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: Biomedicine & Pharmacotherapy, Elsevier BV, Vol. 156 ( 2022-12), p. 113974-
    Type of Medium: Online Resource
    ISSN: 0753-3322
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 1501510-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2573-2573
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2573-2573
    Abstract: The ruthenium-based compound KP1339 (trans-(tetrachlorobis(1H-indazole)ruthenate(III)), which is currently in Phase I/II trial, is already known for its promising anticancer activity. In course of a recent screening using diverse cancer cell lines of different origin (n = 25), a small subgroup with exceptional sensitivity to KP1339-treatment was discovered. The aim of the here presented study was to identify factors that underlie this sensitivity or which are connected to resistance to KP1339. Due to the fact that KP1339 comprises a metal center, interpretations of it's mode of action tend towards either generation of ROS or DNA binding. To avoid a biased approach, whole genome expression arrays were performed with mRNA isolated from hypersensitive and normal responsive cells that were either untreated or treated with KP1339 for 3 h and 6 h. Subsequent bioinformatic analysis of the gained data indicated that while hypersensitive cell lines activated pathways indicative for response to chemical stimuli (“metal ion”, “organic compound”), in normal responsive ones preferentially genes involved in pathways controlling cell cycle, DNA repair and metabolism were found. Subsequent investigations confirmed that upon KP1339-treatment hypersensitive cell lines showed pronounced apoptosis induction (indicated by caspase-mediated PARP cleavage and appearance of apoptotic nuclei visualized by DAPI staining). In contrast, normal responsive cells rather reacted with prolonged duration of the G2 phase to KP1339 treatment (video evaluation). This was confirmed by FACS analysis of ethanol-fixed and propidium iodide-stained cells indicating a profound increase in the percentage of cells with double DNA content upon KP1339 treatment. Moreover, in accordance to previous studies, KP1339 induced strong phosphorylation of ERK, p38 and JNK especially in normal responsive cell lines. This is of interest as p38 and JNK activation are known to be pro-survival factors within the extrinsic pathway of apoptosis. The relevance of this particular pathway is supported by preliminary results of activation of caspase 8 upon KP1339 treatment in hypersensitive cells. This knowledge can help to better understand the mechanisms underlying the promising anticancer activity of KP1339. Citation Format: Beatrix Alte, Christine Pirker, Thomas Mohr, Kushtrim Kryeziu, Bernhard K. Keppler, Petra Heffeter, Walter Berger. Investigation of factors involved in the hypersensitivity to KP1339-treatment. [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 2573. doi:10.1158/1538-7445.AM2015-2573
    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 ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 3541-3541
    Abstract: Aim of this study was to test the in vitro and in vivo activity of KP1339, a promising ruthenium-based anti-cancer compound, in combination with the tyrosine kinase inhibitor sorafenib against human hepatoma cells. As a first approach, the drugs were tested together against a hepatoma cell line panel (n=7) by MTT assay. Combination of KP1339 with sorafenib induced additive to synergistic effects in all cell lines tested. Especially at higher sorafenib concentrations (10 µM) strong synergism with CI values between 0.1 and 0.5 were observed. Notably, synergistic activity with KP1339 was found in sorafenib-resistant as well as sorafenib-responsive cell lines. Recently, we have presented that KP1339 treatment induced phosphorylation of P38, JNK and erk and led to cell cycle arrest in G2/M Phase. Consequently, the effects of sorafenib on KP1339-induced P38 phosphorylation were investigated. Western blot analyses revealed that sorafenib strongly reduced phosphorylation of P38 and erk after KP1339 treatment. Moreover, addition of sorafenib reduced KP1339-induced G2/M arrest. Finally, the activity of the KP1339/sorafenib combination was evaluated in xenograft experiments (Hep3B cells). KP1339 monotreatment led to a 2.4-fold increase in life span (mean survival 80 days vs. 33 days in control) and thus was superior to sorafenib monotherapy, which induced a 1.9-fold survival increase (60 days). Combination of KP1339 with sorafenib increased the mean survival by 3.9-fold to 96 days. Together, our data indicate that the combination of KP1339 with Sorafenib is very promising in vitro and in vivo especially against human hepatoma cells. 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 3541. doi:10.1158/1538-7445.AM2011-3541
    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
    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 ...
  • 5
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 1992
    In:  Journal of Cancer Research and Clinical Oncology Vol. 118, No. 3 ( 1992-3), p. 195-200
    In: Journal of Cancer Research and Clinical Oncology, Springer Science and Business Media LLC, Vol. 118, No. 3 ( 1992-3), p. 195-200
    Type of Medium: Online Resource
    ISSN: 0171-5216 , 1432-1335
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 1992
    detail.hit.zdb_id: 1459285-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: European Journal of Cancer, Elsevier BV, Vol. 49, No. 15 ( 2013-10), p. 3366-3375
    Type of Medium: Online Resource
    ISSN: 0959-8049
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2013
    detail.hit.zdb_id: 1120460-6
    detail.hit.zdb_id: 1468190-0
    detail.hit.zdb_id: 82061-1
    Location Call Number Limitation 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. 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 ...
  • 8
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4395-4395
    Abstract: Chemotherapeutic drugs exert their activity by directly killing tumor cells through a variety of cellular targets eventually leading to apoptosis. Apoptosis is regarded as non-inflammatory and non-immunogenic, however, recent data confirm that the induction of damage-associated molecular patterns by dying cancer cells treated with certain chemotherapeutic agents, such as oxaliplatin, lead to their recognition by the immune system, and result in dendritic cell maturation and immune responses, a mechanism described as Immunogenic Cell Death (ICD). The main hallmarks of ICD are: Calreticulin (CRT) exposure on the cell membrane and release of high mobility group box-1 (HMGB-1) and ATP from dying cells. We have recently shown that the mode of action of KP-1339, a clinically investigated ruthenium-based metal complex, involves the enhancement of endoplasmic reticulum stress (ERS) and the induction of apoptosis-like events. We next investigated the ability of KP-1339 to trigger ICD. There is currently no reliable method to study ICD in vitro, and therefore we aimed at a more suitable model that better mimics the tumor microenvironment in comparison to 2D monolayer cultures. For this reason, ICD was assessed in human colorectal cancer HCT-116 3D multicellular spheroids in vitro. We demonstrate that spheroids treated with KP-1339 activate the PERK/Phospho-eIF-2alpha pathway of the ERS, a prerequisite for CRT exposure on the cell membrane. HMGB1 protein levels decrease considerably during a 72 hours drug treatment, as shown by Immunoblotting, suggesting the depletion of the protein. Additionally, treated spheroids released significantly more ATP than untreated controls. Immunofluorescence of cryosections of KP-1339 treated spheroids stained for CRT and HMGB1 confirmed the above observations and, clearly showed the translocation of CRT to the cell membrane. In conclusion, we have established a 3D tumor spheroid model as an alternative to the 2D monolayer cultures to study ICD in vitro. This model more closely resembles the tumor microenvironment and recapitulates relevant aspects of the in vivo situation. We successfully employed this model to demonstrate that the clinically investigated compound KP-1339 triggers the main hallmarks of ICD, which suggests enhanced induction anti-tumor immunity in colorectal cancer. Citation Format: Debora Wernitznig, Giorgia Del Favero, Konstantinos Kiakos, Nathalie Harrer, Herwig Machat, Doris Marko, Michael Jakupec, Wolfgang Sommergruber, Bernhard K. Keppler. KP-1339 (IT-139) induces the hallmarks of immunogenic cell death in a colon cancer 3D model in vitro [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4395.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    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 ...
  • 10
    In: The Lancet, Elsevier BV, Vol. 400, No. 10363 ( 2022-11), p. 1607-1617
    Type of Medium: Online Resource
    ISSN: 0140-6736
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 2067452-1
    detail.hit.zdb_id: 3306-6
    detail.hit.zdb_id: 1476593-7
    SSG: 5,21
    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...