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
  • 1
    In: Neoplasia, Elsevier BV, Vol. 18, No. 12 ( 2016-12), p. 785-794
    Type of Medium: Online Resource
    ISSN: 1476-5586
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2016
    detail.hit.zdb_id: 2008231-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Molecular Cancer Research Vol. 10, No. 9 ( 2012-09-01), p. 1228-1239
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 10, No. 9 ( 2012-09-01), p. 1228-1239
    Abstract: Mutation of KRAS is a common initiating event in pancreatic ductal adenocarcinoma (PDAC). Yet, the specific roles of KRAS-stimulated signaling pathways in the transformation of pancreatic ductal epithelial cells (PDEC), putative cells of origin for PDAC, remain unclear. Here, we show that KRASG12D and BRAFV600E enhance PDEC proliferation and increase survival after exposure to apoptotic stimuli in a manner dependent on MEK/ERK and PI3K/AKT signaling. Interestingly, we find that activation of PI3K/AKT signaling occurs downstream of MAP–ERK kinase (MEK), and is dependent on the autocrine activation of the insulin-like growth factor (IGF) receptor (IGF1R) by IGF2. Importantly, IGF1R inhibition impairs KRASG12D- and BRAFV600E-induced survival, whereas ectopic IGF2 expression rescues KRASG12D- and BRAFV600E-mediated survival downstream of MEK inhibition. Moreover, we show that KRASG12D- and BRAFV600E-induced tumor formation in an orthotopic model requires IGF1R. Interestingly, we show that while individual inhibition of MEK or IGF1R does not sensitize PDAC cells to apoptosis, their concomitant inhibition reduces survival. Our findings identify a novel mechanism of PI3K/AKT activation downstream of activated KRAS, illustrate the importance of MEK/ERK, PI3K/AKT, and IGF1R signaling in pancreatic tumor initiation, and suggest potential therapeutic strategies for this malignancy. Mol Cancer Res; 10(9); 1228–39. ©2012 AACR.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 24_Supplement ( 2016-12-15), p. B25-B25
    Abstract: Pancreatic ductal adenocarcinoma (PDAC), a poor prognostic cancer, commonly develops following activating mutations in the KRAS oncogene. Activation of the Wnt signaling pathway is also commonly observed in PDAC. To ascertain the impact of postnatal activation of the Wnt signaling pathway in PDAC development, we combined the elastase-tva-based RCAS-TVA pancreatic cancer model with the established LSL-KrasG12D, Ptf1a-cre model. Delivery of RCAS viruses encoding β-cateninS37A and Wnt1 stimulated the progression of premalignant PanIN and PDAC development. Indeed, mice injected with RCAS-β-cateninS37A and Wnt1 had reduced survival relative to RCAS-GFP controls (P & lt; 0.05). Active β-catenin or its DNA-binding partner TCF4 enhanced PDAC cell proliferation, soft-agar colony formation, as well as migration and invasion activity. In contrast, these phenotypes were significantly blocked by the introduction of Icat, an inhibitor of the β-catenin/TCF4 interaction. Meanwhile, we identified Cyr61/CCN1 as a target molecule for Wnt/β-catenin signaling pathway in PDAC. Nuclear β-catenin and Cyr61/CCN1 expression were predominantly detected in moderately to poorly differentiated murine and human PDAC. Indeed, nuclear β-catenin and Cyr61/CCN1-positivity independently correlated with poor prognosis (P & lt; 0.01). Knockdown of Cyr61/CCN1 in a β-catenin-activated PDAC cell line reduced soft agar, migration and invasion activity. Together, these data suggest that the Wnt/β-catenin signaling pathway enhances pancreatic cancer development and malignancy in part via up-regulation of Cyr61/CCN1. Citation Format: Makoto Sano, David R. Driscoll, Wilfredo E. DeJesus-Monge, Brian Quattrochi, Victoria A. Appleman, Jianhong Ou, Lihua Julie Zhu, Nao Yoshida, Shintaro Yamazaki, Tadatoshi Takayama, Masahiko Sugitani, Norimichi Nemoto, David S. Klimstra, Brian C. Lewis.{Authors}. Activation of Wnt/β-catenin signaling pathway enhances pancreatic cancer development and the malignant potential via up-regulation of Cyr61/CCN1. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Adv ances in Science and Clinical Care; 2016 May 12-15; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2016;76(24 Suppl):Abstract nr B25.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 974-974
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 974-974
    Abstract: TAK-164 is an antibody drug conjugate comprised of a full length, fully human IgG1 monoclonal antibody (mAb) directed towards the human extra-cellular domain of guanylyl cyclase C (GCC). The mAb is conjugated using ImmunoGen's peptide-linked indolinobenzodiazepine DNA alkylator DGN549. Previously presented preclinical studies using TAK-164 have demonstrated single agent activity of TAK-164 in a panel of patient derived xenograft models of colorectal cancer grown in immunodeficient mice. With emerging evidence demonstrating the immunostimulatory activity of cytotoxic antibody drug conjugates we sought to explore the potential of TAK-164 in immune competent mice. A CT26 model was engineered to express human GCC, allowing us to test the ability of TAK-164 to modulate the tumor microenvironment, thereby potentiating the activity of checkpoint inhibitors. huGCC-CT26 tumor cells were implanted in the right flank of 6-8 week old BALB/c female mice and randomized when tumors were between 50-150mm3 and treated with a single intravenous administration of TAK-164 alone or in combination with checkpoint inhibition administered Intraperitoneally biweekly for 2 weeks and monitored for tumor growth. To understand if responses observed were driven by a response capable of inducing immunological memory we separated mice with complete responses and rechallenged with CT26 tumor cells on the opposite flank. While memory was observed in the mice that achieved complete response in the single agent groups, both frequency of complete response and tumor rejection was increased in groups treated with a combination of TAK-164 and immune checkpoint inhibition. We performed immunophenotyping studies which revealed broad immunomodulation of lymphoid and myeloid cells by TAK-164 and combination regimens. Collectively these data support further interrogation of immune checkpoint inhibition in combination with TAK-164. Citation Format: Victoria A. Appleman, Michael D. Smith, Michelle Ganno, Tiquella Hatten, Maria Borland, Adnan O. Abu-Yousif. Immune modulation following combination of TAK-164 with checkpoint inhibitors [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 974.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 109, No. 17 ( 2012-04-24)
    Abstract: Our results have broad clinical implications. Small-molecule inhibitors targeting the Smo signaling molecule are being evaluated currently for the treatment of pancreatic cancer ( 5 ). However, the activity of these Smo inhibitors likely is limited to the stromal compartment, having no effect on epithelial cells. Our genetic data identifying an alternative genetic pathway involving Gli argue strongly for the clinical development of Gli inhibitors, because these molecules contribute critical function in the tumor epithelium as well as the tumor stroma. We next investigated which downstream genes mediated Gli-driven phenotypes in pancreatic cancer cells. We performed gene-expression profiling analysis in human pancreatic ductal adenocarcinoma cells in which Gli-mediated transcription had been inhibited by Gli3T. Interestingly, we found that Gli activation in pancreatic cancer cells imposed a unique transcriptional program. Specifically, we found that the expression of the IκB kinase IKBKE (IKKε) was regulated by Gli proteins. These results suggest that IKBKE may play a critical role in pancreatic tumorigenesis. In general, kinases are enzymes that affect other proteins by adding phosphate molecules to these proteins. IKBKE is a noncanonical IkB kinase involved in stimulating the NF-κB signaling pathway. We showed that IKBKE is highly expressed in pancreatic tumors that are characterized by both Kras and Gli activation. We also found that Gli regulates NF-κB activity in vitro and in vivo. Furthermore, we demonstrated that Gli-dependent IKBKE expression in pancreatic cancer cells is essential for maintaining their cell survival and transformation characteristics, which are fundamental properties of cancerous cells. These data establish a connection between Gli transcription factors and the IKBKE and NF-κB activity in tumorigenesis, providing a molecular basis for the observed requirement of Gli transcription in the development of pancreatic ductal adenocarcinoma in vivo. In addition, we evaluated the tumorigenic potential of Gli1 in pancreatic cancer by ectopically expressing Gli1 in the mouse pancreas. We found that Gli1 activation significantly enhanced the ability of Kras to promote tumor development. Thus, both our loss-of-function and gain-of-function analyses highlight the functional importance of epithelial Gli activity in Kras-induced pancreatic tumorigenesis in vivo. However, using a primary pancreatic ductal epithelial cell (PDEC) culture, we showed that Gli-mediated transcription activation is required for Kras-induced PDEC proliferation and survival. Additionally, we combined this model with another in which oncogenic Kras activation in the mouse pancreatic epithelium drove tumor initiation and progression, mimicking the pathogenesis of human pancreatic cancer. Significantly, when these mice were genetically modified to exhibit both activation of Kras and inhibition of Gli transcription in the pancreatic epithelium, we found that the development of pancreatic precursor lesions and pancreatic ductal adenocarcinoma initiated by Kras activation was blocked. This inhibition happened even when the p53 tumor suppressor, which commonly is inactivated in this disease, was deleted. These results provide genetic evidence that Gli activity is required for Kras-driven transformation of the pancreatic epithelium. To investigate the role of Gli in pancreatic tumorigenesis, we generated a mouse model of pancreatic epithelium-specific inhibition of Gli-mediated transcription. This model was achieved by the ectopic expression of a dominant Gli3 repressor allele, Gli3T, which is capable of inhibiting Gli1- and Gli2-mediated activation of transcription. We found that inhibition of Gli transcriptional activity did not affect differentiation of the pancreatic epithelial cell lineages. This lack of effect suggests that epithelial Gli activity is largely dispensable for the proper development of the mouse pancreas. The Gli proteins Gli1, Gli2, and Gli3 are transcription factors. They operate in a cellular signaling pathway called the “Hedgehog” (Hh) pathway that often is activated in human tumors. The Hh pathway is mediated by activation of a cell-surface molecule, Smoothened (Smo) ( 3 ). Stimulation of the Hh pathway results in the activation of Gli proteins. However, Gli proteins may be regulated by other oncogenic pathways. For example, Gli1 gene expression is maintained in pancreatic cancer cells despite deletion of the Smo gene and is regulated in part by Kras independently of Hh ligand input ( 4 ). However, Gli's role in pancreatic tumorigenesis in vivo remains unexplored. Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer-related mortality in the United States ( 1 ). This disease arises from precursor lesions in pancreatic epithelium, characterized by mutations in a potential cancer-causing gene called the “ KRAS oncogene” ( 1 , 2 ). However, limited understanding of the molecular and genetic mechanisms underlying Kras-dependent pancreatic tumorigenesis has impeded the development of early diagnostic and treatment tools. Here, using mouse genetics and cellular biology, we demonstrated that the ability of Gli proteins to activate gene transcription is critical for the development of Kras-driven pancreatic cancer ( Fig. P1 ). Significantly, we identified a key mechanism that operates after Gli transcription activity during pancreatic tumorigenesis: the induction of the expression of the atypical IkB kinase IKBKE and the activity of the NF-κB pathway in the tumor epithelium ( Fig. P1 ).
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2012
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    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...