Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    Walter de Gruyter GmbH ; 2001
    In:  Biological Chemistry Vol. 382, No. 1 ( 2001-01-06)
    In: Biological Chemistry, Walter de Gruyter GmbH, Vol. 382, No. 1 ( 2001-01-06)
    Type of Medium: Online Resource
    ISSN: 1431-6730
    Language: Unknown
    Publisher: Walter de Gruyter GmbH
    Publication Date: 2001
    detail.hit.zdb_id: 1466062-3
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Rockefeller University Press ; 2007
    In:  The Journal of Cell Biology Vol. 177, No. 1 ( 2007-04-09), p. 127-137
    In: The Journal of Cell Biology, Rockefeller University Press, Vol. 177, No. 1 ( 2007-04-09), p. 127-137
    Abstract: We demonstrate a role for protein kinase casein kinase 2 (CK2) in the phosphorylation and regulation of the M3-muscarinic receptor in transfected cells and cerebellar granule neurons. On agonist occupation, specific subsets of receptor phosphoacceptor sites (which include the SASSDEED motif in the third intracellular loop) are phosphorylated by CK2. Receptor phosphorylation mediated by CK2 specifically regulates receptor coupling to the Jun-kinase pathway. Importantly, other phosphorylation-dependent receptor processes are regulated by kinases distinct from CK2. We conclude that G protein–coupled receptors (GPCRs) can be phosphorylated in an agonist-dependent fashion by protein kinases from a diverse range of kinase families, not just the GPCR kinases, and that receptor phosphorylation by a defined kinase determines a specific signalling outcome. Furthermore, we demonstrate that the M3-muscarinic receptor can be differentially phosphorylated in different cell types, indicating that phosphorylation is a flexible regulatory process where the sites that are phosphorylated, and hence the signalling outcome, are dependent on the cell type in which the receptor is expressed.
    Type of Medium: Online Resource
    ISSN: 1540-8140 , 0021-9525
    RVK:
    Language: English
    Publisher: Rockefeller University Press
    Publication Date: 2007
    detail.hit.zdb_id: 1421310-2
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Immunopharmacology, Elsevier BV, Vol. 35, No. 1 ( 1996-10), p. 23-26
    Type of Medium: Online Resource
    ISSN: 0162-3109
    Language: English
    Publisher: Elsevier BV
    Publication Date: 1996
    detail.hit.zdb_id: 1483922-2
    Library Location Call Number Volume/Issue/Year 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. 1786-1786
    Abstract: The receptor tyrosine kinase MET is a prime target in clinical oncology because of its aberrant activation in a broad spectrum of malignancies. Here, we tested the anti tumor activity of a novel MET inhibitor, EMD1214063, on cancer cells overexpressing MET and in eight transfected NIH3T3 cell lines expressing different activating mutants of the MET kinase. Our results demonstrate a dose-dependent decrease in MET autophosphorylation in both MET overexpressing cell lines (IC50 of 20nM and 30nM for GTL-16 and H1993, respectively) and in six of the eight cell lines transfected with MET activating mutants (IC50 5-18nM). Blockade of MET by EMD1214063 resulted in reduced downstream activation of AKT, ERK and PLCγ. In contrast, lack of MET inhibition by EMD1214063 in the resistant Y1248H and L1213V MET mutants was associated with high levels of AKT, ERK and PLCγ activation. In all sensitive mutants, EMD1214063 attenuated MET-dependent cellular proliferation and significantly altered cell cycle distribution profiles (increase in G1 and concomitant decrease of S phase). Furthermore, EMD1214063 strongly affected MET-driven biological functions, such as cellular morphology, motility and anchorage-independent growth. To assess the in vivo efficacy of EMD1214063, we established tumor transplantation models exploiting NIH3T3 cells expressing the H1112L or L1213V MET mutants, respectively sensitive and resistant, to in vitro treatment with EMD1214063. Animals bearing H1112L or L1213V tumors were randomized for the treatment with EMD1214063 (50mg/kg/day) or vehicle only. Remarkably, 5 days of treatment with EMD1214063 induced complete regression of sensitive H1112L tumors, while tumor growth remained unaffected in mice injected with resistant L1213V tumor cells and in vehicle-treated mice. Taken together, these results underscore the concordance between in vitro and in vivo effects of EMD1214063 and strongly support its efficacy in inhibiting MET-driven tumor cell growth. 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 1786. doi:1538-7445.AM2012-1786
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 925-925
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 925-925
    Abstract: The mesenchymal-epithelial transition factor (c-Met) receptor, also known as hepatocyte growth factor receptor (HGFR), controls morphogenesis, a process physiologically required for embryonic development and tissue repair. Aberrant c-Met activation is associated with human malignancies, including hepatocellular carcinomas (HCC). The aim of this study was to evaluate the effect of a novel, highly selective c-Met inhibitor, MSC2156119J (EMD 1214063), in hepatocellular cancer models. MHCC97H is a human HCC cell line co-expressing c-Met and HGF, and exhibiting high metastatic potential. The effect of MSC2156119J on MHCC97H was evaluated in a Balb/c nu/nu murine xenograft model in a subcutaneous and orthotopic setting. Tumor-bearing mice were orally treated with 100 mg/kg MSC2156119J (5 days on/2 days off). In the subcutaneous setting, MSC2156119J resulted in tumor regression in 10/10 mice, achieving tumor eradication in 9/10 mice. In the orthotopic model, MHCC97H tumor fragments proliferate in the liver and invariably metastasize to the lung (100%). MSC2156119J treatment started 7 days after tumor fragment implantation. After 5 weeks of treatment, mice were necropsied and a series of parameters were assessed. Primary tumor size and weight, and circulating Alpha-Feto-Protein levels were significantly lower in mice treated with MSC2156119J compared to controls (p & lt;0.001). Notably, treatment with MSC2156119J resulted in a reduced number of mice with visible lung metastasis (6/9), and correlated with a significantly lower number of metastatic foci in the lungs compared to control mice (p & lt;0.01). To better predict the clinical efficacy of MSC2156119J, 9 patient-derived primary HCC explants models were used. The explants were subdivided into 3 groups, based on their c-Met levels and HGF expression. Each group comprised 3 tumors expressing high, intermediate, and low levels of c-Met and HGF, respectively. Mice were treated with MSC2156119J, sorafenib, or a combination of both. MSC2156119J (100 mg/kg/5 out of 7 days) strongly inhibited tumor growth in 4/9 models (TGD range of 370% to 41%). Analysis of intratumoral c-Met, phospho c-Met and HGF levels indicated that explants with high levels of c-Met and HGF were more sensitive to MSC2156119J than low-expressing models. In 3/4 responsive models, MSC2156119J exhibited a better anti-tumor activity than sorafenib, while sorafenib was more efficacious in a model characterized by intermediate c-Met/HGF expression. MSC2156119J was not efficacious in models exhibiting low or no signs of c-Met signalling. When used in combination, MSC2156119J did not enhance the activity of sorafenib. MSC2156119J monotherapy was well tolerated while sorafenib alone and in combination with MSC2156119J induced significant body weight loss. Overall, these data indicate that MSC2156119J may be a valuable therapeutic option for liver cancers with high levels of c-Met expression/activation. Citation Format: Friedhelm Bladt, Andree Blaukat, Dieter Dorsch, Manja Friese-Hamim, Michael Meyring, Oliver Schadt. The c-Met inhibitor MSC2156119J effectively inhibits growth of liver cancer models. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 925. doi:10.1158/1538-7445.AM2013-925
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Molecular Cancer Research Vol. 17, No. 12 ( 2019-12-01), p. 2457-2468
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 17, No. 12 ( 2019-12-01), p. 2457-2468
    Abstract: Inhibition of DNA double-strand break (DSB) repair in cancer cells has been proposed as a new therapeutic strategy for potentiating the anticancer effects of radiotherapy. M3814 is a novel, selective pharmacologic inhibitor of the serine/threonine kinase DNA-dependent protein kinase (DNA-PK), a key driver of nonhomologous end-joining, one of the main DSB-repair pathways, currently under clinical investigation. Here, we show that M3814 effectively blocks the repair of radiation-induced DSBs and potently enhances p53 phosphorylation and activation. In p53 wild-type cells, ataxia telangiectasia–mutated (ATM) and its targets, p53 and checkpoint kinase 2 (CHK2), were more strongly activated by combination treatment with M3814 and radiation than by radiation alone, leading to a complete p53-dependent cell-cycle block and premature cell senescence. Cancer cells with dysfunctional p53 were unable to fully arrest their cell cycle and entered S and M phases with unrepaired DNA, leading to mitotic catastrophe and apoptotic cell death. Isogenic p53-null/wild-type A549 and HT-1080 cell lines were generated and used to demonstrate that p53 plays a critical role in determining the response to ionizing radiation and M3814. Time-lapse imaging of cell death and measuring apoptosis in panels of p53 wild-type and p53-null/mutant cancer lines confirmed the clear differences in cell fate, dependent on p53 status. Implications: Our results identify p53 as a possible biomarker for response of cancer cells to combination treatment with radiation and a DNA-PK inhibitor and suggest that p53 mutation status should be considered in the design of future clinical trials. Visual Overview: http://mcr.aacrjournals.org/content/molcanres/17/12/2457/F1.large.jpg.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2786-2786
    Abstract: The involvement of the mesenchymal endothelial transition factor (c-Met) in the primary event of oncogenic transformation and the secondary ability to mediate metastatic spread has been convincingly demonstrated in preclinical and early clinical settings. The clinical benefits of c-Met kinase inhibitors with various modes of actions and selectivity profiles are currently under investigation hoping that inhibitors of c-Met might emerge as valuable cancer therapeutics in the future. During an HTS run 3-(diethylamino)propyl N-[3-[[5-(3,4-dimethoxyphenyl)-2-oxo-6H-1,3,4-thiadiazin-3-yl]methyl] phenyl]carbamate was identified as an attractive lead structure with an interesting overall profile (clogD (7.4): 2.5, S (pH 7.4): & gt;100 µg/ml, IC50 (cMet enzyme): 30 nM, IC50 (cMet A549): 800 nM) providing a valid starting point. The co-crystal structure of 1 revealed the binding mode and the essential structural features. The initial HTS hit was bound in a DFG-in conformation interacting with the main chain nitrogen atom of Met1160 within the hinge region and with the main chain nitrogen of Asp1222. After subsequent optimization of potency, efficacy, PK properties and the safety profile of thiadiazinone 1, EMD1204831 was identified as development compound. EMD1204831 is currently being investigated in a phase 1 clinical trial. The pyridazinone EMD1204831 inhibits enzymatic and cellular c-Met kinase activity with IC50 values of 12 nM and 15 nM, respectively. EMD1204831 displayed an exquisite selectivity when tested in vitro against a panel of more than 400 potential off-targets, including kinases, GPCRs, ion channels, transporters and various enzymes. EMD 1204831 demonstrated excellent anti-tumor activity in vivo in a variety of xenograft models, including U87-MG glioblastoma cells (autocrine HGF expression), TPR-Met-transformed mouse fibroblasts (oncogenic Met fusion protein) or Hs746T gastric cancer cells (c-Met gene amplification and HGF-independent activation). Depending on the particular model, complete regressions were observed with doses as low as 6 mg/kg/d administered per os. PK/PD analysis revealed efficient, dose- and time-dependent inhibition of c-Met phosphorylation, reduction of IL-8 and cyclin D1 expression as well as an induction of the cell cycle inhibitor p27. The overall profile of EMD1204831 including first time structural disclosure, some structure activity relationships, in vitro potency, selectivity profile and in-vivo data will be presented. 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 2786. doi:10.1158/1538-7445.AM2011-2786
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 6212-6212
    Abstract: Ataxia telangiectasia and Rad3 related (ATR) and ataxia telangiectasia mutated (ATM) protein kinases play key roles in the DNA damage response (DDR) by responding to replication stress and double-strand DNA breaks respectively to pause the cell cycle and promote DNA repair. ATR kinase inhibition in cancer cells disrupts cell cycle control and causes unrepaired DNA lesions and cytotoxicity. Accordingly, several ATR inhibitors (ATRi) are in clinical development as monotherapies and in combination with DNA damaging chemotherapies and poly(ADP-ribose) polymerase (PARP) inhibitors. Here we show that while treatment of cancer cells with an ATRi inhibits the ATR pathway, it simultaneously activates the ATM signaling pathway as shown by increased levels of p-ATM, p-CHK2, p-KAP1 and p-p53. In p53 wild-type cancer cells, ATR inhibition causes an ATM-mediated G1 cell cycle arrest which diminishes the DNA lesions and cytotoxic effects of ATR inhibition. Combination of an ATRi with a selective ATM inhibitor (ATMi) synergistically potentiated efficacy in cancer cells in vitro and increased efficacy in vivo at doses that did not show overt toxicities. In a panel of patient-derived xenograft (PDX) models of triple-negative breast cancer treated with an ATRi/ATMi combination, substantial improvement in efficacy was observed. Thus, activation of the ATM pathway by an ATRi acts as a compensatory resistance pathway for ATR inhibition. These results suggest a novel and efficacious combination approach for cancer therapy by dual inhibition of these two key DDR kinases. Citation Format: Audrey Turchick, Astrid Zimmermann, Li-Ya Chiu, Heike Dahmen, Brian Elenbaas, Frank T. Zenke, Andree Blaukat, Lyubomir T. Vassilev. Inhibition of ATM-dependent checkpoint control and DNA double-strand break repair enhances the efficacy of ATR inhibitors. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 6212.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2724-2724
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2724-2724
    Abstract: Introduction: MSC2156119J, a highly selective, potent, reversible, ATP-competitive c-Met inhibitor currently under clinical testing, efficiently inhibits c-Met phosphorylation and downstream signaling in vivo and induces regression of established tumors in xenograft models in preclinical studies (Bladt et al. Clin Cancer Res. 2013;19:2941-51). One initial event following c-Met activation is Y1234/35 phosphorylation in the activation loop of the kinase domain. This results in kinase activation and triggers phosphorylation of tyrosine residues in the c-Met C-terminal tail (eg, Y1349), creating multifunctional docking sites for intracellular adapters. The development of an assay allowing the measurement of these phospho-c-Met epitopes in tumor biopsies will be crucial for establishing an optimal biologic dose for MSC2156119J. Methods: We describe here the development of a highly sensitive Luminex assay that can reliably and reproducibly measure the phosphorylation state of Y1234/35 and Y1349 residues of the c-Met receptor. The high sensitivity of the assay was confirmed by measuring the phospho-c-Met levels in biopsies from patients before and during MSC2156119J treatment. Results: We first assessed the stability of phosphorylated Y1234/35 and Y1349 epitopes in preclinical tumor samples. Analyses revealed that the total c-Met protein is rather stable over time, while the phospho-c-Met epitopes Y1234/35 and Y1349 are relatively unstable; their detection requires rapid processing of tumor samples derived from preclinical tumors or from patients. In tumor samples derived from human xenografts treated with MSC2156119J, Y1349 phosphorylation inhibition varied in different tumor models and did not show strong dose dependence. In contrast, phosphorylation of Y1234/35 residues was effectively inhibited by MSC2156119J in all tumor models tested. Based on these findings, phosphorylation of c-Met Y1234/35 was also assessed in tumor biopsies from patients treated with MSC2156119J in the first-in-man trial (NCT01014936; Falchook et al. J Clin Oncol. 2013;31(suppl):2506). When compared to pretreatment tumor biopsies, on-treatment samples showed effective, dose-dependent inhibition of & gt;90% of c-Met Y1234/35 phosphorylation. Conclusions: We successfully developed an assay capable of detecting phosphorylation of the c-Met Y1234/35 epitope, which is crucial for c-Met activation. Using this assay on tumor samples from xenograft models and on patient-derived tumor biopsies, we demonstrated that MSC2156119J inhibits phosphorylation of c-Met Y1234/35 in a dose-dependent fashion. Therefore, phosphorylation of c-Met Y1234/35 can be used as a pharmacodynamic biomarker of c-Met inhibition and will be an important and valuable element for the selection of the optimal biologic dose of MSC2156119J. Citation Format: Friedhelm Bladt, Frank Jaehrling, Manja Friese-Hamim, Gerald S. Falchook, Hesham M. Amin, Manfred B. Klevesath, Andree Blaukat. Development of a sensitive assay for measuring pharmacodynamic modulation of c-Met in biopsies. [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 2724. doi:10.1158/1538-7445.AM2014-2724
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4183-4183
    Abstract: M3814 is a potent and selective inhibitor of DNA-PK, one of the key cellular regulators of DNA damage induced by ionizing radiation or certain cytostatics used in the treatment malignant disease. One of these drugs, Etoposide, induces double strand breaks (DSB) in cellular DNA. DSBs are most difficult to repair and, if left unrepaired, can lead to induction of cell cycle arrest and/or apoptosis and ultimately cell death. DNA-PK plays a critical role in the repair of DSB via the non-homologous end-joining pathway. M3814 was tested for activity in combination with Etoposide in a panel of 98 cancer cell lines derived from lung cancer. A broad potentiation effect of DNA-PK inhibitor was observed in most cancer cell lines. As a rule, cell lines sensitive to Etoposide demonstrated increased sensitivity to the combination. M3814 did not show significant effect on cancer cell growth/viability in combination with Cisplatin compared to Cisplatin alone at concentrations that effectively inhibit DNA-PK activity. At the same time, M3814 did not negatively affect the antitumor activity of Cisplatin. The therapeutic effect of M3814 in combination with the standard of care (SoC) regimen of Etoposide and Cisplatin was tested in the human small cell lung cancer xenograft model, NCI-H520. Triple combination of M3814, Etoposide and Cisplatin resulted in increased efficacy compared to SoC treatment arm. Since myeloid and lymphoid suppression is one of the dose limiting toxicities of the SoC regimen in patients, the effect of the triple combination on myeloid and lymphoid blood cells was investigated in immunocompetent mice. Whereas the SoC treatment showed reduction of the myeloid and lymphoid compartments, the addition of M3814 did not additionally reduce these cells neither in the treatment nor the recovery phase. Our results warrant further investigations to explore the potential of the combination in the clinical setting. Citation Format: Christian Sirrenberg, Astrid Zimmermann, Thomas Grombacher, Lyubomir T. Vassilev, Lars Damstrup, Frank T. Zenke, Andree Blaukat. A novel selective DNA-PK inhibitor, M3814, as a potential combination partner of Etoposide and Cisplatin in the therapy of lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4183. doi:10.1158/1538-7445.AM2017-4183
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages