Network


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

Hotspot


Dive into the research topics where Claire Ward is active.

Publication


Featured researches published by Claire Ward.


Clinical Cancer Research | 2009

Antibody-Mediated Inhibition of Cathepsin S Blocks Colorectal Tumor Invasion and Angiogenesis

Roberta Burden; Julie Gormley; Thomas Jaquin; Donna M. Small; Derek J. Quinn; Shauna Hegarty; Claire Ward; Brian Walker; James A. Johnston; Shane Olwill; Christopher J. Scott

Purpose: Cathepsin S is a cysteine protease that promotes the invasion of tumor and endothelial cells during cancer progression. Here we investigated the potential to target cathepsin S using an antagonistic antibody, Fsn0503, to block these tumorigenic effects. Experimental Design: A panel of monoclonal antibodies was raised to human cathepsin S. The effects of a selected antibody were subsequently determined using invasion and proteolysis assays. Endothelial cell tube formation and aorta sprouting assays were done to examine antiangiogenic effects. In vivo effects were also evaluated using HCT116 xenograft studies. Results: A selected cathepsin S antibody, Fsn0503, significantly blocked invasion of a range of tumor cell lines, most significantly HCT116 colorectal carcinoma cells, through inhibition of extracellular cathepsin S–mediated proteolysis. We subsequently found enhanced expression of cathepsin S in colorectal adenocarcinoma biopsies when compared with normal colon tissue. Moreover, Fsn0503 blocked endothelial cell capillary tube formation and aortic microvascular sprouting. We further showed that administration of Fsn0503 resulted in inhibition of tumor growth and neovascularization of HCT116 xenograft tumors. Conclusions: These results show that blocking the invasive and proangiogenic effects of cathepsin S with antibody inhibitors may have therapeutic utility upon further preclinical and clinical evaluation. (Clin Cancer Res 2009;15(19):6042–51)


Gene Therapy | 2007

p21 (WAF1)-mediated transcriptional targeting of inducible nitric oxide synthase gene therapy sensitizes tumours to fractionated radiotherapy

H. McCarthy; Jenny Worthington; Elaine Barrett; E. Cosimo; Marie Boyd; Robert J. Mairs; Claire Ward; Stephanie R. McKeown; David Hirst; Tracy Robson

Cancer gene therapy that utilizes toxic transgene products requires strict transcriptional targeting to prevent adverse normal tissue effects. We report on the use of a promoter derived from the cyclin dependent kinase inhibitor, p21(WAF1), to control transgene expression. We demonstrate that this promoter is relatively silent in normal cells (L132, FSK, HMEC-1) compared to the almost constitutive expression obtained in tumour cells (DU145, LNCaP, HT29 and MCF-7) of varying p53 status, a characteristic that will be important in gene therapy protocols. In addition, we found that the p21(WAF1) promoter could be further induced by both external beam radiation (up to eight-fold in DU145 cells), intracellular-concentrated radionuclides ([211At]MABG) (up to 3.5-fold in SK-N-BE(2c) cells) and hypoxia (up to four-fold in DU145 cells). We have previously achieved significant radiosensitization of tumour cells both in vitro and in vivo by using inducible nitric oxide synthase (iNOS) gene therapy to generate the potent radiosensitizer, nitric oxide (NO•). Here, we report that a clinically relevant schedule of p21(WAF1)-driven iNOS gene therapy significantly sensitized both p53 wild-type RIF-1 tumours and p53 mutant HT29 tumours to fractionated radiotherapy. Our data highlight the utility of this p21(WAF1)/iNOS-targeted approach.


PLOS ONE | 2010

Antibody targeting of cathepsin S inhibits angiogenesis and synergistically enhances anti-VEGF.

Claire Ward; Diana Kuehn; Roberta Burden; Julie Gormley; Thomas Jaquin; Mihaela Gazdoiu; Donna M. Small; Roy Bicknell; J D Johnston; Christopher J. Scott; Shane Olwill

Background Angiogenesis is a key hallmark of tumourigenesis and its inhibition is a proven strategy for the development of novel anti-cancer therapeutics. An important aspect of early angiogenesis is the co-ordinated migration and invasion of endothelial cells through the hypoxic tumour tissue. Cathepsin S has been shown to play an important role in angiogenesis as has vascular endothelial growth factor (VEGF). We sought to assess the anti-angiogenic effect of Fsn0503, a novel cathepsin S inhibitory antibody, when combined with anti-VEGF on vascular development. Methodology/Principal Findings Cathepsin S expression and secretion from endothelial cells was characterised using RT-PCR and western blotting. We further show that cathepsin S promotes pericellular hydrolysis of extracellular matrix components in the tumour microenvironment and facilitates endothelial invasion. The cathepsin S inhibitory antibody, Fsn0503, blocks extracellular proteolysis, inhibiting endothelial invasion and tube formation in cell-based assays. The anti-angiogenic effects of Fsn0503 were also shown in vivo where it significantly retarded the development of vasculature in human xenograft models. Furthermore, when Fsn0503 was combined with an anti-VEGF antibody, a synergistic inhibition of microvascular development was observed. Conclusions/Significance Taken together, this data demonstrates that the antibody-mediated targeting of cathepsin S represents a novel method of inhibiting angiogenesis. Furthermore, when used in combination with anti-VEGF therapies, Fsn0503 has the potential to significantly enhance current treatments of tumour neovascularisation and may also be of use in the treatment of other conditions associated with inappropriate angiogenesis.


Cancer Gene Therapy | 2006

Tumor-selective drug activation: a GDEPT approach utilizing cytochrome P450 1A1 and AQ4N.

Anita Yakkundi; Verna McErlane; Margaret Murray; H. McCarthy; Claire Ward; Ciara Hughes; Laurence H. Patterson; David Hirst; Stephanie R. McKeown; Tracy Robson

Drug metabolizing transgene products, which activate bioreductive cytotoxins, can be used to target treatment-resistant hypoxic tumors. The prodrug AQ4N is bioreduced in hypoxic cells by cytochrome P450s (CYPs) to the cytotoxin AQ4. Previously we have shown that intra-tumoral injection of CYP3A4 and CYP2B6 transgenes with AQ4N and radiation inhibits tumor growth. Here we examine the ability of other CYPs, in particular CYP1A1, to metabolize AQ4N, and to enhance radiosensitization. Metabolism of AQ4N was assessed using microsomes prepared from baculovirus-infected cells transfected with various CYP isoforms. AQ4N metabolism was most efficient with CYP1A1 (66.7 nmol/min/pmol) and 2B6 (34.4 nmol/min/pmol). Transient transfection of human CYP1A1±CYP reductase (CYPRED) was investigated in hypoxic RIF-1 mouse cells in vitro using the alkaline comet assay. There was a significant increase in DNA damage following transient transfection of CYP1A1 compared to non-transfected cells; inclusion of CYPRED provided no additional effect. In vivo, a single intra-tumoral injection of a CYP1A1 construct in combination with AQ4N (100 mg/kg i.p.) and 20 Gy X-rays caused a 16-day delay in tumor regrowth compared to tumors receiving AQ4N plus radiation and empty vector (P=0.0344). The results show the efficacy of a CYP1A1-mediated GDEPT strategy for bioreduction of AQ4N.


International Journal of Cancer | 2013

Androgen deprivation results in time-dependent hypoxia in LNCaP prostate tumours; informed scheduling of the bioreductive drug AQ4N improves treatment response

Louise Ming; Niall M. Byrne; Sarah Nicole Camac; Christopher A. Mitchell; Claire Ward; David Waugh; Stephanie R. McKeown; Jenny Worthington

Androgen withdrawal induces hypoxia in androgen‐sensitive tissue; this is important as in the tumour microenvironment, hypoxia is known to drive malignant progression. Our study examined the time‐dependent effect of androgen deprivation therapy (ADT) on tumour oxygenation and investigated the role of ADT‐induced hypoxia on malignant progression in prostate tumours. LNCaP xenografted tumours were treated with anti‐androgens and tumour oxygenation measured. Dorsal skin fold (DSF) chambers were used to image tumour vasculature in vivo. Quantitative PCR (QPCR) identified differential gene expression following treatment with bicalutamide. Bicalutamide‐treated and vehicle‐only‐treated tumours were re‐established in vitro, and invasion and sensitivity to docetaxel were measured. Tumour growth delay was calculated following treatment with bicalutamide combined with the bioreductive drug AQ4N. Tumour oxygenation measurements showed a precipitate decrease following initiation of ADT. A clinically relevant dose of bicalutamide (2 mg/kg/day) decreased tumour oxygenation by 45% within 24 hr, reaching a nadir of 0.09% oxygen (0.67 ± 0.06 mmHg) by Day 7; this persisted until Day 14 when it increased up to Day 28. Using DSF chambers, LNCaP tumours treated with bicalutamide showed loss of small vessels at Days 7 and 14 with revascularisation occurring by Day 21. QPCR showed changes in gene expression consistent with the vascular changes and malignant progression. Cells from bicalutamide‐treated tumours were more malignant than vehicle‐treated controls. Combining bicalutamide with AQ4N (50 mg/kg, single dose) caused greater tumour growth delay than bicalutamide alone. Our study shows that bicalutamide‐induced hypoxia selects for cells that show malignant progression; targeting hypoxic cells may provide greater clinical benefit.


Molecular Cancer | 2011

Antibody targeting of Cathepsin S induces antibody-dependent cellular cytotoxicity

Hang Fai Kwok; Richard Buick; Diana Kuehn; Julie Gormley; Declan Doherty; Thomas Jaquin; Angela McClurg; Claire Ward; Teresa Byrne; Jacob Jaworski; Ka Lai Leung; Philip Snoddy; Christine McAnally; Roberta Burden; Breena Gray; Jenny Lowry; Isabelle Sermadiras; Natalia Gruszka; Nigel Courtenay-Luck; Adrien Kissenpfennig; Christopher J. Scott; James A. Johnston; Shane Olwill

BackgroundProteolytic enzymes have been implicated in driving tumor progression by means of their cancer cell microenvironment activity where they promote proliferation, differentiation, apoptosis, migration, and invasion. Therapeutic strategies have focused on attenuating their activity using small molecule inhibitors, but the association of proteases with the cell surface during cancer progression opens up the possibility of targeting these using antibody dependent cellular cytotoxicity (ADCC). Cathepsin S is a lysosomal cysteine protease that promotes the growth and invasion of tumour and endothelial cells during cancer progression. Our analysis of colorectal cancer patient biopsies shows that cathepsin S associates with the cell membrane indicating a potential for ADCC targeting.ResultsHere we report the cell surface characterization of cathepsin S and the development of a humanized antibody (Fsn0503h) with immune effector function and a stable in vivo half-life of 274 hours. Cathepsin S is expressed on the surface of tumor cells representative of colorectal and pancreatic cancer (23%-79% positive expression). Furthermore the binding of Fsn0503h to surface associated cathepsin S results in natural killer (NK) cell targeted tumor killing. In a colorectal cancer model Fsn0503h elicits a 22% cytotoxic effect.ConclusionsThis data highlights the potential to target cell surface associated enzymes, such as cathepsin S, as therapeutic targets using antibodies capable of elicitingADCC in tumor cells.


Clinical Cancer Research | 2008

Evaluation of the Antiangiogenic Potential of AQ4N

Martin O'Rourke; Claire Ward; Jenny Worthington; Julie McKenna; Andrea Valentine; Tracy Robson; David Hirst; Stephanie R. McKeown

Purpose: A number of cytotoxic chemotherapy agents tested at low concentrations show antiangiogenic properties with limited cytotoxicity, e.g., cyclophosphamide, tirapazamine, and mitoxantrone. AQ4N is a bioreductive alkylaminoanthraquinone that is cytotoxic when reduced to AQ4; hence, it can be used to target hypoxic tumor cells. AQ4N is structurally similar to mitoxantrone and was evaluated for antiangiogenic properties without the need for bioreduction. Experimental Design: The effect of AQ4N and fumagillin on human microvascular endothelial cells (HMEC-1) was measured using a variety of in vitro assays, i.e., 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, wound scrape, tubule formation, rat aortic ring, and invasion assays. Low-dose AQ4N (20 mg/kg) was also given in vivo to mice bearing a tumor in a dorsal skin flap. Results: AQ4N (10−11 to 10−5 mol/L) had no effect on HMEC-1 viability. AQ4N (10−9 to 10−5mol/L) caused a sigmoidal dose-dependent inhibition of endothelial cell migration in the wound scrape model. Fumagillin showed a similar response over a lower dose range (10−13 to 10−9 mol/L); however, the maximal inhibition was less (25% versus 43% for AQ4N). AQ4N inhibited HMEC-1 cell contacts on Matrigel (10−8 to 10−5 mol/L), HMEC-1 cell invasion, and sprouting in rat aorta explants. Immunofluorescence staining with tubulin, vimentim, dynein, and phalloidin revealed that AQ4N caused disruption to the cell cytoskeleton. When AQ4N (20 mg/kg) was given in vivo for 5 days, microvessels disappeared in LNCaP tumors grown in a dorsal skin flap. Conclusions: This combination of assays has shown that AQ4N possesses antiangiogenic effects in normoxic conditions, which could potentially contribute to antitumor activity.


PLOS ONE | 2018

Characterization of the naive murine antibody repertoire using unamplified high-throughput sequencing

Trisha A. Rettig; Claire Ward; Bailey A. Bye; Michael J. Pecaut; Stephen K. Chapes

Antibody specificity and diversity are generated through the enzymatic splicing of genomic gene segments within each B cell. Antibodies are heterodimers of heavy- and light-chains encoded on separate loci. We studied the antibody repertoire from pooled, splenic tissue of unimmunized, adult female C57BL/6J mice, using high-throughput sequencing (HTS) without amplification of antibody transcripts. We recovered over 90,000 heavy-chain and over 135,000 light-chain immunoglobulin sequences. Individual V-, D-, and J-gene segment usage was uniform among the three mouse pools, particularly in highly abundant gene segments, with low frequency V-gene segments not being detected in all pools. Despite the similar usage of individual gene segments, the repertoire of individual B-cell CDR3 amino acid sequences in each mouse pool was highly varied, affirming the combinatorial diversity in the B-cell pool that has been previously demonstrated. There also was some skewing in the V-gene segments that were detected depending on chromosomal location. This study presents a unique, non-primer biased glimpse of the conventionally housed, unimmunized antibody repertoire of the C57BL6/J mouse.


Molecular Cancer Therapeutics | 2009

Abstract B17: Fsn0503: A novel cathepsin S‐specific antibody that blocks tumor invasion and angiogenesis

Claire Ward; Roberta Burden; Julie Gormley; Donna M. Small; Thomas Jacquin; Mihaela Gazdoiu; Roy Bicknell; James A. Johnston; Christopher J. Scott; Shane Olwill

AACR Annual Meeting-- Apr 18-22, 2009; Denver, CO Antibody based drugs represent a major class of therapeutics which have contributed greatly to improved prognostic outcome for patients suffering from many types of cancer. The characteristic target specificity, long half life and reduced toxicity make antibodies attractive modalities for the treatment of neoangiogenesis where long term administration is often necessary. Traditionally antibodies have been developed against targets such as membrane receptors or ligands where they evoke an agonistic / antagonistic response. More recently some groups, including ours, have explored their application in targeting biomarkers, present in the tumour microenvironment, which may originate from more than one tumour associated cell type. Cathepsin S (CatS) is a cysteine protease which has been implicated in tumour angiogenesis with KO models showing an attenuation of tumour growth and reduced vessel formation. During tumour development CatS, normally confined to the lysosomes of professional antigen presenting cells, is secreted into the tumour microenvironment where it is involved in extracellular matrix remodelling. We have developed an antibody, Fsn0503, which specifically targets and inhibits extracellular CatS and have demonstrated efficacy in a range of angiogenesis models. The CatS inhibitory antibody significantly impaired microtubule formation in the in vitro HUVEC assay and ex vivo rat aortic ring assay (p<0.01) . The antibody attenuated HCT116 tumour cell invasion by 64% (p: <0.0001), preventing CatS-mediated extracellular matrix hydrolysis and led to a reduction in tumour mass and metastatic burden in preclinical models. Our data indicates that Fsn0503 is an exciting experimental therapeutic which has great clinical potential. Citation Information: In: Proc Am Assoc Cancer Res; 2009 Apr 18-22; Denver, CO. Philadelphia (PA): AACR; 2009. Abstract nr LB-152.


Biochimie | 2012

Inhibition of Cathepsin S by Fsn0503 enhances the efficacy of chemotherapy in colorectal carcinomas

Roberta Burden; Julie Gormley; Diana Kuehn; Claire Ward; Hang Fai Kwok; Mihaela Gazdoiu; Angela McClurg; Thomas Jaquin; James A. Johnston; Christopher J. Scott; Shane Olwill

Collaboration


Dive into the Claire Ward's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roberta Burden

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Donna M. Small

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

David Hirst

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar

Roy Bicknell

University of Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Angela McClurg

Queen's University Belfast

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrea Valentine

Queen's University Belfast

View shared research outputs
Researchain Logo
Decentralizing Knowledge