Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Sci Rep ; 10(1): 3836, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32123273

ABSTRACT

There is mounting evidence of androgen receptor signaling inducing genome instability and changing DNA repair capacity in prostate cancer cells. Expression of genes associated with base excision repair (BER) is increased with prostate cancer progression and correlates with poor prognosis. Poly(ADP-ribose) polymerase (PARP) and poly(ADP-ribose) glycohydrolase (PARG) are key enzymes in BER that elongate and degrade PAR polymers on target proteins. While PARP inhibitors have been tested in clinical trials and are a promising therapy for prostate cancer patients with TMPRSS2-ERG fusions and mutations in DNA repair genes, PARG inhibitors have not been evaluated. We show that PARG is a direct androgen receptor (AR) target gene. AR is recruited to the PARG locus and induces PARG expression. Androgen ablation combined with PARG inhibition synergistically reduces BER capacity in independently derived LNCaP and LAPC4 prostate cancer cell lines. A combination of PARG inhibition with androgen ablation or with the DNA damaging drug, temozolomide, significantly reduces cellular proliferation and increases DNA damage. PARG inhibition alters AR transcriptional output without changing AR protein levels. Thus, AR and PARG are engaged in reciprocal regulation suggesting that the success of androgen ablation therapy can be enhanced by PARG inhibition in prostate cancer patients.


Subject(s)
Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Repair/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Glycoside Hydrolases/metabolism , Humans , Male , Molecular Targeted Therapy
3.
Cancer Cell ; 35(3): 519-533.e8, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30889383

ABSTRACT

Inhibitors of poly(ADP-ribose) polymerase (PARP) have demonstrated efficacy in women with BRCA-mutant ovarian cancer. However, only 15%-20% of ovarian cancers harbor BRCA mutations, therefore additional therapies are required. Here, we show that a subset of ovarian cancer cell lines and ex vivo models derived from patient biopsies are sensitive to a poly(ADP-ribose) glycohydrolase (PARG) inhibitor. Sensitivity is due to underlying DNA replication vulnerabilities that cause persistent fork stalling and replication catastrophe. PARG inhibition is synthetic lethal with inhibition of DNA replication factors, allowing additional models to be sensitized by CHK1 inhibitors. Because PARG and PARP inhibitor sensitivity are mutually exclusive, our observations demonstrate that PARG inhibitors have therapeutic potential to complement PARP inhibitor strategies in the treatment of ovarian cancer.


Subject(s)
DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , Ovarian Neoplasms/genetics , Cell Line, Tumor , Checkpoint Kinase 1 , Female , Glycoside Hydrolases/antagonists & inhibitors , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Quinazolinones/pharmacology
4.
Bioorg Med Chem Lett ; 29(4): 560-562, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30616904

ABSTRACT

Fluorination of metabolic hotspots in a molecule is a common medicinal chemistry strategy to improve in vivo half-life and exposure and, generally, this strategy offers significant benefits. Here, we report the application of this strategy to a series of poly-ADP ribose glycohydrolase (PARG) inhibitors, resulting in unexpected in vivo toxicity which was attributed to this single-atom modification.


Subject(s)
Cyclopropanes/pharmacology , Glycoside Hydrolases/toxicity , Microsomes, Liver/drug effects , Administration, Oral , Animals , Cyclopropanes/administration & dosage , Cyclopropanes/chemistry , Cyclopropanes/pharmacokinetics , Glycoside Hydrolases/administration & dosage , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/pharmacokinetics , Half-Life , Humans , Mice , Microsomes, Liver/metabolism
5.
J Med Chem ; 61(23): 10767-10792, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30403352

ABSTRACT

DNA damage repair enzymes are promising targets in the development of new therapeutic agents for a wide range of cancers and potentially other diseases. The enzyme poly(ADP-ribose) glycohydrolase (PARG) plays a pivotal role in the regulation of DNA repair mechanisms; however, the lack of potent drug-like inhibitors for use in cellular and in vivo models has limited the investigation of its potential as a novel therapeutic target. Using the crystal structure of human PARG in complex with the weakly active and cytotoxic anthraquinone 8a, novel quinazolinedione sulfonamides PARG inhibitors have been identified by means of structure-based virtual screening and library design. 1-Oxetan-3-ylmethyl derivatives 33d and 35d were selected for preliminary investigations in vivo. X-ray crystal structures help rationalize the observed structure-activity relationships of these novel inhibitors.


Subject(s)
DNA Repair , Drug Design , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Quinazolinones/chemistry , Quinazolinones/pharmacology , Administration, Oral , Animals , Biological Availability , Catalytic Domain , Glycoside Hydrolase Inhibitors/administration & dosage , Glycoside Hydrolase Inhibitors/pharmacokinetics , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/metabolism , HeLa Cells , Humans , Male , Mice , Models, Molecular , Quinazolinones/administration & dosage , Quinazolinones/pharmacokinetics , Structure-Activity Relationship
6.
Cancer Cell ; 33(6): 1078-1093.e12, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894693

ABSTRACT

Inhibitors of poly(ADP-ribose) (PAR) polymerase (PARPi) have recently entered the clinic for the treatment of homologous recombination (HR)-deficient cancers. Despite the success of this approach, drug resistance is a clinical hurdle, and we poorly understand how cancer cells escape the deadly effects of PARPi without restoring the HR pathway. By combining genetic screens with multi-omics analysis of matched PARPi-sensitive and -resistant Brca2-mutated mouse mammary tumors, we identified loss of PAR glycohydrolase (PARG) as a major resistance mechanism. We also found the presence of PARG-negative clones in a subset of human serous ovarian and triple-negative breast cancers. PARG depletion restores PAR formation and partially rescues PARP1 signaling. Importantly, PARG inactivation exposes vulnerabilities that can be exploited therapeutically.


Subject(s)
Glycoside Hydrolases/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Synthetic Lethal Mutations , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/metabolism , Homologous Recombination/drug effects , Homologous Recombination/genetics , Humans , Mice, 129 Strain , Mice, Knockout , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly ADP Ribosylation/drug effects
7.
Bioorg Med Chem Lett ; 27(20): 4755-4759, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28927796

ABSTRACT

As part of our ongoing efforts to develop reversible inhibitors of LSD1, we identified a series of 4-(pyrrolidin-3-yl)benzonitrile derivatives that act as successful scaffold-hops of the literature inhibitor GSK-690. The most active compound, 21g, demonstrated a Kd value of 22nM and a biochemical IC50 of 57nM. In addition, this compound displayed improved selectivity over the hERG ion channel compared to GSK-690, and no activity against the related enzymes MAO-A and B. In human THP-1 acute myeloid leukaemia cells, 21g was found to increase the expression of the surrogate cellular biomarker CD86. This work further demonstrates the versatility of scaffold-hopping asa method to develop structurally diverse, potent inhibitors of LSD1.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Histone Demethylases/antagonists & inhibitors , Nitriles/chemistry , Nitriles/pharmacology , Binding Sites , Cell Line, Tumor , Drug Design , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Histone Demethylases/metabolism , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Nitriles/chemical synthesis , Protein Structure, Tertiary , Pyrrolidines/chemistry , Stereoisomerism , Structure-Activity Relationship
8.
F1000Res ; 5: 962, 2016.
Article in English | MEDLINE | ID: mdl-27703665

ABSTRACT

We present IncucyteDRC, an R package for the analysis of data from live cell imaging cell proliferation experiments carried out on the Essen Biosciences IncuCyte ZOOM instrument. The package provides a simple workflow for summarising data into a form that can be used to calculate dose response curves and EC50 values for small molecule inhibitors. Data from different cell lines, or cell lines grown under different conditions, can be normalised as to their doubling time. A simple graphical web interface, implemented using shiny, is provided for the benefit of non-R users. The software is potentially useful to any research group studying the impact of small molecule inhibitors on cell proliferation using the IncuCyte ZOOM.

9.
ACS Chem Biol ; 11(11): 3179-3190, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27689388

ABSTRACT

The enzyme poly(ADP-ribose) glycohydrolase (PARG) performs a critical role in the repair of DNA single strand breaks (SSBs). However, a detailed understanding of its mechanism of action has been hampered by a lack of credible, cell-active chemical probes. Herein, we demonstrate inhibition of PARG with a small molecule, leading to poly(ADP-ribose) (PAR) chain persistence in intact cells. Moreover, we describe two advanced, and chemically distinct, cell-active tool compounds with convincing on-target pharmacology and selectivity. Using one of these tool compounds, we demonstrate pharmacology consistent with PARG inhibition. Further, while the roles of PARG and poly(ADP-ribose) polymerase (PARP) are closely intertwined, we demonstrate that the pharmacology of a PARG inhibitor differs from that observed with the more thoroughly studied PARP inhibitor olaparib. We believe that these tools will facilitate a wider understanding of this important component of DNA repair and may enable the development of novel therapeutic agents exploiting the critical dependence of tumors on the DNA damage response (DDR).


Subject(s)
DNA Repair , Glycoside Hydrolases/chemistry , Molecular Probes/chemistry , Phthalazines/pharmacology , Piperazines/pharmacology , Enzyme Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , HeLa Cells , Humans , Surface Plasmon Resonance
10.
F1000Res ; 5: 736, 2016.
Article in English | MEDLINE | ID: mdl-27610220

ABSTRACT

After a DNA damage signal multiple polymers of ADP ribose attached to poly(ADP) ribose (PAR) polymerases (PARPs) are broken down by the enzyme poly(ADP) ribose glycohydrolase (PARG). Inhibition of PARG leads to a failure of DNA repair and small molecule inhibition of PARG has been a goal for many years. To determine whether biochemical inhibitors of PARG are active in cells we have designed an immunofluorescence assay to detect nuclear PAR after DNA damage. This 384-well assay is suitable for medium throughput high-content screening and can detect cell-permeable inhibitors of PARG from nM to µM potency. In addition, the assay has been shown to work in murine cells and in a variety of human cancer cells. Furthermore, the assay is suitable for detecting the DNA damage response induced by treatment with temozolomide and methylmethane sulfonate (MMS). Lastly, the assay has been shown to be robust over a period of several years.

11.
F1000Res ; 5: 1005, 2016.
Article in English | MEDLINE | ID: mdl-27429741

ABSTRACT

RET (REarranged during Transfection) is a receptor tyrosine kinase, which plays pivotal roles in regulating cell survival, differentiation, proliferation, migration and chemotaxis. Activation of RET is a mechanism of oncogenesis in medullary thyroid carcinomas where both germline and sporadic activating somatic mutations are prevalent. At present, there are no known specific RET inhibitors in clinical development, although many potent inhibitors of RET have been opportunistically identified through selectivity profiling of compounds initially designed to target other tyrosine kinases. Vandetanib and cabozantinib, both multi-kinase inhibitors with RET activity, are approved for use in medullary thyroid carcinoma, but additional pharmacological activities, most notably inhibition of vascular endothelial growth factor - VEGFR2 (KDR), lead to dose-limiting toxicity. The recent identification of RET fusions present in ~1% of lung adenocarcinoma patients has renewed interest in the identification and development of more selective RET inhibitors lacking the toxicities associated with the current treatments. In an earlier publication [Newton et al, 2016; 1] we reported the discovery of a series of 2-substituted phenol quinazolines as potent and selective RET kinase inhibitors. Here we describe the development of the robust screening cascade which allowed the identification and advancement of this chemical series.  Furthermore we have profiled a panel of RET-active clinical compounds both to validate the cascade and to confirm that none display a RET-selective target profile.

12.
Anal Biochem ; 503: 58-64, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27036617

ABSTRACT

Poly(ADP-ribose) (PAR) polymers are transient post-translational modifications, and their formation is catalyzed by poly(ADP-ribose) polymerase (PARP) enzymes. A number of PARP inhibitors are in advanced clinical development for BRCA-mutated breast cancer, and olaparib has recently been approved for BRCA-mutant ovarian cancer; however, there has already been evidence of developed resistance mechanisms. Poly(ADP-ribose) glycohydrolase (PARG) catalyzes the hydrolysis of the endo- and exo-glycosidic bonds within the PAR polymers. As an alternative strategy, PARG is a potentially attractive therapeutic target. There is only one PARG gene, compared with 17 known PARP family members, and therefore a PARG inhibitor may have wider application with fewer compensatory mechanisms. Prior to the initiation of this project, there were no known existing cell-permeable small molecule PARG inhibitors for use as tool compounds to assess these hypotheses and no suitable high-throughput screening (HTS)-compatible biochemical assays available to identify start points for a drug discovery project. The development of this newly described high-throughput homogeneous time-resolved fluorescence (HTRF) assay has allowed HTS to proceed and, from this, the identification and advancement of multiple validated series of tool compounds for PARG inhibition.


Subject(s)
Fluorescence , Glycoside Hydrolases/metabolism , High-Throughput Screening Assays/methods , Luminescent Measurements/methods , Cell Line , Enzyme Inhibitors/pharmacology , Glycoside Hydrolases/analysis , Glycoside Hydrolases/antagonists & inhibitors , Humans , Structure-Activity Relationship , Time Factors
13.
Biochem J ; 473(13): 1869-79, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27099339

ABSTRACT

Tyrosyl-DNA phosphodiesterase 2 (TDP2) is a 5'-tyrosyl DNA phosphodiesterase important for the repair of DNA adducts generated by non-productive (abortive) activity of topoisomerase II (TOP2). TDP2 facilitates therapeutic resistance to topoisomerase poisons, which are widely used in the treatment of a range of cancer types. Consequently, TDP2 is an interesting target for the development of small molecule inhibitors that could restore sensitivity to topoisomerase-directed therapies. Previous studies identified a class of deazaflavin-based molecules that showed inhibitory activity against TDP2 at therapeutically useful concentrations, but their mode of action was uncertain. We have confirmed that the deazaflavin series inhibits TDP2 enzyme activity in a fluorescence-based assay, suitable for high-throughput screen (HTS)-screening. We have gone on to determine crystal structures of these compounds bound to a 'humanized' form of murine TDP2. The structures reveal their novel mode of action as competitive ligands for the binding site of an incoming DNA substrate, and point the way to generating novel and potent inhibitors of TDP2.


Subject(s)
Phosphoric Diester Hydrolases/metabolism , Riboflavin/chemistry , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Activation/drug effects , Humans , Mice , Phosphoric Diester Hydrolases/chemistry , Protein Binding , Protein Denaturation , Protein Structure, Secondary , Protein Structure, Tertiary , Riboflavin/analogs & derivatives , Riboflavin/pharmacology , Temperature
14.
Bioorg Med Chem Lett ; 26(11): 2724-9, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27086121

ABSTRACT

We have previously reported a series of anilinoquinazoline derivatives as potent and selective biochemical inhibitors of the RET kinase domain. However, these derivatives displayed diminished cellular potency. Herein we describe further optimisation of the series through modification of their physicochemical properties, delivering improvements in cell potency. However, whilst cellular selectivity against key targets could be maintained, combining cell potency and acceptable pharmacokinetics proved challenging.


Subject(s)
Aniline Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Quinazolines/pharmacology , Aniline Compounds/chemical synthesis , Aniline Compounds/chemistry , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-ret/metabolism , Quinazolines/chemical synthesis , Quinazolines/chemistry , Structure-Activity Relationship
15.
Eur J Med Chem ; 112: 20-32, 2016 Apr 13.
Article in English | MEDLINE | ID: mdl-26874741

ABSTRACT

Deregulation of the receptor tyrosine kinase RET has been implicated in medullary thyroid cancer, a small percentage of lung adenocarcinomas, endocrine-resistant breast cancer and pancreatic cancer. There are several clinically approved multi-kinase inhibitors that target RET as a secondary pharmacology but additional activities, most notably inhibition of KDR, lead to dose-limiting toxicities. There is, therefore, a clinical need for more specific RET kinase inhibitors. Herein we report our efforts towards identifying a potent and selective RET inhibitor using vandetanib 1 as the starting point for structure-based drug design. Phenolic anilinoquinazolines exemplified by 6 showed improved affinities towards RET but, unsurprisingly, suffered from high metabolic clearance. Efforts to mitigate the metabolic liability of the phenol led to the discovery that a flanking substituent not only improved the hepatocyte stability, but could also impart a significant gain in selectivity. This culminated in the identification of 36; a potent RET inhibitor with much improved selectivity against KDR.


Subject(s)
Piperidines/chemistry , Piperidines/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Quinazolines/chemistry , Quinazolines/pharmacology , Animals , Cell Line , Drug Design , Humans , Mice , Molecular Docking Simulation , Piperidines/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-ret/metabolism , Quinazolines/pharmacokinetics
16.
Drug Discov Today ; 20(5): 525-35, 2015 May.
Article in English | MEDLINE | ID: mdl-25542353

ABSTRACT

The contraction in research within pharma has seen a renaissance in drug discovery within the academic setting. Often, groups grow organically from academic research laboratories, exploiting a particular area of novel biology or new technology. However, increasingly, new groups driven by industrial staff are emerging with demonstrable expertise in the delivery of medicines. As part of a strategic review by Cancer Research UK (CR-UK), the drug discovery team at the Manchester Institute was established to translate novel research from the Manchester cancer research community into drug discovery programmes. From a standing start, we have taken innovative approaches to solve key issues faced by similar groups, such as hit finding and target identification. Herein, we share our lessons learnt and successful strategies.


Subject(s)
Academies and Institutes , Antineoplastic Agents/therapeutic use , Drug Discovery/methods , Neoplasms/drug therapy , Translational Research, Biomedical/methods , Academies and Institutes/organization & administration , Antineoplastic Agents/chemistry , Cooperative Behavior , Drug Discovery/organization & administration , England , Humans , Models, Organizational , Molecular Targeted Therapy , Neoplasms/metabolism , Neoplasms/pathology , Program Development , Research Personnel/organization & administration , Signal Transduction/drug effects , Time Factors , Translational Research, Biomedical/organization & administration , Workflow
18.
J Med Chem ; 56(16): 6352-70, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23859074

ABSTRACT

The recently discovered enzyme tyrosyl-DNA phosphodiesterase 2 (TDP2) has been implicated in the topoisomerase-mediated repair of DNA damage. In the clinical setting, it has been hypothesized that TDP2 may mediate drug resistance to topoisomerase II (topo II) inhibition by etoposide. Therefore, selective pharmacological inhibition of TDP2 is proposed as a novel approach to overcome intrinsic or acquired resistance to topo II-targeted drug therapy. Following a high-throughput screening (HTS) campaign, toxoflavins and deazaflavins were identified as the first reported sub-micromolar and selective inhibitors of this enzyme. Toxoflavin derivatives appeared to exhibit a clear structure-activity relationship (SAR) for TDP2 enzymatic inhibition. However, we observed a key redox liability of this series, and this, alongside early in vitro drug metabolism and pharmacokinetics (DMPK) issues, precluded further exploration. The deazaflavins were developed from a singleton HTS hit. This series showed distinct SAR and did not display redox activity; however low cell permeability proved to be a challenge.


Subject(s)
Phosphoric Diester Hydrolases/drug effects , Pyrimidinones/pharmacology , Topoisomerase II Inhibitors/pharmacology , Triazines/pharmacology , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemistry
19.
Anal Biochem ; 440(1): 1-5, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23688965

ABSTRACT

Tyrosyl-DNA phosphodiesterase 1 (Tdp1) catalyzes the hydrolysis of phosphodiester bonds between the DNA 3'-phosphate and tyrosine residues and plays a major role in the repair of stalled topoisomerase I-DNA covalent complexes. Given this role, Tdp1 is of interest as a potential target for anticancer therapy. Inhibiting Tdp1 in combination with clinically used Top1 inhibitors may potentiate the effects of the latter and help to overcome some of the chemoresistance issues currently observed. In addition, Tdp1 can function during DNA repair to remove a variety of other 3' adducts from DNA such as phosphoglycolates and abasic or apurinic/apyrimidinic (AP) sites. Here we describe a new mix-and-read homogeneous fluorogenic assay for the measurement of the AP-site cleavage activity of Tdp1 that is compatible with high-throughput screening. The application of such an assay will open up further avenues for the discovery of novel Tdp1 inhibitors.


Subject(s)
DNA Cleavage , DNA Repair , Enzyme Assays/methods , Fluorescence , High-Throughput Screening Assays/methods , Phosphoric Diester Hydrolases/chemistry , Humans , Purines/chemistry , Pyrimidines/chemistry
20.
Cancer Chemother Pharmacol ; 71(4): 1021-32, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23355042

ABSTRACT

PURPOSE: Cediranib (AZD2171) is a highly potent inhibitor of all three vascular endothelial growth factor receptors. The aim of this preclinical study was to examine the effect of combining cediranib with mechanistically distinct anti-tumour therapies. METHODS: Cediranib (1.5 or 3 mg/kg/day) was evaluated alone and in combination with either gefitinib, imatinib, ZD6126, saracatinib, selumetinib, bevacizumab, 5-fluorouracil (5-FU), docetaxel, oxaliplatin, gemcitabine, pemetrexed, irinotecan or cisplatin in human tumour xenograft models. Anti-tumour activity was measured by assessing the change in tumour volume following treatment compared with vehicle-treated time-matched controls. RESULTS: In all cases, the combination regimens, at tolerated doses and schedules, inhibited tumour growth to a greater extent than the corresponding monotherapy treatments. Compared with cediranib alone, statistically significant enhancements in anti-tumour activity were observed with all combination regimens. Notably, after 14 days of treatment, the combination of cediranib with ZD6126 induced substantial tumour regression (60 % compared with pre-treatment volume), whilst treatment with each agent alone led only to partial growth inhibition. A combination of cediranib with gefitinib also induced tumour regressions, and cediranib combined with either gemcitabine or irinotecan was found to inhibit tumour growth profoundly (by 99 and 98 %, respectively). CONCLUSIONS: Combining cediranib with selected cytotoxic or targeted agents proved efficacious in a range of human tumour xenograft models.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neovascularization, Pathologic/drug therapy , Quinazolines/therapeutic use , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Humans , Mice , Quinazolines/administration & dosage , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...