File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Antitumor Activity of a Mitochondrial-Targeted HSP90 Inhibitor in Gliomas

TitleAntitumor Activity of a Mitochondrial-Targeted HSP90 Inhibitor in Gliomas
Authors
Issue Date2022
Citation
Clinical Cancer Research, 2022, v. 28, n. 10, p. 2180-2195 How to Cite?
AbstractPurpose: To investigate the antitumor activity of a mitochondrial-localized HSP90 inhibitor, Gamitrinib, in multiple glioma models, and to elucidate the antitumor mechanisms of Gamitrinib in gliomas. Experimental Design: A broad panel of primary and temozolomide (TMZ)-resistant human glioma cell lines were screened by cell viability assays, flow cytometry, and crystal violet assays to investigate the therapeutic efficacy of Gamitrinib. Seahorse assays were used to measure the mitochondrial respiration of glioma cells. Integrated analyses of RNA sequencing (RNAseq) and reverse phase protein array (RPPA) data were performed to reveal the potential antitumor mechanisms of Gamitrinib. Neurospheres, patient-derived organoids (PDO), cell line–derived xenografts (CDX), and patient-derived xenografts (PDX) models were generated to further evaluate the therapeutic efficacy of Gamitrinib. Results: Gamitrinib inhibited cell proliferation and induced cell apoptosis and death in 17 primary glioma cell lines, 6 TMZ-resistant glioma cell lines, 4 neurospheres, and 3 PDOs. Importantly, Gamitrinib significantly delayed the tumor growth and improved survival of mice in both CDX and PDX models in which tumors were either subcutaneously or intracranially implanted. Integrated computational analyses of RNAseq and RPPA data revealed that Gamitrinib exhibited its antitumor activity via (i) suppressing mitochondrial biogenesis, OXPHOS, and cell-cycle progression and (ii) activating the energy-sensing AMP-activated kinase, DNA damage, and stress response. Conclusions: These preclinical findings established the therapeutic role of Gamitrinib in gliomas and revealed the inhibition of mitochondrial biogenesis and tumor bioenergetics as the primary antitumor mechanisms in gliomas.
Persistent Identifierhttp://hdl.handle.net/10722/319010
ISSN
2021 Impact Factor: 13.801
2020 SCImago Journal Rankings: 5.427
ISI Accession Number ID

 

DC FieldValueLanguage
dc.contributor.authorWei, Shiyou-
dc.contributor.authorYin, Delong-
dc.contributor.authorYu, Shengnan-
dc.contributor.authorLin, Xiang-
dc.contributor.authorSavani, Milan R.-
dc.contributor.authorDu, Kuang-
dc.contributor.authorKu, Yin-
dc.contributor.authorWu, Di-
dc.contributor.authorLi, Shasha-
dc.contributor.authorLiu, Hao-
dc.contributor.authorTian, Meng-
dc.contributor.authorChen, Yaohui-
dc.contributor.authorBowie, Michelle-
dc.contributor.authorHariharan, Seethalakshmi-
dc.contributor.authorWaitkus, Matthew-
dc.contributor.authorKeir, Stephen T.-
dc.contributor.authorSugarman, Eric T.-
dc.contributor.authorDeek, Rebecca A.-
dc.contributor.authorLabrie, Marilyne-
dc.contributor.authorKhasraw, Mustafa-
dc.contributor.authorLu, Yiling-
dc.contributor.authorMills, Gordon B.-
dc.contributor.authorHerlyn, Meenhard-
dc.contributor.authorWu, Kongming-
dc.contributor.authorLiu, Lunxu-
dc.contributor.authorWei, Zhi-
dc.contributor.authorFlaherty, Keith T.-
dc.contributor.authorAbdullah, Kalil-
dc.contributor.authorZhang, Gao-
dc.contributor.authorAshley, David M.-
dc.date.accessioned2022-10-11T12:25:04Z-
dc.date.available2022-10-11T12:25:04Z-
dc.date.issued2022-
dc.identifier.citationClinical Cancer Research, 2022, v. 28, n. 10, p. 2180-2195-
dc.identifier.issn1078-0432-
dc.identifier.urihttp://hdl.handle.net/10722/319010-
dc.description.abstractPurpose: To investigate the antitumor activity of a mitochondrial-localized HSP90 inhibitor, Gamitrinib, in multiple glioma models, and to elucidate the antitumor mechanisms of Gamitrinib in gliomas. Experimental Design: A broad panel of primary and temozolomide (TMZ)-resistant human glioma cell lines were screened by cell viability assays, flow cytometry, and crystal violet assays to investigate the therapeutic efficacy of Gamitrinib. Seahorse assays were used to measure the mitochondrial respiration of glioma cells. Integrated analyses of RNA sequencing (RNAseq) and reverse phase protein array (RPPA) data were performed to reveal the potential antitumor mechanisms of Gamitrinib. Neurospheres, patient-derived organoids (PDO), cell line–derived xenografts (CDX), and patient-derived xenografts (PDX) models were generated to further evaluate the therapeutic efficacy of Gamitrinib. Results: Gamitrinib inhibited cell proliferation and induced cell apoptosis and death in 17 primary glioma cell lines, 6 TMZ-resistant glioma cell lines, 4 neurospheres, and 3 PDOs. Importantly, Gamitrinib significantly delayed the tumor growth and improved survival of mice in both CDX and PDX models in which tumors were either subcutaneously or intracranially implanted. Integrated computational analyses of RNAseq and RPPA data revealed that Gamitrinib exhibited its antitumor activity via (i) suppressing mitochondrial biogenesis, OXPHOS, and cell-cycle progression and (ii) activating the energy-sensing AMP-activated kinase, DNA damage, and stress response. Conclusions: These preclinical findings established the therapeutic role of Gamitrinib in gliomas and revealed the inhibition of mitochondrial biogenesis and tumor bioenergetics as the primary antitumor mechanisms in gliomas.-
dc.languageeng-
dc.relation.ispartofClinical Cancer Research-
dc.titleAntitumor Activity of a Mitochondrial-Targeted HSP90 Inhibitor in Gliomas-
dc.typeArticle-
dc.description.naturelink_to_subscribed_fulltext-
dc.identifier.doi10.1158/1078-0432.CCR-21-0833-
dc.identifier.pmid35247901-
dc.identifier.scopuseid_2-s2.0-85130634563-
dc.identifier.volume28-
dc.identifier.issue10-
dc.identifier.spage2180-
dc.identifier.epage2195-
dc.identifier.eissn1557-3265-
dc.identifier.isiWOS:000799639000001-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats