throbber
Cancer Cell
`
`Article
`
`Integrative Genomic Profiling
`of Human Prostate Cancer
`
`Barry S. Taylor,1,8 Nikolaus Schultz,1,8 Haley Hieronymus,2,8 Anuradha Gopalan,3 Yonghong Xiao,3 Brett S. Carver,4
`Vivek K. Arora,2 Poorvi Kaushik,1 Ethan Cerami,1 Boris Reva,1 Yevgeniy Antipin,1 Nicholas Mitsiades,5 Thomas Landers,2
`Igor Dolgalev,2 John E. Major,6 Manda Wilson,6 Nicholas D. Socci,6 Alex E. Lash,6 Adriana Heguy,2 James A. Eastham,4
`Howard I. Scher,5 Victor E. Reuter,3 Peter T. Scardino,4 Chris Sander,1 Charles L. Sawyers,2,7,* and William L. Gerald2,3,9
`1Program in Computational Biology
`2Program in Human Oncology and Pathogenesis (HOPP)
`3Department of Pathology
`4Department of Urology
`5Department of Medicine
`6Bioinformatics Core
`Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
`7Howard Hughes Medical Institute, Chevy Chase, MD 20815-6789, USA
`8These authors contributed equally to this work
`9Deceased
`*Correspondence: sawyersc@mskcc.org
`DOI 10.1016/j.ccr.2010.05.026
`
`SUMMARY
`
`Annotation of prostate cancer genomes provides a foundation for discoveries that can impact disease under-
`standing and treatment. Concordant assessment of DNA copy number, mRNA expression, and focused exon
`resequencing in 218 prostate cancer tumors identified the nuclear receptor coactivator NCOA2 as an onco-
`gene in 11% of tumors. Additionally, the androgen-driven TMPRSS2-ERG fusion was associated with
`a previously unrecognized, prostate-specific deletion at chromosome 3p14 that implicates FOXP1, RYBP,
`and SHQ1 as potential cooperative tumor suppressors. DNA copy-number data from primary tumors
`revealed that copy-number alterations robustly define clusters of low- and high-risk disease beyond that
`achieved by Gleason score. The genomic and clinical outcome data from these patients are now made avail-
`able as a public resource.
`
`INTRODUCTION
`
`Prostate cancer is the most common malignancy in males with
`190,000 new cases diagnosed per year in the United States
`and 27,000 deaths. Prostate tumors show tremendous biolog-
`ical heterogeneity, with some patients dying of metastatic
`disease within 2–3 years of diagnosis whereas others can live
`for 10–20 years with organ-confined disease, likely a reflection
`of underlying genomic diversity. Large-scale cancer genome
`characterization projects studying glioblastoma, lung, colon,
`pancreas, and breast cancers have provided critical new insights
`
`into the molecular classification of cancers and have the poten-
`tial to identify new therapeutic targets (Cancer Genome Atlas
`Research Network, 2008; Ding et al., 2008; Jones et al., 2008;
`Parsons et al., 2008; Sjoblom et al., 2006; Weir et al., 2007;
`Wood et al., 2007). Prostate cancer presents special challenges
`for such large-scale multicenter genomics projects because of
`the relatively small tumor size and admixture with stroma that
`requires careful pathologist-guided dissection.
`A number of groups have reported analyses of transcriptomes
`and copy-number alterations (CNAs)
`in prostate cancer,
`but rarely from the same samples and typically from modest
`
`Significance
`
`Current knowledge of prostate cancer genomes is largely based on small patient cohorts using single modality platforms.
`We present an integrated oncogenomic analysis of 218 primary and metastatic prostate cancers as well as 12 cell lines and
`xenografts. Mutations in known, commonly mutated oncogenes and tumor suppressor genes such as PIK3CA, KRAS,
`BRAF, and TP53 are present but rare. However, integrative analysis of mutations, copy-number alterations, and expression
`changes revealed changes in the PI3K, RAS/RAF, and androgen receptor (AR) pathways in nearly all metastatic samples and
`a high frequency of primary samples. These data clarify the role of several known cancer pathways in prostate cancer, impli-
`cate several new ones, and provide a blueprint for clinical development of pathway inhibitors.
`
`Cancer Cell 18, 11–22, July 13, 2010 ª2010 Elsevier Inc. 11
`
`AVENTIS EXHIBIT 2124
`Mylan v. Aventis, IPR2016-00712
`
`
`

`
`Genomic Characterization of Human Prostate Cancers
`
`Cancer Cell
`
`Table 1. Summary of Clinical Characteristics for the Study
`Cohort
`
`Characteristic
`
`Primary Tumorsa
`
`Metastasesb
`
`Age
`
`Median
`
`Mean
`
`Standard deviation
`
`58.3
`
`58.3
`
`7
`
`60
`
`60
`
`8.6
`
`Min-max
`
`37.3-83
`
`41-82
`
`PSA at Diagnosis (ng/ml)
`
`Median
`
`<4
`
`4–10
`
`>10
`
`6 (IQR 4.4, 9)
`
`17 (IQR 8.6, 46.6)
`
`31 (17.2%)
`
`105 (58.3%)
`
`44 (24.5%)
`
`4 (12.5%)
`
`6 (18.75%)
`
`22 (68.75%)
`
`Initial Biopsy Gleason Score
`
`5
`
`6
`
`7
`
`8
`
`9
`
`Initial Clinical Stage
`
`cT1c
`
`cT2
`
`cT3
`
`cT4
`
`Not available
`
`Ethnicity
`
`Black
`
`Asian
`
`White Hispanic
`
`2 (1%)
`
`101 (56%)
`
`61 (34%)
`
`11 (6%)
`
`6 (3%)
`
`95 (52.4%)
`
`76 (42%)
`
`9 (5%)
`
`–
`
`–
`
`29 (16.1%)
`
`4 (2.2%)
`
`0 (0%)
`
`–
`
`2 (6%)
`
`16 (46%)
`
`8 (23%)
`
`9 (25%)
`
`8 (22%)
`
`12 (33%)
`
`3 (8%)
`
`1 (3%)
`
`9 (25%)
`
`2 (5.6%)
`
`0 (0%)
`
`2 (5.6%)
`
`White non-Hispanic
`
`142 (78.5%)
`
`32 (88.9%)
`
`Unknown
`
`6 (3.3%)
`
`0 (0%)
`
`The cohort is composed of
`a primary tumors (n = 181; one patient with metastasis and primary
`analyzed) and
`b metastases (n = 37; one patient with two metastases analyzed), cell
`lines (n = 7; CWR22RV1, DU145, PC3, VCaP, LNCaP, LNCaP104R,
`LNCaP104S), and xenografts (n = 5; LAPC9, LNM971, LuCaP35,
`LAPC3, and LAPC4 [for both, two samples at different passages]). See
`also Figure S1 and Table S1.
`
`possibility that our stringent tumor selection criteria might bias
`our data set toward larger, more aggressive prostate cancers,
`we compared the clinical outcome of the 181 primary tumors
`(Table S1) in this data set with 3437 consecutive men with pros-
`tate cancer treated by prostatectomy at MSKCC from 2000 to
`2006. The time to biochemical relapse, defined as an increase
`in serum prostate-specific antigen, was somewhat shorter in
`this study cohort (Table 1; Figure S1A). While this indicates
`that genomic findings from these samples may be biased toward
`larger, more aggressive prostate cancers (selected to ensure
`sufficient nucleic acid yield), this cohort nevertheless includes
`patients with favorable long-term clinical outcome (24% of
`patients have >5 years of recurrence-free survival).
`Global analysis of CNA data from 194 tumors and 12 cell
`lines/xenografts revealed broad diversity in alteration levels.
`
`numbers of tumors (50–100 samples) or with lower resolution
`platforms (Kim et al., 2007; Lapointe et al., 2004, 2007; Lieber-
`farb et al., 2003; Perner et al., 2006; Singh et al., 2002). Con-
`sistent and common findings from these reports include the
`TMPRSS2-ERG fusion in 50%, 8p loss in 30%–50%, and
`8q gain in 20%–40% of cases. The data implicating ERG as
`a prostate cancer gene are clear (Tomlins et al., 2005), but there
`has been less progress in defining specific genes targeted by
`various common amplifications and deletions, in part due to
`limited availability of complementary transcriptome and exon
`resequencing data on sufficient patients to narrow the focus to
`a small list of candidate genes. Numerous transcriptome studies
`have defined general prostate cancer signatures, but, unlike
`breast cancer (Paik et al., 2004; van de Vijver et al., 2002), these
`analyses have not identified robust subtypes of prostate cancer
`with different prognoses (Febbo and Sellers, 2003; Lapointe
`et al., 2004; Singh et al., 2002).
`Here, we adopted a comprehensive approach to define tran-
`scriptomes and CNAs in 218 prostate tumors (181 primaries,
`37 metastases) and 12 prostate cancer cell lines and xenografts,
`as well as complete exon resequencing and/or focused mutation
`detection for 157 high interest genes in 80 tumors and 11 cell/
`xenograft lines (Table 1). After generating a map of CNAs across
`the data set, we used matching mRNA and microRNA transcrip-
`tome and exon resequencing data to define the frequency of
`alterations in several common signal transduction pathways,
`explore various candidate genes within a few selected regions
`of copy-number gain and loss, and correlate genomic alter-
`ations to clinical outcome. These data serve as a valuable
`resource for the cancer genomics community, prostate cancer
`scientists and clinicians and is readily and freely available
`through a user-friendly web-based portal
`(http://cbio.mskcc.
`org/prostate-portal/).
`
`RESULTS
`
`Global Copy-Number and Transcriptome Profiles Define
`Core Pathway Alterations
`We applied rigorous criteria for selecting tumors for genomic
`analysis that have become the standard in large genomic studies
`(Cancer Genome Atlas Research Network, 2008) but adapted to
`address unique challenges posed by prostate cancers. All 218
`samples had at least 70% tumor content (Table 1; Figure S1
`and Table S1 available online). Transcriptome (mRNA and micro-
`RNA) and CNA profiling were conducted without amplification,
`with the exception of exon resequencing, which required
`whole-genome amplification. Because we did not
`impose
`a stringent tumor size requirement, the small size of some tumors
`precluded concurrent analysis across all four platforms (Table 2;
`Table S2).
`Analysis of known prostate cancer alterations in our data set
`indicates successful tumor selection criteria (Figure S1). For
`example, the frequency of ERG alteration was 52% (see Exper-
`imental Procedures), consistent with other studies, and chromo-
`some 8p loss and 8q gain were easily detected (Figure 1A). Overt
`CNAs were observed in 89% of tumors, also indicative of high
`tumor content. Additional histologic and molecular analysis of
`those tumors without CNA confirmed high tumor content (e.g.,
`detection of TMPRSS2-ERG translocations). To address the
`
`12 Cancer Cell 18, 11–22, July 13, 2010 ª2010 Elsevier Inc.
`
`

`
`Cancer Cell
`
`Genomic Characterization of Human Prostate Cancers
`
`Table 2. Number of Primary and Metastatic Tumors Analyzed by
`Each Platform
`
`Data Type
`
`aCGH
`
`mRNA
`
`miRNA
`Sequencingb
`aCGH, mRNAc
`aCGH, mRNA, miRNAc
`
`aCGH, miRNA,
`sequencingb,c
`
`aCGH, mRNA, miRNA,
`sequencingb,c
`
`Primariesa Metastases
`
`Cell Lines/
`Xenografts
`
`157
`
`131 (29)
`
`99 (28)
`
`75
`
`109
`
`79
`
`72
`
`61
`
`37
`
`19
`
`14
`
`5
`
`19
`
`13
`
`2
`
`1
`
`13
`
`6
`
`0
`
`11
`
`5
`
`2
`
`0
`
`0
`
`All
`
`207
`
`156
`
`113
`
`98
`
`133
`
`94
`
`74
`
`62
`
`Summary of tumors and characterization platforms:
`a number of samples in parentheses refers to the count of matching
`normal prostate expression;
`b sequencing category includes only tumors with matched normals and
`cell/xenografts lines; and
`c mRNA and miRNA refers to expression profiling data. See also
`Tables S2 and S5.
`
`Metastatic tumors and cell/xenograft lines harbored the greatest
`number of whole chromosome, chromosome arm, and focal
`amplifications and deletions, but primary tumors also displayed
`a wide range of alteration levels, from tumors appearing meta-
`static-like in profile to those with fundamentally diploid genomes
`(Figure S1B). Regions of recurrent CNA were identified using the
`statistical method RAE (Taylor et al., 2008), revealing 30 focal
`amplifications and 36 focal deletions as well as recurrent gains
`and losses of seven chromosome arms (Figure 1A; Tables S3
`and S4). The most frequent alteration in the prostate oncoge-
`nome was loss of chromosome 8p, a common abnormality in
`many epithelial tumors that harbors NKX3.1 (He et al., 1997).
`Interestingly, NKX3.1 mRNA expression did not correlate with
`copy-number loss, suggesting the possibility of alternative tumor
`suppressors in this region. Consistent with prior studies, we also
`found peaks of deletion targeting PTEN on 10q23.31, RB1 on
`13q14.2, TP53 on 17p31.1, and the interstitial 21q22.2-3 deletion
`spanning ERG and TMPRSS2. Other broader deletions included
`12p13.31-p12.3, which spans ETV6 and DUSP16 in addition to
`CDKN1B, the previously reported target of this genomic deletion
`(Lapointe et al., 2007). The most common amplified loci included
`MYC on 8q24.21 and a previously unreported NCOA2 amplifica-
`tion on 8q13.3 (discussed further below). Focal amplifications of
`AR (Xq12) were also common but restricted to metastatic
`tumors. Other gains span discontinuous regions of 7q, including
`genes such as BRAF and EZH2, for which we were unable to
`localize individual target genes. We observed less frequent gains
`of 5p13.3-p13.1 spanning AMACR, RICTOR, and SKP2 as well
`as 47 other genes and two microRNAs.
`Eighty tumors were examined for somatic mutations in 138
`genes by exon sequencing (Figure 1A; Tables S5 and S6). These
`and an additional 76 tumors were also profiled for well-known
`oncogenic mutations in 22 genes by mass spectrometry using
`the iPLEX Sequenom assay (Table S5). In total, 84 confirmed
`somatic mutations were detected in 57 different genes
`
`(Table S6). Thirty-seven percent of the missense mutations we
`detected are predicted to affect protein function (Table S6)
`based on an algorithm that uses a combination of evolutionary
`information from protein-family sequence alignments and
`residue placement
`in known or homology-deduced three-
`dimensional protein and complex structures (B.R., Y.A., C.S.,
`unpublished data; http://www.mutationassessor.org/). Among
`all mutated genes, including those bearing previously known
`mutations, the most commonly mutated gene was the androgen
`receptor (AR), with four samples, all metastases. Mutations in 21
`other genes were detected in two or more samples, but no single
`gene other than AR had mutations in more than three samples.
`We also confirmed prior data suggesting that common, broadly
`mutated oncogenes such as PIK3CA, KRAS, and BRAF are
`not commonly mutated in prostate cancer (two tumors had
`H1047R and E545K PIK3CA mutations, two had G12V and
`Q61H/L KRAS mutations, and one tumor had a BRAF V600E
`mutation). Mutations in other more recently identified oncogenes
`such as IDH1 and IDH2 were similarly rare, with only one tumor
`bearing an IDH2 R172K mutation. Curiously, one tumor with
`a mutation in the mismatch repair gene MSH6 (V250A) had 11
`confirmed somatic mutations versus an average of two somatic
`mutations per
`tumor, suggestive of a mutator phenotype.
`Mutations in two other DNA repair genes, BLM and XPC, were
`each found in a single tumor, but not in association with an
`increased number of other mutations. Only two tumors had
`missense mutations in TP53 and none had mutations in PTEN,
`but both tumor suppressors were commonly altered through
`hetero- or homozygous copy-number loss (24% and 21%,
`respectively). Comparison of synonymous and nonsynonymous
`changes detected in these samples suggests a low mutation rate
`in prostate cancer (0.31 mutations/Mb). Consistent with this
`notion, the frequency of mutations recovered in our analysis
`did not exceed the expected background rate (Ding et al.,
`2008), although the modest number of genes and samples
`sequenced limits this analysis.
`We next integrated the CNA, transcriptome, and mutation data
`to conduct a core pathway analysis, based on the success of
`this approach in revealing common pathway alterations in glio-
`blastoma (Cancer Genome Atlas Research Network, 2008).
`Three well-known cancer pathways were commonly altered,
`PI3K, RAS/RAF, and RB, with frequencies ranging from 34% to
`43% in primary tumors versus 74% to 100% in metastases
`(Figure 1B). In this analysis, a tumor was considered altered if
`one or more genes in the pathway were mutated or significantly
`deregulated at the expression level (outlier expression compared
`to the distribution of expression in normal prostate samples, see
`Experimental Procedures). As in glioblastoma, the extremely
`high frequency of alteration in these pathways became evident
`only through examination of multiple genes in each pathway
`since individual genes are affected less commonly. Of particular
`interest is the PI3K pathway, which was altered in nearly half of
`primaries and all metastases examined. Loss of PTEN function,
`through deletion, mutation, or reduced expression, has been
`well documented in prostate cancer with an estimated frequency
`of 40% (Pourmand et al., 2007), consistent with our findings
`here. The frequency of PI3K pathway alteration rises substan-
`tially when PTEN alterations are considered together with alter-
`ations in the INPP4B and PHLPP phosphatases recently
`
`Cancer Cell 18, 11–22, July 13, 2010 ª2010 Elsevier Inc. 13
`
`

`
`Genomic Characterization of Human Prostate Cancers
`
`Cancer Cell
`
`A
`
`B
`
`AKT
`
`Figure 1. A Global View of the Prostate Cancer Genome
`(A) Significant genomic aberrations in the prostate oncogenome. Regions of amplification (red) or deletion (blue) with FDR %10% are plotted, with chromosomes
`indicated at the center and centromeres in red. Genes in which we detected somatic nonsynonymous mutations are listed on top (black). Additional genes of
`interest targeted by copy-number alterations alone are also indicated (gray).
`(B) Three of the most commonly altered pathways in both primary and metastatic prostate cancers: RB, PI3K, and RAS/RAF signaling. Alteration frequencies are
`shown for individual genes and for the entire pathway in primary and metastatic tumors. Alterations are defined as those having outlier expression (significant up-
`or downregulation) compared with the distribution of expression in normal prostate samples (outlier analysis described in further detail in Experimental Proce-
`dures), or by somatic mutations, and are interpreted as activation (red) or inactivation (blue) of protein function. See also Figure S1 and Tables S3 and S6.
`
`implicated in PI3K regulation, the PIK3CA gene itself, and the
`PIK3CA regulatory subunits PIK3R1 and PIK3R3 (Cancer
`Genome Atlas Research Network, 2008; Gao et al., 2005; Gewin-
`ner et al., 2009; Jaiswal et al., 2009; Ueki et al., 2003). These data
`provide strong rationale for exploring the clinical activity of PI3K
`pathway inhibitors, many of which are now in early clinical devel-
`opment, in prostate cancer.
`
`Common Genomic Alterations in Androgen Receptor
`Signaling Pathway Members
`We also conducted a core pathway analysis of AR, which is
`essential for growth and differentiation of the normal prostate
`and is responsible for treatment failure in castration-resistant,
`metastatic disease (Chen et al., 2004; Tran et al., 2009). As
`expected, alteration of AR through mutation, gene amplification,
`
`14 Cancer Cell 18, 11–22, July 13, 2010 ª2010 Elsevier Inc.
`
`

`
`Cancer Cell
`
`Genomic Characterization of Human Prostate Cancers
`
`A
`
`C
`
`B
`
`100%100%
`
`D
`
`E
`
`Figure 2. Diversity of Androgen Signaling Pathway Alterations in Primary and Metastatic Prostate Cancers
`(A) Alterations in androgen signaling components where frequencies are shown for AR and selected modulators in primary and metastatic tumors. Alterations are
`defined as those having outlier expression (as described in Figure 1B and Experimental Procedures), or by somatic mutations, and interpreted as activation (red)
`or inactivation (blue).
`(B) Overall genomic alteration rates in androgen signaling genes. The 11 genes in the AR pathway (A) had somatic mutations (black, red) and/or outlier expression
`(over- or underexpression, see Experimental Procedures), a subset of which was the result of copy-number alterations (gray, broad and focal gain or loss of one or
`more copies, see the Supplemental Information). Primary tumors show moderate levels of alteration in at least one of these 11 AR pathway genes (left), preceding
`the generally higher alteration rates in metastatic tumors (right).
`(C) The steroid receptor coactivator NCOA2 had two novel somatic point mutations in primary tumors. These clustered near sites of known NCOA2 point muta-
`tions in melanoma (G435S in the serine/threonine-rich [S/T] regulatory domain) and lung cancer (S1024N in the transcriptional activation domain 1 [AD1]).
`(D) Increasing levels of NCOA2 induce increasing androgen-dependent AR transcriptional activity. Increasing amounts of NCOA2 plasmid were transfected into
`LNCaP cells, resulting in NCOA2 protein expression (inset western blot) and ARE-luciferase reporter activity. Error bars representing the SEM (standard error of
`the mean) are displayed but represent a very small portion of the signal and are therefore not visible.
`(E) Noncastrate primary tumors with NCOA2 gain (defined as copy-number amplification greater than single-copy gain, outlier overexpression, or mutation) have
`higher androgen signaling (Student’s t test, p < 0.0001), as assessed by an independent signature of 29 androgen-responsive genes (Hieronymus et al., 2006).
`See also Figure S2.
`
`and/or overexpression was common but occurred exclusively in
`metastatic samples (58%, Figure 2A). However, AR pathway
`analysis (including several known AR coactivators and corepres-
`sors) revealed alteration in 56% of primaries and 100% in metas-
`tases (Figure 2A; Figure S2). Among AR pathway genes, the most
`striking finding was a peak of copy-number gain on 8q13.3
`(57 Mb away from the peak at 8q24 commonly attributed to
`MYC, and of even greater significance) that spans the nuclear
`receptor coactivator gene NCOA2 (also known as SRC2/TIF2/
`GRIP1). Seventeen percent of tumors had broad gains of the
`region spanning NCOA2 on 8q, whereas 6.2% of tumors (1.9%
`and 24.3% of primary and metastases, respectively) harbored
`focal or high-level amplifications of the locus and these were
`
`significantly correlated with elevated NCOA2 transcript levels
`(p < 1016, Figure S2A). In addition to copy-number and expres-
`sion changes, AR pathway alterations included mutations in
`NCOA2 (two confirmed somatic) as well as in NCOR2 (three
`tumors), NRIP1, TNK2, and EP300 (one tumor each). Overall,
`8% of primary tumors and 37% of metastases had NCOA2
`gain of expression (determined to be outlier expression as
`described in Experimental Procedures) or mutation (Figures 2A
`and 2B).
`Including broader gains of 8q,
`the frequency of
`NCOA2 alteration may be as high as 20% and 63% in primary
`and metastatic tumors, respectively. Of note, NCOA2 mutations
`have also been reported in melanoma and lung cancer and, in
`conjunction with the prostate mutations detected here, cluster
`
`Cancer Cell 18, 11–22, July 13, 2010 ª2010 Elsevier Inc. 15
`
`

`
`Genomic Characterization of Human Prostate Cancers
`
`Cancer Cell
`
`ERG in transgenic mice and in a prostate tissue reconstitution
`model (Carver et al., 2009; King et al., 2009; Zong et al., 2009).
`The 3p14 deletion, whose association with TMPRSS2-ERG
`was even more significant, has not been previously reported
`and spans only eight genes. Further interrogation of 2550 tumors
`and cell
`lines spanning 14 tumor types (acute lymphoid leu-
`kemia, breast, colorectal, esophageal, glioma, hepatocellular,
`non-small cell
`lung, squamous lung, medulloblastoma, mela-
`noma, myeloproliferative, ovarian, renal, and prostate) for CNAs
`in this region suggests this deletion is only found in prostate
`cancer (Beroukhim et al., 2010). Indeed, the only other focal
`signal found in this region is an amplicon in melanoma that
`includes microphthalmia-associated transcription factor (MITF),
`a previously reported finding (Garraway et al., 2005).
`Closer inspection of the 3p14 deletion in our prostate cohort
`revealed two distinct peaks of association within the region that,
`together with expression data and the focal deletion patterns,
`implicates only three genes: FOXP1, RYBP, and SHQ1 (Figures
`3B and 3C). Deletions in some tumors spanned FOXP1 only,
`whereas others included RYBP and SHQ1, but spared FOXP1.
`FOXP1 encodes a forkhead box transcription factor and functions
`in motor neuron specification in the spinal cord, as well as early
`thymocyte development, in collaboration with various HOX genes
`(Arber, 2008; Pfaff, 2008). A role for FOXP1 in cancer has been
`proposed based on reduced expression in breast and other
`cancers, increased expression in some lymphoid malignancies
`and, remarkably, by translocation-mediated fusion to the ERG
`homolog ETV1 in at least one prostate cancer (Goatly et al.,
`2008; Hermans et al., 2008; Koon et al., 2007). Furthermore,
`recent evidence implicates the FoxP family member FOXP3 as
`a potential tumor suppressor in prostate cancer (Wang et al.,
`2009). RYBP (Ring and YY1 Binding Protein) encodes a polycomb
`group transcriptional repressor implicated in homeotic develop-
`ment and, potentially, as a tumor suppressor through inhibition
`of MDM2 and subsequent p53 stabilization (Chen et al., 2009).
`SHQ1 encodes an accessory factor for the assembly of H/ACA
`ribonucleoproteins (RNP)
`through direct binding to NAP57,
`a core RNP subunit. Missense mutations in NAP57 that disrupt
`interaction with SHQ1 are associated with the bone marrow
`failure syndrome dyskeratosis congenita, raising a potential link
`to precancer syndromes (Grozdanov et al., 2009).
`To gather further evidence for a potential tumor suppressor
`role of either of these genes, we searched for point mutations
`through exon resequencing. We found no mutations in FOXP1
`or RYBP, but detected a confirmed somatic mutation in SHQ1
`(P22S) in a highly conserved region of the CS domain that is
`required for SHQ1 function (Singh et al., 2009). A second tumor
`had a deletion targeting the middle of the SHQ1 gene that,
`consequently, resulted in production of an aberrant mRNA
`species truncated at exon 6 (Figure 3D). Although these data
`further implicate SHQ1 as a tumor suppressor in this locus, the
`fact that some tumors with 3p14 loss spare SHQ1 (Figure 3B) rai-
`ses the possibility of multiple tumor suppressors in this region.
`
`Unsupervised Clustering of CNAs Reveals Distinct
`Subgroups with Differing Risk of Relapse
`after Prostatectomy
`Given the pressing need for biomarkers that distinguish indolent
`from aggressive prostate cancer, we also examined the genomic
`
`in two highly conserved regions. These include a serine/threo-
`nine-rich stretch (S/T) known to be phosphorylated and an acti-
`vation domain (AD1) that mediates binding with histone acetyl-
`transferases such as CBP and p300 (Huang and Cheng, 2004)
`(Figure 2C). A third patient had an A407S NCOA2 substitution
`that was potentially germline (detected at low frequency in adja-
`cent normal prostate). Interestingly, noncastrate patients with
`primary tumors harboring NCOA2 mutation, overexpression, or
`high-level amplification had significantly higher rates of recur-
`rence (Figure S2B).
`The combination of the high frequency of NCOA2 gain in
`primary tumors and its known role as an AR coactivator (Agoulnik
`et al., 2005) suggests that these two genes might collaborate in
`early prostate cancer progression by enhancing AR transcrip-
`tional output. We addressed this possibility by expressing
`increasing levels of NCOA2 in prostate cancer cells with a fixed
`endogenous level of nonamplified AR. The range in NCOA2
`protein levels is shown by Western blot (Figure 2D) and is
`similar to the 2- to 4-fold increase in NCOA2 mRNA levels
`over the mean level seen in the overall prostate cancer cohort.
`As expected from other work, AR transcriptional output (mea-
`sured using an androgen-responsive reporter construct) was
`increased when cells were treated with dihydrotestosterone
`(DHT)
`in a dose-dependent fashion, but reached a plateau
`between 1 and 10 mM (Figure 2D). Increasing levels of NCOA2
`shifted the DHT dose-response curve leftward and upward, indi-
`cating that NCOA2 can not only prime AR to respond to lower
`androgen concentrations but can also boost the total magnitude
`of AR transcriptional output. One prediction from these in vitro
`data is that the AR transcriptional output in prostate cancers
`with NCOA2 gene amplification should be greater than those
`without. Based on a 29-gene signature of AR transcriptional
`output previously used to conduct small molecule screens for
`novel antiandrogens (Hieronymus et al., 2006), NCOA2-ampli-
`fied primary tumors displayed an increase in AR signaling (Fig-
`ure 2E). Collectively, the genomic and functional data suggest
`that NCOA2 functions as a driver oncogene in primary tumors
`by increasing AR signaling, which is known to play a critical
`role in early and late stage prostate cancer. In contrast, AR
`amplification, which is largely restricted to castration-resistant
`metastatic disease, is more likely a mechanism of drug resis-
`tance rather than a natural step in tumor progression. We also
`propose that NCOA2 and MYC both function as driver onco-
`genes on the 8q13 and 8q24 amplicons, respectively.
`
`Genetic Alterations Highly Associated
`with TMPRSS2-ERG
`The TMPRSS2-ERG fusion is the single most prevalent molec-
`ular lesion in prostate cancer (Tomlins et al., 2005). Functional
`studies of TMPRSS2-ERG, including transgenic expression in
`the mouse prostate, have shown modest evidence of oncogenic
`activity (Carver et al., 2009; King et al., 2009; Klezovitch et al.,
`2008; Tomlins et al., 2008), which raises the possibility that coop-
`erating events are required.
`Analysis of 194 tumors for CNAs associated with TMPRSS2-
`ERG fusion revealed three significant regions of copy-number
`loss: two spanning the tumor suppressors PTEN and TP53 and
`a third spanning the multigenic region at 3p14 (Figure 3A).
`PTEN loss was recently shown to cooperate with TMPRSS2-
`
`16 Cancer Cell 18, 11–22, July 13, 2010 ª2010 Elsevier Inc.
`
`

`
`Cancer Cell
`
`Genomic Characterization of Human Prostate Cancers
`
`A
`
`C
`
`B
`
`D
`
`Figure 3. TMPRSS2-ERG-Associated Deletion of 3p14.1-p13
`(A) TMPRSS2-ERG gene fusion co-occurs with genomic aberrations in the prostate cancer genome including deletions of loci encoding TP53 (17p13.1) and PTEN
`(10q23.31), as well as focal deletion of 3p14.1-p13 (genes listed in genomic order; green lines represent statistically significant associations, FDR %1%). Chro-
`mosomes are shown around the ring.
`(B) Diverse genomic deletions (heterozygous and homozygous deletions, light and dark blue, respectively) target a 2.2 Mb region of 3p14.1-p13 encoding eight
`genes (indicated at bottom). Tumors are rows and those harboring TMPRSS2-ERG fusion or PTEN/TP53 deletion (heterozygous and homozygous deletions are
`black and gray, respectively) are identified at right. Tumors are sorted according to their locus of deletion with focal losses preferentially affecting FOXP1 (top),
`RYBP and the adjacent gene SHQ1 (bottom), or both loci simultaneously (middle). Inset indicates the pattern of significance of total genomic deletion juxtaposed
`to the significance of TMPRSS2-ERG-associated deletion (black and dotted blue, respectively).
`(C) Transcript expression according to copy-number status for the three genes targeted by the 3p14.1-p13 deletion: FOXP1, RYBP, and SHQ1, all three of which
`are correlated (p values as indicated, ANOVA). EIF4E3 and PPP4R2 expression and copy-number loss were also correlated, but neither of these two genes was
`focally targeted by 3p14.1-p13 deletion.
`(D) Along with whole-gene deletions of SHQ1 (B), we detected a single tumor with a P22S somatic mutation of the CS domain, indicated in red in the three-dimen-
`sional structure of the SHQ1 yeast homolog (3eud) and in linear representation of the protein (top right). Also, intragenic deletions (shown here in metastatic
`sample PCA0187) confer exon-specific loss of expression indicating a truncation event (bottom right).
`
`data for prognostic significance. It is estimated that 30%–50% of
`men diagnosed with prostate cancer could avoid surgery or radi-
`ation (and instead be followed by watchful waiting) because they
`have good-prognosis tumors that are unlikely to progress (Coop-
`erberg et al., 2005). Whereas transcriptome analysis defines
`breast cancer subgroups with distinct prognoses and treatment
`outcomes that have changed clinical practice (Paik et al., 2004;
`van de Vijver et al., 2002), similar studies in prostate cancer have
`been less clinically useful (Mucci et al., 2008a, 2008b). The 5 year
`median clinical follow-up linked to this tumor set provided an
`opportunity to address the prognosis question using various
`
`forms of oncogenomic data. While unsupervised hierarchical
`clustering of mRNA and microRNA data failed to identify robust
`clusters of patients with significant differences in prognosis, the
`CNA data revealed distinct subgroups with substantial differ-
`ences in time to biochemical (PSA) relapse (Figures 4A and 4B;
`Figure S3A–S3C). Further attempts to identify individual genes
`whose expression has prognostic impact through outlier anal-
`ysis (1766 genes with over- or underexpressing outliers relative
`to normal prostate) were only modestly successful and these
`associations were weak relative to those observed using the
`CNA data.
`
`Cancer Cell 18, 11–22, July 13, 2010 ª2010 Elsevier Inc. 17
`
`

`
`Genomic Characterization of Human Prostate Cancers
`
`Cancer Cell
`
`A
`
`B
`
`C
`
`*
`
`Figure 4. Genomic Aberrations Identify Clinically Distinct Subtypes of Prostate Cancer
`(A) Unsupervised hierarchical clustering of copy-number alterations (heat map; red represents amplification, white represents copy-neutral, blue represents dele-
`tion) indicates six groups of prostate cancers exist. Samples are ordered on the basis of their group membership (dendrogram, groups are colored; metastatic
`samples are indicated by hashes). Selected genomic regi

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket