Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Cytometry, Wiley, Vol. 45, No. 2 ( 2001-10-01), p. 87-95
    Type of Medium: Online Resource
    ISSN: 0196-4763 , 1097-0320
    URL: Issue
    RVK:
    Language: English
    Publisher: Wiley
    Publication Date: 2001
    detail.hit.zdb_id: 2180639-1
    detail.hit.zdb_id: 1474272-X
    detail.hit.zdb_id: 2180651-2
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Elsevier BV ; 2001
    In:  Current Opinion in Chemical Biology Vol. 5, No. 1 ( 2001-2), p. 74-77
    In: Current Opinion in Chemical Biology, Elsevier BV, Vol. 5, No. 1 ( 2001-2), p. 74-77
    Type of Medium: Online Resource
    ISSN: 1367-5931
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2001
    detail.hit.zdb_id: 2019216-2
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Oncotarget, Impact Journals, LLC, Vol. 11, No. 3 ( 2020-01-21), p. 265-281
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2020
    detail.hit.zdb_id: 2560162-3
    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. 73, No. 8_Supplement ( 2013-04-15), p. 2744-2744
    Abstract: Background: Neuroblastoma is a pediatric embryonal cancer for which the survival of patients with high-risk disease is less than 50% and has not dramatically changed over the last several years. Recently, a number of cell cycle genes_particularly those within the Cyclin D/CDK4/CDK6/RB network_have been identified as oncogenic vulnerabilities in neuroblastoma, suggesting that their therapeutic exploitation might improve survivability. Indeed, genomic amplifications of CDK4, CDK6, and CCND1 have been reported in primary neuroblastomas, and we have previously shown via an unbiased loss of function screen that CDK4 depletion is associated with potent anti-tumor activity (Cole, PNAS 2011). Here, we sought to translate these findings into novel therapies for children with neuroblastoma by evaluating the effect of pharmacologic Cdk4/Cdk6 inhibition on neuroblastoma viability. Methods: We analyzed the effect of combined Cdk4/6 inhibition in a comprehensive panel of human-derived neuroblastoma cell lines using LEE011, a highly specific Cdk4/6 small molecule inhibitor. Anti-tumor activity was also determined in vivo in three neuroblastoma xenograft models, and integrative genomics was used to identify biomarkers of drug sensitivity. Results: Treatment with LEE011 significantly inhibited proliferation in 10 of 15 human neuroblastoma-derived cell lines by inducing cytostasis at nanomolar concentrations (mean IC50 = 361 ± 97 nM, considering sensitive lines only), as evidenced by significant cell cycle arrest and senescence that were likely attributed to dose-dependent decreases in phosphorylated RB and FOXM1. In addition, responsiveness of neuroblastoma xenografts to LEE011 was reflective of in vitro data in that there was a direct correlation of IC50 values with degree of subcutaneous xenograft growth delay, with the most sensitive lines in vitro showing profound growth inhibition in vivo. While our data indicate that neuroblastomas sensitive to LEE011 were more likely to contain genomic amplification of MYCN (p= 0.04, student's t test), a supervised hierarchical clustering of gene expression data identified several potential gene signatures that could explain the observed differential sensitivity to Cdk4/6 inhibition. Conclusions: Our data show that LEE011 is highly active in a large subset of neuroblastoma cell lines and xenograft models, and therefore support the clinical development of LEE011 as a therapy for neuroblastoma as well as efforts to validate biomarkers of drug activity. Citation Format: JulieAnn Rader, Lori Hart, Mike Russell, Michael Nakazawa, Lili Belcastro, Daniel Martinez, Erica Carpenter, Sunkyu Kim, Sudha Parasuraman, Giordano Caponigro, Robert Schnepp, Andrew Wood, Bruce Pawel, Deborah Watson, Patrick Warren, Kristina Cole, John Maris. CDK4/CDK6 inhibition is potently active in a definable subset of human neuroblastomas. [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 2744. doi:10.1158/1538-7445.AM2013-2744
    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 ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 2058-2058
    Abstract: In recent years great advances have been made in developing targeted cancer therapeutics that produce dramatic responses in a subset of rationally selected patients. The initial breakthrough of the targeted design concept was established by the treatment of chronic myelogenous leukemia with Abl inhibitors and has been expanded to other cancer indications. As a consequence of this early success in CML, the identification and targeting of genetic lesions that confer cancer dependence has become an established strategy for drug discovery efforts. However, this approach has met with mixed degrees of success as confounding factors, such as tumor heterogeneity, have often resulted in partial responses and/or the emergence of resistance when targeted therapies were employed as single agents. To improve the therapeutic benefit in cancer, rationally-devised novel combinations of two or more agents are being explored clinically. To discover combinations that may be more effective therapies, an unbiased, systematic approach was used to identify drug combinations in vitro, using a panel of genetically diverse, and well characterized cell lines from the cancer cell line encyclopedia (CCLE: Barretina et al. Nature 2012). For three cancer indications, all pairwise combinations of 18 selected drugs (both novel inhibitors and standards of care) were tested as dose matrices in a proliferation assay. Synergistic interactions were scored using isobologram/Loewe's excess inhibition and synergistic concentration ranges for each agent were identified. However, the clinical translation of positive combinations from in vitro matrix-based screens into clinically-relevant doses and schedules are challenging, due to host biology, tumor-stroma interactions, and the pharmacokinetic and pharmacodynamics of drug delivery. To explore this higher complexity, we evaluated the in vitro to in vivo translation of drug synergies in immune-compromised mouse tumor xenograft models. To recapitulate the pharmacological combination effects in vivo, mouse pharmacokinetic data and simulation was used to determine single agent doses that would result in the desired compound plasma concentration range and duration of action. Pharmakokinetics, pharmacodynamics, antitumor activity and tolerability of the combinations were then tested in tumor-bearing mice. Observed combination effects in vivo could in some cases be explained by either the expected biological pathway interactions or partially by physiological effects relating to drug-drug interactions. Citation Format: Marion Wiesmann, Mark Stump, Giordano Caponigro, David Duhl, Brant Firestone, Tom Gesner, Bjoern Gruenenfelder, Daniel Alexander Guthy, Jocelyn Holash, Fred King, Joseph Lehar, Christophe Leroy, Manway Liu, Lilli Petruzelli, Dale Porter, Paul McSheehy, Daniel Menezes, Anupama Reddy, Johannes Roesel, Christian Schnell, Timothy R. Smith, Markus Wartmann. Systematic evaluation of drug combinations in vitro and in vivo. [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 2058. doi:10.1158/1538-7445.AM2013-2058
    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) ; 2011
    In:  Cancer Research Vol. 71, No. 18_Supplement ( 2011-09-15), p. C70-C70
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 18_Supplement ( 2011-09-15), p. C70-C70
    Abstract: We describe an integrative analysis approach leveraging the publicly available Cancer Cell Line Encyclopedia to uncover regulatory pathways in cancer. Six different lines of evidence are considered in identifying candidate factors associated with disease: 1) gene mutation status, 2) copy number variations, 3) RNAi growth inhibitions, 4) pharmacological growth inhibitions, 5) baseline gene expression data, and 6) physical protein-protein interactions. As each of these evidence sources measure a different aspect of cellular regulation, they may be considered in combination to uncover regulatory mechanisms associated with tumorigenesis. For example, gene mutation status or copy number variation data may be combined with baseline gene expressions to identify genes that are differentially expressed in cell lines with aberrant or deactivated versions of specific tumor suppressors and oncogenes. To assess our approach, we applied it to identification of genes involved in the well-known RAS-RAF regulatory signaling pathway in both tissuespecific and nonspecific contexts. The RAS-RAF signaling pathway plays a key role in cellular differentiation and proliferation and is frequently associated with tumorigenesis. Specifically, we identified differentially expressed genes in BRAF mutant vs. wild-type cell lines, genes associated with high and low RAF copy number alteration cell lines, genes correlating in expression to growth inhibition by RAF-targeting compounds, and genes that either rescue or sensitize cell lines to BRAF knockdown by RNA interference. We further considered these genes within the overall context of physical interactions between their protein products. Highlighting the significant, research potential of our approach, our RAF-focused analysis identified both well-known and novel genes, supported by different lines of evidence, that mediate RAS-RAF signaling across multiple tissues. Positive control genes include members of the classical pathway, such as MAPK1, MAP2K1, and PI3K. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the Second AACR International Conference on Frontiers in Basic Cancer Research; 2011 Sep 14-18; San Francisco, CA. Philadelphia (PA): AACR; Cancer Res 2011;71(18 Suppl):Abstract nr C70.
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 5455-5455
    Abstract: Comprehensive genomic characterization of cancer is proceeding at a rapidly accelerating pace, mainly due to the expanded use of massively parallel sequencing. Despite the promise of cancer genomics, many cancer drugs still fail in the clinic due to nonresponsive patients and this translates into a significant unmet medical need. Accurate predictions of which patients are more likely to respond to drugs in development could speed clinical trials and personalize treatments. Here we propose the use of a compendium of experimentally tractable cancer model systems, ∼1000 human genomically-annotated cancer cell lines (at the level of gene expression, DNA copy number alterations and mutations), coupled with pharmacological profiling, to systematically link genetic and transcriptional features to drug response. This resource, the Cancer Cell Line Encyclopedia (CCLE), is available online at www.broadinstitute.org/ccle. Through computational predictive modeling we have both rediscovered molecular features that predict response to several drugs and also uncovered a number of novel potential biomarkers of sensitivity and resistance to targeted agents and chemotherapy drugs. For instance, we have found that response to topoisomerase 1 inhibitors seem to be driven by expression of a single gene. We have also observed that tissue lineage is a key predictor for sensitivity to certain compounds, providing rationale for clinical trials of these drugs in particular cancer types. Our cell line-based platform provides a valuable tool for the development of personalized cancer medicine, revealing critical tumor dependencies and helping to stratify patients for clinical trials. 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 5455. doi:10.1158/1538-7445.AM2011-5455
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 2337-2337
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 2337-2337
    Abstract: Selective RAF inhibitors have a significant role in the treatment of patients with metastatic melanoma whose tumors express BRAFV600E with the majority of patients experiencing significant tumor regression. However in patients with colorectal cancer (CRC) whose tumors express BRAFV600E, response-rates appear to be much lower, with only a few patients reported to experience a partial tumor response to vemurafenib (Kopetz et al., 2010) or the combination of dabrafenib plus trametinib (Corcoran et al., 2012). LGX818 is a highly potent RAF inhibitor with selective anti-proliferative activity in cells expressing BRAFV600E. A distinguishing feature of LGX818 is its very long dissociation half-life from BRAFV600E which leads to strong and sustained target inhibition even following drug wash-out. In addition, LGX818 has a very wide therapeutic index, with tumor regression observed at doses as low as 3 mg/kg bid and excellent tolerability up to 300 mg/kg bid in melanoma xenograft models. In the CRC cell line Colo205 (BRAFV600E), LGX818 inhibits proliferation with an EC50 = 0.005 μM and in vivo leads to significant tumor regression at doses as low as 20 mg/kg. However, other BRAFV600E CRC cell lines do not appear to be as sensitive (EC50 = 0.018 to & gt;2.7 μM) and this intrinsic resistance translates in vivo in xenograft models generated from these cell lines. Based on clinical and preclinical data, it is clear that combination strategies will be required if RAF inhibitors are going to play a significant role in the treatment of CRC. Recent publications have implicated feedback-mediated activation of EGFR in BRAFV600E CRC cells treated with RAF and MEK inhibitors and this led us to test combinations of LGX818 with anti-EGFR therapies such as cetuximab and erlotinib. In vitro and in vivo studies indicated that the combination of LGX818 with cetuximab or erlotinib can be highly synergistic, leading to enhanced cell killing. In vivo the combination of LGX818 + cetuximab led to complete inhibition of tumor growth at doses where no single-agent activity was observed. Since PIK3CA mutations often co-occur with BRAFV600E in CRC tumors, we also tested the alpha-selective PI3K inhibitor BYL719 in combination with LGX818 and cetuximab as a triple combination. In vitro, this triple combination produced synergistic anti-proliferative effects and in vivo resulted in tumor regression. These preclinical data led to the initiation of an ongoing Phase I/II trial to evaluate the triple combination of LGX818, cetuximab and BYL719 in BRAFV600E CRC. Based on the preclinical results described above it is anticipated that the combination of a RAF inhibitor with anti-EGFR therapy and a PI3K inhibitor will have superior efficacy to single-agent RAF inhibitor in BRAFV600E CRC. Citation Format: Giordano Caponigro, Z A. Cao, Xiaobin Zhang, Hui Q. Wang, Christine M. Fritsch, Darrin D. Stuart. Efficacy of the RAF/PI3Kα/anti-EGFR triple combination LGX818 + BYL719 + cetuximab in BRAFV600E colorectal tumor 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 2337. doi:10.1158/1538-7445.AM2013-2337
    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 ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. LB-114-LB-114
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. LB-114-LB-114
    Abstract: RAS mutant tumors have proven largely refractory to inhibitors of the MAPK pathway in clinic. In contrast, selective inhibitors of BRAFV600E but not wild type RAF have been successfully developed in BRAFV600E mutant melanoma, NSCLC, and thyroid cancers. Success of these drugs results largely from the therapeutic index imparted by lack of activity against wild RAFs; however, this selectivity also prevents their utility in indications beyond BRAFV600E disease, such as RAS-mutant tumors. In contrast, MEK1/2 inhibitors, while effective pre-clinically in RAS mutant models, have proven ineffective against RAS mutant tumors due at least in part to the poor therapeutic index of these molecules that results from pathway inhibition in normal cells. Thus, to inhibit effectively MAPK pathway signaling in tumors, inhibitors with improved tolerability via differential pathway inhibition in tumor vs normal tissues are required. Recent data in mice conditionally lacking different RAF paralogs suggest that inhibitors biased towards CRAF might retain anti-tumor properties, with improved tolerability. LXH254 is a RAF inhibitor with high selectivity and nanomolar potency for BRAF and CRAF. Profiling of LXH254 activity in Ras mutant lines lacking either ARAF, BRAF or CRAF by CRISPR-Cas9 modification revealed that loss of ARAF, but not BRAF or CRAF, sensitized cells to LXH254. This increased sensitivity to LXH254 from ARAF knockout was rescued by over-expression of wild-type ARAF but not by either ARAF that harbored mutations that impaired kinase activity (K336M, D447A) or its ability to dimerize (R362H). Comparing the effect of LXH254 on MAPK pathway signaling in RAS mutant cells engineered to express only one of the three RAF paralogs indicated that significantly higher concentrations of LXH254 are required to inhibit MAPK signaling in cells expressing only ARAF. Furthermore, at low concentrations of LXH254, cells expressing only ARAF displayed paradoxical activation of MAPK signaling in a manner similar to the BRAFV600E selective inhibitor dabrafenib. Thus, both LXH254 and dabrafenib inhibit mutationally activated monomeric BRAF, and promote/stabilize RAS-dependent dimerization of WT RAFs. However, because dabrafenib is only able to effectively inhibit one of the two RAF protomers in each dimer it paradoxically activates MAPK signaling in a RAS-dependent manner. In contrast, LXH254 retains activity against dimerized B-and C-RAF (but not ARAF) thereby theoretically extending its utility beyond BRAF mutant disease with better tolerability relative to, e.g. MEK1/2 inhibitors. Citation Format: Kelli-Ann Monaco, Scott Delach, Paul Fordjour, Salonee Parikh, Yun Feng, Mariela Jaskelioff, Darrin Stuart, Giordano Caponigro. RAF inhibitor LXH254 effectively inhibits B-and-CRAF, but not ARAF [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr LB-114.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 23, No. 11 ( 2017-06-01), p. 2856-2868
    Abstract: Purpose: Anaplastic lymphoma kinase (ALK) is the most frequently mutated oncogene in the pediatric cancer neuroblastoma. We performed an in vitro screen for synergistic drug combinations that target neuroblastomas with mutations in ALK to determine whether drug combinations could enhance antitumor efficacy. Experimental Design: We screened combinations of eight molecularly targeted agents against 17 comprehensively characterized human neuroblastoma-derived cell lines. We investigated the combination of ceritinib and ribociclib on in vitro proliferation, cell cycle, viability, caspase activation, and the cyclin D/CDK4/CDK6/RB and pALK signaling networks in cell lines with representative ALK status. We performed in vivo trials in CB17 SCID mice bearing conventional and patient-derived xenograft models comparing ceritinib alone, ribociclib alone, and the combination, with plasma pharmacokinetics to evaluate for drug–drug interactions. Results: The combination of ribociclib, a dual inhibitor of cyclin-dependent kinase (CDK) 4 and 6, and the ALK inhibitor ceritinib demonstrated higher cytotoxicity (P = 0.008) and synergy scores (P = 0.006) in cell lines with ALK mutations as compared with cell lines lacking mutations or alterations in ALK. Compared with either drug alone, combination therapy enhanced growth inhibition, cell-cycle arrest, and caspase-independent cell death. Combination therapy achieved complete regressions in neuroblastoma xenografts with ALK-F1174L and F1245C de novo resistance mutations and prevented the emergence of resistance. Murine ribociclib and ceritinib plasma concentrations were unaltered by combination therapy. Conclusions: This preclinical combination drug screen with in vivo validation has provided the rationale for a first-in-children trial of combination ceritinib and ribociclib in a molecularly selected pediatric population. Clin Cancer Res; 23(11); 2856–68. ©2016 AACR.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    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