Imperial College London

ProfessorIainMcNeish

Faculty of MedicineDepartment of Surgery & Cancer

Chair in Oncology
 
 
 
//

Contact

 

+44 (0)20 7594 2185i.mcneish Website

 
 
//

Assistant

 

Ms Sophie Lions +44 (0)20 7594 2792

 
//

Location

 

G036Institute of Reproductive and Developmental BiologyHammersmith Campus

//

Summary

 

Publications

Publication Type
Year
to

328 results found

Goranova T, Ennis D, Piskorz AM, Macintyre G, Lewsley LA, Stobo J, Wilson C, Kay D, Glasspool RM, Lockley M, Brockbank E, Montes A, Walther A, Sundar S, Edmondson R, Hall GD, Clamp A, Gourley C, Hall M, Fotopoulou C, Gabra H, Freeman S, Moore L, Jimenez-Linan M, Paul J, Brenton JD, McNeish IA, BriTROC investigatorset al., 2019, Correction: Safety and utility of image-guided research biopsies in relapsed high-grade serous ovarian carcinoma-experience of the BriTROC consortium., British Journal of Cancer, Vol: 120, ISSN: 0007-0920

This article was originally published under a CC BY NC SA License, but has now been made available under a CC BY 4.0 License.

Journal article

Bagnoli M, Shi TY, Gourley C, Speiser P, Reuss A, Nijman HW, Creutzberg CL, Scholl S, Negrouk A, Brady MF, Hasegawa K, Oda K, McNeish IA, Kohn EC, Oza AM, MacKay H, Millan D, Bennett K, Scott C, Mezzanzanica Det al., 2019, Gynecological cancers translational, research implementation, and harmonization: Gynecologic Cancer InterGroup consensus and still open questions, Cells, Vol: 8, ISSN: 2073-4409

In the era of personalized medicine, the introduction of translational studies in clinical trials has substantially increased their costs, but provides the possibility of improving the productivity of trials with a better selection of recruited patients. With the overall goal of creating a roadmap to improve translational design for future gynecological cancer trials and of defining translational goals, a main discussion was held during a brainstorming day of the Gynecologic Cancer InterGroup (GCIG) Translational Research Committee and overall conclusions are here reported. A particular emphasis was dedicated to the new frontier of the immunoprofiling of gynecological cancers. The discussion pointed out that to maximize patients’ benefit, translational studies should be integral to clinical trial design with standardization and optimization of procedures including a harmonization program of Standard Operating Procedures. Pathology-reviewed sample collection should be mandatory and ensured by dedicated funding. Biomarker validation and development should be made public and transparent to ensure rapid progresses with positive outcomes for patients. Guidelines/templates for patients’ informed consent are needed. Importantly for the public, recognized goals are to increase the involvement of advocates and to improve the reporting of translational data in a forum accessible to patients.

Journal article

Buckley MA, Woods NT, Tyrer JP, Mendoza-Fandino G, Lawrenson K, Hazelett DJ, Najafabadi HS, Gjyshi A, Carvalho RS, Lyra PC, Coetzee SG, Shen HC, Yang AW, Earp MA, Yoder SJ, Risch H, Chenevix-Trench G, Ramus SJ, Phelan CM, Coetzee GA, Noushmehr H, Hughes TR, Sellers TA, Goode EL, Pharoah PD, Gayther SA, Monteiro ANA, Chen YA, Fridley BL, Aben KKH, Kiemeney LA, Anton-Culver H, Ziogas A, Bruinsma F, Milne RL, Bandera EV, Giles GG, Bean YT, Pejovic T, Beckmann MW, Hein A, Bjorge L, Fasching PA, Thomsen LCV, Kopperud RK, Bischof K, Bogdanova N, Doek T, Hillemanns P, Brinton LA, Wentzensen N, Yang H, Brooks-Wilson A, Bunker CH, Butzow R, Nevanlinna H, Pelttari LM, Campbell IG, Southey MC, Modugno F, Carty K, Glasspool R, McNeish I, Paul J, Siddiqui N, Chang-Claude J, Rudolph A, Chang-Claude J, Cook LS, Cramer DW, Terry KL, Cunningham JM, Cybulski C, Gronwald J, Jakubowska A, Lubinski J, Dansonka-Mieszkowska A, Kupryjanczyk J, Rzepecka IK, du Bois A, Harter P, Dicks E, Song H, Doherty JA, Rossing MA, Duerst M, Easton DF, Eccles DM, Edwards RP, Ekici AB, Fasching PA, Gao Y-T, Milne RL, Gentry-Maharaj A, Giles GG, Goodman MT, Thompson PJ, Hasmad HN, Teo S-H, Hildebrandt MAT, Wu X, Hogdall E, Jensen A, Kjaer SK, Hogdall E, Iversen ES, Karlan BY, Lester J, Orsulic S, Walsh CS, Kelley JL, Lambrechts D, Lambrechts S, Vergote I, Lee AW, Levine DA, Liang D, Lissowska J, Lu K, Lundvall L, Kjaer SK, Massuger LFAG, van Altena AM, Matsuo K, McGuire V, McLaughlin JR, Menon U, Moysich KB, Ness RB, Odunsi K, Olson SH, Orlow I, Pike MC, Pearce CL, Wu AH, Permuth JB, Tsai Y-Y, Tworoger SS, Poole EM, Rosen B, Shu X-O, Shvetsov YB, Wilkens LR, Sieh W, Spiewankiewicz B, Sucheston-Campbell L, Thomsen L, Wang-Gohrke S, Whittemore AS, Woo Y-L, Zheng W, Berchuck A, Chenevix-Trench G, Schildkraut JM, Kelemen LE, Freedman MLet al., 2019, Functional Analysis and Fine Mapping of the 9p22.2 Ovarian Cancer Susceptibility Locus, CANCER RESEARCH, Vol: 79, Pages: 467-481, ISSN: 0008-5472

Journal article

Yang Y, Wu L, Shu X, Lu Y, Shu X-O, Cai Q, Beeghly-Fadiel A, Li B, Ye F, Berchuck A, Anton-Culver H, Banerjee S, Benitez J, Bjørge L, Brenton JD, Butzow R, Campbell IG, Chang-Claude J, Chen K, Cook LS, Cramer DW, DeFazio A, Dennis J, Doherty JA, Dork T, Eccles DM, Velez Edwards D, Fasching PA, Fortner RT, Gayther SA, Giles GG, Glasspool RM, Goode EL, Goodman MT, Gronwald J, Harris HR, Heitz F, Hildebrandt MAT, Høgdall E, Høgdall CK, Huntsman DG, Kar SP, Karlan BY, Kelemen LE, Kiemeney LA, Kjaer SK, Koushik A, Lambrechts D, Le ND, Levine DA, Massuger LFAG, Matsuo K, May T, McNeish IA, Menon U, Modugno F, Monteiro AN, Moorman PG, Moysich KB, Ness RB, Nevanlinna H, Olsson H, Onland-Moret NC, Park SK, Paul J, Pearce CL, Pejovic T, Phelan CM, Pike MC, Ramus SJ, Riboli E, Rodríguez-Antona C, Romieu I, Sandler DP, Schildkraut JM, Setiawan VW, Shan K, Siddiqui N, Sieh W, Stampfer MJ, Sutphen R, Swerdlow AJ, Szafron LM, Teo SH, Tworoger SS, Tyrer JP, Webb PM, Wentzensen N, White E, Willett WC, Wolk A, Woo YL, Wu AH, Yan L, Yannoukakos D, Chenevix-Trench G, Sellers TA, Pharoah PDP, Zheng W, Long Jet al., 2019, Genetic data from nearly 63,000 women of European descent predicts DNA methylation biomarkers and epithelial ovarian cancer risk, Cancer Research, Vol: 79, Pages: 505-517, ISSN: 1538-7445

DNA methylation is instrumental for gene regulation. Global changes in the epigenetic landscape have been recognized as a hallmark of cancer. However, the role of DNA methylation in epithelial ovarian cancer (EOC) remains unclear. In this study, high density genetic and DNA methylation data in white blood cells from the Framingham Heart Study (N=1,595) were used to build genetic models to predict DNA methylation levels. These prediction models were then applied to the summary statistics of a genome-wide association study (GWAS) of ovarian cancer including 22,406 EOC cases and 40,941 controls to investigate genetically predicted DNA methylation levels in association with EOC risk. Among 62,938 CpG sites investigated, genetically predicted methylation levels at 89 CpG were significantly associated with EOC risk at a Bonferroni-corrected threshold of P<7.94×10-7. Of them, 87 were located at GWAS-identified EOC susceptibility regions and two resided in a genomic region not previously reported to be associated with EOC risk. Integrative analyses of genetic, methylation, and gene expression data identified consistent directions of associations across 12 CpG, five genes, and EOC risk, suggesting that methylation at these 12 CpG may influence EOC risk by regulating expression of these five genes, namely MAPT, HOXB3, ABHD8, ARHGAP27 and SKAP1. We identified novel DNA methylation markers associated with EOC risk and propose that methylation at multiple CpG may affect EOC risk via regulation of gene expression.

Journal article

Lin KK, Harrell MI, Oza AM, Oaknin A, Ray-Coquard I, Tinker AV, Helman E, Radke MR, Say C, Vo L-T, Mann E, Isaacson JD, Maloney L, O'Malley DM, Chambers SK, Kaufmann SH, Scott CL, Konecny GE, Coleman RL, Sun JX, Giordano H, Brenton JD, Harding TC, McNeish IA, Swisher EMet al., 2019, BRCA reversion mutations in circulating tumor DNA predict primary and acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma, Cancer Discovery, Vol: 9, Pages: 210-219, ISSN: 2159-8274

A key resistance mechanism to platinum-based chemotherapies and PARP inhibitors in BRCA-mutant cancers is the acquisition of BRCA reversion mutations that restore protein function. To estimate the prevalence of BRCA reversion mutations in high-grade ovarian carcinoma (HGOC), we performed targeted next-generation sequencing of circulating cell-free DNA (cfDNA) extracted from pretreatment and postprogression plasma in patients with deleterious germline or somatic BRCA mutations treated with the PARP inhibitor rucaparib. BRCA reversion mutations were identified in pretreatment cfDNA from 18% (2/11) of platinum-refractory and 13% (5/38) of platinum-resistant cancers, compared to 2% (1/48) of platinum-sensitive cancers (P = 0.049). Patients without BRCA reversion mutations detected in pretreatment cfDNA had significantly longer rucaparib progression-free survival than those with reversion mutations (median, 9.0 vs. 1.8 months; HR, 0.12; P < 0.0001). To study acquired resistance, we sequenced 78 postprogression cfDNA, identifying eight additional patients with BRCA reversion mutations not found in pretreatment cfDNA.

Journal article

Macintyre G, Piskorz A, Ross E, Morse D, Yuan K, Ennis D, Pike J, Goranova T, McNeish I, Brenton J, Markowetz Fet al., 2018, frenchFISH: Poisson models for quantifying DNA copy-number from fluorescence in situ hybridisation of tissue sections

Chromosomal aberration and DNA copy number change are robust hallmarks of cancer. Imaging of spots generated using fluorescence in situ hybridisation (FISH) of locus specific probes is routinely used to detect copy number changes in tumour nuclei. However, it often does not perform well on solid tumour tissue sections, where partially represented or overlapping nuclei are common. To overcome these challenges, we have developed a computational approach called FrenchFISH, which comprises a nuclear volume correction method coupled with two types of Poisson models: either a Poisson model for improved manual spot counting without the need for control probes; or a homogenous Poisson Point Process model for automated spot counting. We benchmarked the performance of FrenchFISH against previous approaches in a controlled simulation scenario and exemplify its use in 12 ovarian cancer FFPE-tissue sections, for which we assess copy number alterations in three loci (c-Myc, hTERC and SE7). We show that FrenchFISH outperforms standard spot counting approaches and that the automated spot counting is significantly faster than manual without loss of performance. FrenchFISH is a general approach that can be used to enhance clinical diagnosis on sections of any tissue.

Working paper

Vergote I, Coens C, Nankivell M, Kristensen GB, Parmar MKB, Ehlen T, Jayson GC, Johnson N, Swart AM, Verheijen R, McCluggage WG, Perren T, Panici PB, Kenter G, Casado A, Mendiola C, Stuart G, Reed NS, Kehoe S, EORTC, MRC CHORUS study investigatorset al., 2018, Neoadjuvant chemotherapy versus debulking surgery in advanced tubo-ovarian cancers: pooled analysis of individual patient data from the EORTC 55971 and CHORUS trials, The Lancet Oncology, Vol: 19, Pages: 1680-1687, ISSN: 1213-9432

BACKGROUND: Individual patient data from two randomised trials comparing neoadjuvant chemotherapy with upfront debulking surgery in advanced tubo-ovarian cancer were analysed to examine long-term outcomes for patients and to identify any preferable therapeutic approaches for subgroup populations. METHODS: We did a per-protocol pooled analysis of individual patient data from the European Organisation for Research and Treatment of Cancer (EORTC) 55971 trial (NCT00003636) and the Medical Research Council Chemotherapy Or Upfront Surgery (CHORUS) trial (ISRCTN74802813). In the EORTC trial, eligible women had biopsy-proven International Federation of Gynecology and Obstetrics (FIGO) stage IIIC or IV invasive epithelial tubo-ovarian carcinoma. In the CHORUS trial, inclusion criteria were similar to those of the EORTC trial, and women with apparent FIGO stage IIIA and IIIB disease were also eligible. The main aim of the pooled analysis was to show non-inferiority in overall survival with neoadjuvant chemotherapy compared with upfront debulking surgery, using the reverse Kaplan-Meier method. Tests for heterogeneity were based on Cochran's Q heterogeneity statistic. FINDINGS: Data for 1220 women were included in the pooled analysis, 670 from the EORTC trial and 550 from the CHORUS trial. 612 women were randomly allocated to receive upfront debulking surgery and 608 to receive neoadjuvant chemotherapy. Median follow-up was 7·6 years (IQR 6·0-9·6; EORTC, 9·2 years [IQR 7·3-10·4]; CHORUS, 5·9 years [IQR 4·3-7·4]). Median age was 63 years (IQR 56-71) and median size of the largest metastatic tumour at diagnosis was 8 cm (IQR 4·8-13·0). 55 (5%) women had FIGO stage II-IIIB disease, 831 (68%) had stage IIIC disease, and 230 (19%) had stage IV disease, with staging data missing for 104 (9%) women. In the entire population, no difference in median overall survival was noted between patients who underwent neo

Journal article

Sierra Gonzalez P, OPrey J, Cardaci S, Barthet V, Sakamaki J-I, Beaumatin F, Roseweir A, Gay D, Mackay G, Malviya G, Kania E, Ritchie S, Baudot A, Zunino B, Mrowinska A, Nixon C, Ennis D, Hoyle A, Millan D, McNeish I, Sansom O, Edwards J, Ryan Ket al., 2018, Mannose impairs tumour growth and enhances chemotherapy, Nature, Vol: 563, Pages: 719-723, ISSN: 0028-0836

It is now well established that tumours undergo changes in cellular metabolism1. As this can reveal tumour cell vulnerabilities and because many tumours exhibit enhanced glucose uptake2, we have been interested in how tumour cells respond to different forms of sugar. Here we report that the monosaccharide mannose causes growth retardation in several tumour types in vitro, and enhances cell death in response to major forms of chemotherapy. We then show that these effects also occur in vivo in mice following the oral administration of mannose, without significantly affecting the weight and health of the animals. Mechanistically, mannose is taken up by the same transporter(s) as glucose3 but accumulates as mannose-6-phosphate in cells, and this impairs the further metabolism of glucose in glycolysis, the tricarboxylic acid cycle, the pentose phosphate pathway and glycan synthesis. As a result, the administration of mannose in combination with conventional chemotherapy affects levels of anti-apoptotic proteins of the Bcl-2 family, leading to sensitization to cell death. Finally we show that susceptibility to mannose is dependent on the levels of phosphomannose isomerase (PMI). Cells with low levels of PMI are sensitive to mannose, whereas cells with high levels are resistant, but can be made sensitive by RNA-interference-mediated depletion of the enzyme. In addition, we use tissue microarrays to show that PMI levels also vary greatly between different patients and different tumour types, indicating that PMI levels could be used as a biomarker to direct the successful administration of mannose. We consider that the administration of mannose could be a simple, safe and selective therapy in the treatment of cancer, and could be applicable to multiple tumour types.

Journal article

McNeish IA, 2018, Neoantigens in ovarian cancer: embarrassment of riches or needles in a haystack?, Clinical Cancer Research, Vol: 24, Pages: 5493-5495, ISSN: 1078-0432

Comprehensive genomic and transcriptomic analysis demonstrates that tumor-infiltrating T lymphocytes that react to mutated neo-epitopes could be identified in recurrent ovarian cancer. Two of these T cell populations reacted against TP53 hotspot missense mutations that are present in a wide variety of malignancies.

Journal article

Mcneish IA, Lin KK, Radke MR, Oza AM, Oaknin A, Ray-Coquard I, Tinker AV, Helman E, Isaacson J, Maloney L, O'malley DM, Chambers SK, Kaufmann SH, Scott C, Konecny GE, Coleman RL, Giordano H, Brenton JD, Harding TC, Swisher EMet al., 2018, BRCA reversion mutations in circulating cell-free tumour DNA predict primary and acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma (HGOC), 30th EORTC-NCI-AACR Symposium, Publisher: ELSEVIER SCI LTD, Pages: E28-E28, ISSN: 0959-8049

Conference paper

Rust K, Spiliopoulou P, Tang CY, Bell C, Stirling D, Phang THF, Davidson R, Mackean M, Nussey F, Glasspool R, Reed N, Sadozye A, Porteous M, McGoldrick T, Ferguson M, Miedzybrodzka Z, McNeish IA, Gourley Cet al., 2018, Routine germline BRCA1 and BRCA2 testing in ovarian carcinoma patients: analysis of the Scottish real life experience, BJOG: An International Journal of Obstetrics and Gynaecology, Vol: 125, Pages: 1451-1548, ISSN: 1470-0328

Objective:To determine the rate of germline BRCA1 and BRCA2 mutations in Scottish ovarian cancer patients before and after a change in testing policy.Design:Retrospective cohort study.Setting:Four cancer/genetics centres in Scotland.Population:Ovarian cancer patients undergoing germline BRCA1 and BRCA2 (gBRCA1/2) gene sequencing before 2013 (‘old criteria’; selection based solely on family history), after 2013 (‘new criteria’; sequencing offered to newly presenting non-mucinous ovarian cancer patients) and the ‘prevalent population’ (who presented before 2013, were not eligible for sequencing under the old criteria but were sequenced under the new criteria).Methods:Clinicopathological and sequence data were collected before and for 18 months after this change in selection criteria.Main Outcome Measures:Frequency of germline BRCA1, BRCA2, RAD51C and RAD51D mutations.Results:Of 599 patients sequenced, 205, 236 and 158 were in the ‘old criteria’, ‘new criteria’ and ‘prevalent’ populations respectively. The frequency of gBRCA1/2 mutations was 30.7%, 13.1% and 12.7% respectively. The annual rate of gBRCA1/2 mutation detection was 4.2 before and 20.7 after the policy change. 48% (15/31) ‘new criteria’ patients with gBRCA1/2 mutations had a Manchester score <15 and would not have been offered sequencing based on family history criteria. In addition, 20 gBRCA1/2 patients were identified in the prevalent population. The prevalence of gBRCA1/2 mutations in patients >70 years was 8.2%.Conclusions:Sequencing all non-mucinous ovarian cancer patients produces much higher annual gBRCA1/2 mutation detection with the frequency of positive tests still exceeding the 10% threshold upon which many family history based models operate.

Journal article

Ennis D, Cooke S, Evers L, Dowson S, Chan MY, Paul J, Hirschowitz L, Glasspool R, Singh N, Bell S, Day E, Kiochman A, Wilkinson N, Beer P, Martin S, Millan D, Biankin AV, McNeish IAet al., 2018, The driver mutational landscape of ovarian squamous cell carcinomas arising in mature cystic teratoma, Publisher: BMJ PUBLISHING GROUP, Pages: 56-56, ISSN: 1048-891X

Conference paper

Clamp AR, McNeish IA, Dean A, Gallardo-Rincon D, Kim J-W, O'Donnell DM, Hook J, Blagden S, Brenton JD, Naik R, Perren TJ, Sundar S, Cook AD, James EC, Gabra H, Lord R, Hall M, Dark G, Kaplan RS, Ledermann JAet al., 2018, Response to neoadjuvant chemotherapy in ICON8: A GCIG phase III randomised trial evaluating weekly dose-dense chemotherapy integration in first-line epithelial ovarian/fallopian tube/primary peritoneal carcinoma (EOC) treatment, 43rd ESMO Congress (ESMO), Publisher: OXFORD UNIV PRESS, Pages: 336-336, ISSN: 0923-7534

Conference paper

Leary A, Ledermann JA, Oaknin A, Oza AM, Lorusso D, Colombo N, Dean A, Clamp A, Scambia G, Casado Herraez A, Amenedo Gancedo M, Maria Guerra E, Balmana J, Floquet A, Fong PC, Holloway RW, Poelcher M, Roxburgh P, Shapira-Frommer R, Tamberi S, Tinker AV, Cameron T, Maloney L, Goble S, Kristeleit RS, Swisher EM, McNeish IA, Coleman RLet al., 2018, Use of the Poly (ADP-Ribose) Polymerase Inhibitor Rucaparib in Women with Recurrent Ovarian Carcinoma with Endometrioid and Other Nonserous Histopathologic Subtypes, Publisher: BMJ PUBLISHING GROUP, Pages: 200-201, ISSN: 1048-891X

Conference paper

McNeish IA, Kondrashova O, Topp M, Nesic K, Lieschke E, Ho G-Y, Harrell MI, Zapparoli GV, Hadley A, Holian R, Boehm E, Heong V, Sanij E, Pearson RB, Krais JJ, Johnson N, McNally O, Ananda S, Alsop K, Hutt KJ, Kaufmann SH, Lin KK, Harding TC, Traficante N, DeFazio A, Bowtell DD, Swisher EM, Dobrovic A, Wakefield MJ, Scott CLet al., 2018, Methylation of all BRCA1 copies predicts response to the PARP inhibitor rucaparib in ovarian carcinoma, Nature Communications, Vol: 9, ISSN: 2041-1723

Accurately identifying patients with high-grade serous ovarian carcinoma (HGSOC) who respond to poly(ADP-ribose) polymerase inhibitor (PARPi) therapy is of great clinical importance. Here we show that quantitative BRCA1 methylation analysis provides new insight into PARPi response in preclinical models and ovarian cancer patients. The response of 12 HGSOC patient-derived xenografts (PDX) to the PARPi rucaparib was assessed, with variable dose-dependent responses observed in chemo-naïve BRCA1/2-mutated PDX, and no responses in PDX lacking DNA repair pathway defects. Among BRCA1-methylated PDX, silencing of all BRCA1 copies predicts rucaparib response, whilst heterozygous methylation is associated with resistance. Analysis of 21 BRCA1-methylated platinum-sensitive recurrent HGSOC (ARIEL2 Part 1 trial) confirmed that homozygous or hemizygous BRCA1 methylation predicts rucaparib clinical response, and that methylation loss can occur after exposure to chemotherapy. Accordingly, quantitative BRCA1methylation analysis in a pre-treatment biopsy could allow identification of patients most likely to benefit and facilitate tailoring of PARPi therapy.

Journal article

Monk BJ, Coleman RL, Fujiwara K, Oza AM, Westin SN, O'Malley DM, Herzog TJ, Marme F, Oaknin A, Eskander R, Lorusso D, Korach J, Lin KK, Shih D, Goble S, Grechko N, Hume S, Maloney L, McNeish I, Kristeleit RSet al., 2018, ATHENA (GOG-3020/ENGOT-OV45): A RANDOMISED, DOUBLE-BLIND, PLACEBO-CONTROLLED, PHASE 3 STUDY OF RUCAPARIB plus NIVOLUMAB FOLLOWING FRONT-LINE PLATINUM-BASED CHEMOTHERAPY IN OVARIAN CANCER, Publisher: BMJ PUBLISHING GROUP, Pages: 1019-1020, ISSN: 1048-891X

Conference paper

Farquharson M, Walton J, Dowson S, Ennis D, Mason S, Clark W, Bailey P, Upstill-Goddard R, Blyth K, McNeish Iet al., 2018, REVERSAL OF THE IMMUNOSUPPRESSIVE TUMOUR MICROENVIRONMENT INDUCED BY PTEN LOSS AFTER TREATMENT WITH A PI3Kβ INHIBITOR (AZD8186), Publisher: BMJ PUBLISHING GROUP, Pages: 153-153, ISSN: 1048-891X

Conference paper

Lu Y, Beeghly-Fadiel A, Wu L, Guo X, Li B, Schildkraut JM, Im HK, Chen YA, Permuth JB, Reid BM, Teer JK, Moysich KB, Andrulis IL, Anton-Culver H, Arun BK, Bandera EV, Barkardottir RB, Barnes DR, Benitez J, Bjørge L, Brenton J, Butzow R, Caldes T, Caligo MA, Campbell IG, Chang-Claude J, Claes KBM, Couch FJ, Cramer DW, Daly MB, DeFazio A, Dennis J, Díez O, Domchek SM, Dork T, Easton DF, Eccles DM, Fasching PA, Fortner RT, Fountzilas G, Friedman E, Ganz PA, Garber J, Giles GG, Godwin AK, Goldgar DE, Goodman MT, Greene MH, Gronwald J, Hamann U, Heitz F, Hildebrandt MAT, Høgdall CK, Hollestelle A, Hulick PJ, Huntsman DG, Imyanitov EN, Isaacs C, Jakubowska A, James P, Karlan BY, Kelemen LE, Kiemeney LA, Kjaer SK, Kwong A, Le ND, Leslie G, Lesueur F, Levine DA, Mattiello A, May T, McGuffog L, McNeish IA, Merritt MA, Modugno F, Montagna M, Neuhausen SL, Nevanlinna H, Cilius Nielsen FC, Nikitina-Zake L, Nussbaum RL, Offit K, Olah E, Olopade OI, Olson SH, Olsson H, Osorio A, Park SK, Parsons MT, Peeters PHM, Pejovic T, Peterlongo P, Phelan CM, Pujana MA, Ramus SJ, Rennert G, Risch H, Rodriguez GC, Rodríguez-Antona C, Romieu I, Rookus MA, Rossing MA, Rzepecka IK, Sandler DP, Schmutzler RK, Setiawan VW, Sharma P, Sieh W, Simard J, Singer CF, Song H, Southey MC, Spurdle AB, Sutphen R, Swerdlow AJ, Teixeira MR, Teo SH, Thomassen M, Tischkowitz M, Toland AE, Trichopoulou A, Tung N, Tworoger SS, van Rensburg EJ, Vanderstichele A, Vega A, Velez Edwards D, Webb PM, Weitzel JN, Wentzensen N, White E, Wolk A, Wu AH, Yannoukakos D, Zorn KK, Gayther SA, Antoniou AC, Berchuck A, Goode EL, Chenevix-Trench G, Sellers TA, Pharoah PDP, Zheng W, Long Jet al., 2018, A transcriptome-wide association study among 97,898 women to identify candidate susceptibility genes for epithelial ovarian cancer risk, Cancer Research, Vol: 78, Pages: 5419-5430, ISSN: 1538-7445

Large-scale genome-wide association studies (GWAS) have identified approximately 35 loci associated with epithelial ovarian cancer (EOC) risk. The majority of GWAS-identified disease susceptibility variants are located in non-coding regions, and causal genes underlying these associations remain largely unknown. Here we performed a transcriptome-wide association study to search for novel genetic loci and plausible causal genes at known GWAS loci. We used RNA sequencing data (68 normal ovarian-tissue samples from 68 individuals and 6,124 cross-tissue samples from 369 individuals) and high-density genotyping data from European descendants of the Genotype-Tissue Expression (GTEx V6) project to build ovarian and cross-tissue models of genetically regulated expression using elastic net methods. We evaluated 17,121 genes for their cis-predicted gene expression in relation to EOC risk using summary statistics data from GWAS of 97,898 women, including 29,396 EOC cases. With a Bonferroni-corrected significance level of P<2.2×10-6, we identified 35 genes including FZD4 at 11q14.2 (Z=5.08, P=3.83×10-7, the cross-tissue model; 1 Mb away from any GWAS-identified EOC risk variant), a potential novel locus for EOC risk. All other 34 significantly-associated genes were located within 1 Mb of known GWAS-identified loci, including 23 genes at 6 loci not previously linked to EOC risk. Upon conditioning on nearby known EOC GWAS-identified variants, the associations for 31 genes disappeared and 3 genes remained (P<1.47 x 10-3). These data identify one novel locus (FZD4) and 34 genes at 13 known EOC risk loci associated with EOC risk, providing new insights into EOC carcinogenesis.

Journal article

Kelemen LE, Earp M, Fridley BL, Chenevix-Trench G, Australian Ovarian Cancer Study Group, Fasching PA, Beckmann MW, Ekici AB, Hein A, Lambrechts D, Lambrechts S, Van Nieuwenhuysen E, Vergote I, Rossing MA, Doherty JA, Chang-Claude J, Behrens S, Moysich KB, Cannioto R, Lele S, Odunsi K, Goodman MT, Shvetsov YB, Thompson PJ, Wilkens LR, Dörk T, Antonenkova N, Bogdanova N, Hillemanns P, Runnebaum IB, du Bois A, Harter P, Heitz F, Schwaab I, Butzow R, Pelttari LM, Nevanlinna H, Modugno F, Edwards RP, Kelley JL, Ness RB, Karlan BY, Lester J, Orsulic S, Walsh C, Kjaer SK, Jensen A, Cunningham JM, Vierkant RA, Giles GG, Bruinsma F, Southey MC, Hildebrandt MAT, Liang D, Lu K, Wu X, Sellers TA, Levine DA, Schildkraut JM, Iversen ES, Terry KL, Cramer DW, Tworoger SS, Poole EM, Bandera EV, Olson SH, Orlow I, Vestrheim Thomsen LC, Bjorge L, Krakstad C, Tangen IL, Kiemeney LA, Aben KKH, Massuger LFAG, van Altena AM, Pejovic T, Bean Y, Kellar M, Cook LS, Le ND, Brooks-Wilson A, Gronwald J, Cybulski C, Jakubowska A, Lubiński J, Wentzensen N, Brinton LA, Lissowska J, Hogdall E, Engelholm SA, Hogdall C, Lundvall L, Nedergaard L, Pharoah PDP, Dicks E, Song H, Tyrer JP, McNeish I, Siddiqui N, Carty K, Glasspool R, Paul J, Campbell IG, Eccles D, Whittemore AS, McGuire V, Rothstein JH, Sieh W, Narod SA, Phelan CM, McLaughlin JR, Risch HA, Anton-Culver H, Ziogas A, Menon U, Gayther SA, Gentry-Maharaj A, Ramus SJ, Wu AH, Pearce CL, Lee AW, Pike MC, Kupryjanczyk J, Podgorska A, Plisiecka-Halasa J, Sawicki W, Goode EL, Berchuck A, Ovarian Cancer Association Consortiumet al., 2018, rs495139 in the TYMS-ENOSF1 region and risk of Ovarian carcinoma of mucinous histologo, International Journal of Molecular Sciences, Vol: 19, ISSN: 1422-0067

Thymidylate synthase (TYMS) is a crucial enzyme for DNA synthesis. TYMS expression is regulated by its antisense mRNA, ENOSF1. Disrupted regulation may promote uncontrolled DNA synthesis and tumor growth. We sought to replicate our previously reported association between rs495139 in the TYMS-ENOSF1 3' gene region and increased risk of mucinous ovarian carcinoma (MOC) in an independent sample. Genotypes from 24,351 controls to 15,000 women with invasive OC, including 665 MOC, were available. We estimated per-allele odds ratios (OR) and 95% confidence intervals (CI) using unconditional logistic regression, and meta-analysis when combining these data with our previous report. The association between rs495139 and MOC was not significant in the independent sample (OR = 1.09; 95% CI = 0.97⁻1.22; p = 0.15; N = 665 cases). Meta-analysis suggested a weak association (OR = 1.13; 95% CI = 1.03⁻1.24; p = 0.01; N = 1019 cases). No significant association with risk of other OC histologic types was observed (p = 0.05 for tumor heterogeneity). In expression quantitative trait locus (eQTL) analysis, the rs495139 allele was positively associated with ENOSF1 mRNA expression in normal tissues of the gastrointestinal system, particularly esophageal mucosa (r = 0.51, p = 1.7 × 10-28), and nonsignificantly in five MOC tumors. The association results, along with inconclusive tumor eQTL findings, suggest that a true effect of rs495139 might be small.

Journal article

Macintyre G, Goranova TE, De Silva D, Ennis D, Piskorz AM, Eldridge M, Sie D, Lewsley L-A, Hanif A, Wilson C, Dowson S, Glasspool RM, Lockley M, Brockbank E, Montes A, Walther A, Sundar SS, Edmondson R, Hall GD, Clamp A, Gourley C, Hall M, Fotopoulou C, Gabra H, Paul J, Supernat A, Millan D, Hoyle A, Bryson G, Nourse C, Mincarelli L, Navarro Sanchez L, Ylstra B, Jimenez-Linan M, Moore L, Hofmann O, Markowetz F, McNeish IA, Brenton JDet al., 2018, Copy-number signatures and mutational processes in ovarian carcinoma, Nature Genetics, Vol: 50, Pages: 1262-1270, ISSN: 1061-4036

The genomic complexity of profound copy-number aberration has prevented effective molecular stratification of ovarian cancers. To decode this complexity, we derived copy-number signatures from shallow whole genome sequencing of 117 high-grade serous ovarian cancer (HGSOC) cases, which were validated on 527 independent cases. We show that HGSOC comprises a continuum of genomes shaped by multiple mutational processes that result in known patterns of genomic aberration. Copy-number signature exposures at diagnosis predict both overall survival and the probability of platinum-resistant relapse. Measuring signature exposures provides a rational framework to choose combination treatments that target multiple mutational processes.

Journal article

Leung EYL, Ennis DP, Athineos D, Kennedy PR, Hansell CA, Blyth K, Davis DM, Graham GJ, McNeish IAet al., 2018, Oncolytic adenovirus infection leads to contact-dependent activation of natural killer cells and augments virotherapy effectiveness for ovarian cancer., AACR Special Conference on Addressing Critical Questions in Ovarian Cancer Research and Treatment, Publisher: AMER ASSOC CANCER RESEARCH, Pages: 97-98, ISSN: 1078-0432

Conference paper

Spiliopoulou P, Walton J, Dowson S, Binks A, Maddocks O, Adams P, McNeish Iet al., 2018, Epigenetic modification of ovarian cancer immunogenicity., AACR Special Conference on Addressing Critical Questions in Ovarian Cancer Research and Treatment, Publisher: AMER ASSOC CANCER RESEARCH, Pages: 57-58, ISSN: 1078-0432

Conference paper

Spiliopoulou P, Zubairi I, Gibson S, Brown J, McNeish I, Glasspool Ret al., 2018, Influence of germline BRCA mutation on response to oral cyclophosphamide in relapsed heavily pretreated ovarian cancer., AACR Special Conference on Addressing Critical Questions in Ovarian Cancer Research and Treatment, Publisher: AMER ASSOC CANCER RESEARCH, Pages: 82-83, ISSN: 1078-0432

Conference paper

Ennis D, Cooke SL, Evers L, Dowson S, Chan MY, Paul J, Hirschowitz L, Glasspool R, Singh N, Bell S, Day E, Kochman A, Wilkinson N, Beer P, Martin S, Millan D, Biankin AV, McNeish IAet al., 2018, The driver mutational landscape of ovarian squamous cell carcinomas arising in mature cystic teratoma., AACR Special Conference on Addressing Critical Questions in Ovarian Cancer Research and Treatment, Publisher: AMER ASSOC CANCER RESEARCH, Pages: 34-+, ISSN: 1078-0432

Conference paper

Earp M, Tyrer JP, Winham SJ, Lin H-Y, Chornokur G, Dennis J, Aben KKH, Anton-Culver H, Antonenkova N, Bandera EV, Bean YT, Beckmann MW, Bjorge L, Bogdanova N, Brinton LA, Brooks-Wilson A, Bruinsma F, Bunker CH, Butzow R, Campbell IG, Carty K, Chang-Claude J, Cook LS, Cramer DW, Cunningham JM, Cybulski C, Dansonka-Mieszkowska A, Despierre E, Doherty JA, Dörk T, du Bois A, Dürst M, Easton DF, Eccles DM, Edwards RP, Ekici AB, Fasching PA, Fridley BL, Gentry-Maharaj A, Giles GG, Glasspool R, Goodman MT, Gronwald J, Harter P, Hein A, Heitz F, Hildebrandt MAT, Hillemanns P, Hogdall CK, Høgdall E, Hosono S, Iversen ES, Jakubowska A, Jensen A, Ji B-T, Jung AY, Karlan BY, Kellar M, Kiemeney LA, Kiong Lim B, Kjaer SK, Krakstad C, Kupryjanczyk J, Lambrechts D, Lambrechts S, Le ND, Lele S, Lester J, Levine DA, Li Z, Liang D, Lissowska J, Lu K, Lubinski J, Lundvall L, Massuger LFAG, Matsuo K, McGuire V, McLaughlin JR, McNeish I, Menon U, Milne RL, Modugno F, Moysich KB, Ness RB, Nevanlinna H, Odunsi K, Olson SH, Orlow I, Orsulic S, Paul J, Pejovic T, Pelttari LM, Permuth JB, Pike MC, Poole EM, Rosen B, Rossing MA, Rothstein JH, Runnebaum IB, Rzepecka IK, Schernhammer E, Schwaab I, Shu X-O, Shvetsov YB, Siddiqui N, Sieh W, Song H, Southey MC, Spiewankiewicz B, Sucheston-Campbell L, Tangen IL, Teo S-H, Terry KL, Thompson PJ, Thomsen L, Tworoger SS, van Altena AM, Vergote I, Vestrheim Thomsen LC, Vierkant RA, Walsh CS, Wang-Gohrke S, Wentzensen N, Whittemore AS, Wicklund KG, Wilkens LR, Woo Y-L, Wu AH, Wu X, Xiang Y-B, Yang H, Zheng W, Ziogas A, Lee AW, Pearce CL, Berchuck A, Schildkraut JM, Ramus SJ, Monteiro ANA, Narod SA, Sellers TA, Gayther SA, Kelemen LE, Chenevix-Trench G, Risch HA, Pharoah PDP, Goode EL, Phelan CMet al., 2018, Variants in genes encoding small GTPases and association with epithelial ovarian cancer susceptibility, PLoS ONE, Vol: 13, Pages: 1-13, ISSN: 1932-6203

Epithelial ovarian cancer (EOC) is the fifth leading cause of cancer mortality in American women. Normal ovarian physiology is intricately connected to small GTP binding proteins of the Ras superfamily (Ras, Rho, Rab, Arf, and Ran) which govern processes such as signal transduction, cell proliferation, cell motility, and vesicle transport. We hypothesized that common germline variation in genes encoding small GTPases is associated with EOC risk. We investigated 322 variants in 88 small GTPase genes in germline DNA of 18,736 EOC patients and 26,138 controls of European ancestry using a custom genotype array and logistic regression fitting log-additive models. Functional annotation was used to identify biofeatures and expression quantitative trait loci that intersect with risk variants. One variant, ARHGEF10L (Rho guanine nucleotide exchange factor 10 like) rs2256787, was associated with increased endometrioid EOC risk (OR=1.33, p=4.46 x 10-6). Other variants of interest included another in ARHGEF10L, rs10788679, which was associated with invasive serous EOC risk (OR=1.07, p=0.00026) and two variants in AKAP6 (A-kinase anchoring protein 6) which were associated with risk of invasive EOC (rs1955513, OR=0.90, p=0.00033; rs927062, OR =0.94, p=0.00059). Functional annotation revealed that the two ARHGEF10L variants were located in super-enhancer regions and that AKAP6 rs927062 was associated with expression of GTPase gene ARHGAP5 (Rho GTPase activating protein 5). Inherited variants in ARHGEF10L and AKAP6, with potential transcriptional regulatory function and association with EOC risk, warrant investigation in independent EOC study populations.

Journal article

Oaknin A, Ledermann JA, Oza AM, Lorusso D, Aghajanian C, Dean AP, Colombo N, Weberpals JI, Clamp AR, Scambia G, Leary A, Holloway RW, O'Malley DM, McNeish IA, Swisher EM, Cameron T, Goble S, Sun J, Lin KK, Coleman RLet al., 2018, Exploratory analysis of percentage of genomic loss of heterozygosity (LOH) in patients with platinum-sensitive recurrent ovarian carcinoma (rOC) in ARIEL3, Publisher: AMER SOC CLINICAL ONCOLOGY, ISSN: 0732-183X

Conference paper

Walton JB, Farquharson M, Mason S, Port J, Kruspig B, Dowson S, Stevenson D, Murphy D, Matzuk M, Kim J, Coffelt S, Blyth K, McNeish IAet al., 2018, Author correction: CRISPR/Cas9-derived models of ovarian high grade serous carcinoma targeting Brca1, Pten and Nf1, and correlation with platinum sensitivity, Scientific Reports, Vol: 8, ISSN: 2045-2322

Journal article

Nesic K, Wakefield M, Kondrashova O, Scott CL, McNeish IAet al., 2018, Targeting DNA repair: the genome as a potential biomarker, Journal of Pathology, Vol: 244, Pages: 586-597, ISSN: 0022-3417

Genomic instability and mutations are fundamental aspects of human malignancies, leading to progressive accumulation of the hallmarks of cancer. For some time, it has been clear that key mutations may be used both as prognostic and predictive biomarkers, the best-known examples being the presence of germline BRCA1 or BRCA2 mutations, which are not only associated with improved prognosis in ovarian cancer, but are also predictive of response to poly(ADP-ribose) polymerase (PARP) inhibitors. Although biomarkers as specific and powerful as these are rare in human malignancies, next generation sequencing and improved bioinformatic analyses are revealing mutational signatures, broader patterns of alterations in the cancer genome that have the power to reveal information about underlying driver mutational processes. Thus, the cancer genome can act as a stratification factor in clinical trials and, ultimately, will be used to drive personalised treatment decisions. In this review, we will use ovarian high grade serous carcinoma (HGSC) as an example of a disease of extreme genomic complexity that is marked by widespread copy number alterations, but which lacks powerful driver oncogene mutations. Understanding of the genomics of HGSC has led to the routine introduction of germline and somatic BRCA1/2 testing, as well as testing of mutations in other homologous recombination genes, widening the range of patients who may benefit from PARP inhibitors. We will discuss how whole genome-wide analyses, including loss of heterozygosity quantification and whole genome sequencing, may extend this paradigm to allow all patients to benefit from effective targeted therapies.

Journal article

Ong J-S, Hwang L-D, Cuellar-Partida G, Martin NG, Chenevix-Trench G, Quinn MCJ, Cornelis MC, Gharahkhani P, Webb PM, MacGregor Set al., 2018, Assessment of moderate coffee consumption and risk of epithelial ovarian cancer: a Mendelian randomization study, INTERNATIONAL JOURNAL OF EPIDEMIOLOGY, Vol: 47, Pages: 450-459, ISSN: 0300-5771

Journal article

This data is extracted from the Web of Science and reproduced under a licence from Thomson Reuters. You may not copy or re-distribute this data in whole or in part without the written consent of the Science business of Thomson Reuters.

Request URL: http://wlsprd.imperial.ac.uk:80/respub/WEB-INF/jsp/search-html.jsp Request URI: /respub/WEB-INF/jsp/search-html.jsp Query String: limit=30&id=00401895&person=true&page=5&respub-action=search.html