Network


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

Hotspot


Dive into the research topics where Dra Uges is active.

Publication


Featured researches published by Dra Uges.


British Journal of Cancer | 2002

Reduced 5-FU clearance in a patient with low DPD activity due to heterozygosity for a mutant allele of the DPYD gene.

Jan Gerard Maring; A. B. P. van Kuilenburg; Janet Haasjes; H. Piersma; Hjm Groen; Dra Uges; A. H. van Gennip; E.G.E. de Vries

5-fluorouracil pharmacokinetics, dihydropyrimidine dehydrogenase-activity and DNA sequence analysis were compared between a patient with extreme 5-fluorouracil induced toxicity and six control patients with normal 5-fluorouracil related symptoms. Patients were treated for colorectal cancer and received chemotherapy consisting of leucovorin 20u2009mgu2009m−2 plus 5-fluorouracil 425u2009mgu2009m−2. Blood sampling was carried out on day 1 of the first cycle. The 5-fluorouracil area under the curve0 → 3h in the index patient was 24.1u2009mgu2009hu2009l−1 compared to 9.8±3.6 (range 5.4–15.3) mgu2009hu2009l−1 in control patients. The 5-fluorouracil clearance was 520u2009mlu2009min−1 vs 1293±302 (range 980–1780) mlu2009min−1 in controls. The activity of dihydropyrimidine dehydrogenase in mononuclear cells was lower in the index patient (5.5u2009nmolu2009mgu2009h−1) compared to the six controls (10.3±1.6, range 8.0–11.7u2009nmolu2009mgu2009h−1). Sequence analysis of the dihydropyrimidine dehydrogenase gene revealed that the index patient was heterozygous for a IVS14+1G>A point mutation. Our results indicate that the inactivation of one dihydropyrimidine dehydrogenase allele can result in a strong reduction in 5-fluorouracil clearance, causing severe 5-fluorouracil induced toxicity.


British Journal of Cancer | 1996

Differential expression of DNA topoisomerase II alpha and -beta in P-gp and MRP-negative VM26, mAMSA and mitoxantrone-resistant sublines of the human SCLC cell line GLC4.

Sebo Withoff; E.G.E. de Vries; W. N. Keith; Edith F. Nienhuis; W.T.A. van der Graaf; Dra Uges; Nh Mulder

Sublines of the human small-cell lung carcinoma (SCLC) cell line GLC4 with acquired resistance to teniposide, amsacrine and mitoxantrone (GLC4/VM20x, GLC4/AM3x and GLC4/MIT60x, respectively) were derived to study the contribution of DNA topoisomerase II alpha and -beta (TopoII alpha and -beta) to resistance for TopoII-targeting drugs. The cell lines did not overexpress P-glycoprotein or the multidrug resistance-associated protein but were cross-resistant to other TopoII drugs. GLC4/VM20x showed a major decrease in TopoII alpha protein (54%; for all assays presented in this paper the GLC4 level was defined to be 100%) without reduction in TopoII beta protein; GLC4/AM3x showed only a major decrease in TopoII beta protein (to 18%) and not in TopoII alpha. In GLC4/MIT60x, the TopoII alpha and -beta protein levels were both decreased (TopoII alpha to 31%; TopoII beta protein was undetectable). The decrease in TopoII alpha protein in GLC4/VM20x and GLC4/MIT60x, was mediated by decreased TopoII alpha mRNA levels. Loss of TopoII alpha gene copies contributed to the mRNA decrease in these cell lines. Only in the GLC4/MIT60x cell line was an accumulation defect observed for the drug against which the cell line was made resistant. In conclusion, TopoII alpha and -beta levels were decreased differentially in the resistant cell lines, suggesting that resistance to these drugs may be mediated by a decrease in a specific isozyme.


British Journal of Cancer | 1995

PHASE-II AND PHARMACOKINETIC STUDY OF LOBAPLATIN IN PATIENTS WITH RELAPSED OVARIAN-CANCER

J. A. Gietema; Gj Veldhuis; Hj Guchelaar; Phb Willemse; Dra Uges; A. Cats; Wta vanderGraaf; Dt Sleijfer; Ege Devries; Nh Mulder; J. Boonstra

In phase I studies, lobaplatin showed activity in ovarian cancer patients pretreated with platinum. A phase II trial with lobaplatin was performed in patients with refractory or relapsed ovarian cancer to define activity and pharmacokinetics. Twenty-two patients were treated with lobaplatin administered as an intravenous bolus every 4 weeks. Dependent on creatinine clearance (CRCL) patients received 30 or 50 mg m-2 lobaplatin as the starting dose. Twenty-two patients received 78 courses (median 3, range 1-6). In eight patients total platinum (TPt) in plasma and urine, free platinum (FPt) in plasma ultrafiltrate (both measured by atomic absorption spectrometry) and lobaplatin in plasma ultrafiltrate measured (by high-performance liquid chromatography) were measured. Toxicity was confined to mild nausea and vomiting, mild leucocytopenia (WHO grade 3 in 18% of the courses), and renal function-related thrombocytopenia (WHO grade 3/4 in 53% of the courses). A correlation was found between CRCL and reduction in platelet count (r = -0.77; P < 0.01). No renal toxicity was encountered. Five of 21 evaluable patients (24%) achieved a response (four complete remissions and one partial remission). Remissions occurred mainly in patients who relapsed more than 6 months after primary treatment. The median survival from start of lobaplatin treatment was 8 months. The mean areas under the curve (AUCs) were 4.2 +/- 0.5, 3.0 +/- 0.6, and 3.2 +/- 1.1 h mgl-1 for TPt, FPt and lobaplatin respectively. The free platinum fraction (FPt/TPt) was initially very high, indicating low protein binding. FPt was essentially present as intact lobaplatin. Four hours after infusion 54 +/- 5% and 24 h after infusion 74 +/- 3% of the lobaplatin dose was excreted in the urine. In conclusion, lobaplatin is a platinum compound with anti-tumour activity in patients with relapsed ovarian cancer, especially in those who have platinum-sensitive tumours. The main toxicity of lobaplatin is thrombocytopenia and its dose should be corrected according to renal function.


British Journal of Cancer | 1999

Phase I and pharmacokinetic study of the topoisomerase II catalytic inhibitor fostriecin

R.S. de Jong; Nh Mulder; Dra Uges; D.Th. Sleijfer; F J P Höppener; Hjm Groen; Phb Willemse; W.T.A. van der Graaf; E.G.E. de Vries

SummaryWe conducted a phase I and pharmacokinetic study of the topoisomerase II catalytic inhibitor fostriecin. Fostriecin was administered intravenously over 60 min on days 1–5 at 4-week intervals. Dose was escalated from 2 mg m–2day–1to 20 mg m–2day–1in 20 patients. Drug pharmacokinetics was analysed with high performance liquid chromatography with UV-detection. Plasma collected during drug administration was tested in vitro for growth inhibition of a teniposide-resistant small-cell lung cancer (SCLC) cell line. The predominant toxicities were elevated liver transaminases (maximum common toxicity criteria (CTC) grade 4) and serum creatinine (maximum CTC grade 2). These showed only a limited increase with increasing doses, often recovered during drug administration and were fully reversible. Duration of elevated alanine–amino transferase (ALT) was dose-limiting in one patient at 20 mg m–2. Other frequent toxicities were grade 1–2 nausea/vomiting, fever and mild fatigue. Mean fostriecin plasma half-life was 0.36 h (initial; 95% CI, 0–0.76 h) and 1.51 h (terminal; 95% CI, 0.41–2.61 h). A metabolite, most probably dephosphorylated fostriecin, was detected in plasma and urine. No tumour responses were observed, but the plasma concentrations reached in the patients were insufficient to induce significant growth inhibition in vitro. The maximum tolerated dose (MTD) has not been reached, because drug supply was stopped at the 20 mg m–2dose level. However, further escalation seems possible and is warranted to achieve potentially effective drug levels. Fostriecin has a short plasma half-life and longer duration of infusion should be considered.


British Journal of Cancer | 1992

Cisplatin and platinum pharmacokinetics during hyperthermic isolated limb perfusion for human tumours of the extremities.

Hj Guchelaar; Hj Hoekstra; Ege Devries; Dra Uges; Jw Oosterhuis; Hs Koops

Cisplatin and platinum pharmacokinetics during hyperthermic isolated limb perfusion for human tumours of the extremities


British Journal of Cancer | 2000

A phase I study of a new polyamine biosynthesis inhibitor, SAM486A, in cancer patients with solid tumours

Robert Paridaens; Dra Uges; N Barbet; Les Choi; M Seeghers; W.T.A. van der Graaf; Hjm Groen; Herlinde Dumez; I Van Buuren; Frits A.J. Muskiet; Renaud Capdeville; A. van Oosterom; E.G.E. de Vries

Because tumour cell proliferation is highly dependent upon up-regulation of de-novo polyamine synthesis, inhibition of the polyamine synthesis pathway represents a potential target for anticancer therapy. SAM486A (CGP 48664) is a new inhibitor of the polyamine biosynthetic enzyme S-adenosylmethionine decarboxylase (SAMDC), more potent and specific than the first-generation SAMDC inhibitor methylglyoxal (bis) guanylhydrazone (MGBG). Preclinical testing confirmed promising antiproliferative activity. In this phase I study, SAM486A was given 4-weekly as a 120 h infusion. 39 adult cancer patients were enrolled with advanced/refractory disease not amenable to established treatments, PS ≤ 2, adequate marrow, liver, renal and cardiac function. Doses were escalated in 100% increments without toxicity in 24 pts from 3 mg m–2 cycle–1 up to 400 mg m–2 cycle–1. At 550 and 700 mg m–2 cycle–1 reversible dose-limiting neutropenia occurred. Other toxicities included mild fatigue, nausea and vomiting. No objective remission was seen. Pharmakokinetic analysis showed a terminal half-life of approximately 2 days. AUC and Cmax were related to dose; neutropenia correlated with AUC. The recommended dose for further phase II studies on this schedule is 400 mg m–2 cycle–1.


British Journal of Cancer | 1997

Randomized comparison of etoposide pharmacokinetics after oral etoposide phosphate and oral etoposide

Rs deJong; Nh Mulder; Dra Uges; S Kaul; B Winograd; Dt Sleijfer; Hjm Groen; Phb Willemse; Wta vanderGraaf; Ege Devries

Etoposide phosphate is a water-soluble prodrug of etoposide. The plasma pharmacokinetics of etoposide following oral administration of etoposide phosphate or oral etoposide were compared. Seventeen patients with solid tumours were enrolled to receive oral etoposide phosphate 125 mg m(-2) on days 1-5 every 3 weeks, with escalation to 175 mg m(-2) from course 3 when possible. Patients were randomized to receive oral etoposide phosphate or oral etoposide on day 1 of course 1 and the alternative compound on day 1 of course 2. Fifteen patients received two or more courses and were evaluable for pharmacokinetic comparisons. The median AUC(inf) (area under the concentration vs time curve from zero to infinity) of etoposide was 77.7 mg l(-1) h after etoposide phosphate (95% CI 61.3-100.5) and 62.0 mg l(-1) h after oral etoposide (95% CI 52.2-76.9). The difference in favour of etoposide phosphate was borderline significant: median 9.9 mg l(-1) h (95% CI 0.1-32.8 mg l(-1) h; P = 0.05). However, the inter-patient variability of etoposide AUC(inf) was not improved (coefficients of variation 42.3% and 48.4%). Etoposide phosphate was undetectable in plasma after oral administration. Toxicities of oral etoposide phosphate were not different from those known for etoposide. In conclusion, oral etoposide phosphate does not offer a clinically relevant benefit over oral etoposide.


Cancer Chemotherapy and Pharmacology | 2001

Pharmacokinetics of MEN-10755, a novel anthracycline disaccharide analogue, in two phase I studies in adults with advanced solid tumours

A M E Bos; E.G.E. de Vries; P. Dombernovsky; Steinar Aamdal; Dra Uges; D. Schrijvers; J. Wanders; M. Roelvink; A. R. Hanauske; S. Bortini; A. Capriati; A. E. G. Crea; J. B. Vermorken

Abstract. The doxorubicin analogue MEN-10755 has been identified as a compound with promising antitumour activity based on structure-activity studies of a new series of anthracycline disaccharides. The high antitumour activity of MEN-10755 in human tumour xenografts, including doxorubicin-resistant xenografts, and its unique pharmacological and biological properties made this novel disaccharide analogue an interesting candidate for clinical evaluation. Two pharmacokinetic phase I studies with different dosing schedules were performed in adults with solid refractory malignancies. The pharmacokinetics of MEN-10755 were studied after a 15-min i.v. infusion given once every 3xa0weeks or once every week for 3xa0weeks followed by 1xa0week rest. Plasma and urine levels of MEN-10755 were measured by HPLC with fluorescent detection. It was possible to combine the pharmacokinetic results of the two studies because there was no accumulation of MEN-10755 before the next infusion of MEN-10755 in the weekly study with 1xa0week rest. The administered dose levels on dayxa01 in this study were all in the lower range from the 3-weekly study. The postinfusion plasma kinetics of MEN-10755 were best described by a triexponential model. The plasma peak levels (Cmax) of MEN-10755 showed a linear relationship with the administered dose. Peak plasma MEN-10755 levels ranged between 474 and 21,587xa0µg/l. The mean elimination half-life (T1/2γ) was 20.7±9.0xa0h. The AUC0–∞ was proportional to the administered dose. The mean plasma clearance of MEN-10755 was 6.0±2.2xa0l/h per m2 with a mean volume of distribution (Vss) of 95.6±43.4xa0l/m2. The mean renal excretion of unchanged drug within 24xa0h was 4.3±1.8%. Compared to epirubicin and doxorubicin, the pharmacokinetics of MEN-10755 were characterized by an approximately twofold shorter terminal half-life, a much lower total plasma clearance and a much smaller volume of distribution.


British Journal of Cancer | 1997

Conversion of the prodrug etoposide phosphate to etoposide in gastric juice and bile.

Rs deJong; Eam Slijfer; Dra Uges; Nh Mulder; Ege Devries

Etoposide phosphate is a water-soluble prodrug of etoposide. It was expected that this prodrug could be used to overcome the solubility limitations and erratic bioavailability of oral etoposide. To investigate the possibility of prodrug conversion to etoposide within the gastrointestinal lumen, etoposide phosphate was dissolved in water and incubated with human gastric juice or human bile in vitro. Samples were collected during 150 min and analysed for etoposide concentration with high-performance liquid chromatography. Conversion of prodrug to etoposide during incubation with gastric juice was negligible. There was significant conversion during incubation with bile at pH 7-8. The percentage of prodrug converted to etoposide at pH 8 after 60 min was 78 +/- 18% (mean +/- S.D.) for a 0.1 mg ml-1 prodrug solution and 36 +/- 26% for 0.5 mg ml-1. At pH 7, after 60 min 22% of prodrug was converted to etoposide when incubated at 0.1 mg ml-1 and 10% at 0.5 mg ml-1. No conversion was found after inactivation of alkaline phosphate (AP) by overnight heating of bile at 65 degrees C or by the addition of disodium edetate to the bile. In conclusion, because of AP in bile, variable conversion of etoposide phosphate to etoposide can be expected within the intestinal lumen after oral administration. This could have important pharmacokinetic consequences.


Pharmacy World & Science | 1988

CONTINUOUS INFUSION OF LOW-DOSE DOXORUBICIN, EPIRUBICIN AND MITOXANTRONE IN CANCER-CHEMOTHERAPY - A REVIEW

Janke Greidanus; Phb Willemse; Dra Uges; Ethgj Oremus; Zj Delangen; Ege Devries

With the recent development of reliable portable pumps and safe venous access systems, continuous infusion of chemotherapeutic agents on an out-patient basis has become feasible. Advantages of continuous infusion are the long-term exposure of tumour cells to the drug and the fact that most toxic effects are reduced for doxorubicin, epirubicin and mitoxantrone due to elimination of the high peak plasma levels. Preliminary data for doxorubicin suggest that its antitumour activity is maintained. Pharmacokinetic studies with epirubicin and mitoxantrone showed a linear relationship between drug dose infused and the steady-state plasma level for these drugs. The area under the curve for leukocytes drug level was higher during continuous infusion than after an equitoxic bolus injection of epirubicin and mitoxantrone. Well-randomized clinical trials will be necessary to investigate the role of continuous infusion of antracyclines and mitoxantrone in cancer chemotherapy in the future.

Collaboration


Dive into the Dra Uges's collaboration.

Top Co-Authors

Avatar

Nh Mulder

University of Groningen

View shared research outputs
Top Co-Authors

Avatar

E.G.E. de Vries

University Medical Center Groningen

View shared research outputs
Top Co-Authors

Avatar

Ege Devries

University of Groningen

View shared research outputs
Top Co-Authors

Avatar

Phb Willemse

University of Groningen

View shared research outputs
Top Co-Authors

Avatar

Dt Sleijfer

University of Groningen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

W.T.A. van der Graaf

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

A M E Bos

University of Groningen

View shared research outputs
Top Co-Authors

Avatar

A. van Oosterom

Radboud University Nijmegen

View shared research outputs
Top Co-Authors

Avatar

Coby Meijer

University Medical Center Groningen

View shared research outputs
Researchain Logo
Decentralizing Knowledge