Network


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

Hotspot


Dive into the research topics where Sian Tomiko Taylor is active.

Publication


Featured researches published by Sian Tomiko Taylor.


Cancer Research | 2005

AZD2171: A Highly Potent, Orally Bioavailable, Vascular Endothelial Growth Factor Receptor-2 Tyrosine Kinase Inhibitor for the Treatment of Cancer

Stephen R. Wedge; Jane Kendrew; Laurent Francois Andre Hennequin; Paula J. Valentine; Simon T. Barry; Sandra R. Brave; Neil R. Smith; Neil H. James; Michael Dukes; Jon Owen Curwen; Rosemary Chester; Janet A. Jackson; Sarah J. Boffey; Lyndsey L. Kilburn; Sharon Barnett; Graham Richmond; Peter F. Wadsworth; Michael D. Walker; Alison L. Bigley; Sian Tomiko Taylor; Lee A. D. Cooper; Sarah Beck; Juliane M. Jürgensmeier; Donald J. Ogilvie

Inhibition of vascular endothelial growth factor-A (VEGF) signaling is a promising therapeutic approach that aims to stabilize the progression of solid malignancies by abrogating tumor-induced angiogenesis. This may be accomplished by inhibiting the kinase activity of VEGF receptor-2 (KDR), which has a key role in mediating VEGF-induced responses. The novel indole-ether quinazoline AZD2171 is a highly potent (IC50 < 1 nmol/L) ATP-competitive inhibitor of recombinant KDR tyrosine kinase in vitro. Concordant with this activity, in human umbilical vein endothelial cells, AZD2171 inhibited VEGF-stimulated proliferation and KDR phosphorylation with IC50 values of 0.4 and 0.5 nmol/L, respectively. In a fibroblast/endothelial cell coculture model of vessel sprouting, AZD2171 also reduced vessel area, length, and branching at subnanomolar concentrations. Once-daily oral administration of AZD2171 ablated experimental (VEGF-induced) angiogenesis in vivo and inhibited endochondral ossification in bone or corpora luteal development in ovary; physiologic processes that are highly dependent upon neovascularization. The growth of established human tumor xenografts (colon, lung, prostate, breast, and ovary) in athymic mice was inhibited dose-dependently by AZD2171, with chronic administration of 1.5 mg per kg per day producing statistically significant inhibition in all models. A histologic analysis of Calu-6 lung tumors treated with AZD2171 revealed a reduction in microvessel density within 52 hours that became progressively greater with the duration of treatment. These changes are indicative of vascular regression within tumors. Collectively, the data obtained with AZD2171 are consistent with potent inhibition of VEGF signaling, angiogenesis, neovascular survival, and tumor growth. AZD2171 is being developed clinically as a once-daily oral therapy for the treatment of cancer.


Molecular Cancer Therapeutics | 2011

Assessing the Activity of Cediranib, a VEGFR-2/-3 tyrosine kinase inhibitor, against VEGFR-1 and members of the structurally related PDGFR-family

Sandra R. Brave; Kirsty Ratcliffe; Zena Wilson; Neil H. James; Susan Ashton; Anna Wainwright; Jane Kendrew; Philippa Dudley; Nicola Broadbent; Graham Sproat; Sian Tomiko Taylor; Claire Barnes; Charles Farnsworth; Laurent Francois Andre Hennequin; Donald J. Ogilvie; Juliane M. Jürgensmeier; Stephen R. Wedge; Simon T. Barry

Cediranib is a potent inhibitor of the VEGF receptor (VEGFR)-2 and VEGFR-3 tyrosine kinases. This study assessed the activity of cediranib against the VEGFR-1 tyrosine kinase and the platelet-derived growth factor receptor (PDGFR)-associated kinases c-Kit, PDGFR-α, and PDGFR-β. Cediranib inhibited VEGF-A–stimulated VEGFR-1 activation in AG1-G1-Flt1 cells (IC50 = 1.2 nmol/L). VEGF-A induced greatest phosphorylation of VEGFR-1 at tyrosine residues Y1048 and Y1053; this was reversed by cediranib. Potency against VEGFR-1 was comparable with that previously observed versus VEGFR-2 and VEGFR-3. Cediranib also showed significant activity against wild-type c-Kit in cellular phosphorylation assays (IC50 = 1–3 nmol/L) and in a stem cell factor–induced proliferation assay (IC50 = 13 nmol/L). Furthermore, phosphorylation of wild-type c-Kit in NCI-H526 tumor xenografts was reduced markedly following oral administration of cediranib (≥1.5 mg/kg/d) to tumor-bearing nude mice. The activity of cediranib against PDGFR-β and PDGFR-α was studied in tumor cell lines, vascular smooth muscle cells (VSMC), and a fibroblast line using PDGF-AA and PDGF-BB ligands. Both receptor phosphorylation (IC50 = 12–32 nmol/L) and PDGF-BB–stimulated cellular proliferation (IC50 = 32 nmol/L in human VSMCs; 64 nmol/L in osteosarcoma cells) were inhibited. In vivo, ligand-induced PDGFR-β phosphorylation in murine lung tissue was inhibited by 55% following treatment with cediranib at 6 mg/kg but not at 3 mg/kg or less. In contrast, in C6 rat glial tumor xenografts in mice, ligand-induced phosphorylation of both PDGFR-α and PDGFR-β was reduced by 46% to 61% with 0.75 mg/kg cediranib. Additional selectivity was showed versus Flt-3, CSF-1R, EGFR, FGFR1, and FGFR4. Collectively, these data indicate that cediranib is a potent pan-VEGFR kinase inhibitor with similar activity against c-Kit but is significantly less potent than PDGFR-α and PDGFR-β. Mol Cancer Ther; 10(5); 861–73. ©2011 AACR.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of AZD2932, a new Quinazoline Ether Inhibitor with high affinity for VEGFR-2 and PDGFR tyrosine kinases.

Patrick Ple; Frederic Henri Jung; Sue Ashton; Laurent Francois Andre Hennequin; Romuald Laine; Rémy Morgentin; Georges Pasquet; Sian Tomiko Taylor

A new series of Quinazoline Ether Inhibitor which potently inhibits VEGFR-2 and PDGFR tyrosine kinases is described here. In vitro, pharmacokinetics and in vivo evaluations led to the selection of AZD2932.


Bioorganic & Medicinal Chemistry Letters | 2012

Discovery of new quinoline ether inhibitors with high affinity and selectivity for PDGFR tyrosine kinases.

Patrick Ple; Frederic Henri Jung; Sue Ashton; Laurent Francois Andre Hennequin; Romuald Laine; Christine Lambert-van der Brempt; Rémy Morgentin; Georges Pasquet; Sian Tomiko Taylor

A new series of quinoline ether inhibitors, which potently and selectively inhibit PDGFR tyrosine kinases, is described in this Letter. Compounds 23 and 33 are selective, low nanomolar inhibitors of PDGFRα and β, display good pharmacokinetics in rat and dog and are active in vivo at low doses when given orally twice daily. Further evaluation of these compounds is warranted.


Archive | 2003

N-(-3-methoxy-5-methylpyrazin-2-yl)-2-(4-'1,3,4-oxadiazol-2-yl!phenyl)pyridine-3 sulphonamide as an anticancer agent

David William Tonge; Sian Tomiko Taylor; Francis Thomas Boyle; Andrew Hughes; Donna Johnstone; Marianne Ashford; Nigel Charles Barrass


Archive | 2009

Use of n-(3-methoxy-5-methylpyrazin-2-yl)-2-(4-[1,3,4-oxadiazol-2-yl]phenyl)pyridine-3-sulphonamide in the treatment of cancer

Marianne Ashford; Nigel Charles Barrass; Francis Thomas Boyle; Andrew Hughes; Donna Johnstone; Sian Tomiko Taylor; David William Tonge


Archive | 2003

Therapeutische behandlung Therapeutic treatment

Francis Thomas AstraZeneca Macclesfield Boyle; Jon Owen Curwen; Neil Gallagher; Ursula Joy AstraZeneca Macclesfield Hancox; Andrew Hughes; Donna Johnstone; Sian Tomiko Taylor; David William Tonge


Archive | 2003

Use of N- (3-methoxy-5-methylpyrazin-2-yl) -2- (4-ä1,3,4-oxadiazol-2-ylüphenyl) pyridine-3-sulphonamide in tumor therapy

David William Tonge; Sian Tomiko Taylor; Francis Thomas Boyle; Andrew Hughes; Donna Johnstone; Marianne Ashford; Nigel Charles Barrass


Archive | 2003

N-(3-methoxy-5-methylpyrazin-2-yl)-2-(4-[1,3,4-oxadiazol-2-yl]phenyl)pyridine-3 sulfonamide as analgetic agent

Marianne Ashford; Nigel Charles Barrass; Francis Thomas Boyle; Andrew Hughes; Donna Johnstone; Sian Tomiko Taylor; David William Tonge


Archive | 2003

Verwendung von n-(3-methoxy-5-methylpyrazin-2-yl)-2-(4-ä1,3,4-oxadiazol-2-ylüphenyl)pyridin-3-sulphonamid in der tumortherapie Use of N- (3-methoxy-5-methylpyrazin-2-yl) -2- (4-ä1,3,4-oxadiazol-2-ylüphenyl) pyridine-3-sulphonamide in tumor therapy

David William Tonge; Sian Tomiko Taylor; Francis Thomas Boyle; Andrew Hughes; Donna Johnstone; Marianne Ashford; Nigel Charles Barrass

Collaboration


Dive into the Sian Tomiko Taylor's collaboration.

Researchain Logo
Decentralizing Knowledge