Jackie C. Bloomer
GlaxoSmithKline
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Jackie C. Bloomer.
Drug Metabolism and Disposition | 2009
Zoe Riches; Emma L. Stanley; Jackie C. Bloomer; Michael W.H. Coughtrie
Expression levels of the major human sulfotransferases (SULTs) involved in xenobiotic detoxification in a range of human tissues (i.e., SULT “pies”) are not available in a form allowing comparison between tissues and individuals. Here we have determined, by quantitative immunoblotting, expression levels for the five principal human SULTs—SULT1A1, SULT1A3/4, SULT1B1, SULT1E1, and SULT2A1—and determined the kinetic properties toward probe substrates, where available, for these enzymes in cytosol samples from a bank of adult human liver, small intestine, kidney, and lung. We produced new isoform-selective antibodies against SULT1B1 and SULT2A1, which were used alongside antibodies against SULT1A3 and SULT1A1 previously produced in our laboratory or available commercially (SULT1E1). Expression levels were derived using purified recombinant enzymes to construct standard curves for each individual isoform and immunoblot. Substantial intertissue and interindividual differences in expression were observed. SULT1A1 was the major enzyme (>50% of total, range 420-4900 ng/mg cytosol protein) in the liver, followed by SULT2A1, SULT1B1, and SULT1E1. SULT1A3 was completely absent from this tissue. In contrast, the small intestine contained the largest overall amount of SULT of any of the tissues, with SULT1B1 the major enzyme (36%), closely followed by SULT1A3 (31%), and SULT1A1, SULT1E1, and SULT2A1 more minor forms (19, 8, and 6% of total, respectively). The kidney and lung contained low levels of SULT. We provide a unique data set that will add value to the study of the role and contribution of sulfation to drug and xenobiotic metabolism in humans.
Bioorganic & Medicinal Chemistry Letters | 2003
Josie A. Blackie; Jackie C. Bloomer; Murray J.B. Brown; Hung-Yuan Cheng; Beverley Hammond; Deirdre Mary Bernadette Hickey; Robert J. Ife; Colin A. Leach; V.Ann Lewis; Colin H. Macphee; Kevin J. Milliner; Kitty Moores; Ivan Leo Pinto; Stephen A. Smith; Ian G. Stansfield; Steven James Stanway; Maxine A. Taylor; Colin J. Theobald
Modification of the pyrimidone 5-substituent in clinical candidate SB-435495 has given a series of inhibitors of recombinant lipoprotein-associated phospholipase A(2) with sub-nanomolar potency. Cyclopentyl fused derivative 21, SB-480848, showed an enhanced in vitro and in vivo profile versus SB-435495 and has been selected for progression to man.
Chemical Research in Toxicology | 2012
Melanie Z. Sakatis; Melinda J. Reese; Andrew W. Harrell; Maxine A. Taylor; Ian Baines; Liangfu Chen; Jackie C. Bloomer; Eric Yang; Harma Ellens; Jeffrey L. Ambroso; Cerys A. Lovatt; Andrew Ayrton; Stephen E. Clarke
Drug-induced liver injury is the most common cause of market withdrawal of pharmaceuticals, and thus, there is considerable need for better prediction models for DILI early in drug discovery. We present a study involving 223 marketed drugs (51% associated with clinical hepatotoxicity; 49% non-hepatotoxic) to assess the concordance of in vitro bioactivation data with clinical hepatotoxicity and have used these data to develop a decision tree to help reduce late-stage candidate attrition. Data to assess P450 metabolism-dependent inhibition (MDI) for all common drug-metabolizing P450 enzymes were generated for 179 of these compounds, GSH adduct data generated for 190 compounds, covalent binding data obtained for 53 compounds, and clinical dose data obtained for all compounds. Individual data for all 223 compounds are presented here and interrogated to determine what level of an alert to consider termination of a compound. The analysis showed that 76% of drugs with a daily dose of <100 mg were non-hepatotoxic (p < 0.0001). Drugs with a daily dose of ≥100 mg or with GSH adduct formation, marked P450 MDI, or covalent binding ≥200 pmol eq/mg protein tended to be hepatotoxic (∼ 65% in each case). Combining dose with each bioactivation assay increased this association significantly (80-100%, p < 0.0001). These analyses were then used to develop the decision tree and the tree tested using 196 of the compounds with sufficient data (49% hepatotoxic; 51% non-hepatotoxic). The results of these outcome analyses demonstrated the utility of the tree in selectively terminating hepatotoxic compounds early; 45% of the hepatotoxic compounds evaluated using the tree were recommended for termination before candidate selection, whereas only 10% of the non-hepatotoxic compounds were recommended for termination. An independent set of 10 GSK compounds with known clinical hepatotoxicity status were also assessed using the tree, with similar results.
Bioorganic & Medicinal Chemistry Letters | 2002
Josie A. Blackie; Jackie C. Bloomer; Murray J.B. Brown; Hung-Yuan Cheng; Richard L. Elliott; Beverley Hammond; Deirdre Mary Bernadette Hickey; Robert J. Ife; Colin A. Leach; V.Ann Lewis; Colin H. Macphee; Kevin J. Milliner; Kitty Moores; Ivan Leo Pinto; Stephen A. Smith; Ian G. Stansfield; Steven James Stanway; Maxine A. Taylor; Colin J. Theobald; Caroline M. Whittaker
The introduction of a functionalised amido substituent into a series of 1-(biphenylmethylacetamido)-pyrimidones has given a series of inhibitors of recombinant lipoprotein-associated phospholipase A(2) with sub-nanomolar potency and very encouraging developability properties. Diethylaminoethyl derivative 32, SB-435495, was selected for progression to man.
Drug Metabolism and Disposition | 2011
Carole Shardlow; Grant T. Generaux; Christopher MacLauchlin; Nicoletta Pons; Konstantine W. Skordos; Jackie C. Bloomer
Several reports in the literature present the utility and value of in vitro drug-metabolizing enzyme inhibition data to predict in vivo drug-drug interactions in humans. A retrospective analysis has been conducted for 26 GlaxoSmithKline (GSK) drugs and drug candidates for which in vitro inhibition parameters have been determined, and clinical drug interaction information, from a total of 46 studies, is available. The dataset, for drugs with a diverse range of physiochemical properties, included both reversible and potentially irreversible cytochrome P450 inhibitors for which in vitro inhibition parameters (IC50 or KI/kinact as appropriate) were determined using standardized methodologies. Mechanistic static models that differentiated reversible and metabolism-dependent inhibition, and also considered the contribution of intestinal metabolism for CYP3A4 substrates, were applied to estimate the magnitude of the interactions. Several pharmacokinetic parameters, including total Cmax, unbound Cmax, as well as estimates of hepatic inlet and liver concentration, were used as surrogates for the inhibitor concentration at the enzyme active site. The results suggest that estimated unbound liver concentration or unbound hepatic inlet concentration, with consideration of intestinal contribution, offered the most accurate predictions of drug-drug interactions (occurrence and magnitude) for the drugs in this dataset. When used with epidemiological information on comedication profiles for a given therapeutic area, these analyses offer a quantitative risk assessment strategy to inform the necessity of excluding specific comedications in early clinical studies and the ultimate requirement for clinical drug-drug interaction studies. This strategy has significantly reduced the number of clinical drug interaction studies performed at GSK.
Xenobiotica | 2009
Zoe Riches; Jackie C. Bloomer; Patel A; A Nolan; Michael W.H. Coughtrie
Cultured cryopreserved human hepatocytes are extensively used as a model system for studying drug metabolism, although they remain poorly characterized in respect of the major conjugation reactions glucuronidation and sulfation. Using paracetamol (acetaminophen), we assessed eleven samples of cryopreserved human hepatocytes for their suitability to investigate the simultaneous glucuronidation and sulfation of xenobiotics and evaluated inhibitors of conjugation. Kinetic characterization showed broadly similar values for paracetamol conjugation by hepatocytes (as reported in the literature for in vitro systems), with Km values of approximately 6 mM and 0.3 mM for glucuronidation and sulfation, respectively. Substantial interindividual differences were observed. The hepatocytes demonstrated a strong dose-dependent switch from a preponderance of sulfation at low concentrations of paracetamol to glucuronidation at higher doses, consistent with routes of clearance in vivo. A number of drugs, some of which such as probenecid and sulfinpyrazone are known to interact with paracetamol in vivo, were demonstrated to inhibit the sulfation and/or glucuronidation of paracetamol in hepatocytes, demonstrating the potential application of this model system for studying drug–drug interactions involving conjugation.
British Journal of Clinical Pharmacology | 2013
Jackie C. Bloomer; Mike Nash; Alison Webb; Aili L. Lazaar; Claire Beaumont; William J. Guiney
Characterization of the biliary disposition of GSK1325756, using a non‐invasive bile sampling technique and spectrometric analyses, to inform the major routes of metabolic elimination and to enable an assessment of victim drug interaction risk.
Bioorganic & Medicinal Chemistry Letters | 2001
Jackie C. Bloomer; Helen F. Boyd; Deirdre Mary Bernadette Hickey; Robert J. Ife; Colin A. Leach; Colin H. Macphee; Kevin J. Milliner; Ivan Leo Pinto; D. Anthony Rawlings; Stephen A. Smith; Ian G. Stansfield; Steven James Stanway; Maxine A. Taylor; Colin J. Theobald; Caroline M. Whittaker
The lipophilic 1-substituent in a series of 1-((amidolinked)-alkyl)-pyrimidones, inhibitors of recombinant lipoprotein-associated phospholipase A(2), has been modified to give inhibitors of high potency in human plasma and enhanced physicochemical properties. Phenylpiperazineacetamide derivative 23 shows very promising oral activity.
Expert Opinion on Drug Metabolism & Toxicology | 2013
Jackie C. Bloomer; Geo Derimanov; Etienne Dumont; Harma Ellens; Christopher Matheny
Introduction: Pharmacokinetic drug interactions resulting from the inhibition of drug elimination mechanisms are of concern in drug development due to the clinical risk associated with elevated drug concentrations. Advances in understanding the mechanistic basis of these drug interactions has resulted in the widespread application of mechanistic in vitro assays and the conduct of clinical drug interaction studies to predict and quantify the risks in drug development. Areas covered: The authors investigate co-medication prescriptions in target patient populations and characterize the mechanistic basis and clinical impact of known co-medication drug interactions. This has enabled identification of critical mechanistic in vitro studies and provided options to manage co-medication use in clinical studies. Furthermore, they demonstrate, for the pharmaceutical scientist, how improved understanding of the drug interactions risks associated with key medications in a target therapeutic area, can help prioritize the conduct of in vitro data and optimize the timing of the clinical drug interaction studies required to characterize drug interaction risks. Expert opinion: This approach provides a more targeted strategy to drug interaction data generation, as routine application of assays may provide limited impact on drug progression decisions. Assessing co-medications in the target patient population enables early discharge of the safety risks associated with drug interactions and reduced investment in drug interaction studies.
European Journal of Pharmaceutics and Biopharmaceutics | 2017
Jackie C. Bloomer; Claire Ambery; Paul Connolly; Helen Garden; Nick Henley; Neil Hodnett; Sarah Keel; James L. Kreindler; Richard Lloyd; Wayne Matthews; John Yonchuk; Aili L. Lazaar
Graphical abstract Figure. No caption available. ABSTRACT The natural variability of gastric pH or gastric acid reducing medications can result in lower and more variable clinical pharmacokinetics for basic compounds in patient populations. Progressing alternative salt forms with improved solubility and dissolution properties can minimise this concern. This manuscript outlines a nonclinical approach comprising multiple biopharmaceutical, in vitro and physiologically based pharmacokinetic model (PBPK) modelling studies to enable selection of an alternative salt form for danirixin (DNX, GSK1325756), a pharmaceutical agent being developed for chronic obstructive pulmonary disease (COPD). The hydrobromide salt of DNX was identified as having superior biopharmaceutical properties compared to the free base (FB) form in clinical development and the impact of switching to the hydrobromide salt (HBr) was predicted by integrating the nonclinical data in a PBPK model (using GastroPlus™) to enable simulation of clinical drug exposure with FB and HBr salts in the absence and presence of a gastric acid reducing comedication (omeprazole, a proton pump inhibitor (PPI)). Subsequent investigation of DNX pharmacokinetics in a Phase 1 clinical study comparing FB with HBr salt forms confirmed that DNX HBr had reduced the variability of drug exposure and that exposure was not affected by PPI co‐administration with DNX HBr. This case study therefore adds to the surprisingly few examples of a more soluble salt of a weak base translating to an improvement in human pharmacokinetics and illustrates a clear clinical benefit of salt selection during drug development.