Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 285
Filter
1.
Oncogene ; 39(24): 4780, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32427987

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Blood Cancer J ; 7(3): e549, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28362441

ABSTRACT

Myeloma is heterogeneous at the molecular level with subgroups of patients characterised by features of epigenetic dysregulation. Outcomes for myeloma patients have improved over the past few decades except for molecularly defined high-risk patients who continue to do badly. Novel therapeutic approaches are, therefore, required. A growing number of epigenetic inhibitors are now available including EZH2 inhibitors that are in early-stage clinical trials for treatment of haematological and other cancers with EZH2 mutations or in which overexpression has been correlated with poor outcomes. For the first time, we have identified and validated a robust and independent deleterious effect of high EZH2 expression on outcomes in myeloma patients. Using two chemically distinct small-molecule inhibitors, we demonstrate a reduction in myeloma cell proliferation with EZH2 inhibition, which leads to cell cycle arrest followed by apoptosis. This is mediated via upregulation of cyclin-dependent kinase inhibitors associated with removal of the inhibitory H3K27me3 mark at their gene loci. Our results suggest that EZH2 inhibition may be a potential therapeutic strategy for the treatment of myeloma and should be investigated in clinical studies.


Subject(s)
Cell Cycle Checkpoints/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression , Multiple Myeloma/genetics , Multiple Myeloma/mortality , Apoptosis/genetics , Biomarkers , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Epigenesis, Genetic , Gene Expression Profiling , Histones/metabolism , Humans , Kaplan-Meier Estimate , Mesenchymal Stem Cells/metabolism , Multiple Myeloma/diagnosis , Multiple Myeloma/therapy , Phenotype , Prognosis , Proportional Hazards Models , RNA, Messenger/genetics
3.
Psychol Med ; 46(15): 3065-3080, 2016 11.
Article in English | MEDLINE | ID: mdl-27605134

ABSTRACT

BACKGROUND: It is well established that people with irritable bowel syndrome (IBS) have higher levels of anxiety and depression compared with controls. However, the role of these as risk factors is less clearly established. The aims of this systematic review were to investigate: (1) whether anxiety and/or depression predict IBS onset; (2) the size of the relative risk (RR) of anxiety versus depression in IBS onset. Subgroup analyses explored if methodological factors affected the overall findings. METHOD: Prospective cohort or case-control studies were included if they: (1) focused on the development of IBS in population-based or gastroenteritis cohorts; (2) explored the effects of anxiety and/or depression at baseline as predictors of IBS onset at a future point. In all, 11 studies were included of which eight recruited participants with a gastrointestinal infection. Meta-analyses were conducted. RESULTS: The risk of developing IBS was double for anxiety cases at baseline compared with those who were not [RR 2.38, 95% confidence interval (CI) 1.58-3.60]. Similar results were found for depression (RR 2.06, 95% CI 1.44-2.96). Anxiety and depression seemed to play a stronger role in IBS onset in individuals with a gastrointestinal infection although this could be attributed to other differences in methodology, such as use of diagnostic interviews rather than self-report. CONCLUSIONS: The findings suggest that self-reported anxiety and depression provide a twofold risk for IBS onset. There is less support for the role of anxiety or depressive disorder diagnosed using clinical interview. These findings may have implications for the development of interventions focused on IBS prevention and treatment.


Subject(s)
Anxiety Disorders/epidemiology , Anxiety/epidemiology , Depression/epidemiology , Depressive Disorder/epidemiology , Irritable Bowel Syndrome/epidemiology , Anxiety/psychology , Anxiety Disorders/psychology , Depression/psychology , Depressive Disorder/psychology , Humans , Irritable Bowel Syndrome/psychology , Risk , Risk Factors
4.
Oncogene ; 34(1): 15-26, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-24292678

ABSTRACT

The HSP90 molecular chaperone plays a key role in the maturation, stability and activation of its clients, including many oncogenic proteins. Kinases are a substantial and important subset of clients requiring the key cochaperone CDC37. We sought an improved understanding of protein kinase chaperoning by CDC37 in cancer cells. CDC37 overexpression in human colon cancer cells increased CDK4 protein levels, which was negated upon CDC37 knockdown. Overexpressing CDC37 increased CDK4 protein half-life and enhanced binding of HSP90 to CDK4, consistent with CDC37 promoting kinase loading onto chaperone complexes. Against expectation, expression of C-terminus-truncated CDC37 (ΔC-CDC37) that lacks HSP90 binding capacity did not affect kinase client expression or activity; moreover, as with wild-type CDC37 overexpression, it augmented CDK4-HSP90 complex formation. However, although truncation blocked binding to HSP90 in cells, ΔC-CDC37 also showed diminished client protein binding and was relatively unstable. CDC37 mutants with single and double point mutations at residues M164 and L205 showed greatly reduced binding to HSP90, but retained association with client kinases. Surprisingly, these mutants phenocopied wild-type CDC37 overexpression by increasing CDK4-HSP90 association and CDK4 protein levels in cells. Furthermore, expression of the mutants was sufficient to protect kinase clients CDK4, CDK6, CRAF and ERBB2 from depletion induced by silencing endogenous CDC37, indicating that CDC37's client stabilising function cannot be inactivated by substantially reducing its direct interaction with HSP90. However, CDC37 could not compensate for loss of HSP90 function, showing that CDC37 and HSP90 have their own distinct and non-redundant roles in maintaining kinase clients. Our data substantiate the important function of CDC37 in chaperoning protein kinases. Furthermore, we demonstrate that CDC37 can stabilise kinase clients by a mechanism that is not dependent on a substantial direct interaction between CDC37 and HSP90, but nevertheless requires HSP90 activity. These results have significant implications for therapeutic targeting of CDC37.


Subject(s)
Cell Cycle Proteins/metabolism , Chaperonins/metabolism , Colonic Neoplasms/metabolism , HSP90 Heat-Shock Proteins/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mutation , Point Mutation , Protein Binding , Proto-Oncogene Proteins c-raf/metabolism , RNA, Small Interfering/metabolism , Receptor, ErbB-2/metabolism
5.
Br J Cancer ; 109(6): 1562-9, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23942066

ABSTRACT

BACKGROUND: Non-invasive imaging biomarkers underpin the development of molecularly targeted anti-cancer drugs. This study evaluates tumour apparent diffusion coefficient (ADC), measured by diffusion-weighted magnetic resonance imaging (DW-MRI), as a biomarker of response to the MEK1/2 inhibitor selumetinib (AZD6244, ARRY-142886) in human tumour xenografts. METHODS: Nude mice bearing human BRAF(V600D) WM266.4 melanoma or BRAF(V600E) Colo205 colon carcinoma xenografts were treated for 4 days with vehicle or selumetinib. DW-MRI was performed before and 2 h after the last dose and excised tumours analysed for levels of phospho-ERK1/2, cleaved caspase 3 (CC3) and necrosis. RESULTS: Selumetinib treatment induced tumour stasis and reduced ERK1/2 phosphorylation in both WM266.4 and Colo205 tumour xenografts. Relative to day 0, mean tumour ADC was unchanged in the control groups but was significantly increased by up to 1.6-fold in selumetinib-treated WM266.4 and Colo205 tumours. Histological analysis revealed a significant increase in necrosis in selumetinib-treated WM266.4 and Colo205 xenografts and CC3 staining in selumetinib-treated Colo205 tumours relative to controls. CONCLUSION: Changes in ADC following treatment with the MEK1/2 inhibitor selumetinib in responsive human tumour xenografts were concomitant with induction of tumour cell death. ADC may provide a useful non-invasive pharmacodynamic biomarker for early clinical assessment of response to selumetinib and other MEK-ERK1/2 signalling-targeted therapies.


Subject(s)
Benzimidazoles/pharmacology , Colonic Neoplasms/drug therapy , Diffusion Magnetic Resonance Imaging/methods , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Melanoma/drug therapy , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line, Tumor , Colonic Neoplasms/enzymology , Female , Humans , Melanoma/enzymology , Mice , Mice, Nude , Molecular Targeted Therapy , Xenograft Model Antitumor Assays
6.
Clin Pharmacol Ther ; 93(3): 252-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23361103

ABSTRACT

The progressive elucidation of the molecular pathogenesis of cancer has fueled the rational development of targeted drugs for patient populations stratified by genetic characteristics. Here we discuss general challenges relating to molecular diagnostics and describe predictive biomarkers for personalized cancer medicine. We also highlight resistance mechanisms for epidermal growth factor receptor (EGFR) kinase inhibitors in lung cancer. We envisage a future requiring the use of longitudinal genome sequencing and other omics technologies alongside combinatorial treatment to overcome cellular and molecular heterogeneity and prevent resistance caused by clonal evolution.


Subject(s)
Biomarkers, Tumor/analysis , Drug Resistance, Neoplasm , Medical Oncology , Neoplasms/drug therapy , Pathology, Molecular , Precision Medicine , Humans , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors
7.
Leukemia ; 26(7): 1462-70, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22354205

ABSTRACT

Acquired resistance to selective FLT3 inhibitors is an emerging clinical problem in the treatment of FLT3-ITD(+) acute myeloid leukaemia (AML). The paucity of valid pre-clinical models has restricted investigations to determine the mechanism of acquired therapeutic resistance, thereby limiting the development of effective treatments. We generated selective FLT3 inhibitor-resistant cells by treating the FLT3-ITD(+) human AML cell line MOLM-13 in vitro with the FLT3-selective inhibitor MLN518, and validated the resistant phenotype in vivo and in vitro. The resistant cells, MOLM-13-RES, harboured a new D835Y tyrosine kinase domain (TKD) mutation on the FLT3-ITD(+) allele. Acquired TKD mutations, including D835Y, have recently been identified in FLT3-ITD(+) patients relapsing after treatment with the novel FLT3 inhibitor, AC220. Consistent with this clinical pattern of resistance, MOLM-13-RES cells displayed high relative resistance to AC220 and Sorafenib. Furthermore, treatment of MOLM-13-RES cells with AC220 lead to loss of the FLT3 wild-type allele and the duplication of the FLT3-ITD-D835Y allele. Our FLT3-Aurora kinase inhibitor, CCT137690, successfully inhibited growth of FLT3-ITD-D835Y cells in vitro and in vivo, suggesting that dual FLT3-Aurora inhibition may overcome selective FLT3 inhibitor resistance, in part due to inhibition of Aurora kinase, and may benefit patients with FLT3-mutated AML.


Subject(s)
Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Tandem Repeat Sequences/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Apoptosis/drug effects , Aurora Kinases , Benzenesulfonates/pharmacology , Benzothiazoles/pharmacology , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Female , Humans , Imidazoles/pharmacology , Mice , Mice, Nude , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Piperazines/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Quinazolines/pharmacology , Sorafenib , Tumor Cells, Cultured , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/metabolism
8.
Curr Med Chem ; 18(18): 2686-714, 2011.
Article in English | MEDLINE | ID: mdl-21649578

ABSTRACT

The phosphoinositide 3-kinases (PI3Ks) constitute an important family of lipid kinase enzymes that control a range of cellular processes through their regulation of a network of signal transduction pathways, and have emerged as important therapeutic targets in the context of cancer, inflammation and cardiovascular diseases. Since the mid-late 1990s, considerable progress has been made in the discovery and development of small molecule ATP-competitive PI3K inhibitors, a number of which have entered early phase human trials over recent years from which key clinical results are now being disclosed. This review summarizes progress made to date, primarily on the discovery and characterization of class I and dual class I/IV subtype inhibitors, together with advances that have been made in translational and clinical research, notably in cancer.


Subject(s)
Cardiovascular Diseases/drug therapy , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Clinical Medicine/methods , Inflammation/drug therapy , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Cardiovascular Diseases/enzymology , Class I Phosphatidylinositol 3-Kinases/metabolism , Clinical Medicine/trends , Humans , Inflammation/enzymology , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology
10.
Br J Cancer ; 102(11): 1555-77, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20502460

ABSTRACT

Animal experiments remain essential to understand the fundamental mechanisms underpinning malignancy and to discover improved methods to prevent, diagnose and treat cancer. Excellent standards of animal care are fully consistent with the conduct of high quality cancer research. Here we provide updated guidelines on the welfare and use of animals in cancer research. All experiments should incorporate the 3Rs: replacement, reduction and refinement. Focusing on animal welfare, we present recommendations on all aspects of cancer research, including: study design, statistics and pilot studies; choice of tumour models (e.g., genetically engineered, orthotopic and metastatic); therapy (including drugs and radiation); imaging (covering techniques, anaesthesia and restraint); humane endpoints (including tumour burden and site); and publication of best practice.


Subject(s)
Animal Experimentation/standards , Animal Welfare/standards , Neoplasms/pathology , Neoplasms/therapy , Practice Guidelines as Topic , Algorithms , Animal Experimentation/ethics , Animal Welfare/ethics , Animal Welfare/organization & administration , Animals , Biomarkers, Pharmacological/analysis , Biomedical Research/ethics , Biomedical Research/legislation & jurisprudence , Biomedical Research/organization & administration , Biomedical Research/standards , Cell Line, Transformed , Diagnostic Imaging , Disease Models, Animal , Female , Humans , Male , Mice , Neoplasm Transplantation/methods , Neoplasm Transplantation/pathology , Neoplasm Transplantation/standards , Neoplasms/diagnosis , Neoplasms/genetics , Treatment Outcome , Xenograft Model Antitumor Assays
11.
Br J Cancer ; 101(11): 1860-8, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19935799

ABSTRACT

BACKGROUND: SR4554 is a fluorine-containing 2-nitroimidazole, designed as a hypoxia marker detectable with 19F magnetic resonance spectroscopy (MRS). In an initial phase I study of SR4554, nausea/vomiting was found to be dose-limiting, and 1400 mg m(-2) was established as MTD. Preliminary MRS studies demonstrated some evidence of 19F retention in tumour. In this study we investigated higher doses of SR4554 and intratumoral localisation of the 19F MRS signal. METHODS: Patients had tumours > or = 3 cm in diameter and < or = 4 cm deep. Measurements were performed using 1H/19F surface coils and localised 19F MRS acquisition. SR4554 was administered at 1400 mg m(-2), with subsequent increase to 2600 mg m(-2) using prophylactic metoclopramide. Spectra were obtained immediately post infusion (MRS no. 1), at 16 h (MRS no. 2) and 20 h (MRS no. 3), based on the SR4554 half-life of 3.5 h determined from a previous study. 19Fluorine retention index (%) was defined as (MRS no. 2/MRS no. 1)*100. RESULTS: A total of 26 patients enrolled at: 1400 (n=16), 1800 (n=1), 2200 (n=1) and 2600 mg m(-2) (n=8). SR4554 was well tolerated and toxicities were all < or = grade 1; mean plasma elimination half-life was 3.7+/-0.9 h. SR4554 signal was seen on both unlocalised and localised MRS no. 1 in all patients. Localised 19F signals were detected at MRS no. 2 in 5 out of 9 patients and 4 out of 5 patients at MRS no. 3. The mean retention index in tumour was 13.6 (range 0.6-43.7) compared with 4.1 (range 0.6-7.3) for plasma samples taken at the same times (P=0.001) suggesting (19)F retention in tumour and, therefore, the presence of hypoxia. CONCLUSION: We have demonstrated the feasibility of using 19F MRS with SR4554 as a potential method of detecting hypoxia. Certain patients showed evidence of 19F retention in tumour, supporting further development of this technique for detection of tumour hypoxia.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Neoplasms/metabolism , Nitroimidazoles/pharmacokinetics , Adult , Aged , Aged, 80 and over , Cell Hypoxia/physiology , Female , Humans , Male , Middle Aged , Nitroimidazoles/adverse effects , Oxygen/metabolism , Partial Pressure , Young Adult
12.
Clin Pharmacol Ther ; 85(2): 131-3, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19151637

ABSTRACT

Compared with conventional chemotherapy, rationally designed molecularly targeted agents may be more likely to have antitumor activity in selected tumor subgroups driven by the oncogenic signals targeted by these compounds and a different side-effect profile. The use of biomarkers in the early clinical trials of these new anticancer agents has the potential to increase study participants' benefit from early clinical trials, accelerate the drug development process, maximize the ability to generate important biological information about human cancer, and decrease the risk of late and costly drug attrition.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Clinical Trials as Topic/trends , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Biomarkers, Tumor/metabolism , Clinical Trials as Topic/methods , Darkness , Humans , Light , Neoplasms/genetics , Time Factors
13.
Oncogene ; 28(2): 157-69, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18931700

ABSTRACT

The cochaperone CDC37 promotes the association of HSP90 with the protein kinase subset of client proteins to maintain their stability and signalling functions. HSP90 inhibitors induce depletion of clients, which include several oncogenic kinases. We hypothesized that the targeting of CDC37 using siRNAs would compromise the maturation of these clients and increase the sensitivity of cancer cells to HSP90 inhibitors. Here, we show that silencing of CDC37 in human colon cancer cells diminished the association of kinase clients with HSP90 and reduced levels of the clients ERBB2, CRAF, CDK4 and CDK6, as well as phosphorylated AKT. CDC37 silencing promoted the proteasome-mediated degradation of kinase clients, suggesting a degradation pathway independent from HSP90 binding. Decreased cell signalling through kinase clients was also demonstrated by reduced phosphorylation of downstream substrates and colon cancer cell proliferation was subsequently reduced by the inhibition of the G1/S-phase transition. Furthermore, combining CDC37 silencing with the HSP90 inhibitor 17-AAG induced more extensive and sustained depletion of kinase clients and potentiated cell cycle arrest and apoptosis. These results support an essential role for CDC37 in concert with HSP90 in maintaining oncogenic protein kinase clients and endorse the therapeutic potential of targeting CDC37 in cancer.


Subject(s)
Adenocarcinoma/pathology , Breast Neoplasms/pathology , Cell Cycle Proteins/physiology , Chaperonins/physiology , Colonic Neoplasms/pathology , HSP90 Heat-Shock Proteins/physiology , Neoplasm Proteins/physiology , Prostatic Neoplasms/pathology , Protein Kinases/metabolism , RNA Interference , RNA, Small Interfering/pharmacology , Adenocarcinoma/metabolism , Apoptosis/drug effects , Benzoquinones/pharmacology , Breast Neoplasms/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Division/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Chaperonins/antagonists & inhibitors , Chaperonins/genetics , Colonic Neoplasms/metabolism , Female , G1 Phase/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Targeting , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic/pharmacology , Male , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Prostatic Neoplasms/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational/drug effects , Pyrazoles/pharmacology , Signal Transduction/drug effects
14.
Oncogene ; 27(8): 1036-44, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-17700519

ABSTRACT

The PPM1D gene is aberrantly amplified in a range of common cancers and encodes a protein phosphatase that is a potential therapeutic target. However, the issue of whether inhibition of PPM1D in human tumour cells that overexpress this protein compromises their viability has not yet been fully addressed. We show here, using an RNA interference (RNAi) approach, that inhibition of PPM1D can indeed reduce the viability of human tumour cells and that this effect is selective; tumour cell lines that overexpress PPM1D are sensitive to PPM1D inhibition whereas cell lines with normal levels are not. Loss of viability associated with PPM1D RNAi in human tumour cells occurs via the activation of the kinase P38. To identify chemical inhibitors of PPM1D, a high-throughput screening of a library of small molecules was performed. This strategy successfully identified a compound that selectively reduces viability of human tumour cell lines that overexpress PPM1D. As expected of a specific inhibitor, the toxicity to PPM1D overexpressing cell lines after inhibitor treatment is P38 dependent. These results further validate PPM1D as a therapeutic target and identify a proof-of-principle small molecule inhibitor.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cyclopentanes/pharmacology , Growth Inhibitors/pharmacology , Phosphoprotein Phosphatases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Phosphoprotein Phosphatases/biosynthesis , Phosphoprotein Phosphatases/genetics , Protein Phosphatase 2C
15.
Br J Cancer ; 96(1): 29-37, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17179992

ABSTRACT

Seliciclib (CYC202; R-roscovitine) is the first selective, orally bioavailable inhibitor of cyclin-dependent kinases 1, 2, 7 and 9 to enter clinical trial. Preclinical studies showed antitumour activity in a broad range of human tumour xenografts. A phase I trial was performed with a 7-day b.i.d. p.o. schedule. Twenty-one patients (median age 62 years, range: 39-73 years) were treated with doses of 100, 200 and 800 b.i.d. Dose-limiting toxicities were seen at 800 mg b.i.d.; grade 3 fatigue, grade 3 skin rash, grade 3 hyponatraemia and grade 4 hypokalaemia. Other toxicities included reversible raised creatinine (grade 2), reversible grade 3 abnormal liver function and grade 2 emesis. An 800 mg portion was investigated further in 12 patients, three of whom had MAG3 renograms. One patient with a rapid increase in creatinine on day 3 had a reversible fall in renal perfusion, with full recovery by day 14, and no changes suggestive of renal tubular damage. Further dose escalation was precluded by hypokalaemia. Seliciclib reached peak plasma concentrations between 1 and 4 h and elimination half-life was 2-5 h. Inhibition of retinoblastoma protein phosphorylation was not demonstrated in peripheral blood mononuclear cells. No objective tumour responses were noted, but disease stabilisation was recorded in eight patients; this lasted for a total of six courses (18 weeks) in a patient with ovarian cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Enzyme Inhibitors/administration & dosage , Neoplasms/drug therapy , Purines/administration & dosage , Administration, Oral , Adult , Aged , Antineoplastic Agents/adverse effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Administration Schedule , Enzyme Inhibitors/adverse effects , Female , Humans , Hyperglycemia/chemically induced , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Purines/adverse effects , Roscovitine , Treatment Outcome
16.
Handb Exp Pharmacol ; (172): 331-58, 2006.
Article in English | MEDLINE | ID: mdl-16610366

ABSTRACT

Specific inhibitors of Hsp90 have recently entered human clinical trials. At the time of writing, trials have been initiated only in metastatic cancer, although a rationale exists for using these agents in a variety of human diseases where protein (mis)folding is involved in the disease pathophysiology. Hsp90 inhibitors offer a unique anti-cancer opportunity because they provide simultaneous combinatorial blockade of multiple oncogenic pathways. The first compound in this class, 17-AAG, has completed phase I trials and phase II trials are in progress. The toxicity has been manageable and evidence of possible clinical activity has been seen in metastatic melanoma, prostate cancer and multiple myeloma. Other inhibitors with improved properties are approaching clinical trials. This chapter presents an update of the current clinical trials using Hsp90 inhibitors, focussing on the areas that will be increasingly relevant in the next 5 years.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Benzoquinones , Clinical Trials as Topic , Drug Design , Humans , Lactams, Macrocyclic , Male , Melanoma/drug therapy , Multiple Myeloma/drug therapy , Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Quinones/therapeutic use , Rifabutin/analogs & derivatives , Rifabutin/therapeutic use
17.
Anal Biochem ; 350(2): 202-13, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16460658

ABSTRACT

Hsp90 encodes a ubiquitous molecular chaperone protein conserved among species which acts on multiple substrates, many of which are important cell-signaling proteins. Inhibition of Hsp90 function has been promoted as a mechanism to degrade client proteins involved in tumorigenesis and disease progression. Several assays to monitor inhibition of Hsp90 function currently exist but are limited in their use for a drug discovery campaign. Using data from the crystal structure of an initial hit compound, we have developed a fluorescence polarization assay to monitor binding of compounds to the ATP-binding site of Hsp90. This assay is very robust (Z' > 0.9) and can detect affinity of compounds with IC50s to 40 nM. We have used this assay in conjunction with cocrystal structures of small molecules to drive a structure-based design program aimed at the discovery and optimization of a novel class of potent Hsp90 inhibitors.


Subject(s)
Coumarins/chemistry , Coumarins/pharmacology , Fluorescence Polarization/methods , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Pyrazoles/chemistry , Pyrazoles/pharmacology , Adenosine Triphosphatases/antagonists & inhibitors , Binding Sites , Crystallography, X-Ray , Inhibitory Concentration 50 , Resorcinols/chemistry , Saccharomyces cerevisiae/enzymology , Structure-Activity Relationship
18.
Br J Cancer ; 93(9): 1011-8, 2005 Oct 31.
Article in English | MEDLINE | ID: mdl-16249792

ABSTRACT

In this study we investigated the in vitro time dependence of radiosensitisation, pharmacokinetics and metabolism of NU7026, a novel inhibitor of the DNA repair enzyme DNA-dependent protein kinase (DNA-PK). At a dose of 10 muM, which is nontoxic to cells per se, a minimum NU7026 exposure of 4 h in combination with 3 Gy radiation is required for a significant radiosensitisation effect in CH1 human ovarian cancer cells. Following intravenous administration to mice at 5 mg kg(-1), NU7026 underwent rapid plasma clearance (0.108 l h(-1)) and this was largely attributed to extensive metabolism. Bioavailability following interperitoneal (i.p.) and p.o. administration at 20 mg kg(-1) was 20 and 15%, respectively. Investigation of NU7026 metabolism profiles in plasma and urine indicated that the compound undergoes multiple hydroxylations. A glucuronide conjugate of a bis-hydroxylated metabolite represented the major excretion product in urine. Identification of the major oxidation site as C-2 of the morpholine ring was confirmed by the fact that the plasma clearance of NU7107 (an analogue of NU7026 methylated at C-2 and C-6 of the morpholine ring) was four-fold slower than that of NU7026. The pharmacokinetic simulations performed predict that NU7026 will have to be administered four times per day at 100 mg kg(-1) i.p. in order to obtain the drug exposure required for radiosensitisation.


Subject(s)
Chromones/metabolism , Chromones/pharmacokinetics , DNA-Activated Protein Kinase/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacokinetics , Morpholines/metabolism , Morpholines/pharmacokinetics , Ovarian Neoplasms/metabolism , Animals , Biological Availability , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Drug Evaluation, Preclinical , Female , Gamma Rays , Humans , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/radiotherapy , Radiation Tolerance , Tumor Stem Cell Assay
19.
Br J Cancer ; 93(8): 876-83, 2005 Oct 17.
Article in English | MEDLINE | ID: mdl-16222321

ABSTRACT

SU5416 (Z-3-[(2,4-dimethylpyrrol-5-yl)methylidenyl]-2-indolinone; semaxanib) is a small molecule inhibitor of the vascular endothelial growth factor receptor (VEGFR2). A Phase I dose escalation study was performed. Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was used as a pharmacodynamic assessment tool. In all, 27 patients were recruited. SU5416 was administered twice weekly by fixed rate intravenous infusion. Patients were treated in sequential cohorts of three patients at 48, 65, 85 110 and 145 mg m-2. A further dose level of 190 mg m-2 after a 2-week lead in period at a lower dose was completed; thereafter, the cohort at 145 mg m-2 was expanded. SU5416 showed linear pharmacokinetics to 145 mg m-2 with a large volume of distribution and rapid clearance. A significant degree of interpatient variability was seen. SU5416 was well tolerated, by definition a maximum-tolerated dose was not defined. No reproducible changes were seen in DCE-MRI end points. Serial assessments of VEGF in a cohort of patients treated at 145 mg m-2 did not show a statistically significant treatment-related change. Parallel assessments of the impact of SU5416 on coagulation profiles in six patients showed a transient effect within the fibrinolytic pathway. Clinical experience showed that patients who had breaks of therapy longer than a week could not have treatment reinitiated at a dose of 190 mg m-2 without unacceptable toxicity. The 145 mg m-2 dose level is thus the recommended dose for future study.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacokinetics , Indoles/adverse effects , Indoles/pharmacokinetics , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Adolescent , Adult , Aged , Angiogenesis Inhibitors/therapeutic use , Drug Administration Schedule , Endpoint Determination , Female , Humans , Indoles/therapeutic use , Infusions, Intravenous , Magnetic Resonance Imaging , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/drug therapy , Neoplasms/pathology , Pyrroles/therapeutic use
20.
Br J Cancer ; 92(9): 1599-610, 2005 May 09.
Article in English | MEDLINE | ID: mdl-15870830

ABSTRACT

Vascular and angiogenic processes provide an important target for novel cancer therapeutics. Dynamic contrast-enhanced magnetic resonance imaging is being used increasingly to noninvasively monitor the action of these therapeutics in early-stage clinical trials. This publication reports the outcome of a workshop that considered the methodology and design of magnetic resonance studies, recommending how this new tool might best be used.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Magnetic Resonance Imaging , Neoplasms/blood supply , Neoplasms/drug therapy , Clinical Trials as Topic , Evaluation Studies as Topic , Reproducibility of Results , Terminology as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...