Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Biomol Screen ; 18(3): 298-308, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23139381

ABSTRACT

Inositol-requiring enzyme 1 alpha (IRE1α) is a transmembrane sensor protein with both kinase and ribonuclease activity, which plays a crucial role in the unfolded protein response (UPR). Protein misfolding in the endoplasmic reticulum (ER) lumen triggers dimerization and subsequent trans-autophosphorylation of IRE1α. This leads to the activation of its endoribonuclease (RNase) domain and splicing of the mRNA of the transcriptional activator XBP1, ultimately generating an active XBP1 (XBP1s) implicated in multiple myeloma survival. Previously, we have identified human IRE1α as a target for the development of kinase inhibitors that could modulate the UPR in human cells, which has particular relevance for multiple myeloma and other secretory malignancies. Here we describe the development and validation of a 384-well high-throughput screening assay using DELFIA technology that is specific for IRE1α autophosphorylation. Using this format, a focused library of 2312 potential kinase inhibitors was screened, and several novel IRE1α kinase inhibitor scaffolds were identified that could potentially be developed toward new therapies to treat multiple myeloma.


Subject(s)
Endoribonucleases/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line , Endoribonucleases/chemistry , Endoribonucleases/metabolism , High-Throughput Screening Assays/methods , Humans , Insecta , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/enzymology , Multiple Myeloma/metabolism , Phosphorylation/drug effects , Protein Folding/drug effects , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Unfolded Protein Response/drug effects
2.
Clin Cancer Res ; 17(6): 1561-70, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21278242

ABSTRACT

PURPOSE: A phase I study to define toxicity and recommend a phase II dose of the HSP90 inhibitor alvespimycin (17-DMAG; 17-dimethylaminoethylamino-17-demethoxygeldanamycin). Secondary endpoints included evaluation of pharmacokinetic profile, tumor response, and definition of a biologically effective dose (BED). PATIENTS AND METHODS: Patients with advanced solid cancers were treated with weekly, intravenous (i.v.) 17-DMAG. An accelerated titration dose escalation design was used. The maximum tolerated dose (MTD) was the highest dose at which ≤ 1/6 patients experienced dose limiting toxicity (DLT). Dose de-escalation from the MTD was planned with mandatory, sequential tumor biopsies to determine a BED. Pharmacokinetic and pharmacodynamic assays were validated prior to patient accrual. RESULTS: Twenty-five patients received 17-DMAG (range 2.5-106 mg/m(2)). At 106 mg/m(2) of 17-DMAG 2/4 patients experienced DLT, including one treatment-related death. No DLT occurred at 80 mg/m(2). Common adverse events were gastrointestinal, liver function changes, and ocular. Area under the curve and mean peak concentration increased proportionally with 17-DMAG doses 80 mg/m(2) or less. In peripheral blood mononuclear cells significant (P < 0.05) HSP72 induction was detected (≥ 20 mg/m(2)) and sustained for 96 hours (≥ 40 mg/m(2)). Plasma HSP72 levels were greatest in the two patients who experienced DLT. At 80 mg/m(2) client protein (CDK4, LCK) depletion was detected and tumor samples from 3 of 5 patients confirmed HSP90 inhibition. Clinical activity included complete response (castration refractory prostate cancer, CRPC 124 weeks), partial response (melanoma, 159 weeks), and stable disease (chondrosarcoma, CRPC, and renal cancer for 28, 59, and 76 weeks, respectively). CONCLUSIONS: The recommended phase II dose of 17-DMAG is 80 mg/m(2) weekly i.v.


Subject(s)
Benzoquinones/pharmacology , Lactams, Macrocyclic/pharmacology , Neoplasms/drug therapy , Adult , Aged , Biopsy , Blotting, Western , Cohort Studies , Enzyme-Linked Immunosorbent Assay/methods , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Leukocytes, Mononuclear/cytology , Male , Maximum Tolerated Dose , Middle Aged
3.
EMBO J ; 30(5): 894-905, 2011 Mar 02.
Article in English | MEDLINE | ID: mdl-21317875

ABSTRACT

Ire1 (Ern1) is an unusual transmembrane protein kinase essential for the endoplasmic reticulum (ER) unfolded protein response (UPR). Activation of Ire1 by association of its N-terminal ER luminal domains promotes autophosphorylation by its cytoplasmic kinase domain, leading to activation of the C-terminal ribonuclease domain, which splices Xbp1 mRNA generating an active Xbp1s transcriptional activator. We have determined the crystal structure of the cytoplasmic portion of dephosphorylated human Ire1α bound to ADP, revealing the 'phosphoryl-transfer' competent dimeric face-to-face complex, which precedes and is distinct from the back-to-back RNase 'active' conformation described for yeast Ire1. We show that the Xbp1-specific ribonuclease activity depends on autophosphorylation, and that ATP-competitive inhibitors staurosporin and sunitinib, which inhibit autophosphorylation in vitro, also inhibit Xbp1 splicing in vivo. Furthermore, we demonstrate that activated Ire1α is a competent protein kinase, able to phosphorylate a heterologous peptide substrate. These studies identify human Ire1α as a target for development of ATP-competitive inhibitors that will modulate the UPR in human cells, which has particular relevance for myeloma and other secretory malignancies.


Subject(s)
Cell Nucleus/genetics , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum/metabolism , Endoribonucleases/chemistry , Endoribonucleases/metabolism , Gene Expression Regulation , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , Transcription Factors/metabolism , Unfolded Protein Response/physiology , Blotting, Western , Crystallography, X-Ray , Cytoplasm , DNA-Binding Proteins/genetics , Endoribonucleases/genetics , Humans , Membrane Proteins/genetics , Phosphorylation , Protein Folding , Protein Multimerization , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription, Genetic , X-Box Binding Protein 1
4.
Neuro Oncol ; 13(1): 99-108, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21163810

ABSTRACT

Angiogenesis plays a key role in glioblastoma biology and antiangiogenic agents are under clinical investigation with promising results. However, the angiogenic profiles of patients with glioblastoma and their clinical significance are not well understood. Here we characterize the serum angiogenic profile of patients with glioblastoma, and examine the prognostic significance of individual angiogenic factors. Serum samples from 36 patients with glioblastoma were collected on admission and simultaneously assayed for 48 angiogenic factors using protein microarrays. The data were analyzed using hierarchical cluster analysis. Vessel morphology was assessed histologically after immunostaining for the pan-endothelial marker CD31. Tumor samples were also immunostained for tissue inhibitor of metalloproteinase-1 (TIMP-1). Cluster analysis of the serum angiogenic profiles revealed 2 distinct subtypes of glioblastoma. The 2 subtypes had markedly different tumor microvessel densities. A low serum level of TIMP-1 was associated with significantly longer survival independent of patient age, performance status, or treatment. The serum angiogenic profile in patients with glioblastoma mirrors tumor biology and has prognostic value. Our data suggest the serum TIMP-1 level as an independent predictor of survival.


Subject(s)
Angiogenesis Inducing Agents/blood , Biomarkers, Tumor/blood , Brain Neoplasms/classification , Glioblastoma/classification , Neovascularization, Pathologic/blood , Tissue Inhibitor of Metalloproteinase-1/blood , Adult , Aged , Brain Neoplasms/blood supply , Brain Neoplasms/pathology , Case-Control Studies , Female , Glioblastoma/blood supply , Glioblastoma/pathology , Humans , Immunoenzyme Techniques , Male , Microcirculation , Middle Aged , Protein Array Analysis
5.
J Med Chem ; 53(24): 8663-78, 2010 Dec 23.
Article in English | MEDLINE | ID: mdl-21080647

ABSTRACT

A novel series of HDAC inhibitors demonstrating class I subtype selectivity and good oral bioavailability is described. The compounds are potent enzyme inhibitors (IC50 values less than 100 nM), and improved activity in cell proliferation assays was achieved by modulation of polar surface area (PSA) through the introduction of novel linking groups. Employing oral pharmacokinetic studies in mice, comparing drug levels in spleen to plasma, we selected compounds that were tested for efficacy in human tumor xenograft studies based on their potential to distribute into tumor. One compound, 21r (CHR-3996), showed good oral activity in these models, including dose-related activity in a LoVo xenograft. In addition 21r showed good activity in combination with other anticancer agents in in vitro studies. On the basis of these results, 21r was nominated for clinical development.


Subject(s)
Antineoplastic Agents/chemical synthesis , Azabicyclo Compounds/chemical synthesis , Histone Deacetylase Inhibitors/chemical synthesis , Pyrimidines/chemical synthesis , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Azabicyclo Compounds/pharmacokinetics , Azabicyclo Compounds/pharmacology , Cell Line, Tumor , Dogs , Drug Screening Assays, Antitumor , Drug Synergism , Female , Histone Deacetylase Inhibitors/pharmacokinetics , Histone Deacetylase Inhibitors/pharmacology , Humans , In Vitro Techniques , Mice , Mice, Nude , Microsomes, Liver/metabolism , Models, Molecular , Neoplasm Transplantation , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship , Tissue Distribution , Transplantation, Heterologous
6.
Bioorg Med Chem Lett ; 20(22): 6657-60, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20884208

ABSTRACT

Inhibition of histone deacetylase activity represents a promising new modality in the treatment of a number of cancers. A novel HDAC series demonstrating inhibitory activity in cell proliferation assays is described. Optimisation based on the introduction of basic amine linkers to effect good drug distribution to tumour led to the identification of a compound with oral activity in a human colon cancer xenograft study associated with increased histone H3 acetylation in tumour tissue.


Subject(s)
Drug Design , Histone Deacetylase Inhibitors/chemical synthesis , Hydroxamic Acids/chemical synthesis , Pyrimidines/chemistry , Cell Line, Tumor , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/chemistry , Hydroxamic Acids/pharmacology , Transplantation, Heterologous
7.
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
8.
Mol Cancer Ther ; 6(4): 1198-211, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17431102

ABSTRACT

Although the heat shock protein 90 (HSP90) inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) shows clinical promise, potential limitations encourage development of alternative chemotypes. We discovered the 3,4-diarylpyrazole resorcinol CCT018159 by high-throughput screening and used structure-based design to generate more potent pyrazole amide analogues, exemplified by VER-49009. Here, we describe the detailed biological properties of VER-49009 and the corresponding isoxazole VER-50589. X-ray crystallography showed a virtually identical HSP90 binding mode. However, the dissociation constant (K(d)) of VER-50589 was 4.5 +/- 2.2 nmol/L compared with 78.0 +/- 10.4 nmol/L for VER-49009, attributable to higher enthalpy for VER-50589 binding. A competitive binding assay gave a lower IC(50) of 21 +/- 4 nmol/L for VER-50589 compared with 47 +/- 9 nmol/L for VER-49009. Cellular uptake of VER-50589 was 4-fold greater than for VER-49009. Mean cellular antiproliferative GI(50) values for VER-50589 and VER-49009 for a human cancer cell line panel were 78 +/- 15 and 685 +/- 119 nmol/L, respectively, showing a 9-fold potency gain for the isoxazole. Unlike 17-AAG, but as with CCT018159, cellular potency of these analogues was independent of NAD(P)H:quinone oxidoreductase 1/DT-diaphorase and P-glycoprotein expression. Consistent with HSP90 inhibition, VER-50589 and VER-49009 caused induction of HSP72 and HSP27 alongside depletion of client proteins, including C-RAF, B-RAF, and survivin, and the protein arginine methyltransferase PRMT5. Both caused cell cycle arrest and apoptosis. Extent and duration of pharmacodynamic changes in an orthotopic human ovarian carcinoma model confirmed the superiority of VER-50589 over VER-49009. VER-50589 accumulated in HCT116 human colon cancer xenografts at levels above the cellular GI(50) for 24 h, resulting in 30% growth inhibition. The results indicate the therapeutic potential of the resorcinylic pyrazole/isoxazole amide analogues as HSP90 inhibitors.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/pharmacology , Pyrazoles/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Resistance, Neoplasm/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Female , HCT116 Cells , HSP90 Heat-Shock Proteins/chemistry , HT29 Cells , Humans , Isoxazoles/chemistry , Isoxazoles/pharmacokinetics , Mice , Mice, Nude , NAD(P)H Dehydrogenase (Quinone)/metabolism , Protein Binding/drug effects , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 6(3): 1112-22, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17363504

ABSTRACT

Histone deacetylases (HDACs), histone acetyltransferases (HATs), and the molecular chaperone heat shock protein 90 (HSP90) are attractive anticancer drug targets. High-throughput screening plays a pivotal role in modern molecular mechanism-based drug discovery. Cell-based screens are particularly useful in that they identify compounds that are permeable and active against the selected target or pathway in a cellular context. We have previously developed time-resolved fluorescence cell immunosorbent assays (TRF-Cellisas) for compound screening and pharmacodynamic studies. These assays use a primary antibody to the single protein of interest and a matched secondary immunoglobulin labeled with an europium chelate (Eu). The availability of species-specific secondary antibodies labeled with different lanthanide chelates provides the potential for multiplexing this type of assay. The approach has been applied to the development of a 384-well duplexed cell-based screen to simultaneously detect compounds that induce the co-chaperone HSP70 as a molecular marker of potential inhibitors of HSP90 together with those that modulate cellular acetylation (i.e., potential inhibitors of histone deacetylase or histone acetyltransferase activity). The duplexed assay proved reliable in high-throughput format and approximately 64,000 compounds were screened. Following evaluation in secondary assays, 3 of 13 hits from the HSP70 arm were confirmed. Two of these directly inhibited the intrinsic ATPase activity of HSP90 whereas the third seems to have a different mechanism of action. In the acetylation arm, two compounds increased cellular acetylation, one of which inhibited histone deacetylase activity. A third compound decreased cellular histone acetylation, potentially through a novel mechanism of action.


Subject(s)
Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Histone Acetyltransferases/antagonists & inhibitors , Histone Deacetylase Inhibitors , Protein Processing, Post-Translational/drug effects , Acetylation/drug effects , Adenosine Triphosphatases/metabolism , Cell Extracts/analysis , HCT116 Cells/drug effects , HSP90 Heat-Shock Proteins/metabolism , Heterocyclic Compounds, 2-Ring/pharmacology , Histone Acetyltransferases/metabolism , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Immunoassay , Molecular Chaperones
10.
Assay Drug Dev Technol ; 3(3): 273-85, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15971989

ABSTRACT

High-throughput screening of chemical libraries and the subsequent rapid progress of hit compounds through an iterative developmental test cascade are essential parts of modern molecular mechanism-based drug discovery. These processes depend on the use of efficient assay technologies and equipment. Enzyme-linked immunosorbent assays have historically been carried out in 96-well microtitre plates. Improvements in reagents and assay technologies mean that solid-phase immunoassays can be adapted for higher throughput to play an important role in modern drug discovery. The molecular chaperone heat-shock protein (Hsp) 90 is an important anticancer drug target because it maintains the conformation, stability, and function of many important oncogenic client proteins, including those involved with signal transduction, cell proliferation, survival, differentiation, motility angiogenesis, and metastasis. Using the standard inhibitors of the adenosine triphosphatase (ATPase) activity of Hsp90, geldanamycin (GA) and 17-allylamino-17- demethoxygeldanamycin (17AAG), novel solid-phase immunoassays have been validated using a time-resolved fluorescence (TRF) end point. Their utility for confirming the mechanism of action of Hsp90 inhibition in secondary cell-based assays has been shown and applied to the novel Hsp90 inhibitor CCT018159. Adaptation of these assays for later studies using human tumour xenografts and samples obtained from a Phase 1 trial of 17AAG is also described. Finally, comparison is made between the use and applicability of this type of immunoassay and other techniques such as western blotting, immunohistochemistry, and flow cytometry analysis.


Subject(s)
Drug Design , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Immunoassay/methods , Animals , Benzoquinones , Blotting, Western , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Extracts/analysis , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , HCT116 Cells , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , HT29 Cells , Heterocyclic Compounds, 2-Ring/pharmacology , Humans , Intracellular Signaling Peptides and Proteins , Lactams, Macrocyclic , Lymphocytes/chemistry , Lymphocytes/drug effects , Lymphocytes/metabolism , Mice , Neoplasm Transplantation , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/metabolism , Pyrazoles/pharmacology , Quinones/pharmacology , Rifabutin/analogs & derivatives , Rifabutin/pharmacology , Technology, Pharmaceutical/methods , Technology, Pharmaceutical/trends , Transplantation, Heterologous , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/metabolism
11.
Anal Biochem ; 320(1): 66-74, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12895470

ABSTRACT

The tumor suppressor protein, pRb, regulates progression through the G1 phase of the cell cycle by its ability to bind to and regulate the activity of a variety of transcription factors. This function of pRb is disabled through its phosphorylation by the cyclin-dependent kinase (CDK) family of serine/threonine kinases. In many human cancers, genetic alteration such as loss of CDK inhibitor function and deregulated G1 cyclin expression leads to inappropriate phosphorylation and hence inactivation of this tumor suppressor. Identification of cell-permeable small molecules that block pRb phosphorylation in these tumors could therefore lead to development of an effective anticancer treatment. As a result, we have developed a high-throughput assay to detect changes in the level of pRb phosphorylation in cells. Signal detection is by a time-resolved fluorescence-based cellular immunosorbant assay on a fixed monolayer of cells. This comprises a mouse monoclonal antibody that recognizes the phosphorylated form of serine 608 on pRb, a known site of CDK phosphorylation, and a Europium-labeled secondary antibody for signal detection. The assay is reproducible and amenable to automation and has been used to screen 2000 compounds in a search for cell-permeable small molecules that will block pRb phosphorylation.


Subject(s)
Fluoroimmunoassay/methods , Retinoblastoma Protein/metabolism , Animals , Antibodies, Monoclonal/metabolism , Cell Line, Tumor , Cell Membrane/chemistry , Drug Evaluation, Preclinical/methods , Female , Humans , Mice , Phosphorylation , Purines/pharmacology , Retinoblastoma Protein/antagonists & inhibitors , Roscovitine
13.
Clin Cancer Res ; 9(2): 792-801, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12576452

ABSTRACT

PURPOSE: The purpose of this research was to evaluate the predictive value of expression of thymidylate synthase (TS) and other genes for response to raltitrexed (RTX). EXPERIMENTAL DESIGN: Twenty-five patients with metastatic colorectal cancer received RTX 3 mg/m(2) 3-weekly. Pretreatment tumor biopsies were analyzed for TS, dihydropyrimidine dehydrogenase (DPD), thymidine phosphorylase (TP), folylpolyglutamate synthetase, and reduced folate carrier mRNA expression by real-time reverse transcription-PCR. TS protein expression was evaluated by immunohistochemistry using a polyclonal TS antibody. RESULTS: Twenty patients were evaluable for response and gene expression. Six of 20 (30%) achieved a partial response. Median TS/beta-actin was 5.7 x 10(3) (range, 2.2-42 x 10(3)). Median TS/beta-actin was 3.7 x 10(3) in responding patients and 6.1 x 10(3) in nonresponders (P = 0.048). Five of 6 patients with TS/beta-actin

Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Quinazolines/therapeutic use , Thiophenes/therapeutic use , Thymidylate Synthase/genetics , Antimetabolites, Antineoplastic/blood , Base Sequence , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/pathology , DNA Primers , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation, Enzymologic/drug effects , Humans , Predictive Value of Tests , Protein Biosynthesis , Quinazolines/blood , RNA, Messenger/genetics , Radiography , Reverse Transcriptase Polymerase Chain Reaction , Thiophenes/blood , Thymidylate Synthase/antagonists & inhibitors , Transcription, Genetic , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...