Network


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

Hotspot


Dive into the research topics where Fiona Blows is active.

Publication


Featured researches published by Fiona Blows.


PLOS Medicine | 2010

Subtyping of breast cancer by immunohistochemistry to investigate a relationship between subtype and short and long term survival: a collaborative analysis of data for 10,159 cases from 12 studies

Fiona Blows; Kristy Driver; Marjanka K. Schmidt; Annegien Broeks; Flora E. van Leeuwen; Jelle Wesseling; Maggie Cheang; Karen A. Gelmon; Torsten O. Nielsen; Carl Blomqvist; Päivi Heikkilä; Tuomas Heikkinen; Heli Nevanlinna; Lars A. Akslen; Louis R. Bégin; William D. Foulkes; Fergus J. Couch; Xianshu Wang; Vicky Cafourek; Janet E. Olson; Laura Baglietto; Graham G. Giles; Gianluca Severi; Catriona McLean; Melissa C. Southey; Emad A. Rakha; Andrew R. Green; Ian O. Ellis; Mark E. Sherman; Jolanta Lissowska

Paul Pharoah and colleagues evaluate the prognostic significance of immunohistochemical subtype classification in more than 10,000 breast cancer cases with early disease, and examine the influence of a patients survival time on the prediction of future survival.


Annals of Oncology | 2014

Association between CD8+ T-cell infiltration and breast cancer survival in 12 439 patients

Hamid Raza Ali; Elena Provenzano; S-J Dawson; Fiona Blows; Bin Liu; Mitulkumar Nandlal Shah; Helena M. Earl; Christopher J. Poole; Louise Hiller; Janet A. Dunn; Sarah Bowden; C. Twelves; Jms Bartlett; Sma Mahmoud; Emad A. Rakha; Ian O. Ellis; Suzanne Liu; Dongxia Gao; Torsten O. Nielsen; Paul Pharoah; Carlos Caldas

BACKGROUND T-cell infiltration in estrogen receptor (ER)-negative breast tumours has been associated with longer survival. To investigate this association and the potential of tumour T-cell infiltration as a prognostic and predictive marker, we have conducted the largest study of T cells in breast cancer to date. PATIENTS AND METHODS Four studies totalling 12 439 patients were used for this work. Cytotoxic (CD8+) and regulatory (forkhead box protein 3, FOXP3+) T cells were quantified using immunohistochemistry (IHC). IHC for CD8 was conducted using available material from all four studies (8978 samples) and for FOXP3 from three studies (5239 samples)-multiple imputation was used to resolve missing data from the remaining patients. Cox regression was used to test for associations with breast cancer-specific survival. RESULTS In ER-negative tumours [triple-negative breast cancer and human epidermal growth factor receptor 2 (human epidermal growth factor receptor 2 (HER2) positive)], presence of CD8+ T cells within the tumour was associated with a 28% [95% confidence interval (CI) 16% to 38%] reduction in the hazard of breast cancer-specific mortality, and CD8+ T cells within the stroma with a 21% (95% CI 7% to 33%) reduction in hazard. In ER-positive HER2-positive tumours, CD8+ T cells within the tumour were associated with a 27% (95% CI 4% to 44%) reduction in hazard. In ER-negative disease, there was evidence for greater benefit from anthracyclines in the National Epirubicin Adjuvant Trial in patients with CD8+ tumours [hazard ratio (HR) = 0.54; 95% CI 0.37-0.79] versus CD8-negative tumours (HR = 0.87; 95% CI 0.55-1.38). The difference in effect between these subgroups was significant when limited to cases with complete data (P heterogeneity = 0.04) and approached significance in imputed data (P heterogeneity = 0.1). CONCLUSIONS The presence of CD8+ T cells in breast cancer is associated with a significant reduction in the relative risk of death from disease in both the ER-negative [supplementary Figure S1, available at Annals of Oncology online] and the ER-positive HER2-positive subtypes. Tumour lymphocytic infiltration may improve risk stratification in breast cancer patients classified into these subtypes. NEAT ClinicalTrials.gov: NCT00003577.


Genetics | 2007

The DrosDel Deletion Collection: A Drosophila Genomewide Chromosomal Deficiency Resource

Edward Ryder; Michael Ashburner; Rosa Bautista-Llacer; Jenny Drummond; Jane Webster; Glynnis Johnson; Terri Morley; Yuk Sang Chan; Fiona Blows; Darin Coulson; Gunter Reuter; Heiko Baisch; Christian Apelt; Andreas Kauk; Thomas Rudolph; Maria Kube; Melanie Klimm; Claudia Nickel; János Szidonya; Péter Maróy; Margit Pál; Åsa Rasmuson-Lestander; Karin Ekström; Hugo Stocker; Christoph Hugentobler; Ernst Hafen; David Gubb; Gert O. Pflugfelder; Christian Dorner; Bernard M. Mechler

We describe a second-generation deficiency kit for Drosophila melanogaster composed of molecularly mapped deletions on an isogenic background, covering ∼77% of the Release 5.1 genome. Using a previously reported collection of FRT-bearing P-element insertions, we have generated 655 new deletions and verified a set of 209 deletion-bearing fly stocks. In addition to deletions, we demonstrate how the P elements may also be used to generate a set of custom inversions and duplications, particularly useful for balancing difficult regions of the genome carrying haplo-insufficient loci. We describe a simple computational resource that facilitates selection of appropriate elements for generating custom deletions. Finally, we provide a computational resource that facilitates selection of other mapped FRT-bearing elements that, when combined with the DrosDel collection, can theoretically generate over half a million precisely mapped deletions.


British Journal of Cancer | 2010

BCL2 in breast cancer: a favourable prognostic marker across molecular subtypes and independent of adjuvant therapy received

Sarah-Jane Dawson; Nikita Makretsov; Fiona Blows; Kristy Driver; Elena Provenzano; J. Le Quesne; Laura Baglietto; Gianluca Severi; Graham G. Giles; Catriona McLean; Grace Callagy; Andrew R. Green; Ian O. Ellis; Karen A. Gelmon; Gulisa Turashvili; Scy Leung; Sam Aparicio; David Huntsman; Carlos Caldas; P Pharoah

BACKGROUND Breast cancer is heterogeneous and the existing prognostic classifiers are limited in accuracy, leading to unnecessary treatment of numerous women. B-cell lymphoma 2 (BCL2), an antiapoptotic protein, has been proposed as a prognostic marker, but this effect is considered to relate to oestrogen receptor (ER) status. This study aimed to test the clinical validity of BCL2 as an independent prognostic marker. METHODS Five studies of 11 212 women with early-stage breast cancer were analysed. Individual patient data included tumour size, grade, lymph node status, endocrine therapy, chemotherapy and mortality. BCL2, ER, progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) levels were determined in all tumours. A Cox model incorporating the time-dependent effects of each variable was used to explore the prognostic significance of BCL2. RESULTS In univariate analysis, ER, PR and BCL2 positivity was associated with improved survival and HER2 positivity with inferior survival. For ER and PR this effect was time dependent, whereas for BCL2 and HER2 the effect persisted over time. In multivariate analysis, BCL2 positivity retained independent prognostic significance (hazard ratio (HR) 0.76, 95% confidence interval (CI) 0.66-0.88, P<0.001). BCL2 was a powerful prognostic marker in ER- (HR 0.63, 95% CI 0.54-0.74, P<0.001) and ER+ disease (HR 0.56, 95% CI 0.48-0.65, P<0.001), and in HER2- (HR 0.55, 95% CI 0.49-0.61, P<0.001) and HER2+ disease (HR 0.70, 95% CI 0.57-0.85, P<0.001), irrespective of the type of adjuvant therapy received. Addition of BCL2 to the Adjuvant! Online prognostic model, for a subset of cases with a 10-year follow-up, improved the survival prediction (P=0.0039). CONCLUSIONS BCL2 is an independent indicator of favourable prognosis for all types of early-stage breast cancer. This study establishes the rationale for introduction of BCL2 immunohistochemistry to improve prognostic stratification. Further work is now needed to ascertain the exact way to apply BCL2 testing for risk stratification and to standardise BCL2 immunohistochemistry for this application.Background:Breast cancer is heterogeneous and the existing prognostic classifiers are limited in accuracy, leading to unnecessary treatment of numerous women. B-cell lymphoma 2 (BCL2), an antiapoptotic protein, has been proposed as a prognostic marker, but this effect is considered to relate to oestrogen receptor (ER) status. This study aimed to test the clinical validity of BCL2 as an independent prognostic marker.Methods:Five studies of 11 212 women with early-stage breast cancer were analysed. Individual patient data included tumour size, grade, lymph node status, endocrine therapy, chemotherapy and mortality. BCL2, ER, progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) levels were determined in all tumours. A Cox model incorporating the time-dependent effects of each variable was used to explore the prognostic significance of BCL2.Results:In univariate analysis, ER, PR and BCL2 positivity was associated with improved survival and HER2 positivity with inferior survival. For ER and PR this effect was time dependent, whereas for BCL2 and HER2 the effect persisted over time. In multivariate analysis, BCL2 positivity retained independent prognostic significance (hazard ratio (HR) 0.76, 95% confidence interval (CI) 0.66–0.88, P<0.001). BCL2 was a powerful prognostic marker in ER− (HR 0.63, 95% CI 0.54–0.74, P<0.001) and ER+ disease (HR 0.56, 95% CI 0.48–0.65, P<0.001), and in HER2− (HR 0.55, 95% CI 0.49–0.61, P<0.001) and HER2+ disease (HR 0.70, 95% CI 0.57–0.85, P<0.001), irrespective of the type of adjuvant therapy received. Addition of BCL2 to the Adjuvant! Online prognostic model, for a subset of cases with a 10-year follow-up, improved the survival prediction (P=0.0039).Conclusions:BCL2 is an independent indicator of favourable prognosis for all types of early-stage breast cancer. This study establishes the rationale for introduction of BCL2 immunohistochemistry to improve prognostic stratification. Further work is now needed to ascertain the exact way to apply BCL2 testing for risk stratification and to standardise BCL2 immunohistochemistry for this application.


Annals of Oncology | 2015

PD-L1 protein expression in breast cancer is rare, enriched in basal-like tumours and associated with infiltrating lymphocytes

Hamid Raza Ali; S-E Glont; Fiona Blows; Elena Provenzano; S-J Dawson; Bin Liu; Louise Hiller; Janet A. Dunn; Christopher J. Poole; Sarah Bowden; Helena M. Earl; Paul Pharoah; Carlos Caldas

BACKGROUND Expression of programmed death ligand 1 (PD-L1) in solid tumours has been shown to predict whether patients are likely to respond to anti-PD-L1 therapies. To estimate the therapeutic potential of PD-L1 inhibition in breast cancer, we evaluated the prevalence and significance of PD-L1 protein expression in a large collection of breast tumours. PATIENTS AND METHODS Correlations between CD274 (PD-L1) copy number, transcript and protein levels were evaluated in tumours from 418 patients recruited to the METABRIC genomic study. Immunohistochemistry was used to detect PD-L1 protein in breast tumours in tissue microarrays from 5763 patients recruited to the SEARCH population-based study (N = 4079) and the NEAT randomised, controlled trial (N = 1684). RESULTS PD-L1 protein data was available for 3916 of the possible 5763 tumours from the SEARCH and NEAT studies. PD-L1 expression by immune cells was observed in 6% (235/3916) of tumours and expression by tumour cells was observed in just 1.7% (66/3916). PD-L1 was most frequently expressed in basal-like tumours. This was observed both where tumours were subtyped by combined copy number and expression profiling [39% (17/44) of IntClust 10 i.e. basal-like tumours were PD-L1 immune cell positive; P < 0.001] and where a surrogate IHC-based classifier was used [19% (56/302) of basal-like tumours were PD-L1 immune cell positive; P < 0.001]. Moreover, CD274 (PD-L1) amplification was observed in five tumours of which four were IntClust 10. Expression of PD-L1 by either tumour cells or infiltrating immune cells was positively correlated with infiltration by both cytotoxic and regulatory T cells (P < 0.001). There was a nominally significant association between PD-L1 and improved disease-specific survival (hazard ratio 0.53, 95% confidence interval 0.26-1.07; P = 0.08) in ER-negative disease. CONCLUSIONS Expression of PD-L1 is rare in breast cancer, markedly enriched in basal-like tumours and is correlated with infiltrating lymphocytes. PD-L1 inhibition may benefit the 19% of patients with basal-like tumours in which the protein is expressed. NEAT CLINICALTRIALSGOV NCT00003577.


British Journal of Cancer | 2012

PREDICT Plus: development and validation of a prognostic model for early breast cancer that includes HER2

Gordon Wishart; Chris Bajdik; Ed Dicks; Elena Provenzano; Marjanka K. Schmidt; Mark E. Sherman; David C Greenberg; Andrew R. Green; Karen A. Gelmon; Veli-Matti Kosma; Janet E. Olson; Matthias W. Beckmann; Robert Winqvist; Simon S. Cross; Gianluca Severi; David Huntsman; K Pylkas; Ian O. Ellis; Torsten O. Nielsen; Graham G. Giles; Carl Blomqvist; Peter A. Fasching; Fergus J. Couch; Emad A. Rakha; William D. Foulkes; Fiona Blows; Louis R. Bégin; L van't Veer; Melissa C. Southey; Heli Nevanlinna

Background:Predict (www.predict.nhs.uk) is an online, breast cancer prognostication and treatment benefit tool. The aim of this study was to incorporate the prognostic effect of HER2 status in a new version (Predict+), and to compare its performance with the original Predict and Adjuvant!.Methods:The prognostic effect of HER2 status was based on an analysis of data from 10 179 breast cancer patients from 14 studies in the Breast Cancer Association Consortium. The hazard ratio estimates were incorporated into Predict. The validation study was based on 1653 patients with early-stage invasive breast cancer identified from the British Columbia Breast Cancer Outcomes Unit. Predicted overall survival (OS) and breast cancer-specific survival (BCSS) for Predict+, Predict and Adjuvant! were compared with observed outcomes.Results:All three models performed well for both OS and BCSS. Both Predict models provided better BCSS estimates than Adjuvant!. In the subset of patients with HER2-positive tumours, Predict+ performed substantially better than the other two models for both OS and BCSS.Conclusion:Predict+ is the first clinical breast cancer prognostication tool that includes tumour HER2 status. Use of the model might lead to more accurate absolute treatment benefit predictions for individual patients.


Cancer Research | 2011

Common Breast Cancer Susceptibility Loci Are Associated with Triple-Negative Breast Cancer

Kristen N. Stevens; Celine M. Vachon; Adam Lee; Susan L. Slager; Timothy G. Lesnick; Curtis Olswold; Peter A. Fasching; Penelope Miron; Diana Eccles; Jane Carpenter; Andrew K. Godwin; Christine B. Ambrosone; Robert Winqvist; Hiltrud Brauch; Marjanka K. Schmidt; Angela Cox; Simon S. Cross; Elinor Sawyer; Arndt Hartmann; Matthias W. Beckmann; Rud̈iger Schulz-Wendtland; Arif B. Ekici; William Tapper; Susan M. Gerty; Lorraine Durcan; Nikki Graham; Rebecca Hein; Stephan Nickels; Dieter Flesch-Janys; Judith Heinz

Triple-negative breast cancers are an aggressive subtype of breast cancer with poor survival, but there remains little known about the etiologic factors that promote its initiation and development. Commonly inherited breast cancer risk factors identified through genome-wide association studies display heterogeneity of effect among breast cancer subtypes as defined by the status of estrogen and progesterone receptors. In the Triple Negative Breast Cancer Consortium (TNBCC), 22 common breast cancer susceptibility variants were investigated in 2,980 Caucasian women with triple-negative breast cancer and 4,978 healthy controls. We identified six single-nucleotide polymorphisms, including rs2046210 (ESR1), rs12662670 (ESR1), rs3803662 (TOX3), rs999737 (RAD51L1), rs8170 (19p13.1), and rs8100241 (19p13.1), significantly associated with the risk of triple-negative breast cancer. Together, our results provide convincing evidence of genetic susceptibility for triple-negative breast cancer.


British Journal of Cancer | 2009

Molecular characteristics of screen-detected vs symptomatic breast cancers and their impact on survival

Sarah-Jane Dawson; Stephen W. Duffy; Fiona Blows; Kristy Driver; Elena Provenzano; J LeQuesne; D C Greenberg; Paul Pharoah; Carlos Caldas; G C Wishart

Background:Several recent studies have shown that screen detection remains an independent prognostic factor after adjusting for disease stage at presentation. This study compares the molecular characteristics of screen-detected with symptomatic breast cancers to identify if differences in tumour biology may explain some of the survival benefit conferred by screen detection.Methods:A total of 1379 women (aged 50–70 years) with invasive breast cancer from a large population-based case–control study were included in the analysis. Individual patient data included tumour size, grade, lymph node status, adjuvant therapy, mammographic screening status and mortality. Immunohistochemistry was performed on tumour samples using 11 primary antibodies to define five molecular subtypes. The effect of screen detection compared with symptomatic diagnosis on survival was estimated after adjustment for grade, nodal status, Nottingham Prognostic Index (NPI) and the molecular markers.Results:Fifty-six per cent of the survival benefit associated with screen-detected breast cancer was accounted for by a shift in the NPI, a further 3–10% was explained by the biological variables and more than 30% of the effect remained unexplained.Conclusion:Currently known biomarkers remain limited in their ability to explain the heterogeneity of breast cancer fully. A more complete understanding of the biological profile of breast tumours will be necessary to assess the true impact of tumour biology on the improvement in survival seen with screen detection.


The Journal of Pathology | 2012

A Ki67/BCL2 index based on immunohistochemistry is highly prognostic in ER-positive breast cancer.

H. Raza Ali; Sarah-Jane Dawson; Fiona Blows; Elena Provenzano; Samuel Leung; Torsten O. Nielsen; Paul Pharoah; Carlos Caldas

There is an urgent need to improve prognostic classifiers in breast cancer. Ki67 and B‐cell lymphoma 2 protein (BCL2) are established prognostic markers which have traditionally been assessed separately, in a dichotomous manner. This study was conducted to test the hypothesis that combinatorial assessment of these markers would provide superior prognostic information and improve their clinical utility. Tissue microarrays were used to assess the expression of Ki67 and BCL2 in 2749 cases of invasive breast cancer. We devised a Ki67/BCL2 index representing the relative expression of each protein and assessed its association with breast cancer‐specific survival (BCSS) using a Cox proportional‐hazards model. Based on our findings, an independent cohort of 3992 cases was used to validate the prognostic significance of the Ki67/BCL2 index. All survival analyses were conducted on complete data as well as data where missing values were resolved using multiple imputation. This study complied with reporting recommendations for tumour marker prognostic studies (REMARK) criteria. The Ki67/BCL2 index showed a significant association with BCSS at 10 years in estrogen receptor (ER)‐positive disease. In multivariate analysis, adjusting for major clinical and molecular markers, the Ki67/BCL2 index retained prognostic significance, robustly classifying cases into three risk groups [intermediate‐ versus low‐risk hazard ratio (HR), 1.5; 95% confidence interval (95% CI), 1.0–2.0; p = 0.031; high‐ versus low‐risk HR, 2.6; 95% CI, 1.3–5.0; p = 0.005]. This finding was validated in an independent cohort of 3992 tumours containing 2761 ER‐positive tumours (intermediate‐ versus low‐risk HR, 1.7; 95% CI, 1.3–2.1; p < 0.001; high‐ versus low‐risk HR, 2.0; 95% CI, 1.4–2.9; p < 0.001). Ki67 and BCL2 can be effectively combined to produce an index which is an independent predictor of BCSS in ER‐positive breast cancer, enhancing their potential prognostic utility. Copyright


The Journal of Pathology | 2012

Biological and prognostic associations of miR-205 and let-7b in breast cancer revealed by in situ hybridization analysis of micro-RNA expression in arrays of archival tumour tissue.

John Le Quesne; Julia Jones; Joanna Warren; Sarah-Jane Dawson; H. Raza Ali; Helen Bardwell; Fiona Blows; Paul Pharoah; Carlos Caldas

Micro‐RNAs (miRNAs) are frequently dysregulated in a range of human malignancies, many have been shown to act either as tumour supressors or oncogenes and several have been implicated in breast cancer. However, breast cancer is a diverse disease and little is known about the relationships between miRNA expression, clinical outcome and tumour subtype. We used locked nucleic acid probe in situ hybridization (LNA‐ISH) to visualize, in tissue micro‐arrays (TMAs) of 2919 formalin‐fixed paraffin‐embedded (FFPE) archival breast tumours, the expression of two key miRNAs that are frequently lost in a range of solid malignancies, let‐7b and miR‐205. These miRNAs were also quantified by quantitative reverse transcription PCR in cores of FFPE tissue from 40 of these cases, demonstrating that LNA‐ISH is semi‐quantitative. The tumours in the TMAs were assigned to subtypes based on their immunohistochemical (IHC) staining with ER, PR, HER2, CK5/6 and EGFR. let‐7b expression was shown to be associated with luminal tumours and to have an independent significant positive prognostic value in this group. miR‐205 is associated with tumours of ductal morphology and is of significant positive prognostic value within these tumours. We propose that the expression of miR‐205 may contribute to ductal tumour morphology. Copyright

Collaboration


Dive into the Fiona Blows's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elena Provenzano

Cambridge University Hospitals NHS Foundation Trust

View shared research outputs
Top Co-Authors

Avatar

Paul Pharoah

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

H. Raza Ali

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar

Penny Coulson

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Sarah-Jane Dawson

Peter MacCallum Cancer Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mark E. Sherman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge