Network


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

Hotspot


Dive into the research topics where Malcolm D. Tingle is active.

Publication


Featured researches published by Malcolm D. Tingle.


Biochemical Pharmacology | 2002

UDP-glucuronosyltransferase activity, expression and cellular localization in human placenta at term

Abby C. Collier; Natalie A. Ganley; Malcolm D. Tingle; Marion Blumenstein; Keith W. Marvin; James W. Paxton; Murray D. Mitchell; Jeffrey A. Keelan

The activity, expression and localization of the UDP-glucuronosyltransferases (UGTs) were investigated in human placenta at term. UGT activity (measured with the substrate 4-methylumbelliferone (4-MU)) was observed in all 25 placentas sampled and maximum velocity (V(max)) ranged 13-fold from 5.1+/-0.9 to 66.9+/-17.5 nmol/min/mg protein (mean+/-SD). Substrate affinity (K(m)) ranged 5-fold from 246+/-24 to 1124+/-422 microM. Using reverse transcriptase-polymerase chain reaction (RT-PCR), expression of the isoforms UGT2B4, 2B7, 2B10, 2B11 and 2B15 was observed in all (12/12) placentas sampled and expression of UGT2B17 was noted in 8/12 placentas. Northern analysis of the UGT2B7 isoform in 12 placentas revealed a 10-fold difference in expression with RT-PCR variability and the 13-fold variation observed in UGT activity. The presence of UGT2B4 and 2B7 proteins (52 and 56kDa, respectively) was demonstrated by Western blotting. The sites of placental UGT2B transcription (in situ hybridization) and protein expression (immunohistochemistry) were located in the syncytium of the placental trophoblasts bordering the placental villi. UGT1A proteins could not be observed with immunohistochemistry or Western blotting and expression could not be observed with RT-PCR. Our discovery of UGT expression and activity at the site of maternal-fetal exchange is consistent with a role for UGTs in detoxification of exogenous and endogenous ligands and the maintenance of placental function through clearance and regulation of steroid hormones.


Clinical Cancer Research | 2004

Thalidomide pharmacokinetics and metabolite formation in mice, rabbits, and multiple myeloma patients.

Francisco Chung; Jun Lu; Brian D. Palmer; Philip Kestell; Peter Browett; Bruce C. Baguley; Malcolm D. Tingle; Lai-Ming Ching

Purpose: Thalidomide has a variety of biological effects that vary considerably according to the species tested. We sought to establish whether differences in pharmacokinetics could form a basis for the species-specific effects of thalidomide. Experimental Design: Mice and rabbits were administered thalidomide (2 mg/kg) p.o. or i.v., and plasma concentrations of thalidomide were measured after drug administration using high performance liquid chromotography. Plasma samples from five multiple myeloma patients over 24 hours after their first dose of thalidomide (200 mg) were similarly analyzed and all data were fitted to a one-compartment model. Metabolites of thalidomide in plasma were identified simultaneously using liquid chromatography-mass spectrometry. Results: Plasma concentration-time profiles for the individual patients were very similar to each other, but widely different pharmacokinetic properties were found between patients compared with those in mice or rabbits. Area under the concentration curve values for mice, rabbits, and multiple myeloma patients were 4, 8, and 81 μmol/L · hour, respectively, and corresponding elimination half-lives were 0.5, 2.2, and 7.3 hours, respectively. Large differences were also observed between the metabolite profiles from the three species. Hydrolysis products were detected for all species, and the proportion of hydroxylated metabolites was higher in mice than in rabbits and undetectable in patients. Conclusions: Our results show major interspecies differences in the pharmacokinetics of thalidomide that are related to the altered degree of metabolism. We suggest that the interspecies differences in biological effects of thalidomide may be attributable, at least in part, to the differences in its metabolism and hence pharmacokinetics.


Mutation Research | 2000

Antimutagenic effects of wheat bran diet through modification of xenobiotic metabolising enzymes

Nuala A. Helsby; Shuotun Zhu; Amira Pearson; Malcolm D. Tingle; Lynnette R. Ferguson

Diets containing wheat bran (WB) protect against cancers of the colon or breast in rats, and may be beneficial in humans. In a previous study of rats treated with the carcinogen 2-amino-3-methylimidazo[4,5-f]quinoline (IQ), inclusion of 10% wheat bran in the diet led to an apparent reduction in IQ metabolites but not of intact IQ in plasma. In the present study, male Wistar rats were fed diets containing 0, 10 or 20% wheat bran, and effects on xenobiotic metabolising enzymes compared. Wheat bran-supplementation showed differential effects on phase I enzymes, significantly increasing the activity of hepatic cytochrome P450 isozyme CYP3A2, but slightly reducing the activity of CYP1A1/2. The activities of both hepatic phase II detoxification enzymes glutathione-S-transferase and glucuronosyl transferase were also reduced. Western blotting revealed similar effects on expression of the proteins. Interestingly, the expression of xenobiotic metabolising enzymes (XME) in the colon appeared to be modulated independently of hepatic XME. Although the wheat bran-supplemented diet still led to an increased expression of CYP3A, it now slightly increased CYP1A in the colon. However, 20% wheat bran significantly increased the expression of both glutathione transferase isozymes, GST A1 & A2, in the colon. Natures Gold (NG) is a commercial wheat bran derivative which is lower than wheat bran in dietary fibre, but enriched in vitamins, minerals and various phytochemicals. Dietary supplementation with 20% Natures Gold led to similar trends as seen in wheat bran-fed rats, but more potent effects in both hepatic and colonic enzymes. The significance of these changes for activation of carcinogens to mutagenic metabolites was investigated using the Salmonella/mammalian microsome mutagenicity test. The activation of IQ and benzo[a]pyrene, but not cyclophosphamide, to a mutagen by hepatic S9 from wheat bran-fed or Natures Gold-fed rats was significantly reduced compared with S9 from animals on a diet lacking wheat bran. We suggest that modulation of xenobiotic metabolising enzymes may be an important component of cancer protection by wheat bran, and this effect may relate to micronutrients or cancer-protective non-nutrient phytochemicals rather more than to dietary fibre.


British Journal of Clinical Pharmacology | 2010

The combined impact of CYP2C19 and CYP2B6 pharmacogenetics on cyclophosphamide bioactivation

Nuala A. Helsby; Chung‐Yee Hui; Michael A. Goldthorpe; Janet K. Coller; May Ching Soh; Peter Gow; Janak de Zoysa; Malcolm D. Tingle

AIMS The role of CYP pharmacogenetics in the bioactivation of cyclophosphamide is still controversial. Recent clinical studies have suggested a role for either CYP2C19 or CYP2B6. The aim of this study was to clarify the role of these pharmacogenes. METHODS We used a combined in vitro-in vivo approach to determine the role of these pharmacogenes in the bioactivation of the prodrug to 4-hydroxy cyclophosphamide (4-OHCP). Cyclophosphamide metabolism was determined in a human liver biobank (n= 14) and in patients receiving the drug for treatment of lupus nephritis (n= 16) RESULTS In livers of known CYP2C19 and CYP2B6 genotype and protein expression we observed that there was a combined role for both CYP2C19 and CYP2B6 in the bioactivation of cyclophosphamide in vitro. The presence of at least one loss of function (LoF) allele at either CYP2C19 or CYP2B6 resulted in a significant decrease in both V(max) (P= 0.028) and CL(int) (P= 0.0017) compared with livers with no LoF alleles. This dual genotype relationship was also observed in a preliminary clinical study, with patients who had ≥1 LoF allele at either CYP2C19 or CYP2B6 also displaying significantly (P= 0.0316) lower bioactivation of cyclophosphamide. The mean 4-OHCP : CP bioactivation ratio was 0.0014 (95% CI 0.0007, 0.002) compared with 0.0071 (95% CI 0.0001, 0.014) in patients with no LoF alleles at either of these genes. CONCLUSIONS The presence of ≥1 LoF allele(s) at either CYP2B6 or CYP2C19 appeared to result in decreased bioactivation of cyclophosphamide both in vitro and in patients. Further clinical studies to confirm this relationship are warranted.


Journal of Forensic Sciences | 2010

Validation of an LC–MS Method for the Detection and Quantification of BZP and TFMPP and their Hydroxylated Metabolites in Human Plasma and its Application to the Pharmacokinetic Study of TFMPP in Humans*

Ushtana Antia; Malcolm D. Tingle; Bruce R. Russell

Abstract:  An LC–MS method was developed for benzylpiperazine (BZP) and trifluoromethylphenylpiperazine (TFMPP), constituents of “party pills” or “legal herbal highs,” and their metabolites in human blood plasma. Compounds were resolved using a mixture of ammonium formate (pH 4.5, 0.01 M) and acetonitrile (flow rate of 1.0 mL/min) with a C18 column. Calibration curves were linear from 1 to 50 ng/mL (R2 > 0.99); the lower limit of quantification (LLOQ) was 5 ng/mL; the accuracy was >90%; the intra‐ and interday relative standard deviations (R.S.D) were <5% and <10%, respectively. Human plasma concentrations of TFMPP were measured in blood samples taken from healthy adults (n = 6) over 24 h following a 60‐mg oral dose of TFMPP: these peaked at 24.10 ng/mL (±1.8 ng/mL) (Cmax) after 90 min (Tmax). Plasma concentrations of 1‐(3‐trifluoromethyl‐4‐hydroxyphenyl) piperazine peaked at 20.2 ng/mL (±4.6 ng/mL) after 90 min. TFMPP had two disposition phases (t½ = 2.04 h (±0.19 h) and 5.95 h (±1.63 h). Apparent clearance (Cl/F) was 384 L/h (±45 L/h).


Xenobiotica | 2001

Identification and reactivity of the major metabolite (ß-1-glucuronide) of the anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) in humans

Shu-Feng Zhou; James W. Paxton; Malcolm D. Tingle; Phillip Kestell; M. B. Jameson; P. I. Thompson; Bruce C. Baguley

1. The novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) is extensively metabolized by glucuronidation and 6-methylhydroxylation, resulting in DMXAA acyl glucuronide (DMXAA-G) and 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA). 2. The major human urinary metabolite of DMXAA was isolated and purified by a solid-phase extraction (SPE) method. The isolated metabolite was hydrolysed to free DMXAA by strong base, and by β-glucuronidase. Liquid chromatography-mass spectrometry (LC-MS) and spectral data indicated the presence of a molecular ion [M + 1]+ at m/z 459, which was consistent with the molecular weight of protonated DMXAA-G. 3. The glucuronide was unstable in buffer at physiological pH, plasma and blood with species variability in half-life. Hydrolysis and intramolecular migration were major degradation pathways. 4. In vitro and in vivo formation of DMXAA-protein adducts was observed. The formation of DMXAA-protein adducts in cancer patients receiving DMXAA was significantly correlated with plasma DMXAA-G concentration and maximum plasma DMXAA concentration. 5. At least five metabolites of DMXAA were observed in patient urine, with up to 60% of the total dose excreted as DMXAA-G, 5.5% as 6-OH-MXAA and 4.5% as the glucuronide of 6-OH-MXAA. 6. These data suggest that the major metabolite in patients’ urine is DMXAA β-1- glucuronide, which may undergo hydrolysis, molecular rearrangement and covalent binding to plasma protein. The reactive properties of DMXAA-G may have important implications for the pharmacokinetics, pharmacodynamics and toxicity of DMXAA.


Clinical and Experimental Pharmacology and Physiology | 2008

COMPARATIVE PROTEIN BINDING, STABILITY AND DEGRADATION OF SATRAPLATIN, JM118 AND CISPLATIN IN HUMAN PLASMA IN VITRO

Deanna N. Bell; Johnson J. Liu; Malcolm D. Tingle; Benno Rattel; Thorsten Meyer; Mark J. McKeage

1 Satraplatin is an investigational orally administered platinum‐based antitumour drug. The present study compared the plasma protein binding, stability and degradation of satraplatin with that of its active metabolite JM118 and cisplatin. 2 The platinum complexes were incubated in human plasma for up to 2 h at 37°C and quantified in plasma fractions by inductively coupled plasma–mass spectrometry on‐ or off‐line to high‐performance liquid chromatography. 3 All three platinum drugs became irreversibly bound to plasma proteins and showed negligible reversible protein binding. They were also unstable in plasma and generated one or more platinum‐containing degradation products during their incubation. However, the three platinum complexes differed in the kinetics of their instability and protein binding, as well as in the number of degradation products formed during their incubation. 4 In conclusion, the plasma protein binding, instability and degradation of satraplatin and its active metabolite JM118 are qualitatively similar to that of cisplatin and other clinically approved platinum‐based drugs. Quantitative differences in their irreversible protein binding and degradation were related to their respective physiochemical properties and bioactivation mechanisms.


Journal of Pharmacy and Pharmacology | 2001

Reversible binding of the novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid to plasma proteins and its distribution into blood cells in various species.

Shu-Feng Zhou; James W. Paxton; Philip Kestell; Malcolm D. Tingle

The plasma protein binding and distribution in blood cells of the novel anti‐tumour agent 5,6‐dimethylxanthenone‐4‐acetic acid (DMXAA) has been investigated in‐vitro using filtration and an HPLC method to measure DMXAA. DMXAA (500 μm) was extensively bound in plasma from all species with an unbound fraction (fu) of 4.61 ± 1.10 (mouse), 2.59 ± 0.32 (rat), 2.02 ± 0.48 (rabbit) and 2.07 ± 0.23% (human). The binding was concentration dependent with DMXAA concentrations ≥ 1000 μm markedly increasing the fu in the plasma from all species. The estimated number of binding sites in plasma were 2.4 ± 0.2 (mouse), 1.7 ± 0.2 (rat), 0.8 ± 0.1 (rabbit) and 2.1 ± 0.2 (human). The major binding protein in human plasma was albumin, with negligible binding to γ‐globulin and α1‐acid glycoprotein. There was a significant linear relationship between the bound:free DMXAA concentration ratio (Cb/Cu) and albumin concentration in human serum albumin solution (r = 0.955; P < 0.05) and in healthy human plasma (r = 0.998; P < 0.05), but not in plasma from cancer patients (n = 5), nor across species. In cancer patients (n = 5) DMXAA had a significantly higher (P < 0.05) fu (4.60 ± 0.42%) compared with healthy human plasma (2.07 ± 0.23%). In human plasma, the fu of DMXAA (500 μm) was significantly reduced by 500 μm diazepam (P < 0.05), but not by warfarin, phenylbutazone, salicylic acid, ibuprofen or clofibric acid at that concentration. DMXAA significantly reduced the binding of dansylsarcosine (a Site‐II binder) to HSA, but significantly increased the binding of dansylamide (a Site‐I binder). Within species, the blood:plasma concentration ratio (CBL/CP) of DMXAA was relatively constant (mouse, 0.581 ± 0.005; rat, 0.667 ± 0.025; rabbit, 0.637 ± 0.019; human, 0.673 ± 0.103) over the range 50–1000 μm, but increased significantly at DMXAA concentrations > 1000 μm in all species except the rabbit. These results indicate that significant alterations in DMXAA plasma binding and distribution into blood cells occur with increasing concentrations of DMXAA in all species, and also that significant interspecies differences exist. It would be more appropriate to compare plasma unbound concentrations when assessing DMXAA exposure in cancer patients or when extrapolating across species.


Journal of Chromatography B: Biomedical Sciences and Applications | 1999

Determination of two major metabolites of the novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid in hepatic microsomal incubations by high-performance liquid chromatography with fluorescence detection

Shu-Feng Zhou; James W. Paxton; Malcolm D. Tingle; John L. McCall; Philip Kestell

High-performance liquid chromatographic methods have been developed and validated for the glucuronidated and oxidative metabolites of the novel anti-tumour agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA), produced in human liver microsomal incubations. Calibration curves for DMXAA acyl glucuronide (DMXAA-Glu) and 6-hydroxymethyl-5-methylxanthenone-4-acetic acid (6-OH-MXAA) were constructed over the concentration ranges of 0.25 to 20 and 0.5 to 40 μM, respectively. Assay performance was determined by intra- and inter-day accuracy and precision of quality control (QC) samples. The difference between the theoretical and measured concentration, and the coefficient of variation, were less than 15% at low QC concentrations, and less than 10% at medium and high QC concentrations for both analytes. The methods presented good accuracy, precision and sensitivity for use in kinetic studies of the glucuronidated and oxidative metabolites of DMXAA in human liver microsomes.


Drugs & Aging | 2002

Thalidomide in Cancer Treatment

Shu-Feng Zhou; Philip Kestell; Malcolm D. Tingle; James W. Paxton

There is increased interest in the treatment of cancer with thalidomide because of its antiangiogenic, immunomodulating and sedative effects. In animal models, the antitumour activity of thalidomide is dependent on the species, route of administration and coadministration of other drugs. For example, thalidomide has shown antitumour effects as a single agent in rabbits, but not in mice. In addition, the antitumour effects of the conventional cytotoxic drug cyclophosphamide and the tumour necrosis factor inducer 5,6-dimethylxanthenone-4-acetic acid (DMXAA) were found to be potentiated by thalidomide in mice bearing colon 38 adenocarcinoma tumours. Further studies have revealed that thalidomide upregulates intratumoral production of tumour necrosis factor-α 10-fold over that induced by DMXAA alone. Coadministration of thalidomide also significantly reduced the plasma clearance of DMXAA and cyclophosphamide. All these effects of thalidomide may contribute to the enhanced antitumour activity.Recent clinical trials of thalidomide have indicated that it has minimal anticancer activity for most patients with solid tumours when used as a single agent, although it was well tolerated. However, improved responses have been reported in patients with multiple myeloma. Palliative effects of thalidomide on cancer-related symptoms have also been observed, especially for geriatric patients with prostate cancer. Thalidomide also eliminates the dose-limiting gastrointestinal toxic effects of irinotecan. There is preliminary evidence indicating that the clearance of thalidomide may be reduced in the elderly.The exact role of thalidomide in the treatment of cancer and cancer cachexia in the elderly remains to be elucidated. However, it may have some value as part of a multimodality anticancer therapy, rather than as a single agent.

Collaboration


Dive into the Malcolm D. Tingle's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shu-Feng Zhou

University of South Florida

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Abby C. Collier

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jun Lu

Auckland University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge