mln-8237 and Brain-Neoplasms

mln-8237 has been researched along with Brain-Neoplasms* in 9 studies

Trials

1 trial(s) available for mln-8237 and Brain-Neoplasms

ArticleYear
Phase I trial of alisertib with concurrent fractionated stereotactic re-irradiation for recurrent high grade gliomas.
    Radiotherapy and oncology : journal of the European Society for Therapeutic Radiology and Oncology, 2019, Volume: 132

    We conducted a phase I trial of alisertib, an oral aurora kinase inhibitor, with fractionated stereotactic re-irradiation therapy (FSRT) for patients with recurrent high grade glioma (HGG).. Adult patients with recurrent HGG were enrolled from Feb 2015 to Feb 2017. Patients were treated with concurrent FSRT and alisertib followed by maintenance alisertib. Concurrent alisertib dose was escalated from 20 mg to 50 mg twice daily (BID).. 17 patients were enrolled. Median follow-up was 11 months. Median FSRT dose was 35 Gy. There were 6, 6, 3, and 2 patients enrolled in 20 mg, 30 mg, 40 mg, and 50 mg cohort, respectively. Only one DLT was observed. One patient in the 20 mg cohort had severe headache (Grade 3) resolved with steroids. There was no non-hematological grade 3 or higher toxicity. There were two Grade 4 late toxicities (one with grade 4 neutropenia and leukopenia, one with pulmonary embolism). One patient developed radiation necrosis (Grade 3). Sixteen patients finished concurrent treatment and received maintenance therapy (median cycles was 3, range 1-9). OS for all cohorts at 6 months was 88.2% with median survival time of 11.1 months. PFS at 6 months was 35.3% with median time to progression of 4.9 months. The trial stopped early due to closure of alisertib program with only 2 of 3 planned patients enrolled in the 50 mg cohort.. Re-irradiation with FSRT combined with alisertib is safe and well tolerated for HGG with doses up to 40 mg BID. Although no DLT observed in the 50 mg cohort, this cohort was not fully enrolled and MTD was not reached. Clinical outcomes appear comparable to historical results. (NCT02186509).

    Topics: Adult; Aged; Azepines; Brain Neoplasms; Chemoradiotherapy; Cohort Studies; Female; Glioma; Humans; Male; Middle Aged; Neoplasm Recurrence, Local; Protein Kinase Inhibitors; Pyrimidines; Radiosurgery; Re-Irradiation; Treatment Outcome

2019

Other Studies

8 other study(ies) available for mln-8237 and Brain-Neoplasms

ArticleYear
Murine Central Nervous System and Bone Marrow Distribution of the Aurora A Kinase Inhibitor Alisertib: Pharmacokinetics and Exposure at the Sites of Efficacy and Toxicity.
    The Journal of pharmacology and experimental therapeutics, 2022, Volume: 383, Issue:1

    Important challenges in developing drugs that target central nervous system (CNS) tumors include overcoming barriers for CNS delivery and reducing systemic side effects. Alisertib, an aurora A kinase inhibitor, has been examined for treatment of several CNS tumors in preclinical and clinical studies. In this study, we investigated the distribution of alisertib into the CNS, the site of efficacy for brain tumors, and into the bone marrow, the site of dose-limiting toxicity leading to myelosuppression. Mechanisms influencing site-specific distribution, such as active transport mediated by the efflux proteins, p-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp), were examined. Alisertib exposure to the brain in wild-type mice was less than 1% of that in the plasma, and was evenly distributed throughout various brain regions and the spinal cord. Studies using transporter knockout mice and pharmacological inhibition show that alisertib CNS distribution is influenced by P-gp, but not Bcrp. Conversely, upon systemic administration, alisertib distribution to the bone marrow occurred rapidly, was not significantly limited by efflux transporters, and reached higher concentrations than in the CNS. This study demonstrates that, given an equivalent distributional driving force exposure in plasma, the exposure of alisertib in the brain is significantly less than that in the bone marrow, suggesting that targeted delivery may be necessary to guarantee therapeutic efficacy with minimal risk for adverse events.Therefore, these data suggest that, to improve the therapeutic index when using alisertib for brain tumors, a localized regional delivery, such as convection-enhanced delivery, may be warranted. SIGNIFICANCE STATEMENT: The CNS penetration of alisertib is limited with uniform distribution in various regions of the brain, and P-gp efflux is an important mechanism limiting that CNS distribution. Alisertib rapidly distributes into the bone marrow, a site of toxicity, with a greater exposure than in the CNS, a possible site of efficacy. These results suggest a need to design localized delivery strategies to improve the CNS exposure of alisertib and limit systemic toxicities in the treatment of brain tumors.

    Topics: Animals; ATP Binding Cassette Transporter, Subfamily B; ATP Binding Cassette Transporter, Subfamily B, Member 1; ATP Binding Cassette Transporter, Subfamily G, Member 2; Aurora Kinase A; Azepines; Bone Marrow; Brain Neoplasms; Central Nervous System; Mice; Mice, Knockout; Neoplasm Proteins; Protein Kinase Inhibitors

2022
Alisertib demonstrates significant antitumor activity in bevacizumab resistant, patient derived orthotopic models of glioblastoma.
    Journal of neuro-oncology, 2017, Volume: 131, Issue:1

    Topics: Animals; Antineoplastic Agents, Immunological; Aurora Kinase A; Azepines; Bevacizumab; Brain Neoplasms; Cell Line, Tumor; Disease Models, Animal; Dose-Response Relationship, Drug; Glioblastoma; Histones; Humans; Inhibitory Concentration 50; Mice; Mice, Inbred BALB C; Mice, Nude; Protein Kinase Inhibitors; Pyrimidines; Survival Analysis; Xenograft Model Antitumor Assays

2017
miR-124 suppresses glioblastoma growth and potentiates chemosensitivity by inhibiting AURKA.
    Biochemical and biophysical research communications, 2017, 04-22, Volume: 486, Issue:1

    Glioblastoma (GBM) accounts for about half of all malignant brain cancers. Although the treatment strategies for glioblastoma develop rapidly, a considerable number of patients could not benefit from temozolomide (TMZ)-based chemotherapy. Here, we revealed a miR-124-AURKA axis that regulated glioblastoma growth and chemosensitivity. Mechanistically, AURKA was up-regulated in glioblastoma tissues and associated with poor overall survival. While overexpression of AURKA enhanced tumor growth, genetic or pharmacological inhibition of AURKA led to growth-inhibitory and chemopotentiating effects in glioblastoma. AURKA was further identified as a target of miR-124. Furthermore, our data showed that miR-124 down-regulated AURKA expression and subsequently suppressed cell growth. Re-expression of AURKA significantly rescued miR124-mediated proliferation repression and chemosensitivity. In conclusion, our results demonstrated that miR-124 inhibited glioblastoma growth and potentiated chemosensitivity by targeting AURKA, which may represent promising targets and rational therapeutic options for glioblastoma.

    Topics: 3' Untranslated Regions; Aged; Antineoplastic Combined Chemotherapy Protocols; Aurora Kinase A; Azepines; Base Sequence; Blotting, Western; Brain Neoplasms; Cell Line, Tumor; Cell Proliferation; Dacarbazine; Female; Gene Expression Regulation, Neoplastic; Glioblastoma; Humans; Kaplan-Meier Estimate; Male; MicroRNAs; Middle Aged; Pyrimidines; Reverse Transcriptase Polymerase Chain Reaction; RNA Interference; Sequence Homology, Nucleic Acid; Temozolomide; Tumor Burden; Xenograft Model Antitumor Assays

2017
Alisertib is active as single agent in recurrent atypical teratoid rhabdoid tumors in 4 children.
    Neuro-oncology, 2015, Volume: 17, Issue:6

    Aurora Kinase A (AURKA) encodes a protein that regulates the formation and stability of the mitotic spindle and is highly active in atypical teratoid rhabdoid tumors (ATRT) through loss of the INI1 tumor suppressor gene. Alisertib (MLN8237) inhibits AURKA in vitro and in vivo. Given the strong preclinical data supporting the use of alisertib for ATRT patients, we sought and obtained permission to use alisertib in single patient treatment plans for 4 recurrent pediatric ATRT patients.. Patients with recurrent or progressive ATRT received alisertib 80 mg/m(2) by mouth once daily for 7 days of a 21-day treatment cycle. Disease evaluation (MRI of brain and spine and lumbar puncture) was done after 2 cycles of alisertib and every 2-3 cycles thereafter for as long as the patients remained free from tumor progression.. Four patients with median age of 2.5 years (range, 1.39-4.87 y) at diagnosis received alisertib 80 mg/m(2) by mouth once daily for 7 days of a 21-day treatment cycle, and all 4 patients had disease stabilization and/or regression after 3 cycles of alisertib therapy. Two patients continued to have stable disease regression for 1 and 2 years, respectively, on therapy.. Single-agent alisertib produced marked and durable regression in disease burden, as detected by brain and spine MRI and by evaluation of spinal fluid cytology. Alisertib has moderate but manageable toxicities, and its chronic administration appears feasible in this pediatric population. These novel data support the incorporation of alisertib in future therapeutic trials for children with ATRT.

    Topics: Antineoplastic Agents; Aurora Kinase A; Azepines; Brain; Brain Neoplasms; Child, Preschool; Female; Humans; Infant; Male; Pyrimidines; Rhabdoid Tumor; Teratoma; Treatment Outcome

2015
Targeted delivery of silver nanoparticles and alisertib: in vitro and in vivo synergistic effect against glioblastoma.
    Nanomedicine (London, England), 2014, Volume: 9, Issue:6

    Targeted biocompatible nanoplatforms presenting multiple therapeutic functions have great potential for the treatment of cancer.. Multifunctional nanocomposites formed by polymeric nanoparticles (PNPs) containing two cytotoxic agents - the drug alisertib and silver nanoparticles - were synthesized. These PNPs have been conjugated with a chlorotoxin, an active targeting 36-amino acid-long peptide that specifically binds to MMP-2, a receptor overexpressed by brain cancer cells.. The individual and synergistic activity of these two cytotoxic agents against glioblastoma multiforme was tested both in vitro and in vivo. The induced cytotoxicity in a human glioblastoma-astrocytoma epithelial-like cell line (U87MG) was studied in vitro through a trypan blue exclusion test after 48 and 72 h of exposure. Subsequently, the PNPs' biodistribution in healthy animals and their effect on tumor reduction in tumor-bearing mice were studied using PNPs radiolabeled with (99m)Tc.. Tumor reduction was achieved in vivo when using silver/alisertib@PNPs-chlorotoxin.

    Topics: Animals; Antineoplastic Agents; Azepines; Brain Neoplasms; Cell Line, Tumor; Drug Delivery Systems; Drug Synergism; Glioblastoma; Humans; Mice; Nanoparticles; Polymers; Pyrimidines; Scorpion Venoms; Silver; Tissue Distribution

2014
Differential sensitivity of Glioma stem cells to Aurora kinase A inhibitors: implications for stem cell mitosis and centrosome dynamics.
    Stem cell research, 2014, Volume: 13, Issue:1

    Glioma stem-cell-like cells are considered to be responsible for treatment resistance and tumour recurrence following chemo-radiation in glioblastoma patients, but specific targets by which to kill the cancer stem cell population remain elusive. A characteristic feature of stem cells is their ability to undergo both symmetric and asymmetric cell divisions. In this study we have analysed specific features of glioma stem cell mitosis. We found that glioma stem cells appear to be highly prone to undergo aberrant cell division and polyploidization. Moreover, we discovered a pronounced change in the dynamic of mitotic centrosome maturation in these cells. Accordingly, glioma stem cell survival appeared to be strongly dependent on Aurora A activity. Unlike differentiated cells, glioma stem cells responded to moderate Aurora A inhibition with spindle defects, polyploidization and a dramatic increase in cellular senescence, and were selectively sensitive to Aurora A and Plk1 inhibitor treatment. Our study proposes inhibition of centrosomal kinases as a novel strategy to selectively target glioma stem cells.

    Topics: Animals; Aurora Kinase A; Azepines; Brain Neoplasms; Cell Line, Tumor; Centrosome; Glioblastoma; HeLa Cells; Humans; Mice; Mitosis; Neoplastic Stem Cells; Polyploidy; Protein Kinase Inhibitors; Pyrimidines; Xenograft Model Antitumor Assays

2014
Aurora kinases as targets in drug-resistant neuroblastoma cells.
    PloS one, 2014, Volume: 9, Issue:9

    Aurora kinase inhibitors displayed activity in pre-clinical neuroblastoma models. Here, we studied the effects of the pan-aurora kinase inhibitor tozasertib (VX680, MK-0457) and the aurora kinase inhibitor alisertib (MLN8237) that shows some specificity for aurora kinase A over aurora kinase B in a panel of neuroblastoma cell lines with acquired drug resistance. Both compounds displayed anti-neuroblastoma activity in the nanomolar range. The anti-neuroblastoma mechanism included inhibition of aurora kinase signalling as indicated by decreased phosphorylation of the aurora kinase substrate histone H3, cell cycle inhibition in G2/M phase, and induction of apoptosis. The activity of alisertib but not of tozasertib was affected by ABCB1 expression. Aurora kinase inhibitors induced a p53 response and their activity was enhanced in combination with the MDM2 inhibitor and p53 activator nutlin-3 in p53 wild-type cells. In conclusion, aurora kinases are potential drug targets in therapy-refractory neuroblastoma, in particular for the vast majority of p53 wild-type cases.

    Topics: Antineoplastic Agents; Apoptosis; ATP Binding Cassette Transporter, Subfamily B; Aurora Kinase A; Aurora Kinase B; Azepines; Brain Neoplasms; Cell Cycle Checkpoints; Cell Proliferation; Drug Resistance, Neoplasm; Drug Synergism; Gene Expression Regulation, Neoplastic; Histones; Humans; Imidazoles; Neuroblastoma; Phosphorylation; Piperazines; Primary Cell Culture; Protein Kinase Inhibitors; Proto-Oncogene Proteins c-mdm2; Pyrimidines; Signal Transduction; Tumor Suppressor Protein p53

2014
Aurora A is differentially expressed in gliomas, is associated with patient survival in glioblastoma and is a potential chemotherapeutic target in gliomas.
    Cell cycle (Georgetown, Tex.), 2012, Feb-01, Volume: 11, Issue:3

    Aurora A is critical for mitosis and is overexpressed in several neoplasms. Its overexpression transforms cultured cells, and both its overexpression and knockdown cause genomic instability. In transgenic mice, Aurora A haploinsufficiency, not overexpression, leads to increased malignant tumor formation. Aurora A thus appears to have both tumor-promoting and tumor-suppressor functions. Here, we report that Aurora A protein, measured by quantitative protein gel blotting, is differentially expressed in major glioma types in lineage-specific patterns. Aurora A protein levels in WHO grade II oligodendrogliomas (n=16) and grade III anaplastic oligodendrogliomas (n=16) are generally low, similar to control epilepsy cerebral tissue (n=11). In contrast, pilocytic astrocytomas (n=6) and ependymomas (n=12) express high Aurora A levels. Among grade II to grade III astrocytomas (n=7, n=14, respectively) and grade IV glioblastomas (n=31), Aurora A protein increases with increasing tumor grade. We also found that Aurora A expression is induced by hypoxia in cultured glioblastoma cells and is overexpressed in hypoxic regions of glioblastoma tumors. Retrospective Kaplan-Meier analysis revealed that both lower Aurora A protein measured by quantitative protein gel blot (n=31) and Aurora A mRNA levels measured by real-time quantitative RT-PCR (n=58) are significantly associated with poorer patient survival in glioblastoma. Furthermore, we report that the selective Aurora A inhibitor MLN8237 is potently cytotoxic to glioblastoma cells, and that MLN8237 cytotoxicty is potentiated by ionizing radiation. MLN8237 also appeared to induce senescence and differentiation of glioblastoma cells. Thus, in addition to being significantly associated with survival in glioblastoma, Aurora A is a potential new drug target for the treatment of glioblastoma and possibly other glial neoplasms.

    Topics: Adolescent; Adult; Aged; Aurora Kinase A; Aurora Kinases; Azepines; Biomarkers; Brain Neoplasms; Cell Differentiation; Cell Line, Tumor; Cellular Senescence; Child; Female; Gene Expression Regulation, Neoplastic; Glioblastoma; Glioma; Humans; Kaplan-Meier Estimate; Male; Middle Aged; Neoplasm Staging; Protein Serine-Threonine Kinases; Pyrimidines; Radiation, Ionizing; Retrospective Studies; RNA, Messenger; Young Adult

2012