64 results on '"Schumacher SE"'
Search Results
2. Characterizing genetic intra-tumor heterogeneity across 2,658 human cancer genomes
- Author
-
Dentro, SC, Leshchiner, I, Haase, K, Tarabichi, M, Wintersinger, J, Deshwar, AG, Yu, K, Rubanova, Y, Macintyre, G, Demeulemeester, J, Vazquez-Garcia, I, Kleinheinz, K, Livitz, DG, Malikic, S, Donmez, N, Sengupta, S, Anur, P, Jolly, C, Cmero, M, Rosebrock, D, Schumacher, SE, Fan, Y, Fittall, M, Drews, RM, Yao, X, Watkins, TBK, Lee, J, Schlesner, M, Zhu, H, Adams, DJ, McGranahan, N, Swanton, C, Getz, G, Boutros, PC, Imielinski, M, Beroukhim, R, Sahinalp, SC, Ji, Y, Peifer, M, Martincorena, I, Markowetz, F, Mustonen, V, Yuan, K, Gerstung, M, Spellman, PT, Wang, W, Morris, QD, Wedge, DC, Van Loo, P, Dentro, SC, Leshchiner, I, Haase, K, Tarabichi, M, Wintersinger, J, Deshwar, AG, Yu, K, Rubanova, Y, Macintyre, G, Demeulemeester, J, Vazquez-Garcia, I, Kleinheinz, K, Livitz, DG, Malikic, S, Donmez, N, Sengupta, S, Anur, P, Jolly, C, Cmero, M, Rosebrock, D, Schumacher, SE, Fan, Y, Fittall, M, Drews, RM, Yao, X, Watkins, TBK, Lee, J, Schlesner, M, Zhu, H, Adams, DJ, McGranahan, N, Swanton, C, Getz, G, Boutros, PC, Imielinski, M, Beroukhim, R, Sahinalp, SC, Ji, Y, Peifer, M, Martincorena, I, Markowetz, F, Mustonen, V, Yuan, K, Gerstung, M, Spellman, PT, Wang, W, Morris, QD, Wedge, DC, and Van Loo, P
- Abstract
Intra-tumor heterogeneity (ITH) is a mechanism of therapeutic resistance and therefore an important clinical challenge. However, the extent, origin, and drivers of ITH across cancer types are poorly understood. To address this, we extensively characterize ITH across whole-genome sequences of 2,658 cancer samples spanning 38 cancer types. Nearly all informative samples (95.1%) contain evidence of distinct subclonal expansions with frequent branching relationships between subclones. We observe positive selection of subclonal driver mutations across most cancer types and identify cancer type-specific subclonal patterns of driver gene mutations, fusions, structural variants, and copy number alterations as well as dynamic changes in mutational processes between subclonal expansions. Our results underline the importance of ITH and its drivers in tumor evolution and provide a pan-cancer resource of comprehensively annotated subclonal events from whole-genome sequencing data.
- Published
- 2021
3. Comprehensive analysis of chromothripsis in 2,658 human cancers using whole-genome sequencing
- Author
-
Cortes-Ciriano, I, Lee, JJ-K, Xi, R, Jain, D, Jung, YL, Yang, L, Gordenin, D, Klimczak, LJ, Zhang, C-Z, Pellman, DS, Park, PJ, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Beroukhim, R, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Campbell, PJ, Chan, K, Chen, K, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gerstein, M, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Ju, YS, Kazanov, MD, Koh, Y, Korbel, JO, Kumar, K, Lee, EA, Li, Y, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Pearson, J, Puiggros, M, Rippe, K, Roberts, ND, Roberts, SA, Rodriguez-Martin, B, Schumacher, SE, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Wala, JA, Weischenfeldt, J, Yao, X, Yoon, S-S, Zamora, J, Alsop, K, Christie, EL, Fereday, S, Mileshkin, L, Mitchell, C, Thorne, H, Traficante, N, Cmero, M, Cowin, PA, Hamilton, A, Mir Arnau, G, Vedururu, R, Grimmond, SM, Hofmann, O, Morrison, C, Oien, KA, Pairojkul, C, Waring, PM, van de Vijver, MJ, Behren, A, Cortes-Ciriano, I, Lee, JJ-K, Xi, R, Jain, D, Jung, YL, Yang, L, Gordenin, D, Klimczak, LJ, Zhang, C-Z, Pellman, DS, Park, PJ, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Beroukhim, R, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Campbell, PJ, Chan, K, Chen, K, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gerstein, M, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Ju, YS, Kazanov, MD, Koh, Y, Korbel, JO, Kumar, K, Lee, EA, Li, Y, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Pearson, J, Puiggros, M, Rippe, K, Roberts, ND, Roberts, SA, Rodriguez-Martin, B, Schumacher, SE, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Wala, JA, Weischenfeldt, J, Yao, X, Yoon, S-S, Zamora, J, Alsop, K, Christie, EL, Fereday, S, Mileshkin, L, Mitchell, C, Thorne, H, Traficante, N, Cmero, M, Cowin, PA, Hamilton, A, Mir Arnau, G, Vedururu, R, Grimmond, SM, Hofmann, O, Morrison, C, Oien, KA, Pairojkul, C, Waring, PM, van de Vijver, MJ, and Behren, A
- Abstract
Chromothripsis is a mutational phenomenon characterized by massive, clustered genomic rearrangements that occurs in cancer and other diseases. Recent studies in selected cancer types have suggested that chromothripsis may be more common than initially inferred from low-resolution copy-number data. Here, as part of the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA), we analyze patterns of chromothripsis across 2,658 tumors from 38 cancer types using whole-genome sequencing data. We find that chromothripsis events are pervasive across cancers, with a frequency of more than 50% in several cancer types. Whereas canonical chromothripsis profiles display oscillations between two copy-number states, a considerable fraction of events involve multiple chromosomes and additional structural alterations. In addition to non-homologous end joining, we detect signatures of replication-associated processes and templated insertions. Chromothripsis contributes to oncogene amplification and to inactivation of genes such as mismatch-repair-related genes. These findings show that chromothripsis is a major process that drives genome evolution in human cancer.
- Published
- 2020
4. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes
- Author
-
Rheinbay, E, Nielsen, MM, Abascal, F, Wala, JA, Shapira, O, Tiao, G, Hornshoj, H, Hess, JM, Juul, RI, Lin, Z, Feuerbach, L, Sabarinathan, R, Madsen, T, Kim, J, Mularoni, L, Shuai, S, Lanzos, A, Herrmann, C, Maruvka, YE, Shen, C, Amin, SB, Bandopadhayay, P, Bertl, J, Boroevich, KA, Busanovich, J, Carlevaro-Fita, J, Chakravarty, D, Chan, CWY, Craft, D, Dhingra, P, Diamanti, K, Fonseca, NA, Gonzalez-Perez, A, Guo, Q, Hamilton, MP, Haradhvala, NJ, Hong, C, Isaev, K, Johnson, TA, Juul, M, Kahles, A, Kahraman, A, Kim, Y, Komorowski, J, Kumar, K, Kumar, S, Lee, D, Lehmann, K-V, Li, Y, Liu, EM, Lochovsky, L, Park, K, Pich, O, Roberts, ND, Saksena, G, Schumacher, SE, Sidiropoulos, N, Sieverling, L, Sinnott-Armstrong, N, Stewart, C, Tamborero, D, Tubio, JMC, Umer, HM, Uuskula-Reimand, L, Wadelius, C, Wadi, L, Yao, X, Zhang, C-Z, Zhang, J, Haber, JE, Hobolth, A, Imielinski, M, Kellis, M, Lawrence, MS, von Mering, C, Nakagawa, H, Raphael, BJ, Rubin, MA, Sander, C, Stein, LD, Stuart, JM, Tsunoda, T, Wheeler, DA, Johnson, R, Reimand, J, Gerstein, M, Khurana, E, Campbell, PJ, Lopez-Bigas, N, Weischenfeldt, J, Beroukhim, R, Martincorena, I, Pedersen, JS, Getz, G, Bader, GD, Barenboim, J, Brunak, S, Chen, K, Choi, JK, Deu-Pons, J, Fink, JL, Frigola, J, Gambacorti-Passerini, C, Garsed, DW, Gut, IG, Haan, D, Harmanci, AO, Helmy, M, Hodzic, E, Izarzugaza, JMG, Kim, JK, Korbel, JO, Larsson, E, Li, S, Li, X, Lou, S, Marchal, K, Martinez-Fundichely, A, McGillivray, PD, Meyerson, W, Muinos, F, Paczkowska, M, Pons, T, Pulido-Tamayo, S, Reyes-Salazar, I, Reyna, MA, Rubio-Perez, C, Sahinalp, SC, Salichos, L, Shackleton, M, Shrestha, R, Valencia, A, Vazquez, M, Verbeke, LPC, Wang, J, Warrell, J, Waszak, SM, Wu, G, Yu, J, Zhang, X, Zhang, Y, Zhao, Z, Zou, L, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, CortesCiriano, I, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Frenkel-Morgenstern, M, Gordenin, DA, Hutter, B, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Meyerson, M, Miyano, S, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, J, Puiggros, M, Rippe, K, Roberts, SA, RodriguezMartin, B, Scully, R, Torrents, D, Villasante, I, Waddell, N, Yang, L, Yoon, S-S, Zamora, J, Rheinbay, E, Nielsen, MM, Abascal, F, Wala, JA, Shapira, O, Tiao, G, Hornshoj, H, Hess, JM, Juul, RI, Lin, Z, Feuerbach, L, Sabarinathan, R, Madsen, T, Kim, J, Mularoni, L, Shuai, S, Lanzos, A, Herrmann, C, Maruvka, YE, Shen, C, Amin, SB, Bandopadhayay, P, Bertl, J, Boroevich, KA, Busanovich, J, Carlevaro-Fita, J, Chakravarty, D, Chan, CWY, Craft, D, Dhingra, P, Diamanti, K, Fonseca, NA, Gonzalez-Perez, A, Guo, Q, Hamilton, MP, Haradhvala, NJ, Hong, C, Isaev, K, Johnson, TA, Juul, M, Kahles, A, Kahraman, A, Kim, Y, Komorowski, J, Kumar, K, Kumar, S, Lee, D, Lehmann, K-V, Li, Y, Liu, EM, Lochovsky, L, Park, K, Pich, O, Roberts, ND, Saksena, G, Schumacher, SE, Sidiropoulos, N, Sieverling, L, Sinnott-Armstrong, N, Stewart, C, Tamborero, D, Tubio, JMC, Umer, HM, Uuskula-Reimand, L, Wadelius, C, Wadi, L, Yao, X, Zhang, C-Z, Zhang, J, Haber, JE, Hobolth, A, Imielinski, M, Kellis, M, Lawrence, MS, von Mering, C, Nakagawa, H, Raphael, BJ, Rubin, MA, Sander, C, Stein, LD, Stuart, JM, Tsunoda, T, Wheeler, DA, Johnson, R, Reimand, J, Gerstein, M, Khurana, E, Campbell, PJ, Lopez-Bigas, N, Weischenfeldt, J, Beroukhim, R, Martincorena, I, Pedersen, JS, Getz, G, Bader, GD, Barenboim, J, Brunak, S, Chen, K, Choi, JK, Deu-Pons, J, Fink, JL, Frigola, J, Gambacorti-Passerini, C, Garsed, DW, Gut, IG, Haan, D, Harmanci, AO, Helmy, M, Hodzic, E, Izarzugaza, JMG, Kim, JK, Korbel, JO, Larsson, E, Li, S, Li, X, Lou, S, Marchal, K, Martinez-Fundichely, A, McGillivray, PD, Meyerson, W, Muinos, F, Paczkowska, M, Pons, T, Pulido-Tamayo, S, Reyes-Salazar, I, Reyna, MA, Rubio-Perez, C, Sahinalp, SC, Salichos, L, Shackleton, M, Shrestha, R, Valencia, A, Vazquez, M, Verbeke, LPC, Wang, J, Warrell, J, Waszak, SM, Wu, G, Yu, J, Zhang, X, Zhang, Y, Zhao, Z, Zou, L, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, CortesCiriano, I, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Frenkel-Morgenstern, M, Gordenin, DA, Hutter, B, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Meyerson, M, Miyano, S, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, J, Puiggros, M, Rippe, K, Roberts, SA, RodriguezMartin, B, Scully, R, Torrents, D, Villasante, I, Waddell, N, Yang, L, Yoon, S-S, and Zamora, J
- Abstract
The discovery of drivers of cancer has traditionally focused on protein-coding genes1-4. Here we present analyses of driver point mutations and structural variants in non-coding regions across 2,658 genomes from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium5 of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). For point mutations, we developed a statistically rigorous strategy for combining significance levels from multiple methods of driver discovery that overcomes the limitations of individual methods. For structural variants, we present two methods of driver discovery, and identify regions that are significantly affected by recurrent breakpoints and recurrent somatic juxtapositions. Our analyses confirm previously reported drivers6,7, raise doubts about others and identify novel candidates, including point mutations in the 5' region of TP53, in the 3' untranslated regions of NFKBIZ and TOB1, focal deletions in BRD4 and rearrangements in the loci of AKR1C genes. We show that although point mutations and structural variants that drive cancer are less frequent in non-coding genes and regulatory sequences than in protein-coding genes, additional examples of these drivers will be found as more cancer genomes become available.
- Published
- 2020
5. Cancer LncRNA Census reveals evidence for deep functional conservation of long noncoding RNAs in tumorigenesis
- Author
-
Carlevaro-Fita, J, Lanzos, A, Feuerbach, L, Hong, C, Mas-Ponte, D, Pedersen, JS, Johnson, R, Abascal, F, Amin, SB, Bader, GD, Barenboim, J, Beroukhim, R, Bertl, J, Boroevich, KA, Brunak, S, Campbell, PJ, Chakravarty, D, Chan, CWY, Chen, K, Choi, JK, Deu-Pons, J, Dhingra, P, Diamanti, K, Fink, JL, Fonseca, NA, Frigola, J, Gambacorti-Passerini, C, Garsed, DW, Gerstein, M, Getz, G, Gonzalez-Perez, A, Guo, Q, Gut, IG, Haan, D, Hamilton, MP, Haradhvala, NJ, Harmanci, AO, Helmy, M, Herrmann, C, Hess, JM, Hobolth, A, Hodzic, E, Hornshoj, H, Isaev, K, Izarzugaza, JMG, Johnson, TA, Juul, M, Juul, RI, Kahles, A, Kahraman, A, Kellis, M, Khurana, E, Kim, J, Kim, JK, Kim, Y, Komorowski, J, Korbel, JO, Kumar, S, Larsson, E, Lawrence, MS, Lee, D, Lehmann, K-V, Li, S, Li, X, Lin, Z, Liu, EM, Lochovsky, L, Lou, S, Madsen, T, Marchal, K, Martincorena, I, Martinez-Fundichely, A, Maruvka, YE, McGillivray, PD, Meyerson, W, Muinos, F, Mularoni, L, Nakagawa, H, Nielsen, MM, Paczkowska, M, Park, K, Pich, O, Pons, T, Pulido-Tamayo, S, Raphael, BJ, Reimand, J, Reyes-Salazar, I, Reyna, MA, Rheinbay, E, Rubin, MA, Rubio-Perez, C, Sabarinathan, R, Sahinalp, SC, Saksena, G, Salichos, L, Sander, C, Schumacher, SE, Shackleton, M, Shapira, O, Shen, C, Shrestha, R, Shuai, S, Sidiropoulos, N, Sieverling, L, Sinnott-Armstrong, N, Stein, LD, Stuart, JM, Tamborero, D, Tiao, G, Tsunoda, T, Umer, HM, Uuskula-Reimand, L, Valencia, A, Vazquez, M, Verbeke, LPC, Wadelius, C, Wadi, L, Wang, J, Warrell, J, Waszak, SM, Weischenfeldt, J, Wheeler, DA, Wu, G, Yu, J, Zhang, J, Zhang, X, Zhang, Y, Zhao, Z, Zou, L, von Mering, C, Carlevaro-Fita, J, Lanzos, A, Feuerbach, L, Hong, C, Mas-Ponte, D, Pedersen, JS, Johnson, R, Abascal, F, Amin, SB, Bader, GD, Barenboim, J, Beroukhim, R, Bertl, J, Boroevich, KA, Brunak, S, Campbell, PJ, Chakravarty, D, Chan, CWY, Chen, K, Choi, JK, Deu-Pons, J, Dhingra, P, Diamanti, K, Fink, JL, Fonseca, NA, Frigola, J, Gambacorti-Passerini, C, Garsed, DW, Gerstein, M, Getz, G, Gonzalez-Perez, A, Guo, Q, Gut, IG, Haan, D, Hamilton, MP, Haradhvala, NJ, Harmanci, AO, Helmy, M, Herrmann, C, Hess, JM, Hobolth, A, Hodzic, E, Hornshoj, H, Isaev, K, Izarzugaza, JMG, Johnson, TA, Juul, M, Juul, RI, Kahles, A, Kahraman, A, Kellis, M, Khurana, E, Kim, J, Kim, JK, Kim, Y, Komorowski, J, Korbel, JO, Kumar, S, Larsson, E, Lawrence, MS, Lee, D, Lehmann, K-V, Li, S, Li, X, Lin, Z, Liu, EM, Lochovsky, L, Lou, S, Madsen, T, Marchal, K, Martincorena, I, Martinez-Fundichely, A, Maruvka, YE, McGillivray, PD, Meyerson, W, Muinos, F, Mularoni, L, Nakagawa, H, Nielsen, MM, Paczkowska, M, Park, K, Pich, O, Pons, T, Pulido-Tamayo, S, Raphael, BJ, Reimand, J, Reyes-Salazar, I, Reyna, MA, Rheinbay, E, Rubin, MA, Rubio-Perez, C, Sabarinathan, R, Sahinalp, SC, Saksena, G, Salichos, L, Sander, C, Schumacher, SE, Shackleton, M, Shapira, O, Shen, C, Shrestha, R, Shuai, S, Sidiropoulos, N, Sieverling, L, Sinnott-Armstrong, N, Stein, LD, Stuart, JM, Tamborero, D, Tiao, G, Tsunoda, T, Umer, HM, Uuskula-Reimand, L, Valencia, A, Vazquez, M, Verbeke, LPC, Wadelius, C, Wadi, L, Wang, J, Warrell, J, Waszak, SM, Weischenfeldt, J, Wheeler, DA, Wu, G, Yu, J, Zhang, J, Zhang, X, Zhang, Y, Zhao, Z, Zou, L, and von Mering, C
- Abstract
Long non-coding RNAs (lncRNAs) are a growing focus of cancer genomics studies, creating the need for a resource of lncRNAs with validated cancer roles. Furthermore, it remains debated whether mutated lncRNAs can drive tumorigenesis, and whether such functions could be conserved during evolution. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, we introduce the Cancer LncRNA Census (CLC), a compilation of 122 GENCODE lncRNAs with causal roles in cancer phenotypes. In contrast to existing databases, CLC requires strong functional or genetic evidence. CLC genes are enriched amongst driver genes predicted from somatic mutations, and display characteristic genomic features. Strikingly, CLC genes are enriched for driver mutations from unbiased, genome-wide transposon-mutagenesis screens in mice. We identified 10 tumour-causing mutations in orthologues of 8 lncRNAs, including LINC-PINT and NEAT1, but not MALAT1. Thus CLC represents a dataset of high-confidence cancer lncRNAs. Mutagenesis maps are a novel means for identifying deeply-conserved roles of lncRNAs in tumorigenesis.
- Published
- 2020
6. Disruption of chromatin folding domains by somatic genomic rearrangements in human cancer
- Author
-
Akdemir, KC, Le, VT, Chandran, S, Li, Y, Verhaak, RG, Beroukhim, R, Campbell, PJ, Chin, L, Dixon, JR, Futreal, PA, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, Chen, K, Cortes-Ciriano, I, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gerstein, M, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Korbel, JO, Kumar, K, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, JV, Puiggros, M, Rippe, K, Roberts, ND, Roberts, SA, Rodriguez-Martin, B, Schumacher, SE, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Wala, JA, Weischenfeldt, J, Yang, L, Yao, X, Yoon, S-S, Zamora, J, Zhang, C-Z, Akdemir, KC, Le, VT, Chandran, S, Li, Y, Verhaak, RG, Beroukhim, R, Campbell, PJ, Chin, L, Dixon, JR, Futreal, PA, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, Chen, K, Cortes-Ciriano, I, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gerstein, M, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Korbel, JO, Kumar, K, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, JV, Puiggros, M, Rippe, K, Roberts, ND, Roberts, SA, Rodriguez-Martin, B, Schumacher, SE, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Wala, JA, Weischenfeldt, J, Yang, L, Yao, X, Yoon, S-S, Zamora, J, and Zhang, C-Z
- Abstract
Chromatin is folded into successive layers to organize linear DNA. Genes within the same topologically associating domains (TADs) demonstrate similar expression and histone-modification profiles, and boundaries separating different domains have important roles in reinforcing the stability of these features. Indeed, domain disruptions in human cancers can lead to misregulation of gene expression. However, the frequency of domain disruptions in human cancers remains unclear. Here, as part of the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA), which aggregated whole-genome sequencing data from 2,658 cancers across 38 tumor types, we analyzed 288,457 somatic structural variations (SVs) to understand the distributions and effects of SVs across TADs. Notably, SVs can lead to the fusion of discrete TADs, and complex rearrangements markedly change chromatin folding maps in the cancer genomes. Notably, only 14% of the boundary deletions resulted in a change in expression in nearby genes of more than twofold.
- Published
- 2020
7. Patterns of somatic structural variation in human cancer genomes
- Author
-
Li, Y, Roberts, ND, Wala, JA, Shapira, O, Schumacher, SE, Kumar, K, Khurana, E, Waszak, S, Korbel, JO, Haber, JE, Imielinski, M, Weischenfeldt, J, Beroukhim, R, Campbell, PJ, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, Chen, K, Cortes-Ciriano, I, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gerstein, M, Gordenin, DA, Haan, D, Hess, JM, Hutter, B, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, J, Puiggros, M, Rippe, K, Roberts, SA, Rodriguez-Martin, B, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Yang, L, Yao, X, Yoon, S-S, Zamora, J, Zhang, C-Z, Li, Y, Roberts, ND, Wala, JA, Shapira, O, Schumacher, SE, Kumar, K, Khurana, E, Waszak, S, Korbel, JO, Haber, JE, Imielinski, M, Weischenfeldt, J, Beroukhim, R, Campbell, PJ, Akdemir, KC, Alvarez, EG, Baez-Ortega, A, Boutros, PC, Bowtell, DDL, Brors, B, Burns, KH, Chan, K, Chen, K, Cortes-Ciriano, I, Dueso-Barroso, A, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gerstein, M, Gordenin, DA, Haan, D, Hess, JM, Hutter, B, Jones, DTW, Ju, YS, Kazanov, MD, Klimczak, LJ, Koh, Y, Lee, EA, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martincorena, I, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Navarro, FCP, Ossowski, S, Park, PJ, Pearson, J, Puiggros, M, Rippe, K, Roberts, SA, Rodriguez-Martin, B, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Torrents, D, Tubio, JMC, Villasante, I, Waddell, N, Yang, L, Yao, X, Yoon, S-S, Zamora, J, and Zhang, C-Z
- Abstract
A key mutational process in cancer is structural variation, in which rearrangements delete, amplify or reorder genomic segments that range in size from kilobases to whole chromosomes1-7. Here we develop methods to group, classify and describe somatic structural variants, using data from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA), which aggregated whole-genome sequencing data from 2,658 cancers across 38 tumour types8. Sixteen signatures of structural variation emerged. Deletions have a multimodal size distribution, assort unevenly across tumour types and patients, are enriched in late-replicating regions and correlate with inversions. Tandem duplications also have a multimodal size distribution, but are enriched in early-replicating regions-as are unbalanced translocations. Replication-based mechanisms of rearrangement generate varied chromosomal structures with low-level copy-number gains and frequent inverted rearrangements. One prominent structure consists of 2-7 templates copied from distinct regions of the genome strung together within one locus. Such cycles of templated insertions correlate with tandem duplications, and-in liver cancer-frequently activate the telomerase gene TERT. A wide variety of rearrangement processes are active in cancer, which generate complex configurations of the genome upon which selection can act.
- Published
- 2020
8. Pan-cancer analysis of whole genomes identifies driver rearrangements promoted by LINE-1 retrotransposition
- Author
-
Rodriguez-Martin, B, Alvarez, EG, Baez-Ortega, A, Zamora, J, Supek, F, Demeulemeester, J, Santamarina, M, Ju, YS, Temes, J, Garcia-Souto, D, Detering, H, Li, Y, Rodriguez-Castro, J, Dueso-Barroso, A, Bruzos, AL, Dentro, SC, Blanco, MG, Contino, G, Ardeljan, D, Tojo, M, Roberts, ND, Zumalave, S, Edwards, PAW, Weischenfeldt, J, Puiggros, M, Chong, Z, Chen, K, Lee, EA, Wala, JA, Raine, K, Butler, A, Waszak, SM, Navarro, FCP, Schumacher, SE, Monlong, J, Maura, F, Bolli, N, Bourque, G, Gerstein, M, Park, PJ, Wedge, DC, Beroukhim, R, Torrents, D, Korbel, JO, Martincorena, I, Fitzgerald, RC, Van Loo, P, Kazazian, HH, Burns, KH, Campbell, PJ, Tubio, JMC, Akdemir, KC, Boutros, PC, Bowtell, DDL, Brors, B, Chan, K, Cortes-Ciriano, I, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Kazanov, MD, Klimczak, LJ, Koh, Y, Kumar, K, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Ossowski, S, Pearson, J, Rippe, K, Roberts, SA, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Villasante, I, Waddell, N, Yang, L, Yao, X, Yoon, S-S, Zhang, C-Z, Rodriguez-Martin, B, Alvarez, EG, Baez-Ortega, A, Zamora, J, Supek, F, Demeulemeester, J, Santamarina, M, Ju, YS, Temes, J, Garcia-Souto, D, Detering, H, Li, Y, Rodriguez-Castro, J, Dueso-Barroso, A, Bruzos, AL, Dentro, SC, Blanco, MG, Contino, G, Ardeljan, D, Tojo, M, Roberts, ND, Zumalave, S, Edwards, PAW, Weischenfeldt, J, Puiggros, M, Chong, Z, Chen, K, Lee, EA, Wala, JA, Raine, K, Butler, A, Waszak, SM, Navarro, FCP, Schumacher, SE, Monlong, J, Maura, F, Bolli, N, Bourque, G, Gerstein, M, Park, PJ, Wedge, DC, Beroukhim, R, Torrents, D, Korbel, JO, Martincorena, I, Fitzgerald, RC, Van Loo, P, Kazazian, HH, Burns, KH, Campbell, PJ, Tubio, JMC, Akdemir, KC, Boutros, PC, Bowtell, DDL, Brors, B, Chan, K, Cortes-Ciriano, I, Dunford, AJ, Edwards, PA, Estivill, X, Etemadmoghadam, D, Feuerbach, L, Fink, JL, Frenkel-Morgenstern, M, Garsed, DW, Gordenin, DA, Haan, D, Haber, JE, Hess, JM, Hutter, B, Imielinski, M, Jones, DTW, Kazanov, MD, Klimczak, LJ, Koh, Y, Kumar, K, Lee, JJ-K, Lynch, AG, Macintyre, G, Markowetz, F, Martinez-Fundichely, A, Meyerson, M, Miyano, S, Nakagawa, H, Ossowski, S, Pearson, J, Rippe, K, Roberts, SA, Scully, R, Shackleton, M, Sidiropoulos, N, Sieverling, L, Stewart, C, Villasante, I, Waddell, N, Yang, L, Yao, X, Yoon, S-S, and Zhang, C-Z
- Abstract
About half of all cancers have somatic integrations of retrotransposons. Here, to characterize their role in oncogenesis, we analyzed the patterns and mechanisms of somatic retrotransposition in 2,954 cancer genomes from 38 histological cancer subtypes within the framework of the Pan-Cancer Analysis of Whole Genomes (PCAWG) project. We identified 19,166 somatically acquired retrotransposition events, which affected 35% of samples and spanned a range of event types. Long interspersed nuclear element (LINE-1; L1 hereafter) insertions emerged as the first most frequent type of somatic structural variation in esophageal adenocarcinoma, and the second most frequent in head-and-neck and colorectal cancers. Aberrant L1 integrations can delete megabase-scale regions of a chromosome, which sometimes leads to the removal of tumor-suppressor genes, and can induce complex translocations and large-scale duplications. Somatic retrotranspositions can also initiate breakage-fusion-bridge cycles, leading to high-level amplification of oncogenes. These observations illuminate a relevant role of L1 retrotransposition in remodeling the cancer genome, with potential implications for the development of human tumors.
- Published
- 2020
9. Comprehensive and Integrated Genomic Characterization of Adult Soft Tissue Sarcomas
- Author
-
Lazar, AJ, McLellan, MD, Bailey, MH, Miller, CA, Appelbaum, EL, Cordes, MG, Fronick, CC, Fulton, LA, Fulton, RS, Mardis, ER, Schmidt, HK, Wong, W, Wilson, RK, Yellapantula, V, Radenbaugh, AJ, Hoadley, KA, Hayes, DN, Parker, JS, Wilkerson, MD, Auman, JT, Balu, S, Bodenheimer, T, Hoyle, AP, Jefferys, SR, Jones, CD, Lehmann, K-V, Meng, S, Mieczkowski, PA, Mose, LE, Perou, CM, Roach, J, Senbabaoglu, Y, Shi, Y, Simons, JV, Skelly, T, Soloway, MG, Tan, D, Veluvolu, U, Davis, IJ, Hepperla, AJ, Brohl, AS, Kasaian, K, Mungall, K, Sadeghi, S, Barthel, FP, Verhaak, R, Hu, X, Chibon, F, Cherniack, AD, Shih, J, Beroukhim, R, Meyerson, M, Cibulskis, C, Gabriel, SB, Saksena, G, Schumacher, SE, Gao, Q, Wyczalkowski, M, Bowlby, R, Robertson, AG, Ally, A, Balasundaram, M, Brooks, D, Carlsen, R, Chuah, E, Dhalla, N, Holt, RA, Jones, SJM, Lee, D, Li, I, Ma, Y, Marra, MA, Mayo, M, Moore, RA, Mungall, AJ, Schein, JE, Sipahimalani, P, Tam, A, Thiessen, N, Wong, T, Danilova, L, Cope, L, Baylin, SB, Bootwalla, MS, Lai, PH, Laird, PW, Maglinte, DT, Van Den Berg, DJ, Weisenberger, DJ, Wrangle, J, Drill, E, Shen, R, Iype, L, Reynolds, SM, Shmulevich, I, Yau, C, Armenia, J, Liu, EM, Benz, C, Pastore, A, Sanchez-Vega, F, Schultz, N, Akbani, R, Hegde, AM, Liu, W, Lu, Y, Mills, GB, Weinstein, JN, Roszik, J, Anur, P, Spellman, P, Abeshouse, A, Chen, H-W, Gao, J, Heins, Z, Kundra, R, Larsson, E, Ochoa, A, Sander, C, Socci, N, Zhang, H, Noble, MS, Heiman, DI, Kim, J, Chin, L, Getz, G, Cho, J, Defreitas, T, Frazer, S, Gehlenborg, N, Lawrence, MS, Lin, P, Meier, S, Voet, D, Byers, L, Diao, L, Gay, CM, Wang, J, Newton, Y, Cooper, LAD, Gutman, DA, Lee, S, Nalisnik, M, Bowen, J, Gastier-Foster, JM, Gerken, M, Helsel, C, Hobensack, S, Leraas, KM, Lichtenberg, TM, Ramirez, NC, Wise, L, Zmuda, E, Anderson, ML, Castro, P, Ittmann, M, Gordienko, E, Paklina, O, Setdikova, G, Raut, CP, Karlan, BY, Lester, J, Belyaev, D, Fulidou, V, Potapova, O, Voronina, O, Demetri, GD, Ramalingam, SS, Behera, M, Delman, K, Owonikoko, TK, Sica, GL, Boyd, J, Magliocco, A, Salner, A, Bennett, J, Iacocca, M, Swanson, P, Dottino, P, Kalir, T, Pereira, E, Akeredolu, T, Crain, D, Curley, E, Gardner, J, Mallery, D, Morris, S, Paulauskis, J, Penny, R, Shelton, C, Shelton, T, Thompson, E, Hoon, DB, Parfitt, J, Birrer, M, Karseladze, A, Mariamidze, A, Dao, F, Levine, DA, Olvera, N, Maki, RG, Bartlett, J, Eschbacher, J, Dubina, M, Mozgovoy, E, Fedosenko, K, Manikhas, G, Sekhon, H, Ramirez, N, Ingram, DR, Torres, KE, DiSaia, P, Godwin, AK, Godwin, EM, Kuo, H, Madan, R, Reilly, C, Adebamowo, C, Adebamowo, SN, Bocklage, T, Higgins, K, Martinez, C, Boice, L, Grilley-Olson, JE, Huang, M, Perou, AH, Thorne, LB, Rathmell, WK, Gutmann, DH, Singer, S, Chudamani, S, Liu, J, Lolla, L, Naresh, R, Pihl, T, Sun, Q, Wan, Y, Wu, Y, Felau, I, Zenklusen, JC, Demchok, JA, Ferguson, ML, Hutter, CM, Sofia, HJ, Tarnuzzer, R, Wang, Z, Yang, L, Zhang, JJ, Demicco, EG, Doyle, LA, Hornick, JL, Rubin, BP, de Rijn, MV, Baker, L, Riedel, RF, Ding, L, Ladanyi, M, Novak, JE, Van Tine, BA, Davis, LE, Grilley-Olsen, JE, Pollock, RE, Jones, KB, Martignetti, JA, Tong, P, and Network, CGAR
- Subjects
0301 basic medicine ,Leiomyosarcoma ,Adult ,Epigenomics ,DNA Copy Number Variations ,Genomics ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Undifferentiated Pleomorphic Sarcoma ,Article ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,medicine ,Cluster Analysis ,Humans ,ATRX ,Aged ,Comparative genomics ,Aged, 80 and over ,Genome, Human ,Sarcoma ,Middle Aged ,medicine.disease ,Synovial sarcoma ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,DNA methylation ,Mutation ,Cancer research ,Genome-Wide Association Study - Abstract
Summary Sarcomas are a broad family of mesenchymal malignancies exhibiting remarkable histologic diversity. We describe the multi-platform molecular landscape of 206 adult soft tissue sarcomas representing 6 major types. Along with novel insights into the biology of individual sarcoma types, we report three overarching findings: (1) unlike most epithelial malignancies, these sarcomas (excepting synovial sarcoma) are characterized predominantly by copy-number changes, with low mutational loads and only a few genes ( TP53 , ATRX , RB1 ) highly recurrently mutated across sarcoma types; (2) within sarcoma types, genomic and regulomic diversity of driver pathways defines molecular subtypes associated with patient outcome; and (3) the immune microenvironment, inferred from DNA methylation and mRNA profiles, associates with outcome and may inform clinical trials of immune checkpoint inhibitors. Overall, this large-scale analysis reveals previously unappreciated sarcoma-type-specific changes in copy number, methylation, RNA, and protein, providing insights into refining sarcoma therapy and relationships to other cancer types.
- Published
- 2017
- Full Text
- View/download PDF
10. Integrated genomic characterization of oesophageal carcinoma
- Author
-
Kim, J, Bowlby, R, Mungall, AJ, Robertson, AG, Odze, RD, Cherniack, AD, Shih, J, Pedamallu, CS, Cibulskis, C, Dunford, A, Meier, SR, Raphael, BJ, Wu, H-T, Wong, AM, Willis, JE, Bass, AJ, Derks, S, Garman, K, McCall, SJ, Wiznerowicz, M, Pantazi, A, Parfenov, M, Thorsson, V, Shmulevich, I, Dhankani, V, Miller, M, Sakai, R, Wang, K, Schultz, N, Shen, R, Arora, A, Weinhold, N, Sanchez-Vega, F, Kelsen, DP, Zhang, J, Felau, I, Demchok, J, Rabkin, CS, Camargo, MC, Zenklusen, JC, Bowen, J, Leraas, K, Lichtenberg, TM, Curtis, C, Seoane, JA, Ojesina, AI, Beer, DG, Gulley, ML, Pennathur, A, Luketich, JD, Zhou, Z, Weisenberger, DJ, Akbani, R, Lee, J-S, Liu, W, Mills, GB, Zhang, W, Reid, BJ, Hinoue, T, Laird, PW, Shen, H, Piazuelo, MB, Schneider, BG, McLellan, M, Taylor-Weiner, A, Lawrence, M, Cibulskis, K, Stewart, C, Getz, G, Lander, E, Gabriel, SB, Ding, L, McLellan, MD, Miller, CA, Appelbaum, EL, Cordes, MG, Fronick, CC, Fulton, LA, Mardis, ER, Wilson, RK, Schmidt, HK, Fulton, RS, Ally, A, Balasundaram, M, Carlsen, R, Chuah, E, Dhalla, N, Holt, RA, Jones, SJM, Kasaian, K, Brooks, D, Li, HI, Ma, Y, Marra, MA, Mayo, M, Moore, RA, Mungall, KL, Schein, JE, Sipahimalani, P, Tam, A, Thiessen, N, Wong, T, Beroukhim, R, Bullman, S, Murray, BA, Saksena, G, Schumacher, SE, Gabriel, S, Meyerson, M, Hadjipanayis, A, Kucherlapati, R, Ren, X, Park, PJ, Lee, S, Kucherlapati, M, Yang, L, Baylin, SB, Hoadley, KA, Bootwalla, MS, Lai, PH, Van den Berg, DJ, Berrios, M, Holbrook, A, Hwang, J-E, Jang, H-J, Weinstein, JN, Lu, Y, Sohn, BH, Mills, G, Seth, S, Protopopov, A, Bristow, CA, Mahadeshwar, HS, Tang, J, Song, X, Cho, J, Defrietas, T, Frazer, S, Gehlenborg, N, Heiman, DI, Lawrence, MS, Lin, P, Noble, MS, Doug, V, Zhang, H, Polak, P, Chin, L, Bernard, B, Iype, L, Reynolds, SM, Abeshouse, A, Armenia, J, Kundra, R, Ladanyi, M, Kjong-Van, L, Gao, J, Sander, C, Chakravarty, D, Radenbaugh, A, Hegde, A, Penny, R, Crain, D, Gardner, J, Curley, E, Mallery, D, Morris, S, Paulauskis, J, Shelton, T, Shelton, C, Frick, J, Gastier-Foster, JM, Gerken, M, Leraas, KM, Ramirez, NC, Wise, L, Zmuda, E, Tarvin, K, Saller, C, Park, YS, Button, M, Carvalho, AL, Reis, RM, Matsushita, MM, Lucchesi, F, de Oliveira, AT, Le, X, Paklina, O, Setdikova, G, Lee, J-H, Bennett, J, Iacocca, M, Huelsenbeck-Dill, L, Potapova, CO, Voronina, O, Liu, O, Fulidou, V, Cates, C, Sharp, A, Behera, M, Force, S, Khuri, F, Owonikoko, T, Pickens, A, Ramalingam, S, Sica, G, Dinjens, W, van Nistelrooij, A, Wijnhoven, B, Sandusky, G, Stepa, S, Juhl, IH, Zornig, C, Kwon, SY, Kelsen, D, Kim, GHK, Bartlett, J, Parfitt, J, Chetty, R, Darling, G, Knox, J, Wong, R, El-Zimaity, H, Liu, G, Boussioutas, A, Park, DY, Kemp, R, Carlotti, CG, da Cunha Tirapelli, DP, Saggioro, FP, Sankarankutty, AK, Noushmehr, H, dos Santos, JS, Trevisan, FA, Eschbacher, J, Dubina, M, Mozgovoy, E, Carey, F, Chalmers, S, Forgie, I, Godwin, A, Reilly, C, Madan, R, Naima, Z, Ferrer-Torres, D, Rathmell, WK, Dhir, R, Luketich, J, Ajani, JA, Janjigian, Y, Tang, L, Cheong, J-H, Chudamani, S, Liu, J, Lolla, L, Naresh, R, Pihl, T, Sun, Q, Wan, Y, Wu, Y, Demchok, JA, Ferguson, ML, Shaw, KRM, sheth, M, Tarnuzzer, R, Wang, Z, Hutter, CM, Sofia, HJ, Kim, J, Bowlby, R, Mungall, AJ, Robertson, AG, Odze, RD, Cherniack, AD, Shih, J, Pedamallu, CS, Cibulskis, C, Dunford, A, Meier, SR, Raphael, BJ, Wu, H-T, Wong, AM, Willis, JE, Bass, AJ, Derks, S, Garman, K, McCall, SJ, Wiznerowicz, M, Pantazi, A, Parfenov, M, Thorsson, V, Shmulevich, I, Dhankani, V, Miller, M, Sakai, R, Wang, K, Schultz, N, Shen, R, Arora, A, Weinhold, N, Sanchez-Vega, F, Kelsen, DP, Zhang, J, Felau, I, Demchok, J, Rabkin, CS, Camargo, MC, Zenklusen, JC, Bowen, J, Leraas, K, Lichtenberg, TM, Curtis, C, Seoane, JA, Ojesina, AI, Beer, DG, Gulley, ML, Pennathur, A, Luketich, JD, Zhou, Z, Weisenberger, DJ, Akbani, R, Lee, J-S, Liu, W, Mills, GB, Zhang, W, Reid, BJ, Hinoue, T, Laird, PW, Shen, H, Piazuelo, MB, Schneider, BG, McLellan, M, Taylor-Weiner, A, Lawrence, M, Cibulskis, K, Stewart, C, Getz, G, Lander, E, Gabriel, SB, Ding, L, McLellan, MD, Miller, CA, Appelbaum, EL, Cordes, MG, Fronick, CC, Fulton, LA, Mardis, ER, Wilson, RK, Schmidt, HK, Fulton, RS, Ally, A, Balasundaram, M, Carlsen, R, Chuah, E, Dhalla, N, Holt, RA, Jones, SJM, Kasaian, K, Brooks, D, Li, HI, Ma, Y, Marra, MA, Mayo, M, Moore, RA, Mungall, KL, Schein, JE, Sipahimalani, P, Tam, A, Thiessen, N, Wong, T, Beroukhim, R, Bullman, S, Murray, BA, Saksena, G, Schumacher, SE, Gabriel, S, Meyerson, M, Hadjipanayis, A, Kucherlapati, R, Ren, X, Park, PJ, Lee, S, Kucherlapati, M, Yang, L, Baylin, SB, Hoadley, KA, Bootwalla, MS, Lai, PH, Van den Berg, DJ, Berrios, M, Holbrook, A, Hwang, J-E, Jang, H-J, Weinstein, JN, Lu, Y, Sohn, BH, Mills, G, Seth, S, Protopopov, A, Bristow, CA, Mahadeshwar, HS, Tang, J, Song, X, Cho, J, Defrietas, T, Frazer, S, Gehlenborg, N, Heiman, DI, Lawrence, MS, Lin, P, Noble, MS, Doug, V, Zhang, H, Polak, P, Chin, L, Bernard, B, Iype, L, Reynolds, SM, Abeshouse, A, Armenia, J, Kundra, R, Ladanyi, M, Kjong-Van, L, Gao, J, Sander, C, Chakravarty, D, Radenbaugh, A, Hegde, A, Penny, R, Crain, D, Gardner, J, Curley, E, Mallery, D, Morris, S, Paulauskis, J, Shelton, T, Shelton, C, Frick, J, Gastier-Foster, JM, Gerken, M, Leraas, KM, Ramirez, NC, Wise, L, Zmuda, E, Tarvin, K, Saller, C, Park, YS, Button, M, Carvalho, AL, Reis, RM, Matsushita, MM, Lucchesi, F, de Oliveira, AT, Le, X, Paklina, O, Setdikova, G, Lee, J-H, Bennett, J, Iacocca, M, Huelsenbeck-Dill, L, Potapova, CO, Voronina, O, Liu, O, Fulidou, V, Cates, C, Sharp, A, Behera, M, Force, S, Khuri, F, Owonikoko, T, Pickens, A, Ramalingam, S, Sica, G, Dinjens, W, van Nistelrooij, A, Wijnhoven, B, Sandusky, G, Stepa, S, Juhl, IH, Zornig, C, Kwon, SY, Kelsen, D, Kim, GHK, Bartlett, J, Parfitt, J, Chetty, R, Darling, G, Knox, J, Wong, R, El-Zimaity, H, Liu, G, Boussioutas, A, Park, DY, Kemp, R, Carlotti, CG, da Cunha Tirapelli, DP, Saggioro, FP, Sankarankutty, AK, Noushmehr, H, dos Santos, JS, Trevisan, FA, Eschbacher, J, Dubina, M, Mozgovoy, E, Carey, F, Chalmers, S, Forgie, I, Godwin, A, Reilly, C, Madan, R, Naima, Z, Ferrer-Torres, D, Rathmell, WK, Dhir, R, Luketich, J, Ajani, JA, Janjigian, Y, Tang, L, Cheong, J-H, Chudamani, S, Liu, J, Lolla, L, Naresh, R, Pihl, T, Sun, Q, Wan, Y, Wu, Y, Demchok, JA, Ferguson, ML, Shaw, KRM, sheth, M, Tarnuzzer, R, Wang, Z, Hutter, CM, and Sofia, HJ
- Abstract
Oesophageal cancers are prominent worldwide; however, there are few targeted therapies and survival rates for these cancers remain dismal. Here we performed a comprehensive molecular analysis of 164 carcinomas of the oesophagus derived from Western and Eastern populations. Beyond known histopathological and epidemiologic distinctions, molecular features differentiated oesophageal squamous cell carcinomas from oesophageal adenocarcinomas. Oesophageal squamous cell carcinomas resembled squamous carcinomas of other organs more than they did oesophageal adenocarcinomas. Our analyses identified three molecular subclasses of oesophageal squamous cell carcinomas, but none showed evidence for an aetiological role of human papillomavirus. Squamous cell carcinomas showed frequent genomic amplifications of CCND1 and SOX2 and/or TP63, whereas ERBB2, VEGFA and GATA4 and GATA6 were more commonly amplified in adenocarcinomas. Oesophageal adenocarcinomas strongly resembled the chromosomally unstable variant of gastric adenocarcinoma, suggesting that these cancers could be considered a single disease entity. However, some molecular features, including DNA hypermethylation, occurred disproportionally in oesophageal adenocarcinomas. These data provide a framework to facilitate more rational categorization of these tumours and a foundation for new therapies.
- Published
- 2017
11. Integrated genomic characterization of endometrial carcinoma
- Author
-
Getz, G, Gabriel, SB, Cibulskis, K, Lander, E, Sivachenko, A, Sougnez, C, Lawrence, M, Kandoth, C, Dooling, D, Fulton, R, Fulton, L, Kalicki-Veizer, J, McLellan, MD, O'Laughlin, M, Schmidt, H, Wilson, RK, Ye, K, Li, D, Ally, A, Balasundaram, M, Birol, I, Butterfield, YSN, Carlsen, R, Carter, C, Chu, A, Chuah, E, Chun, HJE, Dhalla, N, Guin, R, Hirst, C, Holt, RA, Jones, SJM, Lee, D, Li, HI, Marra, MA, Mayo, M, Moore, RA, Mungall, AJ, Plettner, P, Schein, JE, Sipahimalani, P, Tam, A, Varhol, RJ, Gordon Robertson, A, Cherniack, AD, Pashtan, I, Saksena, G, Onofrio, RC, Schumacher, SE, Tabak, B, Carter, SL, Hernandez, B, Gentry, J, Salvesen, HB, Ardlie, K, Winckler, W, Beroukhim, R, Meyerson, M, Hadjipanayis, A, Lee, S, Mahadeshwar, HS, Park, P, Protopopov, A, Ren, X, Seth, S, Song, X, Tang, J, Xi, R, Yang, L, Dong, Z, Kucherlapati, R, Chin, L, Zhang, J, Todd Auman, J, Balu, S, Bodenheimer, T, Buda, E, Neil Hayes, D, Hoyle, AP, Jefferys, SR, Jones, CD, Meng, S, Mieczkowski, PA, Mose, LE, Parker, JS, and Perou, CM
- Subjects
endocrine system diseases - Abstract
We performed an integrated genomic, transcriptomic and proteomic characterization of 373 endometrial carcinomas using array-and sequencing-based technologies. Uterine serous tumours and ∼25% of high-grade endometrioid tumours had extensive copy number alterations, few DNA methylation changes, low oestrogen receptor/progesterone receptor levels, and frequent TP53 mutations. Most endometrioid tumours had few copy number alterations or TP53 mutations, but frequent mutations in PTEN, CTNNB1, PIK3CA, ARID1A and KRAS and novel mutations in the SWI/SNF chromatin remodelling complex gene ARID5B. A subset of endometrioid tumours that we identified had a markedly increased transversion mutation frequency and newly identified hotspot mutations in POLE. Our results classified endometrial cancers into four categories: POLE ultramutated, microsatellite instability hypermutated, copy-number low, and copy-number high. Uterine serous carcinomas share genomic features with ovarian serous and basal-like breast carcinomas. We demonstrated that the genomic features of endometrial carcinomas permit a reclassification that may affect post-surgical adjuvant treatment for women with aggressive tumours. © 2013 Macmillan Publishers Limited. All rights reserved.
- Published
- 2013
12. Subgroup-specific structural variation across 1,000 medulloblastoma genomes
- Author
-
Northcott, PA, Shih, DJH, Peacock, J, Garzia, L, Sorana Morrissy, A, Zichner, T, Stútz, AM, Korshunov, A, Reimand, J, Schumacher, SE, Beroukhim, R, Ellison, DW, Marshall, CR, Lionel, AC, MacK, S, Dubuc, A, Yao, Y, Ramaswamy, V, Luu, B, Rolider, A, Cavalli, FMG, Wang, X, Remke, M, Wu, X, Chiu, RYB, Chu, A, Chuah, E, Corbett, RD, Hoad, GR, Jackman, SD, Li, Y, Lo, A, Mungall, KL, Ming Nip, K, Qian, JQ, Raymond, AGJ, Thiessen, N, Varhol, RJ, Birol, I, Moore, RA, Mungall, AJ, Holt, R, Kawauchi, D, Roussel, MF, Kool, M, Jones, DTW, Witt, H, Fernandez-L, A, Kenney, AM, Wechsler-Reya, RJ, Dirks, P, Aviv, T, Grajkowska, WA, Perek-Polnik, M, Haberler, CC, Delattre, O, Reynaud, SS, Doz, FF, Pernet-Fattet, SS, Cho, BK, Kim, SK, Wang, KC, Scheurlen, W, Eberhart, CG, Fèvre-Montange, M, Jouvet, A, Pollack, IF, Fan, X, Muraszko, KM, Yancey Gillespie, G, Di Rocco, C, Massimi, L, Michiels, EMC, Kloosterhof, NK, French, PJ, Kros, JM, Olson, JM, Ellenbogen, RG, Zitterbart, K, Kren, L, Thompson, RC, and Cooper, MK
- Subjects
Oncogene Proteins, Fusion ,DNA Copy Number Variations ,Genome, Human ,General Science & Technology ,NF-kappa B ,Genes, myc ,Proteins ,Nerve Tissue Proteins ,Genomics ,Translocation, Genetic ,Transforming Growth Factor beta ,Gene Duplication ,Genomic Structural Variation ,MD Multidisciplinary ,Humans ,Hedgehog Proteins ,RNA, Long Noncoding ,Cerebellar Neoplasms ,Carrier Proteins ,Child ,Medulloblastoma ,Signal Transduction - Abstract
Medulloblastoma, the most common malignant paediatric brain tumour, is currently treated with nonspecific cytotoxic therapies including surgery, whole-brain radiation, and aggressive chemotherapy. As medulloblastoma exhibits marked intertumoural heterogeneity, with at least four distinct molecular variants, previous attempts to identify targets for therapy have been underpowered because of small samples sizes. Here we report somatic copy number aberrations (SCNAs) in 1,087 unique medulloblastomas. SCNAs are common in medulloblastoma, and are predominantly subgroup-enriched. The most common region of focal copy number gain is a tandem duplication of SNCAIP, a gene associated with Parkinson's disease, which is exquisitely restricted to Group 4α. Recurrent translocations of PVT1, including PVT1-MYC and PVT1-NDRG1, that arise through chromothripsis are restricted to Group 3. Numerous targetable SCNAs, including recurrent events targeting TGF-β signalling in Group 3, and NF-κB signalling in Group 4, suggest future avenues for rational, targeted therapy. © 2012 Macmillan Publishers Limited. All rights reserved.
- Published
- 2012
13. Comprehensive molecular characterization of gastric adenocarcinoma
- Author
-
Bass, AJ, Thorsson, V, Shmulevich, I, Reynolds, SM, Miller, M, Bernard, B, Hinoue, T, Laird, PW, Curtis, C, Shen, H, Weisenberger, DJ, Schultz, N, Shen, R, Weinhold, N, Keiser, DP, Bowlby, R, Sipahimalani, P, Cherniack, AD, Getz, G, Liu, Y, Noble, MS, Pedamallu, C, Sougnez, C, Taylor-Weiner, A, Akbani, R, Lee, J-S, Liu, W, Mills, GB, Yang, D, Zhang, W, Pantazi, A, Parfenov, M, Gulley, M, Piazuelo, MB, Schneider, BG, Kim, J, Boussioutas, A, Sheth, M, Demchok, JA, Rabkin, CS, Willis, JE, Ng, S, Garman, K, Beer, DG, Pennathur, A, Raphael, BJ, Wu, H-T, Odze, R, Kim, HK, Bowen, J, Leraas, KM, Lichtenberg, TM, Weaver, L, McLellan, M, Wiznerowicz, M, Sakai, R, Lawrence, MS, Cibulskis, K, Lichtenstein, L, Fisher, S, Gabriel, SB, Lander, ES, Ding, L, Niu, B, Ally, A, Balasundaram, M, Birol, I, Brooks, D, Butterfield, YSN, Carlsen, R, Chu, A, Chu, J, Chuah, E, Chun, H-JE, Clarke, A, Dhalla, N, Guin, R, Holt, RA, Jones, SJM, Kasaian, K, Lee, D, Li, HA, Lim, E, Ma, Y, Marra, MA, Mayo, M, Moore, RA, Mungall, AJ, Mungall, KL, Nip, KM, Robertson, AG, Schein, JE, Tam, A, Thiessen, N, Beroukhim, R, Carter, SL, Cho, J, DiCara, D, Frazer, S, Gehlenborg, N, Heiman, DI, Jung, J, Lin, P, Meyerson, M, Ojesina, AI, Pedamallu, CS, Saksena, G, Schumacher, SE, Stojanov, P, Tabak, B, Voet, D, Rosenberg, M, Zack, TI, Zhang, H, Zou, L, Protopopov, A, Santoso, N, Lee, S, Zhang, J, Mahadeshwar, HS, Tang, J, Ren, X, Seth, S, Yang, L, Xu, AW, Song, X, Xi, R, Bristow, CA, Hadjipanayis, A, Seidman, J, Chin, L, Park, PJ, Kucherlapati, R, Ling, S, Rao, A, Weinstein, JN, Kim, S-B, Lu, Y, Mills, G, Bootwalla, MS, Lai, PH, Triche, T, Van Den Berg, DJ, Baylin, SB, Herman, JG, Murray, BA, Askoy, BA, Ciriello, G, Dresdner, G, Gao, J, Gross, B, Jacobsen, A, Lee, W, Ramirez, R, Sander, C, Senbabaoglu, Y, Sinha, R, Sumer, SO, Sun, Y, Iype, L, Kramer, RW, Kreisberg, R, Rovira, H, Tasman, N, Haussler, D, Stuart, JM, Verhaak, RGW, Leiserson, MDM, Taylor, BS, Black, AD, Carney, JA, Gastier-Foster, JM, Helsel, C, McAllister, C, Ramirez, NC, Tabler, TR, Wise, L, Zmuda, E, Penny, R, Crain, D, Gardner, J, Lau, K, Curely, E, Mallery, D, Morris, S, Paulauskis, J, Shelton, T, Shelton, C, Sherman, M, Benz, C, Lee, J-H, Fedosenko, K, Manikhas, G, Voronina, O, Belyaev, D, Dolzhansky, O, Rathmell, WK, Brzezinski, J, Ibbs, M, Korski, K, Kycler, W, Lazniak, R, Leporowska, E, Mackiewicz, A, Murawa, D, Murawa, P, Spychala, A, Suchorska, WM, Tatka, H, Teresiak, M, Abdel-Misih, R, Bennett, J, Brown, J, Iacocca, M, Rabeno, B, Kwon, S-Y, Kemkes, A, Curley, E, Alexopoulou, I, Engel, J, Bartlett, J, Albert, M, Park, D-Y, Dhir, R, Luketich, J, Landreneau, R, Janjigian, YY, Kelsen, DP, Cho, E, Ladanyi, M, Tang, L, McCall, SJ, Park, YS, Cheong, J-H, Ajani, J, Camargo, MC, Alonso, S, Ayala, B, Jensen, MA, Pihl, T, Raman, R, Walton, J, Wan, Y, Eley, G, Shaw, KRM, Tarnuzzer, R, Wang, Z, Zenklusen, JC, Davidsen, T, Hutter, CM, Sofia, HJ, Burton, R, Chudamani, S, Liu, J, Bass, AJ, Thorsson, V, Shmulevich, I, Reynolds, SM, Miller, M, Bernard, B, Hinoue, T, Laird, PW, Curtis, C, Shen, H, Weisenberger, DJ, Schultz, N, Shen, R, Weinhold, N, Keiser, DP, Bowlby, R, Sipahimalani, P, Cherniack, AD, Getz, G, Liu, Y, Noble, MS, Pedamallu, C, Sougnez, C, Taylor-Weiner, A, Akbani, R, Lee, J-S, Liu, W, Mills, GB, Yang, D, Zhang, W, Pantazi, A, Parfenov, M, Gulley, M, Piazuelo, MB, Schneider, BG, Kim, J, Boussioutas, A, Sheth, M, Demchok, JA, Rabkin, CS, Willis, JE, Ng, S, Garman, K, Beer, DG, Pennathur, A, Raphael, BJ, Wu, H-T, Odze, R, Kim, HK, Bowen, J, Leraas, KM, Lichtenberg, TM, Weaver, L, McLellan, M, Wiznerowicz, M, Sakai, R, Lawrence, MS, Cibulskis, K, Lichtenstein, L, Fisher, S, Gabriel, SB, Lander, ES, Ding, L, Niu, B, Ally, A, Balasundaram, M, Birol, I, Brooks, D, Butterfield, YSN, Carlsen, R, Chu, A, Chu, J, Chuah, E, Chun, H-JE, Clarke, A, Dhalla, N, Guin, R, Holt, RA, Jones, SJM, Kasaian, K, Lee, D, Li, HA, Lim, E, Ma, Y, Marra, MA, Mayo, M, Moore, RA, Mungall, AJ, Mungall, KL, Nip, KM, Robertson, AG, Schein, JE, Tam, A, Thiessen, N, Beroukhim, R, Carter, SL, Cho, J, DiCara, D, Frazer, S, Gehlenborg, N, Heiman, DI, Jung, J, Lin, P, Meyerson, M, Ojesina, AI, Pedamallu, CS, Saksena, G, Schumacher, SE, Stojanov, P, Tabak, B, Voet, D, Rosenberg, M, Zack, TI, Zhang, H, Zou, L, Protopopov, A, Santoso, N, Lee, S, Zhang, J, Mahadeshwar, HS, Tang, J, Ren, X, Seth, S, Yang, L, Xu, AW, Song, X, Xi, R, Bristow, CA, Hadjipanayis, A, Seidman, J, Chin, L, Park, PJ, Kucherlapati, R, Ling, S, Rao, A, Weinstein, JN, Kim, S-B, Lu, Y, Mills, G, Bootwalla, MS, Lai, PH, Triche, T, Van Den Berg, DJ, Baylin, SB, Herman, JG, Murray, BA, Askoy, BA, Ciriello, G, Dresdner, G, Gao, J, Gross, B, Jacobsen, A, Lee, W, Ramirez, R, Sander, C, Senbabaoglu, Y, Sinha, R, Sumer, SO, Sun, Y, Iype, L, Kramer, RW, Kreisberg, R, Rovira, H, Tasman, N, Haussler, D, Stuart, JM, Verhaak, RGW, Leiserson, MDM, Taylor, BS, Black, AD, Carney, JA, Gastier-Foster, JM, Helsel, C, McAllister, C, Ramirez, NC, Tabler, TR, Wise, L, Zmuda, E, Penny, R, Crain, D, Gardner, J, Lau, K, Curely, E, Mallery, D, Morris, S, Paulauskis, J, Shelton, T, Shelton, C, Sherman, M, Benz, C, Lee, J-H, Fedosenko, K, Manikhas, G, Voronina, O, Belyaev, D, Dolzhansky, O, Rathmell, WK, Brzezinski, J, Ibbs, M, Korski, K, Kycler, W, Lazniak, R, Leporowska, E, Mackiewicz, A, Murawa, D, Murawa, P, Spychala, A, Suchorska, WM, Tatka, H, Teresiak, M, Abdel-Misih, R, Bennett, J, Brown, J, Iacocca, M, Rabeno, B, Kwon, S-Y, Kemkes, A, Curley, E, Alexopoulou, I, Engel, J, Bartlett, J, Albert, M, Park, D-Y, Dhir, R, Luketich, J, Landreneau, R, Janjigian, YY, Kelsen, DP, Cho, E, Ladanyi, M, Tang, L, McCall, SJ, Park, YS, Cheong, J-H, Ajani, J, Camargo, MC, Alonso, S, Ayala, B, Jensen, MA, Pihl, T, Raman, R, Walton, J, Wan, Y, Eley, G, Shaw, KRM, Tarnuzzer, R, Wang, Z, Zenklusen, JC, Davidsen, T, Hutter, CM, Sofia, HJ, Burton, R, Chudamani, S, and Liu, J
- Abstract
Gastric cancer is a leading cause of cancer deaths, but analysis of its molecular and clinical characteristics has been complicated by histological and aetiological heterogeneity. Here we describe a comprehensive molecular evaluation of 295 primary gastric adenocarcinomas as part of The Cancer Genome Atlas (TCGA) project. We propose a molecular classification dividing gastric cancer into four subtypes: tumours positive for Epstein-Barr virus, which display recurrent PIK3CA mutations, extreme DNA hypermethylation, and amplification of JAK2, CD274 (also known as PD-L1) and PDCD1LG2 (also known as PD-L2); microsatellite unstable tumours, which show elevated mutation rates, including mutations of genes encoding targetable oncogenic signalling proteins; genomically stable tumours, which are enriched for the diffuse histological variant and mutations of RHOA or fusions involving RHO-family GTPase-activating proteins; and tumours with chromosomal instability, which show marked aneuploidy and focal amplification of receptor tyrosine kinases. Identification of these subtypes provides a roadmap for patient stratification and trials of targeted therapies.
- Published
- 2014
14. Effectiveness of stabilising exercise in pregnancy-associated lumbar-pelvic pain: systematic literature review.
- Author
-
Theermann C, Schumacher SE, and van der Wurff P
- Published
- 2007
15. Subgroup-specific structural variation across 1,000 medulloblastoma genomes
- Author
-
Jenny Q. Qian, Darell D. Bigner, Miklós Garami, Shaun D. Jackman, Wiesława Grajkowska, Nalin Gupta, Johan M. Kros, Poul H. Sorensen, Anna Kenney, Stéphanie Reynaud, Byung Kyu Cho, Ian F. Pollack, Marcel Kool, Steven C. Clifford, Kyu-Chang Wang, Inanc Birol, Tzvi Aviv, Hendrick Witt, Gemma Hoad, Martine F. Roussel, Christine Haberler, Pim J. French, Betty Luu, Cynthia Hawkins, Claudia C. Faria, Richard A. Moore, Karin M. Muraszko, Yuan Yao, Nanne K. Kloosterhof, Rameen Beroukhim, Leos Kren, Erna M.C. Michiels, Jan O. Korbel, Paul A. Northcott, Stefan M. Pfister, Marc Remke, Nina Thiessen, Jennifer A. Chan, Adam M. Fontebasso, Maryam Fouladi, Shin Jung, Richard G. Ellenbogen, Richard Corbett, László Bognár, Yoon Jae Cho, Massimo Zollo, Robert J. Wechsler-Reya, Steven E. Schumacher, Xing Fan, Michael J. Levy, Wolfram Scheurlen, Young Shin Ra, Adrian M. Stütz, William A. Weiss, Simon Bailey, Rajeev Vibhakar, Giuseppe Cinalli, Toshihiro Kumabe, Marco A. Marra, Christian R. Marshall, Eric Bouffet, Luca Massimi, Scott L. Pomeroy, Sarah S. Pernet-Fattet, Andrew J. Mungall, James T. Rutka, G. Yancey Gillespie, Charles G. Eberhart, Peter Hauser, Andy Chu, Jüri Reimand, Xiaochong Wu, Adi Rolider, Xin Wang, Stephen W. Scherer, Reid C. Thompson, Ka Ming Nip, Anne Jouvet, Timothy E. Van Meter, Robert A. Holt, Anthony Raymond, Livia Garzia, Teiji Tominaga, Erwin G. Van Meir, John Peacock, Michael D. Taylor, Achille Iolascon, Roger E. McLendon, Andrey Korshunov, Stephen C. Mack, Nada Jabado, Readman Chiu, Africa Fernandez-L, Eric Chuah, Richard Varhol, Hideo Nakamura, Samer K. Elbabaa, Uri Tabori, Peter B. Dirks, Gary D. Bader, Linda M. Liau, François Doz, Allan Lo, Janet C. Lindsey, Adrian M. Dubuc, Michelle Fèvre-Montange, David T.W. Jones, Carlos Gilberto Carlotti, Ali G. Saad, Steffen Albrecht, Michael K. Cooper, Karen Mungall, Daisuke Kawauchi, A. Sorana Morrissy, Boleslaw Lach, Karel Zitterbart, Joshua B. Rubin, Matthew Meyerson, Florence M.G. Cavalli, Yisu Li, Shenandoah Robinson, Marta Perek-Polnik, Olivier Delattre, David Malkin, Almos Klekner, James M. Olson, Steven J.M. Jones, Thomas Zichner, David W. Ellison, Seung-Ki Kim, Vijay Ramaswamy, Anath C. Lionel, David Shih, Jeffrey R. Leonard, Concezio Di Rocco, Pulmonary Medicine, Pediatrics, Neurology, Pathology, Northcott, Pa, Shih, Dj, Peacock, J, Garzia, L, Morrissy, A, Zichner, T, Stütz, Am, Korshunov, A, Reimand, J, Schumacher, Se, Beroukhim, R, Ellison, Dw, Marshall, Cr, Lionel, Ac, Mack, S, Dubuc, A, Yao, Y, Ramaswamy, V, Luu, B, Rolider, A, Cavalli, Fm, Wang, X, Remke, M, Wu, X, Chiu, Ry, Chu, A, Chuah, E, Corbett, Rd, Hoad, Gr, Jackman, Sd, Li, Y, Lo, A, Mungall, Kl, Nip, Km, Qian, Jq, Raymond, Ag, Thiessen, Nt, Varhol, Rj, Birol, I, Moore, Ra, Mungall, Aj, Holt, R, Kawauchi, D, Roussel, Mf, Kool, M, Jones, Dt, Witt, H, Fernandez L., A, Kenney, Am, Wechsler Reya, Rj, Dirks, P, Aviv, T, Grajkowska, Wa, Perek Polnik, M, Haberler, Cc, Delattre, O, Reynaud, S, Doz, Ff, Pernet Fattet, S, Cho, Bk, Kim, Sk, Wang, Kc, Scheurlen, W, Eberhart, Cg, Fèvre Montange, M, Jouvet, A, Pollack, If, Fan, X, Muraszko, Km, Gillespie, Gy, Di Rocco, C, Massimi, L, Michiels, Em, Kloosterhof, Nk, French, Pj, Kros, Jm, Olson, Jm, Ellenbogen, Rg, Zitterbart, K, Kren, L, Thompson, Rc, Cooper, Mk, Lach, B, Mclendon, Re, Bigner, Dd, Fontebasso, A, Albrecht, S, Jabado, N, Lindsey, Jc, Bailey, S, Gupta, N, Weiss, Wa, Bognár, L, Klekner, A, Van Meter, Te, Kumabe, T, Tominaga, T, Elbabaa, Sk, Leonard, Jr, Rubin, Jb, Liau, Lm, Van Meir, Eg, Fouladi, M, Nakamura, H, Cinalli, G, Garami, M, Hauser, P, Saad, Ag, Iolascon, Achille, Jung, S, Carlotti, Cg, Vibhakar, R, Ra, Y, Robinson, S, Zollo, Massimo, Faria, Cc, Chan, Ja, Levy, Ml, Sorensen, Ph, Meyerson, M, Pomeroy, Sl, Cho, Yj, Bader, Gd, Tabori, U, Hawkins, Ce, Bouffet, E, Scherer, Sw, Rutka, Jt, Malkin, D, Clifford, Sc, Jones, Sj, Korbel, Jo, Pfister, Sm, Marra, Ma, and Taylor, M. D.
- Subjects
DNA Copy Number Variations ,Oncogene Proteins, Fusion ,medicine.medical_treatment ,Genes, myc ,Nerve Tissue Proteins ,Biology ,Bioinformatics ,medulloblastoma ,Article ,Translocation, Genetic ,Targeted therapy ,Structural variation ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,Gene Duplication ,Gene duplication ,medicine ,Humans ,Hedgehog Proteins ,Cerebellar Neoplasms ,Child ,030304 developmental biology ,Medulloblastoma ,0303 health sciences ,Multidisciplinary ,Chromothripsis ,PROTEÍNAS DE TRANSPORTE (GENÉTICA) ,Genome, Human ,NF-kappa B ,Cancer ,Proteins ,Genomics ,medicine.disease ,Human genetics ,3. Good health ,PVT1 ,030220 oncology & carcinogenesis ,Genomic Structural Variation ,RNA, Long Noncoding ,Carrier Proteins ,Signal Transduction - Abstract
Medulloblastoma, the most common malignant paediatric brain tumour, is currently treated with nonspecific cytotoxic therapies including surgery, whole-brain radiation, and aggressive chemotherapy. As medulloblastoma exhibits marked intertumoural heterogeneity, with at least four distinct molecular variants, previous attempts to identify targets for therapy have been underpowered because of small samples sizes. Here we report somatic copy number aberrations (SCNAs) in 1,087 unique medulloblastomas. SCNAs are common in medulloblastoma, and are predominantly subgroup-enriched. The most common region of focal copy number gain is a tandem duplication of SNCAIP, a gene associated with Parkinson's disease, which is exquisitely restricted to Group 4 alpha. Recurrent translocations of PVT1, including PVT1-MYC and PVT1-NDRG1, that arise through chromothripsis are restricted to Group 3. Numerous targetable SCNAs, including recurrent events targeting TGF-beta signalling in Group 3, and NF-kappa B signalling in Group 4, suggest future avenues for rational, targeted therapy.
- Published
- 2012
16. Author Correction: Pan-cancer analysis of whole genomes identifies driver rearrangements promoted by LINE-1 retrotransposition.
- Author
-
Rodriguez-Martin B, Alvarez EG, Baez-Ortega A, Zamora J, Supek F, Demeulemeester J, Santamarina M, Ju YS, Temes J, Garcia-Souto D, Detering H, Li Y, Rodriguez-Castro J, Dueso-Barroso A, Bruzos AL, Dentro SC, Blanco MG, Contino G, Ardeljan D, Tojo M, Roberts ND, Zumalave S, Edwards PA, Weischenfeldt J, Puiggròs M, Chong Z, Chen K, Lee EA, Wala JA, Raine KM, Butler A, Waszak SM, Navarro FCP, Schumacher SE, Monlong J, Maura F, Bolli N, Bourque G, Gerstein M, Park PJ, Wedge DC, Beroukhim R, Torrents D, Korbel JO, Martincorena I, Fitzgerald RC, Van Loo P, Kazazian HH, Burns KH, Campbell PJ, and Tubio JMC
- Published
- 2023
- Full Text
- View/download PDF
17. Author Correction: Patterns of somatic structural variation in human cancer genomes.
- Author
-
Li Y, Roberts ND, Wala JA, Shapira O, Schumacher SE, Kumar K, Khurana E, Waszak S, Korbel JO, Haber JE, Imielinski M, Weischenfeldt J, Beroukhim R, and Campbell PJ
- Published
- 2023
- Full Text
- View/download PDF
18. Author Correction: Analyses of non-coding somatic drivers in 2,658 cancer whole genomes.
- Author
-
Rheinbay E, Nielsen MM, Abascal F, Wala JA, Shapira O, Tiao G, Hornshøj H, Hess JM, Juul RI, Lin Z, Feuerbach L, Sabarinathan R, Madsen T, Kim J, Mularoni L, Shuai S, Lanzós A, Herrmann C, Maruvka YE, Shen C, Amin SB, Bandopadhayay P, Bertl J, Boroevich KA, Busanovich J, Carlevaro-Fita J, Chakravarty D, Chan CWY, Craft D, Dhingra P, Diamanti K, Fonseca NA, Gonzalez-Perez A, Guo Q, Hamilton MP, Haradhvala NJ, Hong C, Isaev K, Johnson TA, Juul M, Kahles A, Kahraman A, Kim Y, Komorowski J, Kumar K, Kumar S, Lee D, Lehmann KV, Li Y, Liu EM, Lochovsky L, Park K, Pich O, Roberts ND, Saksena G, Schumacher SE, Sidiropoulos N, Sieverling L, Sinnott-Armstrong N, Stewart C, Tamborero D, Tubio JMC, Umer HM, Uusküla-Reimand L, Wadelius C, Wadi L, Yao X, Zhang CZ, Zhang J, Haber JE, Hobolth A, Imielinski M, Kellis M, Lawrence MS, von Mering C, Nakagawa H, Raphael BJ, Rubin MA, Sander C, Stein LD, Stuart JM, Tsunoda T, Wheeler DA, Johnson R, Reimand J, Gerstein M, Khurana E, Campbell PJ, López-Bigas N, Weischenfeldt J, Beroukhim R, Martincorena I, Pedersen JS, and Getz G
- Published
- 2023
- Full Text
- View/download PDF
19. Tangent normalization for somatic copy-number inference in cancer genome analysis.
- Author
-
Gao GF, Oh C, Saksena G, Deng D, Westlake LC, Hill BA, Reich M, Schumacher SE, Berger AC, Carter SL, Cherniack AD, Meyerson M, Tabak B, Beroukhim R, and Getz G
- Subjects
- Humans, Algorithms, DNA Copy Number Variations, High-Throughput Nucleotide Sequencing methods, Neoplasms genetics, Software
- Abstract
Motivation: Somatic copy-number alterations (SCNAs) play an important role in cancer development. Systematic noise in sequencing and array data present a significant challenge to the inference of SCNAs for cancer genome analyses. As part of The Cancer Genome Atlas, the Broad Institute Genome Characterization Center developed the Tangent normalization method to generate copy-number profiles using data from single-nucleotide polymorphism (SNP) arrays and whole-exome sequencing (WES) technologies for over 10 000 pairs of tumors and matched normal samples. Here, we describe the Tangent method, which uses a unique linear combination of normal samples as a reference for each tumor sample, to subtract systematic errors that vary across samples. We also describe a modification of Tangent, called Pseudo-Tangent, which enables denoising through comparisons between tumor profiles when few normal samples are available., Results: Tangent normalization substantially increases signal-to-noise ratios (SNRs) compared to conventional normalization methods in both SNP array and WES analyses. Tangent and Pseudo-Tangent normalizations improve the SNR by reducing noise with minimal effect on signal and exceed the contribution of other steps in the analysis such as choice of segmentation algorithm. Tangent and Pseudo-Tangent are broadly applicable and enable more accurate inference of SCNAs from DNA sequencing and array data., Availability and Implementation: Tangent is available at https://github.com/broadinstitute/tangent and as a Docker image (https://hub.docker.com/r/broadinstitute/tangent). Tangent is also the normalization method for the copy-number pipeline in Genome Analysis Toolkit 4 (GATK4)., Supplementary Information: Supplementary data are available at Bioinformatics online., (© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
20. Characterizing genetic intra-tumor heterogeneity across 2,658 human cancer genomes.
- Author
-
Dentro SC, Leshchiner I, Haase K, Tarabichi M, Wintersinger J, Deshwar AG, Yu K, Rubanova Y, Macintyre G, Demeulemeester J, Vázquez-García I, Kleinheinz K, Livitz DG, Malikic S, Donmez N, Sengupta S, Anur P, Jolly C, Cmero M, Rosebrock D, Schumacher SE, Fan Y, Fittall M, Drews RM, Yao X, Watkins TBK, Lee J, Schlesner M, Zhu H, Adams DJ, McGranahan N, Swanton C, Getz G, Boutros PC, Imielinski M, Beroukhim R, Sahinalp SC, Ji Y, Peifer M, Martincorena I, Markowetz F, Mustonen V, Yuan K, Gerstung M, Spellman PT, Wang W, Morris QD, Wedge DC, and Van Loo P
- Subjects
- DNA Copy Number Variations, DNA, Neoplasm chemistry, DNA, Neoplasm metabolism, Databases, Genetic, Drug Resistance, Neoplasm genetics, Humans, Neoplasms pathology, Polymorphism, Single Nucleotide, Whole Genome Sequencing, Genetic Heterogeneity, Neoplasms genetics
- Abstract
Intra-tumor heterogeneity (ITH) is a mechanism of therapeutic resistance and therefore an important clinical challenge. However, the extent, origin, and drivers of ITH across cancer types are poorly understood. To address this, we extensively characterize ITH across whole-genome sequences of 2,658 cancer samples spanning 38 cancer types. Nearly all informative samples (95.1%) contain evidence of distinct subclonal expansions with frequent branching relationships between subclones. We observe positive selection of subclonal driver mutations across most cancer types and identify cancer type-specific subclonal patterns of driver gene mutations, fusions, structural variants, and copy number alterations as well as dynamic changes in mutational processes between subclonal expansions. Our results underline the importance of ITH and its drivers in tumor evolution and provide a pan-cancer resource of comprehensively annotated subclonal events from whole-genome sequencing data., Competing Interests: Declaration of interests G.M. and F.M. are cofounders and shareholders of Tailor Bio. R.B. owns equity in Ampressa Therapeutics. G.G. receives research funds from IBM and Pharmacyclics and is an inventor on patent applications related to MuTect, ABSOLUTE, MutSig, MSMuTect, and POLYSOLVER. I.L. is a consultant for PACT Pharma. B.J.R. is a consultant at and has ownership interest (including stock and patents) in Medley Genomics. N.M. has stock options in and has consulted for Achilles Therapeutics. C.S. acknowledges grant support from Pfizer, AstraZeneca, Bristol Myers Squibb, Roche-Ventana, Boehringer-Ingelheim, Archer Dx, and Ono Pharmaceutical; is an AstraZeneca Advisory Board Member and Chief Investigator for the MeRmaiD-1 clinical trial; has consulted for Pfizer, Novartis, GlaxoSmithKline, MSD, Bristol Myers Squibb, Celgene, AstraZeneca, Illumina, Amgen, Genentech, Roche-Ventana, GRAIL, Medicxi, Bicycle Therapeutics, and the Sarah Cannon Research Institute; has stock options in Apogen Biotechnologies, Epic Bioscience, and GRAIL; and has stock options and is co-founder of Achilles Therapeutics., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
21. Pan-cancer analysis of whole genomes identifies driver rearrangements promoted by LINE-1 retrotransposition.
- Author
-
Rodriguez-Martin B, Alvarez EG, Baez-Ortega A, Zamora J, Supek F, Demeulemeester J, Santamarina M, Ju YS, Temes J, Garcia-Souto D, Detering H, Li Y, Rodriguez-Castro J, Dueso-Barroso A, Bruzos AL, Dentro SC, Blanco MG, Contino G, Ardeljan D, Tojo M, Roberts ND, Zumalave S, Edwards PA, Weischenfeldt J, Puiggròs M, Chong Z, Chen K, Lee EA, Wala JA, Raine KM, Butler A, Waszak SM, Navarro FCP, Schumacher SE, Monlong J, Maura F, Bolli N, Bourque G, Gerstein M, Park PJ, Wedge DC, Beroukhim R, Torrents D, Korbel JO, Martincorena I, Fitzgerald RC, Van Loo P, Kazazian HH, Burns KH, Campbell PJ, and Tubio JMC
- Subjects
- Humans, Neoplasms pathology, Carcinogenesis genetics, Gene Rearrangement genetics, Genome, Human genetics, Long Interspersed Nucleotide Elements genetics, Neoplasms genetics, Retroelements genetics
- Abstract
About half of all cancers have somatic integrations of retrotransposons. Here, to characterize their role in oncogenesis, we analyzed the patterns and mechanisms of somatic retrotransposition in 2,954 cancer genomes from 38 histological cancer subtypes within the framework of the Pan-Cancer Analysis of Whole Genomes (PCAWG) project. We identified 19,166 somatically acquired retrotransposition events, which affected 35% of samples and spanned a range of event types. Long interspersed nuclear element (LINE-1; L1 hereafter) insertions emerged as the first most frequent type of somatic structural variation in esophageal adenocarcinoma, and the second most frequent in head-and-neck and colorectal cancers. Aberrant L1 integrations can delete megabase-scale regions of a chromosome, which sometimes leads to the removal of tumor-suppressor genes, and can induce complex translocations and large-scale duplications. Somatic retrotranspositions can also initiate breakage-fusion-bridge cycles, leading to high-level amplification of oncogenes. These observations illuminate a relevant role of L1 retrotransposition in remodeling the cancer genome, with potential implications for the development of human tumors.
- Published
- 2020
- Full Text
- View/download PDF
22. Analyses of non-coding somatic drivers in 2,658 cancer whole genomes.
- Author
-
Rheinbay E, Nielsen MM, Abascal F, Wala JA, Shapira O, Tiao G, Hornshøj H, Hess JM, Juul RI, Lin Z, Feuerbach L, Sabarinathan R, Madsen T, Kim J, Mularoni L, Shuai S, Lanzós A, Herrmann C, Maruvka YE, Shen C, Amin SB, Bandopadhayay P, Bertl J, Boroevich KA, Busanovich J, Carlevaro-Fita J, Chakravarty D, Chan CWY, Craft D, Dhingra P, Diamanti K, Fonseca NA, Gonzalez-Perez A, Guo Q, Hamilton MP, Haradhvala NJ, Hong C, Isaev K, Johnson TA, Juul M, Kahles A, Kahraman A, Kim Y, Komorowski J, Kumar K, Kumar S, Lee D, Lehmann KV, Li Y, Liu EM, Lochovsky L, Park K, Pich O, Roberts ND, Saksena G, Schumacher SE, Sidiropoulos N, Sieverling L, Sinnott-Armstrong N, Stewart C, Tamborero D, Tubio JMC, Umer HM, Uusküla-Reimand L, Wadelius C, Wadi L, Yao X, Zhang CZ, Zhang J, Haber JE, Hobolth A, Imielinski M, Kellis M, Lawrence MS, von Mering C, Nakagawa H, Raphael BJ, Rubin MA, Sander C, Stein LD, Stuart JM, Tsunoda T, Wheeler DA, Johnson R, Reimand J, Gerstein M, Khurana E, Campbell PJ, López-Bigas N, Weischenfeldt J, Beroukhim R, Martincorena I, Pedersen JS, and Getz G
- Subjects
- DNA Breaks, Databases, Genetic, Gene Expression Regulation, Neoplastic, Genome-Wide Association Study, Humans, INDEL Mutation, Genome, Human genetics, Mutation genetics, Neoplasms genetics
- Abstract
The discovery of drivers of cancer has traditionally focused on protein-coding genes
1-4 . Here we present analyses of driver point mutations and structural variants in non-coding regions across 2,658 genomes from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium5 of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA). For point mutations, we developed a statistically rigorous strategy for combining significance levels from multiple methods of driver discovery that overcomes the limitations of individual methods. For structural variants, we present two methods of driver discovery, and identify regions that are significantly affected by recurrent breakpoints and recurrent somatic juxtapositions. Our analyses confirm previously reported drivers6,7 , raise doubts about others and identify novel candidates, including point mutations in the 5' region of TP53, in the 3' untranslated regions of NFKBIZ and TOB1, focal deletions in BRD4 and rearrangements in the loci of AKR1C genes. We show that although point mutations and structural variants that drive cancer are less frequent in non-coding genes and regulatory sequences than in protein-coding genes, additional examples of these drivers will be found as more cancer genomes become available.- Published
- 2020
- Full Text
- View/download PDF
23. Patterns of somatic structural variation in human cancer genomes.
- Author
-
Li Y, Roberts ND, Wala JA, Shapira O, Schumacher SE, Kumar K, Khurana E, Waszak S, Korbel JO, Haber JE, Imielinski M, Weischenfeldt J, Beroukhim R, and Campbell PJ
- Subjects
- Gene Rearrangement genetics, Genomics, Humans, Mutagenesis, Insertional, Telomerase genetics, Genetic Variation, Genome, Human genetics, Neoplasms genetics
- Abstract
A key mutational process in cancer is structural variation, in which rearrangements delete, amplify or reorder genomic segments that range in size from kilobases to whole chromosomes
1-7 . Here we develop methods to group, classify and describe somatic structural variants, using data from the Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium of the International Cancer Genome Consortium (ICGC) and The Cancer Genome Atlas (TCGA), which aggregated whole-genome sequencing data from 2,658 cancers across 38 tumour types8 . Sixteen signatures of structural variation emerged. Deletions have a multimodal size distribution, assort unevenly across tumour types and patients, are enriched in late-replicating regions and correlate with inversions. Tandem duplications also have a multimodal size distribution, but are enriched in early-replicating regions-as are unbalanced translocations. Replication-based mechanisms of rearrangement generate varied chromosomal structures with low-level copy-number gains and frequent inverted rearrangements. One prominent structure consists of 2-7 templates copied from distinct regions of the genome strung together within one locus. Such cycles of templated insertions correlate with tandem duplications, and-in liver cancer-frequently activate the telomerase gene TERT. A wide variety of rearrangement processes are active in cancer, which generate complex configurations of the genome upon which selection can act.- Published
- 2020
- Full Text
- View/download PDF
24. Author Correction: Targeting wild-type KRAS-amplified gastroesophageal cancer through combined MEK and SHP2 inhibition.
- Author
-
Wong GS, Zhou J, Bin Liu J, Wu Z, Xu X, Li T, Xu D, Schumacher SE, Puschhof J, McFarland J, Zou C, Dulak A, Henderson L, Xu P, O'Day E, Rendak R, Liao WL, Cecchi F, Hembrough T, Schwartz S, Szeto C, Rustgi AK, Wong KK, Diehl JA, Jensen K, Graziano F, Ruzzo A, Fereshetian S, Mertins P, Carr SA, Beroukhim R, Nakamura K, Oki E, Watanabe M, Baba H, Imamura Y, Catenacci D, and Bass AJ
- Abstract
In the Supplementary Information originally published with this article, a lane was missing in the β-actin blot in Supplementary Fig. 2. The lane has been added. The error has been corrected in the Supplementary Information associated with this article.
- Published
- 2018
- Full Text
- View/download PDF
25. SETD2 Is Recurrently Mutated in Whole-Exome Sequenced Canine Osteosarcoma.
- Author
-
Sakthikumar S, Elvers I, Kim J, Arendt ML, Thomas R, Turner-Maier J, Swofford R, Johnson J, Schumacher SE, Alföldi J, Axelsson E, Couto CG, Kisseberth WC, Pettersson ME, Getz G, Meadows JRS, Modiano JF, Breen M, Kierczak M, Forsberg-Nilsson K, Marinescu VD, and Lindblad-Toh K
- Subjects
- Animals, DNA Copy Number Variations, DNA Mutational Analysis, Disease Models, Animal, Dog Diseases pathology, Dogs, Female, Genetic Predisposition to Disease, Humans, Male, Mutation, Osteosarcoma pathology, Tumor Suppressor Protein p53 genetics, Exome Sequencing, Dog Diseases genetics, Histone-Lysine N-Methyltransferase genetics, Osteosarcoma genetics
- Abstract
Osteosarcoma is a debilitating bone cancer that affects humans, especially children and adolescents. A homologous form of osteosarcoma spontaneously occurs in dogs, and its differential incidence observed across breeds allows for the investigation of tumor mutations in the context of multiple genetic backgrounds. Using whole-exome sequencing and dogs from three susceptible breeds (22 golden retrievers, 21 Rottweilers, and 23 greyhounds), we found that osteosarcoma tumors show a high frequency of somatic copy-number alterations (SCNA), affecting key oncogenes and tumor-suppressor genes. The across-breed results are similar to what has been observed for human osteosarcoma, but the disease frequency and somatic mutation counts vary in the three breeds. For all breeds, three mutational signatures (one of which has not been previously reported) and 11 significantly mutated genes were identified. TP53 was the most frequently altered gene (83% of dogs have either mutations or SCNA in TP53 ), recapitulating observations in human osteosarcoma. The second most frequently mutated gene, histone methyltransferase SETD2 , has known roles in multiple cancers, but has not previously been strongly implicated in osteosarcoma. This study points to the likely importance of histone modifications in osteosarcoma and highlights the strong genetic similarities between human and dog osteosarcoma, suggesting that canine osteosarcoma may serve as an excellent model for developing treatment strategies in both species. Significance: Canine osteosarcoma genomics identify SETD2 as a possible oncogenic driver of osteosarcoma, and findings establish the canine model as a useful comparative model for the corresponding human disease. Cancer Res; 78(13); 3421-31. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
26. Targeting wild-type KRAS-amplified gastroesophageal cancer through combined MEK and SHP2 inhibition.
- Author
-
Wong GS, Zhou J, Liu JB, Wu Z, Xu X, Li T, Xu D, Schumacher SE, Puschhof J, McFarland J, Zou C, Dulak A, Henderson L, Xu P, O'Day E, Rendak R, Liao WL, Cecchi F, Hembrough T, Schwartz S, Szeto C, Rustgi AK, Wong KK, Diehl JA, Jensen K, Graziano F, Ruzzo A, Fereshetian S, Mertins P, Carr SA, Beroukhim R, Nakamura K, Oki E, Watanabe M, Baba H, Imamura Y, Catenacci D, and Bass AJ
- Subjects
- Animals, Cell Line, Tumor, Disease Models, Animal, Esophageal Neoplasms pathology, Humans, Mice, Mitogen-Activated Protein Kinase Kinases metabolism, Piperidines pharmacology, Protein Kinase Inhibitors pharmacology, Protein Tyrosine Phosphatase, Non-Receptor Type 11 metabolism, Pyridones pharmacology, Pyrimidines pharmacology, Pyrimidinones pharmacology, Stomach Neoplasms pathology, Esophageal Neoplasms genetics, Gene Amplification, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Protein Tyrosine Phosphatase, Non-Receptor Type 11 antagonists & inhibitors, Proto-Oncogene Proteins p21(ras) genetics, Stomach Neoplasms genetics
- Abstract
The role of KRAS, when activated through canonical mutations, has been well established in cancer
1 . Here we explore a secondary means of KRAS activation in cancer: focal high-level amplification of the KRAS gene in the absence of coding mutations. These amplifications occur most commonly in esophageal, gastric and ovarian adenocarcinomas2-4 . KRAS-amplified gastric cancer models show marked overexpression of the KRAS protein and are insensitive to MAPK blockade owing to their capacity to adaptively respond by rapidly increasing KRAS-GTP levels. Here we demonstrate that inhibition of the guanine-exchange factors SOS1 and SOS2 or the protein tyrosine phosphatase SHP2 can attenuate this adaptive process and that targeting these factors, both genetically and pharmacologically, can enhance the sensitivity of KRAS-amplified models to MEK inhibition in both in vitro and in vivo settings. These data demonstrate the relevance of copy-number amplification as a mechanism of KRAS activation, and uncover the therapeutic potential for targeting of these tumors through combined SHP2 and MEK inhibition.- Published
- 2018
- Full Text
- View/download PDF
27. Genomic and Functional Approaches to Understanding Cancer Aneuploidy.
- Author
-
Taylor AM, Shih J, Ha G, Gao GF, Zhang X, Berger AC, Schumacher SE, Wang C, Hu H, Liu J, Lazar AJ, Cherniack AD, Beroukhim R, and Meyerson M
- Subjects
- Cell Cycle, Cell Proliferation, Chromosome Aberrations, Chromosome Deletion, Chromosomes, Human, Pair 3 genetics, Databases, Genetic, Humans, Mutation Rate, Aneuploidy, Carcinoma, Squamous Cell genetics, Genomics methods, Tumor Suppressor Protein p53 genetics
- Abstract
Aneuploidy, whole chromosome or chromosome arm imbalance, is a near-universal characteristic of human cancers. In 10,522 cancer genomes from The Cancer Genome Atlas, aneuploidy was correlated with TP53 mutation, somatic mutation rate, and expression of proliferation genes. Aneuploidy was anti-correlated with expression of immune signaling genes, due to decreased leukocyte infiltrates in high-aneuploidy samples. Chromosome arm-level alterations show cancer-specific patterns, including loss of chromosome arm 3p in squamous cancers. We applied genome engineering to delete 3p in lung cells, causing decreased proliferation rescued in part by chromosome 3 duplication. This study defines genomic and phenotypic correlates of cancer aneuploidy and provides an experimental approach to study chromosome arm aneuploidy., (Copyright © 2018 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
28. Erratum: Genomic landscape of high-grade meningiomas.
- Author
-
Bi WL, Greenwald NF, Abedalthagafi M, Wala J, Gibson WJ, Agarwalla PK, Horowitz P, Schumacher SE, Esaulova E, Mei Y, Chevalier A, A Ducar M, Thorner AR, van Hummelen P, O Stemmer-Rachamimov A, Artyomov M, Al-Mefty O, Dunn GP, Santagata S, Dunn IF, and Beroukhim R
- Abstract
[This corrects the article DOI: 10.1038/s41525-017-0014-7.].
- Published
- 2017
- Full Text
- View/download PDF
29. Clinical targeted exome-based sequencing in combination with genome-wide copy number profiling: precision medicine analysis of 203 pediatric brain tumors.
- Author
-
Ramkissoon SH, Bandopadhayay P, Hwang J, Ramkissoon LA, Greenwald NF, Schumacher SE, O'Rourke R, Pinches N, Ho P, Malkin H, Sinai C, Filbin M, Plant A, Bi WL, Chang MS, Yang E, Wright KD, Manley PE, Ducar M, Alexandrescu S, Lidov H, Delalle I, Goumnerova LC, Church AJ, Janeway KA, Harris MH, MacConaill LE, Folkerth RD, Lindeman NI, Stiles CD, Kieran MW, Ligon AH, Santagata S, Dubuc AM, Chi SN, Beroukhim R, and Ligon KL
- Subjects
- Brain Neoplasms diagnosis, Child, Comparative Genomic Hybridization, Gene Dosage, Humans, Mutation, Polymorphism, Single Nucleotide, Sequence Analysis, DNA, Brain Neoplasms genetics, DNA Copy Number Variations, Exome, Genomics methods, Precision Medicine methods
- Abstract
Background: Clinical genomics platforms are needed to identify targetable alterations, but implementation of these technologies and best practices in routine clinical pediatric oncology practice are not yet well established., Methods: Profile is an institution-wide prospective clinical research initiative that uses targeted sequencing to identify targetable alterations in tumors. OncoPanel, a multiplexed targeted exome-sequencing platform that includes 300 cancer-causing genes, was used to assess single nucleotide variants and rearrangements/indels. Alterations were annotated (Tiers 1-4) based on clinical significance, with Tier 1 alterations having well-established clinical utility. OncoCopy, a clinical genome-wide array comparative genomic hybridization (aCGH) assay, was also performed to evaluate copy number alterations and better define rearrangement breakpoints., Results: Cancer genomes of 203 pediatric brain tumors were profiled across histological subtypes, including 117 samples analyzed by OncoPanel, 146 by OncoCopy, and 60 tumors subjected to both methodologies. OncoPanel revealed clinically relevant alterations in 56% of patients (44 cancer mutations and 20 rearrangements), including BRAF alterations that directed the use of targeted inhibitors. Rearrangements in MYB-QKI, MYBL1, BRAF, and FGFR1 were also detected. Furthermore, while copy number profiles differed across histologies, the combined use of OncoPanel and OncoCopy identified subgroup-specific alterations in 89% (17/19) of medulloblastomas., Conclusion: The combination of OncoPanel and OncoCopy multiplex genomic assays can identify critical diagnostic, prognostic, and treatment-relevant alterations and represents an effective precision medicine approach for clinical evaluation of pediatric brain tumors., (© The Author(s) 2017. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com)
- Published
- 2017
- Full Text
- View/download PDF
30. Corrigendum: Recurrent hormone-binding domain truncated ESR1 amplifications in primary endometrial cancers suggest their implication in hormone independent growth.
- Author
-
Holst F, Hoivik EA, Gibson WJ, Taylor-Weiner A, Schumacher SE, Asmann YW, Grossmann P, Trovik J, Necela BM, Thompson EA, Meyerson M, Beroukhim R, Salvesen HB, and Cherniack AD
- Abstract
This corrects the article DOI: 10.1038/srep25521.
- Published
- 2017
- Full Text
- View/download PDF
31. Somatic copy number alterations in gastric adenocarcinomas among Asian and Western patients.
- Author
-
Schumacher SE, Shim BY, Corso G, Ryu MH, Kang YK, Roviello F, Saksena G, Peng S, Shivdasani RA, Bass AJ, and Beroukhim R
- Subjects
- Humans, Adenocarcinoma genetics, Asian People genetics, DNA Copy Number Variations, Stomach Neoplasms genetics, White People genetics
- Abstract
Gastric cancer, a leading worldwide cause of cancer mortality, shows high geographic and ethnic variation in incidence rates, which are highest in East Asia. The anatomic locations and clinical behavior also differ by geography, leading to the controversial idea that Eastern and Western forms of the disease are distinct. In view of these differences, we investigated whether gastric cancers from Eastern and Western patients show distinct genomic profiles. We used high-density profiling of somatic copy-number aberrations to analyze the largest collection to date of gastric adenocarcinomas and utilized genotyping data to rigorously annotate ethnic status. The size of this collection allowed us to accurately identify regions of significant copy-number alteration and separately to evaluate tumors arising in Eastern and Western patients. Among molecular subtypes classified by The Cancer Genome Atlas, the frequency of gastric cancers showing chromosomal instability was modestly higher in Western patients. After accounting for this difference, however, gastric cancers arising in Easterners and Westerners have highly similar somatic copy-number patterns. Only one genomic event, focal deletion of the phosphatase gene PTPRD, was significantly enriched in Western cases, though also detected in Eastern cases. Thus, despite the different risk factors and clinical features, gastric cancer appears to be a fundamentally similar disease in both populations and the divergent clinical outcomes cannot be ascribed to different underlying structural somatic genetic aberrations.
- Published
- 2017
- Full Text
- View/download PDF
32. Landscape of Genomic Alterations in Pituitary Adenomas.
- Author
-
Bi WL, Horowitz P, Greenwald NF, Abedalthagafi M, Agarwalla PK, Gibson WJ, Mei Y, Schumacher SE, Ben-David U, Chevalier A, Carter S, Tiao G, Brastianos PK, Ligon AH, Ducar M, MacConaill L, Laws ER Jr, Santagata S, Beroukhim R, and Dunn IF
- Subjects
- Adult, Aged, Aged, 80 and over, Brain Neoplasms pathology, DNA Copy Number Variations genetics, Female, Gene Expression Regulation, Neoplastic genetics, Genome, Human, Humans, INDEL Mutation genetics, Male, Middle Aged, Mutation, Pituitary Neoplasms pathology, Brain Neoplasms genetics, Chromosomes, Human genetics, Pituitary Neoplasms genetics, Exome Sequencing
- Abstract
Purpose: Pituitary adenomas are the second most common primary brain tumor, yet their genetic profiles are incompletely understood. Experimental Design: We performed whole-exome sequencing of 42 pituitary macroadenomas and matched normal DNA. These adenomas included hormonally active and inactive tumors, ones with typical or atypical histology, and ones that were primary or recurrent. Results: We identified mutations, insertions/deletions, and copy-number alterations. Nearly one-third of samples (29%) had chromosome arm-level copy-number alterations across large fractions of the genome. Despite such widespread genomic disruption, these tumors had few focal events, which is unusual among highly disrupted cancers. The other 71% of tumors formed a distinct molecular class, with somatic copy number alterations involving less than 6% of the genome. Among the highly disrupted group, 75% were functional adenomas or atypical null-cell adenomas, whereas 87% of the less-disrupted group were nonfunctional adenomas. We confirmed this association between functional subtype and disruption in a validation dataset of 87 pituitary adenomas. Analysis of previously published expression data from an additional 50 adenomas showed that arm-level alterations significantly impacted transcript levels, and that the disrupted samples were characterized by expression changes associated with poor outcome in other cancers. Arm-level losses of chromosomes 1, 2, 11, and 18 were significantly recurrent. No significantly recurrent mutations were identified, suggesting no genes are altered by exonic mutations across large fractions of pituitary macroadenomas. Conclusions: These data indicate that sporadic pituitary adenomas have distinct copy-number profiles that associate with hormonal and histologic subtypes and influence gene expression. Clin Cancer Res; 23(7); 1841-51. ©2016 AACR ., (©2016 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
33. Pan-Cancer Analysis Links PARK2 to BCL-XL-Dependent Control of Apoptosis.
- Author
-
Gong Y, Schumacher SE, Wu WH, Tang F, Beroukhim R, and Chan TA
- Subjects
- Apoptosis Regulatory Proteins genetics, Apoptosis Regulatory Proteins metabolism, Cell Line, Tumor, Gene Expression Regulation, Neoplastic, Genes, Tumor Suppressor, Humans, Mitochondria metabolism, Mutation, Neoplasms genetics, Proteasome Endopeptidase Complex metabolism, Protein Binding, Proteolysis, Ubiquitin-Protein Ligases genetics, bcl-X Protein genetics, Apoptosis genetics, Neoplasms metabolism, Ubiquitin-Protein Ligases metabolism, bcl-X Protein metabolism
- Abstract
Mutation of the PARK2 gene can promote both Parkinson's Disease and cancer, yet the underlying mechanisms of how PARK2 controls cellular physiology is incompletely understood. Here, we show that the PARK2 tumor suppressor controls the apoptotic regulator BCL-XL and modulates programmed cell death. Analysis of approximately 10,000 tumor genomes uncovers a striking pattern of mutual exclusivity between PARK2 genetic loss and amplification of BCL2L1, implicating these genes in a common pathway. PARK2 directly binds to and ubiquitinates BCL-XL. Inactivation of PARK2 leads to aberrant accumulation of BCL-XL both in vitro and in vivo, and cancer-specific mutations in PARK2 abrogate the ability of the ubiquitin E3 ligase to target BCL-XL for degradation. Furthermore, PARK2 modulates mitochondrial depolarization and apoptosis in a BCL-XL-dependent manner. Thus, like genes at the nodal points of growth arrest pathways such as p53, the PARK2 tumor suppressor is able to exert its antiproliferative effects by regulating both cell cycle progression and programmed cell death., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
34. Genomic landscape of high-grade meningiomas.
- Author
-
Bi WL, Greenwald NF, Abedalthagafi M, Wala J, Gibson WJ, Agarwalla PK, Horowitz P, Schumacher SE, Esaulova E, Mei Y, Chevalier A, Ducar M, Thorner AR, van Hummelen P, Stemmer-Rachamimov A, Artyomov M, Al-Mefty O, Dunn GP, Santagata S, Dunn IF, and Beroukhim R
- Abstract
High-grade meningiomas frequently recur and are associated with high rates of morbidity and mortality. To determine the factors that promote the development and evolution of these tumors, we analyzed the genomes of 134 high-grade meningiomas and compared this information with data from 587 previously published meningiomas. High-grade meningiomas had a higher mutation burden than low-grade meningiomas but did not harbor any statistically significant mutated genes aside from NF2 . High-grade meningiomas also possessed significantly elevated rates of chromosomal gains and losses, especially among tumors with monosomy 22. Meningiomas previously treated with adjuvant radiation had significantly more copy number alterations than radiation-induced or radiation-naïve meningiomas. Across serial recurrences, genomic disruption preceded the emergence of nearly all mutations, remained largely uniform across time, and when present in low-grade meningiomas, correlated with subsequent progression to a higher grade. In contrast to the largely stable copy number alterations, mutations were strikingly heterogeneous across tumor recurrences, likely due to extensive geographic heterogeneity in the primary tumor. While high-grade meningiomas harbored significantly fewer overtly targetable alterations than low-grade meningiomas, they contained numerous mutations that are predicted to be neoantigens, suggesting that immunologic targeting may be of therapeutic value.
- Published
- 2017
- Full Text
- View/download PDF
35. Tumor-suppressor genes that escape from X-inactivation contribute to cancer sex bias.
- Author
-
Dunford A, Weinstock DM, Savova V, Schumacher SE, Cleary JP, Yoda A, Sullivan TJ, Hess JM, Gimelbrant AA, Beroukhim R, Lawrence MS, Getz G, and Lane AA
- Subjects
- Female, Humans, Male, Chromosomes, Human, X genetics, Genes, Tumor Suppressor, Genes, X-Linked genetics, Mutation genetics, Neoplasms genetics, Sexism statistics & numerical data, X Chromosome Inactivation genetics
- Abstract
There is a striking and unexplained male predominance across many cancer types. A subset of X-chromosome genes can escape X-inactivation, which would protect females from complete functional loss by a single mutation. To identify putative 'escape from X-inactivation tumor-suppressor' (EXITS) genes, we examined somatic alterations from >4,100 cancers across 21 tumor types for sex bias. Six of 783 non-pseudoautosomal region (PAR) X-chromosome genes (ATRX, CNKSR2, DDX3X, KDM5C, KDM6A, and MAGEC3) harbored loss-of-function mutations more frequently in males (based on a false discovery rate < 0.1), in comparison to zero of 18,055 autosomal and PAR genes (Fisher's exact P < 0.0001). Male-biased mutations in genes that escape X-inactivation were observed in combined analysis across many cancers and in several individual tumor types, suggesting a generalized phenomenon. We conclude that biallelic expression of EXITS genes in females explains a portion of the reduced cancer incidence in females as compared to males across a variety of tumor types.
- Published
- 2017
- Full Text
- View/download PDF
36. Genomic evolution and chemoresistance in germ-cell tumours.
- Author
-
Taylor-Weiner A, Zack T, O'Donnell E, Guerriero JL, Bernard B, Reddy A, Han GC, AlDubayan S, Amin-Mansour A, Schumacher SE, Litchfield K, Turnbull C, Gabriel S, Beroukhim R, Getz G, Carter SL, Hirsch MS, Letai A, Sweeney C, and Van Allen EM
- Subjects
- Apoptosis, Disease Progression, Evolution, Molecular, Exome genetics, Genomics, Humans, Loss of Heterozygosity, Male, Mitochondria metabolism, Mutation, Nanog Homeobox Protein deficiency, Neoplasm Metastasis genetics, Neoplasm Metastasis pathology, Neoplasms, Germ Cell and Embryonal metabolism, Neoplasms, Germ Cell and Embryonal pathology, Octamer Transcription Factor-3 deficiency, Phylogeny, Proto-Oncogene Proteins p21(ras) genetics, Teratoma genetics, Testicular Neoplasms drug therapy, Testicular Neoplasms genetics, Testicular Neoplasms metabolism, Testicular Neoplasms pathology, Transcriptome genetics, Tumor Suppressor Protein p53 genetics, Drug Resistance, Neoplasm, Genome, Human genetics, Neoplasms, Germ Cell and Embryonal drug therapy, Neoplasms, Germ Cell and Embryonal genetics
- Abstract
Germ-cell tumours (GCTs) are derived from germ cells and occur most frequently in the testes. GCTs are histologically heterogeneous and distinctly curable with chemotherapy. Gains of chromosome arm 12p and aneuploidy are nearly universal in GCTs, but specific somatic genomic features driving tumour initiation, chemosensitivity and progression are incompletely characterized. Here, using clinical whole-exome and transcriptome sequencing of precursor, primary (testicular and mediastinal) and chemoresistant metastatic human GCTs, we show that the primary somatic feature of GCTs is highly recurrent chromosome arm level amplifications and reciprocal deletions (reciprocal loss of heterozygosity), variations that are significantly enriched in GCTs compared to 19 other cancer types. These tumours also acquire KRAS mutations during the development from precursor to primary disease, and primary testicular GCTs (TGCTs) are uniformly wild type for TP53. In addition, by functional measurement of apoptotic signalling (BH3 profiling) of fresh tumour and adjacent tissue, we find that primary TGCTs have high mitochondrial priming that facilitates chemotherapy-induced apoptosis. Finally, by phylogenetic analysis of serial TGCTs that emerge with chemotherapy resistance, we show how TGCTs gain additional reciprocal loss of heterozygosity and that this is associated with loss of pluripotency markers (NANOG and POU5F1) in chemoresistant teratomas or transformed carcinomas. Our results demonstrate the distinct genomic features underlying the origins of this disease and associated with the chemosensitivity phenotype, as well as the rare progression to chemoresistance. These results identify the convergence of cancer genomics, mitochondrial priming and GCT evolution, and may provide insights into chemosensitivity and resistance in other cancers.
- Published
- 2016
- Full Text
- View/download PDF
37. Corrigendum: Recurrent hormone-binding domain truncated ESR1 amplifications in primary endometrial cancers suggest their implication in hormone independent growth.
- Author
-
Holst F, Hoivik EA, Gibson WJ, Taylor-Weiner A, Schumacher SE, Asmann YW, Grossmann P, Trovik J, Necela BM, Thompson EA, Meyerson M, Beroukhim R, Salvesen HB, and Cherniack AD
- Published
- 2016
- Full Text
- View/download PDF
38. Recurrent hormone-binding domain truncated ESR1 amplifications in primary endometrial cancers suggest their implication in hormone independent growth.
- Author
-
Holst F, Hoivik EA, Gibson WJ, Taylor-Weiner A, Schumacher SE, Asmann YW, Grossmann P, Trovik J, Necela BM, Thompson EA, Meyerson M, Beroukhim R, Salvesen HB, and Cherniack AD
- Abstract
The estrogen receptor alpha (ERα) is highly expressed in both endometrial and breast cancers, and represents the most prevalent therapeutic target in breast cancer. However, anti-estrogen therapy has not been shown to be effective in endometrial cancer. Recently it has been shown that hormone-binding domain alterations of ERα in breast cancer contribute to acquired resistance to anti-estrogen therapy. In analyses of genomic data from The Cancer Genome Atlas (TCGA), we observe that endometrial carcinomas manifest recurrent ESR1 gene amplifications that truncate the hormone-binding domain encoding region of ESR1 and are associated with reduced mRNA expression of exons encoding the hormone-binding domain. These findings support a role for hormone-binding alterations of ERα in primary endometrial cancer, with potentially important therapeutic implications.
- Published
- 2016
- Full Text
- View/download PDF
39. MYB-QKI rearrangements in angiocentric glioma drive tumorigenicity through a tripartite mechanism.
- Author
-
Bandopadhayay P, Ramkissoon LA, Jain P, Bergthold G, Wala J, Zeid R, Schumacher SE, Urbanski L, O'Rourke R, Gibson WJ, Pelton K, Ramkissoon SH, Han HJ, Zhu Y, Choudhari N, Silva A, Boucher K, Henn RE, Kang YJ, Knoff D, Paolella BR, Gladden-Young A, Varlet P, Pages M, Horowitz PM, Federation A, Malkin H, Tracy AA, Seepo S, Ducar M, Van Hummelen P, Santi M, Buccoliero AM, Scagnet M, Bowers DC, Giannini C, Puget S, Hawkins C, Tabori U, Klekner A, Bognar L, Burger PC, Eberhart C, Rodriguez FJ, Hill DA, Mueller S, Haas-Kogan DA, Phillips JJ, Santagata S, Stiles CD, Bradner JE, Jabado N, Goren A, Grill J, Ligon AH, Goumnerova L, Waanders AJ, Storm PB, Kieran MW, Ligon KL, Beroukhim R, and Resnick AC
- Subjects
- Carcinogenesis genetics, Cell Line, Tumor, Child, Comparative Genomic Hybridization, Exome genetics, Gene Expression Regulation, Neoplastic, Gene Rearrangement, Glioma pathology, High-Throughput Nucleotide Sequencing, Humans, Mutation, Oncogene Proteins v-myb biosynthesis, Oncogene Proteins, Fusion biosynthesis, RNA-Binding Proteins biosynthesis, Glioma genetics, Oncogene Proteins v-myb genetics, Oncogene Proteins, Fusion genetics, RNA-Binding Proteins genetics
- Abstract
Angiocentric gliomas are pediatric low-grade gliomas (PLGGs) without known recurrent genetic drivers. We performed genomic analysis of new and published data from 249 PLGGs, including 19 angiocentric gliomas. We identified MYB-QKI fusions as a specific and single candidate driver event in angiocentric gliomas. In vitro and in vivo functional studies show that MYB-QKI rearrangements promote tumorigenesis through three mechanisms: MYB activation by truncation, enhancer translocation driving aberrant MYB-QKI expression and hemizygous loss of the tumor suppressor QKI. To our knowledge, this represents the first example of a single driver rearrangement simultaneously transforming cells via three genetic and epigenetic mechanisms in a tumor.
- Published
- 2016
- Full Text
- View/download PDF
40. Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma.
- Author
-
Linehan WM, Spellman PT, Ricketts CJ, Creighton CJ, Fei SS, Davis C, Wheeler DA, Murray BA, Schmidt L, Vocke CD, Peto M, Al Mamun AA, Shinbrot E, Sethi A, Brooks S, Rathmell WK, Brooks AN, Hoadley KA, Robertson AG, Brooks D, Bowlby R, Sadeghi S, Shen H, Weisenberger DJ, Bootwalla M, Baylin SB, Laird PW, Cherniack AD, Saksena G, Haake S, Li J, Liang H, Lu Y, Mills GB, Akbani R, Leiserson MD, Raphael BJ, Anur P, Bottaro D, Albiges L, Barnabas N, Choueiri TK, Czerniak B, Godwin AK, Hakimi AA, Ho TH, Hsieh J, Ittmann M, Kim WY, Krishnan B, Merino MJ, Mills Shaw KR, Reuter VE, Reznik E, Shelley CS, Shuch B, Signoretti S, Srinivasan R, Tamboli P, Thomas G, Tickoo S, Burnett K, Crain D, Gardner J, Lau K, Mallery D, Morris S, Paulauskis JD, Penny RJ, Shelton C, Shelton WT, Sherman M, Thompson E, Yena P, Avedon MT, Bowen J, Gastier-Foster JM, Gerken M, Leraas KM, Lichtenberg TM, Ramirez NC, Santos T, Wise L, Zmuda E, Demchok JA, Felau I, Hutter CM, Sheth M, Sofia HJ, Tarnuzzer R, Wang Z, Yang L, Zenklusen JC, Zhang J, Ayala B, Baboud J, Chudamani S, Liu J, Lolla L, Naresh R, Pihl T, Sun Q, Wan Y, Wu Y, Ally A, Balasundaram M, Balu S, Beroukhim R, Bodenheimer T, Buhay C, Butterfield YS, Carlsen R, Carter SL, Chao H, Chuah E, Clarke A, Covington KR, Dahdouli M, Dewal N, Dhalla N, Doddapaneni HV, Drummond JA, Gabriel SB, Gibbs RA, Guin R, Hale W, Hawes A, Hayes DN, Holt RA, Hoyle AP, Jefferys SR, Jones SJ, Jones CD, Kalra D, Kovar C, Lewis L, Li J, Ma Y, Marra MA, Mayo M, Meng S, Meyerson M, Mieczkowski PA, Moore RA, Morton D, Mose LE, Mungall AJ, Muzny D, Parker JS, Perou CM, Roach J, Schein JE, Schumacher SE, Shi Y, Simons JV, Sipahimalani P, Skelly T, Soloway MG, Sougnez C, Tam A, Tan D, Thiessen N, Veluvolu U, Wang M, Wilkerson MD, Wong T, Wu J, Xi L, Zhou J, Bedford J, Chen F, Fu Y, Gerstein M, Haussler D, Kasaian K, Lai P, Ling S, Radenbaugh A, Van Den Berg D, Weinstein JN, Zhu J, Albert M, Alexopoulou I, Andersen JJ, Auman JT, Bartlett J, Bastacky S, Bergsten J, Blute ML, Boice L, Bollag RJ, Boyd J, Castle E, Chen YB, Cheville JC, Curley E, Davies B, DeVolk A, Dhir R, Dike L, Eckman J, Engel J, Harr J, Hrebinko R, Huang M, Huelsenbeck-Dill L, Iacocca M, Jacobs B, Lobis M, Maranchie JK, McMeekin S, Myers J, Nelson J, Parfitt J, Parwani A, Petrelli N, Rabeno B, Roy S, Salner AL, Slaton J, Stanton M, Thompson RH, Thorne L, Tucker K, Weinberger PM, Winemiller C, Zach LA, and Zuna R
- Subjects
- Carcinoma, Papillary genetics, CpG Islands physiology, DNA Methylation, Humans, Kidney Neoplasms genetics, MicroRNAs chemistry, NF-E2-Related Factor 2 genetics, Phenotype, Proto-Oncogene Proteins c-met chemistry, Proto-Oncogene Proteins c-met genetics, RNA, Messenger chemistry, RNA, Neoplasm chemistry, Sequence Analysis, RNA, Signal Transduction physiology, Carcinoma, Papillary metabolism, Kidney Neoplasms metabolism, Mutation, NF-E2-Related Factor 2 metabolism, Proto-Oncogene Proteins c-met metabolism
- Abstract
Background: Papillary renal-cell carcinoma, which accounts for 15 to 20% of renal-cell carcinomas, is a heterogeneous disease that consists of various types of renal cancer, including tumors with indolent, multifocal presentation and solitary tumors with an aggressive, highly lethal phenotype. Little is known about the genetic basis of sporadic papillary renal-cell carcinoma, and no effective forms of therapy for advanced disease exist., Methods: We performed comprehensive molecular characterization of 161 primary papillary renal-cell carcinomas, using whole-exome sequencing, copy-number analysis, messenger RNA and microRNA sequencing, DNA-methylation analysis, and proteomic analysis., Results: Type 1 and type 2 papillary renal-cell carcinomas were shown to be different types of renal cancer characterized by specific genetic alterations, with type 2 further classified into three individual subgroups on the basis of molecular differences associated with patient survival. Type 1 tumors were associated with MET alterations, whereas type 2 tumors were characterized by CDKN2A silencing, SETD2 mutations, TFE3 fusions, and increased expression of the NRF2-antioxidant response element (ARE) pathway. A CpG island methylator phenotype (CIMP) was observed in a distinct subgroup of type 2 papillary renal-cell carcinomas that was characterized by poor survival and mutation of the gene encoding fumarate hydratase (FH)., Conclusions: Type 1 and type 2 papillary renal-cell carcinomas were shown to be clinically and biologically distinct. Alterations in the MET pathway were associated with type 1, and activation of the NRF2-ARE pathway was associated with type 2; CDKN2A loss and CIMP in type 2 conveyed a poor prognosis. Furthermore, type 2 papillary renal-cell carcinoma consisted of at least three subtypes based on molecular and phenotypic features. (Funded by the National Institutes of Health.).
- Published
- 2016
- Full Text
- View/download PDF
41. Bias modification training can alter approach bias and chocolate consumption.
- Author
-
Schumacher SE, Kemps E, and Tiggemann M
- Subjects
- Adolescent, Adult, Body Mass Index, Cues, Female, Humans, Taste, Young Adult, Candy, Chocolate, Choice Behavior, Food Preferences psychology, Health Behavior
- Abstract
Recent evidence has demonstrated that bias modification training has potential to reduce cognitive biases for attractive targets and affect health behaviours. The present study investigated whether cognitive bias modification training could be applied to reduce approach bias for chocolate and affect subsequent chocolate consumption. A sample of 120 women (18-27 years) were randomly assigned to an approach-chocolate condition or avoid-chocolate condition, in which they were trained to approach or avoid pictorial chocolate stimuli, respectively. Training had the predicted effect on approach bias, such that participants trained to approach chocolate demonstrated an increased approach bias to chocolate stimuli whereas participants trained to avoid such stimuli showed a reduced bias. Further, participants trained to avoid chocolate ate significantly less of a chocolate muffin in a subsequent taste test than participants trained to approach chocolate. Theoretically, results provide support for the dual process model's conceptualisation of consumption as being driven by implicit processes such as approach bias. In practice, approach bias modification may be a useful component of interventions designed to curb the consumption of unhealthy foods., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
42. MECP2 Is a Frequently Amplified Oncogene with a Novel Epigenetic Mechanism That Mimics the Role of Activated RAS in Malignancy.
- Author
-
Neupane M, Clark AP, Landini S, Birkbak NJ, Eklund AC, Lim E, Culhane AC, Barry WT, Schumacher SE, Beroukhim R, Szallasi Z, Vidal M, Hill DE, and Silver DP
- Subjects
- 5-Methylcytosine analogs & derivatives, Alternative Splicing, Animals, Cell Line, Tumor, Cytosine metabolism, Epigenesis, Genetic, Humans, Methyl-CpG-Binding Protein 2 metabolism, Mice, Neoplasm Transplantation, Protein Isoforms metabolism, Signal Transduction, Cytosine analogs & derivatives, Gene Amplification, Methyl-CpG-Binding Protein 2 genetics, Neoplasms genetics, ras Proteins genetics
- Abstract
Unlabelled: An unbiased genome-scale screen for unmutated genes that drive cancer growth when overexpressed identified methyl cytosine-guanine dinucleotide (CpG) binding protein 2 (MECP2) as a novel oncogene. MECP2 resides in a region of the X-chromosome that is significantly amplified across 18% of cancers, and many cancer cell lines have amplified, overexpressed MECP2 and are dependent on MECP2 expression for growth. MECP2 copy-number gain and RAS family member alterations are mutually exclusive in several cancer types. The MECP2 splicing isoforms activate the major growth factor pathways targeted by activated RAS, the MAPK and PI3K pathways. MECP2 rescued the growth of a KRAS(G12C)-addicted cell line after KRAS downregulation, and activated KRAS rescues the growth of an MECP2-addicted cell line after MECP2 downregulation. MECP2 binding to the epigenetic modification 5-hydroxymethylcytosine is required for efficient transformation. These observations suggest that MECP2 is a commonly amplified oncogene with an unusual epigenetic mode of action., Significance: MECP2 is a commonly amplified oncogene in human malignancies with a unique epigenetic mechanism of action. Cancer Discov; 6(1); 45-58. ©2015 AACR.This article is highlighted in the In This Issue feature, p. 1., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
43. Exome sequencing of lymphomas from three dog breeds reveals somatic mutation patterns reflecting genetic background.
- Author
-
Elvers I, Turner-Maier J, Swofford R, Koltookian M, Johnson J, Stewart C, Zhang CZ, Schumacher SE, Beroukhim R, Rosenberg M, Thomas R, Mauceli E, Getz G, Palma FD, Modiano JF, Breen M, Lindblad-Toh K, and Alföldi J
- Subjects
- Animals, B-Lymphocytes metabolism, Cell Cycle Proteins genetics, DNA Copy Number Variations, Disease Models, Animal, F-Box Proteins genetics, F-Box-WD Repeat-Containing Protein 7, Humans, Lymphoma, B-Cell diagnosis, Mutation, Protein Serine-Threonine Kinases genetics, Sequence Alignment, Shelterin Complex, T-Lymphocytes metabolism, TNF Receptor-Associated Factor 3 genetics, Telomere-Binding Proteins genetics, Ubiquitin-Protein Ligases genetics, NF-kappaB-Inducing Kinase, Dogs genetics, Exome, Genetic Background, Lymphoma, B-Cell genetics
- Abstract
Lymphoma is the most common hematological malignancy in developed countries. Outcome is strongly determined by molecular subtype, reflecting a need for new and improved treatment options. Dogs spontaneously develop lymphoma, and the predisposition of certain breeds indicates genetic risk factors. Using the dog breed structure, we selected three lymphoma predisposed breeds developing primarily T-cell (boxer), primarily B-cell (cocker spaniel), and with equal distribution of B- and T-cell lymphoma (golden retriever), respectively. We investigated the somatic mutations in B- and T-cell lymphomas from these breeds by exome sequencing of tumor and normal pairs. Strong similarities were evident between B-cell lymphomas from golden retrievers and cocker spaniels, with recurrent mutations in TRAF3-MAP3K14 (28% of all cases), FBXW7 (25%), and POT1 (17%). The FBXW7 mutations recurrently occur in a specific codon; the corresponding codon is recurrently mutated in human cancer. In contrast, T-cell lymphomas from the predisposed breeds, boxers and golden retrievers, show little overlap in their mutation pattern, sharing only one of their 15 most recurrently mutated genes. Boxers, which develop aggressive T-cell lymphomas, are typically mutated in the PTEN-mTOR pathway. T-cell lymphomas in golden retrievers are often less aggressive, and their tumors typically showed mutations in genes involved in cellular metabolism. We identify genes with known involvement in human lymphoma and leukemia, genes implicated in other human cancers, as well as novel genes that could allow new therapeutic options., (© 2015 Elvers et al. Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2015
- Full Text
- View/download PDF
44. Expression profiles of 151 pediatric low-grade gliomas reveal molecular differences associated with location and histological subtype.
- Author
-
Bergthold G, Bandopadhayay P, Hoshida Y, Ramkissoon S, Ramkissoon L, Rich B, Maire CL, Paolella BR, Schumacher SE, Tabak B, Ferrer-Luna R, Ozek M, Sav A, Santagata S, Wen PY, Goumnerova LC, Ligon AH, Stiles C, Segal R, Golub T, Grill J, Ligon KL, Chan JA, Kieran MW, and Beroukhim R
- Subjects
- Adolescent, Child, Child, Preschool, Cluster Analysis, Female, Gene Expression Profiling, Humans, Immunohistochemistry, In Situ Hybridization, Fluorescence, Infant, Infant, Newborn, Male, Neoplasm Grading, Oligonucleotide Array Sequence Analysis, Brain Neoplasms genetics, Brain Neoplasms pathology, Glioma genetics, Glioma pathology, Transcriptome
- Abstract
Background: Pediatric low-grade gliomas (PLGGs), the most frequent pediatric brain tumor, comprise a heterogeneous group of diseases. Recent genomic analyses suggest that these tumors are mostly driven by mitogene-activated protein kinase (MAPK) pathway alterations. However, little is known about the molecular characteristics inherent to their clinical and histological heterogeneity., Methods: We performed gene expression profiling on 151 paraffin-embedded PLGGs from different locations, ages, and histologies. Using unsupervised and supervised analyses, we compared molecular features with age, location, histology, and BRAF genomic status. We compared molecular differences with normal pediatric brain expression profiles to observe whether those patterns were mirrored in normal brain., Results: Unsupervised clustering distinguished 3 molecular groups that correlated with location in the brain and histological subtype. "Not otherwise specified" (NOS) tumors did not constitute a unified class. Supratentorial pilocytic astrocytomas (PAs) were significantly enriched with genes involved in pathways related to inflammatory activity compared with infratentorial tumors. Differences based on tumor location were not mirrored in location-dependent differences in expression within normal brain tissue. We identified significant differences between supratentorial PAs and diffuse astrocytomas as well as between supratentorial PAs and dysembryoplastic neuroepithelial tumors but not between supratentorial PAs and gangliogliomas. Similar expression patterns were observed between childhood and adolescent PAs. We identified differences between BRAF-duplicated and V600E-mutated tumors but not between primary and recurrent PLGGs., Conclusion: Expression profiling of PLGGs reveals significant differences associated with tumor location, histology, and BRAF genomic status. Supratentorial PAs, in particular, are enriched in inflammatory pathways that appear to be tumor-related., (© The Author(s) 2015. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
45. Clinical implementation of integrated whole-genome copy number and mutation profiling for glioblastoma.
- Author
-
Ramkissoon SH, Bi WL, Schumacher SE, Ramkissoon LA, Haidar S, Knoff D, Dubuc A, Brown L, Burns M, Cryan JB, Abedalthagafi M, Kang YJ, Schultz N, Reardon DA, Lee EQ, Rinne ML, Norden AD, Nayak L, Ruland S, Doherty LM, LaFrankie DC, Horvath M, Aizer AA, Russo A, Arvold ND, Claus EB, Al-Mefty O, Johnson MD, Golby AJ, Dunn IF, Chiocca EA, Trippa L, Santagata S, Folkerth RD, Kantoff P, Rollins BJ, Lindeman NI, Wen PY, Ligon AH, Beroukhim R, Alexander BM, and Ligon KL
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Child, Child, Preschool, Comparative Genomic Hybridization, Female, Gene Expression Profiling, Genotype, Humans, Infant, Isocitrate Dehydrogenase genetics, Male, Middle Aged, PTEN Phosphohydrolase genetics, Prospective Studies, Tumor Suppressor Protein p53 genetics, Young Adult, Brain Neoplasms diagnosis, Brain Neoplasms genetics, DNA Copy Number Variations, Genome-Wide Association Study, Glioblastoma diagnosis, Glioblastoma genetics, Mutation
- Abstract
Background: Multidimensional genotyping of formalin-fixed paraffin-embedded (FFPE) samples has the potential to improve diagnostics and clinical trials for brain tumors, but prospective use in the clinical setting is not yet routine. We report our experience with implementing a multiplexed copy number and mutation-testing program in a diagnostic laboratory certified by the Clinical Laboratory Improvement Amendments., Methods: We collected and analyzed clinical testing results from whole-genome array comparative genomic hybridization (OncoCopy) of 420 brain tumors, including 148 glioblastomas. Mass spectrometry-based mutation genotyping (OncoMap, 471 mutations) was performed on 86 glioblastomas., Results: OncoCopy was successful in 99% of samples for which sufficient DNA was obtained (n = 415). All clinically relevant loci for glioblastomas were detected, including amplifications (EGFR, PDGFRA, MET) and deletions (EGFRvIII, PTEN, 1p/19q). Glioblastoma patients ≤40 years old had distinct profiles compared with patients >40 years. OncoMap testing reliably identified mutations in IDH1, TP53, and PTEN. Seventy-seven glioblastoma patients enrolled on trials, of whom 51% participated in targeted therapeutic trials where multiplex data informed eligibility or outcomes. Data integration identified patients with complete tumor suppressor inactivation, albeit rarely (5% of patients) due to lack of whole-gene coverage in OncoMap., Conclusions: Combined use of multiplexed copy number and mutation detection from FFPE samples in the clinical setting can efficiently replace singleton tests for clinical diagnosis and prognosis in most settings. Our results support incorporation of these assays into clinical trials as integral biomarkers and their potential to impact interpretation of results. Limited tumor suppressor variant capture by targeted genotyping highlights the need for whole-gene sequencing in glioblastoma., (© The Author(s) 2015. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
46. Comprehensive, Integrative Genomic Analysis of Diffuse Lower-Grade Gliomas.
- Author
-
Brat DJ, Verhaak RG, Aldape KD, Yung WK, Salama SR, Cooper LA, Rheinbay E, Miller CR, Vitucci M, Morozova O, Robertson AG, Noushmehr H, Laird PW, Cherniack AD, Akbani R, Huse JT, Ciriello G, Poisson LM, Barnholtz-Sloan JS, Berger MS, Brennan C, Colen RR, Colman H, Flanders AE, Giannini C, Grifford M, Iavarone A, Jain R, Joseph I, Kim J, Kasaian K, Mikkelsen T, Murray BA, O'Neill BP, Pachter L, Parsons DW, Sougnez C, Sulman EP, Vandenberg SR, Van Meir EG, von Deimling A, Zhang H, Crain D, Lau K, Mallery D, Morris S, Paulauskis J, Penny R, Shelton T, Sherman M, Yena P, Black A, Bowen J, Dicostanzo K, Gastier-Foster J, Leraas KM, Lichtenberg TM, Pierson CR, Ramirez NC, Taylor C, Weaver S, Wise L, Zmuda E, Davidsen T, Demchok JA, Eley G, Ferguson ML, Hutter CM, Mills Shaw KR, Ozenberger BA, Sheth M, Sofia HJ, Tarnuzzer R, Wang Z, Yang L, Zenklusen JC, Ayala B, Baboud J, Chudamani S, Jensen MA, Liu J, Pihl T, Raman R, Wan Y, Wu Y, Ally A, Auman JT, Balasundaram M, Balu S, Baylin SB, Beroukhim R, Bootwalla MS, Bowlby R, Bristow CA, Brooks D, Butterfield Y, Carlsen R, Carter S, Chin L, Chu A, Chuah E, Cibulskis K, Clarke A, Coetzee SG, Dhalla N, Fennell T, Fisher S, Gabriel S, Getz G, Gibbs R, Guin R, Hadjipanayis A, Hayes DN, Hinoue T, Hoadley K, Holt RA, Hoyle AP, Jefferys SR, Jones S, Jones CD, Kucherlapati R, Lai PH, Lander E, Lee S, Lichtenstein L, Ma Y, Maglinte DT, Mahadeshwar HS, Marra MA, Mayo M, Meng S, Meyerson ML, Mieczkowski PA, Moore RA, Mose LE, Mungall AJ, Pantazi A, Parfenov M, Park PJ, Parker JS, Perou CM, Protopopov A, Ren X, Roach J, Sabedot TS, Schein J, Schumacher SE, Seidman JG, Seth S, Shen H, Simons JV, Sipahimalani P, Soloway MG, Song X, Sun H, Tabak B, Tam A, Tan D, Tang J, Thiessen N, Triche T Jr, Van Den Berg DJ, Veluvolu U, Waring S, Weisenberger DJ, Wilkerson MD, Wong T, Wu J, Xi L, Xu AW, Yang L, Zack TI, Zhang J, Aksoy BA, Arachchi H, Benz C, Bernard B, Carlin D, Cho J, DiCara D, Frazer S, Fuller GN, Gao J, Gehlenborg N, Haussler D, Heiman DI, Iype L, Jacobsen A, Ju Z, Katzman S, Kim H, Knijnenburg T, Kreisberg RB, Lawrence MS, Lee W, Leinonen K, Lin P, Ling S, Liu W, Liu Y, Liu Y, Lu Y, Mills G, Ng S, Noble MS, Paull E, Rao A, Reynolds S, Saksena G, Sanborn Z, Sander C, Schultz N, Senbabaoglu Y, Shen R, Shmulevich I, Sinha R, Stuart J, Sumer SO, Sun Y, Tasman N, Taylor BS, Voet D, Weinhold N, Weinstein JN, Yang D, Yoshihara K, Zheng S, Zhang W, Zou L, Abel T, Sadeghi S, Cohen ML, Eschbacher J, Hattab EM, Raghunathan A, Schniederjan MJ, Aziz D, Barnett G, Barrett W, Bigner DD, Boice L, Brewer C, Calatozzolo C, Campos B, Carlotti CG Jr, Chan TA, Cuppini L, Curley E, Cuzzubbo S, Devine K, DiMeco F, Duell R, Elder JB, Fehrenbach A, Finocchiaro G, Friedman W, Fulop J, Gardner J, Hermes B, Herold-Mende C, Jungk C, Kendler A, Lehman NL, Lipp E, Liu O, Mandt R, McGraw M, Mclendon R, McPherson C, Neder L, Nguyen P, Noss A, Nunziata R, Ostrom QT, Palmer C, Perin A, Pollo B, Potapov A, Potapova O, Rathmell WK, Rotin D, Scarpace L, Schilero C, Senecal K, Shimmel K, Shurkhay V, Sifri S, Singh R, Sloan AE, Smolenski K, Staugaitis SM, Steele R, Thorne L, Tirapelli DP, Unterberg A, Vallurupalli M, Wang Y, Warnick R, Williams F, Wolinsky Y, Bell S, Rosenberg M, Stewart C, Huang F, Grimsby JL, Radenbaugh AJ, and Zhang J
- Subjects
- Adolescent, Adult, Aged, Chromosomes, Human, Pair 1, Chromosomes, Human, Pair 19, Cluster Analysis, Female, Glioblastoma genetics, Glioma metabolism, Glioma mortality, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Neoplasm Grading, Proportional Hazards Models, Sequence Analysis, DNA, Signal Transduction, DNA, Neoplasm analysis, Genes, p53, Glioma genetics, Mutation
- Abstract
Background: Diffuse low-grade and intermediate-grade gliomas (which together make up the lower-grade gliomas, World Health Organization grades II and III) have highly variable clinical behavior that is not adequately predicted on the basis of histologic class. Some are indolent; others quickly progress to glioblastoma. The uncertainty is compounded by interobserver variability in histologic diagnosis. Mutations in IDH, TP53, and ATRX and codeletion of chromosome arms 1p and 19q (1p/19q codeletion) have been implicated as clinically relevant markers of lower-grade gliomas., Methods: We performed genomewide analyses of 293 lower-grade gliomas from adults, incorporating exome sequence, DNA copy number, DNA methylation, messenger RNA expression, microRNA expression, and targeted protein expression. These data were integrated and tested for correlation with clinical outcomes., Results: Unsupervised clustering of mutations and data from RNA, DNA-copy-number, and DNA-methylation platforms uncovered concordant classification of three robust, nonoverlapping, prognostically significant subtypes of lower-grade glioma that were captured more accurately by IDH, 1p/19q, and TP53 status than by histologic class. Patients who had lower-grade gliomas with an IDH mutation and 1p/19q codeletion had the most favorable clinical outcomes. Their gliomas harbored mutations in CIC, FUBP1, NOTCH1, and the TERT promoter. Nearly all lower-grade gliomas with IDH mutations and no 1p/19q codeletion had mutations in TP53 (94%) and ATRX inactivation (86%). The large majority of lower-grade gliomas without an IDH mutation had genomic aberrations and clinical behavior strikingly similar to those found in primary glioblastoma., Conclusions: The integration of genomewide data from multiple platforms delineated three molecular classes of lower-grade gliomas that were more concordant with IDH, 1p/19q, and TP53 status than with histologic class. Lower-grade gliomas with an IDH mutation either had 1p/19q codeletion or carried a TP53 mutation. Most lower-grade gliomas without an IDH mutation were molecularly and clinically similar to glioblastoma. (Funded by the National Institutes of Health.).
- Published
- 2015
- Full Text
- View/download PDF
47. Pan-cancer genetic analysis identifies PARK2 as a master regulator of G1/S cyclins.
- Author
-
Gong Y, Zack TI, Morris LG, Lin K, Hukkelhoven E, Raheja R, Tan IL, Turcan S, Veeriah S, Meng S, Viale A, Schumacher SE, Palmedo P, Beroukhim R, and Chan TA
- Subjects
- Animals, Cell Line, Tumor, Cyclin-Dependent Kinase Inhibitor p16 metabolism, G1 Phase, Gene Deletion, Gene Expression Profiling, Genes, Tumor Suppressor, Genome, Human, Genomics, Humans, Insecta, RNA, Small Interfering metabolism, S Phase, Cell Cycle, Cyclin D1 metabolism, Cyclin E metabolism, Cyclin-Dependent Kinase 4 metabolism, Gene Expression Regulation, Neoplastic, Oncogene Proteins metabolism, Ubiquitin-Protein Ligases genetics
- Abstract
Coordinate control of different classes of cyclins is fundamentally important for cell cycle regulation and tumor suppression, yet the underlying mechanisms are incompletely understood. Here we show that the PARK2 tumor suppressor mediates this coordination. The PARK2 E3 ubiquitin ligase coordinately controls the stability of both cyclin D and cyclin E. Analysis of approximately 5,000 tumor genomes shows that PARK2 is a very frequently deleted gene in human cancer and uncovers a striking pattern of mutual exclusivity between PARK2 deletion and amplification of CCND1, CCNE1 or CDK4-implicating these genes in a common pathway. Inactivation of PARK2 results in the accumulation of cyclin D and acceleration of cell cycle progression. Furthermore, PARK2 is a component of a new class of cullin-RING-containing ubiquitin ligases targeting both cyclin D and cyclin E for degradation. Thus, PARK2 regulates cyclin-CDK complexes, as does the CDK inhibitor p16, but acts as a master regulator of the stability of G1/S cyclins.
- Published
- 2014
- Full Text
- View/download PDF
48. BET bromodomain inhibition of MYC-amplified medulloblastoma.
- Author
-
Bandopadhayay P, Bergthold G, Nguyen B, Schubert S, Gholamin S, Tang Y, Bolin S, Schumacher SE, Zeid R, Masoud S, Yu F, Vue N, Gibson WJ, Paolella BR, Mitra SS, Cheshier SH, Qi J, Liu KW, Wechsler-Reya R, Weiss WA, Swartling FJ, Kieran MW, Bradner JE, Beroukhim R, and Cho YJ
- Subjects
- Animals, Cell Cycle Proteins, Cell Line, Tumor, Cell Survival, G1 Phase Cell Cycle Checkpoints, Gene Amplification, Gene Dosage, Humans, Medulloblastoma metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Nuclear Proteins metabolism, Signal Transduction, Transcription Factors metabolism, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Azepines pharmacology, Medulloblastoma drug therapy, Nuclear Proteins antagonists & inhibitors, Proto-Oncogene Proteins c-myc genetics, Transcription Factors antagonists & inhibitors, Triazoles pharmacology
- Abstract
Purpose: MYC-amplified medulloblastomas are highly lethal tumors. Bromodomain and extraterminal (BET) bromodomain inhibition has recently been shown to suppress MYC-associated transcriptional activity in other cancers. The compound JQ1 inhibits BET bromodomain-containing proteins, including BRD4. Here, we investigate BET bromodomain targeting for the treatment of MYC-amplified medulloblastoma., Experimental Design: We evaluated the effects of genetic and pharmacologic inhibition of BET bromodomains on proliferation, cell cycle, and apoptosis in established and newly generated patient- and genetically engineered mouse model (GEMM)-derived medulloblastoma cell lines and xenografts that harbored amplifications of MYC or MYCN. We also assessed the effect of JQ1 on MYC expression and global MYC-associated transcriptional activity. We assessed the in vivo efficacy of JQ1 in orthotopic xenografts established in immunocompromised mice., Results: Treatment of MYC-amplified medulloblastoma cells with JQ1 decreased cell viability associated with arrest at G1 and apoptosis. We observed downregulation of MYC expression and confirmed the inhibition of MYC-associated transcriptional targets. The exogenous expression of MYC from a retroviral promoter reduced the effect of JQ1 on cell viability, suggesting that attenuated levels of MYC contribute to the functional effects of JQ1. JQ1 significantly prolonged the survival of orthotopic xenograft models of MYC-amplified medulloblastoma (P < 0.001). Xenografts harvested from mice after five doses of JQ1 had reduced the expression of MYC mRNA and a reduced proliferative index., Conclusion: JQ1 suppresses MYC expression and MYC-associated transcriptional activity in medulloblastomas, resulting in an overall decrease in medulloblastoma cell viability. These preclinical findings highlight the promise of BET bromodomain inhibitors as novel agents for MYC-amplified medulloblastoma., (©2013 AACR)
- Published
- 2014
- Full Text
- View/download PDF
49. Widespread genetic heterogeneity in multiple myeloma: implications for targeted therapy.
- Author
-
Lohr JG, Stojanov P, Carter SL, Cruz-Gordillo P, Lawrence MS, Auclair D, Sougnez C, Knoechel B, Gould J, Saksena G, Cibulskis K, McKenna A, Chapman MA, Straussman R, Levy J, Perkins LM, Keats JJ, Schumacher SE, Rosenberg M, Getz G, and Golub TR
- Subjects
- Blotting, Western, Gene Dosage, Humans, Mutation, Sequence Analysis, DNA, Genetic Heterogeneity, Multiple Myeloma genetics
- Abstract
We performed massively parallel sequencing of paired tumor/normal samples from 203 multiple myeloma (MM) patients and identified significantly mutated genes and copy number alterations and discovered putative tumor suppressor genes by determining homozygous deletions and loss of heterozygosity. We observed frequent mutations in KRAS (particularly in previously treated patients), NRAS, BRAF, FAM46C, TP53, and DIS3 (particularly in nonhyperdiploid MM). Mutations were often present in subclonal populations, and multiple mutations within the same pathway (e.g., KRAS, NRAS, and BRAF) were observed in the same patient. In vitro modeling predicts only partial treatment efficacy of targeting subclonal mutations, and even growth promotion of nonmutated subclones in some cases. These results emphasize the importance of heterogeneity analysis for treatment decisions., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
50. Pan-cancer patterns of somatic copy number alteration.
- Author
-
Zack TI, Schumacher SE, Carter SL, Cherniack AD, Saksena G, Tabak B, Lawrence MS, Zhsng CZ, Wala J, Mermel CH, Sougnez C, Gabriel SB, Hernandez B, Shen H, Laird PW, Getz G, Meyerson M, and Beroukhim R
- Subjects
- Epigenesis, Genetic, Gene Regulatory Networks, Genetic Association Studies, Genetic Predisposition to Disease, Humans, Mutagenesis, Ploidies, DNA Copy Number Variations, Neoplasms genetics
- Abstract
Determining how somatic copy number alterations (SCNAs) promote cancer is an important goal. We characterized SCNA patterns in 4,934 cancers from The Cancer Genome Atlas Pan-Cancer data set. Whole-genome doubling, observed in 37% of cancers, was associated with higher rates of every other type of SCNA, TP53 mutations, CCNE1 amplifications and alterations of the PPP2R complex. SCNAs that were internal to chromosomes tended to be shorter than telomere-bounded SCNAs, suggesting different mechanisms underlying their generation. Significantly recurrent focal SCNAs were observed in 140 regions, including 102 without known oncogene or tumor suppressor gene targets and 50 with significantly mutated genes. Amplified regions without known oncogenes were enriched for genes involved in epigenetic regulation. When levels of genomic disruption were accounted for, 7% of region pairs were anticorrelated, and these regions tended to encompass genes whose proteins physically interact, suggesting related functions. These results provide insights into mechanisms of generation and functional consequences of cancer-related SCNAs.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.