Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer

Himisha Beltran, Davide Prandi, Juan Miguel Mosquera, Matteo Benelli, Loredana Puca, Joanna Cyrta, Clarisse Marotz, Eugenia Giannopoulou, Balabhadrapatruni V S K Chakravarthi, Sooryanarayana Varambally, Scott A Tomlins, David M Nanus, Scott T Tagawa, Eliezer M Van Allen, Olivier Elemento, Andrea Sboner, Levi A Garraway, Mark A Rubin, Francesca Demichelis, Himisha Beltran, Davide Prandi, Juan Miguel Mosquera, Matteo Benelli, Loredana Puca, Joanna Cyrta, Clarisse Marotz, Eugenia Giannopoulou, Balabhadrapatruni V S K Chakravarthi, Sooryanarayana Varambally, Scott A Tomlins, David M Nanus, Scott T Tagawa, Eliezer M Van Allen, Olivier Elemento, Andrea Sboner, Levi A Garraway, Mark A Rubin, Francesca Demichelis

Abstract

An increasingly recognized resistance mechanism to androgen receptor (AR)-directed therapy in prostate cancer involves epithelial plasticity, in which tumor cells demonstrate low to absent AR expression and often have neuroendocrine features. The etiology and molecular basis for this 'alternative' treatment-resistant cell state remain incompletely understood. Here, by analyzing whole-exome sequencing data of metastatic biopsies from patients, we observed substantial genomic overlap between castration-resistant tumors that were histologically characterized as prostate adenocarcinomas (CRPC-Adeno) and neuroendocrine prostate cancer (CRPC-NE); analysis of biopsy samples from the same individuals over time points to a model most consistent with divergent clonal evolution. Genome-wide DNA methylation analysis revealed marked epigenetic differences between CRPC-NE tumors and CRPC-Adeno, and also designated samples of CRPC-Adeno with clinical features of AR independence as CRPC-NE, suggesting that epigenetic modifiers may play a role in the induction and/or maintenance of this treatment-resistant state. This study supports the emergence of an alternative, 'AR-indifferent' cell state through divergent clonal evolution as a mechanism of treatment resistance in advanced prostate cancer.

Figures

Figure 1. Clinical and mutational profile of…
Figure 1. Clinical and mutational profile of the cohort
(a) Schematic illustrating sites of biopsy for CRPC-NE (dark pink) and CRPC-Adeno (light pink) subgroups. Numbers in circles indicate numerosity of samples from each site. (b) AR signaling (right) based on abundance of mRNA transcripts included in the AR signaling signature described in ref 19. Violin plots show the density of AR signaling. Each dot represents a sample; diamonds and solid lines represent the mean and 95% confidence interval, respectively. Representative immunohistochemistry (left) shows AR protein expression. Scale bars, 50 μm. (c) Significantly mutated genes. Each row represents a gene and each column an individual subject. Top light green bars correspond to the total number of non-silent SNVs in an individual. Left light green bars indicate the number of subjects harboring non silent corresponding mutations in the genes indicated on the right. Bottom panel reports the copy number status of selected genes. (d) Genomic location of AR mutations in samples from SU2C-2015 and this study. (e) Copy number status of AR locus. Color intensity and location are indicative of level and focality of amplification. (f) Frequency of copy number aberrations; concordant fractions (gray), CRPC-NE specific (dark pink) and CRPC-Adeno specific (light pink). Data adjusted for tumor ploidy and purity. Highlighted genes are significantly preferentially aberrant in one class and demonstrate concordant differential mRNA levels (for DNA and mRNA: FDR <= 10% for deletions and p-value <= 1% for amplifications).
Figure 2. Tracing CRPC-NE emergence through allele…
Figure 2. Tracing CRPC-NE emergence through allele specific analysis
(a) Potential models of evolution that occur during prostate cancer progression towards the neuroendocrine phenotype: linear progression from primary untreated adenocarcinoma to CRPC-Adeno to CRPC-NE; independent progression of two distinct clonal populations within the primary or metastatic CRPC-Adeno towards either CRPC-Adeno or CRPC-NE; divergent clonal evolution of CRPC-NE from either primary adenocarcinoma or CRPC-Adeno. *indicates favored model. (b) Allele specific analysis of primary prostate adenocarcinoma and local lymph node metastasis removed at time of radical prostatectomy (RP) and two metastatic CRPC-NE (treated) tumors (3 years after RP) from subject WCMC7520. H&E pathology images and intervening therapies are shown in the timeline. Scale bars, 100 μm. (c) Allele specific analysis of tumors at three time points from patient subject WCMC161 during castration resistance: lymph node (CRPC-Adeno), bone biopsy (CRPC-Adeno), and liver biopsy (small cell CRPC-NE). H&E pathology images and intervening therapies are shown in the timeline. ADT= androgen deprivation therapy; EP= etoposide and cisplatin chemotherapy; Abi= abiraterone acetate with prednisone. Circos plots summarize genome-wide allele specific DNA quantity in tumor cells. Individual's tumor phylogeny sketched upon allele-specific analysis including genome-wide amplification and ploidy assessment. Scale bars, 100 μm.
Figure 3. Methylation analysis of CRPC-NE and…
Figure 3. Methylation analysis of CRPC-NE and CRPC-Adeno
(a) Hierarchical clustering of 28 eRRBS samples data using (1 - Pearson's correlation) as distance measure on unselected sites. Clinical features of outlier cases are described. (b) Left, pie chart showing the number of differentially methylated genes, identified by annotating hyper- and hypo- methylated loci (number is reported between parentheses) on GENCODE version 19. Right, table shows a selection of terms enriched by differentially methylated genes. (c) Top, genome track of SPDEF. Hyper-methylated loci are reported in the annotation track. Bottom, box plot of expression levels of SPDEF samples for This Study (left) and SU2C/PCF 2015 (right) cohorts. (d) Bar plots highlight the effect of EZH2 transcription activity across 487 samples with different pathology classification. The bars are relative to the mRNA level fold (with respect to benign prostate tissue samples) of homeobox genes under-expressed in CRPC-NE versus CRPC-Adeno (FDR < 0.1); a selection of EZH2 target genes (DKK1, NKD1, AMD1, HOXA13, HOXA11, NKX3-1); DNA methyltransferase genes - indicated as DNMTs (DNMT1, DNMT3B, DNMT3A, DNMT3L); EZH2. Significance of differences between CRPC-NE and CRPC-Adeno subgroups are shown (max P = 3*10−5 for DNMTs). When significant, p-values in SU2C/PCF cohort are shown. The number of samples for each pathology classification is reported inside the square symbols of the legend. (e) Cell viability in prostate adenocarcinoma cell lines (DU145, LNCaP) the neuroendocrine prostate cell line NCI-H660 assessed at 48 hours after treatment with escalating does of the EZH2 inhibitor GSK343 (5, 7.5, 10uM).
Figure 4. Integrative DNA, RNA and Methylation…
Figure 4. Integrative DNA, RNA and Methylation analysis
(a) Weighted Venn diagram with the number of protein-coding genes significantly differentially observed in the three data layers. The superimposed pie chart reports the estimation of the impact of each layer upon the following priority rule: methylation overall and DNA over RNA. (b) Integrated NEPC score analysis across 604 samples from four different RNA-Seq prostate cancer datasets (This Study, SU2C/PCF 2015, WCMC 2011/2014 and TCGA). Samples are ordered by decreasing values of Integrated NEPC score (only a fraction of data is shown, entire data are reported in Supplementary Fig. 10). Top, annotation tracks report original dataset and pathology classification. Middle, plot reports Integrated NEPC score (black line) and AR signaling (grey line) across samples. Bottom, heat map of normalized FPKMs for a selection of the 70 genes (in rows) across samples (in columns). (c) Prediction accuracy of CRPC-NE samples by precision and recall statistics for Integrated NEPC Score (circles), AR signaling (squares), mRNA level of SPDEF (diamonds), AR (triangles) in RNA-seq datasets: This Study (green), SU2C/PCF 2015 (orange), WCMC 2011/14 (violet) and all datasets (black). Grey curves represent F-measure levels, defined as the harmonic mean of precision and recall. Due to the absence of CRPC-NE samples (positive events), TCGA data were not reported here. (d) AR signaling versus Integrated NEPC score across 730 samples from five independent prostate datasets using transcriptome data as proxy. The old-rose shaded area refers to significant values of Integrated NEPC Score. Predicted CRPC-NE percentages calculated by excluding benign samples.

References

    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA: a cancer journal for clinicians. 2015;65:5–29. doi:10.3322/caac.21254.
    1. Huggins C, Hodges CV. Studies on prostatic cancer: I. The effect of castration, of estrogen and of androgen injection on serum phosphatases in metastatic carcinoma of the prostate. 1941. The Journal of urology. 2002;168:9–12.
    1. Chen CD, et al. Molecular determinants of resistance to antiandrogen therapy. Nature medicine. 2004;10:33–39. doi:10.1038/nm972.
    1. Knudsen KE, Scher HI. Starving the addiction: new opportunities for durable suppression of AR signaling in prostate cancer. Clinical cancer research : an official journal of the American Association for Cancer Research. 2009;15:4792–4798. doi:10.1158/1078-0432.CCR-08-2660.
    1. Scher HI, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. The New England journal of medicine. 2012;367:1187–1197. doi:10.1056/NEJMoa1207506.
    1. de Bono JS, et al. Abiraterone and increased survival in metastatic prostate cancer. The New England journal of medicine. 2011;364:1995–2005. doi:10.1056/NEJMoa1014618.
    1. Logothetis CJ, et al. Molecular classification of prostate cancer progression: foundation for marker-driven treatment of prostate cancer. Cancer discovery. 2013;3:849–861. doi:10.1158/-12-0460.
    1. Epstein JI, et al. Proposed morphologic classification of prostate cancer with neuroendocrine differentiation. The American journal of surgical pathology. 2014;38:756–767. doi:10.1097/PAS.0000000000000208.
    1. Beltran H, et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer Discov. 2011;1:487–495. doi:10.1158/-11-0130.
    1. Yao JL, et al. Small cell carcinoma of the prostate: an immunohistochemical study. Am J Surg Pathol. 2006;30:705–712. doi:00000478-200606000-00005 [pii]
    1. Beltran H, et al. Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets. Cancer discovery. 2011;1:487–495. doi:10.1158/-11-0130.
    1. Beltran H, et al. Challenges in recognizing treatment-related neuroendocrine prostate cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30:e386–389. doi:10.1200/JCO.2011.41.5166.
    1. Aparicio A, Tzelepi V. Neuroendocrine (Small-Cell) Carcinomas: Why They Teach Us Essential Lessons About Prostate Cancer. Oncology. 2014;28
    1. Lin D, et al. High fidelity patient-derived xenografts for accelerating prostate cancer discovery and drug development. Cancer research. 2014;74:1272–1283. doi:10.1158/0008-5472.CAN-13-2921-T.
    1. Wang HT, et al. Neuroendocrine Prostate Cancer (NEPC) progressing from conventional prostatic adenocarcinoma: factors associated with time to development of NEPC and survival from NEPC diagnosis-a systematic review and pooled analysis. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2014;32:3383–3390. doi:10.1200/JCO.2013.54.3553.
    1. Tagawa ST. Neuroendocrine prostate cancer after hormonal therapy: knowing is half the battle. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2014;32:3360–3364. doi:10.1200/JCO.2014.57.5100.
    1. Tan HL, et al. Rb loss is characteristic of prostatic small cell neuroendocrine carcinoma. Clin Cancer Res. 2014;20:890–903. doi:1078-0432.CCR-13-1982 [pii] 10.1158/1078-0432.CCR-13-1982.
    1. Hieronymus H, et al. Gene expression signature-based chemical genomic prediction identifies a novel class of HSP90 pathway modulators. Cancer cell. 2006;10:321–330. doi:10.1016/j.ccr.2006.09.005.
    1. Chen H, et al. Pathogenesis of prostatic small cell carcinoma involves the inactivation of the P53 pathway. Endocrine-related cancer. 2012;19:321–331. doi:10.1530/ERC-11-0368.
    1. Zhou Z, et al. Synergy of p53 and Rb deficiency in a conditional mouse model for metastatic prostate cancer. Cancer research. 2006;66:7889–7898. doi:10.1158/0008-5472.CAN-06-0486.
    1. Grasso CS, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature. 2012;487:239–243. doi:10.1038/nature11125.
    1. Beltran H, et al. Targeted next-generation sequencing of advanced prostate cancer identifies potential therapeutic targets and disease heterogeneity. European urology. 2013;63:920–926. doi:10.1016/j.eururo.2012.08.053.
    1. Robinson D VAE, Wu Y, Schultz N, Lonigro RJ, Mosquera JM, Montgomery R, Taplin ME, Pritchard CC, Attard G, Beltran H, Abida W, Bradley RK, Vinson J, Cao X, Vats P, Kunju L, Hussain M, Feng FY, Tomlins SA, Cooney KA, Smith DC, Brennan C, Siddiqui J, Mehra R, Chen Y, Rathkopf DE, Morris MJ, Solomon SB, Durack JC, Reuter VE, Gopalan A, Gao J, Loda M, Lis RT, Bowden M, Balk SP, Gaviolaa G, Sougnez C, Gupta M, Yu EU, Mostaghel EA, Cheng HH, Mulcahy H, True LD, Plymate SR, Dvinge H, Ferraldeschi R, Flohr P, Miranda S, Zafeiriou Z, Tunari, Mateo J, Lopez-Perez R, Demichelis F, Robinson BD, Schiffman M, Nanus DM, Tagawa ST, Sigaras A, Eng KW, Elemento O, Sboner A, Heathman EI, Scher HI, Pienta KJ, Kantoff P, de Bono J, Rubin MA, Nelson PS, Garraway LA, Sawyers CL, Chinnaiyan AM. Integrative Clinical Genomics of Advanced Prostate Cancer. Cell. 2015;161:1215–1228.
    1. Baca SC, et al. Punctuated evolution of prostate cancer genomes. Cell. 2013;153:666–677. doi:10.1016/j.cell.2013.03.021.
    1. Barbieri CE, et al. Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nature genetics. 2012;44:685–689. doi:10.1038/ng.2279.
    1. Robinson D, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161:1215–1228. doi:10.1016/j.cell.2015.05.001.
    1. Pietanza MC, Ladanyi M. Bringing the genomic landscape of small-cell lung cancer into focus. Nat Genet. 2012;44:1074–1075. doi:ng.2415 [pii] 10.1038/ng.2415.
    1. Banck MS, et al. The genomic landscape of small intestine neuroendocrine tumors. J Clin Invest. 2013;123:2502–2508. doi:67963 [pii] 10.1172/JCI67963.
    1. Comprehensive molecular characterization of human colon and rectal cancer. Nature. 2012;487:330–337. doi:nature11252 [pii] 10.1038/nature11252.
    1. Prandi D, et al. Unraveling the clonal hierarchy of somatic genomic aberrations. Genome Biol. 2014;15:439. doi:s13059-014-0439-6 [pii] 10.1186/s13059-014-0439-6.
    1. Forbes SA, et al. COSMIC: exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015;43:D805–811. doi:gku1075 [pii] 10.1093/nar/gku1075.
    1. Rajan N, et al. The cylindromatosis gene product, CYLD, interacts with MIB2 to regulate notch signalling. Oncotarget. 2014;5:12126–12140. doi:2573 [pii]
    1. Lim JH, et al. CYLD negatively regulates transforming growth factor-beta-signalling via deubiquitinating Akt. Nat Commun. 2012;3:771. doi:ncomms1776 [pii] 10.1038/ncomms1776.
    1. Theurillat JP, et al. Prostate cancer. Ubiquitylome analysis identifies dysregulation of effector substrates in SPOP-mutant prostate cancer. Science. 2014;346:85–89. doi:346/6205/85 [pii] 10.1126/science.1250255.
    1. Lin L, et al. Mechanisms underlying cancer growth and apoptosis by DEK overexpression in colorectal cancer. PLoS One. 2014;9:e111260. doi:10.1371/journal.pone.0111260 PONE-D-14-27910 [pii]
    1. Nowell PC. The clonal evolution of tumor cell populations. Science. 1976;194:23–28.
    1. Maley CC, et al. Genetic clonal diversity predicts progression to esophageal adenocarcinoma. Nat Genet. 2006;38:468–473. doi:ng1768 [pii] 10.1038/ng1768.
    1. Hansel DE, et al. Shared TP53 gene mutation in morphologically and phenotypically distinct concurrent primary small cell neuroendocrine carcinoma and adenocarcinoma of the prostate. Prostate. 2009;69:603–609. doi:10.1002/pros.20910.
    1. Gundem G, et al. The evolutionary history of lethal metastatic prostate cancer. Nature. 2015;520:353–357. doi:nature14347 [pii] 10.1038/nature14347.
    1. Smith BA, et al. A basal stem cell signature identifies aggressive prostate cancer phenotypes. Proceedings of the National Academy of Sciences of the United States of America. 2015;112:E6544–6552. doi:10.1073/pnas.1518007112.
    1. Borno ST, et al. Genome-wide DNA methylation events in TMPRSS2-ERG fusion-negative prostate cancers implicate an EZH2-dependent mechanism with miR-26a hypermethylation. Cancer discovery. 2012;2:1024–1035. doi:10.1158/-12-0041.
    1. Varambally S, et al. Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer. Science. 2008;322:1695–1699. doi:10.1126/science.1165395.
    1. Yu J, et al. A polycomb repression signature in metastatic prostate cancer predicts cancer outcome. Cancer research. 2007;67:10657–10663. doi:10.1158/0008-5472.CAN-07-2498.
    1. Karanikolas BD, Figueiredo ML, Wu L. Comprehensive evaluation of the role of EZH2 in the growth, invasion, and aggression of a panel of prostate cancer cell lines. The Prostate. 2010;70:675–688. doi:10.1002/pros.21112.
    1. Clermont PL, et al. Polycomb-mediated silencing in neuroendocrine prostate cancer. Clin Epigenetics. 2015;7:40. doi:10.1186/s13148-015-0074-4 74 [pii]
    1. Cheng AS, et al. EZH2-mediated concordant repression of Wnt antagonists promotes beta-catenin-dependent hepatocarcinogenesis. Cancer Res. 2011;71:4028–4039. doi:0008-5472.CAN-10-3342 [pii] 10.1158/0008-5472.CAN-10-3342.
    1. Kanduri M, et al. A key role for EZH2 in epigenetic silencing of HOX genes in mantle cell lymphoma. Epigenetics. 2013;8:1280–1288. doi:26546 [pii] 10.4161/epi.26546.
    1. Akamatsu S, et al. The Placental Gene PEG10 Promotes Progression of Neuroendocrine Prostate Cancer. Cell Rep. 2015;12:922–936. doi:S2211-1247(15)00752-4 [pii] 10.1016/j.celrep.2015.07.012.
    1. The Molecular Taxonomy of Primary Prostate Cancer. Cell. 2015;163:1011–1025. doi:S0092-8674(15)01339-2 [pii] 10.1016/j.cell.2015.10.025.
    1. Chakravarty D, et al. The oestrogen receptor alpha-regulated lncRNA NEAT1 is a critical modulator of prostate cancer. Nat Commun. 2014;5:5383. doi:ncomms6383 [pii] 10.1038/ncomms6383.
    1. Carreira S, et al. Tumor clone dynamics in lethal prostate cancer. Sci Transl Med. 2014;6:254ra125. doi:6/254/254ra125 [pii] 10.1126/scitranslmed.3009448.
    1. Beltran H, et al. Whole-Exome Sequencing of Metastatic Cancer and Biomarkers of Treatment Response. JAMA Oncol. 2015;1:466–474. doi:2294965 [pii] 10.1001/jamaoncol.2015.1313.
    1. Esgueva R, et al. Next-generation prostate cancer biobanking: toward a processing protocol amenable for the International Cancer Genome Consortium. Diagn Mol Pathol. 2012;21:61–68. doi:10.1097/PDM.0b013e31823b6da6.
    1. Van Allen EM, et al. Whole-exome sequencing and clinical interpretation of formalin-fixed, paraffin-embedded tumor samples to guide precision cancer medicine. Nature medicine. 2014;20:682–688. doi:10.1038/nm.3559.
    1. Cibulskis K, et al. ContEst: estimating cross-contamination of human samples in next-generation sequencing data. Bioinformatics. 2011;27:2601–2602. doi:10.1093/bioinformatics/btr446.
    1. Demichelis F, et al. SNP panel identification assay (SPIA): a genetic-based assay for the identification of cell lines. Nucleic Acids Res. 2008;36:2446–2456. doi:gkn089 [pii] 10.1093/nar/gkn089.
    1. Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013;31:213–219. doi:nbt.2514 [pii] 10.1038/nbt.2514.
    1. Ramos AH, et al. Oncotator: cancer variant annotation tool. Hum Mutat. 2015;36:E2423–2429. doi:10.1002/humu.22771.
    1. Lawrence MS, et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature. 2013;499:214–218. doi:nature12213 [pii] 10.1038/nature12213.
    1. Romanel A, Lago S, Prandi D, Sboner A, Demichelis F. ASEQ: fast allele-specific studies from next-generation sequencing data. BMC Med Genomics. 2015;8:9. doi:10.1186/s12920-015-0084-2 10.1186/s12920-015-0084-2 [pii]
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21. doi:bts635 [pii] 10.1093/bioinformatics/bts635.
    1. Habegger L, et al. RSEQtools: a modular framework to analyze RNA-Seq data using compact, anonymized data summaries. Bioinformatics. 2011;27:281–283. doi:btq643 [pii] 10.1093/bioinformatics/btq643.
    1. Park K, et al. Antibody-based detection of ERG rearrangement-positive prostate cancer. Neoplasia. 2010;12:590–598.
    1. Sboner A, et al. FusionSeq: a modular framework for finding gene fusions by analyzing paired-end RNA-sequencing data. Genome Biol. 2010;11:R104. doi:gb-2010-11-10-r104 [pii] 10.1186/gb-2010-11-10-r104.
    1. Mosquera JM, et al. Concurrent AURKA and MYCN gene amplifications are harbingers of lethal treatment-related neuroendocrine prostate cancer. Neoplasia. 2013;15:1–10.
    1. Lin PC, et al. Epigenomic alterations in localized and advanced prostate cancer. Neoplasia. 2013;15:373–383.
    1. Akalin A, et al. methylKit: a comprehensive R package for the analysis of genome-wide DNA methylation profiles. Genome Biol. 2012;13:R87. doi:gb-2012-13-10-r87 [pii] 10.1186/gb-2012-13-10-r87.
    1. Dale RK, Pedersen BS, Quinlan AR. Pybedtools: a flexible Python library for manipulating genomic datasets and annotations. Bioinformatics. 2011;27:3423–3424. doi:btr539 [pii] 10.1093/bioinformatics/btr539.
    1. Chen J, Bardes EE, Aronow BJ, Jegga AG. ToppGene Suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res. 2009;37:W305–311. doi:gkp427 [pii] 10.1093/nar/gkp427.
    1. Vire E, et al. The Polycomb group protein EZH2 directly controls DNA methylation. Nature. 2006;439:871–874. doi:nature04431 [pii] 10.1038/nature04431.
    1. Cerami E, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–404. doi:2/5/401 [pii] 10.1158/-12-0095.

Source: PubMed

3
구독하다