting EGFR inhibition. Microb Cell Fact 2025; 24:99. [PMID: 40329373 PMCID: PMC12054202 DOI: 10.1186/s12934-025-02717-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Staphyloxanthin (STX) is a secondary metabolite pigment associated with membrane structures, recognized for its significant antioxidant properties. It plays a crucial role in combating reactive oxygen species (ROS), positioning it as a promising and effective alternative in cancer treatment. This study focused on enhancing the production of STX pigment by employing statistical optimization of media components, alongside the evaluation of its safety and anticancer properties. RESULTS A total of 59 Staphylococcus aureus isolates were screened and quantitatively estimated for STX production. The best pigment-producing isolate was identified based on molecular phylogenetic analysis as S. aureus A2, with accession number PP197164. A Box-Wilson central composite design was employed to evaluate the intricate interactions among six variables affecting the pigment yield. The most optimal conditions resulted in the highest production of STX of OD456 = 0.328, which is approximately 1.5-fold greater than the yield (OD456 = 0.215) obtained from OFAT optimization. The final response surface model fitting the data achieved a R² of 0.8748. STX exhibited marked cytotoxicity against the A549 NSCLC cell line with IC50 of 57.3 µg/mL, a safe dose in normal Vero cells. The anticancer activity of STX was predominantly mediated by the apoptotic pathway, as confirmed by confocal microscopy, the annexin V-FITC apoptosis assay, and the overexpression of caspase-3. Moreover, STX disrupted cell cycle at pre-G1 and G0/G1 phases in lung cancer. Intriguingly, STX exhibited its antitumor activity through reducing the EGFR expression. The molecular docking study revealed the potential binding interactions and affinities within the active sites of both wild-type and mutant EGFR. CONCLUSION The bioprocess for optimized production, combined with the biological profiling and low cytotoxicity, substantiates the potential application of STX pigment in combating lung cancer.
Collapse
Affiliation(s)
- Ahmed M Nosair
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Ahmed A Abdelaziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amal M Abo-Kamer
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Lamiaa A Al-Madboly
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Mahmoud H Farghali
- Department of Microbiology and Immunology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
15
|
Wang Y, Zhu X, Li L, Su D, Ai L, Xie H, Zhou D, Yang H, Li B. Fibroblast EGFR signaling mediates ricin toxin-induced acute lung injury via EGR1/CXCL1 axis. Arch Toxicol 2025:10.1007/s00204-025-04067-3. [PMID: 40317338 DOI: 10.1007/s00204-025-04067-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025]
Abstract
Ricin toxin (RT), a highly potent plant-derived toxin, represents a critical threat due to its capacity to induce fatal acute lung injury (ALI) upon inhalation. While the epidermal growth factor receptor (EGFR), a receptor tyrosine kinase predominantly expressed on epithelial cells and fibroblasts, regulates cellular processes such as growth, proliferation, differentiation and inflammation, its involvement in RT-induced ALI remains unexplored. This study investigates this relationship using a mouse model of ALI induced by aerosolized RT at a dose of 2.0 × LD50 (approximately 0.01 mg kg -1). The results demonstrate that damage to alveolar epithelial type II (AT2) cells leads to the release of heparin-binding epidermal growth factor-like growth factor (HB-EGF), which activates EGFR on fibroblasts, exacerbating lung injury pathology and reducing survival. Mechanistically, EGFR activation in fibroblasts induces the early growth response protein 1 (EGR1), which subsequently enhances chemokine C-X-C motif ligand 1 (CXCL1) secretion 24 h post-exposure, promoting neutrophil infiltration in the lung. RNA sequencing analysis corroborates these findings. Notably, pharmacological inhibition of EGFR phosphorylation using Erlotinib (ERL) significantly mitigates the inflammatory response in RT-induced ALI. These results not only illuminate the immune response in lung tissue but also highlight EGFR signaling in fibroblasts as a pivotal mediator of RT-induced ALI. This study identifies a novel therapeutic strategy targeting EGFR signaling in fibroblasts for the treatment of inflammatory lung diseases.
Collapse
Affiliation(s)
- Yuqing Wang
- Department of Clinical Laboratory, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Graduate School of PLA General Hospital, Beijing, 100853, China
| | - Xiaoyu Zhu
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Lu Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Duo Su
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Lingli Ai
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Hao Xie
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Dongsheng Zhou
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China
| | - Huiying Yang
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, 100071, China.
| | - Boan Li
- Department of Clinical Laboratory, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
16
|
Kuang Y, Zhao Y, Miao Z, Xu Y, Yang Q. Oxaloacetate stimulates phosphorylation of epidermal growth factor receptor in epithelial cells in vitro. Growth Factors 2025:1-13. [PMID: 40314117 DOI: 10.1080/08977194.2025.2499634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/16/2025] [Indexed: 05/03/2025]
Abstract
Oxaloacetate (OA) is a pivotal endogenous metabolite. Within our investigation, we ascertained that OA functions as an agonist for the epidermal growth factor receptor (EGFR), a key protagonist in the genesis of diverse tumours. We substantiated that escalating concentrations of OA initially enhanced the cellular viability of several cancer cells, followed by subsequent attenuation, which is similar to the effect of EGF. Furthermore, the protein phosphorylation profile in HepG2 cells exposed to OA closely paralleled that induced by epidermal growth factor (EGF). Additional findings underscored the capability of OA to induce the generation of EGFR dimers. Finally, our observations revealed that OA governs the activation of AKT and Erk, the typical downstream signalling proteins of EGFR. We postulate that the endogenous metabolite OA can function as either an agonist or inhibitor of EGFR at specific concentrations to modulate tumour proliferation, and provide new insights into the regulation of EGFR activation.
Collapse
Affiliation(s)
- Ye Kuang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| | - Yuxiang Zhao
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| | - Zeyu Miao
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| | - Yang Xu
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| | - Qing Yang
- Department of Pathogenobiology, College of Basic Medical Sciences, Jilin University, Changchun City, China
| |
Collapse
|
17
|
Liu K, Hu S, Wufuer R, Zhang Q, Qiu L, Zhang Z, Wang M, Zhang Y. Deficiency of DDI2 suppresses liver cancer progression by worsening cell survival conditions. Free Radic Biol Med 2025; 232:200-213. [PMID: 40049338 DOI: 10.1016/j.freeradbiomed.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/25/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
The levels of reactive oxygen species (ROS) and the extent of ensuing DNA damage significantly influence cancer initiation and progression. Of crucial importance, the aspartate protease DDI2 has been proposed to play a pivotal role in monitoring intracellular ROS levels (to trigger oxidative eustress or distress), as well as in the oxidative DNA damage repair, through redox homeostasis-determining factor Nrf1 (encoded by NFE2L1). However, the specific role of DDI2 in the multi-step process resulting in the development and progression of liver cancer remains elusive to date. In the present study, we employed the CRISPR/Cas9 gene editing system to create two nuanced lines of DDI2 knockout (i.e., DDI2-/- and DDI2insG/-) from liver cancer cells. Subsequent experiments indicate that the knockout of DDI2 leads to increased ROS levels in hepatoma cells by downregulating two major antioxidant transcription factors Nrf1 and Nrf2 (encoded by NFE2L2), exacerbating endogenous DNA damages caused by ROS and not-yet-identified factors, thereby inhibiting cell proliferation and promoting apoptosis, and ultimately hindering in vivo malignant growth of xenograft tumor cells. Conversely, the restoration of DDI2 expression reverses the accumulation of ROS and associated DNA damage caused by DDI2 knockout, eliminating the subsequent inhibitory effects of DDI2 deficiency on both in vitro and in vivo growth of liver cancer cells. Collectively, these findings demonstrate that DDI2 deficiency impedes liver tumor growth by disrupting its survival environment, suggesting that DDI2 may serve as a novel therapeutic target for anti-cancer strategies aimed at modulating ROS or DNA damage processes.
Collapse
Affiliation(s)
- Keli Liu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Shaofan Hu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Jinfeng Laboratory, No. 313 Jinyue Road, Chongqing High-tech District, 401329, China; Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba, Chongqing, 400038, China
| | - Reziyamu Wufuer
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; School of Parmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Xinjiang, 830017, China
| | - Qun Zhang
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China
| | - Lu Qiu
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Department of Pathology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Zhengwen Zhang
- Laboratory of Neuroscience, Institute of Cognitive Neuroscience and School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, England, United Kingdom
| | - Meng Wang
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; Jinfeng Laboratory, No. 313 Jinyue Road, Chongqing High-tech District, 401329, China; Department of Pathophysiology, College of High Altitude Military Medicine, Third Military Medical University (Army Medical University), No. 30 Gaotanyan Street, Shapingba, Chongqing, 400038, China.
| | - Yiguo Zhang
- The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China; School of Life and Health Sciences, Fuyao University of Science and Technology, No. 104 Wisdom Avenue, Nanyu Town, Minhou County High-Tech District, Fuzhou, 350109, Fujian, China.
| |
Collapse
|
18
|
Hajikarimloo B, Mohammadzadeh I, Tos SM, Habibi MA, Hashemi R, Hezaveh EB, Najari D, Hasanzade A, Hooshmand M, Bana S. Machine learning in prediction of epidermal growth factor receptor status in non-small cell lung cancer brain metastases: a systematic review and meta-analysis. BMC Cancer 2025; 25:818. [PMID: 40312289 PMCID: PMC12044993 DOI: 10.1186/s12885-025-14221-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) mutations are present in 10-60% of all non-small cell lung cancer (NSCLC) patients and are associated with dismal prognosis. Lung cancer brain metastases (LCBM) are a common complication of lung cancer. Predictions of EGFR can help physicians in decision-making and, through optimizing treatment strategies, can result in more favorable outcomes. This systematic review and meta-analysis evaluated the predictive performance of machine learning (ML)-based models in EGFR status in NSCLC patients with brain metastasis. METHODS On December 20, 2024, the four electronic databases, Pubmed, Embase, Scopus, and Web of Science, were systematically searched. Studies that evaluated EGFR status in patients with brain metastasis from NSCLC were included. RESULTS Twenty studies with 3517 patients with 6205 NSCLC brain metastatic lesions were included. The majority of the best-performance models were ML-based (70%, 7/10), and deep learning (DL)-based models comprised 30% (6/20) of models. The area under the curve (AUC) and accuracy (ACC) of the best-performance models ranged from 0.765 to 1 and 0.69 to 0.93, respectively. The meta-analysis of the best-performance model revealed a pooled AUC of 0.91 (95%CI: 0.88-0.93) and ACC of 0.82 (95%CI: 0.79-0.86) along with a pooled sensitivity of 0.87 (95%CI: 0.83-0.9), specificity of 0.86 (95%CI: 0.79-0.9), and diagnostic odds ratio (DOR) of 35.2 (95%CI: 21.2-58.4). The subgroup analysis did not show significant differences between ML and DL models. CONCLUSION ML-based models demonstrated promising predictive outcomes in predicting EGFR status. Applying ML-based models in daily clinical practice can optimize treatment strategies and enhance clinical and radiological outcomes.
Collapse
Affiliation(s)
- Bardia Hajikarimloo
- Department of Neurological Surgery, Shohada Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ibrahim Mohammadzadeh
- Skull Base Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Salem M Tos
- Department of Neurological Surgery, University of Virginia, Charlottesville, VA, USA
| | - Mohammad Amin Habibi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Rana Hashemi
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Bahrami Hezaveh
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Dorsa Najari
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arman Hasanzade
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Hooshmand
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Bana
- Department of Neurosurgery, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Wang T, Shen Z, Yang L, Zhang X, Yu M, Yu S, Zhao B. The coagulation and tumor system are directly linked through the proteolysis and activation of epidermal growth factor receptor by thrombin. Oncogene 2025; 44:1153-1166. [PMID: 39910317 DOI: 10.1038/s41388-025-03296-1] [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: 10/23/2024] [Revised: 01/10/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025]
Abstract
Cancer cachexia and cancer-associated thrombosis are potentially fatal outcomes of advanced cancer. Unfortunately, this knowledge has not yet led to any breakthrough in cancer therapy. Thrombin is the key enzyme of blood coagulation system. The identification of a direct link between thrombin and the tumor progression remains unknown. We illustrated thrombin expression in lung adenocarcinoma (LUAD) was closely related to clinicopathological features, prognosis, and chemotherapy outcome of patients via TCGA and clinical pathological analysis. Using genetic and pharmacological approaches, we showed a direct link between thrombin catalytic activity and lung cancer progression in vitro and in vivo. Furthermore, we revealed that thrombin cleaves epidermal growth factor receptor (EGFR) at a GRG motif perfectly conserved across disparate species, indicating functional importance, which results in activation of EGFR/AKT/mTOR signaling pathway. Last we found the mutual interaction between thrombin and chemotherapy resistance. Combination therapy of thrombin inhibitor and chemotherapy results in improved anti-tumor efficacy. Together, our data firstly revealed a mechanism of cancer progression and chemotherapy resistance that involves thrombin-mediated EGFR cleavage. We propose that thrombin could be a prognostic biomarker for lung cancer, blockade of thrombin is a valuable therapeutic strategy to overcome cancer's resistance to chemotherapy.
Collapse
Affiliation(s)
- Tianfa Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhiyuan Shen
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Liu Yang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaohan Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Min Yu
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Qidong-Fudan Innovative Institute of Medical Sciences, Nantong, Jiangsu Province, China.
| | - Sanjian Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Bing Zhao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Qidong-Fudan Innovative Institute of Medical Sciences, Nantong, Jiangsu Province, China.
| |
Collapse
|
20
|
Manivannan HP, Veeraraghavan VP, Francis AP. Prediction of Multi-targeting Pharmacological Activity of Bioactive Compounds from Medicinal Plants Against Hepatocellular Carcinoma Through Advanced Network Pharmacology and Bioinformatics-Based Investigation. Appl Biochem Biotechnol 2025; 197:2979-3007. [PMID: 39820926 DOI: 10.1007/s12010-024-05150-8] [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] [Accepted: 12/24/2024] [Indexed: 01/19/2025]
Abstract
The primary objective of this study was to identify bioactive compounds from four medicinal plants with multi-targeting activity against hepatocellular carcinoma (HCC). A comprehensive analysis led to the identification of a subset of compounds possessing favorable drug-likeness, pharmacokinetics, and absence of toxicity profiles. Target analysis for 42 phytochemicals revealed 210 potential targets associated with HCC. Protein-protein interaction (PPI) analysis of these targets uncovered five critical hub genes, STAT3, SRC, AKT1, MAPK3, and EGFR, in our study. Correlation analysis of these hub genes indicated a strong positive correlation between EGFR, MAPK3, and SRC expression highlighting their interconnected roles in HCC. Survival analysis underscored the significant prognostic role of these hub genes in HCC underscoring their potential as biomarkers. The co-expression analysis unveiled an intricate network of interactions among the hub genes, while the enrichment analysis demonstrated their enrichment in diverse biological and signaling pathways related to HCC. Molecular docking analysis between the seven phytochemicals and five identified targets revealed that bauerenol exhibited good affinity towards all the targets. Subsequent molecular dynamics (MD) simulations demonstrated that bauerenol formed stable complexes with STAT3, AKT1, EGFR, and MAPK3, suggesting its potential as a multi-targeted inhibitor. Our research suggests that bauerenol shows promise as an inhibitor for HCC targets and stands out as a notable lead compound. However, further experimental studies are necessary to confirm its activity and to evaluate its potential as a therapeutic agent for HCC.
Collapse
Affiliation(s)
- Hema Priya Manivannan
- Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India
| | - Arul Prakash Francis
- Centre of Molecular Medicine and Diagnostics (COMManD), Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India.
| |
Collapse
|
21
|
Jiang F, Yang X, Shan L, Miao H, Shi C. 15-Deoxy-Δ-12,14-Prostaglandin J2 Represses Immune Escape of Lung Adenocarcinoma by Polarizing Macrophages Through Epidermal Growth Factor Receptor/Ras/Raf Pathway. J Immunother 2025; 48:119-126. [PMID: 39711151 DOI: 10.1097/cji.0000000000000546] [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: 09/11/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024]
Abstract
Lung adenocarcinoma (LUAD) is a widespread and deadly form of cancer. Prostaglandin 15-deoxy-Δ-12,14-prostaglandin J2 (15d-PGJ2) possesses antioxidant, anti-inflammatory, and anticancer properties. However, it is unclear whether this effect on LUAD progression stems from its ability to influence macrophage polarization. By utilizing 3- (4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), flow cytometry, colony formation, transwell assays, and enzyme linked immunosorbent assay (ELISA), we investigated how 15d-PGJ2 affects A549 cell viability, proliferation, apoptosis, and invasion, as well as levels of interleukin (IL)-4, IL-13, and IL-17. Human monocytic cell line THP-1 was induced into M2 macrophages using phorbol 12-myristate 13-acetate and IL-4/IL-13, followed by treatment with 15d-PGJ2. The study employed flow cytometry to observe the polarization of macrophages, quantitative reverse transcription polymerase chain reaction (qRT-PCR) to identify epidermal growth factor receptor (EGFR) expression, western blot for identifying expression of macrophage marker proteins, and examining EGFR/rat sarcoma (Ras)/rapidly accelerated fibrosarcoma (Raf) activation. In a coculture setup, CD8 + T cells were shown to have a proliferation capacity by carboxifluorescein diacetate succinimidyl ester (CFSE), a killing ability by lactate dehydrogenase, and an analysis of their interferon gamma and tumor necrosis factor alpha levels by ELISA. 15d-PGJ2 reduced invasion capacity and expression of inflammatory cytokines, lowered A549 cell viability in a dose-dependent way, and promoted apoptosis. 15d-PGJ2 facilitated the transition of M2 macrophages to the M1 type, inhibited Ras/Raf pathway activation, reduced EGFR expression in macrophages, and stimulated CD8 + T cells to enhance anti-tumor immunity. 15d-PGJ2 repressed M2 macrophage polarization and LUAD immune evasion by targeting the EGFR/Ras/Raf pathway in macrophages.
Collapse
Affiliation(s)
- Fan Jiang
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenling, China
| | - Xiaoxiao Yang
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenling, China
| | - Liqun Shan
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenling, China
| | - Huiwen Miao
- Department of Thoracic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chaohong Shi
- Department of Rehabilitation Medicine, The First People's Hospital of Wenling, Wenling, China
| |
Collapse
|
22
|
Wang R, Hu B, Pan Z, Mo C, Zhao X, Liu G, Hou P, Cui Q, Xu Z, Wang W, Yu Z, Zhao L, He M, Wang Y, Fu C, Wei M, Yu L. Antibody-Drug Conjugates (ADCs): current and future biopharmaceuticals. J Hematol Oncol 2025; 18:51. [PMID: 40307936 PMCID: PMC12044742 DOI: 10.1186/s13045-025-01704-3] [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: 12/26/2024] [Accepted: 04/13/2025] [Indexed: 05/02/2025] Open
Abstract
Antibody-drug conjugates (ADCs) represent a novel class of biopharmaceuticals comprising monoclonal antibodies covalently conjugated to cytotoxic agents via engineered chemical linkers. This combination enables targeted delivery of cytotoxic agents to tumor site through recognizing target antigens by antibody while minimizing off-target effects on healthy tissues. Clinically, ADCs overcome the limitations of traditional chemotherapy, which lacks target specificity, and enhance the therapeutic efficacy of monoclonal antibodies, providing higher efficacy and fewer toxicity anti-tumor biopharmaceuticals. ADCs have ushered in a new era of targeted cancer therapy, with 15 drugs currently approved for clinical use. Additionally, ADCs are being investigated as potential therapeutic candidates for autoimmune diseases, persistent bacterial infections, and other challenging indications. Despite their therapeutic benefits, the development and application of ADCs face significant challenges, including antibody immunogenicity, linker instability, and inadequate control over the release of cytotoxic agent. How can ADCs be designed to be safer and more efficient? What is the future development direction of ADCs? This review provides a comprehensive overview of ADCs, summarizing the structural and functional characteristics of the three core components, antibody, linker, and payload. Furthermore, we systematically assess the advancements and challenges associated with the 15 approved ADCs in cancer therapy, while also exploring the future directions and ongoing challenges. We hope that this work will provide valuable insights into the design and optimization of next-generation ADCs for wider clinical applications.
Collapse
Grants
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- No. U20A20413, China NSFC-Liaoning joint fund key program
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- 2023JH2/20200126 Liaoning Province Scientific Research Foundation
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
- NSFC, No. 81903658, 82272797, 82304564, China National Natural Science Foundation of China
Collapse
Affiliation(s)
- Ruili Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Baohui Hu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Ziyu Pan
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Chongxia Mo
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Xin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Guojia Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Ping Hou
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Qi Cui
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Zhao Xu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Wenjia Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
- Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, 110122, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
- Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, 110122, China
| | - Yan Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China
- Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, 110122, China
| | - Chen Fu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
- Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, 110122, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
- Liaoning Medical Diagnosis and Treatment Center, Shenyang, 110000, China.
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
- Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
23
|
Mei H, Yang J, Hao J, Ding Y, Wan X, Dong M, Zhang X, Luo L, Xiong T, Wang L, Yang T, Huang C. Mechanism of ethyl acetate fraction of Amorphophallus konjac against breast cancer based on network pharmacology, molecular docking and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119648. [PMID: 40107477 DOI: 10.1016/j.jep.2025.119648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/19/2025] [Accepted: 03/16/2025] [Indexed: 03/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Amorphophallus konjac (Sheliugu), a medicinal and edible herb from East China and regions south of the Yangtze River, exhibits significant antitumor activity. However, its active constituents and mechanisms remain poorly understood. AIMS OF THE STUDY This study explores the therapeutic effects of the konjac ethyl acetate fraction (KEAF) in triple-negative breast cancer (TNBC) and elucidates its underlying mechanisms. MATERIALS AND METHODS UPLC-MS/MS identified KEAF's chemical components, and network pharmacology determined its key targets in TNBC. Survival curves of essential genes were analyzed using UALCAN, bc-GenExMiner, and Kaplan-Meier plotter databases. Protein expression and prognostic data identified TNBC-linked genes. Molecular docking assessed binding affinities of KEAF's bioactive components with these genes. In vitro experiments validated KEAF's effects on proliferation, migration, cell cycle regulation, and apoptosis. RESULTS KEAF contained 15 active compounds and 146 principal targets, with eight key targets identified: TP53, EGFR, AKT1, MYC, STAT3, HIF1A, ESR1, and JUN. GO and KEGG enrichment analyses highlighted the PI3K/Akt signaling pathway as central to KEAF's therapeutic effects. Protein expression and prognostic studies confirmed EGFR and ESR1 as critical in TNBC progression. Molecular docking revealed strong binding of scutellarein and genistein to EGFR and ESR1 (T score >5). In vitro, KEAF inhibited MDA-MB-231 cell proliferation and migration, modulated the cell cycle, and induced apoptosis by downregulating PI3K/Akt signaling. CONCLUSION Scutellarein and genistein in KEAF exhibit therapeutic potential against TNBC by targeting EGFR and ESR1 and suppressing the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Huimin Mei
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Jinglong Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Jiapeng Hao
- Bijie Hospital of Traditional Chinese Medicine, Bijie, 551700, PR China
| | - Yushan Ding
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Xinliang Wan
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Minghong Dong
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Xudong Zhang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Liying Luo
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Tongtong Xiong
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Lu Wang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China
| | - Tianming Yang
- Bijie Hospital of Traditional Chinese Medicine, Bijie, 551700, PR China.
| | - Cong Huang
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, PR China; Anshun Hospital of Traditional Chinese Medicine, Anshun, 561000, PR China.
| |
Collapse
|
24
|
Zhu F, Qiu J, Ye H, Su W, Wang R, Fu Y. The Prognostic Significance of Epidermal Growth Factor Receptor Amplification and Epidermal Growth Factor Receptor Variant III Mutation in Glioblastoma: A Systematic Review and Meta-Analysis with Implications for Targeted Therapy. Int J Mol Sci 2025; 26:3539. [PMID: 40331985 PMCID: PMC12027172 DOI: 10.3390/ijms26083539] [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: 03/04/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Glioblastoma (GBM) is the most aggressive and heterogeneous neoplasm among central nervous system tumors, with a dismal prognosis and a high recurrence rate. Among the various genetic alterations found in GBM, the amplification of epidermal growth factor receptor (EGFR) and the EGFR variant III (EGFRvIII) mutation are among the most common, though their prognostic value remains controversial. This systematic review and meta-analysis aimed to provide a comprehensive evaluation of the diagnostic and prognostic significance of EGFR amplification and the EGFRvIII mutation in GBM patients, incorporating recent studies published in the past few years to offer a more complete and up-to-date analysis. An extensive search of the PubMed, Web of Science, and Scopus databases was conducted, including studies that reported on EGFR and/or the EGFRvIII mutation status with detailed survival data. A total of 32 studies with 4208 GBM patients were included. The results indicated that EGFR amplification significantly correlated with worse OS (Overall survival) (HR = 1.27, 95% CI: 1.03-1.57), suggesting that EGFR amplification is an independent prognostic marker. The prognostic value of EGFRvIII was inconclusive, with a pooled hazard ratio for overall survival of 1.13 (95% CI: 0.94-1.36), indicating no significant effect on survival in the general population. However, a subgroup analysis suggested that EGFRvIII may be associated with poorer outcomes, particularly in recurrent GBM patients, where its prognostic significance became more evident. Furthermore, subgroup analyses based on geographic region revealed significant heterogeneity in the prognostic impact of EGFR amplification across different populations. In American cohorts, EGFR amplification was strongly associated with an increased risk of mortality (HR = 1.53, 95% CI: 1.28-1.84, p = 0.001), suggesting that it serves as a more reliable prognostic marker in this region. In contrast, no significant prognostic impact of EGFR amplification was observed in Asian (HR = 0.64, 95% CI: 0.35-1.17) or European (HR = 0.98, 95% CI: 0.80-1.19) populations. Overall, this study underscores the potential of EGFR amplification as a prognostic marker in GBM, while further research is needed to fully elucidate the role of the EGFRvIII mutation, particularly in specific patient subgroups. Clarifying these associations could offer important insights for targeted treatment strategies, improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Renxi Wang
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, No. 10 Xitoutiao, Beijing 100069, China; (F.Z.); (J.Q.); (H.Y.); (W.S.)
| | - Yi Fu
- Laboratory of Brain Disorders, Beijing Institute of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, No. 10 Xitoutiao, Beijing 100069, China; (F.Z.); (J.Q.); (H.Y.); (W.S.)
| |
Collapse
|
25
|
Mulliqi E, Khelwatty S, Bagwan I, Kamaludin A, Morgan A, Long N, Ashkan K, Modjtahedi H. The Co-Expression and Cellular Location of HER Family Members, EGFRvIII, Putative Cancer Stem Cell Biomarkers CD44 and CD109 in Patients with Glioblastoma, and Their Impacts on Prognosis. Cancers (Basel) 2025; 17:1221. [PMID: 40227788 PMCID: PMC11987930 DOI: 10.3390/cancers17071221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES The aberrant expression and activation of HER family members is a known major oncogenic pathway for the proliferation, progression, and metastasis of a wide range of human malignancies. In this study, our aim was to examine the relative expression and prognostic significance of all members of the HER family, the type III EGFR mutant (EGFRvIII), and the putative stem cell markers CD44 and CD109 in patients with glioblastoma. METHODS The expression levels of wild-type EGFR (wtEGFR), HER2, HER3, HER4, EGFRvIII, CD44, and CD109 were determined in tumour specimens from 80 patients by immunohistochemistry. The staining was scored based on the percentage of positive tumour cells, the intensity, and the cellular location of immunostaining. The association between the expression level of the biomarkers and patient overall survival was evaluated using Chi-squared, Kaplan-Meier survival curves, and log-rank tests. RESULTS At a cut-off value of ≥5% with positive staining, 46% (wtEGFR), 75% (HER2), 19% (HER3), 71% (HER4), 85% (EGFRvIII), 95% (CD44), and 16% (CD109) of the cases were positive for these biomarkers. Interestingly, at the same cut-off value, the expression of wtEGFR in these patients was accompanied by co-expression with HER2 (35%), HER3 (0%), HER4 (30%), EGFRvIII (36%), CD44 (44%), HER2/EGFRvIII (28%), HER2/CD44 (31%), and EGFRvIII/CD44 (36%). In addition, the expression of EGFRvIII was accompanied by co-expression with HER2 (65%), HER3 (15%), HER4 (63%), CD44 (83%), CD109 (16%), wtEGFR/HER2 (28%), and 55% of the cases had co-expression of EGFRvIII/HER2/HER4/CD44. With the exception of HER2 expression, at cut-off values of ≥5% of tumour cells with positive staining, which was associated with better overall survival [HR = 0.57 (p = 0.038), HR = 0.56 (p = 0.034)], there was no significant association between the expression of other members of the HER family, EGFRvIII, CD44, and CD109 on the overall survival in both univariate and multivariate analysis. Conclusions Our results suggest that the co-expression of different members of the HER family, with EGFRvIII, CD44, and CD109, occurs in patients with glioblastoma. As the results of therapy with EGFR inhibitors have not been encouraging in patients with a brain tumour, further investigation should determine whether the co-expression of such biomarkers can be of predictive value for the response to the therapy with various types of HER inhibitors and their potential as therapeutic targets for co-targeted therapy.
Collapse
Affiliation(s)
- Ermira Mulliqi
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| | - Said Khelwatty
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| | - Izhar Bagwan
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
- Berkshire Surrey Pathology Services, Royal Surrey Hospital, Guildford GU2 7XX, UK
| | - Ahmad Kamaludin
- Department of Neurosurgery, Kings College Hospital, Denmark Hill, London SE5 9RS, UK; (A.K.); (N.L.); (K.A.)
| | - Anna Morgan
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| | - Natalie Long
- Department of Neurosurgery, Kings College Hospital, Denmark Hill, London SE5 9RS, UK; (A.K.); (N.L.); (K.A.)
| | - Keyoumars Ashkan
- Department of Neurosurgery, Kings College Hospital, Denmark Hill, London SE5 9RS, UK; (A.K.); (N.L.); (K.A.)
| | - Helmout Modjtahedi
- School of Life Science, Pharmacy and Chemistry, Faculty of Health, Science, Social Care and Education, Kingston University London, Kingston-upon-Thames KT1 2EE, UK; (E.M.); (S.K.); (I.B.); (A.M.)
| |
Collapse
|
26
|
Li D, Gao Z, Zhang Z, Chen H, Tang R, Zhou L, Ye Y, Lin J, Zhou P, Wang C, Feng X, He Y, Meng Z, Zheng M, Lu W, Feng Z, Wang L, Pei Y, Yang J, Tao T, Zhang X, Jiang L. Suprabasin promotes gastric cancer liver metastasis via hepatic stellate cells-mediated EGF/CCL2/JAK2 intercellular signaling pathways. Oncogene 2025:10.1038/s41388-025-03370-8. [PMID: 40181153 DOI: 10.1038/s41388-025-03370-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Gastric cancer is among the most prevalent gastrointestinal tumors, with liver metastasis significantly worsening patient outcomes. While hepatic stellate cell activation is crucial in hepatocellular carcinoma progression and liver metastasis, its role in gastric cancer liver metastasis is not well understood. In this study, we identified Suprabasin (SBSN) as a key oncogene driving gastric cancer liver metastasis. SBSN was upregulated in gastric cancer tissues and further elevated in liver metastasis, correlating with poor prognosis. Mechanistically, SBSN promoted proliferation, migration, and invasion of gastric cancer cells by activating the STAT3 signaling pathway, as shown in vitro and in vivo. Using a co-culture model of gastric cancer cells and hepatic stellate cell line LX-2, we found that increased SBSN expression in gastric cancer cells triggered EGF secretion, activating LX-2 cells through the EGF/EGFR axis. Activated LX-2 cells then secreted CCL2, initiating the CCL2/CCR2/JAK2 signaling pathway in gastric cancer cells, facilitating their migration to the liver and promoting colonization and growth. Our findings highlight the prognostic significance of SBSN in gastric cancer and liver metastasis, suggesting it as a potential biomarker for disease progression. The SBSN-mediated EGF/EGFR and CCL2/CCR2/JAK2 signaling axes are critical for LX-2 activation and gastric cancer cell migration, offering a rationale for targeting SBSN in treating gastric cancer liver metastasis.
Collapse
Affiliation(s)
- Difeng Li
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
- Department of Pathology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhiqing Gao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
- Department of Molecular Medicine, School of Basic Medicine, Southern Medical University, Guangzhou, China
| | - Zhuojun Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Han Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Ruiming Tang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Lihuan Zhou
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Yingmin Ye
- Department of Pathogen Biology and Immunology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiaqian Lin
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Ping Zhou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Chanjuan Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China
| | - Xiaoli Feng
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Yaoming He
- Department of Gastrointestinal Surgery, Jiangmen Central Hospital, Jiangmen, China
| | - Zijie Meng
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Central Hospital, Jiangmen, China
| | - Mingzhu Zheng
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Central Hospital, Jiangmen, China
| | - Wenjie Lu
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Central Hospital, Jiangmen, China
| | - Zhengfu Feng
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China
| | - Lan Wang
- Department of Pathogen Biology and Immunology, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuanyuan Pei
- Department of Central Laboratory, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, China
| | - Jianan Yang
- Department of Urologic Oncosurgery, Guangzhou Institute of Cancer Research, The Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tianyu Tao
- Cancer Institute, Southern Medical University, Guangzhou, China
| | - Xin Zhang
- Clinical Experimental Center, Jiangmen Engineering Technology Research Center of Clinical Biobank and Translational Research, Jiangmen Central Hospital, Jiangmen, China.
| | - Lili Jiang
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, China.
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
27
|
Topalan E, Büyükgüngör A, Çiğdem M, Güra S, Sever B, Otsuka M, Fujita M, Demirci H, Ciftci H. A Structural Insight Into Two Important ErbB Receptors (EGFR and HER2) and Their Relevance to Non-Small Cell Lung Cancer. Arch Pharm (Weinheim) 2025; 358:e2400992. [PMID: 40194950 PMCID: PMC11975551 DOI: 10.1002/ardp.202400992] [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: 12/31/2024] [Revised: 02/23/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
The epidermal growth factor receptor (EGFR) family, comprising receptor tyrosine kinases (RTK) such as EGFR and HER2, plays a critical role in various signaling pathways related to cell proliferation, differentiation, and growth. EGFR overactivation due to aberrant signaling can lead to various cancers, including non-small cell lung cancer (NSCLC). To develop treatment for EGFR-related NSCLC, several tyrosine kinase inhibitors (TKIs) were designed: gefitinib, erlotinib, as first-generation; neratinib, dacomitinib as second-generation; osimertinib, lazertinib as third-generation, as examples. However, due to the acquired resistance by the mutations such as EGFRT790M and EGFRC797S together with the exon 20 insertion mutations, these drugs do not provide promising results for NSCLC patients. The development of fourth-generation inhibitors like EAI045 and further innovative drugs to overcome this resistance problem is a must to cure EGFR-related NSCLC. Among these, pyrazoline-thiazole scaffolds are found effective as EGFR-HER2 inhibitors against NSCLC, making them promising drug candidates. Although structures obtained so far for the EGFR family provide meaningful insights into the mechanisms, the quality and the quantity of the EGFR family structures are insufficient to elucidate the complete structures and functions to overcome NSCLC. This review evaluates the structures of EGFR-HER2 and investigates their relation to NSCLC.
Collapse
Affiliation(s)
- Edanur Topalan
- Department of Molecular Biology and GeneticsKoc UniversityIstanbulTürkiye
| | - Ahmet Büyükgüngör
- Department of Molecular Biology and GeneticsKoc UniversityIstanbulTürkiye
- Department of Molecular Biology and GeneticsIstanbul Technical UniversityIstanbulTürkiye
| | - Melih Çiğdem
- Department of Molecular Biology and GeneticsKoc UniversityIstanbulTürkiye
- Department of Biological SciencesMiddle East Technical UniversityAnkaraTürkiye
| | - Sinan Güra
- Department of Molecular Biology and GeneticsKoc UniversityIstanbulTürkiye
- Graduate School of Biology & HealthUniversité Paris SaclayOrsayFrance
| | - Belgin Sever
- Department of Pharmaceutical Chemistry, Faculty of PharmacyAnadolu UniversityEskisehirTürkiye
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Masami Otsuka
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
- Department of Drug DiscoveryScience Farm Ltd.KumamotoJapan
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Hasan Demirci
- Department of Molecular Biology and GeneticsKoc UniversityIstanbulTürkiye
| | - Halilibrahim Ciftci
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
- Department of Drug DiscoveryScience Farm Ltd.KumamotoJapan
- Department of Molecular Biology and GeneticsMehmet Akif Ersoy UniversityBurdurTürkiye
- Department of Bioengineering SciencesIzmir Katip Celebi UniversityIzmirTürkiye
| |
Collapse
|
28
|
Yan Y, Zhang Y, Liu J, Chen B, Wang Y. Emerging magic bullet: subcellular organelle-targeted cancer therapy. MEDICAL REVIEW (2021) 2025; 5:117-138. [PMID: 40224364 PMCID: PMC11987508 DOI: 10.1515/mr-2024-0044] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/27/2024] [Indexed: 04/15/2025]
Abstract
The therapeutic efficacy of anticancer drugs heavily relies on their concentration and retention at the corresponding target site. Hence, merely increasing the cellular concentration of drugs is insufficient to achieve satisfactory therapeutic outcomes, especially for the drugs that target specific intracellular sites. This necessitates the implementation of more precise targeting strategies to overcome the limitations posed by diffusion distribution and nonspecific interactions within cells. Consequently, subcellular organelle-targeted cancer therapy, characterized by its exceptional precision, have emerged as a promising approach to eradicate cancer cells through the specific disruption of subcellular organelles. Owing to several advantages including minimized dosage and side effect, optimized efficacy, and reversal of multidrug resistance, subcellular organelle-targeted therapies have garnered significant research interest in recent years. In this review, we comprehensively summarize the distribution of drug targets, targeted delivery strategies at various levels, and sophisticated strategies for targeting specific subcellular organelles. Additionally, we highlight the significance of subcellular targeting in cancer therapy and present essential considerations for its clinical translation.
Collapse
Affiliation(s)
- Yue Yan
- Department of Central Laboratory, Peking University First Hospital, Beijing, China
| | - Yimeng Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jianxiong Liu
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Binlong Chen
- Department of Central Laboratory, Peking University First Hospital, Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery System, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Chemical Biology Center, Peking University, Beijing, China
| |
Collapse
|
29
|
Banu H, Al-Shammari E, Shahanawaz S, Azam F, Patel M, Alarifi NA, Ahmad MF, Adnan M, Ashraf SA. Insights into the Therapeutic Targets and Molecular Mechanisms of Eruca sativa Against Colorectal Cancer: An Integrated Approach Combining Network Pharmacology, Molecular Docking and Dynamics Simulation. Pharmaceuticals (Basel) 2025; 18:453. [PMID: 40283891 PMCID: PMC12030293 DOI: 10.3390/ph18040453] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/12/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: This study presents a novel and comprehensive investigation into the anti-colorectal cancer (CRC) effects and underlying mechanisms of Eruca sativa (E. sativa) using an integrated approach combining network pharmacology, molecular docking and molecular dynamics simulation. Methods: Using an integrated approach, six bioactive compounds and 40 potential targets were identified. A compound-target network was constructed, and enrichment analysis was performed to explore the key pathways influenced by E. sativa. Molecular docking analysis was used to evaluate the binding interactions between the identified compounds and key CRC-related targets (AKT1, PGR, MMP9, and PTGS2). Furthermore, molecular dynamics simulation was utilized to confirm the stability and reliability of these interactions. Results: The study found that E. sativa exhibits strong anticancer potential, particularly through major compounds such as β-ionone, 1-octanol, isorhamnetin, 2-hexenal, propionic acid, and quercetin. Molecular docking revealed favorable binding interactions between these compounds and key CRC targets, with quercetin and isorhamnetin showing the highest binding affinities. Additionally, molecular dynamics simulations validated the stability of these interactions, reinforcing their therapeutic relevance. Conclusions: This study provides valuable insights into the pharmacological mechanisms of E. sativa against CRC, highlighting its potential as a natural anticancer agent. These findings pave the way for future clinical studies to validate the efficacy and safety of E. sativa and its bioactive compounds, potentially contributing to the development of novel, plant-based therapeutic strategies for CRC treatment.
Collapse
Affiliation(s)
- Humera Banu
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (H.B.)
| | - Eyad Al-Shammari
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (H.B.)
| | - Syed Shahanawaz
- Department of Physiotherapy, College of Applied Medical Sciences, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia
| | - Faizul Azam
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia;
| | - Mitesh Patel
- Research and Development Cell, Parul University, Vadodara 391760, Gujarat, India;
- Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, Gujarat, India
| | | | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia;
| | - Syed Amir Ashraf
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Ha’il, Ha’il P.O. Box 2440, Saudi Arabia; (H.B.)
| |
Collapse
|
30
|
Wang H, Wang H, Wang R, Li Y, Wang Z, Zhou W, Deng L, Li X, Zou L, Yang Q, Lai R, Qi X, Nie J, Jiao B. Discovery of a molecular glue for EGFR degradation. Oncogene 2025; 44:545-556. [PMID: 39627505 DOI: 10.1038/s41388-024-03241-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 02/19/2025]
Abstract
Aberrant expression of epidermal growth factor receptor (EGFR) plays a critical role in the pathogenesis of various tumors, potentially representing a target for therapeutic intervention. Nonetheless, EGFR remains a challenging protein to target pharmacologically in triple-negative breast cancer (TNBC). An emerging approach to address the removal of such proteins is the application of molecular glue (MG) degraders. These compounds facilitate protein-protein interactions between a target protein and an E3-ubiquitin ligase, subsequently leading to protein degradation. Herein, we identified a new MG (CDDO-Me, C-28 methyl ester of 2-cyano-3, 12-dioxooleana-1, 9(11)-dien-28-oic acid), which orchestrated binding between EGFR and KEAP1 (an E3-ubiquitin ligase adapter), thereby initiating the ubiquitination and degradation of EGFR. CDDO-Me directly interacted with the tyrosine kinase (TK) domain of EGFR, resulting in its degradation via an autophagy-dependent lysosomal pathway. Knockdown of KEAP1 decreased the degradation of EGFR by reducing its K63-linked ubiquitination, leading to diminished EGFR colocalization in autophagosomes and lysosomes. Notably, CDDO-Me attenuates TNBC progression by accelerating EGFR degradation in cell-derived xenografts and patient-derived organoid models, highlighting its clinical application potential. Consequently, induction of EGFR degradation through MG degraders represents a viable therapeutic strategy for TNBC.
Collapse
Affiliation(s)
- Hairui Wang
- Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Hui Wang
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| | - Rui Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Yuanzhen Li
- Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Zhipeng Wang
- China West Normal University, Nanchong, Sichuan, China
| | - Wenshen Zhou
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Li Deng
- Jianyang City People's Hospital, Chengdu, Sichuan, China
| | - Xiyin Li
- Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Li Zou
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Qin Yang
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Ren Lai
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, China.
| | - Jianyun Nie
- Department of Breast Cancer, Third Affiliated Hospital, Kunming Medical University, Kunming, Yunnan, China.
| | - Baowei Jiao
- National Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
- KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.
| |
Collapse
|
31
|
Zhang M, Zhou G, Xu Y, Wei B, Liu Q, Zhang G, Chang R. Immunogenic cell death signature predicts survival and reveals the role of VEGFA + Mast cells in lung adenocarcinoma. Sci Rep 2025; 15:7213. [PMID: 40021802 PMCID: PMC11871002 DOI: 10.1038/s41598-025-91401-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
Lung cancer is prevalent worldwide and is a major cause of cancer-related mortality. Despite being the primary model for immunotherapy research, the response rates of lung cancer patients to immunotherapy are unsatisfactory. Furthermore, research on immunogenic cell death (ICD) in lung cancer is limited, which limits the development of strategies that combine ICD-related therapies with immunotherapy. In this study, we compiled and summarized 69 genes associated with ICD and developed an IRS. Across seven independent datasets, the IRS was identified as an independent prognostic factor. IRS was positively associated with multiple tumor proliferation pathways and negatively associated with immune-related pathways. Additionally, IRS negatively correlated with the infiltration of various immune cells, supporting its association with survival outcomes. Based on the correlation between IRS and immune activity, we validated the ability of IRS to predict immunotherapy efficacy across seven immunotherapy datasets and demonstrated that patients who respond to immunotherapy tend to have a lower IRS. Moreover, utilizing single-cell RNA sequencing, we revealed the role of mast cells in the TME with the highest IRS. Through interactions with various receptors on macrophages, endothelial cells, and tumor cells, mast cells promote tumor progression, providing a comprehensive explanation for poor prognosis and lack of response to immunotherapy in patients with high IRS. Our study offers new guidance for combination therapies in lung adenocarcinoma patients and elucidated the mechanism by which mast cells contribute to cancer development within the TME.
Collapse
Affiliation(s)
- Meng Zhang
- The Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Guowei Zhou
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yantao Xu
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Benliang Wei
- Big Data Institute, Central South University, Changsha, Hunan, China
| | - Qian Liu
- Big Data Institute, Central South University, Changsha, Hunan, China
| | - Guanxiong Zhang
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Ruimin Chang
- The Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
| |
Collapse
|
32
|
Liu A, Wen J, Zhao K, Jiang L, Meng X. Case Report: Grade 4 pneumonitis occurred after thoracic radiotherapy and dacomitinib in a patient with lung adenocarcinoma. Front Oncol 2025; 15:1436134. [PMID: 40071087 PMCID: PMC11893403 DOI: 10.3389/fonc.2025.1436134] [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: 05/21/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Osimertinib combined with chest radiotherapy has a high incidence of pneumonia, dacomitinib is widely used in clinical practice, but there are no studies reporting the pulmonary safety of dacomitinib in combinating with radiotherapy. Here we report a case of radiation pneumonitis occurring by dacomitinib and thoracic radiotherapy (TRT). The patient was a 55-year-old woman with lung adenocarcinoma. She had received surgery and adjuvant chemotherapy. The patient presented with bilateral intramammary and para-aortic metastatic lymphadenopathy, which was confirmed as metastasis, and subsequently received treatment with dacomitinib. Radiotherapy started after 4 months of dacomitinib. The Clinical Target Volume (CTV) was metastatic lymph nodes area. The prescription dose was 60 Gy/30F. The mean lung dose (MLD), V20, and V5 were 8.16Gy, 16%, and 34.5%. Despite the lung V20 and mean lung dose being exceptionally low, the patient exhibited respiratory symptoms, and a CT chest scan revealed grade 4 radiation pneumonitis two weeks following the conclusion of radiotherapy. The radiotherapy and dacomitinib were discontinued, and immediate initiation of pulmonary anti-inflammatory treatment ensued. The concurrent administration of dacomitinib and RT carries the risk of inducing serious pneumonia. This case highlights the potential risk of severe pneumonia associated with this combination therapy, emphasizing the need for further research to clarify its safety and develop effective management strategies.
Collapse
Affiliation(s)
- Ailing Liu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Junxu Wen
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Kaikai Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Liyang Jiang
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
33
|
Hong Z, Huang X, Xia L, Liang T, Bai X. Reciprocal regulation of MMP-28 and EGFR is required for sustaining proliferative signaling in PDAC. J Exp Clin Cancer Res 2025; 44:68. [PMID: 39994761 PMCID: PMC11849219 DOI: 10.1186/s13046-025-03323-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUD Sustaining proliferation signaling is the top hallmarks of cancer, driving continuous tumor growth and resistance to drug treatments. Blocking proliferation signaling has shown limited benefit in clinical treatment of pancreatic ductal adenocarcinoma, highlighting the urgent need to deeply understand proliferation signaling and develop new therapeutic strategies. METHODS By leveraging clinical data and data from the TCGA and GDSC datasets, we investigated the association between MMP-28 expression and the sensitivity to EGFR inhibitors as well as the prognosis of PDAC. Transcriptomic and biological experiments explore the regulatory role of MMP-28 on the EGFR signaling pathway. Additionally, in vitro and in vivo studies are employed to evaluate MMP-28 as a biomarker for sensitivity to EGFR inhibitors. RESULTS We found that MMP-28, a metalloproteinase, was significantly associated with the sensitivity to EGFR inhibitors. Furthermore, MMP-28 could promote PDAC growth and metastasis. Mechanistically, MMP-28 facilitated the maturation and release of the TGF-α precursor, thus promoting EGFR activation. In return, EGFR upregulated MMP-28 through AP-1-mediated transcription, forming a positive feedback loop that provided sustaining proliferation signaling for PDAC. Subsequently, MMP-28 was identified to predict the response to EGFR inhibitors and recognize responsive patients. CONCLUSIONS Our findings revealed the role of MMP-28 and EGFR in generation of sustaining proliferation signaling and provided a new therapy strategy for PDAC.
Collapse
Affiliation(s)
- Zhengtao Hong
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xing Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Zhejiang University Cancer Center, Hangzhou, 310063, China.
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, 310003, China.
| | - Linghao Xia
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Zhejiang Province, Hangzhou, 31003, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China.
- Zhejiang University Cancer Center, Hangzhou, 310063, China.
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, 310003, China.
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Zhejiang Province, Hangzhou, 31003, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China.
- Zhejiang University Cancer Center, Hangzhou, 310063, China.
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, 310003, China.
| |
Collapse
|
34
|
Vo DK, Trinh KTL. Polymerase Chain Reaction Chips for Biomarker Discovery and Validation in Drug Development. MICROMACHINES 2025; 16:243. [PMID: 40141854 PMCID: PMC11944077 DOI: 10.3390/mi16030243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025]
Abstract
Polymerase chain reaction (PCR) chips are advanced, microfluidic platforms that have revolutionized biomarker discovery and validation because of their high sensitivity, specificity, and throughput levels. These chips miniaturize traditional PCR processes for the speed and precision of nucleic acid biomarker detection relevant to advancing drug development. Biomarkers, which are useful in helping to explain disease mechanisms, patient stratification, and therapeutic monitoring, are hard to identify and validate due to the complexity of biological systems and the limitations of traditional techniques. The challenges to which PCR chips respond include high-throughput capabilities coupled with real-time quantitative analysis, enabling researchers to identify novel biomarkers with greater accuracy and reproducibility. More recent design improvements of PCR chips have further expanded their functionality to also include digital and multiplex PCR technologies. Digital PCR chips are ideal for quantifying rare biomarkers, which is essential in oncology and infectious disease research. In contrast, multiplex PCR chips enable simultaneous analysis of multiple targets, therefore simplifying biomarker validation. Furthermore, single-cell PCR chips have made it possible to detect biomarkers at unprecedented resolution, hence revealing heterogeneity within cell populations. PCR chips are transforming drug development, enabling target identification, patient stratification, and therapeutic efficacy assessment. They play a major role in the development of companion diagnostics and, therefore, pave the way for personalized medicine, ensuring that the right patient receives the right treatment. While this tremendously promising technology has exhibited many challenges regarding its scalability, integration with other omics technologies, and conformity with regulatory requirements, many still prevail. Future breakthroughs in chip manufacturing, the integration of artificial intelligence, and multi-omics applications will further expand PCR chip capabilities. PCR chips will not only be important for the acceleration of drug discovery and development but also in raising the bar in improving patient outcomes and, hence, global health care as these technologies continue to mature.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea;
| | - Kieu The Loan Trinh
- Bionano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
35
|
Szerenyi D, Jarvas G, Guttman A. Multifaceted Approaches in Epithelial Cell Adhesion Molecule-Mediated Circulating Tumor Cell Isolation. Molecules 2025; 30:976. [PMID: 40076201 PMCID: PMC11901967 DOI: 10.3390/molecules30050976] [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: 11/30/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Circulating tumor cells (CTCs) are pivotal in cancer metastasis and serve as valuable biomarkers for diagnosis, prognosis, and treatment monitoring. Traditional CTC capture methods predominantly utilize the epithelial cell adhesion molecule (EpCAM) as a marker for isolation. However, the heterogeneity of these circulating cells and the epithelial-to-mesenchymal transition process (wherein epithelial cells acquire mesenchymal characteristics) limit the efficacy of EpCAM-based capture techniques. In this paper, we critically review the role of the EpCAM in CTC capture, explore the impact of epithelial-to-mesenchymal transition on EpCAM expression, and discuss alternative biomarkers and strategies to enhance CTC isolation. By evaluating the limitations of EpCAM-mediated capture and the challenges posed by epithelial-to-mesenchymal transition, we aim to provide insights into the development of more comprehensive liquid biopsy approaches for cancer management.
Collapse
Affiliation(s)
- Dora Szerenyi
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, 8200 Veszprem, Hungary;
| | - Gabor Jarvas
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, 8200 Veszprem, Hungary;
- CAPTEC Medical Ltd., 8200 Veszprem, Hungary
| | - Andras Guttman
- Research Institute of Biomolecular and Chemical Engineering, Faculty of Engineering, University of Pannonia, 8200 Veszprem, Hungary;
- CAPTEC Medical Ltd., 8200 Veszprem, Hungary
- Horváth Csaba Memorial Laboratory of Bioseparation Sciences, Research Center for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
36
|
Xia H, Tai XJ, Cheng W, Wu Y, He D, Wang LF, Liu H, Zhang SY, Sun YT, Liu HZ, Liu DD, Zhao HZ, Ji FY, Li XH. Metformin inhibits the growth of SCLC cells by inducing autophagy and apoptosis via the suppression of EGFR and AKT signalling. Sci Rep 2025; 15:6081. [PMID: 39971923 PMCID: PMC11839993 DOI: 10.1038/s41598-025-87537-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Small cell lung cancer (SCLC) is a therapeutically challenging disease. Metformin, an effective agent for the treatment of type 2 diabetes, has been shown to have antitumour effects on many cancers, including non-small cell lung cancer (NSCLC) and breast cancer. Currently, the antitumour effects of metformin on SCLC and the underlying molecular mechanisms remain unclear. CCK-8, EdU, colony formation, flow cytometry, immunofluorescence, molecular docking, western blotting, nude mouse transplanted tumour model, and immunohistochemistry experiments were conducted to analyse gene functions and the underlying mechanism involved. In vitro experiments demonstrated that metformin inhibited the growth of SCLC cells (H446, H526, H446/DDP and H526/DDP), which was confirmed in xenograft mouse models in vivo. Additionally, metformin induced cell cycle arrest, apoptosis, and autophagy in these SCLC cells. The molecular docking results indicated that metformin has a certain binding affinity for EGFR. The western blotting results revealed that metformin decreased the expression of EGFR, p-EGFR, AKT, and p-AKT, which could be reversed by EGF and SC79. Moreover, metformin activated AMPK and inactivated mTOR, and compound C and SC79 increased the levels of p-mTOR. Metformin can not only enhance the antitumour effect of cisplatin but also alleviate the toxic effects of cisplatin on the organs of xenograft model animals. In summary, the current study revealed that metformin inhibits the growth of SCLC by inducing autophagy and apoptosis via suppression of the EGFR/AKT/AMPK/mTOR pathway. Metformin might be a promising candidate drug for combination therapy of SCLC.
Collapse
Affiliation(s)
- Hong Xia
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Xue-Jiao Tai
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Wang Cheng
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Yi Wu
- Department of Oncology, Taihe Hospital, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Dan He
- School of Biomedical Engineering, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Li-Feng Wang
- School of Biomedical Engineering, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Hao Liu
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Shen-Yi Zhang
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Yu-Ting Sun
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Hang-Zhi Liu
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Dan-Dan Liu
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Hu-Zi Zhao
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China
| | - Fu-Yun Ji
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China.
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing, 400039, China.
| | - Xi-Hua Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Science, Hubei University of Medicine, 30 Renmin Road, Shiyan, 442000, Hubei, China.
| |
Collapse
|
37
|
Zhao C, Cheng L, Li A, Wang H, Li X, Xu J. EGFR-Mutant Lung Adenocarcinoma Cell-Derived Exosomal miR-651-5p Induces CD8+ T Cell Apoptosis via Downregulating BCL2 Expression. Biomedicines 2025; 13:482. [PMID: 40002895 PMCID: PMC11852681 DOI: 10.3390/biomedicines13020482] [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: 12/18/2024] [Revised: 02/08/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Background: The efficacy of programmed cell death 1 (PD-1) or ligand 1 (PD-L1) inhibitors in epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC) patients is not satisfactory. Studies have indicated that the ratio of CD8+ tumor infiltration lymphocytes (TILs) was associated with immunotherapy efficacy; however, it was significantly lower in EGFR-mutant than wild type patients. The underlying mechanisms need to be studied. Methods: Database analysis, clinical specimens, small RNA sequencing, and single-cell sequencing were used to analyze miRNA expression and immune cell infiltration. Cell co-culture and flow cytometry were conducted to detect immune cell apoptosis. The mouse model was performed to analyze the influence of miR-651-5p antagomirs on the tumor microenvironment. Results: The miR-651-5p was found to be highly expressed in EGFR-mutant lung adenocarcinoma cell-derived exosomes, which could promote CD8+ T cell apoptosis, while the miR-651-5p inhibitor decreased the ratio of PC9-secreted exosomes and induced apoptosis. Mechanistically, the EGFR signaling pathway promoted the expression of miR-651-5p by activating the transcription factor Fos proto-oncogene (FOS) in EGFR-mutant lung adenocarcinoma cell lines. B-cell lymphoma 2 (BCL2) was the target of miR-651-5p, and miR-651-5p could promote T cell apoptosis by inhibiting BCL2 expression. In addition, the miR-651-5p antagomir increased T cell infiltration and enhanced the efficacy of the PD-1 inhibitor treating the EGFR-mutant lung adenocarcinoma humanized mouse model. Conclusions: EGFR-mutant lung adenocarcinoma promotes T cell apoptosis through exosomal miR-651-5p. miR-651-5p antagonists increase immune cell infiltration and enhance the anti-tumor effect of PD-1 inhibitor, suggesting a new combination therapy to improve the efficacy of immunotherapy in EGFR-mutant NSCLC patients.
Collapse
Affiliation(s)
- Chao Zhao
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; (C.Z.); (L.C.)
| | - Lei Cheng
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; (C.Z.); (L.C.)
| | - Aiwu Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Haowei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Xuefei Li
- Department of Lung Cancer and Immunology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China; (C.Z.); (L.C.)
| | - Jun Xu
- Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| |
Collapse
|
38
|
Yadav M, Sharma A, Patne K, Tabasum S, Suryavanshi J, Rawat L, Machaalani M, Eid M, Singh RP, Choueiri TK, Pal S, Sabarwal A. AXL signaling in cancer: from molecular insights to targeted therapies. Signal Transduct Target Ther 2025; 10:37. [PMID: 39924521 PMCID: PMC11808115 DOI: 10.1038/s41392-024-02121-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/02/2024] [Accepted: 12/19/2024] [Indexed: 02/11/2025] Open
Abstract
AXL, a member of the TAM receptor family, has emerged as a potential target for advanced-stage human malignancies. It is frequently overexpressed in different cancers and plays a significant role in various tumor-promoting pathways, including cancer cell proliferation, invasion, metastasis, epithelial-mesenchymal transition (EMT), angiogenesis, stemness, DNA damage response, acquired therapeutic resistance, immunosuppression, and inflammatory responses. Beyond oncology, AXL also facilitates viral infections, including SARS-CoV-2 and Zika highlighting its importance in both cancer and virology. In preclinical models, small-molecule kinase inhibitors targeting AXL have shown promising anti-tumorigenic potential. This review primarily focuses on the induction, regulation and biological functions of AXL in mediating these tumor-promoting pathways. We discuss a range of therapeutic strategies, including recently developed small-molecule tyrosine kinase inhibitors (TKIs), monoclonal antibodies, and antibody-drug conjugates (ADCs), anti-AXL-CAR, and combination therapies. These interventions are being examined in both preclinical and clinical studies, offering the potential for improved drug sensitivity and therapeutic efficacy. We further discuss the mechanisms of acquired therapeutic resistance, particularly the crosstalk between AXL and other critical receptor tyrosine kinases (RTKs) such as c-MET, EGFR, HER2/HER3, VEGFR, PDGFR, and FLT3. Finally, we highlight key research areas that require further exploration to enhance AXL-mediated therapeutic approaches for improved clinical outcomes.
Collapse
Affiliation(s)
- Monika Yadav
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
- Laboratory of Nanotechnology and Chemical Biology, Regional Center for Biotechnology, Faridabad, Haryana, India
| | - Akansha Sharma
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ketki Patne
- Chromatin Remodeling Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Saba Tabasum
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Jyoti Suryavanshi
- Department of Cell Biology and Anatomy, New York Medical College, Valhalla, NY, USA
| | - Laxminarayan Rawat
- Harvard Medical School, Boston, MA, USA
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
| | - Marc Machaalani
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Marc Eid
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, Delhi, India
| | - Toni K Choueiri
- Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Soumitro Pal
- Harvard Medical School, Boston, MA, USA.
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA.
| | - Akash Sabarwal
- Harvard Medical School, Boston, MA, USA.
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
39
|
Liu J, Deng Y, Pu Z, Miao Y, Hao Z, Wang H, Zhang S, Liu H, Wang J, Lv Y, Hu B, Wan H, Zhuang Z, Sun T, Hao S, Ji N, Feng J. The AURKA inhibitor alters the immune microenvironment and enhances targeting B7-H3 immunotherapy in glioblastoma. JCI Insight 2025; 10:e173700. [PMID: 39928563 PMCID: PMC11949004 DOI: 10.1172/jci.insight.173700] [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: 07/05/2023] [Accepted: 01/22/2025] [Indexed: 02/12/2025] Open
Abstract
Glioblastoma (GBM) is one of the most lethal adult brain tumors with limited effective therapeutic options. Immunotherapy targeting B7-H3 (CD276) has shown promising efficacy in the treatment of gliomas. However, the response to this treatment varies among glioma patients due to individual differences. It's necessary to find an effective strategy to improve the efficacy of targeting B7-H3 immunotherapy for nonresponders. In this study, we demonstrated a strong correlation between aurora kinase A (AURKA) and CD276 expression in glioma tissue samples. Additionally, both AURKA knockdown and overexpression resulted in parallel changes in B7-H3 expression levels in glioma cells. Mechanistically, AURKA elevated B7-H3 expression by promoting epidermal growth factor receptor (EGFR) phosphorylation, which was validated in glioma cell lines and primary GBM cells. What's more, the combination of AURKA inhibitor (alisertib) and anti-B7-H3 antibody markedly reduced tumor size and promoted CD8+ T cell infiltration and activation in mouse orthotopic syngeneic glioma models. To our knowledge, this study is the first to demonstrate AURKA-mediated B7-H3 upregulation in glioma cells; moreover, it proposes a promising therapeutic strategy combining the AURKA inhibitor alisertib with B7-H3-specific blocking mAbs.
Collapse
Affiliation(s)
- Jinqiu Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yuxuan Deng
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhuonan Pu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Yazhou Miao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhaonian Hao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Herui Wang
- National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Shaodong Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Hanjie Liu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiejun Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yifan Lv
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Boyi Hu
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Hong Wan
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | | | - Tai Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuyu Hao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Nan Ji
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Jie Feng
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Xiao Y, He Z, Li W, Chen D, Niu X, Yang X, Zeng W, Wang M, Qian Y, Su Y, Luo F, Chen G, Liu J, Sui X, Zhou X, Gao Y. A covalent peptide-based lysosome-targeting protein degradation platform for cancer immunotherapy. Nat Commun 2025; 16:1388. [PMID: 39910101 PMCID: PMC11799215 DOI: 10.1038/s41467-025-56648-6] [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: 07/05/2024] [Accepted: 01/27/2025] [Indexed: 02/07/2025] Open
Abstract
The lysosome-targeting chimera (LYTAC) strategy provided a very powerful tool for the degradation of membrane proteins. However, the synthesis of LYTACs, antibody-small molecule conjugates, is challenging. The ability of antibody-based LYTACs to penetrate solid tumor is limited as well, especially to cross the blood-brain barrier (BBB). Here, we propose a covalent chimeric peptide-based targeted degradation platform (Pep-TACs) by introducing a long flexible aryl sulfonyl fluoride group, which allows proximity-enabled cross-linking upon binding with the protein of interest. The Pep-TACs platform facilitates the degradation of target proteins through the mechanism of recycling transferrin receptor (TFRC)-mediated lysosomal targeted endocytosis. Biological experiments demonstrate that covalent Pep-TACs can significantly degrade the expression of PD-L1 on tumor cells, dendritic cells and macrophages, especially under acidic conditions, and markedly enhance the function of T cells and tumor phagocytosis by macrophages. Furthermore, both in anti-PD-1-responsive and -resistant tumor models, the Pep-TACs exert significant anti-tumor immune response. It is noteworthy that Pep-TACs can cross the BBB and prolong the survival of mice with in situ brain tumor. As a proof-of-concept, this study introduces a modular TFRC-based covalent peptide degradation platform for the degradation of membrane protein, and especially for the immunotherapy of brain tumors.
Collapse
Affiliation(s)
- Youmei Xiao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Zhuoying He
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Wanqiong Li
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Danhong Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Xiaoshuang Niu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Xin Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Wenxuan Zeng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Mengfan Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Yuzhen Qian
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, Henan Province, China
| | - Ye Su
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Feiyu Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Guanyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Juan Liu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Xinghua Sui
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China
| | - Xiuman Zhou
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China.
| | - Yanfeng Gao
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, China.
| |
Collapse
|
41
|
Zhao J, He C, Xie H, Zou Y, Yan Z, Deng J, Du Y, Yang W, Zhang X. Latent Association Between Diets and Glioma Risk: A Mendelian Randomization Analysis. Nutrients 2025; 17:582. [PMID: 39940440 PMCID: PMC11819737 DOI: 10.3390/nu17030582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Gliomas, particularly high-grade gliomas such as glioblastoma, represent a major challenge due to their poor prognosis. While dietary factors have been proposed as potential modulators of glioma risk, causal inference has been hindered by confounding and reverse causality in observational studies. This study employs Mendelian randomization to investigate the causal relationship between dietary factors and glioma risk. METHODS A two-sample MR framework was applied, utilizing genome-wide association study data for 22 dietary exposures and glioma risks, including both GBM and non-GBM subtypes. Instrumental variables (genetic variants) were identified for each dietary factor to address confounding and pleiotropy. Causal inference was conducted using inverse-variance weighted regression, complemented by MR-Egger and MR-PRESSO analyses to assess and correct for potential pleiotropy. RESULTS A positive causal association was observed between the intake of cooked vegetables and the GBM risk (OR = 6.55, 95% CI: 1.86-23.12, p = 0.00350). While alcohol intake demonstrated a protective effect for non-GBM risk (OR = 0.770, 95% CI: 0.61-0.97, p = 0.029), beer was substantially linked to an increased risk of non-GBM gliomas (OR = 4.82, 95% CI: 1.84-12.59, p = 0.0014). Other dietary factors did not exhibit significant causal associations. CONCLUSIONS These findings suggest that certain dietary factors, including cooked vegetable intake, beer consumption, and alcohol intake, may exert a causal influence on glioma risk. This study provides new insights into the potential dietary determinants of glioma and underscores the need for further investigation into modifiable risk factors for glioma prevention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiangheng Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou 510060, China; (J.Z.); (C.H.); (H.X.); (Y.Z.); (Z.Y.); (J.D.); (Y.D.); (W.Y.)
| |
Collapse
|
42
|
Han JM, Oh KY, Choi SJ, Lee WW, Jin BH, Kim JH, Yu HJ, Kim RJY, Yoon HJ, Lee JI, Hong SD, Cho SD. Antitumor activity of afatinib in EGFR T790M-negative human oral cancer therapeutically targets mTOR/Mcl-1 signaling axis. Cell Oncol (Dordr) 2025; 48:123-138. [PMID: 38888847 PMCID: PMC11850456 DOI: 10.1007/s13402-024-00962-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2024] [Indexed: 06/20/2024] Open
Abstract
PURPOSE This study investigates the role and effectiveness of the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) in oral cancer, focusing on the clinical relevance of EGFR and myeloid cell leukemia-1 (Mcl-1) in head and neck cancers (HNCs). It aims to explore the molecular mechanism of afatinib, a TKI, in treating human oral cancer. METHODS We conducted an in silico analysis using databases like The Cancer Genome Atlas, Gene Expression Omnibus, and Clinical Proteomic Tumor Analysis Consortium, along with immunohistochemistry staining, to study EGFR and Mcl-1 expression in HNCs. For investigating afatinib's anticancer properties, we performed various in vitro and in vivo analyses, including trypan blue exclusion assay, Western blotting, 4'-6-diamidino-2-phenylindole staining, flow cytometry, quantitative real-time PCR, Mitochondrial membrane potential assay, overexpression vector construction, transient transfection, and a tumor xenograft model. RESULTS Higher expression levels of EGFR and Mcl-1 were observed in HNC patient tissues compared to normal tissues, with their co-expression significantly linked to poor prognosis. There was a strong correlation between EGFR and Mcl-1 expressions in oral cancer patients. Afatinib treatment induced apoptosis and suppressed Mcl-1 in oral cancer cell lines without the EGFR T790M mutation. The mechanism of afatinib-induced apoptosis involved the EGFR/mTOR/Mcl-1 axis, as shown by the effects of mTOR activator MHY1485 and inhibitor rapamycin. Afatinib also increased Bim expression, mitochondrial membrane permeabilization, and cytochrome c release. It significantly lowered tumor volume without affecting body, liver, and kidney weights. CONCLUSION Afatinib, targeting the EGFR/mTOR/Mcl-1 axis, shows promise as a therapeutic strategy for oral cancer, especially in patients with high EGFR and Mcl-1 expressions.
Collapse
Affiliation(s)
- Jung-Min Han
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Kyu-Young Oh
- Department of Oral Pathology, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Su-Jung Choi
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Won-Woo Lee
- Laboratory Animal Center, CHA University, CHA Biocomplex, Sampyeong-Dong, Seongnam, 13488, Republic of Korea
| | - Bo-Hwan Jin
- Laboratory Animal Center, CHA University, CHA Biocomplex, Sampyeong-Dong, Seongnam, 13488, Republic of Korea
| | - Ji-Hoon Kim
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hyun-Ju Yu
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Ryan Jin Young Kim
- Department of Dental Science, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Hye-Jung Yoon
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Jae-Il Lee
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea
| | - Seong-Doo Hong
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
| | - Sung-Dae Cho
- Department of Oral Pathology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, 03080, Republic of Korea.
| |
Collapse
|
43
|
Wang Y, Han J, Zhu Y, Huang N, Qu N. New advances in the therapeutic strategy of head and neck squamous cell carcinoma: A review of latest therapies and cutting-edge research. Biochim Biophys Acta Rev Cancer 2025; 1880:189230. [PMID: 39608621 DOI: 10.1016/j.bbcan.2024.189230] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a common and aggressive malignancy with a poor prognosis, particularly when diagnosed at advanced stages. Despite progress in surgical, chemotherapeutic, and radiotherapeutic interventions, the five-year survival rate remains low due to high rates of recurrence and therapeutic resistance. This review explores recent advances in therapeutic strategies for HNSCC, focusing on targeted therapies, immunotherapy, and innovative drug delivery systems. Targeted therapies, such as EGFR inhibitors and PI3K/AKT/mTOR pathway inhibitors, offer promising options for overcoming HNSCC, though resistance challenges persist. Emerging treatments, including dual-target inhibitors and personalized therapeutic approaches, show potential in addressing these limitations. Immunotherapy, particularly PD-1/PD-L1 blockade, has achieved positive outcomes in a subset of patients, though overall response rates remain modest. Strategies aimed at enhancing immune responses, such as combination therapies and nanotechnology-based drug delivery systems, are actively being investigated to improve efficacy. This review also underscores the critical role of the tumor microenvironment and epithelial-mesenchymal transition (EMT) in HNSCC progression and therapeutic resistance. Novel approaches, including smart drug delivery systems utilizing nanotechnology and immune modulation, are opening new avenues for more personalized and effective treatments. Ongoing interdisciplinary research into molecular targets and advanced drug delivery techniques holds great promise for significantly improving patient outcomes in HNSCC.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing Han
- Department of Oral and Maxillofacial Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Rd., Huangpu District, Shanghai 200011, China
| | - Yongxue Zhu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Naisi Huang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ning Qu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
44
|
Long D, Ding Y, Wang P, Wei L, Ma K. Multi-Omics Analysis Reveals Immune Infiltration and Clinical Significance of Phosphorylation Modification Enzymes in Lung Adenocarcinoma. Int J Mol Sci 2025; 26:1066. [PMID: 39940833 PMCID: PMC11817228 DOI: 10.3390/ijms26031066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Protein phosphorylation is a dynamic and reversible modification involved in almost all cellular processes. Numerous investigations have shown that protein phosphorylation modification enzymes (PPMEs) that regulate protein phosphorylation play an important role in the occurrence and treatment of tumors. However, there is still a lack of effective insights into the value of PPMEs in the classification and treatment of patients with lung adenocarcinoma (LUAD). Here, four topological algorithms identified 15 hub PPMEs from a protein-protein interaction (PPI) network. This PPI network was constructed using 124 PPMEs significantly correlated with 35 cancer hallmark-related pathways. Our study illustrates that these hub PPMEs can affect the survival of patients with LUAD in the form of somatic mutation or expression perturbation. Consistency clustering based on hub PPMEs recognized two phosphorylation modification subtypes (namely cluster1 and cluster2) from LUAD. Compared with patients in cluster1, the survival prognosis of patients in cluster2 is worse. This disparity is probably attributed to the higher tumor mutation burden, the higher male proportion, and the more significant expression disturbance in patients in cluster2. Moreover, phosphorylation modification subtypes also have different characteristics in terms of immune activity, immune infiltration level, immunotherapy response, and drug sensitivity. We constructed a PSig scoring system by using a principal component analysis algorithm to estimate the level of phosphorylation modification in individual LUAD patients. Patients in the high and low PSig score groups demonstrated different characteristics in terms of survival rate, tumor mutation burden, somatic gene mutation rate, immune cell abundance, and sensitivity to immunotherapy and drug treatment. This work reveals that phosphorylation plays a non-negligible role in the tumor microenvironment and immunotherapy of LUAD. Evaluating the phosphorylation status of individual LUAD patients by the PSig score can contribute to enhancing our cognition of the tumor microenvironment and guiding the formulation of more effective personalized treatment strategies.
Collapse
Affiliation(s)
- Deyu Long
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University Medical College, Shihezi 832000, China
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Yanheng Ding
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Peng Wang
- Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Lili Wei
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University Medical College, Shihezi 832000, China
| | - Ketao Ma
- The Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University Medical College, Shihezi 832000, China
| |
Collapse
|
45
|
Fu MJ, Jin H, Wang SP, Shen L, Liu HM, Liu Y, Zheng YC, Dai XJ. Unleashing the Power of Covalent Drugs for Protein Degradation. Med Res Rev 2025. [PMID: 39834319 DOI: 10.1002/med.22101] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Targeted protein degradation (TPD) has emerged as a significant therapeutic approach for a variety of diseases, including cancer. Advances in TPD techniques, such as molecular glue (MG) and lysosome-dependent strategies, have shown substantial progress since the inception of the first PROTAC in 2001. The PROTAC methodology represents the forefront of TPD technology, with ongoing evaluation in more than 20 clinical trials for the treatment of diverse medical conditions. Two prominent PROTACs, ARV-471 and ARV-110, are currently undergoing phase III and II clinical trials, respectively. Traditional PROTACs are encountering obstacles such as limited binding affinity and a restricted range of E3 ligase ligands for facilitating the protein of interest (POI) degradation. Covalent medicines offer the potential to enhance PROTAC efficacy by enabling the targeting of previously considered "undruggable" shallow binding sites. Strategic alterations allow PROTAC to establish covalent connections with particular target proteins, including Kirsten rat sarcoma viral oncogene homolog (KRAS), Bruton's tyrosine kinase (BTK), epidermal growth factor receptor (EGFR), as well as E3 ligases such as DDB1 and CUL4 associated factor 16 (DCAF16) and Kelch-like ECH-associated protein 1 (Keap1). The concept of covalent degradation has also been utilized in various new forms of degraders, including covalent molecule glue (MG), in-cell click-formed proteolysis targeting chimera (CLIPTAC), HaloPROTAC, lysosome-targeting chimera (LYTAC) and GlueTAC. This review focuses on recent advancements in covalent degraders beyond covalent PROTACs and examines obstacles and future directions pertinent to this field.
Collapse
Affiliation(s)
- Meng-Jie Fu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hang Jin
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shao-Peng Wang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Liang Shen
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Hong-Min Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Liu
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi-Chao Zheng
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xing-Jie Dai
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Henan Province for Drug Quality and Evaluation; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Cardio-Cerebrovascular Drug, China Meheco Topfond Pharmaceutical Company, Zhumadian, Henan, China
| |
Collapse
|
46
|
Cao W, Jin D, Min W, Li H, Wang R, Zhang J, Gou Y. Prognostic values of intracellular cell-related genes in esophageal cancer and their regulatory mechanisms. BMC Cancer 2025; 25:105. [PMID: 39833728 PMCID: PMC11744837 DOI: 10.1186/s12885-025-13483-8] [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: 05/22/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Esophageal cancer is a grave malignant condition. While radiotherapy, often in conjunction with chemotherapy, serves as a cornerstone in the management of locally advanced or metastatic cases, patient tolerance and treatment resistance frequently hinder its efficacy. Cell-in-cell structures, prevalent in various tumors, have been linked to prognosis. Hence, investigating the prognostic significance and regulatory mechanisms of genes related to these intracellular structures in esophageal cancer is imperative. The Cancer Genome Atlas (TCGA) Esophageal Cancer (ESCA) dataset served as the training set for the analysis. Differentially expressed genes (DEGs) in ESCA samples were identified, with those related to intercellular structures designated cell-in-cell-related differential expression genes (CIC-related DEGs). Cox regression analysis was employed to identify prognostic genes, categorizing samples into high- and low-risk groups based on median risk scores. Validation was conducted using the GSE53624 risk model. Established methodologies included morphological mapping, enrichment analysis, immune infiltration analysis, prognostic gene expression validation, molecular docking, and Reverse Transcription Polymerase Chain Reaction (RT-PCR) validation. Thirty-eight intersecting genes were identified between the disease and normal groups in ESCA samples. Stepwise multivariate Cox analysis pinpointed three prognostic genes: androgen receptor (AR), C-X-C motif chemokine ligand 8 (CXCL8), and epidermal growth factor receptor (EGFR). The risk model's applicability was confirmed in the GSE53624 dataset, revealing eight significantly different immune-related gene sets. Prognostic gene expression validation demonstrated significant differences between the disease and normal groups in both datasets. The proteins corresponding to the three prognostic genes interacted with gefitinib and osimertinib. RT-PCR results corroborated the differential expression of prognostic genes in esophageal cancer tissues. This study identified AR, CXCL8, and EGFR as prognostic genes and demonstrated their molecular interactions with gefitinib and osimertinib, providing a foundation for ESCA diagnosis and treatment.
Collapse
Affiliation(s)
- Wei Cao
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Dacheng Jin
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Weirun Min
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Haochi Li
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Rong Wang
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
| | - Jinlong Zhang
- First Clinical Medical School, Gansu University of Chinese Medicine, Lanzhou, China
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China
| | - Yunjiu Gou
- Chest Clinic Center, Gansu Provincial People's Hospital, Lanzhou, China.
- First Department of Thoracic Surgery, Gansu Provincial People's Hospital, Lanzhou, China.
| |
Collapse
|
47
|
Wang H, Fan S, Zhan Y, Xu Y, Du Y, Luo J, Zang H, Peng S, Wang W. Targeting EGFR-binding protein SLC7A11 enhancing antitumor immunity of T cells via inducing MHC-I antigen presentation in nasopharyngeal carcinoma. Cell Death Dis 2025; 16:21. [PMID: 39820491 PMCID: PMC11739652 DOI: 10.1038/s41419-024-07327-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 12/08/2024] [Accepted: 12/27/2024] [Indexed: 01/19/2025]
Abstract
Approximately 80% of nasopharyngeal carcinoma (NPC) patients exhibit EGFR overexpression. The overexpression of EGFR has been linked to its potential role in modulating major histocompatibility complex class I (MHC-I) molecules. We discovered that EGFR, operating in a kinase-independent manner, played a role in stabilizing the expression of SLC7A11, which subsequently inhibited MHC-I antigen presentation. This mechanism, in turn, provided protection to NPC cells against T cell-mediated cytotoxicity. The underlying molecular processes revealed that the high and stable expression of SLC7A11 hindered the nuclear entry of GR, thereby suppressing TAP1 transcription and the presentation of MHC-I molecules. Additionally, elevated SLC7A11 expression led to an increase in FAF2 expression and triggered ERAD-dependent degradation of MHC-I, resulting in a reduction of MHC-I molecules on the cell membrane. The NPC patients exhibiting high EGFR and low MHC-I expression, combined with a scarcity of CD8+ T cells (EGFRhighMHC-IlowCD8few phenotype), experienced considerably shorter overall survival times compared to other situations. What is more, our study demonstrated that sorafenib had the capability to enhance the MHC-I antigen presentation process, thereby facilitating T cell-mediated killing of NPC cells via targeting SLC7A11. Consequently, targeting SLC7A11 with sorafenib emerges as a promising therapeutic strategy for the treatment of NPC.
Collapse
Affiliation(s)
- Haihua Wang
- Department of Pathology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Department of Gastroenterology, The Second Affiliated Hospital of Hainan Medical University, 570216, Haikou, Hainan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, 410011, Changsha, Hunan, China
| | - Yuting Zhan
- Department of Pathology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, 410011, Changsha, Hunan, China
| | - Yue Xu
- Department of Pathology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, 410011, Changsha, Hunan, China
| | - Yao Du
- Department of Pathology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, 410011, Changsha, Hunan, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, 410011, Changsha, Hunan, China
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital, Central South University, 410011, Changsha, Hunan, China
- Hunan Clinical Medical Research Center for Cancer Pathogenic Genes Testing and Diagnosis, 410011, Changsha, Hunan, China
| | - Shuping Peng
- Cancer Research Institute, School of Basic Medical Science, Central South University, 410078, Changsha, Hunan, China
| | - Weiyuan Wang
- Department of Pathology, The Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
| |
Collapse
|
48
|
Allen D, Szoo MJ, van Bergen TD, Seppelin A, Oh J, Saad MA. Near-infrared photoimmunotherapy: mechanisms, applications, and future perspectives in cancer research. Antib Ther 2025; 8:68-85. [PMID: 39958565 PMCID: PMC11826922 DOI: 10.1093/abt/tbaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/01/2024] [Accepted: 01/16/2025] [Indexed: 02/18/2025] Open
Abstract
Photoimmunotherapy (PIT) involves the targeted delivery of a photosensitizer through antibody conjugation, which, upon binding to its cellular target and activation by external irradiation, induces localized toxicity. This approach addresses several limitations of conventional cancer therapies, such as chemo- and radiotherapies, which result in off-target effects that significantly reduce patient quality of life. Furthermore, PIT improves on the challenges encountered with photodynamic therapy (PDT), such as nonspecific localization of the photosensitizer, which often results in unintended toxicities. Although PIT was first proposed in the early 1980s, its clinical applications have been constrained by limitations in antibody engineering, conjugation chemistries, and optical technologies. However, recent advances in antibody-drug conjugate (ADC) research and the emergence of sophisticated laser technologies have greatly benefited the broader applicability of PIT. Notably, the first near-infrared photoimmunotherapy (NIR-PIT) treatment for head and neck cancer has been approved in Japan and is currently in phase III clinical trials in the USA. A significant advantage of PIT over traditional ADCs in cancer management is the agnostic nature of PDT, making it more adaptable to different tumor types. Specifically, PIT can act on cancer stem cells and cancer cells displaying treatment resistance and aggressive phenotypes-a capability beyond the scope of ADCs alone. This review provides an overview of the mechanism of action of NIR-PIT, highlighting its adaptability and application in cancer therapeutics, and concludes by exploring the potential of PIT in advancing cancer treatments.
Collapse
Affiliation(s)
- Derek Allen
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Madeline JoAnna Szoo
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, United States
- Department of Biochemistry, Northeastern University, Boston, MA 02115, United States
| | - Tessa D van Bergen
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| | - Ani Seppelin
- Department of Biochemistry, Northeastern University, Boston, MA 02115, United States
| | - Jeonghyun Oh
- Department of Physics, Northeastern University, Boston, MA 02115, United States
| | - Mohammad A Saad
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, United States
| |
Collapse
|
49
|
Liu X, Li T, Wang Y, Gao X, Wang F, Chen Y, Wang K, Luo W, Kong F, Kou Y, You H, Kong D, Zhang Q, Tang R. Delta-Like Homolog 2 Facilitates Malignancy of Hepatocellular Carcinoma via Activating EGFR/PKM2 Signaling Pathway. Mol Carcinog 2025; 64:176-191. [PMID: 39467107 DOI: 10.1002/mc.23836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/24/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024]
Abstract
Delta-like homolog 2 (DLK2) plays a crucial role in adipogenesis, chondrogenic differentiation, and the progression of certain cancers. However, the key roles of DLK2 underlying the progression of hepatocellular carcinoma (HCC) remain ambiguous. In the current study, we demonstrate that DLK2 is upregulated in HCC, significantly correlated with clinicopathological variables and serves as an independent diagnostic marker. Functional assays reveal that DLK2 facilitates malignant progression of HCC in vitro and in vivo models. Mechanistically, DLK2 binds to EGFR resulting in its auto-phosphorylation, which activates NF-κB pathway leading to P65-dependent transcriptional upregulation of PKM2. Furthermore, that elevates both enzyme-dependent and -independent activities of PKM2 contributing to cancer proliferation and metastasis. In summary, our findings demonstrate a novel pro-tumoral role and mechanism of DLK2 in the regulation of HCC malignant progression, suggesting its potential as a clinical diagnostic marker and therapeutic target.
Collapse
Affiliation(s)
- Xiangye Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Tingting Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Yuting Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Feitong Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Yang Chen
- School of Life Science, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Kaisheng Wang
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Weiming Luo
- Department of General Surgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Fanyun Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Yanbo Kou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Hongjuan You
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Delong Kong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| | - Renxian Tang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, People's Republic of China
| |
Collapse
|
50
|
Chen X, Nie X, Lin X, Wang Y, Zhang L, Chen Z, Lin M. Regulation of keratinocyte barrier function and inflammatory response by the EGFR-STAT3 Pathway: Potential therapeutic implications of osimertinib and afatinib. Cytokine 2025; 185:156802. [PMID: 39612655 DOI: 10.1016/j.cyto.2024.156802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/29/2024] [Accepted: 11/02/2024] [Indexed: 12/01/2024]
Abstract
The epidermal growth factor receptor (EGFR) signaling pathway is crucial for skin barrier integrity and immune response. This study explores the impact of EGFR inhibitors, osimertinib and afatinib, on keratinocyte function, focusing on keratin (KRT1, KRT17) and tight junction protein (CLDN1, CLDN2, CLDN4) expression in HaCaT cells. Osimertinib significantly increased the mRNA and protein levels of keratins and inflammatory markers, IL-6 and TNF-α, via activation of the EGFR-STAT3 signaling pathway. Co-treatment with recombinant human EGF reversed these changes, suggesting the pathway's modulatory role. These findings underscore the potential therapeutic applications of targeting the EGFR-STAT3 axis in skin barrier dysfunction and inflammatory skin disorders.
Collapse
Affiliation(s)
- Xin Chen
- Ningde Clinical Medical College of Fujian Medical University, Ningde, Fujian 352100, China
| | - Xuekun Nie
- Ningde Clinical Medical College of Fujian Medical University, Ningde, Fujian 352100, China; Clinical Pharmacy, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian 352100, China; Collaborative Innovation Center of Active Ingredient Research of She Medicine(2020Z03), Ningde Normal University, Ningde, Fujian 352100, China; Innovation Center of Clinical Pharmacy Service(2023T06), Ningde Normal University, Ningde, Fujian 352100, China
| | - Xiaohui Lin
- Ningde Clinical Medical College of Fujian Medical University, Ningde, Fujian 352100, China; Clinical Pharmacy, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian 352100, China; Collaborative Innovation Center of Active Ingredient Research of She Medicine(2020Z03), Ningde Normal University, Ningde, Fujian 352100, China; Innovation Center of Clinical Pharmacy Service(2023T06), Ningde Normal University, Ningde, Fujian 352100, China
| | - Yujia Wang
- Ningde Clinical Medical College of Fujian Medical University, Ningde, Fujian 352100, China; Clinical Pharmacy, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian 352100, China; Collaborative Innovation Center of Active Ingredient Research of She Medicine(2020Z03), Ningde Normal University, Ningde, Fujian 352100, China; Innovation Center of Clinical Pharmacy Service(2023T06), Ningde Normal University, Ningde, Fujian 352100, China
| | - Lingling Zhang
- Ningde Clinical Medical College of Fujian Medical University, Ningde, Fujian 352100, China; Clinical Pharmacy, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian 352100, China; Collaborative Innovation Center of Active Ingredient Research of She Medicine(2020Z03), Ningde Normal University, Ningde, Fujian 352100, China; Innovation Center of Clinical Pharmacy Service(2023T06), Ningde Normal University, Ningde, Fujian 352100, China
| | - Zichun Chen
- Ningde Clinical Medical College of Fujian Medical University, Ningde, Fujian 352100, China; Clinical Pharmacy, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian 352100, China; Collaborative Innovation Center of Active Ingredient Research of She Medicine(2020Z03), Ningde Normal University, Ningde, Fujian 352100, China; Innovation Center of Clinical Pharmacy Service(2023T06), Ningde Normal University, Ningde, Fujian 352100, China.
| | - Minhua Lin
- Ningde Clinical Medical College of Fujian Medical University, Ningde, Fujian 352100, China; Clinical Pharmacy, Ningde Municipal Hospital of Ningde Normal University, Ningde, Fujian 352100, China; Collaborative Innovation Center of Active Ingredient Research of She Medicine(2020Z03), Ningde Normal University, Ningde, Fujian 352100, China; Innovation Center of Clinical Pharmacy Service(2023T06), Ningde Normal University, Ningde, Fujian 352100, China.
| |
Collapse
|