250 results on '"Grimmond, SM"'
Search Results
2. qmotif: determination of telomere content from whole-genome sequence data
- Author
-
Stamatakis, A, Holmes, O, Nones, K, Tang, YH, Loffler, KA, Lee, M, Patch, A-M, Dagg, RA, Lau, LMS, Leonard, C, Wood, S, Xu, Q, Pickett, HA, Reddel, RR, Barbour, AP, Grimmond, SM, Waddell, N, Pearson, J, Stamatakis, A, Holmes, O, Nones, K, Tang, YH, Loffler, KA, Lee, M, Patch, A-M, Dagg, RA, Lau, LMS, Leonard, C, Wood, S, Xu, Q, Pickett, HA, Reddel, RR, Barbour, AP, Grimmond, SM, Waddell, N, and Pearson, J
- Abstract
MOTIVATION: Changes in telomere length have been observed in cancer and can be indicative of mechanisms involved in carcinogenesis. Most methods used to estimate telomere length require laboratory analysis of DNA samples. Here, we present qmotif, a fast and easy tool that determines telomeric repeat sequences content as an estimate of telomere length directly from whole-genome sequencing. RESULTS: qmotif shows similar results to quantitative PCR, the standard method for high-throughput clinical telomere length quantification. qmotif output correlates strongly with the output of other tools for determining telomere sequence content, TelSeq and TelomereHunter, but can run in a fraction of the time-usually under a minute. AVAILABILITY AND IMPLEMENTATION: qmotif is implemented in Java and source code is available at https://github.com/AdamaJava/adamajava, with instructions on how to build and use the application available from https://adamajava.readthedocs.io/en/latest/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics Advances online.
- Published
- 2022
3. Comprehensive genomic and tumour immune profiling reveals potential therapeutic targets in malignant pleural mesothelioma
- Author
-
Creaney, J, Patch, A-M, Addala, V, Sneddon, SA, Nones, K, Dick, IM, Lee, YCG, Newell, F, Rouse, EJ, Naeini, MM, Kondrashova, O, Lakis, V, Nakas, A, Waller, D, Sharkey, A, Mukhopadhyay, P, Kazakoff, SH, Koufariotis, LT, Davidson, AL, Ramarao-Milne, P, Holmes, O, Xu, Q, Leonard, C, Wood, S, Grimmond, SM, Bueno, R, Fennell, DA, Pearson, J, Robinson, BW, Waddell, N, Creaney, J, Patch, A-M, Addala, V, Sneddon, SA, Nones, K, Dick, IM, Lee, YCG, Newell, F, Rouse, EJ, Naeini, MM, Kondrashova, O, Lakis, V, Nakas, A, Waller, D, Sharkey, A, Mukhopadhyay, P, Kazakoff, SH, Koufariotis, LT, Davidson, AL, Ramarao-Milne, P, Holmes, O, Xu, Q, Leonard, C, Wood, S, Grimmond, SM, Bueno, R, Fennell, DA, Pearson, J, Robinson, BW, and Waddell, N
- Abstract
BACKGROUND: Malignant pleural mesothelioma (MPM) has a poor overall survival with few treatment options. Whole genome sequencing (WGS) combined with the immune features of MPM offers the prospect of identifying changes that could inform future clinical trials. METHODS: We analysed somatic mutations from 229 MPM samples, including previously published data and 58 samples that had undergone WGS within this study. This was combined with RNA-seq analysis to characterize the tumour immune environment. RESULTS: The comprehensive genome analysis identified 12 driver genes, including new candidate genes. Whole genome doubling was a frequent event that correlated with shorter survival. Mutational signature analysis revealed SBS5/40 were dominant in 93% of samples, and defects in homologous recombination repair were infrequent in our cohort. The tumour immune environment contained high M2 macrophage infiltrate linked with MMP2, MMP14, TGFB1 and CCL2 expression, representing an immune suppressive environment. The expression of TGFB1 was associated with overall survival. A small subset of samples (less than 10%) had a higher proportion of CD8 T cells and a high cytolytic score, suggesting a 'hot' immune environment independent of the somatic mutations. CONCLUSIONS: We propose accounting for genomic and immune microenvironment status may influence therapeutic planning in the future.
- Published
- 2022
4. Methyl-CpG binding domain 4, DNA glycosylase (MBD4)-associated neoplasia syndrome associated with a homozygous missense variant in MBD4: Expansion of an emerging phenotype
- Author
-
Blombery, P, Ryland, GL, Fox, LC, Stark, Z, Wall, M, Jarmolowicz, A, Roesley, A, Thompson, ER, Grimmond, SM, Panicker, S, Kwok, F, Blombery, P, Ryland, GL, Fox, LC, Stark, Z, Wall, M, Jarmolowicz, A, Roesley, A, Thompson, ER, Grimmond, SM, Panicker, S, and Kwok, F
- Published
- 2022
5. Enhancer retargeting of CDX2 and UBTF::ATXN7L3 define a subtype of high-risk B-progenitor acute lymphoblastic leukemia
- Author
-
Kimura, S, Montefiori, L, Iacobucci, I, Zhao, Y, Gao, Q, Paietta, EM, Haferlach, C, Laird, AD, Mead, PE, Gu, Z, Stock, W, Litzow, M, Rowe, JM, Luger, SM, Hunger, SP, Ryland, GL, Schmidt, B, Ekert, PG, Oshlack, A, Grimmond, SM, Rehn, J, Breen, J, Yeung, D, White, DL, Aldoss, I, Jabbour, EJ, Pui, C-H, Meggendorfer, M, Walter, W, Kern, W, Haferlach, T, Brady, S, Zhang, J, Roberts, KG, Blombery, P, Mullighan, CG, Kimura, S, Montefiori, L, Iacobucci, I, Zhao, Y, Gao, Q, Paietta, EM, Haferlach, C, Laird, AD, Mead, PE, Gu, Z, Stock, W, Litzow, M, Rowe, JM, Luger, SM, Hunger, SP, Ryland, GL, Schmidt, B, Ekert, PG, Oshlack, A, Grimmond, SM, Rehn, J, Breen, J, Yeung, D, White, DL, Aldoss, I, Jabbour, EJ, Pui, C-H, Meggendorfer, M, Walter, W, Kern, W, Haferlach, T, Brady, S, Zhang, J, Roberts, KG, Blombery, P, and Mullighan, CG
- Abstract
Transcriptome sequencing has identified multiple subtypes of B-progenitor acute lymphoblastic leukemia (B-ALL) of prognostic significance, but a minority of cases lack a known genetic driver. Here, we used integrated whole-genome (WGS) and -transcriptome sequencing (RNA-seq), enhancer mapping, and chromatin topology analysis to identify previously unrecognized genomic drivers in B-ALL. Newly diagnosed (n = 3221) and relapsed (n = 177) B-ALL cases with tumor RNA-seq were studied. WGS was performed to detect mutations, structural variants, and copy number alterations. Integrated analysis of histone 3 lysine 27 acetylation and chromatin looping was performed using HiChIP. We identified a subset of 17 newly diagnosed and 5 relapsed B-ALL cases with a distinct gene expression profile and 2 universal and unique genomic alterations resulting from aberrant recombination-activating gene activation: a focal deletion downstream of PAN3 at 13q12.2 resulting in CDX2 deregulation by the PAN3 enhancer and a focal deletion of exons 18-21 of UBTF at 17q21.31 resulting in a chimeric fusion, UBTF::ATXN7L3. A subset of cases also had rearrangement and increased expression of the PAX5 gene, which is otherwise uncommon in B-ALL. Patients were more commonly female and young adult with median age 35 (range,12-70 years). The immunophenotype was characterized by CD10 negativity and immunoglobulin M positivity. Among 16 patients with known clinical response, 9 (56.3%) had high-risk features including relapse (n = 4) or minimal residual disease >1% at the end of remission induction (n = 5). CDX2-deregulated, UBTF::ATXN7L3 rearranged (CDX2/UBTF) B-ALL is a high-risk subtype of leukemia in young adults for which novel therapeutic approaches are required.
- Published
- 2022
6. CAF hierarchy driven by pancreatic cancer cell p53-status creates a pro-metastatic and chemoresistant environment via perlecan
- Author
-
Vennin, C, Melenec, P, Rouet, R, Nobis, M, Cazet, As, Murphy, Kj, Herrmann, D, Reed, Da, Lucas, Mc, Warren, Sc, Elgundi, Z, Pinese, M, Kalna, G, Roden, D, Samuel, M, Zaratzian, A, Grey, St, Da Silva, A, Leung, W, Mathivanan, S, Wang, Yx, Braithwaite, Aw, Christ, D, Benda, A, Parkin, A, Phillips, Pa, Whitelock, Jm, Gill, Aj, Sansom, Oj, Croucher, Dr, Parker, Bl, Pajic, M, Morton, Jp, Cox, Tr, Timpson, P, Johns, Al, Chantrill, La, Chou, A, Steinmann, A, Arshi, M, Dwarte, T, Froio, D, Pereira, B, Ritchie, S, Chambers, Cr, Metcalf, X, Waddell, N, Pearson, Jv, Patch, Am, Nones, K, Newell, F, Mukhopadhyay, P, Addala, V, Kazakoff, S, Holmes, O, Leonard, C, Wood, S, Grimmond, Sm, Hofmann, O, Christ, A, Bruxner, T, Samra, Js, Pavlakis, N, High, Ha, Asghari, R, Merrett, Nd, Pavey, D, Das, A, Cosman, Ph, Ismail, K, O'Connnor, C, Stoita, A, Williams, D, Spigellman, A, Lam, Vw, Mcleod, D, Kirk, J, Kench, Jg, Grimison, P, Cooper, Cl, Sandroussi, C, Goodwin, A, Mead, Rs, Tucker, K, Andrews, L, Texler, M, Forest, C, Epari, Kp, Ballal, M, Fletcher, Dr, Mukhedkar, S, Zeps, N, Beilin, M, Feeney, K, Nguyen, Nq, Ruszkiewicz, Ar, Worthley, C, Chen, J, Brooke-Smith, Me, Papangelis, V, Clouston, Ad, Barbour, Ap, O'Rourke, Tj, Fawcett, Jw, Slater, K, Hatzifotis, M, Hodgkinson, P, Nikfarjam, M, Eshleman, Jr, Hruban, Rh, Wolfgang, Cl, Lawlor, Rt, Beghelli, S, Corbo, V, Scardoni, M, Bassi, C, Biankin, Av, Dixon, J, Jamieson, Nb, and Chang, Dk
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Drug Resistance ,General Physics and Astronomy ,02 engineering and technology ,Mice ,Cancer-Associated Fibroblasts ,Cell Movement ,lcsh:Science ,Inbred BALB C ,Cancer ,education.field_of_study ,Mice, Inbred BALB C ,Multidisciplinary ,Tumor ,021001 nanoscience & nanotechnology ,3. Good health ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,0210 nano-technology ,Pancreas ,Signal Transduction ,Cancer microenvironment ,Cell biology ,Science ,Population ,Perlecan ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Cell Line ,03 medical and health sciences ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Humans ,Animals ,Neoplasm Invasiveness ,education ,Cell Proliferation ,Neoplastic ,fungi ,General Chemistry ,Immunotherapy ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,Gene Expression Regulation ,Drug Resistance, Neoplasm ,Cancer cell ,Cancer research ,biology.protein ,Neoplasm ,lcsh:Q ,Stromal Cells ,Tumor Suppressor Protein p53 ,Heparan Sulfate Proteoglycans - Abstract
Heterogeneous subtypes of cancer-associated fibroblasts (CAFs) coexist within pancreatic cancer tissues and can both promote and restrain disease progression. Here, we interrogate how cancer cells harboring distinct alterations in p53 manipulate CAFs. We reveal the existence of a p53-driven hierarchy, where cancer cells with a gain-of-function (GOF) mutant p53 educate a dominant population of CAFs that establish a pro-metastatic environment for GOF and null p53 cancer cells alike. We also demonstrate that CAFs educated by null p53 cancer cells may be reprogrammed by either GOF mutant p53 cells or their CAFs. We identify perlecan as a key component of this pro-metastatic environment. Using intravital imaging, we observe that these dominant CAFs delay cancer cell response to chemotherapy. Lastly, we reveal that depleting perlecan in the stroma combined with chemotherapy prolongs mouse survival, supporting it as a potential target for anti-stromal therapies in pancreatic cancer., Subtypes of cancer associated fibroblasts can both promote and suppress tumorigenesis. Here, the authors investigate how p53 status in pancreatic cancer cells affects their interaction with cancer associated fibroblasts, and report perlecan as a mediator of the pro-metastatic environment.
- Published
- 2019
7. Targeting DNA Damage Response and Replication Stress in Pancreatic Cancer
- Author
-
Dreyer, SB, Upstill-Goddard, R, Paulus-Hock, V, Paris, C, Lampraki, EM, Dray, E, Serrels, B, Caligiuri, G, Rebus, S, Plenker, D, Galluzzo, Z, Brunton, H, Cunningham, R, Tesson, M, Nourse, C, Bailey, UM, Jones, MD, Moran-Jones, K, Wright, DW, Duthie, F, Oien, K, Evers, L, McKay, CJ, McGregor, GA, Gulati, A, Brough, R, Bajrami, I, Pettitt, S, Dziubinski, ML, Candido, J, Balkwill, F, Barry, ST, Grützmann, R, Rahib, L, Allison, S, Bailey, PJ, Biankin, AV, Beraldi, D, Cameron, E, Chang, DK, Cooke, SL, Grimwood, P, Kelly, S, Marshall, J, Martin, S, McDade, B, McElroy, D, Musgrove, EA, Ramsay, D, Wright, D, Hair, J, Jamieson, NB, Westwood, P, Williams, N, Johns, AL, Mawson, A, Scarlett, CJ, Brancato, MAL, Rowe, SJ, Simpson, SH, Martyn-Smith, M, Thomas, MT, Chantrill, LA, Chin, VT ; https://orcid.org/0000-0002-4630-4451, Chou, A ; https://orcid.org/0000-0002-8129-7170, Cowley, MJ ; https://orcid.org/0000-0002-9519-5714, Humphris, JL, Mead, RS, Nagrial, AM, Pajic, M ; https://orcid.org/0000-0002-3871-3829, Pettit, J, Pinese, M ; https://orcid.org/0000-0001-5078-6687, Rooman, I, Wu, J, Tao, J, DiPietro, R, Watson, C, Steinmann, A, Lee, HC, Wong, R, Pinho, AV, Giry-Laterriere, M, Daly, RJ, Sutherland, RL, Grimmond, SM, Waddell, N, Kassahn, KS, Miller, DK, Wilson, PJ, Patch, AM, Song, S, Harliwong, I, Idrisoglu, S, Nourbakhsh, E, Manning, S, Wani, S, Gongora, M, Anderson, M, Holmes, O, Leonard, C, Biankin, Andrew, Toon, Christopher Chien Wei ; https://orcid.org/0000-0002-0750-918X, Pavey, Darren ; https://orcid.org/0000-0001-5351-5567, Merrett, Neil ; https://orcid.org/0000-0002-8370-0293, asghari, Ray, Chang, David, Pereira, Brooke ; https://orcid.org/0000-0003-3513-1214, Dreyer, SB, Upstill-Goddard, R, Paulus-Hock, V, Paris, C, Lampraki, EM, Dray, E, Serrels, B, Caligiuri, G, Rebus, S, Plenker, D, Galluzzo, Z, Brunton, H, Cunningham, R, Tesson, M, Nourse, C, Bailey, UM, Jones, MD, Moran-Jones, K, Wright, DW, Duthie, F, Oien, K, Evers, L, McKay, CJ, McGregor, GA, Gulati, A, Brough, R, Bajrami, I, Pettitt, S, Dziubinski, ML, Candido, J, Balkwill, F, Barry, ST, Grützmann, R, Rahib, L, Allison, S, Bailey, PJ, Biankin, AV, Beraldi, D, Cameron, E, Chang, DK, Cooke, SL, Grimwood, P, Kelly, S, Marshall, J, Martin, S, McDade, B, McElroy, D, Musgrove, EA, Ramsay, D, Wright, D, Hair, J, Jamieson, NB, Westwood, P, Williams, N, Johns, AL, Mawson, A, Scarlett, CJ, Brancato, MAL, Rowe, SJ, Simpson, SH, Martyn-Smith, M, Thomas, MT, Chantrill, LA, Chin, VT ; https://orcid.org/0000-0002-4630-4451, Chou, A ; https://orcid.org/0000-0002-8129-7170, Cowley, MJ ; https://orcid.org/0000-0002-9519-5714, Humphris, JL, Mead, RS, Nagrial, AM, Pajic, M ; https://orcid.org/0000-0002-3871-3829, Pettit, J, Pinese, M ; https://orcid.org/0000-0001-5078-6687, Rooman, I, Wu, J, Tao, J, DiPietro, R, Watson, C, Steinmann, A, Lee, HC, Wong, R, Pinho, AV, Giry-Laterriere, M, Daly, RJ, Sutherland, RL, Grimmond, SM, Waddell, N, Kassahn, KS, Miller, DK, Wilson, PJ, Patch, AM, Song, S, Harliwong, I, Idrisoglu, S, Nourbakhsh, E, Manning, S, Wani, S, Gongora, M, Anderson, M, Holmes, O, Leonard, C, Biankin, Andrew, Toon, Christopher Chien Wei ; https://orcid.org/0000-0002-0750-918X, Pavey, Darren ; https://orcid.org/0000-0001-5351-5567, Merrett, Neil ; https://orcid.org/0000-0002-8370-0293, asghari, Ray, Chang, David, and Pereira, Brooke ; https://orcid.org/0000-0003-3513-1214
- Abstract
Background & Aims: Continuing recalcitrance to therapy cements pancreatic cancer (PC) as the most lethal malignancy, which is set to become the second leading cause of cancer death in our society. The study aim was to investigate the association between DNA damage response (DDR), replication stress, and novel therapeutic response in PC to develop a biomarker-driven therapeutic strategy targeting DDR and replication stress in PC. Methods: We interrogated the transcriptome, genome, proteome, and functional characteristics of 61 novel PC patient–derived cell lines to define novel therapeutic strategies targeting DDR and replication stress. Validation was done in patient-derived xenografts and human PC organoids. Results: Patient-derived cell lines faithfully recapitulate the epithelial component of pancreatic tumors, including previously described molecular subtypes. Biomarkers of DDR deficiency, including a novel signature of homologous recombination deficiency, cosegregates with response to platinum (P <.001) and PARP inhibitor therapy (P <.001) in vitro and in vivo. We generated a novel signature of replication stress that predicts response to ATR (P <.018) and WEE1 inhibitor (P <.029) treatment in both cell lines and human PC organoids. Replication stress was enriched in the squamous subtype of PC (P <.001) but was not associated with DDR deficiency. Conclusions: Replication stress and DDR deficiency are independent of each other, creating opportunities for therapy in DDR-proficient PC and after platinum therapy.
- Published
- 2021
8. The Diverse Applications of Pancreatic Ductal Adenocarcinoma Organoids
- Author
-
Low, RRJ, Lim, WW, Nguyen, PM, Lee, B, Christie, M, Burgess, AW, Gibbs, P, Grimmond, SM, Hollande, F, Putoczki, TL, Low, RRJ, Lim, WW, Nguyen, PM, Lee, B, Christie, M, Burgess, AW, Gibbs, P, Grimmond, SM, Hollande, F, and Putoczki, TL
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal solid malignancies. While immortalized cancer cell lines and genetically engineered murine models have increased our understanding of PDAC tumorigenesis, they do not recapitulate inter- and intra-patient heterogeneity. PDAC patient derived organoid (PDO) biobanks have overcome this hurdle, and provide an opportunity for the high throughput screening of potential new therapies. This review provides a summary of the PDAC PDO biobanks established to date, and discusses how they have advanced our understanding of PDAC biology. Looking forward, the development of coculturing techniques for specific immune or stromal cell populations will enable a better understanding of the crosstalk that occurs within the tumor microenvironment, and the impact of this crosstalk on treatment response.
- Published
- 2021
9. DNA methylation patterns identify subgroups of pancreatic neuroendocrine tumors with clinical association
- Author
-
Lakis, V, Lawlor, RT, Newell, F, Patch, A-M, Mafficini, A, Sadanandam, A, Koufariotis, LT, Johnston, RL, Leonard, C, Wood, S, Rusev, B, Corbo, V, Luchini, C, Cingarlini, S, Landoni, L, Salvia, R, Milella, M, Chang, D, Bailey, P, Jamieson, NB, Duthie, F, Gingras, M-C, Muzny, DM, Wheeler, DA, Gibbs, RA, Milione, M, Pederzoli, P, Samra, JS, Gill, AJ, Johns, AL, Pearson, J, Biankin, A, Grimmond, SM, Waddell, N, Nones, K, Scarpa, A, Lakis, V, Lawlor, RT, Newell, F, Patch, A-M, Mafficini, A, Sadanandam, A, Koufariotis, LT, Johnston, RL, Leonard, C, Wood, S, Rusev, B, Corbo, V, Luchini, C, Cingarlini, S, Landoni, L, Salvia, R, Milella, M, Chang, D, Bailey, P, Jamieson, NB, Duthie, F, Gingras, M-C, Muzny, DM, Wheeler, DA, Gibbs, RA, Milione, M, Pederzoli, P, Samra, JS, Gill, AJ, Johns, AL, Pearson, J, Biankin, A, Grimmond, SM, Waddell, N, Nones, K, and Scarpa, A
- Abstract
Here we report the DNA methylation profile of 84 sporadic pancreatic neuroendocrine tumors (PanNETs) with associated clinical and genomic information. We identified three subgroups of PanNETs, termed T1, T2 and T3, with distinct patterns of methylation. The T1 subgroup was enriched for functional tumors and ATRX, DAXX and MEN1 wild-type genotypes. The T2 subgroup contained tumors with mutations in ATRX, DAXX and MEN1 and recurrent patterns of chromosomal losses in half of the genome with no association between regions with recurrent loss and methylation levels. T2 tumors were larger and had lower methylation in the MGMT gene body, which showed positive correlation with gene expression. The T3 subgroup harboured mutations in MEN1 with recurrent loss of chromosome 11, was enriched for grade G1 tumors and showed histological parameters associated with better prognosis. Our results suggest a role for methylation in both driving tumorigenesis and potentially stratifying prognosis in PanNETs.
- Published
- 2021
10. Intravital imaging technology guides FAK-mediated priming in pancreatic cancer precision medicine according to Merlin status
- Author
-
Murphy, KJ, Reed, DA, Vennin, C, Conway, JRW, Nobis, M, Yin, JX, Chambers, CR, Pereira, BA, Lee, V, Filipe, EC, Trpceski, M, Ritchie, S, Lucas, MC, Warren, SC, Skhinas, JN, Magenau, A, Metcalf, XL, Stoehr, J, Major, G, Parkin, A, Bidanel, R, Lyons, RJ, Zaratzian, A, Tayao, M, Da Silva, A, Abdulkhalek, L, Gill, AJ, Johns, AL, Biankin, A, Samra, J, Grimmond, SM, Chou, A, Goetz, JG, Samuel, MS, Lyons, JG, Burgess, A, Caldon, CE, Horvath, LG, Daly, RJ, Gadegaard, N, Wang, Y, Sansom, OJ, Morton, JP, Cox, TR, Pajic, M, Herrmann, D, Timpson, P, Murphy, KJ, Reed, DA, Vennin, C, Conway, JRW, Nobis, M, Yin, JX, Chambers, CR, Pereira, BA, Lee, V, Filipe, EC, Trpceski, M, Ritchie, S, Lucas, MC, Warren, SC, Skhinas, JN, Magenau, A, Metcalf, XL, Stoehr, J, Major, G, Parkin, A, Bidanel, R, Lyons, RJ, Zaratzian, A, Tayao, M, Da Silva, A, Abdulkhalek, L, Gill, AJ, Johns, AL, Biankin, A, Samra, J, Grimmond, SM, Chou, A, Goetz, JG, Samuel, MS, Lyons, JG, Burgess, A, Caldon, CE, Horvath, LG, Daly, RJ, Gadegaard, N, Wang, Y, Sansom, OJ, Morton, JP, Cox, TR, Pajic, M, Herrmann, D, and Timpson, P
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic, chemoresistant malignancy and is characterized by a dense, desmoplastic stroma that modulates PDAC progression. Here, we visualized transient manipulation of focal adhesion kinase (FAK), which integrates bidirectional cell-environment signaling, using intravital fluorescence lifetime imaging microscopy of the FAK-based Förster resonance energy transfer biosensor in mouse and patient-derived PDAC models. Parallel real-time quantification of the FUCCI cell cycle reporter guided us to improve PDAC response to standard-of-care chemotherapy at primary and secondary sites. Critically, micropatterned pillar plates and stiffness-tunable matrices were used to pinpoint the contribution of environmental cues to chemosensitization, while fluid flow–induced shear stress assessment, patient-derived matrices, and personalized in vivo models allowed us to deconstruct how FAK inhibition can reduce PDAC spread. Last, stratification of PDAC patient samples via Merlin status revealed a patient subset with poor prognosis that are likely to respond to FAK priming before chemotherapy.
- Published
- 2021
11. PRMT5: An Emerging Target for Pancreatic Adenocarcinoma
- Author
-
Lee, MKC, Grimmond, SM, McArthur, GA, Sheppard, KE, Lee, MKC, Grimmond, SM, McArthur, GA, and Sheppard, KE
- Abstract
The overall survival of pancreatic ductal adenocarcinoma (PDAC) remains poor and its incidence is rising. Targetable mutations in PDAC are rare, thus novel therapeutic approaches are needed. Protein arginine methyltransferase 5 (PRMT5) overexpression is associated with worse survival and inhibition of PRMT5 results in decreased cancer growth across multiple cancers, including PDAC. Emerging evidence also suggests that altered RNA processing is a driver in PDAC tumorigenesis and creates a partial dependency on this process. PRMT5 inhibition induces altered splicing and this vulnerability can be exploited as a novel therapeutic approach. Three possible biological pathways underpinning the action of PRMT5 inhibitors are discussed; c-Myc regulation appears central to its action in the PDAC setting. Whilst homozygous MTAP deletion and symmetrical dimethylation levels are associated with increased sensitivity to PRMT5 inhibition, neither measure robustly predicts its growth inhibitory response. The immunomodulatory effect of PRMT5 inhibitors on the tumour microenvironment will also be discussed, based on emerging evidence that PDAC stroma has a significant bearing on disease behaviour and response to therapy. Lastly, with the above caveats in mind, current knowledge gaps and the implications and rationales for PRMT5 inhibitor development in PDAC will be explored.
- Published
- 2021
12. Sex differences in oncogenic mutational processes
- Author
-
Li, CH, Prokopec, SD, Sun, RX, Yousif, F, Schmitz, N, Al-Shahrour, F, Atwal, G, Bailey, PJ, Biankin, AV, Boutros, PC, Campbell, PJ, Chang, DK, Cooke, SL, Deshpande, V, Faltas, BM, Faquin, WC, Garraway, L, Getz, G, Grimmond, SM, Haider, S, Hoadley, KA, Jiao, W, Kaiser, VB, Karlić, R, Kato, M, Kübler, K, Lazar, AJ, Louis, DN, Margolin, A, Martin, S, Nahal-Bose, HK, Nielsen, GP, Nik-Zainal, S, Omberg, L, P’ng, C, Perry, MD, Polak, P, Rheinbay, E, Rubin, MA, Semple, CA, Sgroi, DC, Shibata, T, Siebert, R, Smith, J, Stein, LD, Stobbe, MD, Thai, K, Wright, DW, Wu, CL, Yuan, K, Zhang, J, Aaltonen, LA, Abascal, F, Abeshouse, A, Aburatani, H, Adams, DJ, Agrawal, N, Ahn, KS, Ahn, SM, Aikata, H, Akbani, R, Akdemir, KC, Al-Ahmadie, H, Al-Sedairy, ST, Alawi, M, Albert, M, Aldape, K, Alexandrov, LB, Ally, A, Alsop, K, Alvarez, EG, Amary, F, Amin, SB, Aminou, B, Ammerpohl, O, Anderson, MJ, Ang, Y, Antonello, D, Anur, P, Aparicio, S, Appelbaum, EL, Arai, Y, Aretz, A, Arihiro, K, Ariizumi, SI, Armenia, J, Arnould, L, Asa, S, Assenov, Y, Aukema, S, Auman, JT, Aure, MR, Awadalla, P, Aymerich, M, Bader, GD, Baez-Ortega, A, Pajic, Marina ; https://orcid.org/0000-0002-3871-3829, Merrett, Neil ; https://orcid.org/0000-0002-8370-0293, Pinese, Mark ; https://orcid.org/0000-0001-5078-6687, Lau, Loretta ; https://orcid.org/0000-0002-3172-0970, Gill, Anthony, Toon, Christopher Chien Wei ; https://orcid.org/0000-0002-0750-918X, Chou, Angela ; https://orcid.org/0000-0002-8129-7170, Johns, Amber, Li, CH, Prokopec, SD, Sun, RX, Yousif, F, Schmitz, N, Al-Shahrour, F, Atwal, G, Bailey, PJ, Biankin, AV, Boutros, PC, Campbell, PJ, Chang, DK, Cooke, SL, Deshpande, V, Faltas, BM, Faquin, WC, Garraway, L, Getz, G, Grimmond, SM, Haider, S, Hoadley, KA, Jiao, W, Kaiser, VB, Karlić, R, Kato, M, Kübler, K, Lazar, AJ, Louis, DN, Margolin, A, Martin, S, Nahal-Bose, HK, Nielsen, GP, Nik-Zainal, S, Omberg, L, P’ng, C, Perry, MD, Polak, P, Rheinbay, E, Rubin, MA, Semple, CA, Sgroi, DC, Shibata, T, Siebert, R, Smith, J, Stein, LD, Stobbe, MD, Thai, K, Wright, DW, Wu, CL, Yuan, K, Zhang, J, Aaltonen, LA, Abascal, F, Abeshouse, A, Aburatani, H, Adams, DJ, Agrawal, N, Ahn, KS, Ahn, SM, Aikata, H, Akbani, R, Akdemir, KC, Al-Ahmadie, H, Al-Sedairy, ST, Alawi, M, Albert, M, Aldape, K, Alexandrov, LB, Ally, A, Alsop, K, Alvarez, EG, Amary, F, Amin, SB, Aminou, B, Ammerpohl, O, Anderson, MJ, Ang, Y, Antonello, D, Anur, P, Aparicio, S, Appelbaum, EL, Arai, Y, Aretz, A, Arihiro, K, Ariizumi, SI, Armenia, J, Arnould, L, Asa, S, Assenov, Y, Aukema, S, Auman, JT, Aure, MR, Awadalla, P, Aymerich, M, Bader, GD, Baez-Ortega, A, Pajic, Marina ; https://orcid.org/0000-0002-3871-3829, Merrett, Neil ; https://orcid.org/0000-0002-8370-0293, Pinese, Mark ; https://orcid.org/0000-0001-5078-6687, Lau, Loretta ; https://orcid.org/0000-0002-3172-0970, Gill, Anthony, Toon, Christopher Chien Wei ; https://orcid.org/0000-0002-0750-918X, Chou, Angela ; https://orcid.org/0000-0002-8129-7170, and Johns, Amber
- Abstract
Sex differences have been observed in multiple facets of cancer epidemiology, treatment and biology, and in most cancers outside the sex organs. Efforts to link these clinical differences to specific molecular features have focused on somatic mutations within the coding regions of the genome. Here we report a pan-cancer analysis of sex differences in whole genomes of 1983 tumours of 28 subtypes as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium. We both confirm the results of exome studies, and also uncover previously undescribed sex differences. These include sex-biases in coding and non-coding cancer drivers, mutation prevalence and strikingly, in mutational signatures related to underlying mutational processes. These results underline the pervasiveness of molecular sex differences and strengthen the call for increased consideration of sex in molecular cancer research.
- Published
- 2020
13. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing
- Author
-
Cortes-Ciriano, I, Lee, JJ-K, Xi, R, Jain, D, Jung, YL, Yang, L, Gordenin, D, Klimczak, LJ, Zhang, C-Z, Pellman, DS, Park, PJ, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Beroukhim, R, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Campbell, PJ, Chan, K, Chen, K, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gerstein, M, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Ju, YS, Kazanov, MD, Koh, Y, Korbel, JO, Kumar, K, Lee, EA, Li, Y, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Pearson, J, Puiggros, M, Rippe, K, Roberts, ND, Roberts, SA, Rodriguez-Martin, B, Schumacher, SE, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Wala, JA, Weischenfeldt, J, Yao, X, Yoon, S-S, Zamora, J, Alsop, K, Christie, EL, Fereday, S, Mileshkin, L, Mitchell, C, Thorne, H, Traficante, N, Cmero, M, Cowin, PA, Hamilton, A, Mir Arnau, G, Vedururu, R, Grimmond, SM, Hofmann, O, Morrison, C, Oien, KA, Pairojkul, C, Waring, PM, van de Vijver, MJ, Behren, A, Cortes-Ciriano, I, Lee, JJ-K, Xi, R, Jain, D, Jung, YL, Yang, L, Gordenin, D, Klimczak, LJ, Zhang, C-Z, Pellman, DS, Park, PJ, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Beroukhim, R, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Campbell, PJ, Chan, K, Chen, K, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gerstein, M, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Ju, YS, Kazanov, MD, Koh, Y, Korbel, JO, Kumar, K, Lee, EA, Li, Y, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Pearson, J, Puiggros, M, Rippe, K, Roberts, ND, Roberts, SA, Rodriguez-Martin, B, Schumacher, SE, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Wala, JA, Weischenfeldt, J, Yao, X, Yoon, S-S, Zamora, J, Alsop, K, Christie, EL, Fereday, S, Mileshkin, L, Mitchell, C, Thorne, H, Traficante, N, Cmero, M, Cowin, PA, Hamilton, A, Mir Arnau, G, Vedururu, R, Grimmond, SM, Hofmann, O, Morrison, C, Oien, KA, Pairojkul, C, Waring, PM, van de Vijver, MJ, and Behren, A
- Abstract
Chromothripsis is a mutational phenomenon characterized by massive, clustered genomic rearrangements that occurs in cancer and other diseases. Recent studies in selected cancer types have suggested that chromothripsis may be more common than initially inferred from low-resolution copy-number data. Here, as part of the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA), we analyze patterns of chromothripsis across 2,658 tumors from 38 cancer types using whole-genome sequencing data. We find that chromothripsis events are pervasive across cancers, with a frequency of more than 50% in several cancer types. Whereas canonical chromothripsis profiles display oscillations between two copy-number states, a considerable fraction of events involve multiple chromosomes and additional structural alterations. In addition to non-homologous end joining, we detect signatures of replication-associated processes and templated insertions. Chromothripsis contributes to oncogene amplification and to inactivation of genes such as mismatch-repair-related genes. These findings show that chromothripsis is a major process that drives genome evolution in human cancer.
- Published
- 2020
14. Targeting enhancer switching overcomes non-genetic drug resistance in acute myeloid leukaemia
- Author
-
Bell, CC, Fenne, KA, Chan, Y-C, Rambow, F, Yeung, MM, Vassiliadis, D, Lara, L, Yeh, P, Martelotto, LG, Rogiers, A, Kremer, BE, Barbash, O, Mohammad, HP, Johanson, TM, Burr, ML, Dhar, A, Karpinich, N, Tian, L, Tyler, DS, MacPherson, L, Shi, J, Pinnawala, N, Fong, CY, Papenfuss, AT, Grimmond, SM, Dawson, S-J, Allan, RS, Kruger, RG, Vakoc, CR, Goode, DL, Naik, SH, Gilan, O, Lam, EYN, Marine, J-C, Prinjha, RK, Dawson, MA, Bell, CC, Fenne, KA, Chan, Y-C, Rambow, F, Yeung, MM, Vassiliadis, D, Lara, L, Yeh, P, Martelotto, LG, Rogiers, A, Kremer, BE, Barbash, O, Mohammad, HP, Johanson, TM, Burr, ML, Dhar, A, Karpinich, N, Tian, L, Tyler, DS, MacPherson, L, Shi, J, Pinnawala, N, Fong, CY, Papenfuss, AT, Grimmond, SM, Dawson, S-J, Allan, RS, Kruger, RG, Vakoc, CR, Goode, DL, Naik, SH, Gilan, O, Lam, EYN, Marine, J-C, Prinjha, RK, and Dawson, MA
- Abstract
Non-genetic drug resistance is increasingly recognised in various cancers. Molecular insights into this process are lacking and it is unknown whether stable non-genetic resistance can be overcome. Using single cell RNA-sequencing of paired drug naïve and resistant AML patient samples and cellular barcoding in a unique mouse model of non-genetic resistance, here we demonstrate that transcriptional plasticity drives stable epigenetic resistance. With a CRISPR-Cas9 screen we identify regulators of enhancer function as important modulators of the resistant cell state. We show that inhibition of Lsd1 (Kdm1a) is able to overcome stable epigenetic resistance by facilitating the binding of the pioneer factor, Pu.1 and cofactor, Irf8, to nucleate new enhancers that regulate the expression of key survival genes. This enhancer switching results in the re-distribution of transcriptional co-activators, including Brd4, and provides the opportunity to disable their activity and overcome epigenetic resistance. Together these findings highlight key principles to help counteract non-genetic drug resistance.
- Published
- 2019
15. Platypus Induced Pluripotent Stem Cells: the Unique Pluripotency Signature of a Monotreme
- Author
-
Whitworth, DJ, primary, Limnios, IJ, additional, Gauthier, M-E, additional, Weeratunga, P, additional, Ovchinnikov, DA, additional, Baillie, G, additional, Grimmond, SM, additional, Marshall Graves, JA, additional, and Wolvetang, EJ, additional
- Published
- 2018
- Full Text
- View/download PDF
16. Telomere sequence content can be used to determine ALT activity in tumours
- Author
-
Lee, M, Teber, ET, Holmes, O, Nones, K, Patch, A-M, Dagg, RA, Lau, LMS, Lee, JH, Napier, CE, Arthur, JW, Grimmond, SM, Hayward, NK, Johansson, PA, Mann, GJ, Scolyer, RA, Wilmott, JS, Reddel, RR, Pearson, JV, Waddell, N, Pickett, HA, Lee, M, Teber, ET, Holmes, O, Nones, K, Patch, A-M, Dagg, RA, Lau, LMS, Lee, JH, Napier, CE, Arthur, JW, Grimmond, SM, Hayward, NK, Johansson, PA, Mann, GJ, Scolyer, RA, Wilmott, JS, Reddel, RR, Pearson, JV, Waddell, N, and Pickett, HA
- Abstract
The replicative immortality of human cancer cells is achieved by activation of a telomere maintenance mechanism (TMM). To achieve this, cancer cells utilise either the enzyme telomerase, or the Alternative Lengthening of Telomeres (ALT) pathway. These distinct molecular pathways are incompletely understood with respect to activation and propagation, as well as their associations with clinical outcomes. We have identified significant differences in the telomere repeat composition of tumours that use ALT compared to tumours that do not. We then employed a machine learning approach to stratify tumours according to telomere repeat content with an accuracy of 91.6%. Importantly, this classification approach is applicable across all tumour types. Analysis of pathway mutations that were under-represented in ALT tumours, across 1,075 tumour samples, revealed that the autophagy, cell cycle control of chromosomal replication, and transcriptional regulatory network in embryonic stem cells pathways are involved in the survival of ALT tumours. Overall, our approach demonstrates that telomere sequence content can be used to stratify ALT activity in cancers, and begin to define the molecular pathways involved in ALT activation.
- Published
- 2018
17. Tailored first-line and second-line CDK4-targeting treatment combinations in mouse models of pancreatic cancer
- Author
-
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, 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, and Pajic, M
- Abstract
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.
- Published
- 2018
18. Lost in translation: Returning germline genetic results in genome-scale cancer research
- Author
-
Johns, AL, McKay, SH, Humphris, JL, Pinese, M ; https://orcid.org/0000-0001-5078-6687, Chantrill, LA ; https://orcid.org/0000-0002-5790-0208, Mead, RS, Tucker, K ; https://orcid.org/0000-0002-6452-9427, Andrews, L, Goodwin, A, Leonard, C, High, HA, Nones, K, Waddell, N, Patch, AM, Merrett, ND ; https://orcid.org/0000-0002-8370-0293, Pavlakis, N, Kassahn, KS, Samra, JS, Miller, DK, Chang, DK, Pajic, M ; https://orcid.org/0000-0002-3871-3829, Pearson, JV, Grimmond, SM, Zeps, N, Gill, AJ, Biankin, AV, Chin, VT ; https://orcid.org/0000-0002-4630-4451, Chou, A ; https://orcid.org/0000-0002-8129-7170, Steinmann, A, Arshi, M, Drury, A, Froio, D, Morgan, A, Timpson, P, Hermann, D, Vennin, C, Warren, S ; https://orcid.org/0000-0002-5253-7147, Wu, J, Pinho, AV, Newell, F, Mukhopadhyay, P, Addala, V, Kazakoff, S, Holmes, O, Wood, S, Xu, C, Hofmann, O, Wilson, PJ, Christ, A, Bruxner, T, Samra, S, Arena, J, Mittal, A, Asghari, R, Pavey, D ; https://orcid.org/0000-0001-5351-5567, Das, A, Cosman, PH, Ismail, K, O'Connnor, C, Williams, D, Spigellman, A, Lam, W, McLeod, D, Nagrial, AM, Kirk, J, James, V, Grimison, P, Cooper, CL, Sandroussi, C, Forest, C, Epari, KP, Ballal, M, Fletcher, DR, Mukhedkar, S, Beilin, M, Feeney, K, Nguyen, NQ, Ruszkiewicz, AR, Worthley, C, Brooke-Smith, ME, Papangelis, V, Clouston, AD, Martin, P, Barbour, AP, O'Rourke, TJ, Fawcett, JW, Slater, K, Hatzifotis, M, Hodgkinson, P, Hruban, RH, Wolfgang, CL, Hodgin, M, Lawlor, RT, Beghelli, S, Corbo, V, Scardoni, M, Bassi, C, Bailey, P, Martin, S, Musgrove, EA, Herrmann, David ; https://orcid.org/0000-0002-9514-7501, Johns, AL, McKay, SH, Humphris, JL, Pinese, M ; https://orcid.org/0000-0001-5078-6687, Chantrill, LA ; https://orcid.org/0000-0002-5790-0208, Mead, RS, Tucker, K ; https://orcid.org/0000-0002-6452-9427, Andrews, L, Goodwin, A, Leonard, C, High, HA, Nones, K, Waddell, N, Patch, AM, Merrett, ND ; https://orcid.org/0000-0002-8370-0293, Pavlakis, N, Kassahn, KS, Samra, JS, Miller, DK, Chang, DK, Pajic, M ; https://orcid.org/0000-0002-3871-3829, Pearson, JV, Grimmond, SM, Zeps, N, Gill, AJ, Biankin, AV, Chin, VT ; https://orcid.org/0000-0002-4630-4451, Chou, A ; https://orcid.org/0000-0002-8129-7170, Steinmann, A, Arshi, M, Drury, A, Froio, D, Morgan, A, Timpson, P, Hermann, D, Vennin, C, Warren, S ; https://orcid.org/0000-0002-5253-7147, Wu, J, Pinho, AV, Newell, F, Mukhopadhyay, P, Addala, V, Kazakoff, S, Holmes, O, Wood, S, Xu, C, Hofmann, O, Wilson, PJ, Christ, A, Bruxner, T, Samra, S, Arena, J, Mittal, A, Asghari, R, Pavey, D ; https://orcid.org/0000-0001-5351-5567, Das, A, Cosman, PH, Ismail, K, O'Connnor, C, Williams, D, Spigellman, A, Lam, W, McLeod, D, Nagrial, AM, Kirk, J, James, V, Grimison, P, Cooper, CL, Sandroussi, C, Forest, C, Epari, KP, Ballal, M, Fletcher, DR, Mukhedkar, S, Beilin, M, Feeney, K, Nguyen, NQ, Ruszkiewicz, AR, Worthley, C, Brooke-Smith, ME, Papangelis, V, Clouston, AD, Martin, P, Barbour, AP, O'Rourke, TJ, Fawcett, JW, Slater, K, Hatzifotis, M, Hodgkinson, P, Hruban, RH, Wolfgang, CL, Hodgin, M, Lawlor, RT, Beghelli, S, Corbo, V, Scardoni, M, Bassi, C, Bailey, P, Martin, S, Musgrove, EA, and Herrmann, David ; https://orcid.org/0000-0002-9514-7501
- Abstract
Background: The return of research results (RoR) remains a complex and well-debated issue. Despite the debate, actual data related to the experience of giving individual results back, and the impact these results may have on clinical care and health outcomes, is sorely lacking. Through the work of the Australian Pancreatic Cancer Genome Initiative (APGI) we: (1) delineate the pathway back to the patient where actionable research data were identified; and (2) report the clinical utilisation of individual results returned. Using this experience, we discuss barriers and opportunities associated with a comprehensive process of RoR in large-scale genomic research that may be useful for others developing their own policies. Methods: We performed whole-genome (n = 184) and exome (n = 208) sequencing of matched tumour-normal DNA pairs from 392 patients with sporadic pancreatic cancer (PC) as part of the APGI. We identified pathogenic germline mutations in candidate genes (n = 130) with established predisposition to PC or medium-high penetrance genes with well-defined cancer associated syndromes or phenotypes. Variants from candidate genes were annotated and classified according to international guidelines. Variants were considered actionable if clinical utility was established, with regard to prevention, diagnosis, prognostication and/or therapy. Results: A total of 48,904 germline variants were identified, with 2356 unique variants undergoing annotation and in silico classification. Twenty cases were deemed actionable and were returned via previously described RoR framework, representing an actionable finding rate of 5.1%. Overall, 1.78% of our cohort experienced clinical benefit from RoR. Conclusion: Returning research results within the context of large-scale genomics research is a labour-intensive, highly variable, complex operation. Results that warrant action are not infrequent, but the prevalence of those who experience a clinical difference as a result of returning
- Published
- 2017
19. Mitochondrial mutations and metabolic adaptation in pancreatic cancer
- Author
-
Hardie, R-A, van Dam, E, Cowley, M, Han, T-L, Balaban, S, Pajic, M, Pinese, M, Iconomou, M, Shearer, RF, McKenna, J, Miller, D, Waddell, N, Pearson, JV, Grimmond, SM, Sazanov, L, Biankin, AV, Villas-Boas, S, Hoy, AJ, Turner, N, Saunders, DN, Hardie, R-A, van Dam, E, Cowley, M, Han, T-L, Balaban, S, Pajic, M, Pinese, M, Iconomou, M, Shearer, RF, McKenna, J, Miller, D, Waddell, N, Pearson, JV, Grimmond, SM, Sazanov, L, Biankin, AV, Villas-Boas, S, Hoy, AJ, Turner, N, and Saunders, DN
- Abstract
BACKGROUND: Pancreatic cancer has a five-year survival rate of ~8%, with characteristic molecular heterogeneity and restricted treatment options. Targeting metabolism has emerged as a potentially effective therapeutic strategy for cancers such as pancreatic cancer, which are driven by genetic alterations that are not tractable drug targets. Although somatic mitochondrial genome (mtDNA) mutations have been observed in various tumors types, understanding of metabolic genotype-phenotype relationships is limited. METHODS: We deployed an integrated approach combining genomics, metabolomics, and phenotypic analysis on a unique cohort of patient-derived pancreatic cancer cell lines (PDCLs). Genome analysis was performed via targeted sequencing of the mitochondrial genome (mtDNA) and nuclear genes encoding mitochondrial components and metabolic genes. Phenotypic characterization of PDCLs included measurement of cellular oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) using a Seahorse XF extracellular flux analyser, targeted metabolomics and pathway profiling, and radiolabelled glutamine tracing. RESULTS: We identified 24 somatic mutations in the mtDNA of 12 patient-derived pancreatic cancer cell lines (PDCLs). A further 18 mutations were identified in a targeted study of ~1000 nuclear genes important for mitochondrial function and metabolism. Comparison with reference datasets indicated a strong selection bias for non-synonymous mutants with predicted functional effects. Phenotypic analysis showed metabolic changes consistent with mitochondrial dysfunction, including reduced oxygen consumption and increased glycolysis. Metabolomics and radiolabeled substrate tracing indicated the initiation of reductive glutamine metabolism and lipid synthesis in tumours. CONCLUSIONS: The heterogeneous genomic landscape of pancreatic tumours may converge on a common metabolic phenotype, with individual tumours adapting to increased anabolic demands via different ge
- Published
- 2017
20. Recurrent noncoding regulatory mutations in pancreatic ductal adenocarcinoma
- Author
-
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, 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, and Tuveson, DA
- Abstract
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.
- Published
- 2017
21. Whole Exome Sequencing in Patients with White Matter Abnormalities
- Author
-
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, 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, and Taft, RJ
- Abstract
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.
- Published
- 2016
22. Dual targeting of p53 and c-MYC selectively eliminates leukaemic stem cells
- Author
-
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, 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, and Holyoake, TL
- Abstract
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.
- Published
- 2016
23. Identification of the CIMP-like subtype and aberrant methylation of members of the chromosomal segregation and spindle assembly pathways in esophageal adenocarcinoma
- Author
-
Krause, L, Nones, K, Loffler, KA, Nancarrow, D, Oey, H, Tang, YH, Wayte, NJ, Patch, AM, Patel, K, Brosda, S, Manning, S, Lampe, G, Clouston, A, Thomas, J, Stoye, J, Hussey, DJ, Watson, DI, Lord, RV, Phillips, WA, Gotley, D, Smithers, BM, Whiteman, DC, Hayward, NK, Grimmond, SM, Waddell, N, Barbour, AP, Krause, L, Nones, K, Loffler, KA, Nancarrow, D, Oey, H, Tang, YH, Wayte, NJ, Patch, AM, Patel, K, Brosda, S, Manning, S, Lampe, G, Clouston, A, Thomas, J, Stoye, J, Hussey, DJ, Watson, DI, Lord, RV, Phillips, WA, Gotley, D, Smithers, BM, Whiteman, DC, Hayward, NK, Grimmond, SM, Waddell, N, and Barbour, AP
- Abstract
The incidence of esophageal adenocarcinoma (EAC) has risen significantly over recent decades. Although survival has improved, cure rates remain poor, with <20% of patients surviving 5 years. This is the first study to explore methylome, transcriptome and ENCODE data to characterize the role of methylation in EAC. We investigate the genome-wide methylation profile of 250 samples including 125 EAC, 19 Barrett's esophagus (BE), 85 squamous esophagus and 21 normal stomach. Transcriptome data of 70 samples (48 EAC, 4 BE and 18 squamous esophagus) were used to identify changes in methylation associated with gene expression. BE and EAC showed similar methylation profiles, which differed from squamous tissue. Hypermethylated sites in EAC and BE were mainly located in CpG-rich promoters. A total of 18575 CpG sites associated with 5538 genes were differentially methylated, 63% of these genes showed significant correlation between methylation and mRNA expression levels. Pathways involved in tumorigenesis including cell adhesion, TGF and WNT signaling showed enrichment for genes aberrantly methylated. Genes involved in chromosomal segregation and spindle formation were aberrantly methylated. Given the recent evidence that chromothripsis may be a driver mechanism in EAC, the role of epigenetic perturbation of these pathways should be further investigated. The methylation profiles revealed two EAC subtypes, one associated with widespread CpG island hypermethylation overlapping H3K27me3 marks and binding sites of the Polycomb proteins. These subtypes were supported by an independent set of 89 esophageal cancer samples. The most hypermethylated tumors showed worse patient survival.
- Published
- 2016
24. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling
- Author
-
Nones, K, Waddell, N, Song, S, Patch, Am, Miller, D, Johns, A, Wu, J, Kassahn, Ks, Wood, D, Bailey, P, Fink, L, Manning, S, Christ, An, Nourse, C, Kazakoff, S, Taylor, D, Leonard, C, Chang, Dk, Jones, Md, Thomas, M, Watson, C, Pinese, M, Cowley, M, Rooman, I, Pajic, M, Butturini, Giovanni, Malpaga, A, Corbo, Vincenzo, Crippa, Stefano, Falconi, M, Zamboni, Giuseppe, Castelli, P, Lawlor, Rita Teresa, Gill, Aj, Scarpa, Aldo, Pearson, Jv, Biankin, Av, Grimmond, Sm, Apgi, Nones, K, Waddell, N, Song, S, Patch, Am, Miller, D, Johns, A, Wu, J, Kassahn, K, Wood, D, Bailey, P, Fink, L, Manning, S, Christ, An, Nourse, C, Kazakoff, S, Taylor, D, Leonard, C, Chang, Dk, Jones, Md, Thomas, M, Watson, C, Pinese, M, Cowley, M, Rooman, I, Pajic, M, Apgi, Butturini, G, Malpaga, A, Corbo, V, Falconi, Massimo, Zamboni, G, Castelli, P, Lawlor, Rt, Gill, Aj, Scarpa, A, Pearson, Jv, Biankin, Av, Grimmond, S. M., Crippa, Stefano, Basic (bio-) Medical Sciences, and Laboratory for Medical and Molecular Oncology
- Subjects
Adult ,Male ,pancreatic cancer ,Integrin alpha2 ,Nerve Tissue Proteins ,Signal transduction ,Epigenesis, Genetic ,axon-guidance ,Transforming Growth Factor beta ,stellate cell activation ,Humans ,RNA, Messenger ,Receptors, Immunologic ,Promoter Regions, Genetic ,Aged ,DNA methylation ,Base Sequence ,SLIT-ROBO pathway ,Gene Expression Profiling ,Pancreatic Stellate Cells ,Pancreatic Ducts ,Membrane Proteins ,cell adhesion ,Sequence Analysis, DNA ,Middle Aged ,Proto-Oncogene Proteins c-met ,SLIT-ROBO pathway, axon-guidance, methylation, pancreatic cancer, stellate cell activation ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,Wnt Proteins ,integrins ,Intercellular Signaling Peptides and Proteins ,Female ,methylation ,aged, 80 and over ,Carcinoma, Pancreatic Ductal - Abstract
The importance of epigenetic modifications such as DNA methylation in tumorigenesis is increasingly being appreciated. To define the genome-wide pattern of DNA methylation in pancreatic ductal adenocarcinomas (PDAC), we captured the methylation profiles of 167 untreated resected PDACs and compared them to a panel of 29 adjacent nontransformed pancreata using high-density arrays. A total of 11,634 CpG sites associated with 3,522 genes were significantly differentially methylated (DM) in PDAC and were capable of segregating PDAC from non-malignant pancreas, regardless of tumor cellularity. As expected, PDAC hypermethylation was most prevalent in the 5' region of genes (including the proximal promoter, 5'UTR and CpG islands). Approximately 33% DM genes showed significant inverse correlation with mRNA expression levels. Pathway analysis revealed an enrichment of aberrantly methylated genes involved in key molecular mechanisms important to PDAC: TGF-β, WNT, integrin signaling, cell adhesion, stellate cell activation and axon guidance. Given the recent discovery that SLIT-ROBO mutations play a clinically important role in PDAC, the role of epigenetic perturbation of axon guidance was pursued in more detail. Bisulfite amplicon deep sequencing and qRT-PCR expression analyses confirmed recurrent perturbation of axon guidance pathway genes SLIT2, SLIT3, ROBO1, ROBO3, ITGA2 and MET and suggests epigenetic suppression of SLIT-ROBO signaling and up-regulation of MET and ITGA2 expression. Hypomethylation of MET and ITGA2 correlated with high gene expression, which was associated with poor survival. These data suggest that aberrant methylation plays an important role in pancreatic carcinogenesis affecting core signaling pathways with potential implications for the disease pathophysiology and therapy.
- Published
- 2014
25. RON is not a prognostic marker for resectable pancreatic cancer
- Author
-
Tactacan, Carole M, Chang, David K, Cowley, Mark J, Humphrey, Emily S, Jianmin, Wu, Gill, Anthony J, Chou, Angela, Nones, Katia, Grimmond, Sean M, Sutherland, Robert L, Biankin, Andrew V, Daly, Roger J, Biankin, Av, Johns, Al, Mawson, A, Chang, Dk, Scarlett, Cj, Brancato, Ma, Rowe, Sj, Simpson, Sl, Martyn-Smith, M, Chantrill, La, Chin, Vt, Chou, A, Cowley, Mj, Caplan, W, Humphris, Jl, Jones, Md, Mead, R, Nagrial, Am, Pajic, M, Pettit, J, Pinese, M, Rooman, I, Wu, Jun, Daly, Rj, Musgrove, Ea, Sutherland, Rl, Grimmond, Sm, Waddell, N, Kassahn, Ks, Miller, Dk, Wilson, Pj, Patch, Am, Song, S, Harliwong, I, Idrisoglu, S, Nourse, C, Nourbakhsh, E, Manning, S, Wani, S, Gongora, M, Anderson, Michela, Holmes, O, Leonard, C, Taylor, D, Wood, JOHN STEPHAN, Xu, C, Nones, K, Fink, Julika, Christ, A, Bruxner, T, Cloonan, N, Newell, F, Pearson, Jv, Samra, Js, Gill, Aj, Nickpavlakis, Guminski, A, Toon, C, Asghari, R, Merrett, Nd, Pavey, Da, Das, A, Cosman, Ph, Ismail, K, O'Connor, C, Lam, Vw, Mcleod, D, Pleass, Hc, James, V, Kench, Jg, Cooper, Cl, Joseph, D, Sandroussi, C, Crawford, M, Texler, M, Forrest, C, Laycock, A, Epari, Kp, Ballal, M, Fletcher, Dr, Mukhedkar, S, Spry, Na, Deboer, B, Chai, M, Feeney, K, Zeps, N, Beilin, M, Nguyen, Nq, Ruszkiewicz, Ar, Worthley, C, Tan, Cp, Debrencini, T, Chen, J, Brooke-Smith, Me, Papangelis, V, Tang, H, Barbour, Ap, Clouston, Ad, Martin, P, O'Rourke, Tj, Chiang, A, Fawcett, Jw, Slater, K, Yeung, S, Hatzifotis, M, Hodgkinson, P, Christophi, C, Nikfarjam, M, Eshleman, Jr, Hruban, Rh, Maitra, A, Iacobuzio-Donahue, Ca, Schulick, Rd, Wolfgang, Cl, Morgan, Ra, Scarpa, A, Lawlor, Rt, Beghelli, S, Corbo, V, Scardoni, M, Bassi, C, Tempero, Ma., and Basic (bio-) Medical Sciences
- Subjects
Oncology ,Male ,Receptor Protein-Tyrosine Kinases/analysis ,Cancer Research ,Receptor tyrosine kinase ,Kaplan-Meier Estimate ,Surgical oncology ,80 and over ,Prospective cohort study ,Pancreatic Neoplasms/metabolism ,Biomarker ,Gemcitabine ,Chemotherapy ,Adenocarcinoma ,Adult ,Aged ,Aged, 80 and over ,Biomarkers, Tumor ,Blotting, Western ,Female ,Humans ,Immunohistochemistry ,Middle Aged ,Pancreatic Neoplasms ,Prognosis ,Proportional Hazards Models ,Receptor Protein-Tyrosine Kinases ,Tissue Array Analysis ,Tumor ,Blotting ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Adenocarcinoma/metabolism ,CA19-9 ,Western ,Research Article ,medicine.drug ,medicine.medical_specialty ,lcsh:RC254-282 ,Biomarkers, Tumor/analysis ,Breast cancer ,Internal medicine ,Pancreatic cancer ,medicine ,Genetics ,business.industry ,Cancer ,medicine.disease ,business ,aged, 80 and over ,Biomarkers - Abstract
Background The receptor tyrosine kinase RON exhibits increased expression during pancreatic cancer progression and promotes migration, invasion and gemcitabine resistance of pancreatic cancer cells in experimental models. However, the prognostic significance of RON expression in pancreatic cancer is unknown. Methods RON expression was characterized in several large cohorts, including a prospective study, totaling 492 pancreatic cancer patients and relationships with patient outcome and clinico-pathologic variables were assessed. Results RON expression was associated with outcome in a training set, but this was not recapitulated in the validation set, nor was there any association with therapeutic responsiveness in the validation set or the prospective study. Conclusions Although RON is implicated in pancreatic cancer progression in experimental models, and may constitute a therapeutic target, RON expression is not associated with prognosis or therapeutic responsiveness in resected pancreatic cancer.
- Published
- 2012
26. A comprehensive assessment of somatic mutation detection in cancer using whole-genome sequencing
- Author
-
Alioto, TS, Buchhalter, I, Derdak, S, Hutter, B, Eldridge, MD, Hovig, E, Heisler, LE, Beck, TA, Simpson, JT, Tonon, L, Sertier, A-S, Patch, A-M, Jaeger, N, Ginsbach, P, Drews, R, Paramasivam, N, Kabbe, R, Chotewutmontri, S, Diessl, N, Previti, C, Schmidt, S, Brors, B, Feuerbach, L, Heinold, M, Groebner, S, Korshunov, A, Tarpey, PS, Butler, AP, Hinton, J, Jones, D, Menzies, A, Raine, K, Shepherd, R, Stebbings, L, Teague, JW, Ribeca, P, Giner, FC, Beltran, S, Raineri, E, Dabad, M, Heath, SC, Gut, M, Denroche, RE, Harding, NJ, Yamaguchi, TN, Fujimoto, A, Nakagawa, H, Quesada, C, Valdes-Mas, R, Nakken, S, Vodak, D, Bower, L, Lynch, AG, Anderson, CL, Waddell, N, Pearson, JV, Grimmond, SM, Peto, M, Spellman, P, He, M, Kandoth, C, Lee, S, Zhang, J, Letourneau, L, Ma, S, Seth, S, Torrents, D, Xi, L, Wheeler, DA, Lopez-Otin, C, Campo, E, Campbell, PJ, Boutros, PC, Puente, XS, Gerhard, DS, Pfister, SM, McPherson, JD, Hudson, TJ, Schlesner, M, Lichter, P, Eils, R, Jones, DTW, Gut, IG, Alioto, TS, Buchhalter, I, Derdak, S, Hutter, B, Eldridge, MD, Hovig, E, Heisler, LE, Beck, TA, Simpson, JT, Tonon, L, Sertier, A-S, Patch, A-M, Jaeger, N, Ginsbach, P, Drews, R, Paramasivam, N, Kabbe, R, Chotewutmontri, S, Diessl, N, Previti, C, Schmidt, S, Brors, B, Feuerbach, L, Heinold, M, Groebner, S, Korshunov, A, Tarpey, PS, Butler, AP, Hinton, J, Jones, D, Menzies, A, Raine, K, Shepherd, R, Stebbings, L, Teague, JW, Ribeca, P, Giner, FC, Beltran, S, Raineri, E, Dabad, M, Heath, SC, Gut, M, Denroche, RE, Harding, NJ, Yamaguchi, TN, Fujimoto, A, Nakagawa, H, Quesada, C, Valdes-Mas, R, Nakken, S, Vodak, D, Bower, L, Lynch, AG, Anderson, CL, Waddell, N, Pearson, JV, Grimmond, SM, Peto, M, Spellman, P, He, M, Kandoth, C, Lee, S, Zhang, J, Letourneau, L, Ma, S, Seth, S, Torrents, D, Xi, L, Wheeler, DA, Lopez-Otin, C, Campo, E, Campbell, PJ, Boutros, PC, Puente, XS, Gerhard, DS, Pfister, SM, McPherson, JD, Hudson, TJ, Schlesner, M, Lichter, P, Eils, R, Jones, DTW, and Gut, IG
- Abstract
As whole-genome sequencing for cancer genome analysis becomes a clinical tool, a full understanding of the variables affecting sequencing analysis output is required. Here using tumour-normal sample pairs from two different types of cancer, chronic lymphocytic leukaemia and medulloblastoma, we conduct a benchmarking exercise within the context of the International Cancer Genome Consortium. We compare sequencing methods, analysis pipelines and validation methods. We show that using PCR-free methods and increasing sequencing depth to ∼ 100 × shows benefits, as long as the tumour:control coverage ratio remains balanced. We observe widely varying mutation call rates and low concordance among analysis pipelines, reflecting the artefact-prone nature of the raw data and lack of standards for dealing with the artefacts. However, we show that, using the benchmark mutation set we have created, many issues are in fact easy to remedy and have an immediate positive impact on mutation detection accuracy.
- Published
- 2015
27. Towards the Systematic Mapping and Engineering of the Protein Prenylation Machinery in Saccharomyces cerevisiae
- Author
-
Uversky, VN, Stein, V, Kubala, MH, Steen, J, Grimmond, SM, Alexandrov, K, Uversky, VN, Stein, V, Kubala, MH, Steen, J, Grimmond, SM, and Alexandrov, K
- Abstract
Protein prenylation is a widespread and highly conserved eukaryotic post-translational modification that endows proteins with the ability to reversibly attach to intracellular membranes. The dynamic interaction of prenylated proteins with intracellular membranes is essential for their signalling functions and is frequently deregulated in disease processes such as cancer. As a result, protein prenylation has been pharmacologically targeted by numerous drug discovery programs, albeit with limited success. To a large extent, this can be attributed to an insufficient understanding of the interplay of different protein prenyltransferases and the combinatorial diversity of the prenylatable sequence space. Here, we report a high-throughput, growth-based genetic selection assay in Saccharomyces cerevisiae based on the Ras Recruitment System which, for the first time, has allowed us to create a comprehensive map of prenylatable protein sequences in S. cerevisiae. We demonstrate that potential prenylatable space is sparsely (6.2%) occupied leaving room for creation of synthetic orthogonal prenylatable sequences. To experimentally demonstrate that, we used the developed platform to engineer mutant farnesyltransferases that efficiently prenylate substrate motives that are not recognised by endogenous protein prenyltransferases. These uncoupled mutants can now be used as starting points for the systematic engineering of the eukaryotic protein prenylation machinery.
- Published
- 2015
28. Recommendations for Accurate Resolution of Gene and Isoform Allele-Specific Expression in RNA-Seq Data
- Author
-
Jordan, IK, Wood, DLA, Nones, K, Steptoe, A, Christ, A, Harliwong, I, Newell, F, Bruxner, TJC, Miller, D, Cloonan, N, Grimmond, SM, Jordan, IK, Wood, DLA, Nones, K, Steptoe, A, Christ, A, Harliwong, I, Newell, F, Bruxner, TJC, Miller, D, Cloonan, N, and Grimmond, SM
- Abstract
Genetic variation modulates gene expression transcriptionally or post-transcriptionally, and can profoundly alter an individual's phenotype. Measuring allelic differential expression at heterozygous loci within an individual, a phenomenon called allele-specific expression (ASE), can assist in identifying such factors. Massively parallel DNA and RNA sequencing and advances in bioinformatic methodologies provide an outstanding opportunity to measure ASE genome-wide. In this study, matched DNA and RNA sequencing, genotyping arrays and computationally phased haplotypes were integrated to comprehensively and conservatively quantify ASE in a single human brain and liver tissue sample. We describe a methodological evaluation and assessment of common bioinformatic steps for ASE quantification, and recommend a robust approach to accurately measure SNP, gene and isoform ASE through the use of personalized haplotype genome alignment, strict alignment quality control and intragenic SNP aggregation. Our results indicate that accurate ASE quantification requires careful bioinformatic analyses and is adversely affected by sample specific alignment confounders and random sampling even at moderate sequence depths. We identified multiple known and several novel ASE genes in liver, including WDR72, DSP and UBD, as well as genes that contained ASE SNPs with imbalance direction discordant with haplotype phase, explainable by annotated transcript structure, suggesting isoform derived ASE. The methods evaluated in this study will be of use to researchers performing highly conservative quantification of ASE, and the genes and isoforms identified as ASE of interest to researchers studying those loci.
- Published
- 2015
29. A biphasic epigenetic switch controls immunoevasion, virulence and niche adaptation in non-typeable Haemophilus influenzae
- Author
-
Atack, JM, Srikhanta, YN, Fox, KL, Jurcisek, JA, Brockman, KL, Clark, TA, Boitano, M, Power, PM, Jen, FE-C, McEwan, AG, Grimmond, SM, Smith, AL, Barenkamp, SJ, Korlach, J, Bakaletz, LO, Jennings, MP, Atack, JM, Srikhanta, YN, Fox, KL, Jurcisek, JA, Brockman, KL, Clark, TA, Boitano, M, Power, PM, Jen, FE-C, McEwan, AG, Grimmond, SM, Smith, AL, Barenkamp, SJ, Korlach, J, Bakaletz, LO, and Jennings, MP
- Abstract
Non-typeable Haemophilus influenzae contains an N(6)-adenine DNA-methyltransferase (ModA) that is subject to phase-variable expression (random ON/OFF switching). Five modA alleles, modA2, modA4, modA5, modA9 and modA10, account for over two-thirds of clinical otitis media isolates surveyed. Here, we use single molecule, real-time (SMRT) methylome analysis to identify the DNA-recognition motifs for all five of these modA alleles. Phase variation of these alleles regulates multiple proteins including vaccine candidates, and key virulence phenotypes such as antibiotic resistance (modA2, modA5, modA10), biofilm formation (modA2) and immunoevasion (modA4). Analyses of a modA2 strain in the chinchilla model of otitis media show a clear selection for ON switching of modA2 in the middle ear. Our results indicate that a biphasic epigenetic switch can control bacterial virulence, immunoevasion and niche adaptation in an animal model system.
- Published
- 2015
30. Using the MCF10A/MCF10CA1a Breast Cancer Progression Cell Line Model to Investigate the Effect of Active, Mutant Forms of EGFR in Breast Cancer Development and Treatment Using Gefitinib
- Author
-
Lee, JW, Bessette, DC, Tilch, E, Seidens, T, Quinn, MCJ, Wiegmans, AP, Shi, W, Cocciardi, S, McCart-Reed, A, Saunus, JM, Simpson, PT, Grimmond, SM, Lakhani, SR, Khanna, KK, Waddell, N, Al-Ejeh, F, Chenevix-Trench, G, Lee, JW, Bessette, DC, Tilch, E, Seidens, T, Quinn, MCJ, Wiegmans, AP, Shi, W, Cocciardi, S, McCart-Reed, A, Saunus, JM, Simpson, PT, Grimmond, SM, Lakhani, SR, Khanna, KK, Waddell, N, Al-Ejeh, F, and Chenevix-Trench, G
- Abstract
BACKGROUND: Basal-like and triple negative breast cancer (TNBC) share common molecular features, poor prognosis and a propensity for metastasis to the brain. Amplification of epidermal growth factor receptor (EGFR) occurs in ~50% of basal-like breast cancer, and mutations in the epidermal growth factor receptor (EGFR) have been reported in up to ~ 10% of Asian TNBC patients. In non-small cell lung cancer several different mutations in the EGFR tyrosine kinase domain confer sensitivity to receptor tyrosine kinase inhibitors, but the tumourigenic potential of EGFR mutations in breast cells and their potential for targeted therapy is unknown. MATERIALS AND METHODS: Constructs containing wild type, G719S or E746-A750 deletion mutant forms of EGFR were transfected into the MCF10A breast cells and their tumorigenic derivative, MCF10CA1a. The effects of EGFR over-expression and mutation on proliferation, migration, invasion, response to gefitinib, and tumour formation in vivo was investigated. Copy number analysis and whole exome sequencing of the MCF10A and MCF10CA1a cell lines were also performed. RESULTS: Mutant EGFR increased MCF10A and MCF10CA1a proliferation and MCF10A gefitinib sensitivity. The EGFR-E746-A750 deletion increased MCF10CA1a cell migration and invasion, and greatly increased MCF10CA1a xenograft tumour formation and growth. Compared to MCF10A cells, MCF10CA1a cells exhibited large regions of gain on chromosomes 3 and 9, deletion on chromosome 7, and mutations in many genes implicated in cancer. CONCLUSIONS: Mutant EGFR enhances the oncogenic properties of MCF10A cell line, and increases sensitivity to gefitinib. Although the addition of EGFR E746-A750 renders the MCF10CA1a cells more tumourigenic in vivo it is not accompanied by increased gefitinib sensitivity, perhaps due to additional mutations, including the PIK3CA H1047R mutation, that the MCF10CA1a cell line has acquired. Screening TNBC/basal-like breast cancer for EGFR mutations may prove useful fo
- Published
- 2015
31. The transcriptional network that controls growth arrest and differentiation in a human myeloid leukemia cell line
- Author
-
FANTOM Consortium, Suzuki H, Forrest AR, van Nimwegen E, Daub CO, Balwierz PJ, Irvine KM, Lassmann T, Ravasi T, Hasegawa Y, de Hoon MJ, Katayama S, Schroder K, Carninci P, Tomaru Y, Kanamori-Katayama M, Kubosaki A, Akalin A, Ando Y, Arner E, Asada M, Asahara H, Bailey T, Bajic VB, Bauer D, Beckhouse AG, Bertin N, Bjxf6rkegren J, Brombacher F, Bulger E, Chalk AM, Chiba J, Cloonan N, Dawe A, Dostie J, Engstrxf6m PG, Essack M, Faulkner GJ, Fink JL, Fredman D, Fujimori K, Furuno M, Gojobori T, Gough J, and Grimmond SM
- Published
- 2009
32. An epigenomic roadmap to induced pluripotency reveals DNA methylation as a reprogramming modulator
- Author
-
Lee, D-S, Shin, J-Y, Tonge, PD, Puri, MC, Lee, S, Park, H, Lee, W-C, Hussein, SMI, Bleazard, T, Yun, J-Y, Kim, J, Li, M, Cloonan, N, Wood, D, Clancy, JL, Mosbergen, R, Yi, J-H, Yang, K-S, Kim, H, Rhee, H, Wells, CA, Preiss, T, Grimmond, SM, Rogers, IM, Nagy, A, Seo, J-S, Lee, D-S, Shin, J-Y, Tonge, PD, Puri, MC, Lee, S, Park, H, Lee, W-C, Hussein, SMI, Bleazard, T, Yun, J-Y, Kim, J, Li, M, Cloonan, N, Wood, D, Clancy, JL, Mosbergen, R, Yi, J-H, Yang, K-S, Kim, H, Rhee, H, Wells, CA, Preiss, T, Grimmond, SM, Rogers, IM, Nagy, A, and Seo, J-S
- Abstract
Reprogramming of somatic cells to induced pluripotent stem cells involves a dynamic rearrangement of the epigenetic landscape. To characterize this epigenomic roadmap, we have performed MethylC-seq, ChIP-seq (H3K4/K27/K36me3) and RNA-Seq on samples taken at several time points during murine secondary reprogramming as part of Project Grandiose. We find that DNA methylation gain during reprogramming occurs gradually, while loss is achieved only at the ESC-like state. Binding sites of activated factors exhibit focal demethylation during reprogramming, while ESC-like pluripotent cells are distinguished by extension of demethylation to the wider neighbourhood. We observed that genes with CpG-rich promoters demonstrate stable low methylation and strong engagement of histone marks, whereas genes with CpG-poor promoters are safeguarded by methylation. Such DNA methylation-driven control is the key to the regulation of ESC-pluripotency genes, including Dppa4, Dppa5a and Esrrb. These results reveal the crucial role that DNA methylation plays as an epigenetic switch driving somatic cells to pluripotency.
- Published
- 2014
33. Rapid Identification of a Novel Complex I MT-ND3 m.10134C>A Mutation in a Leigh Syndrome Patient
- Author
-
Whitworth, AJ, Miller, DK, Menezes, MJ, Simons, C, Riley, LG, Cooper, ST, Grimmond, SM, Thorburn, DR, Christodoulou, J, Taft, RJ, Whitworth, AJ, Miller, DK, Menezes, MJ, Simons, C, Riley, LG, Cooper, ST, Grimmond, SM, Thorburn, DR, Christodoulou, J, and Taft, RJ
- Abstract
Leigh syndrome (LS) is a rare progressive multi-system neurodegenerative disorder, the genetics of which is frequently difficult to resolve. Rapid determination of the genetic etiology of LS in a 5-year-old girl facilitated inclusion in Edison Pharmaceutical's phase 2B clinical trial of EPI-743. SNP-arrays and high-coverage whole exome sequencing were performed on the proband, both parents and three unaffected siblings. Subsequent multi-tissue targeted high-depth mitochondrial sequencing was performed using custom long-range PCR amplicons. Tissue-specific mutant load was also assessed by qPCR. Complex I was interrogated by spectrophotometric enzyme assays and Western Blot. No putatively causal mutations were identified in nuclear-encoded genes. Analysis of low-coverage off-target mitochondrial reads revealed a previously unreported mitochondrial mutation in the proband in MT-ND3 (m.10134C>A, p.Q26K), a Complex I mitochondrial gene previously associated with LS. Targeted investigations demonstrated that this mutation was 1% heteroplasmic in the mother's blood and homoplasmic in the proband's blood, fibroblasts, liver and muscle. Enzyme assays revealed decreased Complex I activity. The identification of this novel LS MT-ND3 variant, the genomics of which was accomplished in less than 3.5 weeks, indicates that rapid genomic approaches may prove useful in time-sensitive cases with an unresolved genetic diagnosis.
- Published
- 2014
34. Targeting mTOR dependency in pancreatic cancer
- Author
-
Morran, DC, Wu, J, Jamieson, NB, Mrowinska, A, Kalna, G, Karim, SA, Au, AYM, Scarlett, CJ, Chang, DK, Pajak, MZ, Oien, KA, McKay, CJ, Carter, CR, Gillen, G, Champion, S, Pimlott, SL, Anderson, KI, Evans, TRJ, Grimmond, SM, Biankin, AV, Sansom, OJ, Morton, JP, Morran, DC, Wu, J, Jamieson, NB, Mrowinska, A, Kalna, G, Karim, SA, Au, AYM, Scarlett, CJ, Chang, DK, Pajak, MZ, Oien, KA, McKay, CJ, Carter, CR, Gillen, G, Champion, S, Pimlott, SL, Anderson, KI, Evans, TRJ, Grimmond, SM, Biankin, AV, Sansom, OJ, and Morton, JP
- Abstract
OBJECTIVE: Pancreatic cancer is a leading cause of cancer-related death in the Western world. Current chemotherapy regimens have modest survival benefit. Thus, novel, effective therapies are required for treatment of this disease. DESIGN: Activating KRAS mutation almost always drives pancreatic tumour initiation, however, deregulation of other potentially druggable pathways promotes tumour progression. PTEN loss leads to acceleration of Kras(G12D)-driven pancreatic ductal adenocarcinoma (PDAC) in mice and these tumours have high levels of mammalian target of rapamycin (mTOR) signalling. To test whether these KRAS PTEN pancreatic tumours show mTOR dependence, we compared response to mTOR inhibition in this model, to the response in another established model of pancreatic cancer, KRAS P53. We also assessed whether there was a subset of pancreatic cancer patients who may respond to mTOR inhibition. RESULTS: We found that tumours in KRAS PTEN mice exhibit a remarkable dependence on mTOR signalling. In these tumours, mTOR inhibition leads to proliferative arrest and even tumour regression. Further, we could measure response using clinically applicable positron emission tomography imaging. Importantly, pancreatic tumours driven by activated KRAS and mutant p53 did not respond to treatment. In human tumours, approximately 20% of cases demonstrated low PTEN expression and a gene expression signature that overlaps with murine KRAS PTEN tumours. CONCLUSIONS: KRAS PTEN tumours are uniquely responsive to mTOR inhibition. Targeted anti-mTOR therapies may offer clinical benefit in subsets of human PDAC selected based on genotype, that are dependent on mTOR signalling. Thus, the genetic signatures of human tumours could be used to direct pancreatic cancer treatment in the future.
- Published
- 2014
35. Imperfect centered miRNA binding sites are common and can mediate repression of target mRNAs
- Author
-
Martin, HC, Wani, S, Steptoe, AL, Krishnan, K, Nones, K, Nourbakhsh, E, Vlassov, A, Grimmond, SM, Cloonan, N, Martin, HC, Wani, S, Steptoe, AL, Krishnan, K, Nones, K, Nourbakhsh, E, Vlassov, A, Grimmond, SM, and Cloonan, N
- Abstract
BACKGROUND: MicroRNAs (miRNAs) bind to mRNAs and target them for translational inhibition or transcriptional degradation. It is thought that most miRNA-mRNA interactions involve the seed region at the 5' end of the miRNA. The importance of seed sites is supported by experimental evidence, although there is growing interest in interactions mediated by the central region of the miRNA, termed centered sites. To investigate the prevalence of these interactions, we apply a biotin pull-down method to determine the direct targets of ten human miRNAs, including four isomiRs that share centered sites, but not seeds, with their canonical partner miRNAs. RESULTS: We confirm that miRNAs and their isomiRs can interact with hundreds of mRNAs, and that imperfect centered sites are common mediators of miRNA-mRNA interactions. We experimentally demonstrate that these sites can repress mRNA activity, typically through translational repression, and are enriched in regions of the transcriptome bound by AGO. Finally, we show that the identification of imperfect centered sites is unlikely to be an artifact of our protocol caused by the biotinylation of the miRNA. However, the fact that there was a slight bias against seed sites in our protocol may have inflated the apparent prevalence of centered site-mediated interactions. CONCLUSIONS: Our results suggest that centered site-mediated interactions are much more frequent than previously thought. This may explain the evolutionary conservation of the central region of miRNAs, and has significant implications for decoding miRNA-regulated genetic networks, and for predicting the functional effect of variants that do not alter protein sequence.
- Published
- 2014
36. Genomic catastrophes frequently arise in esophageal adenocarcinoma and drive tumorigenesis
- Author
-
Nones, K, Waddell, N, Wayte, N, Patch, A-M, Bailey, P, Newell, F, Holmes, O, Fink, JL, Quinn, MCJ, Tang, YH, Lampe, G, Quek, K, Loffler, KA, Manning, S, Idrisoglu, S, Miller, D, Xu, Q, Wilson, PJ, Bruxner, TJC, Christ, AN, Harliwong, I, Nourse, C, Nourbakhsh, E, Anderson, M, Kazakoff, S, Leonard, C, Wood, S, Simpson, PT, Reid, LE, Krause, L, Hussey, DJ, Watson, DI, Lord, RV, Nancarrow, D, Phillips, WA, Gotley, D, Smithers, BM, Whiteman, DC, Hayward, NK, Campbell, PJ, Pearson, JV, Grimmond, SM, Barbour, AP, Nones, K, Waddell, N, Wayte, N, Patch, A-M, Bailey, P, Newell, F, Holmes, O, Fink, JL, Quinn, MCJ, Tang, YH, Lampe, G, Quek, K, Loffler, KA, Manning, S, Idrisoglu, S, Miller, D, Xu, Q, Wilson, PJ, Bruxner, TJC, Christ, AN, Harliwong, I, Nourse, C, Nourbakhsh, E, Anderson, M, Kazakoff, S, Leonard, C, Wood, S, Simpson, PT, Reid, LE, Krause, L, Hussey, DJ, Watson, DI, Lord, RV, Nancarrow, D, Phillips, WA, Gotley, D, Smithers, BM, Whiteman, DC, Hayward, NK, Campbell, PJ, Pearson, JV, Grimmond, SM, and Barbour, AP
- Abstract
Oesophageal adenocarcinoma (EAC) incidence is rapidly increasing in Western countries. A better understanding of EAC underpins efforts to improve early detection and treatment outcomes. While large EAC exome sequencing efforts to date have found recurrent loss-of-function mutations, oncogenic driving events have been underrepresented. Here we use a combination of whole-genome sequencing (WGS) and single-nucleotide polymorphism-array profiling to show that genomic catastrophes are frequent in EAC, with almost a third (32%, n=40/123) undergoing chromothriptic events. WGS of 22 EAC cases show that catastrophes may lead to oncogene amplification through chromothripsis-derived double-minute chromosome formation (MYC and MDM2) or breakage-fusion-bridge (KRAS, MDM2 and RFC3). Telomere shortening is more prominent in EACs bearing localized complex rearrangements. Mutational signature analysis also confirms that extreme genomic instability in EAC can be driven by somatic BRCA2 mutations. These findings suggest that genomic catastrophes have a significant role in the malignant transformation of EAC.
- Published
- 2014
37. Returning individual research results for genome sequences of pancreatic cancer
- Author
-
Johns, AL, Miller, DK, Simpson, SH, Gill, AJ, Kassahn, KS, Humphris, JL, Samra, JS, Tucker, K, Andrews, L, Chang, DK, Waddell, N, Pajic, M, Pearson, JV, Grimmond, SM, Biankin, AV, Zeps, N, Johns, AL, Miller, DK, Simpson, SH, Gill, AJ, Kassahn, KS, Humphris, JL, Samra, JS, Tucker, K, Andrews, L, Chang, DK, Waddell, N, Pajic, M, Pearson, JV, Grimmond, SM, Biankin, AV, and Zeps, N
- Abstract
BACKGROUND: Disclosure of individual results to participants in genomic research is a complex and contentious issue. There are many existing commentaries and opinion pieces on the topic, but little empirical data concerning actual cases describing how individual results have been returned. Thus, the real life risks and benefits of disclosing individual research results to participants are rarely if ever presented as part of this debate. METHODS: The Australian Pancreatic Cancer Genome Initiative (APGI) is an Australian contribution to the International Cancer Genome Consortium (ICGC), that involves prospective sequencing of tumor and normal genomes of study participants with pancreatic cancer in Australia. We present three examples that illustrate different facets of how research results may arise, and how they may be returned to individuals within an ethically defensible and clinically practical framework. This framework includes the necessary elements identified by others including consent, determination of the significance of results and which to return, delineation of the responsibility for communication and the clinical pathway for managing the consequences of returning results. RESULTS: Of 285 recruited patients, we returned results to a total of 25 with no adverse events to date. These included four that were classified as medically actionable, nine as clinically significant and eight that were returned at the request of the treating clinician. Case studies presented depict instances where research results impacted on cancer susceptibility, current treatment and diagnosis, and illustrate key practical challenges of developing an effective framework. CONCLUSIONS: We suggest that return of individual results is both feasible and ethically defensible but only within the context of a robust framework that involves a close relationship between researchers and clinicians.
- Published
- 2014
38. SnapShot-Seq: A Method for Extracting Genome-Wide, In Vivo mRNA Dynamics from a Single Total RNA Sample
- Author
-
Zuo, Z, Gray, JM, Harmin, DA, Boswell, SA, Cloonan, N, Mullen, TE, Ling, JJ, Miller, N, Kuersten, S, Ma, Y-C, McCarroll, SA, Grimmond, SM, Springer, M, Zuo, Z, Gray, JM, Harmin, DA, Boswell, SA, Cloonan, N, Mullen, TE, Ling, JJ, Miller, N, Kuersten, S, Ma, Y-C, McCarroll, SA, Grimmond, SM, and Springer, M
- Abstract
mRNA synthesis, processing, and destruction involve a complex series of molecular steps that are incompletely understood. Because the RNA intermediates in each of these steps have finite lifetimes, extensive mechanistic and dynamical information is encoded in total cellular RNA. Here we report the development of SnapShot-Seq, a set of computational methods that allow the determination of in vivo rates of pre-mRNA synthesis, splicing, intron degradation, and mRNA decay from a single RNA-Seq snapshot of total cellular RNA. SnapShot-Seq can detect in vivo changes in the rates of specific steps of splicing, and it provides genome-wide estimates of pre-mRNA synthesis rates comparable to those obtained via labeling of newly synthesized RNA. We used SnapShot-Seq to investigate the origins of the intrinsic bimodality of metazoan gene expression levels, and our results suggest that this bimodality is partly due to spillover of transcriptional activation from highly expressed genes to their poorly expressed neighbors. SnapShot-Seq dramatically expands the information obtainable from a standard RNA-Seq experiment.
- Published
- 2014
39. Clinical and molecular characterization of HER2 amplified-pancreatic cancer
- Author
-
Chou, A ; https://orcid.org/0000-0002-8129-7170, Waddell, N, Cowley, MJ ; https://orcid.org/0000-0002-9519-5714, Gill, AJ, Chang, DK, Patch, AM, Nones, K, Wu, J, Pinese, M ; https://orcid.org/0000-0001-5078-6687, Johns, AL, Miller, DK, Kassahn, KS, Nagrial, AM, Wasan, H, Goldstein, D ; https://orcid.org/0000-0001-6142-3291, Toon, CW ; https://orcid.org/0000-0002-0750-918X, Chin, V ; https://orcid.org/0000-0002-4630-4451, Chantrill, L ; https://orcid.org/0000-0002-5790-0208, Humphris, J, Mead, RS, Rooman, I, Samra, JS, Pajic, M ; https://orcid.org/0000-0002-3871-3829, Musgrove, EA, Pearson, JV, Morey, AL, Grimmond, SM, Biankin, AV, Chou, A ; https://orcid.org/0000-0002-8129-7170, Waddell, N, Cowley, MJ ; https://orcid.org/0000-0002-9519-5714, Gill, AJ, Chang, DK, Patch, AM, Nones, K, Wu, J, Pinese, M ; https://orcid.org/0000-0001-5078-6687, Johns, AL, Miller, DK, Kassahn, KS, Nagrial, AM, Wasan, H, Goldstein, D ; https://orcid.org/0000-0001-6142-3291, Toon, CW ; https://orcid.org/0000-0002-0750-918X, Chin, V ; https://orcid.org/0000-0002-4630-4451, Chantrill, L ; https://orcid.org/0000-0002-5790-0208, Humphris, J, Mead, RS, Rooman, I, Samra, JS, Pajic, M ; https://orcid.org/0000-0002-3871-3829, Musgrove, EA, Pearson, JV, Morey, AL, Grimmond, SM, and Biankin, AV
- Abstract
Background: Pancreatic cancer is one of the most lethal and molecularly diverse malignancies. Repurposing of therapeutics that target specific molecular mechanisms in different disease types offers potential for rapid improvements in outcome. Although HER2 amplification occurs in pancreatic cancer, it is inadequately characterized to exploit the potential of anti-HER2 therapies. Methods: HER2 amplification was detected and further analyzed using multiple genomic sequencing approaches. Standardized reference laboratory assays defined HER2 amplification in a large cohort of patients (n = 469) with pancreatic ductal adenocarcinoma (PDAC). Results: An amplified inversion event (1 MB) was identified at the HER2 locus in a patient with PDAC. Using standardized laboratory assays, we established diagnostic criteria for HER2 amplification in PDAC, and observed a prevalence of 2%. Clinically, HER2- amplified PDAC was characterized by a lack of liver metastases, and a preponderance of lung and brain metastases. Excluding breast and gastric cancer, the incidence of HER2-amplified cancers in the USA is >22,000 per annum. Conclusions: HER2 amplification occurs in 2% of PDAC, and has distinct features with implications for clinical practice. The molecular heterogeneity of PDAC implies that even an incidence of 2% represents an attractive target for anti-HER2 therapies, as options for PDAC are limited. Recruiting patients based on HER2 amplification, rather than organ of origin, could make trials of anti-HER2 therapies feasible in less common cancer types. © 2013 Chou et al.; licensee BioMed Central Ltd.
- Published
- 2013
40. Signatures of mutational processes in human cancer
- Author
-
Alexandrov, LB, Nik-Zainal, Serena, Wedge, DC, Aparicio, SA, Behjati, S, Biankin, AV, Bignell, GR, Bolli, N, Borg, A, Børresen-Dale, A L, Boyault, S, Burkhardt, B, Butler, AP, Caldas, Carlos, Davies, HR, Desmedt, Christine, Eils, Roland, Eyfjörd, Jórunn Erla, Foekens, John A, Greaves, M, Hosoda, F, Hutter, B, Ilicic, T, Imbeaud, S, Imielinski, M, Jäger, N, Jones, DT, Jones, Wendell, Knappskog, Stian, Kool, M, Lakhani, Sunil R, López-Otín, C, Martin, S, Munshi, NC, Nakamura, H, Northcott, PA, Pajic, M, Papaemmanuil, Elli, Paradiso, A, Pearson, JV, Puente, XS, Raine, K, Ramakrishna, M, Richardson, Andrea L, Richter, J, Rosenstiel, P, Schlesner, M, Schumacher, TN, Span, Paul, Teague, JW, Totoki, Y, Tutt, AN, Valdés-Mas, R, Van Buuren, MM, Van't Veer, Laura, Vincent-Salomon, Anne, Waddell, N, Yates, LR, Zucman-Rossi, J, Futreal, P Andrew, McDermott, U, Lichter, P, Meyerson, M, Grimmond, SM, Siebert, R, Campo, E, Shibata, T, Pfister, SM, Campbell, Peter J, Stratton, Michael R, Alexandrov, LB, Nik-Zainal, Serena, Wedge, DC, Aparicio, SA, Behjati, S, Biankin, AV, Bignell, GR, Bolli, N, Borg, A, Børresen-Dale, A L, Boyault, S, Burkhardt, B, Butler, AP, Caldas, Carlos, Davies, HR, Desmedt, Christine, Eils, Roland, Eyfjörd, Jórunn Erla, Foekens, John A, Greaves, M, Hosoda, F, Hutter, B, Ilicic, T, Imbeaud, S, Imielinski, M, Jäger, N, Jones, DT, Jones, Wendell, Knappskog, Stian, Kool, M, Lakhani, Sunil R, López-Otín, C, Martin, S, Munshi, NC, Nakamura, H, Northcott, PA, Pajic, M, Papaemmanuil, Elli, Paradiso, A, Pearson, JV, Puente, XS, Raine, K, Ramakrishna, M, Richardson, Andrea L, Richter, J, Rosenstiel, P, Schlesner, M, Schumacher, TN, Span, Paul, Teague, JW, Totoki, Y, Tutt, AN, Valdés-Mas, R, Van Buuren, MM, Van't Veer, Laura, Vincent-Salomon, Anne, Waddell, N, Yates, LR, Zucman-Rossi, J, Futreal, P Andrew, McDermott, U, Lichter, P, Meyerson, M, Grimmond, SM, Siebert, R, Campo, E, Shibata, T, Pfister, SM, Campbell, Peter J, and Stratton, Michael R
- Abstract
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, SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2013
41. Novel cancer drivers: mining the kinome
- Author
-
Biankin, AV, Grimmond, SM, Biankin, AV, and Grimmond, SM
- Abstract
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.
- Published
- 2013
42. Clinical and molecular characterization of HER2 amplified-pancreatic cancer
- Author
-
Chou, A, Waddell, N, Cowley, MJ, Gill, AJ, Chang, DK, Patch, A-M, Nones, K, Wu, J, Pinese, M, Johns, AL, Miller, DK, Kassahn, KS, Nagrial, AM, Wasan, H, Goldstein, D, Toon, CW, Chin, V, Chantrill, L, Humphris, J, Mead, RS, Rooman, I, Samra, JS, Pajic, M, Musgrove, EA, Pearson, JV, Morey, AL, Grimmond, SM, Biankin, AV, Chou, A, Waddell, N, Cowley, MJ, Gill, AJ, Chang, DK, Patch, A-M, Nones, K, Wu, J, Pinese, M, Johns, AL, Miller, DK, Kassahn, KS, Nagrial, AM, Wasan, H, Goldstein, D, Toon, CW, Chin, V, Chantrill, L, Humphris, J, Mead, RS, Rooman, I, Samra, JS, Pajic, M, Musgrove, EA, Pearson, JV, Morey, AL, Grimmond, SM, and Biankin, AV
- Abstract
BACKGROUND: Pancreatic cancer is one of the most lethal and molecularly diverse malignancies. Repurposing of therapeutics that target specific molecular mechanisms in different disease types offers potential for rapid improvements in outcome. Although HER2 amplification occurs in pancreatic cancer, it is inadequately characterized to exploit the potential of anti-HER2 therapies. METHODS: HER2 amplification was detected and further analyzed using multiple genomic sequencing approaches. Standardized reference laboratory assays defined HER2 amplification in a large cohort of patients (n = 469) with pancreatic ductal adenocarcinoma (PDAC). RESULTS: An amplified inversion event (1 MB) was identified at the HER2 locus in a patient with PDAC. Using standardized laboratory assays, we established diagnostic criteria for HER2 amplification in PDAC, and observed a prevalence of 2%. Clinically, HER2- amplified PDAC was characterized by a lack of liver metastases, and a preponderance of lung and brain metastases. Excluding breast and gastric cancer, the incidence of HER2-amplified cancers in the USA is >22,000 per annum. CONCLUSIONS: HER2 amplification occurs in 2% of PDAC, and has distinct features with implications for clinical practice. The molecular heterogeneity of PDAC implies that even an incidence of 2% represents an attractive target for anti-HER2 therapies, as options for PDAC are limited. Recruiting patients based on HER2 amplification, rather than organ of origin, could make trials of anti-HER2 therapies feasible in less common cancer types.
- Published
- 2013
43. Somatic Point Mutation Calling in Low Cellularity Tumors
- Author
-
Jordan, IK, Kassahn, KS, Holmes, O, Nones, K, Patch, A-M, Miller, DK, Christ, AN, Harliwong, I, Bruxner, TJ, Xu, Q, Anderson, M, Wood, S, Leonard, C, Taylor, D, Newell, F, Song, S, Idrisoglu, S, Nourse, C, Nourbakhsh, E, Manning, S, Wani, S, Steptoe, A, Pajic, M, Cowley, MJ, Pinese, M, Chang, DK, Gill, AJ, Johns, AL, Wu, J, Wilson, PJ, Fink, L, Biankin, AV, Waddell, N, Grimmond, SM, Pearson, JV, Jordan, IK, Kassahn, KS, Holmes, O, Nones, K, Patch, A-M, Miller, DK, Christ, AN, Harliwong, I, Bruxner, TJ, Xu, Q, Anderson, M, Wood, S, Leonard, C, Taylor, D, Newell, F, Song, S, Idrisoglu, S, Nourse, C, Nourbakhsh, E, Manning, S, Wani, S, Steptoe, A, Pajic, M, Cowley, MJ, Pinese, M, Chang, DK, Gill, AJ, Johns, AL, Wu, J, Wilson, PJ, Fink, L, Biankin, AV, Waddell, N, Grimmond, SM, and Pearson, JV
- Abstract
Somatic mutation calling from next-generation sequencing data remains a challenge due to the difficulties of distinguishing true somatic events from artifacts arising from PCR, sequencing errors or mis-mapping. Tumor cellularity or purity, sub-clonality and copy number changes also confound the identification of true somatic events against a background of germline variants. We have developed a heuristic strategy and software (http://www.qcmg.org/bioinformatics/qsnp/) for somatic mutation calling in samples with low tumor content and we show the superior sensitivity and precision of our approach using a previously sequenced cell line, a series of tumor/normal admixtures, and 3,253 putative somatic SNVs verified on an orthogonal platform.
- Published
- 2013
44. Signatures of mutational processes in human cancer
- Author
-
Alexandrov, Ludmil B, Nik-Zainal, Serena, Wedge, DC, Aparicio, SA, Behjati, S, Biankin, AV, Bignell, GR, Bolli, N, Borg, A, Børresen-Dale, A L, Boyault, S, Burkhardt, B, Butler, AP, Caldas, Carlos, Davies, HR, Desmedt, Christine, Eils, Roland, Eyfjörd, Jórunn Erla, Foekens, John A, Greaves, M, Hosoda, F, Hutter, B, Ilicic, T, Imbeaud, S, Imielinski, M, Jäger, N, Jones, DT, Jones, Wendell, Knappskog, Stian, Kool, M, Lakhani, Sunil R, López-Otín, C, Martin, S, Munshi, NC, Nakamura, H, Northcott, PA, Pajic, M, Papaemmanuil, Elli, Paradiso, A, Pearson, JV, Puente, XS, Raine, K, Ramakrishna, M, Richardson, Andrea L, Richter, J, Rosenstiel, P, Schlesner, M, Schumacher, TN, Span, Paul, Teague, JW, Totoki, Y, Tutt, AN, Valdés-Mas, R, Van Buuren, MM, Van't Veer, Laura, Vincent-Salomon, Anne, Waddell, N, Yates, LR, Zucman-Rossi, J, Futreal, P Andrew, McDermott, U, Lichter, P, Meyerson, M, Grimmond, SM, Siebert, R, Campo, E, Shibata, T, Pfister, SM, Campbell, Peter J, Stratton, Michael R, Alexandrov, Ludmil B, Nik-Zainal, Serena, Wedge, DC, Aparicio, SA, Behjati, S, Biankin, AV, Bignell, GR, Bolli, N, Borg, A, Børresen-Dale, A L, Boyault, S, Burkhardt, B, Butler, AP, Caldas, Carlos, Davies, HR, Desmedt, Christine, Eils, Roland, Eyfjörd, Jórunn Erla, Foekens, John A, Greaves, M, Hosoda, F, Hutter, B, Ilicic, T, Imbeaud, S, Imielinski, M, Jäger, N, Jones, DT, Jones, Wendell, Knappskog, Stian, Kool, M, Lakhani, Sunil R, López-Otín, C, Martin, S, Munshi, NC, Nakamura, H, Northcott, PA, Pajic, M, Papaemmanuil, Elli, Paradiso, A, Pearson, JV, Puente, XS, Raine, K, Ramakrishna, M, Richardson, Andrea L, Richter, J, Rosenstiel, P, Schlesner, M, Schumacher, TN, Span, Paul, Teague, JW, Totoki, Y, Tutt, AN, Valdés-Mas, R, Van Buuren, MM, Van't Veer, Laura, Vincent-Salomon, Anne, Waddell, N, Yates, LR, Zucman-Rossi, J, Futreal, P Andrew, McDermott, U, Lichter, P, Meyerson, M, Grimmond, SM, Siebert, R, Campo, E, Shibata, T, Pfister, SM, Campbell, Peter J, and Stratton, Michael R
- Abstract
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, SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2013
45. Conservation and divergence in Toll-like receptor 4-regulated gene expression in primary human versus mouse macrophages
- Author
-
Schroder, K, Irvine, KM, Taylor, MS, Bokil, NJ, Le Cao, KA, Masterman, KA, Labzin, LI, Semple, CA, Kapetanovic, R, Fairbairn, L, Akalin, A, Faulkner, GJ, Baillie, JK, Gongora, M, Daub, CO, Kawaji, H, McLachlan, GJ, Goldman, N, Grimmond, SM, Carninci, P, Suzuki, H, Hayashizaki, Y, Lenhard, B, Hume, DA, Sweet, MJ, Schroder, K, Irvine, KM, Taylor, MS, Bokil, NJ, Le Cao, KA, Masterman, KA, Labzin, LI, Semple, CA, Kapetanovic, R, Fairbairn, L, Akalin, A, Faulkner, GJ, Baillie, JK, Gongora, M, Daub, CO, Kawaji, H, McLachlan, GJ, Goldman, N, Grimmond, SM, Carninci, P, Suzuki, H, Hayashizaki, Y, Lenhard, B, Hume, DA, and Sweet, MJ
- Published
- 2012
46. RON is not a prognostic marker for resectable pancreatic cancer
- Author
-
Tactacan, CM, Chang, DK, Cowley, MJ, Humphrey, ES, Wu, J, Gill, AJ, Chou, A, Nones, K, Grimmond, SM, Sutherland, RL, Biankin, AV, Daly, RJ, Tactacan, CM, Chang, DK, Cowley, MJ, Humphrey, ES, Wu, J, Gill, AJ, Chou, A, Nones, K, Grimmond, SM, Sutherland, RL, Biankin, AV, and Daly, RJ
- Abstract
BACKGROUND: The receptor tyrosine kinase RON exhibits increased expression during pancreatic cancer progression and promotes migration, invasion and gemcitabine resistance of pancreatic cancer cells in experimental models. However, the prognostic significance of RON expression in pancreatic cancer is unknown. METHODS: RON expression was characterized in several large cohorts, including a prospective study, totaling 492 pancreatic cancer patients and relationships with patient outcome and clinico-pathologic variables were assessed. RESULTS: RON expression was associated with outcome in a training set, but this was not recapitulated in the validation set, nor was there any association with therapeutic responsiveness in the validation set or the prospective study. CONCLUSIONS: Although RON is implicated in pancreatic cancer progression in experimental models, and may constitute a therapeutic target, RON expression is not associated with prognosis or therapeutic responsiveness in resected pancreatic cancer.
- Published
- 2012
47. The deubiquitinase USP9X suppresses pancreatic ductal adenocarcinoma
- Author
-
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, 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, 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, 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, and Tuveson, DA
- Abstract
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.
- Published
- 2012
48. PINA v2.0: mining interactome modules
- Author
-
Cowley, MJ, Pinese, M, Kassahn, KS, Waddell, N, Pearson, JV, Grimmond, SM, Biankin, AV, Hautaniemi, S, Wu, J, Cowley, MJ, Pinese, M, Kassahn, KS, Waddell, N, Pearson, JV, Grimmond, SM, Biankin, AV, Hautaniemi, S, and Wu, J
- Abstract
The Protein Interaction Network Analysis (PINA) platform is a comprehensive web resource, which includes a database of unified protein-protein interaction data integrated from six manually curated public databases, and a set of built-in tools for network construction, filtering, analysis and visualization. The second version of PINA enhances its utility for studies of protein interactions at a network level, by including multiple collections of interaction modules identified by different clustering approaches from the whole network of protein interactions ('interactome') for six model organisms. All identified modules are fully annotated by enriched Gene Ontology terms, KEGG pathways, Pfam domains and the chemical and genetic perturbations collection from MSigDB. Moreover, a new tool is provided for module enrichment analysis in addition to simple query function. The interactome data are also available on the web site for further bioinformatics analysis. PINA is freely accessible at http://cbg.garvan.unsw.edu.au/pina/.
- Published
- 2012
49. Identification of Novel Markers of Mouse Fetal Ovary Development
- Author
-
Orban, L, Chen, H, Palmer, JS, Thiagarajan, RD, Dinger, ME, Lesieur, E, Chiu, H, Schulz, A, Spiller, C, Grimmond, SM, Little, MH, Koopman, P, Wilhelm, D, Orban, L, Chen, H, Palmer, JS, Thiagarajan, RD, Dinger, ME, Lesieur, E, Chiu, H, Schulz, A, Spiller, C, Grimmond, SM, Little, MH, Koopman, P, and Wilhelm, D
- Abstract
In contrast to the developing testis, molecular pathways driving fetal ovarian development have been difficult to characterise. To date no single master regulator of ovarian development has been identified that would be considered the female equivalent of Sry. Using a genomic approach we identified a number of novel protein-coding as well as non-coding genes that were detectable at higher levels in the ovary compared to testis during early mouse gonad development. We were able to cluster these ovarian genes into different temporal expression categories. Of note, Lrrc34 and AK015184 were detected in XX but not XY germ cells before the onset of sex-specific germ cell differentiation marked by entry into meiosis in an ovary and mitotic arrest in a testis. We also defined distinct spatial expression domains of somatic cell genes in the developing ovary. Our data expands the set of markers of early mouse ovary differentiation and identifies a classification of early ovarian genes, thus providing additional avenues with which to dissect this process.
- Published
- 2012
50. qpure: A Tool to Estimate Tumor Cellularity from Genome-Wide Single-Nucleotide Polymorphism Profiles
- Author
-
Ting, AH, Song, S, Nones, K, Miller, D, Harliwong, I, Kassahn, KS, Pinese, M, Pajic, M, Gill, AJ, Johns, AL, Anderson, M, Holmes, O, Leonard, C, Taylor, D, Wood, S, Xu, Q, Newell, F, Cowley, MJ, Wu, J, Wilson, P, Fink, L, Biankin, AV, Waddell, N, Grimmond, SM, Pearson, JV, Ting, AH, Song, S, Nones, K, Miller, D, Harliwong, I, Kassahn, KS, Pinese, M, Pajic, M, Gill, AJ, Johns, AL, Anderson, M, Holmes, O, Leonard, C, Taylor, D, Wood, S, Xu, Q, Newell, F, Cowley, MJ, Wu, J, Wilson, P, Fink, L, Biankin, AV, Waddell, N, Grimmond, SM, and Pearson, JV
- Abstract
Tumour cellularity, the relative proportion of tumour and normal cells in a sample, affects the sensitivity of mutation detection, copy number analysis, cancer gene expression and methylation profiling. Tumour cellularity is traditionally estimated by pathological review of sectioned specimens; however this method is both subjective and prone to error due to heterogeneity within lesions and cellularity differences between the sample viewed during pathological review and tissue used for research purposes. In this paper we describe a statistical model to estimate tumour cellularity from SNP array profiles of paired tumour and normal samples using shifts in SNP allele frequency at regions of loss of heterozygosity (LOH) in the tumour. We also provide qpure, a software implementation of the method. Our experiments showed that there is a medium correlation 0.42 ([Formula: see text]-value=0.0001) between tumor cellularity estimated by qpure and pathology review. Interestingly there is a high correlation 0.87 ([Formula: see text]-value [Formula: see text] 2.2e-16) between cellularity estimates by qpure and deep Ion Torrent sequencing of known somatic KRAS mutations; and a weaker correlation 0.32 ([Formula: see text]-value=0.004) between IonTorrent sequencing and pathology review. This suggests that qpure may be a more accurate predictor of tumour cellularity than pathology review. qpure can be downloaded from https://sourceforge.net/projects/qpure/.
- Published
- 2012
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.