90 results on '"Pleasance E"'
Search Results
2. Response to angiotensin blockade with irbesartan in a patient with metastatic colorectal cancer
- Author
-
Jones, M.R., Schrader, K.A., Shen, Y., Pleasance, E., Ch'ng, C., Dar, N., Yip, S., Renouf, D.J., Schein, J.E., Mungall, A.J., Zhao, Y., Moore, R., Ma, Y., Sheffield, B.S., Ng, T., Jones, S.J.M., Marra, M.A., Laskin, J., and Lim, H.J.
- Published
- 2016
- Full Text
- View/download PDF
3. Successful targeting of the NRG1 pathway indicates novel treatment strategy for metastatic cancer
- Author
-
Jones, M R, Lim, H, Shen, Y, Pleasance, E, Chʼng, C, Reisle, C, Leelakumari, S, Zhao, C, Yip, S, Ho, J, Zhong, E, Ng, T, Ionescu, D, Schaeffer, D F, Mungall, A J, Mungall, K L, Zhao, Y, Moore, R A, Ma, Y, Chia, S, Ho, C, Renouf, D J, Gelmon, K, Jones, S J M, Marra, M A, and Laskin, J
- Published
- 2017
- Full Text
- View/download PDF
4. ORegAnno: an open access database and curation system for literature-derived promoters, transcription factor binding sites and regulatory variation
- Author
-
Montgomery, S. B., Griffith, O. L., Sleumer, M. C., Bergman, C. M., Bilenky, M., Pleasance, E. D., Prychyna, Y., Zhang, X., and Jones, S. J. M.
- Published
- 2006
5. cisRED: a database system for genome-scale computational discovery of regulatory elements
- Author
-
Robertson, G., Bilenky, M., Lin, K., He, A., Yuen, W., Dagpinar, M., Varhol, R., Teague, K., Griffith, O. L., Zhang, X., Pan, Y., Hassel, M., Sleumer, M. C., Pan, W., Pleasance, E. D., Chuang, M., Hao, H., Li, Y. Y., Robertson, N., Fjell, C., Li, B., Montgomery, S. B., Astakhova, T., Zhou, J., Sander, J., Siddiqui, A. S., and Jones, S. J. M.
- Published
- 2006
6. Predictive markers of checkpoint inhibitor activity in adult metastatic solid tumours
- Author
-
Pender, A., primary, Titmuss, E., additional, Pleasance, E., additional, Fan, K., additional, Pearson, H., additional, Bonakdar, M., additional, Taylor, G., additional, Mungall, K., additional, Moore, R., additional, Lavoie, J.-M., additional, Yip, S., additional, Lim, H., additional, Renouf, D., additional, Jones, S.J., additional, Marra, M.A., additional, and Laskin, J.J., additional
- Published
- 2019
- Full Text
- View/download PDF
7. Personalized oncogenomics in the management of gastrointestinal carcinomas&mdash
- Author
-
Sheffield, B.S., Tessier-Cloutier, B., Li-Chang, H., Shen, Y., Pleasance, E., Kasaian, K., Li, Y., Jones, S.J.M., Lim, H.J., Renouf, D.J., Huntsman, D.G., Yip, S., Laskin, J., Marra, M., and Schaeffer, D.F.
- Subjects
Oncogenomics ,colonic adenocarcinoma ,appendiceal adenocarcinoma ,genomics ,personalized medicine ,bevacizumab ,cholangiocarcinoma ,targeted therapy ,digestive system diseases - Abstract
Gastrointestinal carcinomas are genomically complex cancers that are lethal in the metastatic setting. Whole-genome and transcriptome sequencing allow for the simultaneous characterization of multiple oncogenic pathways. We report 3 cases of metastatic gastrointestinal carcinoma in patients enrolled in the Personalized Onco-Genomics program at the BC Cancer Agency. Real-time genomic profiling was combined with clinical expertise to diagnose a carcinoma of unknown primary, to explore treatment response to bevacizumab in a colorectal cancer, and to characterize an appendiceal adenocarcinoma. In the first case, genomic profiling revealed an IDH1 somatic mutation, supporting the diagnosis of cholangiocarcinoma in a malignancy of unknown origin, and further guided therapy by identifying epidermal growth factor receptor amplification. In the second case, a BRAF V600E mutation and wild-type KRAS profile justified the use of targeted therapies to treat a colonic adenocarcinoma. The third case was an appendiceal adenocarcinoma defined by a p53 inactivation, Ras/raf/mek, Akt/mtor, Wnt, and notch pathway activation, and overexpression of ret, erbb2 (her2), erbb3, met, and cell cycle regulators. We show that whole-genome and transcriptome sequencing can be achieved within clinically effective timelines, yielding clinically useful and actionable information.
- Published
- 2016
- Full Text
- View/download PDF
8. FUS-NFATc2 sarcoma of bone, a novel molecular entity with aggressive behavior: Clinical and molecular pathology findings of two cases
- Author
-
Lacambra, M.D., primary, Loong, H., additional, To, K.F., additional, Feng, X., additional, Taylor, G., additional, Pleasance, E., additional, Laskin, J., additional, Marra, M., additional, Griffith, J., additional, Yeung, H.Y.M., additional, Wong, K.-C., additional, Chow, C., additional, Kumta, S., additional, Ng, W.H.A., additional, Tse, T., additional, Tong, C., additional, and Ng, T., additional
- Published
- 2018
- Full Text
- View/download PDF
9. 29 Unraveling molecular drivers of brain cancers at the clinical setting
- Author
-
Wong, D., primary, Shen, Y., additional, Levine, A. B., additional, Hoon Lee, T., additional, Pleasance, E., additional, Jones, M., additional, Thiessen, B., additional, Toyota, B., additional, Laskin, C.M, J., additional, Jones, S., additional, Marra, M. A, additional, and Yip, S., additional
- Published
- 2018
- Full Text
- View/download PDF
10. 1256P - Predictive markers of checkpoint inhibitor activity in adult metastatic solid tumours
- Author
-
Pender, A., Titmuss, E., Pleasance, E., Fan, K., Pearson, H., Bonakdar, M., Taylor, G., Mungall, K., Moore, R., Lavoie, J.-M., Yip, S., Lim, H., Renouf, D., Jones, S.J., Marra, M.A., and Laskin, J.J.
- Published
- 2019
- Full Text
- View/download PDF
11. 410P - FUS-NFATc2 sarcoma of bone, a novel molecular entity with aggressive behavior: Clinical and molecular pathology findings of two cases
- Author
-
Lacambra, M.D., Loong, H., To, K.F., Feng, X., Taylor, G., Pleasance, E., Laskin, J., Marra, M., Griffith, J., Yeung, H.Y.M., Wong, K.-C., Chow, C., Kumta, S., Ng, W.H.A., Tse, T., Tong, C., and Ng, T.
- Published
- 2018
- Full Text
- View/download PDF
12. 232 Management of germline findings revealed throughout the course of tumor-normal whole genome sequencing in oncology
- Author
-
Lim, H., primary, Schrader, K., additional, Young, S., additional, Fok, A., additional, Pleasance, E., additional, Jones, M., additional, Shen, Y., additional, Armstrong, L., additional, Virani, A., additional, Rassekh, S., additional, Deyell, R., additional, Yip, S., additional, Roscoe, R., additional, Karsan, A., additional, Marra, M., additional, and Laskin, J., additional
- Published
- 2015
- Full Text
- View/download PDF
13. 231 Whole genome analysis in a population-based cancer system: Results from sequencing >100 metastatic cancer patients
- Author
-
Lim, H., primary, Renouf, D., additional, Sun, S., additional, Ho, C., additional, Gelmon, K., additional, Chia, S., additional, Pleasance, E., additional, Jones, M., additional, Shen, Y., additional, Eirew, P., additional, Rassekh, S., additional, Deyell, R., additional, Yip, S., additional, Huntsman, D., additional, Roscoe, R., additional, Fok, A., additional, Ma, Y., additional, and Jones, S., additional
- Published
- 2015
- Full Text
- View/download PDF
14. Practical Guidelines for Ethical and Policy Issues that Arise from the Clinical Application of Whole Genome Sequencing in Cancer Patients
- Author
-
Lim, H., primary, Virani, A., additional, Fok, A., additional, Karsan, A., additional, Renouf, D., additional, Gelmon, K.A., additional, Yip, S., additional, Chia, S., additional, Sun, S., additional, Tinker, A., additional, Lee, S.J., additional, Rassekh, R., additional, Deyell, R., additional, Roscoe, R., additional, Jones, S., additional, Pleasance, E., additional, Marra, M., additional, and Laskin, J., additional
- Published
- 2014
- Full Text
- View/download PDF
15. Comprehensivemolecular characterization of clear cell renal cell carcinoma
- Author
-
Creighton, C., Morgan, M., Gunaratne, P., Wheeler, D., Gibbs, R., Robertson, A., Chu, A., Beroukhim, R., Cibulskis, K., Signoretti, S., Vandin, F., Wu, H., Raphael, B., Verhaak, R., Tamboli, P., Torres-Garcia, W., Akbani, R., Weinstein, J., Reuter, V., Hsieh, J., Brannon, A., Hakimi, A., Jacobsen, A., Ciriello, G., Reva, B., Ricketts, C., Linehan, W., Stuart, J., Rathmell, W., Shen, H., Laird, P., Muzny, D., Davis, C., Xi, L., Chang, K., Kakkar, N., Trevino, L., Benton, S., Reid, J., Morton, D., Doddapaneni, H., Han, Y., Lewis, L., Dinh, H., Kovar, C., Zhu, Y., Santibanez, J., Wang, M., Hale, W., Kalra, D., Getz, G., Lawrence, M., Sougnez, C., Carter, S., Sivachenko, A., Lichtenstein, L., Stewart, C., Voet, D., Fisher, S., Gabriel, S., Lander, E., Schumacher, S., Tabak, B., Saksena, G., Onofrio, R., Cherniack, A., Gentry, J., Ardlie, K., Meyerson, M., Chun, H., Mungall, A., Sipahimalani, P., Stoll, D., Ally, A., Balasundaram, M., Butterfield, Y., Carlsen, R., Carter, C., Chuah, E., Coope, R., Dhalla, N., Gorski, S., Guin, R., Hirst, C., Hirst, M., Holt, R., Lebovitz, C., Lee, D., Li, H., Mayo, M., Moore, R., Pleasance, E., Plettner, P., Schein, J., Shafiei, A., Slobodan, J., Tam, A., Thiessen, N., Varhol, Richard, Wye, N., Zhao, Y., Birol, I., Jones, S., Marra, M., Auman, J., Tan, D., Jones, C., Hoadley, K., Mieczkowski, P., Mose, L., Jefferys, S., Topal, M., Liquori, C., Turman, Y., Shi, Y., Waring, S., Buda, E., Walsh, J., Wu, J., Bodenheimer, T., Hoyle, A., Simons, J., Soloway, M., Balu, S., Parker, J., Hayes, D., Perou, C., Kucherlapati, R., Park, P., Triche, T., Weisenberger, D., Lai, P., Bootwalla, M., Maglinte, D., Mahurkar, S., Berman, B., Van den Berg, D., Cope, L., Baylin, S., Noble, M., DiCara, D., Zhang, H., Cho, J., Heiman, D., Gehlenborg, N., Mallard, W., Lin, P., Frazer, S., Stojanov, P., Liu, Y., Zhou, L., Kim, J., Chin, L., Benz, C., Yau, C., Reynolds, S., Shmulevich, I., Vegesna, R., Kim, H., Zhang, W., Cogdell, D., Jonasch, E., Ding, Z., Lu, Y., Zhang, N., Unruh, A., Casasent, T., Wakefield, C., Tsavachidou, D., Mills, G., Schultz, N., Antipin, Y., Gao, J., Cerami, E., Gross, B., Aksoy, B., Sinha, R., Weinhold, N., Sumer, S., Taylor, B., Shen, R., Ostrovnaya, I., Berger, M., Ladanyi, M., Sander, C., Fei, S., Stout, A., Spellman, P., Rubin, D., Liu, T., Sam, N., Paull, E., Carlin, D., Goldstein, T., Waltman, P., Ellrott, K., Zhu, J., Haussler, D., Xiao, W., Shelton, C., Gardner, J., Penny, R., Sherman, M., Mallery, D., Morris, S., Paulauskis, J., Burnett, K., Shelton, T., Kaelin, W., Choueiri, T., Atkins, M., Curley, E., Tickoo, S., Thorne, L., Boice, L., Huang, M., Fisher, J., Vocke, C., Peterson, J., Worrell, R., Merino, M., Schmidt, L., Czerniak, B., Aldape, K., Wood, C., Boyd, J., Weaver, J., Iacocca, M., Petrelli, N., Witkin, G., Brown, J., Czerwinski, C., Huelsenbeck-Dill, L., Rabeno, B., Myers, J., Morrison, C., Bergsten, J., Eckman, J., Harr, J., Smith, C., Tucker, K., Zach, L., Bshara, W., Gaudioso, C., Dhir, R., Maranchie, J., Nelson, J., Parwani, A., Potapova, O., Fedosenko, K., Cheville, J., Thompson, R., Mosquera, J., Rubin, M., Blute, M., Pihl, T., Jensen, M., Sfeir, R., Kahn, A., Kothiyal, P., Snyder, E., Pontius, J., Ayala, B., Backus, M., Walton, J., Baboud, J., Berton, D., Nicholls, M., Srinivasan, D., Raman, R., Girshik, S., Kigonya, P., Alonso, S., Sanbhadti, R., Barletta, S., Pot, D., Sheth, M., Demchok, J., Davidsen, T., Wang, Z., Yang, L., Tarnuzzer, R., Zhang, J., Eley, G., Ferguson, M., Shaw, K., Guyer, M., Ozenberger, B., Sofia, H., Creighton, C., Morgan, M., Gunaratne, P., Wheeler, D., Gibbs, R., Robertson, A., Chu, A., Beroukhim, R., Cibulskis, K., Signoretti, S., Vandin, F., Wu, H., Raphael, B., Verhaak, R., Tamboli, P., Torres-Garcia, W., Akbani, R., Weinstein, J., Reuter, V., Hsieh, J., Brannon, A., Hakimi, A., Jacobsen, A., Ciriello, G., Reva, B., Ricketts, C., Linehan, W., Stuart, J., Rathmell, W., Shen, H., Laird, P., Muzny, D., Davis, C., Xi, L., Chang, K., Kakkar, N., Trevino, L., Benton, S., Reid, J., Morton, D., Doddapaneni, H., Han, Y., Lewis, L., Dinh, H., Kovar, C., Zhu, Y., Santibanez, J., Wang, M., Hale, W., Kalra, D., Getz, G., Lawrence, M., Sougnez, C., Carter, S., Sivachenko, A., Lichtenstein, L., Stewart, C., Voet, D., Fisher, S., Gabriel, S., Lander, E., Schumacher, S., Tabak, B., Saksena, G., Onofrio, R., Cherniack, A., Gentry, J., Ardlie, K., Meyerson, M., Chun, H., Mungall, A., Sipahimalani, P., Stoll, D., Ally, A., Balasundaram, M., Butterfield, Y., Carlsen, R., Carter, C., Chuah, E., Coope, R., Dhalla, N., Gorski, S., Guin, R., Hirst, C., Hirst, M., Holt, R., Lebovitz, C., Lee, D., Li, H., Mayo, M., Moore, R., Pleasance, E., Plettner, P., Schein, J., Shafiei, A., Slobodan, J., Tam, A., Thiessen, N., Varhol, Richard, Wye, N., Zhao, Y., Birol, I., Jones, S., Marra, M., Auman, J., Tan, D., Jones, C., Hoadley, K., Mieczkowski, P., Mose, L., Jefferys, S., Topal, M., Liquori, C., Turman, Y., Shi, Y., Waring, S., Buda, E., Walsh, J., Wu, J., Bodenheimer, T., Hoyle, A., Simons, J., Soloway, M., Balu, S., Parker, J., Hayes, D., Perou, C., Kucherlapati, R., Park, P., Triche, T., Weisenberger, D., Lai, P., Bootwalla, M., Maglinte, D., Mahurkar, S., Berman, B., Van den Berg, D., Cope, L., Baylin, S., Noble, M., DiCara, D., Zhang, H., Cho, J., Heiman, D., Gehlenborg, N., Mallard, W., Lin, P., Frazer, S., Stojanov, P., Liu, Y., Zhou, L., Kim, J., Chin, L., Benz, C., Yau, C., Reynolds, S., Shmulevich, I., Vegesna, R., Kim, H., Zhang, W., Cogdell, D., Jonasch, E., Ding, Z., Lu, Y., Zhang, N., Unruh, A., Casasent, T., Wakefield, C., Tsavachidou, D., Mills, G., Schultz, N., Antipin, Y., Gao, J., Cerami, E., Gross, B., Aksoy, B., Sinha, R., Weinhold, N., Sumer, S., Taylor, B., Shen, R., Ostrovnaya, I., Berger, M., Ladanyi, M., Sander, C., Fei, S., Stout, A., Spellman, P., Rubin, D., Liu, T., Sam, N., Paull, E., Carlin, D., Goldstein, T., Waltman, P., Ellrott, K., Zhu, J., Haussler, D., Xiao, W., Shelton, C., Gardner, J., Penny, R., Sherman, M., Mallery, D., Morris, S., Paulauskis, J., Burnett, K., Shelton, T., Kaelin, W., Choueiri, T., Atkins, M., Curley, E., Tickoo, S., Thorne, L., Boice, L., Huang, M., Fisher, J., Vocke, C., Peterson, J., Worrell, R., Merino, M., Schmidt, L., Czerniak, B., Aldape, K., Wood, C., Boyd, J., Weaver, J., Iacocca, M., Petrelli, N., Witkin, G., Brown, J., Czerwinski, C., Huelsenbeck-Dill, L., Rabeno, B., Myers, J., Morrison, C., Bergsten, J., Eckman, J., Harr, J., Smith, C., Tucker, K., Zach, L., Bshara, W., Gaudioso, C., Dhir, R., Maranchie, J., Nelson, J., Parwani, A., Potapova, O., Fedosenko, K., Cheville, J., Thompson, R., Mosquera, J., Rubin, M., Blute, M., Pihl, T., Jensen, M., Sfeir, R., Kahn, A., Kothiyal, P., Snyder, E., Pontius, J., Ayala, B., Backus, M., Walton, J., Baboud, J., Berton, D., Nicholls, M., Srinivasan, D., Raman, R., Girshik, S., Kigonya, P., Alonso, S., Sanbhadti, R., Barletta, S., Pot, D., Sheth, M., Demchok, J., Davidsen, T., Wang, Z., Yang, L., Tarnuzzer, R., Zhang, J., Eley, G., Ferguson, M., Shaw, K., Guyer, M., Ozenberger, B., and Sofia, H.
- Abstract
Genetic changes underlying clear cell renal cell carcinoma(ccRCC) include alterations in genes controlling cellularoxygen sensing (for example, VHL) and the maintenance of chromatin states (for example, PBRM1). We surveyed more than 400 tumours using different genomic platforms and identified 19 significantly mutated genes. The PI(3)K/AKT pathway was recurrently mutated, suggesting this pathway as a potential therapeutic target. Widespread DNA hypomethylation was associated with mutation of the H3K36 methyltransferase SETD2, and integrative analysis suggested that mutations involving the SWI/SNF chromatin remodelling complex (PBRM1, ARID1A, SMARCA4) could have far-reaching effects on other pathways. Aggressive cancers demonstrated evidence of a metabolic shift, involving downregulation of genes involved in the TCA cycle, decreasedAMPK and PTEN protein levels, upregulation of the pentose phosphate pathway and the glutamine transporter genes, increased acetyl-CoA carboxylase protein, and altered promoter methylation of miR-21 (also known as MIR21) and GRB10. Remodelling cellular metabolism thus constitutes a recurrent pattern in ccRCC that correlates with tumour stage and severity and offers new views on the opportunities for disease treatment. © 2013 Macmillan Publishers Limited. All rights reserved.
- Published
- 2013
16. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia
- Author
-
Ley, T., Miller, C., Ding, L., Raphael, B., Mungall, A., Robertson, G., Hoadley, K., Triche, T., Laird, P., Baty, J., Fulton, L., Fulton, R., Heath, S., Kalicki-Veizer, J., Kandoth, C., Klco, J., Koboldt, D., Kanchi, K., Kulkarni, S., Lamprecht, T., Larson, D., Lin, G., Lu, C., McLellan, M., McMichael, J., Payton, J., Schmidt, H., Spencer, D., Tomasson, M., Wallis, J., Wartman, L., Watson, M., Welch, J., Wendl, M., Ally, A., Balasundaram, M., Birol, I., Butterfield, Y., Chiu, R., Chu, A., Chuah, E., Chun, H., Corbett, R., Dhalla, N., Guin, R., He, A., Hirst, C., Hirst, M., Holt, R., Jones, S., Karsan, A., Lee, D., Li, H., Marra, M., Mayo, M., Moore, R., Mungall, K., Parker, J., Pleasance, E., Plettner, P., Schein, J., Stoll, D., Swanson, L., Tam, A., Thiessen, N., Varhol, Richard, Wye, N., Zhao, Y., Gabriel, S., Getz, G., Sougnez, C., Zou, L., Leiserson, M., Vandin, F., Wu, H., Applebaum, F., Baylin, S., Akbani, R., Broom, B., Chen, K., Motter, T., Nguyen, K., Weinstein, J., Zhang, N., Ferguson, M., Adams, C., Black, A., Bowen, J., Gastier-Foster, J., Grossman, T., Lichtenberg, T., Wise, L., Davidsen, T., Demchok, J., Mills Shaw, K., Sheth, M., Sofia, H., Yang, L., Downing, J., Eley, G., Ley, T., Miller, C., Ding, L., Raphael, B., Mungall, A., Robertson, G., Hoadley, K., Triche, T., Laird, P., Baty, J., Fulton, L., Fulton, R., Heath, S., Kalicki-Veizer, J., Kandoth, C., Klco, J., Koboldt, D., Kanchi, K., Kulkarni, S., Lamprecht, T., Larson, D., Lin, G., Lu, C., McLellan, M., McMichael, J., Payton, J., Schmidt, H., Spencer, D., Tomasson, M., Wallis, J., Wartman, L., Watson, M., Welch, J., Wendl, M., Ally, A., Balasundaram, M., Birol, I., Butterfield, Y., Chiu, R., Chu, A., Chuah, E., Chun, H., Corbett, R., Dhalla, N., Guin, R., He, A., Hirst, C., Hirst, M., Holt, R., Jones, S., Karsan, A., Lee, D., Li, H., Marra, M., Mayo, M., Moore, R., Mungall, K., Parker, J., Pleasance, E., Plettner, P., Schein, J., Stoll, D., Swanson, L., Tam, A., Thiessen, N., Varhol, Richard, Wye, N., Zhao, Y., Gabriel, S., Getz, G., Sougnez, C., Zou, L., Leiserson, M., Vandin, F., Wu, H., Applebaum, F., Baylin, S., Akbani, R., Broom, B., Chen, K., Motter, T., Nguyen, K., Weinstein, J., Zhang, N., Ferguson, M., Adams, C., Black, A., Bowen, J., Gastier-Foster, J., Grossman, T., Lichtenberg, T., Wise, L., Davidsen, T., Demchok, J., Mills Shaw, K., Sheth, M., Sofia, H., Yang, L., Downing, J., and Eley, G.
- Abstract
BACKGROUND: Many mutations that contribute to the pathogenesis of acute myeloid leukemia (AML) are undefined. The relationships between patterns of mutations and epigenetic phenotypes are not yet clear. METHODS: We analyzed the genomes of 200 clinically annotated adult cases of de novo AML, using either whole-genome sequencing (50 cases) or whole-exome sequencing (150 cases), along with RNA and microRNA sequencing and DNA-methylation analysis. RESULTS: AML genomes have fewer mutations than most other adult cancers, with an average of only 13 mutations found in genes. Of these, an average of 5 are in genes that are recurrently mutated in AML. A total of 23 genes were significantly mutated, and another 237 were mutated in two or more samples. Nearly all samples had at least 1 nonsynonymous mutation in one of nine categories of genes that are almost certainly relevant for pathogenesis, including transcription-factor fusions (18% of cases), the gene encoding nucleophosmin (NPM1) (27%), tumor-suppressor genes (16%), DNA-methylation-related genes (44%), signaling genes (59%), chromatin-modifying genes (30%), myeloid transcription-factor genes (22%), cohesin-complex genes (13%), and spliceosome-complex genes (14%). Patterns of cooperation and mutual exclusivity suggested strong biologic relationships among several of the genes and categories. CONCLUSIONS: We identified at least one potential driver mutation in nearly all AML samples and found that a complex interplay of genetic events contributes to AML pathogenesis in individual patients. The databases from this study are widely available to serve as a foundation for further investigations of AML pathogenesis, classification, and risk stratification. (Funded by the National Institutes of Health.) Copyright © 2013 Massachusetts Medical Society.
- Published
- 2013
17. Comprehensive molecular portraits of human breast tumours
- Author
-
Koboldt, D., Fulton, R., McLellan, M., Schmidt, H., Kalicki-Veizer, J., McMichael, J., Fulton, L., Dooling, D., Ding, L., Mardis, E., Wilson, R., Ally, A., Balasundaram, M., Butterfield, Y., Carlsen, R., Carter, C., Chu, A., Chuah, E., Chun, H., Coope, R., Dhalla, N., Guin, R., Hirst, C., Hirst, M., Holt, R., Lee, D., Li, H., Mayo, M., Moore, R., Mungall, A., Pleasance, E., Robertson, A., Schein, J., Shafiei, A., Sipahimalani, P., Slobodan, J., Stoll, D., Tam, A., Thiessen, N., Varhol, Richard, Wye, N., Zeng, T., Zhao, Y., Birol, I., Jones, S., Marra, M., Cherniack, A., Saksena, G., Onofrio, R., Pho, N., Carter, S., Schumacher, S., Tabak, B., Hernandez, B., Gentry, J., Nguyen, H., Crenshaw, A., Ardlie, K., Beroukhim, R., Winckler, W., Getz, G., Gabriel, S., Meyerson, M., Chin, L., Kucherlapati, R., Hoadley, K., Auman, J., Fan, C., Turman, Y., Shi, Y., Li, L., Topal, M., He, X., Chao, H., Prat, A., Silva, G., Iglesia, M., Koboldt, D., Fulton, R., McLellan, M., Schmidt, H., Kalicki-Veizer, J., McMichael, J., Fulton, L., Dooling, D., Ding, L., Mardis, E., Wilson, R., Ally, A., Balasundaram, M., Butterfield, Y., Carlsen, R., Carter, C., Chu, A., Chuah, E., Chun, H., Coope, R., Dhalla, N., Guin, R., Hirst, C., Hirst, M., Holt, R., Lee, D., Li, H., Mayo, M., Moore, R., Mungall, A., Pleasance, E., Robertson, A., Schein, J., Shafiei, A., Sipahimalani, P., Slobodan, J., Stoll, D., Tam, A., Thiessen, N., Varhol, Richard, Wye, N., Zeng, T., Zhao, Y., Birol, I., Jones, S., Marra, M., Cherniack, A., Saksena, G., Onofrio, R., Pho, N., Carter, S., Schumacher, S., Tabak, B., Hernandez, B., Gentry, J., Nguyen, H., Crenshaw, A., Ardlie, K., Beroukhim, R., Winckler, W., Getz, G., Gabriel, S., Meyerson, M., Chin, L., Kucherlapati, R., Hoadley, K., Auman, J., Fan, C., Turman, Y., Shi, Y., Li, L., Topal, M., He, X., Chao, H., Prat, A., Silva, G., and Iglesia, M.
- Abstract
We analysed primary breast cancers by genomic DNA copy number arrays, DNA methylation, exome sequencing, messenger RNA arrays, microRNA sequencing and reverse-phase protein arrays. Our ability to integrate information across platforms provided key insights into previously defined gene expression subtypes and demonstrated the existence of four main breast cancer classes when combining data from five platforms, each of which shows significant molecular heterogeneity. Somatic mutations in only three genes (TP53, PIK3CA and GATA3) occurred at.10% incidence across all breast cancers; however, there were numerous subtype-associated and novel gene mutations including the enrichment of specific mutations in GATA3, PIK3CA and MAP3K1 with the luminal A subtype. We identified two novel protein-expression-defined subgroups, possibly produced by stromal/microenvironmental elements, and integrated analyses identified specific signalling pathways dominant in each molecular subtype including a HER2/phosphorylated HER2/EGFR/phosphorylated EGFR signature within the HER2-enriched expression subtype. Comparison of basal-like breast tumours with high-grade serous ovarian tumours showed many molecular commonalities, indicating a related aetiology and similar therapeutic opportunities. The biological finding of the four main breast cancer subtypes caused by different subsets of genetic and epigenetic abnormalities raises the hypothesis that much of the clinically observable plasticity and heterogeneity occurs within, and not across, these major biological subtypes of breast cancer. © 2012 Macmillan Publishers Limited. All rights reserved.
- Published
- 2012
18. Comprehensive molecular characterization of human colon and rectal cancer
- Author
-
Muzny, D., Bainbridge, M., Chang, K., Dinh, H., Drummond, J., Fowler, G., Kovar, C., Lewis, L., Morgan, M., Newsham, I., Reid, J., Santibanez, J., Shinbrot, E., Trevino, L., Wu, Y., Wang, M., Gunaratne, P., Donehower, L., Creighton, C., Wheeler, D., Gibbs, R., Lawrence, M., Voet, D., Jing, R., Cibulskis, K., Sivachenko, A., Stojanov, P., McKenna, A., Lander, E., Gabriel, S., Ding, L., Fulton, R., Koboldt, D., Wylie, T., Walker, J., Dooling, D., Fulton, L., Delehaunty, K., Fronick, C., Demeter, R., Mardis, E., Wilson, R., Chu, A., Chun, H., Mungall, A., Pleasance, E., Gordon Robertson, A., Stoll, D., Balasundaram, M., Birol, I., Butterfield, Y., Chuah, E., Coope, R., Dhalla, N., Guin, R., Hirst, C., Hirst, M., Holt, R., Lee, D., Li, H., Mayo, M., Moore, R., Schein, J., Slobodan, J., Tam, A., Thiessen, N., Varhol, Richard, Zeng, T., Zhao, Y., Jones, S., Marra, M., Bass, A., Ramos, A., Saksena, G., Cherniack, A., Schumacher, S., Tabak, B., Carter, S., Pho, N., Nguyen, H., Onofrio, R., Crenshaw, A., Ardlie, K., Muzny, D., Bainbridge, M., Chang, K., Dinh, H., Drummond, J., Fowler, G., Kovar, C., Lewis, L., Morgan, M., Newsham, I., Reid, J., Santibanez, J., Shinbrot, E., Trevino, L., Wu, Y., Wang, M., Gunaratne, P., Donehower, L., Creighton, C., Wheeler, D., Gibbs, R., Lawrence, M., Voet, D., Jing, R., Cibulskis, K., Sivachenko, A., Stojanov, P., McKenna, A., Lander, E., Gabriel, S., Ding, L., Fulton, R., Koboldt, D., Wylie, T., Walker, J., Dooling, D., Fulton, L., Delehaunty, K., Fronick, C., Demeter, R., Mardis, E., Wilson, R., Chu, A., Chun, H., Mungall, A., Pleasance, E., Gordon Robertson, A., Stoll, D., Balasundaram, M., Birol, I., Butterfield, Y., Chuah, E., Coope, R., Dhalla, N., Guin, R., Hirst, C., Hirst, M., Holt, R., Lee, D., Li, H., Mayo, M., Moore, R., Schein, J., Slobodan, J., Tam, A., Thiessen, N., Varhol, Richard, Zeng, T., Zhao, Y., Jones, S., Marra, M., Bass, A., Ramos, A., Saksena, G., Cherniack, A., Schumacher, S., Tabak, B., Carter, S., Pho, N., Nguyen, H., Onofrio, R., Crenshaw, A., and Ardlie, K.
- Abstract
To characterize somatic alterations in colorectal carcinoma, we conducted a genome-scale analysis of 276 samples, analysing exome sequence, DNA copy number, promoter methylation and messenger RNA and microRNA expression. A subset of these samples (97) underwent low-depth-of-coverage whole-genome sequencing. In total, 16% of colorectal carcinomas were found to be hypermutated: three-quarters of these had the expected high microsatellite instability, usually with hypermethylation and MLH1 silencing, and one-quarter had somatic mismatch-repair gene and polymerase µ (POLE) mutations. Excluding the hypermutated cancers, colon and rectum cancers were found to have considerably similar patterns of genomic alteration. Twenty-four genes were significantly mutated, and in addition to the expected APC, TP53, SMAD4, PIK3CA and KRAS mutations, we found frequent mutations in ARID1A, SOX9 and FAM123B. Recurrent copy-number alterations include potentially drug-targetable amplifications of ERBB2 and newly discovered amplification of IGF2. Recurrent chromosomal translocations include the fusion of NAV2 and WNT pathway member TCF7L1. Integrative analyses suggest new markers for aggressive colorectal carcinoma and an important role for MYC-directed transcriptional activation and repression. © 2012 Macmillan Publishers Limited. All rights reserved.
- Published
- 2012
19. Comprehensive genomic characterization of squamous cell lung cancers
- Author
-
Hammerman, P., Voet, D., Lawrence, M., Jing, R., Cibulskis, K., Sivachenko, A., Stojanov, P., McKenna, A., Lander, E., Gabriel, S., Getz, G., Imielinski, M., Helman, E., Hernandez, B., Pho, N., Meyerson, M., Chu, A., Hye-Chun, J., Mungall, A., Pleasance, E., Robertson, A., Sipahimalani, P., Stoll, D., Balasundaram, M., Birol, I., Butterfield, Y., Chuah, E., Coope, R., Corbett, R., Dhalla, N., Guin, R., He, A., Hirst, C., Hirst, M., Holt, R., Lee, D., Li, H., Mayo, M., Moore, R., Mungall, K., Nip, K., Olshen, A., Schein, J., Slobodan, J., Tam, A., Thiessen, N., Varhol, Richard, Zeng, T., Zhao, Y., Jones, S., Marra, M., Saksena, G., Cherniack, A., Schumacher, S., Tabak, B., Carter, S., Nguyen, H., Onofrio, R., Crenshaw, A., Ardlie, K., Beroukhim, R., Winckler, W., Protopopov, A., Zhang, J., Hadjipanayis, A., Lee, S., Xi, R., Yang, L., Ren, X., Zhang, H., Shukla, S., Chen, P., Haseley, P., Lee, E., Chin, L., Hammerman, P., Voet, D., Lawrence, M., Jing, R., Cibulskis, K., Sivachenko, A., Stojanov, P., McKenna, A., Lander, E., Gabriel, S., Getz, G., Imielinski, M., Helman, E., Hernandez, B., Pho, N., Meyerson, M., Chu, A., Hye-Chun, J., Mungall, A., Pleasance, E., Robertson, A., Sipahimalani, P., Stoll, D., Balasundaram, M., Birol, I., Butterfield, Y., Chuah, E., Coope, R., Corbett, R., Dhalla, N., Guin, R., He, A., Hirst, C., Hirst, M., Holt, R., Lee, D., Li, H., Mayo, M., Moore, R., Mungall, K., Nip, K., Olshen, A., Schein, J., Slobodan, J., Tam, A., Thiessen, N., Varhol, Richard, Zeng, T., Zhao, Y., Jones, S., Marra, M., Saksena, G., Cherniack, A., Schumacher, S., Tabak, B., Carter, S., Nguyen, H., Onofrio, R., Crenshaw, A., Ardlie, K., Beroukhim, R., Winckler, W., Protopopov, A., Zhang, J., Hadjipanayis, A., Lee, S., Xi, R., Yang, L., Ren, X., Zhang, H., Shukla, S., Chen, P., Haseley, P., Lee, E., and Chin, L.
- Abstract
Lung squamous cell carcinoma is a common type of lung cancer, causing approximately 400,000 deaths per year worldwide. Genomic alterations in squamous cell lung cancers have not been comprehensively characterized, and no molecularly targeted agents have been specifically developed for its treatment. As part of The Cancer Genome Atlas, here we profile 178 lung squamous cell carcinomas to provide a comprehensive landscape of genomic and epigenomic alterations. We show that the tumour type is characterized by complex genomic alterations, with a mean of 360 exonic mutations, 165 genomic rearrangements, and 323 segments of copy number alteration per tumour. We find statistically recurrent mutations in 11 genes, including mutation of TP53 in nearly all specimens. Previously unreported loss-of-function mutations are seen in the HLA-A class I major histocompatibility gene. Significantly altered pathways included NFE2L2 and KEAP1 in 34%, squamous differentiation genes in 44%, phosphatidylinositol-3-OH kinase pathway genes in 47%, and CDKN2A and RB1 in 72% of tumours. We identified a potential therapeutic target in most tumours, offering new avenues of investigation for the treatment of squamous cell lung cancers. © 2012 Macmillan Publishers Limited. All rights reserved.
- Published
- 2012
20. A systematic, large-scale resequencing screen of X-chromosome coding exons in mental retardation.
- Author
-
Tarpey, P.S., Smith, R., Pleasance, E., Whibley, A., Edkins, S., Hardy, C., O'Meara, S., Latimer, C., Dicks, E., Menzies, A., Stephens, P., Blow, M., Greenman, C., Xue, Y., Tyler-Smith, C., Thompson, D., Gray, K., Andrews, J., Barthorpe, S., Buck, G., Cole, J., Dunmore, R., Jones, D., Maddison, M., Mironenko, T., Turner, R., Turrell, K., Varian, J., West, S., Widaa, S., Wray, P., Teague, J., Butler, A., Jenkinson, A., Jia, M., Richardson, D., Shepherd, R., Wooster, R., Tejada, M.I., Martinez, F., Carvill, G., Goliath, R., Brouwer, A.P.M. de, Bokhoven, H. van, Esch, H. van, Chelly, J., Raynaud, M., Ropers, H.H., Abidi, F.E., Srivastava, A.K., Cox, J., Luo, Y., Mallya, U., Moon, J., Parnau, J., Mohammed, S., Tolmie, J.L., Shoubridge, C., Corbett, M., Gardner, A., Haan, E., Rujirabanjerd, S., Shaw, M.A., Vandeleur, L., Fullston, T., Easton, D.F., Boyle, J., Partington, M., Hackett, A., Field, M., Skinner, C., Stevenson, R.E., Bobrow, M., Turner, G., Schwartz, C.E., Gecz, J., Raymond, F.L., Futreal, P.A., Stratton, M.R., Tarpey, P.S., Smith, R., Pleasance, E., Whibley, A., Edkins, S., Hardy, C., O'Meara, S., Latimer, C., Dicks, E., Menzies, A., Stephens, P., Blow, M., Greenman, C., Xue, Y., Tyler-Smith, C., Thompson, D., Gray, K., Andrews, J., Barthorpe, S., Buck, G., Cole, J., Dunmore, R., Jones, D., Maddison, M., Mironenko, T., Turner, R., Turrell, K., Varian, J., West, S., Widaa, S., Wray, P., Teague, J., Butler, A., Jenkinson, A., Jia, M., Richardson, D., Shepherd, R., Wooster, R., Tejada, M.I., Martinez, F., Carvill, G., Goliath, R., Brouwer, A.P.M. de, Bokhoven, H. van, Esch, H. van, Chelly, J., Raynaud, M., Ropers, H.H., Abidi, F.E., Srivastava, A.K., Cox, J., Luo, Y., Mallya, U., Moon, J., Parnau, J., Mohammed, S., Tolmie, J.L., Shoubridge, C., Corbett, M., Gardner, A., Haan, E., Rujirabanjerd, S., Shaw, M.A., Vandeleur, L., Fullston, T., Easton, D.F., Boyle, J., Partington, M., Hackett, A., Field, M., Skinner, C., Stevenson, R.E., Bobrow, M., Turner, G., Schwartz, C.E., Gecz, J., Raymond, F.L., Futreal, P.A., and Stratton, M.R.
- Abstract
Contains fulltext : 79687.pdf (publisher's version ) (Closed access), Large-scale systematic resequencing has been proposed as the key future strategy for the discovery of rare, disease-causing sequence variants across the spectrum of human complex disease. We have sequenced the coding exons of the X chromosome in 208 families with X-linked mental retardation (XLMR), the largest direct screen for constitutional disease-causing mutations thus far reported. The screen has discovered nine genes implicated in XLMR, including SYP, ZNF711 and CASK reported here, confirming the power of this strategy. The study has, however, also highlighted issues confronting whole-genome sequencing screens, including the observation that loss of function of 1% or more of X-chromosome genes is compatible with apparently normal existence.
- Published
- 2009
21. Serial analysis of gene expression in the southern cattle tick following acaricide treatment of larvae from organophosphate resistant and susceptible strains
- Author
-
Guerrero, F. D., primary, Bendele, K. G., additional, Chen, A. C., additional, Li, A. Y., additional, Miller, R. J., additional, Pleasance, E., additional, Varhol, R., additional, Rousseau, M.‐E., additional, and Nene, V. M., additional
- Published
- 2007
- Full Text
- View/download PDF
22. 1615P - Practical Guidelines for Ethical and Policy Issues that Arise from the Clinical Application of Whole Genome Sequencing in Cancer Patients
- Author
-
Lim, H., Virani, A., Fok, A., Karsan, A., Renouf, D., Gelmon, K.A., Yip, S., Chia, S., Sun, S., Tinker, A., Lee, S.J., Rassekh, R., Deyell, R., Roscoe, R., Jones, S., Pleasance, E., Marra, M., and Laskin, J.
- Published
- 2014
- Full Text
- View/download PDF
23. cis-Regulatory Element Prediction in Mammalian Genomes
- Author
-
Siddiqui, A., primary, Robertson, G., additional, Bilenky, M., additional, Astakhova, T., additional, Griffith, O.L., additional, Hassel, M., additional, Lin, K., additional, Montgomery, S., additional, Oveisi, M., additional, Pleasance, E., additional, Robertson, N., additional, Sleumer, M.C., additional, Teague, K., additional, Varhol, R., additional, Zhang, M., additional, and Jones, S., additional
- Full Text
- View/download PDF
24. NTRK2 Fusion Driven Pediatric Glioblastoma: Identification of key molecular drivers by personalized oncology
- Author
-
Levine, Shen, Y, Mungall, K, Serrano, J, Snuderl, M, Pleasance, E, Jones, SJM, Laskin, J, Marra, MA, Rassekh, R, Deyell, R, Yip, S, Cheng, S, and Dunham, C
- Abstract
We describe the case of an 11-month-old girl with a rare cerebellar glioblastoma driven by a NACC2-NTRK2(Nucleus Accumbens Associated Protein 2-Neurotrophic Receptor Tyrosine Kinase 2) fusion. Initial workup of our case demonstrated homozygous CDKN2Adeletion, but immunohistochemistry for other driver mutations, including IDH1 R132H, BRAF V600E, and H3F3A K27M were negative, and ATRX was retained. Tissue was subsequently submitted for personalized oncogenomic analysis, including whole genome and whole transcriptome sequencing, which demonstrated an activating NTRK2fusion, as well as high PD-L1 expression, which was subsequently confirmed by immunohistochemistry. Furthermore, H3and IDHdemonstrated wildtype status. These findings suggested the possibility of treatment with either NTRK- or immune checkpoint- inhibitors through active clinical trials. Ultimately, the family pursued standard treatment that involved Head Start III chemotherapy and proton radiotherapy. Notably, at most recent follow upapproximately two years from initial diagnosis, the patient is in disease remission and thriving, suggesting favorable biology despite histologic malignancy. This case illustrates the value of personalized oncogenomics, as the molecular profiling revealed two actionable changes that would not have been apparent through routine diagnostics. NTRKfusions are known oncogenic drivers in a range of cancer types, but this is the first report of a NACC2-NTRK2fusion in a glioblastoma.LEARNING OBJECTIVESThis presentation will enable the learner to:1.Explore the current molecular landscape of pediatric high grade gliomas2.Recognize the value of personalized oncogenomic analysis, particularly in rare and/or aggressive tumors3.Discuss the current status of NTRK inhibitor clinical trials
- Published
- 2019
- Full Text
- View/download PDF
25. cis-Regulatory element prediction in mammalian genomes.
- Author
-
Siddiqui, A., Robertson, G., Bilenky, M., Astakhova, T., Griffith, O.L., Hassel, M., Lin, K., Montgomery, S., Oveisi, M., Pleasance, E., Robertson, N., Sleumer, M.C., Teague, K., Varhol, R., Zhang, M., and Jones, S.
- Published
- 2005
- Full Text
- View/download PDF
26. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer
- Author
-
Lucy Stebbings, Syd Barthorpe, Sarah O’Meara, Michael R. Stratton, Richard J. Kahnoski, Lee Mulderrig, Bin Tean Teh, Ronald A. DePinho, Laura Mudie, Mark Maddison, Catherine Leroy, Giovanni Tonon, Philip J. Stephens, Jenny Andrews, David J. McBride, Yu-Tzu Tai, John Wong, Sok Kean Khoo, Meng-Lay Lin, Tatiana Mironenko, Aaron Massie, Claire Hardy, Rachel Turner, David T. Jones, Calli Latimer, Jennifer Cole, Sarah Edkins, Dave Beare, Sofie West, Peter J. Campbell, V. Peter Collins, Helen Davies, Sara Widaa, Graham R. Bignell, Mingming Jia, Patrick S. Tarpey, Gijs van Haaften, Jennifer Varian, Gurpreet Tang, Adam Butler, Chai Yin Kok, Simon Law, Gillian L. Dalgliesh, Raffaella Smith, Koichi Ichimura, Rebecca Shepherd, Jon W. Teague, Erin Pleasance, Kirsten McLay, Simon Maquire, Gemma Buck, Suet Yi Leung, Paul Wray, Andrew Menzies, Simon A. Forbes, Christopher Greenman, P. Andrew Futreal, Kelly Turrell, Jonathan Hinton, Lina Chen, Siu Tsan Yuen, Kenneth C. Anderson, van Haaften, G, Dalgliesh, Gl, Davies, H, Chen, L, Bignell, G, Greenman, C, Edkins, S, Hardy, C, O'Meara, S, Teague, J, Butler, A, Hinton, J, Latimer, C, Andrews, J, Barthorpe, S, Beare, D, Buck, G, Campbell, Pj, Cole, J, Forbes, S, Jia, M, Jones, D, Kok, Cy, Leroy, C, Lin, Ml, Mcbride, Dj, Maddison, M, Maquire, S, Mclay, K, Menzies, A, Mironenko, T, Mulderrig, L, Mudie, L, Pleasance, E, Shepherd, R, Smith, R, Stebbings, L, Stephens, P, Tang, G, Tarpey, P, Turner, R, Turrell, K, Varian, J, West, S, Widaa, S, Wray, P, Collins, Vp, Ichimura, K, Law, S, Wong, J, Yuen, St, Leung, Sy, Tonon, G, Depinho, Ra, Tai, Yt, Anderson, Kc, Kahnoski, Rj, Massie, A, Khoo, Sk, Teh, Bt, Stratton, Mr, and Futreal, Pa.
- Subjects
Jumonji Domain-Containing Histone Demethylases ,Methyltransferase ,medicine.disease_cause ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,Germline mutation ,Neoplasms ,Genetics ,medicine ,Humans ,Epigenetics ,030304 developmental biology ,0303 health sciences ,Mutation ,biology ,Oxidoreductases, N-Demethylating ,Methylation ,Histone ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Demethylase - Abstract
Somatically acquired epigenetic changes are present in many cancers. Epigenetic regulation is maintained via post-translational modifications of core histones. Here, we describe inactivating somatic mutations in the histone lysine demethylase gene UTX, pointing to histone H3 lysine methylation deregulation in multiple tumor types. UTX reintroduction into cancer cells with inactivating UTX mutations resulted in slowing of proliferation and marked transcriptional changes. These data identify UTX as a new human cancer gene.
- Published
- 2009
27. Long-read sequencing of an advanced cancer cohort resolves rearrangements, unravels haplotypes, and reveals methylation landscapes.
- Author
-
O'Neill K, Pleasance E, Fan J, Akbari V, Chang G, Dixon K, Csizmok V, MacLennan S, Porter V, Galbraith A, Grisdale CJ, Culibrk L, Dupuis JH, Corbett R, Hopkins J, Bowlby R, Pandoh P, Smailus DE, Cheng D, Wong T, Frey C, Shen Y, Lewis E, Paulin LF, Sedlazeck FJ, Nelson JMT, Chuah E, Mungall KL, Moore RA, Coope R, Mungall AJ, McConechy MK, Williamson LM, Schrader KA, Yip S, Marra MA, Laskin J, and Jones SJM
- Abstract
The Long-Read Personalized OncoGenomics (POG) dataset comprises a cohort of 189 patient tumors and 41 matched normal samples sequenced using the Oxford Nanopore Technologies PromethION platform. This dataset from the POG program and the Marathon of Hope Cancer Centres Network includes DNA and RNA short-read sequence data, analytics, and clinical information. We show the potential of long-read sequencing for resolving complex cancer-related structural variants, viral integrations, and extrachromosomal circular DNA. Long-range phasing facilitates the discovery of allelically differentially methylated regions (aDMRs) and allele-specific expression, including recurrent aDMRs in the cancer genes RET and CDKN2A. Germline promoter methylation in MLH1 can be directly observed in Lynch syndrome. Promoter methylation in BRCA1 and RAD51C is a likely driver behind homologous recombination deficiency where no coding driver mutation was found. This dataset demonstrates applications for long-read sequencing in precision medicine and is available as a resource for developing analytical approaches using this technology., Competing Interests: Declaration of interests The following authors disclose relevant potential competing interests: K.O.N., V.P., L.F.P., K.D., J.L., and S.J.M.J. received travel funding from Oxford Nanopore Technologies to present at conferences in 2022 and 2023., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
28. Whole genome and transcriptome integrated analyses guide clinical care of pediatric poor prognosis cancers.
- Author
-
Deyell RJ, Shen Y, Titmuss E, Dixon K, Williamson LM, Pleasance E, Nelson JMT, Abbasi S, Krzywinski M, Armstrong L, Bonakdar M, Ch'ng C, Chuah E, Dunham C, Fok A, Jones M, Lee AF, Ma Y, Moore RA, Mungall AJ, Mungall KL, Rogers PC, Schrader KA, Virani A, Wee K, Young SS, Zhao Y, Jones SJM, Laskin J, Marra MA, and Rassekh SR
- Subjects
- Humans, Child, Female, Adolescent, Male, Child, Preschool, Prognosis, Infant, Transcriptome, Young Adult, Whole Genome Sequencing, Germ-Line Mutation, Mutation, Genome, Human genetics, Genetic Predisposition to Disease, Neoplasms genetics, Neoplasms therapy, Gene Expression Profiling methods, DNA Copy Number Variations
- Abstract
The role for routine whole genome and transcriptome analysis (WGTA) for poor prognosis pediatric cancers remains undetermined. Here, we characterize somatic mutations, structural rearrangements, copy number variants, gene expression, immuno-profiles and germline cancer predisposition variants in children and adolescents with relapsed, refractory or poor prognosis malignancies who underwent somatic WGTA and matched germline sequencing. Seventy-nine participants with a median age at enrollment of 8.8 y (range 6 months to 21.2 y) are included. Germline pathogenic/likely pathogenic variants are identified in 12% of participants, of which 60% were not known prior. Therapeutically actionable variants are identified by targeted gene report and whole genome in 32% and 62% of participants, respectively, and increase to 96% after integrating transcriptome analyses. Thirty-two molecularly informed therapies are pursued in 28 participants with 54% achieving a clinical benefit rate; objective response or stable disease ≥6 months. Integrated WGTA identifies therapeutically actionable variants in almost all tumors and are directly translatable to clinical care of children with poor prognosis cancers., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
29. The benefit of a complete reference genome for cancer structural variant analysis.
- Author
-
Paulin LF, Fan J, O'Neill K, Pleasance E, Porter VL, Jones SJM, and Sedlazeck FJ
- Abstract
The complexities of cancer genomes are becoming more easily interpreted due to advancements in sequencing technologies and improved bioinformatic analysis. Structural variants (SVs) represent an important subset of somatic events in tumors. While detection of SVs has been markedly improved by the development of long-read sequencing, somatic variant identification and annotation remains challenging. We hypothesized that use of a completed human reference genome (CHM13-T2T) would improve somatic SV calling. Our findings in a tumour/normal matched benchmark sample and two patient samples show that the CHM13-T2T improves SV detection and prioritization accuracy compared to GRCh38, with a notable reduction in false positive calls. We also overcame the lack of annotation resources for CHM13-T2T by lifting over CHM13-T2T-aligned reads to the GRCh38 genome, therefore combining both improved alignment and advanced annotations. In this process, we assessed the current SV benchmark set for COLO829/COLO829BL across four replicates sequenced at different centers with different long-read technologies. We discovered instability of this cell line across these replicates; 346 SVs (1.13%) were only discoverable in a single replicate. We identify 49 somatic SVs, which appear to be stable as they are consistently present across the four replicates. As such, we propose this consensus set as an updated benchmark for somatic SV calling and include both GRCh38 and CHM13-T2T coordinates in our benchmark. The benchmark is available at: 10.5281/zenodo.10819636 Our work demonstrates new approaches to optimize somatic SV prioritization in cancer with potential improvements in other genetic diseases., Competing Interests: The following authors disclose relevant potential competing interests: Kieran O’Neill, Vanessa Porter, Luis F Paulin and Steven J.M. Jones received travel funding from Oxford Nanopore Technologies to present at conferences in 2022 and/or 2023. Fritz J Sedlazeck receives research support from ONT, Pacbio, Illumina and Genentech. Luis F Paulin received research support from Genentech from 2021 to 2023.
- Published
- 2024
- Full Text
- View/download PDF
30. Comprehensive Immune Profiling Unveils a Subset of Leiomyosarcoma with "Hot" Tumor Immune Microenvironment.
- Author
-
Feng X, Tonon L, Li H, Darbo E, Pleasance E, Macagno N, Dufresne A, Brahmi M, Bollard J, Ducimetière F, Karanian M, Meurgey A, Pérot G, Valentin T, Chibon F, and Blay JY
- Abstract
Purpose: To investigate the immune biomarker in Leiomyosarcoma (LMS), which is rare and recognized as an immune cold cancer showing a poor response rate (<10%) to immune checkpoint inhibitors (ICIs). However, durable response and clinical benefit to ICIs has been observed in a few cases of LMS, including, but not only, LMS with tertiary lymphoid structure (TLS) structures. Patients and methods: We used comprehensive transcriptomic profiling and a deconvolution method extracted from RNA-sequencing gene expression data in two independent LMS cohorts, the International Cancer Genome Consortium (ICGC, N = 146) and The Cancer Genome Atlas (TCGA, N = 75), to explore tumor immune microenvironment (TIME) in LMS. Results: Unsupervised clustering analysis using the previously validated two methods, 90-gene signature and Cell-type Identification by Estimating Relative Subsets of RNA Transcripts (CIBERSORT), identified immune hot (I-H) and immune high (I-Hi) LMS, respectively, in the ICGC cohort. Similarly, immune active groups (T-H, T-Hi) were identified in the TCGA cohort using these two methods. These immune active ("hot") clusters were significantly associated, but not completely overlapping, with several validated immune signatures such as sarcoma immune class (SIC) classification and TLS score, T cell inflamed signature (TIS) score, immune infiltration score (IIS), and macrophage score (M1/M2), with more patients identified by our clustering as potentially immune hot. Conclusions: Comprehensive immune profiling revealed a subset of LMS with a distinct active ("hot") TIME, consistently associated with several validated immune signatures in other cancers. This suggests that the methodologies that we used in this study warrant further validation and development, which can potentially help refine our current immune biomarkers to select the right LMS patients for ICIs in clinical trials.
- Published
- 2023
- Full Text
- View/download PDF
31. Homologous recombination deficiency signatures in gastrointestinal and thoracic cancers correlate with platinum therapy duration.
- Author
-
Tsang ES, Csizmok V, Williamson LM, Pleasance E, Topham JT, Karasinska JM, Titmuss E, Schrader I, Yip S, Tessier-Cloutier B, Mungall K, Ng T, Sun S, Lim HJ, Loree JM, Laskin J, Marra MA, Jones SJM, Schaeffer DF, and Renouf DJ
- Abstract
There is emerging evidence about the predictive role of homologous recombination deficiency (HRD), but this is less defined in gastrointestinal (GI) and thoracic malignancies. We reviewed whole genome (WGS) and transcriptomic (RNA-Seq) data from advanced GI and thoracic cancers in the Personalized OncoGenomics trial (NCT02155621) to evaluate HRD scores and single base substitution (SBS)3, which is associated with BRCA1/2 mutations and potentially predictive of defective HRD. HRD scores were calculated by sum of loss of heterozygosity, telomeric allelic imbalance, and large-scale state transitions scores. Regression analyses examined the association between HRD and time to progression on platinum (TTPp). We included 223 patients with GI (n = 154) or thoracic (n = 69) malignancies. TTPp was associated with SBS3 (p < 0.01) but not HRD score in patients with GI malignancies, whereas neither was associated with TTPp in thoracic malignancies. Tumors with gBRCA1/2 mutations and a somatic second alteration exhibited high SBS3 and HRD scores, but these signatures were also present in several tumors with germline but no somatic second alterations, suggesting silencing of the wild-type allele or BRCA1/2 haploinsufficiency. Biallelic inactivation of an HR gene, including loss of XRCC2 and BARD1, was identified in BRCA1/2 wild-type HRD tumors and these patients had prolonged response to platinum. Thoracic cases with high HRD score were associated with high RECQL5 expression (p ≤ 0.025), indicating another potential mechanism of HRD. SBS3 was more strongly associated with TTPp in patients with GI malignancies and may be complementary to using HRD and BRCA status in identifying patients who benefit from platinum therapy., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
32. A p53 transcriptional signature in primary and metastatic cancers derived using machine learning.
- Author
-
Keshavarz-Rahaghi F, Pleasance E, Kolisnik T, and Jones SJM
- Abstract
The tumor suppressor gene, TP53 , has the highest rate of mutation among all genes in human cancer. This transcription factor plays an essential role in the regulation of many cellular processes. Mutations in TP53 result in loss of wild-type p53 function in a dominant negative manner. Although TP53 is a well-studied gene, the transcriptome modifications caused by the mutations in this gene have not yet been explored in a pan-cancer study using both primary and metastatic samples. In this work, we used a random forest model to stratify tumor samples based on TP53 mutational status and detected a p53 transcriptional signature. We hypothesize that the existence of this transcriptional signature is due to the loss of wild-type p53 function and is universal across primary and metastatic tumors as well as different tumor types. Additionally, we showed that the algorithm successfully detected this signature in samples with apparent silent mutations that affect correct mRNA splicing. Furthermore, we observed that most of the highly ranked genes contributing to the classification extracted from the random forest have known associations with p53 within the literature. We suggest that other genes found in this list including GPSM2, OR4N2, CTSL2, SPERT, and RPE65 protein coding genes have yet undiscovered linkages to p53 function. Our analysis of time on different therapies also revealed that this signature is more effective than the recorded TP53 status in detecting patients who can benefit from platinum therapies and taxanes. Our findings delineate a p53 transcriptional signature, expand the knowledge of p53 biology and further identify genes important in p53 related pathways., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Keshavarz-Rahaghi, Pleasance, Kolisnik and Jones.)
- Published
- 2022
- Full Text
- View/download PDF
33. The impact of whole genome and transcriptome analysis (WGTA) on predictive biomarker discovery and diagnostic accuracy of advanced malignancies.
- Author
-
Tessier-Cloutier B, Grewal JK, Jones MR, Pleasance E, Shen Y, Cai E, Dunham C, Hoang L, Horst B, Huntsman DG, Ionescu D, Karnezis AN, Lee AF, Lee CH, Lee TH, Twa DD, Mungall AJ, Mungall K, Naso JR, Ng T, Schaeffer DF, Sheffield BS, Skinnider B, Smith T, Williamson L, Zhong E, Regier DA, Laskin J, Marra MA, Gilks CB, Jones SJ, and Yip S
- Subjects
- Algorithms, Biomarkers, Tumor genetics, Gene Expression Profiling, Humans, Neoplasms diagnosis, Neoplasms drug therapy, Neoplasms genetics
- Abstract
In this study, we evaluate the impact of whole genome and transcriptome analysis (WGTA) on predictive molecular profiling and histologic diagnosis in a cohort of advanced malignancies. WGTA was used to generate reports including molecular alterations and site/tissue of origin prediction. Two reviewers analyzed genomic reports, clinical history, and tumor pathology. We used National Comprehensive Cancer Network (NCCN) consensus guidelines, Food and Drug Administration (FDA) approvals, and provincially reimbursed treatments to define genomic biomarkers associated with approved targeted therapeutic options (TTOs). Tumor tissue/site of origin was reassessed for most cases using genomic analysis, including a machine learning algorithm (Supervised Cancer Origin Prediction Using Expression [SCOPE]) trained on The Cancer Genome Atlas data. WGTA was performed on 652 cases, including a range of primary tumor types/tumor sites and 15 malignant tumors of uncertain histogenesis (MTUH). At the time WGTA was performed, alterations associated with an approved TTO were identified in 39 (6%) cases; 3 of these were not identified through routine pathology workup. In seven (1%) cases, the pathology workup either failed, was not performed, or gave a different result from the WGTA. Approved TTOs identified by WGTA increased to 103 (16%) when applying 2021 guidelines. The histopathologic diagnosis was reviewed in 389 cases and agreed with the diagnostic consensus after WGTA in 94% of non-MTUH cases (n = 374). The remainder included situations where the morphologic diagnosis was changed based on WGTA and clinical data (0.5%), or where the WGTA was non-contributory (5%). The 15 MTUH were all diagnosed as specific tumor types by WGTA. Tumor board reviews including WGTA agreed with almost all initial predictive molecular profile and histopathologic diagnoses. WGTA was a powerful tool to assign site/tissue of origin in MTUH. Current efforts focus on improving therapeutic predictive power and decreasing cost to enhance use of WGTA data as a routine clinical test., (© 2022 The Authors. The Journal of Pathology: Clinical Research published by The Pathological Society of Great Britain and Ireland & John Wiley & Sons, Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
34. A platform for oncogenomic reporting and interpretation.
- Author
-
Reisle C, Williamson LM, Pleasance E, Davies A, Pellegrini B, Bleile DW, Mungall KL, Chuah E, Jones MR, Ma Y, Lewis E, Beckie I, Pham D, Matiello Pletz R, Muhammadzadeh A, Pierce BM, Li J, Stevenson R, Wong H, Bailey L, Reisle A, Douglas M, Bonakdar M, Nelson JMT, Grisdale CJ, Krzywinski M, Fisic A, Mitchell T, Renouf DJ, Yip S, Laskin J, Marra MA, and Jones SJM
- Subjects
- Biomarkers, Tumor, Databases, Genetic, Genetic Variation, Genomics, Humans, Knowledge Bases, Precision Medicine, Carcinogenesis genetics, Neoplasms genetics
- Abstract
Manual interpretation of variants remains rate limiting in precision oncology. The increasing scale and complexity of molecular data generated from comprehensive sequencing of cancer samples requires advanced interpretative platforms as precision oncology expands beyond individual patients to entire populations. To address this unmet need, we introduce a Platform for Oncogenomic Reporting and Interpretation (PORI), comprising an analytic framework that facilitates the interpretation and reporting of somatic variants in cancer. PORI integrates reporting and graph knowledge base tools combined with support for manual curation at the reporting stage. PORI represents an open-source platform alternative to commercial reporting solutions suitable for comprehensive genomic data sets in precision oncology. We demonstrate the utility of PORI by matching 9,961 pan-cancer genome atlas tumours to the graph knowledge base, calculating therapeutically informative alterations, and making available reports describing select individual samples., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
35. Clinical response to nivolumab in an INI1-deficient pediatric chordoma correlates with immunogenic recognition of brachyury.
- Author
-
Williamson LM, Rive CM, Di Francesco D, Titmuss E, Chun HE, Brown SD, Milne K, Pleasance E, Lee AF, Yip S, Rosenbaum DG, Hasselblatt M, Johann PD, Kool M, Harvey M, Dix D, Renouf DJ, Holt RA, Nelson BH, Hirst M, Jones SJM, Laskin J, Rassekh SR, Deyell RJ, and Marra MA
- Abstract
Poorly differentiated chordoma (PDC) is a recently recognized subtype of chordoma characterized by expression of the embryonic transcription factor, brachyury, and loss of INI1. PDC primarily affects children and is associated with a poor prognosis and limited treatment options. Here we describe the molecular and immune tumour microenvironment profiles of two paediatric PDCs produced using whole-genome, transcriptome and whole-genome bisulfite sequencing (WGBS) and multiplex immunohistochemistry. Our analyses revealed the presence of tumour-associated immune cells, including CD8+ T cells, and expression of the immune checkpoint protein, PD-L1, in both patient samples. Molecular profiling provided the rationale for immune checkpoint inhibitor (ICI) therapy, which resulted in a clinical and radiographic response. A dominant T cell receptor (TCR) clone specific for a brachyury peptide-MHC complex was identified from bulk RNA sequencing, suggesting that targeting of the brachyury tumour antigen by tumour-associated T cells may underlie this clinical response to ICI. Correlative analysis with rhabdoid tumours, another INI1-deficient paediatric malignancy, suggests that a subset of tumours may share common immune phenotypes, indicating the potential for a therapeutically targetable subgroup of challenging paediatric cancers., (© 2021. Crown.)
- Published
- 2021
- Full Text
- View/download PDF
36. Rearrangement-mediated cis-regulatory alterations in advanced patient tumors reveal interactions with therapy.
- Author
-
Zhang Y, Chen F, Pleasance E, Williamson L, Grisdale CJ, Titmuss E, Laskin J, Jones SJM, Cortes-Ciriano I, Marra MA, and Creighton CJ
- Subjects
- Chromosome Aberrations, DNA Copy Number Variations genetics, Databases, Genetic, Gene Rearrangement genetics, Genome, Human, Genomics, Humans, Oncogenes, Sequence Analysis, RNA methods, Translocation, Genetic genetics, Exome Sequencing methods, Whole Genome Sequencing methods, Gene Expression Regulation, Neoplastic genetics, Genomic Structural Variation genetics, Neoplasm Recurrence, Local genetics
- Abstract
The global impact of somatic structural variants (SVs) on gene regulation in advanced tumors with complex treatment histories has been mostly uncharacterized. Here, using whole-genome and RNA sequencing from 570 recurrent or metastatic tumors, we report the altered expression of hundreds of genes in association with nearby SV breakpoints, including oncogenes and G-protein-coupled receptor-related genes such as PLEKHG2. A significant fraction of genes with SV-expression associations correlate with worse patient survival in primary and advanced cancers, including SRD5A1. In many instances, SV-expression associations involve retrotransposons being translocated near genes. High overall SV burden is associated with treatment with DNA alkylating agents or taxanes and altered expression of metabolism-associated genes. SV-expression associations within tumors from topoisomerase I inhibitor-treated patients include chromatin-related genes. Within anthracycline-treated tumors, SV breakpoints near chromosome 1p genes include PDE4B. Patient treatment and history can help understand the widespread SV-mediated cis-regulatory alterations found in cancer., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
37. Clinical and cost outcomes following genomics-informed treatment for advanced cancers.
- Author
-
Weymann D, Pollard S, Chan B, Titmuss E, Bohm A, Laskin J, Jones SJM, Pleasance E, Nelson J, Fok A, Lim H, Karsan A, Renouf DJ, Schrader KA, Sun S, Yip S, Schaeffer DF, Marra MA, and Regier DA
- Subjects
- Breast Neoplasms, Costs and Cost Analysis, Female, Gastrointestinal Neoplasms genetics, Gastrointestinal Neoplasms mortality, Gastrointestinal Neoplasms pathology, Gastrointestinal Neoplasms therapy, Genome-Wide Association Study, Genomics economics, Genomics methods, Humans, Logistic Models, Male, Middle Aged, Neoplasms mortality, Neoplasms pathology, Precision Medicine methods, Retrospective Studies, Treatment Outcome, Withholding Treatment, Neoplasms genetics, Neoplasms therapy, Precision Medicine economics, Sequence Analysis, DNA
- Abstract
Background: Single-arm trials are common in precision oncology. Owing to the lack of randomized counterfactual, resultant data are not amenable to comparative outcomes analyses. Difference-in-difference (DID) methods present an opportunity to generate causal estimates of time-varying treatment outcomes. Using DID, our study estimates within-cohort effects of genomics-informed treatment versus standard care on clinical and cost outcomes., Methods: We focus on adults with advanced cancers enrolled in the single-arm BC Cancer Personalized OncoGenomics program between 2012 and 2017. All individuals had a minimum of 1-year follow up. Logistic regression explored baseline differences across patients who received a genomics-informed treatment versus a standard care treatment after genomic sequencing. DID estimated the incremental effects of genomics-informed treatment on time to treatment discontinuation (TTD), time to next treatment (TTNT), and costs. TTD and TTNT correlate with improved response and survival., Results: Our study cohort included 346 patients, of whom 140 (40%) received genomics-informed treatment after sequencing and 206 (60%) received standard care treatment. No significant differences in baseline characteristics were detected across treatment groups. DID estimated that the incremental effect of genomics-informed versus standard care treatment was 102 days (95% CI: 35, 167) on TTD, 91 days (95% CI: -9, 175) on TTNT, and CAD$91,098 (95% CI: $46,848, $176,598) on costs. Effects were most pronounced in gastrointestinal cancer patients., Conclusions: Genomics-informed treatment had a statistically significant effect on TTD compared to standard care treatment, but at increased treatment costs. Within-cohort evidence generated through this single-arm study informs the early-stage comparative effectiveness of precision oncology., (© 2021 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
38. NTRK2 Fusion driven pediatric glioblastoma: Identification of oncogenic Drivers via integrative Genome and transcriptome profiling.
- Author
-
Britton HM, Levine AB, Shen Y, Mungall K, Serrano J, Snuderl M, Pleasance E, Jones SJM, Laskin J, Marra MA, Rassekh SR, Deyell R, Yip S, Cheng S, and Dunham C
- Abstract
This is the first report of a NACC2-NTRK2 fusion in a histological glioblastoma. Oncogenomic analysis revealed this actionable fusion oncogene in a pediatric cerebellar glioblastoma, which would not have been identified through routine diagnostics, demonstrating the value of clinical genome profiling in cancer care., Competing Interests: The authors declare no competing interests., (© 2021 The Authors. Clinical Case Reports published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
39. Uncovering Clinically Relevant Gene Fusions with Integrated Genomic and Transcriptomic Profiling of Metastatic Cancers.
- Author
-
Tsang ES, Grisdale CJ, Pleasance E, Topham JT, Mungall K, Reisle C, Choo C, Carreira M, Bowlby R, Karasinska JM, MacMillan D, Williamson LM, Chuah E, Moore RA, Mungall AJ, Zhao Y, Tessier-Cloutier B, Ng T, Sun S, Lim HJ, Schaeffer DF, Renouf DJ, Yip S, Laskin J, Marra MA, Jones SJM, and Loree JM
- Subjects
- Humans, Neoplasm Metastasis, Neoplasms drug therapy, Neoplasms pathology, RNA-Seq methods, Retrospective Studies, Treatment Outcome, Exome Sequencing methods, Gene Expression Profiling methods, Gene Fusion, Genomics methods, Neoplasms genetics, Oncogene Proteins, Fusion genetics
- Abstract
Purpose: Gene fusions are important oncogenic drivers and many are actionable. Whole-genome and transcriptome (WGS and RNA-seq, respectively) sequencing can discover novel clinically relevant fusions., Experimental Design: Using WGS and RNA-seq, we reviewed the prevalence of fusions in a cohort of 570 patients with cancer, and compared prevalence to that predicted with commercially available panels. Fusions were annotated using a consensus variant calling pipeline (MAVIS) and required that a contig of the breakpoint could be constructed and supported from ≥2 structural variant detection approaches., Results: In 570 patients with advanced cancer, MAVIS identified 81 recurrent fusions by WGS and 111 by RNA-seq, of which 18 fusions by WGS and 19 by RNA-seq were noted in at least 3 separate patients. The most common fusions were EML4-ALK in thoracic malignancies (9/69, 13%), and CMTM8-CMTM7 in colorectal cancer (4/73, 5.5%). Combined genomic and transcriptomic analysis identified novel fusion partners for clinically relevant genes, such as NTRK2 (novel partners: SHC3, DAPK1) , and NTRK3 (novel partners: POLG, PIBF1 )., Conclusions: Utilizing WGS/RNA-seq facilitates identification of novel fusions in clinically relevant genes, and detected a greater proportion than commercially available panels are expected to find. A significant benefit of WGS and RNA-seq is the innate ability to retrospectively identify variants that becomes clinically relevant over time, without the need for additional testing, which is not possible with panel-based approaches., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
40. Improved structural variant interpretation for hereditary cancer susceptibility using long-read sequencing.
- Author
-
Thibodeau ML, O'Neill K, Dixon K, Reisle C, Mungall KL, Krzywinski M, Shen Y, Lim HJ, Cheng D, Tse K, Wong T, Chuah E, Fok A, Sun S, Renouf D, Schaeffer DF, Cremin C, Chia S, Young S, Pandoh P, Pleasance S, Pleasance E, Mungall AJ, Moore R, Yip S, Karsan A, Laskin J, Marra MA, Schrader KA, and Jones SJM
- Subjects
- Base Sequence, Genome, High-Throughput Nucleotide Sequencing, Humans, Genetic Predisposition to Disease, Neoplasms
- Abstract
Purpose: Structural variants (SVs) may be an underestimated cause of hereditary cancer syndromes given the current limitations of short-read next-generation sequencing. Here we investigated the utility of long-read sequencing in resolving germline SVs in cancer susceptibility genes detected through short-read genome sequencing., Methods: Known or suspected deleterious germline SVs were identified using Illumina genome sequencing across a cohort of 669 advanced cancer patients with paired tumor genome and transcriptome sequencing. Candidate SVs were subsequently assessed by Oxford Nanopore long-read sequencing., Results: Nanopore sequencing confirmed eight simple pathogenic or likely pathogenic SVs, resolving three additional variants whose impact could not be fully elucidated through short-read sequencing. A recurrent sequencing artifact on chromosome 16p13 and one complex rearrangement on chromosome 5q35 were subsequently classified as likely benign, obviating the need for further clinical assessment. Variant configuration was further resolved in one case with a complex pathogenic rearrangement affecting TSC2., Conclusion: Our findings demonstrate that long-read sequencing can improve the validation, resolution, and classification of germline SVs. This has important implications for return of results, cascade carrier testing, cancer screening, and prophylactic interventions.
- Published
- 2020
- Full Text
- View/download PDF
41. Establishing a Framework for the Clinical Translation of Germline Findings in Precision Oncology.
- Author
-
Dixon K, Young S, Shen Y, Thibodeau ML, Fok A, Pleasance E, Zhao E, Jones M, Aubert G, Armstrong L, Virani A, Regier D, Gelmon K, Renouf D, Chia S, Bosdet I, Rassekh SR, Deyell RJ, Yip S, Fisic A, Titmuss E, Abadi S, Jones SJM, Sun S, Karsan A, Marra M, Laskin J, Lim H, and Schrader KA
- Abstract
Inherited genetic variation has important implications for cancer screening, early diagnosis, and disease prognosis. A role for germline variation has also been described in shaping the molecular landscape, immune response, microenvironment, and treatment response of individual tumors. However, there is a lack of consensus on the handling and analysis of germline information that extends beyond known or suspected cancer susceptibility in large-scale cancer genomics initiatives. As part of the Personalized OncoGenomics program in British Columbia, we performed whole-genome and transcriptome sequencing in paired tumor and normal tissues from advanced cancer patients to characterize the molecular tumor landscape and identify putative targets for therapy. Overall, our experience supports a multidisciplinary and integrative approach to germline data management. This includes a need for broader definitions and standardized recommendations regarding primary and secondary germline findings in precision oncology. Here, we propose a framework for identifying, evaluating, and returning germline variants of potential clinical significance that may have indications for health management beyond cancer risk reduction or prevention in patients and their families., (© The Author(s) 2020. Published by Oxford University Press.)
- Published
- 2020
- Full Text
- View/download PDF
42. Pan-cancer analysis of advanced patient tumors reveals interactions between therapy and genomic landscapes.
- Author
-
Pleasance E, Titmuss E, Williamson L, Kwan H, Culibrk L, Zhao EY, Dixon K, Fan K, Bowlby R, Jones MR, Shen Y, Grewal JK, Ashkani J, Wee K, Grisdale CJ, Thibodeau ML, Bozoky Z, Pearson H, Majounie E, Vira T, Shenwai R, Mungall KL, Chuah E, Davies A, Warren M, Reisle C, Bonakdar M, Taylor GA, Csizmok V, Chan SK, Zong Z, Bilobram S, Muhammadzadeh A, D'Souza D, Corbett RD, MacMillan D, Carreira M, Choo C, Bleile D, Sadeghi S, Zhang W, Wong T, Cheng D, Brown SD, Holt RA, Moore RA, Mungall AJ, Zhao Y, Nelson J, Fok A, Ma Y, Lee MKC, Lavoie JM, Mendis S, Karasinska JM, Deol B, Fisic A, Schaeffer DF, Yip S, Schrader K, Regier DA, Weymann D, Chia S, Gelmon K, Tinker A, Sun S, Lim H, Renouf DJ, Laskin J, Jones SJM, and Marra MA
- Subjects
- Humans, Neoplasms drug therapy
- Abstract
Advanced and metastatic tumors with complex treatment histories drive cancer mortality. Here we describe the POG570 cohort, a comprehensive whole-genome, transcriptome and clinical dataset, amenable for exploration of the impacts of therapies on genomic landscapes. Previous exposure to DNA-damaging chemotherapies and mutations affecting DNA repair genes, including POLQ and genes encoding Polζ, were associated with genome-wide, therapy-induced mutagenesis. Exposure to platinum therapies coincided with signatures SBS31 and DSB5 and, when combined with DNA synthesis inhibitors, signature SBS17b. Alterations in ESR1, EGFR, CTNNB1, FGFR1, VEGFA and DPYD were consistent with drug resistance and sensitivity. Recurrent noncoding events were found in regulatory region hotspots of genes including TERT, PLEKHS1, AP2A1 and ADGRG6. Mutation burden and immune signatures corresponded with overall survival and response to immunotherapy. Our data offer a rich resource for investigation of advanced cancers and interpretation of whole-genome and transcriptome sequencing in the context of a cancer clinic., (© 2020. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
43. Fluorouracil sensitivity in a head and neck squamous cell carcinoma with a somatic DPYD structural variant.
- Author
-
Majounie E, Wee K, Williamson LM, Jones MR, Pleasance E, Lim HJ, Ho C, Renouf DJ, Yip S, Jones SJM, Marra MA, and Laskin J
- Subjects
- Aged, Alleles, Amino Acid Substitution, Antimetabolites, Antineoplastic therapeutic use, Biopsy, Fluorouracil therapeutic use, Humans, Male, Phenotype, Polymorphism, Single Nucleotide, Sequence Analysis, DNA, Squamous Cell Carcinoma of Head and Neck diagnosis, Squamous Cell Carcinoma of Head and Neck drug therapy, Antimetabolites, Antineoplastic pharmacology, Dihydrouracil Dehydrogenase (NADP) genetics, Drug Resistance, Neoplasm genetics, Fluorouracil pharmacology, Genetic Variation, Squamous Cell Carcinoma of Head and Neck genetics
- Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most common cancers worldwide and represents a heterogeneous group of tumors, the majority of which are treated with a combination of surgery, radiation, and chemotherapy. Fluoropyrimidine (5-FU) and its oral prodrug, capecitabine, are commonly prescribed treatments for several solid tumor types including HNSCC. 5-FU-associated toxicity is observed in ∼30% of treated patients and is largely caused by germline polymorphisms in DPYD , which encodes dihydropyrimidine dehydrogenase, a key enzyme of 5-FU catabolism and deactivation. Although the association of germline DPYD alterations with toxicity is well-described, the potential contribution of somatic DPYD alterations to 5-FU sensitivity has not been explored. In a patient with metastatic HNSCC, in-depth genomic and transcriptomic integrative analysis on a biopsy from a metastatic neck lesion revealed alterations in genes that are associated with 5-FU uptake and metabolism. These included a novel somatic structural variant resulting in a partial deletion affecting DPYD , a variant of unknown significance affecting SLC29A1 , and homozygous deletion of MTAP There was no evidence of deleterious germline polymorphisms that have been associated with 5-FU toxicity, indicating a potential vulnerability of the tumor to 5-FU therapy. The discovery of the novel DPYD variant led to the initiation of 5-FU treatment that resulted in a rapid response lasting 17 wk, with subsequent relapse due to unknown resistance mechanisms. This suggests that somatic alterations present in this tumor may serve as markers for tumor sensitivity to 5-FU, aiding in the selection of personalized treatment strategies., (© 2020 Majounie et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2020
- Full Text
- View/download PDF
44. Therapeutic Implication of Genomic Landscape of Adult Metastatic Sarcoma.
- Author
-
Feng X, Pleasance E, Zhao EY, Ng T, Grewal JK, Mohammad N, Taylor SK, Simmons C, Srikanthan A, Rassekh SR, Deyell R, Rauw J, Knowling M, Khoo K, Lee U, Noonan K, Hart J, Tonseth RP, Shen Y, Titmuss E, Jones M, Bonakdar M, Reisle C, Taylor GA, Chan S, Mungall K, Chuah E, Zhao Y, Mungall A, Moore R, Lim H, Renouf DJ, Gelmon K, Yip S, Jones SJM, Marra M, and Laskin J
- Abstract
Purpose: This study investigated therapeutic potential of integrated genome and transcriptome profiling of metastatic sarcoma, a rare but extremely heterogeneous group of aggressive mesenchymal malignancies with few systemic therapeutic options., Methods: Forty-three adult patients with advanced or metastatic non-GI stromal tumor sarcomas of various histology subtypes who were enrolled in the Personalized OncoGenomics program at BC Cancer were included in this study. Fresh tumor tissues along with blood samples underwent whole-genome and transcriptome sequencing., Results: The most frequent genomic alterations in this cohort are large-scale structural variation and somatic copy number variation. Outlier RNA expression as well as somatic copy number variations, structural variations, and small mutations together suggest the presence of one or more potential therapeutic targets in the majority of patients in our cohort. Point mutations or deletions in known targetable cancer genes are rare; for example, tuberous sclerosis complex 2 provides a rationale for targeting the mammalian target of rapamycin pathway, resulting in a few patients with exceptional clinical benefit from everolimus. In addition, we observed recurrent 17p11-12 amplifications, which seem to be a sarcoma-specific event. This may suggest that this region harbors an oncogene(s) that is significant for sarcoma tumorigenesis. Furthermore, some sarcoma tumors carrying a distinct mutational signature suggestive of homologous recombination deficiency seem to demonstrate sensitivity to double-strand DNA-damaging agents., Conclusion: Integrated large-scale genomic analysis may provide insights into potential therapeutic targets as well as novel biologic features of metastatic sarcomas that could fuel future experimental and clinical research and help design biomarker-driven basket clinical trials for novel therapeutic strategies.
- Published
- 2019
- Full Text
- View/download PDF
45. The pivotal role of sampling recurrent tumors in the precision care of patients with tumors of the central nervous system.
- Author
-
Wong D, Shen Y, Levine AB, Pleasance E, Jones M, Mungall K, Thiessen B, Toyota B, Laskin J, Jones SJM, Marra MA, and Yip S
- Subjects
- Adult, Female, Genomics methods, Humans, Male, Middle Aged, Neoplasm Recurrence, Local genetics, Prognosis, Exome Sequencing methods, Central Nervous System Neoplasms genetics, Central Nervous System Neoplasms therapy, Neoplasm Recurrence, Local diagnosis, Precision Medicine methods
- Abstract
Effective management of brain and spine tumors relies on a multidisciplinary approach encompassing surgery, radiation, and systemic therapy. In the era of personalized oncology, the latter is complemented by various molecularly targeting agents. Precise identification of cellular targets for these drugs requires comprehensive profiling of the cancer genome coupled with an efficient analytic pipeline, leading to an informed decision on drug selection, prognosis, and confirmation of the original pathological diagnosis. Acquisition of optimal tumor tissue for such analysis is paramount and often presents logistical challenges in neurosurgery. Here, we describe the experience and results of the Personalized OncoGenomics (POG) program with a focus on tumors of the central nervous system (CNS). Patients with recurrent CNS tumors were consented and enrolled into the POG program prior to accrual of tumor and matched blood followed by whole-genome and transcriptome sequencing and processing through the POG bioinformatic pipeline. Sixteen patients were enrolled into POG. In each case, POG analyses identified genomic drivers including novel oncogenic fusions, aberrant pathways, and putative therapeutic targets. POG has highlighted that personalized oncology is truly a multidisciplinary field, one in which neurosurgeons must play a vital role if these programs are to succeed and benefit our patients., (© 2019 Wong et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2019
- Full Text
- View/download PDF
46. NRG1 Gene Fusions Are Recurrent, Clinically Actionable Gene Rearrangements in KRAS Wild-Type Pancreatic Ductal Adenocarcinoma.
- Author
-
Jones MR, Williamson LM, Topham JT, Lee MKC, Goytain A, Ho J, Denroche RE, Jang G, Pleasance E, Shen Y, Karasinska JM, McGhie JP, Gill S, Lim HJ, Moore MJ, Wong HL, Ng T, Yip S, Zhang W, Sadeghi S, Reisle C, Mungall AJ, Mungall KL, Moore RA, Ma Y, Knox JJ, Gallinger S, Laskin J, Marra MA, Schaeffer DF, Jones SJM, and Renouf DJ
- Subjects
- Female, Gene Fusion, Gene Rearrangement, Humans, Neuregulin-1, Oncogene Proteins, Fusion genetics, Positron Emission Tomography Computed Tomography, Proto-Oncogene Proteins p21(ras), Lung Neoplasms genetics, Pancreatic Neoplasms
- Abstract
Purpose: Gene fusions involving neuregulin 1 ( NRG1 ) have been noted in multiple cancer types and have potential therapeutic implications. Although varying results have been reported in other cancer types, the efficacy of the HER-family kinase inhibitor afatinib in the treatment of NRG1 fusion-positive pancreatic ductal adenocarcinoma is not fully understood., Experimental Design: Forty-seven patients with pancreatic ductal adenocarcinoma received comprehensive whole-genome and transcriptome sequencing and analysis. Two patients with gene fusions involving NRG1 received afatinib treatment, with response measured by pretreatment and posttreatment PET/CT imaging., Results: Three of 47 (6%) patients with advanced pancreatic ductal adenocarcinoma were identified as KRAS wild type by whole-genome sequencing. All KRAS wild-type tumors were positive for gene fusions involving the ERBB3 ligand NRG1 . Two of 3 patients with NRG1 fusion-positive tumors were treated with afatinib and demonstrated a significant and rapid response while on therapy., Conclusions: This work adds to a growing body of evidence that NRG1 gene fusions are recurrent, therapeutically actionable genomic events in pancreatic cancers. Based on the clinical outcomes described here, patients with KRAS wild-type tumors harboring NRG1 gene fusions may benefit from treatment with afatinib. See related commentary by Aguirre, p. 4589 ., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
47. Base excision repair deficiency signatures implicate germline and somatic MUTYH aberrations in pancreatic ductal adenocarcinoma and breast cancer oncogenesis.
- Author
-
Thibodeau ML, Zhao EY, Reisle C, Ch'ng C, Wong HL, Shen Y, Jones MR, Lim HJ, Young S, Cremin C, Pleasance E, Zhang W, Holt R, Eirew P, Karasinska J, Kalloger SE, Taylor G, Majounie E, Bonakdar M, Zong Z, Bleile D, Chiu R, Birol I, Gelmon K, Lohrisch C, Mungall KL, Mungall AJ, Moore R, Ma YP, Fok A, Yip S, Karsan A, Huntsman D, Schaeffer DF, Laskin J, Marra MA, Renouf DJ, Jones SJM, and Schrader KA
- Subjects
- Age of Onset, DNA Glycosylases deficiency, Female, Germ-Line Mutation, Humans, Loss of Heterozygosity, Middle Aged, Proto-Oncogene Proteins p21(ras) genetics, Breast Neoplasms genetics, Carcinoma, Pancreatic Ductal genetics, DNA Glycosylases genetics, Mutation, Pancreatic Neoplasms genetics
- Abstract
We report a case of early-onset pancreatic ductal adenocarcinoma in a patient harboring biallelic MUTYH germline mutations, whose tumor featured somatic mutational signatures consistent with defective MUTYH -mediated base excision repair and the associated driver KRAS transversion mutation p.Gly12Cys. Analysis of an additional 730 advanced cancer cases ( N = 731) was undertaken to determine whether the mutational signatures were also present in tumors from germline MUTYH heterozygote carriers or if instead the signatures were only seen in those with biallelic loss of function. We identified two patients with breast cancer each carrying a pathogenic germline MUTYH variant with a somatic MUTYH copy loss leading to the germline variant being homozygous in the tumor and demonstrating the same somatic signatures. Our results suggest that monoallelic inactivation of MUTYH is not sufficient for C:G>A:T transversion signatures previously linked to MUTYH deficiency to arise ( N = 9), but that biallelic complete loss of MUTYH function can cause such signatures to arise even in tumors not classically seen in MUTYH -associated polyposis ( N = 3). Although defective MUTYH is not the only determinant of these signatures, MUTYH germline variants may be present in a subset of patients with tumors demonstrating elevated somatic signatures possibly suggestive of MUTYH deficiency (e.g., COSMIC Signature 18, SigProfiler SBS18/SBS36, SignatureAnalyzer SBS18/SBS36)., (© 2019 Thibodeau et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2019
- Full Text
- View/download PDF
48. Clinical outcomes after whole-genome sequencing in patients with metastatic non-small-cell lung cancer.
- Author
-
Tsang ES, Shen Y, Chooback N, Ho C, Jones M, Renouf DJ, Lim H, Sun S, Yip S, Pleasance E, Ionescu DN, Mungall K, Kasaian K, Ma Y, Zhao Y, Mungall A, Moore R, Jones SJM, Marra M, and Laskin J
- Subjects
- Adenocarcinoma diagnosis, Aged, Carcinoma, Non-Small-Cell Lung diagnosis, Carcinoma, Squamous Cell diagnosis, Carcinoma, Squamous Cell genetics, Female, Humans, Lung Neoplasms diagnosis, Male, Middle Aged, Progression-Free Survival, Whole Genome Sequencing, Adenocarcinoma genetics, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms genetics, Mutation
- Abstract
The Personalized Onco-Genomics (POG) program at BC Cancer integrates whole-genome (DNA) and RNA sequencing into practice for metastatic malignancies. We examined the subgroup of patients with metastatic non-small-cell lung cancer (NSCLC) and report the prevalence of actionable targets, treatments, and outcomes. We identified patients who were enrolled in the POG program between 2012 and 2016 who had a tumor biopsy and blood samples with comprehensive DNA (80×, 40× normal) and RNA sequencing followed by in-depth bioinformatics to identify potential cancer drivers and actionable targets. In NSCLC cases, we compared the progression-free survival (PFS) of "POG-informed therapies" with the PFS of the last regimen prior to POG (PFS ratio). In 29 NSCLC cases, 11 were male (38%), the median age was 60.2 yr (range: 39.4-72.6), and histologies included were adenocarcinoma (93%) and squamous cell carcinoma (7%). Potential molecular targets (i.e., cancer drivers including TP53 mutations) were identified in 26 (90%), and 21 (72%) had actionable targets. Therapies based on standard-of-care mutation analysis, such as EGFR mutations, were not considered POG-informed therapies. Thirteen received POG-informed therapies, of which three had no therapy before POG; therefore a comparator PFS could not be obtained. Of 10 patients with POG-informed therapy, median PFS ratio was 0.94 (IQR 0.2-3.4). Three (30%) had a PFS ratio ≥1.3, and three (30%) had a PFS ratio ≥0.8 and <1.3. In this small cohort of NSCLC, 30% demonstrated longer PFS with POG-informed therapies. Larger studies will help clarify the role of whole-genome analysis in clinical practice., (© 2019 Tsang et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2019
- Full Text
- View/download PDF
49. Temporal Dynamics of Genomic Alterations in a BRCA1 Germline-Mutated Pancreatic Cancer With Low Genomic Instability Burden but Exceptional Response to Fluorouracil, Oxaliplatin, Leucovorin, and Irinotecan.
- Author
-
Wong HL, Zhao EY, Jones MR, Reisle CR, Eirew P, Pleasance E, Grande BM, Karasinska JM, Kalloger SE, Lim HJ, Shen Y, Yip S, Morin RD, Laskin J, Marra MA, Jones SJM, Schrader KA, Schaeffer DF, and Renouf DJ
- Abstract
Competing Interests: The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center. Hui-li WongResearch Funding: Roche Australia (Inst)Eric Y. ZhaoNo relationship to discloseMartin R. JonesNo relationship to discloseCaralyn R. ReisleNo relationship to disclosePeter EirewNo relationship to discloseErin PleasanceNo relationship to discloseBruno M. GrandeNo relationship to discloseJoanna M. KarasinskaNo relationship to discloseSteve E. KallogerNo relationship to discloseHoward J. LimHonoraria: LEO Pharma, Eli Lilly, ImClone Systems, Ipsen, Merck, Roche Canada, Eisai, Taiho PharmaceuticalYaoqing ShenNo relationship to discloseStephen YipNo relationship to discloseRyan D. MorinConsulting or Advisory Role: EpizymeJanessa LaskinHonoraria: Boehringer Ingelheim, Roche Canada, AstraZeneca, Pfizer Research Funding: AstraZeneca (Inst), Roche Canada (Inst)Marco A. MarraNo relationship to discloseSteven J.M. JonesNo relationship to discloseKasmintan A. SchraderConsulting or Advisory Role: Eli Lilly (Inst) Research Funding: Eli Lilly (Inst)David F. SchaefferNo relationship to discloseDaniel J. RenoufHonoraria: Celgene, Shire Consulting or Advisory Role: Celgene, Shire Research Funding: Bayer AG (Inst), AstraZeneca (Inst)
- Published
- 2018
- Full Text
- View/download PDF
50. Personalized oncogenomic analysis of metastatic adenoid cystic carcinoma: using whole-genome sequencing to inform clinical decision-making.
- Author
-
Chahal M, Pleasance E, Grewal J, Zhao E, Ng T, Chapman E, Jones MR, Shen Y, Mungall KL, Bonakdar M, Taylor GA, Ma Y, Mungall AJ, Moore RA, Lim H, Renouf D, Yip S, Jones SJM, Marra MA, and Laskin J
- Subjects
- Adult, Biomarkers, Carcinoma, Adenoid Cystic diagnosis, Carcinoma, Adenoid Cystic mortality, Clinical Decision-Making, Female, Gene Expression, Humans, Immunohistochemistry, Male, Middle Aged, Mutation, Neoplasm Staging, Oncogene Proteins, Fusion, Precision Medicine methods, Prognosis, Biomarkers, Tumor, Carcinoma, Adenoid Cystic genetics, Genome, Human, Genomics methods, Oncogenes
- Abstract
Metastatic adenoid cystic carcinomas (ACCs) can cause significant morbidity and mortality. Because of their slow growth and relative rarity, there is limited evidence for systemic therapy regimens. Recently, molecular profiling studies have begun to reveal the genetic landscape of these poorly understood cancers, and new treatment possibilities are beginning to emerge. The objective is to use whole-genome and transcriptome sequencing and analysis to better understand the genetic alterations underlying the pathology of metastatic and rare ACCs and determine potentially actionable therapeutic targets. We report five cases of metastatic ACC, not originating in the salivary glands, in patients enrolled in the Personalized Oncogenomics (POG) Program at the BC Cancer Agency. Genomic workup included whole-genome and transcriptome sequencing, detailed analysis of tumor alterations, and integration with existing knowledge of drug-target combinations to identify potential therapeutic targets. Analysis reveals low mutational burden in these five ACC cases, and mutation signatures that are commonly observed in multiple cancer types. Notably, the only recurrent structural aberration identified was the well-described MYB-NFIB fusion that was present in four of five cases, and one case exhibited a closely related MYBL1-NFIB fusion. Recurrent mutations were also identified in BAP1 and BCOR, with additional mutations in individual samples affecting NOTCH1 and the epigenetic regulators ARID2, SMARCA2, and SMARCB1. Copy changes were rare, and they included amplification of MYC and homozygous loss of CDKN2A in individual samples. Genomic analysis revealed therapeutic targets in all five cases and served to inform a therapeutic choice in three of the cases to date., (© 2018 Chahal et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.