Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Cancer ; 116(9): 1166-1176, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28334731

ABSTRACT

BACKGROUND: The main role of the cell cycle is to enable error-free DNA replication, chromosome segregation and cytokinesis. One of the best characterised checkpoint pathways is the spindle assembly checkpoint, which prevents anaphase onset until the appropriate attachment and tension across kinetochores is achieved. MPS1 kinase activity is essential for the activation of the spindle assembly checkpoint and has been shown to be deregulated in human tumours with chromosomal instability and aneuploidy. Therefore, MPS1 inhibition represents an attractive strategy to target cancers. METHODS: To evaluate CCT271850 cellular potency, two specific antibodies that recognise the activation sites of MPS1 were used and its antiproliferative activity was determined in 91 human cancer cell lines. DLD1 cells with induced GFP-MPS1 and HCT116 cells were used in in vivo studies to directly measure MPS1 inhibition and efficacy of CCT271850 treatment. RESULTS: CCT271850 selectively and potently inhibits MPS1 kinase activity in biochemical and cellular assays and in in vivo models. Mechanistically, tumour cells treated with CCT271850 acquire aberrant numbers of chromosomes and the majority of cells divide their chromosomes without proper alignment because of abrogation of the mitotic checkpoint, leading to cell death. We demonstrated a moderate level of efficacy of CCT271850 as a single agent in a human colorectal carcinoma xenograft model. CONCLUSIONS: CCT271850 is a potent, selective and orally bioavailable MPS1 kinase inhibitor. On the basis of in vivo pharmacodynamic vs efficacy relationships, we predict that more than 80% inhibition of MPS1 activity for at least 24 h is required to achieve tumour stasis or regression by CCT271850.


Subject(s)
Cell Cycle Proteins/genetics , Heterocyclic Compounds, 4 or More Rings/administration & dosage , M Phase Cell Cycle Checkpoints/drug effects , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Animals , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , HCT116 Cells , Humans , Mice , Neoplasms/genetics , Neoplasms/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
2.
J Med Chem ; 59(11): 5221-37, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27167172

ABSTRACT

Multiparameter optimization of a series of 5-((4-aminopyridin-2-yl)amino)pyrazine-2-carbonitriles resulted in the identification of a potent and selective oral CHK1 preclinical development candidate with in vivo efficacy as a potentiator of deoxyribonucleic acid (DNA) damaging chemotherapy and as a single agent. Cellular mechanism of action assays were used to give an integrated assessment of compound selectivity during optimization resulting in a highly CHK1 selective adenosine triphosphate (ATP) competitive inhibitor. A single substituent vector directed away from the CHK1 kinase active site was unexpectedly found to drive the selective cellular efficacy of the compounds. Both CHK1 potency and off-target human ether-a-go-go-related gene (hERG) ion channel inhibition were dependent on lipophilicity and basicity in this series. Optimization of CHK1 cellular potency and in vivo pharmacokinetic-pharmacodynamic (PK-PD) properties gave a compound with low predicted doses and exposures in humans which mitigated the residual weak in vitro hERG inhibition.


Subject(s)
4-Aminopyridine/analogs & derivatives , Checkpoint Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyrazines/pharmacology , 4-Aminopyridine/chemical synthesis , 4-Aminopyridine/chemistry , 4-Aminopyridine/pharmacology , Checkpoint Kinase 1/metabolism , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrazines/chemical synthesis , Pyrazines/chemistry , Structure-Activity Relationship
3.
Oncotarget ; 7(3): 2329-42, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26295308

ABSTRACT

CCT245737 is the first orally active, clinical development candidate CHK1 inhibitor to be described. The IC50 was 1.4 nM against CHK1 enzyme and it exhibited>1,000-fold selectivity against CHK2 and CDK1. CCT245737 potently inhibited cellular CHK1 activity (IC50 30-220 nM) and enhanced gemcitabine and SN38 cytotoxicity in multiple human tumor cell lines and human tumor xenograft models. Mouse oral bioavailability was complete (100%) with extensive tumor exposure. Genotoxic-induced CHK1 activity (pS296 CHK1) and cell cycle arrest (pY15 CDK1) were inhibited both in vitro and in human tumor xenografts by CCT245737, causing increased DNA damage and apoptosis. Uniquely, we show CCT245737 enhanced gemcitabine antitumor activity to a greater degree than for higher doses of either agent alone, without increasing toxicity, indicating a true therapeutic advantage for this combination. Furthermore, development of a novel ELISA assay for pS296 CHK1 autophosphorylation, allowed the quantitative measurement of target inhibition in a RAS mutant human tumor xenograft of NSCLC at efficacious doses of CCT245737. Finally, CCT245737 also showed significant single-agent activity against a MYC-driven mouse model of B-cell lymphoma. In conclusion, CCT245737 is a new CHK1 inhibitor clinical development candidate scheduled for a first in man Phase I clinical trial, that will use the novel pS296 CHK1 ELISA to monitor target inhibition.


Subject(s)
4-Aminopyridine/analogs & derivatives , Carcinoma, Non-Small-Cell Lung/drug therapy , Checkpoint Kinase 1/drug effects , Lung Neoplasms/drug therapy , Lymphoma, B-Cell/drug therapy , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Pyrazines/pharmacology , Xenograft Model Antitumor Assays , 4-Aminopyridine/pharmacokinetics , 4-Aminopyridine/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , CDC2 Protein Kinase , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA Damage/drug effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Synergism , HT29 Cells , Humans , Irinotecan , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, Transgenic , Pyrazines/pharmacokinetics , Gemcitabine
4.
J Med Chem ; 55(22): 10229-40, 2012 Nov 26.
Article in English | MEDLINE | ID: mdl-23082860

ABSTRACT

Inhibitors of checkpoint kinase 1 (CHK1) are of current interest as potential antitumor agents, but the most advanced inhibitor series reported to date are not orally bioavailable. A novel series of potent and orally bioavailable 3-alkoxyamino-5-(pyridin-2-ylamino)pyrazine-2-carbonitrile CHK1 inhibitors was generated by hybridization of two lead scaffolds derived from fragment-based drug design and optimized for CHK1 potency and high selectivity using a cell-based assay cascade. Efficient in vivo pharmacokinetic assessment was used to identify compounds with prolonged exposure following oral dosing. The optimized compound (CCT244747) was a potent and highly selective CHK1 inhibitor, which modulated the DNA damage response pathway in human tumor xenografts and showed antitumor activity in combination with genotoxic chemotherapies and as a single agent.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Neuroblastoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Pyrimidines/pharmacology , Administration, Oral , Aminopyridines/chemical synthesis , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemical synthesis , Checkpoint Kinase 1 , Child , Colonic Neoplasms/enzymology , DNA Damage/drug effects , Drug Design , Humans , Mice , Mice, Nude , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/enzymology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemical synthesis , Protein Kinases/metabolism , Pyrimidines/chemical synthesis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 18(20): 5650-61, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22929806

ABSTRACT

PURPOSE: Many tumors exhibit defective cell-cycle checkpoint control and increased replicative stress. CHK1 is critically involved in the DNA damage response and maintenance of replication fork stability. We have therefore discovered a novel potent, highly selective, orally active ATP-competitive CHK1 inhibitor, CCT244747, and present its preclinical pharmacology and therapeutic activity. EXPERIMENTAL DESIGN: Cellular CHK1 activity was assessed using an ELISA assay, and cytotoxicity a SRB assay. Biomarker modulation was measured using immunoblotting, and cell-cycle effects by flow cytometry analysis. Single-agent oral CCT244747 antitumor activity was evaluated in a MYCN-driven transgenic mouse model of neuroblastoma by MRI and in genotoxic combinations in human tumor xenografts by growth delay. RESULTS: CCT244747 inhibited cellular CHK1 activity (IC(50) 29-170 nmol/L), significantly enhanced the cytotoxicity of several anticancer drugs, and abrogated drug-induced S and G(2) arrest in multiple tumor cell lines. Biomarkers of CHK1 (pS296 CHK1) activity and cell-cycle inactivity (pY15 CDK1) were induced by genotoxics and inhibited by CCT244747 both in vitro and in vivo, producing enhanced DNA damage and apoptosis. Active tumor concentrations of CCT244747 were obtained following oral administration. The antitumor activity of both gemcitabine and irinotecan were significantly enhanced by CCT244747 in several human tumor xenografts, giving concomitant biomarker modulation indicative of CHK1 inhibition. CCT244747 also showed marked antitumor activity as a single agent in a MYCN-driven neuroblastoma. CONCLUSION: CCT244747 represents the first structural disclosure of a highly selective, orally active CHK1 inhibitor and warrants further evaluation alone or combined with genotoxic anticancer therapies.


Subject(s)
Aminopyridines/administration & dosage , Neoplasms, Experimental , Neuroblastoma , Protein Kinases , Pyrimidines/administration & dosage , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Checkpoints/drug effects , Checkpoint Kinase 1 , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Transgenic , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinases/genetics , Protein Kinases/metabolism
6.
Clin Cancer Res ; 18(14): 3912-23, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22781553

ABSTRACT

PURPOSE: Deregulated phosphatidylinositol 3-kinase pathway signaling through AGC kinases including AKT, p70S6 kinase, PKA, SGK and Rho kinase is a key driver of multiple cancers. The simultaneous inhibition of multiple AGC kinases may increase antitumor activity and minimize clinical resistance compared with a single pathway component. EXPERIMENTAL DESIGN: We investigated the detailed pharmacology and antitumor activity of the novel clinical drug candidate AT13148, an oral ATP-competitive multi-AGC kinase inhibitor. Gene expression microarray studies were undertaken to characterize the molecular mechanisms of action of AT13148. RESULTS: AT13148 caused substantial blockade of AKT, p70S6K, PKA, ROCK, and SGK substrate phosphorylation and induced apoptosis in a concentration and time-dependent manner in cancer cells with clinically relevant genetic defects in vitro and in vivo. Antitumor efficacy in HER2-positive, PIK3CA-mutant BT474 breast, PTEN-deficient PC3 human prostate cancer, and PTEN-deficient MES-SA uterine tumor xenografts was shown. We show for the first time that induction of AKT phosphorylation at serine 473 by AT13148, as reported for other ATP-competitive inhibitors of AKT, is not a therapeutically relevant reactivation step. Gene expression studies showed that AT13148 has a predominant effect on apoptosis genes, whereas the selective AKT inhibitor CCT128930 modulates cell-cycle genes. Induction of upstream regulators including IRS2 and PIK3IP1 as a result of compensatory feedback loops was observed. CONCLUSIONS: The clinical candidate AT13148 is a novel oral multi-AGC kinase inhibitor with potent pharmacodynamic and antitumor activity, which shows a distinct mechanism of action from other AKT inhibitors. AT13148 will now be assessed in a first-in-human phase I trial.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms , Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Pyrimidines/administration & dosage , Pyrroles/administration & dosage , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 13(23): 7191-8, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18056200

ABSTRACT

PURPOSE: The effective treatment of ovarian cancer is hampered by the development of drug resistance, which may be mediated by members of the Bcl-2 family of apoptosis regulators. ABT-737 is a recently described inhibitor of members of this family. We investigated whether this compound could sensitize ovarian cancer cells to chemotherapeutic agents. EXPERIMENTAL DESIGN: The sensitivity of ovarian cancer cell lines to ABT-737 in combination with either carboplatin or paclitaxel was tested either in vitro by assessing cell growth/survival and apoptosis or in xenograft studies. RESULTS: As a single agent, ABT-737 inhibited the growth of eight ovarian cancer cell lines, although with relatively poor potency. However, ABT-737, but not a less active enantiomer, increased the sensitivity of several cell lines to carboplatin. The increased sensitivity to carboplatin was accompanied by a decrease in time at which apoptosis was observed when assessed according to the number of attached cells, PARP cleavage, and nucleosome formation. ABT-737 was more effective at sensitizing IGROV-1 cells when ABT-737 was administered after carboplatin. In addition, ABT-737 significantly enhanced the activity of carboplatin in one of three primary cultures derived directly from ascitic tumor cells in patients recently treated with chemotherapy. Small interfering RNA directed to Bcl-X(L) also increased the sensitivity of ovarian cancer cell lines to carboplatin. ABT-737 was also able to augment the inhibition of IGROV-1 tumor xenograft growth beyond that obtained with carboplatin alone. CONCLUSIONS: These data suggest that ABT-737, in combination with carboplatin, may find utility in the treatment of patients with ovarian cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biphenyl Compounds/pharmacology , Carboplatin/pharmacology , Nitrophenols/pharmacology , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , Animals , Apoptosis/drug effects , Biphenyl Compounds/administration & dosage , Carboplatin/administration & dosage , Cell Line, Tumor , Drug Administration Schedule , Drug Synergism , Female , Humans , Mice , Nitrophenols/administration & dosage , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Piperazines/administration & dosage , Piperazines/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays , bcl-X Protein/genetics
8.
Assay Drug Dev Technol ; 5(3): 391-401, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17638539

ABSTRACT

In this age of molecularly targeted drug discovery, robust techniques are required to measure pharmacodynamic (PD) responses in tumors so that drug exposures can be associated with their effects on molecular biomarkers and efficacy. Our aim was to develop a rapid screen to monitor PD responses within xenografted human tumors as an important step towards a clinically applicable technology. Currently there are various methods available to measure PD end points, including immunohistochemistry, enzyme-linked immunosorbent assay (ELISA), reverse transcription-polymerase chain reaction, gene expression profiling, and western blotting. These may require relatively large samples of tumor, surrogate tissue, or peripheral blood lymphocytes with subsequent analyses taking several days. The phosphoinositide 3-kinase (PI3-kinase) pathway is frequently deregulated in cancer and is also important in diabetes and autoimmune conditions. In this paper, optimization of the Meso Scale Discovery (MSD) (Gaithersburg, MD) platform to quantify changes in phospho-AKT and phospho-glycogen synthase kinase-3beta in response to a PI3-kinase inhibitor, LY294002, is described, initially in vitro and then within xenografted solid tumors. This method is highly practical with high throughput since large number of samples can be run simultaneously in 96-well format. The assays are robust (coefficient of variation for phospho-AKT 13.4%) and offer significant advantages (in terms of speed and quantitation) over western blots. This optimized procedure can be used for both in vitro and in vivo analysis, unlike an established fixed-cell ELISA with a time-resolved fluorescent end point.


Subject(s)
Chromones/therapeutic use , Glycogen Synthase Kinase 3/metabolism , Morpholines/therapeutic use , Neoplasms, Experimental/chemistry , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/analysis , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line, Tumor , Female , Glycogen Synthase Kinase 3 beta , Humans , Mice , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Reproducibility of Results , Specimen Handling , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...