Catherine Harper-Wynne
Maidstone and Tunbridge Wells NHS Trust
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Catherine Harper-Wynne.
Cancer Research | 2015
Andrew Tutt; P. Ellis; Lucy Kilburn; Cheryl Gilett; Sarah Pinder; Jacinta Abraham; Sophie Barrett; Peter Barrett-Lee; Stephen Chan; Maggie Cheang; Mitch Dowsett; Lisa Fox; Patrycja Gazinska; Anita Grigoriadis; Alexander Gutin; Catherine Harper-Wynne; M.Q. Hatton; Sarah Kernaghan; Jerry S. Lanchbury; James Morden; Julie Owen; Jyoti Parikh; Peter J. Parker; Nazneen Rahman; Rebecca Roylance; Adam Shaw; Ian E. Smith; Rose Thompson; Kirsten Timms; Holly Tovey
Introduction: Subgroups within sporadic triple negative breast cancers (TNBCs) appear to share impaired DNA damage response mechanisms with BRCA1/2 mutation-associated breast cancers. This has been hypothesised to confer particular sensitivity to DNA-damaging platinum chemotherapy. The TNT trial, a randomized phase III trial in women with metastatic or recurrent locally advanced TNBC or BRCA1/2 mutation-associated breast cancer, aimed to test this hypothesis and examine treatment effect in biological subgroups. Patients & Methods: Eligible patients had either ER-, PR-, HER2- breast cancer or were known BRCA1/2 carriers (any ER/PR/HER2). Patients were randomized (1:1) to receive either C (AUC 6 q3wk) or D (100mg/m2 q3wk) for 6-8 cycles or until disease progression if sooner and could cross over to the alternative treatment on confirmed progression. Ineligible patients included those who had ECOG performance status >2, received adjuvant taxane therapy in the last 12 mths, any previous treatment with a platinum chemotherapy, or previous non-anthracycline chemotherapy for metastatic disease. For consenting patients a blood sample and archived tissue samples were obtained for BRCA1/2 genotyping and central biomarker analysis (primary tumour, lymph nodes and recurrent tumour biopsy if available) of subtypes within TNBC and biomarkers of DNA repair deficiency. The primary endpoint was RECIST objective tumour response up to cycle 6 of randomised treatment. Secondary endpoints included toxicity, progression free survival (PFS), time to progression and overall survival. TNT aimed to detect a 15% improvement in ORR with C compared to D, with planned target sample size range of 370-450 depending on assumed ORR in D patients (2-sided α=0.05, power=90%). 376 (188 C, 188 D) were recruited from 74 UK centres between Apr 08 and Mar 14. Results: A snapshot of the data was taken on 30/5/14 at which point 336 (89.4%) patients had experienced a PFS event, with overall median PFS time of 4.4 mths. Median age of patients was 55 yrs (IQR 48-63). 366/376 (97%) patients had TNBC of whom 18 were also known BRCA1/2 mutation carriers, with the remaining 10 patients receptor +ve and BRCA1/2 carriers. 338/376 (90%) had metastatic and 38/376 (10%) recurrent locally advanced disease. 53% had liver or lung metastases affecting the parenchyma and 34% had received previous adjuvant taxane therapy. Median time from initial diagnosis to entering TNT was 2.2 yrs (IQR 1.5-3.5). Primary tumour tissue has currently been received for 277 patients, blood from 286 patients and recurrent tumour tissue from 85 patients. Discussion: TNT will report evidence on the activity of single agent platinum chemotherapy compared with single agent taxane in patients with TNBC and BRCA1/2 associated breast cancer. Correlative analyses of BRCA1/2 mutation status, subtypes and DNA repair biomarkers will also be reported. TNT will be the first randomised trial to report the activity of platinum compared with standard chemotherapy within TNBC subtypes and in relation to BRCA1/2 mutation status and DNA repair biomarkers. Safety, tolerability and response to crossover treatment will also be presented. Citation Format: Andrew Tutt, Paul Ellis, Lucy Kilburn, Cheryl Gilett, Sarah Pinder, Jacinta Abraham, Sophie Barrett, Peter Barrett-Lee, Stephen Chan, Maggie Cheang, Mitch Dowsett, Lisa Fox, Patrycja Gazinska, Anita Grigoriadis, Alexander Gutin, Catherine Harper-Wynne, Matthew Hatton, Sarah Kernaghan, Jerry Lanchbury, James Morden, Julie Owen, Jyoti Parikh, Peter Parker, Nazneen Rahman, Rebecca Roylance, Adam Shaw, Ian Smith, Rose Thompson, Kirsten Timms, Holly Tovey, Andrew Wardley, Gregory Wilson, Mark Harries, Judith Bliss. The TNT trial: A randomized phase III trial of carboplatin (C) compared with docetaxel (D) for patients with metastatic or recurrent locally advanced triple negative or BRCA1/2 breast cancer (CRUK/07/012) [abstract]. In: Proceedings of the Thirty-Seventh Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2014 Dec 9-13; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2015;75(9 Suppl):Abstract nr S3-01.
Clinical Cancer Research | 2015
Alexandra Leary; Abigail Evans; Stephen R. D. Johnston; Roger A'Hern; Judith M. Bliss; Rashmita Sahoo; Simone Detre; Benjamin P. Haynes; Margaret Hills; Catherine Harper-Wynne; N.J. Bundred; Gill Coombes; Ian E. Smith; Mitch Dowsett
Purpose: Not all breast cancers respond to lapatinib. A change in Ki67 after short-term exposure may elucidate a biomarker profile for responsive versus nonresponsive tumors. Experimental Design: Women with primary breast cancer were randomized (3:1) to 10 to 14 days of preoperative lapatinib or placebo in a multicenter phase II trial (ISRCTN68509377). Biopsies pre-/posttreatment were analyzed for Ki67, apoptosis, HER2, EGFR, ER, PgR, pAKT, pERK, and stathmin by IHC. Further markers were measured by RT-PCR. Primary endpoint was change in Ki67. HER2+ was defined as 2+/3+ by IHC and FISH+. Results: One hundred twenty-one patients (lapatinib, 94; placebo, 27) were randomized; of these, 21% were HER2+, 78% were HER2− nonamplified, 26% were EGFR+. Paired samples containing tumor were obtained for 98% (118 of 121). Ki67 fell significantly with lapatinib (−31%; P < 0.001), but not with placebo (−3%). Whereas Ki67 reduction with lapatinib was greatest in HER2+ breast cancer (−46%; P = 0.003), there was a significant Ki67 decrease in HER2− breast cancer (−27%; P = 0.017) with 14% of HER2− breast cancer demonstrating ≥50% Ki67 reduction with lapatinib. Among HER2+ patients, the only biomarker predictive of Ki67 response was the EGFR/HER4 ligand epiregulin (EREG) (rho = −0.7; P = 0.002). Among HER2− tumors, only HER3 mRNA levels were significantly associated with Ki67 response on multivariate analysis (P = 0.01). In HER2− breast cancer, HER2 and HER3 mRNA levels were highly correlated (rho = 0.67, P < 0.001), with all Ki67 responders having elevated HER3 and HER2 expression. Conclusions: Lapatinib has antiproliferative effects in a subgroup of HER2− nonamplified tumors characterized by high HER3 expression. The possible role of high HER2:HER3 heterodimers in predicting response to lapatinib merits investigation in HER2− tumors. Clin Cancer Res; 21(13); 2932–40. ©2014 AACR. See related commentary by Campbell and Moasser, p. 2886
Cancer Epidemiology, Biomarkers & Prevention | 2008
Anne Kendall; Helen Anderson; Anita K. Dunbier; Alan Mackay; Tim Dexter; Ander Urruticoechea; Catherine Harper-Wynne; Mitch Dowsett
Aromatase inhibitors play a key role in the clinical management of hormone receptor–positive breast cancer and have potential utility as chemopreventive agents. Further understanding of the molecular effects of estrogen and its deprivation in normal breast tissue may allow the development of biomarkers of risk of breast cancer and help to predict the value of chemoprevention with aromatase inhibitors. Core biopsies of normal breast tissue were taken before and after letrozole treatment from postmenopausal women in the LITMaS pilot prevention study. RNA was extracted from these samples and used for cDNA microarray analysis. Gene expression changes induced by letrozole treatment were much less extensive than observed in estrogen receptor–positive malignant tissue; however, overall, they correlated to a highly significant degree (ρ = 0.511; P < 10−20). As well as some classically estrogen-associated genes, many genes associated with extracellular matrix remodeling were affected by estrogen deprivation in the normal breast in vivo. These data indicate for the first time that gene expression of normal breast tissue remains dependent on endogenous estrogens after the menopause. The modest degree of gene change suggests that intermediate markers of chemoprevention may be difficult to identify. (Cancer Epidemiol Biomarkers Prev 2008;17(4):855–63)
Cancer Research | 2016
M Ferreira; S Dubey; M Zaiss; Catherine Harper-Wynne; A Makris; V Brown; H Kristeleit; Gargi Patel; A Perelló; A Jones; N Mithal; I Ruiz; S Kümmel; Am Brunt; Ja Guerra; M Gonzalez Cao; C Saura; Kelly Mousa; S-J Sarker; C Coetzee; R Swann; J Cortes
Background: Resistance to endocrine therapy remains a major clinical challenge with aberrant PI3K/ mTOR pathway activation being one of the main drivers. Randomised clinical trials have demonstrated a substantial benefit of adding everolimus to endocrine therapy. Vistusertib (AZD2014), a dual inhibitor of mTORC1 and mTORC2, has shown a broader range of activity in preclinical ER+ breast cancer models, showing superior activity to everolimus (EVE) both in hormone-sensitive and resistant models. The MANTA trial was desgined to evaluate the safety and efficacy of vistusertib (VIS) in combination with fulvestrant (FULV) relative to FULV alone or FULV + EVE. In addition to a continuous (cont) daily schedule of VIS, the study also explored an intermittent (int) schedule to assess the potential of short-term, maximum target inhibition. Methods: MANTA is an investigator-led, randomised, open-label phase II trial. Postmenopausal women with estrogen-receptor (ER)-positive breast cancer were eligible if they had disease recurrence while on or within 12 months of end of adjuvant treatment with an aromatase inhibitor (AI), or progression while on or within one month of end of AI treatment for locally advanced or metastatic breast cancer. Patients were randomly assigned (2:3:3:2) to receive either FULV (500 mg intramuscular injection on day 1, followed by 500 mg doses on days 15 and 29, and then every 28 days); FULV + daily VIS (50mg BD), FULV + intermittent VIS (2 days on, 5 days off; 125mg BD); or FULV + EVE (10mg OD). Treatment was given until disease progression (RECIST 1.1) or intolerable toxicity. Patients were stratified by disease measurability and response to prior endocrine therapy. The primary endpoint was investigator-assessed progression-free survival (PFS). Secondary objectives included objective response, clinical benefit rate, duration of response and clinical benefit, overall survival and safety. Results: Between 04/2014 and 10/2016, a total of 333 patients were randomised at 88 sites in 9 countries. 66 patients were assigned to receive FULV; 101 to FULV+VIS (cont), 95 to FULV+VIS (int); and 64 to FULV+EVE. Median PFS was 4.6 months (95% CI 3.4–6.9) in patients assigned to FULV; 7.5 months (95% CI 5.6–9.4) in those assigned to FULV+VIS (cont); 7.6 months (95% CI 5.5–9.6) in those assigned to FULV+VIS (int); and 12.2 months (95% CI 7.5–14.3) in those assigned to FULV+EVE. No significant difference was recorded between the patients assigned to FULV+VIS (cont) and FULV (hazard ratio 0.87, 95% CI 0.62-1.23; log-rank p=0.42); FULV+VIS (int) and FULV (HR 0.78, 95% CI 0.55-1.12; log-rank p=0.16); and FULV+VIS (cont) and FULV+VIS (int) (HR 1.11, 95% CI 0.81-1.52; log-rank p=0.52). PFS was significantly longer in patients assigned to FULV+EVE compared to FULV+VIS (cont) (HR 0.64, 95% CI 0.45-0.91; log-rank p=0.01) and FULV+EVE compared to FULV (HR 0.64, 95% CI 0.43-0.94; log-rank p=0.02). Conclusion: The trial failed to demonstrate a benefit of adding the TORC1/2 inhibitor vistusertib (AZD2014) to FULV. The combination FULV+EVE demonstrated significantly longer PFS compared to FULV+VIS or FULV. Citation Format: Schmid P, Zaiss M, Harper-Wynne C, Ferreira M, Dubey S, Chan S, Makris A, Nemsadze G, Brunt AM, Kuemmel S, Ruiz Cabrero I, Perello A, Kendall A, Brown J, Kristeleit H, Conibear J, Saura C, Grenier J, Mahr K, Schenker M, Sohn JH, Lee KS, Sarker S-J, Coetzee C, Mousa K, Cortes Castan J. MANTA - A randomized phase II study of fulvestrant in combination with the dual mTOR inhibitor AZD2014 or everolimus or fulvestrant alone in estrogen receptor-positive advanced or metastatic breast cancer [abstract]. In: Proceedings of the 2017 San Antonio Breast Cancer Symposium; 2017 Dec 5-9; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2018;78(4 Suppl):Abstract nr GS2-07.
Cancer Research | 2016
Duncan Wheatley; R Baird; Stephen Chan; J Abraham; Andrew Tutt; H Kristeleit; Gargi Patel; U Bathakur; J Bishop; Catherine Harper-Wynne; E Sims; E Copson; Timothy J. Perren; Robert Stein; Christopher J. Poole; H Cartwright; S-J Sarker; Kelly Mousa; Nicholas C. Turner
Management of metastatic TNBC remains a challenge. Chemotherapy is the mainstay of treatment but benefits are frequently short-lived with rapid development of resistance. The PI3K/AKT/mTOR pathway has been implicated in many ways in TNBC, making inhibition of AKT an attractive therapeutic target. Based on downstream pathway activation signatures, PI3K pathway activation appears higher in TNBC compared to other molecular subtypes, despite a relatively low percentage of activating PI3K mutations. Alternative means of activating the PI3K pathway have been identified in TNBC, including loss or mutation of PTEN (up to 35%) and INPP4B (up to 30%) and/or amplification of PIK3CA, AKT2 or AKT3, resulting in increased activation of AKT. Induction of AKT by chemotherapy can be an early compensatory mechanism that can be exploited therapeutically to increase the efficacy of chemotherapy. Preclinical TNBC models with activated AKT signalling have been shown to be highly sensitive to AKT inhibitors. AZD5363 is a potent pan-AKT inhibitor with good oral bioavailability. Multiple lines of investigation have demonstrated strong synergistic effects between AKT inhibition and taxane chemotherapy in models of TNBC both in vitro and in vivo, providing rationale for the combination of AZD5363 and paclitaxel in TNBC. PAKT is designed to test the hypothesis that inhibition of AKT will increase the anti-tumour activity of paclitaxel chemotherapy in TNBC. The study will try to characterize those patients who may benefit from this treatment to identify potential predictors of sensitivity. PAKT is an international investigator led and sponsored, double-blind, placebo controlled, randomised phase II trial. Patients are randomised 1:1 to receive paclitaxel weekly (90mg/m2) on days 1,8, and 15 plus AZD5363 (400mgBD) or placebo (400mgBD) on days 2-5, 9-12, 15-19 (28 day treatment cycles). Patients are stratified by the number of metastatic sites and the interval from the end of adjuvant chemotherapy. Treatment is given until disease progression (RECIST 1.1), intolerable toxicity or elective withdrawal. Tumour assessments are carried out every 8 weeks. PAKT enrols patients with histologically documented locally advanced/metastatic TNBC (ER≤Allred2, PR≤Allred2, HER2=0,1+or2+), no prior systemic therapy for advanced TNBC, ECOG PS 0-2 and measurable disease per RECIST v1.1. Patients with brain metastases, significant cardiovascular disease, motor polyneuropathy are excluded. The primary endpoint is progression-free survival. Secondary endpoints are objective response rate, change in tumour size, clinical benefit rate, overall survival, duration of response, and patient reported outcomes. Archival tumour tissue must be available to evaluate potential biomarkers associated with therapeutic response and resistance. PFS will be compared between treatment arms by the stratified log-rank test. HR for disease progression/death will be estimated using a stratified Cox proportional hazards model. Kaplan-Meier methodology will be used to estimate the median PFS for each arm. Approximately 140 patients will be enrolled at ≈65 sites in the UK, France, Hungary, Romania, Georgia & South Korea. Citation Format: Schmid P, Wheatley D, Baird R, Chan S, Abraham J, Tutt A, Kristeleit H, Patel G, Bathakur U, Bishop J, Harper-Wynne C, Sims E, Copson E, Perren T, Stein R, Poole C, Cartwright H, Sarker S-J, Mousa K, Turner N. A phase II, double blind, randomised, placebo-controlled study of the AKT Inhibitor AZD5363 in combination with paclitaxel in triple-negative advanced or metastatic breast cancer (TNBC)(NCT02423603). [abstract]. In: Proceedings of the Thirty-Eighth Annual CTRC-AACR San Antonio Breast Cancer Symposium: 2015 Dec 8-12; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2016;76(4 Suppl):Abstract nr OT1-03-13.
Cancer Research | 2012
M. Dowsett; Alexandra Leary; Abigail Evans; Roger A'Hern; Judith M. Bliss; Rashmita Sahoo; Simone Detre; Margaret Hills; B Haynes; Catherine Harper-Wynne; N.J. Bundred; Gill Coombes; I. E. Smith; S. Johnston
Aim: To identify pretreatment biomarker predictors of Ki67 response to lapatinib in women with HER2− primary breast cancer. Background: Lapatinib is an EGFR/HER2 inhibitor. Its clinical use is restricted to HER2 overexpressing disease. The MAPLE (Molecular Antiproliferative Predictors of Lapatinib9s Effects) presurgical window of opportunity study of lapatinib vs placebo was conducted in women with HER2-amplified (HER2+) or HER2− primary disease. Ki67 (primary end-point) was reduced by a geomean 46% (95%CI 23–63%, p = 0.002) and 27% (95%CI 8–42%, p = 0.008) in HER2+ and HER2− disease, respectively (Leary et al, AACR 2012). We have now assessed whether predictive biomarkers of the antiproliferative response in HER2− disease could be identified. Methods: 121 primary breast cancer patients were randomized (3:1) to 14 days of 1500mg/d lapatinib or placebo before surgery. Biopsies were taken before treatment and at surgery. Ki67 responders were defined as having a >/=50% reduction in Ki67 compared to baseline (Ellis, P et al, Breast Cancer Res Treat 1998, 48, 107). ER, PgR, HER2, EGFR, pAKT, pERK1/2 (nuclear and cytoplasmic), stathmin and apoptosis (TUNEL) were assessed by IHC (+FISH for HER2[all cases]) and scored visually by continuous methods. HER2, HER3, epiregulin (epir), amphiregulin (amphir) and neuregulin (neur) were assessed by qrtPCR. Results: Three of the 121 patients were excluded because of inadequate biopsy material. Ninety-one of the remaining 118 patients received lapatinib: 7/19 (37%) HER2+ cases and 10/72 (14%) HER2− cases were Ki67 responders. Thus while the proportion of Ki67 responders was higher for HER2+ disease there was a similar or higher absolute number of responders with HER2− disease. All of the following relates to patients with HER2− disease. None of the pretreatment levels of ER, PgR, pAKT, pERK1/2, EGFR, epir, amphir or neur were associated with Ki67 response (p > 0.20). However, HER3 (p = 0.01) and HER2 (p = 0.06) mRNA levels were associated with greater Ki67 response. There was a tendency for Ki67 response to be greater with lower baseline Ki67 (p = 0.07). Multivariate analysis showed only HER3 mRNA levels to be independently significant. HER2 and HER3 mRNA levels were highly correlated (rho = 0.67, p Conclusions: Lapatinib is antiproliferative in a subgroup of HER2− tumours. This exploratory analysis indicates that they are characterized by high HER3 expression. The possible importance of high HER2:HER3 heterodimers in predicting this response is supported by the relationship between HER2 and HER3 expression. Further exploration of lapatinib is merited in HER2− cases with high HER3 expression. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr PD07-07.
Cancer Research | 2001
Mitch Dowsett; Catherine Harper-Wynne; Irene Boeddinghaus; Janine Salter; Margaret Hills; Mike Dixon; Steve R. Ebbs; Gerald Gui; Nigel Sacks; Ian E. Smith
Cancer Epidemiology, Biomarkers & Prevention | 2002
Catherine Harper-Wynne; Gillian Ross; Nigel Sacks; Janine Salter; Nazar Nasiri; Jhangir Iqbal; Roger A'Hern; Mitch Dowsett
Nature Medicine | 2018
Andrew Tutt; H Tovey; Maggie Cheang; Sarah Kernaghan; Lucy Kilburn; Patrycja Gazinska; Julie Owen; J Abraham; Sophie Barrett; Peter Barrett-Lee; Robert Brown; Stephen Chan; Mitch Dowsett; James M. Flanagan; Lisa Fox; Anita Grigoriadis; Alexander Gutin; Catherine Harper-Wynne; M.Q. Hatton; K. A Hoadley; Jyoti Parikh; Peter J. Parker; Charles M. Perou; R Roylance; Shah; Adam Shaw; Ian E. Smith; Kirsten Timms; Andrew M Wardley; Godfrey Wilson
Cancer Epidemiology and Prevention Biomarkers | 2002
Catherine Harper-Wynne; Gillian Ross; Nigel Sacks; Janine Salter; Nazar Nasiri; Jhangir Iqbal; Roger A’Hern; Mitch Dowsett