Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
2.
Cancer Chemother Pharmacol ; 91(2): 191-201, 2023 02.
Article in English | MEDLINE | ID: mdl-36694044

ABSTRACT

INTRODUCTION: Glioblastoma (GBM) has a very poor prognosis despite current treatment. We previously found cytotoxic synergy between the AURKA inhibitor alisertib and the CNS-penetrating taxane TPI 287 against GBM tumor cells in vitro. METHODS: We used an orthotopic human GBM xenograft mouse model to test if TPI 287 potentiates alisertib in vivo. Western blotting, immunohistochemistry, siRNA knockdown, annexin V binding, and 3-dimensional Matrigel invasion assays were used to investigate potential mechanisms of alisertib and TPI 287 treatment interactions. RESULTS: Alisertib + TPI 287 combination therapy significantly prolonged animal survival compared to vehicle (p = 0.011), but only marginally compared to alisertib alone. Alisertib, TPI 287, and combined alisertib + TPI 287 reduced animal tumor volume compared to vehicle-treated controls. This was statistically significant for the combination therapy at 4 weeks (p < 0.0001). Alisertib + TPI 287 treatment decreased anti-apoptotic Bcl-2 protein levels in vivo and in vitro. Expression of the pro-apoptotic protein Bak was significantly increased by combination treatment (p < 0.0001). Pro-apoptotic Bim and Bak knockdown by siRNA decreased apoptosis by alisertib + TPI 287 in GB9, GB30, and U87 cells (p = 0.0005 to 0.0381). Although alisertib and TPI 287 significantly reduced GBM cell invasion (p < 0.0001), their combination was no more effective than TPI 287 alone. CONCLUSIONS: Results suggest that apoptosis is the dominant mechanism of potentiation of GBM growth inhibition by alisertib + TPI 287, in part through effects on Bcl-2 family proteins, providing a rationale for further laboratory testing of an AURKA inhibitor plus TPI 287 as a potential therapy against GBM.


Subject(s)
Aurora Kinase A , Glioblastoma , Humans , Animals , Mice , Cell Line, Tumor , Azepines/therapeutic use , Apoptosis , Taxoids/therapeutic use , Glioblastoma/drug therapy , Apoptosis Regulatory Proteins , Proto-Oncogene Proteins c-bcl-2 , RNA, Small Interfering , Xenograft Model Antitumor Assays
3.
Nat Commun ; 13(1): 2083, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440587

ABSTRACT

Astroblastomas (ABs) are rare brain tumors of unknown origin. We performed an integrative genetic and epigenetic analysis of AB-like tumors. Here, we show that tumors traceable to neural stem/progenitor cells (radial glia) that emerge during early to later brain development occur in children and young adults, respectively. Tumors with MN1-BEND2 fusion appear to present exclusively in females and exhibit overexpression of genes expressed prior to 25 post-conception weeks (pcw), including genes enriched in early ventricular zone radial glia and ependymal tumors. Other, histologically classic ABs overexpress or harbor mutations of mitogen-activated protein kinase pathway genes, outer and truncated radial glia genes, and genes expressed after 25 pcw, including neuronal and astrocyte markers. Findings support that AB-like tumors arise in the context of epigenetic and genetic changes in neural progenitors. Selective gene fusion, variable imprinting and/or chromosome X-inactivation escape resulting in biallelic overexpression may contribute to female predominance of AB molecular subtypes.


Subject(s)
Neoplasms, Neuroepithelial , Neural Stem Cells , Cell Lineage/genetics , Child , Ependymoglial Cells , Female , Humans , Male , Neuroglia , X Chromosome Inactivation/genetics , Young Adult
4.
Diagn Pathol ; 16(1): 98, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34706741

ABSTRACT

BACKGROUND: Aurora-A kinase is important for cellular proliferation and is implicated in the tumorigenesis of several malignancies, including of the ovary. Information regarding the expression patterns of Aurora-A in normal Müllerian epithelium as well as benign, borderline and malignant epithelial ovarian neoplasms is limited. METHODS: We investigated Aurora-A expression by immunohistochemistry in 15 benign, 19 borderline and 17 malignant ovarian serous tumors, and 16 benign, 8 borderline, and 2 malignant ovarian mucinous tumors. Twelve fimbriae from seven patients served as normal Müllerian epithelium controls. We also examined Aurora-A protein expression by western blot in normal fimbriae and tumor specimens. RESULTS: All normal fimbriae (n = 12) showed nuclear but not cytoplasmic Aurora-A immunoreactivity by immunohistochemistry. Benign ovarian tumors also showed strong nuclear Aurora-A immunoreactivity. Forty-eight percent (13/27) of borderline tumors demonstrated nuclear Aurora-A immunoreactivity, while the remainder (52%, 14/27) lacked Aurora-A staining. Nuclear Aurora-A immunoreactivity was absent in all malignant serous tumors, however, 47% (8/17) demonstrated perinuclear cytoplasmic staining. These results were statistically significant when tumor class (benign/borderline/malignant) was compared to immunoreactivity localization or intensity (Fisher Exact Test, p < 0.01). Western blot analysis confirmed the greater nuclear Aurora-A expression in control Müllerian epithelium compared to borderline and malignant tumors. CONCLUSION: Aurora-A kinase is differentially expressed across normal Müllerian epithelium, benign and borderline serous and mucinous ovarian epithelial neoplasms and malignant serous ovarian tumors., with nuclear expression of unphosphorylated Aurora-A being present in normal and benign neoplastic epithelium, and lost in malignant serous neoplasms. Further studies of the possible biological and clinical implications of the loss of nuclear Aurora-A expression in ovarian tumors, and its role in ovarian carcinogenesis are warranted.


Subject(s)
Aurora Kinase A/biosynthesis , Carcinoma, Ovarian Epithelial/enzymology , Cystadenocarcinoma, Mucinous/enzymology , Cystadenocarcinoma, Serous/enzymology , Ovary/enzymology , Carcinoma, Ovarian Epithelial/pathology , Cell Nucleus/enzymology , Cystadenocarcinoma, Mucinous/pathology , Cystadenocarcinoma, Serous/pathology , Cytoplasm/enzymology , Epithelium/enzymology , Female , Humans
5.
J Neurooncol ; 143(2): 231-240, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31011934

ABSTRACT

INTRODUCTION: Glioblastoma remains difficult to treat and patients whose tumors express high levels of O6-methylguanine DNA methyltransferase (MGMT) usually respond poorly to standard temozolomide chemotherapy. We have previously shown that the selective AURKA inhibitor alisertib potently inhibits growth of glioblastoma cells. METHODS: We used colony formation assays, annexin V binding, and western blotting to examine the effects of alisertib on the antiproliferative capabilities of carboplatin and irinotecan in glioblastoma cells. RESULTS: In colony formation assays, alisertib potentiated the antiproliferative effects of both carboplatin and irinotecan, often synergistically, including against glioblastoma tumor stem-like cells, as demonstrated by Chou-Talalay and Bliss statistical analyses. Western blotting showed that high MGMT expression in cell lines correlated with more pronounced potentiation of carboplatin's growth inhibitory effects by alisertib, while low MGMT expression correlated with stronger potentiation of irinotecan by alisertib. This pattern was also observed when these drug combinations were tested for their ability to induce apoptosis via annexin V binding assays. MGMT knockdown increased apoptosis caused by combined alisertib and irinotecan, while exogenous MGMT overexpression increased apoptosis from alisertib and carboplatin combination treatment. CONCLUSIONS: These results suggest that tumor MGMT expression levels may be predictive of patient response to these drug combinations, and importantly that the combination of alisertib and carboplatin may be selectively effective in glioblastoma patients with high tumor MGMT who are resistant to standard therapy. Since clinical experience with alisertib, carboplatin and irinotecan as single agents already exists, these findings may provide rationale for the design of clinical trials for their use in combination treatment regimens.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Drug Synergism , Glioblastoma/drug therapy , Glioblastoma/pathology , Tumor Suppressor Proteins/metabolism , Azepines/administration & dosage , Carboplatin/administration & dosage , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/genetics , DNA Repair Enzymes/antagonists & inhibitors , DNA Repair Enzymes/genetics , Glioblastoma/metabolism , Humans , Irinotecan/administration & dosage , Pyrimidines/administration & dosage , RNA, Small Interfering/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
6.
J Neurooncol ; 137(3): 481-492, 2018 May.
Article in English | MEDLINE | ID: mdl-29396807

ABSTRACT

Glioblastoma is a highly malignant disease in critical need of expanded treatment options. The AURKA inhibitor alisertib exhibits antiproliferative activity against glioblastoma in vitro and in vivo. Unlike current clinically used taxane drugs, the novel taxane TPI 287 penetrates the CNS. We tested for interactions between three selective AURKA inhibitors and TPI 287 against standard U87 and U1242 cells and primary glioblastoma neurospheres using colony formation assays. Bliss and Chou-Talalay analyses were utilized to statistically test for synergism. Morphological analysis, flow cytometry and annexin V binding were employed to examine cell cycle and apoptotic effects of these drug combinations. TPI 287 not only potentiated the cytotoxicity of the AURKA inhibitors alisertib, MLN8054 and TC-A2317, but was often potently synergistic. Morphologic and biochemical analysis of the combined effects of alisertib and TPI 287 consistently revealed synergistic induction of apoptosis. While each agent alone induces a mitotic block, slippage occurs allowing some tumor cells to avoid apoptosis. Combination treatment greatly attenuated mitotic slippage, committing the majority of cells to apoptosis. Alisertib and TPI 287 demonstrate significant synergism against glioblastoma cells largely attributable to a synergistic effect in inducing apoptosis. These results provide compelling rationale for clinical testing of alisertib and/or other AURKA inhibitors for potential combination use with TPI 287 against glioblastoma and other CNS neoplasms.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aurora Kinase A/antagonists & inhibitors , Azepines/pharmacology , Glioblastoma/drug therapy , Pyrimidines/pharmacology , Taxoids/pharmacology , Apoptosis/physiology , Aurora Kinase A/metabolism , Benzazepines/pharmacology , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , Drug Synergism , Glioblastoma/enzymology , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/physiology , Protein Kinase Inhibitors/pharmacology , Tumor Stem Cell Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...