Network


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

Hotspot


Dive into the research topics where Janet S. Macpherson is active.

Publication


Featured researches published by Janet S. Macpherson.


Biochemical Pharmacology | 1986

Evidence for the metabolism of mitozantrone by microsomal glutathione transferases and 3-methylcholanthrene-inducible glucuronosyl transferases

C. Roland Wolf; Janet S. Macpherson; John F. Smyth

The metabolism of mitozantrone, a chemotherapeutic agent used in the treatment of breast cancer, has been studied in vitro using rat liver subcellular fractions. This compound would appear to be metabolized by two interesting pathways. One involves conjugation with glucuronic acid, catalyzed most effectively by a 3-methylcholanthrene-inducible glucuronosyl transferase. The other pathway appears to be a glutathione conjugation reaction which requires prior metabolism by cytochrome P-450. The reaction with glutathione appears to be enzymatic as 1-chloro-2,4-dinitrobenzene was a potent inhibitor of this reaction. Liver cytosol did not enhance the microsomal rate of glutathione-conjugate formation, suggesting an important role for the microsomal glutathione transferases in the disposition of this compound. The relationship between these reactions and the mode of action of mitozantrone is discussed.


Biochemical Pharmacology | 2002

Enhanced clearance of topoisomerase I inhibitors from human colon cancer cells by glucuronidation

Jeffrey Cummings; Gary Boyd; Brian T. Ethell; Janet S. Macpherson; Brian Burchell; John F. Smyth; Duncan I. Jodrell

As part of a program to identify novel mechanisms of resistance to topoisomerase I (topo I) inhibitors, the cellular pharmacology of 7-ethyl-10-hydroxycamptothecin (SN-38), the active metabolite of clinically used irinotecan (CPT-11) and NU/ICRF 505, an anthraquinone-tyrosine conjugate, has been investigated in two human colorectal cancer (CRC) cell lines. Two novel metabolites of NU/ICRF 505 (M1 and M2) and a single metabolite of SN-38 (M1) were detected by high performance liquid chromatography in the culture medium of HT29 cells but were absent in HCT116 cells. Identities of all three metabolites were established by a combination of biochemical and physicochemical techniques. M1 of SN-38 was the C10-(beta)-glucuronide of the parent lactone while M1 of NU/ICRF 505 was the C4-O-glucuronide and M2 the tyrosine-O-glucuronide, both of the parent compound. Drug transport studies revealed that by 24hr HT29 cells had effectively cleared 82.5% of NU/ICRF 505 (10 microM) into the culture medium as the two glucuronides. In contrast, intracellular concentrations of NU/ICRF 505 were maintained in HCT116 cells in the absence of glucuronidation at a level 550 times greater than in HT29 cells. HT29 cells cleared 40.9% of SN-38 (1 microM) as the glucuronide to the culture medium, while the parent drug was maintained at a level 2-fold greater in HCT116 cells. Enhanced drug clearance due to glucuronidation may contribute to intrinsic drug resistance of human CRC.


Cancer Chemotherapy and Pharmacology | 1986

The clinical pharmacology of mitozantrone.

John F. Smyth; Janet S. Macpherson; Pamela S. Warrington; Robert C. F. Leonard; C. Roland Wolf

SummaryThe pharmacological disposition of the anthracenedione mitozantrone has been measured in 11 patients with six different tumour types. Administered at 14 mg/m2 as a 30-min infusion, the drug was assayed by a high-pressure liquid chromatographic technique sensitive to 1 ng mitozantrone/ml plasma. The mean half-lives for mitozantrone in plasma were as follows: α, 9.4 min; β, 1.6 h; γ, 23h. The mean volume of distribution (Vd) was 1565 l. For two patients with impaired liver function the T 1/2 γ and Vd were 63.1 h and 4853 l, respectively. Less than 5% of the administered drug was excreted in urine, but two urinary metabolites were identified. These were not influenced by pre incubation of urine samples with β-glucuronidase or sulphatase, suggesting that neither metabolite is a glucuronide or a sulphate conjugate of mitozantrone. Hepatic metabolism is the major route of elimination of mitozantrone, and caution should be exercised when using this drug for patients with hepatic dysfunction.


Biochemical Pharmacology | 1996

Development of anthracenyl-amino acid conjugates as topoisomerase I and II inhibitors that circumvent drug resistance

Jeffrey Cummings; Janet S. Macpherson; Ian Meikle; John F. Smyth

Anthracenyl-amino acid conjugates (AAC) represent a novel class of topoisomerase (topo) inhibitor. The relationship between mechanism of enzyme inhibition and in vitro cytotoxicity has been investigated in a panel of 5 Chinese hamster ovary (CHO) and 2 human ovarian cancer cell lines (A2780) shown to possess different drug resistance phenotypes associated with altered expression of topo I and topo II. From a total of 13 compounds, 4 displayed broad-spectrum activity (IC50 ranging from 3.5-29.7 microM). NU/ICRF 500 (topo II catalytic inhibitor) was 1.4-fold more active against CHO ADR-1, which overexpresses topo II and was essentially noncross-resistant in CHO ADR-r (13.9-fold resistant to doxorubicin (DOX)) and 2780AD (1,460-fold resistant to DOX). NU/ICRF 505, which stabilises topo I cleavable complexes, was noncross-resistant in CHO ADR-3 (3,4-fold resistant to camptothecin) and only 1.8-fold cross-resistant in 2780AD. Hypersensitivity was recorded in ADR-r that overexpresses topo I. The most active compound was NU/ICRF 506, a dual catalytic inhibitor of topo I and II. Hypersensitivity was observed in ADR-r (1.4-fold) but not ADR-1, indicating that topo I is the likely nuclear target, and a low level of resistance was seen in the CHO ADR-6 drug transport mutant and 2780AD. The topo II catalytic inhibitor NU/ICRF 513 only produced hypersensitivity in ADR-r. These data suggest that NU/ICRF 500, 505, and 506 induce cell death, at least partly, through topo inhibition. NU/ICRF 513 appears to be cytotoxic via a nontopo mechanism of action. In addition, NU/ICRF 505 significantly inhibited the growth of two human xenografts (HT-29 colon cancer and NX002 nonsmall-cell lung cancer) in nude mice after i.p. administration at a dose of 25 mg/kg. The important properties of noncross-resistance and in vivo antitumour activity merit further development of AAC as potential new anticancer drugs.


Molecular Cancer Therapeutics | 2006

Time-dependent cytotoxicity induced by SJG-136 (NSC 694501): influence of the rate of interstrand cross-link formation on DNA damage signaling

Stéphanie Arnould; Victoria J. Spanswick; Janet S. Macpherson; John A. Hartley; David E. Thurston; Duncan I. Jodrell; Sylvie Guichard

SJG-136 is a new pyrrolobenzodiazepine dimer inducing time-dependent cytotoxicity. HCT 116 cells were exposed to 50 nmol/L of SJG-136 for 1 hour or 1 nmol/L of SJG-136 for 24 hours to achieve similar levels of interstrand cross-links (ICL). The short exposure led to a rapid formation of ICLs (1 hour), early H2AX foci formation (4 hours), prominent S phase arrest, and greater phosphorylation of Nbs1 (on serine 343) and Chk1 (on serine 317) than a 24-hour exposure. The prolonged exposure at low concentrations of SJG-136 induced a gradual formation of ICLs (up to 24 hours) which was associated with a limited S phase arrest and delayed Nbs1 phosphorylation. Prolonged exposure was also associated with a reduced phosphorylation of p53 on serines 15 and 20, a limited and delayed phosphorylation on serine 392, and a less prominent increase in p21 levels. These data suggest that the 24-hour exposure to a low concentration of SJG-136 led to delayed and reduced DNA damage signaling compared with a higher concentration of SJG-136 for 1 hour, resulting in greater cytotoxicity and contributing to the time-dependent cytotoxic effect of SJG-136. [Mol Cancer Ther 2006;5(6):1602–9]


Biochemical Pharmacology | 1995

Biochemistry of topoisomerase I and II inhibition by anthracenyl-amino acid conjugates.

Ian Meikle; Jeffrey Cummings; Janet S. Macpherson; John A. Hadfield; John F. Smyth

Mono-conjugation of an anthraquinone nucleus with a range of naturally occurring amino acids chemically modified at their C-terminus has been adopted as a synthetic approach in the rational design of novel topoisomerase (topo) inhibitors. The biochemistry of topo I and II inhibition has been investigated for a series of 16 new compounds (NU/ICRF 500-515) from which structure-activity relationships have been investigated. Only three compounds could be demonstrated to bind to DNA: two serine derivatives (NU/ICRFs 500 and 506) and an arginine derivative (NU/ICRF 510). In decatenation and relaxation assays with purified enzyme, several compounds were shown to be potent catalytic inhibitors of topo II (100% inhibition at 5 micrograms/mL (10-15 microM) or less) without stabilizing cleavable complex formation. These included the three DNA binding species (of which NU/ICRF 506 was the most active) and a dihydroxyphenylalanine analogue (NU/ICRF 513). Both NU/ICRFs 500 and 506 were further shown to antagonize DNA cleavage induced by amsacrine. Only NU/ICRF 506 unequivocally inhibited the catalytic activity of topo I without induction of DNA cleavage, and was the only combined topo I and II catalytic inhibitor. One compound, NU/ICRF 505 (tyrosine conjugate), stabilized topo I cleavable complexes without inhibiting the catalytic activity of topo I and II. Modifications to the structure of NU/ICRF 505 revealed that the presence of an unhindered hydroxyl on the tyrosine ring and a more hydrophobic ethyl ester at the amino acid C-terminal were both essential, suggesting a highly specific interaction between drug, enzyme and DNA in the ternary complex. Molecular modelling studies suggested that the observed differences in topo inhibition are a consequence of major conformational alterations brought about by small changes in the amino acid substituent, and confirmed a rigid structural requirement for the induction of topo I cleavage, in addition to a less rigid structural requirement for topo II inhibition. A strong correlation was observed between topo inhibition and in vitro cytotoxicity against the human ovarian cancer cell line A2780 (IC50 range 3.4-11.6 microM), suggesting a mechanism of cell kill, at least in part, involving topo inhibition.


European Journal of Cancer and Clinical Oncology | 1987

Phase I study of TCNU, a novel nitrosourea

John F. Smyth; Janet S. Macpherson; Pamela S. Warrington; M.E. Kerr; J.M. Whelan; Michael A. Cornbleet; Robert C. F. Leonard

TCNU is a chloroethyl nitrosourea based on the endogenous amino acid taurine. This paper reports its first evaluation in man. Eighty-four patients with refractory cancer received 12 dose escalations from 10-150 mg/m2 TCNU administered orally every 6 weeks. Clinical side-effects were predominantly gastro-intestinal but dose-limiting toxicity was thrombocytopenia. Pharmacokinetic monitoring with an HPLC assay sensitive to the nanogram range demonstrated unchanged TCNU in plasma for up to 8 h following administration. The mean half-life was 60 min. Clinical responses were seen in melanoma (four patients), lung cancer (two squamous, one small cell) and one patient each with renal and stomach cancer. These responses, together with the unusual pharmacokinetic profile of TCNU, warrant exploration in disease-orientated phase II studies at a recommended dose of 130 mg/m2 p.o. q 5 weeks.


Mutation Research\/genetic Toxicology | 1995

Cytogenetic evaluation of the mechanism of cell death induced by the novel anthracenyl-amino acid topoisomerase II catalytic inhibitor NU / ICRF 500

Jeffrey Cummings; Adrian T. Sumner; Anne Slavotinek; Ian Meikle; Janet S. Macpherson; John F. Smyth

Anthracenyl-amino acid/dipeptides are novel topoisomerase (topo) inhibitors which can be actively cytotoxic in the low microM range. The present studies have been performed to determine whether cells treated with the topo II catalytic inhibitor NU/ICRF 500 (serine derivative) would manifest cytogenetic lesions consistent with its proposed mechanism of enzyme inhibition. Three other compounds were included for comparison: NU/ICRF 505 (tyrosine) which stabilises topo I cleavable complexes, NU/ICRF 602 (gly-gly) a non-cytotoxic catalytic inhibitor of topo I and II and NU/ICRF 502 (alanine) a non-cytotoxic non-topo inhibitor. Chromosomal damage was measured using the micronucleus test. NU/ICRF 500 (7.5-30 microM) induced an increase in CREST negative micronuclei (11-15 per 500 cells) in human lymphocytes (HL) and blocked the traverse of HL through the cell cycle, with cells accumulating in G2/M at 15 microM drug and G1/S at 30 microM drug. NU/ICRF 502 was without effect in the micronucleus test. NU/ICRF 500 and 602 (90-150 microM) caused no block in passage of synchronised metaphase Chinese hamster ovary cells through mitosis whereas NU/ICRF 505 produced a significant delay. DNA measurements of post-mitotic cells revealed that after NU/ICRF 500 treatment nuclei had a 4C DNA content, indicative of a lack of chromosomal segregation. Normal (2C) DNA content was observed with NU/ICRF 505 and 602. Overall, the data for NU/ICRF 500 are consistent with the cytogenetic modifications expected after catalytic inhibition of topo II and suggest that cell death may be mediated, at least in part, through this mechanism.


Cancer Chemotherapy and Pharmacology | 1995

Accumulation of anthracenyl-amino acid topoisomerase I and II inhibitors in drug-sensitive and drug-resistant human ovarian cancer cell lines determined by high-performance liquid chromatography.

Jeffrey Cummings; Ian Meikle; Janet S. Macpherson; John F. Smyth

Anthracenyl amino acid/dipeptide conjugates (AADC) represent novel structures rationally designed for their DNA-binding properties. A high-performance liquid chromatography method is described for simultaneous determination of five compounds that exhibit novel mechanisms of action as topoisomerase I and II inhibitors. The method uses an Apex ODS-2 column and a mobile phase of 0.25M ammonium acetate/trifluoroacetic acid (pH 3) in methanol with gradient elution. Selective detection is achieved by monitoring at 545 nm, with limits of detection ranging between 2 and 4 ng on the column. AADC are recovered from cell sonicates by solid-phase extraction using C2 cartridges, with extraction efficiencies ranging from 84% to 95%. Drug uptake studies were performed with three active compounds in the human ovarian cancer cell line A2780 and its multi-drug-resistant counterpart 2780AD. Marked differences were observed in the pattern of cellular accumulation produced by each compound. NU/ICRF 505 (tyrosine derivative) was taken up most avidly, reaching plateau levels of 4000 pmol/106 cells after 2 h, with no difference being apparent between A2780 and 2780AD. NU/ICRF 510 (arginine derivative) accumulated slowly in A2780, failing to achieve an equilibrium after 4 h, and appeared to be completely excluded from 2780AD. NU/ICRF 500 (serine derivative) was most rapidly taken up by A2780, producing a plateau of 800 pmol/106 cells after only 30 min with approximately 3-fold less accumulation in 2780AD. These results are correlated to the chemosensitivity of the two cell lines to the three compounds.


European Journal of Cancer | 1997

Cell cycle effects of the novel topoisomerase I inhibitor NU/ICRF 505 in a panel of Chinese hamster ovary cell lines.

Janet S. Macpherson; Jeffrey Cummings; Ian Meikle; E P Miller; John F. Smyth

The effect of the novel topoisomerase I inhibitor NU/ICRF 505 (20 microM, approximate IC50 concentration) on the cell cycle was studied by flow cytometry in four Chinese hamster ovary (CHO) cell lines. Postdrug treated cells were incubated with optimal concentrations of cytochalasin B to prevent re-entry of daughter cells into the cell cycle. NU/ICRF 505 had no significant effect on cell cycle distribution in the parent cell line (CHO-K1) and two mutants hypersensitive to topo II inhibitors (ADR-1, ADR-3), all of which express similar levels of topo I protein. In the drug-resistant variant ADR-r, which overexpresses topo I 2-fold, a significant accumulation of cells in G1 phase was recorded. These results are broadly consistent with the cell cycle effects expected in CHO cells by a classic topo I poison (camptothecin) and add weight to the view that NU/ICRF 505 induces cell death primarily through topo I inhibition.

Collaboration


Dive into the Janet S. Macpherson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ian Meikle

Western General Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gary Boyd

Western General Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Roland Wolf

Western General Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge