Deterministic Evolutionary Trajectories Influence Primary Tumor Growth: TRACERx Renal

Samra Turajlic, Hang Xu, Kevin Litchfield, Andrew Rowan, Stuart Horswell, Tim Chambers, Tim O'Brien, Jose I Lopez, Thomas B K Watkins, David Nicol, Mark Stares, Ben Challacombe, Steve Hazell, Ashish Chandra, Thomas J Mitchell, Lewis Au, Claudia Eichler-Jonsson, Faiz Jabbar, Aspasia Soultati, Simon Chowdhury, Sarah Rudman, Joanna Lynch, Archana Fernando, Gordon Stamp, Emma Nye, Aengus Stewart, Wei Xing, Jonathan C Smith, Mickael Escudero, Adam Huffman, Nik Matthews, Greg Elgar, Ben Phillimore, Marta Costa, Sharmin Begum, Sophia Ward, Max Salm, Stefan Boeing, Rosalie Fisher, Lavinia Spain, Carolina Navas, Eva Grönroos, Sebastijan Hobor, Sarkhara Sharma, Ismaeel Aurangzeb, Sharanpreet Lall, Alexander Polson, Mary Varia, Catherine Horsfield, Nicos Fotiadis, Lisa Pickering, Roland F Schwarz, Bruno Silva, Javier Herrero, Nick M Luscombe, Mariam Jamal-Hanjani, Rachel Rosenthal, Nicolai J Birkbak, Gareth A Wilson, Orsolya Pipek, Dezso Ribli, Marcin Krzystanek, Istvan Csabai, Zoltan Szallasi, Martin Gore, Nicholas McGranahan, Peter Van Loo, Peter Campbell, James Larkin, Charles Swanton, TRACERx Renal Consortium, Samra Turajlic, Hang Xu, Kevin Litchfield, Andrew Rowan, Stuart Horswell, Tim Chambers, Tim O'Brien, Jose I Lopez, Thomas B K Watkins, David Nicol, Mark Stares, Ben Challacombe, Steve Hazell, Ashish Chandra, Thomas J Mitchell, Lewis Au, Claudia Eichler-Jonsson, Faiz Jabbar, Aspasia Soultati, Simon Chowdhury, Sarah Rudman, Joanna Lynch, Archana Fernando, Gordon Stamp, Emma Nye, Aengus Stewart, Wei Xing, Jonathan C Smith, Mickael Escudero, Adam Huffman, Nik Matthews, Greg Elgar, Ben Phillimore, Marta Costa, Sharmin Begum, Sophia Ward, Max Salm, Stefan Boeing, Rosalie Fisher, Lavinia Spain, Carolina Navas, Eva Grönroos, Sebastijan Hobor, Sarkhara Sharma, Ismaeel Aurangzeb, Sharanpreet Lall, Alexander Polson, Mary Varia, Catherine Horsfield, Nicos Fotiadis, Lisa Pickering, Roland F Schwarz, Bruno Silva, Javier Herrero, Nick M Luscombe, Mariam Jamal-Hanjani, Rachel Rosenthal, Nicolai J Birkbak, Gareth A Wilson, Orsolya Pipek, Dezso Ribli, Marcin Krzystanek, Istvan Csabai, Zoltan Szallasi, Martin Gore, Nicholas McGranahan, Peter Van Loo, Peter Campbell, James Larkin, Charles Swanton, TRACERx Renal Consortium

Abstract

The evolutionary features of clear-cell renal cell carcinoma (ccRCC) have not been systematically studied to date. We analyzed 1,206 primary tumor regions from 101 patients recruited into the multi-center prospective study, TRACERx Renal. We observe up to 30 driver events per tumor and show that subclonal diversification is associated with known prognostic parameters. By resolving the patterns of driver event ordering, co-occurrence, and mutual exclusivity at clone level, we show the deterministic nature of clonal evolution. ccRCC can be grouped into seven evolutionary subtypes, ranging from tumors characterized by early fixation of multiple mutational and copy number drivers and rapid metastases to highly branched tumors with >10 subclonal drivers and extensive parallel evolution associated with attenuated progression. We identify genetic diversity and chromosomal complexity as determinants of patient outcome. Our insights reconcile the variable clinical behavior of ccRCC and suggest evolutionary potential as a biomarker for both intervention and surveillance.

Trial registration: ClinicalTrials.gov NCT03226886.

Keywords: branched evolution; cancer evolution; chromosome instability; deterministic evolution; intratumor heterogeneity; linear evolution; metastasis; punctuated evolution; renal cell cancer; tumor diversity.

Copyright © 2018 Francis Crick Institute. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure S1
Figure S1
Consort Diagram, Related to STAR Methods (A and B) (A) shows the Consort diagram for the filtering steps leading to the reported cohort; (B) shows the summary of Driver Panel, Whole Exome and Whole Genome Sequencing in the TRACERx Renal 101 Cohort.
Figure 1
Figure 1
Overview (A) Overview of somatic driver alterations, including SNVs, DNVs, INDELs, and SCNAs, detected in the tumors of 101 TRACERx Renal cases. Rectangles and triangles indicate clonal and subclonal alterations, respectively. Parallel evolution is indicated in orange with a split indicating 2 or more parallel events. Five bilateral/multi-focal cases are shown on the right; distinct VHL mutations within tumor pairs are indicated with an asterisk. (B) Mutational frequency in 14 key driver genes in the TRACERx Renal cohort and three single biopsy ccRCC studies (TCGA KIRC, Sato et al. [2013], and Scelo et al. [2014]). Clonal mutations are shown in the darker shade, subclonal in lighter. (C) Frequency of SCNAs in the TRACERx Renal cohort. Copy number gains and losses are indicated in red and blue respectively. Clonal SCNAs are shown in darker and subclonal SCNAs in lighter shade of color. Putative driver copy number altered regions are annotated. The dotted line indicates the frequency of the same SCNAs in the TCGA KIRC cohort. See also Tables S1 and S2 and Data S1, S2, S3, and S4.
Figure 2
Figure 2
Driver Phylogenetic Trees Driver phylogenetic trees for each tumor (or multiple tumors from the same patient) are shown. The trees are ordered by the overall tumors stage: I–IV. The founding clone is indicated in light blue, with subsequent sub clones shown in distinct colors. The size of each node represents the number of SCNAs detected within that subclone. The length of lines connecting tumor subclones does not contain information. See also Data S2.
Figure 3
Figure 3
Parallel Evolution Table shows driver gene events with >10 subclonal mutations across the cohort. These genes were tested for evidence of parallel evolution using a permutation model accounting for overall gene mutation frequency and the number of biopsies per tumor (see STAR Methods). BAP1, SETD2, and PTEN were found to show significant evidence of parallel evolution (p < 0.05, FDR < 0.1). Example driver trees and accompanying tumor sampling images are presented for each significant gene: BAP1, PTEN, and SETD2. Parallel events are marked on the driver trees and clone color is matched from the tree to the corresponding sampled tumor region. See also Data S3.
Figure 4
Figure 4
Conserved Features of ccRCC Evolution (A) Event co-occurrence analysis, with red indicating enrichment for co-occurrence and blue for mutual exclusivity. Values are log2(observed no. of co-occurrences/expected no. of co-occurrences, STAR Methods), with significant patterns marked according to the legend. Data are shown for event co-occurrence/mutually exclusivity, in first truncal clones only per case (bottom left) and second all terminal subclones (top right) such that all clonal and subclonal interactions are considered (see STAR Methods). p values are calculated under a probabilistic model, as implemented in R package “co-occur,” with only interactions significant in both “clonal” and “clonal + subclonal” analyses are considered significant. (B) Molecular clock timing analysis from the whole genome sequenced cohort, with time from the most recent common ancestor (MRCA) to tumor diagnosis plotted on the x axis. On the y axis are cases split into three groups, based on having one, two or three clonal driver events. VHL wild type cases (n = 2) are excluded on account of their distinct etiological and phenotypic profile. p value is assessed using a linear model, adjusting for the total clonal mutation burden per tumor. (C) Same y axis patient groups as (B), but plotted on the x axis is tumor size (mm). p value is based on Kruskal-Wallis test. (D) On the y axis, all cases from the 100-patient cohort, again VHL wild-type cases were then excluded, and remaining cases were split into three groups based on one, two, or three clonal driver mutations. Multi-region data on % of cells staining positive for proliferation marker Ki67 is shown on the x axis. p value is based on a linear mixed effect model to account for non-independence of multiple observations per tumor. (E) Left: an illustrative schematic tree to demonstrate the method used to trace each tumor’s evolutionary paths. Right: results from the event ordering analysis for all pairs of events with n = 10 or more observations. Plotted are the counts of instances where: event 1 was found to precede event 2, and event 1 was found to follow event 2. Significance was tested using a binomial test with p values shown after correction for multiple testing using Benjamini-Hochberg procedure. See also Figure S2 and Table S3.
Figure S2
Figure S2
SCNAs Co-occurring with Mutational Driver Events, Related to Figure 4 (A–D) (A) shows SCNAs co-occurring with mutational driver events in TRACERx Renal cohort. (B) shows SCNA co-occurrence in TCGA KIRC cohort. (C) shows 14q loss co-occurring with the other SCNAs. 14q loss is shown on X-axis and on Y-axis is log(p-value) for co-occurrence. (D) shows observed versus expected co-occurrence frequencies.
Figure 5
Figure 5
Evolutionary Subtypes Cases grouped by evolutionary subtype, with the following parameters also annotated: presence of clonal wGII (blue > median, white ≤ median), presence of subclonal wGII (blue > median, white ≤ median), ITH index score (red > median, white ≤ median), and tumor size (mm) (range [18–180], white = low, black = high). Occurrences of parallel evolution are denoted in the heatmap with “P.” Plotted next is the distribution of stages per subtype, followed by grade, colored as per the legend, and then a further six metrics are summarized as the average values for each group: (1) mean number of tumor clones, (2) % of patients with grade 4 disease, (3) % of patients with microvascular invasion, (4) mean % of cells staining positive for Ki67 proliferation index (mean calculated first per class and then across the cohort), (5) % of patients with disease relapse/progression, and (6) relapse/progression time. Shown next are relapse/progression-free survival plots per group, and shown last are three example driver phylogenetic trees from each group. See also Figure S3.
Figure S3
Figure S3
TRACERx Renal Cohort Unsupervised Clustering Analysis of Evolutionary Features, Related to Figure 5 and STAR Methods On the x-axis are the rule based evolutionary subtype groups, and on the y-axis are group assignments based on unsupervised clustering. Shown below the x-axis is the percentage of members, from each evolutionary subtype, which are assigned to the same unsupervised cluster. Colours have no meaning except to denote different groups.
Figure 6
Figure 6
Intratumor Heterogeneity Index and Saturation Analysis (A) Number of tumor biopsies profiled (x axis) versus the number of driver events (i.e. all gene mutations and SCNAs shown in Figure 1A) discovered (y axis) for densely sampled (20+ biopsies) cases. (B) Saturation curves for all cases with ≥15 biopsies, with biopsy number plotted on x axis and proportion of the total driver events detected (from all biopsies) on y axis, increasing with each additional biopsy taken. Data are shown for all cases and tumors split based on low and high ITH (above/below median). (C) Boxplot summary of the absolute number (top) of biopsies needed to detect ≥0.75 of driver events for tumors grouped by evolutionary subtype. Also shown (bottom) is the proportion of biopsies needed (out of the total number taken from each tumor) to normalize for absolute biopsy count. (D) Illustration of the potential errors arising from a two-site biopsy approach: considering all pairs of biopsies, plotted on the x axis is the mean number of subclonal driver events misidentified as clonal (illusion of clonality), on y axis is the number of subclonal driver events missed entirely. Data are shown for three clinical scenarios. Left: small renal masses (size,

Figure 7

Clinical Endpoints (A) Kaplan-Meier plots…

Figure 7

Clinical Endpoints (A) Kaplan-Meier plots for progression free survival (PFS) in the TRACERx…

Figure 7
Clinical Endpoints (A) Kaplan-Meier plots for progression free survival (PFS) in the TRACERx Renal cohort (three plots in top row) and for overall survival (OS) in TCGA KIRC cohort (three plots in bottom row). Three groupings are plotted for each cohort. Left: high (>median) versus low ITH index. Middle: high (>median) versus low wGII. Right: four group high/low combination groupings of the two metrics. Log-rank and adjusted (for stage and grade as covariates in a Cox proportional hazard model) p values are stated. (B) Proportion of cases, within each of the high/low four groups, that progressed to disseminated versus solitary metastases, based on each patient’s first progression event. Counts in the highest group “low ITH, high wGII,” were compared to all other groups through Fisher’s exact test. (C) Cancer-related deaths OS analysis (as opposed to PFS shown in A) for the TRACERx Renal cohort, with patients grouped using the four-category high/low ITH/wGII system. Log-rank and adjusted (for stage and grade as covariates in a Cox proportional hazard model) p values are stated. See also Table S4.
All figures (11)
Figure 7
Figure 7
Clinical Endpoints (A) Kaplan-Meier plots for progression free survival (PFS) in the TRACERx Renal cohort (three plots in top row) and for overall survival (OS) in TCGA KIRC cohort (three plots in bottom row). Three groupings are plotted for each cohort. Left: high (>median) versus low ITH index. Middle: high (>median) versus low wGII. Right: four group high/low combination groupings of the two metrics. Log-rank and adjusted (for stage and grade as covariates in a Cox proportional hazard model) p values are stated. (B) Proportion of cases, within each of the high/low four groups, that progressed to disseminated versus solitary metastases, based on each patient’s first progression event. Counts in the highest group “low ITH, high wGII,” were compared to all other groups through Fisher’s exact test. (C) Cancer-related deaths OS analysis (as opposed to PFS shown in A) for the TRACERx Renal cohort, with patients grouped using the four-category high/low ITH/wGII system. Log-rank and adjusted (for stage and grade as covariates in a Cox proportional hazard model) p values are stated. See also Table S4.

References

    1. Arai E., Sakamoto H., Ichikawa H., Totsuka H., Chiku S., Gotoh M., Mori T., Nakatani T., Ohnami S., Nakagawa T. Multilayer-omics analysis of renal cell carcinoma, including the whole exome, methylome and transcriptome. Int. J. Cancer. 2014;135:1330–1342.
    1. Beroukhim R., Brunet J.P., Di Napoli A., Mertz K.D., Seeley A., Pires M.M., Linhart D., Worrell R.A., Moch H., Rubin M.A. Patterns of gene expression and copy-number alterations in von-hippel lindau disease-associated and sporadic clear cell carcinoma of the kidney. Cancer Res. 2009;69:4674–4681.
    1. Bex A., Ljungberg B., van Poppel H., Powles T., European Association of Urology The role of cytoreductive nephrectomy: European Association of Urology recommendations in 2016. Eur. Urol. 2016;70:901–905.
    1. Brastianos P.K., Carter S.L., Santagata S., Cahill D.P., Taylor-Weiner A., Jones R.T., Van Allen E.M., Lawrence M.S., Horowitz P.M., Cibulskis K. Genomic characterization of brain metastases reveals branched evolution and potential therapeutic targets. Cancer Discov. 2015;5:1164–1177.
    1. Cancer Genome Atlas Research Network Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499:43–49.
    1. Carter S.L., Cibulskis K., Helman E., McKenna A., Shen H., Zack T., Laird P.W., Onofrio R.C., Winckler W., Weir B.A. Absolute quantification of somatic DNA alterations in human cancer. Nat. Biotechnol. 2012;30:413–421.
    1. Chen F., Zhang Y., Şenbabaoğlu Y., Ciriello G., Yang L., Reznik E., Shuch B., Micevic G., De Velasco G., Shinbrot E. Multilevel genomics-based taxonomy of renal cell carcinoma. Cell Rep. 2016;14:2476–2489.
    1. Cibulskis K., Lawrence M.S., Carter S.L., Sivachenko A., Jaffe D., Sougnez C., Gabriel S., Meyerson M., Lander E.S., Getz G. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 2013;31:213–219.
    1. da Costa W.H., Moniz R.R., da Cunha I.W., Fonseca F.P., Guimaraes G.C., de Cássio Zequi S. Impact of renal vein invasion and fat invasion in pT3a renal cell carcinoma. BJU Int. 2012;109:544–548.
    1. Dalgliesh G.L., Furge K., Greenman C., Chen L., Bignell G., Butler A., Davies H., Edkins S., Hardy C., Latimer C. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature. 2010;463:360–363.
    1. Davis C.F., Ricketts C.J., Wang M., Yang L., Cherniack A.D., Shen H., Buhay C., Kang H., Kim S.C., Fahey C.C. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell. 2014;26:319–330.
    1. De Sano L., Caravagna G., Ramazzotti D., Graudenzi A., Mauri G., Mishra B., Antoniotti M. TRONCO: an R package for the inference of cancer progression models from heterogeneous genomic data. Bioinformatics. 2016;32:1911–1913.
    1. Delahunt B., Cheville J.C., Martignoni G., Humphrey P.A., Magi-Galluzzi C., McKenney J., Egevad L., Algaba F., Moch H., Grignon D.J., Members of the ISUP Renal Tumor Panel The International Society of Urological Pathology (ISUP) grading system for renal cell carcinoma and other prognostic parameters. Am. J. Surg. Pathol. 2013;37:1490–1504.
    1. Eldredge N., Gould S.J. On punctuated equilibria. Science. 1997;276:338–341.
    1. Endesfelder D., Burrell R., Kanu N., McGranahan N., Howell M., Parker P.J., Downward J., Swanton C., Kschischo M. Chromosomal instability selects gene copy-number variants encoding core regulators of proliferation in ER+ breast cancer. Cancer Res. 2014;74:4853–4863.
    1. Fang H., Bergmann E.A., Arora K., Vacic V., Zody M.C., Iossifov I., O’Rawe J.A., Wu Y., Jimenez Barron L.T., Rosenbaum J. Indel variant analysis of short-read sequencing data with Scalpel. Nat. Protoc. 2016;11:2529–2548.
    1. Gabadinho A., Ritschard G., Muller N.S., Studer M. Analyzing and visualizing state sequences in R with TraMineR. J. Stat. Softw. 2011;40:1–37.
    1. Abecasis G.R., Altshuler D., Auton A., Brooks L.D., Durbin R.M., Gibbs R.A., Hurles M.E., McVean G.A., 1000 Genomes Project Consortium A map of human genome variation from population-scale sequencing. Nature. 2010;467:1061–1073.
    1. Gerlinger M., Horswell S., Larkin J., Rowan A.J., Salm M.P., Varela I., Fisher R., McGranahan N., Matthews N., Santos C.R. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet. 2014;46:225–233.
    1. Goldschmidt R. Yale University Press; New Haven, CT: 1940. The Material Basis of Evolution.
    1. Griffith D.M., Veech J.A., Marsh C.J. cooccur: probabilistic species co-occurrence analysis in R. J. Stat. Softw. 2016;69:1–17.
    1. Gu Y.F., Cohn S., Christie A., McKenzie T., Wolff N., Do Q.N., Madhuranthakam A.J., Pedrosa I., Wang T., Dey A. Modeling renal cell carcinoma in mice: Bap1 and Pbrm1 inactivation drive tumor grade. Cancer Discov. 2017;7:900–917.
    1. Hakimi A.A., Ostrovnaya I., Reva B., Schultz N., Chen Y.B., Gonen M., Liu H., Takeda S., Voss M.H., Tickoo S.K., ccRCC Cancer Genome Atlas (KIRC TCGA) Research Network investigators Adverse outcomes in clear cell renal cell carcinoma with mutations of 3p21 epigenetic regulators BAP1 and SETD2: a report by MSKCC and the KIRC TCGA research network. Clin. Cancer Res. 2013;19:3259–3267.
    1. Hakimi A.A., Tickoo S.K., Jacobsen A., Sarungbam J., Sfakianos J.P., Sato Y., Morikawa T., Kume H., Fukayama M., Homma Y. TCEB1-mutated renal cell carcinoma: a distinct genomic and morphological subtype. Mod. Pathol. 2015;28:845–853.
    1. Ito T., Pei J., Dulaimi E., Menges C., Abbosh P.H., Smaldone M.C., Chen D.Y., Greenberg R.E., Kutikov A., Viterbo R. Genomic copy number alterations in renal cell carcinoma with sarcomatoid features. J. Urol. 2016;195:852–858.
    1. Jamal-Hanjani M., Wilson G.A., McGranahan N., Birkbak N.J., Watkins T.B.K., Veeriah S., Shafi S., Johnson D.H., Mitter R., Rosenthal R., TRACERx Consortium Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 2017;376:2109–2121.
    1. Jewett M.A., Mattar K., Basiuk J., Morash C.G., Pautler S.E., Siemens D.R., Tanguay S., Rendon R.A., Gleave M.E., Drachenberg D.E. Active surveillance of small renal masses: progression patterns of early stage kidney cancer. Eur. Urol. 2011;60:39–44.
    1. Joseph R.W., Kapur P., Serie D.J., Parasramka M., Ho T.H., Cheville J.C., Frenkel E., Parker A.S., Brugarolas J. Clear cell renal cell carcinoma subtypes identified by BAP1 and PBRM1 expression. J. Urol. 2016;195:180–187.
    1. Kane C.J., Mallin K., Ritchey J., Cooperberg M.R., Carroll P.R. Renal cell cancer stage migration: analysis of the National Cancer Data Base. Cancer. 2008;113:78–83.
    1. Kapur P., Peña-Llopis S., Christie A., Zhrebker L., Pavía-Jiménez A., Rathmell W.K., Xie X.J., Brugarolas J. Effects on survival of BAP1 and PBRM1 mutations in sporadic clear-cell renal-cell carcinoma: a retrospective analysis with independent validation. Lancet Oncol. 2013;14:159–167.
    1. Koboldt D.C., Chen K., Wylie T., Larson D.E., McLellan M.D., Mardis E.R., Weinstock G.M., Wilson R.K., Ding L. VarScan: variant detection in massively parallel sequencing of individual and pooled samples. Bioinformatics. 2009;25:2283–2285.
    1. Kojima T., Shimazui T., Hinotsu S., Joraku A., Oikawa T., Kawai K., Horie R., Suzuki H., Nagashima R., Yoshikawa K. Decreased expression of CXXC4 promotes a malignant phenotype in renal cell carcinoma by activating Wnt signaling. Oncogene. 2009;28:297–305.
    1. La Rochelle J., Klatte T., Dastane A., Rao N., Seligson D., Said J., Shuch B., Zomorodian N., Kabbinavar F., Belldegrun A., Pantuck A.J. Chromosome 9p deletions identify an aggressive phenotype of clear cell renal cell carcinoma. Cancer. 2010;116:4696–4702.
    1. Leibovich B.C., Blute M.L., Cheville J.C., Lohse C.M., Frank I., Kwon E.D., Weaver A.L., Parker A.S., Zincke H. Prediction of progression after radical nephrectomy for patients with clear cell renal cell carcinoma: a stratification tool for prospective clinical trials. Cancer. 2003;97:1663–1671.
    1. Li H., Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. Malouf G.G., Ali S.M., Wang K., Balasubramanian S., Ross J.S., Miller V.A., Stephens P.J., Khayat D., Pal S.K., Su X. Genomic characterization of renal cell carcinoma with sarcomatoid dedifferentiation pinpoints recurrent genomic alterations. Eur. Urol. 2016;70:348–357.
    1. Martinez P., Birkbak N.J., Gerlinger M., McGranahan N., Burrell R.A., Rowan A.J., Joshi T., Fisher R., Larkin J., Szallasi Z., Swanton C. Parallel evolution of tumour subclones mimics diversity between tumours. J. Pathol. 2013;230:356–364.
    1. Mitchell T.J., Turajlic S., Rowan A., Nicol D., Farmery J.H.R., O’Brien T., Martincorena I., Tarpey P., Angelopoulos N., Yates L. Timing the Landmark Events in the Evolution of Clear Cell Renal Cell Cancer. Cell. 2018;173:611–623. this issue.
    1. Monzon F.A., Alvarez K., Peterson L., Truong L., Amato R.J., Hernandez-McClain J., Tannir N., Parwani A.V., Jonasch E. Chromosome 14q loss defines a molecular subtype of clear-cell renal cell carcinoma associated with poor prognosis. Mod. Pathol. 2011;24:1470–1479.
    1. Nik-Zainal S., Van Loo P., Wedge D.C., Alexandrov L.B., Greenman C.D., Lau K.W., Raine K., Jones D., Marshall J., Ramakrishna M., Breast Cancer Working Group of the International Cancer Genome Consortium The life history of 21 breast cancers. Cell. 2012;149:994–1007.
    1. Nilsen G., Liestøl K., Van Loo P., Moen Vollan H.K., Eide M.B., Rueda O.M., Chin S.F., Russell R., Baumbusch L.O., Caldas C. Copynumber: Efficient algorithms for single- and multi-track copy number segmentation. BMC Genomics. 2012;13:591.
    1. Noorbakhsh J., Chuang J.H. Uncertainties in tumor allele frequencies limit power to infer evolutionary pressures. Nat. Genet. 2017;49:1288–1289.
    1. Ortmann C.A., Kent D.G., Nangalia J., Silber Y., Wedge D.C., Grinfeld J., Baxter E.J., Massie C.E., Papaemmanuil E., Menon S. Effect of mutation order on myeloproliferative neoplasms. N. Engl. J. Med. 2015;372:601–612.
    1. Peña-Llopis S., Christie A., Xie X.J., Brugarolas J. Cooperation and antagonism among cancer genes: the renal cancer paradigm. Cancer Res. 2013;73:4173–4179.
    1. Peng J., Ma J., Li W., Mo R., Zhang P., Gao K., Jin X., Xiao J., Wang C., Fan J. Stabilization of MCRS1 by BAP1 prevents chromosome instability in renal cell carcinoma. Cancer Lett. 2015;369:167–174.
    1. Pengelly R.J., Gibson J., Andreoletti G., Collins A., Mattocks C.J., Ennis S. A SNP profiling panel for sample tracking in whole-exome sequencing studies. Genome Med. 2013;5:89.
    1. Perrino C.M., Hucthagowder V., Evenson M., Kulkarni S., Humphrey P.A. Genetic alterations in renal cell carcinoma with rhabdoid differentiation. Hum. Pathol. 2015;46:9–16.
    1. Przybycin C.G., McKenney J.K., Reynolds J.P., Campbell S., Zhou M., Karafa M.T., Magi-Galluzzi C. Rhabdoid differentiation is associated with aggressive behavior in renal cell carcinoma: a clinicopathologic analysis of 76 cases with clinical follow-up. Am. J. Surg. Pathol. 2014;38:1260–1265.
    1. Quinlan A.R., Hall I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26:841–842.
    1. Reiter J.G., Bozic I., Allen B., Chatterjee K., Nowak M.A. The effect of one additional driver mutation on tumor progression. Evol. Appl. 2013;6:34–45.
    1. Rini B.I., Dorff T.B., Elson P., Rodriguez C.S., Shepard D., Wood L., Humbert J., Pyle L., Wong Y.N., Finke J.H. Active surveillance in metastatic renal-cell carcinoma: a prospective, phase 2 trial. Lancet Oncol. 2016;17:1317–1324.
    1. Roth A., Khattra J., Yap D., Wan A., Laks E., Biele J., Ha G., Aparicio S., Bouchard-Côté A., Shah S.P. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods. 2014;11:396–398.
    1. Sato Y., Yoshizato T., Shiraishi Y., Maekawa S., Okuno Y., Kamura T., Shimamura T., Sato-Otsubo A., Nagae G., Suzuki H. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat. Genet. 2013;45:860–867.
    1. Scelo G., Riazalhosseini Y., Greger L., Letourneau L., Gonzàlez-Porta M., Wozniak M.B., Bourgey M., Harnden P., Egevad L., Jackson S.M. Variation in genomic landscape of clear cell renal cell carcinoma across Europe. Nat. Commun. 2014;5:5135.
    1. Shen C., Beroukhim R., Schumacher S.E., Zhou J., Chang M., Signoretti S., Kaelin W.G., Jr. Genetic and functional studies implicate HIF1α as a 14q kidney cancer suppressor gene. Cancer Discov. 2011;1:222–235.
    1. Smittenaar C.R., Petersen K.A., Stewart K., Moitt N. Cancer incidence and mortality projections in the UK until 2035. Br. J. Cancer. 2016;115:1147–1155.
    1. Srigley J.R., Delahunt B., Eble J.N., Egevad L., Epstein J.I., Grignon D., Hes O., Moch H., Montironi R., Tickoo S.K., ISUP Renal Tumor Panel The International Society of Urological Pathology (ISUP) Vancouver Classification of Renal Neoplasia. Am. J. Surg. Pathol. 2013;37:1469–1489.
    1. Talevich E., Shain A.H., Botton T., Bastian B.C. CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput. Biol. 2016;12:e1004873.
    1. Tomasetti C., Marchionni L., Nowak M.A., Parmigiani G., Vogelstein B. Only three driver gene mutations are required for the development of lung and colorectal cancers. Proc. Natl. Acad. Sci. USA. 2015;112:118–123.
    1. Turajlic S., Swanton C. TRACERx Renal: tracking renal cancer evolution through therapy. Nat. Rev. Urol. 2017;14:575–576.
    1. Turajlic S., Xu H., Litchfield K., Rowan A., Chambers T., Lopez J.I., Nicol D., O'Brien T., Larkin J., Horswell S. Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal. Cell. 2018;173:581–594. this issue.
    1. Van Loo P., Nordgard S.H., Lingjærde O.C., Russnes H.G., Rye I.H., Sun W., Weigman V.J., Marynen P., Zetterberg A., Naume B. Allele-specific copy number analysis of tumors. Proc. Natl. Acad. Sci. USA. 2010;107:16910–16915.
    1. Varela I., Tarpey P., Raine K., Huang D., Ong C.K., Stephens P., Davies H., Jones D., Lin M.L., Teague J. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011;469:539–542.
    1. Wang K., Li M., Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 2010;38:e164.
    1. Wang Z., Kim T.B., Peng B., Karam J., Creighton C., Joon A., Kawakami F., Trevisan P., Jonasch E., Chow C.W. Sarcomatoid renal cell carcinoma has a distinct molecular pathogenesis, driver mutation profile and transcriptional landscape. Clin. Cancer Res. 2017;23:6686–6696.
    1. Welch H.G., Skinner J.S., Schroeck F.R., Zhou W., Black W.C. Regional variation of computed tomographic imaging in the United States and the risk of nephrectomy. JAMA Intern. Med. 2017;178:221–227.
    1. Williamson S.R., Kum J.B., Goheen M.P., Cheng L., Grignon D.J., Idrees M.T. Clear cell renal cell carcinoma with a syncytial-type multinucleated giant tumor cell component: implications for differential diagnosis. Hum. Pathol. 2014;45:735–744.
    1. Zack T.I., Schumacher S.E., Carter S.L., Cherniack A.D., Saksena G., Tabak B., Lawrence M.S., Zhsng C.Z., Wala J., Mermel C.H. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 2013;45:1134–1140.
    1. Zhang B.Y., Thompson R.H., Lohse C.M., Leibovich B.C., Boorjian S.A., Cheville J.C., Costello B.A. A novel prognostic model for patients with sarcomatoid renal cell carcinoma. BJU Int. 2015;115:405–411.
    1. Znaor A., Lortet-Tieulent J., Laversanne M., Jemal A., Bray F. International variations and trends in renal cell carcinoma incidence and mortality. Eur. Urol. 2015;67:519–530.

Source: PubMed

3
Abonnere