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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4397-4397
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4397-4397
    Abstract: Chemotherapy and therapy with small targeted molecules are two major strategies for therapy of human cancer at the disseminated stage. During the last decades, thousands of compounds have been developed and, consequently, have improved therapy effectiveness. Especially the development of receptor tyrosine kinase inhibitors such as erlotinib or imatinib were major step forwards in cancer treatment. However, despite its success EGFR inhibitor therapy is still limited by strong side effects, resistance development and insufficient tumor accumulation. Aim of the here presented study was the development of novel EGFR inhibitors, which are specifically activated in the malignant tissue. To this end a cobalt(III)-based prodrug strategy was used, which allows targeted release of the active EGFR inhibitor triggered by hypoxic conditions of the solid tumor. As a first step, new inhibitors with bis-chelating moieties were prepared and tested for their efficacy against several cell models with differing EGFR status. The most promising lead candidate was selected based on potent kinase inhibition (confirmed by in vitro kinase assays as well as Western blotting) resulting in activity against EGFR-driven cells in the nM range (MTT assay). Subsequently, the respective cobalt complex was prepared and its activity tested in hypoxia vs. normoxia revealing that the new complex was distinctly more active under hypoxic conditions. Finally, the anticancer activity of the new complex was tested in two xenografts indicationg potent and hypoxia-dependent anticancer activity also in vivo. Summarizing, cobalt(III)-based tumor-targeting represents a promising strategy to reduce the side effects of tyrosine kinase inhibitors such as erlotinib. Acknowledgements. This work was performed in course of the research platform “Translational Cancer Therapy Research” Vienna, Austria and the COST action CM1105 and supported by “Fonds der Stadt Wien für innovative interdisziplinäre Krebsforschung”. Citation Format: Petra Heffeter, Claudia Karnthaler-Benbakka, Diana Groza, Kushtrim Kryeziu, Walter Berger, Bernhard K. Keppler, Christian R. Kowol. Preclinical development of a novel hypoxia-activated EGFR inhibitor using a cobalt(III)-based prodrug design. [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 4397. doi:10.1158/1538-7445.AM2015-4397
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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. 71, No. 8_Supplement ( 2011-04-15), p. 2637-2637
    Abstract: The α-N-heterocyclic thiosemicarbazone Triapine (3-aminopyridine-2-carboxaldehyde thiosemicarbazone) has been studied in several phase I & II clinical trials and it is known as a potent ribonucleotide reductase inhibitor. We synthesized Triapine (3-AP) and the corresponding terminally N4-dimethylated derivative (3-AP-Me2). Previously, we have shown that dimethylation of the terminal amino group leads to significant enhancement of cytotoxicity. In this study we utilized the intrinsic fluorescence properties of these compounds to investigate their intracellular distribution and studied the ability of both compounds to induce endoplasmic reticulum (ER) stress in SW480 colon carcinoma cells. Fluorescence microscopy was performed on viable SW480 cells (colon carcinoma) treated with 3-AP and 3-AP-Me2, showing a strong affinity of the compounds to the nuclear membrane and to cytosolic structures. Co-localization studies with organelle-specific staining revealed that both agents are associated with structures of ER and mitochondria. Therefore, we hypothesized an interference with ER functions. Immunoblotting showed that treatment with the dimethylated compound (3-AP-Me2) results in increased expression of BIP/GRP78 and induction of CHOP. Both are relevant stress markers for unfolded protein response (UPR), and CHOP indicates induction of apoptosis via the mitochondrial pathway. In contrast, treatment with 3-AP leads to an inhibition of BIP/GRP78 expression and does not induce CHOP expression. However, both compounds cause splicing of XBP-1 mRNA indicating activation of the IRE1-α pathway, which triggers apoptosis by activation of the ASK1-JNK pathway. In accordance with these findings 3-AP as well as 3-AP-Me2 activates c-Jun N-terminal kinase (JNK), which is commonly associated with cellular stress such as UPR. A dichlorofluorescein assay showed no generation of reactive oxygen species (ROS) neither by 3-AP nor by 3-AP-Me2, suggesting that the initiation of the ER stress pathway is ROS independent. In conclusion, dimethylation of the terminal amino group leads to up regulation of ER stress mechanism by increase of BIP/GRP79 expression and induction of CHOP. Activation of CHOP indicates induction of apoptosis and may be crucial for the higher cytotoxicity of dimethylated derivates of thiosemicarbazones. XBP-1 splicing and phosphorylation of JNK suggest an activation of the IRE1α-ASK1-JNK signaling pathway, which is connected to cell death involving proteins of the BCL-2 family. The induction of ER stress in a ROS independent manner is a novel mechanism of action relevant for the anti-neoplastic activity of this class of compounds. 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 2637. doi:10.1158/1538-7445.AM2011-2637
    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 ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 1939-1939
    Abstract: Triapine, which is currently tested in a clinical phase III trial, is the best-studied thiosemicarbazone (TSC) for anticancer therapy. With regard to the mode of action, anticancer activity of TSCs is frequently linked to their ability to chelate essential metal ions such as copper and iron. As Triapine monotherapy showed promising results mainly against hematological diseases, novel TSC derivatives have been developed and clinically investigated for their activity against solid tumors. These novel TSCs belong to a subclass with enhanced anticancer activity in vitro and in vivo which were recently discovered to induce paraptosis, a form of programmed but caspase-independent cell death. As these TSCs are characterized by a up to 1000-fold higher activity in cell culture compared to Triapine, the aim of this study was to elucidate the mechanism of action as well as the underlying structural and chemical requirements of these effects. For this purpose, a panel of structurally related Triapine derivatives was examined for anticancer activity (as metal-free ligand and copper(II) complex), paraptosis-inducing potential as well as solution stability and redox properties of their copper(II) complexes. Correlation studies between these chemical and biological properties revealed that the increased anticancer activity and paraptosis-inducing potential of the nanomolar active TSCs is related to a higher copper(II) complex solution stability and slower reduction rate. Unexpectedly, the TSCs with lower activity produced higher superoxide levels in a cell-free setting. This paradox could be explained by their lower copper(II) complex stability and increased readiness to be reduced, which resulted in a fast reduction of intracellular complexes and release of the metal-free ligand. Although this process resulted in the generation of superoxide, cell damage seemed to be prevented by rapid upregulation of the superoxide dismutase (in vitro and in vivo). In contrast, the copper complexes of the highly active TSCs are stable enough to reach intracellular targets such as the ER-resident protein disulfide isomerase, whose inhibition is crucial for paraptosis induction by TSCs. In conclusion, copper complex stability is a crucial parameter of TSC activity, influencing the (intracellular) formation or dissociation of copper complexes. This intracellular stability of complexes, affects their mechanism of action as well as cell death induction. Overall, this study points out the importance of the redox parameters in order to understand and predict the TSC anticancer activity as well as their mechanism of action and, thereby, will pave the way for the development of improved anticancer agents. Citation Format: Sonja Hager, Veronika F. Pape, Vivien Pósa, Bianca Montsch, Lukas Uhlik, Gergely Szakàcs, Szilárd Tóth, Nikolett Jabronka, Bernhard K. Keppler, Walter Berger, Christian R. Kowol, Éva A. Enyedy, Petra Heffeter. Improved activity and paraptosis-induction of anticancer thiosemicarbazones requires high copper(II) complex stability and slow reduction kinetics [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 1939.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...