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.
Cell Cycle ; 15(5): 730-9, 2016.
Article in English | MEDLINE | ID: mdl-26890478

ABSTRACT

In order to determine the relative contribution of checkpoint abrogation and subsequent aberrant mitotic entry to gemcitabine chemosensitization by CHK1 inhibition, we established a model utilizing the CDK inhibitors roscovitine or purvalanol A to re-establish cell cycle arrest and prevent aberrant mitotic entry in pancreatic cancer cells treated with gemcitabine and the CHK inhibitor AZD7762. In this study, we report that the extent of aberrant mitotic entry, as determined by flow cytometry for the mitotic marker phospho-Histone H3 (Ser10), did not reflect the relative sensitivities of pancreatic cancer cell lines to gemcitabine chemosensitization by AZD7762. In addition, re-establishing gemcitabine-induced cell cycle arrest either pharmacologically, with roscovitine or purvalanol A, or genetically, with cyclin B1 siRNA, did not inhibit chemosensitization uniformly across the cell lines. Furthermore, we found that AZD7762 augmented high-intensity γH2AX signaling in gemcitabine-treated cells, suggesting the presence of replication stress when CHK1 is inhibited. Finally, the ability of roscovitine to prevent chemosensitization correlated with its ability to inhibit AZD7762-induced high-intensity γH2AX, but not aberrant pHH3, suggesting that the effects of AZD7762 on DNA replication or repair rather than aberrant mitotic entry determine gemcitabine chemosensitization in pancreatic cancer cells.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Deoxycytidine/analogs & derivatives , Thiophenes/pharmacology , Urea/analogs & derivatives , Cell Cycle Checkpoints , Cell Line, Tumor , Cyclin B1/metabolism , DNA Damage , Deoxycytidine/pharmacology , Drug Screening Assays, Antitumor , G2 Phase Cell Cycle Checkpoints , Humans , Mitosis , Nocodazole/pharmacology , Pancreatic Neoplasms/drug therapy , Urea/pharmacology , Gemcitabine
2.
Neoplasia ; 14(6): 519-25, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22787433

ABSTRACT

Checkpoint kinase 1 (Chk1) inhibition sensitizes pancreatic cancer cells and tumors to gemcitabine. We hypothesized that Chk1 inhibition would sensitize pancreatic cancer stem cells to gemcitabine. We tested this hypothesis by using two patient-derived xenograft models (designated J and F) and the pancreatic cancer stem cell markers CD24, CD44, and ESA. We determined the percentage of marker-positive cells and their tumor-initiating capacity (by limiting dilution assays) after treatment with gemcitabine and the Chk1 inhibitor, AZD7762. We found that marker-positive cells were significantly reduced by the combination of gemcitabine and AZD7762. In addition, secondary tumor initiation was significantly delayed in response to primary tumor treatment with gemcitabine + AZD7762 compared with control, gemcitabine, or AZD7762 alone. Furthermore, for the same number of stem cells implanted from gemcitabine- versus gemcitabine + AZD7762-treated primary tumors, secondary tumor initiation at 10 weeks was 83% versus 43%, respectively. We also found that pS345 Chk1, which is a measure of DNA damage, was induced in marker-positive cells but not in the marker-negative cells. These data demonstrate that Chk1 inhibition in combination with gemcitabine reduces both the percentage and the tumor-initiating capacity of pancreatic cancer stem cells. Furthermore, the finding that the Chk1-mediated DNA damage response was greater in stem cells than in non-stem cells suggests that Chk1 inhibition may selectively sensitize pancreatic cancer stem cells to gemcitabine, thus making Chk1 a potential therapeutic target for improving pancreatic cancer therapy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Animals , Antimetabolites, Antineoplastic/administration & dosage , Checkpoint Kinase 1 , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays , Gemcitabine
3.
Clin Cancer Res ; 17(11): 3706-15, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21482692

ABSTRACT

PURPOSE: Chk1 inhibitors, such as AZD7762, are in clinical development in combination with cytotoxic agents for the treatment of solid tumors, including pancreatic cancers. To maximize the likelihood of their clinical success, it is essential to optimize drug scheduling as well as pharmacodynamic biomarkers in preclinical models. EXPERIMENTAL DESIGN: We tested multiple schedules of administration of gemcitabine and AZD7762 on the survival of pancreatic cancer cells. Potential pharmacodynamic biomarkers including pChk1, pChk2, pHistone H3, and caspase-3 were evaluated in vitro, followed by assessment of promising candidate biomarkers in vivo. We then went on to determine the contributions of PP2A and DNA damage to the mechanism(s) of induction of the identified biomarker, pS345 Chk1. RESULTS: AZD7762 given during and after or after gemcitabine administration produced maximum chemosensitization. In vivo, AZD7762 significantly inhibited the growth of pancreatic tumor xenografts in response to gemcitabine. Of the biomarkers assessed, pS345 Chk1 was most consistently increased in response to gemcitabine and AZD7762 in tumors and normal tissues (hair follicles). pS345 Chk1 induction in response to gemcitabine and AZD7762 occurred in the presence of PP2A inhibition and in association with elevated γH2AX, suggesting that DNA damage is an underlying mechanism. CONCLUSIONS: AZD7762 sensitizes pancreatic cancer cells and tumors to gemcitabine in association with induction of pS345 Chk1. Together these data support the clinical investigation of AZD7762 with gemcitabine in pancreatic cancer under a dosing schedule in which gemcitabine is administered concurrent with or before AZD7762 and in conjunction with skin biopsies to measure pS345 Chk1.


Subject(s)
Deoxycytidine/analogs & derivatives , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Thiophenes/pharmacology , Urea/analogs & derivatives , Animals , Biomarkers/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Checkpoint Kinase 1 , DNA Damage , Deoxycytidine/pharmacology , Histones/metabolism , Humans , Mice , Pancreatic Neoplasms/drug therapy , Phosphorylation/drug effects , Transplantation, Heterologous , Urea/pharmacology , Xenograft Model Antitumor Assays , Gemcitabine
4.
Cancer Res ; 70(12): 4972-81, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20501833

ABSTRACT

The median survival for patients with locally advanced pancreatic cancer treated with gemcitabine and radiation is approximately 1 year. To develop improved treatment, we have combined a Chk1/2-targeted agent, AZD7762, currently in phase I clinical trials, with gemcitabine and ionizing radiation in preclinical pancreatic tumor models. We found that in vitro AZD7762 alone or in combination with gemcitabine significantly sensitized MiaPaCa-2 cells to radiation. AZD7762 inhibited Chk1 autophosphorylation (S296 Chk1), stabilized Cdc25A, and increased ATR/ATM-mediated Chk1 phosphorylation (S345 Chk1). Radiosensitization by AZD7762 was associated with abrogation of the G(2) checkpoint as well as with inhibition of Rad51 focus formation, inhibition of homologous recombination repair, and persistent gamma-H2AX expression. AZD7762 was also a radiation sensitizer in multiple tumor xenograft models. In both MiaPaCa-2- and patient-derived xenografts, AZD7762 significantly prolonged the median time required for tumor volume doubling in response to gemcitabine and radiation. Together, our findings suggest that G(2) checkpoint abrogation and homologous recombination repair inhibition both contribute to sensitization by Chk1 inhibition. Furthermore, they support the clinical use of AZD7762 in combination with gemcitabine and radiation for patients with locally advanced pancreatic cancer.


Subject(s)
DNA Repair/drug effects , G2 Phase/drug effects , Pancreatic Neoplasms/radiotherapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Thiophenes/pharmacology , Urea/analogs & derivatives , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Blotting, Western , Cell Line, Tumor , Checkpoint Kinase 1 , Checkpoint Kinase 2 , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/radiation effects , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Therapy, Combination , Flow Cytometry , Fluorescent Antibody Technique , G2 Phase/radiation effects , Gamma Rays , Humans , Immunoblotting , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Protein Kinases/chemistry , Protein Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rad51 Recombinase/metabolism , Recombination, Genetic/drug effects , Recombination, Genetic/radiation effects , Reverse Transcriptase Polymerase Chain Reaction , Urea/pharmacology , Xenograft Model Antitumor Assays , Gemcitabine
5.
Clin Cancer Res ; 16(7): 2076-84, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20233881

ABSTRACT

PURPOSE: Inhibition of checkpoint kinase 1 has been shown to enhance the cytotoxicity of DNA-damaging targeted chemotherapy through cell cycle checkpoint abrogation and impaired DNA damage repair. A novel checkpoint kinase 1/2 inhibitor, AZD7762, was evaluated for potential enhancement of radiosensitivity for human tumor cells in vitro and in vivo xenografts. EXPERIMENTAL DESIGN: Survival of both p53 wild-type and mutant human cell lines was evaluated by clonogenic assay. Dose modification factors (DMF) were determined from survival curves (ratio of radiation doses for control versus drug treated at 10% survival). Flow cytometry, Western blot, and radiation-induced tumor regrowth delay assays were conducted. RESULTS: AZD7762 treatment enhanced the radiosensitivity of p53-mutated tumor cell lines (DMFs ranging from 1.6-1.7) to a greater extent than for p53 wild-type tumor lines (DMFs ranging from 1.1-1.2). AZD7762 treatment alone exhibited little cytotoxicity to any of the cell lines and did not enhance the radiosensitivity of normal human fibroblasts (1522). AZD7762 treatment abrogated radiation-induced G(2) delay, inhibited radiation damage repair (assessed by gamma-H2AX), and suppressed radiation-induced cyclin B expression. HT29 xenografts exposed to five daily radiation fractions and to two daily AZD7762 doses exhibited significant radiation enhancement compared with radiation alone. CONCLUSIONS: AZD7762 effectively enhanced the radiosensitivity of mutated p53 tumor cell lines and HT29 xenografts and was without untoward toxicity when administered alone or in combination with radiation. The results of this study support combining AZD7762 with radiation in clinical trials.


Subject(s)
Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Thiophenes/therapeutic use , Urea/analogs & derivatives , Animals , Cells, Cultured , Checkpoint Kinase 1 , DNA Damage/drug effects , DNA Damage/radiation effects , Dose-Response Relationship, Drug , Female , G2 Phase/drug effects , G2 Phase/radiation effects , HT29 Cells , Humans , Mice , Mice, Nude , Mitotic Index , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/metabolism , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , Thiophenes/pharmacology , Urea/pharmacology , Urea/therapeutic use , Xenograft Model Antitumor Assays
6.
Mol Cancer Ther ; 7(9): 2955-66, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18790776

ABSTRACT

Insights from cell cycle research have led to the hypothesis that tumors may be selectively sensitized to DNA-damaging agents resulting in improved antitumor activity and a wider therapeutic margin. The theory relies on the observation that the majority of tumors are deficient in the G1-DNA damage checkpoint pathway resulting in reliance on S and G2 checkpoints for DNA repair and cell survival. The S and G2 checkpoints are regulated by checkpoint kinase 1, a serine/threonine kinase that is activated in response to DNA damage; thus, inhibition of checkpoint kinase 1 signaling impairs DNA repair and increases tumor cell death. Normal tissues, however, have a functioning G1 checkpoint signaling pathway allowing for DNA repair and cell survival. Here, we describe the preclinical profile of AZD7762, a potent ATP-competitive checkpoint kinase inhibitor in clinical trials. AZD7762 has been profiled extensively in vitro and in vivo in combination with DNA-damaging agents and has been shown to potentiate response in several different settings where inhibition of checkpoint kinase results in the abrogation of DNA damage-induced cell cycle arrest. Dose-dependent potentiation of antitumor activity, when AZD7762 is administered in combination with DNA-damaging agents, has been observed in multiple xenograft models with several DNA-damaging agents, further supporting the potential of checkpoint kinase inhibitors to enhance the efficacy of both conventional chemotherapy and radiotherapy and increase patient response rates in a variety of settings.


Subject(s)
DNA Damage , DNA, Neoplasm/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Thiophenes/pharmacology , Urea/analogs & derivatives , Animals , Biological Assay , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Checkpoint Kinase 1 , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Synergism , G2 Phase/drug effects , HCT116 Cells , HT29 Cells , Humans , Male , Mice , Mutation/genetics , Protein Kinase Inhibitors/analysis , Protein Kinase Inhibitors/chemistry , Rats , Thiophenes/analysis , Thiophenes/chemistry , Topotecan/pharmacology , Tumor Suppressor Protein p53/metabolism , Urea/analysis , Urea/chemistry , Urea/pharmacology , Xenograft Model Antitumor Assays , Gemcitabine
8.
Cancer Cell ; 6(1): 33-43, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15261140

ABSTRACT

Homeostasis under hypoxic conditions is maintained through a coordinated transcriptional response mediated by the hypoxia-inducible factor (HIF) pathway and requires coactivation by the CBP and p300 transcriptional coactivators. Through a target-based high-throughput screen, we identified chetomin as a disrupter of HIF binding to p300. At a molecular level, chetomin disrupts the structure of the CH1 domain of p300 and precludes its interaction with HIF, thereby attenuating hypoxia-inducible transcription. Systemic administration of chetomin inhibited hypoxia-inducible transcription within tumors and inhibited tumor growth. These results demonstrate a therapeutic window for pharmacological attenuation of HIF activity and further establish the feasibility of disrupting a signal transduction pathway by targeting the function of a transcriptional coactivator with a small molecule.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA-Binding Proteins , Nuclear Proteins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Carcinoma, Hepatocellular/pathology , Cell Hypoxia/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Disulfides , E1A-Associated p300 Protein , Erythropoietin/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit , Indole Alkaloids , Liver Neoplasms/pathology , Luciferases/metabolism , Male , Mice , Mice, Nude , Nuclear Proteins/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Protein Binding/drug effects , Receptors, Aryl Hydrocarbon/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Trans-Activators/genetics , Transcription Factors/genetics , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...