Network


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

Hotspot


Dive into the research topics where Ann L. Jackman is active.

Publication


Featured researches published by Ann L. Jackman.


European Journal of Cancer | 1981

A potent antitumour quinazoline inhibitor of thymidylate synthetase: Synthesis, biological properties and therapeutic results in mice

T.R. Jones; A.H. Calvert; Ann L. Jackman; S.J. Brown; M. Jones; Kenneth R. Harrap

Abstract We describe the synthesis and some biological properties of N-(4-N-[2-amino-4-hydroxy-6-quinazolinyl)methyl]prop-2-ynylaminobenzoyl)- l -glutamic acid (CB 3717), a potent antifolate inhibitor of thymidylate synthetase (EC 2.1.1.45). This compound inhibited thymidylate synthetase from L1210 cells competitively with respect to the substrate methylene-tetrahydrofolate. The inhibitor constant (Ki) was approximately lnM. Toxicity to L1210 cells in suspension culture could be reversed more effectively by co-incubation with thymidine than with folinic acid. A cultured subline of L1210 cells resistant to methotrexate by virtue of increased cellular dihydrofolate reductase (EC 1.5.1.4) content was not cross-resistant to CB 3717. Treatment of animals bearing the L1210 tumour with CB 3717 at 125 or 200 mg/kg/day for 5 days resulted in ‘cures’ (>120 day survival) in 90% of animals. These doses did not cause obvious toxicity or significant weight loss in animals. The studies in tissue culture and the enzymology suggest that thymidylate synthetase is the cytotoxic locus of action of this compound. The high therapeutic efficacy of CB 3717 may be due to preservation of de novo purine synthesis, which is inhibited by other antifolates. The in vitro results further suggest that CB 3717 could be active against methotrexate-resistant tumours.


Arthritis & Rheumatism | 2009

Folate receptor beta as a potential delivery route for novel folate antagonists to macrophages in the synovial tissue of rheumatoid arthritis patients

Joost W. van der Heijden; Ruud Oerlemans; Ben A. C. Dijkmans; Huiling Qi; Conny J. van der Laken; Willem F. Lems; Ann L. Jackman; Maarten C. Kraan; Paul P. Tak; Manohar Ratnam; Gerrit Jansen

OBJECTIVE To determine the expression of folate receptor beta (FRbeta) in synovial biopsy tissues and peripheral blood lymphocytes from rheumatoid arthritis (RA) patients and to identify novel folate antagonists that are more selective in the targeting and internalization of FRbeta than methotrexate (MTX). METHODS Immunohistochemistry and computer-assisted digital imaging analyses were used for the detection of FRbeta protein expression on immunocompetent cells in synovial biopsy samples from RA patients with active disease and in noninflammatory control synovial tissues. FRbeta messenger RNA (mRNA) levels were determined by reverse transcription-polymerase chain reaction analysis. Binding affinities of FRbeta for folate antagonists were assessed by competition experiments for 3H-folic acid binding on FRbeta-transfected cells. Efficacy of FRbeta-mediated internalization of folate antagonists was evaluated by assessment of antiproliferative effects against FRbeta-transfected cells. RESULTS Immunohistochemical staining of RA synovial tissue showed high expression of FRbeta on macrophages in the intimal lining layer and synovial sublining, whereas no staining was observed in T cell areas or in control synovial tissue. Consistently, FRbeta mRNA levels were highest in synovial tissue extracts and RA monocyte-derived macrophages, but low in peripheral blood T cells and monocytes. Screening of 10 new-generation folate antagonists revealed 4 compounds for which FRbeta had a high binding affinity (20-77-fold higher than for MTX). One of these, the thymidylate synthase inhibitor BCG 945, displayed selective targeting against FRbeta-transfected cells. CONCLUSION Abundant FRbeta expression on activated macrophages in synovial tissue from RA patients deserves further exploration for selective therapeutic interventions with high-affinity-binding folate antagonists, of which BCG 945 may be a prototypical representative.


British Journal of Cancer | 1995

Mechanisms of acquired resistance to the quinazoline thymidylate synthase inhibitor ZD1694 (Tomudex) in one mouse and three human cell lines

Ann L. Jackman; Lloyd R. Kelland; R. Kimbell; M. Brown; W. Gibson; G. W. Aherne; Anthea Hardcastle; F. T. Boyle

Four cell lines, the mouse L1210 leukaemia, the human W1L2 lymphoblastoid and two human ovarian (CH1 and 41M) cell lines, were made resistant to ZD1694 (Tomudex) by continual exposure to incremental doses of the drug. A 500-fold increase in thymidylate synthase (TS) activity is the primary mechanism of resistance to ZD1694 in the W1L2:RD1694 cell line, which is consequently highly cross-resistant to other folate-based TS inhibitors, including BW1843U89, LY231514 and AG337, but sensitive to antifolates with other enzyme targets. The CH1:RD1694 cell line is 14-fold resistant to ZD1694, largely accounted for by the 4.2-fold increase in TS activity. Cross-resistance was observed to other TS inhibitors, including 5-fluorodeoxyuridine (FdUrd). 41M:RD1694 cells, when exposed to 0.1 microM [3H]ZD1694, accumulated approximately 20-fold less 3H-labelled material over 24 h than the parental line. Data are consistent with this being the result of impaired transport of the drug via the reduced folate/methotrexate carrier. Resistance was therefore observed to methotrexate but not to CB3717, a compound known to use this transport mechanism poorly. The mouse L1210:RD1694 cell line does not accumulate ZD1694 or Methotrexate (MTX) polyglutamates. Folylpolyglutamate synthetase substrate activity (using ZD1694 as the substrate) was decreased to approximately 13% of that observed in the parental line. Cross-resistance was found to those compounds known to be active through polyglutamation.


Molecular Cancer Therapeutics | 2006

Potentiation of paclitaxel activity by the HSP90 inhibitor 17-allylamino-17-demethoxygeldanamycin in human ovarian carcinoma cell lines with high levels of activated AKT

Nivedita Sain; Bhavani Krishnan; Michael G. Ormerod; Assunta De Rienzo; Wai M. Liu; Stanley B. Kaye; Paul Workman; Ann L. Jackman

Activation of the phosphatidylinositol-3-kinase (PI3K)/AKT survival pathway is a mechanism of cytotoxic drug resistance in ovarian cancer, and inhibitors of this pathway can sensitize to cytotoxic drugs. The HSP90 inhibitor 17-allylamino-17-demethoxygeldanamycin (17-AAG) depletes some proteins involved in PI3K/AKT signaling, e.g., ERBB2, epidermal growth factor receptor (EGFR), and phosphorylated AKT (p-AKT). 17-AAG and paclitaxel were combined (at a fixed 1:1 ratio of their IC50) in four ovarian cancer cell lines that differ in expression of p-AKT, EGFR, and ERBB2. The EGFR-overexpressing A431 and KB epidermoid cell lines were also included. Combination indices (CI) were calculated using the median-effect equation and interpreted in the context of 17-AAG-mediated inhibition of PI3K signaling. Synergy was observed in IGROV-1- and ERBB2-overexpressing SKOV-3 ovarian cancer cells that express a high level of constitutively activated p-AKT [CI at fraction unaffected (fu)0.5 = 0.50 and 0.53, respectively]. Slight synergy was observed in A431 cells (moderate p-AKT/overexpressed EGFR; CI at fu0.5 = 0.76) and antagonism in CH1 (moderate p-AKT), HX62 cells (low p-AKT), and KB cells (low p-AKT/overexpressed EGFR; CI at fu50 = 3.0, 3.5, and 2.0, respectively). The observed effects correlated with changes in the rate of apoptosis induction. 17-AAG induced a decrease in HSP90 client proteins (e.g., C-RAF, ERBB2, and p-AKT) or in downstream markers of their activity (e.g., phosphorylated extracellular signal-regulated kinase or p-AKT) in SKOV-3, IGROV-1, and CH1 cells at IC50 concentrations. A non–growth-inhibitory concentration (6 nmol/L) reduced the phosphorylation of AKT (but not extracellular signal-regulated kinase) and sensitized SKOV-3 cells to paclitaxel. In conclusion, 17-AAG may sensitize a subset of ovarian cancer to paclitaxel, particularly those tumors in which resistance is driven by ERBB2 and/or p-AKT. [Mol Cancer Ther 2006;5(5):1197–208]


European Journal of Cancer | 1995

ZD1694 (Tomudex): A new thymidylate synthase inhibitor with activity in colorectal cancer

Ann L. Jackman; D.C Farrugia; William Gibson; Rosemary Kimbell; Kenneth R. Harrap; T.C Stephens; M Azab; F.T Boyle

ZD1694 (Tomudex) is a new antifolate which is a specific inhibitor of thymidylate synthase (TS). Evidence suggests that ZD1694 has a spectrum of activity that only partially overlaps with 5-fluorouracil (modulated with leucovorin) against colon tumours in vitro. Potent cytotoxic activity is dependent upon active uptake into cells via the reduced folate/methotrexate cell membrane carrier (RFC) and subsequent metabolism to polyglutamated forms (tri, tetra and pentaglutamates). These polyglutamates are approximately 60-fold more active as TS inhibitors and are not effluxed readily from cells. Extensive polyglutamation also occurs in various mouse tissues (e.g. small intestinal epithelium, liver and kidney), resulting in high tissue/plasma drug ratios which persist for a prolonged period. ZD1694 has antitumour activity in mice, although the high plasma thymidine in this species complicates: (1) the interpretation of therapeutic index; (2) tumour types in which activity is likely to be observed; and (3) translation of doses and schedules for clinical evaluation. ZD1694 entered clinical study and has completed Phase I and II evaluation, with activity observed in several tumour types. Appreciable activity in the Phase II colorectal study (29% objective response rate on interim analysis) led to the current Phase III study, randomised against 5-fluorouracil/leucovorin.


Biochemical Pharmacology | 1984

Modulation of anti-metabolite effects: Effects of thymidine on the efficacy of the quinazoline-based thymidylate synthetase inhibitor, cb3717

Ann L. Jackman; Taylor Ga; Calvert Ah; Kenneth R. Harrap

CB3717 (N-(4-(N-((2-amino-4- hydroxy-6-quinazolinyl)methyl)prop-2-ynylamino ) benzoyl)-L-glutamic acid) is an antitumour agent that inhibits thymidylate synthetase (TS). A dose-dependent fall in plasma thymidine (dThd) (1.43 microM to 0.47 microM) occurred in non-tumour-bearing mice following the administration of CB3717. Further, in mice carrying the L1210/CBRI tumour, the drugs antitumour properties were ablated by co-administration of dThd, an effect consistent with TS being the cytotoxic locus. In vitro studies of protection by dThd against CB3717 cytotoxicity were carried out in an attempt to quantify this reversal. The metabolism of [14C]-dThd was measured in cultures of L1210 cells (10(4)/ml) exposed to a completely cytotoxic dose of CB3717 (50 microM). The cytotoxicity of the drug was only expressed when the dThd concentration (0.5-2 microM) had fallen to less than 0.1 microM in the media. This reduction was due to: (1) dThd incorporation into DNA, (2) catabolism of dThd to thymine. By reducing the initial cell concentration to 10(3)/ml the depletion of dThd was substantially reduced and consequently cells continued to grow for a longer period. The critical concentration of dThd, below which growth in the presence of CB3717 could not be supported was estimated to be between 0.026 and 0.1 microM. Thus even the minimum level of dThd achieved in vivo was still in excess of that required for protection from CB3717 toxicity in vitro. There was a small accumulation of deoxyuridine (dUrd) (approximately 2-fold) in mouse plasma 24 hr after completion of a 5-day course of CB3717 (200 mg/kg) but in vitro studies demonstrated that this was unlikely to modulate CB3717 toxicity in the presence of dThd. We caution against the use of rodent tumour models (or human tumour xenografts) for antitumour or toxicity testing of compounds designed to inhibit the de novo synthesis of thymidylate; they may be misleading because the high dThd levels found in these animals compared with man may mask the cytotoxic effects of these drugs.


Cancer Chemotherapy and Pharmacology | 1980

The clinical pharmacology of the adenosine deaminase inhibitor 2'-deoxycoformycin.

John F. Smyth; Rosanne M. Paine; Ann L. Jackman; Kenneth R. Harrap; Marvin M. Chassin; Richard H. Adamson; David G. Johns

Summary2′-deoxycoformycin (2′-dCF; Pentostatin), a stoichiometric inhibitor of mammalian adenosine deaminase (ado deaminase), exhibits immunosuppressive and antilymphocytic activity in animal test systems. A clinical pharmacology/phase I study of 2′-dCF administered as a single agent has been completed (18 patients). Dose levels ranged from 0.1 mg/kgx1 to 0.25 mg/kg/dayx5; ado deaminase and 2′-dCF were measured spectrophotometrically. Plasma decay curves were bi-exponential (α and βt1/2 values about 1 and 10 h respectively). Recovery of unchanged 2′-dCF from urine (48 h) was 32%–48% of the administered drug. Major toxic manifestations were lymphocytopenia (all patients) and urate nephropathy (1 patient, with subsequent patients in the series receiving allopurinol, 300 mg/day). Three partial responses were seen in seven patients with acute lymphocytic leukaemia receiving 0.25 mg 2′-dCF/kg/dayx5.


Journal of Clinical Oncology | 1996

Phase I trial of ZD1694, a new folate-based thymidylate synthase inhibitor, in patients with solid tumors.

S J Clarke; Janet Hanwell; M. P. de Boer; André Planting; Jaap Verweij; M Walker; Robert S. Smith; Ann L. Jackman; L R Hughes; Kenneth R. Harrap; G T Kennealey; Ian Judson

PURPOSE To perform a phase I clinical and pharmacologic study of ZD1694 (Tomudex, Alderley Park, United Kingdom), a new folate-based thymidylate synthase (TS) inhibitor, in patients with advanced malignancy. PATIENTS AND METHODS From February 1991 to January 1993, 61 patients with a range of solid tumor received 161 courses of ZD1694 given as a single 15-minute intravenous infusion every 3 weeks, at escalating doses from 0.1 to 3.5 mg/m2. Pharmacokinetic (PK) analysis was performed with the first two courses of treatment. There were 33 men and 28 women with a median age of 53 years (range, 21 to 73). Fifty-five patients (90%) had previously received chemotherapy. RESULTS Reversible liver toxicity and dose-related gastrointestinal (GI) and bone marrow toxicity occurred at > or = 1.6 mg/m2. Liver function usually returned to normal with repeated treatment, but GI and bone marrow toxicities generally became more severe. No renal toxicity was observed. The maximum-tolerated dose (MTD) was 3.5 mg/m2, at which, in addition to antiproliferative toxicities, four of six patients (67%) developed severe malaise that consisted of anorexia, nausea, and asthenia, with rapidly decreasing performance status that limited re-treatment. Abnormal liver function was also seen in four patients (67%). At 3.0 mg/m2, grades III and IV diarrhea were seen in six of 23 patients (26%) and grade IV myelosuppression in two others. Liver toxicity was self-limiting and not associated with severe malaise. Two patients had a partial response to treatment. PK analysis showed that plasma elimination was triexponential, with pronounced variability in the mean terminal half-life (t1/2gamma) for a given dose ranging from 8.2 to 105 hours. There was a linear relationship between dose and both the area under the concentration-time curve (AUC) and maximum concentration (Cmax), but no clear association between these parameters and response or toxicity. CONCLUSION The dose of ZD1694 recommended for phase II trials is 3.0 mg/m2.


Vitamins and Hormones Series | 2008

Exploitation of the folate receptor in the management of cancer and inflammatory disease.

Christopher P. Leamon; Ann L. Jackman

Over the last 25+ years, the folate receptor (FR) has emerged as an attractive tumor biomarker with the potential to be exploited for therapeutic purposes. Increasing evidence suggests that this endocytosing protein can functionally mediate the cellular uptake and retention of natural folates, certain antifolates, and folate-drug conjugates; the consequences of the latter two events could result in biological modulation, including (but not limited to) tumor-targeted cytotoxicity. Because its tissue expression profile appears to be somewhat limited to either tissues responsible for whole body retention of folates (e.g., kidney and placenta), or certain pathologic tissues (e.g., tumors or activated macrophages), the FR is believed to be a useful biological target for disease management. Indeed, recent years have been peppered with reports of novel FR-targeted therapies, and many have demonstrated impressive in vivo potency, particularly against tumor xenografts, without the undesirable toxicity that often accompanies nontargeted drug regimens. This chapter will provide essential details on the properties of the FR, including where it is expressed and how it has been successfully manipulated for therapeutic benefit.


Clinical Cancer Research | 2004

The Role of α-Folate Receptor-Mediated Transport in the Antitumor Activity of Antifolate Drugs

Davinder S. Theti; Ann L. Jackman

Purpose: Raltitrexed, pemetrexed, lometrexol, and ZD9331 are antifolate drugs transported into cells via the ubiquitously expressed reduced-folate carrier. They display also high affinity for the α-folate receptor (α-FR), a low capacity folate transporter that is highly overexpressed in some epithelial tumors. The role of α-FR in the activity of the antifolates has been evaluated in two α-FR-overexpressing cell lines grown in a physiological concentration of folate (20 nm R,S-Leucovorin). Experimental Design and Results: A431-FBP cells (transfected with the α-FR) were 3–5-fold more sensitive to the antifolates than A431 cells. KB cells (constitutive α-FR overexpression) were less sensitive to the drugs when coexposed to 1 μm folic acid to competitively inhibit binding to the α-FR. Raltitrexed, pemetrexed, and lometrexol are polyglutamated in cells leading to drug retention, e.g., the raltitrexed 4- and 24-h IC50s in A431 cells were ∼0.6 and 0.008 μm, respectively, compared with 0.003 μm for 72-h continuous exposure. A431-FBP cells were ∼3-fold more sensitive to raltitrexed and pemetrexed at all exposure times. ZD9331 is not polyglutamated, and the 4- and 24-h IC50s in A431 cells were >100 and ∼100 μm, respectively, reducing to 2 and 0.1 μm, respectively, in A431-FBP cells. The ZD9331 4- and 24-h IC50s in KB cells were 20 and 1 μm, respectively, and reversible by coaddition of 1 μm folic acid. An in situ thymidylate synthase assay demonstrated continued thymidylate synthase inhibition after ZD9331-treated A431-FBP and KB, but not A431, cells were placed in drug-free medium for 16 h. A model is proposed in which the antifolates accumulate in the α-FR/endosomal apparatus, leading to slow release into the cytoplasm. In particular, this leads to cellular retention of the nonpolyglutamatable ZD9331. Conclusions: Antifolate drugs, particularly ZD9331, have the potential for increased efficacy in tumors that highly overexpress the α-FR.

Collaboration


Dive into the Ann L. Jackman's collaboration.

Top Co-Authors

Avatar

Rosemary Kimbell

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Ian Judson

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Vassilios Bavetsias

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Kenneth R. Harrap

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anthea Hardcastle

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Graham M.F. Bisset

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Lorraine Skelton

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

William Gibson

Institute of Cancer Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge