Whole-genome plasma sequencing reveals focal amplifications as a driving force in metastatic prostate cancer

Peter Ulz, Jelena Belic, Ricarda Graf, Martina Auer, Ingrid Lafer, Katja Fischereder, Gerald Webersinke, Karl Pummer, Herbert Augustin, Martin Pichler, Gerald Hoefler, Thomas Bauernhofer, Jochen B Geigl, Ellen Heitzer, Michael R Speicher, Peter Ulz, Jelena Belic, Ricarda Graf, Martina Auer, Ingrid Lafer, Katja Fischereder, Gerald Webersinke, Karl Pummer, Herbert Augustin, Martin Pichler, Gerald Hoefler, Thomas Bauernhofer, Jochen B Geigl, Ellen Heitzer, Michael R Speicher

Abstract

Genomic alterations in metastatic prostate cancer remain incompletely characterized. Here we analyse 493 prostate cancer cases from the TCGA database and perform whole-genome plasma sequencing on 95 plasma samples derived from 43 patients with metastatic prostate cancer. From these samples, we identify established driver aberrations in a cancer-related gene in nearly all cases (97.7%), including driver gene fusions (TMPRSS2:ERG), driver focal deletions (PTEN, RYBP and SHQ1) and driver amplifications (AR and MYC). In serial plasma analyses, we observe changes in focal amplifications in 40% of cases. The mean time interval between new amplifications was 26.4 weeks (range: 5-52 weeks), suggesting that they represent rapid adaptations to selection pressure. An increase in neuron-specific enolase is accompanied by clonal pattern changes in the tumour genome, most consistent with subclonal diversification of the tumour. Our findings suggest a high plasticity of prostate cancer genomes with newly occurring focal amplifications as a driving force in progression.

Figures

Figure 1. Overall characterization of SCNAs in…
Figure 1. Overall characterization of SCNAs in TCGA and plasma samples.
(a) Circos plot illustrating the relative frequency of SCNAs (gains in red and losses in blue) of 493 prostate cancer cases from the TCGA database (inner circle) and 95 plasma samples from patients with metastasized prostate cancer (middle circle). The outer circle shows ideograms of the respective chromosomes. (b) Partial chromosome 5 circos plot of plasma samples without (inner segment) and with (middle segment) TMPRSS2:ERG fusion, the ellipse marks the CHD1 region. (c) Scatter plot of 30 genes commonly involved in amplifications (left panel; green bar: 20 Mb; red bar: copy number of 4) and 42 genes frequently involved in focal deletions (right panel; green bar: 20 Mb; red bar: copy number of 1) derived from 5,737 cases of the TCGA pan-cancer data set demonstrating an inverse relationship between amplification and deletion size, respectively, as well as copy numbers.
Figure 2. Focal SCNAs in TCGA and…
Figure 2. Focal SCNAs in TCGA and plasma prostate cancer samples.
(a) Circos plot of focal SCNAs in the TCGA database (inner circle) and the plasma samples (middle circle) depicting the relative frequency (Y axis limit for amplifications and deletions: 50%) of focal events (gains in red and losses in blue). The outer circle shows ideograms of the respective chromosomes. Target genes of focal SCNAs are annotated if they were observed in >5% of TCGA/plasma samples. (b) Boxplot and probability densities of focal SCNA calls for T-stage (top), and N-stage (centre), and Gleason score (bottom). Box comprises data from the first to the third quartile (interquartile range, IQR, in blue) and whiskers (black) extend to the 1.5 × IQR from the box. The median is displayed in red. Probability densities (based on Kernel density estimates) are displayed in ochre and data ranges are displayed in red. P values were calculated using Mann–Whitney U-tests. (c) Boxplot and probability densities of focal copy-number changes in the TCGA and plasma datasets (Whiskers, horizontal lines and statistical test as in b). (d) Boxplot and probability densities of focal TCGA and plasma samples for focal deletions (top) and for focal amplifications (bottom; Whiskers, horizontal lines and statistical test as in b). (e) Bar charts depicting the frequency of driver aberrations (green) and relative frequencies of the most frequently observed focal driver amplifications (red) and deletions (blue) in TCGA and plasma samples. (f) Co-occurrence of MYC amplification and TP53 losses in patients P19, P55, P119 and P148. Gains (log2-ratio>0.2) are shown in red and losses (log2-ratio less than −0.2) are shown in blue. Green indicates balanced regions.
Figure 3. Serial analyses revealed de novo…
Figure 3. Serial analyses revealed de novo occurrence of focal amplifications.
(a) Hierarchical clustering with serial plasma samples demonstrating that samples derived from the same patient tend to cluster together (Clustering-Manhattan average). (b) Genome-wide log2-ratio plots of plasma samples from P40 obtained at a castration-sensitive stage (upper panel) and 10 months later after development of CRPC. The inset illustrates enlarged log2-ratio plots of the X chromosome, the bottom sample shows gain of chromosome X material with the highest copy-number gain on Xq12, the region that harbours the AR gene. In this and in the subsequent panels, the grey arrows indicate the time intervals between the sample collections. Copy-number gains are depicted in red and copy-number losses in blue. (c) P106's tumour genome developed AR amplification within 12 months and an additional amplicon at Xq23-q24 within the subsequent 6 weeks. (d) The first plasma sample of P147 had two high-level amplifications, that is, on chromosomes Xq12 (AR) and 5q14.3 (EDIL3), which had not been observed in the primary tumour. Within the next 4 months, a further amplicon evolved on chromosome 10q11.21 (RET). The quality of the analysis of the primary tumour was not optimal due to the fixation conditions of the tissue. The ‘peak' on chromosome 10 in the primary tumour does not involve the RET region and is instead most likely an artefact. (e) The plasma sample from prostate cancer patient P112 displayed an amplicon on 1q21.3 including SETDB1, which had not been present in the analysed part of the primary tumour.
Figure 4. Changing proportions of SCNAs associated…
Figure 4. Changing proportions of SCNAs associated with increasing NSE.
(a) Genome-wide log2-ratio plots of plasma samples P148_1 (upper panel) and P148_3 (centre panel), which was obtained 12 months later. Between these two samples, relative copy-number losses and gains for ∼42.2% and ∼23.3%, respectively, of chromosomal regions (overlay plots in bottom panel; black: first analysis (P148_1); blue: third analysis (P148_3) were observed; regions with different log2-ratios >0.2 are marked with blue and red bars below or above the respective regions). (b) Enlarged X-chromosome profiles demonstrating that the AR amplification is only present in sample P148_1. (c) Confirmation of the AR copy-number status by quantitative PCR in duplicates. Y axis represents RQ (relative quantity) compared with a pooled male reference DNA sample; error bars indicate minimum and maximum values. (d) Plots of P170 illustrating changing chromosomal copy-number patterns over a period of 15 weeks (black: first analysis (P170_1); blue: second analysis (P170_2)). Between the two analyses 26.7% of the genome differs in terms of copy-number status (235 and 590 Mb were gained or lost, respectively, in P170_2 compared with P170_1). Log2-ratios of sample P170_1 have been adjusted to correct for different tumour ratios. Estimated tumour DNA content ratio: 1:4.34 (P170_1/P170_4). (e) Comparison between first and last analysed plasma samples from P179 (black: first analysis (P179_1); blue: second analysis (P179_4)). These analyses do not show major changes on the autosomes, but do show loss of the AR amplicon (indicated as blue bar below the X chromosome).

References

    1. Barbieri C. E. & Tomlins S. A. The prostate cancer genome: perspectives and potential. Urol. Oncol. 32, 53.e15–53.e22 (2014).
    1. Baca S. C. et al.. Punctuated evolution of prostate cancer genomes. Cell 153, 666–677 (2013).
    1. Barbieri C. E. et al.. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat. Genet. 44, 685–689 (2012).
    1. Berger M. F. et al.. The genomic complexity of primary human prostate cancer. Nature 470, 214–220 (2011).
    1. Grasso C. S. et al.. The mutational landscape of lethal castration-resistant prostate cancer. Nature 487, 239–243 (2012).
    1. Gundem G. et al.. The evolutionary history of lethal metastatic prostate cancer. Nature 520, 353–357 (2015).
    1. Hong M. K. et al.. Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer. Nat. Commun. 6, 6605 (2015).
    1. Robinson D. et al.. Integrative clinical genomics of advanced prostate cancer. Cell 161, 1215–1228 (2015).
    1. Taylor B. S. et al.. Integrative genomic profiling of human prostate cancer. Cancer Cell 18, 11–22 (2010).
    1. Heitzer E. et al.. Tumor-associated copy number changes in the circulation of patients with prostate cancer identified through whole-genome sequencing. Genome Med. 5, 30 (2013).
    1. Mohan S. et al.. Changes in colorectal carcinoma genomes under anti-EGFR therapy identified by whole-genome plasma DNA sequencing. PLoS Genet. 10, e1004271 (2014).
    1. Heidary M. et al.. The dynamic range of circulating tumor DNA in metastatic breast cancer. Breast Cancer Res. 16, 421 (2014).
    1. Heitzer E., Auer M., Ulz P., Geigl J. B. & Speicher M. R. Circulating tumor cells and DNA as liquid biopsies. Genome Med. 5, 73 (2013).
    1. Heitzer E., Ulz P. & Geigl J. B. Circulating tumor DNA as a liquid biopsy for cancer. Clin. Chem. 61, 112–123 (2015).
    1. Mermel C. H. et al.. GISTIC2.0 facilitates sensitive and confident localization of the targets of focal somatic copy-number alteration in human cancers. Genome Biol. 12, R41 (2011).
    1. Robbins C. M. et al.. Copy number and targeted mutational analysis reveals novel somatic events in metastatic prostate tumors. Genome Res. 21, 47–55 (2011).
    1. Tomlins S. A. et al.. Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310, 644–648 (2005).
    1. Attard G. et al.. Prostate cancer. Lancet 387, 70–82 (2016).
    1. Romanel A. et al.. Plasma AR and abiraterone-resistant prostate cancer. Sci. Transl. Med. 7, 312re10 (2015).
    1. Azad A. A. et al.. Androgen receptor gene aberrations in circulating cell-free dna: biomarkers of therapeutic resistance in castration-resistant prostate cancer. Clin. Cancer Res. 21, 2315–2324 (2015).
    1. Carreira S. et al.. Tumor clone dynamics in lethal prostate cancer. Sci. Transl. Med. 6, 254ra125 (2014).
    1. Shen M. M. & Abate-Shen C. Molecular genetics of prostate cancer: new prospects for old challenges. Genes Dev. 24, 1967–2000 (2010).
    1. Visakorpi T. et al.. In vivo amplification of the androgen receptor gene and progression of human prostate cancer. Nat. Genet. 9, 401–406 (1995).
    1. Santarius T., Shipley J., Brewer D., Stratton M. R. & Cooper C. S. A census of amplified and overexpressed human cancer genes. Nat. Rev. Cancer 10, 59–64 (2010).
    1. Beroukhim R. et al.. The landscape of somatic copy-number alteration across human cancers. Nature 463, 899–905 (2010).
    1. Zack T. I. et al.. Pan-cancer patterns of somatic copy number alteration. Nat. Genet. 45, 1134–1140 (2013).
    1. Ulz P., Heitzer E. & Speicher M. R. Co-occurrence of MYC amplification and TP53 mutations in human cancer. Nat. Genet. 48, 104–106 (2016).
    1. Bernard D., Pourtier-Manzanedo A., Gil J. & Beach D. H. Myc confers androgen-independent prostate cancer cell growth. J. Clin. Invest. 112, 1724–1731 (2003).
    1. Malik R. et al.. Targeting the MLL complex in castration-resistant prostate cancer. Nat. Med. 21, 344–352 (2015).
    1. Luo J. H. et al.. Transcriptomic and genomic analysis of human hepatocellular carcinomas and hepatoblastomas. Hepatology 44, 1012–1024 (2006).
    1. Zou X. et al.. Downregulation of developmentally regulated endothelial cell locus-1 inhibits the growth of colon cancer. J. Biomed. Sci. 16, 33 (2009).
    1. Schilsky R. L. Implementing personalized cancer care. Nat. Rev. Clin. Oncol. 11, 432–438 (2014).
    1. Sun Y. et al.. Histone methyltransferase SETDB1 is required for prostate cancer cell proliferation, migration and invasion. Asian J. Androl. 16, 319–324 (2014).
    1. Beltran H. et al.. Challenges in recognizing treatment-related neuroendocrine prostate cancer. J. Clin. Oncol. 30, e386–e389 (2012).
    1. Mosquera J. M. et al.. Concurrent AURKA and MYCN gene amplifications are harbingers of lethal treatment-related neuroendocrine prostate cancer. Neoplasia 15, 1–10 (2013).
    1. Lallous N. et al.. Functional analysis of androgen receptor mutations that confer anti-androgen resistance identified in circulating cell-free DNA from prostate cancer patients. Genome Biol. 17, 10 (2016).
    1. Bettegowda C. et al.. Detection of circulating tumor DNA in early- and late-stage human malignancies. Sci. Transl. Med. 6, 224ra24 (2014).
    1. Dawson S. J. et al.. Analysis of circulating tumor DNA to monitor metastatic breast cancer. N. Engl. J. Med. 368, 1199–1209 (2013).
    1. Forshew T. et al.. Noninvasive identification and monitoring of cancer mutations by targeted deep sequencing of plasma DNA. Sci. Transl. Med. 4, 136ra68 (2012).
    1. Leary R. J. et al.. Detection of chromosomal alterations in the circulation of cancer patients with whole-genome sequencing. Sci. Transl. Med. 4, 162ra154 (2012).
    1. Murtaza M. et al.. Non-invasive analysis of acquired resistance to cancer therapy by sequencing of plasma DNA. Nature 497, 108–112 (2013).
    1. Aryee M. J. et al.. DNA methylation alterations exhibit intraindividual stability and interindividual heterogeneity in prostate cancer metastases. Sci. Transl. Med. 5, 169ra10 (2013).
    1. Liu W. et al.. Copy number analysis indicates monoclonal origin of lethal metastatic prostate cancer. Nat. Med. 15, 559–565 (2009).
    1. Rubin M. A., Maher C. A. & Chinnaiyan A. M. Common gene rearrangements in prostate cancer. J. Clin. Oncol. 29, 3659–3668 (2011).
    1. Arora V. K. et al.. Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade. Cell 155, 1309–1322 (2013).
    1. Asangani I. A. et al.. Therapeutic targeting of BET bromodomain proteins in castration-resistant prostate cancer. Nature 510, 278–282 (2014).
    1. Manning G., Whyte D. B., Martinez R., Hunter T. & Sudarsanam S. The protein kinase complement of the human genome. Science 298, 1912–1934 (2002).
    1. Rick F. G., Block N. L. & Schally A. V. An update on the use of degarelix in the treatment of advanced hormone-dependent prostate cancer. Onco Targets Ther. 6, 391–402 (2013).
    1. Li H. & Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).
    1. McKenna A. et al.. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 20, 1297–1303 (2010).
    1. Wang K., Li M. & Hakonarson H. ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data. Nucleic Acids Res. 38, e164 (2010).
    1. Baslan T. et al.. Genome-wide copy number analysis of single cells. Nat. Protoc. 7, 1024–1041 (2012).
    1. Lai W., Choudhary V. & Park P. J. CGHweb: a tool for comparing DNA copy number segmentations from multiple algorithms. Bioinformatics 24, 1014–1015 (2008).
    1. Cazier J. B., Holmes C. C. & Broxholme J. GREVE: Genomic Recurrent Event ViEwer to assist the identification of patterns across individual cancer samples. Bioinformatics 28, 2981–2982 (2012).
    1. Krzywinski M. et al.. Circos: an information aesthetic for comparative genomics. Genome Res. 19, 1639–1645 (2009).
    1. van Bokhoven A. et al.. Molecular characterization of human prostate carcinoma cell lines. Prostate 57, 205–225 (2003).
    1. Hay C. W. & McEwan I. J. The impact of point mutations in the human androgen receptor: classification of mutations on the basis of transcriptional activity. PLoS ONE 7, e32514 (2012).

Source: PubMed

3
Subscribe