1
|
Yang Y, Sun X, Luo L, Peng R, Yang R, Cheng Z, Lv Y, Li H, Tang Q, Zhu W, Qiao D, Xu S. Discovery of novel potent PI3K/mTOR dual-target inhibitors based on scaffold hopping: Design, synthesis, and antiproliferative activity. Arch Pharm (Weinheim) 2023; 356:e2300403. [PMID: 37840368 DOI: 10.1002/ardp.202300403] [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: 07/24/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/17/2023]
Abstract
The PI3K/AKT/mTOR pathway is one of the most common dysregulated signaling cascade responses in human cancers, playing a crucial role in cell proliferation and angiogenesis. Therefore, the development of anticancer drugs targeting the PI3K and mTOR pathways has become a research hotspot in cancer treatment. In this study, the PI3K selective inhibitor GDC-0941 was selected as a lead compound, and 28 thiophenyl-triazine derivatives with aromatic urea structures were synthesized based on scaffold hopping, serving as a novel class of PI3K/mTOR dual inhibitors. The most promising compound Y-2 was obtained through antiproliferative activity evaluation, kinase inhibition, and toxicity assays. The results showed that Y-2 demonstrated potential inhibitory effects on both PI3K kinase and mTOR kinase, with IC50 values of 171.4 and 10.2 nM, respectively. The inhibitory effect of Y-2 on mTOR kinase was 52 times greater than that of the positive drug GDC-0941. Subsequently, the antitumor activity of Y-2 was verified through pharmacological experiments such as AO staining, cell apoptosis, scratch assays, and cell colony formation. The antitumor mechanism of Y-2 was further investigated through JC-1 experiments, real-time quantitative PCR, and Western blot analysis. Based on the above experiments, Y-2 can be identified as a potent PI3K/mTOR dual inhibitor for cancer treatment.
Collapse
Affiliation(s)
- Yang Yang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Xin Sun
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Leixuan Luo
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Rujue Peng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Ruiqing Yang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Zhenjie Cheng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Yao Lv
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Hongfeng Li
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Qidong Tang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Dan Qiao
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, China
| |
Collapse
|
2
|
Luo L, Sun X, Yang Y, Xia L, Wang S, Fu Y, Zhu Y, Xu S, Zhu W. A Novel Dual PI3K/mTOR Inhibitor, XIN-10, for the Treatment of Cancer. Int J Mol Sci 2023; 24:14821. [PMID: 37834269 PMCID: PMC10573424 DOI: 10.3390/ijms241914821] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
An imbalance in PI3K/AKT/mTOR pathway signaling in humans often leads to cancer. Therefore, the investigation of anti-cancer medications that inhibit PI3K and mTOR has emerged as a significant area of research. The aim of this study was to explore the effect of XIN-10, a dual PI3K/mTOR inhibitor, on the growth as well as antiproliferation of tumor cells and to investigate the anti-tumor mechanism of XIN-10 by further exploration. We screened three cell lines for more in-depth exploration by MTT experiments. From the AO staining, cell cycle and apoptosis, we found that XIN-10 had a more obvious inhibitory effect on the MCF-7 breast cancer cell line and used this as a selection for more in-depth experiments. A series of in vitro and in vivo experiments showed that XIN-10 has superior antiproliferative activity compared with the positive drug GDC-0941. Meanwhile, through the results of protein blotting and PCR experiments, we concluded that XIN-10 can block the activation of the downstream pathway of mTOR by inhibiting the phosphorylation of AKT(S473) as well as having significant inhibitory effects on the gene exons of PI3K and mTOR. These results indicate that XIN-10 is a highly potent inhibitor with low toxicity and has a strong potential to be developed as a novel PI3Kα/mTOR dual inhibitor candidate for the treatment of positive breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shan Xu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang 330013, China; (L.L.); (X.S.); (Y.Y.); (L.X.); (S.W.); (Y.F.); (Y.Z.)
| | - Wufu Zhu
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, School of Pharmacy, Jiangxi Science & Technology Normal University, 605 Fenglin Road, Nanchang 330013, China; (L.L.); (X.S.); (Y.Y.); (L.X.); (S.W.); (Y.F.); (Y.Z.)
| |
Collapse
|
3
|
Xiao Y, Yu Y, Hu L, Yang Y, Yuan Y, Zhang W, Luo J, Yu L. Matrine Alleviates Sepsis-Induced Myocardial Injury by Inhibiting Ferroptosis and Apoptosis. Inflammation 2023; 46:1684-1696. [PMID: 37219694 DOI: 10.1007/s10753-023-01833-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/29/2023] [Accepted: 05/06/2023] [Indexed: 05/24/2023]
Abstract
Matrine is a Sophora alkaloid that exerts antitumor effects on a variety of diseases, but few studies have investigated the role of matrine in sepsis-induced myocardial injury. In the present study, we investigated the effects of matrine on septic myocardial injury and the potential mechanisms. Network pharmacology approaches were used to predict the targets of matrine in the treatment of sepsis-induced myocardial injury. A mouse sepsis-induced myocardial injury model was established to determine the effect of matrine. Mouse cardiac function was evaluated by ultrasonography, and cardiac morphology and cardiomyocyte apoptosis were evaluated by HE and TUNEL staining. Oxidative stress was assessed by measuring ROS levels and MDA and SOD activity. Bax, Bcl2, GPX4, ACSL4, PI3K, and AKT protein levels were evaluated by immunohistochemical staining and western blotting. Bioinformatics analysis identified that the potential therapeutic effect of matrine on sepsis-induced myocardial injury is closely related to ferroptosis and apoptosis regulation and showed significant involvement of the PI3K/AKT signaling pathway. In vivo, the matrine group showed improved myocardial function, morphology, and apoptosis ratio and alleviated oxidative stress compared with the LPS group, whereas 25 mg/kg matrine exerted the optimal inhibitory effect. Matrine alleviated LPS-induced cardiomyocyte ferroptosis and apoptosis, resulting in upregulation of Bax/Bcl2 and GPX4 expression and downregulation of ferroptosis marker protein (ACSL4) expression, as shown by immunohistochemistry and western blotting. Moreover, matrine increased PI3K/AKT pathway-related molecule expression and thus modulated ferroptosis and apoptosis. Matrine regulates PI3K/AKT pathway activity to inhibit apoptosis and ferroptosis and thereby alleviates sepsis-induced myocardial injury.
Collapse
Affiliation(s)
- Yuhong Xiao
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yun Yu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Longlong Hu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yuhui Yang
- HuanKui Academy of Nanchang University, Nanchang, Jiangxi, China
| | - Ye Yuan
- HuanKui Academy of Nanchang University, Nanchang, Jiangxi, China
| | - Wenjun Zhang
- Department of Rehabilitation Medicine, Ganzhou Hospital of Nanchang University, Ganzhou, Jiangxi, China
| | - Jun Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Lingling Yu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
4
|
Corr B, Cosgrove C, Spinosa D, Guntupalli S. Endometrial cancer: molecular classification and future treatments. BMJ MEDICINE 2022; 1:e000152. [PMID: 36936577 PMCID: PMC9978763 DOI: 10.1136/bmjmed-2022-000152] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 09/15/2022] [Indexed: 12/31/2022]
Abstract
The treatment for endometrial cancer is rapidly evolving with the development of molecular analysis and novel strategies. Surgical resection, cytotoxic chemotherapy, endocrine or hormonal treatment, and radiation have been the staples of treatment for decades. However, precision based approaches for tumours are rapidly becoming a part of these strategies. Biomarker driven treatments are now a part of primary and recurrent treatment algorithms. This review aims to describe the current state of molecular analysis and treatment for endometrial cancer as well as to elucidate potential approaches for the near future.
Collapse
|
5
|
Smith PG, Roque D, Ching MM, Fulton A, Rao G, Reader JC. The Role of Eicosanoids in Gynecological Malignancies. Front Pharmacol 2020; 11:1233. [PMID: 32982722 PMCID: PMC7479818 DOI: 10.3389/fphar.2020.01233] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
Eicosanoids, bio-active lipid molecules, evoke a multitude of biological effects that directly affect cancer cells and indirectly affect tumor microenvironment. An emerging role has been shown for eicosanoids in the pathogenesis of gynecological malignancies which include cancers of the vulva, vagina, cervix, uterine, and ovary. Eicosanoid biosynthesis pathways start at the metabolism of phospholipids by phospholipase A2 then proceeding to one of three pathways: the cyclooxygenase (COX), lipoxygenase (LOX), or P450 epoxygenase pathways. The most studied eicosanoid pathways include COX and LOX; however, more evidence is appearing to support further study of the P450 epoxygenase pathway in gynecologic cancers. In this review, we present the current knowledge of the role of COX, LOX and P450 pathways in the pathogenesis of gynecologic malignancies. Vulvar and vaginal cancer, the rarest subtypes, there is association of COX-2 expression with poor disease specific survival in vulvar cancer and, in vaginal cancer, COX-2 expression has been found to play a role in mucosal inflammation leading to disease susceptibility and transmission. Cervical cancer is associated with COX-2 levels 7.4 times higher than in healthy tissues. Additionally, HPV elevates COX-2 levels through the EGFR pathway and HIV promotes elevated COX-2 levels in cervical tissue as well as increases PGE2 levels eliciting inflammation and progression of cancer. Evidence supports significant roles for both the LOX and COX pathways in uterine cancer. In endometrial cancer, there is increased expression of 5-LOX which is associated with adverse outcomes. Prostanoids in the COX pathway PGE2 and PGF2α have been shown to play a significant role in uterine cancer including alteration of proliferation, adhesion, migration, invasion, angiogenesis, and the inflammatory microenvironment. The most studied gynecological malignancy in regard to the potential role of eicosanoids in tumorigenesis is ovarian cancer in which all three pathways have shown to be associated or play a role in ovarian tumorigenesis directly on the tumor cell or through modulation of the tumor microenvironment. By identifying the gaps in knowledge, additional pathways and targets could be identified in order to obtain a better understanding of eicosanoid signaling in gynecological malignancies and identify potential new therapeutic approaches.
Collapse
Affiliation(s)
- Paige G. Smith
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Dana Roque
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Mc Millan Ching
- Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Amy Fulton
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, United States
- Baltimore Veterans Administration Medical Center, Baltimore, MD, United States
| | - Gautam Rao
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Jocelyn C. Reader
- Department of Obstetrics, Gynecology and Reproductive Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
6
|
Significance of EGFR/HER2 Expression and PIK3CA Mutations in Giant Cell Tumour of Bone Development. BIOMED RESEARCH INTERNATIONAL 2020. [DOI: 10.1155/2020/2931784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Giant Cell Tumour of Bone (GCTB) is a rare bone tumour. Locally aggressive and recurrent, it might evolve into pulmonary metastases. Our present work is aimed at investigating the involvement of the epidermal growth factor receptor (ErbB) family and its downstream effectors in the development and recurrence of GCTB. For this purpose, we used a cohort of 32 GCTB patients and we evaluated the clinicohistological features and the expression of RANKL, EGFR, and HER2. The mutation status of KRAS, PI3KCA, and PTEN gene as potential oncogene involved in GCTB was also evaluated. We found a significant correlation between advanced histological stages, overexpression of EGFR/HER2, and tumour recurrence. Moreover, two mutations were found in the PIK3CA gene: a missense mutation, 1634A>C, detected for the first time in GCTB patients, without influencing the stability of the protein, and a frameshift mutation, c.1658_1659delGTinsC, causing the loss of the protein kinase domain. Altogether, these results suggest that overexpression of HER2/EGFR, Campanacci, and histological stages could be used as a novel prognostic marker for GCTB recurrence.
Collapse
|
7
|
ARID1A and PI3-kinase pathway mutations in the endometrium drive epithelial transdifferentiation and collective invasion. Nat Commun 2019; 10:3554. [PMID: 31391455 PMCID: PMC6686004 DOI: 10.1038/s41467-019-11403-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 07/03/2019] [Indexed: 01/06/2023] Open
Abstract
ARID1A and PI3-Kinase (PI3K) pathway alterations are common in neoplasms originating from the uterine endometrium. Here we show that monoallelic loss of ARID1A in the mouse endometrial epithelium is sufficient for vaginal bleeding when combined with PI3K activation. Sorted mutant epithelial cells display gene expression and promoter chromatin signatures associated with epithelial-to-mesenchymal transition (EMT). We further show that ARID1A is bound to promoters with open chromatin, but ARID1A loss leads to increased promoter chromatin accessibility and the expression of EMT genes. PI3K activation partially rescues the mesenchymal phenotypes driven by ARID1A loss through antagonism of ARID1A target gene expression, resulting in partial EMT and invasion. We propose that ARID1A normally maintains endometrial epithelial cell identity by repressing mesenchymal cell fates, and that coexistent ARID1A and PI3K mutations promote epithelial transdifferentiation and collective invasion. Broadly, our findings support a role for collective epithelial invasion in the spread of abnormal endometrial tissue. PIK3CA mutations and ARID1A loss co-exist in endometrial neoplasms. Here, the authors show that these co-mutations drive gene expression profiles correlated with differential chromatin accessibility and ARID1A binding in the endometrial epithelium, resulting in partial EMT and myometrial invasion.
Collapse
|
8
|
Lu X, Zhang L, Zhao H, Chen C, Wang Y, Liu S, Lin X, Wang Y, Zhang Q, Lu T, Yan F. Molecular classification and subtype-specific drug sensitivity research of uterine carcinosarcoma under multi-omics framework. Cancer Biol Ther 2018; 20:227-235. [PMID: 30359167 DOI: 10.1080/15384047.2018.1523853] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Uterine carcinosarcomas (UCSs) are aggressive rare tumors recognized as malignancies composed of metaplastic transformation of epithelial elements. Nay no comprehensive molecular classification has been applied to UCS to guide targeted therapies so far, which motivated us to subtyping UCS by aggregating multiple genomic platform data. METHODS We classified UCS into three distinct subtypes with different clinicopathologic and molecular characterization by using similarity network fusion under consensus clustering framework (SNFCC+) to aggregate four genomic data platforms of 55 UCS patients. Differences across subtypes were extracted by functional enrichment, gene mutations and clinical features. Subtypes were further distinguished by putative biomarkers. We also determined associations between individual oncogenes and chemotherapeutics to discuss subtype-specific drug sensitivity. RESULTS Functional enrichment analysis of the subtype-specific differential expression genes endowed three subtypes new designation: Myo, Cell and Hormone. Mutations in PTEN, PIK3CA, ARID1A and PPP2R1A altered across subtypes. The epithelial-to-mesenchymal transition (EMT) score distinguished Myo from another two subtypes whereby a high EMT scores prevalently existed and each case was judged as M (mesenchymal) phenotype in Myo subtype. Through the drug sensitivity analysis, we found that the response to - tinib drugs is quite different across subtypes according to expression level. Additionally, different subtypes' response to broad-spectrum anti-cancer drug paclitaxel may be also different. CONCLUSIONS In this study, we identified three distinct molecular subtypes of UCS with different features. Subtypes were also revealed to have different sensitivity to existing chemotherapy drugs, which may support in-depth study of subtype-specific dosing regimens.
Collapse
Affiliation(s)
- Xiaofan Lu
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| | - Liya Zhang
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| | - Huiling Zhao
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| | - Chen Chen
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| | - Yaoyan Wang
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| | - Shengjie Liu
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| | - Xiao Lin
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| | - Yue Wang
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| | - Qianyuan Zhang
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| | - Tao Lu
- b State Key Laboratory of Natural Medicine , China Pharmaceutical University , Nanjing , P.R. China
| | - Fangrong Yan
- a Research Center of Biostatistics and Computational Pharmacy , China Pharmaceutical University , Nanjing , P.R.China
| |
Collapse
|
9
|
Ahmed RSI, Liu G, Renzetti A, Farshi P, Yang H, Soave C, Saed G, El-Ghoneimy AA, El-Banna HA, Foldes R, Chan TH, Dou QP. Biological and Mechanistic Characterization of Novel Prodrugs of Green Tea Polyphenol Epigallocatechin Gallate Analogs in Human Leiomyoma Cell Lines. J Cell Biochem 2016; 117:2357-69. [DOI: 10.1002/jcb.25533] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 03/03/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Reda Saber Ibrahim Ahmed
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine; Wayne State University; Detroit Michigan
- Faculty of Veterinary Medicine, Department of Pharmacology; South Valley University; Qena Egypt
| | - Gang Liu
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine; Wayne State University; Detroit Michigan
| | - Andrea Renzetti
- Department of Chemistry; McGill University; Montreal Quebec Canada
| | - Pershang Farshi
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine; Wayne State University; Detroit Michigan
| | - Huanjie Yang
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine; Wayne State University; Detroit Michigan
| | - Claire Soave
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine; Wayne State University; Detroit Michigan
| | - Ghassan Saed
- Departments of Obstetrics & Gynecology and Anatomy & Cell Biology; School of Medicine; Wayne State University; Detroit Michigan
| | | | - Hossny Awad El-Banna
- Faculty of Veterinary Medicine, Department of Pharmacology; Cairo University; Giza Egypt
| | - Robert Foldes
- Viteava Pharmaceuticals Inc.; Toronto Ontario Canada
| | - Tak-Hang Chan
- Department of Chemistry; McGill University; Montreal Quebec Canada
| | - Q. Ping Dou
- Barbara Ann Karmanos Cancer Institute, and Departments of Oncology, Pharmacology and Pathology, School of Medicine; Wayne State University; Detroit Michigan
| |
Collapse
|
10
|
Brany D, Dvorska D, Nachajova M, Slavik P, Burjanivova T. Malignant tumors of the uterine corpus: molecular background of their origin. Tumour Biol 2015; 36:6615-21. [DOI: 10.1007/s13277-015-3824-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/20/2015] [Indexed: 12/21/2022] Open
|
11
|
Treilleux I, Arnedos M, Cropet C, Wang Q, Ferrero JM, Abadie-Lacourtoisie S, Levy C, Legouffe E, Lortholary A, Pujade-Lauraine E, Bourcier AV, Eymard JC, Spaeth D, Bachelot T. Translational studies within the TAMRAD randomized GINECO trial: evidence for mTORC1 activation marker as a predictive factor for everolimus efficacy in advanced breast cancer. Ann Oncol 2015; 26:120-125. [PMID: 25361980 DOI: 10.1093/annonc/mdu497] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Everolimus is an agent frequently associated with specific toxicities. Predictive markers of efficacy are needed to help define which patients could benefit from it. The goal of this exploratory study was to identify potential predictive biomarkers in the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) activation pathway using primary tumor samples collected during the phase II tamoxifen plus everolimus (TAMRAD) trial. PATIENTS AND METHODS Tumor tissues were collected retrospectively from the TAMRAD trial. Immunohistochemistry was carried out using specific antibodies directed toward proteins that result in mTORC1 activation [canonical phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mTOR or alternative pathways]. DNA was extracted from the tumor tissue; mutation screening in the PIK3CA gene (exons 9 and 20) and the KRAS gene (exons 2 and 3) was first carried out using Sanger direct sequencing, and then completed by next-generation sequencing for PIK3CA. An exploratory analysis of everolimus efficacy in terms of a time-to-progression (TTP) increase was carried out in each biomarker subgroup (high versus low expression referring to the median percentage of marked cells). RESULTS A total of 55 primary tumor samples from the TAMRAD trial—25 from the tamoxifen-alone group and 30 from the tamoxifen/everolimus group—were evaluated for biomarkers. The subgroups most likely to have an improvement in TTP with tamoxifen/everolimus therapy, compared with tamoxifen alone, were patients with high p4EBP1, low 4EBP1, low liver kinase B1, low pAkt, and low PI3K. Among the 45 samples screened for mutation status, nine samples (20%; 95% CI 9.6-34.6) had a PIK3CA mutation. KRAS mutation was observed in one patient. CONCLUSIONS A positive correlation between late effectors of mTORC1 activation, a positive correlation between Akt-independent mTORC1 activation, and an inverse correlation between canonical PI3K/Akt/mTOR pathway and everolimus efficacy were observed in this exploratory analysis. However, these correlations need to be validated in larger studies before applying the findings to routine clinical practice.
Collapse
Affiliation(s)
- I Treilleux
- Department of Anatomopathology, Centre Léon Bérard, Lyon
| | - M Arnedos
- Oncology Department, Gustave Roussy, Villejuif
| | - C Cropet
- Biostatistics and Therapeutic Evaluation Unit
| | - Q Wang
- Genomic Platform-Translational Research Laboratory, Centre Léon Bérard, Lyon
| | - J-M Ferrero
- Medical Oncology Department, Centre Antoine Lacassagne, Nice
| | | | - C Levy
- Oncology Department, Centre François Baclesse, Caen
| | - E Legouffe
- Hematology and Oncology Department, Clinique de Valdegour, Nimes
| | - A Lortholary
- Oncology Department, Centre Catherine de Sienne, Nantes
| | - E Pujade-Lauraine
- Oncology Department, Université Paris Descartes, AP-HP, Hôpitaux Universitaires Paris Centre, Site Hôtel-Dieu, Paris
| | - A-V Bourcier
- Hematology and Oncology Department, Centre Hospitalier Départemental Les Oudairies, La Roche-Sur-Yon
| | - J-C Eymard
- Department of Medicine, Institut Jean Godinot, Reims
| | - D Spaeth
- Oncology Department, Centre d'Oncologie de Gentilly, Nancy
| | - T Bachelot
- 2B North Department, Department of Medical Oncology and Cancer Research Center of Lyon, Centre Léon Bérard, Lyon, France.
| |
Collapse
|
12
|
Sun Y, Ding H, Liu X, Li X, Li L. INPP4B overexpression enhances the antitumor efficacy of PARP inhibitor AG014699 in MDA-MB-231 triple-negative breast cancer cells. Tumour Biol 2014; 35:4469-77. [DOI: 10.1007/s13277-013-1589-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Accepted: 12/19/2013] [Indexed: 12/21/2022] Open
|
13
|
Women 50 Years Or Younger With Endometrial Cancer: The Argument for Universal Mismatch Repair Screening and Potential for Targeted Therapeutics. Int J Gynecol Cancer 2013; 23:853-60. [DOI: 10.1097/igc.0b013e31828eed9c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
ObjectiveTo identify young patients with endometrial cancer with potential Lynch-related DNA mismatch repair (MMR) protein expression defects and stathmin (STMN1) expression (part of the phosphoinositol 3-kinase pathway) and to correlate clinical data.MethodsThis retrospective study included women with endometrial cancer who were 50 years or younger at diagnosis. Clinical data were abstracted from chart review. Immunohistochemistry for MMR protein expression, STMN1, and pSTMN1 was performed and univariate analyses performed.ResultsThe mean age of 111 patients was 43 years, and the mean body mass index was 39.6 kg/m2. The majority of the endometrial cancers were endometrioid histology (87.4%), International Federation of Gynecology and Obstetrics stage I (73%) and grade 1 (58.6%). Loss of at least one MMR protein on immunohistochemistry was identified in 26% to 41% of patients depending on stringency. Women with loss of MMR protein expression were compared to women with intact tumor protein expression and were less likely to be stage I (58.6% vs 78.0%; P = 0.043), more likely to have grade 3 tumors (32.1% vs 13.9%; P = 0.034), had larger tumors (6.2 vs 3.7 cm; P < 0.001), had positive lymph nodes more often (24.1% vs 3.7%; P < 0.001), and more often reported a first-degree relative with colon cancer (17.2% vs 1.2%; P < 0.001). There were no significant differences in age, weight, body mass index, medical comorbidities, recurrence, or survival. Women with high STMN1 staining had significantly more grade 3 tumors (56.3% vs 15.8%; P = 0.001), more stage III/IV disease (37.5% vs 15.8%; P = 0.04), had higher mean percentage of myometrial invasion (38.9% vs 16.7%; P = 0.003), and more lymphovascular space invasion (43.8% vs 13.7%; P = 0.004).ConclusionsClinical factors failed to differentiate between patients with intact or missing MMR protein expression, which supports universal screening for Lynch-associated protein defects in young women with endometrial cancer. Additionally, STMN1 staining may identify more aggressive tumors, which might benefit from more aggressive treatments or targeted treatment options.
Collapse
|
14
|
Chen LL, Huang M, Tan JY, Chen XT, Lei LH, Wu YM, Zhang DY. PI3K/AKT pathway involvement in the osteogenic effects of osteoclast culture supernatants on preosteoblast cells. Tissue Eng Part A 2013; 19:2226-32. [PMID: 23617625 DOI: 10.1089/ten.tea.2012.0469] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This study aimed to investigate the ability of osteoclasts during bone resorption activities to regulate the differentiation and calcification of osteoblast precursor cells. The bone resorption model was established using in vitro cortical bone slices and mouse RAW264.7 cells, which were differentiated into osteoclasts by stimulation with the receptor activator of nuclear factor-κB ligand and macrophage colony-stimulating factor. Tartrate-resistant acid phosphatase (TRAP) staining, reverse transcriptase-polymerase chain reaction (RT-PCR), and scanning electron microscopy (SEM) were used to detect osteoclast differentiation. The osteoblast precursor cell line MC3T3-E1 was cultured with the bone resorption supernatant (BRS). Involvement of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway in osteogenesis was evaluated by Western blotting, RT-PCR, and ELISA analysis of markers of the early (runt-related transcription factor-2 and alkaline phosphatase) and late (osteocalcin [OCN]) stages of osteogenesis, and Alizarin Red S staining of matrix mineralization. TRAP staining, RT-PCR, and SEM analysis demonstrated the successful establishment of the bone resorption model. Osteoclast BRS effectively increased the differentiation and calcification of MC3T3-E1 cells. Western blot analysis indicated that the BRS enhanced AKT and p-AKT expression levels in MC3T3-E1 cells. Following AKT2 knockdown and treatment with the PI3K/AKT pathway inhibitor LY294002, the expression of OCN in MC3T3-E1 cells was decreased (p<0.05), as was the calcification area (p<0.05). The data obtained in this study indicated that the osteoclast bone resorption medium promoted the differentiation and calcification of MC3T3-E1 cells and that the PI3K/AKT pathway played a role in this process.
Collapse
Affiliation(s)
- Li-Li Chen
- Department of Oral Medicine, Second Affiliated Hospital, School of Medicine, Zhejiang University, No. 88 Jiefang Road, Hangzhou, China
| | | | | | | | | | | | | |
Collapse
|
15
|
Chordoma characterization of significant changes of the DNA methylation pattern. PLoS One 2013; 8:e56609. [PMID: 23533570 PMCID: PMC3606365 DOI: 10.1371/journal.pone.0056609] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 01/15/2013] [Indexed: 02/07/2023] Open
Abstract
Chordomas are rare mesenchymal tumors occurring exclusively in the midline from clivus to sacrum. Early tumor detection is extremely important as these tumors are resistant to chemotherapy and irradiation. Despite continuous research efforts surgical excision remains the main treatment option. Because of the often challenging anatomic location early detection is important to enable complete tumor resection and to reduce the high incidence of local recurrences. The aim of this study was to explore whether DNA methylation, a well known epigenetic marker, may play a role in chordoma development and if hypermethylation of specific CpG islands may serve as potential biomarkers correlated with SNP analyses in chordoma. The study was performed on tumor samples from ten chordoma patients. We found significant genomic instability by Affymetrix 6.0. It was interesting to see that all chordomas showed a loss of 3q26.32 (PIK 3CA) and 3q27.3 (BCL6) thus underlining the potential importance of the PI3K pathway in chordoma development. By using the AITCpG360 methylation assay we elucidated 20 genes which were hyper/hypomethylated compared to normal blood. The most promising candidates were nine hyper/hypomethylated genes C3, XIST, TACSTD2, FMR1, HIC1, RARB, DLEC1, KL, and RASSF1. In summary, we have shown that chordomas are characterized by a significant genomic instability and furthermore we demonstrated a characteristic DNA methylation pattern. These findings add new insights into chordoma development, diagnosis and potential new treatment options.
Collapse
|
16
|
Current World Literature. Curr Opin Obstet Gynecol 2013; 25:81-9. [DOI: 10.1097/gco.0b013e32835cc6b6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
17
|
Sun Y, Gao C, Luo M, Wang W, Gu C, Zu Y, Li J, Efferth T, Fu Y. Aspidin PB, a phloroglucinol derivative, induces apoptosis in human hepatocarcinoma HepG2 cells by modulating PI3K/Akt/GSK3β pathway. Chem Biol Interact 2013. [DOI: 10.1016/j.cbi.2012.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
18
|
Current world literature. Curr Opin Oncol 2012; 24:587-95. [PMID: 22886074 DOI: 10.1097/cco.0b013e32835793f1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Zhao D, Sun X, Tong J, Ma J, Bu X, Xu R, Fan R. A novel multifunctional nanocomposite C225-conjugated Fe3O4/Ag enhances the sensitivity of nasopharyngeal carcinoma cells to radiotherapy. Acta Biochim Biophys Sin (Shanghai) 2012; 44:678-84. [PMID: 22710262 DOI: 10.1093/abbs/gms051] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Radiotherapy is the major treatment for nasopharyngeal carcinoma, a malignant tumor of epithelial origin. In this process, a tracer with high sensitivity is pivotal for diagnostic imaging in radiotherapy. Here, we designed a novel multifunctional magnetic silver nanocomposite, Fe(3)O(4)/Ag conjugated to an epidermal growth factor receptor-specific antibody (C225), which can be potentially used for synchronous cancer therapy and diagnosis via magnetic resonance imaging. Characteristics of Fe(3)O(4)/Ag/C225 were determined by transmission electron microscopy, energy dispersive X-ray spectroscopy, ultraviolet spectra, and dynamic light scattering. The results demonstrated that Fe(3)O(4)/Ag/C225 nanoparticles were spherical and dispersed well in water. The activity of C225 was preserved ∼80% in the Fe(3)O(4)/Ag/C225 nanoparticles. Futhermore, we tested the cytotoxicity and radiosensitivity of the nanocomposite for human nasopharyngeal carcinoma cell lines (CNEs) in vitro. MTT analysis revealed that Fe(3)O(4)/Ag/C225 could inhibit the proliferation of CNEs in a dose- and time-dependent manner. The clonogenic assay indicated that Fe(3)O(4)/Ag/C225 combined with X-ray treatment could increase the sensitivity of CNEs to irradiation. In a summary, the novel multifunctional nanocomposite Fe(3)O(4)/Ag/C225 might be a potential radiosensitizer for treating malign tumors in the clinic.
Collapse
Affiliation(s)
- Di Zhao
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Yang L, Li N, Wang H, Jia X, Wang X, Luo J. Altered microRNA expression in cisplatin-resistant ovarian cancer cells and upregulation of miR-130a associated with MDR1/P-glycoprotein-mediated drug resistance. Oncol Rep 2012; 28:592-600. [PMID: 22614869 DOI: 10.3892/or.2012.1823] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/02/2012] [Indexed: 01/18/2023] Open
Abstract
microRNAs (miRNAs) are short non-coding RNA molecules which are involved in the regulation of various biological processes. Drug resistance has become a major obstacle to successful chemotherapy of ovarian cancer. The aim of this study was to investigate microRNA expression profiles in cisplatin-resistant ovarian cancer cells and the role of miR-130a in regulating drug resistance. Analysis of differentially expressed miRNAs between SKOV3 and SKOV3/CIS cells was assessed by miRNA microarrays. Target prediction of miRNAs was determined with the help of PicTar or TargetScan. Among these miRNAs, the expression of miR‑130a was verified using qRT-PCR. The expression of MDR1 mRNA and P-glycoprotein (P-gp) after cellular transfection was examined using qRT-PCR and western blotting, respectively. Cisplatin sensitivity was detected by the MTT assay. We indentified 35 downregulated and 54 upregulated miRNAs in SKOV3/CIS compared to those in SKOV3. We found that miR-130a was upregulated in SKOV3/CIS compared to the parental SKOV3 cells, and PTEN was predicted to be the potential target of miR-130a. Moreover, downregulation of miR-130a could inhibit MDR1 mRNA and P-gp expression and overcome the cisplatin resistance in SKOV3/CIS cells, which indicated that miR-130a may be associated with MDR1/P-gp-mediated drug resistance and plays the role of an intermediate in drug-resistance pathways of PI3K/Akt/PTEN/mTOR and ABC superfamily drug transporters in SKOV3/CIS cells. This study provides important information for the development of targeted gene therapy for reversing cisplatin resistance in ovarian cancer.
Collapse
Affiliation(s)
- Lingyun Yang
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | | | | | | | | | | |
Collapse
|