D. Timothy Bishop

D. Timothy  Bishop

University of Leeds

H-index: 112

Europe-United Kingdom

About D. Timothy Bishop

D. Timothy Bishop, With an exceptional h-index of 112 and a recent h-index of 54 (since 2020), a distinguished researcher at University of Leeds, specializes in the field of genetic epidemiology, melanoma.

His recent articles reflect a diverse array of research interests and contributions to the field:

The tumor suppressive role of methylthioadenosine phosphorylase in melanoma

Genome-Wide Gene–Environment Interaction Analyses to Understand the Relationship between Red Meat and Processed Meat Intake and Colorectal Cancer Risk

Fine-mapping analysis including over 254,000 East Asian and European descendants identifies 136 putative colorectal cancer susceptibility genes

Genome-wide association study and Mendelian randomization analyses provide insights into the causes of early-onset colorectal cancer

Systemic inflammation, the peripheral blood transcriptome and primary melanoma

Observational and genetic associations between cardiorespiratory fitness and cancer: a UK Biobank and international consortia study

Genetic risk impacts the association of menopausal hormone therapy with colorectal cancer risk

Immune subtyping of melanoma whole slide images using multiple instance learning

D. Timothy Bishop Information

University

University of Leeds

Position

Professor of Epidemiology

Citations(all)

56054

Citations(since 2020)

11653

Cited By

48097

hIndex(all)

112

hIndex(since 2020)

54

i10Index(all)

383

i10Index(since 2020)

186

Email

University Profile Page

University of Leeds

D. Timothy Bishop Skills & Research Interests

genetic epidemiology

melanoma

Top articles of D. Timothy Bishop

The tumor suppressive role of methylthioadenosine phosphorylase in melanoma

Authors

Jennifer J Hsiao,Pedro Andreu-Perez,Lajos V Kemény,Miroslav Hejna,Jérémie Nsengimana,Jennifer Allouche,Abdullah Al Emran,D Timothy Bishop,Göran B Jönsson,Julia Newton-Bishop,Jun Song,David E Fisher

Journal

Cancer Research

Published Date

2024/3/22

Methylthioadenosine phosphorylase (MTAP) is an enzyme that is expressed in virtually all tissues but lost in many cancers. MTAP deficiency can be due to either deletion of the MTAP gene or methylation of the MTAP promoter. In normal cells, MTAP catalyzes the conversion of methylthioadenosine (MTA), produced during polyamine biosynthesis, to adenine and 5-methylthioribose-1-phosphate, which is subsequently converted to methionine. MTAP deficiency results in intracellular accumulation of MTA. Although recent genome wide association studies (GWAS) have associated the MTAP locus with melanoma risk, the molecular mechanisms linking MTAP loss to increased tumorigenesis are not yet fully understood. In this study, we hypothesized that loss of MTAP and the resulting accumulation of MTA would dysregulate microphthalmia transcription factor (MITF), a master transcriptional regulator of melanocytes …

Genome-Wide Gene–Environment Interaction Analyses to Understand the Relationship between Red Meat and Processed Meat Intake and Colorectal Cancer Risk

Authors

Mariana C Stern,Joel Sanchez Mendez,Andre E Kim,Mireia Obón-Santacana,Ferran Moratalla-Navarro,Vicente Martín,Victor Moreno,Yi Lin,Stephanie A Bien,Conghui Qu,Yu-Ru Su,Emily White,Tabitha A Harrison,Jeroen R Huyghe,Catherine M Tangen,Polly A Newcomb,Amanda I Phipps,Claire E Thomas,Eric S Kawaguchi,Juan Pablo Lewinger,John L Morrison,David V Conti,Jun Wang,Duncan C Thomas,Elizabeth A Platz,Kala Visvanathan,Temitope O Keku,Christina C Newton,Caroline Y Um,Anshul Kundaje,Anna Shcherbina,Neil Murphy,Marc J Gunter,Niki Dimou,Nikos Papadimitriou,Stéphane Bézieau,Franzel JB van Duijnhoven,Satu Männistö,Gad Rennert,Alicja Wolk,Michael Hoffmeister,Hermann Brenner,Jenny Chang-Claude,Yu Tian,Loïc Le Marchand,Michelle Cotterchio,Konstantinos K Tsilidis,D Timothy Bishop,Yohannes Adama Melaku,Brigid M Lynch,Daniel D Buchanan,Cornelia M Ulrich,Jennifer Ose,Anita R Peoples,Andrew J Pellatt,Li Li,Matthew AM Devall,Peter T Campbell,Demetrius Albanes,Stephanie J Weinstein,Sonja I Berndt,Stephen B Gruber,Edward Ruiz-Narvaez,Mingyang Song,Amit D Joshi,David A Drew,Jessica L Petrick,Andrew T Chan,Marios Giannakis,Ulrike Peters,Li Hsu,W James Gauderman

Journal

Cancer Epidemiology, Biomarkers & Prevention

Published Date

2024/3/1

Background High red meat and/or processed meat consumption are established colorectal cancer risk factors. We conducted a genome-wide gene–environment (GxE) interaction analysis to identify genetic variants that may modify these associations. Methods A pooled sample of 29,842 colorectal cancer cases and 39,635 controls of European ancestry from 27 studies were included. Quantiles for red meat and processed meat intake were constructed from harmonized questionnaire data. Genotyping arrays were imputed to the Haplotype Reference Consortium. Two-step EDGE and joint tests of GxE interaction were utilized in our genome-wide scan. Results Meta-analyses confirmed positive associations between increased consumption of red meat and processed meat with colorectal cancer risk [per quartile red meat OR = 1.30; 95% confidence interval …

Fine-mapping analysis including over 254,000 East Asian and European descendants identifies 136 putative colorectal cancer susceptibility genes

Authors

Zhishan Chen,Xingyi Guo,Ran Tao,Jeroen R Huyghe,Philip J Law,Ceres Fernandez-Rozadilla,Jie Ping,Guochong Jia,Jirong Long,Chao Li,Quanhu Shen,Yuhan Xie,Maria N Timofeeva,Minta Thomas,Stephanie L Schmit,Virginia Díez-Obrero,Matthew Devall,Ferran Moratalla-Navarro,Juan Fernandez-Tajes,Claire Palles,Kitty Sherwood,Sarah EW Briggs,Victoria Svinti,Kevin Donnelly,Susan M Farrington,James Blackmur,Peter G Vaughan-Shaw,Xiao-Ou Shu,Yingchang Lu,Peter Broderick,James Studd,Tabitha A Harrison,David V Conti,Fredrick R Schumacher,Marilena Melas,Gad Rennert,Mireia Obón-Santacana,Vicente Martín-Sánchez,Jae Hwan Oh,Jeongseon Kim,Sun Ha Jee,Keum Ji Jung,Sun-Seog Kweon,Min-Ho Shin,Aesun Shin,Yoon-Ok Ahn,Dong-Hyun Kim,Isao Oze,Wanqing Wen,Keitaro Matsuo,Koichi Matsuda,Chizu Tanikawa,Zefang Ren,Yu-Tang Gao,Wei-Hua Jia,John L Hopper,Mark A Jenkins,Aung Ko Win,Rish K Pai,Jane C Figueiredo,Robert W Haile,Steven Gallinger,Michael O Woods,Polly A Newcomb,David Duggan,Jeremy P Cheadle,Richard Kaplan,Rachel Kerr,David Kerr,Iva Kirac,Jan Böhm,Jukka-Pekka Mecklin,Pekka Jousilahti,Paul Knekt,Lauri A Aaltonen,Harri Rissanen,Eero Pukkala,Johan G Eriksson,Tatiana Cajuso,Ulrika Hänninen,Johanna Kondelin,Kimmo Palin,Tomas Tanskanen,Laura Renkonen-Sinisalo,Satu Männistö,Demetrius Albanes,Stephanie J Weinstein,Edward Ruiz-Narvaez,Julie R Palmer,Daniel D Buchanan,Elizabeth A Platz,Kala Visvanathan,Cornelia M Ulrich,Erin Siegel,Stefanie Brezina,Andrea Gsur,Peter T Campbell,Jenny Chang-Claude,Michael Hoffmeister,Hermann Brenner,Martha L Slattery,John D Potter,Kostas K Tsilidis,Matthias B Schulze,Marc J Gunter,Neil Murphy,Antoni Castells,Sergi Castellví-Bel,Leticia Moreira,Volker Arndt,Anna Shcherbina,D Timothy Bishop,Graham G Giles,Melissa C Southey,Gregory E Idos,Kevin J McDonnell,Zomoroda Abu-Ful,Joel K Greenson,Katerina Shulman,Flavio Lejbkowicz,Kenneth Offit,Yu-Ru Su,Robert Steinfelder,Temitope O Keku,Bethany van Guelpen,Thomas J Hudson,Heather Hampel,Rachel Pearlman,Sonja I Berndt,Richard B Hayes,Marie Elena Martinez,Sushma S Thomas,Paul DP Pharoah,Susanna C Larsson,Yun Yen,Heinz-Josef Lenz,Emily White,Li Li,Kimberly F Doheny,Elizabeth Pugh,Tameka Shelford,Andrew T Chan,Marcia Cruz-Correa,Annika Lindblom,David J Hunter,Amit D Joshi,Clemens Schafmayer,Peter C Scacheri,Anshul Kundaje,Robert E Schoen

Journal

Nature Communications

Published Date

2024/4/26

Genome-wide association studies (GWAS) have identified more than 200 common genetic variants independently associated with colorectal cancer (CRC) risk, but the causal variants and target genes are mostly unknown. We sought to fine-map all known CRC risk loci using GWAS data from 100,204 cases and 154,587 controls of East Asian and European ancestry. Our stepwise conditional analyses revealed 238 independent association signals of CRC risk, each with a set of credible causal variants (CCVs), of which 28 signals had a single CCV. Our cis-eQTL/mQTL and colocalization analyses using colorectal tissue-specific transcriptome and methylome data separately from 1299 and 321 individuals, along with functional genomic investigation, uncovered 136 putative CRC susceptibility genes, including 56 genes not previously reported. Analyses of single-cell RNA-seq data from colorectal tissues revealed 17 …

Genome-wide association study and Mendelian randomization analyses provide insights into the causes of early-onset colorectal cancer

Authors

RS Laskar,C Qu,JR Huyghe,T Harrison,RB Hayes,Y Cao,PT Campbell,R Steinfelder,FR Talukdar,H Brenner,S Ogino,S Brendt,DT Bishop,DD Buchanan,AT Chan,M Cotterchio,SB Gruber,A Gsur,B van Guelpen,MA Jenkins,TO Keku,BM Lynch,L Le Marchand,RM Martin,K McCarthy,V Moreno,R Pearlman,M Song,KK Tsilidis,P Vodička,MO Woods,K Wu,L Hsu,MJ Gunter,U Peters,N Murphy,Colorectal Transdisciplinary CORECT Study,Colon Cancer Family Registry,Genetics and Epidemiology of Colorectal Cancer Consortium

Journal

Annals of Oncology

Published Date

2024/2/24

BackgroundThe incidence of early-onset colorectal cancer (EOCRC; diagnosed <50 years of age) is rising globally; however, the causes underlying this trend are largely unknown. Colorectal cancer (CRC) has strong genetic and environmental determinants, yet common genetic variants and causal modifiable risk factors underlying EOCRC are unknown. We conducted the first EOCRC-specific genome-wide association study (GWAS) and Mendelian randomization analyses to explore germline genetic and causal modifiable risk factors associated with EOCRC.Patients and methodsWe conducted a genome-wide association study meta-analysis of 6,176 EOCRC cases and 65,829 controls from Genetics and Epidemiology of Colorectal Cancer Consortium (GECCO), Colorectal Transdisciplinary study (CORECT), Colon Cancer Family Registry (CCFR), and UK Biobank. We then used the EOCRC GWAS to investigate …

Systemic inflammation, the peripheral blood transcriptome and primary melanoma

Authors

Juliette Randerson-Moor,John Davies,Mark Harland,Jérémie Nsengimana,Theophile Bigirumurame,Christopher Walker,Jon Laye,S Elizabeth,Graham Ball,Graham P Cook,D Timothy Bishop,Robert J Salmond,Julia Newton-Bishop

Journal

Journal of Investigative Dermatology

Published Date

2024/4/5

Peripheral blood transcriptomes from 383 newly-diagnosed melanoma patients were subjected to differential gene expression analysis. The hypotheses were that impaired systemic immunity is associated with poorer prognosis (thicker tumors and fewer tumor infiltrating lymphocytes (TILs)) and evidence of systemic inflammation (high-sensitivity C-reactive protein (hsCRP) and fibrinogen levels). Higher fibrinogen levels were associated with thicker primaries. In single gene analysis hsCRP levels were significantly associated with higher blood CD274 expression, (coding for PD-L1), but each was independently prognostic, with hsCRP associated with increased mortality, and higher CD274 protective, independent of age. Pathway analysis identified downregulation of immune cell signalling pathways in the blood of people with thicker tumors and notable upregulation of STAT1 in people with brisk TILs …

Observational and genetic associations between cardiorespiratory fitness and cancer: a UK Biobank and international consortia study

Authors

Eleanor L Watts,Tomas I Gonzales,Tessa Strain,Pedro F Saint-Maurice,D Timothy Bishop,Stephen J Chanock,Mattias Johansson,Temitope O Keku,Loic Le Marchand,Victor Moreno,Polly A Newcomb,Christina C Newton,Rish K Pai,Mark P Purdue,Cornelia M Ulrich,Karl Smith-Byrne,Bethany Van Guelpen,Henrik Grönberg,Nora Pashayan,Johanna Schleutker,Demetrius Albanes,Stephanie J Weinstein,Alicja Wolk,Catharine ML West,Lorelei A Mucci,Géraldine Cancel-Tassin,Stella Koutros,Karina Dalsgaard Sørensen,Eli Marie Grindedal,David E Neal,Freddie C Hamdy,Jenny L Donovan,Ruth C Travis,Robert J Hamilton,Sue Ann Ingles,Barry S Rosenstein,Yong-Jie Lu,Graham G Giles,Robert J MacInnis,Adam S Kibel,Ana Vega,Manolis Kogevinas,Kathryn L Penney,Jong Y Park,Janet L Stanford,Cezary Cybulski,Børge G Nordestgaard,Sune F Nielsen,Hermann Brenner,Christiane Maier,Jeri Kim,Esther M John,Manuel R Teixeira,Susan L Neuhausen,Kim De Ruyck,Azad Razack,Lisa F Newcomb,Davor Lessel,Radka Kaneva,Nawaid Usmani,Frank Claessens,Paul A Townsend,Jose Esteban Castelao,Monique J Roobol,Florence Menegaux,Kay-Tee Khaw,Lisa Cannon-Albright,Hardev Pandha,Stephen N Thibodeau,David J Hunter,Peter Kraft,William J Blot,Elio Riboli

Journal

British Journal of Cancer

Published Date

2024/1/31

BackgroundThe association of fitness with cancer risk is not clear.MethodsWe used Cox proportional hazards models to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for risk of lung, colorectal, endometrial, breast, and prostate cancer in a subset of UK Biobank participants who completed a submaximal fitness test in 2009-12 (N = 72,572). We also investigated relationships using two-sample Mendelian randomisation (MR), odds ratios (ORs) were estimated using the inverse-variance weighted method.ResultsAfter a median of 11 years of follow-up, 4290 cancers of interest were diagnosed. A 3.5 ml O2⋅min−1⋅kg−1 total-body mass increase in fitness (equivalent to 1 metabolic equivalent of task (MET), approximately 0.5 standard deviation (SD)) was associated with lower risks of endometrial (HR = 0.81, 95% CI: 0.73–0.89), colorectal (0.94, 0.90–0.99), and breast cancer (0.96, 0.92–0.99). In …

Genetic risk impacts the association of menopausal hormone therapy with colorectal cancer risk

Authors

Yu Tian,Yi Lin,Conghui Qu,Volker Arndt,James W Baurley,Sonja I Berndt,Stephanie A Bien,D Timothy Bishop,Hermann Brenner,Daniel D Buchanan,Arif Budiarto,Peter T Campbell,Robert Carreras-Torres,Graham Casey,Andrew T Chan,Rui Chen,Xuechen Chen,David V Conti,Virginia Díez-Obrero,Niki Dimou,David A Drew,Jane C Figueiredo,Steven Gallinger,Graham G Giles,Stephen B Gruber,Marc J Gunter,Sophia Harlid,Tabitha A Harrison,Akihisa Hidaka,Michael Hoffmeister,Jeroen R Huyghe,Mark A Jenkins,Kristina M Jordahl,Amit D Joshi,Temitope O Keku,Eric Kawaguchi,Andre E Kim,Anshul Kundaje,Susanna C Larsson,Loic Le Marchand,Juan Pablo Lewinger,Li Li,Victor Moreno,John Morrison,Neil Murphy,Hongmei Nan,Rami Nassir,Polly A Newcomb,Mireia Obón-Santacana,Shuji Ogino,Jennifer Ose,Bens Pardamean,Andrew J Pellatt,Anita R Peoples,Elizabeth A Platz,John D Potter,Ross L Prentice,Gad Rennert,Edward A Ruiz-Narvaez,Lori C Sakoda,Robert E Schoen,Anna Shcherbina,Mariana C Stern,Yu-Ru Su,Stephen N Thibodeau,Duncan C Thomas,Konstantinos K Tsilidis,Franzel JB van Duijnhoven,Bethany Van Guelpen,Kala Visvanathan,Emily White,Alicja Wolk,Michael O Woods,Anna H Wu,Ulrike Peters,W James Gauderman,Li Hsu,Jenny Chang-Claude

Journal

British journal of cancer

Published Date

2024/4/1

BackgroundMenopausal hormone therapy (MHT), a common treatment to relieve symptoms of menopause, is associated with a lower risk of colorectal cancer (CRC). To inform CRC risk prediction and MHT risk-benefit assessment, we aimed to evaluate the joint association of a polygenic risk score (PRS) for CRC and MHT on CRC risk.MethodsWe used data from 28,486 postmenopausal women (11,519 cases and 16,967 controls) of European descent. A PRS based on 141 CRC-associated genetic variants was modeled as a categorical variable in quartiles. Multiplicative interaction between PRS and MHT use was evaluated using logistic regression. Additive interaction was measured using the relative excess risk due to interaction (RERI). 30-year cumulative risks of CRC for 50-year-old women according to MHT use and PRS were calculated.ResultsThe reduction in odds ratios by MHT use was larger in women …

Immune subtyping of melanoma whole slide images using multiple instance learning

Authors

Lucy Godson,Navid Alemi,Jérémie Nsengimana,Graham P Cook,Emily L Clarke,Darren Treanor,D Timothy Bishop,Julia Newton-Bishop,Ali Gooya,Derek Magee

Journal

Medical Image Analysis

Published Date

2024/4/1

Determining early-stage prognostic markers and stratifying patients for effective treatment are two key challenges for improving outcomes for melanoma patients. Previous studies have used tumour transcriptome data to stratify patients into immune subgroups, which were associated with differential melanoma specific survival and potential predictive biomarkers. However, acquiring transcriptome data is a time-consuming and costly process. Moreover, it is not routinely used in the current clinical workflow. Here, we attempt to overcome this by developing deep learning models to classify gigapixel haematoxylin and eosin (H&E) stained pathology slides, which are well established in clinical workflows, into these immune subgroups. We systematically assess six different multiple instance learning (MIL) frameworks, using five different image resolutions and three different feature extraction methods. We show that …

Multi-level Graph Representations of Melanoma Whole Slide Images for Identifying Immune Subgroups

Authors

Lucy Godson,Navid Alemi,Jérémie Nsengimana,Graham P Cook,Emily L Clarke,Darren Treanor,D Timothy Bishop,Julia Newton-Bishop,Derek Magee

Published Date

2023/10/8

Stratifying melanoma patients into immune subgroups is important for understanding patient outcomes and treatment options. Current weakly supervised classification methods often involve dividing digitised whole slide images into patches, which leads to the loss of important contextual diagnostic information. Here, we propose using graph attention neural networks, which utilise graph representations of whole slide images, to introduce context to classifications. In addition, we present a novel hierarchical graph approach, which leverages histopathological features from multiple resolutions to improve on state-of-the-art (SOTA) multiple instance learning (MIL) methods. We achieve a mean test area under the curve metric of 0.80 for classifying low and high immune melanoma subtypes, using multi-level and 20x patch graph representations of whole slide images, compared to 0.77 when using SOTA MIL methods …

Population-based analysis of POT1 variants in a cutaneous melanoma case–control cohort

Authors

Irving Simonin-Wilmer,Raul Ossio,Emmett M Leddin,Mark Harland,Karen A Pooley,Mauricio Gerardo Martil de la Garza,Sofia Obolenski,James Hewinson,Chi C Wong,Vivek Iyer,John C Taylor,Julia A Newton-Bishop,D Timothy Bishop,Gerardo Andrés Cisneros,Mark M Iles,David J Adams,Carla Daniela Robles-Espinoza

Journal

Journal of medical genetics

Published Date

2023/7/1

Pathogenic germline variants in the protection of telomeres 1 gene (POT1) have been associated with predisposition to a range of tumour types, including melanoma, glioma, leukaemia and cardiac angiosarcoma. We sequenced all coding exons of the POT1 gene in 2928 European-descent melanoma cases and 3298 controls, identifying 43 protein-changing genetic variants. We performed POT1-telomere binding assays for all missense and stop-gained variants, finding nine variants that impair or disrupt protein–telomere complex formation, and we further define the role of variants in the regulation of telomere length and complex formation through molecular dynamics simulations. We determine that POT1 coding variants are a minor contributor to melanoma burden in the general population, with only about 0.5% of melanoma cases carrying germline pathogenic variants in this gene, but should be screened in …

Supplementary Figure from Cancer Prevention with Resistant Starch in Lynch Syndrome Patients in the CAPP2-Randomized Placebo Controlled Trial: Planned 10-Year Follow-up

Authors

John C Mathers,Faye Elliott,Finlay Macrae,Jukka-Pekka Mecklin,Gabriela Möslein,Fiona E McRonald,Lucio Bertario,D Gareth Evans,Anne-Marie Gerdes,Judy WC Ho,Annika Lindblom,Patrick J Morrison,Jem Rashbass,Raj S Ramesar,Toni T Seppälä,Huw JW Thomas,Harsh J Sheth,Kirsi Pylvänäinen,Lynn Reed,Gillian M Borthwick,D Timothy Bishop,John Burn

Published Date

2023/4/3

Supplementary Figure from Cancer Prevention with Resistant Starch in Lynch Syndrome Patients in the CAPP2-Randomized Placebo Controlled Trial: Planned 10-Year Follow-up — Research Explorer The University of Manchester Skip to main navigation Skip to search Skip to main content Research Explorer The University of Manchester Home Research Explorer The University of Manchester Logo Home Research Profiles Research units Research output Projects Impacts Activities Press/Media Prizes Equipment Datasets Student theses Search by expertise, name or affiliation Supplementary Figure from Cancer Prevention with Resistant Starch in Lynch Syndrome Patients in the CAPP2-Randomized Placebo Controlled Trial: Planned 10-Year Follow-up John C. Mathers, Faye Elliott, Finlay Macrae, Jukka-Pekka Mecklin, Gabriela Möslein, Fiona E. McRonald, Lucio Bertario, D. Gareth Evans, Anne-Marie Gerdes, …

Combining Asian and European genome-wide association studies of colorectal cancer improves risk prediction across racial and ethnic populations

Authors

Minta Thomas,Yu-Ru Su,Elisabeth A Rosenthal,Lori C Sakoda,Stephanie L Schmit,Maria N Timofeeva,Zhishan Chen,Ceres Fernandez-Rozadilla,Philip J Law,Neil Murphy,Robert Carreras-Torres,Virginia Diez-Obrero,Franzel JB Van Duijnhoven,Shangqing Jiang,Aesun Shin,Alicja Wolk,Amanda I Phipps,Andrea Burnett-Hartman,Andrea Gsur,Andrew T Chan,Ann G Zauber,Anna H Wu,Annika Lindblom,Caroline Y Um,Catherine M Tangen,Chris Gignoux,Christina Newton,Christopher A Haiman,Conghui Qu,D Timothy Bishop,Daniel D Buchanan,David R Crosslin,David V Conti,Dong-Hyun Kim,Elizabeth Hauser,Emily White,Erin Siegel,Fredrick R Schumacher,Gad Rennert,Graham G Giles,Heather Hampel,Hermann Brenner,Isao Oze,Jae Hwan Oh,Jeffrey K Lee,Jennifer L Schneider,Jenny Chang-Claude,Jeongseon Kim,Jeroen R Huyghe,Jiayin Zheng,Jochen Hampe,Joel Greenson,John L Hopper,Julie R Palmer,Kala Visvanathan,Keitaro Matsuo,Koichi Matsuda,Keum Ji Jung,Li Li,Loic Le Marchand,Ludmila Vodickova,Luis Bujanda,Marc J Gunter,Marco Matejcic,Mark A Jenkins,Martha L Slattery,Mauro D’amato,Meilin Wang,Michael Hoffmeister,Michael O Woods,Michelle Kim,Mingyang Song,Motoki Iwasaki,Mulong Du,Natalia Udaltsova,Norie Sawada,Pavel Vodicka,Peter T Campbell,Polly A Newcomb,Qiuyin Cai,Rachel Pearlman,Rish K Pai,Robert E Schoen,Robert S Steinfelder,Robert W Haile,Rosita Vandenputtelaar,Ross L Prentice,Sébastien Küry,Sergi Castellví-Bel,Shoichiro Tsugane,Sonja I Berndt,Soo Chin Lee,Stefanie Brezina,Stephanie J Weinstein,Stephen J Chanock,Sun Ha Jee,Sun-Seog Kweon,Susan Vadaparampil,Tabitha A Harrison,Taiki Yamaji,Temitope O Keku,Veronika Vymetalkova,Volker Arndt,Wei-Hua Jia,Xiao-Ou Shu,Yi Lin,Yoon-Ok Ahn,Zsofia K Stadler,Bethany Van Guelpen,Cornelia M Ulrich,Elizabeth A Platz,John D Potter,Christopher I Li,Reinier Meester,Victor Moreno,Jane C Figueiredo,Graham Casey,Iris Lansdorp Vogelaar,Malcolm G Dunlop,Stephen B Gruber,Richard B Hayes,Paul DP Pharoah,Richard S Houlston,Gail P Jarvik,Ian P Tomlinson,Wei Zheng,Douglas A Corley,Ulrike Peters,Li Hsu

Journal

Nature communications

Published Date

2023/10/2

Polygenic risk scores (PRS) have great potential to guide precision colorectal cancer (CRC) prevention by identifying those at higher risk to undertake targeted screening. However, current PRS using European ancestry data have sub-optimal performance in non-European ancestry populations, limiting their utility among these populations. Towards addressing this deficiency, we expand PRS development for CRC by incorporating Asian ancestry data (21,731 cases; 47,444 controls) into European ancestry training datasets (78,473 cases; 107,143 controls). The AUC estimates (95% CI) of PRS are 0.63(0.62-0.64), 0.59(0.57-0.61), 0.62(0.60-0.63), and 0.65(0.63-0.66) in independent datasets including 1681-3651 cases and 8696-115,105 controls of Asian, Black/African American, Latinx/Hispanic, and non-Hispanic White, respectively. They are significantly better than the European-centric PRS in all four major US …

Genome-wide analyses characterize shared heritability among cancers and identify novel cancer susceptibility regions

Authors

Sara Lindström,Lu Wang,Helian Feng,Arunabha Majumdar,Sijia Huo,James Macdonald,Tabitha Harrison,Constance Turman,Hongjie Chen,Nicholas Mancuso,Theo Bammler,Breast Cancer Association Consortium (BCAC),Steve Gallinger,Stephen B Gruber,Marc J Gunter,Loic Le Marchand,Victor Moreno,Kenneth Offit,Colorectal Transdisciplinary Study (CORECT),Colon Cancer Family Registry Study (CCFR),Genetics And Epidemiology Of Colorectal Cancer Consortium (GECCO),Immaculata De Vivo,Tracy A O’Mara,Amanda B Spurdle,Ian Tomlinson,Endometrial Cancer Association Consortium (ECAC),Rebecca Fitzgerald,Puya Gharahkhani,Ines Gockel,Janusz Jankowski,Stuart Macgregor,Johannes Schumacher,Jill Barnholtz-Sloan,Melissa L Bondy,Richard S Houlston,Robert B Jenkins,Beatrice Melin,Margaret Wrensch,Paul Brennan,David C Christiani,Mattias Johansson,James Mckay,Melinda C Aldrich,Christopher I Amos,Maria Teresa Landi,Adonina Tardon,International Lung Cancer Consortium (ILCCO),D Timothy Bishop,Florence Demenais,Alisa M Goldstein,Mark M Iles,Peter A Kanetsky,Matthew H Law,Ovarian Cancer Association Consortium (OCAC),Laufey T Amundadottir,Rachael Stolzenberg-Solomon,Brian M Wolpin,Pancreatic Cancer Cohort Consortium (Panscan),Alison Klein,Gloria Petersen,Harvey Risch,Pancreatic Cancer Case-Control Consortium (Panc4),The PRACTICAL Consortium,Stephen J Chanock,Mark P Purdue,Ghislaine Scelo,Paul Pharoah,Siddhartha Kar,Rayjean J Hung,Bogdan Pasaniuc,Peter Kraft

Journal

JNCI: Journal of the National Cancer Institute

Published Date

2023/6/1

Background The shared inherited genetic contribution to risk of different cancers is not fully known. In this study, we leverage results from 12 cancer genome-wide association studies (GWAS) to quantify pairwise genome-wide genetic correlations across cancers and identify novel cancer susceptibility loci. Methods We collected GWAS summary statistics for 12 solid cancers based on 376 759 participants with cancer and 532 864 participants without cancer of European ancestry. The included cancer types were breast, colorectal, endometrial, esophageal, glioma, head and neck, lung, melanoma, ovarian, pancreatic, prostate, and renal cancers. We conducted cross-cancer GWAS and transcriptome-wide association studies to discover novel cancer susceptibility loci. Finally, we assessed the extent of variant-specific pleiotropy among cancers at known and newly identified …

Validation of a genetic-enhanced risk prediction model for colorectal cancer in a large community-based cohort

Authors

Yu-Ru Su,Lori C Sakoda,Jihyoun Jeon,Minta Thomas,Yi Lin,Jennifer L Schneider,Natalia Udaltsova,Jeffrey K Lee,Iris Lansdorp-Vogelaar,Elisabeth FP Peterse,Ann G Zauber,Jiayin Zheng,Yingye Zheng,Elizabeth Hauser,John A Baron,Elizabeth L Barry,D Timothy Bishop,Hermann Brenner,Daniel D Buchanan,Andrea Burnett-Hartman,Peter T Campbell,Graham Casey,Sergi Castellví-Bel,Andrew T Chan,Jenny Chang-Claude,Jane C Figueiredo,Steven J Gallinger,Graham G Giles,Stephen B Gruber,Andrea Gsur,Marc J Gunter,Jochen Hampe,Heather Hampel,Tabitha A Harrison,Michael Hoffmeister,Xinwei Hua,Jeroen R Huyghe,Mark A Jenkins,Temitope O Keku,Loic Le Marchand,Li Li,Annika Lindblom,Victor Moreno,Polly A Newcomb,Paul DP Pharoah,Elizabeth A Platz,John D Potter,Conghui Qu,Gad Rennert,Robert E Schoen,Martha L Slattery,Mingyang Song,Fränzel JB van Duijnhoven,Bethany Van Guelpen,Pavel Vodicka,Alicja Wolk,Michael O Woods,Anna H Wu,Richard B Hayes,Ulrike Peters,Douglas A Corley,Li Hsu

Journal

Cancer Epidemiology, Biomarkers & Prevention

Published Date

2023/3/6

Background Polygenic risk scores (PRS) which summarize individuals’ genetic risk profile may enhance targeted colorectal cancer screening. A critical step towards clinical implementation is rigorous external validations in large community-based cohorts. This study externally validated a PRS-enhanced colorectal cancer risk model comprising 140 known colorectal cancer loci to provide a comprehensive assessment on prediction performance. Methods The model was developed using 20,338 individuals and externally validated in a community-based cohort (n = 85,221). We validated predicted 5-year absolute colorectal cancer risk, including calibration using expected-to-observed case ratios (E/O) and calibration plots, and discriminatory accuracy using time-dependent AUC. The PRS-related improvement in AUC, sensitivity and specificity were assessed in individuals of age …

Elucidating the risk of colorectal cancer for variants in hereditary colorectal cancer genes

Authors

Khalid Mahmood,Minta Thomas,Conghui Qu,Xiaoliang Wang,Jeroen R Huyghe,Jihoon E Joo,Peter Georgeson,Volker Arndt,Sonja I Berndt,Stéphane Bézieau,Stephanie A Bien,D Timothy Bishop,Hermann Brenner,Stefanie Brezina,Andrea Burnett-Hartman,Peter T Campbell,Graham Casey,Sergi Castellví-Bel,Andrew T Chan,Jenny Chang-Claude,Xuechen Chen,David V Conti,Chiara Cremolini,Brenda Diergaarde,Jane C Figueiredo,Liesel M FitzGerald,Manuela Gago-Dominguez,Steven Gallinger,Graham G Giles,Andrea Gsu,Marc J Gunter,Jochen Hampe,Heather Hampel,Tabitha A Harrison,Michael Hoffmeister,Temitope O Keku,Anshul Kundaje,Loic Le Marchand,Heinz-Josef Lenz,Christopher I Li,Li Li,Yi Lin,Annika Lindblom,Victor Moreno,Neil Murphy,Polly A Newcomb,Christina C Newton,Mireia Obón-Santacana,Shuji Ogino,Rish K Pai,Julie R Palmer,Rachel Pearlman,Paul DP Pharoah,Amanda I Phipps,Elizabeth A Platz,John D Potter,Gad Rennert,Lori C Sakoda,Clemens Schafmayer,Stephanie L Schmit,Robert E Schoen,Martha L Slattery,Zsofia K Stadler,Robert S Steinfelder,Stephen N Thibodeau,Cornelia M Ulrich,Caroline Y Um,Franzel JB van Duijnhoven,Bethany Van Guelpen,Kala Visvanathan,Pavel Vodicka,Ludmila Vodickova,Veronika Vymetalkova,Stephanie J Weinstein,Emily White,Ingrid M Winship,Alicja Wolk,Stephen B Gruber,Mark A Jenkins,Li Hsu,Daniel D Buchanan,Ulrike Peters

Journal

Gastroenterology

Published Date

2023/10/1

The Colon Cancer Family Registry (CCFR, www. coloncfr. org) is supported in part by funding from the National Cancer Institute (NCI), National Institutes of Health (NIH)(award U01 CA167551). Support for case ascertainment was provided in part from the Surveillance, Epidemiology, and End Results (SEER) Program and the following US state cancer registries: AZ, CO, MN, NC, NH; and by the Victoria Cancer Registry (Australia) and Ontario Cancer Registry (Canada). The CCFR Set-1 (Illumina 1M/1M-Duo) and Set-2 (Illumina Omni1-Quad) scans were supported by NIH awards U01 CA122839 and R01 CA143237 (to GC). The CCFR Set-3 (Affymetrix Axiom CORECT Set array) was supported by NIH award U19 CA148107 and R01 CA81488 (to SBG). The CCFR Set-4 (Illumina OncoArray 600K SNP array) was supported by NIH award U19 CA148107 (to SBG) and by the Center for Inherited Disease Research …

Genome-wide Interaction Study with Smoking for Colorectal Cancer Risk Identifies Novel Genetic Loci Related to Tumor Suppression, Inflammation, and Immune Response

Authors

Robert Carreras-Torres,Andre E Kim,Yi Lin,Virginia Diez-Obrero,Stephanie A Bien,Conghui Qu,Jun Wang,Niki Dimou,Elom K Aglago,Emmanouil Bouras,Peter T Campbell,Graham Casey,Jenny Chang-Claude,David A Drew,Marc Gunter,Kristina M Jordahl,Eric Kawaguchi,Anshul Kundaje,John L Morrison,Neil Murphy,Polly Newcomb,Mireia Obon-Santacana,Nikos Papadimitriou,Anita R Peoples,Edward Ruiz-Narvaez,Anna Shcherbina,Mariana C Stern,Yu-Ru Su,Yu Tian,Konstantinos K Tsilidis,Franzel JB van Duijnhoven,Li Hsu,Ulrike Peters,Victor Moreno,W James Gauderman

Journal

Cancer Epidemiology, Biomarkers & Prevention

Published Date

2024/1/1

Background Tobacco smoking is an established risk factor for colorectal cancer. However, genetically defined population subgroups may have increased susceptibility to smoking-related effects on colorectal cancer. Methods A genome-wide interaction scan was performed including 33,756 colorectal cancer cases and 44,346 controls from three genetic consortia. Results Evidence of an interaction was observed between smoking status (ever vs. never smokers) and a locus on 3p12.1 (rs9880919, P = 4.58 × 10−8), with higher associated risk in subjects carrying the GG genotype [OR, 1.25; 95% confidence interval (CI), 1.20–1.30] compared with the other genotypes (OR <1.17 for GA and AA). Among ever smokers, we observed interactions between smoking intensity (increase in 10 cigarettes smoked per day) and two loci on 6p21.33 (rs4151657, P = 1.72 × …

Association of germline variants in telomere maintenance genes (POT1, TERF2IP, ACD, and TERT) with spitzoid morphology in familial melanoma: A multi-center case series

Authors

Alisa M Goldstein,Richard Qin,Emily Y Chu,David E Elder,Daniela Massi,David J Adams,Paul W Harms,Carla Daniela Robles-Espinoza,Julia A Newton-Bishop,D Timothy Bishop,Mark Harland,Elizabeth A Holland,Anne E Cust,Helen Schmid,Graham J Mann,Susana Puig,Miriam Potrony,Llucia Alos,Eduardo Nagore,David Millán-Esteban,Nicholas K Hayward,Natasa Broit,Jane M Palmer,Vaishnavi Nathan,Elizabeth G Berry,Esteban Astiazaran-Symonds,Xiaohong R Yang,Margaret A Tucker,Maria Teresa Landi,Ruth M Pfeiffer,Michael R Sargen

Journal

JAAD international

Published Date

2023/6/1

BackgroundSpitzoid morphology in familial melanoma has been associated with germline variants in POT1, a telomere maintenance gene (TMG), suggesting a link between telomere biology and spitzoid differentiation.ObjectiveTo assess if familial melanoma cases associated with germline variants in TMG (POT1, ACD, TERF2IP, and TERT) commonly exhibit spitzoid morphology.MethodsIn this case series, melanomas were classified as having spitzoid morphology if at least 3 of 4 dermatopathologists reported this finding in ≥25% of tumor cells. Logistic regression was used to calculate odds ratios (OR) of spitzoid morphology compared to familial melanomas from unmatched noncarriers that were previously reviewed by a National Cancer Institute dermatopathologist.ResultsSpitzoid morphology was observed in 77% (23 of 30), 75% (3 of 4), 50% (2 of 4), and 50% (1 of 2) of melanomas from individuals with …

Probing the diabetes and colorectal cancer relationship using gene–environment interaction analyses

Authors

Niki Dimou,Andre E Kim,Orlagh Flanagan,Neil Murphy,Virginia Diez-Obrero,Anna Shcherbina,Elom K Aglago,Emmanouil Bouras,Peter T Campbell,Graham Casey,Steven Gallinger,Stephen B Gruber,Mark A Jenkins,Yi Lin,Victor Moreno,Edward Ruiz-Narvaez,Mariana C Stern,Yu Tian,Kostas K Tsilidis,Volker Arndt,Elizabeth L Barry,James W Baurley,Sonja I Berndt,Stéphane Bézieau,Stephanie A Bien,D Timothy Bishop,Hermann Brenner,Arif Budiarto,Robert Carreras-Torres,Tjeng Wawan Cenggoro,Andrew T Chan,Jenny Chang-Claude,Stephen J Chanock,Xuechen Chen,David V Conti,Christopher H Dampier,Matthew Devall,David A Drew,Jane C Figueiredo,Graham G Giles,Andrea Gsur,Tabitha A Harrison,Akihisa Hidaka,Michael Hoffmeister,Jeroen R Huyghe,Kristina Jordahl,Eric Kawaguchi,Temitope O Keku,Susanna C Larsson,Loic Le Marchand,Juan Pablo Lewinger,Li Li,Bharuno Mahesworo,John Morrison,Polly A Newcomb,Christina C Newton,Mireia Obon-Santacana,Jennifer Ose,Rish K Pai,Julie R Palmer,Nikos Papadimitriou,Bens Pardamean,Anita R Peoples,Paul DP Pharoah,Elizabeth A Platz,John D Potter,Gad Rennert,Peter C Scacheri,Robert E Schoen,Yu-Ru Su,Catherine M Tangen,Stephen N Thibodeau,Duncan C Thomas,Cornelia M Ulrich,Caroline Y Um,Franzel JB Van Duijnhoven,Kala Visvanathan,Pavel Vodicka,Ludmila Vodickova,Emily White,Alicja Wolk,Michael O Woods,Conghui Qu,Anshul Kundaje,Li Hsu,W James Gauderman,Marc J Gunter,Ulrike Peters

Journal

British journal of cancer

Published Date

2023/8/24

BackgroundDiabetes is an established risk factor for colorectal cancer. However, the mechanisms underlying this relationship still require investigation and it is not known if the association is modified by genetic variants. To address these questions, we undertook a genome-wide gene-environment interaction analysis.MethodsWe used data from 3 genetic consortia (CCFR, CORECT, GECCO; 31,318 colorectal cancer cases/41,499 controls) and undertook genome-wide gene-environment interaction analyses with colorectal cancer risk, including interaction tests of genetics(G)xdiabetes (1-degree of freedom; d.f.) and joint testing of Gxdiabetes, G-colorectal cancer association (2-d.f. joint test) and G-diabetes correlation (3-d.f. joint test).ResultsBased on the joint tests, we found that the association of diabetes with colorectal cancer risk is modified by loci on chromosomes 8q24.11 (rs3802177, SLC30A8 – ORAA: 1.62 …

CAPP2 RCT: Resistant Starch Planned Long-term Follow-up—Reply

Authors

John C Mathers,Faye Elliott,D Timothy Bishop,John Burn

Journal

Cancer Prevention Research

Published Date

2023/1/4

We thank Drs. Nielsen and van Duijnhoven for their interest in our paper (1) and for their recognition of the importance of ourfindings as a basis for dietary advice for patients with Lynch syndrome (LS). Participants in the CAPP2 Study, randomized to receive supplemental resistant starch (RS) during the intervention trial (mean 29 months), had significantly lower risk of extracolonic cancers, notably lower risk of cancers of the upper gastrointestinal (GI) tract, when followed up 10 to 20 years later (1). This finding is especially significant given that such cancers are more difficult to manage, and that 10-year survival is much lower for upper GI cancer (58%), than for colorectal cancer (91%; ref. 2). Drs. Nielsen and van Duijnhoven ask two questions:(i) Does RS have similar beneficial effects in lowering the risk of extracolonic cancers in patients with LS with different germline mutations? and ii) How can these findings be …

PTPN11 mosaicism causes a spectrum of pigmentary and vascular neurocutaneous disorders and predisposes to melanoma

Authors

Satyamaanasa Polubothu,Nicole Bender,Siobhan Muthiah,Davide Zecchin,Charalambos Demetriou,Sara Barberan Martin,Sony Malhotra,Jana Travnickova,Zhiqiang Zeng,Markus Böhm,Sebastien Barbarot,Catherine Cottrell,Olivia Davies,Eulalia Baselga,Nigel P Burrows,Virginie Carmignac,Joey Santiago Diaz,Christine Fink,Holger A Haenssle,Rudolf Happle,Mark Harland,Jacquelyn Majerowski,Pierre Vabres,Marie Vincent,Julia A Newton-Bishop,D Tim Bishop,Dawn Siegel,E Elizabeth Patton,Maya Topf,Neil Rajan,Beth Drolet,Veronica A Kinsler

Journal

Journal of Investigative Dermatology

Published Date

2023/6/1

Phakomatosis pigmentovascularis is a diagnosis that denotes the coexistence of pigmentary and vascular birthmarks of specific types, accompanied by variable multisystem involvement, including CNS disease, asymmetrical growth, and a predisposition to malignancy. Using a tight phenotypic group and high-depth next-generation sequencing of affected tissues, we discover here clonal mosaic variants in gene PTPN11 encoding SHP2 phosphatase as a cause of phakomatosis pigmentovascularis type III or spilorosea. Within an individual, the same variant is found in distinct pigmentary and vascular birthmarks and is undetectable in blood. We go on to show that the same variants can cause either the pigmentary or vascular phenotypes alone, and drive melanoma development within pigmentary lesions. Protein structure modeling highlights that although variants lead to loss of function at the level of the …

See List of Professors in D. Timothy Bishop University(University of Leeds)

D. Timothy Bishop FAQs

What is D. Timothy Bishop's h-index at University of Leeds?

The h-index of D. Timothy Bishop has been 54 since 2020 and 112 in total.

What are D. Timothy Bishop's top articles?

The articles with the titles of

The tumor suppressive role of methylthioadenosine phosphorylase in melanoma

Genome-Wide Gene–Environment Interaction Analyses to Understand the Relationship between Red Meat and Processed Meat Intake and Colorectal Cancer Risk

Fine-mapping analysis including over 254,000 East Asian and European descendants identifies 136 putative colorectal cancer susceptibility genes

Genome-wide association study and Mendelian randomization analyses provide insights into the causes of early-onset colorectal cancer

Systemic inflammation, the peripheral blood transcriptome and primary melanoma

Observational and genetic associations between cardiorespiratory fitness and cancer: a UK Biobank and international consortia study

Genetic risk impacts the association of menopausal hormone therapy with colorectal cancer risk

Immune subtyping of melanoma whole slide images using multiple instance learning

...

are the top articles of D. Timothy Bishop at University of Leeds.

What are D. Timothy Bishop's research interests?

The research interests of D. Timothy Bishop are: genetic epidemiology, melanoma

What is D. Timothy Bishop's total number of citations?

D. Timothy Bishop has 56,054 citations in total.

    academic-engine

    Useful Links