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)  (6)
  • Medicine  (6)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 350-350
    Abstract: Traditionally, tumors have been classified based on their tissue of origin and their histopathological characteristics and consequently patients are divided into groups where they are treated identically despite substantial differences in genetic profiles. Identification of genes causally implicated in cancer development has led to the development of ‘stratified’ medicine, which adjusts a patient’s therapy based on biomarkers that identify some of the patients expected not respond to a specific therapy. However, many tumor genome sequencing projects have shown that many more gene mutations drive the development of cancer than previously thought, making every tumor unique. These results in low success rates and thereby high cost in drug approval as the identified drugs are only effective for particular patient groups while large patient cohorts have no clear clinical benefit. In the early process of drug development, drugs are therefore screened on large cancer cell line collections to define drug applicability and to determine potential tumor targets. Here, we report the results of a computational modeling platform, ModCell, allowing the prediction of individual drug effects using large-scale genomic and transcriptomic data to virtualize such cell line screening. We validated this approach two-fold: on publicly available data from the cancer cell line encyclopedia, that comprises pharmacological profiles for anti-cancer drugs across a library of cancer cell lines as well as on cancer cell line culture experiments. We provide evidence that ModCell is able to reproducibly predict the effects of individual drugs with a confounding 80% accuracy, to predict combinatory drug action and to identify new applications for existing drugs. Thus, computational modeling using ModCell can improve todays drug development by accelerating and partly replacing work which would have otherwise be conducted in the laboratory and in the clinic. Citation Format: Alexander Kuehn, Felix Dreher, Svetlana Peycheva, Reha Yildiriman, Verena Lehmann, Thomas Kessler, Christoph Wierling, Hans Lehrach, Bodo MH Lange. Virtualization of drug testing by predictive systems biology modeling for optimal drug treatment of cancer cells and drug repositioning. [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 350. doi:10.1158/1538-7445.AM2014-350
    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
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 16_Supplement ( 2018-08-15), p. A021-A021
    Abstract: Background and Rationale: While impressive improvements in immunology have led to the identification of immune checkpoint proteins such as CTLA-4 and PD-1/PD-L1 that trigger inhibitory T cell antitumor activity, these methods do not work well in PCa patients, underscoring the need to devise novel, safe ways to stimulate the immune system to attack tumors. Recent data shows that PCa tumors use lipid to grow and secrete signals that inactivate the immune cells, so they cannot expand, activate, and attack the tumors. Specifically, activation of lipid oxidation via CPT1A in T cells leads to decreased expansion and anticancer action, suggesting that blocking the ability of tumor and infiltrated T cells to burn lipid will likely stimulate the immune system. Our study goal was to study systemic therapies in mice that block the ability of cells to burn lipid in a safe, nontoxic manner but with significant anticancer effects. Experimental Approach: In order to study the role of the immune system, we used TRAMPC1 cells, a mouse PCa cell line that is syngeneic with C57BL/6 mice and grows well in allografts. Additionally, we generated Cpt1a Knockdown (KD) TRAMPC1 cells and cocultured them with mouse splenocytes to investigate the role of cytokines in the tumor-immune cell. We used etomoxir and perhexiline as inhibitors of CPT1. Ranolazine, an FDA-approved partial fat oxidation inhibitor, was also used in the studies. Infiltrating tumor T-cell phenotypes were examined by flow cytometry and immunofluorescence, staining for CD3, CD4, CD8, T-cell checkpoint molecules PD-1. Results: In order to study the role of the immune system, we used TRAMPC1 PCa cells, which we have found to express abundant Cpt1a and have been used successfully for PCa studies We have found that TRAMPC1 cells are sensitive to treatments with fat oxidation inhibitors (ranolazine, etomoxir, and perhexiline), resulting in a decrease of colony growth over 14 days (50% decrease in growth, p & lt; 0.01 compared to vehicle). Furthermore, in vitro studies incubating T cells with CPT1 inhibitors showed a dose-dependent decrease in CD4+ T cells with minimal effects on CD8+ T cells, suggesting that these drugs may reduce CD4+ T regulatory T cells and promote cytotoxic attack to the tumors. To study these metabolic effects in vivo, we generated tumor allografts with TRAMPC1 cells. We observed significant infiltration of T cells (CD3+) into the tumors, and significant decrease in tumor volume when the mice were systemically treated with ranolazine (40 mg/kg/day) for 21 days. Lastly, to gain mechanistic insight of the role of fat oxidation in this paradigm, we generated TRAMPC1 Cpt1aKD cells using lentiviral shRNAs. We found that coculturing Cpt1aKD cancer cells with murine splenocytes significantly decreased the PD1 receptor in the T cells (40% and 50% decrease vs. controls for CD4+ and CD8+ T cells, respectively, p ≤ 0.05) indicating a potential boost in antitumor activity with decreased fat oxidation mediated by Cpt1a. Ongoing studies are exploring these mechanisms. Conclusions: The inability to mount an efficient immune response that restricts cancer progression is partially due to the presence of nonactive effector T cells present in tumors. Our results indicate that PCa tumors use lipid to grow and secrete signals that inactivate the immune cells, so they cannot expand and attack tumors. Concomitantly, activation of lipid oxidation in T cells leads to decreased expansion and anticancer action, suggesting that blocking both, the ability of tumors and infiltrated T cells to burn lipid, will likely stimulate the immune system to reduce tumor burden. Thus, nontoxic strategies that modify the lipid bioenergetics of immune cells and/or tumor cells themselves offer strong potential to improve or synergize with other forms of therapy, and increase the efficacy of treatments against PCa. Citation Format: Amanda M. Guth, Maren Salzmann-Sullivan, Camille Beaton, Amir Goldkorn, Emily A. Gibson, Elizabeth Kessler, Elaine Lam, Thomas Flaig, Rajesh Agarwal, Isabel R. Schlaepfer. CPT1A-mediated fat oxidation and its role in the immune response to prostate cancer [abstract]. In: Proceedings of the AACR Special Conference: Prostate Cancer: Advances in Basic, Translational, and Clinical Research; 2017 Dec 2-5; Orlando, Florida. Philadelphia (PA): AACR; Cancer Res 2018;78(16 Suppl):Abstract nr A021.
    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 ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 3091-3091
    Abstract: Myc is an oncogenic transcription factor frequently dysregulated in human malignancies. While the transcriptional programs and other functions of Myc have been intensively studied, there remains no effective strategy for inhibiting Myc in patients. To search for pathways that support the Myc oncogenic program, we employed a next-generation RNAi screen for Myc-synthetic lethal (MySL) genes. Using this strategy, we have identified several cellular processes required to tolerate oncogenic Myc. Key among these is the core sumoylation machinery, and we define the Sumo-activating enzyme (SAE) as a central component in this MySL network. Loss of SAE drives synthetic lethality with Myc, and the enzymatic activity of SAE is required to support the Myc oncogenic state. Inactivation of SAE leads to mitotic catastrophe and cell death selectively upon Myc hyper-activation. Mechanistically, depletion of SAE switches a subprogram of Myc transcriptional targets governing mitotic spindle function from activated to repressed, a subprogram we term Sumoylation-dependent Myc Switchers, or SMS genes. Notably, SMS genes are required to tolerate Myc hyper-activation, and SAE and the SMS program are required for Myc-dependent breast cancer cell survival in vitro and tumor growth and progression in vivo. Importantly, patient survival significantly correlates with levels of SAE and SMS gene expression in Myc-high tumors. Collectively, these studies reveal a mitotic vulnerability of Myc-driven cancers and demonstrate that inhibiting sumoylation can selectively impair mitosis and survival in an oncogenic Myc-dependent manner. We propose that drugs targeting SAE and its downstream SMS targets may have therapeutic benefits for patients with Myc-driven cancer. 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 3091. doi:1538-7445.AM2012-3091
    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 ...
  • 4
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 25, No. 1 ( 2019-01-01), p. 253-265
    Abstract: Resistance is an obstacle of glioma therapy. Despite targeted interventions, tumors harbor primary resistance or become resistant over short course of treatment. This study examined the mouse double minute 2 (MDM2) inhibitor RG7388 together with radiotherapy and analyzed strategies to overcome acquired MDM2 inhibitor resistance in glioblastoma. Experimental Design: Effects of RG7388 and radiotherapy were analyzed in p53 wild-type glioblastoma cell lines and glioma-initiating cells. RG7388 resistant cells were generated by increasing RG7388 doses over 3 months. Regulated pathways were investigated by microarray, qRT-PCR, and immunoblot analysis and specifically inhibited to evaluate rational salvage therapies at RG7388 resistance. Effects of RG7388 and trametinib treatment were challenged in an orthotopical mouse model with RG7388 resistant U87MG glioblastoma cells. Results: MDM2 inhibition required functional p53 and showed synergistic activity with radiotherapy in first-line treatment. Long-term exposure to RG7388 induced resistance by activation of the extracellular signal-regulated kinases 1/2 (ERK1/2)–insulin growth factor binding protein 1 (IGFBP1) signaling cascade, which was specifically overcome by ERK1/2 pathway inhibition with trametinib and knockdown of IGFBP1. Combining trametinib with continued RG7388 treatment enhanced antitumor effects at RG7388 resistance in vitro and in vivo. Conclusions: These data provide a rationale for combining RG7388 and radiotherapy as first-line therapy with a specific relevance for tumors insensitive to alkylating standard chemotherapy and for the addition of trametinib to continued RG7388 treatment as salvage therapy after acquired resistance against RG7388 for clinical practice.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. LB-080-LB-080
    Abstract: The innate immune response plays a critical role in modulating the efficacy of and mechanisms conferring resistance to immune checkpoint blockade (ICB) therapies in humans. A major negative regulator of the Interferon (IFN) stimulated gene (ISG) pathway, is the ubiquitin-specific protease 18 (USP18). USP18 is the predominant human deubiquitylating enzyme that processes Interferon Stimulated Gene 15 ISG15, a ubiquitin-like protein that covalently modifies protein substrates, a tightly regulated process in the context of innate immunity. In this study, using advanced mass spectrometry and chemical biology tools, we defined the USP18 Interactome and ISG15ylome in chronic myeloid leukemia (CML)-derived cells (HAP1) treated with Interferon alpha (IFNα). Novel ISG15ylation targets were characterized that reduce the sensing of innate ligands and secretion of cytokines. In addition, we show that USP18 deletion leads to enhanced ISG15ylation profiles. Furthermore, we demonstrate that CML USP18-/- cells are more antigenic, leading to increased activation of cytotoxic T lymphocytes (CTLs), and are more susceptible to irradiation. Our results reinforce the role of USP18 as a key “brake” for inflammatory signals in tumor cells. As USP18 expression is upregulated in lung, breast and colon cancers, USP18 pharmacological inhibition may reflect a target opportunity in cancer immunotherapy. Citation Format: Adan Pinto-Fernandez, Helene Greenwood, Mariolina Salio, Jianzhou Chen, Thomas Partridge, George Vere, Hannah C. Scott, Andreas Damianou, Persephone Borrow, Ruth Muschel, Vincenzo Cerundolo, Benedikt M. Kessler. USP18 modulates the ISGylome, immune signaling and sensitizes tumor cells to irradiation and CTL recognition [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 LB-080.
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 5 ( 2016-03-01), p. 1078-1088
    Abstract: A series of critical pathways are responsible for the detection, signaling, and restart of replication forks that encounter blocks during S-phase progression. Small base lesions may obstruct replication fork progression and processing, but the link between repair of small lesions and replication forks is unclear. In this study, we investigated a hypothesized role for DNA-PK, an important enzyme in DNA repair, in cellular responses to DNA replication stress. The enzyme catalytic subunit DNA-PKcs was phosphorylated on S2056 at sites of stalled replication forks in response to short hydroxyurea treatment. Using DNA fiber experiments, we found that catalytically active DNA-PK was required for efficient replication restart of stalled forks. Furthermore, enzymatically active DNA-PK was also required for PARP-dependent recruitment of XRCC1 to stalled replication forks. This activity was enhanced by preventing Mre11-dependent DNA end resection, suggesting that XRCC1 must be recruited early to an unresected stalled fork. We also found that XRCC1 was required for effective restart of a subset of stalled replication forks. Overall, our work suggested that DNA-PK and PARP-dependent recruitment of XRCC1 is necessary to effectively protect, repair, and restart stalled replication forks, providing new insight into how genomic stability is preserved. Cancer Res; 76(5); 1078–88. ©2015 AACR.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...