John R. Foster
AstraZeneca
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by John R. Foster.
Clinical Cancer Research | 2007
R. Wilkinson; Rajesh Odedra; Simon P. Heaton; Stephen R. Wedge; Nicholas Keen; Claire Crafter; John R. Foster; Madeleine C. Brady; Alison L. Bigley; Elaine Brown; Kate Byth; Nigel Charles Barrass; Kirsten E. Mundt; Kevin Michael Foote; Nicola Murdoch Heron; Frederic Henri Jung; Andrew Austen Mortlock; F. Thomas Boyle; Stephen J. Green
Purpose: In the current study, we examined the in vivo effects of AZD1152, a novel and specific inhibitor of Aurora kinase activity (with selectivity for Aurora B). Experimental Design: The pharmacodynamic effects and efficacy of AZD1152 were determined in a panel of human tumor xenograft models. AZD1152 was dosed via several parenteral (s.c. osmotic mini-pump, i.p., and i.v.) routes. Results: AZD1152 potently inhibited the growth of human colon, lung, and hematologic tumor xenografts (mean tumor growth inhibition range, 55% to ≥100%; P < 0.05) in immunodeficient mice. Detailed pharmacodynamic analysis in colorectal SW620 tumor-bearing athymic rats treated i.v. with AZD1152 revealed a temporal sequence of phenotypic events in tumors: transient suppression of histone H3 phosphorylation followed by accumulation of 4N DNA in cells (2.4-fold higher compared with controls) and then an increased proportion of polyploid cells (>4N DNA, 2.3-fold higher compared with controls). Histologic analysis showed aberrant cell division that was concurrent with an increase in apoptosis in AZD1152-treated tumors. Bone marrow analyses revealed transient myelosuppression with the drug that was fully reversible following cessation of AZD1152 treatment. Conclusions: These data suggest that selective targeting of Aurora B kinase may be a promising therapeutic approach for the treatment of a range of malignancies. In addition to the suppression of histone H3 phosphorylation, determination of tumor cell polyploidy and apoptosis may be useful biomarkers for this class of therapeutic agent. AZD1152 is currently in phase I trials.
Toxicologic Pathology | 2012
Anthony P. Hall; C. R. Elcombe; John R. Foster; Takanori Harada; Wolfgang Kaufmann; A. Knippel; K. Küttler; David E. Malarkey; R. R. Maronpot; A. Nishikawa; Thomas Nolte; A. Schulte; V. Strauss; Malcolm York
Preclinical toxicity studies have demonstrated that exposure of laboratory animals to liver enzyme inducers during preclinical safety assessment results in a signature of toxicological changes characterized by an increase in liver weight, hepatocellular hypertrophy, cell proliferation, and, frequently in long-term (life-time) studies, hepatocarcinogenesis. Recent advances over the last decade have revealed that for many xenobiotics, these changes may be induced through a common mechanism of action involving activation of the nuclear hormone receptors CAR, PXR, or PPARα. The generation of genetically engineered mice that express altered versions of these nuclear hormone receptors, together with other avenues of investigation, have now demonstrated that sensitivity to many of these effects is rodent-specific. These data are consistent with the available epidemiological and empirical human evidence and lend support to the scientific opinion that these changes have little relevance to man. The ESTP therefore convened an international panel of experts to debate the evidence in order to more clearly define for toxicologic pathologists what is considered adverse in the context of hepatocellular hypertrophy. The results of this workshop concluded that hepatomegaly as a consequence of hepatocellular hypertrophy without histologic or clinical pathology alterations indicative of liver toxicity was considered an adaptive and a non-adverse reaction. This conclusion should normally be reached by an integrative weight of evidence approach.
Biochemical Pharmacology | 1986
John R. Foster; Clifford R. Elcombe; Alan R. Boobis; Donald S. Davies; Dorothea Sesardic; Jamie McQuade; Richard T. Robson; Christopher F. Hayward; Edward A. Lock
The cellular distribution of cytochrome P-450 has been studied in the liver and a number of extrahepatic tissues in the rat by immunocytochemistry, using an antibody raised against cytochrome P-450 form c. Immunoreactive cytochrome P-450, most probably form c, was found in the proximal tubules of the kidney, in the Clara cells of the lung, and in the olfactory epithelium and Bowmans glands of the olfactory tissue, in addition to its location in the liver. Immunoreactive cytochrome P-450 was not found in the small intestine, the testes or the adrenal gland, although these organs are known to contain isoenzymes of cytochrome P-450. The use of antibody titration enabled the effects of phenobarbitone, beta-naphthoflavone and clofibrate on the content and distribution of immunoreactive cytochrome P-450 to be studied in both the liver and in the other organs discussed. Phenobarbitone induces epitope-specific cytochrome P-450 in the centrilobular cells of the liver but has no effect in any of the other tissues studied. Clofibrate is without effect on the levels of immunoreactive cytochrome P-450 in any of the tissues studied. In contrast, beta-naphthoflavone induces immunoreactive cytochrome P-450 in the periportal region of the liver, and also in the Clara cells of the lung, in the enterocytes of the small intestine and in the proximal tubules of the kidney. Of all of the tissues studied, in which immunoreactive cytochrome P-450 could be detected, only the olfactory epithelium failed to undergo enzyme induction following treatment with beta-naphthoflavone.
Toxicology Letters | 1982
Tim J.B. Gray; J.A. Beamand; Brian G. Lake; John R. Foster; Sharat D. Gangolli
Adult rat hepatocytes cultured for 48 h in the presence of 0.2 mM clofibrate, mono-(2-ethylhexyl)-phthalate (MEHP) or 2-ethylhexanol (2-EHA) contained increased numbers of peroxisomes. In keeping with the effects of these compounds in vivo, the peroxisome proliferation was associated with marked increases (up to 15-fold) in the activity of carnitine acetyltransferase. No such effects were produced by n-hexanol or two microsomal enzyme inducers, phenobarbital and 1,2-benzanthracene. These results suggest that cultured hepatocytes may provide a useful model system for studying chemically induced peroxisome proliferation.
Toxicology and Applied Pharmacology | 1982
Paul M. D. Foster; John R. Foster; Melvyn W. Cook; Lucy V. Thomas; Sharat D. Gangolli
Di-n-pentyl phthalate has previously been shown in our laboratory to produce testicular atrophy in young male rats when administered orally at 2.2 g/kg body wt/day for 4 days. Development of testicular damage by this compound was also associated with an increased urinary excretion of zinc and decreased testicular content of this element. A study was made of ultrastructural changes and the histochemical localization of zinc in the testis of normal rats and at different times after administration of phthalate until testicular atrophy was evident. The earliest morphological changes to the testis in the treated group occurred 6 hr after a single administration when severe vacuolation of Sertoli cell cytoplasm was apparent. Degradative changes to mitochondria were observed in Sertoli cells, spermatocytes, and spermatids 24 hr following a single phthalate dose. Histochemical localization of zinc in normal testis was most clearly seen in spermatids, where it appeared to be bound to the membranes of the Golgi complex and smooth endoplasmic reticulum. The first changes in zinc distribution were also observed at 6 hr after a single phthalate dose. Loss of zinc in the Golgi region of spermatids was clearly seen prior to any evidence of morphological damage in these cells. At 24 hr a decrease in electron-dense deposits bound to the smooth endoplasmic reticulum of spermatids was also observed. As time of exposure to phthalate increased, larger number of osmiophilic (lipid) bodies were evident in the Sertoli and Leydig cells.
Toxicology | 2012
Clifford R. Elcombe; Barbara M. Elcombe; John R. Foster; Shu-Ching Chang; David J. Ehresman; John L. Butenhoff
The present study investigated the potential role for activation of PPARα and CAR/PXR by potassium PFOS (K⁺ PFOS) with respect to the etiology of hepatic hypertrophy and hepatocellular adenoma in rats. Male Sprague-Dawley rats were fed K⁺ PFOS (20 or 100 ppm) for either 1, 7, or 28 days. Wyeth 14,643 (Wy 14,643, 50 ppm) and phenobarbital (PB, 500 ppm) were the controls for PPARα and CAR/PXR activation, respectively. Measurements included: plasma ALT, AST, cholesterol, triglycerides, and glucose; liver protein and DNA content; liver activities of palmitoyl CoA oxidase (ACOX), Cyp4A, CYP2B, and CYP3A; induction of liver CYP4A1, CYP2E1, CYP2B1/2, and CYP3A1 proteins (SDS-PAGE and Western blots); liver and thyroid microscopic histopathology, apoptotic index, and cell proliferation index. Terminal body weight was decreased by K⁺ PFOS (100 ppm) and Wy 14,643. All test-compound treatments increased liver weight. Plasma lipids were decreased by both PFOS and Wy 14,643. After treatment for 1 day, K⁺ PFOS (100 ppm), PB, and Wy 14,643 increased mean hepatic DNA concentration and total hepatic DNA, and total DNA remained elevated after treatment for 7 days and 28 days (PB and Wy 14,643 only). Hepatic P450 concentration was elevated after 7 and 28 days by K⁺ PFOS and by PB. K⁺ PFOS and Wy 14,643 increased liver activities of ACOX and CYP4A as well as increased liver CYP4A1 protein. By 28 days of treatment, K⁺ PFOS and PB increased liver activities of CYP2B and CYP3A as well as increased liver CYP2B1/2 and CYP3A1 proteins, and Wy 14,643 increased CYP2B enzyme activity to a slight extent. All test compounds increased the liver cell proliferative index and decreased the liver apoptotic index. No histological changes of the thyroid were noted; however, PB and WY increased thyroid follicular cell proliferation index (seven-day treatment only), while K⁺ PFOS did not. The thyroid follicular cell apoptotic index did not differ between groups. The hepatomegaly and hepatocellular adenoma observed after dietary exposure of Sprague-Dawley rats to K⁺ PFOS likely are due to the increased expression of xenosensor nuclear receptors PPARα and CAR/PXR. Given the markedly lower or absent response of human hepatocytes to the proliferative stimulus from activation of PPARα and CAR/PXR, the hepatocellular proliferative response from activation of these receptors by PFOS observed in rats is not expected to be of human relevance.
Science Translational Medicine | 2016
Susan Ashton; Young Ho Song; Jim Nolan; Elaine Cadogan; Jim Murray; Rajesh Odedra; John R. Foster; Peter A. Hall; Susan Low; Paula Taylor; Rebecca Ellston; Urszula M. Polanska; Joanne Wilson; Colin Howes; Aaron Smith; Richard J. A. Goodwin; John G. Swales; Nicole Strittmatter; Zoltan Takats; Anna Nilsson; Per E. Andrén; Dawn Trueman; Mike Walker; Corinne Reimer; Greg Troiano; Donald Parsons; David De Witt; Marianne Ashford; Jeff Hrkach; Stephen E. Zale
A nanoparticle formulation of an Aurora B kinase inhibitor uses ion pairing to achieve controlled release and efficacious, nontoxic target inhibition in tumors. Accurin nanoparticles dutifully deliver drug A class of drugs, called kinase inhibitors, could stop cancer in its tracks…if only these drugs could reach the tumors, stay for a while, and not be toxic. Hypothesizing that a nanoparticle formulation would solve the inhibitors’ woes, Ashton and colleagues investigated several different compositions of so-called Accurins—polymeric particles that encapsulate charged drugs through ion pairing. An Aurora B kinase, once formulated in Accurins, demonstrated a much-improved therapeutic index and preclinical efficacy compared with its parent molecule, when administered to rats and mice bearing human tumors. The Accurins allowed for sustained release of the drug over days, and did not have the same blood toxicity seen with the parent drug. A phase 1 trial is the next step for this nanomedicine, and additional preclinical studies will reveal whether such nanoformulations can improve the tolerability and efficacy of the broader class of molecularly targeted cancer therapeutics, including cell cycle inhibitors. Efforts to apply nanotechnology in cancer have focused almost exclusively on the delivery of cytotoxic drugs to improve therapeutic index. There has been little consideration of molecularly targeted agents, in particular kinase inhibitors, which can also present considerable therapeutic index limitations. We describe the development of Accurin polymeric nanoparticles that encapsulate the clinical candidate AZD2811, an Aurora B kinase inhibitor, using an ion pairing approach. Accurins increase biodistribution to tumor sites and provide extended release of encapsulated drug payloads. AZD2811 nanoparticles containing pharmaceutically acceptable organic acids as ion pairing agents displayed continuous drug release for more than 1 week in vitro and a corresponding extended pharmacodynamic reduction of tumor phosphorylated histone H3 levels in vivo for up to 96 hours after a single administration. A specific AZD2811 nanoparticle formulation profile showed accumulation and retention in tumors with minimal impact on bone marrow pathology, and resulted in lower toxicity and increased efficacy in multiple tumor models at half the dose intensity of AZD1152, a water-soluble prodrug of AZD2811. These studies demonstrate that AZD2811 can be formulated in nanoparticles using ion pairing agents to give improved efficacy and tolerability in preclinical models with less frequent dosing. Accurins specifically, and nanotechnology in general, can increase the therapeutic index of molecularly targeted agents, including kinase inhibitors targeting cell cycle and oncogenic signal transduction pathways, which have to date proved toxic in humans.
Cell Biology and Toxicology | 2011
Kevin J. Randall; John Turton; John R. Foster
The gastrointestinal (GI) tract is an important target organ for the toxicity of xenobiotics. The toxic effects of xenobiotics on this complex, heterogeneous structure have been difficult to model in vitro and have traditionally been assessed in vivo. The explant culture of GI tissue offers an alternative approach. Historically, the organotypic culture of the GI tract proved far more challenging than the culture of other tissues, and it was not until the late 1960s that Browning and Trier described the means by which intestinal tissues could be successfully cultured. This breakthrough provided a tool researchers could utilise, and adapt, to investigate topics such as the pathogenesis of inflammatory intestinal diseases, the effect of growth factors and cytokines on intestinal proliferation and differentiation, and the testing of novel xenobiotics for efficacy and safety. This review considers that intestinal explant culture shows much potential for the application of a relatively under-used procedure in future biomedical research. Furthermore, there appear to be many instances where the technique may provide experimental solutions where both cell culture and in vivo models have been unable to deliver conclusive and convincing findings.
Archives of Toxicology | 1994
Kulwinder K. Banger; John R. Foster; Edward A. Lock; Celia J. Reed
Many xenobiotics induce lesions within the nasal cavity of experimental animals which are site specific. This site selectivity may be due to regional deposition within the nasal cavity and/or the localisation of biotransformation enzymes. We have developed methodology which allows immunohistochemical localisation of xenobiotic biotransformation enzymes in transverse sections of the rat nasal cavity identical to those normally taken for pathological examination. We report the application of this methodology to six isoenzymes of the glutathione S-transferases (GSTs). All six isoenzymes were predominantly located within olfactory epithelium covering the ethmoturbinates (levels III and IV) and extending forwards into the dorsal meatus (level II). Squamous and transitional epithelia showed little or no staining while respiratory epithelium was weakly stained. Within the respiratory epithelium only the ciliated columnar cells and, to a lesser extent, some of the seromucous glands contained GSTs. Within olfactory epithelium the sustentacular cells, basal cells and subepithelial glands all stained positive for GSTs. The different cell types of olfactory epithelium preferentially express different GST isoenzymes: 1-1 and 2-2 were predominantly located in the subepithelial glands; 3-3, 4-4 and 8-8 in sustentacular and basal cells; 7-7 in basal cells.
Journal of Histochemistry and Cytochemistry | 2004
Rachel E. Thomson; Alison L. Bigley; John R. Foster; Ian R. Jowsey; Clifford R. Elcombe; Terry C. Orton; John D. Hayes
Class kappa glutathione S-transferases are a poorly characterized family of detoxication enzymes whose localization has not been defined. In this study we investigated the tissue, cellular, and subcellular distribution of mouse glutathione S-transferase class kappa 1 (mGSTK1) protein using a variety of immunolocalization techniques. Western blotting analysis of mouse tissue homogenates demonstrated that mGSTK1 is expressed at relatively high levels in liver and stomach. Moderate expression was observed in kidney, heart, large intestine, testis, and lung, whereas sparse or essentially no mGSTK1 protein was detected in small intestine, brain, spleen, and skeletal muscle. Immunohistochemical (IHC) analysis for mGSTK1 revealed granular staining of hepatocytes throughout the liver, consistent with organelle staining. IHC analysis of murine kidney localized GSTK1 to the straight portion of the proximal convoluted tubule (pars recta). Staining was consistent with regions rich in mitochondria. Electron microscopy, using indirect immunocolloidal gold staining, clearly showed that mGSTK1 was localized in mitochondria in both mouse liver and kidney. These results are consistent with a role for mGST K1–1 in detoxification, and the confirmation of the intramitochondrial localization of this enzyme implies a unique role for GST class kappa as an antioxidant enzyme.