Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Cancer Chemother Pharmacol ; 61(1): 23-32, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17909809

ABSTRACT

Demonstration of pharmacodynamic activity of new, targeted cancer drugs in tumour tissue is potentially important in guiding early drug development. However, delays between tumour sampling and sample fixation may result in variability of pharmacodynamic biomarkers. The aim of this study, was to assess the impact of delays in fixation on biomarkers of Src kinase activity. A total of 20 patients with locally advanced breast cancer and 5 with early bladder cancer had multiple tissue samples taken which were fixed at documented time points up to 60 min after biopsy. These were examined to determine if the amount of Paxillin, phospho-Paxillin, phospho-focal adhesion kinase (FAK) and total phospho-Tyrosine changed over time, using a quantitative lysate immunoassay. In breast cancer, there was an increase in the amount of phospho-Paxillin (60% per h; P = 0.019) up to 60 min after biopsy. The amount of total Paxillin decreased (28% per h; P = 0.034) over the same time course. In early bladder cancer, no changes were noted in any endpoints up to 45 min. Standardisation of the time taken between biopsy and fixation may be critical, particularly in studies using phosphorylated protein biomarkers.


Subject(s)
Breast Neoplasms/enzymology , Tissue Fixation/methods , Urinary Bladder Neoplasms/enzymology , src-Family Kinases/metabolism , Aged , Aged, 80 and over , Biomarkers , Biopsy , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Freezing , Humans , Immunoassay , Male , Middle Aged , Paxillin/metabolism , Phosphorylation , Phosphotyrosine/metabolism , Time Factors
2.
J Thorac Oncol ; 12(10): 1588-1594, 2017 10.
Article in English | MEDLINE | ID: mdl-28751247

ABSTRACT

INTRODUCTION: Osimertinib is an oral, potent, irreversible EGFR tyrosine kinase inhibitor (TKI) selective for EGFR TKI and T790M resistance mutations. To enhance understanding of osimertinib's mechanism of action, we aimed to evaluate the modulation of key molecular biomarkers after osimertinib treatment in paired clinical samples from the phase I AURA trial. METHODS: Paired tumor biopsy samples were collected before the study and after 15 plus or minus 7 days of osimertinib treatment (80 or 160 mg daily). Clinical efficacy outcomes were assessed according to whether viable paired biopsy samples could be collected; safety was also assessed. Immunohistochemical analyses assessed key pathway and tumor/immune-relevant markers (phospho-EGFR, phospho-S6, phospho-AKT, programmed death ligand 1, and CD8), with samples scored by image analysis or a pathologist blinded to treatment allocation. RESULTS: Predose tumor biopsy samples were collected from 61 patients with EGFR T790M tumors; 29 patients had no viable postdose biopsy sample because of tumor regression or insufficient tumor sample. Evaluable predose and postdose tumor biopsy samples were collected from 24 patients. Objective response rate (ORR) and median progression-free survival (mPFS) were improved in patients from whom a postdose biopsy sample could not be collected (ORR 62% and mPFS 9.7 months [p = 0.027]) compared with those from whom paired samples were collected (ORR 29% and mPFS 6.6 months). Osimertinib modulated key EGFR signaling pathways and led to increased immune cell infiltration. CONCLUSIONS: Collection of paired biopsy samples was challenging because of rapid tumor regression after osimertinib treatment, highlighting the difficulties of performing on-study biopsies in patients treated with highly active drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/metabolism , Biopsy/methods , Piperazines/therapeutic use , Acrylamides , Aniline Compounds , Antineoplastic Agents/pharmacology , Cohort Studies , Disease-Free Survival , Humans , Piperazines/pharmacology
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
4.
Mol Cancer Ther ; 14(11): 2441-51, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26351323

ABSTRACT

AKT1(E17K) mutations occur at low frequency in a variety of solid tumors, including those of the breast and urinary bladder. Although this mutation has been shown to transform rodent cells in culture, it was found to be less oncogenic than PIK3CA mutations in breast epithelial cells. Moreover, the therapeutic potential of AKT inhibitors in human tumors with an endogenous AKT1(E17K) mutation is not known. Expression of exogenous copies of AKT1(E17K) in MCF10A breast epithelial cells increased phosphorylation of AKT and its substrates, induced colony formation in soft agar, and formation of lesions in the mammary fat pad of immunodeficient mice. These effects were inhibited by the allosteric and catalytic AKT inhibitors MK-2206 and AZD5363, respectively. Both AKT inhibitors caused highly significant growth inhibition of breast cancer explant models with AKT1(E17K) mutation. Furthermore, in a phase I clinical study, the catalytic Akt inhibitor AZD5363 induced partial responses in patients with breast and ovarian cancer with tumors containing AKT1(E17K) mutations. In MGH-U3 bladder cancer xenografts, which contain both AKT1(E17K) and FGFR3(Y373C) mutations, AZD5363 monotherapy did not significantly reduce tumor growth, but tumor regression was observed in combination with the FGFR inhibitor AZD4547. The data show that tumors with AKT1(E17K) mutations are rational therapeutic targets for AKT inhibitors, although combinations with other targeted agents may be required where activating oncogenic mutations of other proteins are present in the same tumor.


Subject(s)
Mutation, Missense , Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Doxycycline/pharmacology , Female , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Male , Mice, Nude , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrroles/administration & dosage , Pyrroles/pharmacology , Pyrroles/therapeutic use , Signal Transduction/drug effects , Xenograft Model Antitumor Assays/methods
5.
Cancer Discov ; 4(9): 1046-61, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24893891

ABSTRACT

UNLABELLED: First-generation EGFR tyrosine kinase inhibitors (EGFR TKI) provide significant clinical benefit in patients with advanced EGFR-mutant (EGFRm(+)) non-small cell lung cancer (NSCLC). Patients ultimately develop disease progression, often driven by acquisition of a second T790M EGFR TKI resistance mutation. AZD9291 is a novel oral, potent, and selective third-generation irreversible inhibitor of both EGFRm(+) sensitizing and T790M resistance mutants that spares wild-type EGFR. This mono-anilino-pyrimidine compound is structurally distinct from other third-generation EGFR TKIs and offers a pharmacologically differentiated profile from earlier generation EGFR TKIs. Preclinically, the drug potently inhibits signaling pathways and cellular growth in both EGFRm(+) and EGFRm(+)/T790M(+) mutant cell lines in vitro, with lower activity against wild-type EGFR lines, translating into profound and sustained tumor regression in EGFR-mutant tumor xenograft and transgenic models. The treatment of 2 patients with advanced EGFRm(+) T790M(+) NSCLC is described as proof of principle. SIGNIFICANCE: We report the development of a novel structurally distinct third-generation EGFR TKI, AZD9291, that irreversibly and selectively targets both sensitizing and resistant T790M(+) mutant EGFR while harboring less activity toward wild-type EGFR. AZD9291 is showing promising responses in a phase I trial even at the first-dose level, with first published clinical proof-of-principle validation being presented.


Subject(s)
Acrylamides/therapeutic use , Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation , Protein Kinase Inhibitors/therapeutic use , Acrylamides/chemistry , Acrylamides/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , ErbB Receptors/chemistry , Female , Genes, erbB-2 , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Models, Molecular , Molecular Conformation , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Treatment Outcome , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
6.
Mol Cancer Ther ; 12(9): 1715-27, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23861347

ABSTRACT

Continued androgen receptor (AR) expression and signaling is a key driver in castration-resistant prostate cancer (CRPC) after classical androgen ablation therapies have failed, and therefore remains a target for the treatment of progressive disease. Here, we describe the biological characterization of AZD3514, an orally bioavailable drug that inhibits androgen-dependent and -independent AR signaling. AZD3514 modulates AR signaling through two distinct mechanisms, an inhibition of ligand-driven nuclear translocation of AR and a downregulation of receptor levels, both of which were observed in vitro and in vivo. AZD3514 inhibited testosterone-driven seminal vesicle development in juvenile male rats and the growth of androgen-dependent Dunning R3327H prostate tumors in adult rats. Furthermore, this class of compound showed antitumor activity in the HID28 mouse model of CRPC in vivo. AZD3514 is currently in phase I clinical evaluation.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Antineoplastic Agents/pharmacology , Prostatic Neoplasms, Castration-Resistant/pathology , Pyridazines/pharmacology , Receptors, Androgen/metabolism , Seminal Vesicles/drug effects , Abiraterone Acetate , Androgen Receptor Antagonists/metabolism , Androstadienes/pharmacology , Animals , Antineoplastic Agents/metabolism , Benzamides , Cell Line, Tumor , Disease Models, Animal , Down-Regulation , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Male , Mice , Mice, Nude , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms, Castration-Resistant/drug therapy , Pyridazines/chemical synthesis , Pyridazines/metabolism , Rats , Rats, Wistar , Receptors, Androgen/genetics , Seminal Vesicles/growth & development , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
7.
Mol Imaging Biol ; 14(3): 355-65, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21761255

ABSTRACT

PURPOSE: The aim of this study is to assess the variability of 2-deoxy-2-[(18)F]fluoro-D: -glucose ([(18)F]-FDG) and 3'-deoxy-3'-[(18)F]-fluorothymidine ([(18)F]-FLT) uptake in pre-clinical tumor models and examine the relationship between imaging data and related histological biomarkers. PROCEDURES: [(18)F]-FDG and [(18)F]-FLT studies were carried out in nine human tumor xenograft models in mice. A selection of the models underwent histological analysis for endpoints relevant to radiotracer uptake. Comparisons were made between in vitro uptake, in vivo imaging, and ex vivo histopathology data using quantitative and semi-quantitative analysis. RESULTS: In vitro data revealed that [1-(14)C]-2-deoxy-D: -glucose ([(14)C]-2DG) uptake in the tumor cell lines was variable. In vivo, [(18)F]-FDG and [(18)F]-FLT uptake was highly variable across tumor types and uptake of one tracer was not predictive for the other. [(14)C]-2DG uptake in vitro did not predict for [(18)F]-FDG uptake in vivo. [(18)F]-FDG SUV was inversely proportional to Ki67 and necrosis levels and positively correlated with HKI. [(18)F]-FLT uptake positively correlated with Ki67 and TK1. CONCLUSION: When evaluating imaging biomarkers in response to therapy, the choice of tumor model should take into account in vivo baseline radiotracer uptake, which can vary significantly between models.


Subject(s)
Dideoxynucleosides/pharmacokinetics , Fluorodeoxyglucose F18/pharmacokinetics , Neoplasms, Experimental/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Female , Histocytochemistry , Humans , Mice , Mice, Nude , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/pathology , Positron-Emission Tomography , Research Design , Transplantation, Heterologous , Whole Body Imaging
8.
Mol Oncol ; 3(3): 248-61, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19393585

ABSTRACT

AZD0530, an orally available Src inhibitor, demonstrated potent antimigratory and anti-invasive effects in vitro, and inhibited metastasis in a murine model of bladder cancer. Antiproliferative activity of AZD0530 in vitro varied between cell lines (IC(50) 0.2 ->10µM). AZD0530 inhibited tumor growth in 4/10 xenograft models tested and dynamically inhibited in vivo phosphorylation of Src substrates paxillin and FAK in both growth-inhibition-resistant and -sensitive xenografts. The activity of AZD0530 in NBT-II bladder cancer cells in vitro was consistent with inhibition of cell migration and stabilization of cell-cell adhesion. These data suggest a dominant anti-invasive pharmacology for AZD0530 that may limit tumor progression in a range of cancers. AZD0530 is currently in Phase II clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Benzodioxoles/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Urinary Bladder Neoplasms/drug therapy , Xenograft Model Antitumor Assays , src-Family Kinases/antagonists & inhibitors , Administration, Oral , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Clinical Trials, Phase II as Topic , Focal Adhesion Kinase 1/metabolism , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Paxillin/metabolism , Phosphorylation/drug effects , Rats , Rats, Nude , Transplantation, Heterologous , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/pathology , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL