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.
Br J Cancer ; 119(10): 1233-1243, 2018 11.
Article in English | MEDLINE | ID: mdl-30385821

ABSTRACT

BACKGROUND: AZD0156 and AZD6738 are potent and selective inhibitors of ataxia-telangiectasia-kinase (ATM) and ataxia-telangiectasia-mutated and Rad3-related (ATR), respectively, important sensors/signallers of DNA damage. METHODS: We used multiplexed targeted-mass-spectrometry to select pRAD50(Ser635) as a pharmacodynamic biomarker for AZD0156-mediated ATM inhibition from a panel of 45 peptides, then developed and tested a clinically applicable immunohistochemistry assay for pRAD50(Ser635) detection in FFPE tissue. RESULTS: We found moderate pRAD50 baseline levels across cancer indications. pRAD50 was detectable in 100% gastric cancers (n = 23), 99% colorectal cancers (n = 102), 95% triple-negative-breast cancers (TNBC) (n = 40) and 87.5% glioblastoma-multiformes (n = 16). We demonstrated AZD0156 target inhibition in TNBC patient-derived xenograft models; where AZD0156 monotherapy or post olaparib treatment, resulted in a 34-72% reduction in pRAD50. Similar inhibition of pRAD50 (68%) was observed following ATM inhibitor treatment post irinotecan in a colorectal cancer xenograft model. ATR inhibition, using AZD6738, increased pRAD50 in the ATM-proficient models whilst in ATM-deficient models the opposite was observed, suggesting pRAD50 pharmacodynamics post ATR inhibition may be ATM-dependent and could be useful to determine ATM functionality in patients treated with ATR inhibitors. CONCLUSION: Together these data support clinical utilisation of pRAD50 as a biomarker of AZD0156 and AZD6738 pharmacology to elucidate clinical pharmacokinetic/pharmacodynamic relationships, thereby informing recommended Phase 2 dose/schedule.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Mass Spectrometry/methods , Animals , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/metabolism , Biomarkers/metabolism , Cell Line , DNA Damage , Humans , Immunohistochemistry , Indoles , Irinotecan/pharmacology , Mice , Mice, Nude , Morpholines , Phthalazines/pharmacology , Piperazines/pharmacology , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Signal Transduction , Sulfonamides , Sulfoxides/pharmacology , Sulfoxides/therapeutic use , Triple Negative Breast Neoplasms , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 17(8): 1637-1647, 2018 08.
Article in English | MEDLINE | ID: mdl-29769307

ABSTRACT

Inhibition of ataxia-telangiectasia mutated (ATM) during radiotherapy of glioblastoma multiforme (GBM) may improve tumor control by short-circuiting the response to radiation-induced DNA damage. A major impediment for clinical implementation is that current inhibitors have limited central nervous system (CNS) bioavailability; thus, the goal was to identify ATM inhibitors (ATMi) with improved CNS penetration. Drug screens and refinement of lead compounds identified AZ31 and AZ32. The compounds were then tested in vivo for efficacy and impact on tumor and healthy brain. Both AZ31 and AZ32 blocked the DNA damage response and radiosensitized GBM cells in vitro AZ32, with enhanced blood-brain barrier (BBB) penetration, was highly efficient in vivo as radiosensitizer in syngeneic and human, orthotopic mouse glioma model compared with AZ31. Furthermore, human glioma cell lines expressing mutant p53 or having checkpoint-defective mutations were particularly sensitive to ATMi radiosensitization. The mechanism for this p53 effect involves a propensity to undergo mitotic catastrophe relative to cells with wild-type p53. In vivo, apoptosis was >6-fold higher in tumor relative to healthy brain after exposure to AZ32 and low-dose radiation. AZ32 is the first ATMi with oral bioavailability shown to radiosensitize glioma and improve survival in orthotopic mouse models. These findings support the development of a clinical-grade, BBB-penetrating ATMi for the treatment of GBM. Importantly, because many GBMs have defective p53 signaling, the use of an ATMi concurrent with standard radiotherapy is expected to be cancer-specific, increase the therapeutic ratio, and maintain full therapeutic effect at lower radiation doses. Mol Cancer Ther; 17(8); 1637-47. ©2018 AACR.


Subject(s)
Blood-Brain Barrier/metabolism , Glioma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Administration, Oral , Animals , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Cell Line, Tumor , Humans , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Radiation-Sensitizing Agents/pharmacology
3.
J Immunol ; 171(8): 4203-9, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-14530343

ABSTRACT

It is well established that Janus kinase (JAK) tyrosine kinases play a key role in the activation of STAT6 by IL-4. In this study, we investigated additional molecules involved in this process. We previously found that IL-4 and TNF-alpha cooperate in the activation of STAT6 and NF-kappaB, suggesting that these transcription factors are regulated by common intracellular signaling pathways. To test this hypothesis, we analyzed the effect of known inhibitors of NF-kappaB on the activation of STAT6. We discovered that inhibitors of phosphatidylcholine-specific phospholipase C (PC-PLC), but not other lipases, blocked the activation of STAT6 by IL-4. The activation of PC-PLC seems to be an early event in IL-4 signaling, because its inhibition abrogated JAK activation and STAT6 tyrosine phosphorylation. Interestingly, we found that the effects of pervanadate and sodium orthovanadate on STAT6 activation correspond to their effect on PC-PLC. Thus, pervanadate by itself activated PC-PLC, JAK, and STAT6, whereas sodium orthovanadate suppressed PC-PLC, JAK, and STAT6 activation by IL-4. We further found that PC-PLC activation is necessary but not sufficient to promote STAT6 activation, and therefore, additional intracellular pathways regulated by IL-4 and pervanadate may collaborate with PC-PLC to signal STAT6 activation. It has been reported that IL-4 signals PC-PLC activation; in this study, we provide evidence that this phospholipase plays a key role in IL-4 signaling.


Subject(s)
Phosphatidylcholines/metabolism , Signal Transduction , Trans-Activators/metabolism , Type C Phospholipases/metabolism , Animals , Bridged-Ring Compounds/pharmacology , Cell Line , Cell Line, Tumor , Enzyme Activation/drug effects , Enzyme Activation/immunology , Enzyme Inhibitors/pharmacology , Hydrolysis/drug effects , Interleukin-4/physiology , Janus Kinase 1 , Janus Kinase 3 , Mice , Norbornanes , Protein-Tyrosine Kinases/metabolism , STAT6 Transcription Factor , Signal Transduction/drug effects , Signal Transduction/immunology , Substrate Specificity/drug effects , Substrate Specificity/immunology , Thiocarbamates , Thiones/pharmacology , Trans-Activators/antagonists & inhibitors , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/physiology , Vanadium Compounds/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...