Network


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

Hotspot


Dive into the research topics where Paula Wells is active.

Publication


Featured researches published by Paula Wells.


Cancer Chemotherapy and Pharmacology | 1998

Pharmacokinetic assessment of novel anti-cancer drugs using spectral analysis and positron emission tomography: A feasibility study

Steven R. Meikle; Julian C. Matthews; Cathryn S. Brock; Paula Wells; Robert J.A. Harte; Vincent J. Cunningham; Terry Jones; Patricia M Price

Purpose: The aim of this study was to investigate the feasibility of evaluating the pharmacokinetics of radiolabeled anti-cancer drugs using spectral analysis, a non-compartmental tracer kinetic modeling technique, and positron emission tomography (PET). Methods: Dynamic PET studies were performed on patients receiving tracer doses of 5-fluorouracil (5-[18F]-FU) and two developmental drugs – [11C]-temozolomide and [11C]-acridine carboxamide. Spectral analysis was then used to (a) determine individual and group average pharmacokinetics, (b) predict tumour handling in response to different drug administration regimens, and (c) produce functional parametric images describing regional pharmacokinetics. Results: Spectral analysis could distinguish tumour kinetics from normal tissue kinetics in an individual [11C]-temozolomide study and demonstrated a markedly greater volume of distribution (VD) in glioma than in normal brain, although there was no appreciable difference in mean residence time. Analysis of pooled acridine carboxamide data (n=22) revealed a relatively large VD (and prolonged retention) in the liver and spleen and a markedly lower VD (and initial uptake) in the brain. Continuous infusion of 5-[18F]-FU was predicted to achieve a concentration in colorectal metastases in liver approximately 10 times that achieved in plasma at 10 h after commencement of the infusion. Conclusions: We conclude that spectral analysis provides important pharmacokinetic information about radiolabeled anti-cancer drugs with relatively few model assumptions.


British Journal of Cancer | 2003

Measurement of renal tumour and normal tissue perfusion using positron emission tomography in a phase II clinical trial of razoxane.

H Anderson; Jeffrey T. Yap; Paula Wells; M P Miller; D Propper; Patricia M Price; Adrian L. Harris

Measurement of tumour and normal tissue perfusion in vivo in cancer patients will aid the clinical development of antiangiogenic and antivascular agents. We investigated the potential antiangiogenic effects of the drug razoxane by measuring the changes in parameters estimated from H215O and C15O positron emission tomography (PET) to indicate alterations in vascular physiology. The study comprised 12 patients with primary or metastatic renal tumours >3 cm in diameter enrolled in a Phase II clinical trial of oral razoxane. Perfusion, fractional volume of distribution of water (VD) and blood volume (BV) were measured in tumour and normal tissue before and 4–8 weeks after treatment with 125 mg twice-daily razoxane. Renal tumour perfusion was variable but lower than normal tissue: mean 0.87 ml min−1 ml−1 (range 0.33–1.67) compared to renal parenchyma: mean 1.65 ml min−1 ml−1 (range 1.16–2.88). In eight patients, where parallel measurements were made during the same scan session, renal tumour perfusion was significantly lower than in normal kidney (P=0.0027). There was no statistically significant relationship between pretreatment perfusion and tumour size (r=0.32, n=13). In six patients scanned before and after razoxane administration, there was no statistically significant change in tumour perfusion: mean perfusion pretreatment was 0.81 ml min−1 ml−1 (range 0.46–1.26) and perfusion post-treatment was 0.72 ml min−1 ml−1 (range 0.51–1.15, P=0.15). Tumour VD and BV did not change significantly following treatment: mean pretreatment VD=0.66 (range 0.50–0.87), post-treatment VD=0.71 (range 0.63–0.82, P=0.22); pretreatment BV=0.18 ml ml−1 (range 0.10–0.25), post-treatment BV=0.167 ml ml−1 (range 0.091–0.24, P=0.55). Tumour perfusion, VD and BV did not change significantly with tumour progression. This study has shown that H215O and C15O PET provide useful in vivo physiological measurements, that even highly angiogenic renal cancers have poor perfusion compared to surrounding normal tissue, and that PET can provide valuable information on the in vivo biology of angiogenesis in man and can assess the effects of antiangiogenic therapy.


Archive | 1995

The Use of Radiolabelled Anticancer Drugs in Phase I/II Clinical Trials and the Assessment of Therapeutic Efficacy of New Agents Using PET

Pat Price; Robert J.A. Harte; Owen Tilsley; Julian Matthews; Frank Brady; Sajinder K. Luthra; Safiye Osman; Robert Babarovic; Gavin Brown; Susan O’Reilly; Cathryn Brock; Paula Wells; Terry Jones

PET Oncology is emerging as a new investigational area for cancer research. We present a strategy based on in vivo tracer kinetics of radiolabelled compounds which may be developed to investigate, for the first time, the in vivo molecular interactions, pathways and mechanisms of action of current novel anti-cancer strategies. We outline how PET studies can parallel Phase I & II clinical trials, and could provide new information on tumour and normal tissue pharmacokinetics and functional response. This approach promises to radically assist translation of scientific developments into clinical advances.


Cancer ImagingLung and Breast Carcinomas | 2008

Tumor Proliferation: 2-[11C]-Thymidine Positron Emission Tomography

Paula Wells; Patricia M Price

Publisher Summary [11C]-labeled-thymidine is the first radiotracer used for noninvasive imaging of tumor proliferation and one of the most appropriate pharmacodynamic tools to assess treatment response and assist early validation of novel anticancer agents, because its uptake is directly related to DNA synthesis and correlates with proliferation in human tumors. As a vital component for DNA synthesis, thymidine is essential for cell proliferation and is therefore also a key molecule for measuring tumor proliferation and an important target for cancer therapy. This methodology has been applied to the development of positron-emitting tracers that can provide a measure of cell proliferation in vivo using PET. While the identification of new therapeutic targets is important, the development of more accurate and relevant methods of measuring response to treatment is also vital, especially for the early assessment of new agents. Cancer treatment-induced changes in DNA cell proliferation are more pronounced than changes in glucose metabolism, further supporting the need for a direct marker to image and measure cell proliferation. In contrast, radiolabeled thymidine is incorporated into DNA, and its uptake is a direct measure of cell proliferation rate. Uptake of 2-[11C]-thymidine is related to DNA synthesis and, in comparison with 18F-FDG, has been shown to be a more sensitive discriminator of early clinical response in small cell lung cancer and sarcoma. But the short half-life of 11C and its rapid metabolism in vivo have inhibited its widespread clinical use.


Drug Information Journal | 1997

The Potential of Tracer Kinetic Studies in Drug Development Programs: A New Investigational Area for Cancer Research

Pat Price; Robert J.A. Harte; Paula Wells; C. S. Brock; Julian Matthews; S. Meikle; Colin Steel; J. Luthra; Safiye Osman; Gavin Brown; Frank Brady; C. Prenant; Terry Jones

This paper covers a unique program created in the United Kingdom between the MRC Cyclotron Unit and the CRC Phase I and II drug development committee to investigate the potential of tracer kinetic studies to investigate in vivo tumor and normal tissue kinetics, molecular interactions, pathways, and mechanism of action of novel anti-cancer agents. Positron emission tomography technology offers great potential in streamlining anti-cancer drug development programs, however, significant investment in new methodology and multidisciplinary teams is necessary.


Cancer Research | 2002

Assessment of Proliferation in Vivo Using 2-[11C]Thymidine Positron Emission Tomography in Advanced Intra-abdominal Malignancies

Paula Wells; Roger N. Gunn; Malcolm R. Alison; Colin Steel; Mathew Golding; Alex S. Ranicar; Frank Brady; Safiye Osman; Terry Jones; Patricia M Price


Journal of the National Cancer Institute | 2003

2-[11C]Thymidine Positron Emission Tomography as an Indicator of Thymidylate Synthase Inhibition in Patients Treated With AG337

Paula Wells; Eric O. Aboagye; Roger N. Gunn; Safiye Osman; Alan V. Boddy; Gordon A. Taylor; Imran Rafi; Andrew N. Hughes; A. Hilary Calvert; Patricia M Price; David R. Newell


The Journal of Nuclear Medicine | 2000

A General Method to Correct PET Data for Tissue Metabolites Using a Dual-Scan Approach

Roger N. Gunn; Jeffrey T. Yap; Paula Wells; Safiye Osman; Pat Price; Terry Jones; Vincent J. Cunningham


Clinical Cancer Research | 2003

Assessment of Inter- and Intrapatient Variability in C15O2 Positron Emission Tomography Measurements of Blood Flow in Patients with Intra-abdominal Cancers

Paula Wells; Terry Jones; Patricia M Price


Biochimica et Biophysica Acta | 2004

Measuring tumor pharmacodynamic response using PET proliferation probes: the case for 2-[(11)C]-thymidine.

Paula Wells; Catharine M L West; Terry Jones; Adrian L. Harris; Patricia M Price

Collaboration


Dive into the Paula Wells's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Terry Jones

California Polytechnic State University

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
Top Co-Authors

Avatar

Terry Jones

California Polytechnic State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge