Purpose: Precise detection of copy number aberrations (CNA) from tumor biopsies is critically important to the treatment of metastatic prostate cancer. The use of targeted panel next-generation sequencing (NGS) is inexpensive, high throughput, and easily feasible, allowing single-nucleotide variant calls, but CNA estimation from this remains challenging.

Experimental Design: We evaluated CNVkit for CNA identification from amplicon-based targeted NGS in a cohort of 110 fresh castration-resistant prostate cancer biopsies and used capture-based whole-exome sequencing (WES), array comparative genomic hybridization (aCGH), and FISH to explore the viability of this approach.

Results: We showed that this method produced highly reproducible CNA results (r = 0.92), with the use of pooled germline DNA as a coverage reference supporting precise CNA estimation. CNA estimates from targeted NGS were comparable with WES (r = 0.86) and aCGH (r = 0.7); for key selected genes (BRCA2, MYC, PIK3CA, PTEN, and RB1), CNA estimation correlated well with WES (r = 0.91) and aCGH (r = 0.84) results. The frequency of CNAs in our population was comparable with that previously described (i.e., deep deletions: BRCA2 4.5%; RB1 8.2%; PTEN 15.5%; amplification: AR 45.5%; gain: MYC 31.8%). We also showed, utilizing FISH, that CNA estimation can be impacted by intratumor heterogeneity and demonstrated that tumor microdissection allows NGS to provide more precise CNA estimates.

Conclusions: Targeted NGS and CNVkit-based analyses provide a robust, precise, high-throughput, and cost-effective method for CNA estimation for the delivery of more precise patient care. Clin Cancer Res; 23(20); 6070–7. ©2017 AACR.

Translational Relevance

Detection of oncogenic genomic aberrations has led to novel therapeutic strategies for treating cancer. We recently showed that 20% to 30% of metastatic prostate cancers have deleterious defects in DNA repair machinery affecting genes, such as BRCA2, ATM, PALB2, BRCA1, and CDK12. These aberrations are associated with sensitivity to PARP inhibitors. Robust methods are needed to identify these tumor subtypes. We have previously described a targeted next-generation sequencing method that identifies single-nucleotide variants and copy number aberrations, with precision, from formalin-fixed, paraffin-embedded tumor biopsy samples. We now describe the bioinformatics pipeline for this robust high-throughput, cost-efficient, and rapid solution. We have implemented these methods into clinical trial protocols to enable patient stratification.

Prostate cancer is a highly heterogeneous disease with distinct genomic underpinnings (1) and generally presents a modest number of single point mutations and small insertions and deletions (indels) at approximately four per megabase (2, 3), but frequently has large-scale copy number and structural alterations (4, 5). We recently showed that 20% to 30% of prostate tumors bear defects in DNA repair genes that render them sensitive to treatment with PARP inhibition, and that these are commonly mediated through deletions in these tumor suppressors (6).

High-throughput, inexpensive, multiplex biomarker assays for molecular stratification are needed to guide therapeutic choices and support clinical trial design. Although high-coverage whole-exome or genome sequencing can reliably assess tumor genomics, cost and bioinformatic demands currently restrict their routine clinical implementation to stratify patients for treatment. Targeted next-generation sequencing (NGS) represents an opportunity for implementing genomics in clinical practice with the advantages of a rapid turnaround time, lower cost, and the ability to concurrently analyze multiple genes with a limited bioinformatic analysis burden. Existing approaches to call germline and somatic single-nucleotide variants (SNV) and short indels from targeted sequencing data are well established and have led to a number of FDA biomarker-dependent drug approvals, including the PARP inhibitor olaparib in the past decade (7, 8). However, in the case of PARP inhibition for homologous recombination repair defective cancers, screening for tumors with these DNA repair defects relies on detecting gene copy number changes.

In this study, we identify gene copy number aberrations (CNA), based on targeted amplicon sequencing of a focused biomarker gene panel dedicated to identifying DNA repair defects. We used a patient cohort of metastatic castration-resistant prostate cancer (CRPC) to explore the utility of this approach. This targeted gene panel focused on the study of 113 genes important to DNA repair machinery, including BRCA2, BRCA1, ATM, PALB2, and CDK12 as well as other important potentially actionable prostate cancer genes, including AR, SPOP, PIK3CA, PTEN, AKT1, AKT2, and MYC (1, 6).

Patient selection and sample preparation

Patient tumor (biopsies) and germline (buccal swab and saliva) samples were collected as part of the Royal Marsden ethics committee–approved CCR2472 protocol. Samples were collected, annotated, stored, and reviewed as described previously (6). All samples were collected between July 25, 2015, and October 17, 2016.

Briefly, biopsies were paraffin embedded, DNA from biopsies was extracted using the QIAamp DNA FFPE Tissue Kit (Qiagen), Quant-iT Picogreen High-Sensitivity double-stranded DNA (dsDNA) Assay Kits (Invitrogen, Thermo Fisher Scientific) and QC check by FFPE QC Kit (Illumina).

Targeted panel design

A customized Generead v2 DNAseq Panel (Qiagen) panel was used for library construction; the exonic regions of 113 genes were included in the targeted panel, these being selected for being potentially actionable and/or involved in DNA damage repair processes and/or in prostate carcinogenesis (Supplementary Table S1; ref. 6). The panel covered 564 kb of the human genome, and each of the 113 genes was covered by an average of 18.69 probes (SD, 13.64).

Targeted and exome sequencing

Targeted amplicon sequencing was performed using the Illumina MiSeq platform following the manufacturers' protocol. FASTQ files were generated using the Illumina MiSeq Reporter v2.5.1.3 (9, 10). Reads were aligned to the human reference genome (hg19) using BWA. Whole-exome sequencing (WES) was performed at the University of Michigan (Ann Arbor, MI) as described previously (1).

FastQC (v0.11.2) and Samtools (0.1.19) were used to assess sequencing quality. Targeted sequencing samples were excluded from copy number analysis on the basis of: insufficient (<0.5 million) total read counts, low (<95%) percentage of properly paired reads, and low (<99.99%) percentage of on-target reads. Exome sequencing samples were rejected if the FastQC per-base quality score for 75% of the reads was less than Q20 over the first 80 bases, and alignment quality was monitored with Picard, as described previously (1).

CNAs

We used the software CNVkit (v0.3.5; ref. 11) to analyze sequencing coverage and copy number in the aligned sequencing reads from targeted amplicon sequencing of tumor and germline samples. Sequencing coverage of targeted regions in germline samples was assessed and used to create pooled reference data that included the technical variability at each covered region. Regions that were poorly amplified or mapped were masked from further analysis. The analyses of germline samples were also performed with CNVkit to validate sample quality. The read depths of tumor samples were accessed, normalized (corrected for GC content, target footprint size and spacing, and repetitive sequences), and individually compared with the reference, and the circular binary segmentation (CBS) algorithm (12) was used to infer copy number segments. Very small copy changes (≤3 bins) were treated as artifacts. Poor samples were also identified through high (>0.8) values of the segment interquartile range (IQR), a metric for the spread of bin-level copy ratios within each segment following application of the CBS algorithm. Copy number segments were annotated to genes, and regions bearing a log2 ratio of at least ±0.4 were identified as suggestive of shallow deletions or gains (11). Segments with log2 <−1.2 were classified as deep deletions, and those with log2 >2 were classified as amplifications. Experimental noise was identified as a log2 ratio SD of approximately 0.2.

WES CNA data were extracted from a previously reported Su2C-PCF dataset (1). Briefly, log2 ratios were derived on a per-gene basis from circular-binary segmented, Lowess-normalized log2 transformed coverage ratio between each tumor and matched normal sample.

Array comparative genomic hybridization

Tumor DNA from prostate cancer patients and male reference DNA from Agilent were amplified using Sigma WGA2 Kit (Sigma-Aldrich) according to the manufacturer's recommendations, and the amplified DNA was quantified by the Qubit fluorometric quantitation method (Thermo Fisher Scientific). Amplified tumor DNA (500 ng) was then fluorescently labeled with Cy5, and male reference DNA labeled with Cy3, using the SureTag Complete DNA Labeling Kit (Agilent Technologies). Labeled DNA was subsequently hybridized utilizing the Agilent SurePrint G3 Human CGH Microarray Kit, 4 × 180 K according to the manufacturer's instructions. The slides were then scanned and analyzed using the CytoGenomics Software v 4.0.3.12 (Agilent Technologies). To compare array comparative genomic hybridization (aCGH) and NGS results, we used the log2 ratio of the aCGH segments that overlap with genes present in the NGS panel. Genes not part of a copy-altered segment were included in the analysis by using the mean log2 ratio for probes, if the value was no greater than 0.25. Genes on the X-chromosome were excluded from comparison with results from the male reference–based panel data as a female reference was used for aCGH.

FISH

RB1 FISH was performed using a standard formalin-fixed, paraffin-embedded (FFPE) hybridization method (13) on 3-μm FFPE tissue slices adjacent to hematoxylin and eosin sections that were confirmed to contain a minimum of 50 intact cells. Briefly, RB1 status was determined using Vysis LSI 13 RB1 (13q14) probe (catalog # 08L65-020; Abbott Laboratories) and a reference probe Vysis 13q34 (catalog # 05N34-020; Abbott Laboratories). Nineteen z-stacks were used to assess cell status.

Metastatic prostate cancer sample characteristics

Freshly collected metastatic CRPC (mCRPC) biopsies (n = 110) were collected from patients with progressing disease (Supplementary Table S2) and assessed for copy number changes by sequencing with our targeted panel. Germline DNA samples (buccal swab) were taken from 34 consenting patients to use as a baseline reference and for additional analyses. Tumor biopsies were from: bone (49.1%), lymph nodes (35.5%), liver (9.1%), soft tissue disease (3.6%), and transurethral resection of the prostate (3.6%).

CNA estimation reproducibility

Accurately assessing the depth of coverage of genes can be biased by high GC content and repetitive regions (11, 14), so we first evaluated CNA reproducibility on a per-gene basis between technical replicates. Thirteen samples were sequenced in duplicate to assess technical variation; 12 of these duplicate datasets included repeated library preparation from the same DNA extraction, while one involved resequencing of the same library twice.

We identified 15 genes that produced less concordant results between replicates, including five genes with insufficient number of targeted regions (four or less probes) per gene (NRAS, NFKBIA, FANCF, FAM46C, CDKN1B), and 10 (MAP2K2, CDKN2A, HRAS, RECQL4, NOTCH1, FGFR3, STK11, TSC2, SMARCA4, AXIN1) that contained repetitive or polymorphic exonic regions that impeded accurate read alignment and possessed high coverage variability (Supplementary Fig. S1). These genes were excluded from downstream CNA analyses.

Following removal of the aforementioned genes, CNA estimates between duplicates showed high correlation (Pearson r correlation coefficient = 0.92; Fig. 1A). All genes with deep deletions (log2 ratio, <−1.2) or amplifications (log2 ratio > 2) had a similar (at least ±0.4) CNA estimation replicated in the duplicate sample. A total of 95% of samples with a log2 ratio change of at least ±0.4 had a similar result in the duplicate.

Figure 1.

Copy number estimation results by targeted NGS and the described bioinformatic method were highly reproducible. Correlations of log2 results (overplotted) shown from: replicate samples (A; n = 13); matched germline reference (B; 25 pairs available) against pooled germline reference. Pearson r values presented. Comparing tools for generating informative log2 ratios showed that targeted panel NGS of CRPCs produced valid results for identification of CNAs when compared with orthogonal methods. Shown are correlations of log2 results (overplotted) from: panel data with exome data (C; n = 13); and panel data with aCGH results (D; n = 9). Pearson r values depicted. CNA orthogonal validation focusing on selected genes of therapeutic relevance to CRPC presented as log2 ratios in comparison with exome (E) and aCGH (F) data from the same samples compared with panel data. Colors indicate gene: BRCA2 = red, MYC = orange, PIK3CA = dark blue, PTEN = light blue, and RB1 = yellow. Pearson r values are shown.

Figure 1.

Copy number estimation results by targeted NGS and the described bioinformatic method were highly reproducible. Correlations of log2 results (overplotted) shown from: replicate samples (A; n = 13); matched germline reference (B; 25 pairs available) against pooled germline reference. Pearson r values presented. Comparing tools for generating informative log2 ratios showed that targeted panel NGS of CRPCs produced valid results for identification of CNAs when compared with orthogonal methods. Shown are correlations of log2 results (overplotted) from: panel data with exome data (C; n = 13); and panel data with aCGH results (D; n = 9). Pearson r values depicted. CNA orthogonal validation focusing on selected genes of therapeutic relevance to CRPC presented as log2 ratios in comparison with exome (E) and aCGH (F) data from the same samples compared with panel data. Colors indicate gene: BRCA2 = red, MYC = orange, PIK3CA = dark blue, PTEN = light blue, and RB1 = yellow. Pearson r values are shown.

Close modal

A matched germline sample is commonly used for identifying deviations in coverage ratios, so we sought to compare this with our pooled reference approach. For the pooled reference, we sequenced 34 unmatched germline samples and assessed the depth of coverage as described previously (11). Regions with low variability were identified in the pooled reference and used to weight downstream CNA estimates (11). This approach produced a highly similar result (overall Pearson r = 0.93) when compared with the CNA estimation approach using a matched normal as reference, when matched normal samples were available (Fig. 1B); however, the CNA estimation confidence score (IQR value) was improved when using pooled germline samples (paired t test P = 0.05). This was in line with other reports (14) that indicated that using pooled germline sequencing data as reference offered a robust method to identify gene CNAs.

CNA validation by exome sequencing and aCGH

We cross-compared copy number log2 ratios for all evaluable genes on the targeted panel with log2 ratios generated by exome sequencing (n = 13; Fig. 1C), resulting in a Pearson r correlation coefficient of 0.86. Moreover, all the genes with a log2 ratio <−1.2 by targeted sequencing, in keeping with putative “deep” deletions, exhibited a similar (at least ±0.4) result in the exome sequencing. For example, one sample bore AR amplification (log2 ratio 5.7 in targeted NGS and 4.91 in WES) alongside WRN deletion (−1.53 vs. −1.17) and ATM deletion (−0.68 vs. −0.56). We also compared log2 ratios from aCGH segments (n = 9) with gene copy number estimations by panel NGS among the genes covered by both platforms and found log2 ratios were also concordant (Fig. 1D), with a Pearson r value of 0.7.

We then sought to further validate copy number calls for specific genes of clinical relevance, such as BRCA2, MYC, PIK3CA, PTEN, and RB1 (Fig. 1E and F). For these genes, we found that our panel produced results highly concordant with exome sequencing and aCGH data, with Pearson r values of 0.91 and 0.84, respectively. All clinically relevant deletions of BRCA2 in samples with exome or aCGH data were also identified in our panel results (Table 1).

Table 1.

Validation of BRCA2 results from targeted panel CNV analysis with either whole-exome sequencing or array comparative genome hybridization (log2 ratios are shown)

Validation of BRCA2 results from targeted panel CNV analysis with either whole-exome sequencing or array comparative genome hybridization (log2 ratios are shown)
Validation of BRCA2 results from targeted panel CNV analysis with either whole-exome sequencing or array comparative genome hybridization (log2 ratios are shown)

CNA profiles in CRPC

In the analyses of this cohort of advanced prostate cancer samples, the prevalence of the CNAs detected with this assay was consistent with that of previously reported studies (Fig. 2). CNAs of AR (amplification; log2 > 2) were found in 45.5% (50/110) of tumor samples. Broad deletions of chromosome 13 (1) involving the shared loss of BRCA2 and RB1 were detected as previously described with 23 of 26 (88%) samples with any BRCA2 loss also having some loss of RB1, while overall 23 of 66 (34.8%) samples with RB1 loss had loss of BRCA2 (Fig. 2A).

Figure 2.

A, Binned heatmap of copy number aberrations for all evaluated (n = 110) samples with colors defined by log2 ratio thresholds (dark blue, <−1.2; light blue, <−0.4; pink, >0.4; red, >2). Rows and columns clustered by complete Euclidean distance for visualization purposes only. Commonly deleted genes, such as PTEN, RB1, and BRCA2, cluster toward the left, while commonly amplified genes, such as AR, MYC, BRAF, and KRAS, trend to the right. Genes that are closely located in chromosomal coordinates (e.g., CHEK2 and NF2, BLM and MAP2K1, RB1 and BRCA2) often share similar copy number aberrations. B, Scatter plot showing that the percentage of samples with specific copy-number changes in our targeted sequencing cohort was similar to two recently published studies of CNAs in CRPC (1, 15). Colors indicate type of aberration (red = amplification, pink = gain, blue = deletion). Shape of points indicate study data source. Data were derived from copy number frequency data publicly available from cBioPortal (21, 22), retrieved on November 17, 2016.

Figure 2.

A, Binned heatmap of copy number aberrations for all evaluated (n = 110) samples with colors defined by log2 ratio thresholds (dark blue, <−1.2; light blue, <−0.4; pink, >0.4; red, >2). Rows and columns clustered by complete Euclidean distance for visualization purposes only. Commonly deleted genes, such as PTEN, RB1, and BRCA2, cluster toward the left, while commonly amplified genes, such as AR, MYC, BRAF, and KRAS, trend to the right. Genes that are closely located in chromosomal coordinates (e.g., CHEK2 and NF2, BLM and MAP2K1, RB1 and BRCA2) often share similar copy number aberrations. B, Scatter plot showing that the percentage of samples with specific copy-number changes in our targeted sequencing cohort was similar to two recently published studies of CNAs in CRPC (1, 15). Colors indicate type of aberration (red = amplification, pink = gain, blue = deletion). Shape of points indicate study data source. Data were derived from copy number frequency data publicly available from cBioPortal (21, 22), retrieved on November 17, 2016.

Close modal

Overall, the evaluation of the deep deletions of commonly aberrant CRPC genes revealed results in keeping with previous reports from two recent studies of CRPC genomics (1, 15); PTEN genomic deep deletions were detected in 17 of 110 (15.5%), BRCA2 deep deletion in 5 of 110 (4.5%), and RB1 deep deletion in 9 of 110 (8.2%) CRPC samples. In addition, gain of the MYC locus was found in 36 of 110 samples (32.7%). We found that the copy number alteration frequencies in our cohort were in line with expected CRPC frequencies.

Exploring tumor purity and heterogeneity

To further explore how tumor purity altered the capacity of the assay to detect CNAs, we pursued the serial dilution of tumor DNA acquired from a sample estimated by pathology review as having 80% tumor content, with same-patient germline DNA. This sample harbored a somatic, BRCA2 homozygous deletion (independently confirmed WES). We then sequenced the resulting dilutions (Fig. 3A) and found that homozygous deletions were easily distinguishable with tumor content purities >60% and that clonal CNAs were detectable with purity as low as 30%.

Figure 3.

A, Serial dilution of a tumor sample with matched germline DNA and resequencing highlights functional limits of copy number detection. Homozygously deleted (BRCA2, PTEN – dark blue), hemizygously deleted (RB1 – light blue), copy-neutral (SPOP – gray), and copy-gained (PIK3CA – pink) genes are shown. B, Microdissection of a biopsy to enrich tumor content emphasizes a BRCA2 deletion, as the segment log2 ratio (orange line) shifts from −1.15 to −2.82. Also shown are individual exon-level copy ratios (blue points), from which the segment is assigned. C, Stacked bar plot showing RB1-FISH results for 18 samples (50 cells each), with colors indicating observable copies of RB1 for individual cells. Navy, 0 copies; light blue, 1 copy; white, 2 copies; pink, >2 copies. Black bar, samples that had a deletion of any kind reported by our targeted NGS protocol.

Figure 3.

A, Serial dilution of a tumor sample with matched germline DNA and resequencing highlights functional limits of copy number detection. Homozygously deleted (BRCA2, PTEN – dark blue), hemizygously deleted (RB1 – light blue), copy-neutral (SPOP – gray), and copy-gained (PIK3CA – pink) genes are shown. B, Microdissection of a biopsy to enrich tumor content emphasizes a BRCA2 deletion, as the segment log2 ratio (orange line) shifts from −1.15 to −2.82. Also shown are individual exon-level copy ratios (blue points), from which the segment is assigned. C, Stacked bar plot showing RB1-FISH results for 18 samples (50 cells each), with colors indicating observable copies of RB1 for individual cells. Navy, 0 copies; light blue, 1 copy; white, 2 copies; pink, >2 copies. Black bar, samples that had a deletion of any kind reported by our targeted NGS protocol.

Close modal

We also performed microdissection of a tumor biopsy (pathologist tumor purity estimate, ∼40%) bearing borderline BRCA2 homozygous deletion (log2 ratio of −1.15) and resequenced the resulting tumor-enriched sample (purity estimate, ∼80%). This step further shifted the CNA estimates away from normal, with the log2 ratio for BRCA2 dropping to −2.82, an unambiguous deletion (Fig. 3B).

The log2 copy ratios estimated from sequencing a bulk of cells, as in our protocol, can be biased by tumor sample heterogeneity. To assess this relationship, we performed FISH assays of the RB1 gene, counting the number of cells where each copy number (0 through 3+) was observed (Fig. 3C). Our results showed an association between the proportion of cells with 1 or 0 copies of RB1 (hemizygous or homozygous deletion) and putative copy loss inferred from our targeted sequencing panel (unpaired t test P = 0.02, n = 18, df = 15.884). However, the full complexity of aneuploidy in individual tumor cells is masked by bulk tissue sequencing, which effectively averages the estimated DNA content across all cells in a sample. FISH analyses revealed that many tumor samples contained a mixture of cells with different copy number changes at the RB1 locus, in highly heterogeneous cell populations (Supplementary Fig. S2).

The ability to rapidly, inexpensively, and accurately identify cancers with actionable genomic defects in tumor biopsies is now becoming critically important for advanced prostate cancer care. We have previously shown that a proportion of metastatic CRPCs have DNA repair defects and are sensitive to treatment with PARP inhibitors (6), with emerging studies of platinum-based chemotherapies also indicating important antitumor activity in this population (16). These and other data support further studies of genomic stratification for treatment selection of patients suffering from mCRPC. Here, we demonstrate the analytic validation and clinical use of a targeted NGS assay that allows for copy number screening for predictive biomarkers of response, enabling the integration of more precise patient stratification into trial design.

Our method reliably estimates gene copy number changes from amplicon-based targeted NGS with the acquired results being comparable with widely used assays, including exome sequencing, aCGH, digital droplet PCR, and FISH (17). Critically, the data processing time for this panel-based CNA estimation is substantially shorter of that for exome sequencing and can be easily completed even with a personal computer. In addition, we have confirmed that using pooled germline samples as a reference can produce equivalent and robust results to utilizing matched normal samples. This feature provides the possibility of diagnosing patients with only tumor samples for whom germline DNA is not available, and can dramatically reduce the overall sequencing costs of large studies.

This approach is currently being applied in the TOPARP-B (NCT01682772) study as part of an ongoing attempt to clinically qualify DNA repair pathway aberrations as a predictive biomarker for treatment with PARP inhibition with olaparib (16). However, this assay has utility beyond this study. The gene panel we utilized covers several key genomic aberrations that are of emerging interest as possible clinical predictive biomarkers that may be actionable in prostate cancer, including loss of PTEN, gain of PIK3CA (both may sensitize tumors to PI3K/AKT pathway inhibition; refs. 18, 19), and amplification of MYC (MYC-driven tumors have been reported to sensitize to AURA and BET inhibition and can result in replication stress; ref. 20). Prospectively identifying tumors bearing these CNAs and other deleterious mutations, utilizing such validated methods, is likely to become critically important in defining future treatment strategies for this commonest of male cancers.

We explored the functional limits of assessing CNAs and found that our pipeline can confidently estimate deletion when tumor content is reduced to 60% (homozygous and heterozygous). As tumor content decreases, the pipeline struggles to differentiate between homozygous and heterozygous deletions, and we estimate that interpreting results from samples with <30% purity may be extremely challenging for targeted NGS panels. We have shown, however, that the use of microdissection to enrich tumor content offers a possible solution to resolve these issues.

This approach also suffers from the nature of amplicon-based NGS, including difficulty identifying precise breakpoints, very focal gains or deletions, unequal amplification among genes, and coverage variation across polymorphic repeat regions. Furthermore, because the number of SNVs identified in targeted panels will be much lower when compared with exome or whole-genome sequencing, SNV allele frequencies may not be sufficient to assist with copy number estimation or to detect allelic imbalance.

It is important to note that we did not systematically compare software tools in this study and that different clinical requirements will require individual optimizations. As the field develops, other software tools will emerge to improve the precision of CNA identification in targeted sequencing, and independent validations will be critical in guiding further work.

In conclusion, we have described the validation and clinical application of an assay evaluating 100 genes utilized to assess both deleterious tumor gene copy number, and mutations, from targeted NGS of prostate cancer samples from the most common metastatic sites, including bone biopsies. We have validated these results by orthogonal methods, including not only exome sequencing, but also aCGH and FISH. Such targeted panel NGS assays are likely to become central to future patient care in cancer therapy.

S. Sandhu is a consultant/advisory board member for Amgen, Bristol-Myers Squibb, and Janssen. J.S. de Bono is a consultant/advisory board member for AstraZeneca. No potential conflicts of interest were disclosed by the other authors.

Conception and design: G. Seed, W. Yuan, J. Mateo, S. Carreira, G. Boysen, S. Miranda, S. Sandhu, A.M. Chinnaiyan, J.S. de Bono

Development of methodology: G. Seed, S. Carreira, S. Miranda, E. Talevich, S. Sandhu, A.M. Chinnaiyan, J.S. de Bono

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G. Seed, J. Mateo, S. Carreira, C. Bertan, R. Ferraldeschi, M. Crespo, D.N. Rodrigues, D.R. Robinson, Y.-M. Wu, S. Sandhu, J.S. de Bono

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): G. Seed, W. Yuan, J. Mateo, S. Carreira, M. Lambros, D.N. Rodrigues, E. Talevich, D.R. Robinson, L.P. Kunju, Y.-M. Wu, R. Lonigro, J.S. de Bono

Writing, review, and/or revision of the manuscript: G. Seed, W. Yuan, J. Mateo, S. Carreira, M. Lambros, G. Boysen, S. Miranda, I. Figueiredo, R. Riisnaes, D.N. Rodrigues, E. Talevich, R. Lonigro, S. Sandhu, A.M. Chinnaiyan, J.S. de Bono

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): G. Seed, I. Figueiredo, D.N. Rodrigues, J.S. de Bono

Study supervision: W. Yuan, J.S. de Bono

We thank the staff at the Prostate Targeted Therapy Group and all the patients who agreed to participate in this study.

This study was supported by Cancer Research UK, Prostate Cancer UK, and the Stand Up To Cancer – Prostate Cancer Foundation Prostate Dream Team Translational Research Grant (grant number: SU2C-AACR-DT0712). Research grant administered by the American Association for Cancer Research, the Scientific Partner of SU2C. Support was also provided by an Experimental Cancer Medicine Centre grant and a Medical Research Council/Prostate Cancer UK and PCF Young Investigator Award to J. Mateo. G. Seed was supported by a Prostate Cancer UK PhD studentship (TLD-S15-006), and G. Boysen was supported by a Marie Curie International Incoming Fellowship (625792). This study was also supported by research funding from the EU FP7 project CTCTrap #305341.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Robinson
D
,
Van Allen
EM
,
Wu
YM
,
Schultz
N
,
Lonigro
RJ
,
Mosquera
JM
, et al
Integrative clinical genomics of advanced prostate cancer
.
Cell
2015
;
161
:
1215
28
.
2.
Baca
SC
,
Prandi
D
,
Lawrence
MS
,
Mosquera
JM
,
Romanel
A
,
Drier
Y
, et al
Punctuated evolution of prostate cancer genomes
.
Cell
2013
;
153
:
666
77
.
3.
Barbieri
CE
,
Baca
SC
,
Lawrence
MS
,
Demichelis
F
,
Blattner
M
,
Theurillat
JP
, et al
Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer
.
Nat Genet
2012
;
44
:
685
9
.
4.
Mitchell
T
,
Neal
DE
. 
The genomic evolution of human prostate cancer
.
Br J Cancer
2015
;
113
:
193
8
.
5.
Grasso
CS
,
Wu
YM
,
Robinson
DR
,
Cao
X
,
Dhanasekaran
SM
,
Khan
AP
, et al
The mutational landscape of lethal castration-resistant prostate cancer
.
Nature
2012
;
487
:
239
43
.
6.
Mateo
J
,
Carreira
S
,
Sandhu
S
,
Miranda
S
,
Mossop
H
,
Perez-Lopez
R
, et al
DNA-repair defects and olaparib in metastatic prostate cancer
.
N Engl J Med
2015
;
373
:
1697
708
.
7.
Ledermann
J
,
Harter
P
,
Gourley
C
,
Friedlander
M
,
Vergote
I
,
Rustin
G
, et al
Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised phase 2 trial
.
Lancet Oncol
2014
;
15
:
852
61
.
8.
Fong
PC
,
Boss
DS
,
Yap
TA
,
Tutt
A
,
Wu
P
,
Mergui-Roelvink
M
, et al
Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers
.
N Engl J Med
2009
;
361
:
123
34
.
9.
Ong
M
,
Carreira
S
,
Goodall
J
,
Mateo
J
,
Figueiredo
I
,
Rodrigues
DN
, et al
Validation and utilisation of high-coverage next-generation sequencing to deliver the pharmacological audit trail
.
Br J Cancer
2014
;
111
:
828
36
.
10.
Carreira
S
,
Romanel
A
,
Goodall
J
,
Grist
E
,
Ferraldeschi
R
,
Miranda
S
, et al
Tumor clone dynamics in lethal prostate cancer
.
Sci Transl Med
2014
;
6
:
254ra125
.
11.
Talevich
E
,
Shain
AH
,
Botton
T
,
Bastian
BC
. 
CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing
.
PLoS Comput Biol
2016
;
12
:
e1004873
.
12.
Olshen
AB
,
Venkatraman
ES
,
Lucito
R
,
Wigler
M
. 
Circular binary segmentation for the analysis of array-based DNA copy number data
.
Biostatistics
2004
;
5
:
557
72
.
13.
Ferraldeschi
R
,
Nava Rodrigues
D
,
Riisnaes
R
,
Miranda
S
,
Figueiredo
I
,
Rescigno
P
, et al
PTEN protein loss and clinical outcome from castration-resistant prostate cancer treated with abiraterone acetate
.
Eur Urol
2015
;
67
:
795
802
.
14.
Grasso
C
,
Butler
T
,
Rhodes
K
,
Quist
M
,
Neff
TL
,
Moore
S
, et al
Assessing copy number alterations in targeted, amplicon-based next-generation sequencing data
.
J Mol Diagn
2015
;
17
:
53
63
.
15.
Kumar
A
,
Coleman
I
,
Morrissey
C
,
Zhang
X
,
True
LD
,
Gulati
R
, et al
Substantial interindividual and limited intraindividual genomic diversity among tumors from men with metastatic prostate cancer
.
Nat Med
2016
;
22
:
369
78
.
16.
Cheng
HH
,
Pritchard
CC
,
Boyd
T
,
Nelson
PS
,
Montgomery
B
. 
Biallelic inactivation of BRCA2 in platinum-sensitive metastatic castration-resistant prostate cancer
.
Eur Urol
2016
;
69
:
992
5
.
17.
Strom
CM
,
Rivera
S
,
Elzinga
C
,
Angeloni
T
,
Rosenthal
SH
,
Goos-Root
D
, et al
Development and validation of a next-generation sequencing assay for BRCA1 and BRCA2 variants for the clinical laboratory
.
PLoS One
2015
;
10
:
e0136419
.
18.
Yamamoto
Y
,
De Velasco
MA
,
Kura
Y
,
Nozawa
M
,
Hatanaka
Y
,
Oki
T
, et al
Evaluation of in vivo responses of sorafenib therapy in a preclinical mouse model of PTEN-deficient of prostate cancer
.
J Transl Med
2015
;
13
:
150
.
19.
Park
H
,
Kim
Y
,
Sul
JW
,
Jeong
IG
,
Yi
HJ
,
Ahn
JB
, et al
Synergistic anticancer efficacy of MEK inhibition and dual PI3K/mTOR inhibition in castration-resistant prostate cancer
.
Prostate
2015
;
75
:
1747
59
.
20.
Delmore
JE
,
Issa
GC
,
Lemieux
ME
,
Rahl
PB
,
Shi
J
,
Jacobs
HM
, et al
BET bromodomain inhibition as a therapeutic strategy to target c-Myc
.
Cell
2011
;
146
:
904
17
.
21.
Gao
J
,
Aksoy
BA
,
Dogrusoz
U
,
Dresdner
G
,
Gross
B
,
Sumer
SO
, et al
Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal
.
Sci Signal
2013
;
6
:
pl1
.
22.
Cerami
E
,
Gao
J
,
Dogrusoz
U
,
Gross
BE
,
Sumer
SO
,
Aksoy
BA
, et al
The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data
.
Cancer Discov
2012
;
2
:
401
4
.