GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
  • American Association for Cancer Research (AACR)  (57)
Material
Publisher
  • American Association for Cancer Research (AACR)  (57)
Language
Subjects(RVK)
  • 1
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 7, No. 3 ( 2009-03-01), p. 371-382
    Abstract: Functional suppression of spindle checkpoint protein activity results in apoptotic cell death arising from mitotic failure, including defective spindle formation, chromosome missegregation, and premature mitotic exit. The recently identified p31comet protein acts as a spindle checkpoint silencer via communication with the transient Mad2 complex. In the present study, we found that p31comet overexpression led to two distinct phenotypic changes, cellular apoptosis and senescence. Because of a paucity of direct molecular link of spindle checkpoint to cellular senescence, however, the present report focuses on the relationship between abnormal spindle checkpoint formation and p31comet-induced senescence by using susceptible tumor cell lines. p31comet-induced senescence was accompanied by mitotic catastrophe with massive nuclear and chromosomal abnormalities. The progression of the senescence was completely inhibited by the depletion of p21Waf1/Cip1 and partly inhibited by the depletion of the tumor suppressor protein p53. Notably, p21Waf1/Cip1 depletion caused a dramatic phenotypic conversion of p31comet-induced senescence into cell death through mitotic catastrophe, indicating that p21Waf1/Cip1 is a major mediator of p31comet-induced cellular senescence. In contrast to wild-type p31comet, overexpression of a p31 mutant lacking the Mad2 binding region did not cause senescence. Moreover, depletion of Mad2 by small interfering RNA induced senescence. Here, we show that p31comet induces tumor cell senescence by mediating p21Waf1/Cip1 accumulation and Mad2 disruption and that these effects are dependent on a direct interaction of p31comet with Mad2. Our results could be used to control tumor growth. (Mol Cancer Res 2009;7(3):371–82)
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2009
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 6083-6083
    Abstract: Background: There is no study so far that analyzed the patterns of clonal evolution from the initiation of carcinogenesis to distant metastasis in patients with gallbladder adenocarcinoma (GBAC). This study aimed to elucidate the evolutionary trajectories of GBAC using multi-regional and longitudinal tumor samples. Methods: Whole-exome sequencing was conducted on tumors and matched normal samples with the coverage of 300x and 200x, respectively. Using PyClone, CITUP, MapScape, and TimeScape, phylogenetic trees were visualized in each patient considering each tumor’s location and timing of acquisition. Mutational signatures were analyzed using Mutalisk. Results: Between 2013 and 2018, a total of 11 patients (male, 5) including 2 rapid autopsy cases were enrolled. The median age was 70 (range, 59-75) years. The dataset consisted of 11 normal samples, 4 biliary intraepithelial neoplasia (BilIN), 11 primary tumors, and 30 metastatic tumors. The most frequently altered gene was ERBB2 (54.5%), followed by TP53 (45.5%) and FBXW7 (27.3%). Of 6 patients with analyzable tumor ploidy, 2 patients (33.3%) had whole genome doubling (WGD) in both primary and metastatic tumors, and 1 patient (16.7%) had WGD not in the primary tumor but in the liver metastasis. In the BilIN analysis (n=4), most mutations in frequently altered genes in GBAC were detectable from the BilIN stage, but some of them were subclonal. In these 4 patients, the fittest subclone in BilIN underwent linear and branching evolution by acquiring additional subclonal mutations and thus expanded in the primary tumor, suggesting a selective sweep phenomenon. In combined analysis with metastatic tumors (n=11), branching and linear evolution was identified in 9 (81.8%) and 2 (18.2%) patients, respectively. Of the 9 patients with branching evolution, eight (88.9%) had a total of 11 subclones expanded at least 7-fold in regional or distant metastasis. These subclones harbored putative metastasis-driving mutations in tumor suppressor genes such as SMAD4, ROBO1, and DICER1. Metastases were polyclonal in all patients. However, metastatic lesions in the same or adjacent organs showed similar clonal compositions and there was evidence of metastasis-to-metastasis spread. In mutational signature analysis, we identified 6 mutational signatures: signatures 1 (age), 3 (DNA double-strand break-repair), 7 (ultraviolet), 13 (APOBEC), 22 (aristolochic acid), and 24 (aflatoxin) (cosine similarity values ≥ 0.9). Among them, signatures 1 and 13 were dominant at the carcinogenesis stage while signatures 22 and 24 were dominant at the metastasis stage. Conclusions: We have constructed evolutionary trajectories of individual patients, highlighting the role of each clone and the dynamics among clones during carcinogenesis and metastasis. This novel approach may help us move forward to precision medicine that enables early detection of carcinogenesis and metastasis. Citation Format: Minsu Kang, Hee Young Na, Soomin Ahn, Ji-Won Kim, Sejoon Lee, Soyeon Ahn, Ju Hyun Lee, Jeonghwan Youk, Haesook T. Kim, Kui-Jin Kim, Koung Jin Suh, Jun Suh Lee, Se Hyun Kim, Jin Won Kim, Yu Jung Kim, Keun-Wook Lee, Yoo-Seok Yoon, Jee Hyun Kim, Jin-Haeng Chung, Ho-Seong Han, Jong Seok Lee. Evolutionary trajectories during carcinogenesis and metastasis in gallbladder adenocarcinoma [abstract] . In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 6083.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 4 ( 2019-02-15), p. 795-806
    Abstract: Tumor hypoxia and aerobic glycolysis are well-known resistance factors for anticancer therapies. Here, we demonstrate that tumor-associated macrophages (TAM) enhance tumor hypoxia and aerobic glycolysis in mice subcutaneous tumors and in patients with non–small cell lung cancer (NSCLC). We found a strong correlation between CD68 TAM immunostaining and PET 18fluoro-deoxyglucose (FDG) uptake in 98 matched tumors of patients with NSCLC. We also observed a significant correlation between CD68 and glycolytic gene signatures in 513 patients with NSCLC from The Cancer Genome Atlas database. TAM secreted TNFα to promote tumor cell glycolysis, whereas increased AMP-activated protein kinase and peroxisome proliferator-activated receptor gamma coactivator 1-alpha in TAM facilitated tumor hypoxia. Depletion of TAM by clodronate was sufficient to abrogate aerobic glycolysis and tumor hypoxia, thereby improving tumor response to anticancer therapies. TAM depletion led to a significant increase in programmed death-ligand 1 (PD-L1) expression in aerobic cancer cells as well as T-cell infiltration in tumors, resulting in antitumor efficacy by PD-L1 antibodies, which were otherwise completely ineffective. These data suggest that TAM can significantly alter tumor metabolism, further complicating tumor response to anticancer therapies, including immunotherapy. Significance: These findings show that tumor-associated macrophages can significantly modulate tumor metabolism, hindering the efficacy of anticancer therapies, including anti-PD-L1 immunotherapy.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 13 ( 2014-07-01), p. 3556-3566
    Abstract: Identifying immune escape mechanisms used by tumors may define strategies to sensitize them to immunotherapies to which they are otherwise resistant. In this study, we show that the antiapoptotic gene API5 acts as an immune escape gene in tumors by rendering them resistant to apoptosis triggered by tumor antigen-specific T cells. Its RNAi-mediated silencing in tumor cells expressing high levels of API5 restored antigen-specific immune sensitivity. Conversely, introducing API5 into API5low cells conferred immune resistance. Mechanistic investigations revealed that API5 mediated resistance by upregulating FGF2 signaling through a FGFR1/PKCδ/ERK effector pathway that triggered degradation of the proapoptotic molecule BIM. Blockade of FGF2, PKCδ, or ERK phenocopied the effect of API5 silencing in tumor cells expressing high levels of API5 to either murine or human antigen-specific T cells. Our results identify a novel mechanism of immune escape that can be inhibited to potentiate the efficacy of targeted active immunotherapies. Cancer Res; 74(13); 3556–66. ©2014 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Prevention Research Vol. 4, No. 9 ( 2011-09-01), p. 1507-1521
    In: Cancer Prevention Research, American Association for Cancer Research (AACR), Vol. 4, No. 9 ( 2011-09-01), p. 1507-1521
    Abstract: Colitis-associated cancer (CAC) is one of clear examples of inflammation–carcinogenesis sequence, by which the strict control of colitis with potent anti-inflammatory or antioxidative agent offers the chance of cancer prevention. Supported with the facts that Rac1 binds and activates STAT3, which are significantly upregulated in inflammatory bowel disease (IBD) as well as CAC, but 8-hydroxydeoxyguanosine (8-oxo-7,8-dihydrodeoxyguanosine or 8-OHdG) paradoxically can block Rac1 activation and subsequent NADPH oxidase (NOX) inactivation in various inflammation models, we hypothesized that attenuated Rac1–STAT3 and COX–NF-κB pathway by exogenous 8-OHdG administration may ameliorate inflammatory signaling in dextran sodium sulfate (DSS)-induced colitis and can prevent CAC. Before commencing carcinogenesis model, we checked whether exogenous 8-OHdG can alleviate IBD, for which interleukin (IL)-10 knockout mice were designed to ingest 5% DSS for 1 week, and 8-OHdG is given through intraperitoneal route daily. 8-OHdG treatment groups significantly reduced pathologic grade of DSS-induced colitis as well as various inflammatory mediators such as TNF-α, IL-6, COX-2, and iNOS in a dose-dependent manner. To document the cancer prevention effects of 8-OHdG, mice were injected azoxymethane followed by drinking 2.5% DSS for 1 week, after which 8-OHdG–containing diets were given for 20 weeks. As results, mice that consumed 8-OHdG–containing diet significantly reduced both tumor incidence and multiplicity. Rac1 activity and phosphorylated STAT3 level were significantly attenuated in the 8-OHdG–treated group. Significantly decreased levels of malondialdehyde, monocyte chemotactic protein-1, matrix metalloproteinasess, COX-2, NOX4, and β-catenin nuclear accumulation were responsible for cancer prevention effects of exogenous 8-OHdG. In conclusion, we clearly showed cancer-preventive effect of exogenous 8-OHdG against CAC. Cancer Prev Res; 4(9); 1507–21. ©2011 AACR.
    Type of Medium: Online Resource
    ISSN: 1940-6207 , 1940-6215
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2422346-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Prevention Research Vol. 4, No. 10_Supplement ( 2011-10-01), p. B49-B49
    In: Cancer Prevention Research, American Association for Cancer Research (AACR), Vol. 4, No. 10_Supplement ( 2011-10-01), p. B49-B49
    Abstract: Colitis-associated cancer (CAC) is one of clear examples of inflammation-carcinogenesis sequence, by which the strict control of colitis with potent anti-inflammatory or anti-oxidative agent offers the chance of cancer prevention. Supported with the facts that Rac1 binds and activates STAT3 which are significantly up-regulated in inflammatory bowel disease (IBD) as well as CAC, but 8-hydroxydeoxyguanosine (8-OHdG) paradoxically can block Rac1 activation and subsequent NOX inactivation in various inflammation models, we hypothesized that attenuated Rac1-STAT3 and COX-NF-κB pathway by exogenous 8-OHdG administration may ameliorate inflammatory signaling in dextran sodium sulfate (DSS) induced colitis and can prevent CAC. Before commencing carcinogenesis model, we checked whether exogenous 8-OHdG can alleviate IBD, for which IL-10 knock-out mice were designed to ingest 5% DSS for 1 week, and 8-OHdG is given through intraperitoneal route daily. 8-OHdG treatment groups significantly reduced pathologic grade of DSS-induced colitis as well as various inflammatory mediators such as TNF-α, IL-6, COX-2 and iNOS in a dose-dependent manner. In order to document the cancer prevention effects of 8-OHdG, mice were injected azoxymethane followed by drinking 2.5% DSS for 1 week, after which 8-OHdG-contained diets were given for 20 weeks. As results, mice which consumed 8-OHdG-contained-diet significantly reduced both tumor incidence and multiplicity. Rac1 activity and phosphorylated STAT3 level were significantly attenuated in 8-OHdG treated group. Significantly decreased levels of MDA, MCP-1, MMPs, COX-2, NOX4, and β-catenin nuclear accumulation were responsible for cancer prevention effects of exogenous 8-OHdG. In conclusion, we clearly showed cancer preventive effect of exogenous 8-OHdG against CAC. Citation Information: Cancer Prev Res 2011;4(10 Suppl):B49.
    Type of Medium: Online Resource
    ISSN: 1940-6207 , 1940-6215
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2422346-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 21, No. 23 ( 2015-12-01), p. 5391-5403
    Abstract: Purpose: Previous study identified E2F1 as a key mediator of non–muscle-invasive bladder cancer (NMIBC) progression. The aim of this study was to identify the E2F1-related genes associated with poor prognosis and aggressive characteristics of bladder cancer. Experimental Design: Microarray analysis was performed to find E2F1-related genes associated with tumor progression and aggressiveness in the gene expression data from 165 primary patients with bladder cancer. The biologic activity of E2F1-related genes in tumor progression and aggressiveness was confirmed with experimental assays using bladder cancer cells and tumor xenograft assay. Results: The expression of E2F1 was significantly associated with EZH2 and SUZ12. The overexpression of E2F1, EZH2, and SUZ12 enhanced cancer progression including cell colony formation, migration, and invasiveness. Knockdown of these genes reduced motility, blocked invasion, and decreased tumor size in vivo. E2F1 bound the proximal EZH2 and SUZ12 promoter to activate transcription, suggesting that E2F1 and its downstream effectors, EZH2 and SUZ12, could be important mediators for the cancer progression. In addition, we confirmed an association between these genes and aggressive characteristics. Interestingly, the treatment of anticancer drugs to the cells overexpressing E2F1, EZH2, and SUZ12 induced the expression of CD44, KLF4, OCT4, and ABCG2 known as cancer stem cell (CSC)–related genes. Conclusions: The link between E2F1, EZH2, and/or SUZ12 revealed that E2f1 directly regulates transcription of the EZH2 and SUZ12 genes. The signature of E2F1–EZH2–SUZ12 shows a predictive value for prognosis in bladder tumors and the E2F1–EZH2–SUZ12–driven transcriptional events may regulate the cancer aggressiveness and chemo-resistance, which may provide opportunity for development of new treatment modalities. Clin Cancer Res; 21(23); 5391–403. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3051-3051
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3051-3051
    Abstract: Background: The active metabolite of vitamin D, 1, 25-dihydroxyvitamin D3 (calcitriol), inhibits the growth of several types of human cancer cells in vitro, but its therapeutic use is limited because it causes hypercalcemia. Among its analogs, 19-nor-1, 25-dihydroxyvitamin D2 (paricalcitol) induces fewer calcemic effects and exhibits an activity that is equipotential to that of calcitriol in several in vivo and in vitro systems. We investigated the antitumor activity and mechanism of action of paricalcitol in gastric cancer cells. Methods: We examined the effects of paricalcitol on cell proliferation, cell cycle, apoptosis, and inflammation/immunity in the human gastric cancer cell lines AGS, SNU719, and MKN45. Results: In vitro treatment with paricalcitol was associated with the following: (1) a dose-dependent increase in vitamin D receptor (VDR) protein expression; (2) inhibited gastric cell growth and induced apoptosis in AGS, SNU719, and MKN45 cell lines; (3) increased expression of p21 and p27 and decreased expression of CDK2; (4) increased expression of the caspase-3 cleaved protein and decreased expression of the anti-apoptotic Bcl-2 and Bax proteins; (5) promotion of early and late apoptosis; and (6) decreased expression of STAT-3, JAK2, COX2, and NF-κB. Conclusion: The low-calcemic vitamin D analog, paricalcitol, exhibits anticancer activity against gastric cancer cells, and its mechanism of action may be mediated through the VDR. Paricalcitol is a promising agent for the prevention and treatment of gastric cancer. 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 3051. doi:10.1158/1538-7445.AM2011-3051
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2007
    In:  Molecular Cancer Therapeutics Vol. 6, No. 1 ( 2007-01-01), p. 82-92
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 6, No. 1 ( 2007-01-01), p. 82-92
    Abstract: Resistance to anticancer drugs can sometimes be overcome by combination treatment with other therapeutic drugs. Here, we showed that phytosphingosine treatment in combination with arsenic trioxide (As2O3) enhanced cell death of naturally As2O3-resistant human myeloid leukemia cells. The combination treatment induced an increase in intracellular reactive oxygen species level, mitochondrial relocalization of Bax, poly(ADP-ribose) polymerase-1 (PARP-1) activation, and cytochrome c release from the mitochondria. N-acetyl-l-cysteine, a thiol-containing antioxidant, completely blocked Bax relocalization, PARP-1 activation, and cytochrome c release. Pretreatment of 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone, a PARP-1 inhibitor, or PARP-1/small interfering RNA partially attenuated cytochrome c release, whereas the same treatment did not affect Bax relocalization. The combination treatment induced selective activation of p38 mitogen-activated protein kinase (MAPK). Inhibition of p38 MAPK by treatment of SB203580 or expression of domi nant-negative forms of p38 MAPK suppressed the combination treatment–induced Bax relocalization but did not affect PARP-1 activation. In addition, antioxidant N-acetyl-l-cysteine completely blocked p38 MAPK activation. These results indicate that phytosphingosine in combination with As2O3 induces synergistic apoptosis in As2O3-resistant leukemia cells through the p38 MAPK–mediated mitochondrial translocation of Bax and the PARP-1 activation, and that p38 MAPK and PARP-1 activations are reactive oxygen species dependent. The molecular mechanism that we elucidated in this study may provide insight into the design of future combination cancer therapies to cells intrinsically less sensitive to As2O3 treatment. [Mol Cancer Ther 2007;6(1):82–92]
    Type of Medium: Online Resource
    ISSN: 1535-7163 , 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2007
    detail.hit.zdb_id: 2062135-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4252-4252
    Abstract: Background: Class III β tubulin (TUBB3) reduces microtubule stability and confers resistance to microtubule stabilizing taxanes, including paclitaxel and docetaxel. Expression of vascular endothelial growth factor (VEGF) is highly regulated by hypoxia and the regulation of VEGF expression by hypoxia is mediated by a hypoxia inducible factor-1a(HIF-1a), which increase transcription of the VEGF gene. Recent report showed hypoxia induces expression of TUBB3 by HIF-1a. Inhibition of VEGF system may decrease the expression of HIF-1a and TUBB3. This study was undertaken to investigate the synergistic antitumor effects of bevacizumab and palitaxel on TUBB3 expressing gastric cancer cells. Methods: We evaluated the expressions of TUBB3 and HIF-1a under normoxic and hypoxic conditions in human gastric cancer cells. The chemoresistance of TUBB3 to paclitaxel was checked through knockdown of TUBB3. Expressions of TUBB3 and HIF-1a were measured under blocking of VEGFR1/2 with anti-FLT1 and anti-KDR antibodies. The effects of bevacizumab and paclitaxel on expressions of TUBB3 and HIF1a were monitored by western blot and MTT assay. Results: The chemosensitivities to paclitaxel were increased by siTUBB3. Elevated expressions of TUBB3 and HIF-1a in gastric cancer cells under hypoxic conditions were reduced in the prescence of anti-KDR and anti-FLT1 antibodies. When we added the bevacizumab to paclitaxel, the protein expressions of TUBB3 and HIF-1a in gastric cancer cells under hypoxic conditions were significantly decreased as compared with paclitaxel alone. Apoptosis of gastric cancer cells were more increased in a combined treatment with bevacizumab and paclitaxel than paclitaxel alone. Tyrosine phosphorylation of Erk and Akt was also measured in gastric cancer cells under hypoxic conditions in the absence or prescence of anti-KDR, anti FLT-1 and paclitaxels. Hypoxia induced phosphorylation of Erk and Akt in gastric cancer cells was more decreased by addition of anti FLT-1 and anti KDR antibodies to paclitaxel as compared with paclitaxel alone. Conclusion: Our experiments show that a VEGF/VEGFR/pERK and, or pAKT pathway is activated by hypoxic gastric cancer cells to induce HIF-1a and TUBB3 expressions. Combined treatment with paclitaxel and bevacizumab, a recombinant humanized monoclonal antibody that binds to VEGF, synergistically decreased the expressions of HIF-1a and TUBB3 in gastric cancer cells in hypoxic conditions. With increasing use of taxanes in the treatment of gastric cancer, this study showed the potential synergistic antitumor effects of bevacizumab and paclitaxel in a TUBB3, a marker of resistance to paclitaxel, overexpressing gastric cancer. 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 4252. doi:10.1158/1538-7445.AM2011-4252
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...