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)  (5)
  • Mohri, Dai  (5)
Material
Publisher
  • American Association for Cancer Research (AACR)  (5)
Language
Years
Subjects(RVK)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2381-2381
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2381-2381
    Abstract: Patients with pancreatic ductal adenocarcinoma have a poor prognosis. New therapeutic approaches to improve the prognosis are expected. We generated pancreas-specific Tgfbr2 knockout mice in the context of Kras activation, which recapitulated clinical and histopathological manifestations of human pancreatic cancer. Using this model we investigated the anti-tumor effects of gemcitabine and molecular targeted drugs. We treated the mice with EGFR inhibitor elrotinib or VEGFR inhibitor axitinib in combination with gemcitabine. Median survival times were 45, 60, 77, and 74 days for control, gemcitabine alone, gemcitabine + axitinib, and gemcitabine + erlotinib, respectively. In vitro assays, elrotinib inhibited phosphorylation of EGFR and both drugs also inhibited proliferation of human pancreatic cancer cell lines in vitro.This mouse model also recapitulated chemosensitivity of human pancreatic cancer and suggested erlotinib and axitinib in combination with gemcitabine can be superior to gemcitabine alone. This mouse model helps us to investigate the mechanism of the action of drugs and chemoresistance. 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 2381. doi:10.1158/1538-7445.AM2011-2381
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 7 ( 2013-04-01), p. 2221-2234
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly cancers worldwide. Although many regimens have been used for PDAC treatment, the combination of the EGF receptor (EGFR) inhibitor erlotinib with gemcitabine has been the only molecular-targeted drug tested so far that has been superior to gemcitabine alone. The mechanism underlying this effective combinational regimen remains unknown. Here, we show that the combination is superior to gemcitabine alone in blocking progression and prolonging survival in a murine model of PDAC (Kras activation with Tgfbr2 knockout). We found that gemcitabine induced mitogen-activated protein kinase signaling, which was dramatically inhibited by erlotinib even in the Kras-activated PDAC cells in the mouse model. Mechanistic investigations suggested that gemcitabine induces EGFR ligand expression and ERBB2 activation by increasing heterodimer formation with EGFR, thereby maintaining high levels of ERBB2 protein in PDAC cells. Overall, our findings suggest a significant role of ERBB in PDAC treatment. Cancer Res; 73(7); 2221–34. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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. 71, No. 8_Supplement ( 2011-04-15), p. 544-544
    Abstract: Pancreatic ductal adenocarcinoma (PDAC), one of the most lethal neoplasms, is characterized histologically by expanded stromal components with marked fibrosis, which suggests that tumor-stromal interaction might play an important role in PDAC progression. We have established pancreas epithelium-specific transforming growth factor-ß receptor type II (Tgfbr2) knockout mice in the context of active Kras (KrasG12D) expression (Kras+Tgfbr2KO), which developed differentiated PDAC with 100% penetrance and recapitulated histology of human PDAC described above. In the AACR annual meetings 2009-10, we have reported that tumor-stromal interaction in the PDAC model promoted tumor progression and blockade of CXC chemokines/CXCR2 axis inhibited tumor growth and extended survival of the mice. We further examined the underlying mechanisms of tumor-stromal interaction. The CXC chemokines secreted from the PDAC cells did not change autocrine PDAC cell proliferation nor that of stromal fibroblasts, but induced CTGF expression in the stromal fibroblasts and promoted tumor angiogenesis, which resulted in promotion of tumor growth. Expression of the CXC chemokines was downregulated by TGF-β signaling, whereas, NF-κB signaling was required for the expression. Subcutaneous xenografts coinjected with the PDAC cells and fibroblasts grew faster than those with PDAC cells alone, which suggested tumor-promoting tumor-stromal interaction. We further examined the xenograft growth of CXCR2-knocked down (KD) PDAC cells and CXCR2-intact firbroblasts, compared with that of CXCR2-intact PDAC cells and CXCR2-KD fibroblasts. Results suggested that intact CXCR2 in the stromal fibroblasts was required for the tumor-promoting CXC chemokines/CXCR2 axis. In conclusion, blockade of the CXC chemokines/CXCR2 axis might have a key role in modulating the tumor microenvironment and for development of more effective therapeutic strategy for PDAC. 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 544. doi:10.1158/1538-7445.AM2011-544
    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 ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 851-851
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly cancers. New therapeutic approaches to improve the prognosis are highly required. We generated pancreas-specific TGF-beta receptor II (Tgfbr2) knockout mice in the context of Kras activation, which recapitulated clinical and histopathological manifestations of human PDAC, indicating that an impact of blockade of TGF-beta signaling in PDAC progression. An EGFR inhibitor erlotinib showed a superior survival effect in combination with gemictabine, however, the mechanism by which PDAC with extremely frequent KRAS-mutation benefits from EGFR inhibition remains largely unkown. Using the model we investigated the anti-tumor impact and mechanisms of erlotinib in combination with gemcitabine. We treated the mice with elrotinib and gemcitabine. Median survival times were 52.5, 69, and 74 days for control, gemcitabine alone, and gemcitabine + erlotinib, respectively. Gemcitabine alone significantly extended the survival compared to the control. Further, adding erlotinib to gemcitabine prolonged the survival significantly compared to gemcitabine alone. Interestingly, treatment with gemcitabine enhanced the activation of MAPK signaling, which was inhibited by adding erlotinib, even in the KRAS-mutant PDAC in vivo and in vitro. We observed that the gemcitabine-induced MAPK signaling activation was due to induction of EGFR ligands expression and also due to ERBB2 activation in PDAC cells. Erlotinib inhibited the ERBB2 activation, partly by inhibiting heterodimer formation with EGFR and also decreasing ERBB2 protein expression in PDAC cells. This mouse model appears recapitulating chemosensitivity of human PDAC and indicates the significance of ERBBs in PDAC treatment. This model also might provide important insights into the beneficial patient subpopulation and the mechanism of the action of drugs and chemoresistance, in which TGF-beta signaling might be highly involved. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 851. doi:1538-7445.AM2012-851
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 12, No. 12_Supplement ( 2014-12-01), p. B10-B10
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is the most deadly cancer worldwide. Although many regimens have been tried against PDAC, an epidermal growth factor receptor (EGFR) inhibitor erlotinib in combination with gemcitabine is the only molecular target drug superior to gemcitabine alone. However, the mechanism by which PDAC with extremely frequent KRAS-mutation benefits from EGFR inhibition remains largely unknown. In this study, we evaluated the efficacy of erlotinib in combination with gemcitabine using a murine PDAC model with transforming growth factor-beta receptor II knockout plus Kras activation and investigated the mode of action. The mice were treated using the following drug doses and treatment schedules; erlotinib was administered from 3 weeks of age and gemcitabine was administered from 4 weeks of age. We isolated PDAC cells from the murine PDAC tissues. Effects of erlotinib on the proliferation and intracellular signaling of the murine PDAC cells or human PDAC cell lines were examined in vitro. We sacrificed the mice at 7 weeks of age, and excised the pancreatic tissues and processed for western blot analysis and immunohistochemistry. We evaluated the expression of EGFR ligands by real-time PCR and the heterodimer formation of EGFR with ErbB2 by immunoprecipitaiton after incubation with gemcitabine in vitro. We assessed whether the effect of gemcitabine on EGFR/ErbB2 activation is secondary to mitogen activated protein kinase (MAPK) signal activation after incubation with or without MEK inhibitor and gemcitabine by western blot analysis and real-time PCR. Gemcitabine + erlotinib inhibited PDAC progression and significantly prolonged the survival of the PDAC mice compared to gemcitabine alone. Gemcitabine or erlotinib also inhibited in vitro PDAC cell proliferation. Interestingly, Gemcitabine induced MAPK signaling, which was dramatically inhibited by adding erlotinib, even in the Kras-mutant PDAC cells. The suggested mechanisms were that gemcitabine induced EGFR ligand expression and also ErbB2 activation by increasing heterodimer formation with EGFR and maintaining high ErbB2 protein level in PDAC cells. We observed that the gemcitabine-induced MAPK signaling activation was in part due to induction of Egfr ligands(Egf, Tgf-a, Amphiregulin) expression by real-time PCR and ELISA. Using a phospho-RTK antibody array, we also observed that Gem induced Erbb2 activation in PDAC cells, and validated by western blot analysis, real-time PCR, and immunohistochemistry. Erlotinib inhibited the ErbB2 activation, partly by inhibiting heterodimer formation with EGFR and also decreasing ErbB2 protein expression in PDAC cells. We observed that gemcitabine-induced EGFR ligands up-regulation and EGFR/ErbB2 activation require intact MAPK signaling and these are secondary effects of MAPK signal activation and that gemcitabine induced the activation irrespective of KRAS status and gemcitabine sensitivity. This model helps us to evaluate an efficacy of new drugs and to investigate mechanisms of the mode of action and chemoresistance. This study provides clinical insights into potent therapeutic strategies for this difficult cancer. Citation Format: Koji Miyabayashi, Hideaki Ijichi, Ryota Takahashi, Dai Mohri, Keisuke Yamamoto, Yoshinari Asaoka, Tsuneo Ikenoue, Keisuke Tateishi, Harold L. Moses, Kazuhiko Koike. Epidermal growth factor receptor inhibitor prolongs survival in pancreatic cancer by blocking gemcitabine-induced mitogen-activated protein kinase signal. [abstract]. In: Proceedings of the AACR Special Conference on RAS Oncogenes: From Biology to Therapy; Feb 24-27, 2014; Lake Buena Vista, FL. Philadelphia (PA): AACR; Mol Cancer Res 2014;12(12 Suppl):Abstract nr B10. doi: 10.1158/1557-3125.RASONC14-B10
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    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...