Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where R. G. Dickinson is active.

Publication


Featured researches published by R. G. Dickinson.


Chemico-Biological Interactions | 2003

Acyl glucuronide reactivity in perspective: biological consequences.

Mark J. Bailey; R. G. Dickinson

The metabolic conjugation of exogenous and endogenous carboxylic acid substrates with endogenous glucuronic acid, mediated by the uridine diphosphoglucuronosyl transferase (UGT) superfamily of enzymes, leads to the formation of acyl glucuronide metabolites. Since the late 1970s, acyl glucuronides have been increasingly identified as reactive electrophilic metabolites, capable of undergoing three reactions: intramolecular rearrangement, hydrolysis, and intermolecular reactions with proteins leading to covalent drug-protein adducts. This essential dogma has been accepted for over a decade. The key question proposed by researchers, and now the pharmaceutical industry, is: does or can the covalent modification of endogenous proteins, mediated by reactive acyl glucuronide metabolites, lead to adverse drug reactions, perhaps idiosyncratic in nature? This review evaluates the evidence for acyl glucuronide-derived perturbation of homeostasis, particularly that which might result from the covalent modification of endogenous proteins and other macromolecules. Because of the availability of acyl glucuronides for test tube/in vitro experiments, there is now a substantial literature documenting their rearrangement, hydrolysis and covalent modification of proteins in vitro. It is certain from in vitro experiments that serum albumin, dipeptidyl peptidase IV, tubulin and UGTs are covalently modified by acyl glucuronides. However, these in vitro experiments have been specifically designed to amplify any interference with a biological process in order to find biological effects. The in vivo situation is not at all clear. Certainly it must be concluded that all humans taking carboxylate drugs that form reactive acyl glucuronides will form covalent drug-protein adducts, and it must also be concluded that this in itself is normally benign. However, there is enough in vivo evidence implicating acyl glucuronides, which, when backed up by in vivo circumstantial and documented in vitro evidence, supports the view that reactive acyl glucuronides may initiate toxicity/immune responses. In summary, though acyl glucuronide-derived covalent modification of endogenous macromolecules is well-defined, the work ahead needs to provide detailed links between such modification and its possible biological consequences.


Drug Metabolism and Disposition | 2006

IN VITRO CHARACTERIZATION OF LAMOTRIGINE N2-GLUCURONIDATION AND THE LAMOTRIGINE-VALPROIC ACID INTERACTION

Andrew Rowland; David J. Elliot; Andrew Williams; Peter I. Mackenzie; R. G. Dickinson; John O. Miners

Studies were performed to investigate the UDP-glucuronosyltransferase enzyme(s) responsible for the human liver microsomal N2-glucuronidation of the anticonvulsant drug lamotrigine (LTG) and the mechanistic basis for the LTG-valproic acid (VPA) interaction in vivo. LTG N2-glucuronidation by microsomes from five livers exhibited atypical kinetics, best described by a model comprising the expressions for the Hill (1869 ± 1286 μM, n = 0.65 ± 0.16) and Michaelis-Menten (Km 2234 ± 774 μM) equations. The UGT1A4 inhibitor hecogenin abolished the Michaelis-Menten component, without affecting the Hill component. LTG N2-glucuronidation by recombinant UGT1A4 exhibited Michaelis-Menten kinetics, with a Km of 1558 μM. Although recombinant UGT2B7 exhibited only low activity toward LTG, inhibition by zidovudine and fluconazole and activation by bovine serum albumin (BSA) (2%) strongly suggested that this enzyme was responsible for the Hill component of microsomal LTG N2-glucuronidation. VPA (10 mM) abolished the Hill component of microsomal LTG N2-glucuronidation, without affecting the Michaelis-Menten component or UGT1A4-catalyzed LTG metabolism. Ki values for inhibition of the Hill component of LTG N2-glucuronidation by VPA were 2465 ± 370 μM and 387 ± 12 μM in the absence and presence, respectively, of BSA (2%). Consistent with published data for the effect of fluconazole on zidovudine glucuronidation by human liver microsomal UGT2B7, the Ki value generated in the presence of BSA predicted the magnitude of the LTG-VPA interaction reported in vivo. These data indicate that UGT2B7 and UGT1A4 are responsible for the Hill and Michaelis-Menten components, respectively, of microsomal LTG N2-glucuronidation, and the LTG-VPA interaction in vivo arises from inhibition of UGT2B7.


Medical toxicology and adverse drug experience | 1988

Valproate-Associated Hepatotoxicity and its Biochemical Mechanisms

M. J. Eadie; W. D. Hooper; R. G. Dickinson

SummaryIntake of the anticonvulsant drug valproic acid, or its sodium salt, has been associated with occasional instances of severe and sometimes fatal hepatotoxicity. Probably at least 80 cases have occurred worldwide. The syndrome affects perhaps 1 in 10,000 persons taking the drug, and usually develops in the early weeks or months of therapy. Most instances have involved children, usually those receiving more than 1 anticonvulsant. Multiple cases have occurred in 2 families. The typical presentation is of worsening epilepsy, increasing depression of consciousness, and progressive clinical and biochemical evidence of liver failure. The liver has sometimes shown hepatocyte necrosis, and on other occasions widespread microvesicular steatosis, while cholestatic changes have also occurred. The appearances are interpreted as consistent with a drug toxicity reaction.During the hepatotoxicity increased amounts of unsaturated metabolites of valproate, notably 4-en-valproate, have been found in blood and urine. In 4 cases there has been evidence of impaired β-oxidation of valproate with, in 1 case, accumulation of isomers of valproate glucuronide caused by intramolecular rearrangement of the conjugate. There are molecular structural similarities between 4-en-valproate and 2 known hepatotoxins (4-en-pentanoate and methylenecyclopropylacetic acid, the latter being responsible for hypoglycin poisoning). There are also clinical and histopathological similarities between valproate hepatotoxicity and both hypoglycin poisoning and certain spontaneous disorders of isoleucine metabolism (one pathway ofvalproate metabolism is analogous to oxidative degradation of isoleucine). Unsaturated metabolites of valproate, in particular 4-en-valproate, may contribute to the hepatotoxicity of the drug. However, since the hepatotoxicity appears to involve an element of idiosyncrasy, the primary defect in some cases may be an inherited or acquired deficiency in the drug’s β-oxidation. This defect may divert valproate metabolism towards ω-oxidation, with increased formation of the toxin 4-en-valproate, but may also allow increased formation of a toxic metabolite derived from isoleucine, since β-oxidation of isoleucine derivatives will also be impaired.


Biochemical Pharmacology | 1994

Studies on the reactivity of acyl glucuronides. VI: Modulation of reversible and covalent interaction of diflunisal acyl glucuronide and its isomers with human plasma protein in vitro

Adrienne M. Williams; R. G. Dickinson

Acyl glucuronide conjugates are chemically reactive metabolites which can undergo hydrolysis, rearrangement (isomerization via acyl migration) and covalent binding reactions with protein. The present study was undertaken to identify factors modulating the reactivity of diflunisal acyl glucuronide (DAG) with human serum albumin (HSA) in vitro, by comprehensively evaluating the interplay of the three pathways above when DAG and a mixture of its 2-, 3- and 4-isomers (iso-DAG) were incubated with protein. Buffer, plasma, fraction V HSA, fatty acid-free HSA, globulin-free HSA and fatty acid- and globulin-free HSA were investigated at pH 7.4 and 37 degrees, each in the absence and presence of warfarin, diazepam and diflunisal (DF) as reversible binding competitors. DAG and iso-DAG were highly reversibly bound (ca. 98-99.5%) in plasma and HSA solutions. The binding was primarily at the benzodiazepine site, since displacement occurred in the presence of diazepam and fatty acids but not warfarin. DAG degradation, via rearrangement, hydrolysis and covalent adduct formation (in that order of quantitative importance), was retarded in plasma and HSA solutions compared to buffer. The protective effect of protein was afforded by the high reversible binding to the (non-catalytic) benzodiazepine site. The warfarin site appeared to be catalytic for DAG hydrolysis, whereas rearrangement appeared to be hydroxide ion-catalysed only. In contrast to DAG, iso-DAG degradation was greatly accelerated in the presence of protein, through both covalent binding and catalysis of hydrolysis. Covalent binding via DAG was increased in the presence of warfarin but decreased in the presence of diazepam, DF and fatty acids. The opposite effects were found for covalent binding via iso-DAG. The data suggest that covalent binding of DF to HSA via DAG and iso-DAG occurs by different mechanisms (presumably transacylation and glycation, respectively) at different sites (benzodiazepine and warfarin, respectively) whereas reversible binding occurs primarily at the same site (benzodiazepine).


Therapeutic Drug Monitoring | 1989

Urinary excretion of valproate and some metabolites in chronically treated patients

R. G. Dickinson; W. D. Hooper; Paul R. Dunstan; M. J. Eadie

Urinary excretion of the antiepileptic agent valproic acid (VPA) and major metabolites from its glucuronidation, β-oxidation, and ω- and ω1-hydroxylation pathways were studied under steady state conditions in 24 epileptic patients. Some 55 \pm 18% (SD) of the daily dose was recovered in urine, 33 \pm 14% in the form of VPA-glucuronide, 15 \pm 8% as β-oxidation products, and 4 \pm 2% and 2 \pm 1% as products of the ω- and ω1-hydroxylation pathways, respectively. Only 1 \pm 2% of the dose was excreted unchanged. The proportion metabolized by direct glucuronidation tended to increase with dose at the expense of the oxidative pathways, particularly β-oxidation. However, the wide variation in the patterns of urinary metabolite excretion precludes use of routinely collected urinary excretion data as a basis for detecting any but severe noncompliance with VPA therapy or abnormalities of VPA metabolism.


European Journal of Clinical Pharmacology | 1989

First dose and steady-state pharmacokinetics of oxcarbazepine and its 10-hydroxy metabolite

R. G. Dickinson; W. D. Hooper; P. R. Dunstan; M. J. Eadie

SummaryThe pharmacokinetics of oxcarbazepine (a new anticonvulsant which is a congener of carbamazepine) and of its 10-hydroxy metabolite were studied at the outset of therapy in 8 adult epileptics comedicated with other anticonvulsants. The pharmacokinetic study was repeated under steady-state conditions after 3 months of drug intake in 6 of these subjects.The plasma elimination half-life of oxcarbazepine appeared to lie in the range 1.0–2.5 h, and that of its 10-hydroxy metabolite averaged 8.4 h. The apparent oral clearance of the parent drug (averaging 2.51·kg−1·h−1) was high enough to suggest substantial presystemic elimination. The oral clearance fell after 3 months of drug intake, but the half-lives of the drug and metabolite showed no statistically significant change over this time. Steady-state plasma levels of both drug and metabolite were linearly related to drug dose, metabolite levels averaging 9 times those of the parent substance.


Biochemical Pharmacology | 1991

Studies on the reactivity of acyl glucuronides—II: Interaction of diflunisal acyl glucuronide and its isomers with human serum albumin in vitro

R. G. Dickinson; Andrew R. King

A major metabolite of diflunisal (DF) is its reactive acyl glucuronide conjugate (DAG) which can undergo hydrolysis (regeneration of DF), intramolecular rearrangement (isomerization via acyl migration) and intermolecular reactions with nucleophiles. We have compared the fate of DAG and its individual 2-, 3- and 4-O-acyl positional isomers (at ca. 55 micrograms DF equivalents/mL) after incubation with human serum albumin (HSA, 40 mg/mL) at pH 7.4 and 37 degrees. Initial half-lives (T1/2) for DAG and its 2-, 3- and 4-isomers were 53, 75, 61 and 26 min, respectively. DAG was more labile to hydrolysis than any of its isomers but the latter, in particular the 4-isomer, were much better substrates for formation of covalent DF-HSA adducts. After a 2-hr incubation, 2.4, 8.2, 13.7 and 36.6% of substrate DAG and its 2-, 3- and 4-isomers (respectively) were present as DF-HSA adducts. With long term incubation, the concentrations of adducts so generated in situ declined in a biphasic manner, with apparent terminal T1/2 values of ca. 28 days. DAG was much more labile to transacylation with methanol (i.e. formation of DF methyl ester) than an equimolar mixture of its isomers after incubation in a 1:1 methanol:pH 7.4 buffer solution at 37 degrees (T1/2 values of 5 and 70 min, respectively). The data do not support direct transacylation with nucleophilic groups on protein as the predominant mechanism of formation of covalent DF-HSA adducts in vitro.


Epilepsia | 1996

Effect of felbamate on valproic acid disposition in healthy volunteers : inhibition of β-oxidation

W. D. Hooper; M. E. Franklin; P. Glue; C. R. Banfield; E. Radwanski; D. B. McLaughlin; M. E. McIntyre; R. G. Dickinson; M. J. Eadie

Summary: We assessed the effects of felbamate (FBM) on the disposition of valproic acid (VPA) in healthy volunteer men. Eighteen subjects received sodium VPA, 400 mg/day for 21 days. Plasma and urine samples were taken on day 7 to document the steady‐state disposition of VPA alone. From day 8 to day 21, subjects received placebo or FBM at the following doses (mg/day): 1,200, 2,400, 3,000, or 3,600 (n = 2–4 per group). Many adverse events (AE) occurred from about day 10; 2 subjects dropped out and 1 continued on a reduced FBM dose. Pharmacokinetic studies were repeated on day 21 for the 16 subjects who completed the study. FBM was measured in plasma and urine by high‐performance liquid chromatography (HPLC). VPA and its 2–en, 4–en, and 3–oxo metabolites in plasma, and VPA (nonconjugated and total), and its 3–oxo and 4–hydroxy metabolites in urine, were measured by gas chromatography/mass spectrometry (GUMS). Mean plasma FBM trough concentrations on day 21 ranged from 26.9 μg/ml (1,200 mg dose) to 76.8 μg/ml (3,600–mg dose). Mean plasma VPA Cmax values were 32–42 μg/ml in the various subgroups when VPA only was administered. Higher plasma VPA levels were observed when FBM was administered concurrently (55.4–63.8 μg/ml). The excretion of 3–oxo–VPA in urine was significantly lower on day 21 than on day 7, whereas VPA‐glucuronide was significantly increased. The effects of FBM on VPA disposition were dose dependent and were maximal at ∼2400 mg/day. FBM had caused significant inhibition of the β‐oxidation pathway for VPA metabolic clearance, and this had been largely compensated by increased VPA glucuronidation.


Clinical and Experimental Pharmacology and Physiology | 1988

Markers of physical integrity and metabolic viability of the perfused human placental lobule

G.R. Cannell; Ruth M. Kluck; S. E. Hamilton; Robin H. Mortimer; W. D. Hooper; R. G. Dickinson

b


Therapeutic Drug Monitoring | 1985

Extent of urinary excretion of p-hydroxyphenytoin in healthy subjects given phenytoin

R. G. Dickinson; W. D. Hooper; M. Patterson; M. J. Eadie; B. Maguire

Urinary excretion of p-hydroxyphenytoin and its glucuronide conjugate was measured in eight healthy young adults in a comparative bioavailability study of oral sodium phenytoin (approximately 5 mg/kg/dose). Among these subjects the percentage of the phenytoin dose converted to p-hydroxyphenytoin and appearing in urine was relatively similar (mean 79%, range 67-88%). The great majority of the p-hydroxyphenytoin appeared in urine as conjugates; only 1.4-3.4% of the excreted p-hydroxyphenytoin was in the form of unconjugated metabolite. The proportion of a single phenytoin dose excreted in urine as p-hydroxyphenytoin or its conjugate increased from the first dose (mean +/- SD) 74.9 +/- 4.6% to the second dose, given 2 weeks later 79.3 +/- 4.6% (p less than 0.05). This finding suggests that autoinduction of phenytoin metabolism may occur after relatively brief exposure to the drug.

Collaboration


Dive into the R. G. Dickinson's collaboration.

Top Co-Authors

Avatar

W. D. Hooper

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

M. J. Eadie

Royal Brisbane and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

R. S. Addison

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Andrew R. King

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

A.W.E. Wright

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Karine Mardon

University of Queensland

View shared research outputs
Top Co-Authors

Avatar

Min Wang

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

I. Bernus

University of Queensland

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge