Centre for Cancer Research - Research Publications

Permanent URI for this collection

Search Results

Now showing 1 - 10 of 48
  • Item
    Thumbnail Image
    Whole Exome Sequencing in Patients with White Matter Abnormalities
    Vanderver, A ; Simons, C ; Helman, G ; Crawford, J ; Wolf, NI ; Bernard, G ; Pizzino, A ; Schmidt, JL ; Takanohashi, A ; Miller, D ; Khouzam, A ; Rajan, V ; Ramos, E ; Chowdhury, S ; Hambuch, T ; Ru, K ; Baillie, GJ ; Grimmond, SM ; Caldovic, L ; Devaney, J ; Bloom, M ; Evans, SH ; Murphy, JLP ; McNeill, N ; Fogel, BL ; Schiffmann, R ; van der Knaap, MS ; Taft, RJ (WILEY-BLACKWELL, 2016-06)
    Here we report whole exome sequencing (WES) on a cohort of 71 patients with persistently unresolved white matter abnormalities with a suspected diagnosis of leukodystrophy or genetic leukoencephalopathy. WES analyses were performed on trio, or greater, family groups. Diagnostic pathogenic variants were identified in 35% (25 of 71) of patients. Potentially pathogenic variants were identified in clinically relevant genes in a further 7% (5 of 71) of cases, giving a total yield of clinical diagnoses in 42% of individuals. These findings provide evidence that WES can substantially decrease the number of unresolved white matter cases. Ann Neurol 2016;79:1031-1037.
  • Item
    Thumbnail Image
    Recurrent loss of heterozygosity correlates with clinical outcome in pancreatic neuroendocrine cancer
    Lawrence, B ; Blenkiron, C ; Parker, K ; Tsai, P ; Fitzgerald, S ; Shields, P ; Robb, T ; Yeong, ML ; Kramer, N ; James, S ; Black, M ; Fan, V ; Poonawala, N ; Yap, P ; Coats, E ; Woodhouse, B ; Ramsaroop, R ; Yozu, M ; Robinson, B ; Henare, K ; Koea, J ; Johnston, P ; Carroll, R ; Connor, S ; Morrin, H ; Elston, M ; Jackson, C ; Reid, P ; Windsor, J ; MacCormick, A ; Babor, R ; Bartlett, A ; Damianovich, D ; Knowlton, N ; Grimmond, S ; Findlay, M ; Print, C (SPRINGERNATURE, 2018-07-20)
    Pancreatic neuroendocrine tumors (pNETs) are uncommon cancers arising from pancreatic islet cells. Here we report the analysis of gene mutation, copy number, and RNA expression of 57 sporadic well-differentiated pNETs. pNET genomes are dominated by aneuploidy, leading to concordant changes in RNA expression at the level of whole chromosomes and chromosome segments. We observed two distinct patterns of somatic pNET aneuploidy that are associated with tumor pathology and patient prognosis. Approximately 26% of the patients in this series had pNETs with genomes characterized by recurrent loss of heterozygosity (LoH) of 10 specific chromosomes, accompanied by bi-allelic MEN1 inactivation and generally poor clinical outcome. Another ~40% of patients had pNETs that lacked this recurrent LoH pattern but had chromosome 11 LoH, bi-allelic MEN1 inactivation, and universally good clinical outcome. The somatic aneuploidy allowed pathogenic germline variants (e.g., ATM) to be expressed unopposed, with RNA expression patterns showing inactivation of downstream tumor suppressor pathways. No prognostic associations were found with tumor morphology, single gene mutation, or expression of RNAs reflecting the activity of immune, differentiation, proliferative or tumor suppressor pathways. In pNETs, single gene mutations appear to be less important than aneuploidy, with MEN1 the only statistically significant recurrently mutated driver gene. In addition, only one pNET in the series had clearly actionable single nucleotide variants (SNVs) (in PTEN and FLCN) confirmed by corroborating RNA expression changes. The two clinically relevant patterns of LoH described here define a novel oncogenic mechanism and a plausible route to genomic precision oncology for this tumor type.
  • Item
    Thumbnail Image
    Tailored first-line and second-line CDK4-targeting treatment combinations in mouse models of pancreatic cancer
    Chou, A ; Froio, D ; Nagrial, AM ; Parkin, A ; Murphy, KJ ; Chin, VT ; Wohl, D ; Steinmann, A ; Stark, R ; Drury, A ; Walters, SN ; Vennin, C ; Burgess, A ; Pinese, M ; Chantrill, LA ; Cowley, MJ ; Molloy, TJ ; Waddell, N ; Johns, A ; Grimmond, SM ; Chang, DK ; Biankin, AV ; Sansom, OJ ; Morton, JP ; Grey, ST ; Cox, TR ; Turchini, J ; Samra, J ; Clarke, SJ ; Timpson, P ; Gill, AJ ; Pajic, M (BMJ PUBLISHING GROUP, 2018-12)
    OBJECTIVE: Extensive molecular heterogeneity of pancreatic ductal adenocarcinoma (PDA), few effective therapies and high mortality make this disease a prime model for advancing development of tailored therapies. The p16-cyclin D-cyclin-dependent kinase 4/6-retinoblastoma (RB) protein (CDK4) pathway, regulator of cell proliferation, is deregulated in PDA. Our aim was to develop a novel personalised treatment strategy for PDA based on targeting CDK4. DESIGN: Sensitivity to potent CDK4/6 inhibitor PD-0332991 (palbociclib) was correlated to protein and genomic data in 19 primary patient-derived PDA lines to identify biomarkers of response. In vivo efficacy of PD-0332991 and combination therapies was determined in subcutaneous, intrasplenic and orthotopic tumour models derived from genome-sequenced patient specimens and genetically engineered model. Mechanistically, monotherapy and combination therapy were investigated in the context of tumour cell and extracellular matrix (ECM) signalling. Prognostic relevance of companion biomarker, RB protein, was evaluated and validated in independent PDA patient cohorts (>500 specimens). RESULTS: Subtype-specific in vivo efficacy of PD-0332991-based therapy was for the first time observed at multiple stages of PDA progression: primary tumour growth, recurrence (second-line therapy) and metastatic setting and may potentially be guided by a simple biomarker (RB protein). PD-0332991 significantly disrupted surrounding ECM organisation, leading to increased quiescence, apoptosis, improved chemosensitivity, decreased invasion, metastatic spread and PDA progression in vivo. RB protein is prevalent in primary operable and metastatic PDA and may present a promising predictive biomarker to guide this therapeutic approach. CONCLUSION: This study demonstrates the promise of CDK4 inhibition in PDA over standard therapy when applied in a molecular subtype-specific context.
  • Item
    No Preview Available
    Recurrent noncoding regulatory mutations in pancreatic ductal adenocarcinoma
    Feigin, ME ; Garvin, T ; Bailey, P ; Waddell, N ; Chang, DK ; Kelley, DR ; Shuai, S ; Gallinger, S ; McPherson, JD ; Grimmond, SM ; Khurana, E ; Stein, LD ; Biankin, AV ; Schatz, MC ; Tuveson, DA (NATURE PUBLISHING GROUP, 2017-06)
    The contributions of coding mutations to tumorigenesis are relatively well known; however, little is known about somatic alterations in noncoding DNA. Here we describe GECCO (Genomic Enrichment Computational Clustering Operation) to analyze somatic noncoding alterations in 308 pancreatic ductal adenocarcinomas (PDAs) and identify commonly mutated regulatory regions. We find recurrent noncoding mutations to be enriched in PDA pathways, including axon guidance and cell adhesion, and newly identified processes, including transcription and homeobox genes. We identified mutations in protein binding sites correlating with differential expression of proximal genes and experimentally validated effects of mutations on expression. We developed an expression modulation score that quantifies the strength of gene regulation imposed by each class of regulatory elements, and found the strongest elements were most frequently mutated, suggesting a selective advantage. Our detailed single-cancer analysis of noncoding alterations identifies regulatory mutations as candidates for diagnostic and prognostic markers, and suggests new mechanisms for tumor evolution.
  • Item
    No Preview Available
    Mutational signatures in esophageal adenocarcinoma define etiologically distinct subgroups with therapeutic relevance
    Secrier, M ; Li, X ; de Silva, N ; Eldridge, MD ; Contino, G ; Bornschein, J ; MacRae, S ; Grehan, N ; O'Donovan, M ; Miremadi, A ; Yang, T-P ; Bower, L ; Chettouh, H ; Crawte, J ; Galeano-Dalmau, N ; Grabowska, A ; Saunders, J ; Underwood, T ; Waddell, N ; Barbour, AP ; Nutzinger, B ; Achilleos, A ; Edwards, PAW ; Lynch, AG ; Tavare, S ; Fitzgerald, RC (NATURE PUBLISHING GROUP, 2016-10)
    Esophageal adenocarcinoma (EAC) has a poor outcome, and targeted therapy trials have thus far been disappointing owing to a lack of robust stratification methods. Whole-genome sequencing (WGS) analysis of 129 cases demonstrated that this is a heterogeneous cancer dominated by copy number alterations with frequent large-scale rearrangements. Co-amplification of receptor tyrosine kinases (RTKs) and/or downstream mitogenic activation is almost ubiquitous; thus tailored combination RTK inhibitor (RTKi) therapy might be required, as we demonstrate in vitro. However, mutational signatures showed three distinct molecular subtypes with potential therapeutic relevance, which we verified in an independent cohort (n = 87): (i) enrichment for BRCA signature with prevalent defects in the homologous recombination pathway; (ii) dominant T>G mutational pattern associated with a high mutational load and neoantigen burden; and (iii) C>A/T mutational pattern with evidence of an aging imprint. These subtypes could be ascertained using a clinically applicable sequencing strategy (low coverage) as a basis for therapy selection.
  • Item
    No Preview Available
    Dual targeting of p53 and c-MYC selectively eliminates leukaemic stem cells
    Abraham, SA ; Hopcroft, LEM ; Carrick, E ; Drotar, ME ; Dunn, K ; Williamson, AJK ; Korfi, K ; Baquero, P ; Park, LE ; Scott, MT ; Pellicano, F ; Pierce, A ; Copland, M ; Nourse, C ; Grimmond, SM ; Vetrie, D ; Whetton, AD ; Holyoake, TL (NATURE PORTFOLIO, 2016-06-16)
    Chronic myeloid leukaemia (CML) arises after transformation of a haemopoietic stem cell (HSC) by the protein-tyrosine kinase BCR-ABL. Direct inhibition of BCR-ABL kinase has revolutionized disease management, but fails to eradicate leukaemic stem cells (LSCs), which maintain CML. LSCs are independent of BCR-ABL for survival, providing a rationale for identifying and targeting kinase-independent pathways. Here we show--using proteomics, transcriptomics and network analyses--that in human LSCs, aberrantly expressed proteins, in both imatinib-responder and non-responder patients, are modulated in concert with p53 (also known as TP53) and c-MYC regulation. Perturbation of both p53 and c-MYC, and not BCR-ABL itself, leads to synergistic cell kill, differentiation, and near elimination of transplantable human LSCs in mice, while sparing normal HSCs. This unbiased systems approach targeting connected nodes exemplifies a novel precision medicine strategy providing evidence that LSCs can be eradicated.
  • Item
    No Preview Available
    Novel cancer drivers: mining the kinome
    Biankin, AV ; Grimmond, SM (BMC, 2013-02-28)
    Large-scale cancer genome studies are unveiling significant complexity and heterogeneity even in histopathologically indistinguishable cancers. Differentiating 'driver' mutations that are functionally relevant from 'passenger' mutations is a major challenge in cancer genomics. While recurrent mutations in a gene provides supporting evidence of 'driver' status, novel computational methods and model systems are greatly improving our ability to identify genes important in carcinogenesis. Reimand and Bader have recently shown that driver gene discovery in discrete gene classes (in this case the kinome) is possible across multiple cancer types and has the potential to yield new druggable targets and clinically relevant leads.
  • Item
    No Preview Available
    Signatures of mutational processes in human cancer
    Alexandrov, LB ; Nik-Zainal, S ; Wedge, DC ; Aparicio, SAJR ; Behjati, S ; Biankin, AV ; Bignell, GR ; Bolli, N ; Borg, A ; Borresen-Dale, A-L ; Boyault, S ; Burkhardt, B ; Butler, AP ; Caldas, C ; Davies, HR ; Desmedt, C ; Eils, R ; Eyfjord, JE ; Foekens, JA ; Greaves, M ; Hosoda, F ; Hutter, B ; Ilicic, T ; Imbeaud, S ; Imielinsk, M ; Jaeger, N ; Jones, DTW ; Jones, D ; Knappskog, S ; Kool, M ; Lakhani, SR ; Lopez-Otin, C ; Martin, S ; Munshi, NC ; Nakamura, H ; Northcott, PA ; Pajic, M ; Papaemmanuil, E ; Paradiso, A ; Pearson, JV ; Puente, XS ; Raine, K ; Ramakrishna, M ; Richardson, AL ; Richter, J ; Rosenstiel, P ; Schlesner, M ; Schumacher, TN ; Span, PN ; Teague, JW ; Totoki, Y ; Tutt, ANJ ; Valdes-Mas, R ; van Buuren, MM ; van 't Veer, L ; Vincent-Salomon, A ; Waddell, N ; Yates, LR ; Zucman-Rossi, J ; Futreal, PA ; McDermott, U ; Lichter, P ; Meyerson, M ; Grimmond, SM ; Siebert, R ; Campo, E ; Shibata, T ; Pfister, SM ; Campbell, PJ ; Stratton, MR (NATURE PORTFOLIO, 2013-08-22)
    All cancers are caused by somatic mutations; however, understanding of the biological processes generating these mutations is limited. The catalogue of somatic mutations from a cancer genome bears the signatures of the mutational processes that have been operative. Here we analysed 4,938,362 mutations from 7,042 cancers and extracted more than 20 distinct mutational signatures. Some are present in many cancer types, notably a signature attributed to the APOBEC family of cytidine deaminases, whereas others are confined to a single cancer class. Certain signatures are associated with age of the patient at cancer diagnosis, known mutagenic exposures or defects in DNA maintenance, but many are of cryptic origin. In addition to these genome-wide mutational signatures, hypermutation localized to small genomic regions, 'kataegis', is found in many cancer types. The results reveal the diversity of mutational processes underlying the development of cancer, with potential implications for understanding of cancer aetiology, prevention and therapy.
  • Item
    No Preview Available
    The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma
    Perez-Mancera, PA ; Rust, AG ; van der Weyden, L ; Kristiansen, G ; Li, A ; Sarver, AL ; Silverstein, KAT ; Gruetzmann, R ; Aust, D ; Ruemmele, P ; Knoesel, T ; Herd, C ; Stemple, DL ; Kettleborough, R ; Brosnan, JA ; Li, A ; Morgan, R ; Knight, S ; Yu, J ; Stegeman, S ; Collier, LS ; ten Hoeve, JJ ; de Ridder, J ; Klein, AP ; Goggins, M ; Hruban, RH ; Chang, DK ; Biankin, AV ; Grimmond, SM ; Wessels, LFA ; Wood, SA ; Iacobuzio-Donahue, CA ; Pilarsky, C ; Largaespada, DA ; Adams, DJ ; Tuveson, DA (NATURE PUBLISHING GROUP, 2012-06-14)
    Pancreatic ductal adenocarcinoma (PDA) remains a lethal malignancy despite much progress concerning its molecular characterization. PDA tumours harbour four signature somatic mutations in addition to numerous lower frequency genetic events of uncertain significance. Here we use Sleeping Beauty (SB) transposon-mediated insertional mutagenesis in a mouse model of pancreatic ductal preneoplasia to identify genes that cooperate with oncogenic Kras(G12D) to accelerate tumorigenesis and promote progression. Our screen revealed new candidate genes for PDA and confirmed the importance of many genes and pathways previously implicated in human PDA. The most commonly mutated gene was the X-linked deubiquitinase Usp9x, which was inactivated in over 50% of the tumours. Although previous work had attributed a pro-survival role to USP9X in human neoplasia, we found instead that loss of Usp9x enhances transformation and protects pancreatic cancer cells from anoikis. Clinically, low USP9X protein and messenger RNA expression in PDA correlates with poor survival after surgery, and USP9X levels are inversely associated with metastatic burden in advanced disease. Furthermore, chromatin modulation with trichostatin A or 5-aza-2'-deoxycytidine elevates USP9X expression in human PDA cell lines, indicating a clinical approach for certain patients. The conditional deletion of Usp9x cooperated with Kras(G12D) to accelerate pancreatic tumorigenesis in mice, validating their genetic interaction. We propose that USP9X is a major tumour suppressor gene with prognostic and therapeutic relevance in PDA.
  • Item
    No Preview Available
    International network of cancer genome projects
    Hudson, TJ ; Anderson, W ; Aretz, A ; Barker, AD ; Bell, C ; Bernabe, RR ; Bhan, MK ; Calvo, F ; Eerola, I ; Gerhard, DS ; Guttmacher, A ; Guyer, M ; Hemsley, FM ; Jennings, JL ; Kerr, D ; Klatt, P ; Kolar, P ; Kusuda, J ; Lane, DP ; Laplace, F ; Lu, Y ; Nettekoven, G ; Ozenberger, B ; Peterson, J ; Rao, TS ; Remacle, J ; Schafer, AJ ; Shibata, T ; Stratton, MR ; Vockley, JG ; Watanabe, K ; Yang, H ; Yuen, MMF ; Knoppers, M ; Bobrow, M ; Cambon-Thomsen, A ; Dressler, LG ; Dyke, SOM ; Joly, Y ; Kato, K ; Kennedy, KL ; Nicolas, P ; Parker, MJ ; Rial-Sebbag, E ; Romeo-Casabona, CM ; Shaw, KM ; Wallace, S ; Wiesner, GL ; Zeps, N ; Lichter, P ; Biankin, AV ; Chabannon, C ; Chin, L ; Clement, B ; de Alava, E ; Degos, F ; Ferguson, ML ; Geary, P ; Hayes, DN ; Johns, AL ; Nakagawa, H ; Penny, R ; Piris, MA ; Sarin, R ; Scarpa, A ; van de Vijver, M ; Futreal, PA ; Aburatani, H ; Bayes, M ; Bowtell, DDL ; Campbell, PJ ; Estivill, X ; Grimmond, SM ; Gut, I ; Hirst, M ; Lopez-Otin, C ; Majumder, P ; Marra, M ; Ning, Z ; Puente, XS ; Ruan, Y ; Stunnenberg, HG ; Swerdlow, H ; Velculescu, VE ; Wilson, RK ; Xue, HH ; Yang, L ; Spellman, PT ; Bader, GD ; Boutros, PC ; Flicek, P ; Getz, G ; Guigo, R ; Guo, G ; Haussler, D ; Heath, S ; Hubbard, TJ ; Jiang, T ; Jones, SM ; Li, Q ; Lopez-Bigas, N ; Luo, R ; Pearson, JV ; Quesada, V ; Raphael, BJ ; Sander, C ; Speed, TP ; Stuart, JM ; Teague, JW ; Totoki, Y ; Tsunoda, T ; Valencia, A ; Wheeler, DA ; Wu, H ; Zhao, S ; Zhou, G ; Stein, LD ; Lathrop, M ; Ouellette, BFF ; Thomas, G ; Yoshida, T ; Axton, M ; Gunter, C ; McPherson, JD ; Miller, LJ ; Kasprzyk, A ; Zhang, J ; Haider, SA ; Wang, J ; Yung, CK ; Cross, A ; Liang, Y ; Gnaneshan, S ; Guberman, J ; Hsu, J ; Chalmers, DRC ; Hasel, KW ; Kaan, TSH ; Knoppers, BM ; Lowrance, WW ; Masui, T ; Rodriguez, LL ; Vergely, C ; Cloonan, N ; Defazio, A ; Eshleman, JR ; Etemadmoghadam, D ; Gardiner, BA ; Kench, JG ; Sutherland, RL ; Tempero, MA ; Waddell, NJ ; Wilson, PJ ; Gallinger, S ; Tsao, M-S ; Shaw, PA ; Petersen, GM ; Mukhopadhyay, D ; DePinho, RA ; Thayer, S ; Muthuswamy, L ; Shazand, K ; Beck, T ; Sam, M ; Timms, L ; Ballin, V ; Ji, J ; Zhang, X ; Chen, F ; Hu, X ; Yang, Q ; Tian, G ; Zhang, L ; Xing, X ; Li, X ; Zhu, Z ; Yu, Y ; Yu, J ; Tost, J ; Brennan, P ; Holcatova, I ; Zaridze, D ; Brazma, A ; Egevad, L ; Prokhortchouk, E ; Banks, RE ; Uhlen, M ; Viksna, J ; Ponten, F ; Skryabin, K ; Birney, E ; Borg, A ; Borresen-Dale, A-L ; Caldas, C ; Foekens, JA ; Martin, S ; Reis-Filho, JS ; Richardson, AL ; Sotiriou, C ; van't Veer, L ; Birnbaum, D ; Blanche, H ; Boucher, P ; Boyault, S ; Masson-Jacquemier, JD ; Pauporte, I ; Pivot, X ; Vincent-Salomon, A ; Tabone, E ; Theillet, C ; Treilleux, I ; Bioulac-Sage, P ; Decaens, T ; Franco, D ; Gut, M ; Samuel, D ; Zucman-Rossi, J ; Eils, R ; Brors, B ; Korbel, JO ; Korshunov, A ; Landgraf, P ; Lehrach, H ; Pfister, S ; Radlwimmer, B ; Reifenberger, G ; Taylor, MD ; von Kalle, C ; Majumder, PP ; Pederzoli, P ; Lawlor, RT ; Delledonne, M ; Bardelli, A ; Gress, T ; Klimstra, D ; Zamboni, G ; Nakamura, Y ; Miyano, S ; Fujimoto, A ; Campo, E ; de Sanjose, S ; Montserrat, E ; Gonzalez-Diaz, M ; Jares, P ; Himmelbaue, H ; Bea, S ; Aparicio, S ; Easton, DF ; Collins, FS ; Compton, CC ; Lander, ES ; Burke, W ; Green, AR ; Hamilton, SR ; Kallioniemi, OP ; Ley, TJ ; Liu, ET ; Wainwright, BJ (NATURE PORTFOLIO, 2010-04-15)
    The International Cancer Genome Consortium (ICGC) was launched to coordinate large-scale cancer genome studies in tumours from 50 different cancer types and/or subtypes that are of clinical and societal importance across the globe. Systematic studies of more than 25,000 cancer genomes at the genomic, epigenomic and transcriptomic levels will reveal the repertoire of oncogenic mutations, uncover traces of the mutagenic influences, define clinically relevant subtypes for prognosis and therapeutic management, and enable the development of new cancer therapies.