Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Cancer ; 5(1): 66-84, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38151625

ABSTRACT

Chromosomal instability (CIN) is a hallmark of cancer, caused by persistent errors in chromosome segregation during mitosis. Aggressive cancers like high-grade serous ovarian cancer (HGSOC) and triple-negative breast cancer (TNBC) have a high frequency of CIN and TP53 mutations. Here, we show that inhibitors of the KIF18A motor protein activate the mitotic checkpoint and selectively kill chromosomally unstable cancer cells. Sensitivity to KIF18A inhibition is enriched in TP53-mutant HGSOC and TNBC cell lines with CIN features, including in a subset of CCNE1-amplified, CDK4-CDK6-inhibitor-resistant and BRCA1-altered cell line models. Our KIF18A inhibitors have minimal detrimental effects on human bone marrow cells in culture, distinct from other anti-mitotic agents. In mice, inhibition of KIF18A leads to robust anti-cancer effects with tumor regression observed in human HGSOC and TNBC models at well-tolerated doses. Collectively, our results provide a rational therapeutic strategy for selective targeting of CIN cancers via KIF18A inhibition.


Subject(s)
Kinesins , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Kinesins/genetics , Kinesins/metabolism , Mitosis/genetics , Cell Line , M Phase Cell Cycle Checkpoints
2.
J Med Chem ; 65(6): 4972-4990, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35286090

ABSTRACT

Chromosomal instability (CIN) is a hallmark of cancer that results from errors in chromosome segregation during mitosis. Targeting of CIN-associated vulnerabilities is an emerging therapeutic strategy in drug development. KIF18A, a mitotic kinesin, has been shown to play a role in maintaining bipolar spindle integrity and promotes viability of CIN cancer cells. To explore the potential of KIF18A, a series of inhibitors was identified. Optimization of an initial hit led to the discovery of analogues that could be used as chemical probes to interrogate the role of KIF18A inhibition. Compounds 23 and 24 caused significant mitotic arrest in vivo, which was sustained for 24 h. This would be followed by cell death either in mitosis or in the subsequent interphase. Furthermore, photoaffinity labeling experiments reveal that this series of inhibitors binds at the interface of KIF18A and tubulin. This study represents the first disclosure of KIF18A inhibitors with in vivo activity.


Subject(s)
Kinesins , Neoplasms , Cell Death , Humans , Mitosis , Neoplasms/drug therapy , Neoplasms/metabolism , Spindle Apparatus/metabolism , Tubulin/metabolism
3.
Mol Cancer Ther ; 17(12): 2575-2585, 2018 12.
Article in English | MEDLINE | ID: mdl-30266802

ABSTRACT

Aurora kinase A and B have essential and non-overlapping roles in mitosis, with elevated expression in a subset of human cancers, including acute myeloid leukemia (AML). In this study, pan-aurora kinase inhibitor (AKI) AMG 900 distinguishes itself as an anti-leukemic agent that is more uniformly potent against a panel of AML cell lines than are isoform-selective AKIs and classic AML drugs. AMG 900 inhibited AML cell growth by inducing polyploidization and/or apoptosis. AMG 900 and aurora-B-selective inhibitor AZD1152-hQPA showed comparable cellular effects on AML lines that do not harbor a FLT3-ITD mutation. AMG 900 was active against P-glycoprotein-expressing AML cells resistant to AZD1152-hQPA and was effective at inducing expression of megakaryocyte-lineage markers (CD41, CD42) on human CHRF-288-11 cells and mouse Jak2 V617F cells. In MOLM-13 cells, inhibition of p-histone H3 by AMG 900 was associated with polyploidy, extra centrosomes, accumulation of p53 protein, apoptosis, and cleavage of Bcl-2 protein. Co-administration of cytarabine (Ara-C) with AMG 900 potentiated cell killing in a subset of AML lines, with evidence of attenuated polyploidization. AMG 900 inhibited the proliferation of primary human bone marrow cells in culture, with a better proliferation recovery profile relative to classic antimitotic drug docetaxel. In vivo, AMG 900 significantly reduced tumor burden in a systemic MOLM-13 xenograft model where we demonstrate the utility of 3'-deoxy-3'-18F-fluorothymidine [18F]FLT positron emission tomographic (PET)-CT imaging to measure the antiproliferative effects of AMG 900 in skeletal tissues in mice.


Subject(s)
Aurora Kinases/antagonists & inhibitors , Leukemia, Myeloid, Acute/pathology , Mitosis/drug effects , Phthalazines/pharmacology , Animals , Apoptosis/drug effects , Aurora Kinases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , G1 Phase/drug effects , Humans , Megakaryocytes/drug effects , Megakaryocytes/metabolism , Megakaryocytes/pathology , Mice , Organophosphates/pharmacology , Polyploidy , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Quinazolines/pharmacology , Tumor Burden , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
4.
J Med Chem ; 58(13): 5189-207, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-25970324

ABSTRACT

Efforts to improve upon the physical properties and metabolic stability of Aurora kinase inhibitor 14a revealed that potency against multidrug-resistant cell lines was compromised by increased polarity. Despite its high in vitro metabolic intrinsic clearance, 23r (AMG 900) showed acceptable pharmacokinetic properties and robust pharmacodynamic activity. Projecting from in vitro data to in vivo target coverage was not practical due to disjunctions between enzyme and cell data, complex and apparently contradictory indicators of binding kinetics, and unmeasurable free fraction in plasma. In contrast, it was straightforward to relate pharmacokinetics to pharmacodynamics and efficacy by following the time above a threshold concentration. On the basis of its oral route of administration, a selectivity profile that favors Aurora-driven pharmacology and its activity against multidrug-resistant cell lines, 23r was identified as a potential best-in-class Aurora kinase inhibitor. In phase 1 dose expansion studies with G-CSF support, 23r has shown promising single agent activity.


Subject(s)
Aurora Kinases/antagonists & inhibitors , Drug Discovery , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Cell Proliferation/drug effects , Female , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms/enzymology , Neoplasms/pathology , Rats , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Mol Cancer Ther ; 12(11): 2356-66, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23990115

ABSTRACT

Breast cancer is the most prevalent malignancy affecting women and ranks second in cancer-related deaths, in which death occurs primarily from metastatic disease. Triple-negative breast cancer (TNBC) is a more aggressive and metastatic subtype of breast cancer that is initially responsive to treatment of microtubule-targeting agents (MTA) such as taxanes. Recently, we reported the characterization of AMG 900, an orally bioavailable, potent, and highly selective pan-Aurora kinase inhibitor that is active in multidrug-resistant cell lines. In this report, we investigate the activity of AMG 900 alone and in combination with two distinct classes of MTAs (taxanes and epothilones) in multidrug-resistant TNBC cell lines and xenografts. In TNBC cells, AMG 900 inhibited phosphorylation of histone H3 on Ser(10), a proximal substrate of Aurora-B, and induced polyploidy and apoptosis. Furthermore, AMG 900 potentiated the antiproliferative effects of paclitaxel and ixabepilone at low nanomolar concentrations. In mice, AMG 900 significantly inhibited the growth of MDA-MB-231 (F(11); parental), MDA-MB-231 (F(11)) PTX-r (paclitaxel-resistant variant), and DU4475 xenografts. The combination of AMG 900 with docetaxel enhanced tumor inhibition in MDA-MB-231 (F(11)) xenografts compared with either monotherapy. Notably, combining AMG 900 with ixabepilone resulted in regressions of MDA-MB-231 (F(11)) PTX-r xenografts, in which more than 50% of the tumors failed to regrow 75 days after the cessation of drug treatment. These findings suggest that AMG 900, alone and in combination with MTAs, may be an effective intervention strategy for the treatment of metastatic breast cancer and provide potential therapeutic options for patients with multidrug-resistant tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinases/antagonists & inhibitors , Neoplasm Metastasis/pathology , Phthalazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Triple Negative Breast Neoplasms/pathology , Animals , Antineoplastic Combined Chemotherapy Protocols , Aurora Kinases/metabolism , Cell Death/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Epothilones/pharmacology , Female , Humans , Mammary Neoplasms, Experimental , Mice , Mice, Nude , Neoplasm Metastasis/drug therapy , Paclitaxel/pharmacology , Phosphorylation/drug effects , Polyploidy , Triple Negative Breast Neoplasms/drug therapy , Tubulin Modulators/pharmacology , Xenograft Model Antitumor Assays
6.
J Med Chem ; 53(17): 6368-77, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20684549

ABSTRACT

The discovery of aurora kinases as essential regulators of cell division has led to intense interest in identifying small molecule aurora kinase inhibitors for the potential treatment of cancer. A high-throughput screening effort identified pyridinyl-pyrimidine 6a as a moderately potent dual inhibitor of aurora kinases -A and -B. Optimization of this hit resulted in an anthranilamide lead (6j) that possessed improved enzyme and cellular activity and exhibited a high level of kinase selectivity. However, this anthranilamide and subsequent analogues suffered from a lack of oral bioavailability. Converting the internally hydrogen-bonded six-membered pseudo-ring of the anthranilamide to a phthalazine (8a-b) led to a dramatic improvement in oral bioavailability (38-61%F) while maintaining the potency and selectivity characteristics of the anthranilamide series. In a COLO 205 tumor pharmacodynamic assay measuring phosphorylation of the aurora-B substrate histone H3 at serine 10 (p-histone H3), oral administration of 8b at 50 mg/kg demonstrated significant reduction in tumor p-histone H3 for at least 6 h.


Subject(s)
Antineoplastic Agents/chemical synthesis , Phthalazines/chemical synthesis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/chemical synthesis , Pyrimidines/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Aurora Kinase B , Aurora Kinases , Biological Availability , Blood Proteins/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Histones/metabolism , Humans , In Vitro Techniques , Male , Mice , Mice, Nude , Microsomes, Liver/metabolism , Models, Molecular , Neoplasm Transplantation , Phthalazines/pharmacokinetics , Phthalazines/pharmacology , Protein Binding , Pyridines/pharmacokinetics , Pyridines/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Transplantation, Heterologous
7.
J Biol Chem ; 279(8): 6213-6, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14699147

ABSTRACT

The neonatal Fc receptor (FcRn) plays an important role in regulating the serum half-lives of IgG antibodies. A correlation has been established between the pH-dependent binding affinity of IgG antibodies to FcRn and their serum half-lives in mice. In this study, molecular modeling was used to identify Fc positions near the FcRn binding site in a human IgG antibody that, when mutated, might alter the binding affinity of IgG to FcRn. Following mutagenesis, several IgG2 mutants with increased binding affinity to human FcRn at pH 6.0 were identified at Fc positions 250 and 428. These mutants do not bind to human FcRn at pH 7.5. A pharmacokinetics study of two mutant IgG2 antibodies with increased FcRn binding affinity indicated that they had serum half-lives in rhesus monkeys approximately 2-fold longer than the wild-type antibody.


Subject(s)
Immunoglobulin G/blood , Immunoglobulin G/chemistry , Animals , Antibodies/chemistry , Binding Sites , Binding Sites, Antibody , Binding, Competitive , Cell Line , Cloning, Molecular , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Half-Life , Histocompatibility Antigens Class I , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Inhibitory Concentration 50 , Kidney/cytology , Macaca mulatta , Models, Molecular , Mutagenesis , Mutation , Protein Binding , Receptors, Fc/chemistry , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...