(3S-5S-6E)-7-[3-(4-fluorophenyl)-1-(propan-2-yl)-1H-indol-2-yl]-3-5-dihydroxyhept-6-enoic-acid and Lung-Neoplasms

(3S-5S-6E)-7-[3-(4-fluorophenyl)-1-(propan-2-yl)-1H-indol-2-yl]-3-5-dihydroxyhept-6-enoic-acid has been researched along with Lung-Neoplasms* in 14 studies

Other Studies

14 other study(ies) available for (3S-5S-6E)-7-[3-(4-fluorophenyl)-1-(propan-2-yl)-1H-indol-2-yl]-3-5-dihydroxyhept-6-enoic-acid and Lung-Neoplasms

ArticleYear
Therapeutic trial of fluvastatin in a cell line xenograft model of canine mammary gland cancer.
    Veterinary and comparative oncology, 2023, Volume: 21, Issue:4

    The Hippo signalling pathway is involved in breast cancer and canine mammary tumour (CMT). This study sought to evaluate the efficacy of fluvastatin on the Hippo pathway and its main effectors, YAP and TAZ, in vivo in a murine CMT cell line xenograft model. On treatment day 1, mice were divided into four groups: vehicle, fluvastatin, doxorubicin or a combination therapy. Tumour volumes were monitored with callipers and tissues harvested on day 28th of treatment. Histopathological examination of tumour tissues and major organs was performed as well as tumour evaluation of necrosis, apoptosis, cellular proliferation, expression of YAP, TAZ and the mRNA levels of four of their target genes (CTGF, CYR61, ANKRD1 and RHAMM2). Results showed a statistically significant variation in tumour volumes only for the combination therapy and final tumour weight only for the doxorubicin group compared to control. There was no significant difference in tumour necrosis, expression of CC3, ki-67, YAP and TAZ measured by immunohistochemistry and in the mRNA levels of the target genes. Unexpectedly, lung metastases were found in the control group (9) and not in the fluvastatin treated group (7). In addition, mass spectrometry-based quantification of fluvastatin reveals concentrations comparable to levels reported to exert therapeutic effects. This study shows that fluvastatin tumours concentration reached therapeutic levels without having an effect on the hippo pathway or various tumour parameters. Interestingly, only the control group had lung metastases. This study is the first to explore the repurposing of statins for cancer treatment in veterinary medicine.

    Topics: Animals; Breast Neoplasms; Cell Line, Tumor; Dog Diseases; Dogs; Doxorubicin; Female; Fluvastatin; Heterografts; Humans; Lung Neoplasms; Mammary Glands, Human; Mammary Neoplasms, Animal; Mice; Necrosis; RNA, Messenger; Rodent Diseases; Transcription Factors

2023
Mevalonate Pathway Inhibition Slows Breast Cancer Metastasis via Reduced
    Cancer research, 2021, 05-15, Volume: 81, Issue:10

    Aberrant

    Topics: Adjuvants, Immunologic; Animals; Apoptosis; Breast Neoplasms; Cell Proliferation; Endoplasmic Reticulum; Epithelial-Mesenchymal Transition; Female; Fluvastatin; Gene Expression Regulation, Neoplastic; Glycosylation; Humans; Lung Neoplasms; Mevalonic Acid; Mice; Mice, SCID; Prognosis; Signal Transduction; Tumor Cells, Cultured; Xenograft Model Antitumor Assays

2021
Comparison of the Effects of Statins on A549 Nonsmall-Cell Lung Cancer Cell Line Lipids Using Fourier Transform Infrared Spectroscopy: Rosuvastatin Stands Out.
    Lipids, 2021, Volume: 56, Issue:3

    Statins are commonly prescribed antilipidemic and anticholesterol class of drugs. In addition to their major role, they have been found to have anticancer effects on in vitro, animal and clinical studies. The aim of this study was to investigate the effects of six different statins (rosuvastatin, pravastatin, simvastatin, lovastatin, fluvastatin, and atorvastatin) on A549 cancer cells lipids by Fourier transform infrared (FTIR) spectroscopy. Proliferation tests were carried out to detect the half-maximal inhibitory concentrations (IC

    Topics: A549 Cells; Atorvastatin; Carcinoma, Non-Small-Cell Lung; Cell Proliferation; Cell Survival; Dose-Response Relationship, Drug; Fluvastatin; Humans; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Lipid Metabolism; Lipids; Lovastatin; Lung Neoplasms; Pravastatin; Rosuvastatin Calcium; Simvastatin; Spectroscopy, Fourier Transform Infrared

2021
Delineation of cell death mechanisms induced by synergistic effects of statins and erlotinib in non-small cell lung cancer cell (NSCLC) lines.
    Scientific reports, 2020, 01-22, Volume: 10, Issue:1

    Hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) have been shown to overcome tyrosine kinase inhibitor (TKI) resistance in epithelial growth factor receptor (EGFR) mutated non-small cell lung cancer (NSCLC) cells in vivo and in vitro. However, little is known about the putative induction of non-apoptotic cell death pathways by statins. We investigated the effects of pitavastatin and fluvastatin alone or in combination with erlotinib in three NSCLC cell lines and examined the activation of different cell death pathways. We assessed apoptosis via fluorometric caspase assay and poly (ADP-ribose) polymerase 1 (PARP) cleavage. Furthermore, annexinV/propidium iodide (PI) flow cytometry was performed. Small molecule inhibitors benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (zVAD), necrostatin 1 (Nec1), ferrostatin 1 (Fer1), Ac-Lys-Lys-Norleucinal (Calp1) were used to characterise cell death pathway(s) putatively (co-)activated by pitavastatin/erlotinib co-treatment. Synergism was calculated by additivity and isobolographic analyses. Pitavastatin and fluvastatin induced cell death in EGFR TKI resistant NSCLC cells lines A549, Calu6 and H1993 as shown by caspase 3 activation and PARP cleavage. Co-treatment of cells with pitavastatin and the EGFR TKI erlotinib resulted in synergistically enhanced cytotoxicity compared to pitavastatin monotherapy. Flow cytometry indicated the induction of alternative regulated cell death pathways. However, only co-treatment with mevalonic acid (Mev) or the pan-caspase inhibitor zVAD could restore cell viability. The results show that cytotoxicity mediated by statin/erlotinib co-treatment is synergistic and can overcome erlotinib resistance in K-ras mutated NSCLC and relies only on apoptosis.

    Topics: Antineoplastic Agents; Apoptosis; Carcinoma, Non-Small-Cell Lung; Cell Death; Cell Line, Tumor; Drug Resistance, Neoplasm; Drug Synergism; Erlotinib Hydrochloride; Fluvastatin; Humans; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Lung Neoplasms; Quinolines

2020
Fluvastatin Inhibits HMG-CoA Reductase and Prevents Non-Small Cell Lung Carcinogenesis.
    Cancer prevention research (Philadelphia, Pa.), 2019, Volume: 12, Issue:12

    Topics: Acyl Coenzyme A; Adult; Aged; Animals; Apoptosis; Carcinogenesis; Carcinogens; Carcinoma, Non-Small-Cell Lung; Cell Line, Tumor; Female; Fluvastatin; Gene Knockdown Techniques; HEK293 Cells; Humans; Hydroxymethylglutaryl CoA Reductases; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Lung; Lung Neoplasms; Male; Mevalonic Acid; Mice; Middle Aged; Neoplasms, Experimental; Nitrosamines; RNA, Small Interfering; Xenograft Model Antitumor Assays

2019
Fluvastatin-mediated down-regulation of SATB1 affects aggressive phenotypes of human non-small-cell lung cancer cell line H292.
    Life sciences, 2019, Apr-01, Volume: 222

    Fluvastatin reduces tumor proliferation and increased apoptotic activity in various cancers. Special AT-rich sequence binding protein 1 (SATB1) is a genome organizer that reprogrammes the gene transcription profiles of tumors to promote growth and metastasis. The antitumor effect and molecular mechanisms of fluvastatin on lung cancer is poorly understood. This study aimed to investigate the antitumor effect of fluvastatin on lung cancer and its possible mechanics.. Cell viability assay was used to examine the inhibition of fluvastatin on proliferation of H292 cells. In order to investigate the antitumor mechanics, SATB1 knock-down H292 cells was constructed by lentiviral transfection. RT-PCR and Western blot were performed to examine the effects of fluvastatin on expression of SATB1 and Wnt/β-catenin signaling components.. Fluvastatin significantly inhibited proliferation and invasion of H292 cells in a time- and dose-dependent manner and promoted the apoptosis (p < 0.05). The expression of SATB1 was down-regulated by fluvastatin in a dose-dependent manner. The proliferation and invasion of SATB1-shRNA cells was significantly suppressed, and the apoptosis was significantly enhanced (p < 0.05). We also show that the common target genes were regulated by SATB1 and Wnt/β-catenin pathway simultaneously. There may be a functional link between SATB1 and Wnt/β-catenin pathway.. We presented a possible mechanism of statins that fluvastatin significantly suppressed the in vitro tumor progression of H292 cells possibly by down-regulation of SATB1 via Wnt/β-catenin pathway, which provided new therapeutic possibilities for more cancers driven by hyperexpression of SATB1 and Wnt/β-catenin pathway.

    Topics: A549 Cells; Carcinoma, Non-Small-Cell Lung; Cell Line, Tumor; Down-Regulation; Fluvastatin; Humans; Lung Neoplasms; Matrix Attachment Region Binding Proteins; Neoplasm Invasiveness; Phenotype

2019
Combination therapy with dacarbazine and statins improved the survival rate in mice with metastatic melanoma.
    Journal of cellular physiology, 2019, Volume: 234, Issue:10

    Malignant melanoma is a highly aggressive skin cancer, and the overall median survival in patients with metastatic melanoma is only 6-9 months. Although molecular targeted therapies have recently been developed and have improved the overall survival, melanoma patients may show no response and acquisition of resistance to these drugs. Thus, other molecular approaches are essential for the treatment of metastatic melanoma. In the present study, we investigated the effect of cotreatment with dacarbazine and statins on tumor growth, metastasis, and survival rate in mice with metastatic melanomas. We found that cotreatment with dacarbazine and statins significantly inhibited tumor growth and metastasis via suppression of the RhoA/RhoC/LIM domain kinase/serum response factor/c-Fos pathway and enhanced p53, p21, p27, cleaved caspase-3, and cleaved poly(ADP-ribose) polymerase 1 expression in vivo. Moreover, the cotreatment significantly improved the survival rate in metastasis-bearing mice. Importantly, treatment with dacarbazine plus 100 mg/kg simvastatin or fluvastatin prevented metastasis-associated death in 4/20 mice that received dacarbazine + simvastatin and in 8/20 mice that received dacarbazine + fluvastatin (survival rates, 20% and 40%, respectively). These results suggested that cotreatment with dacarbazine and statins may thus serve as a new therapeutic approach to control tumor growth and metastasis in melanoma patients.

    Topics: Animals; Antineoplastic Agents, Alkylating; Antineoplastic Combined Chemotherapy Protocols; Cell Adhesion; Cell Line, Tumor; Cell Proliferation; Dacarbazine; Female; Fluvastatin; Gene Expression Regulation; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Lung Neoplasms; Melanoma, Experimental; Mice, Inbred C57BL; Signal Transduction; Simvastatin; Skin Neoplasms; Tumor Burden

2019
Fluvastatin Prevents Lung Adenocarcinoma Bone Metastasis by Triggering Autophagy.
    EBioMedicine, 2017, Volume: 19

    Bone is one of the most preferred sites of metastasis in lung cancer. Currently, bisphosphonates and denosumab are major agents for controlling tumor-associated skeletal-related events (SREs). However, both bisphosphonates and denosumab significantly increase the risk for jaw osteonecrosis. Statins, 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors and the most frequently prescribed cholesterol-lowering agents, have been reported to inhibit tumor progression and induce autophagy in cancer cells. However, the effects of statin and role of autophagy by statin on bone metastasis are unknown. In this study, we report that fluvastatin effectively prevented lung adenocarcinoma bone metastasis in a nude mouse model. We further reveal that fluvastatin-induced anti-bone metastatic property was largely dependent on its ability to induce autophagy in lung adenocarcinoma cells. Atg5 or Atg7 deletion, or 3-methyadenine (3-MA) or Bafilomycin A1 (Baf A1) treatment prevented the fluvastatin-induced suppression of bone metastasis. Furthermore, we reveal that fluvastatin stimulation increased the nuclear p53 expression, and fluvastatin-induced autophagy and anti-bone metastatic activity were mostly dependent on p53.

    Topics: Adenocarcinoma; Adenocarcinoma of Lung; Animals; Antineoplastic Agents; Autophagy; Bone Neoplasms; Cell Line, Tumor; Fatty Acids, Monounsaturated; Female; Fluvastatin; Humans; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Indoles; Lung Neoplasms; Mice, Inbred BALB C; Mice, Nude; Tumor Suppressor Protein p53

2017
Statin suppresses Hippo pathway-inactivated malignant mesothelioma cells and blocks the YAP/CD44 growth stimulatory axis.
    Cancer letters, 2017, 01-28, Volume: 385

    Malignant mesothelioma (MM) frequently exhibits Hippo signaling pathway inactivation (HPI) mainly due to NF2 and/or LATS2 mutations, which leads to the activation of YAP transcriptional co-activator. Here, we show antitumor effects of statin on MM cells with HPI, through the interplay of the mevalonate and Hippo signaling pathways. Statin attenuated proliferation and migration of MM cells harboring NF2 mutation by accelerating YAP phosphorylation/inactivation. CD44 expression was decreased by statin, in parallel with YAP phosphorylation/inactivation. Importantly, we discovered that YAP/TEAD activated CD44 transcription by binding to the CD44 promoter at TEAD-binding sites. On the other hand, CD44 regulated Merlin phosphorylation according to cell density and sequentially promoted YAP transcriptional co-activator, suggesting that CD44 plays two pivotal functional roles as an upstream suppressor of the Hippo pathway and one of downstream targets regulated by YAP/TEAD. Moreover, the YAP/CD44 axis conferred cancer stem cell (CSC)-like properties in MM cells leading to chemoresistance, which was blocked by statin. Together, our findings suggest that YAP mediates CD44 up-regulation at the transcriptional level, conferring CSC-like properties in MM cells, and statin represents a potential therapeutic option against MM by inactivating YAP.

    Topics: Adaptor Proteins, Signal Transducing; Antineoplastic Agents; Binding Sites; Cell Line, Tumor; Cell Movement; Cell Proliferation; Dose-Response Relationship, Drug; Fatty Acids, Monounsaturated; Fluvastatin; Gene Expression Regulation, Neoplastic; Hippo Signaling Pathway; Humans; Hyaluronan Receptors; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Indoles; Lung Neoplasms; Mesothelioma; Mesothelioma, Malignant; Mevalonic Acid; Mutation; Neoplasm Invasiveness; Neoplastic Stem Cells; Neurofibromin 2; Phosphoproteins; Phosphorylation; Promoter Regions, Genetic; Protein Serine-Threonine Kinases; RNA Interference; Signal Transduction; Simvastatin; Time Factors; Transcription Factors; Transcription, Genetic; Transfection; Tumor Suppressor Proteins; Ubiquitin Thiolesterase; YAP-Signaling Proteins

2017
Drosophila Lung Cancer Models Identify Trametinib plus Statin as Candidate Therapeutic.
    Cell reports, 2016, Feb-16, Volume: 14, Issue:6

    We have developed a Drosophila lung cancer model by targeting Ras1(G12V)--alone or in combination with PTEN knockdown--to the Drosophila tracheal system. This led to overproliferation of tracheal tissue, formation of tumor-like growths, and animal lethality. Screening a library of FDA-approved drugs identified several that improved overall animal survival. We explored two hits: the MEK inhibitor trametinib and the HMG-CoA reductase inhibitor fluvastatin. Oral administration of these drugs inhibited Ras and PI3K pathway activity, respectively; in addition, fluvastatin inhibited protein prenylation downstream of HMG-CoA reductase to promote survival. Combining drugs led to synergistic suppression of tumor formation and rescue lethality; similar synergy was observed in human A549 lung adenocarcinoma cells. Notably, fluvastatin acted both within transformed cells and also to reduce whole-body trametinib toxicity in flies. Our work supports and provides further context for exploring the potential of combining statins with MAPK inhibitors such as trametinib to improve overall therapeutic index.

    Topics: Animals; Antineoplastic Agents; Cell Line, Tumor; Disease Models, Animal; Drosophila melanogaster; Drosophila Proteins; Drug Combinations; Drug Screening Assays, Antitumor; Drug Synergism; Fatty Acids, Monounsaturated; Fluvastatin; Gene Expression Regulation, Neoplastic; Humans; Hydroxymethylglutaryl-CoA Reductase Inhibitors; IMP Dehydrogenase; Indoles; Lung Neoplasms; Protein Kinase Inhibitors; PTEN Phosphohydrolase; Pyridones; Pyrimidinones; Signal Transduction; Survival Rate

2016
Genome-wide RNAi analysis reveals that simultaneous inhibition of specific mevalonate pathway genes potentiates tumor cell death.
    Oncotarget, 2015, Sep-29, Volume: 6, Issue:29

    The mevalonate (MVA) pathway is often dysregulated or overexpressed in many cancers suggesting tumor dependency on this classic metabolic pathway. Statins, which target the rate-limiting enzyme of this pathway, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), are promising agents currently being evaluated in clinical trials for anti-cancer efficacy. To uncover novel targets that potentiate statin-induced apoptosis when knocked down, we carried out a pooled genome-wide short hairpin RNA (shRNA) screen. Genes of the MVA pathway were amongst the top-scoring targets, including sterol regulatory element binding transcription factor 2 (SREBP2), 3-hydroxy-3-methylglutaryl-coenzyme A synthase 1 (HMGCS1) and geranylgeranyl diphosphate synthase 1 (GGPS1). Each gene was independently validated and shown to significantly sensitize A549 cells to statin-induced apoptosis when knocked down. SREBP2 knockdown in lung and breast cancer cells completely abrogated the fluvastatin-induced upregulation of sterol-responsive genes HMGCR and HMGCS1. Knockdown of SREBP2 alone did not affect three-dimensional growth of lung and breast cancer cells, yet in combination with fluvastatin cell growth was disrupted. Taken together, these results show that directly targeting multiple levels of the MVA pathway, including blocking the sterol-feedback loop initiated by statin treatment, is an effective and targetable anti-tumor strategy.

    Topics: Antineoplastic Agents; Apoptosis; Breast Neoplasms; Cell Line, Tumor; Cell Proliferation; Dimethylallyltranstransferase; Farnesyltranstransferase; Fatty Acids, Monounsaturated; Female; Fluvastatin; Gene Expression Regulation, Neoplastic; Geranyltranstransferase; Humans; Hydroxymethylglutaryl CoA Reductases; Hydroxymethylglutaryl-CoA Synthase; Indoles; Lung Neoplasms; Mevalonic Acid; Neoplasms; Real-Time Polymerase Chain Reaction; RNA Interference; RNA, Small Interfering; Sterol Regulatory Element Binding Protein 2

2015
Reduction of lung metastasis, cell invasion, and adhesion in mouse melanoma by statin-induced blockade of the Rho/Rho-associated coiled-coil-containing protein kinase pathway.
    Journal of experimental & clinical cancer research : CR, 2010, Sep-16, Volume: 29

    Melanomas are highly malignant and have high metastatic potential; hence, there is a need for new therapeutic strategies to prevent cell metastasis. In the present study, we investigated whether statins inhibit tumor cell migration, invasion, adhesion, and metastasis in the B16BL6 mouse melanoma cell line.. The cytotoxicity of statins toward the B16BL6 cells were evaluated using a cell viability assay. As an experimental model, B16BL6 cells were intravenously injected into C57BL/6 mice. Cell migration and invasion were assessed using Boyden chamber assays. Cell adhesion analysis was performed using type I collagen-, type IV collagen-, fibronectin-, and laminin-coated plates. The mRNA levels, enzyme activities and protein levels of matrix metalloproteinases (MMPs) were determined using RT-PCR, activity assay kits, and Western blot analysis, respectively; the mRNA and protein levels of vary late antigens (VLAs) were also determined. The effects of statins on signal transduction molecules were determined by western blot analyses.. We found that statins significantly inhibited lung metastasis, cell migration, invasion, and adhesion at concentrations that did not have cytotoxic effects on B16BL6 cells. Statins also inhibited the mRNA expressions and enzymatic activities of matrix metalloproteinases (MMPs). Moreover, they suppressed the mRNA and protein expressions of integrin α2, integrin α4, and integrin α5 and decreased the membrane localization of Rho, and phosphorylated LIM kinase (LIMK) and myosin light chain (MLC).. The results indicated that statins suppressed the Rho/Rho-associated coiled-coil-containing protein kinase (ROCK) pathways, thereby inhibiting B16BL6 cell migration, invasion, adhesion, and metastasis. Furthermore, they markedly inhibited clinically evident metastasis. Thus, these findings suggest that statins have potential clinical applications for the treatment of tumor cell metastasis.

    Topics: Administration, Oral; Animals; Antineoplastic Agents; Blotting, Western; Cell Adhesion; Cell Movement; Cell Survival; Extracellular Matrix Proteins; Fatty Acids, Monounsaturated; Female; Fluvastatin; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Neoplastic; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Indoles; Integrins; Lim Kinases; Lung Neoplasms; Matrix Metalloproteinases; Melanoma, Experimental; Mice; Mice, Inbred C57BL; Myosin Light Chains; Neoplasm Invasiveness; Phosphorylation; Protein Kinase Inhibitors; Reverse Transcriptase Polymerase Chain Reaction; rho GTP-Binding Proteins; rho-Associated Kinases; RNA, Messenger; Signal Transduction; Simvastatin; Time Factors

2010
Zoledronic acid (Zometa) enhances the cytotoxic effect of gemcitabine and fluvastatin: in vitro isobologram studies with conventional and nonconventional cytotoxic agents.
    Oncology, 2006, Volume: 70, Issue:2

    To identify synergistic combinations of clinically available agents with zoledronic acid which would enhance antitumor activity as measured by median effect isobologram analysis and apoptosis assays in vitro.. The interaction of zoledronic acid as a doublet with either carboplatin, cisplatin, 5'DFUR, docetaxel, epirubicin, fluvastatin, gemcitabine, imatinib, paclitaxel, trastuzumab, or vinorelbine was studied in a 72-hour in vitro system using defined human cancer cell lines grown as a monolayer in exponential phase. Drug effect on growth was measured by a standard MTT assay. Median effect isobologram analysis was applied to the results to determine the presence of synergism, additive effects, or antagonism of drug combinations. Synergistic combinations were also assayed by a cytoplasmic histone-associated DNA fragmentation apoptosis assay to verify that the effect was not cytostatic.. Zoledronic acid with gemcitabine demonstrated global cytotoxic synergy across 7 of 8 cell lines. Clinically achievable concentrations of fluvastatin with zoledronic acid also demonstrated synergy in 7 of 8 cell lines. All the breast cancer cell lines were sensitive. Zoledronic acid and epirubicin were antagonistic in all 4 breast cell lines studied.. Combinations of zoledronic acid with either gemcitabine or fluvastatin may have a therapeutic role in treatment of bone metastasis of selected malignancies.

    Topics: Antineoplastic Combined Chemotherapy Protocols; Apoptosis; Bone Density Conservation Agents; Bone Neoplasms; Breast Neoplasms; Cell Line, Tumor; Cytoplasm; Deoxycytidine; Diphosphonates; DNA Fragmentation; Drug Antagonism; Drug Synergism; Epirubicin; Fatty Acids, Monounsaturated; Female; Fluvastatin; Gemcitabine; Humans; Imidazoles; Indoles; Lung Neoplasms; Male; Prostatic Neoplasms; Zoledronic Acid

2006
3-hydroxy-3-methylglutaryl-coenzyme a reductase inhibitor, fluvastatin, as a novel agent for prophylaxis of renal cancer metastasis.
    Clinical cancer research : an official journal of the American Association for Cancer Research, 2004, Dec-15, Volume: 10, Issue:24

    Inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, also called statins, are currently used widely as a safe, effective therapeutic in the treatment of hypercholesterolemia. Recently, statins have been recognized for their activity against cancer. In the present study, we examined the effect of a synthetic statin, fluvastatin, on the development of renal cancer.. The effects of fluvastatin on cell viability, cell cycle, in vitro angiogenesis, and invasive properties were examined in murine renal cancer cell Renca. The changes in cell cycle-associated proteins, p21(Waf1/Cip1) and p53, and rac1 phosphorylation were analyzed by Western blotting. The prophylactic efficacy of fluvastatin to murine pulmonary metastasis of Renca was examined.. Fluvastatin inhibited in vitro growth of Renca cells in a time- and dose-dependent manner, with up to 70% inhibition at a concentration of 10 mumol/L. This inhibitory effect was due to cell cycle arrest at the G(1) phase and induction of apoptosis accompanied by up-regulation of p21(Waf1/Cip1) and p53. The invasive properties of Renca cells through Matrigel were inhibited by fluvastatin, with decreased phosphorylation of rac1. In vitro angiogenesis was also inhibited by fluvastatin. Furthermore, oral administration at doses of 1 to 10 mg/kg/d, for 12 days after inoculation of Renca cells via the tail vein, significantly decreased the amount of pulmonary metastasis.. Because our results suggest that fluvastatin may effectively inhibit in vitro tumor growth, invasion, angiogenesis, and metastasis of Renca cells, oral administration of fluvastatin could be a novel, safe, and effective agent for preventing metastasis of renal cancer.

    Topics: Animals; Antibiotic Prophylaxis; Apoptosis; Blotting, Western; Carcinoma, Renal Cell; Cell Cycle Proteins; Cell Proliferation; Cyclin-Dependent Kinase Inhibitor p21; Fatty Acids, Monounsaturated; Fluvastatin; G1 Phase; Humans; Hydroxymethylglutaryl CoA Reductases; Hydroxymethylglutaryl-CoA Reductase Inhibitors; Indoles; Kidney Neoplasms; Lung Neoplasms; Male; Mice; Mice, Inbred BALB C; Neoplasm Invasiveness; Neovascularization, Pathologic; Neuropeptides; Phosphorylation; rac GTP-Binding Proteins; rac1 GTP-Binding Protein; Tumor Cells, Cultured; Tumor Suppressor Protein p53

2004