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: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 27, No. 15 ( 2021-08-01), p. 4338-4352
    Abstract: We investigated whether targeting chromatin stability through a combination of the curaxin CBL0137 with the histone deacetylase (HDAC) inhibitor, panobinostat, constitutes an effective multimodal treatment for high-risk neuroblastoma. Experimental Design: The effects of the drug combination on cancer growth were examined in vitro and in animal models of MYCN-amplified neuroblastoma. The molecular mechanisms of action were analyzed by multiple techniques including whole transcriptome profiling, immune deconvolution analysis, immunofluorescence, flow cytometry, pulsed-field gel electrophoresis, assays to assess cell growth and apoptosis, and a range of cell-based reporter systems to examine histone eviction, heterochromatin transcription, and chromatin compaction. Results: The combination of CBL0137 and panobinostat enhanced nucleosome destabilization, induced an IFN response, inhibited DNA damage repair, and synergistically suppressed cancer cell growth. Similar synergistic effects were observed when combining CBL0137 with other HDAC inhibitors. The CBL0137/panobinostat combination significantly delayed cancer progression in xenograft models of poor outcome high-risk neuroblastoma. Complete tumor regression was achieved in the transgenic Th-MYCN neuroblastoma model which was accompanied by induction of a type I IFN and immune response. Tumor transplantation experiments further confirmed that the presence of a competent adaptive immune system component allowed the exploitation of the full potential of the drug combination. Conclusions: The combination of CBL0137 and panobinostat is effective and well-tolerated in preclinical models of aggressive high-risk neuroblastoma, warranting further preclinical and clinical investigation in other pediatric cancers. On the basis of its potential to boost IFN and immune responses in cancer models, the drug combination holds promising potential for addition to immunotherapies.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Molecular Cancer Therapeutics, American Association for Cancer Research (AACR), Vol. 22, No. 12_Supplement ( 2023-12-01), p. B132-B132
    Abstract: Background: We used the TH-MYCN mouse model of neuroblastoma (NB) to determine the importance of simultaneously inhibiting both polyamine uptake and synthesis when added to standard-of-care chemotherapy/immunotherapy. Polyamines are essential cations frequently upregulated in tumors. We have shown the benefit of adding DFMO, an irreversible polyamine synthesis inhibitor, to standard-of-care chemotherapy in several NB mouse models (Evageliou, Clin Cancer Res 2016). Based on our work, a randomized Phase 2 COG trial of DFMO, added to chemotherapy/anti-GD2 immunotherapy, is ongoing for relapsed NB (NCT03794349). We recently showed in NB mouse models that DFMO efficacy is enhanced by adding the polyamine uptake inhibitor AMXT1501 (Gamble, Sci Transl Med, 2019). DFMO/AMXT1501 is now in adult clinical trial (NCT05500508). Preclinical modelling of polyamine-targeted therapeutics in TH-MYCN mice has been invaluable for supporting clinical trial design but has yet to include anti-GD2 immunotherapy. Methods: TH-MYCN mice were used to optimize DFMO/AMXT1501 therapy combined with temozolomide/irinotecan (TEM/IRI) or cyclophosphamide/topotecan (CYCLO/TOPO), and anti-GD2 (14G2a) antibody. We studied escalating doses of DFMO and AMXT1501 (n=10/group), with or without TEM/IRI or CYCLO/TOPO. 14G2a regimen was titrated toward human-relevant pharmacokinetic exposures to establish suitable conditions for combination therapies. Results: In the absence of chemo-immunotherapy, highest-dose DFMO (1.5%) and AMXT1501 (2.5 mg/kg/d) had greatest efficacy without increased toxicity. Median mouse survival was 2-fold greater compared with lower dose DFMO/AMXT1501 combinations, suggesting that maximising DFMO dose is critical. Similar improved efficacy was observed in combination with TEM/IRI or CYCLO/TOPO. Combining 14G2a (100ug 2x/wk, 18 wks; McNerney, Oncoimmunology, 2022) with either CYCLO/TOPO (single 5-day chemo cycle) or DFMO/AMXT1501 (1.5%/2.5 mg/kg/d) increased survival compared with either treatment alone. Combining all 5 drugs resulted in 100% survival at 1 year, with minimal toxicity. However substantial dose reductions of 14G2a ( & lt;25µg, 2 doses) and TEM/IRI or CYCLO/TOPO (single 2-day chemo cycle) were needed to identify clinically relevant backbones which achieved human-relevant 14G2a pharmacokinetics. Addition of DFMO alone to the clinically relevant CYCLO/TOPO/14G2a backbone did not result in benefit compared to backbone alone. In contrast, addition of AMXT1501 plus DFMO to the backbone significantly extended survival (P & lt; 0.001). Conclusions: We have increased the utility of the TH-MYCN mouse model for preclinical modelling of polyamine inhibition, and other investigational treatments, by pharmacokinetics-driven optimization of anti-GD2 therapy with standard-of-care chemotherapy backbones. This approach will improve optimisation for future NB clinical trials. Our data highlight the importance of adding AMXT1501 to DFMO for polyamine inhibition and support a planned international DFMO/AMXT1501/chemotherapy/anti-GD2 trial for refractory NB.   Citation Format: Jayne Murray, Ruby Pandher, Lin Xiao, Klaartje Somers, Jennifer Brand, Erin Mosmann, Stephanie Alfred, Frances Kusuma, Adam Kearns, Julie R Park, Lynley V Marshall, Chiara Gorrini, Louis Chesler, Michael D Hogarty, Andrew D J Pearson, Mark Burns, Jamie I Fletcher, David Ziegler, Murray D Norris, Michelle Haber. Addition of AMXT1501 (polyamine uptake inhibitor) plus DFMO (polyamine synthesis inhibitor) to standard-of-care chemotherapy/anti-GD2 antibody in the TH-MYCN mouse neuroblastoma model, enhances efficacy compared to addition of DFMO alone [abstract]. In: Proceedings of the AACR-NCI-EORTC Virtual International Conference on Molecular Targets and Cancer Therapeutics; 2023 Oct 11-15; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2023;22(12 Suppl):Abstract nr B132.
    Type of Medium: Online Resource
    ISSN: 1538-8514
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2062135-8
    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...