Abstract
Background:
Primary and metastatic prostate cancers have low mutation rates and recurrent alterations in a small set of genes, enabling targeted sequencing of prostate cancer-associated genes as an efficient approach to characterizing patient samples (compared to whole-exome and whole-genome sequencing). For example, targeted sequencing provides a flexible, rapid, and cost-effective method for genomic assessment of patient-derived cell lines to evaluate fidelity to initial patient tumor samples.
Methods:
We developed a prostate cancer-specific targeted next-generation sequencing (NGS) panel to detect alterations in 62 prostate cancer-associated genes as well as recurring gene fusions with ETS family members, representing the majority of common alterations in prostate cancer. We tested this panel on primary prostate cancer tissues and blood biopsies from patients with metastatic prostate cancer. We generated patient-derived cell lines from primary prostate cancers using conditional reprogramming methods and applied targeted sequencing to evaluate the fidelity of these cell lines to the original patient tumors.
Results:
The prostate cancer-specific panel identified biologically and clinically relevant alterations, including point mutations in driver oncogenes and ETS family fusion genes, in tumor tissues from 29 radical prostatectomy samples. The targeted panel also identified genomic alterations in cell-free DNA and circulating tumor cells from patients with metastatic prostate cancer, and in standard prostate cancer cell lines. We used the targeted panel to sequence our set of patient-derived cell lines; however, no prostate cancer-specific mutations were identified in the tumor-derived cell lines, suggesting preferential outgrowth of normal prostate epithelial cells.
Conclusions:
We evaluated a prostate cancer-specific targeted NGS panel to detect common and clinically relevant alterations (including ETS family gene fusions) in prostate cancer. The panel detected driver mutations in a diverse set of clinical samples of prostate cancer, including fresh-frozen tumors, cell-free DNA, circulating tumor cells, and cell lines. Targeted sequencing of patient-derived cell lines highlights the challenge of deriving cell lines from primary prostate cancers and the importance of genomic characterization to credential candidate cell lines. Our study supports that a prostate cancer-specific targeted sequencing panel provides an efficient, clinically feasible approach to identify genetic alterations across a spectrum of prostate cancer samples and cell lines.
Keywords: Prostate cancer, next-generation sequencing, patient-derived cell lines, cell-free DNA, gene rearrangements
Introduction
The genomic landscape of primary and metastatic prostate cancers consists of a relatively limited number of recurrent somatic alterations, indicating that most common alterations in prostate cancer may be captured by sequencing a limited number of genes. Whole-exome sequencing (WES) and whole-genome sequencing (WGS) studies of large prostate cancer cohorts have identified recurrent somatic mutations including single nucleotide variants (SNVs), copy number changes, and structural rearrangements 1–7. Primary prostate cancers have a lower somatic mutation frequency (~1 mutation/Mb) compared to primary tumors of other cancer types (1–10 mutations/Mb) 8. Among primary prostate cancers, recurrent mutations commonly occur in TP53, SPOP, and FOXA1. Deletions of RB1, PTEN, and CHD1 and amplifications of MYC are common, as are ETS family gene fusions such as TMPRSS2-ERG.
Comprehensive genomic sequencing has also elucidated the landscape of alterations in metastatic castration-resistant prostate cancer (mCRPC) 6,7,9–13. Even within this group of metastatic tumors, a relatively low mutation rate was observed at 3–4 mutations/Mb. Somatic mutations in mCRPC tumors are found in several genes that are uncommonly altered in primary prostate cancers, including PIK3CA, PIK3CB, RSPO, BRAF, RAF1, APC, CTNNB1, and ZBTB16 (PLZF) 9. Alterations in the androgen receptor (AR) are common in mCRPC but are rarely seen in primary treatment-naïve prostate cancer. These alterations are associated with androgen therapy resistance and include SNVs and amplification of the AR gene locus; structural rearrangements of AR and amplifications affecting an AR enhancer are also seen 10,11. Deletions and mutations in PTEN and TP53 are present in 40–50% of cases. A substantial fraction of patients harbor defects in DNA repair proteins, and such patients may exhibit responses to poly(ADP-ribose) polymerase (PARP) inhibitor therapy 14. Furthermore, patients with metastatic prostate cancer have a high frequency (12%) of germline DNA repair alterations, which may have implications for both therapy and prevention of prostate cancer 15.
ETS family gene fusions are present in about 50% of primary and metastatic prostate cancers. Detection of ETS rearrangements typically relies on fluorescence in situ hybridization, whole-genome sequencing, or RT-PCR (which may be complicated by variations in fusion architecture), but these methods are not part of standard clinical practice 16.
Despite progress in the genomic characterization of prostate cancers, insufficient cell line models are available to represent the genetic diversity elucidated in these sequencing studies. Human prostate cancer cells are difficult to grow in cell culture; thus, few prostate cancer cell lines are available 17, and do not encompass the full spectrum of genetic alterations observed in patients, such as TMPRSS2-ERG, SPOP mutations, and CHD1 deletions 18. New methods to generate human prostate cancer cell lines include organoid methods 19–22 and a “conditional reprogramming” method with irradiated mouse fibroblast feeder cells and an inhibitor of Rho-associated kinase 23–26. These methods support the growth of both normal and malignant prostate cells from patients; indeed, growth of normal prostate cells is robust in both conditional reprogramming and organoid systems 26,27. Thus, accurate methods are required to distinguish tumor and normal cells in new patient-derived cultures.
To address these challenges, we designed a prostate cancer-specific targeted next-generation sequencing (NGS) panel and applied it to evaluate genomic features of patient-derived cell line models and patient samples. Our targeted panel incorporates exon sequences of 62 genes frequently altered in prostate cancer as well as tiling across the intronic regions of genes with frequent rearrangements and copy alterations in prostate cancer. This approach allows single-nucleotide variants (SNVs), amplifications/deletions, and rearrangements to be simultaneously detected in the sequencing process, and is flexible in design such that the selection of genes can change over time. The prostate cancer-specific targeted panel was applied to primary prostate tumors as well as other clinical specimen types including circulating tumor cells (CTCs) and cell-free DNA (cfDNA) from metastatic prostate cancer patients. The panel detected driver mutations and/or rearrangements of known prostate cancer-associated genes in the majority of patient samples and in commonly used prostate cancer cell lines. In parallel, we attempted to generate new patient-derived cell lines from normal and malignant prostate tissue from primary prostate cancers using conditional reprogramming. We applied the prostate cancer-specific panel to interrogate the genomic features of the patient tissue samples and patient-derived cell lines.
Methods
Patient samples and cell lines
A schematic overview of the study is shown in Figure 1. We received Institutional Review Board (IRB) and institutional approval from Dana-Farber Cancer Institute (DFCI) and Massachusetts General Hospital (MGH) to collect samples of primary prostate cancers and adjacent normal prostate tissue from prostatectomy surgeries. Genitourinary pathologists at DFCI or MGH selected prostate cancer tissue fragments and adjacent normal-appearing prostate tissue from prostatectomy specimens. Fresh tissues were dissociated, and cell lines were generated according to the conditional reprogramming protocol 23, with modifications as detailed in Supplementary Methods. Briefly, cell suspensions were generated by mechanical dissociation and enzymatic digestion, followed by plating cells in F-medium on a layer of irradiated 3T3-J2 fibroblasts or with conditioned medium from these fibroblasts 28–30. Additional samples from each specimen were snap-frozen or cryopreserved in FBS/10% DMSO. We also acquired DNA samples from 2 tumors previously collected in a whole-genome sequencing study 1. We acquired CTC DNA and cfDNA samples from patients with metastatic prostate cancer that were previously isolated and sequenced under IRB-approved clinical protocols at DFCI 31,32. Prostate cancer cell lines were obtained from collaborators at DFCI and cultured in standard medium.
Figure 1.
Schematic of patient samples. Prostate cancer tissue and adjacent normal tissue from primary prostatectomies were used to generate patient-derived cell lines; both tumors and cell lines were sequenced with a prostate cancer-specific targeted panel. The targeted panel was also applied to circulating tumor cells and cell-free DNA from patients with metastatic prostate cancer.
Prostate cancer targeted panel
The targeted panel used in this study includes 62 genes that are recurrently mutated in prostate cancer and/or relevant to prostate cancer biology, selected based on published studies at the time of design of the bait set (Table 1) 1,7,33. We designed RNA bait sequences (Agilent SureDesign, Custom SureSelect XT platform) targeting the exons of these genes as well as intronic and intergenic regions of ERG, TMPRSS2, ETV1, ETV4, SLC45A3, RAF1, and PTEN; total bait size is 790 kb.
Table 1.
List of prostate cancer-associated genes in the targeted panel
| AKT1 |
| APC |
| AR |
| ARID1A |
| ATM |
| BRAF |
| BRCA2 |
| C14orf49 |
| CDK12 |
| CDKN1B |
| CDKN2A |
| CHD1 |
| CTNNB1 |
| EGFR |
| ELAC2 |
| EPHA7 |
| ERG* |
| ETV1* |
| ETV4* |
| ETV5 |
| EZH2 |
| FGFR3 |
| FOXA1 |
| FOXP1 |
| HOXB13 |
| HRAS |
| IDH1 |
| KDM6A |
| KRAS |
| MED12 |
| MLH3 |
| MLL |
| MLL2 |
| MLL3 |
| MSH6 |
| MSR1 |
| MUTYH |
| MYC |
| NCOA2 |
| NCOR1 |
| NIPA2 |
| NKX3-1 |
| NRAS |
| OR5L1 |
| PIK3CA |
| PIK3R1 |
| PTEN* |
| RAF1* |
| RB1 |
| RNASEL |
| RYBP |
| SCN11A |
| SHQ1 |
| SKP2 |
| SLC45A3* |
| SPOP |
| SPTA1 |
| THSD7B |
| TMPRSS2* |
| TP53 |
| ZFHX3 |
| ZNF595 |
The most commonly altered genes in primary and metastatic prostate cancer were selected for inclusion in the panel. The custom RNA bait set for targeted capture targets the exons of most genes. Selected genes frequently involved in somatic copy number variations and/or rearrangements (indicated with an asterisk) had additional bait tiling across intronic and intergenic regions.
Targeted sequencing with prostate-specific panel of custom RNA baits
Snap-frozen solid tissue pieces were dissociated using TissueRuptor (Qiagen). DNA from these tissue or cell line samples was isolated using Allprep or DNeasy Blood and Tissue kits (Qiagen) and quantitated using Qubit dsDNA HS assay kit. The SureSelectXT Target Enrichment System for Illumina Paired-End Sequencing (Agilent) was used for library preparation and hybrid capture according to the manufacturer’s protocol with 200 ng input. The enriched DNA libraries were PCR-amplified using individual indexing primers containing 8-bp indexes to enable pooling for sequencing and subsequent deconvolution, followed by bead purification. DNA quantity/quality of the final amplified, indexed libraries was assessed by Agilent Bioanalyzer. Samples were pooled for sequencing on an Illumina HiSeq 2500 instrument with 200 paired-end reads.
Sequencing data analysis
Most genomic analysis was conducted in the Firehose environment (http://archive.broadinstitute.org/cancer/cga/firehose). Somatic single-nucleotide variants (SNVs) were called using MuTect 34, and short insertions and deletions (indels) were called using Indelocator (http://archive.broadinstitute.org/cancer/cga/indelocator), except in the case of whole-exome sequenced CTC and cfDNA samples which were called with Strelka 35. Data filtering steps and quality control thresholds are described in Supplementary Methods. To detect gene fusions, somatic rearrangements were identified using dRanger (http://archive.broadinstitute.org/cancer/cga/dranger), and the candidate rearrangement breakpoints were passed into Breakpointer 36. Only candidate gene fusions between TMPRSS2 or SLC45A3 and a defined list of fusion partners (ERG, ETV1, ETV3, ETV4, ETV5, ETV6, ELK4, or BRAF) were selected. Somatic mutations and fusion calls for the two WGS samples were obtained directly from a published WGS study 1. The Cancer Cell Line Encyclopedia (CCLE) data used to compare established cell lines sequenced on the targeted panel was based on a panel of 1651 genes that was used to profile mutations in CCLE cell lines (http://www.broadinstitute.org/ccle) 17. Our prostate cancer-specific targeted panel and the sequencing panel used in CCLE had 39 genes in common. Somatic copy number alterations (SCNAs) in samples sequenced using the targeted panel were assessed through an ad hoc method described in Supplementary Methods. Somatic CNAs in whole-exome sequenced samples were determined using ReCapSeg (http://gatkforums.broadinstitute.org/gatk/categories/recapseg). Each detected somatic alteration was evaluated for significance by manually assessing the frequency in prostate cancers via cBioPortal (http://www.cbioportal.org/) and COSMIC (https://cancer.sanger.ac.uk/cosmic) databases (accessed 3/2018).
Rearrangement analysis from cell-free DNA
An independent set of cfDNA samples from patients with metastatic CRPC underwent sequencing using a modified version of the prostate cancer-specific targeted panel that was adapted to detect common rearrangements and tile across the AR locus. cfDNA was extracted using the Qiagen Circulating DNA kit on the QIAsymphony liquid handling system. Tumor fraction was estimated from ultra-low pass whole genome sequencing by ichorCNA 32. cfDNA samples were quantitated using PicoGreen and their size distribution was analyzed by BioAnalyzer (Qiagen). 20 ng of input cfDNA was used for library construction using KAPA HyperPrep kit. The cfDNA was not fragmented before library prep. All libraries were quantified by sequencing on an Illumina MiSeq and then normalized based on the resulting sequencing metrics. The libraries were then pooled in equal mass to 500 ng and enriched using the bait set with the Agilent SureSelectXT Hybrid Capture kit. Pooled sample reads were de-convoluted and sorted using Picard tools 37, and aligned to the reference genome using BWA and the GATK tool for localized realignment around indel sites 38,39. Translocations were detected using Breakmer 40.
Results
Clinical cohort
Under IRB-approved protocols, 31 tumor specimens from primary prostatectomies were collected (Table 2), along with 29 paired normal prostate tissue specimens from the same surgeries, as assessed by a genitourinary pathologist. The tumor percentage in each tumor tissue piece was estimated by a genitourinary pathologist. One tumor/normal pair was excluded for low (<10%) tumor content in the tumor sample.
Table 2.
Clinical and pathologic data from primary prostatectomy samples
| Sample | Age | Gleason score | Pathologic stage | Tumor % |
|---|---|---|---|---|
| PK01 | 66 | 4+5 | pT2cN0 | 60% |
| PK02 | 66 | 3+4 | pT3aN0 | 90% |
| PK03 | 59 | 3+4 | pT2cN0 | 95% |
| PK04 | 52 | 3+4 | pT3bN0 | 70% |
| PK05 | 63 | 4+4 | pT3aN1 | 80% |
| PK06 | 67 | 4+4 | pT3bN0 | 80% |
| PK07 | 69 | 4+3 | pT3aN0 | 80% |
| PK08 | 56 | 4+5 | pT3bN1 | 100% |
| PK09 | 62 | 3+4 | pT2aN0 | 85% |
| PK010 | 57 | 4+4 | pT3bN0 | 95% |
| PK011 | 54 | 4+5 | pT3bN0 | 90% |
| PK012 | 47 | 3+4 | pT2cNx | 70% |
| PK013 | 64 | 3+4 | pT2cN0 | N/A |
| PK014 | 66 | 4+3 | pT2cN0 | N/A |
| PK015 | 71 | 3+4 | pT2cN0 | N/A |
| PK016 | 54 | 3+4 | pT3aN0 | N/A |
| PK018 | 71 | 4+5 | pT3aN0 | N/A |
| MGH1 | 52 | 3+3 | pT2cN0 | 90% |
| MGH2 | 67 | 4+5 | pT3aN0 | 70% |
| MGH3 | 71 | 4+3 | pT3bN0 | 80% |
| MGH4 | 66 | 3+4 | pT2cN0 | 90% |
| MGH5 | 70 | 3+4 | pT3aN0 | 50% |
| MGH6 | 52 | 3+3+(4) | pT2cNX | 40% |
| MGH7 | 58 | 4+3 | pT2aNx | N/A |
| MGH9 | 65 | 4+4 | pT3aN1 | 60% |
| MGH10 | 60 | 3+4 | pT3aN0 | 85% |
| MGH11 | 62 | 3+4 | pT2cN0 | 25% |
| MGH12 | 74 | 4+3 | pT2aN0 | 80% |
| MGH13 | 58 | 4+4 | pT3aN0 | 95% |
| MGH15 | 70 | 3+4 | pT3aN0 | 90% |
Clinical and pathologic data for primary prostatectomy samples. The fraction of tumor in each tumor tissue piece was estimated by a genitourinary pathologist and the maximum percent tumor is listed. Gleason score and pathologic stage were determined by a genitourinary pathologist. Age indicates the patient age at time of sample collection. N/A, not available
Prostate cancer-specific targeted sequencing panel samples and sequencing metrics
To validate the prostate cancer-specific targeted sequencing panel, we sequenced multiple prostate cancer sample types (Figure 1), including: 1) fresh-frozen tumor and normal tissue from 30 primary prostatectomies; 2) DNA from two samples of primary prostate cancer that had previously undergone WGS 1; 3) DNA from four individual CTCs from each of two patients with metastatic prostate cancer (DNA from the same CTCs and from a biopsy of each patient’s metastatic tumor underwent WES separately 31); 4) cfDNA isolated from the blood of two patients with metastatic prostate cancer (WES of CTCs and metastatic tumor biopsy from each patient was performed previously 32); and 5) eight prostate cell lines including immortalized prostate epithelial cells and tumor cell lines (Supplementary Table 1). In addition, we sequenced new patient-derived cell lines generated by conditional reprogramming from tumor and normal tissues (see below).
Across all samples, the mean target coverage was 118X (as expected, coverage varied depending on sample quality and number of samples pooled per lane). The distribution of coverage by sample and by gene is shown in Supplementary Figure 1. One tumor/normal pair was excluded due to poor coverage of the tumor sample. One normal tissue specimen failed library preparation. Two normal-tissue-derived cell lines were excluded due to poor coverage. Across all samples, the percent bases selected was approximately 20%. The percent selected is relatively low relative to optimized whole-exome sequencing panels, but is within the expected range for custom panels, and was also probably lower due to off-target effects related to the baits tiled across intronic regions.
Prostate cancer-specific targeted panel identifies genomic alterations in primary prostate tumors
Using the prostate cancer-specific panel, we obtained sequencing data from fresh-frozen tissue from 29 primary prostate cancers, along with paired normal tissue from the same individual in most instances. Data was analyzed for single nucleotide variants (SNVs), insertions/deletions, and copy number variations as described in Methods. In general, somatic variants were distinguished from germline by comparison with matched normal tissue. In cases where normal tissue data was not available, the tumor sequencing data was compared to a panel of normal individuals to distinguish somatic vs. likely germline events.
Somatic single nucleotide variants:
Of the 29 sequenced primary prostate cancers, 16 (55%) harbored detectable non-silent SNVs or coding indels in the prostate cancer-associated genes in the targeted sequencing panel (Table 3). Some samples had multiple somatic alterations. Several events were known “hotspot” mutations, including KRAS Q61R, TP53 R273G, and CTNNB1 K335I. Two SPOP mutations were identified – one (F102L) at a recurrent hotspot locus and one (F104C) in close proximity. Three alterations in FOXA1 were also identified: C227R, which has been previously observed in prostate cancer, a frameshift mutation at P205, and a small in-frame deletion at position 265. Alterations in the PI3 kinase pathway included PIK3CA H1047R, a known hotspot mutation, and PIK3R1 I20L, which has not been previously reported. NKX3–1 Y177C has also been observed previously in prostate cancer. Other alterations that have not been reported in prostate cancer to our knowledge based on COSMIC, cBioPortal, or Pubmed include nonsense mutations in KDM6A (C293*), EGFR (E759*), CDKN1B (Q147*) and NCOR1 (Q2170*); missense mutations in NCOA2 (R974P), ATM (F1767S and A2893V), APC (S2497L), and SCN11A (P882T); and deletions in PIK3CA (WGIHLMPP11del) and CDKN1B (GLA143del). Of these, CDKN1B Q147*, ATM A2893V, and APC S2497L have been observed in tumors of other cancer types. These alterations may represent novel events in prostate cancer and may warrant further investigation.
Table 3.
Somatic single nucleotide variants and small indels in primary prostate tumors
| Patient ID | Somatic SNVs/Indels |
|---|---|
| PK01 | PIK3R1 p.I20L (39%) |
| PK02 | None detected |
| PK03 | ZFHX3 p.3527_3528insG (14%) |
| PK04 | None detected |
| PK05 | CDKN1B p.Q147* (23%) EGFR p.E759* (19%) |
| PK06 | NCOA2 p.R974P (30%) |
| PK07 | CTNNB1 p.K335I (35%) |
| PK08 | TP53 p.R273G (20%) |
| PK09 | NKX3–1 p.Y177C (35%) PIK3CA p.WGIHLMPP11del (9%) |
| PK010 | None detected |
| PK011 | None detected |
| PK012 | FOXA1 p.P205fs (10%) |
| PK013 | None detected |
| PK014 | None detected |
| PK015 | CDKN1B p.GLA143del (10%) NCOR1 p.Q2170* (27%) SPOP p.F102L (27%) |
| PK016 | None detected |
| PK018 | PIK3CA p.H1047R (26%) SCN11A p.P882T (9%) |
| MGH1 | None detected |
| MGH2 | None detected |
| MGH3 | None detected |
| MGH4 | FOXA1 p.RFK265del (8%) KDM6A p.C293* (8%) |
| MGH5 | ATM p.A2893V (6%) |
| MGH6 | FOXA1 p.C227R (23%) |
| MGH7 | SPOP p.F104C (16%) |
| MGH9 | KRAS p.Q61R (18%) |
| MGH10 | None detected |
| MGH11 | None detected |
| MGH13 | None detected |
| MGH15 | APC p.S2497L (54%) ATM p.F1767S (19%) |
Somatic alterations in primary prostate cancer detected by targeted panel sequencing. Allelic fraction is an estimate of the fraction of the variant allele. None detected indicates no variants in the panel genes were identified, at the given mean target coverage, within the technical limits of the method.
ETS family transcription factor fusions:
The prostate cancer-specific targeted panel is designed to detect common fusion events with ETS family transcription factors. In our set of 29 evaluable primary prostatectomy specimens, ten individual samples (34%) had evidence of an ETS factor fusion event (Table 4). The structure of these fusions is in accordance with fusions reported in the literature for primary prostate tumors 16. Eight patients had a TMPRSS2-ERG fusion, one had a TMPRSS2-ETV4 fusion, and one had a SLC45A3-ETV1 fusion. Of these, four patients also had other SNVs/indels (Table 3) while for six patients the fusion was the sole detectable alteration among the genes in the panel. Taken together, we detected at least one somatic event—either an SNV/indel or an ETS factor fusion—in 22/29 (76%) of the primary prostate cancer samples.
Table 4.
Transcription factor fusion events in primary prostatectomy specimens
| Patient ID | Gene 1 | Gene 2 | # reads in tumor |
|---|---|---|---|
| PK04 | ETV1 | SLC45A3 | 4 |
| PK06 | ERG | TMPRSS2 | 3 |
| PK07 | ERG | TMPRSS2 | 8 |
| PK08 | ERG | TMPRSS2 | 2 |
| PK09 | ERG | TMPRSS2 | 7 |
| PK011 | ETV4 | TMPRSS2 | 2 |
| PK013 | ERG | TMPRSS2 | 2 |
| MGH1 | ERG | TMPRSS2 | 2 |
| MGH10 | ERG | TMPRSS2 | 2 |
| MGH13 | ERG | TMPRSS2 | 16 |
Samples harboring ETS family gene fusions are listed. The two genes involved in the fusion are listed as Gene 1 (5’) and Gene 2 (3’). The number of junctional reads across the fusion junction is listed. (Note: As expected, the number of junctional reads is small considering these reads have to cross the fusion junction which is likely a rare event; additional supporting reads were available for each fusion.)
We also performed targeted panel sequencing on DNA from two tumors which had previously undergone whole-genome sequencing and were known to contain ETS factor fusions 1. The targeted panel detected a fusion in both tumors (Supplementary Table 2).
Copy number changes in prostate cancer genes:
We predicted the presence of amplifications and deep deletions in commonly altered genes within the prostate cancer-specific targeted panel using a combination of tools (see Methods). In our primary tumor sample set, some (though not all) of the primary prostate tumors showed aberrant copy number patterns (Figure 2). Several deletion events previously reported in prostate cancer were observed, including predicted copy loss at CHD1, PTEN, RB1, and BRCA2. Amplification of MYC was observed in one sample. In contrast, most of the normal tissues sequenced had minimal copy number alterations (Figure 2). The data for established prostate cancer cell lines are shown for comparison and discussed in the next section. Of note, compared to WES, SNP arrays, or other methods, a targeted panel cannot detect the full complement of copy number changes (especially low-level copy gains and losses) and can only include the targeted genes as opposed to genome-wide copy number alterations.
Figure 2.
Heatmap of predicted gene-level copy number amplifications (red) and deletions (blue) with log2(CN) scale in normal tissues (left), tumor tissues (center), and cell lines (right). The computational approach for estimating copy number provides only estimated/predicted strong copy number variations, and only include genes in the targeted panel. Normal and tumor tissues from primary prostatectomies are shown in the left and center panels; the right panel shows commonly used prostate cancer cell lines.
Prostate cancer-specific targeted panel identifies mutations in blood biopsies from metastatic prostate cancer and in prostate cancer cell lines
To validate the prostate cancer-specific panel on independent cohorts of other prostate cancer sample types, we applied the panel to “blood biopsies” (circulating tumor cells and cell-free DNA) from patients with metastatic prostate cancer and to commonly used prostate cancer cell lines.
Prostate cancer circulating tumor cells:
WES can be performed on circulating tumor cells (CTCs) obtained from metastatic prostate cancer patients 31. We used the prostate cancer targeted panel on the same DNA from CTCs that had previously undergone WES (Supplementary Table 3). Nearly all alterations in the selected gene set in the targeted NGS panel were also detected by WES and vice-versa, except for a few mutations that lacked sequencing coverage by one method or the other, showing high concordance between targeted NGS and WES of DNA from CTCs in mCRPC.
Cell-free DNA:
WES of cfDNA from blood of metastatic prostate cancer patients has been reported 32. We used the prostate cancer targeted NGS panel on two samples of cfDNA that had undergone WES using this method (Supplementary Table 4). The prostate cancer targeted NGS panel detected 11 out of 17 SNVs identified by WES in the cfDNA; the non-concordant SNVs were either covered and not detected (2/6) or not covered (4/6) in the targeted panel. Of note, the targeted NGS panel identified a TMPRS22-ERG fusion gene in one patient that was not detected by WES.
To further test the ability of the prostate cancer targeted NGS panel to detect structural rearrangements in cfDNA, we sequenced an additional set of six cfDNA samples from patients with metastatic CRPC using a modified version of the panel that targets common rearrangements and additional baits covering the AR locus. Probes from this bait set were combined with a set of overlapping, tiled baits corresponding to non-repetitive regions of the androgen receptor (AR) locus as recently described in the literature 41 for analysis of circulating cfDNA. Plasma samples were subjected to targeted next-generation sequencing using this combined bait set, and the results were analyzed using BreaKmer 40. Events determined through the presence of ≥2 split reads were detected in five of six samples, including one with tumor fraction of less than 10% as estimated through ichorCNA 32. A total of 23 events were detected – 13 indels and 10 rearrangements – including events in TMPRSS2-ERG, ETV1, and AR (Supplementary Table 5). This analysis demonstrates that a probe set comprised of baits tiled along loci frequently altered in prostate cancer can detect relevant translocation events from cfDNA that would otherwise not be detected through existing commercial platforms.
Prostate cancer cell lines:
We sequenced commonly used prostate cancer cell lines using the prostate cancer-specific targeted panel and compared the results to sequencing data from a 1651-gene targeted hybrid-capture panel in the Cancer Cell Line Encyclopedia project 17. We observed a high rate of concordance in events detected by both our custom prostate cancer-specific panel and the CCLE general cancer panel (Supplementary Table 6).
Collectively, these results indicate that the prostate cancer-specific targeted panel is able to capture the majority of driver events occurring in prostate cancer, in both clinical samples and cell lines, including gene fusions that are not typically detected by standard WES.
Generation of patient-derived cell lines from primary prostate tissue
We used the conditional reprogramming method 23–26 to culture cells from fresh tissue samples of the prostate tumors and matched normal prostate tissues. We initiated cell lines from tumor and normal primary prostate specimens from 26 patients and observed robust cell growth for multiple passages (Supplementary Table 7). Of the samples for which serial passage was attempted, 20/20 normal tissues and 21/21 prostate cancer tissues generated cultures that could be serially passaged at least two times. Cell pellets were collected for DNA isolation and sequencing.
Targeted sequencing of patient-derived cell lines from primary prostatectomy samples
We used the prostate cancer-specific targeted panel to credential normal-tissue and tumor-tissue derived cell lines based on genomic features identified in the primary tumor using the targeted panel (Table 3, 4). From the 29 patients with targeted sequencing of the tumor tissue specimen, we sequenced 25 cell lines derived from primary prostate tumor tissue and 23 cell lines derived from matched normal tissue (of which 24 and 19 lines, respectively, gave rise to evaluable sequence data). In the cell lines derived from adjacent normal tissue, we did not observe any of the SNVs or fusion events that were present in the primary tumors, suggesting that on a genomic basis, these normal cell lines represent normal prostate cells. Although it is possible that adjacent normal prostate tissue might harbor precursor/premalignant cells related to the primary tumor, and thus these “normal” cell lines derived from prostate tumor tissue might harbor tumor-specific mutations, prostate cancer-associated mutations were not detected in any of the 21 sequenced normal cell lines, even at a low frequency, at least by the limits of detection of the targeted panel at the given sequencing coverage. Hence, the panel confirmed the expected absence of cancer-specific driver events in patient-derived cell lines from normal prostate.
Interestingly, for all 24 cell lines derived from tumor tissue that we sequenced, none of the SNVs or fusions observed in a patient’s primary tumor sample were observed (Figure 3). In essence, prostate cancer-associated mutations were not detected in cell lines derived from prostate cancer tissues. This suggests that the tumor-derived cell lines were probably composed mostly or completely of normal cells within the same tumor tissue specimen. It is possible that these normal cells had preferentially grown out in the conditional reprogramming conditions; growth of normal prostate cancer cells in conditional reprogramming medium has been previously reported 26,30. Our experience thus highlights the importance of genomic credentialing of putative cancer-derived cell lines, and the utility of prostate cancer targeted sequencing to achieve this goal.
Figure 3.
Summary of mutations and gene fusions in prostate-cancer associated genes (columns) in the samples sequenced in this study (rows) including patient tumor samples, patient-derived cell lines, cell-free DNA, CTCs, and standard prostate cancer cell lines.
Discussion
To facilitate sequencing of relevant genomic alterations in prostate cancer clinical samples and patient-derived cell lines, we generated a prostate cancer-specific targeted NGS panel detecting somatic alterations in 62 prostate cancer-associated genes, including ETS family gene fusions. Targeted sequencing with the panel successfully detected single-nucleotide variants, insertions/deletions, and fusions in a cohort of primary prostate cancers, circulating tumor cells, and cell-free DNA from patients with metastatic prostate cancer.
In this study, we aimed to generate patient-derived cell lines from primary prostate cancers and to validate these cell lines using the prostate cancer-specific targeted panel. The targeted NGS panel facilitated genomic analysis to credential patient-derived cell lines by confirming whether the cell line DNA was consistent with the appropriate normal or malignant genome from its parent tissue. It was notable that none of 24 cell lines derived from primary prostate tumors that we sequenced harbored prostate cancer-specific mutations, using the same targeted NGS technology that reliably detected such mutations in the primary tumor tissue from which the cell line was derived. It is possible that the specific tissue piece used to generate the cell line was deficient in tumor cells, but a matched tissue piece was evaluated by a genitourinary pathologist and contained at least 25% tumor where this information was available. We suspect that, instead, a net outgrowth of normal prostate epithelial cells occurred during several passages in culture, as has also been previously observed in organoid systems of metastatic prostate cancers 20, perhaps due a growth deficiency of the primary prostate tumor cells and/or a growth advantage of normal prostate epithelial cells. Further studies will be needed to determine the mechanisms underlying the inability to maintain primary prostate cancer cells in culture conditions that support other prostate epithelial cells, and this will be an important barrier to overcome in developing new prostate cancer patient-derived models. These findings also highlight the critical importance of validating new patient-derived cell lines to confirm maintenance of the tumor phenotype in culture, including genomic alterations consistent with the parent tumor from which the lines are derived.
In addition to genomic characterization of patient-derived cell lines, prostate cancer-specific targeted NGS panels may be advantageous for several applications in prostate cancer clinical care. In clinical oncology practice, targeted NGS is feasible and effective for identifying tumor-associated alterations and clinically actionable mutations 42–44, typically using a panel of 50–400 genes commonly mutated across all cancer types. Several studies have reported targeted NGS of prostate cancers 45–50 using such pan-cancer gene sets. However, the genes in these panels vary among institutions and commercial platforms, and the tumor-agnostic panels may omit key genes frequently mutated in prostate cancer but rare in other cancers. We note that FOXA1 and CHD1 are not included in the current versions of several of the commercial genomic panels (e.g. Foundation One, Guardant, Caris, Tempus). Compared to tumor-agnostic panels, selectively targeting genes altered in prostate cancer may increase the detection of biologically and clinically relevant alterations in prostate cancer. Our study is one of several to report a prostate cancer-specific targeted NGS panel covering genes recurrently altered in prostate cancer. For instance, sequencing of tumors and cfDNA from patients with mCRPC using a 72-gene panel of genes relevant to mCRPC showed that targeted sequencing of cfDNA could identify variants associated with clinical outcomes 51–54.
As demonstrated in our patient cohort, targeted sequencing may be particularly valuable for sequencing blood biopsies (CTCs and cfDNA) of metastatic prostate cancer patients, which enable monitoring the diversity and evolution of metastatic tumors without invasive tumor biopsies. WES, WGS, and targeted sequencing are feasible from both CTCs and cfDNA from blood of prostate cancer patients 31,32,52–57. However, compared to WES/WGS, targeted sequencing may be more clinically tractable for plasma samples, especially those with small amounts of DNA and low tumor fraction. Mayrhofer et al applied custom panel targeted sequencing to cfDNA in mCRPC and identified multiple alterations in DNA repair genes, among other findings, noting that targeted panel sequencing allowed cost-efficient deep sequencing to detect rare variants 57. Noninvasive blood biopsies coupled with prostate cancer targeted sequencing could be an attractive approach to monitoring metastatic disease and selecting targeted therapies reflecting diverse metastatic lesions and resistance mechanisms, and is already showing promising results52,53,58,59.
Identifying fusion genes within a targeted NGS assay is a feature that is not achieved with some tumor-agnostic NGS panels nor with standard WES, although such fusions can still be challenging to detect even when specifically targeted due to the requirement to target rare fusion reads. While our study used fresh-frozen tissue samples, targeted NGS of FFPE samples should be feasible using this method, especially given the low DNA input requirements. Including more intronic regions of genes involved in recurrent structural alterations in the panel would likely enable detection of additional fusion genes and structural rearrangements 10.
Although our study did have lower overall sequence coverage than some targeted NGS studies (averaging 118X for all samples; 143X for the tumor/normal tissues that passed with >10X coverage), the prostate cancer-specific panel still detected key alterations in 76% of primary prostate tumors; this percentage may increase with greater sequencing depth and/or more genes associated with prostate cancer added to the panel 6. Depth and accuracy may also be increased by using duplex sequencing methods which have become more technically feasible. Overall, the targeted panel detected many of the common alterations in prostate cancer reported from WES studies, yet can be performed at a lower cost and/or greater depth compared to WES due to the smaller total genomic size (790 kb) of the panel.
Prostate cancer-specific targeted NGS panels can also identify targetable alterations with therapeutic implications in primary and metastatic prostate cancer, including BRCA2 mutations or deletions that may predict sensitivity to PARP inhibitors; CDK12 alterations and mismatch repair deficiency that may suggest sensitivity to DNA repair targeting therapies and immunotherapies; PI3K pathway alterations that may be associated with sensitivity to PI3K/AKT inhibitors; and alterations in AR that may affect sensitivity to anti-androgen therapies 11,14,60,61. Other future clinical applications of prostate cancer-specific targeted NGS panels could be envisioned, such as identifying cancer-associated mutations within a diagnostic algorithm for indeterminate prostate biopsies. Panels could also be modified to include estimates of tumor mutational burden and to identify microsatellite instability, both important biomarkers of response to immunotherapy. Given the flexibility of custom targeted NGS, prostate-cancer specific gene panels can be modified or expanded as further sequencing of large cohorts of prostate cancers reveal additional genes altered in the “long tail” of mutations, such as those described in metastatic prostate cancer 9.
From a research and discovery standpoint, our set of 29 primary prostate cancers harbored at least eight SNVs not previously reported, supporting that targeted panels can identify new somatic mutations in known prostate cancer-associated genes. Further functional analysis will be required to determine if these represent novel gain- or loss-of-function mutations that may have implications for prostate cancer biology.
In summary, a prostate cancer-specific targeted NGS panel can detect mutations, insertions/deletions, copy number changes, and gene fusions in prostate cancer-associated genes in cell lines, primary prostate tumors, and blood biopsies from metastatic prostate cancer patients. Targeted sequencing also revealed normal cellular outgrowth within patient-derived cell lines from primary prostate cancers, emphasizing the critical importance of genomic credentialing of patient-derived models. While the number of genes included in this panel is limited, application of a targeted NGS strategy specifically for prostate cancer can identify known and novel mutations in prostate cancer-associated genes and could be implemented clinically for diagnostic and/or therapeutic decision-making.
Supplementary Material
Acknowledgements
We are grateful to the patients who provided samples for this study. We appreciate technical support from the Broad Institute Genomics Platform, Broad Institute Cancer Cell Line Factory, and the DFCI prostate cancer team. We would like to acknowledge Dr. Scott Dehm for providing details regarding sequencing probes targeting the Androgen Receptor locus, and the Center for Cancer Genome Discovery for analysis of structural alterations from cell-free DNA. We thank the members of the Garraway and Boehm laboratories, the DFCI and MGH genitourinary oncology clinical groups, and the Broad Institute Cancer Program for helpful discussions.
Funding Statement
This work was supported by Stand Up to Cancer, Dana-Farber/Harvard Cancer Center SPORE in prostate cancer, the Gerstner Family Foundation, and the Dana-Farber Medical Oncology Department. Additional funding includes the Friends of Dana-Farber Cancer Institute to E.H.S.; Wong Family Foundation to A.D.C.; NIH K08CA191026 to J.G.L.; PCF Challenge Grant 16CHASO3 to E.M.V.A. and M.E.T.; NIH K08CA218530 to D.Y.T.; NIH grants RO1CA131945, R01CA187918, P50CA211024, Department of Defense grants DoD PC160357 and DoD PC180582, and the Prostate Cancer Foundation to M.L.; P01CA120964 to C.L.W.; Fat Boys and Slim Sisters Pan Mass Challenge Team support to M.E.T.; anonymous donor funding to J.S.B.; and Prostate Cancer Foundation Young Investigator Award, American Society of Clinical Oncology Award, and Department of Defense Prostate Cancer Research Program grant W81XWH-14-1-0514 to F.W.H.
Conflict of Interest Disclosure Statement
The authors declare that they have no competing interests relevant to this study. For completeness, the following conflicts of interest disclosure is provided: A.D.C. has received research funding from Bayer for cell-free DNA analysis in patients with prostate cancer and has participated in advisory boards for Clovis, Dendreon, Bayer, Eli Lilly, AstraZeneca, Astellas, and Blue Earth. V.A.A. is a scientific advisor for ACGT GmbH. J.G.L. is a consultant for the company 4SC. G.V.K is an employee and shareholder of KSQ Therapeutics, and is a shareholder of Illumina, Guardant Health and CRISPR Therapeutics. E.M.V.A. has an advisory/consulting role with Tango Therapeutics, Genome Medical, Invitae, Enara Bio, Janssen, Manifold Bio, and Monte Rosa; has received research support from Novartis and Bristol-Meyers-Squibb and travel reimbursement from Roche/Genentech; is an equity holder in Tango Therapeutics, Genome Medical, Syapse, Enara Bio, Manifold Bio, Monte Rosa, and Microsoft. M.E.T. has served on advisory boards for Clovis, Janssen, Progenics, Astellas, Bayer, Astra-Zeneca, Celgene, and Constellation, and has institutional research funding from Janssen, Bayer and Pfizer. L.A.G. is an employee of Genentech/Roche and previously an employee of Eli Lilly. V.A.A., G.H., and S.S.F. have filed a patent on methods for cfDNA analysis. E.M.V.A. has institutional patents filed on ERCC2 mutations and chemotherapy response, chromatin mutations and immunotherapy response, and methods for clinical interpretation.
Ethics Approval Statement and Patient Consent Statement
We received Institutional Review Board (IRB) and institutional approval from Dana-Farber Cancer Institute (DFCI) and Massachusetts General Hospital (MGH) to collect samples of primary prostate cancers and adjacent normal prostate tissue from prostatectomy surgeries.
List of abbreviations
- AR
androgen receptor
- CCLE
Cancer Cell Line Encyclopedia
- cfDNA
cell-free DNA
- CTC
circulating tumor cell
- FFPE
formalin-fixed paraffin-embedded
- IRB
Institutional Review Board
- mCRPC
metastatic castration-resistant prostate cancer
- NGS
next generation sequencing
- PARP
poly(ADP-ribose)
- SCNA
somatic copy number alteration
- SNVs
single nucleotide variants
- WES
whole exome sequencing
- WGS
whole genome sequencing
Data Availability Statement
The datasets supporting the conclusions of this article are included in the paper and sequencing data will be made available in the NCBI Sequence Read Archive repository.
References
- 1.Baca SC, Prandi D, Lawrence MS, et al. Punctuated evolution of prostate cancer genomes. Cell 2013;153:666–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Barbieri CE, Baca SC, Lawrence MS, et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat Genet 2012;44:685–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Berger MF, Lawrence MS, Demichelis F, et al. The genomic complexity of primary human prostate cancer. Nature 2011;470:214–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cancer Genome Atlas Research N. The Molecular Taxonomy of Primary Prostate Cancer. Cell 2015;163:1011–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Huang FW, Mosquera JM, Garofalo A, et al. Exome Sequencing of African-American Prostate Cancer Reveals Loss-of-Function ERF Mutations. Cancer Discov 2017;7:973–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Armenia J, Wankowicz SAM, Liu D, et al. The long tail of oncogenic drivers in prostate cancer. Nat Genet 2018;50:645–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Grasso CS, Wu YM, Robinson DR, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 2012;487:239–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Lawrence MS, Stojanov P, Mermel CH, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature 2014;505:495–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Robinson D, Van Allen EM, Wu YM, et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015;161:1215–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Quigley DA, Dang HX, Zhao SG, et al. Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer. Cell 2018;174:758–69 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Viswanathan SR, Ha G, Hoff AM, et al. Structural Alterations Driving Castration-Resistant Prostate Cancer Revealed by Linked-Read Genome Sequencing. Cell 2018;174:433–47 e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gundem G, Van Loo P, Kremeyer B, et al. The evolutionary history of lethal metastatic prostate cancer. Nature 2015;520:353–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Wedge DC, Gundem G, Mitchell T, et al. Sequencing of prostate cancers identifies new cancer genes, routes of progression and drug targets. Nat Genet 2018;50:682–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Mateo J, Carreira S, Sandhu S, et al. DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. N Engl J Med 2015;373:1697–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Pritchard CC, Mateo J, Walsh MF, et al. Inherited DNA-Repair Gene Mutations in Men with Metastatic Prostate Cancer. N Engl J Med 2016;375:443–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Weier C, Haffner MC, Mosbruger T, et al. Nucleotide resolution analysis of TMPRSS2 and ERG rearrangements in prostate cancer. J Pathol 2013;230:174–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Barretina J, Caponigro G, Stransky N, et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature 2012;483:603–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Gao D, Chen Y. Organoid development in cancer genome discovery. Curr Opin Genet Dev 2015;30:42–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chua CW, Shibata M, Lei M, et al. Single luminal epithelial progenitors can generate prostate organoids in culture. Nat Cell Biol 2014;16:951–61, 1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gao D, Vela I, Sboner A, et al. Organoid cultures derived from patients with advanced prostate cancer. Cell 2014;159:176–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Karthaus WR, Iaquinta PJ, Drost J, et al. Identification of multipotent luminal progenitor cells in human prostate organoid cultures. Cell 2014;159:163–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Puca L, Bareja R, Prandi D, et al. Patient derived organoids to model rare prostate cancer phenotypes. Nat Commun 2018;9:2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Liu X, Ory V, Chapman S, et al. ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. Am J Pathol 2012;180:599–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Palechor-Ceron N, Suprynowicz FA, Upadhyay G, et al. Radiation induces diffusible feeder cell factor(s) that cooperate with ROCK inhibitor to conditionally reprogram and immortalize epithelial cells. Am J Pathol 2013;183:1862–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Suprynowicz FA, Upadhyay G, Krawczyk E, et al. Conditionally reprogrammed cells represent a stem-like state of adult epithelial cells. Proc Natl Acad Sci U S A 2012;109:20035–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Liu X, Krawczyk E, Suprynowicz FA, et al. Conditional reprogramming and long-term expansion of normal and tumor cells from human biospecimens. Nat Protoc 2017;12:439–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Wang S, Gao D, Chen Y. The potential of organoids in urological cancer research. Nat Rev Urol 2017;14:401–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Yuan H, Myers S, Wang J, et al. Use of reprogrammed cells to identify therapy for respiratory papillomatosis. The New England journal of medicine 2012;367:1220–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Suprynowicz FA, Upadhyay G, Krawczyk E, et al. Conditionally reprogrammed cells represent a stem-like state of adult epithelial cells. Proceedings of the National Academy of Sciences of the United States of America 2012;109:20035–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Liu X, Ory V, Chapman S, et al. ROCK inhibitor and feeder cells induce the conditional reprogramming of epithelial cells. The American journal of pathology 2012;180:599–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lohr JG, Adalsteinsson VA, Cibulskis K, et al. Whole-exome sequencing of circulating tumor cells provides a window into metastatic prostate cancer. Nat Biotechnol 2014;32:479–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Adalsteinsson VA, Ha G, Freeman SS, et al. Scalable whole-exome sequencing of cell-free DNA reveals high concordance with metastatic tumors. Nat Commun 2017;8:1324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Barbieri CE, Baca SC, Lawrence MS, et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nature genetics 2012;44:685–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Cibulskis K, Lawrence MS, Carter SL, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol 2013;31:213–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Saunders CT, Wong WS, Swamy S, Becq J, Murray LJ, Cheetham RK. Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs. Bioinformatics 2012;28:1811–7. [DOI] [PubMed] [Google Scholar]
- 36.Drier Y, Lawrence MS, Carter SL, et al. Somatic rearrangements across cancer reveal classes of samples with distinct patterns of DNA breakage and rearrangement-induced hypermutability. Genome Res 2013;23:228–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 2009;25:1754–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.McKenna A, Hanna M, Banks E, et al. The Genome Analysis Toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res 2010;20:1297–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.DePristo MA, Banks E, Poplin R, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat Genet 2011;43:491–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Abo RP, Ducar M, Garcia EP, et al. BreaKmer: detection of structural variation in targeted massively parallel sequencing data using kmers. Nucleic Acids Res 2015;43:e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Li Y, Hwang TH, Oseth LA, et al. AR intragenic deletions linked to androgen receptor splice variant expression and activity in models of prostate cancer progression. Oncogene 2012;31:4759–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.MacConaill LE, Garcia E, Shivdasani P, et al. Prospective enterprise-level molecular genotyping of a cohort of cancer patients. J Mol Diagn 2014;16:660–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wagle N, Berger MF, Davis MJ, et al. High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing. Cancer Discov 2012;2:82–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zehir A, Benayed R, Shah RH, et al. Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients. Nat Med 2017;23:703–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Robbins CM, Tembe WA, Baker A, et al. Copy number and targeted mutational analysis reveals novel somatic events in metastatic prostate tumors. Genome Res 2011;21:47–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Beltran H, Yelensky R, Frampton GM, et al. Targeted next-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. Eur Urol 2013;63:920–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Manson-Bahr D, Ball R, Gundem G, et al. Mutation detection in formalin-fixed prostate cancer biopsies taken at the time of diagnosis using next-generation DNA sequencing. J Clin Pathol 2015;68:212–7. [DOI] [PubMed] [Google Scholar]
- 48.Hovelson DH, McDaniel AS, Cani AK, et al. Development and validation of a scalable next-generation sequencing system for assessing relevant somatic variants in solid tumors. Neoplasia 2015;17:385–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cheng HH, Klemfuss N, Montgomery B, et al. A Pilot Study of Clinical Targeted Next Generation Sequencing for Prostate Cancer: Consequences for Treatment and Genetic Counseling. Prostate 2016;76:1303–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lo Iacono M, Buttigliero C, Monica V, et al. Retrospective study testing next generation sequencing of selected cancer-associated genes in resected prostate cancer. Oncotarget 2016;7:14394–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Annala M, Struss WJ, Warner EW, et al. Treatment Outcomes and Tumor Loss of Heterozygosity in Germline DNA Repair-deficient Prostate Cancer. Eur Urol 2017;72:34–42. [DOI] [PubMed] [Google Scholar]
- 52.Wyatt AW, Annala M, Aggarwal R, et al. Concordance of Circulating Tumor DNA and Matched Metastatic Tissue Biopsy in Prostate Cancer. J Natl Cancer Inst 2017;109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Annala M, Vandekerkhove G, Khalaf D, et al. Circulating Tumor DNA Genomics Correlate with Resistance to Abiraterone and Enzalutamide in Prostate Cancer. Cancer Discov 2018;8:444–57. [DOI] [PubMed] [Google Scholar]
- 54.Taavitsainen S, Annala M, Ledet E, et al. Evaluation of Commercial Circulating Tumor DNA Test in Metastatic Prostate Cancer. JCO Precis Oncol 2019;3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Choudhury AD, Werner L, Francini E, et al. Tumor fraction in cell-free DNA as a biomarker in prostate cancer. JCI Insight 2018;3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ritch E, Fu SYF, Herberts C, et al. Identification of Hypermutation and Defective Mismatch Repair in ctDNA from Metastatic Prostate Cancer. Clin Cancer Res 2020;26:1114–25. [DOI] [PubMed] [Google Scholar]
- 57.Mayrhofer M, De Laere B, Whitington T, et al. Cell-free DNA profiling of metastatic prostate cancer reveals microsatellite instability, structural rearrangements and clonal hematopoiesis. Genome Med 2018;10:85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wyatt AW, Azad AA, Volik SV, et al. Genomic Alterations in Cell-Free DNA and Enzalutamide Resistance in Castration-Resistant Prostate Cancer. JAMA Oncol 2016;2:1598–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Vandekerkhove G, Struss WJ, Annala M, et al. Circulating Tumor DNA Abundance and Potential Utility in De Novo Metastatic Prostate Cancer. Eur Urol 2019;75:667–75. [DOI] [PubMed] [Google Scholar]
- 60.Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med 2015;372:2509–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wu YM, Cieslik M, Lonigro RJ, et al. Inactivation of CDK12 Delineates a Distinct Immunogenic Class of Advanced Prostate Cancer. Cell 2018;173:1770–82 e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The datasets supporting the conclusions of this article are included in the paper and sequencing data will be made available in the NCBI Sequence Read Archive repository.



