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: Blood, American Society of Hematology, Vol. 125, No. 2 ( 2015-01-08), p. 296-303
    Abstract: IDH2 R140Q expression in TF-1 cells can induce DNA and histone hypermethylation that mirrors human IDH2 mutant acute myeloid leukemia. The hypermethylation can be reversed on treatment with AGI-6780, an IDH2 mutant-specific small-molecule inhibitor.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2015
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Blood, American Society of Hematology, Vol. 140, No. Supplement 1 ( 2022-11-15), p. 6239-6240
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2022
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 120, No. 21 ( 2012-11-16), p. 3509-3509
    Abstract: Abstract 3509 Mutations in the isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2) genes are present in ∼16% of acute myeloid leukemia, and cause a neomorphic enzyme activity that results in the production of 2-hydroxyglutarate (2HG). Mutational and epigenetic profiling of a large patient cohort of acute myeloid leukemia (AML) has revealed that IDH1/2-mutant AMLs display global DNA hypermethylation and an impaired hematopoietic differentiation. To further investigate the intrinsic effect of 2HG on hematopoietic proliferation and differentiation, we transfected an erythroleukemia cell line (TF-1) with either IDH1 or IDH2 mutant alleles. These cells overexpress the mutant enzyme, have high levels of 2HG, and exhibit GM-CSF independent growth. Consistent with clinical observations, overexpression of the IDH mutant proteins led to hypermethylation of both histones and DNA. These results suggest that mutations in IDH1/2 could lead to epigenetic rewiring of cells that could facilitate the gain of function phenotype. To gain a broader understanding of the biological consequence of the IDH1/2 gain of function mutations we have generated small molecules that are capable of selectively inhibiting IDHm enzymes. Upon compound treatment in vitro, we are able to reverse hypermethylation of both histones and DNA in Idhm expressing cells. These compounds are also suitable for use in vivo and upon compound treatment are able to lower 2HG by 〉 90% in tumor xenograft models. These data suggest that an inhibitor of IDH1/2 mutations could correct the altered gene expression patterns seen in IDH1/2 mutant AML tumors and potentially lead to a profound effect on hematopoietic differentiation, proliferation and tumor growth. Disclosures: Yen: Agios Pharmaceuticals: Employment, Equity Ownership. Wang:Agios Pharmaceuticals: Employment, Equity Ownership. Schalm:Agios Pharmaceuticals: Employment, Equity Ownership. Hansen:Agios Pharmaceuticals: Employment, Equity Ownership. Straley:Agios Pharmaceuticals: Employment. Kernytsky:Agios Pharmaceuticals: Employment, Equity Ownership. Choe:Agios Pharmaceuticals: Employment, Equity Ownership. Liu:Agios Pharmaceuticals: Employment, Equity Ownership. Popovici-Muller:Agios Pharmaceuticals: Employment, Equity Ownership. Travins:Agios pharmaceuticals: Employment, Equity Ownership. Yang:Agios Pharmaceuticals: Employment, Equity Ownership. Silverman:Agios Pharmaceuticals: Employment, Equity Ownership. Gross:Agios Pharmaceuticals: Employment, Equity Ownership. Dang:Agios Pharmaceuticals: Employment, Equity Ownership. Salituro:Agios Pharmaceuticals: Consultancy, Equity Ownership. Saunders:Agios Pharmaceuticals: Consultancy, Equity Ownership. Dorsch:Agios Pharmaceuticals: Employment, Equity Ownership. Agresta:Agios Pharmaceuticals: Employment. Schenkein:Agios Pharmaceuticals: Employment, Equity Ownership. Su:Agios Pharmaceuticals: Employment, Equity Ownership. Biller:Agios Pharmaceuticals: Employment, Equity Ownership.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2012
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 2296-2296
    Abstract: Mutations of IDH1 and IDH2 which produce the oncometabolite 2-hydroxyglutarate (2HG), have been identified in several tumors including acute myeloid leukemia (AML). Recent studies have shown that expression of the IDH mutant enzymes results in high levels of 2HG and a block in cellular differentiation that can be reversed with IDH-mutant specific small molecule inhibitors. To further understand the role of IDH mutations in cancer, we conducted mechanistic studies in the TF-1 IDH2 R140Q erythroleukemia model system and found that IDH2 mutant expression caused both histone and genomic DNA methylation changes that can be reversed when IDH2 mutant activity is inhibited. Specifically, histone hypermethylation is rapidly reversed within days whereas reversal of DNA hypermethylation proceeds in a progressive manner over the course of weeks. Pathway enrichment analysis revealed several pathways involved in tumorigenesis of leukemia and lymphoma, indicating a selective modulation of relevant cancer genes by IDH mutations. As methylation of DNA and histones is closely linked to mRNA expression and differentiation, these results indicate that IDH2 mutant inhibition may function as a cancer therapy via histone and DNA demethylation at genes involved in differentiation and tumorigenesis. Citation Format: Andrew Kernytsky, Fang Wang, Erica Hansen, Stefanie Schalm, Kimberly Straley, Camelia Gliser, Hua Yang, Jeremy Travins, Stuart Murray, Marion Dorsch, Sam Agresta, David P. Schenkein, Scott A. Biller, Shinsan M. Su, Wei Liu, Katharine E. Yen. IDH2 mutation induced histone and DNA hypermethylation is progressively reversed by small molecule inhibition. [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 2296. doi:10.1158/1538-7445.AM2014-2296
    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 ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1467-1467
    Abstract: Background: EGFR-activating mutations (L858R and ex19del) are major drivers (15-47%) of lung adenocarcinoma. EGFR TKI development has changed the treatment paradigm for EGFR-mutant (EGFR+) NSCLC. However, tumors often develop acquired resistance mutations to current TKIs leading to clinical progression. EGFR+/T790M is the most prevalent acquired resistance mutation after first- and second-generation TKI treatment. Osimertinib, a third-generation TKI which targets EGFR+/T790M, can overcome this resistance, but faces further resistance within 10-18 months. EGFR+/C797S is the most significant on-target resistance mechanism to osimertinib, leading to EGFR+/T790M/C797S double resistant mutants. There are currently no approved targeted therapies for patients with EGFR+/T790M/C797S NSCLC. BLU-945 is a potent and highly selective inhibitor of the EGFR+/T790M/C797S and EGFR+/T790M mutations, with in vivo activity in subcutaneous and intracranial EGFR-mutant tumor models. Method: In vivo antitumor activity of BLU-945 was evaluated in engineered triple mutant cell line-derived xenograft (CDX) models and osimertinib-resistant patient-derived xenograft (PDX) models of NSCLC. Plasma and tumor samples were collected for future pharmacokinetic and pharmacodynamic analyses. In vivo activity was also evaluated in an intracranial implantation model using luciferase-expressing YU-1097 patient-derived-cells harboring EGFRex19del/T790M/C797S resistance mutations, with the tumor burden of intracranial lesions measured by bioluminescence imaging. Results: Oral administration of BLU-945 to tumor-bearing mice showed potent EGFR pathway inhibition and significant single-agent antitumor activity at well-tolerated doses in the engineered osimertinib-resistant EGFRL858R/T790M/C797S CDX model, and an EGFRex19del/T790M/C797S PDX model. In addition, combination of BLU-945 with osimertinib showed enhanced antitumor activity compared with single-agent treatment in the EGFRex19del/T790M/C797S PDX model. Pharmacodynamic assays revealed treatment with BLU-945 resulted in marked inhibition of EGFR, AKT and ERK phosphorylation in the EGFRL858R/T790M/C797S CDX and EGFRex19del/T790M/C797S PDX models. BLU-945 also demonstrated potent activity in a patient-derived model of EGFRex19del/T790M/C797S implanted intracranially. Conclusion: BLU-945 is a potent, selective, and orally available fourth-generation EGFR inhibitor with robust antitumor activity in osimertinib-resistant NSCLC models grown subcutaneously and intracranially. BLU-945 shows activity as a single agent and in combination with other EGFR inhibitors. Clinical development of BLU-945 is expected in 2021. Citation Format: Sun Min Lim, Chae Won Park, Zhuo Zhang, Rich Woessner, Tom Dineen, Faith Stevison, John Hsieh, Meredith Eno, Doug Wilson, John Campbell, Caitlin Utt, Faris Albayya, Nicolas Lamontagne, Marion Dorsch, Klaus Hoeflich, Byoung Chul Cho, Stefanie Schalm. BLU-945, a fourth-generation, potent and highly selective epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) with intracranial activity, demonstrates robust in vivo antitumor activity in models of osimertinib-resistant non-small cell lung cancer (NSCLC) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1467.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 954-954
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 954-954
    Abstract: Human cancer genomes harbor a variety of alterations leading to the deregulation of key pathways in tumor cells. The genomic characterization of tumors has uncovered numerous genes recurrently mutated, deleted or amplified, but gene fusions have not been studied as extensively. Kinase fusions represent ideal targets for the development of cancer drugs because they often confer oncogenic dependency in hematopoietic and solid malignancies as demonstrated by the success of several kinase inhibitors. For example, imatinib induces remission in leukemia patients who are positive for BCR-ABL1 fusions, and crizotinib and ceritinib have produced significant clinical benefit in patients with lung adenocarcinomas and mesenchymal tumors harboring ALK fusions. We have developed heuristics for reliably detecting gene fusion events in RNA-seq data sets and apply them to nearly 9,000 samples from The Cancer Genome Atlas. Fusions between any two genes were identified based on the number of chimeric reads and split reads. Then a number of filtering criteria were applied to flag false positive and non-functional fusions, including the removal of kinase fusions observed in a panel of more than 3,500 normal samples from diverse origins. Finally, we reviewed all recurrent kinase fusions manually to identify putative oncogenic drivers with distinctive characteristics of functional kinase fusions. We thereby were able to recapitulate most known translocation events in solid tumors (i.e., ALK, BRAF, EGFR, FGFR1, 2 and 3, NTRK1, 2 and 3, PDGFRA, PRKCA, RAF1, RET, ROS1). Interestingly, we identified new tumor types harboring such fusions and discovered several novel fusion partners for these kinases. We also detected several low-frequency, pan-cancer kinase fusion events, for example in the neurotrophic tyrosine receptor kinases NTRK1, NTRK2 and NTRK3, that drive tumorigenesis in a small fraction of multiple cancers, regardless of tissue type. Using our computational pipeline, we identified several novel and recurrent kinase fusions involving the MET proto-oncogene and PIK3CA. These bona fide oncogenes have not been shown previously to be activated by fusion events. Our analysis also uncovered novel, recurrent fusions in kinases with no known tumorigenic genomic alterations (e.g., FGR and PKN1), potentially resulting in active and oncogenic fusion proteins that represent putative targets for drug discovery. These findings have immediate diagnostic and clinical implications and expand the therapeutic options for cancer patients, as approved or exploratory drugs exist for many of these kinases. Citation Format: Nicolas Stransky, Ethan Cerami, Stefanie Schalm, Joseph L. Kim, Klaus Hoeflich, Christoph Lengauer. The landscape of kinase fusions in cancer. [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 954. doi:10.1158/1538-7445.AM2015-954
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. LB-164-LB-164
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. LB-164-LB-164
    Abstract: IDH mutations and tumorgenicity Kate Yen1, Fang Wang1, Sung Choe1, Stefanie Schalm1, Erica Hansen1, Kimberly Straley1, Janeta Popovici-Muller1, Jeremy Travins1, Hua Yang1, Lee Silverman1, William Kaelin2, Stefan Gross1, Lenny Dang1, Frank Salituro1, Jeff Saunders1, David Schenkein1, Michael Su1, Scott Biller1 Mutations in the isocitrate dehydrogenase 1 (IDH1) and 2 (IDH2) genes are present in ∼16% of acute myeloid leukemia, and these mutations cause neomorphic enzyme activity that results in the production of 2-hydroxyglutarate (2HG). Mutational and epigenetic profiling of a large patient cohort of acute myeloid leukemia (AML) revealed that IDH1/2-mutant AMLs display global DNA hypermethylation and a specific hypermethylation signature. To further investigate the intrinsic effect of 2HG on hematopoietic differentiation, we studied erythroleukemia cell lines transfected with IDH1 and IDH2 mutant alleles which overexpress the mutant enzyme, have high levels of 2HG, and exhibit GM-CSF independent growth. Further investigation showed that GATA1, a transcriptional factor known to direct myeloid differentiation, is also down-regulated by IDH2m in these cells along with its downstream direct target SLC4A1. These results demonstrate that the IDH2m can perturb the expression of transcription factors that could lead to alterations in myeloid differentiation. These data suggest that an inhibitor of IDH1/2 mutations could correct for the altered gene expression patterns seen in IDH1/2 mutant AML tumors leading to a profound effect on hematopoietic differentiation, proliferation and tumor growth. We are currently studying the global effects of IDH1/2 mutant overexpression on the methylation of histones and DNA to have a broader understanding of the biological consequence of the IDH1/2 gain of function mutations. Furthermore, treatment of these cell lines with compounds specific for either IDH1 or IDH2 mutant enzymes has a profound effect on their biology. 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 LB-164. doi:1538-7445.AM2012-LB-164
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...