Next Article in Journal
The Efficacy and Safety of Immune Checkpoint Inhibitor and Tyrosine Kinase Inhibitor Combination Therapy for Advanced or Metastatic Renal Cell Carcinoma: A Multicenter Retrospective Real-World Cohort Study
Next Article in Special Issue
Radiobiological Meta-Analysis of the Response of Prostate Cancer to Different Fractionations: Evaluation of the Linear–Quadratic Response at Large Doses and the Effect of Risk and ADT
Previous Article in Journal
Novel Candidate loci and Pathogenic Germline Variants Involved in Familial Hematological Malignancies Revealed by Whole-Exome Sequencing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genomic Prostate Score: A New Tool to Assess Prognosis and Optimize Radiation Therapy Volumes and ADT in Intermediate-Risk Prostate Cancer

1
Department of Radiation Oncology and Henri Mondor Breast Center, Henri Mondor Hospital, APHP, University of Paris Est Créteil (UPEC), IMRB, INSERM U 955, 94000 Créteil, France
2
Department of Pathology, Henri Mondor Hospital, University of Paris Est Créteil (UPEC), IMRB, INSERM U 955, 94000 Créteil, France
3
Department of Medical Oncology, Henri Mondor Hospital, University of Paris Est Créteil (UPEC), 94000 Créteil, France
4
Department of Urology, Henri Mondor Hospital, University of Paris Est Créteil (UPEC), 94000 Créteil, France
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(3), 945; https://doi.org/10.3390/cancers15030945
Submission received: 25 December 2022 / Revised: 28 January 2023 / Accepted: 30 January 2023 / Published: 2 February 2023
(This article belongs to the Special Issue Advances in Prostate Cancer Radiotherapy)

Abstract

:

Simple Summary

Current standard clinical risk-stratification systems do not sufficiently reflect the disease heterogeneity for prostate cancer. Intermediate risk prostate cancer represents a highly heterogeneous risk group with different treatment options available such as surgery, radiation therapy and hormone therapy. Genomic Prostate Score which can obtained from prostate core biopsies could help to personalize treatment for men with intermediate-risk prostate cancer. The main aim of this present prospective study was to assess the impact of Genomic Prostate Score when compared to clinical risk factors alone in this population. This research study demonstrated that use of Genomic Prostate Score in a series of 30 patients with intermediate risk prostate cancer resulted in major shift in risk groups in most patients. This genomic score represents a potential impactful tool for treatment decision and better personalization of treatment for patients with intermediate risk in daily practice.

Abstract

Genomic classifiers such as the Genomic Prostate Score (GPS) could help to personalize treatment for men with intermediate-risk prostate cancer (I-PCa). In this study, we aimed to evaluate the ability of the GPS to change therapeutic decision making in I-PCa. Only patients in the intermediate NCCN risk group with Gleason score 3 + 4 were considered. The primary objective was to assess the impact of the GPS on risk stratification: NCCN clinical and genomic risk versus NCCN clinical risk stratification alone. We also analyzed the predictive role of the GPS for locally advanced disease (≥pT3+) and the potential change in treatment strategy. Thirty patients were tested for their GPS between November 2018 and March 2020, with the median age being 70 (45–79). Twenty-three patients had a clinical T1 stage. Eighteen patients were classified as favorable intermediate risk (FIR) based on the NCCN criteria. The median GPS score was 39 (17–70). Among the 23 patients who underwent a radical prostatectomy, Gleason score 3 + 4 was found in 18 patients. There was a significant correlation between the GPS and the percentage of a Gleason grade 4 or higher pattern in the surgical sample: correlation coefficient r = 0.56; 95% CI = 0.2–0.8; p = 0.005. In this study, the GPS combined with NCCN clinical risk factors resulted in significant changes in risk group.

1. Introduction

Prostate cancer (PCa) is the second-most commonly diagnosed cancer and the sixth leading cause of cancer death among men worldwide, with an estimated 1,276,000 new cases and 359,000 deaths in 2018 [1]. The global PCa burden is expected to rise to almost 2.3 million new cases and 740,000 deaths by 2040 simply due to the growth and aging of the population.
Risk stratification and treatment decision making for localized PCa traditionally relies on a combination of clinical staging, PSA level, and biopsy results (Gleason score) [2,3].
Intermediate-risk PCa consists of a highly heterogeneous population with different treatment options. Evidence shows that favorable intermediate risk (FIR) PCa patients have cancer-specific mortality rates similar to those with low-risk PCa and, thus, may be candidates for active surveillance, dose-escalated radiation therapy (RT) without short-term androgen deprivation therapy (ADT). Conversely, patients with unfavorable intermediate-risk (UIR) PCa have prostate cancer-specific mortality rates similar to those reported in high-risk patients. Theoretically, they would require a more aggressive multimodal approach, including long-term ADT in addition to standard-dose or dose-escalated radiation therapy with a brachytherapy boost, for example.
In a review that focused on intermediate-risk PCa, Zumsteg and Zelefsky presented this entity as a highly heterogenous one. This clinical heterogeneity makes application of a uniform treatment standard difficult, especially knowing that more than 50% of variability in cure rates for PCa is related to clinical factors other than tumor stage, PSA level, and Gleason score, such as the proportion of positive biopsy cores, perineural invasion, pretreatment PSA velocity, and primary Gleason pattern [4,5,6,7].
In this context, the liquid biopsy field in PCa has advanced exponentially, developing prognostic and predictive biomarkers including circulating tumor cells, extracellular vesicles, circulating tumor DNA, RNA, and holding promise for a minimally invasive approach of monitoring tumor response [8].
Similarly, genomic classifiers (GC) have been shown to be promising for the identification of aggressive PCa from tumoral tissue and for guiding treatment decisions with different commercially available profiling panels including Prolaris®, ProMark®, Oncotype DX®, and Decipher® [9]. The bulk of the evidence for the GC is available for improving risk stratification in the postoperative setting and for guidance for adjuvant radiation therapy. Less data exists for patients treated with definitive radiation therapy, but GC might facilitate personalized oncologic treatments in various perspectives in all disease stages.
In this study, we aimed to evaluate the ability of the Genomic Prostate Score (GPS) for potentially modifying treatment decision in intermediate-risk PCa in terms of indication and duration of ADT and volumes of radiation compared to clinical parameters alone.

2. Materials and Methods

2.1. Patients and Study Design

The charts of the patients who underwent diagnostic prostatic core biopsies were all reviewed in the weekly genitourinary multidisciplinary tumor board, with eligible patients for the GPS study prospectively recruited between November 2018 and March 2020. Inclusion criteria were: intermediate NCCN (National Comprehensive Cancer Network) clinical risk group and Gleason grade 2 (Gleason score 3 + 4). Exclusion criteria were: high-risk NCCN features including PSA > 20, clinical T3, or >Gleason grade 2, or the presence of clinical nodal or distant metastases. Other exclusion criteria were: age > 80, prior prostate core biopsies, ongoing active surveillance program for PCa, prior ADT or pelvic irradiation, other active cancer, or any other life-threatening disease. The presence of prostatic intra-epithelial neoplasia was also an exclusion criterion, as this histologic pattern was excluded from GPS studies. GPS score tests were proposed to all eligible patients. The first 30 positive responders who completed and returned the approval form were included in the analysis.Multiparametric prostate MRI, bone scan, and CT scan were systematically performed. Choline PET CT or PET MRI were optional for the detection of distant metastases. GPS tests were provided free of charge by Genomic Health based on the agreement for the Physician Experience Program (PEP). The purpose of the PEP is to allow treating physicians to gain experience and acquaint themselves with the Oncotype DX® Genomic Prostate Score. Treatments were provided blind to the GPS results. The study was conducted in accordance with the Declaration of Helsinki and was approved by the HOPITAL DE MONDOR Institutional Review Board (project number 16169, N° ID-RCB: 2016-A00789-42, 22 September 2016).

2.2. Biopsy and Tumor Selection Process

All patients underwent transrectal ultrasound-guided 18-core biopsies and other additional MRI-targeted biopsies were performed in the case of identifiable lesions. Formalin-fixed, paraffin-embedded specimens obtained from biopsy tissue were reviewed by the pathologist according to the 2016 International Society of Urological Pathology consensus guidelines. Tissues were selected for analysis with the GPS assay from the biopsy core associated with the largest tumor amount with Gleason score 4. Eight unstained slides of 5 µm were retrieved from the most predominant core biopsy with Gleason score 4.
Samples underwent standard, pre-established quality control measures prior to generating the GPS result. Samples were referred to the central laboratory of Genomic Health (Redwood City, CA, USA) using a specimen transportation kit.

2.3. GPS Assay Description

The Oncotype DX® Genomic Prostate Score test is a quantitative reverse transcriptase polymerase chain reaction (RT-PCR) assay that measures the expression levels of 17 genes (12 cancer-related and 5 housekeeper) in Messenger RNA extracted from microdissected tumor tissue obtained from fixed prostate core biopsies. It provides a score scaled from 0 to 100 as a molecular measure of increasing tumor aggressiveness and is correlated with the risk of finding an adverse pathology feature (Gleason = 4 + 3 and/or pT3+) on the radical prostatectomy specimen [10]. A global risk group was provided based on the combination of clinical factors and the genomic score (GPS). Evidence shows that the GPS is also predictive of the biochemical recurrence and of the risk of metastases [11]. The estimated risk of metastases and prostate cancer death at 10 years was provided, along with the GPS results estimated on clinical validation of the GPS based on radical prostatectomy.

2.4. Endpoints

The primary objective was to assess the impact of the GPS on risk stratification: NCCN clinical and genomic risk stratification versus NCCN clinical risk stratification alone. In the surgical cohort, we aimed to analyze the correlation between the GPS and the percentage of a Gleason score 4 higher pattern in the surgical sample using a Pearson correlation test. We also analyzed the predictive role of the GPS for locally advanced disease (=pT3+) using logistic regression. Statistical significance was defined as p < 0.05. Irrespective of the treatment actually offered to the patient, radiation was considered as the definitive treatment and the use of ADT, its duration, and the radiation volumes were recommended by radiation oncologists based on the clinical factors alone (PSA level, clinical stage, percentage of positive biopsies, NCCN risk group) and then including the GPS. The follow-up was estimated between the date of surgery or the first day of the radiation and the last dedicated follow-up consultation. All analyses were performed using R software version 3.5.1 (R project, Vienna, Austria).

3. Results

3.1. Patients’ Selection Process

Between November 2018 and March 2020, 164 patients who underwent prostate core biopsies were identified with Gleason score 3 + 4. Among them, 44 were excluded because of the presence of one of the exclusion criteria (prior prostate core biopsies, age > 80, active cancer, or any other life-threatening disease). The remaining 120 patients were considered eligible and the 30 first responders were selected for the GPS test.

3.2. Patients Characteristics

Among the 30 patients who were tested for the GPS, the median age was 70 (45–79) with a median PSA level of 7 ng/mL (0.9–16) (Table 1). Of the 30 patients, 23 had a clinical T1 stage and 7 had a clinical T2 stage. There were 12 patients who had ≥ 50% of their core biopsies involved, while 18 had < 50% of their core biopsies involved. Surgery was performed in 23 patients and 7 patients underwent definitive radiation combined with a short-term ADT.

3.3. Risk Stratification

A total of 18 patients were classified as favorable intermediate risk (FIR) and 12 patients were classified as unfavorable intermediate risk (UIR), as per the NCCN clinical stratification. The median GPS score based on the gene expression was 39 (17–70). The risk category based on the combination of NCCN clinical parameters and the genomic features was provided by Genomic Health for the 30 patients included in the study. Reclassification was observed in 66% of cases (20/30), with 60% (18/30) associated with a higher risk category and 6% (2/30) associated with a lower risk category. The potential impact on treatment decision based on the consensual opinion of radiation oncologists was also significant, with 80% (24/30) of change consisting of intensification in 77% (23/30) and de-escalation in 3% (1/30). Furthermore, 6 patients were reclassified as FIR, 13 as UIR, and 11 as high risk (the details are presented in Figure 1). After a median follow-up of 11 months, 2 patients experienced biochemical recurrence after radical prostatectomy and underwent salvage radiation associated with ADT.

3.4. Correlation between GPS and Histological Features

Among the 23 patients who underwent radical prostatectomy, Gleason score 3 + 4 was found in 18 patients and Gleason score 4 + 3 was found in 5 patients. The Pearson test showed a significant correlation between the GPS and the percentage of a Gleason grade 4 or higher pattern in the surgical sample: correlation coefficient r = 0.56; 95% CI = 0.2–0.8; p = 0.005. Among the 23 patients who underwent radical prostatectomy, 11 were found to have locally advanced disease in the surgical specimen (=pT3+) (pT3a: 10 cases, pT3b: 1 case) and 12 had a localized disease (pT2). The GPS was not significantly predictive of the locally advanced disease (=pT3+) in the radical prostatectomy sample with the logistic regression model: Odds ratio = 1.07; 95% CI = 0.99–1.14; p = 0.09. Among the 23 patients who underwent radical prostatectomy, 20 patients had a lymph node dissection. One patient was found to have a single positive lymph node metastasis among the 10 lymph nodes removed (1N+/10N).

3.5. GPS IMPACT on Therapeutic Decision Making and RT Volumes

The potential impact of the GPS in treatment decision is presented in Table 2. Among the 30 patients included in the study, 23 would have received treatment intensification (77%), 1 would have received treatment de-escalation (3%), and 6 would have had the treatment unchanged.

4. Discussion

The management of localized PCa is currently based on clinical risk groups. In the era of personalized medicine, several prognostic GC such as Oncotype DX GPS®, Prolaris®, ProMark®, DNA-ploidy, and Decipher® have been investigated in PCa [12]. The investigation of GC for guiding radiation therapy as the primary treatment has been limited, but several ongoing prospective trials are addressing this question (Table 3). Genomic classifiers may provide greater insight into PCa tumor biology and more accurately stratify PCa risk for adverse outcomes prior to definitive radiation therapy. Few retrospective studies investigated the clinical utility of these genomic signatures based on biopsy samples in patients with localized PCa treated with definitive radiotherapy.
Decipher® is a genomic classifier based on the expression of 22 selected RNA markers using formalin-fixed, paraffin-embedded tissue. In the context of definitive radiotherapy for PCa, Decipher® was investigated on biopsy specimens from the NRG biobank from patients enrolled in the NRG/RTOG 9202, 9413, and 9902 phase III randomized radiation therapy trials [13]. With a median follow-up of 11 years, the Decipher score was shown to be a significant prognostic factor for distant metastases, PCa-specific mortality, and overall survival in univariate and multivariate analysis in a high-risk PCa population.
In a prospective study, Berlin et al. analyzed outcomes of 121 patients with NCCN intermediate-risk PCa treated with definitive RT without ADT. Decipher® outperformed all other indices in its prediction of distant metastases (HR = 2.05; 95% CI = 1.24–4.24) [14].
On the other hand, Oncotype DX GPS® is a 17-gene RT-PCR-based assay that evaluates the expression of 12 cancer-related genes and 5 housekeeper genes from prostate core biopsies. Oncotype DX GPS® assay generates a genomic score from 0 to 100, with higher scores indicative of more aggressive disease. Oncotype DX GPS® is recognized as an option for patients with very low and low-risk localized PCa who are candidates for active surveillance according to National Comprehensive Cancer Network (NCCN) and American Society for Clinical Oncology guidelines [12].
Oncotype DX GPS® has also been validated as an independent prognostic factor of adverse pathology, biochemical recurrence, distant metastasis, and prostate cancer-related death in men with localized PCa after radical prostatectomy in NCCN very low, low, and favorable intermediate risk population [10,11]. In a higher risk cohort (UIR patients), Cullen et al. reported that the GPS score was a strong independent predictor of recurrence, metastatic evolution, and death [15]. In that study, GPS > 40 was associated with worse outcomes and may require treatment intensification.
A recently published retrospective study reported outcomes of 238 men with localized PCa treated with radiation therapy in North Carolina from 2000 to 2016. The NCCN very low or low, favorable intermediate, unfavorable intermediate, high and very-high-risk patients represented 8%, 23%, 37%, 21%, and 11%, respectively.
With a median follow-up of 7.6 years. GPS results per 20-unit increase were significantly associated with biochemical failure (HR = 3.62; 95% CI = 2.59–5.02), distant metastases (HR = 4.48; 95% CI = 2.75–7.38), and PCa death (HR = 5.36; 95% CI = 3.06–9.76) in univariable analysis. Moreover, GPS results retained statistical significance in the multivariable analysis [16].
Interestingly, the dichotomization of the GPS with a cut-off of 40 (0–40 vs. 41–100) yielded similar results, with patients with GPS > 40 predictive of worse outcomes.
To the best of our knowledge, our study is the first to focus on intermediate NCCN risk group patients with Gleason grade 2 (Gleason score 3 + 4). Our aim was to assess the potential clinical impact of the GPS on risk stratification (NCCN clinical and genomic risk stratification versus NCCN clinical risk stratification alone). The risk group migration occurred in 66% of patients, with 60% associated with a higher risk category.
The potential impact on treatment decision based on the consensual opinion of radiation oncologists was even more significant, with 80% of change consisting mainly of intensification in 77% (23/30). The median GPS score in our study was 39, ranging from 17 to 70. Interestingly, this level is close to the threshold of 40 from the surgical cohort of Cullen et al. and the radiation cohort associated with poor outcomes [15,16].
Intensification strategies for intermediate unfavorable and high risk include ADT, prophylactic pelvic nodal irradiation, and brachytherapy boost.
In intermediate-risk PCa, the use of ADT is controversial with modern doses of radiation since its benefit is limited to biochemical control with overall survival [4].
Long-term ADT is known to improve disease-free survival and overall survival with a superiority of long over short term for high-risk patients [17]. In a recent phase 3 trial, prophylactic pelvic nodal irradiation improved biochemical free survival and disease survival in high-risk PCa [18].
To our knowledge, this report represents the first prospective study investigating the potential clinical utility of Oncotype DX GPS® in patients with PCa with Gleason score 3 + 4 based on 18-core biopsies. The correlation between the GPS score and the percentage of Gleason grade 4 or higher pattern in the surgical specimen was consistent with prior studies. Major limitations related to the study need to be acknowledged. First, monocentric and observational design, and the small size of the cohort (n = 30), represent notable limitations. Second, recommendations related to ADT and radiation volumes were left to the appreciation of the comity of radiation oncologists based on clinical parameters (PSA level, clinical stage, percentage of positive biopsies, NCCN risk group) and genomic score. Different therapies may be offered for two patients within the same NCCN risk group and some of the treatment suggestions were not in accordance with NCCN guidelines. Third, the use of brachytherapy boost could also have been the part of the treatment suggestion. Finally, the follow-up was insufficient (11 months) to study the correlation between GPS and clinical outcomes.

5. Conclusions

Intermediate-risk PCa represents a highly heterogeneous group with different treatment options available. In this study, the GPS combined with NCCN clinical risk factors resulted in significant changes in risk group definition. From a radiation oncologist point of view, this may have resulted in significant differences in terms of radiation volumes and duration of ADT. Future prospective studies exploring genomic classifiers to further personalize therapy in intermediate-risk PCa should be performed.

Author Contributions

Conceptualization, Y.B., A.D.L.T. and G.L.; methodology, Y.B. and G.L.; software, K.D. and G.L.; validation, Y.B., K.D., G.C., J.B., M.N., N.H.T., M.A.C., C.S., A.I., A.D.L.T. and G.L.; formal analysis, G.L. and J.B.; investigation, M.N., G.L. and Y.B.; resources, A.D.L.T., A.I. and M.N.; writing—original draft preparation, Y.B. and G.L.; writing—review and editing, Y.B., K.D. and G.C.; visualization, Y.B., A.D.L.T. and G.L.; supervision, Y.B., K.D., G.C., J.B., M.N., N.H.T., M.A.C., C.S., A.I., A.D.L.T. and G.L.; project administration, A.D.L.T. and C.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and was approved by the HOPITAL DE MONDOR Institutional Review Board (project number 16169, N° ID-RCB: 2016-A00789-42, 22 September 2016).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

The authors are in possession of the datasets related to the study and are willing to share with the editorial board if needed.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Culp, M.B.; Soerjomataram, I.; Efstathiou, J.A.; Bray, F.; Jemal, A. Recent Global Patterns in Prostate Cancer Incidence and Mortality Rates. Eur. Urol. 2020, 77, 38–52. [Google Scholar] [CrossRef] [PubMed]
  2. Stephenson, A.J.; Scardino, P.T.; Eastham, J.A.; Bianco, F.J.; Dotan, Z.A.; Fearn, P.A.; Kattan, M.W. Preoperative Nomogram Predicting the 10-Year Probability of Prostate Cancer Recurrence after Radical Prostatectomy. J. Natl. Cancer Inst. 2006, 98, 715–717. [Google Scholar] [CrossRef] [PubMed]
  3. Cooperberg, M.R.; Broering, J.M.; Carroll, P.R. Risk Assessment for Prostate Cancer Metastasis and Mortality at the Time of Diagnosis. J. Natl. Cancer Inst. 2009, 101, 878–887. [Google Scholar] [CrossRef] [PubMed]
  4. Zumsteg, Z.S.; Zelefsky, M.J. Short-Term Androgen Deprivation Therapy for Patients with Intermediate-Risk Prostate Cancer Undergoing Dose-Escalated Radiotherapy: The Standard of Care? Lancet Oncol. 2012, 13, e259–e269. [Google Scholar] [CrossRef] [PubMed]
  5. D’Amico, A.V.; Renshaw, A.A.; Cote, K.; Hurwitz, M.; Beard, C.; Loffredo, M.; Chen, M.-H. Impact of the Percentage of Positive Prostate Cores on Prostate Cancer-Specific Mortality for Patients with Low or Favorable Intermediate-Risk Disease. J. Clin. Oncol. 2004, 22, 3726–3732. [Google Scholar] [CrossRef] [PubMed]
  6. Feng, F.Y.; Qian, Y.; Stenmark, M.H.; Halverson, S.; Blas, K.; Vance, S.; Sandler, H.M.; Hamstra, D.A. Perineural Invasion Predicts Increased Recurrence, Metastasis, and Death from Prostate Cancer Following Treatment with Dose-Escalated Radiation Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2011, 81, e361–e367. [Google Scholar] [CrossRef] [PubMed]
  7. D’Amico, A.V.; Renshaw, A.A.; Sussman, B.; Chen, M.-H. Pretreatment PSA Velocity and Risk of Death from Prostate Cancer Following External Beam Radiation Therapy. JAMA 2005, 294, 440–447. [Google Scholar] [CrossRef] [PubMed]
  8. Crocetto, F.; Russo, G.; Di Zazzo, E.; Pisapia, P.; Mirto, B.F.; Palmieri, A.; Pepe, F.; Bellevicine, C.; Russo, A.; La Civita, E.; et al. Liquid Biopsy in Prostate Cancer Management-Current Challenges and Future Perspectives. Cancers 2022, 14, 3272. [Google Scholar] [CrossRef] [PubMed]
  9. Spohn, S.K.B.; Draulans, C.; Kishan, A.U.; Spratt, D.; Ross, A.; Maurer, T.; Tilki, D.; Berlin, A.; Blanchard, P.; Collins, S.; et al. Genomic Classifiers in Personalized Prostate Cancer Radiotherapy Approaches—A Systematic Review and Future Perspectives Based on International Consensus. Int. J. Radiat. Oncol. Biol. Phys. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  10. Klein, E.A.; Cooperberg, M.R.; Magi-Galluzzi, C.; Simko, J.P.; Falzarano, S.M.; Maddala, T.; Chan, J.M.; Li, J.; Cowan, J.E.; Tsiatis, A.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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Van Den Eeden, S.K.; Lu, R.; Zhang, N.; Quesenberry, C.P.; Shan, J.; Han, J.S.; Tsiatis, A.C.; Leimpeter, A.D.; Lawrence, H.J.; Febbo, P.G.; et al. A Biopsy-Based 17-Gene Genomic Prostate Score as a Predictor of Metastases and Prostate Cancer Death in Surgically Treated Men with Clinically Localized Disease. Eur. Urol. 2018, 73, 129–138. [Google Scholar] [CrossRef] [PubMed]
  12. Eggener, S.E.; Rumble, R.B.; Armstrong, A.J.; Morgan, T.M.; Crispino, T.; Cornford, P.; van der Kwast, T.; Grignon, D.J.; Rai, A.J.; Agarwal, N.; et al. Molecular Biomarkers in Localized Prostate Cancer: ASCO Guideline. J. Clin. Oncol. 2020, 38, 1474–1494. [Google Scholar] [CrossRef] [PubMed]
  13. Nguyen, P.L.; Huang, H.-C.R.; Spratt, D.E.; Davicioni, E.; Sandler, H.M.; Shipley, W.U.; Efstathiou, J.A.; Simko, J.P.; Pollack, A.; Dicker, A.P.; et al. Analysis of a Biopsy-Based Genomic Classifier in High-Risk Prostate Cancer: Meta-Analysis of the NRG Oncology/RTOG 9202, 9413, and 9902 Phase III Randomized Trials. Int. J. Radiat. Oncol. Biol. Phys. 2022; in press. [Google Scholar] [CrossRef] [PubMed]
  14. Berlin, A.; Murgic, J.; Hosni, A.; Pintilie, M.; Salcedo, A.; Fraser, M.; Kamel-Reid, S.; Zhang, J.; Wang, Q.; Ch’ng, C.; et al. Genomic Classifier for Guiding Treatment of Intermediate-Risk Prostate Cancers to Dose-Escalated Image Guided Radiation Therapy Without Hormone Therapy. Int. J. Radiat. Oncol. Biol. Phys. 2019, 103, 84–91. [Google Scholar] [CrossRef] [PubMed]
  15. Cullen, J.; Kuo, H.-C.; Shan, J.; Lu, R.; Aboushwareb, T.; Van Den Eeden, S.K. The 17-Gene Genomic Prostate Score Test as a Predictor of Outcomes in Men with Unfavorable Intermediate Risk Prostate Cancer. Urology 2020, 143, 103–111. [Google Scholar] [CrossRef] [PubMed]
  16. Janes, J.L.; Boyer, M.J.; Bennett, J.P.; Thomas, V.M.; De Hoedt, A.M.; Edwards, V.D.K.; Singla, P.K.; Abran, J.M.; Aboushwareb, T.; Salama, J.K.; et al. The 17-Gene Genomic Prostate Score Test Is Prognostic for Outcomes After Primary External Beam Radiation Therapy in Men With Clinically Localized Prostate Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2023, 115, 120–131. [Google Scholar] [CrossRef] [PubMed]
  17. Zapatero, A.; Guerrero, A.; Maldonado, X.; Alvarez, A.; Gonzalez San Segundo, C.; Cabeza Rodríguez, M.A.; Macias, V.; Pedro Olive, A.; Casas, F.; Boladeras, A.; et al. High-Dose Radiotherapy with Short-Term or Long-Term Androgen Deprivation in Localised Prostate Cancer (DART01/05 GICOR): A Randomised, Controlled, Phase 3 Trial. Lancet Oncol. 2015, 16, 320–327. [Google Scholar] [CrossRef] [PubMed]
  18. Murthy, V.; Maitre, P.; Kannan, S.; Panigrahi, G.; Krishnatry, R.; Bakshi, G.; Prakash, G.; Pal, M.; Menon, S.; Phurailatpam, R.; et al. Prostate-Only Versus Whole-Pelvic Radiation Therapy in High-Risk and Very High-Risk Prostate Cancer (POP-RT): Outcomes From Phase III Randomized Controlled Trial. J. Clin. Oncol. 2021, 39, 1234–1242. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Clinical and genomic risk stratification. Classification based on NCCN clinical risk group alone and combined NCCN clinical and genomic risk group (GPS). The combined risk group (based on NCCN risk group and genomic score) was provided in the report by Genomic Health. Each round represents a patient.
Figure 1. Clinical and genomic risk stratification. Classification based on NCCN clinical risk group alone and combined NCCN clinical and genomic risk group (GPS). The combined risk group (based on NCCN risk group and genomic score) was provided in the report by Genomic Health. Each round represents a patient.
Cancers 15 00945 g001
Table 1. Patients’ characteristics.
Table 1. Patients’ characteristics.
CharacteristicsN = 30%
Age (years)
Median (range)70 (45–79)-
PSA (ng/mL)
Median (range)7 (0.9–16)-
Clinical T Stage
T12377
T2723
Prostate Volume (mL)
Median (range)40 (15–75)-
Number of Positive Cores
Median (range)7.5 (1–17)-
Number of Cores Sampled
Median (range)18 (18–22)-
Positive Cores/Cores Sampled Ratio
<50%1860
≥50%1240
NCCN Risk Group
Intermediate favorable1860
Intermediate unfavorable1240
Upfront Treatment
Surgery2377
Radiation therapy723
Table 2. Potential impact of GPS on treatment decision.
Table 2. Potential impact of GPS on treatment decision.
Patient Number NCCN Clinical Risk GroupDecision Radiation + Hormonal Therapy (Based on Clinical Factors)GPSNCCN + GPS Risk GroupEstimated Metastatic Risk at 10 Years (%) Based on GPS Validation StudiesEstimated Prostate Cancer Death at 10 Years (%) Based on GPS Validation StudiesDecision Radiation
Based on NCCN + GPS
Impact of GPS
1Unfavorable intermediateRT prostate alone with ADT (short term)17Favorable intermediate41RT prostate alone
without ADT
De-escalation
2Favorable intermediateRT prostate alone with ADT (short term)53High173RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
3Favorable intermediateRT prostate alone without ADT33Unfavorable intermediate71RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
4Unfavorable intermediateRT prostate alone without ADT37Unfavorable intermediate91RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
5Unfavorable intermediateRT prostate alone without ADT19Favorable intermediate41RT prostate alone
without ADT
No change
6Favorable intermediateRT prostate alone with ADT (short term)39Unfavorable intermediate101RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
7Favorable intermediateRT prostate alone without ADT40Unfavorable intermediate101RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
8Favorable intermediateRT prostate alone without ADT30Unfavorable intermediate71RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
9Favorable intermediateRT prostate alone without ADT46High132RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
10Favorable intermediateRT prostate alone without ADT70High234RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
11Unfavorable intermediateRT prostate alone without ADT25Unfavorable intermediate51RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
12Favorable intermediateRT prostate alone without ADT47High132RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
13Favorable intermediateRT prostate alone without ADT36Unfavorable intermediate81RT prostate alone
without ADT
Intensification
14Unfavorable intermediateRT prostate alone with ADT (short term)39Unfavorable intermediate101RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
15Unfavorable intermediateRT prostate alone without ADT42High111RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
16Favorable intermediateRT prostate alone without ADT19Favorable intermediate41RT prostate alone
without ADT
No change
17Favorable intermediateRT prostate alone without ADT18Favorable intermediate41RT prostate alone
without ADT
No change
18Favorable intermediateRT prostate alone without ADT18Favorable intermediate41RT prostate alone
without ADT
No change
19Unfavorable intermediateRT prostate alone without ADT40Unfavorable intermediate101RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
20Favorable intermediateRT prostate alone without ADT56High193RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
21Favorable intermediateRT prostate alone without ADT26Unfavorable intermediate61RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
22Unfavorable intermediateRT prostate alone without ADT20Unfavorable intermediate41RT prostate alone
with ADT (short term)
Intensification
23Favorable intermediateRT prostate alone without ADT18Favorable intermediate41RT prostate alone
without ADT
no change
24Favorable intermediateRT prostate alone without ADT34Unfavorable intermediate81RT of the pelvic nodes and
the prostate with ADT (short term)
Intensification
25Unfavorable intermediateRT prostate alone with ADT (short term)43High111RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
26Unfavorable intermediateRT prostate alone without ADT58High203RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
27Favorable intermediateRT prostate alone without ADT47High122RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
28Unfavorable intermediateRT prostate alone with ADT (short term)55High183RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
29Favorable intermediateRT prostate alone without ADT53High173RT of the pelvic nodes and
the prostate with ADT (long term)
Intensification
30Unfavorable intermediateRT of the pelvic nodes and the prostate with ADT (short term)39Unfavorable intermediate101RT of the pelvic nodes and
the prostate with ADT (short term)
No change
Table 3. Selected ongoing trials evaluating genomic biomarkers to guide treatment decisions in patients undergoing definitive radiation therapy.
Table 3. Selected ongoing trials evaluating genomic biomarkers to guide treatment decisions in patients undergoing definitive radiation therapy.
Trial NameFull Name of TrialCommon NamePhaseParticipants (Number)Status (July 2022)
NRG GU009
NCT04513717
Two studies for patients with high-risk prostate cancer testing less intense treatment for patients with a low gene risk score and testing a more intense treatment with a high gene risk score.PREDICT-RTIII2478Recruiting
NRG GU010
NCT05050084
Two studies for patients with unfavorable intermediate-risk prostate cancer testing less intense treatment for patients with a low gene risk score and testing a more intense treatment with a high gene risk score.GUIDANCEIII2050Recruiting
NCT04396808Genomics in Michigan to adjust outcomes in prostate cancer for men with newly diagnosed favorable-risk prostate cancer.G-MAJORIII350Recruiting
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Belkacemi, Y.; Debbi, K.; Coraggio, G.; Bendavid, J.; Nourieh, M.; To, N.H.; Cherif, M.A.; Saldana, C.; Ingels, A.; De La Taille, A.; et al. Genomic Prostate Score: A New Tool to Assess Prognosis and Optimize Radiation Therapy Volumes and ADT in Intermediate-Risk Prostate Cancer. Cancers 2023, 15, 945. https://doi.org/10.3390/cancers15030945

AMA Style

Belkacemi Y, Debbi K, Coraggio G, Bendavid J, Nourieh M, To NH, Cherif MA, Saldana C, Ingels A, De La Taille A, et al. Genomic Prostate Score: A New Tool to Assess Prognosis and Optimize Radiation Therapy Volumes and ADT in Intermediate-Risk Prostate Cancer. Cancers. 2023; 15(3):945. https://doi.org/10.3390/cancers15030945

Chicago/Turabian Style

Belkacemi, Yazid, Kamel Debbi, Gabriele Coraggio, Jérome Bendavid, Maya Nourieh, Nhu Hanh To, Mohamed Aziz Cherif, Carolina Saldana, Alexandre Ingels, Alexandre De La Taille, and et al. 2023. "Genomic Prostate Score: A New Tool to Assess Prognosis and Optimize Radiation Therapy Volumes and ADT in Intermediate-Risk Prostate Cancer" Cancers 15, no. 3: 945. https://doi.org/10.3390/cancers15030945

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop