1
|
Wu S, Hu C, Hu P, He W, Hui K, Jiang X. RAF1 promotes anlotinib resistance in non-small cell lung cancer by inhibiting apoptosis. J Cancer Res Clin Oncol 2025; 151:138. [PMID: 40227502 PMCID: PMC11996991 DOI: 10.1007/s00432-025-06175-0] [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: 02/27/2025] [Accepted: 03/16/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Anlotinib is an effective treatment for advanced non-small cell lung cancer (NSCLC), but resistance to it often develops during therapy. RAF1, a serine/threonine kinase involved in cancer progression, has limited research in NSCLC, particularly regarding anlotinib resistance. METHODS Analysis of RAF1 expression in NSCLC and its relationship with targeted therapy resistance and apoptosis through bioinformatics methods. Immunohistochemistry (IHC) was employed to evaluate the relationship between RAF1 expression and anlotinib resistance in NSCLC tissues. Anlotinib-resistant PC9 (PC9/AR) cells were constructed in vitro, and cell viability and apoptosis were assessed using the cell counting kit-8 (CCK-8) assay and flow cytometry. Quantitative real-time PCR (qRT-PCR) was carried out to evaluate RAF1 gene expression levels, and western blot (WB) analysis was conducted to determine the expression of RAF1, Bcl-2-associated X protein (Bax) and B-cell lymphoma 2 (Bcl-2). RESULTS Bioinformatics analysis showed that RAF1 was lowly expressed in lung cancer tissues in TCGA and GEPIA databases. Further pathway analysis indicated that RAF1 expression was positively correlated with targeted therapy resistance and negatively correlated with the expression of the anti-apoptotic protein Bcl-2. Immunohistochemical analysis showed that high RAF1 expression in NSCLC tissues was related to anlotinib resistance (P < 0.05). In vitro experiments demonstrated that RAF1 contributed to anlotinib resistance in NSCLC cells. Overexpression of RAF1 increased cell viability and decreased apoptosis in PC9 and PC9/AR cells, while knockdown of RAF1 had the opposite effects. CONCLUSION RAF1 mediates anlotinib resistance in NSCLC cells by regulating apoptosis and may serve as a predictive marker for anlotinib resistance in advanced lung cancer patients.
Collapse
Affiliation(s)
- Shuo Wu
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, 222000, China
| | - Chenxi Hu
- The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222000, China
| | - Pengwu Hu
- The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222000, China
| | - Wei He
- The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222000, China
| | - Kaiyuan Hui
- The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222000, China.
| | - Xiaodong Jiang
- Lianyungang Clinical College of Nanjing Medical University, Lianyungang, 222000, China.
- The Affiliated Lianyungang Hospital of Xuzhou Medical University, Lianyungang, 222000, China.
| |
Collapse
|
2
|
Long C, Shen H, Li H, Han L. Capivasertib augments chemotherapy via Akt inhibition in preclinical small cell lung cancer models. Fundam Clin Pharmacol 2025; 39:e13042. [PMID: 39501557 DOI: 10.1111/fcp.13042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/14/2024] [Accepted: 10/23/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Small cell lung cancer (SCLC) is a highly aggressive type of lung cancer for which platinum-based chemotherapy is the standard of care. Despite an initial response to this therapy, patients eventually develop resistance to the chemotherapy. OBJECTIVES To investigate the potential of capivasertib, an approved drug for advanced breast cancer, to enhance the efficacy of cisplatin in preclinical SCLC models and explore the underlying mechanisms. METHODS SCLC cell lines were treated with capivasertib and cisplatin, alone or in combination, to assess cell viability, proliferation, colony formation, and apoptosis. Next, capivasertib's effects, alone and combined with cisplatin, were evaluated in an SCLC mouse model. Mechanistic studies focused on Akt and MYC signaling, with constitutively active Akt overexpression used to assess its role. RESULTS Capivasertib is active against a panel of SCLC cell lines regardless of cellular origin and genetic profiling with IC50 at a clinically achievable range. Particularly, capivasertib inhibits proliferation and anchorage-independent colony formation and induces apoptosis in SCLC cells. It significantly augments cisplatin's inhibitory effects in all tested cell lines. Importantly, capivasertib at a non-toxic dose is effective in delaying SCLC growth in mice and its combination with cisplatin achieves nearly complete tumor growth inhibition. Mechanistic studies confirm that capivasertib inhibits Akt and MYC signaling, and furthermore, that overexpression of constitutively active Akt reversed anti-SCLC activity of capivasertib. CONCLUSION Our work is the first to reveal that Akt inhibition can augment chemotherapy in SCLC, and capivasertib is a useful addition to the treatment armamentarium for SCLC.
Collapse
Affiliation(s)
- Cheng Long
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Hui Shen
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Hui Li
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Lan Han
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| |
Collapse
|
3
|
Zheng H, Fan S, Luo J, Wen Q, Zang H. MicroRNA-150-3p enhances the antitumour effects of CGP57380 and is associated with a favourable prognosis in non-small cell lung cancer. Sci Rep 2025; 15:1973. [PMID: 39809860 PMCID: PMC11733271 DOI: 10.1038/s41598-025-85793-7] [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: 06/22/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
MicroRNA (miRNA) dysregulation has been identified in several carcinomas, including non-small cell lung cancer (NSCLC), and is known to play a role in the development and progression of this disease. We initially conducted a miRNA microarray analysis, which revealed that the MNK inhibitor CGP57380 increased the expression of miR-150-3p. A similar analysis was performed using data from The Cancer Genome Atlas (TCGA). Cell proliferation, colony formation and migration assays were validated in A549 and H157 cells treated with miR-150-3p mimics. Quantitative polymerase chain reaction (qPCR) was then used to detect potential target genes. We observed significant downregulation of miR-150-3p in NSCLC samples compared with normal samples (P = 0.035). High miR-150-3p expression was associated with longer overall survival (P = 0.005), as determined via a tissue microarray (TMA). These results were validated in the TCGA and revealed that miR-150-3p was expressed at low levels in NSCLC tissues (P < 0.0001) and that patients with high miR-150-3p expression had a better prognosis (P = 0.042). Moreover, the combination of miR-150-3p and CGP57380 exerted a synergistic inhibitory effect on colony formation, growth, and migration and induced apoptosis in NSCLC cell lines. We investigated the potential targets of miR-150-3p and successfully validated six potential target genes through qPCR analysis. High miR-150-3p expression may enhance the response to immunotherapy, cisplatin and gemcitabine. In summary, this study underscores the promising therapeutic implications of combining miR-150-3p and CGP57380 for NSCLC treatment. Additionally, this study provides valuable insights into the molecular mechanisms underlying the effects of this treatment.
Collapse
Affiliation(s)
- Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiuyuan Wen
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
4
|
Liu L, Zhao T, Zheng S, Tang D, Han H, Yang C, Zheng X, Wang J, Ma J, Wei W, Wang Z, He S, He Q. METTL3 inhibitor STM2457 impairs tumor progression and enhances sensitivity to anlotinib in OSCC. Oral Dis 2024; 30:4243-4254. [PMID: 38376115 DOI: 10.1111/odi.14864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/03/2023] [Accepted: 12/29/2023] [Indexed: 02/21/2024]
Abstract
OBJECTIVES To investigate the inhibitory effects of STM2457, which is a novel METTL3 (m6A writer) inhibitor, both as a monotherapy and in combination with anlotinib, in the treatment of oral squamous cell carcinoma (OSCC) both in vitro and in vivo. MATERIALS AND METHODS The efficacy of STM2457 or STM2457 plus anlotinib was evaluated using two OSCC cell lines by CCK8, transwell, colony formation, would-healing, sphere formation, cell cycle, apoptosis assays, and nude mice tumor xenograft techniques. The molecular mechanism study was carried out by western blotting, qRT-PCR, MeRIP-qPCR, immunofluorescence, and immunohistochemistry. RESULTS STM2457 combined with anlotinib enhanced inhibition of cellular survival/proliferation and promotion of apoptosis in vitro. Moreover, this combinatorial approach exerted a notable reduction in stemness properties and EMT (epithelial-mesenchymal transition) features of OSCC cells. Remarkably, in vivo studies validated the efficacy of the combination treatment. Mechanistically, our investigations revealed that the combined action of STM2457 and anlotinib exerted downregulatory effects on EGFR (epidermal growth factor receptor) expression in OSCC cells. CONCLUSIONS The combination of STM2457 and anlotinib targeting EGFR exerted a multiple anti-tumor effect. In near future, anlotinib combined with STM2457 may provide a novel insight for the treatment of OSCC.
Collapse
Affiliation(s)
- Lianlian Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Tingting Zhao
- College & Hospital of Stomatology, Guangxi Medical University, Nanning, China
| | - Siyi Zheng
- Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Dongxiao Tang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Hui Han
- Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Chunlong Yang
- Clinical Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xin Zheng
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Juan Wang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jieyi Ma
- Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Laboratory of General Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wei Wei
- Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhaoyu Wang
- Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuqi He
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Qianting He
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
5
|
Li H, Yao Y, Hao R, Long C. Selective and effective suppression of pancreatic cancer through MNK inhibition. Immunopharmacol Immunotoxicol 2024:1-11. [PMID: 39138614 DOI: 10.1080/08923973.2024.2391462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 08/04/2024] [Indexed: 08/15/2024]
Abstract
Objective: The study aimed to explore the role of the Wnt/β-catenin signaling pathway in pancreatic cancer progression and chemoresistance, with a focus on identifying specific factors that distinguish between normal and tumor cells, thereby offering potential therapeutic targets. Materials and Methods: We analyzed levels of total and phosphorylated eukaryotic translation initiation factor 4E (eIF4E) and β-catenin in pancreatic cancer and normal pancreatic tissues. Functional assays were used to assess the impact of eIF4E phosphorylation on β-catenin signaling, cell proliferation, and chemoresistance, with MNK kinase involvement determined through gene depletion studies. The MNK kinase inhibitor eFT508 was evaluated for its effects on eIF4E phosphorylation, β-catenin activation, and cell viability in both in vitro and in vivo models of pancreatic cancer. Results: Both total and phosphorylated eIF4E, along with β-catenin, were significantly elevated in pancreatic cancer tissues compared to normal tissues. Phosphorylation of eIF4E at serine 209 was shown to activate β-catenin signaling, enhance cell proliferation, and contribute to chemoresistance in pancreatic cancer. Importantly, these effects were dependent on MNK kinase activity. Depletion of eIF4E reduced cell viability in both pancreatic cancer and normal cells, while depletion of MNK selectively decreased viability in pancreatic cancer cells. Treatment with eFT508 effectively inhibited eIF4E phosphorylation, suppressed β-catenin activation, and reduced pancreatic cancer cell growth and survival in vitro and in vivo, with minimal impact on normal cells. Conclusions: The MNK-eIF4E-β-catenin axis plays a critical role in pancreatic cancer progression and chemoresistance, distinguishing pancreatic cancer cells from normal cells. Targeting MNK kinases with inhibitors like eFT508 presents a promising therapeutic strategy for pancreatic cancer, with potential for selective efficacy and reduced toxicity.
Collapse
Affiliation(s)
- Hui Li
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Yang Yao
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Rui Hao
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, People's Republic of China
| | - Cheng Long
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, People's Republic of China
| |
Collapse
|
6
|
Wang L, Xi C, Liu R, Ye T, Xiang N, Deng J, Li H. Dual targeting of Mcl-1 and Bcl-2 to overcome chemoresistance in cervical and colon cancer. Anticancer Drugs 2024; 35:219-226. [PMID: 37948336 DOI: 10.1097/cad.0000000000001553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
After an initial positive response to chemotherapy, cancer patients often become resistant and experience relapse. Our previous research identified eukaryotic translation initiation factor 4E (eIF4E) as a crucial target to overcome chemoresistance. In this study, we delved further into the role and therapeutic potential of myeloid cell leukemia 1 (Mcl-1), an eIF4E-mediated target, in chemoresistance. We showed that the levels of phosphor and total eIF4E, as well as Mcl-1, were elevated in chemoresistant cervical but not colon cancer cells. Mcl-1 inhibitor S64315 decreased Mcl-1 levels in chemoresistant cancer cells, regardless of Mcl-1 upregulation, decreased viability in chemoresistant cancer cells and acted synergistically with chemotherapy drugs. The combined inhibition of Mcl-1 and B-cell lymphoma 2 (Bcl-2), employing both genetic and pharmacological approaches, led to a markedly more substantial decrease in viability compared with the inhibition of either target individually. The combination of S64315 and Bcl-2 inhibitors reduced tumor growth in chemoresistant cervical and colon cancer models without causing general toxicity in mice. This combination also prolonged overall survival compared with using S64315 or venetoclax alone. Our research highlights the therapeutic potential of inhibiting Mcl-1 and Bcl-2 simultaneously in chemoresistant cancers and provides a rationale for initiating clinical trials to investigate the combination of S64315 and venetoclax for the treatment of advanced colon and cervical cancer.
Collapse
Affiliation(s)
- Ling Wang
- Department of Obstetrics and Gynaecology
| | - Changlei Xi
- Department of Anorectal Surgery, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, China
| | - Rong Liu
- Department of Obstetrics and Gynaecology
| | | | - Ning Xiang
- Department of Obstetrics and Gynaecology
| | | | - Hui Li
- Department of Obstetrics and Gynaecology
| |
Collapse
|
7
|
Zhang Q, Zhao J, Xu T. Inhibition of eukaryotic initiation factor 4E by tomivosertib suppresses angiogenesis, growth, and survival of glioblastoma and enhances chemotherapy's efficacy. Fundam Clin Pharmacol 2023. [PMID: 36691859 DOI: 10.1111/fcp.12877] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/24/2022] [Accepted: 01/21/2023] [Indexed: 01/25/2023]
Abstract
Glioblastoma is characterized by extensive vascularization and is highly resistant to current therapy. Identification of drugs that target tumor directly and angiogenesis processes present an effective therapeutic strategy for glioblastoma. Mnk kinase is required for the activation of eukaryotic initiation factor 4E (eIF4E), which mediates translation of oncogenic proteins. We investigated the effects of tomivosertib, a novel MAPK-interacting kinase (MNK) inhibitor, on glioblastoma angiogenesis, growth, and survival. We found that tomivosertib inhibited growth and induced caspase-dependent apoptosis in various glioblastoma cell lines. Tomivosertib disrupted glioblastoma endothelial cell capillary network formation, growth, and survival. Mechanistically, tomivosertib acted on glioblastoma via suppressing MNK-dependent eIF4E phosphorylation and activation in tumor and endothelial cells. We further found that temozolomide activated eIF4E and this was reversed by tomivosertib. Using glioblastoma xenograft mouse model, we demonstrated that temozolomide and tomivosertib combination had higher efficacy than tomivosertib alone. Of note, tomivosertib inhibited glioblastoma angiogenesis and decreased p-eIF4E level in mice. We finally showed that p-eIF4E activation was a common molecular feature in glioblastoma patients. Our pre-clinical findings suggest that tomivosertib is a useful addition to the treatment armamentarium for glioblastoma and that targeting MNK-eIF4E pathway represents a therapeutic strategy to overcome glioblastoma chemoresistance.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Juan Zhao
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei Province, China
| | - Tingwei Xu
- Department of Neurosurgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, 14 Dong Street, Xiangcheng District, Xiangyang, 441021, Hubei Province, China
| |
Collapse
|