1
|
Yang X, Liu S, Liu Y, Wang Y, Cui D, Lan T, Zhu D, Su Z, Hao E, Qin L, Guo H. Total flavonoids of litchi seed inhibit breast cancer metastasis by regulating the PI3K/AKT/mTOR and MAPKs signaling pathways. PHARMACEUTICAL BIOLOGY 2025; 63:229-249. [PMID: 40231974 DOI: 10.1080/13880209.2025.2488135] [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: 07/08/2024] [Revised: 03/07/2025] [Accepted: 03/30/2025] [Indexed: 04/16/2025]
Abstract
CONTEXT Total flavonoids from Litchi chinensis Sonn. (Sapindaceae) seeds (TFLS) effectively attenuate stem cell-like properties in breast cancer cells. However, their pharmacological effects and mechanisms in suppressing breast cancer metastasis remain unclear. OBJECTIVE This study aimed to elucidate the inhibitory effects and underlying mechanisms of TFLS on breast cancer metastasis. MATERIALS AND METHODS The antiproliferative, migratory, and invasive activities of breast cancer cells following TFLS treatment were evaluated using CCK-8, wound-healing, and transwell assays. The epithelial-mesenchymal transition (EMT) biomarkers were evaluated via Western blot analysis. The anti-metastatic effects of TFLS were further validated in vivo using zebrafish and mouse models. Network pharmacology methodology was utilized to predict potential targets and signaling pathways, which were subsequently corroborated by Western blot. Potential active compounds were identified through molecular docking, and the chemical constituents of TFLS were analyzed and characterized using UPLC-QTOF/MS. RESULTS TFLS suppressed the proliferation of MDA-MB-231 and MDA-MB-468 cells, with IC50 values of 44.47 μg/mL and 37.35 μg/mL at 72 h, respectively. It effectively suppressed breast cancer metastasis in vitro, demonstrated by a marked reduction in cellular motility and invasiveness, alongside the reversal of EMT. Consistent with pathway enrichment analysis, network pharmacology revealed that TFLS reduced the phosphorylation levels of PI3K, AKT, mTOR, JNK, ERK, and p38 in breast cancer cells. Molecular docking identified seven potential active ingredients, and UPLC-MS/MS confirmed the presence of key compounds, including procyanidin A2. DISCUSSION AND CONCLUSION TFLS effectively inhibits breast cancer cell proliferation, migration, and invasion in vitro by reversing the EMT phenotype, while suppressing metastasis in vivo. These effects are likely mediated via the attenuation of the PI3K/AKT/mTOR and MAPK signaling pathways.
Collapse
Affiliation(s)
- Xin Yang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
| | - Shoushi Liu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Ying Liu
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica & College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Yuanshuo Wang
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Dianxin Cui
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Taijin Lan
- School of Preclinical Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Dan Zhu
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Zhiheng Su
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica & College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Lilan Qin
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica & College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, China
| | - Hongwei Guo
- Key Laboratory of Longevity and Aging-related Diseases of Chinese Ministry of Education & Center for Translational Medicine, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation & College of Pharmacy, Guangxi Medical University, Nanning, China
| |
Collapse
|
2
|
Xu Q, Yin Z, Li Y, Zhu X, Lou H, Ni J. Prognostic value of HER2 expression in cervical adenocarcinoma: A retrospective cohort study. Oncol Lett 2025; 29:217. [PMID: 40093868 PMCID: PMC11907399 DOI: 10.3892/ol.2025.14963] [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: 10/15/2024] [Accepted: 01/30/2025] [Indexed: 03/19/2025] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is an important therapeutic target in various types of cancer, although the prognostic value and therapeutic potential of HER2 in cervical adenocarcinoma are still underexplored. The present study aimed to examine the association between HER2 expression levels and prognosis in cervical adenocarcinoma, offering new insights into targeted therapies for HER2-expressing cervical adenocarcinoma. A total of 179 patients with cervical cancer who received surgery were included, and HER2 status in surgical specimens of the included patients were assessed using two classification methods: Immunohistochemistry (IHC) alone and traditional combined IHC/fluorescence in situ hybridization (FISH). IHC alone was used to categorize patients into the HER2 zero expression (IHC 0) and HER2 expression (IHC 1+, 2+ and 3+) groups, while traditional combined IHC/FISH classified the HER2 expression as negative (IHC 0 and 1+ or IHC 2+/FISH-) or positive (IHC 3+ or IHC 2+/FISH+). Kaplan-Meier survival analysis and log-rank tests were used to assess the patients' survival prognosis. A Cox proportional hazards regression model was used to identify independent prognostic factors. The HER2 expression rate was 44.1% (79/179) according to IHC alone, while 5.0% (9/179) were classified as HER2-positive according to the traditional method. HER2 expression was significantly associated with advanced International Federation of Gynecology and Obstetrics stages, higher rates of lymph node metastasis, vascular or perineural invasion, elevated cancer antigen 125 levels and increased recurrence rate (P<0.05). Moreover, HER2 expression was significantly associated with shorter progression-free survival (PFS) time [51.02±2.75 vs. 56.01±2.22 months; hazard ratio (HR), 0.559; 95% confidence interval (CI), 0.313-0.998; P=0.049]. Additionally, programmed death-ligand 1 expression levels were significantly higher in HER2-expressing patients who died (P=0.039). When HER2 status was assessed using the traditional combined IHC/FISH method, HER2 positivity was significantly associated with poorer PFS time (36.44±7.85 vs. 55.17±1.78 months; HR, 0.125; 95% CI, 0.03033-0.5156; P=0.004). In conclusion, classification of HER2 status in patients with cervical adenocarcinoma using IHC alone may provide a promising method for predicting patient outcomes and optimizing therapeutic strategies to improve treatment efficacy.
Collapse
Affiliation(s)
- Qing Xu
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Zhuomin Yin
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Yueqi Li
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Xiu Zhu
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Hanmei Lou
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Juan Ni
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| |
Collapse
|
3
|
Chen C, Li H, Zhang Z, Li H, Li H. F-box/WD Repeat-Containing Protein 5 Promotes Breast Cancer Progression by Regulating Ferroptosis via Enhancing Krüppel-like Factor 13 Ubiquitination Through Phosphoinositide 3-Kinase/Serine/Threonine Protein Kinase Pathway. Rejuvenation Res 2025. [PMID: 40228045 DOI: 10.1089/rej.2024.0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
Breast cancer (BC) is a prevalent malignancy among women. Evidence has indicated that F-box/WD repeat-containing protein 5 (FBXW5) is crucial in oncogenesis and progression. However, the function of FBXW5 in BC remains elusive. This work aims to explore the regulatory mechanisms of FBXW5 in the development of BC. The expression of FBXW5 in pan-cancer and breast invasive carcinoma (BRCA) was analyzed using The Cancer Genome Atlas (TCGA) database. FBXW5 level was enhanced in BC tissues. Besides, FBXW5 inhibition significantly decreased cell viability by 49.05% in MDA-MB-231 cells and 62.30% in MCF-7 cells. FBXW5 inhibition significantly inhibited cell proliferation by 66% in MDA-MB-231 cells and 74% in MCF-7 cells. FBXW5 inhibition significantly suppressed cell migration by 77.2% in MDA-MB-231 cells and 82.15% in MCF-7 cells. FBXW5 inhibition significantly inhibited cell invasion by 64.14% in MDA-MB-231 cells and 71.33% in MCF-7 cells. In vivo, FBXW5 depletion reduced tumor weight by 63.39% and tumor volume by 65.17%. Moreover, FBXW5 silencing restrained lung metastases in vivo. Besides, the impact of FBXW5 on the malignant behavior of BC cells was mediated through the regulation of ferroptosis. Mechanically, FBXW5 facilitated Kruppel-like factor 13 (KLF13) degradation by enhancing its ubiquitination. The addition of FBXW5 facilitated cell proliferation, migration, and invasion and inhibited ferroptosis in MDA-MB-231 and MCF-7 cells, which were neutralized by KLF13 overexpression. Besides, the knockdown of KLF13 led to the activation of the PI3K/AKT pathway. KLF13 silencing counteracted the inhibitory effects of FBXW5 depletion on cell proliferation, migration, and invasion, as well as its promotion of ferroptosis, effects that were reversed by LY294002. In conclusion, targeting FBXW5 may serve as a potential therapeutic strategy for BC by modulating the KLF13/PI3K/AKT axis.
Collapse
Affiliation(s)
- Chen Chen
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Hui Li
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Ziyi Zhang
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Haipeng Li
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Hongtao Li
- Department of Oncological Surgery, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| |
Collapse
|
4
|
Huang Z, Wang H, Pang H, Zeng M, Zhang G, Liu F. The Comprehensive Analysis of Weighted Gene Co-Expression Network Analysis and Machine Learning Revealed Diagnostic Biomarkers for Breast Implant Illness Complicated with Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2025; 17:305-324. [PMID: 40230814 PMCID: PMC11996000 DOI: 10.2147/bctt.s507754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025]
Abstract
Purpose An increasing number of breast cancer (BC) patients choose prosthesis implantation after mastectomy, and the occurrence of breast implant illness (BII) has received increasing attention and the underlying molecular mechanisms have not been clearly elucidated. This study aimed to identify the crosstalk genes between BII and BC and explored their clinical value and molecular mechanism initially. Methods We retrieved the data from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA), and identified the differentially expressed genes (DEG) as well as module genes using Limma and weighted gene co-expression network analysis (WGCNA). Enrichment analysis, the protein-protein interaction network (PPI), and machine learning algorithms were performed to explore the hub genes. We employed a nomogram and receiver operating characteristic curve to evaluate the diagnostic accuracy. Single-cell analysis disclosed variations in the expression of key genes across distinct cellular populations. The expression levels of the key genes were further confirmed in BC cell lines. Immunohistochemical analysis was utilized to examine protein levels from 25 patients with breast cancer undergoing prosthetic implant surgery. Ultimately, we deployed single-sample Gene Set Enrichment Analysis (ssGSEA) to scrutinize the immunological profiles between the normal and BC cohorts, as well as between the non-BII and BII groups. Results WGCNA identified 1137 common genes, whereas DEG analysis found 541 overlapping genes in BII and BC. After constructing the PPI network, 17 key genes were selected, and three potential hub genes include KRT14, KIT, ALB were chosen for nomogram creation and diagnostic assessment through machine learning. The validation of these results was conducted by examining gene expression patterns in the validation dataset, breast cancer cell lines, and BII-BC patients. However, ssGSEA uncovered different immune cell infiltration patterns in BII and BC. Conclusion We pinpointed shared three central genes include KRT14, KIT, ALB and molecular pathways common to BII and BC. Shedding light on the complex mechanisms underlying these conditions and suggesting potential targets for diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Zhenfeng Huang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, People’s Republic of China
| | - Huibo Wang
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong Province, People’s Republic of China
| | - Hui Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, People’s Republic of China
| | - Mengyao Zeng
- Department of Medical Training, Aimiker Technology Development Co., Ltd, Nanjing, Jiangsu Province, People’s Republic of China
| | - Guoqiang Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, People’s Republic of China
| | - Feng Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, People’s Republic of China
| |
Collapse
|
5
|
Wang B, Hao S, Han F, Wu T, Jia S, Ruan X, Zhou Q. Discovery of 5-Phenylthiazol-2-amine Derivatives as Novel PI4KIIIβ Inhibitors with Efficacious Antitumor Activity by Inhibiting the PI3K/AKT Axis. J Med Chem 2025; 68:6270-6291. [PMID: 40047238 DOI: 10.1021/acs.jmedchem.4c02588] [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: 03/28/2025]
Abstract
To develop novel PI4KIIIβ inhibitors and explore their antitumor activity, a series of 5-phenylthiazol-2-amine derivatives were synthesized by structural modifications of PIK93. Biological assay results indicated that compounds 16 and 43 exhibited superior PI4KIIIβ selective inhibitory and antiproliferative activity than PIK93. Mechanistic studies revealed that the two compounds inhibit the PI3K/AKT pathway more effectively, thereby inducing cancer cell apoptosis, cycle arrest in the G2/M phase and autophagy. Importantly, in vivo toxicity and pharmacodynamics studies showed that compounds 16 and 43 exhibited superior safety to that of commercially available PI3K/AKT axis inhibitor alpelisib, and obviously antitumor activity in small cell lung cancer H446 xenograft models. Overall, this work highlights the therapeutic potential and safety of PI4KIIIβ inhibitors 16 and 43 in the treatment of tumors, and provides candidates and viable drug development strategies for the treatment of small cell lung cancer and the development of novel PI3K/AKT axis inhibitors.
Collapse
Affiliation(s)
- Bichuan Wang
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Siyuan Hao
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Fang Han
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Tianzhi Wu
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Shuolei Jia
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Xiuqin Ruan
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | - Qingfa Zhou
- State Key Laboratory of Natural Medicines, Department of Organic Chemistry, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
6
|
Liu Y, Li H, Chai D, Lian B, Bai Z, Gao Y, Li J. LncRNA TCL6 regulates miR-876-5p/MYL2 axis to suppress breast cancer progression. Transl Oncol 2025; 53:102210. [PMID: 39874729 PMCID: PMC11810844 DOI: 10.1016/j.tranon.2024.102210] [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: 04/28/2024] [Revised: 09/26/2024] [Accepted: 11/18/2024] [Indexed: 01/30/2025] Open
Abstract
We explored the influence of the TCL6/miR-876-5p axis on breast cancer cell proliferation and migration. Using The Cancer Genome Atlas (TCGA) database, we evaluated the expression of TCL6 in breast cancer patients and studied its effects on cell proliferation, migration, and the cell cycle in vitro. The regulatory effect of miR-876-5p on myosin light chain-2 (MYL2) 3' untranslated regions (3'UTR) was analyzed through luciferase reporter assays, and rescue experiments confirmed TCL6-driven upregulation of MYL2 via a competitive RNA binding mechanism. Furthermore, we used a mouse subcutaneous tumor model to assess the impact of TCL6 knockdown combined with immune checkpoint blockade therapy. Our results indicated that higher TCL6 expression correlated with a favorable prognosis in breast cancer patients. In vitro experiments showed that knockdown of TCL6 and MYL2 enhanced breast cancer cell proliferation and migration. The luciferase and rescue assays demonstrated that TCL6 interacted with miR-876-5p to upregulate MYL2, thereby inhibiting cell proliferation and migration. Both in vitro and in vivo studies revealed that overexpression of TCL6 suppressed tumor growth and improved the response to PD-1 immunotherapy in tumor-bearing mice. This research highlights the pivotal role of lncRNA TCL6 in breast cancer development via a ceRNA network involving miR-876-5p and MYL2, suggesting a novel molecular target for breast cancer therapy.
Collapse
Affiliation(s)
- YaoBang Liu
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Hong Li
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - DaHai Chai
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Bin Lian
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | | | - YaLi Gao
- Ningxia Medical University, Yinchuan 750004, China
| | - JinPing Li
- Department of Surgical Oncology, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
7
|
Oner M, Cheng YC, Soong SW, Cheng PT, Wang YH, Yang SF, Tsai SCS, Lin H. Dinaciclib Interrupts Cell Cycle and Induces Apoptosis in Oral Squamous Cell Carcinoma: Mechanistic Insights and Therapeutic Potential. Int J Mol Sci 2025; 26:2197. [PMID: 40076816 PMCID: PMC11900514 DOI: 10.3390/ijms26052197] [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: 12/03/2024] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Dinaciclib, a potent cyclin-dependent kinase (CDK) inhibitor, has demonstrated considerable antitumor effects in various malignancies. However, its impact on oral squamous cell carcinoma (OSCC), a predominant and highly aggressive form of head and neck squamous cell carcinoma (HNSC) with limited treatment options, remains underexplored. We conducted gene set enrichment analyses in HNSC patients that reinforced the relevance of these cell cycle-related genes to OSCC pathogenesis. Given the known dysregulation of cell cycle-related genes in HNSC patients, we hypothesized that Dinaciclib may inhibit OSCC growth by targeting overexpressed cyclins and CDKs, thereby disrupting cell cycle progression and inducing apoptosis. This study investigated Dinaciclib's effects on cell proliferation, cell cycle progression, and apoptosis in the OSCC cell lines Ca9-22, OECM-1, and HSC-3. Our results demonstrated that Dinaciclib significantly reduces OSCC cell proliferation in a dose-dependent manner. Flow cytometry and Western blot analyses showed that Dinaciclib induces cell cycle arrest at the G1/S and G2/M transitions by downregulating Cyclins A, B, D, and E, along with CDKs 1 and 2-key regulators of these checkpoints. Furthermore, Dinaciclib treatment upregulated apoptotic markers, such as cleaved-caspase-3 and cleaved-PARP, confirming its pro-apoptotic effects. In conclusion, these findings highlight Dinaciclib's therapeutic promise in OSCC by simultaneously disrupting cell cycle progression and inducing apoptosis. These results support further exploration of Dinaciclib as a viable monotherapy or combination treatment in OSCC and other HNSC subtypes to improve patient outcomes.
Collapse
Affiliation(s)
- Muhammet Oner
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan; (M.O.); (Y.-C.C.); (S.-W.S.); (P.-T.C.)
| | - Yu-Chiao Cheng
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan; (M.O.); (Y.-C.C.); (S.-W.S.); (P.-T.C.)
| | - Shiuan-Woei Soong
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan; (M.O.); (Y.-C.C.); (S.-W.S.); (P.-T.C.)
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Pang-Ting Cheng
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan; (M.O.); (Y.-C.C.); (S.-W.S.); (P.-T.C.)
| | - Yan-Hsiung Wang
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
| | - Shun-Fa Yang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan;
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Stella Chin-Shaw Tsai
- Superintendent Office, Tungs’ Taichung MetroHarbor Hospital, Taichung 43503, Taiwan
- College of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Post-Baccalaureate Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Ho Lin
- Department of Life Sciences, National Chung Hsing University, Taichung 40227, Taiwan; (M.O.); (Y.-C.C.); (S.-W.S.); (P.-T.C.)
| |
Collapse
|
8
|
Nussinov R, Yavuz BR, Jang H. Molecular principles underlying aggressive cancers. Signal Transduct Target Ther 2025; 10:42. [PMID: 39956859 PMCID: PMC11830828 DOI: 10.1038/s41392-025-02129-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/02/2024] [Accepted: 01/07/2025] [Indexed: 02/18/2025] Open
Abstract
Aggressive tumors pose ultra-challenges to drug resistance. Anti-cancer treatments are often unsuccessful, and single-cell technologies to rein drug resistance mechanisms are still fruitless. The National Cancer Institute defines aggressive cancers at the tissue level, describing them as those that spread rapidly, despite severe treatment. At the molecular, foundational level, the quantitative biophysics discipline defines aggressive cancers as harboring a large number of (overexpressed, or mutated) crucial signaling proteins in major proliferation pathways populating their active conformations, primed for their signal transduction roles. This comprehensive review explores highly aggressive cancers on the foundational and cell signaling levels, focusing on the differences between highly aggressive cancers and the more treatable ones. It showcases aggressive tumors as harboring massive, cancer-promoting, catalysis-primed oncogenic proteins, especially through certain overexpression scenarios, as predisposed aggressive tumor candidates. Our examples narrate strong activation of ERK1/2, and other oncogenic proteins, through malfunctioning chromatin and crosslinked signaling, and how they activate multiple proliferation pathways. They show the increased cancer heterogeneity, plasticity, and drug resistance. Our review formulates the principles underlying cancer aggressiveness on the molecular level, discusses scenarios, and describes drug regimen (single drugs and drug combinations) for PDAC, NSCLC, CRC, HCC, breast and prostate cancers, glioblastoma, neuroblastoma, and leukemia as examples. All show overexpression scenarios of master transcription factors, transcription factors with gene fusions, copy number alterations, dysregulation of the epigenetic codes and epithelial-to-mesenchymal transitions in aggressive tumors, as well as high mutation loads of vital upstream signaling regulators, such as EGFR, c-MET, and K-Ras, befitting these principles.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA.
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD, 21702, USA.
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, 69978, Tel Aviv, Israel.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| |
Collapse
|
9
|
Nedeljković M, Vuletić A, Mirjačić Martinović K. Divide and Conquer-Targeted Therapy for Triple-Negative Breast Cancer. Int J Mol Sci 2025; 26:1396. [PMID: 40003864 PMCID: PMC11855393 DOI: 10.3390/ijms26041396] [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: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant type of breast cancer with limited treatment options and poor prognosis. One of the most significant impediments in TNBC treatment is the high heterogeneity of this disease, as highlighted by the detection of several molecular subtypes of TNBC. Each subtype is driven by distinct mutations and pathway aberrations, giving rise to specific molecular characteristics closely connected to clinical behavior, outcomes, and drug sensitivity. This review summarizes the knowledge regarding TNBC molecular subtypes and how it can be harnessed to devise tailored treatment strategies instead of blindly using targeted drugs. We provide an overview of novel targeted agents and key insights about new treatment modalities with an emphasis on the androgen receptor signaling pathway, cancer stem cell-associated pathways, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, growth factor signaling, and immunotherapy.
Collapse
Affiliation(s)
- Milica Nedeljković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (A.V.); (K.M.M.)
| | | | | |
Collapse
|
10
|
Batal A, Garousi S, Finnson KW, Philip A. CD109, a master regulator of inflammatory responses. Front Immunol 2025; 15:1505008. [PMID: 39990858 PMCID: PMC11842317 DOI: 10.3389/fimmu.2024.1505008] [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: 10/01/2024] [Accepted: 12/23/2024] [Indexed: 02/25/2025] Open
Abstract
Inflammation is a complex response to harmful stimuli, crucial for immunity, and linked to chronic diseases and cancer, with TGF-β and NF-κB pathways as key regulators. CD109 is a glycosylphosphatidylinositol (GPI)-anchored protein, that our group has originally identified as a TGF-β co-receptor and inhibitor of TGF-β signaling. CD109 modulates TGF-β and NF-κB pathways, to influence immune responses and inflammation. CD109's multifaceted role in inflammation spans various tissue types, including the skin, lung, bone and bone-related tissues, and various types of cancers. CD109 exerts its effects by modulating processes such as cytokine secretion, immune cell recruitment, macrophage polarization, T helper cell function and cancer cell phenotype and function. Here, we review CD109's regulatory functions in inflammatory responses in these various tissues and cell types. Exploration of CD109's mechanisms of action will enhance our understanding of its contributions to disease pathology and its potential for therapeutic applications.
Collapse
Affiliation(s)
- Adel Batal
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- Surgical and Interventional Sciences Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Setareh Garousi
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- Surgical and Interventional Sciences Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Kenneth W. Finnson
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- Surgical and Interventional Sciences Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Anie Philip
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, QC, Canada
- Surgical and Interventional Sciences Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
11
|
Xu L, Xu P, Wang J, Ji H, Zhang L, Tang Z. Advancements in clinical research and emerging therapies for triple-negative breast cancer treatment. Eur J Pharmacol 2025; 988:177202. [PMID: 39675457 DOI: 10.1016/j.ejphar.2024.177202] [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: 08/02/2024] [Revised: 11/30/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Triple-negative breast cancer (TNBC), defined by the lack of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor 2 (HER2) expression, is acknowledged as the most aggressive form of breast cancer (BC), comprising 15%-20% of all primary cases. Despite the prevalence of TNBC, effective and well-tolerated targeted therapies remain limited, with chemotherapy continuing to be the mainstay of treatment. However, the horizon is brightened by recent advancements in immunotherapy and antibody-drug conjugates (ADCs), which have garnered the U.S. Food and Drug Administration (FDA) approval for various stages of TNBC. Poly (ADP-ribose) polymerase inhibitors (PARPi), particularly for TNBC with BRCA mutations, present a promising avenue, albeit with the challenge of resistance that must be addressed. The success of phosphoinositide-3 kinase (PI3K) pathway inhibitors in hormone receptor (HR)-positive BC suggests potential applicability in TNBC, spurring optimism within the research community. This review endeavors to offer a comprehensive synthesis of both established and cutting-edge targeted therapies for TNBC. We delve into the specifics of PARPi, androgen receptor (AR) inhibitors, Cancer stem cells (CSCs), PI3K/Protein Kinase B (AKT)/mammalian target of rapamycin (mTOR), the transforming growth factor-beta (TGF-β), Ntoch, Wnt/β-catenin, hedgehog (Hh) pathway inhibitors, Epigenetic target-mediated drug delivery, ADCs, immune checkpoint inhibitors (ICIs)and novel immunotherapeutic solutions, contextualizing TNBC within current treatment paradigms. By elucidating the mechanisms of these drugs and their prospective clinical applications, we aim to shed light on the challenges and underscore the beacon of hope that translational research and innovative therapies represent for the oncology field.
Collapse
Affiliation(s)
- Lili Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Pengtao Xu
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Jingsong Wang
- Department of Pharmacy, Guangyuan Central Hospital, Guangyuan, Sichuan, 628000, China
| | - Hui Ji
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Lin Zhang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People's Hospital, Shaoxing, Zhejiang, 312000, China.
| |
Collapse
|
12
|
Effiong ME, Bella-Omunagbe M, Afolabi IS, Chinedu SN. Molecular Docking Appraisal of Pleurotus ostreatus Phytochemicals as Potential Inhibitors of PI3K/Akt Pathway for Breast Cancer Treatment. Bioinform Biol Insights 2025; 19:11779322251316864. [PMID: 39906062 PMCID: PMC11792010 DOI: 10.1177/11779322251316864] [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: 06/09/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025] Open
Abstract
Introduction Breast cancer (BC) is a heterogeneous disease involving a network of numerous extracellular signal transduction pathways. The phosphoinositide 3-kinase (PI3K)/serine/threonine kinase (Akt)/mechanistic target of rapamycin (mTOR) pathway is crucial for understanding the BC development. Phosphoinositide 3-kinase, phosphatase and tensin homolog (PTEN), mTOR, Akt, 3-phosphoinositide-dependent kinase 1 (PDK1), FoxO1, glycogen synthase kinase 3 (GSK-3), mouse double minute 2 (MDM2), H-Ras, and proapoptotic B-cell lymphoma 2 (BCL-2) family protein (BAD) proteins are key drivers of this pathway and potential therapeutic targets. Pleurotus ostreatus is an edible mushroom that is rich in flavonoids and phenols that can serve as potential inhibitors of proteins in the PI3K/Akt/mTOR pathway. Aim This study evaluated the anticancer properties of P ostreatus through a structure-based virtual screening of 22 biologically active compounds present in the mushroom. Method Model optimization was carried out on PI3K, PTEN, mTOR, Akt, PDK1, FoxO1, GSK-3, MDM2, H-Ras, and BAD proteins in the PI3K/Akt/mTOR pathway and molecular docking of compounds/control inhibitors in the binding pocket were simulated AutoDock Vina in PyRx. The drug likeness, pharmacokinetic, and pharmacodynamic features of prospective docking leads were all anticipated. Result Several potent inhibitors of the selected key driver proteins in PI3K/Akt/mTOR pathway were identified from P ostreatus. Ellagic acid with binding affinities of -8.0, -8.0, -8.1, -8.2, -6.2, and -7.1 kcal/mol on PI3K, Akt, PDK1, GSK-3, MDM2, and BAD, respectively, had better binding affinity compared with their reference drugs. Likewise, apigenin (-7.8 kcal/mol), chrysin (-7.8 kcal/mol), quercetin (-6.4 kcal/mol), and chlorogenic acid (-6.2 kcal/mol) had better binding affinities to PTEN, mTOR, FoxO1, and H-Ras proteins, respectively. Conclusion Ellagic acid, apigenin, luteolin, quercetin, chlorogenic acid, chrysin, and naringenin phytochemicals are seen as the better lead molecules due to their ability to strongly bind to the proteins under study in this pathway. Analogs of these compounds can also be designed as potential drugs.
Collapse
Affiliation(s)
- Magdalene Eno Effiong
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Mercy Bella-Omunagbe
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Israel Sunmola Afolabi
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant University Public Health and Wellbeing Research Cluster (CUPHWERC), Covenant University, Ota, Nigeria
| | - Shalom Nwodo Chinedu
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant University Public Health and Wellbeing Research Cluster (CUPHWERC), Covenant University, Ota, Nigeria
| |
Collapse
|
13
|
Richters L, Gluz O, Weber-Lassalle N, Christgen M, Haverkamp H, Kuemmel S, Kayali M, Kates RE, Grischke EM, Altmüller J, Forstbauer H, Thiele H, Braun M, Warm M, Ossowski A, Wuerstlein R, Ernst C, Graeser M, Linn SC, Nitz U, Hauke J, Kreipe HH, Schmutzler RK, Hahnen E, Harbeck N. Genetic Alterations, Therapy Response, and Survival Among Patients With Triple-Negative Breast Cancer: A Secondary Analysis of a Randomized Clinical Trial. JAMA Netw Open 2025; 8:e2461639. [PMID: 40009381 DOI: 10.1001/jamanetworkopen.2024.61639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/27/2025] Open
Abstract
Importance Subgroup definitions for possible deescalation of neoadjuvant cancer treatment are urgently needed in clinical practice. Objective To investigate the effect of BRCA1 and/or BRCA2 tumor pathogenic variants (tPVs) by comparing 2 deescalated neoadjuvant regimens (nab-paclitaxel plus either carboplatin or gemcitabine) on pathologic complete response (pCR), invasive disease-free survival (IDFS), and overall survival (OS) of patients with early-stage triple-negative breast cancer (TNBC). Design, Setting, and Participants This was a preplanned secondary analysis of a phase 2 prospective randomized clinical trial (ADAPT-TN) conducted by the West German Study Group (WSG) at 45 sites in Germany between June 2013 and February 2015. The trial enrolled patients with noninflammatory early-stage TNBC (clinical tumor size ≥1 cm; estrogen receptor and progesterone receptor expression <1%; and ERBB2 negative). DNA samples from pretreatment biopsies were obtained. Genetic analysis was performed between January 2018 and March 2020. Final data analyses took place in September 2023. Exposure Patients were randomized to 12 weeks of treatment with nab-paclitaxel plus either carboplatin or gemcitabine; omission of otherwise mandatory anthracycline-containing chemotherapy was allowed in the case of pCR. tPVs in 20 cancer-associated genes, including BRCA1 and BRCA2, were analyzed using a customized gene panel. Main Outcomes and Measures The prevalence of BRCA1 and/or BRCA2 tPVs and their effect on pCR rate, IDFS, and OS were evaluated using logistic and Cox proportional hazards regression. Results Of the 307 patients with DNA samples from pretreatment biopsies available, tumor next-generation sequencing analyses were successful for 266 patients. The 266 patients included in this analysis were female, with a median age of 51 years (range, 26-76 years). A total of 162 patients (60.9%) had a clinical tumor size of 2 cm or greater, and 70 (26.3%) had clinical node-positive disease. BRCA1 and/or BRCA2 tPVs were detected in 42 patients (15.8%). The highest pCR rate among patients with BRCA1 and/or BRCA2 tPVs was seen in the nab-paclitaxel plus carboplatin group (9 of 14 patients [64.3%]) compared with the nab-paclitaxel plus gemcitabine group (10 of 28 [35.7%]) (odds ratio, 3.24 [95% CI, 0.85-12.36]; P = .08); the highest numeric 5-year IDFS and OS rates (84.4% and 92.9%, respectively) were seen in the nab-paclitaxel plus carboplatin group. Conclusions and Relevance In this secondary analysis of the WSG-ADAPT-TN randomized clinical trial on tPVs, deescalated nab-paclitaxel plus carboplatin was superior to nab-paclitaxel plus gemcitabine, particularly in patients with BRCA1 and/or BRCA2 tPVs. These findings suggest that BRCA1 and/or BRCA2 tPV status could be a candidate marker for a deescalation strategy in early-stage TNBC; however, prospective validation of survival outcomes in larger cohorts with differentiation between germline and somatic pathogenic variants is necessary. Trial Registration ClinicalTrials.gov Identifier: NCT01815242.
Collapse
Affiliation(s)
- Lisa Richters
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Center for Integrated Oncology, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Oleg Gluz
- West German Study Group, Moenchengladbach, Germany
- Ev. Hospital Bethesda, Breast Center Niederrhein, Moenchengladbach, Germany
- Women's Clinic and Breast Center, University Clinics Cologne, Cologne, Germany
| | - Nana Weber-Lassalle
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Center for Integrated Oncology, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | | | - Heinz Haverkamp
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Now with Miltenyi Biomedicine GmbH, Bergisch Gladbach, Germany
| | - Sherko Kuemmel
- West German Study Group, Moenchengladbach, Germany
- Interdisciplinary Breast Center, Kliniken Essen-Mitte, Evang. Hospital Essen-Mitte, Essen, Germany
- Department of Gynecology, Breast Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mohamad Kayali
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Center for Integrated Oncology, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | | | | | - Janine Altmüller
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Core Unit Genomics, Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Michael Braun
- Interdisciplinary Breast Center, Rotkreuz-Clinics Munich, Munich, Germany
| | - Mathias Warm
- Breast Center, Municipal Hospital Holweide, Cologne, Germany
| | - Anna Ossowski
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Rachel Wuerstlein
- West German Study Group, Moenchengladbach, Germany
- Breast Center, Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital of Ludwig Maximilian University, Munich, Germany
| | - Corinna Ernst
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Center for Integrated Oncology, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Monika Graeser
- West German Study Group, Moenchengladbach, Germany
- Ev. Hospital Bethesda, Breast Center Niederrhein, Moenchengladbach, Germany
- Department of Gynecology, University Medical Center Hamburg, Hamburg, Germany
| | - Sabine C Linn
- Department of Molecular Pathology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Ulrike Nitz
- West German Study Group, Moenchengladbach, Germany
| | - Jan Hauke
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Center for Integrated Oncology, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | | | - Rita K Schmutzler
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Center for Integrated Oncology, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Eric Hahnen
- Center for Familial Breast and Ovarian Cancer, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
- Center for Integrated Oncology, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Nadia Harbeck
- West German Study Group, Moenchengladbach, Germany
- Breast Center, Department of Obstetrics and Gynecology, Comprehensive Cancer Center Munich, University Hospital of Ludwig Maximilian University, Munich, Germany
| |
Collapse
|
14
|
De SK. Capivasertib: First Approved AKT inhibitor for the Treatment of Patients with Breast Cancer. Anticancer Agents Med Chem 2025; 25:371-377. [PMID: 39633517 DOI: 10.2174/0118715206360571241126080725] [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: 10/05/2024] [Revised: 11/02/2024] [Accepted: 11/05/2024] [Indexed: 12/07/2024]
Abstract
Breast cancer frequently occurs in women. Among the several types of breast cancers, almost 50% of breast cancers are caused by one or more gene mutations of the PI3K/mTOR/AKT pathway. Capivasertib, the first AKT inhibitor, was authorized by the US FDA on November 16, 2023. It is used for the treatment of adult patients with hormone receptor-positive, human epidermal growth factor receptor 2 negative metastatic breast cancer with at least one alteration on PIK3CA/AKT1/PTEN. In this short perspective, Capivasertib's physicochemical properties, synthesis, mechanism of action, binding mode, pharmacokinetics, drug interaction studies, and treatment-emergent adverse events are discussed.
Collapse
Affiliation(s)
- Surya K De
- Department of Chemistry, Conju-Probe, San Diego, California, USA
| |
Collapse
|
15
|
Pachuau L, Lalremmawia H, Ralte L, Vanlalpeka J, Pautu JL, Chenkual S, Zomuana T, Lalruatfela ST, Zohmingthanga J, Chhakchhuak L, Varma AK, Kumar NS. Uncovering novel pathogenic variants and pathway mutations in triple-negative breast cancer among the endogamous mizo tribe. Breast Cancer Res Treat 2025; 209:375-387. [PMID: 39384723 DOI: 10.1007/s10549-024-07501-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/23/2024] [Indexed: 10/11/2024]
Abstract
PURPOSE The incidence of triple-negative breast cancer (TNBC) in India is higher compared to Western populations. The objective of this study is to identify novel and less reported variants in TNBC in Mizoram, a state with a high cancer incidence in India. METHODS We analysed whole exome sequencing data from triple-negative breast cancer (TNBC) patients in the Mizo population to identify key and novel variants. Moreover, we analysed reported breast cancer-related genes and pathway alterations. RESULTS Somatic mutation analysis revealed that TP53 was the most frequently mutated gene and TP53, CACNA1E, IGSF3, RYR1, and FAM155A as significantly mutated driver genes. Based on the ACMG guidelines, we identified a rare pathogenic germline variant of BRCA1 (p.C1697R) in 13% and a likely pathogenic frameshift insertion in RBMX (p.P106Ffs) in 73% of the patients. We also found that the ATM, STK11, and CDKN2A genes were significantly mutated in germline TNBC samples compared to healthy samples. Moreover, we identified novel somatic variants in CHEK2 (p.K182M) and NF1 (p.C245X), and novel germline variants RB1 (p.D111G), CDH1 (p.A10Gfs), CDKN2A (p.V96G), CDKN2A (p.S12Afs*22), MAP3K1 (CAAdelins0), MSH6 (p.L1226_L1230del), and PMS2 (TTCdelins0). Pathway analysis revealed that most somatic mutations were highly associated with PI3K-Akt signalling pathway and MAPK signalling pathways in TNBC. CONCLUSIONS These findings identified novel variants and key genes contributing to disease development and progression. Further analysis of less studied genes, including RBMX, MRC1, ATM, CTNNB1, and CDKN2A, in TNBC may reveal new potential genes for targeted therapeutic strategies and contribute to clinical advancements in the treatment of TNBC.
Collapse
Affiliation(s)
- Lalawmpuii Pachuau
- Department of Biotechnology, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
- Department of Pathology, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, Mizoram, 796001, India
| | - H Lalremmawia
- Department of Biotechnology, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
| | - Lalengkimi Ralte
- Department of Biotechnology, Mizoram University, Tanhril, Aizawl, Mizoram, 796004, India
| | - Johan Vanlalpeka
- Department of Medicine, Zoram Medical College, Falkawn, Aizawl, Mizoram, 796 005, India
| | - Jeremy Lalrinsanga Pautu
- Department of Medical Oncology, Mizoram State Cancer Institute, Zemabawk, Aizawl, Mizoram, 796017, India
| | - Saia Chenkual
- Department of Surgery, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, 796001, Mizoram, India
- Zoram Medical College, Falkawn, Aizawl, Mizoram, 796 005, India
| | - Thomas Zomuana
- Department of Surgery, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, 796001, Mizoram, India
| | - Sailo Tlau Lalruatfela
- Department of Surgery, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, 796001, Mizoram, India
| | | | - Lalchhandama Chhakchhuak
- Department of Pathology, Department of Health & Family Welfare, Civil Hospital Aizawl, Government of Mizoram, Dawrpui, Aizawl, Mizoram, 796001, India
| | - Ashok K Varma
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Kharghar, Navi Mumbai, Maharashtra, 410210, India
- Homi Bhabha National Institute, Anushaktinagar, Maharastra, 400094, Mumbai, India
| | | |
Collapse
|
16
|
Feng S, Li J, Yan A, Zhu X, Zhang L, Tang D, Liu L. Application of Gene Editing in Triple-Negative Breast Cancer Research. Cell Biochem Funct 2025; 43:e70044. [PMID: 39844394 DOI: 10.1002/cbf.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/24/2025]
Abstract
With the rapid development of gene editing technology, its application in breast cancer has gradually become the focus of research. This article reviews the application of gene editing technology in the treatment of breast cancer, and discusses its challenges and future development directions. The key application areas of gene editing technology in the treatment of breast cancer will be outlined, including the discovery of new therapeutic targets and the development of drugs related to the pathway. Gene editing technology has played an important role in the discovery of new therapeutic targets. Through the use of gene editing technology, breast cancer-related genes are systematically edited to regulate key regulatory factors on related pathways or key tumor suppressor genes such as FOXC1 and BRCA, and the results are analyzed in cell or animal experiments, and the target is obtained from the experimental results, which provides important clues for the development of new drugs. This approach provides an innovative way to find more effective treatment strategies and inhibit tumor growth. In addition, gene editing technology has also promoted the personalization of breast cancer treatment. By analyzing a patient's genomic information, researchers can pinpoint key genetic mutations in a patient's tumor and design personalized treatments. This personalized treatment approach is expected to improve the therapeutic effect and reduce adverse reactions. Finally, the application of gene editing technology also provides support for the development of breast cancer immunotherapy. By editing immune cells to make them more potent against tumors, researchers are trying to develop more effective immunotherapies to bring new treatment options to breast cancer patients.
Collapse
Affiliation(s)
- Shuying Feng
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jixia Li
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Aifen Yan
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Xiangxing Zhu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Ligang Zhang
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Dongsheng Tang
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Lian Liu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| |
Collapse
|
17
|
Bashar MA, Hossain MA, Kavey MRH, Shazib R, Islam MS, Ansari SA, Rahman MH. Network Pharmacology and In silico Elucidation of Phytochemicals Extracted from Ajwa Dates ( Phoenix dactylifera L.) to Inhibit Akt and PI3K Causing Triple Negative Breast Cancer (TNBC). Curr Pharm Des 2025; 31:774-796. [PMID: 39698883 DOI: 10.2174/0113816128348876241017101729] [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: 08/09/2024] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND About 10-15% of all breast cancers comprise triple-negative breast cancer (TNBC), defined as cancer cells that lack ER, PR, and HER2 protein receptors. Due to the absence of these receptors, treating TNBC using conventional chemotherapy is challenging and, therefore, requires the discovery of novel chemotherapeutic agents derived from natural sources. OBJECTIVE The current work was intended to study the potential phytochemicals of Ajwa dates (Phoenix dactylifera L.) with the predicted potential targets (namely, Akt and PI3K) to determine possible TNBC inhibitors. METHODS We harnessed network pharmacology, molecular docking, drug-likeness studies, Molecular Dynamics (MD) simulation, and binding free energy (MM-GBSA) calculation to get phytochemicals with potential effects against TNBC. Firstly, molecular docking was performed on 125 phytochemicals against the Akt and PI3K proteins utilizing PyRx. Then, the phytochemicals with the highest binding affinity (≤ -8.1 kcal/mol) were examined for in silico drug-likeness and toxicity profiles. Finally, phytochemicals with optimal druglikeness and toxicity profiles were studied by Molecular Dynamics (MD) simulation and binding free energy (MM-GBSA) to identify compounds that can form stable complexes. RESULTS The results of the network pharmacology revealed that the Akt and PI3K proteins are potential targets of TNBC for the phytochemicals of Phoenix dactylifera L. used in this study. The outcomes of molecular docking displayed that among 125 phytochemicals, 42 of them (with a binding affinity ≤ -8.1 kcal/mol) have potentially inhibiting effects on both proteins PI3K and Akt expressed in TNBC. Then, the results of in silico drug-likeness identified seven phytochemicals with optimal pharmacokinetic profiles. Furthermore, toxicity studies showed that three phytochemicals (namely, Chrysoeriol, Daidzein, and Glycitein) did not cause any toxicities. Finally, the Molecular Dynamics (MD) simulation studies and binding free energy (MM-GBSA) verified that Daidzein stayed within the binding cavities of both proteins (Akt and PI3K) by establishing a stable protein-ligand complex during simulation. CONCLUSION Taken together, the current work emphasizes the potential effects of Daidzein from Phoenix dactylifera L. against TNBC, and it can be further studied to establish it as a standard chemotherapy for TNBC.
Collapse
Affiliation(s)
- Md Abul Bashar
- Department of Pharmacy, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Md Arju Hossain
- Department of Biotechnology and Genetic Engineering, Mawlana Bhashani Science and Technology University, Santosh, Tangail 1902, Bangladesh
- Department of Microbiology, Primeasia University, Banani, Dhaka 1213, Bangladesh
| | - Md Reduanul Haque Kavey
- Department of Pharmacy, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Rayhanuzzaman Shazib
- Department of Pharmacy, Faculty of Biological Sciences, Islamic University, Kushtia 7003, Bangladesh
| | - Md Shofiqul Islam
- Institute for Intelligent Systems Research and Innovation (IISRI), Deakin University, 75 Pigdons Rd, Warunponds, Victoria 3216, Australia
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O Box 2457, Riyadh 11451, Saudi Arabia
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushtia 7003, Bangladesh
- Center for Advanced Bioinformatics and Artificial Intelligence Research, Islamic University, Kushtia 7003, Bangladesh
| |
Collapse
|
18
|
Teklemariam AB, Muche ZT, Agidew MM, Mulu AT, Zewde EA, Baye ND, Adugna DG, Maru L, Ayele TM. Receptor tyrosine kinases and steroid hormone receptors in breast cancer: Review of recent evidences. Metabol Open 2024; 24:100324. [PMID: 39493231 PMCID: PMC11530601 DOI: 10.1016/j.metop.2024.100324] [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: 07/15/2024] [Revised: 10/09/2024] [Accepted: 10/12/2024] [Indexed: 11/05/2024] Open
Abstract
Breast cancer development and progression are driven by intricate networks involving receptor tyrosine kinases (RTKs) and steroid hormone receptors specifically estrogen receptor (ER) and progesterone receptor (PR). This review examined roles of each receptor under normal physiology and in breast cancer, and explored their multifaceted interactions via signaling pathways, focusing on their contributions to breast cancer progression. Since defining the mechanism by which these two-receptor mediated signaling pathways cooperate is essential for understanding breast cancer progression, we discussed the mechanisms of cross-talk between RTKs and ER and PR and their potential therapeutic implications as well. The crosstalk between RTKs and steroid hormone receptors (ER and PR) in breast cancer can influence the disease's progression and treatment outcomes. Therefore, understanding the functions of the aforementioned receptors and their interactions is crucial for developing effective therapies.
Collapse
Affiliation(s)
| | - Zelalem Tilahun Muche
- Department of Medical Physiology, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Melaku Mekonnen Agidew
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Anemut Tilahun Mulu
- Department of Medical Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Edgeit Abebe Zewde
- Department of Medical Physiology, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Nega Dagnew Baye
- Department of Human Anatomy, School of Medicine, College of Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Dagnew Getnet Adugna
- Department of Human Anatomy, School of Medicine, College of Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Lemlemu Maru
- Department of Medical Physiology, School of Medicine, College of Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Teklie Mengie Ayele
- Department of Pharmacy, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| |
Collapse
|
19
|
Lu X, Zhang W, Yang X, Yan X, Hussain Z, Wu Q, Zhao J, Yuan B, Yao K, Dong Z, Liu K, Jiang Y. Dronedarone hydrochloride inhibits gastric cancer proliferation in vitro and in vivo by targeting SRC. Transl Oncol 2024; 50:102136. [PMID: 39369581 PMCID: PMC11491965 DOI: 10.1016/j.tranon.2024.102136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a significant global concern, ranking as the fifth most prevalent cancer. Unfortunately, the five-year survival rate is less than 30 %. Additionally, approximately 50 % of patients experience a recurrence or metastasis. As a result, finding new drugs to prevent relapse is of utmost importance. METHODS The inhibitory effect of Dronedarone hydrochloride (DH) on gastric cancer cells was examined using proliferation assays and anchorage-dependent assays. The binding of DH with SRC was detected by molecular docking, pull-down assays, and cellular thermal shift assays (CETSA). DH's inhibition of Src kinase activity was confirmed through in vitro kinase assays. The SRC knockout cells, established using the CRISPR-Cas9 system, were used to verify Src's role in GC cell proliferation. Patient-derived xenograft (PDX) models were employed to elucidate that DH suppressed tumor growth in vivo. RESULTS Our research discovered DH inhibited GC cell proliferation in vitro and in vivo. DH bound to the SRC protein to inhibit the SRC/AKT1 signaling pathway in gastric cancer. Additionally, we observed a decrease in the sensitivity of gastric cancer cells to DH upon down-regulation of SRC. Notably, we demonstrated DH's anti-tumor effects were similar to those of Dasatinib, a well-known SRC inhibitor, in GC patient-derived xenograft models. CONCLUSION Our research has revealed that Dronedarone hydrochloride, an FDA-approved drug, is an SRC inhibitor that can suppress the growth of GC cells by blocking the SRC/AKT1 signaling pathway. It provides a scientific basis for use in the clinical treatment of GC.
Collapse
Affiliation(s)
- Xuebo Lu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Weizhe Zhang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Xiaoxiao Yang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Xiao Yan
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Zubair Hussain
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Qiong Wu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Jinmin Zhao
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Baoyin Yuan
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Ke Yao
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Zigang Dong
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, Henan, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, Henan, China
| | - Kangdong Liu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, Henan, China; Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University 450000, Zhengzhou, Henan, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, Henan, China
| | - Yanan Jiang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, Henan, China; Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University 450000, Zhengzhou, Henan, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, Henan, China.
| |
Collapse
|
20
|
Dilber Y, Çeker HT, Öztüzün A, Çırçırlı B, Kırımlıoğlu E, Barut Z, Aslan M. Sparstolonin B Reduces Estrogen-Dependent Proliferation in Cancer Cells: Possible Role of Ceramide and PI3K/AKT/mTOR Inhibition. Pharmaceuticals (Basel) 2024; 17:1564. [PMID: 39770406 PMCID: PMC11677571 DOI: 10.3390/ph17121564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background: The aim of this study was to determine the effect of Sparstolonin B (SsnB) on cell proliferation and apoptosis in human breast cancer (MCF-7) and human ovarian epithelial cancer (OVCAR-3) cell lines in the presence and absence of estradiol hemihydrate (ES). Phosphoinositol-3 kinase (PI3K), phosphorylated protein kinase B alpha (p-AKT), phosphorylated mTOR (mechanistic target of rapamycin) signaling proteins, and sphingomyelin/ceramide metabolites were also measured within the scope of the study. Methods: The anti-proliferative effects of SsnB therapy were evaluated over a range of times and concentrations. Cell proliferation was determined by measuring the Proliferating Cell Nuclear Antigen (PCNA). PCNA was quantified by ELISA and cell distribution was assessed by immunofluorescence microscopy. MTT analysis was used to test the vitality of the cells, while LC-MS/MS was used to analyze the amounts of ceramides (CERs), sphingosine-1-phosphate (S1P), and sphingomyelins (SMs). TUNEL labeling was used to assess apoptosis, while immunofluorescence staining and enzyme-linked immunosorbent assay (ELISA) were used to measure the levels of PI3K, p-AKT, and p-mTOR proteins. Results: Sparstolonin B administration significantly decreased cell viability in MCF-7 and OVCAR-3 cells both in the presence and absence of ES, while it did not cause toxicity in healthy human fibroblasts. In comparison to controls, cancer cells treated with SsnB showed a significant drop in the levels of S1P, PI3K, p-AKT, and p-mTOR. In cancer cells cultured with SsnB, a significant increase in intracellular concentrations of C16-C24 CERs and apoptosis was observed. Conclusions: SsnB downregulated the levels of S1P, PI3K, p-AKT, and p-mTOR while reducing cell proliferation and promoting ceramide buildup and apoptosis.
Collapse
Affiliation(s)
- Yağmur Dilber
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey; (Y.D.); (H.T.Ç.); (A.Ö.)
| | - Hanife Tuğçe Çeker
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey; (Y.D.); (H.T.Ç.); (A.Ö.)
| | - Aleyna Öztüzün
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey; (Y.D.); (H.T.Ç.); (A.Ö.)
| | - Bürke Çırçırlı
- Department of Medical Biotechnology, Institute of Health Sciences, Akdeniz University, Antalya 07070, Turkey;
| | - Esma Kırımlıoğlu
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey;
| | - Zerrin Barut
- Faculty of Dentistry, Antalya Bilim University, Antalya 07070, Turkey;
| | - Mutay Aslan
- Department of Medical Biochemistry, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey; (Y.D.); (H.T.Ç.); (A.Ö.)
- Department of Medical Biotechnology, Institute of Health Sciences, Akdeniz University, Antalya 07070, Turkey;
| |
Collapse
|
21
|
Shu X, Gao J, Xu H, Li Q, Gong Y, Li J. The Effects of Dexamethasone on Human Lens Epithelial Cells and the Analysis of Related Pathways with Transcriptome Sequencing. FRONT BIOSCI-LANDMRK 2024; 29:391. [PMID: 39614453 DOI: 10.31083/j.fbl2911391] [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: 08/06/2024] [Revised: 10/16/2024] [Accepted: 10/28/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND The goal of this study was to investigate the effects of dexamethasone on human lens epithelial cells (HLECs) and the potential mechanisms. METHODS HLECs (HLE-B3) were cultured in vitro to assess the effects of dexamethasone on cell size at different concentrations. Immunofluorescence staining was used to detect specific protein expression in HLE-B3 cells. The cell size was observed using phase-contrast microscopy, and the length and area were quantitatively measured with ImageJ software for statistical analysis. Flow cytometry was used to verify these outcomes. The means of three groups were statistically analyzed using one-way analysis of variance, whereas the means of two groups were statistically analyzed with the parametric Student's t-test. Additionally, high-throughput transcriptome sequencing was performed to compare messenger RNA (mRNA) expression levels between different concentrations of dexamethasone treatment groups and the control group, to identify potential signaling pathways. Subsequently, we performed quantitative Polymerase Chain Reaction (qPCR), immunofluorescence staining, and molecular docking experiments on the key differentially expressed genes. RESULTS Dexamethasone affected the size of HLE-B3 cells. Both 0.25 and 0.5 μmol/L dexamethasone increased cell length and area, exhibiting no significant difference between the two treatment groups. Flow cytometry showed that dexamethasone increased cell size and granularity, with 0.25 μmol/L dexamethasone leading to larger cell areas and higher intracellular granularity. High-throughput transcriptome sequencing revealed significant upregulation of lysophosphatidic acid receptor 1 (LPAR1) and the pathways related to the glucocorticoid (GC) receptor. CONCLUSIONS Certain concentrations of dexamethasone impact the morphology and biological functions of HLECs. As a subtype of G protein-coupled receptors, LPAR1 on the cell membrane may interact with dexamethasone, affecting cell size and inhibiting autophagy via the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway. These discoveries offer crucial biological insights into how dexamethasone influences the morphology and function of HLECs and the pathogenesis of GC-induced cataracts, offering potential molecular targets for future therapeutic strategies.
Collapse
Affiliation(s)
- Xinjie Shu
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, 401331 Chongqing, China
| | - Jiamin Gao
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, 401331 Chongqing, China
- Department of Ophthalmology and Optometry, Chongqing Medical University, 401331 Chongqing, China
| | - Han Xu
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, 401331 Chongqing, China
- Department of Ophthalmology and Optometry, Chongqing Medical University, 401331 Chongqing, China
| | - Qiyou Li
- Department of Ophthalmology, Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), 400038 Chongqing, China
| | - Yu Gong
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, 401331 Chongqing, China
| | - Jiawen Li
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, 401331 Chongqing, China
- Department of Ophthalmology and Optometry, Chongqing Medical University, 401331 Chongqing, China
| |
Collapse
|
22
|
Ding X, Liang J, Sharko AC, Hilimire TA, Li J, Loskutov J, Mack ZT, Ji H, Schools GP, Cai C, Pugacheva EN, Chen M, Roninson IB, Broude EV. Mediator kinase inhibitors suppress triple-negative breast cancer growth and extend tumor suppression by mTOR and AKT inhibitors. Proc Natl Acad Sci U S A 2024; 121:e2414501121. [PMID: 39541354 PMCID: PMC11588072 DOI: 10.1073/pnas.2414501121] [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: 07/18/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Triple-negative breast cancers (TNBC) are treated primarily by chemotherapy and lack clinically validated therapeutic targets. In particular, inhibitors of the PI3K/AKT/mTOR pathway, abnormally activated in many breast cancers, failed to achieve clinical efficacy in TNBC due to the development of adaptive drug resistance, which is largely driven by the transcriptomic plasticity of TNBC. Expression of CDK8/19 Mediator kinases that control transcriptional reprogramming correlates with relapse-free survival and treatment failure in breast cancer patients, including TNBC. We now investigated how CDK8/19 inhibitors affect the growth of TNBC tumors and their response to mTOR and AKT inhibitors. In contrast to the effects of most anticancer drugs, all the tested human TNBC models (including patient-derived xenografts) responded to CDK8/19 inhibitors in vivo even when they did not respond in vitro. Furthermore, CDK8/19 inhibition extended the host survival of established lung metastases in a murine TNBC model, where the primary tumors were not significantly affected. CDK8/19 inhibitors synergized with an mTORC1 inhibitor everolimus and a pan-AKT inhibitor capivasertib in vitro and strongly potentiated these drugs in long-term in vivo studies. Transcriptomic analysis of tumors that responded or became adapted to everolimus revealed that drug adaptation in vivo was associated with major transcriptional changes in both tumor and stromal cells. Combining everolimus with a CDK8/19 inhibitor counteracted many of these changes and induced combination-specific effects on the expression of multiple genes that affect tumor growth. These results warrant the exploration of CDK8/19 Mediator kinase inhibitors as a new type of drugs for TNBC therapy.
Collapse
Affiliation(s)
- Xiaokai Ding
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| | - Jiaxin Liang
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| | - Amanda C. Sharko
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| | - Thomas A. Hilimire
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
- Senex Biotechnology, Inc., Columbia, SC29208
| | - Jing Li
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| | - Jürgen Loskutov
- Department of Biochemistry and Molecular Medicine, West Virginia University Cancer Institute, School of Medicine, Morgantown, WV26506
| | - Zachary T. Mack
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| | - Hao Ji
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| | - Gary P. Schools
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| | - Chao Cai
- Department of Clinical Pharmacy and Outcomes Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| | - Elena N. Pugacheva
- Department of Biochemistry and Molecular Medicine, West Virginia University Cancer Institute, School of Medicine, Morgantown, WV26506
| | - Mengqian Chen
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
- Senex Biotechnology, Inc., Columbia, SC29208
| | - Igor B. Roninson
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| | - Eugenia V. Broude
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC29208
| |
Collapse
|
23
|
Zhang Z, Liu Y, Wu H, Yuan Y, Liu Z, Sulaiman M, Yuan S, Yang M. Netupitant Inhibits the Proliferation of Breast Cancer Cells by Targeting AGK. Cancers (Basel) 2024; 16:3807. [PMID: 39594764 PMCID: PMC11592365 DOI: 10.3390/cancers16223807] [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: 09/20/2024] [Revised: 10/31/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Currently, there is a significant lack of effective pharmacological agents for the treatment of breast cancer. Acylglycerol Kinase (AGK), a lipid kinase, has been found to be aberrantly expressed in breast cancer and is closely associated with tumor proliferation, migration, and invasion. However, no clinical anti-tumor drugs specifically targeting this kinase have been developed. Methods: siRNA was utilized to knock down the AGK gene; CCK8 and colony formation assays were employed to evaluate the in vitro proliferative capacity of tumor cells. Molecular dynamics simulations and BIL assays were conducted to analyze drug binding affinity. Annexin V/PI staining was used to assess apoptotic phenomena; subcutaneous xenograft tumor experiments in nude mice were performed to confirm the in vivo anti-tumor efficacy of the drug. Results: Netupitant exhibited stable binding affinity for AGK and interacted with amino acids within the ATP-binding region of the enzyme. The IC50 values for the SK-BR-3 and MDA-MB-231 cell lines were determined as 16.15 ± 4.25 µmol/L and 24.02 ± 4.19 µmol/L, respectively; at a concentration of 2.5 µmol/L, Netupitant effectively inhibited clonogenic capacity in breast cancer cells; furthermore, treatment with 10 µmol/L significantly induced apoptosis in these cells. Doses of 50 mg/kg and 100 mg/kg Netupitant markedly suppressed growth rates of subcutaneous xenograft tumors in nude mice while also promoting apoptotic processes. Both in vivo and in vitro studies indicated that Netupitant could inhibit the activation of the PI3K/AKT/mTOR signaling pathway. Conclusions: By targeting AGK, Netupitant inhibits its kinase activity, which leads to reduced phosphorylation levels of PTEN, thereby suppressing the activation of the PI3K/AKT/mTOR signaling pathway and ultimately resulting in apoptosis in breast cancer cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shengtao Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (Y.L.); (H.W.); (M.S.)
| | - Mei Yang
- New Drug Screening and Pharmacodynamics Evaluation Center, National Key Laboratory for Multi-Target Natural Drugs, China Pharmaceutical University, Nanjing 210009, China; (Z.Z.); (Y.L.); (H.W.); (M.S.)
| |
Collapse
|
24
|
Poustforoosh A. Scaffold Hopping Method for Design and Development of Potential Allosteric AKT Inhibitors. Mol Biotechnol 2024:10.1007/s12033-024-01307-2. [PMID: 39463205 DOI: 10.1007/s12033-024-01307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
Targeting AKT is a practical strategy for cancer therapy in many cancer types. Targeted inhibitors of AKT are attractive solutions for inhibiting the interconnected signaling pathways, like PI3K/Akt/mTOR. Allosteric inhibitors are more desirable among different classes of AKT inhibitors as they could be more specific with fewer off-target proteins. In this study, a ligand/structure-based pipeline was developed to design new allosteric AKT inhibitors by employing the core hopping method. Triciribine, a traditional allosteric AKT inhibitor was used as the template, and the FDA-approved kinase inhibitors for cancer treatment were considered as the cores. The allosteric site in the crystal structure of AKT1 was used to screen the designed compounds. The results were further evaluated using molecular docking, ADME/T analysis, molecular dynamics (MD) simulation, and binding free energy calculations. The outcomes introduced 24 newly designed inhibitors, amongst which three compounds C6, C20, and C16 showed remarkable binding affinity to AKT1. While the docking scores for triciribine was around - 8.6 kcal/mol, the docking scores of these compounds were about - 11 to - 13 kcal/mol. The MD results indicated that designed compounds target the essential residues of the PH domain and kinase domain of AKT, such as Trp80, Thr211, Tyr272, Asp274, and Asp292. Scaffold hopping is a tremendous tool for designing novel anti-cancer agents by improving already known and potential drug compounds. The designed compounds are worth to be examined by experimental investigation in vitro and in vivo.
Collapse
Affiliation(s)
- Alireza Poustforoosh
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
25
|
Altundag K. PI3K/AKT/mTOR inhibitors for the management of triple-negative breast cancer. Med Oncol 2024; 41:279. [PMID: 39400749 DOI: 10.1007/s12032-024-02515-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/19/2024] [Indexed: 10/15/2024]
Affiliation(s)
- Kadri Altundag
- MKA Breast Cancer Clinic, Tepe Prime, Cankaya, 06800, Ankara, Turkey.
| |
Collapse
|
26
|
Altundag K. Clinical research on triple-negative breast cancer that targets the PIK3CA pathway. Med Oncol 2024; 41:268. [PMID: 39400773 DOI: 10.1007/s12032-024-02493-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024]
Affiliation(s)
- Kadri Altundag
- MKA Breast Cancer Clinic, Tepe Prime, Cankaya, 06800, Ankara, Turkey.
| |
Collapse
|
27
|
Seifati SM, Zare F, Bafghi SAM, Hadinedoushan H. Impact of anti leukemia inhibitory factor antibody on immune related gene expression in breast cancer Balb/c mouse model. Sci Rep 2024; 14:20403. [PMID: 39223212 PMCID: PMC11369080 DOI: 10.1038/s41598-024-71014-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Leukemia inhibitory factor (LIF) is involved in the progression of different cancers. In this study, we investigated the effect of anti-LIF antibodies on immune-related gene expression in the Balb/c mouse model of breast cancer. To immunize mice against LIF, recombinant LIF with Freund adjuvant was injected into the test group, whereas the control group received phosphate-buffered saline with adjuvant. Tumor induction (4T1 cell line) was performed by increasing the antibody titer. The expression of immune-related genes was evaluated by real-time PCR. The anti-LIF titer was significantly increased in the immunized group. The expression of genes related to the differentiation of T helper (Th)-1, Th-2, and Th-17 cells was significantly higher in the immunized group than in the control group. In addition, anti-LIF did not have a significant effect on the expression of genes related to the differentiation of regulatory T cells, and immune checkpoint-associated genes. Additionally, the test group had higher survival and lower tumor development rates. The results demonstrated that the anti-LIF antibody may potentially play a role in the differentiation of immune cells or immune responses. However, further studies utilizing advanced techniques are necessary to validate its function.
Collapse
Affiliation(s)
- Seyed Mohammad Seifati
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Postal Code: 8916181635, Iran
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Zare
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Postal Code: 8916181635, Iran
| | | | - Hossein Hadinedoushan
- Reproductive Immunology Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Postal Code: 8916181635, Iran.
- Department of Immunology, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
28
|
Nava-Tapia DA, Román-Justo NY, Cuenca-Rojo A, Guerrero-Rivera LG, Patrón-Guerrero A, Poblete-Cruz RI, Zacapala-Gómez AE, Sotelo-Leyva C, Navarro-Tito N, Mendoza-Catalán MA. Exploring the potential of tocopherols: mechanisms of action and perspectives in the prevention and treatment of breast cancer. Med Oncol 2024; 41:208. [PMID: 39060448 DOI: 10.1007/s12032-024-02454-x] [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: 05/05/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Currently, breast cancer is the most common cause of mortality caused by neoplasia in women worldwide. The unmet challenges of conventional cancer therapy are chemoresistance and lack of selectivity, which can lead to serious side effects in patients; therefore, new treatments based on natural compounds that serve as adjuvants in breast cancer therapy are urgently needed. Tocopherols are naturally occurring antioxidant compounds that have shown antitumor activity against several types of cancer, including breast cancer. This review summarizes the antitumoral activity of tocopherols, such as the antiproliferative, apoptotic, anti-invasive, and antioxidant effects of tocopherols, through different molecular mechanisms. According to the studies described, α-T, δ-T and γ-T are the most studied in breast tumor cells; however, α-T and γ-T show a more critical antitumor activity and significant potential as a complements to chemotherapeutic drugs against breast cancer, enhancing toxicity against tumor cells and preventing cytotoxicity in nontumor cells. However, the possible relationship between tocopherol intake, related to concentration, and the promotion of cancer in particular cases should not be ruled out, so additional studies are required to determine the correct dose to obtain the desired antitumor effect. Moreover, nanomicelles of D-α-tocopherol have promising potential as pharmaceutical excipients for drug delivery to improve the cytotoxicity and selectivity of first-line chemotherapeutics against breast cancer.
Collapse
Affiliation(s)
- Dania A Nava-Tapia
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Norely Y Román-Justo
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Antonio Cuenca-Rojo
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Lizeth G Guerrero-Rivera
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Annet Patrón-Guerrero
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Ruth I Poblete-Cruz
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Ana E Zacapala-Gómez
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - César Sotelo-Leyva
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico
| | - Napoleón Navarro-Tito
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico.
| | - Miguel A Mendoza-Catalán
- Facultad de Ciencias Químico Biológicas, Autonomous University of Guerrero, Av. Lázaro Cárdenas S/N., 39090, Chilpancingo, Guerrero, Mexico.
| |
Collapse
|
29
|
Reddi KK, Chava S, Chabattula SC, Edwards YJK, Singh K, Gupta R. ASAH1 facilitates TNBC by DUSP5 suppression-driven activation of MAP kinase pathway and represents a therapeutic vulnerability. Cell Death Dis 2024; 15:452. [PMID: 38926346 PMCID: PMC11208621 DOI: 10.1038/s41419-024-06831-2] [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: 12/28/2023] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is prone to metastasis and therapy resistance. Owing to its aggressive nature and limited availability of targeted therapies, TNBC is associated with higher mortality as compared to other forms of breast cancer. In order to develop new therapeutic options for TNBC, we characterized the factors involved in TNBC growth and progression. Here, we demonstrate that N-acylsphingosine amidohydrolase 1 (ASAH1) is overexpressed in TNBC cells and is regulated via p53 and PI3K-AKT signaling pathways. Genetic knockdown or pharmacological inhibition of ASAH1 suppresses TNBC growth and progression. Mechanistically, ASAH1 inhibition stimulates dual-specificity phosphatase 5 (DUSP5) expression, suppressing the mitogen-activated protein kinase (MAPK) pathway. Furthermore, pharmacological cotargeting of the ASAH1 and MAPK pathways inhibits TNBC growth. Collectively, we unmasked a novel role of ASAH1 in driving TNBC and identified dual targeting of the ASAH1 and MAPK pathways as a potential new therapeutic approach for TNBC treatment.
Collapse
Affiliation(s)
- Kiran Kumar Reddi
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suresh Chava
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Siva Chander Chabattula
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yvonne J K Edwards
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kamaljeet Singh
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center at The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
30
|
Marafie SK, Al-Mulla F, Abubaker J. mTOR: Its Critical Role in Metabolic Diseases, Cancer, and the Aging Process. Int J Mol Sci 2024; 25:6141. [PMID: 38892329 PMCID: PMC11173325 DOI: 10.3390/ijms25116141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/21/2024] Open
Abstract
The mammalian target of rapamycin (mTOR) is a pivotal regulator, integrating diverse environmental signals to control fundamental cellular functions, such as protein synthesis, cell growth, survival, and apoptosis. Embedded in a complex network of signaling pathways, mTOR dysregulation is implicated in the onset and progression of a range of human diseases, including metabolic disorders such as diabetes and cardiovascular diseases, as well as various cancers. mTOR also has a notable role in aging. Given its extensive biological impact, mTOR signaling is a prime therapeutic target for addressing these complex conditions. The development of mTOR inhibitors has proven advantageous in numerous research domains. This review delves into the significance of mTOR signaling, highlighting the critical components of this intricate network that contribute to disease. Additionally, it addresses the latest findings on mTOR inhibitors and their clinical implications. The review also emphasizes the importance of developing more effective next-generation mTOR inhibitors with dual functions to efficiently target the mTOR pathways. A comprehensive understanding of mTOR signaling will enable the development of effective therapeutic strategies for managing diseases associated with mTOR dysregulation.
Collapse
Affiliation(s)
- Sulaiman K. Marafie
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Department of Translational Research, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait;
| | - Jehad Abubaker
- Biochemistry and Molecular Biology Department, Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait
| |
Collapse
|