Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 18(5): 909-919, 2019 05.
Article in English | MEDLINE | ID: mdl-30872381

ABSTRACT

Barasertib (AZD1152), a pro-drug of the highly potent and selective Aurora B kinase inhibitor AZD2811, showed promising clinical activity in relapsed/refractory diffuse large B-cell lymphoma (DLBCL) patients administered as a 4-day infusion. To improve potential therapeutic benefit of Aurora B kinase inhibition, a nanoparticle formulation of AZD2811 has been developed to address limitations of repeated intravenous infusion. One of the challenges with the use of nanoparticles for chronic treatment of tumors is optimizing dose and schedule required to enable repeat administration to sustain tumor growth inhibition. AZD2811 gives potent cell growth inhibition across a range of DLBCL cells lines in vitro In vivo, repeat administration of the AZD2811 nanoparticle gave antitumor activity at half the dose intensity of AZD1152. Compared with AZD1152, a single dose of AZD2811 nanoparticle gave less reduction in pHH3, but increased apoptosis and reduction of cells in G1 and G2-M, albeit at later time points, suggesting that duration and depth of target inhibition influence the nature of the tumor cell response to drug. Further exploration of the influence of dose and schedule on efficacy revealed that AZD2811 nanoparticle can be used flexibly with repeat administration of 25 mg/kg administered up to 7 days apart being sufficient to maintain equivalent tumor control. Timing of repeat administration could be varied with 50 mg/kg every 2 weeks controlling tumor control as effectively as 25 mg/kg every week. AZD2811 nanoparticle can be administered with very different doses and schedules to inhibit DLBCL tumor growth, although maximal tumor growth inhibition was achieved with the highest dose intensities.


Subject(s)
Acetanilides/pharmacology , Aurora Kinase B/genetics , Lymphoma, Large B-Cell, Diffuse/drug therapy , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Acetanilides/chemistry , Animals , Aurora Kinase B/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Nanoparticles/chemistry , Protein Kinase Inhibitors/chemistry , Quinazolines/chemistry , Xenograft Model Antitumor Assays
2.
Cancer Discov ; 9(2): 230-247, 2019 02.
Article in English | MEDLINE | ID: mdl-30373918

ABSTRACT

Small cell lung cancer (SCLC) accounts for 15% of lung cancers and is almost always linked to inactivating RB1 and TP53 mutations. SCLC frequently responds, albeit briefly, to chemotherapy. The canonical function of the RB1 gene product RB1 is to repress the E2F transcription factor family. RB1 also plays both E2F-dependent and E2F-independent mitotic roles. We performed a synthetic lethal CRISPR/Cas9 screen in an RB1 -/- SCLC cell line that conditionally expresses RB1 to identify dependencies that are caused by RB1 loss and discovered that RB1 -/- SCLC cell lines are hyperdependent on multiple proteins linked to chromosomal segregation, including Aurora B kinase. Moreover, we show that an Aurora B kinase inhibitor is efficacious in multiple preclinical SCLC models at concentrations that are well tolerated in mice. These results suggest that RB1 loss is a predictive biomarker for sensitivity to Aurora B kinase inhibitors in SCLC and perhaps other RB1 -/- cancers. SIGNIFICANCE: SCLC is rarely associated with actionable protooncogene mutations. We did a CRISPR/Cas9-based screen that showed that RB1 -/- SCLC are hyperdependent on AURKB, likely because both genes control mitotic fidelity, and confirmed that Aurora B kinase inhibitors are efficacious against RB1 -/- SCLC tumors in mice at nontoxic doses.See related commentary by Dick and Li, p. 169.This article is highlighted in the In This Issue feature, p. 151.


Subject(s)
Aurora Kinase B/metabolism , Cell Proliferation , Genes, Tumor Suppressor , Lung Neoplasms/pathology , Mutation , Retinoblastoma Binding Proteins/metabolism , Small Cell Lung Carcinoma/pathology , Ubiquitin-Protein Ligases/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Aurora Kinase B/genetics , CRISPR-Cas Systems , Chromosome Segregation , Drug Resistance, Neoplasm , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , Mice, Nude , Retinoblastoma Binding Proteins/antagonists & inhibitors , Retinoblastoma Binding Proteins/genetics , Signal Transduction , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/metabolism , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
3.
Mol Cancer Ther ; 16(6): 1031-1040, 2017 06.
Article in English | MEDLINE | ID: mdl-28292940

ABSTRACT

Barasertib (AZD1152), a highly potent and selective aurora kinase B inhibitor, gave promising clinical activity in elderly acute myeloid leukemia (AML) patients. However, clinical utility was limited by the requirement for a 7-day infusion. Here we assessed the potential of a nanoparticle formulation of the selective Aurora kinase B inhibitor AZD2811 (formerly known as AZD1152-hQPA) in preclinical models of AML. When administered to HL-60 tumor xenografts at a single dose between 25 and 98.7 mg/kg, AZD2811 nanoparticle treatment delivered profound inhibition of tumor growth, exceeding the activity of AZD1152. The improved antitumor activity was associated with increased phospho-histone H3 inhibition, polyploidy, and tumor cell apoptosis. Moreover, AZD2811 nanoparticles increased antitumor activity when combined with cytosine arabinoside. By modifying dose of AZD2811 nanoparticle, therapeutic benefit in a range of preclinical models was further optimized. At high-dose, antitumor activity was seen in a range of models including the MOLM-13 disseminated model. At these higher doses, a transient reduction in bone marrow cellularity was observed demonstrating the potential for the formulation to target residual disease in the bone marrow, a key consideration when treating AML. Collectively, these data establish that AZD2811 nanoparticles have activity in preclinical models of AML. Targeting Aurora B kinase with AZD2811 nanoparticles is a novel approach to deliver a cell-cycle inhibitor in AML, and have potential to improve on the clinical activity seen with cell-cycle agents in this disease. Mol Cancer Ther; 16(6); 1031-40. ©2017 AACR.


Subject(s)
Antineoplastic Agents/administration & dosage , Aurora Kinase B/antagonists & inhibitors , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Nanoparticles , Organophosphates/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Quinazolines/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Line, Tumor , Cytarabine/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Mice , Organophosphates/pharmacokinetics , Polyploidy , Protein Kinase Inhibitors/pharmacokinetics , Quinazolines/pharmacokinetics , Rats , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...