Main Content

A Guide for Clinicians in the Evaluation of Emerging Molecular Diagnostics for Newly Diagnosed Prostate Cancer

Diagnosis Update

Diagnosis UpDate A Guide for Clinicians in the Evaluation of Emerging Molecular Diagnostics for Newly Diagnosed Prostate Cancer Steven E. Canfield, MD,1 Adam S. Kibel, MD,2 Michael J. Kemeter, MSPAS,3 Phillip G. Febbo, MD,3 H. Jeffrey Lawrence, MD,3 Judd W. Moul, MD4 1Division of Urology, Department of Surgery, The University of Texas Medical School at Houston, Houston, TX; 2Division of Urology, Department of Surgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA; 3Departments of Medical Affairs and Oncology Development, Genomic Health, Inc., Redwood City, CA; 4Division of Urologic Surgery and Duke Cancer Institute, Duke University Medical Center, Durham, NC Prostate-specific antigen (PSA) screening is associated with a decline in prostate cancerrelated mortality. However, screening has also led to overdiagnosis and overtreatment of clinically insignificant tumors. Recently, certain national guidelines (eg, US Preventive Services Task Force) have recommended against PSA screening, which may lead to a reverse-stage migration. Although many prostate tumors are indolent at presentation, others are aggressive and are appropriate targets for treatment interventions. Utilization of molecular markers may improve our ability to measure tumor biology and allow better discrimination of indolent and aggressive tumors at diagnosis. Many emerging commercial molecular diagnostic assays have been designed to provide more accurate risk stratification for newly diagnosed prostate cancer. Unfamiliarity with molecular diagnostics may make it challenging for some clinicians to navigate and interpret the medical literature to ascertain whether particular assays are appropriately developed and validated for clinical use. Herein, the authors provide a framework for practitioners to use when assessing new tissue-based molecular assays. This review outlines aspects of assay development, clinical and analytic validation and clinical utility studies, and regulatory issues, which collectively determine whether tests (1) are actionable for specific clinical indications, (2) measurably influence treatment decisions, and (3) are sufficiently validated to warrant incorporation into clinical practice. [Rev Urol. 2014;16(4):172-180 doi: 10.3909/riu0644] ® © 2014 MedReviews , LLC Key words Adverse pathology • Biomarker • Clinical utility • Clinical validation • Genomic prostate score • Molecular diagnostics • Prostate cancer 172 • Vol. 16 No. 4 • 2014 • Reviews in Urology 4004170006_RIU0644.indd 172 09/12/14 11:28 AM Molecular Diagnostics for Prostate Cancer T here is a growing consensus that many prostate cancers identified by prostate-specific antigen (PSA) screening are indolent and could be appropriately managed with active surveillance. However, a proportion of PSAdiagnosed, early-stage prostate cancers are biologically aggressive and should be considered for immediate treatment.1 The discrimination of these two types of disease is arguably the greatest challenge in the management of newly diagnosed prostate cancer today. Prediction of outcomes and treatment decisions for prostate collectively determine the aggressiveness of a given tumor. The hallmarks of cancer include (1) sustained proliferation, (2) evasion of growth suppressors, (3) resistance of cell death, (4) enabling of replicative immortality (“immortalization”), (5) induction of angiogenesis, and (6) activation of invasion and metastasis.7 In addition, tumor tissues are composed of multiple interacting cell types, including malignant cells and tumor-associated stroma, which jointly participate in the process of tumor growth and progression. Multiplex molecular markers can provide a window on the contri- Molecular diagnostics can provide an objective biologic measure of tumor aggressiveness. cancer are currently based on various nomograms and risk groupings that incorporate clinical stage, serum PSA, and histologic features on diagnostic needle biopsy.2-4 Although these tools have important prognostic value, numerous series have demonstrated that, in approximately 30% of cases, patients are found to have highergrade and/or -stage disease than predicted by their biopsy features, highlighting the risk of undersampling inherent to needle biopsies.5 Conversely, not all patients with biopsies containing Gleason grade 4 disease prove to have aggressive disease at surgery.6 These discrepancies make it challenging for clinicians to select the appropriate treatment with confidence. Molecular diagnostic assays that provide more accurate measures of tumor aggressiveness may help address this unmet medical need. Molecular diagnostics can provide an objective biologic measure of tumor aggressiveness. Prostate cancer is a complex and highly variable disease with multiple genomic alterations that influence a wide range of biologic pathways, which bution of these various biologic processes to the behavior of individual cancers. A number of tissue-based, multigene expression assays are now clinically available for measuring tumor aggressiveness in prostate cancer. This review provides guidance for clinicians on assessing the validity and utility of these types of genomic tests. The gene expression tests that have the most mature data and are commercially available— the Oncotype DX® Prostate Cancer Test (Genomic Prostate Score [GPS] assay; Genomic Health, Redwood City, CA), which assesses diagnostic biopsy samples at the time of diagnosis; the Decipher® test (GenomeDX Biosciences, San Diego, CA), which assesses postprostatectomy specimens; and the Prolaris® test (Myriad Genetics, Salt Lake City, UT), which assesses either specimen type—serve as the examples in this review (Table 1). The key components in assessing any new diagnostic assays for prostate cancer include understanding the following: (1) how the assay was developed, and what genetic pathways and abnormalities are measured by the test; (2) what clinical need is addressed by the assay and what its clinical indications are (is the assay fit-for-purpose?); (3) what type and amount of tissue is required for the assay, and its cost; and (4) what the level of evidence that has been validated is, being mindful of the three key components of assay validation (analytic validation, clinical validation, and clinical utility; Table 2). Molecular diagnostic assays are new to the clinical care of prostate cancer, and no multiplex genomic assays are currently included in the guidelines for prostate cancer of the National Comprehensive Cancer Network (NCCN) or the American Urological Association. Few multiplex gene expression assays are currently in clinical practice for any solid tumors. One example that has been rigorously validated and incorporated into clinical guidelines for breast cancer8 is the 21-gene Recurrence Score® assay (Oncotype DX breast cancer test; Genomic Health, Redwood City, CA), which is indicated for use in patients with early breast cancer to inform decisions regarding adjuvant chemotherapy.9 We refer to this as a “case study” for the development and validation of such a test. Molecular Diagnostics 101: Molecular Diagnostics Terminology A biomarker can be broadly defined as any measurable biologic entity whose presence signifies a disease or biologic condition. Molecular diagnostics can include nucleic acids (RNA or DNA), protein, and metabolite biomarkers, which can be measured in a variety of human specimens (eg, tissue, plasma, or urine). Nucleic acid–based assays may include measurement of gene expression in messenger RNA, detection of genetic abnormalities such Vol. 16 No. 4 • 2014 • Reviews in Urology • 173 4004170006_RIU0644.indd 173 09/12/14 4:16 PM Molecular Diagnostics for Prostate Cancer continued TABLe 1 Gene Expression Assays Company Assay Name Indication Result Genomic Health Inc. (Redwood City, CA) GenomeDX Biosciences (San Diego, CA) Myriad Genetics (Salt Lake city, UT) Oncotype DX® Prostate Cancer assay Positive prostate biopsy Genomic prostate score from 0 to 100 Likelihood of freedom from higher Gleason scorea and/or non–organ-confined disease Decipher® Post radical prostatectomy Risk of metastasis at 5 years post prostatectomy and 3 years post PSA recurrence Prolaris® Positive prostate biopsy or Post radical prostatectomy 10-year PCSM risk or 10-year BCR risk BCR, biochemical recurrence; PCSM, prostate cancer-specific mortality; PSA, prostate-specific antigen. aHigh Gleason score defined as Gleason dominant pattern 4 or any Gleason pattern 5. TABLe 2 Essential Components of Assay Validation Components of Assay Validation Definition Analytic validation Demonstrating that the assay measures the biomarker(s): it is intended to measure in a robust, accurate, and reproducible manner Clinical validation Demonstrating that the assay has a strong association with clinically relevant outcomes (such as adverse pathology at surgery or clinical recurrence) Clinical utility Demonstrating that the assay results meaningfully influence the clinical management of the patient and improve net health outcomes as mutations, gene deletions, and gene fusions, and epigenetic markers such as DNA methylation. The focus of this review is tissue-based, multigene expression assays that measure RNA levels. Commonly used terms are presented in Table 3. Development of TissueBased, Gene Expression Assays Two major technical challenges for the development of multigene expression assays in prostate cancer bear mentioning: the very small amounts of tissue and RNA available in needle biopsies,11 and the patterns observed in the biopsy may not reflect the overall biology of the tumor. The development strategy should optimize assay performance for very small amounts of tumor RNA and mitigate the effects of tumor heterogeneity. A truly powerful biomarker for prostate cancer should be able to predict the same outcomes from any cancer tissue sampled (and perhaps even from adjacent noncancerous tissue). There is exploratory evidence that the Oncotype DX GPS assay and Prolaris test can predict tumor aggressiveness when measured in adjacent tissue that appears normal,12,13 suggesting that these assays are measuring—in part—a more generalized field effect in the gland and, therefore, may be less The development strategy should optimize assay performance for very small amounts of tumor RNA and mitigate the effects of tumor heterogeneity. intrinsic heterogeneity and multifocal nature of most prostate tumors. Given that a standard 12-core prostate needle biopsy samples , 1% of prostate tissue, gene expression susceptible to tumor heterogeneity and multifocality. The tests discussed here are all designed to measure the expression of multiple genes, using RNA 174 • Vol. 16 No. 4 • 2014 • Reviews in Urology 4004170006_RIU0644.indd 174 09/12/14 11:28 AM Molecular Diagnostics for Prostate Cancer TABLe 3 Molecular Diagnostics Terminology Terminology Description Biomarker (also called molecular marker and signature marker) DNA methylation Biological molecule in blood, other body fluids, or tissues that signifies a normal or abnormal process, including a condition or disease; a biomarker may also be used to assess treatment efficacy Attachment of a methyl group to DNA at cytosine bases; DNA methylation is correlated with reduced gene transcription The study of heritable changes to gene activity (activation; deactivation) without any alterations in the underlying DNA sequencea Process by which information that is encoded in a gene is used by a cell to direct the assembly of a specific protein molecule Study of genes and heredity Study of the complete genome of an organism, which includes genes and their functions DNA that is present in cell mitochondria represents a small fraction of the total DNA in cells; mtDNA contains 37 genes; mtDNA is prone to somatic (a type of noninherited) mutations; mtDNA mutations are associated with numerous chronic diseases, including cancer Observable physical and/or biochemical characteristics of gene expressiona The study of the structure, function, and interaction of proteins produced by specific genes. Various techniques (ie, molecular biology, genetics, and biochemistry) may be utilized Process whereby the RT enzyme enables the transcription of RNA to DNA (retroviruses [eg, HIV], which are RNA viruses, utilize RT to replicate) Epigenetics Gene expression Genetics Genomics mtDNA Phenotype Proteomics RT Types of laboratory assays Microarray-based gene expression profiling PCR qPCR RT-PCR (qualitative) qRT-PCR (quantitative) Simultaneous analysis of mRNA expression levels of multiple genes in a tumor specimen; both the Oncotype DX® Prostate Cancer Assay and the Prolaris test utilize a microarray platformb Qualitative amplification of DNA Amplification and quantitation of DNA Following reverse transcription of RNA into cDNA, multiple copies of cDNA are then generated via PCR, enabling qualitative detection of gene expression Sensitive assay that enables both detection and quantitation of gene expression by creating multiple copies of cDNA from RNA, followed by cDNA amplification with specialized probes aData from National Human Genome Research Institute Web site.10 Oncotype Dx® Prostate Cancer Assay is manufactured by Genomic Health (Redwood City, CA); the Prolaris test is manufactured by Myriad Genetics (Salt Lake city, UT). cDNA, complementary DNA; mtDNA, mitochondrial DNA; PCR, polymerase chain reaction; qPCR, quantitative PCR; qRT-PCR, quantitation real-time PCR; RT, reverse transcription. bThe extracted from formalin-fixed tumor tissue, and to provide a biologic measure of tumor aggressiveness. However, assay development and gene selection strategies for each test were different. Given the contribution of cell proliferation to tumor aggressiveness, the Prolaris test was developed by a narrowly focused screening of 126 cell cycle genes and was not intended to measure other biologic pathways, such as tumor invasiveness. Genes were selected based on their correlation with the mean expression of all 126 genes. The resulting commercial assay, which can be performed on needle biopsy material and on radical prostatectomy tissue, consists of 31 highly correlated cell cycle progression (CCP) genes and 15 housekeeping genes (to correct for variable RNA quality and quantity), and provides Vol. 16 No. 4 • 2014 • Reviews in Urology • 175 4004170006_RIU0644.indd 175 09/12/14 11:28 AM Molecular Diagnostics for Prostate Cancer continued a CCP score that ranges from 22 to 16, with higher numbers reflecting increased proliferation.14 The Oncotype DX GPS test was developed based on a screening of 732 cancer-related genes, representing a wide range of biologic pathways, to determine which genes were most predictive of clinical recurrence after surgery.15 Genes were selected based on their ability to predict clinical recurrence, biochemical recurrence, prostate cancer-specific mortality, and adverse pathology, when assessed in two different regions of a given tumor, to mitigate the confounding effects of tumor heterogeneity, and then further selected by their analytic performance in biopsy tissue. The final biopsy-based assay consists of 12 cancer-related genes (representing androgen signaling, stromal response, cellular organization, and proliferation) and 5 reference genes, and provides a GPS on a 100-unit scale. It is currently not intended for use on radical prostatectomy specimens. The genes in the Decipher assay were selected based on a screening of more than 1.4 million probes using a GeneChip® Human Exon ST Array (Affymetrix, Santa Clara, CA).16 A 22-gene genomic classifier was developed, using a random forest machine learning algorithm, as a predictor of clinical recurrence after surgery. The 22 genes selected for the final assay, which is performed on radical prostatectomy tissue, reflect a variety of biologic pathways, such as cell proliferation, differentiation, and motility, and include noncoding RNAs. Analytic Validation For a test to be analytically validated, it must be proven robust and reliable (eg, in the face of various fixatives, variable amounts of input nucleic acid), and measures the analyte it claims to measure. Analytic validation studies determine the test accuracy, precision, and reproducibility (ie, if the results are consistent between duplicate samples, different operators, different instruments, different reagent lots, and over different time periods), and includes the determination of quality control measures for the assay.17 Standards for the conduct and reporting of analytic validation studies have been recently reviewed.18 The analytic validation studies for the Oncotype DX GPS test demonstrated that all of the genes in the test met prespecified criteria for precision and reproducibility.11 to IV), to indicate that retrospective studies of archival tissues can provide Level 1 evidence of clinical validity (Table 4),20 if they are rigorously designed and conducted; have prestated eligibility criteria, specific aims, and statistical analysis plans; and are performed on patient cohorts that are representative of the contemporary target population for the assay. Lastly, for a study to be considered a true clinical validation study, it should be performed using the final, analytically validated commercial-grade version of the biomarker assay. It should be noted that even retrospective studies that assess late end- For an assay to be clinically validated, a high level of evidence must show that the biomarkers that the test measures have a strong association with one or more clinically relevant outcomes. In prostate cancer, this can include the prediction of adverse pathology at surgery, biochemical recurrence, development of metastases, and prostate cancer death. Clinical Validation For an assay to be clinically validated, a high level of evidence must show that the biomarkers that the test measures have a strong association with one or more clinically relevant outcomes. In prostate cancer, this can include the prediction of adverse pathology at surgery, biochemical recurrence, development of metastases, and prostate cancer death. In 1996, a grading system was proposed to define levels of evidence for clinical validation of tumor markers.19 The traditional gold standard for a clinical validation study was a prospective randomized trial designed specifically to determine if an assay was a significant predictor of a clinically relevant endpoint.17 However, given the extended natural history of prostate cancer, prospective trials with long-term clinical endpoints are not practical. In 2009, the authors modified their algorithm, which assigns levels of evidence (from I points are challenging to conduct, as there are few mature cohorts with available archival tissue, and randomized trials in early prostate cancer are extremely uncommon. Patients in many of these older cohorts were not screened or managed in accordance with contemporary practices. Thus, more near-term endpoints, such as biochemical recurrence and adverse pathology (high-grade and/or high-stage disease) at surgery, are more feasible as surrogates for later clinical events. The three assays discussed here can be compared with these standards. The Oncotype DX GPS has been clinically validated, in two independent cohorts, to predict the likelihood of high-grade and/ or non–organ-confined disease at radical prostatectomy (P , .005) in men with NCCN low and intermediate disease when performed on pretreatment diagnostic needle biopsy specimens.21 The assay has 176 • Vol. 16 No. 4 • 2014 • Reviews in Urology 4004170006_RIU0644.indd 176 09/12/14 11:28 AM Molecular Diagnostics for Prostate Cancer TABLe 4 Determination of LOE Using Elements of Tumor Marker Studies LOE LOE Category Study Design Validation Studies Available I III A B B C C Prospective Prospective using archive samples Prospective using archive samples Prospective/observational Prospective/observational IV-V D Prospective/observational None required $ 1 with consistent results None or inconsistent results $ 2 with consistent results None or 1 with consistent results or inconsistent results Not applicablea II aNot applicable because LOE IV and V studies will never be satisfactory for the determination of medical utility. LOE, level of evidence. Reproduced from Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst. 2009;101: 1446-1452, by permission of Oxford University Press.20 also been validated as a significant independent predictor of biochemical recurrence after surgery for localized prostate cancer.22 The Prolaris CCP score has been clinically validated to predict biochemical recurrence after surgery when performed on radical prostatectomy specimens and to predict prostate cancer-specific death when performed on transurethral resection of the prostate14 or diagnostic needle biopsies.23 The Decipher assay has been clinically validated to predict the likelihood of developing metastatic disease after radical prostatectomy when performed on surgical tissue.16 Thus, all three assays can be considered to have Level 1B evidence for clinical validation (ie, $ 1 prospectively designed study of archival tissues [Table 4]). Clinical Utility Does the assay affect patient outcomes and treatment decisions in a meaningful way? An ideal clinical utility study for a cancer assay would demonstrate that use of a particular test is associated with a reduction in cancer-related mortality. The long natural history of prostate cancer makes such studies impractical. Subsequently, most clinical utility studies focus on short-term endpoints, such as changes in the treatment decisions that are likely to improve long-term outcomes. For example, does using the assay drive greater adoption of active surveillance in good candidates, thereby reducing harms associated with overtreatment, without having a negative effect on overall outcome? Such “decision impact” studies for prostate cancer tests have been conducted a number of different ways. Badani and colleagues24 studied the clinical utility of the Decipher test in the postprostatectomy setting with online surveys using hypothetical cases, for which physicians were asked to indicate their treatment recommendations before and after receiving the result of the test. They observed a 43% to 53% post-test change in treatment recommendations regarding adjuvant or salvage therapy. Crawford and colleagues25 assessed the clinical utility of the Prolaris test in a prospective study of newly diagnosed patients, in which their urologists completed questionnaires indicating their treatment recommendations prior to and after receiving the test results. They observed a 65% change in post-test treatment recommendations, with a 37% reduction in recommendations for interventional therapies. Kartha and associates26 conducted a single-institution study of the clinical utility of the Oncotype DX GPS test on risk stratification and treatment decisions for 115 men with newly diagnosed prostate cancer. They observed a 21% change in risk stratification in the total cohort, and a 46% change (44% to very low risk) in men with NCCN low-risk disease. The authors noted that physician treatment recommendations changed in every case in which the risk stratification was altered by the test result and that patients accepted the physician recommendation in all but one case. The results of physician questionnaire–based studies must be interpreted with caution, as they may not reflect real-life clinical practice and may be subject to the Hawthorne effect (also referred to as “observer effect”). The Hawthorne effect is described as subjects’ awareness of their participation in an experiment, which can affect their behavior and, ultimately, the results of an experiment.27 In addition, it is unclear if the post-test recommendation is actually carried out. Strategies to ensure that such Vol. 16 No. 4 • 2014 • Reviews in Urology • 177 4004170006_RIU0644.indd 177 09/12/14 11:28 AM Molecular Diagnostics for Prostate Cancer continued studies reflect real-life clinical decision making include (1) capturing physician-reported levels of decisional confidence and usefulness of the test, (2) including patient preferences and patient-reported measures of decision conflict, and (3) systematic post-test chart reviews to document the actual management course taken in comparison with matched controls of untested patients. To ensure that a test has broad clinical utility, these studies should represent a range of practice types, patient demographics, and geographic locations. Regulatory Issues: What Regulatory Approvals Are Required for Molecular Diagnostics? Although molecular diagnostic tests in the United States are regulated by the US Food and Drug Administration (FDA) and the Centers for Medicare & Medicaid Services (CMS), the FDA has exer- all laboratory testing. The three prostate cancer tests under consideration here all fall under the CLIA umbrella. Whereas FDA clearance requires demonstration of clinical and analytical validation but not clinical utility, all three components are typically required for inclusion in clinical cancer guidelines and for payer reimbursement. A Case Study of a Fully Validated Molecular Diagnostic for Breast Cancer The 21-gene Recurrence Score assay (Genomic Health, Inc., Redwood City, CA) is an example of a mature molecular diagnostics test that has satisfied all three measures of assay validation—analytic validation, clinical validation, and clinical utility—and is used to help guide treatment decisions regarding adjuvant chemotherapy for women Although molecular diagnostic tests in the United States are regulated by the FDA and CMS, the FDA has exercised discretion in its requirements for clearance and approval of in vitro diagnostic assays, limiting them to distributed tests and companion diagnostics required to select patients for specific drug treatments. cised discretion in its requirements for clearance and approval of in vitro diagnostic assays, limiting them to distributed tests (tests sold as kits to commercial laboratories) and companion diagnostics required to select patients for specific drug treatments.28,29 Laboratory-developed assays (in vitro tests developed, validated, and performed in-house by a specific reference laboratory) are required to abide by the Clinical Laboratory Improvement Amendments (CLIA) of 1988, which are administered by CMS. CLIA aims to ensure reliability and accuracy of test results and establish quality standards for with localized breast cancer. As a consequence of its comprehensive validation, the test is included by various cancer guidelines, including those of the American Society of Clinical Oncology, NCCN, and National Institute for Health and Care Excellence.2 The analytic validation studies for the assay demonstrated that the amplification efficiency, precision, linearity, and dynamic range for each gene in the test met prespecified criteria for analytic performance, and the test was reproducible as described earlier30 and has been cited as a model for the conduct and reporting of analytic validation studies.18 The Recurrence Score assay has been clinically validated in women with early breast cancer to predict the likelihood of (1) 10-year distant recurrence in estrogen receptor–positive, tamoxifen-treated patients31 (2) benefit from adjuvant chemotherapy,32 and (3) breast cancerrelated death in the absence of adjuvant chemotherapy.33 In a 2012 analysis of molecular assays for predicting outcomes in patients with early-stage breast cancer, category I level of evidence was achieved only in studies validating this assay.34 For clinical utility, the use of the Recurrence Score has been shown to alter treatment recommendations in 30% to 40% of patients in several decision impact studies.35 In most cases, the shift was from adjuvant chemotherapy/endocrine therapy to endocrine therapy alone, sparing a substantial fraction of women the toxicity and expense of chemotherapy. These studies also demonstrated that physician confidence in their treatment recommendations improved by 45% (P , .001), and patient decisional conflict was reduced by the Recurrence Score results. Additional Considerations and Conclusions Two major technical challenges for the development of multigene expression assays in prostate cancer deserve some mention. They are the very small amounts of tissue and RNA available in needle biopsies,11 and the intrinsic heterogeneity and multifocal nature of most prostate tumors. Given that a standard 12-core prostate needle biopsy samples , 1% of prostate tissue, gene expression patterns observed in the biopsy may not reflect the overall biology of the tumor. The development strategy should optimize assay performance for very small amounts of tumor RNA, and 178 • Vol. 16 No. 4 • 2014 • Reviews in Urology 4004170006_RIU0644.indd 178 09/12/14 11:28 AM Molecular Diagnostics for Prostate Cancer mitigate effects of tumor heterogeneity. A truly powerful biomarker for prostate cancer should be able to predict the same outcomes from any cancer tissue sampled (and perhaps even from adjacent noncancerous tissue). There is exploratory evidence that the Oncotype DX GPS assay and Prolaris test can predict tumor aggressiveness when measured in adjacent normalappearing tissue,12,13 suggesting that these assays are measuring— in part—a more generalized field effect in the gland and may, therefore, be less susceptible to tumor heterogeneity and multifocality. The key attributes of a clinical biomarker are that (1) it has a strong association with clinically meaningful endpoints, be they pathologic or clinical, (2) it provides additional information about tumor biology beyond existing clinical nomograms, and (3) use of the marker has a meaningful influence on physicians’ treatment decisions and patient outcomes. In the end, the assay should permit more accurate risk assessment for patients with early-stage prostate cancer and more individualized treatment decisions. Steven E. Canfield reports financial relationships with Bayer AG (Leverkusen, North Rhine-Westphalia, Germany), and Genomic Health, Inc. (Redwood City, CA); Adam S. Kibel reports financial relationships with Genomic Health, Inc.; Michael J. Kemeter is a full-time employee of Genomic Health, Inc.; Phillip G. Febbo is a full-time employee of Genomic Health, Inc.; H. Jeffrey Lawrence is a full-time employee of Genomic Health, Inc.; Judd W. Moul reports financial relationships with Myriad Genetics (Salt Lake City, UT) and Genomic Health, Inc. KnowledgePoint360 (Lyndhurst, NJ) provided writing, editorial, and graphics support for the development of this manuscript; their participation was funded by Genomic Health, Inc. (Redwood City, CA). No authors received payment for their participation in the development of this manuscript. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. References 1. 2. Johansson JE, Andrén O, Andersson SO, et al. Natural history of early, localized prostate cancer. JAMA. 2004; 291:2713-2719. Shariat SF, Kattan MW, Vickers AJ, et al. Critical review of prostate cancer predictive tools. Future Oncol. 2009;5:1555-1584. 13. 14. Kattan MW, Cuzick J, Fisher G, et al; Transatlantic Prostate Group. Nomogram incorporating PSA level to predict cancer-specific survival for men with clinically localized prostate cancer managed without curative intent. Cancer. 2008;112:69-74. National Comprehensive Cancer Network Web site. NCCN Clinical Practice Guidelines in Oncology: prostate cancer (Version 2.2014). http://www.nccn. org/professionals/physician_gls/f_guidelines.asp. Conti SL, Dall’era M, Fradet V, et al. Pathological outcomes of candidates for active surveillance of prostate cancer. J Urol. 2009;181:1628-1633. Amin A, Partin A, Epstein JI. Gleason score 7 prostate cancer on needle biopsy: relation of primary pattern 3 or 4 to pathological stage and progression after radical prostatectomy. J Urol. 2011;186:1286-1290. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646-674. National Comprehensive Cancer Network Web site: NCCN Clinical Practice Guidelines in Oncology: breast cancer (Version 2.2014). http://www.nccn.org/ professionals/physician_gls/f_guidelines.asp. Paik S, Tang G, Shak S, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol. 2006;24:3726-3734. National Human Genome Research Institute Web site. Talking glossary of genetic terms. http://www.genome. gov/glossary/index.cfm. Accessed November 10, 2014. Knezevic D, Goddard AD, Natraj N, et al. Analytical validation of the Oncotype DX prostate cancer assay a clinical RT-PCR assay optimized for prostate needle biopsies. BMC Genomics. 2013;14:690. Klein EA, Falzarano SM, Zhang N, et al. Evidence for a field effect in early prostate cancer (PCa): gene expression profiles in normal-appearing prostate tissue (NT) adjacent to tumor (T) as predictors of clinical outcome. J Clin Oncol. 2013;31(suppl):5029. Carvalho F, Welbourn W, Reid R, et al. Evidence for a cell cycle proliferation “field effect” in prostate cancer. Eur Urol. 2013;4(suppl):e605. Abstract 1475. Cuzick J, Swanson GP, Fisher G, et al; Transatlantic Prostate Group. Prognostic value of an RNA MAin PoinTs • Many prostate cancers identified by prostate-specific antigen (PSA) screening are indolent and could be appropriately managed with active surveillance. However, some PSA-diagnosed early-stage prostate cancers are biologically aggressive and should be treated immediately. The discrimination of indolent from aggressive of disease is arguably the greatest challenge in the management of newly diagnosed prostate cancer today; molecular diagnostic assays that provide more accurate measures of tumor aggressiveness may help address this unmet medical need. • Assay development, clinical and analytic validation, and clinical utility studies collectively determine whether tests are actionable for specific clinical indications, measurably influence treatment decisions, and are sufficiently validated to warrant incorporation into clinical practice. • A number of tissue-based, multigene expression assays are now clinically available for measuring tumor aggressiveness in prostate cancer. These include the Oncotype DX® Prostate Cancer Test (Genomic Prostate Score assay; Genomic Health, Redwood City, CA); the Decipher® test (GenomeDX Biosciences, San Diego, CA); and the Prolaris® test (Myriad Genetics, Salt Lake City, UT). • Each of these tests is designed to measure the expression of multiple genes using RNA extracted from formalinfixed tumor tissue, and to provide a biologic measure of tumor aggressiveness. • An assay should permit more accurate risk assessment for patients with early-stage prostate cancer and more individualized treatment decisions. Vol. 16 No. 4 • 2014 • Reviews in Urology • 179 4004170006_RIU0644.indd 179 09/12/14 11:28 AM Molecular Diagnostics for Prostate Cancer continued 15. 16. 17. 18. 19. 20. 21. 22. expression signature derived from cell cycle proliferation genes in patients with prostate cancer: a retrospective study. Lancet Oncol. 2011:12:245-255. Klein EA, Maddala T, Millward C, et al. Development of a needle biopsy-based genomic test to improve discrimination of clinically aggressive from indolent prostate cancer. J Clin Oncol. 2012;30(suppl):4560. Erho N, Crisan A, Vergara IA, et al. Discovery and validation of a prostate cancer genomic classifier that predicts early metastasis following radical prostatectomy. PLoS One. 2013;8:e66855. Teutsch SM, Bradley LA, Palomaki GE, et al; EGAPP Working Group. The Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Initiative: methods of the EGAPP Working Group. Genet Med. 2009;11:3-14. McShane LM, Hayes DF. Publication of tumor marker research results: the necessity for complete and transparent reporting. J Clin Oncol. 2012;30:4223-4232. Hayes DF, Bast RC, Desch CE, et al. Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J Natl Cancer Inst. 1996;88:1456-1466. Simon RM, Paik S, Hayes DF. Use of archived specimens in evaluation of prognostic and predictive biomarkers. J Natl Cancer Inst. 2009;101:1446-1452. Klein EA, Cooperberg MR, Magi-Galluzzi C, et al. A 17-gene assay to predict prostate cancer aggressiveness in the context of Gleason grade heterogeneity, tumor multifocality, and biopsy undersampling. Eur Urol. 2014;66:550-560. Cullen J, Rosner I, Brand T, et al. A prospectively designed study to determine the association of a 17-gene genomic prostate score with recurrence 23. 24. 25. 26. 27. 28. following surgery for localized prostate cancer (PCa). Presented at: the European Society for Medical Oncology (ESMO) Congress 2014; September 26-30, 2014; Madrid, Spain. Abstract LBA22. Cuzick J, Berney DM, Fisher G, et al; Transatlantic Prostate Group. Prognostic value of a cell cycle progression signature for prostate cancer death in a conservatively managed needle biopsy cohort. Br J Cancer. 2012;106:1095-1099. Badani K, Thompson DJ, Buerki C, et al. Impact of a genomic classifier of metastatic risk on postoperative treatment recommendations for prostate cancer patients: a report from the DECIDE study group. Oncotarget. 2013;4:600-609. Crawford ED, Scholz MC, Kar AJ, et al. Cell cycle progression score and treatment decisions in prostate cancer: results from an ongoing registry. Curr Med Res Opin. 2014;30:1025-1031. Kartha GK, Nyame Y, Klein EA. Evaluation of the OncotypeDX genomic prostate score for risk stratification in prostate cancer patients considered candidates for active surveillance. J Clin Oncol. 2014;32 (suppl 4):122. McCambridge J, Witton J, Elbourne DR. Systematic review of the Hawthorne effect: new concepts are needed to study research participation effects. J Clin Epidemiol. 2014;67:267-277. Anderson S, Bloom KJ, Vallera DU, et al. Multisite analytic performance studies of a real-time polymerase chain reaction assay for the detection of BRAF V600E mutations in formalin-fixed, paraffin-embedded tissue specimens of malignant melanoma. Arch Pathol Lab Med. 2012;136: 1385-1391. 29. 30. 31. 32. 33. 34. 35. Benlloch S, Botero ML, Beltran-Alamillo J, et al. Clinical validation of a PCR assay for the detection of EGFR mutations in non-small-cell lung cancer: retrospective testing of specimens from the EURTAC trial. PLoS One. 2014;9:e89518. Cronin M, Sangli C, Liu ML, et al. Analytical validation of the Oncotype DX genomic diagnostic test for recurrence prognosis and therapeutic response prediction in node-negative, estrogen receptor-positive breast cancer. Clin Chem. 2007;53: 1084-1091. Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of tamoxifen-treated, nodenegative breast cancer. N Engl J Med. 2004;351:28172826. Paik S, Tang G, Shak S, et al. Gene expression and benefit of chemotherapy in women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol. 2006;24:3726-3734. Habel LA, Shak S, Jacobs MK, et al. A populationbased study of tumor gene expression and risk of breast cancer death among lymph node-negative patients. Breast Cancer Res. 2006;8:R25. Hornberger J, Alvarado MD, Rebecca C, et al. Clinical validity/utility, change in practice patterns, and economic implications of risk stratifiers to predict outcomes for early-stage breast cancer: a systematic review. J Natl Cancer Inst. 2012;104:1068-1079. Eiermann W, Rezai M, Kümmel S, et al. The 21-gene recurrence score assay impacts adjuvant therapy recommendations for ER-positive, node-negative and node-positive early breast cancer resulting in a riskadapted change in chemotherapy use. Ann Oncol. 2012;24:618-624. 180 • Vol. 16 No. 4 • 2014 • Reviews in Urology 4004170006_RIU0644.indd 180 09/12/14 11:28 AM

Side Content