Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal

Samra Turajlic, Hang Xu, Kevin Litchfield, Andrew Rowan, Tim Chambers, Jose I Lopez, David Nicol, Tim O'Brien, James Larkin, Stuart Horswell, Mark Stares, Lewis Au, Mariam Jamal-Hanjani, Ben Challacombe, Ashish Chandra, Steve Hazell, Claudia Eichler-Jonsson, Aspasia Soultati, Simon Chowdhury, Sarah Rudman, Joanna Lynch, Archana Fernando, Gordon Stamp, Emma Nye, Faiz Jabbar, Lavinia Spain, Sharanpreet Lall, Rosa Guarch, Mary Falzon, Ian Proctor, Lisa Pickering, Martin Gore, Thomas B K Watkins, Sophia Ward, Aengus Stewart, Renzo DiNatale, Maria F Becerra, Ed Reznik, James J Hsieh, Todd A Richmond, George F Mayhew, Samantha M Hill, Catherine D McNally, Carol Jones, Heidi Rosenbaum, Stacey Stanislaw, Daniel L Burgess, Nelson R Alexander, Charles Swanton, PEACE, TRACERx Renal Consortium, Samra Turajlic, Hang Xu, Kevin Litchfield, Andrew Rowan, Tim Chambers, Jose I Lopez, David Nicol, Tim O'Brien, James Larkin, Stuart Horswell, Mark Stares, Lewis Au, Mariam Jamal-Hanjani, Ben Challacombe, Ashish Chandra, Steve Hazell, Claudia Eichler-Jonsson, Aspasia Soultati, Simon Chowdhury, Sarah Rudman, Joanna Lynch, Archana Fernando, Gordon Stamp, Emma Nye, Faiz Jabbar, Lavinia Spain, Sharanpreet Lall, Rosa Guarch, Mary Falzon, Ian Proctor, Lisa Pickering, Martin Gore, Thomas B K Watkins, Sophia Ward, Aengus Stewart, Renzo DiNatale, Maria F Becerra, Ed Reznik, James J Hsieh, Todd A Richmond, George F Mayhew, Samantha M Hill, Catherine D McNally, Carol Jones, Heidi Rosenbaum, Stacey Stanislaw, Daniel L Burgess, Nelson R Alexander, Charles Swanton, PEACE, TRACERx Renal Consortium

Abstract

Clear-cell renal cell carcinoma (ccRCC) exhibits a broad range of metastatic phenotypes that have not been systematically studied to date. Here, we analyzed 575 primary and 335 metastatic biopsies across 100 patients with metastatic ccRCC, including two cases sampledat post-mortem. Metastatic competence was afforded by chromosome complexity, and we identify 9p loss as a highly selected event driving metastasis and ccRCC-related mortality (p = 0.0014). Distinct patterns of metastatic dissemination were observed, including rapid progression to multiple tissue sites seeded by primary tumors of monoclonal structure. By contrast, we observed attenuated progression in cases characterized by high primary tumor heterogeneity, with metastatic competence acquired gradually and initial progression to solitary metastasis. Finally, we observed early divergence of primitive ancestral clones and protracted latency of up to two decades as a feature of pancreatic metastases.

Trial registration: ClinicalTrials.gov NCT03226886 NCT03004755.

Keywords: chromosome instability; cytoreductive nephrectomy; evolution of metastasis; loss of 9p; metastasectomy; metastasis; oligometastasis; renal cell cancer; solitary metastasis.

Crown Copyright © 2018. Published by Elsevier Inc. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Figure S1
Figure S1
The flow diagram illustrates the phases of selection of metastatic samples included in the TRACERx Renal Primary-Metastasis cohort
Figure 1
Figure 1
Overview (A) An overview of somatic alterations detected in matched primary and metastatic tumors across 38 TRACERx Renal patients. The top panel shows the proportion of clonal and subclonal alterations. In the middle panel alterations in primary tumors are indicated in a lighter shade and those detected in metastases in a darker shade. Clonal alterations are shown as rectangles and subclonal alterations as triangles. Parallel evolution is indicated in orange with a split indicating multiple events. Abbreviations for tumor sites: P, primary; TT, tumor thrombus; AD, adrenal gland, indirect metastasis; AD(D), direct invasion of adrenal gland; AD(CL), contralateral adrenal gland; Renal(CL), contralateral kidney; Pr, perirenal invasion; and Pf, peri-nephric fat and Gerota’s fascia invasion. (B) The number of clonal and subclonal somatic alterations in primary and metastatic tumors. (C) The number of somatic alterations (1) detected in both primary tumor (P) and the matched metastatic tumor (M), (2) detected in primary tumor but not the matched metastatic tumor, and (3) detected in the metastatic tumor but not the matched primary tumor. (D) The proportions of synchronous and metachronous metastatic tumors profiled in the TRACERx Renal, HUC, and MSK cohorts. (E) The range of the metastatic sites sampled across the TRACERx, HUC, and MSK cohorts. The total number of metastases sampled (n) and the number from each cohort are shown in brackets (Tx represents TRACERx Renal; HUC and MSK are extension cohorts). See also Tables S1 and S6 and Table S2.
Figure S2
Figure S2
Driver Events in HUC and MSK Cohorts, Related to Figure 1 (A and B) This figure shows driver mutations and driver SCNAs detected in matched primary and metastatic tumours in HUC (A) and MSK (B) cohorts. Clonal alterations are shown as rectangles and subclonal alterations as triangles. Parallel evolution is indicated in orange with a split indicating multiple events.
Figure 2
Figure 2
Characterization of a Metastasizing Clone(s) (A) Illustration of the method used to categorize tumor clones. (B) Four violin plots summarizing (starting at the top left and working clockwise): (1) non-synonymous mutation count, (2) wGII, (3) ploidy, and (4) Ki67. Values are compared between tumor clones “not selected” and “selected” in metastasis, with all region/clone values plotted per tumor (excluding MRCA “maintained” clones; see the STAR Methods). A linear mixed effects (LMEs) model was used to determine significance, to account for the non-independence of multiple observations from individual tumors. (C) For each driver event the proportion of times it was observed in “not selected” and “selected” clones for TRACERx, HUC, and MSK cohorts. The far-right panel shows the log10 p value for each event for enrichment in “selected” versus “not selected” clones. Testing was performed using a binomial test with meta-analysis conducted using Fisher’s method of combining p values from independent tests. p values are corrected for multiple testing using Benjamini-Hochberg procedure. (D) Overall survival hazard ratios for events with p 

Figure 3

Tumor Thrombus This figure shows…

Figure 3

Tumor Thrombus This figure shows tumor thrombus (TT) driver trees with primary clones…

Figure 3
Tumor Thrombus This figure shows tumor thrombus (TT) driver trees with primary clones in the lower panels and level I, level II, level III, and level IV TT clones in light green, blue, orange, and red, respectively, in the upper panels. Tumor TNM stage and driver events leading to TT are annotated. Length of branches connecting clones is not informative. See also Figure S3.

Figure S3

Analysis of Tumor Thrombus, Related…

Figure S3

Analysis of Tumor Thrombus, Related to Figure 3 (A and B) (A) shows…

Figure S3
Analysis of Tumor Thrombus, Related to Figure 3 (A and B) (A) shows Ki67 proliferation index data (mean % of cells staining positive for Ki67 across all primary tumour regions) for cases presenting with and without TT. (B) shows cases with TT and distal metastases.

Figure 4

Evolution of Progressive Disease (A)…

Figure 4

Evolution of Progressive Disease (A) Driver trees and the clinical course for cases…

Figure 4
Evolution of Progressive Disease (A) Driver trees and the clinical course for cases with lymph node and distal metastases. Cases were grouped into those with “rapid progression” and “attenuated progression.” The primary tumor evolutionary subtype, primary tumor ITH/wGII classification, and select driver events on the tree (VHL, BAP1, PBRM1, MTOR, SETD2, TSC1, TSC2, chr 9p loss, and chr 14q loss) are annotated for each case. Metastasis seeding subclones and any subclones private to metastasis are highlighted in blue. Clinical course is shown from the time of nephrectomy to death or last follow-up. Pattern of disease progression is classified as multiple new metastases (multiple circles), solitary new metastasis (single circle), and progression of existing metastases (“PD”). Progression and follow-up times are shown in months. Systemic treatments are indicated. Synchronous and metachronous metastatic sites are listed under corresponding time points. Profiled metastases are highlighted in blue boxes. Abbreviation for tumor sites: P, primary; TT, tumor thrombus; AD, adrenal gland; AD(D), direct invasion of adrenal gland; AD(CL), contralateral adrenal gland; renal(CL), contralateral kidney; Pr, perirenal invasion; and Pf, peri-nephric fat and Gerota’s fascia invasion. (B) The number of cases with “rapid progression” or “attenuated progression” in each evolutionary subtype. (C) The maximum wGII and ITH in cases with “rapid progression” and “attenuated progression.” See also Figure S4 and Table S1.

Figure S4

Fishplot Summary of Selected Cases,…

Figure S4

Fishplot Summary of Selected Cases, Related to Figure 4 This figure shows 2…

Figure S4
Fishplot Summary of Selected Cases, Related to Figure 4 This figure shows 2 example cases with distal metastases. Diagrams of the primary tumour and the involved tissue sites are illustrated. Fishplots are used to show disease evolution. Driver events are annotated on each fishplot.

Figure 5

Latent Metastases (A) The distribution…

Figure 5

Latent Metastases (A) The distribution of times from nephrectomy to metastasis resection split…

Figure 5
Latent Metastases (A) The distribution of times from nephrectomy to metastasis resection split by site of metastasis. The circle represents the median value, and the gray lines depict the median average deviation (MAD) value (i.e., plus/minus one MAD). The range (min to max) values are far right in brackets. (B) wGII values per region split by site of metastasis. All regions are shown per metastasis, and a linear mixed effects (LMEs) model was used to determine significance (for pancreas vs. all other), to account for the non-independence of multiple observations from individual tumors. (C) Fishplots for the three cases (SP006, SP023, and SP058) with latent pancreatic metastases. *Case SP058 had additional metastases to skeletal muscle (time = 0) and the small bowel (time = 7). See also Figure S5.

Figure S5

Shown is the driver mutation…

Figure S5

Shown is the driver mutation and SCNA phylogenetic tree and heatmap illustrating the…

Figure S5
Shown is the driver mutation and SCNA phylogenetic tree and heatmap illustrating the clonal relationship between the primary and metastasis for case SP58

Figure 6

Spatial Resolution of Metastases through…

Figure 6

Spatial Resolution of Metastases through Post-Mortem Sampling (A and B) Shows cases K548…

Figure 6
Spatial Resolution of Metastases through Post-Mortem Sampling (A and B) Shows cases K548 (A) and K489 (B), which were sampled at post-mortem with the extent of sampling. The clinical time course (in months) and therapuetic interventions are shown. Metastatic progression is illustrated using fish plots with the select driver events annotated (VHL, BAP1, PBRM1, MTOR, SETD2, TSC1, TSC2, 9p loss, and 14q loss). Metastasizing clone color matches that of the corresponding metastatic site. See also Table S1.

Figure 7

Summary of Key Conclusions from…

Figure 7

Summary of Key Conclusions from the Study

Figure 7
Summary of Key Conclusions from the Study
All figures (13)
Figure 3
Figure 3
Tumor Thrombus This figure shows tumor thrombus (TT) driver trees with primary clones in the lower panels and level I, level II, level III, and level IV TT clones in light green, blue, orange, and red, respectively, in the upper panels. Tumor TNM stage and driver events leading to TT are annotated. Length of branches connecting clones is not informative. See also Figure S3.
Figure S3
Figure S3
Analysis of Tumor Thrombus, Related to Figure 3 (A and B) (A) shows Ki67 proliferation index data (mean % of cells staining positive for Ki67 across all primary tumour regions) for cases presenting with and without TT. (B) shows cases with TT and distal metastases.
Figure 4
Figure 4
Evolution of Progressive Disease (A) Driver trees and the clinical course for cases with lymph node and distal metastases. Cases were grouped into those with “rapid progression” and “attenuated progression.” The primary tumor evolutionary subtype, primary tumor ITH/wGII classification, and select driver events on the tree (VHL, BAP1, PBRM1, MTOR, SETD2, TSC1, TSC2, chr 9p loss, and chr 14q loss) are annotated for each case. Metastasis seeding subclones and any subclones private to metastasis are highlighted in blue. Clinical course is shown from the time of nephrectomy to death or last follow-up. Pattern of disease progression is classified as multiple new metastases (multiple circles), solitary new metastasis (single circle), and progression of existing metastases (“PD”). Progression and follow-up times are shown in months. Systemic treatments are indicated. Synchronous and metachronous metastatic sites are listed under corresponding time points. Profiled metastases are highlighted in blue boxes. Abbreviation for tumor sites: P, primary; TT, tumor thrombus; AD, adrenal gland; AD(D), direct invasion of adrenal gland; AD(CL), contralateral adrenal gland; renal(CL), contralateral kidney; Pr, perirenal invasion; and Pf, peri-nephric fat and Gerota’s fascia invasion. (B) The number of cases with “rapid progression” or “attenuated progression” in each evolutionary subtype. (C) The maximum wGII and ITH in cases with “rapid progression” and “attenuated progression.” See also Figure S4 and Table S1.
Figure S4
Figure S4
Fishplot Summary of Selected Cases, Related to Figure 4 This figure shows 2 example cases with distal metastases. Diagrams of the primary tumour and the involved tissue sites are illustrated. Fishplots are used to show disease evolution. Driver events are annotated on each fishplot.
Figure 5
Figure 5
Latent Metastases (A) The distribution of times from nephrectomy to metastasis resection split by site of metastasis. The circle represents the median value, and the gray lines depict the median average deviation (MAD) value (i.e., plus/minus one MAD). The range (min to max) values are far right in brackets. (B) wGII values per region split by site of metastasis. All regions are shown per metastasis, and a linear mixed effects (LMEs) model was used to determine significance (for pancreas vs. all other), to account for the non-independence of multiple observations from individual tumors. (C) Fishplots for the three cases (SP006, SP023, and SP058) with latent pancreatic metastases. *Case SP058 had additional metastases to skeletal muscle (time = 0) and the small bowel (time = 7). See also Figure S5.
Figure S5
Figure S5
Shown is the driver mutation and SCNA phylogenetic tree and heatmap illustrating the clonal relationship between the primary and metastasis for case SP58
Figure 6
Figure 6
Spatial Resolution of Metastases through Post-Mortem Sampling (A and B) Shows cases K548 (A) and K489 (B), which were sampled at post-mortem with the extent of sampling. The clinical time course (in months) and therapuetic interventions are shown. Metastatic progression is illustrated using fish plots with the select driver events annotated (VHL, BAP1, PBRM1, MTOR, SETD2, TSC1, TSC2, 9p loss, and 14q loss). Metastasizing clone color matches that of the corresponding metastatic site. See also Table S1.
Figure 7
Figure 7
Summary of Key Conclusions from the Study

References

    1. Adey N., Emery D., Bosh D., Callahan S., Schreiner J., Chen Y., Greig A., Geiersbach K., Parry R. A mill based instrument and software system for dissecting slide-mounted tissue that provides digital guidance and documentation. BMC Clin. Pathol. 2013;13:29.
    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. Bashashati A., Ha G., Tone A., Ding J., Prentice L.M., Roth A., Rosner J., Shumansky K., Kalloger S., Senz J. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J. Pathol. 2013;231:21–34.
    1. Becerra M.F., Reznik E., Redzematovic A., Tennenbaum D.M., Kashan M., Ghanaat M., Casuscelli J., Manley B., Jonsson P., DiNatale R.G. Comparative genomic profiling of matched primary and metastatic tumors in renal cell carcinoma. Eur. Urol. Focus. 2017 S2405-4569(17)30237-7.
    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. Bex A., Mulders P., Jewett M.A.S., Wagstaff J., van Velthoven R., Laguna Pes P.M., Wood L., van Melick H.H.E., Soetekouw P., Lattouf J.B. Immediate versus deferred cytoreductive nephrectomy (CN) in patients with synchronous metastatic renal cell carcinoma (mRCC) receiving sunitinib (EORTC 30073 SURTIME) Ann Oncol. 2017;28(Suppl 5):v605–v649.
    1. Bhindi B., Wallis C.J.D., Boorjian S.A., Thompson R.H., Farrell A., Kim S.P., Karam J.A., Capitanio U., Golijanin D., Leibovich B.C., Gershman B. The role of lymph node dissection in the management of renal cell carcinoma: a systematic review and meta-analysis. BJU Int. 2018 Published online March 1, 2018.
    1. Bianchi M., Sun M., Jeldres C., Shariat S.F., Trinh Q.D., Briganti A., Tian Z., Schmitges J., Graefen M., Perrotte P. Distribution of metastatic sites in renal cell carcinoma: a population-based analysis. Ann. Oncol. 2012;23:973–980.
    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. Campbell P.J., Yachida S., Mudie L.J., Stephens P.J., Pleasance E.D., Stebbings L.A., Morsberger L.A., Latimer C., McLaren S., Lin M.L. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature. 2010;467:1109–1113.
    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. Casuscelli J., Weinhold N., Gundem G., Wang L., Zabor E.C., Drill E., Wang P.I., Nanjangud G.J., Redzematovic A., Nargund A.M. Genomic landscape and evolution of metastatic chromophobe renal cell carcinoma. JCI Insight. 2017;2:2.
    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. Dabestani S., Marconi L., Hofmann F., Stewart F., Lam T.B., Canfield S.E., Staehler M., Powles T., Ljungberg B., Bex A. Local treatments for metastases of renal cell carcinoma: a systematic review. Lancet Oncol. 2014;15:e549–e561.
    1. Davoli T., Uno H., Wooten E.C., Elledge S.J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355:355.
    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. Ding L., Ellis M.J., Li S., Larson D.E., Chen K., Wallis J.W., Harris C.C., McLellan M.D., Fulton R.S., Fulton L.L. Genome remodelling in a basal-like breast cancer metastasis and xenograft. Nature. 2010;464:999–1005.
    1. El-Mokadem I., Fitzpatrick J., Bondad J., Rauchhaus P., Cunningham J., Pratt N., Fleming S., Nabi G. Chromosome 9p deletion in clear cell renal cell carcinoma predicts recurrence and survival following surgery. Br. J. Cancer. 2014;111:1381–1390.
    1. Eldredge N., Gould S.J. On punctuated equilibria. Science. 1997;276:338–341.
    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. Field M.G., Durante M.A., Anbunathan H., Cai L.Z., Decatur C.L., Bowcock A.M., Kurtenbach S., Harbour J.W. Punctuated evolution of canonical genomic aberrations in uveal melanoma. Nat. Commun. 2018;9:116.
    1. Gao R., Davis A., McDonald T.O., Sei E., Shi X., Wang Y., Tsai P.C., Casasent A., Waters J., Zhang H. Punctuated copy number evolution and clonal stasis in triple-negative breast cancer. Nat. Genet. 2016;48:1119–1130.
    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. Gerlinger M., Rowan A.J., Horswell S., Math M., Larkin J., Endesfelder D., Gronroos E., Martinez P., Matthews N., Stewart A. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012;366:883–892.
    1. Gershman B., Moreira D.M., Thompson R.H., Boorjian S.A., Lohse C.M., Costello B.A., Cheville J.C., Leibovich B.C. Renal cell carcinoma with isolated lymph node involvement: long-term natural history and predictors of oncologic outcomes following surgical resection. Eur. Urol. 2017;72:300–306.
    1. Giancotti F.G. Mechanisms governing metastatic dormancy and reactivation. Cell. 2013;155:750–764.
    1. Gundem G., Van Loo P., Kremeyer B., Alexandrov L.B., Tubio J.M.C., Papaemmanuil E., Brewer D.S., Kallio H.M.L., Högnäs G., Annala M., ICGC Prostate Group The evolutionary history of lethal metastatic prostate cancer. Nature. 2015;520:353–357.
    1. Haffner M.C., Mosbruger T., Esopi D.M., Fedor H., Heaphy C.M., Walker D.A., Adejola N., Gürel M., Hicks J., Meeker A.K. Tracking the clonal origin of lethal prostate cancer. J. Clin. Invest. 2013;123:4918–4922.
    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. Klatte T., Rao P.N., de Martino M., LaRochelle J., Shuch B., Zomorodian N., Said J., Kabbinavar F.F., Belldegrun A.S., Pantuck A.J. Cytogenetic profile predicts prognosis of patients with clear cell renal cell carcinoma. J. Clin. Oncol. 2009;27:746–753.
    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. Kutikov A., Uzzo R.G., Caraway A., Reese C.T., Egleston B.L., Chen D.Y., Viterbo R., Greenberg R.E., Wong Y.N., Raman J.D., Boorjian S.A. Use of systemic therapy and factors affecting survival for patients undergoing cytoreductive nephrectomy. BJU Int. 2010;106:218–223.
    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. Lee S.Y., Haq F., Kim D., Jun C., Jo H.J., Ahn S.M., Lee W.S. Comparative genomic analysis of primary and synchronous metastatic colorectal cancers. PLoS ONE. 2014;9:e90459.
    1. Li H., Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. McGranahan N., Rosenthal R., Hiley C.T., Rowan A.J., Watkins T.B.K., Wilson G.A., Birkbak N.J., Veeriah S., Van Loo P., Herrero J. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell. 2017;171:1259–1271.e11.
    1. McKay R.R., Kroeger N., Xie W., Lee J.L., Knox J.J., Bjarnason G.A., MacKenzie M.J., Wood L., Srinivas S., Vaishampayan U.N. Impact of bone and liver metastases on patients with renal cell carcinoma treated with targeted therapy. Eur. Urol. 2014;65:577–584.
    1. McPherson A., Roth A., Laks E., Masud T., Bashashati A., Zhang A.W., Ha G., Biele J., Yap D., Wan A. Divergent modes of clonal spread and intraperitoneal mixing in high-grade serous ovarian cancer. Nat. Genet. 2016;48:758–767.
    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. 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. The life history of 21 breast cancers. 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. Notta F., Chan-Seng-Yue M., Lemire M., Li Y., Wilson G.W., Connor A.A., Denroche R.E., Liang S.B., Brown A.M., Kim J.C. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature. 2016;538:378–382.
    1. Psutka S.P., Leibovich B.C. Management of inferior vena cava tumor thrombus in locally advanced renal cell carcinoma. Ther. Adv. Urol. 2015;7:216–229.
    1. Quinlan A.R., Hall I.M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. 2010;26:841–842.
    1. Reese A.C., Whitson J.M., Meng M.V. Natural history of untreated renal cell carcinoma with venous tumor thrombus. Urol. Oncol. 2013;31:1305–1309.
    1. Robinson D.R., Wu Y.M., Lonigro R.J., Vats P., Cobain E., Everett J., Cao X., Rabban E., Kumar-Sinha C., Raymond V. Integrative clinical genomics of metastatic cancer. Nature. 2017;548:297–303.
    1. Santaguida S., Amon A. Short- and long-term effects of chromosome mis-segregation and aneuploidy. Nat. Rev. Mol. Cell Biol. 2015;16:473–485.
    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. Schwarz R.F., Ng C.K., Cooke S.L., Newman S., Temple J., Piskorz A.M., Gale D., Sayal K., Murtaza M., Baldwin P.J. Spatial and temporal heterogeneity in high-grade serous ovarian cancer: a phylogenetic analysis. PLoS Med. 2015;12:e1001789.
    1. Shah S.P., Morin R.D., Khattra J., Prentice L., Pugh T., Burleigh A., Delaney A., Gelmon K., Guliany R., Senz J. Mutational evolution in a lobular breast tumour profiled at single nucleotide resolution. Nature. 2009;461:809–813.
    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. Turajlic S., Xu H., Litchfield K., Rowan A., Horswell S., Chambers T., O’Brien T., Lopez J.I., Watkins T.B.K., Nicol D. Deterministic Evolutionary Trajectories Influence Primary Tumor Growth: TRACERx Renal. Cell. 2018;173:595–610. this issue.
    1. Van Loo P., Nordgard S.H., Lingjaerde 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. PNAS. 2010;107:16910–16915.
    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. Weichselbaum R.R., Hellman S. Oligometastases revisited. Nat. Rev. Clin. Oncol. 2011;8:378–382.
    1. Woodruff D.Y., Van Veldhuizen P., Muehlebach G., Johnson P., Williamson T., Holzbeierlein J.M. The perioperative management of an inferior vena caval tumor thrombus in patients with renal cell carcinoma. Urol. Oncol. 2013;31:517–521.
    1. Wszolek M.F., Wotkowicz C., Libertino J.A. Surgical management of large renal tumors. Nat. Clin. Pract. Urol. 2008;5:35–46.
    1. Yachida S., Jones S., Bozic I., Antal T., Leary R., Fu B., Kamiyama M., Hruban R.H., Eshleman J.R., Nowak M.A. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467:1114–1117.
    1. Yates L.R., Knappskog S., Wedge D., Farmery J.H.R., Gonzalez S., Martincorena I., Alexandrov L.B., Van Loo P., Haugland H.K., Lilleng P.K. Genomic evolution of breast cancer metastasis and relapse. Cancer Cell. 2017;32:169–184.e7.
    1. Zehir A., Benayed R., Shah R.H., Syed A., Middha S., Kim H.R., Srinivasan P., Gao J., Chakravarty D., Devlin S.M. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat. Med. 2017;23:703–713.
    1. Zhang J., Lu A., Li L., Yue J., Lu Y. p16 Modulates VEGF expression via its interaction with HIF-1alpha in breast cancer cells. Cancer Invest. 2010;28:588–597.
    1. Zhao Z.M., Zhao B., Bai Y., Iamarino A., Gaffney S.G., Schlessinger J., Lifton R.P., Rimm D.L., Townsend J.P. Early and multiple origins of metastatic lineages within primary tumors. Proc. Natl. Acad. Sci. USA. 2016;113:2140–2145.

Source: PubMed

3
订阅