Network


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

Hotspot


Dive into the research topics where Michael Burgess is active.

Publication


Featured researches published by Michael Burgess.


Clinical Cancer Research | 2004

A Phase I Trial of a Potent P-Glycoprotein Inhibitor, Zosuquidar.3HCl Trihydrochloride (LY335979), Administered Orally in Combination with Doxorubicin in Patients with Advanced Malignancies

Eric H. Rubin; Dinesh P. de Alwis; Isabelle Pouliquen; Lisa J. Green; Phil Marder; Yong Lin; Rita Musanti; Stephanie Grospe; Sharon Smith; Deborah Toppmeyer; Judy Much; Michael Kane; Ajai K. Chaudhary; Christopher Jordan; Michael Burgess; Christopher A. Slapak

Purpose: Our intention was to (a) to investigate the safety and tolerability of a potent P-glycoprotein modulator, zosuquidar trihydrochloride (LY335979), when administered i.v. alone or in combination with doxorubicin, (b) to determine the pharmacokinetics of zosuquidar and correlate exposure to inhibition of P-glycoprotein function in a surrogate assay, and (c) to compare the pharmacokinetics of doxorubicin in the presence and absence of zosuquidar. Patients and Methods: Patients with advanced malignancies who provided written informed consent received zosuquidar and doxorubicin administered separately during the first cycle of therapy and then concurrently in subsequent cycles. Zosuquidar was given i.v. over 48 h in a cohort-dose escalation manner until the occurrence of dose-limiting toxicity or protocol specified maximum exposure. Doxorubicin doses of 45, 60, 75 mg/m2 were administered during the course of the trial. Results: Dose escalation proceeded through 9 cohorts with a total of 40 patients. The maximal doses administered were 640 mg/m2 of zosuquidar and 75 mg/m2 of doxorubicin. No dose-limiting toxicity of zosuquidar was observed. Pharmacokinetic analysis revealed that, in the presence of zosuquidar at doses that exceeded 500 mg, there was a modest decrease in clearance (17–22%) and modest increase in area under the curve (15–25%) of doxorubicin. This change was associated with an enhanced leukopenia and thrombocytopenia but was without demonstrable clinical significance. The higher doses of zosuquidar were associated with maximal P-glycoprotein inhibition in natural killer cells. Conclusion: Zosuquidar can be safely coadministered with doxorubicin using a 48 h i.v. dosing schedule.


Advanced Drug Delivery Reviews | 2003

Considerations in the design and development of transport inhibitors as adjuncts to drug therapy.

Anne H. Dantzig; Dinesh P. de Alwis; Michael Burgess

With the realization of the importance of drug efflux transporters in disease processes and treatment, development of inhibitors to these transporters has been sought for use as adjuncts to therapy. To date, inhibitors that have been best studied are modulators of P-glycoprotein, a transporter important in the removal of anticancer agents from cells and overexpression of this transporter results in multidrug resistance. There is a delicate balance between efficacy and toxicity. This review summarizes key learning points in the development of P-glycoprotein inhibitors. Topics covered include specificity of the inhibitor for the target transporter, effect on metabolism of coadministered drugs, pharmacokinetic interactions, toxicity and the salient features needed for efficacy. These points will have general application to the development of inhibitors of transporters.


Clinical Cancer Research | 2010

Tumor Survivin Is Downregulated by the Antisense Oligonucleotide LY2181308: A Proof-of-Concept, First-in-Human Dose Study

Denis C. Talbot; Malcolm R Ranson; Joanna Davies; Michael Lahn; Sophie Callies; Valérie André; Sunil Kadam; Michael Burgess; Christopher A. Slapak; Anna Olsen; Peter J. McHugh; Johann S. de Bono; Julian C. Matthews; Azeem Saleem; Patricia M Price

Purpose: Enhanced tumor cell survival through expression of inhibitors of apoptosis (IAP) is a hallmark of cancer. Survivin, an IAP absent from most normal tissues, is overexpressed in many malignancies and associated with a poorer prognosis. We report the first-in-human dose study of LY2181308, a second-generation antisense oligonucleotide (ASO) directed against survivin mRNA. Patients and Methods: A dose-escalation study evaluating the safety, pharmacokinetics, and pharmacodynamics of LY2181308 administered intravenously for 3 hours as a loading dose on 3 consecutive days and followed by weekly maintenance doses. Patients were eligible after signing informed consent, had exhausted approved anticancer therapies and agreed to undergo pre- and posttreatment tumor biopsies to evaluate reduction of survivin protein and gene expression. Results: A total of 40 patients were treated with LY2181308 at doses of 100 to 1,000 mg. Twenty-six patients were evaluated at the recommended phase 2 dose of 750 mg, at which level serial tumor sampling and [11C]LY2183108 PET (positron emission tomography) imaging demonstrated that ASO accumulated within tumor tissue, reduced survivin gene and protein expression by 20% and restored apoptotic signaling in tumor cells in vivo. Pharmacokinetics were consistent with preclinical modeling, exhibiting rapid tissue distribution, and terminal half-life of 31 days. Conclusions: The tumor-specific, molecularly targeted effects demonstrated by this ASO in man underpin confirmatory studies evaluating its therapeutic efficacy in cancer.


Clinical Cancer Research | 2004

Phase I Study of Docetaxel in Combination with the P-Glycoprotein Inhibitor, Zosuquidar, in Resistant Malignancies

Paula M. Fracasso; Lori J. Goldstein; Dinesh P. de Alwis; Janet S. Rader; Matthew A. Arquette; Sherry A. Goodner; Lisa P. Wright; Carole L. Fears; Robert J. Gazak; Valérie André; Michael Burgess; Christopher A. Slapak; Jan H. M. Schellens

Purpose: To determine the maximum tolerated dose, dose-limiting toxicity, and pharmacokinetics of docetaxel infused over 1 hour when given in combination with oral zosuquidar to patients with resistant solid tumors. Experimental Design: In cycle 1, patients received docetaxel alone. In subsequent cycles, zosuquidar was administered with docetaxel, which was escalated from 75 to 100 mg/m2. Zosuquidar was escalated from 100 to 300 mg/m2 every 8 hours on days 1 to 3 for a total of 7 doses, or from 400 to 500 mg every 12 hours for 2 doses administered 2 hours before docetaxel. The pharmacokinetics of docetaxel with and without zosuquidar administration were obtained. Results: Thirty-six of 41 patients completed at least one cycle of docetaxel and zosuquidar. The maximum tolerated dose was docetaxel 100 mg/m2 and zosuquidar 500 mg every 12 hours for 2 doses. The most common toxicity was neutropenia. In 35 patients, zosuquidar produced minimal increases in the docetaxel peak plasma concentrations and area under the curve. Dosing over 3 days with zosuquidar (7 doses) did not show benefit over the 1-day dosing. Of the 36 patients, one patient had a partial response, and 14 patients had disease stabilization. Conclusions: Docetaxel at 75 or 100 mg/m2 and zosuquidar 500 mg 2 hours before docetaxel and 12 hours later is well tolerated. Zosuquidar minimally alters the pharmacokinetics of docetaxel, allowing full dose docetaxel to be given with this P-glycoprotein modulator. A Phase II study with this combination in advanced breast carcinoma is underway.


British Journal of Clinical Pharmacology | 2011

Integrated analysis of preclinical data to support the design of the first in man study of LY2181308, a second generation antisense oligonucleotide.

Sophie Callies; Valérie André; Bharvin Patel; David Waters; Paul C Francis; Michael Burgess; Michael Lahn

AIMS To predict the concentration and target inhibition profiles of the survivin inhibitor antisense oligonucleotide LY2181308 in humans. METHODS An indirect pharmacokinetic/pharmacodynamic (PK/PD) model was built to predict the inhibition of survivin mRNA and protein in humans following LY2181308 dosing. Plasma and tissue PK data from cynomolgus monkeys were analyzed by non-linear mixed effect modelling techniques. Human PK parameters were predicted using allometric scaling. Assumptions about the pharmacodynamic parameters were made based upon the target and tumour growth inhibition data from mouse xenograft models. This enabled the prediction of the clinical PK/PD profiles. RESULTS Following a 750 mg dose, LY2181308 tumour concentrations ranging from 18.8 to 54µgg(-1) were predicted to lead to 50 to 90% target inhibition. In humans, LY2181308 tumour concentrations fro 13.9 to 52.8µgg(-1) (n=4, LY2181308 750mg) were observed associated with a median survivin mRNA and protein inhibition of 20%±34 (SD) (n=9) and 23%±63 (SD) (n=10), respectively. The human PK parameters were adequately estimated: central V(d) , 4.09 l (90% CI, 3.6, 4.95), distribution clearances, 2.54 (2.36, 2.71), 0.0608 (0.033, 0.6) and 1.67 (1.07, 2.00)lh(-1) , peripheral V(d) s, 25 900 (19 070, 37 200), 0.936 (0.745, 2.07) and 2.51 (1.01, 2.922)l, mean elimination clearance 23.1lh(-1) (5.6, 33.4) and mean terminal half-life, 32.7 days (range 22-52 days). CONCLUSION The model reasonably predicted LY2181308 PK in humans. Overall, the integration of preclinical PK/PD data enabled to appropriately predict dose and dosing regimen of LY2181308 in humans with pharmacologically relevant survivin inhibition achieved at 750mg.


Haematologica | 2004

Clinical effects and P-glycoprotein inhibition in patients with acute myeloid leukemia treated with zosuquidar trihydrochloride, daunorubicin and cytarabine

Gareth Gerrard; Em Payne; R Baker; Dt Jones; Michael N. Potter; Hg Prentice; M Ethell; H McCullough; Michael Burgess; Atul Mehta; K Ganeshaguru


Cancer Chemotherapy and Pharmacology | 2003

A population pharmacokinetic model for doxorubicin and doxorubicinol in the presence of a novel MDR modulator, zosuquidar trihydrochloride (LY335979)

Sophie Callies; Dinesh P. de Alwis; James G. Wright; Alan Sandler; Michael Burgess; Leon Aarons


Cancer Chemotherapy and Pharmacology | 2004

Population pharmacokinetic model for daunorubicin and daunorubicinol coadministered with zosuquidar.3HCl (LY335979)

Sophie Callies; Dinesh P. de Alwis; Atul Mehta; Michael Burgess; Leon Aarons


British Journal of Clinical Pharmacology | 2003

A population pharmacokinetic model for paclitaxel in the presence of a novel P-gp modulator, Zosuquidar Trihydrochloride (LY335979)

Sophie Callies; Dinesh P. de Alwis; Adrian Harris; Paul A. Vasey; Jos H. Beijnen; Jan H. M. Schellens; Michael Burgess; Leon Aarons


Blood | 2004

Glucosylceramide Synthetase Inhibitors Sensitise B-CLL Cells to Cytotoxic Agents without Reversing P-gp Functional Activity.

G Gerrard; Terry D. Butters; Atul Mehta; A. V. Hoffbrand; Derralynn Hughes; Michael Burgess; K Ganeshaguru

Collaboration


Dive into the Michael Burgess's collaboration.

Top Co-Authors

Avatar

Atul Mehta

Royal Free London NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael N. Potter

The Royal Marsden NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Leon Aarons

University of Manchester

View shared research outputs
Top Co-Authors

Avatar

R Baker

University College London

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge