GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
Filter
  • American Association for Cancer Research (AACR)  (22)
Materialart
Verlag/Herausgeber
  • American Association for Cancer Research (AACR)  (22)
Sprache
Erscheinungszeitraum
Fachgebiete(RVK)
  • 1
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 23, No. 9 ( 2017-05-01), p. 2335-2345
    Kurzfassung: Purpose: Accumulating evidence indicates that factors secreted by cancer epithelial cells shape the tumor microenvironment to promote cancer invasion and metastasis. Recent studies also shed light on alterations of Rab small GTPase–mediated exocytosis in tumorigenesis. However, the mechanisms for Rab-mediated exocytosis in tumor microenvironment remain elusive. We aimed to investigate the interplay between Rab37-mediated exocytosis and tumor microenvironment, focusing on endothelial cell motility and angiogenesis. Experimental Design: We performed fluorescence IHC for Rab37, thrombospondin-1 (TSP1, an antiangiogenesis factor), and angiogenesis marker CD31 in 183 surgically resected esophageal squamous cell carcinoma (ESCC) patient samples. Cell migration, invasion, angiogenesis, and tumor metastasis were measured. Results: ESCC patients with low expression of Rab37 or TSP1 significantly correlated with high CD31 expression and were associated with worse progression-free survival. The multivariate Cox regression analysis showed that concordant low expression of both Rab37 and TSP1 was an independent prognostic factor of ESCC patients. Rab37-mediated exocytosis of TSP1 led to the inhibition of neovasculature in vitro and in vivo. Secreted TSP1 from cancer cells with Rab37 exocytic function inhibited the p-FAK/p-paxillin/p-ERK migration signaling in both cancer epithelial cells and their surrounding endothelial cells. Dysfunction of Rab37 or loss of TSP1 abrogated the suppressive effects on angiogenesis and metastasis. Conclusions: Our findings suggest that Rab37-mediated TSP1 secretion in cancer cells suppresses metastasis and angiogenesis via a cross-talk with endothelial cells and reveal a novel component of the vesicular exocytic machinery in tumor microenvironment and tumor progression. Dysregulation of Rab37/TSP1 axis has clinical implications for prognosis prediction. Clin Cancer Res; 23(9); 2335–45. ©2016 AACR.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2017
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 74-74
    Kurzfassung: Ovarian cancer is one of the most lethal cancers in the female reproductive system. Previous study suggested that long term treatment of estrogen such as hormonal replacement therapy (HRT) may increase the risk of ovarian cancer, however the role of estrogen in ovarian carcinogenesis is still controversial. To decipher this complicated process, we generated a mathematical model and found that estrogen-mediated up-regulation of E2F6 could upregulate the ovarian cancer stem/initiating marker, c-kit by two means one through epigenetic silencing of their co-targeted miR193a by binding of E2F6 which subsequently recruit EZH2 to miR-193a promoter; and second, by competing endogenous (ceRNA) mechanism. To confirm this model, treatment of E2 or environmental hormone, BPA resulted in upregulation of both E2F6 and c-kit but down-regulation of miR-193a in immortalized ovarian surface epithelial cells. Further bisulfite pyrosequencing, ChIP-qPCR and epigenetic treatment found that miR193a was epigenetically silenced by DNA methylation and H3K27me3 in CP70 but not HeyC2 ovarian cancer cells. Overexpression of miR193a inhibited tumor growth in vitro and in vivo. Depletion of EZH2 or E2F6 in CP70 restored miR-193a expression and decreased the number of “ovo” spheroid by reversing the repressive chromatin status of miR-193a promoter. To further explore the biological significance of this E2F6 ceRNA network, integrative RNA-Seq and computational analysis found that PBX1, a miR-193a target and transcriptional activator of the immunosuppressive cytokine IL-10, was down-regulated in E2F6 and EZH2 knockdown CP70 cells. Overexpression of E2F6 3'UTR containing miR-193a MRE but not MRE mutant increased the expression of PBX1 and IL10 in ovarian cancer cells. Importantly, co-culture of conditional media from E2F6 3'UTR overexpressing CP70 cells inhibited the differentiation of THP-1 monocytes into dendritic cell and the T-cell activating function of this THP-1 derived DC. This phenomenon can be rescued by incubation of anti-IL-10 antibody or pretreatment of CP70 cells with EZH2 inhibitor. Finally, clinical studies demonstrated that patients with higher promoter methylation of miR193a were associated with poor survival. Serum IL10 level was found to be higher in high staged ovarian cancer patients and patients with higher E2F6 mRNA level. Additional analysis from TCGA ovarian cancer expression microarray dataset demonstrated that ovarian cancer patients with low expression of EZH2, showed a positive correlation between E2F6, c-KIT and PBX1 resembling the ceRNA phenomenon between these mRNAs. Taken together, our results showed that estrogen-mediated E2F6 ceRNA network can regulate cancer stemness and anti-tumor immunity of DC through epigenetic silencing of miR-193a. Anti-estrogen therapy together with the EZH2 inhibitor may be a novel strategy against this deadly cancer. Citation Format: Frank Hsueh-Che Cheng, Hon-Yi Lin, Yin-Chen Chen, Tzy-Wei Hwang, Rui-Lan Huang, Chia-Bin Chang, Ru-Inn Lin, Ching-Wen Lin, Gary C.W. Chen, Jora M. J. Lin, Yu-Ming Chuang, Jian-Liang Chou, Chin Li, Alfred S.L. Cheng, Hung-Cheng Lai, Shu-Fen Wu, Je-Chiang Tsai, Michael W.Y. Chan. E2F6-mediated ceRNA and epigenetic silencing of miR193a lead to cancer stemness and anticancer immunity in ovarian cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 74.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2018
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3433-3433
    Kurzfassung: The TGF-β signaling pathway plays an important role in controlling cell growth and differentiation. In advanced ovarian cancer, frequent TGF-β-induced metastasis or epithelial-mesenchymal transition (EMT) can be observed. This phenomenon is often associated with epigenetic silencing of epithelial marker, E-cadherin which can also be observed in ovarian cancer cell lines that demethylation treatment restored E-cadherin expression. We recently hypothesized that long term activation of TGF-β signaling may induce EMT phenotype by epigenetic silencing of E-cadherin and that inhibition of the signaling may restore E-cadherin and reverse EMT in ovarian cancer (Chou et al., Expert Opin Ther Targets 2010). In this study, we cloned the cDNA of the inhibitory SMAD, SMAD7 from a human immortalized ovarian surface epithelial cell, IOSE into pcDNA3.1 mammalian expression vector. The inhibitory effect of this SMAD7 expression vector on TGF-β signaling has been confirmed by reporter assay. We then stably transfected the SMAD7 expression vector into a mesenchymal ovarian cancer cell, CP70 in which E-cadherin is silenced by complete promoter methylation. Cells over-expressing SMAD7 showed up-regulation of SMAD7 and a decrease in SMAD2 phosphorylation while the control cells maintained a hyperphosphorylation of SMAD2 thus suggesting that TGF-β signaling is disrupted in SMAD7-overexpressing cells. We further examined the expression of E-cadherin from passage 5 up to 30 of the stable transfectants. Surprisingly, stable restoration of E-cadherin can only be observed from passage 20 and thereafter, of the SMAD7-overexpressing cells, while E-cadherin remained silence in the control cells. To investigate if this restoration is due to promoter demethylation of E-cadherin, we performed bisulphite pyrosequencing on the E-cadherin promoter CpG island spanning -586 to -12 of the region. Compared with control cells, consistent demethylation of E-cadherin promoter can be observed at 2 CpG sites located at -214 and -235 of the promoter such that gradual demethylation occurred from passage 5 to 30 of the SMAD7-overexpressing cells (passage 20, methylation% control vs SMAD7: 91% vs 67% at -235; 80% vs 52% at -214); while the rest of the CpG sites remained heavily methylated. This demethylation may be due to down-regulation of transcriptional repressor, TWIST after SMAD7 transfection. Additionally, one of the SMAD7 stable expression clones with highest restoration of E-cadherin showed decreased migration and invasion ability as determined by wound healing and invasion assay. Taken together, disruption of TGF-β signaling can induce demethylation of E-cadherin promoter and reverse EMT phenotype in ovarian cancer. The therapeutic potential of targeting TGF-β signaling pathway in inhibiting metastasis of ovarian cancer deserves further investigation. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3433. doi:10.1158/1538-7445.AM2011-3433
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2011
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2007
    In:  Clinical Cancer Research Vol. 13, No. 3 ( 2007-02-01), p. 851-857
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 13, No. 3 ( 2007-02-01), p. 851-857
    Kurzfassung: Purpose: Hepatitis B virus (HBV) reactivation is one unique pathogenesis in Asian carriers with liver toxicity after radiotherapy for hepatobiliary malignancies. This study attempts to delineate the biological mechanism of radiation-induced HBV reactivation. Experimental Design: Primary cultures of hepatocytes (PCC) were prepared from the noncancerous liver tissue removed perioperatively from 12 HBV carriers with hepatocellular carcinoma (HCC). The conditioned medium of irradiated PCCs, HCC, and endothelial cells from patients was transferred to PCCs or HepG2.2.15 cells (a human hepatoblastoma cell line transfected with HBV DNA) before subsequent irradiation. Forty-eight hours after irradiation, HBV DNA was measured by real-time quantitative PCR. Specific cytokines were determined by cytokine array and ELISA analysis. Preradiotherapy and postradiotherapy sera from 10 HBV carriers and 16 non-HBV carriers were analyzed for viral loads and cytokine activities. Results: Radiation induced HBV DNA replication in (a) irradiated PCCs cultured with the conditioned medium from irradiated PCCs (2.74-fold; P = 0.004) and endothelial cells (9.50-fold; P = 3.1 × 10−10), but not from HCCs (1.07-fold), and in (b) irradiated HepG2.2.15 cells (17.7-fold) cocultured with human umbilical vascular endothelial cells. Cytokine assay revealed increased expression of interleukin-6 (IL-6) in conditioned medium from irradiated human umbilical vascular endothelial cells. All 16 patients with liver irradiated had the increased serum IL-6 compared with 3 of 10 patients with irradiation excluding liver (P & lt; 0.001). All nine HBV carriers with liver irradiated had postradiotherapy increases in both HBV DNA and IL-6. Conclusions: Radiation-induced liver toxicity with HBV reactivation is from a bystander effect on irradiated endothelial cells releasing cytokines, including IL-6.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2007
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 83-83
    Kurzfassung: Dysregulation of TGF-β signaling plays a crucial role in ovarian carcinogenesis and maintaining cancer stem cell properties. We have previously identified the TGF-β responsive targets in immortalized ovarian surface epithelial cells (IOSE) by expression microarray and found that ABCA1, a family members of ATP-binding cassette is up-regulated following TGF-β treatment (Qin et al, BMC Syst Biol 2009). Thus, we hypothesize that ABCA1 may be involved in ovarian cancer and its initiation. First, we compared the expression level of ABCA1 in IOSE and a panel of ovarian cancer cell lines and found that ABCA1 was up-regulated in HeyC2, SKOV3, MCP3, and MCP2 ovarian cancer cell lines but down-regulated in A2780 and CP70 ovarian cancer cells. The down-regulation in A2780 and CP70 cells was associated with promoter hypermethylation as demonstrated by bisulphite sequencing and demethylation treatment. To investigate if ABCA1 is involved in ovarian cancer initiating cells (OCIC), we compared the methylation level of ABCA1 in ovarian tumor samples and their corresponding spheroids which is previously found to be enriched with OCIC. Lower methylation of ABCA1 could be detected in 2/6 OCIC samples as compared to their original tumors. We further analyzed the expression and methylation level of ABCA1 in CP70 spheroid (CP70sp) derived from suspension culture of CP70 ovarian cancer cells. Comparing to CP70, up-regulation of stem cell markers Nanog (3.8 fold) and Oct4 (12 fold) was detected in CP70sp cells suggesting that CP70sp may contain OCIC. Interestingly, up-regulation of ABCA1 (5.2 fold) concomitant with promoter hypomethylation was detected in CP70sp cells. Additionally, active demethylation may be involved in the hypomethylation of ABCA1 as expression of the methylcytosine dioxygenase, TET1 was up-regulated in the CP70sp cells (5.3 fold). Furthermore, ABCA1 was involved in drug resistance of ovarian cancer which is a feature of cancer initiating cells, as lenti-viral knockdown of ABCA1 in a platinum resistant MCP2 ovarian cancer cells resensitized the cells to cisplatin (IC50: shABCA1 vs shGFP: 0.267 ug/ml vs 0.402 ug/ml). We further analyzed the methylation level of ABCA1 in 97 ovarian cancer, 46 benign, and 4 normal samples using real-time quantitative MSP assay and found that higher methylation level of ABCA1 was detected in cancer than benign (P=0.051) and normal tissues (P & lt;0.05). Importantly, cancer patients showing lower or no methylation of ABCA1 has significantly shorter progression free survival than patients with higher methylation (log rank, P=0.0463). In conclusion, ABCA1 is hypomethylated in a sub-set of ovarian cancer and its cancer initiating cells and is associated with poor prognosis in cancer patients. The role of ABCA1 in OCIC and the involvement of 5-hydroxymethylcytosine (5hmC) in the active demethylation of ABCA1 deserves further investigation. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 83. doi:10.1158/1538-7445.AM2011-83
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2011
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1476-1476
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1476-1476
    Kurzfassung: Ovarian cancer is one of the most lethal cancers in the female reproductive system. One of the hypotheses suggests that ovarian cancer arises from cancer stem/progenitor cell with surface antigen CD117 (or c-kit). In order to identify microRNAs (miRNAs) that can regulate the expression of c-kit, bioinformatic prediction was performed and identified miR-193a as one of the regulators. Expression analysis showed that miR-193a was up-regulated upon TGF-β treatment in immortalized ovarian surface epithelial cells (IOSE) cells. On the contrary, expression of miR-193a cannot be detected in a panel of ovarian cancer cell lines (SKOV3, A2780, CP70, MCP2, MCP3). Bisulphite pyrosequencing revealed that promoter CpG island of miR-193a was not methylated in IOSE cell but hypermethylated in the ovarian cancer cells showing down-regulation of miR-193a. Treatment of 5azaDC, a DNMT inhibitor, partially restored miR-193a expression in the ovarian cancer cells, thus suggesting DNA hypermethylation may be responsible for the down-regulation of miR-193a in ovarian cancer. Further clinical studies using bisulphite pyrosequencing found that promoter region of miR-193a was heavily methylated in 109 ovarian cancer patient samples but not in normal OSE samples. Kaplan-meier analysis revealed that patients with higher methylation of miR-193a was significantly associated with shorter overall survival and progression-free survival. Taken together, miR-193a may be a TGF-β regulated tumor suppressor micro-RNA and is epigenetically silenced in ovarian cancer. Methylation of miR-193a may be able to predict prognosis in ovarian cancer patients. Citation Format: Frank Hsueh-Che Cheng, Gary C.W. Chen, Jian-Liang Chou, Ya-Wen Lin, Lin-Yu Chen, Hung-Cheng Lai, Chin Li, Michael W.Y. Chan. Hypermethylation of a TGF-β regulated micro-RNA, miR-193a, predicts prognosis in ovarian cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1476. doi:10.1158/1538-7445.AM2014-1476
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2014
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1950-1950
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 1950-1950
    Kurzfassung: Lung cancer remains the leading cause of cancer mortality in the world. Metastatic property is one of the major reasons for treatment failure in lung cancer patients. Tumor microenvironment plays a key role in shaping tumor progression. In solid tumors, a decreased oxygen and nutrient supply creates a hypoxic microenvironment in the central region, which elicits a set of genes to endow cancer cells with increased survival or proliferation abilities. On the other hand, TGF-beta, which is often released by tumor stroma cells, attenuates cell proliferation but induces invasion of lung cancer. How cancer cells are shaped by tumor microenvironment with elevated proliferation and invasion abilities are elusive. Here, we report that CITED2, a MYC interacting transcriptional modulator, responds to hypoxia induction and TGF-beta suppression to orchestrate cellular proliferation and invasion, respectively. We observed that hypoxia induced CITED2 expression in a group of lung cancer cells, the proliferation and survival of which were dependent on CITED2 signaling. TGF-beta stimulation inhibited CITED2 expression, causing decreased cell proliferation. Nonetheless, hypoxia potentiated TGF-beta mediated invasion and rendered cell resistant to TGF-beta induced cell growth arrest by elevating CITED2 dependent signaling. Our findings provide that CITED2 functions as an oncogeneic switch for hypoxia and TGF-beta mediated proliferation and invasion in lung cancer, deserving additional evaluation as a biomarker for lung cancer progression. Citation Format: Ming-Han Kuo, Cheng-Han Hsieh, Yuan-Hung Wang, Cheng-Wen Wu, Yu-Ting Chou. Emerging role of CITED2 in hypoxia and TGF-beta mediated proliferation and invasion in lung cancer. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1950. doi:10.1158/1538-7445.AM2015-1950
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2015
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 2 ( 2013-01-15), p. 953-966
    Kurzfassung: Chromosomal instability (CIN) is widely considered a hallmark of cancer, but its precise roles in cancer stem cells (CSC) and malignant progression remain uncertain. BMI1 is a member of the Polycomb group of chromatin-modifier proteins that is essential for stem cell self-renewal. In human cancers, BMI1 overexpression drives stem-like properties associated with induction of epithelial–mesenchymal transition (EMT) that promotes invasion, metastasis, and poor prognosis. Here, we report that BMI1 mediates its diverse effects through upregulation of the mitotic kinase Aurora A, which is encoded by the AURKA gene. Two mechanisms were found to be responsible for BMI1-induced AURKA expression. First, BMI1 activated the Akt pathway, thereby upregulating AURKA expression through activation of the β-catenin/TCF4 transcription factor complex. Second, BMI1 repressed miRNA let-7i through a Polycomb complex-dependent mechanism, thereby relieving AURKA expression from let-7i suppression. AURKA upregulation by BMI1 exerts several effects, including centrosomal amplification and aneuploidy, antiapoptosis, and cell-cycle progression through p53 degradation and EMT through stabilization of Snail. Inhibiting Aurora A kinase activity attenuated BMI1-induced tumor growth in vivo. In clinical specimens of head and neck cancer, we found that coamplification of BMI1 and AURKA correlated with poorer prognosis. Together, our results link CSCs, EMT, and CIN through the BMI1–AURKA axis and suggest therapeutic use from inhibiting Aurora A in head and neck cancers, which overexpress BMI1. Cancer Res; 73(2); 953–66. ©2012 AACR.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2013
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5796-5796
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5796-5796
    Kurzfassung: Introduction The p53 gene is well known as an important tumor suppressor gene. Mutant p53 genes account for about half of all lung cancer cases. There is increasing evidence for the anti-tumor effects of statins via inhibition of the mevalonate pathway. However, the cancer prevention and protection effects of statinsin population-based studies are controversial. Previous population-based studies have shown protective effects of statins in advanced lung cancer patients; however, the effects in early lung cancer patients are unclear. Methods We investigated the correlation between statin use and lung cancer prognosis using the Taiwanese National Health Insurance Research Database retrospectively, mainly focusing on early-stage lung cancer. Cell viability, lipid raft changes, and motile activity from simvastatin-treated cell lines were examined to determine the effect of simvastatin under different p53 mutations. Apoptotic pathways and autophagy in lung cancer cells treated with simvastatin were analyzed using a western blot assay. Results Statin treatments reduced the 5-year mortality (odds ratio, 0.43; 95% confidence interval [CI], 0.37-0.49;P & lt; 0.001) in this population-based study. Simvastatin inhibited cell growth, especially in mutant p53 lung cancer cell lines. Significantly higher levels of cellular apoptosis and decreased lipid rafts were shown in mutant p53 lung cancer cells treated with simvastatin. Further, simvastatin increased activity of the caspase-dependent apoptotic pathway in lung cancer cells containing mutant p53. In addition, simvastatin reduced migration in cells with a mutated p53 gene. Conclusion This study reports the protective effects of statin use in early-stage lung cancer patients regardless of chemotherapy. Simvastatin induced cellular apoptosis and regulated lipid raft content; further, motile activity was reduced in lung cancer cells with p53 missense mutations. These data suggest that statin use in selected lung cancer patients may have clinical benefits. Citation Format: Cheng-wei Chou, Ching-Heng Lin, Tzu-Hung Hsiao, Chia-Chien Lo, Chih-Ying Hsieh, Cheng-Chung Huang, Yuh-Pyng Sher. Therapeutic effects of statins against lung adenocarcinoma via mutant p53 mediated apoptosis [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5796.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2020
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2010
    In:  Clinical Cancer Research Vol. 16, No. 14_Supplement ( 2010-07-15), p. A37-A37
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 16, No. 14_Supplement ( 2010-07-15), p. A37-A37
    Kurzfassung: Radiotherapy is one of the treatment options for hepatocellular carcinoma (HCC), but the unsatisfactory results are mainly due to insufficient dose to the tumor from hepatic intolerance. As compared to advanced technology of radiotherapy, the experience in the combined use of the effective molecular target and radiotherapy remains limited. This study was aimed to assess the effect of combining a novel Aurora kinase inhibitor, VE465, with radiotherapy, by testing the in vitro and in vivo models of human HCC. Human HCC cell line Huh-7, was treated with combined irradiation and the low- toxic dose of VE465. A significant effect on enhancing the radiation induced cell death was found in clonogenic assay. VE-465 induced proliferation blockade, histone H3 (Ser10) dephosphorylation, mitotic disturbance, and apoptosis in Huh-7 cells. The in vivo efficacy of combining VE465 with radiotherapy was investigated in the ectopic xenograft subcutaneous tumor and orthotopic liver tumor models of severe combined immunodeficient mice. Pretreatment with daily i.p. administration of VE465 (20 mg/kg) significantly enhanced the therapeutic effect of radiotherapy (5 Gy per day for 5 days) on Huh-7 derived ectopic xenograft tumor model by 54%. This differential tumor suppression correlated with the modulation of intratumoral biomarker stainings associated with histone 3 phosphorylation inhibition and apoptosis regulation. Furthermore, VE465 improved intrahepatic tumor control and survival of radiotherapy (4 Gy per day for 5 days) treated Huh-7 derived orthotopic liver tumor model by 17.2%, reduction in tumor size. The synergistic effect of combining VE465 with radiotherapy in HCC was associated with histone3 phosphorylation inhibition and cell cycle regulation. We conclude that VE465 is a potent inhibitor of Aurora kinase with clinical value in therapeutic strategies for radiotherapy to HCC. Citation Information: Clin Cancer Res 2010;16(14 Suppl):A37.
    Materialart: Online-Ressource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2010
    ZDB Id: 1225457-5
    ZDB Id: 2036787-9
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...