Discovery and validation of a prostate cancer genomic classifier that predicts early metastasis following radical prostatectomy

Nicholas Erho, Anamaria Crisan, Ismael A Vergara, Anirban P Mitra, Mercedeh Ghadessi, Christine Buerki, Eric J Bergstralh, Thomas Kollmeyer, Stephanie Fink, Zaid Haddad, Benedikt Zimmermann, Thomas Sierocinski, Karla V Ballman, Timothy J Triche, Peter C Black, R Jeffrey Karnes, George Klee, Elai Davicioni, Robert B Jenkins, Nicholas Erho, Anamaria Crisan, Ismael A Vergara, Anirban P Mitra, Mercedeh Ghadessi, Christine Buerki, Eric J Bergstralh, Thomas Kollmeyer, Stephanie Fink, Zaid Haddad, Benedikt Zimmermann, Thomas Sierocinski, Karla V Ballman, Timothy J Triche, Peter C Black, R Jeffrey Karnes, George Klee, Elai Davicioni, Robert B Jenkins

Abstract

Purpose: Clinicopathologic features and biochemical recurrence are sensitive, but not specific, predictors of metastatic disease and lethal prostate cancer. We hypothesize that a genomic expression signature detected in the primary tumor represents true biological potential of aggressive disease and provides improved prediction of early prostate cancer metastasis.

Methods: A nested case-control design was used to select 639 patients from the Mayo Clinic tumor registry who underwent radical prostatectomy between 1987 and 2001. A genomic classifier (GC) was developed by modeling differential RNA expression using 1.4 million feature high-density expression arrays of men enriched for rising PSA after prostatectomy, including 213 who experienced early clinical metastasis after biochemical recurrence. A training set was used to develop a random forest classifier of 22 markers to predict for cases--men with early clinical metastasis after rising PSA. Performance of GC was compared to prognostic factors such as Gleason score and previous gene expression signatures in a withheld validation set.

Results: Expression profiles were generated from 545 unique patient samples, with median follow-up of 16.9 years. GC achieved an area under the receiver operating characteristic curve of 0.75 (0.67-0.83) in validation, outperforming clinical variables and gene signatures. GC was the only significant prognostic factor in multivariable analyses. Within Gleason score groups, cases with high GC scores experienced earlier death from prostate cancer and reduced overall survival. The markers in the classifier were found to be associated with a number of key biological processes in prostate cancer metastatic disease progression.

Conclusion: A genomic classifier was developed and validated in a large patient cohort enriched with prostate cancer metastasis patients and a rising PSA that went on to experience metastatic disease. This early metastasis prediction model based on genomic expression in the primary tumor may be useful for identification of aggressive prostate cancer.

Conflict of interest statement

Competing Interests: The authors have read the journal's policy and have the following conflicts: NE, AC, IV, MG, CB, ZH, BZ, TS, TT, and ED are employees of GenomeDx Biosciences Inc. ED and TT own stock in GenomeDx Biosciences Inc. ED has received research funding from GenomeDx Biosciences Inc. and the National Research Council - Industrial Research Assistance Program. PB has received research funding from GenomeDx Biosciences Inc. GK has received research funding from Beckman Coulter. KB, EB, RC, SF, RK, RJ, and TK have declared that no competing interests exist. This does not alter the authors adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Consort diagram.
Figure 1. Consort diagram.
Study breakdown into cases and controls. Training and validation sets are shown.
Figure 2. Multidimensional scaling plot of (A)…
Figure 2. Multidimensional scaling plot of (A) the training and (B) the validation sets.
Controls are indicated in blue and cases in red. In both the training and validation sets the controls tend to cluster on the left of the plot and the cases on the right of the plot. In this manner, most of the biological differences are expressed in the first dimension of the scaling. Random forest proximity [http://www.stat.berkeley.edu/~breiman/] was used to measure the 22 marker distance between samples.
Figure 3. Performance of classifiers and individual…
Figure 3. Performance of classifiers and individual clinicopathologic variables.
For each predictor, the AUC obtained in the training and validation sets, as well as the 95% Confidence Interval for this metric is shown. CC: clinical-only classifier. GC: genomic classifier. GCC: combined genomic-clinical classifier.
Figure 4. Score distributions of multivariable classifiers…
Figure 4. Score distributions of multivariable classifiers in cases and controls in validation set.
Distributions of scores are plotted for A) CC B) GC and C) GCC for controls and cases. Median scores and 95% confidence intervals are represented by a horizontal black line and notches, respectively. Non-overlapping notches indicate that differences in the distribution of scores between cases and controls are statistically significant. Outliers are represented as points beyond the boxplot whiskers.
Figure 5. Distribution of GC scores among…
Figure 5. Distribution of GC scores among pathologic GS categories in validation.
GC scores are plotted with a jitter so as to more easily differentiate the patients among each pathologic GS (x-axis) groups. Case (red) and controls patients (blue) are shown for each category. The dashed black line indicates the GC cutoff of 0.5. Trends show the patients with high GC scores tend to have high GS as well.
Figure 6. Kaplan Meier estimates for all…
Figure 6. Kaplan Meier estimates for all Cases with (A) PCSM and (B) OS endpoints.
Cases were separated into high (>0.5) or low risk according to GC score. Log-rank p-values are shown in the upper right corner. Time to PCSM and OS is measured from BCR in years.
Figure 7. Performance of external signatures in…
Figure 7. Performance of external signatures in training and validation sets.
For each signature, the institution associated to it, year of publication, lead author, the AUC obtained in the training and validation sets, as well as the 95% Confidence Interval for this metric is shown.

References

    1. Siegel R, DeSantis C, Virgo K, Stein K, Mariotto A, et al. (2012) Cancer treatment and survivorship statistics, 2012. CA Cancer J Clin 62: 220–241.
    1. Hull GW, Rabbani F, Abbas F, Wheeler TM, Kattan MW, et al. (2002) Cancer control with radical prostatectomy alone in 1,000 consecutive patients. J Urol 167: 528–534.
    1. Patel AR, Stephenson AJ (2011) Radiation therapy for prostate cancer after prostatectomy: adjuvant or salvage? Nat Rev Urol 8: 385–392.
    1. Mishra MV, Champ CE, Den RB, Scher ED, Shen X, et al. (2011) Postprostatectomy radiation therapy: an evidence-based review. Future Oncol 7: 1429–1440.
    1. Swanson GP, Basler JW (2010) Prognostic factors for failure after prostatectomy. J Cancer 2: 1–19.
    1. Bolla M, van Poppel H, Tombal B, Vekemans K, Da Pozzo L, et al. (2012) Postoperative radiotherapy after radical prostatectomy for high-risk prostate cancer: long-term results of a randomised controlled trial (EORTC trial 22911). Lancet 380: 2018–2027.
    1. Bolla M, van Poppel H, Collette L, van Cangh P, Vekemans K, et al. (2005) Postoperative radiotherapy after radical prostatectomy: a randomised controlled trial (EORTC trial 22911). Lancet 366: 572–578.
    1. Thompson IM Jr, Tangen CM, Paradelo J, Lucia MS, Miller G, et al. (2006) Adjuvant radiotherapy for pathologically advanced prostate cancer: a randomized clinical trial. JAMA 296: 2329–2335.
    1. Wiegel T, Bottke D, Steiner U, Siegmann A, Golz R, et al. (2009) Phase III postoperative adjuvant radiotherapy after radical prostatectomy compared with radical prostatectomy alone in pT3 prostate cancer with postoperative undetectable prostate-specific antigen: ARO 96–02/AUO AP 09/95. J Clin Oncol 27: 2924–2930.
    1. Thompson IM, Tangen CM, Paradelo J, Lucia MS, Miller G, et al. (2009) Adjuvant radiotherapy for pathological T3N0M0 prostate cancer significantly reduces risk of metastases and improves survival: long-term followup of a randomized clinical trial. J Urol 181: 956–962.
    1. Agell L, Hernandez S, Nonell L, Lorenzo M, Puigdecanet E, et al. (2012) A 12-gene expression signature is associated with aggressive histological in prostate cancer: SEC14L1 and TCEB1 genes are potential markers of progression. Am J Pathol 181: 1585–1594.
    1. Bibikova M, Chudin E, Arsanjani A, Zhou L, Garcia EW, et al. (2007) Expression signatures that correlated with Gleason score and relapse in prostate cancer. Genomics 89: 666–672.
    1. Bismar TA, Demichelis F, Riva A, Kim R, Varambally S, et al. (2006) Defining aggressive prostate cancer using a 12-gene model. Neoplasia 8: 59–68.
    1. Cheville JC, Karnes RJ, Therneau TM, Kosari F, Munz JM, et al. (2008) Gene panel model predictive of outcome in men at high-risk of systemic progression and death from prostate cancer after radical retropubic prostatectomy. J Clin Oncol 26: 3930–3936.
    1. Cuzick J, Swanson GP, Fisher G, Brothman AR, Berney DM, et al. (2011) Prognostic value of an RNA expression signature derived from cell cycle proliferation genes in patients with prostate cancer: a retrospective study. Lancet Oncol 12: 245–255.
    1. Glinsky GV, Berezovska O, Glinskii AB (2005) Microarray analysis identifies a death-from-cancer signature predicting therapy failure in patients with multiple types of cancer. J Clin Invest 115: 1503–1521.
    1. Glinsky GV, Glinskii AB, Stephenson AJ, Hoffman RM, Gerald WL (2004) Gene expression profiling predicts clinical outcome of prostate cancer. J Clin Invest 113: 913–923.
    1. Lapointe J, Li C, Higgins JP, van de Rijn M, Bair E, et al. (2004) Gene expression profiling identifies clinically relevant subtypes of prostate cancer. Proc Natl Acad Sci U S A 101: 811–816.
    1. Larkin SE, Holmes S, Cree IA, Walker T, Basketter V, et al. (2012) Identification of markers of prostate cancer progression using candidate gene expression. Br J Cancer 106: 157–165.
    1. Nakagawa T, Kollmeyer TM, Morlan BW, Anderson SK, Bergstralh EJ, et al. (2008) A tissue biomarker panel predicting systemic progression after PSA recurrence post-definitive prostate cancer therapy. PLoS One 3: e2318.
    1. Olmos D, Brewer D, Clark J, Danila DC, Parker C, et al. (2012) Prognostic value of blood mRNA expression signatures in castration-resistant prostate cancer: a prospective, two-stage study. Lancet Oncol 13: 1114–1124.
    1. Penney KL, Sinnott JA, Fall K, Pawitan Y, Hoshida Y, et al. (2011) mRNA expression signature of Gleason grade predicts lethal prostate cancer. J Clin Oncol 29: 2391–2396.
    1. Ramaswamy S, Ross KN, Lander ES, Golub TR (2003) A molecular signature of metastasis in primary solid tumors. Nat Genet 33: 49–54.
    1. Ross RW, Galsky MD, Scher HI, Magidson J, Wassmann K, et al. (2012) A whole-blood RNA transcript-based prognostic model in men with castration-resistant prostate cancer: a prospective study. Lancet Oncol 13: 1105–1113.
    1. Saal LH, Johansson P, Holm K, Gruvberger-Saal SK, She QB, et al. (2007) Poor prognosis in carcinoma is associated with a gene expression signature of aberrant PTEN tumor suppressor pathway activity. Proc Natl Acad Sci U S A 104: 7564–7569.
    1. Singh D, Febbo PG, Ross K, Jackson DG, Manola J, et al. (2002) Gene expression correlates of clinical prostate cancer behavior. Cancer Cell 1: 203–209.
    1. Stephenson AJ, Smith A, Kattan MW, Satagopan J, Reuter VE, et al. (2005) Integration of gene expression profiling and clinical variables to predict prostate carcinoma recurrence after radical prostatectomy. Cancer 104: 290–298.
    1. Talantov D, Jatkoe TA, Bohm M, Zhang Y, Ferguson AM, et al. (2010) Gene based prediction of clinically localized prostate cancer progression after radical prostatectomy. J Urol 184: 1521–1528.
    1. Varambally S, Yu J, Laxman B, Rhodes DR, Mehra R, et al. (2005) Integrative genomic and proteomic analysis of prostate cancer reveals signatures of metastatic progression. Cancer Cell 8: 393–406.
    1. Yu J, Rhodes DR, Tomlins SA, Cao X, Chen G, et al. (2007) A polycomb repression signature in metastatic prostate cancer predicts cancer outcome. Cancer Res 67: 10657–10663.
    1. Pound CR, Partin AW, Eisenberger MA, Chan DW, Pearson JD, et al. (1999) Natural history of progression after PSA elevation following radical prostatectomy. JAMA 281: 1591–1597.
    1. Vergara IA, Erho N, Triche TJ, Ghadessi M, Crisan A, et al. (2012) Genomic "Dark Matter" in Prostate Cancer: Exploring the Clinical Utility of ncRNA as Biomarkers. Front Genet 3: 23.
    1. Prensner JR, Iyer MK, Balbin OA, Dhanasekaran SM, Cao Q, et al. (2011) Transcriptome sequencing across a prostate cancer cohort identifies PCAT-1, an unannotated lincRNA implicated in disease progression. Nat Biotechnol 29: 742–749.
    1. Lockstone HE (2011) Exon array data analysis using Affymetrix power tools and R statistical software. Brief Bioinform 12: 634–644.
    1. McCall MN, Bolstad BM, Irizarry RA (2010) Frozen robust multiarray analysis (fRMA). Biostatistics 11: 242–253.
    1. Leek JT, Scharpf RB, Bravo HC, Simcha D, Langmead B, et al. (2010) Tackling the widespread and critical impact of batch effects in high-throughput data. Nat Rev Genet 11: 733–739.
    1. Crisan A, Ghadessi M, Buerki C, Vergara IA, Thompson DJS, et al... (2012) Clinical and genomic analysis of metastatic disease progression in a background of biochemical recurrence. J Clin Oncol. ASCO Meeting.
    1. Zou H, Hastie T (2005) Regularization and variable selection via the elastic net. JR Statist Soc B 67: 301–320.
    1. Friedman J, Hastie T, Tibshirani R (2010) Regularization Paths for Generalized Linear Models via Coordinate Descent. J Stat Softw 33: 1–22.
    1. Breiman L (2001) Random Forests. In: Shapire RE, editor. Machine Learning: Kluwer Academic Publishers. 5–32.
    1. Liaw A, Wiener M (2002) Classification and Regression by randomForest.. R news 2: 18–22.
    1. Meyer D, Dimitriadou E, Hornik K, Weingessel A, Leisch F (2012) e1071: Misc Functions of the Department of Statistics (e1071), TU Wien.
    1. Deftos LJ (1998) Granin-A, parathyroid hormone-related protein, and calcitonin gene products in neuroendocrine prostate cancer. Prostate Suppl 8: 23–31.
    1. Chen H, Tu SW, Hsieh JT (2005) Down-regulation of human DAB2IP gene expression mediated by polycomb Ezh2 complex and histone deacetylase in prostate cancer. J Biol Chem 280: 22437–22444.
    1. Kristiansen G, Fritzsche FR, Wassermann K, Jager C, Tolls A, et al. (2008) GOLPH2 protein expression as a novel tissue biomarker for prostate cancer: implications for tissue-based diagnostics. Br J Cancer 99: 939–948.
    1. Veeramani S, Yuan TC, Chen SJ, Lin FF, Petersen JE, et al. (2005) Cellular prostatic acid phosphatase: a protein tyrosine phosphatase involved in androgen-independent proliferation of prostate cancer. Endocr Relat Cancer 12: 805–822.
    1. Tomlins SA, Rhodes DR, Perner S, Dhanasekaran SM, Mehra R, et al. (2005) Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer. Science 310: 644–648.
    1. Berney DM, Gopalan A, Kudahetti S, Fisher G, Ambroisine L, et al. (2009) Ki-67 and outcome in clinically localised prostate cancer: analysis of conservatively treated prostate cancer patients from the Trans-Atlantic Prostate Group study. Br J Cancer 100: 888–893.
    1. Stamey TA, Yang N, Hay AR, McNeal JE, Freiha FS, et al. (1987) Prostate-specific antigen as a serum marker for adenocarcinoma of the prostate. N Engl J Med 317: 909–916.
    1. Reiter RE, Gu Z, Watabe T, Thomas G, Szigeti K, et al. (1998) Prostate stem cell antigen: a cell surface marker overexpressed in prostate cancer. Proc Natl Acad Sci U S A 95: 1735–1740.
    1. Perner S, Hofer MD, Kim R, Shah RB, Li H, et al. (2007) Prostate-specific membrane antigen expression as a predictor of prostate cancer progression. Hum Pathol 38: 696–701.
    1. Rubin MA, Zhou M, Dhanasekaran SM, Varambally S, Barrette TR, et al. (2002) alpha-Methylacyl coenzyme A racemase as a tissue biomarker for prostate cancer. JAMA 287: 1662–1670.
    1. Lee WH, Morton RA, Epstein JI, Brooks JD, Campbell PA, et al. (1994) Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis. Proc Natl Acad Sci U S A 91: 11733–11737.
    1. Bussemakers MJ, van Bokhoven A, Verhaegh GW, Smit FP, Karthaus HF, et al. (1999) DD3: a new prostate-specific gene, highly overexpressed in prostate cancer. Cancer Res 59: 5975–5979.
    1. Roth TJ, Sheinin Y, Lohse CM, Kuntz SM, Frigola X, et al. (2007) B7-H3 ligand expression by prostate cancer: a novel marker of prognosis and potential target for therapy. Cancer Res 67: 7893–7900.
    1. Yang G, Truong LD, Timme TL, Ren C, Wheeler TM, et al. (1998) Elevated expression of caveolin is associated with prostate and breast cancer. Clin Cancer Res 4: 1873–1880.
    1. Korn EL (1986) Censoring distributions as a measure of follow-up in survival analysis. Stat Med 5: 255–260.
    1. Brimo F, Montironi R, Egevad L, Erbersdobler A, Lin DW, et al. (2013) Contemporary grading for prostate cancer: implications for patient care. Eur Urol 63: 892–901.
    1. Singh AP, Bafna S, Chaudhary K, Venkatraman G, Smith L, et al. (2008) Genome-wide expression profiling reveals transcriptomic variation and perturbed gene networks in androgen-dependent and androgen-independent prostate cancer cells. Cancer Lett 259: 28–38.
    1. LaTulippe E, Satagopan J, Smith A, Scher H, Scardino P, et al. (2002) Comprehensive gene expression analysis of prostate cancer reveals distinct transcriptional programs associated with metastatic disease. Cancer Res 62: 4499–4506.
    1. Gorlov IP, Byun J, Gorlova OY, Aparicio AM, Efstathiou E, et al. (2009) Candidate pathways and genes for prostate cancer: a meta-analysis of gene expression data. BMC Med Genomics 2: 48.
    1. Van Den Berg C, Guan XY, Von Hoff D, Jenkins R, Bittner, et al (1995) DNA sequence amplification in human prostate cancer identified by chromosome microdissection: potential prognostic implications. Clin Cancer Res 1: 11–18.
    1. Levin AM, Machiela MJ, Zuhlke KA, Ray AM, Cooney KA, et al. (2008) Chromosome 17q12 variants contribute to risk of early-onset prostate cancer. Cancer Res 68: 6492–6495.
    1. Viticchie G, Lena AM, Latina A, Formosa A, Gregersen LH, et al. (2011) MiR-203 controls proliferation, migration and invasive potential of prostate cancer cell lines. Cell Cycle 10: 1121–1131.
    1. Bera TK, Das S, Maeda H, Beers R, Wolfgang CD, et al. (2004) NGEP, a gene encoding a membrane protein detected only in prostate cancer and normal prostate. Proc Natl Acad Sci U S A 101: 3059–3064.
    1. Chung EY, Liu J, Homma Y, Zhang Y, Brendolan A, et al. (2007) Interleukin-10 expression in macrophages during phagocytosis of apoptotic cells is mediated by homeodomain proteins Pbx1 and Prep-1. Immunity 27: 952–964.
    1. Horton R, Wilming L, Rand V, Lovering RC, Bruford EA, et al. (2004) Gene map of the extended human MHC. Nat Rev Genet 5: 889–899.
    1. Barcellos LF, May SL, Ramsay PP, Quach HL, Lane JA, et al. (2009) High-density SNP screening of the major histocompatibility complex in systemic lupus erythematosus demonstrates strong evidence for independent susceptibility regions. PLoS Genet 5: e1000696.
    1. Chen Q, Watson JT, Marengo SR, Decker KS, Coleman I, et al. (2006) Gene expression in the LNCaP human prostate cancer progression model: progression associated expression in vitro corresponds to expression changes associated with prostate cancer progression in vivo. Cancer Lett 244: 274–288.
    1. Sreekumar A, Poisson LM, Rajendiran TM, Khan AP, Cao Q, et al. (2009) Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression. Nature 457: 910–914.
    1. Wishart DS, Jewison T, Guo AC, Wilson M, Knox C, et al. (2013) HMDB 3.0–The Human Metabolome Database in 2013. Nucleic Acids Res 41: D801–807.
    1. Poliseno L, Salmena L, Zhang J, Carver B, Haveman WJ, et al. (2010) A coding-independent function of gene and pseudogene mRNAs regulates tumour biology. Nature 465: 1033–1038.
    1. Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, et al. (2010) Integrative genomic profiling of human prostate cancer. Cancer Cell 18: 11–22.

Source: PubMed

3
Subscribe