ArticlePDF Available

Genomic classifiers in personalized prostate cancer radiotherapy approaches – a systematic review and future perspectives based on international consensus

Authors:

Figures

Content may be subject to copyright.
Journal Pre-proof
Genomic classifiers in personalized prostate cancer radiotherapy
approaches a systematic review and future perspectives based on
international consensus
Simon K.B. Spohn MD , C´
edric Draulans MD ,
Amar U. Kishan MD , Daniel Spratt MD , Ashley Ross MD, PhD ,
Tobias Maurer Prof. MD , Derya Tilki Prof., MD ,
Alejandro Berlin MD , Pierre Blanchard MD, PhD ,
Sean Collins MD , Peter Bronsert MD , Ronald Chen MD, PhD ,
Alan Dal Pra MD , Gert de Meerleer MD, Prof. ,
Thomas Eade MD, Prof. , Karin Haustermans MD, Prof. ,
Tobias H¨
olscher MD , Stefan H¨
ocht MD, Prof. ,
Pirus Ghadjar MD, Prof. , Elai Davicioni PhD , Matthias Heck MD ,
Linda G.W. Kerkmeijer MD, PhD , Simon Kirste MD ,
Nikolaos Tselis MD , Phuoc T. Tran MD, PhD ,
Michael Pinkawa MD, Prof. , Pascal Pommier MD ,
Constantinos Deltas PhD , Nina-Sophie Schmidt-Hegemann MD ,
Thomas Wiegel MD, Prof. , Thomas Zilli MD , Alison C. Tree MD ,
Xuefeng Qiu MD , Vedang Murthy MD ,
Jonathan I. Epstein MD, Prof. , Christian Graztke MD, Prof. ,
Xin Gao MD , Anca L. Grosu MD, Prof. , Sophia C. Kamran MD ,
Constantinos Zamboglou MD
PII: S0360-3016(22)03691-4
DOI: https://doi.org/10.1016/j.ijrobp.2022.12.038
Reference: ROB 27992
To appear in: International Journal of Radiation Oncology, Biology, Physics
Received date: 2 June 2022
Revised date: 9 December 2022
Accepted date: 24 December 2022
Please cite this article as: Simon K.B. Spohn MD , C´
edric Draulans MD ,
Amar U. Kishan MD , Daniel Spratt MD , Ashley Ross MD, PhD , Tobias Maurer Prof. MD ,
Derya Tilki Prof., MD , Alejandro Berlin MD , Pierre Blanchard MD, PhD , Sean Collins MD ,
Peter Bronsert MD , Ronald Chen MD, PhD , Alan Dal Pra MD , Gert de Meerleer MD, Prof. ,
Thomas Eade MD, Prof. , Karin Haustermans MD, Prof. , Tobias H¨
olscher MD ,
Stefan H¨
ocht MD, Prof. , Pirus Ghadjar MD, Prof. , Elai Davicioni PhD , Matthias Heck MD ,
Linda G.W. Kerkmeijer MD, PhD , Simon Kirste MD , Nikolaos Tselis MD , Phuoc T. Tran MD, PhD ,
Michael Pinkawa MD, Prof. , Pascal Pommier MD , Constantinos Deltas PhD ,
Nina-Sophie Schmidt-Hegemann MD , Thomas Wiegel MD, Prof. , Thomas Zilli MD ,
Alison C. Tree MD , Xuefeng Qiu MD , Vedang Murthy MD , Jonathan I. Epstein MD, Prof. ,
Christian Graztke MD, Prof. , Xin Gao MD , Anca L. Grosu MD, Prof. , Sophia C. Kamran MD ,
Constantinos Zamboglou MD , Genomic classifiers in personalized prostate cancer radio-
therapy approaches a systematic review and future perspectives based on interna-
tional consensus, International Journal of Radiation Oncology, Biology, Physics (2022), doi:
https://doi.org/10.1016/j.ijrobp.2022.12.038
This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition
of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of
record. This version will undergo additional copyediting, typesetting and review before it is published
in its final form, but we are providing this version to give early visibility of the article. Please note that,
during the production process, errors may be discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.
©2022 Published by Elsevier Inc.
1
Title: Genomic classifiers in personalized prostate cancer radiotherapy
approaches a systematic review and future perspectives based on
international consensus
Short Running Title: Genomic Classifier Prostate Cancer Radiation
Simon K.B. Spohn, MD a,b,c, Cédric Draulans, MD d, Amar U. Kishan, MD e, Daniel
Spratt, MD f, Ashley Ross MD, PhD g, Tobias Maurer, Prof. MD h,i, Derya Tilki Prof.,
MD h,i,j, Alejandro Berlin, MD k, Pierre Blanchard, MD, PhD l, Sean Collins, MD m,
Peter Bronsert, MD n, Ronald Chen, MD, PhD o, Alan Dal Pra, MD p, Gert de
Meerleer, MD, Prof. d, Thomas Eade, MD, Prof. q, Karin Haustermans, MD, Prof. d,
Tobias Hölscher, MD r, Stefancht, MD, Prof. s, Pirus Ghadjar, MD, Prof. t, Elai
Davicioni, PhD u, Matthias Heck, MD v, Linda G.W. Kerkmeijer, MD, PhD w, Simon
Kirste, MD a,b, Nikolaos Tselis, MD x, Phuoc T. Tran, MD, PhD y, Michael Pinkawa,
MD, Prof. z, Pascal Pommier, MD aa, Constantinos Deltas, PhD bb, Nina-Sophie
Schmidt-Hegemann, MD cc, Thomas Wiegel, MD, Prof. dd, Thomas Zilli, MD ee, Alison
C. Tree, MD ff, Xuefeng Qiu, MD gg, Vedang Murthy, MDhh, Jonathan I. Epstein, MD,
Prof. ii, Christian Graztke, MD, Prof. jj, Xin Gao, MD kk, Anca L. Grosu, MD, Prof. a,b,
Sophia C. Kamran, MD ll,mm *, Constantinos Zamboglou, MD a,b,c,nn*
(a) Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine. University of
Freiburg, Freiburg, Germany
(b) German Cancer Consortium (DKTK). Partner Site Freiburg, Freiburg, Germany
(c) Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
(d) Department of Radiation Oncology, University Hospitals Leuven, Belgium; Department of Oncology, KU
Leuven, Belgium
(e) Department of Radiation Oncology; Department of Urology, University of California, Los Angeles, Los
Angeles, CA, USA
(f) Department of Radiation Oncology, UH Seidman Cancer Center, Case Western Reserve University, USA
(g) Department of Urology, Northwestern Feinberg School of Medicine, USA
(h) Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
(i) Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
(j) Department of Urology, Koc University Hospital, Istanbul, Turkey
(k) Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto; Radiation
Medicine Program, Princess Margaret Cancer Centre, University Health Network. Toronto, Canada.
(l) Department of Radiation Oncology, Gustave Roussy, Oncostat U1018, Inserm, Paris-Saclay University,
Villejuif, France
(m) Department of Radiation Medicine, Medstar Georgetown University Hospital, Washington, DC USA
(n) Institute for Surgical Pathology, University Medical Center Freiburg, Faculty of Medicine. University of
Freiburg,Freiburg, Germany
(o) Department of Radiation Oncology, University of Kansas Cancer Center, Kansas City, KS 66160, USA.
(p) Department of Radiation Oncology, University of Miami, Miller School of Medicine, USA
(q) Northern Sydney Cancer Centre, Radiation Oncology Unit, Royal North Shore Hospital, Sydney, New
South Wales, Australia.
(r) Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl
Gustav Carus, Technische Universit Dresden, Dresden, Germany
(s) Xcare Practices Dept. Radiotherapy, Saarlouis, Germany
(t) Department of Radiation Oncology, Chari Universitätsmedizin Berlin, corporate member of Freie
Universität Berlin and Humboldt-Universit zu Berlin
(u) Veracyte, Inc, San Diego, CA, USA
(v) Department of Urology, Rechts der Isar Medical Center, Technical University of Munich, Germany
(w) Department of Radiation Oncology, Radboud University Medical Center, The Netherlands
(x) Department of Radiation Oncology, University Hospital Johann Wolfgang Goethe University, Frankfurt,
Germany.
(y) Department of Radiation Oncology, University of Maryland, USA
(z) Department of Radiation Oncology, MediClin Robert Janker Klinik Bonn, Germany
(aa) Department of Radiation Oncology, Centre Léon Bérard, Lyon, France.
(bb) Molecular Medicine Research Center and Laboratory of Molecular and Medical Genetics, Department
of Biological Sciences, University of Cyprus, Nicosia, Cyprus.
(cc) Department of Radiation Oncology, University Hospital LMU Munich, Munich, Germany.
(dd) Department of Radiation Oncology, University Hospital Ulm, Ulm, Germany
(ee) Department of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland
(ff) Department of Radiotherapy, The Royal Marsden Hospital and the Institute of Cancer Research, London,
UK
2
(gg) Department of Urology, Medical School of Nanjing University, Affiliated Drum Tower Hospital, Nanjing,
China
(hh) Department of Radiation Oncology, ACTREC, Tata Memorial Centre, Homi Bhabha National University,
India
(ii) Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
(jj) Department of Urology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg,
Freiburg, Germany
(kk) Department of Internal Medicine, Division of Hematology and Oncology, Massachusetts General
Hospital, Boston, MA, USA.
(ll) Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston,
MA
(mm) Broad Institute of MIT and Harvard, Cambridge, MA
(nn) German Oncology Center, European University of Cyprus, Limassol, Cyprus
* Contributed equally
Corresponding author
Simon K.B. Spohn
Robert-Koch-Straße 3, 79106 Freiburg, Germany
Tel: +49 761 270 94610; Fax: +49 761 270 94720
Simon.Spohn@uniklinik-freiburg.de
Word count: 3907
Author contribution
Conceptualization: SKBS, SCK and CZ; Methodology: SKBS and CZ; Resources:
SKBS, CD and CZ, Investigation: CD. (Leuven), AUK, DS, AR, RM, DT, AB, PB
(Paris), SC, PB (Freiburg), RC, ADP, GM, TE, KH, TH, SH, PG, ED, MH, LGWK, SK,
NT, PTT, MP, PP, NS, TW, TZ, ACT, XQ, VM, JIE; Data curation: SKBS, SCK and
CZ, Formal analysis: SKBS SCK and CZ, Supervision: CD (Cyprus), CG, XG, ALG;
Writing-Original draft: SKBS, SCK and CZ; Writing Review & Editing: All authors
Author responsible for statistical analysis
Simon Spohn
Email: Simon.Spohn@uniklinik-freiburg.de
Disclosures
A.U.K. reports funding support from grant P50CA09213 from the Prostate Cancer
National Institutes of Health Specialized Programs of Research Excellence and grant
W81XWH-22-1-0044 from the Department of Defense, as well as grant RSD1836
from the Radiological Society of North America, the STOP Cancer organization, the
Jonsson Comprehensive Cancer Center, and the Prostate Cancer Foundation.
C.Z. received founding from the Klaus Tschira foundation, Naslund Medical and from
the German research foundation. CZ received speaker fees from Johnson and
Johnson and from Novocure, all outside the submitted work.
E.D. is an employee of Veracyte, manufacturer of Decipher.
A.R. is a consultant and / or speaker for astellas, bayer, pfizer, blue earth, lantheus,
janssen, tempus, veracyte
X.G. is in the consulting/advisory board for Bayer, Myovant, Guardant Health.
Funding
This study is supported by the German Federal Ministry of Education and Research
(BMBF) as part of the ERA PER Joint Funding Call 2019.
Grant No.: Med-Call / JTC2019-299 01KU2015
Role of the funder
The funders had no role in study design, data collection and analysis, decision to
publish, or preparation of the manuscript
3
Data Availability Statement
The results of the systematic review are included in the manuscript. All data
generated during the DELPHI consensus are included in the manuscript and
supplementary material. The corresponding author is available for any further
questions on the data.
Acknowledgements
AT acknowledges support from Cancer Research UK (C33589/A28284 and
C7224/A28724 CRUK RadNet). This project represents independent research
supported by the National Institute for Health research (NIHR) Biomedical Research
Centre at The Royal Marsden NHS Foundation Trust and the Institute of Cancer
Research, London. The views expressed are those of the authors and not
necessarily those of the NIHR or the Department of Health and Social Care.
We thank Fotini Miltiadous for the administrative support.
Abstract
Background:
Current risk-stratification systems for prostate cancer (PCa) do not sufficiently reflect
the disease heterogeneity. Genomic classifiers (GC) enable improved risk-
stratification after surgery, but less data exists for patients treated with definitive
radiotherapy (RT) or RT in oligo-/metastatic disease stages. In order to guide future
perspectives of GCs for RT, we conducted (i) a systematic review on the evidence of
GCs for patients treated with RT and (ii) a survey of experts using the DELPHI
method, addressing the role of GCs in personalized treatments to identify relevant
fields of future clinical and translational research.
Methods:
We performed a systematic review and screened ongoing clinical trials on
“clinicaltrials.gov”. Based on these results a multidisciplinary international team of
experts received an adapted DELPHI method survey. 31 and 30 experts answered
round 1 and round 2, respectively. Questions with 75% agreement were considered
as relevant and included into the qualitative synthesis.
4
Results:
Evidence for GCs as predictive biomarkers is mainly available to the postoperative
RT setting. Validation of GCs as prognostic markers in the definitive RT settings is
emerging. Experts used GCs in PCa patients with extensive metastases (30%), in
postoperative settings (27%) and newly diagnosed PCa (23%). 47% of experts do not
currently use GCs in clinical practice. Expert consensus demonstrates that GCs are
promising tools to improve risk-stratification in primary and oligo-/metastatic patients
in addition to existing classifications. Experts were convinced that GCs might guide
treatment decisions in terms of RT-field definition and intensification/de-intensification
in various disease stages.
Conclusions:
This work confirms the value of GCs and the promising evidence of GC utility in the
setting of RT. Additional studies of GCs as prognostic biomarkers are anticipated and
form the basis for future studies addressing predictive capabilities of GCs to optimize
RT and systemic therapy. The expert consensus points out future directions for GC
research in the management of PCa.
5
1. Introduction
Prostate cancer (PCa) is the second most common malignancy in men worldwide [1].
Improvements in screening and diagnostics have led to an increased number of
patients diagnosed with all stages of PCa [2, 3]. Radiotherapy (RT) plays a central
role in the management of PCa patients and can be applied in a curative setting or as
part of a palliative treatment concept. However, current risk stratification systems are
imperfect the use of novel prognostic or predictive biomarkers are urgently needed
to enable better patient selection for appropriate treatment in the future. Several
biomarkers evaluating blood, urine, or tissue have been developed to aid with risk
stratification. Genomic classifiers (GC), or mRNA-based gene expression profiles
from tissue, have shown promise to reliably enable identification of aggressive PCa
and guide treatment decisions with different commercially available profiling panels,
including Prolaris, Oncotype DX and Decipher for overview see [4, 5]). Additionally,
the PAM50 classifier has been demonstrated to differentiate between luminal and
basal PCa, with luminal B tumours being associated with favorable response to
postoperative ADT [6].
Most evidence is available for the Decipher GC after radical prostatectomy (RP),
improving risk stratification and consequently guiding postoperative disease
management [7]. Less data exists for patients treated with definitive RT or RT in the
(oligo)metastatic setting, but GC might facilitate personalized oncologic treatments in
various perspectives in all disease stages. The aim of this work is to (i) summarize
the role of GCs for PCa patients in all disease stages treated with RT, since this is
aspect has not previously been highlighted and (ii) point out relevant clinical and
translational issues for future fields of research. We therefore conducted (i) a
systematic review and (ii) a survey of experts and key opinion leaders using the
DELPHI method. See Figure 1 for a summarizing overview.
6
2. Methodology
2.1 Systematic Review
Studies eligible for inclusion were original articles on GC in PCa in the setting of RT,
comprising primary definitive, post-operative, as well as metastasis-directed (MDT)
RT. In general, 3 types of manuscript were included: (A) manuscripts on oncologic
outcomes after RT, (B) the role of GCs in RT decision process and (C) correlation
studies between other biomarkers and GCs. Inclusion criteria were: A1) patients
treated with RT (definitive, postoperative, MDT); A2) clinical results with the following
endpoints: clinical recurrence (CR), biochemical recurrence (BR), distant metastases
(DM), prostate-cancer specific mortality (PCSM), overall survival (OS). A3) Articles
with retrospective and prospective data were allowed (n patients > 50); B1)
Translational work addressing correlation of GCs with imaging, biomarkers or
biological features (radioresistance, androgen signaling etc); C1) Impact of GCs in
RT treatment decision processes. Exclusion criteria were: 1) articles not written in
English 2) non-original articles. SKBS and CZ performed a PubMed/Medline,
EMBASE and Cochrane Library database search for the terms:
PubMed:
(prostatic neoplasms [MeSH Terms6) or (prostatic neoplas*[tiab OR prostate
neoplas*[tiab] OR prostatic cancer*[tiab] OR prostate cancer*[tiab] OR prostatic
carcinoma*[tiab] OR prostate carcinoma*[tiab] OR prostatic adenocarcinoma*[tiab]
OR prostate adenocarcinoma*[tiab] OR prostatic tumor*[tiab] OR prostate
tumor*[tiab] OR prostatic tumour*[tiab] OR prostate tumour*[tiab]) AND
(Radiotherapy"[8]) or (radiotherapy [Subheading]) or (radiotherap*[tiab] OR
radiati*[tiab] OR irradiati*[tiab] OR "x ray therapy" [tiab] OR "x ray therapies" [tiab] OR
radioimmunotherap*[tiab] OR immunoradiotherap*[tiab]) AND (genomic* classif* [tiab]
OR decipher* [tiab])
7
EMBASE and Cochrane:
('prostate cancer'/exp OR 'prostate cancer') AND ('radiotherapy'/exp OR
'radiotherapy') AND ('genomic classifier'/exp OR 'genomic classifier')
Mapped terms ''genomic classifier'' mapped to 'genomic classifier'.
In case of discrepant findings (n=3), a third reviewer (CeDr) provided a final decision.
The time period considered in this review was from June 6th 2013 until December 1st
2021. One hundred twenty-six articles were identified and 32 duplicates removed.
After applying inclusion and exclusion criteria, 26 studies were included for qualitative
review according to PRISMA [9] (Figure 2). This version was sent to the experts for
the first round of the survey. Between round 1 and 2 of the survey, a second round of
literature research was performed considering articles until the December 31st 2021.
No additional studies matching in- and exclusion criteria were found. During the peer-
review process an update of the literature search was performed:5 more studies and
one more clinical trial were included by considering a time period from June 6th 2013
until December 1st 2022.
2.2 Ongoing clinical trials
In order to provide an overview of clinical trials implementing GCs in treatment
decision, which serves to classify results of the expert survey, ongoing clinical trials
were screened on “clinicaltrials.gov”. Studies needed to be ongoing trials on GCs in
PCa in the setting of RT. SKBS performed the search for the terms (“Condition or
disease: prostate cancer” AND genomic classifier” OR “radiotherapy”). Five clinical
trials on GCs in RT were located.
2.3 Expert opinion
The multidisciplinary team of expert professionals included radiation oncologists,
urological oncologists and pathologists. Experts were characterized by long-time
8
experience in care and/or clinical trials of PCa patients, scientific research and their
role as key opinion leaders. An adapted DELPHI method was used to identify the
most relevant questions for future perspectives of GC. Since predictive biomarkers
are ultimately warranted to guide personalized treatments, we focused on the
putative capability of GCs to identity patients who might benefit from a certain
treatment. In round 1 (R1) preliminary results of literature search and key questions
were prepared by SKBS and CZ and emailed to 46 PCa experts, receiving 27 replies.
The survey was designed using the online tool Surveymonkey. Based on the
recommendations of the participants, we sent additional 9 invitations, of which 4
replied. In total 31 experts answered R1 (response rate: 56%). After completion of
R1, SKBS and CZ consolidated questionnaires and prepared round 2 (R2), in which
participant’s feedback and questions that did not reach consensus (defined as 50-
75% of votes) were included. These results were prepared by SKBS and CZ and
distributed to all participants (n=31). 30 experts provided answers in R2 (response
rate: 97%). Finally, only questions with ≥75% agreement were considered as relevant
and included into the qualitative synthesis. The detailed results of the adapted Delphi
rounds can be found in the supplementary information.
3. Results
3.1 GC in the Literature methodological aspects
Literature search revealed 31 original papers addressing GCs in the setting of RT
(see Table 1 for details). Most of the studies (n=26) included retrospectively collected
patient collectives whilst 5 studies analyzed GC in prospectively collected patient
cohorts. Only two studies performed an external validation [11][36]. With 20 (65%)
studies, the Decipher test was used in the vast majority [10-29].
In 24 (77%) studies RP specimens were used to obtain tissue for genomic analyses
[10-24, 26, 28, 30-36]. In one study (3%) the Decipher test was applied to both RP-
9
and biopsy-specimens [26], whilst six studies (19%) solely used biopsy specimens for
further analyses [27-29, 37-39]. Most studies (n=18, 58%) investigated the
associations between GCs and oncological outcomes such as DM (n=12, 39%) [12-
14, 26-32, 38, 39], prostate cancer specific mortality (PCSM) (n=3, 10%) [14, 28, 38],
overall survival (OS) (n=1, 3%)[14], clinical recurrence (CR) (n=3, 10%) [10, 11, 15]
and biochemical recurrence (BR) (n=6, 19%) [13, 15, 27, 33, 37, 38]. The other
studies reported on the role of GCs on treatment decision making (n=9, 29%) [16-24]
or correlated GCs with other biomarkers (n=4, 13%) [25, 34-36].
All studies included in this review reported on the role of GC as prognostic
biomarkers for PCa patients. To correctly assess the predictive value of a biomarker
in a study, at least two comparison groups must be available (in the best case, two
treatment arms in a RCT) [40]. This pre-requirement was not fulfilled by any study in
this review. However, five studies suggested a predictive role for GC in the setting of
adjuvant RT after surgery and one study supposed a predictive role of GC in the
response to androgen deprivation therapy (ADT) in the definitive RT setting [11-13,
30, 31, 39].
3.2 GC in the Literature GCs for outcome prediction in RT for primary
localized PCa
In total seven studies included 1551 patients treated with definitive RT [26-29, 37-39].
Tosoian et al. performed a retrospective analysis of 405 men with high-risk PCa, of
which 80 were treated with definitive RT +/- ADT. A subset analysis showed that GC
was an independent prognosticator for patients treated with RT (HR 1.61, 95%CI
1.082.40,) [26]. Berlin et al. analyzed 121 patients with National Comprehensive
Cancer Network (NCCN) intermediate-risk PCa treated with definitive RT without
ADT. The GC outperformed all other indices in prediction of DM (HR 2.05, 95%CI
10
1.24 4.24) [27]. Nguyen et al. investigated retrospectively the Decipher biopsy test
as a prognosticator for DM and PCSM in intermediate- and high-risk patients treated
with RP or RT +/- ADT, respectively. In the mixed cohort the GC test was a
significant predictor for DM (HR 1.37 per 0.1 score increase, 95% CI: 1.061.78) and
PCSM (HR 1.57 per 0.1 score increase, 95%CI: 1.032.48) [28]. Another study by
Nguyen et al. included patients with intermediate- and high-risk PCa. Each GC score
increase was a significant predictor for DM in multivariate analysis (HR 1.36, 95%CI:
1.041.83). Furthermore, patients with a GC>0.6 (high-risk) had a 20% cumulative
incidence of metastasis at 5 years after RT, whereas patients with a low-risk GC
score of 0.2 had 0% cumulative incidence [29]. Tward et al. showed that a clinical
cell-cycle risk score (CAPRA score + Prolaris GC) prognosticated DM with a HR per
unit score of 2.22 (95%CI: 1.71-2.89) after dose-escalated RT +/- ADT in
intermediate- and high-risk patients [39]. Additionally, the authors suggested a
multimodality threshold defining men in which adding ADT may not significantly
reduce their risk of DM. Freedland et al. included patients with low-, intermediate-
and high-risk PCa and evaluated the prognostic utility of the Polaris score for BR
after primary RT +/- ADT. In the multivariate analysis the GC was a significant
predictor for BR (HR 2.11, 95%CI: 1.05-4.25) [37]. Comparable results in a similar
collective were observed by the study from Janes et al. by also considering the
endpoints DM (HR 4.28, 95%CI:2.43 7.75) and PCSM (HR 6.11, 95%CI:2.93
14.33) [38].
3.3 GC in the Literature GCs for outcome prediction in RT for postoperative
PCa
In total 10 studies (ART or SRT: n=7, SRT: n=3) with 9792 patients evaluated the
role of GC in the postoperative RT setting [10-15, 30-33]. Dalela et al. [11] proposed
11
a nomogram for the prediction of clinical progression in the postoperative RT setting.
By including the Decipher score in the model a C-index of 0.85 was obtained. Lee et
al. observed a C-Index of 0.84 in an external validation of this model [10]. The group
by Den et al. evaluated the prognostic role of the Decipher score for DM prediction in
the postoperative setting and observed a HR of 1.61 (95%CI: 1.2 2.15) and of 0.78
(95%CI: 0.64 0.91) in multicentric and monocentric retrospective cohorts,
respectively [12, 13]. Both studies suggested that patients with low GC scores are
best treated with SRT, whereas those with high GC scores benefit from ART. Similar
results for DM prediction after postoperative RT were observed by other studies
incorporating PORTOS [31], the genomic expression of stromal infiltration markers
[30] and the clinical genomic risk [32]. Feng et al. showed a significant impact of the
Decipher score on DM (HR, 1.17 95%CI: 1.05 1.32) and PCSM (HR 1.39,
95%CI1.20 1.63) in the prospective RTOG 9601 trial cohort treated with SRT +/-
ADT [14]. Dal Pra et al. examined the prognostic impact of the Decipher score in the
SAKK09/10 study collective which was treated with SRT for recurrent PCa after
surgery and observed a HR of 2.21 (95%CI: 1.41 3.47) for BR [15].
3.4 GC in the Literature GCs for outcome prediction in RT for oligometastatic
PCa
Deek et al. analyzed the impact of genetic features on outcomes in a pooled cohort
of the STOMP and ORIOLE trial [41]. Patients without a high-risk mutational status
experienced favorable progression-free survival rates (HR 0.57, 95%CI: 0.32 1.03).
The authors observed a potential larger benefit for MDT in patients with high-risk
mutations.
3.5 GC in the Literature GCs for treatment decision making
12
Eight studies evaluated the effect of the 22-gene Decipher score on postoperative
treatment decision making and showed that high GC risk scores were associated
with intensification of treatment in terms of admission of ADT, RT dose and
expansion of RT fields, independent from clinicopathological factors [16-23]. Five
studies assessed treatment recommendations before and after addition of GC score
information in patients treated with RP and adverse pathological features such as
pT3 stage and positive margins [16-20]. Post-Decipher recommendations changed in
up to 40% [20], the number needed to test for a change in recommendation varied
between 3 and 4 [16, 17]. Badani et al. showed similar results in a retrospective
cohort of low-, intermediate- and high-risk patients according to D’Amico risk
classification [21]. Furthermore, implementation of GC testing and its results in
clinical practice decreased cancer specific anxiety [16, 18] and decisional conflict
scores [18, 19]. Nguyen et al. assessed the treatment recommendations from 20 US
board certified urologists and 26 radiation oncologists with high rates of
recommendation change, identifying the GC risk score as the strongest influencing
factor [22]. Lobo et al. developed a Markov Model for decision of postoperative
treatment decision [23] and for cost effectiveness, which demonstrated improved cost
effectiveness and quality adjusted life years (QALYs) [24].
3.6 GC in the Literature GCs in correlation with other biomarkers
In addition to the 22-gene Decipher score, the Genomics Resource for Intelligent
Discovery (GRID) database provides comprehensive transcriptomic profiles and thus
enables additional genomic studies. Ben-Salem et al. analyzed androgen receptor
(AR) target genes in the GRID database and validated their results in smaller cohorts
and could identify a baseline heterogeneity in AR action and found that specific up-
or downregulation of AR genes was associated with treatment response prediction
13
[34]. Two studies additionally assessed the immune content score (ICS) derived from
immune cell-specific genes [25, 35]. Yamoah et al. showed that patients with high
Decipher GC and ICS scores have a higher risk of DM and PCSM and are
associated with genes correlated to radiosensitivity [25]. Awashti et al. assessed
difference in immune specific genes between African-American (AAM) and
European-American (EAM) patients and identified that PCa of AAM patients exhibit
higher ICS scores, lower DNA damage repair and higher radiosensitivity [35].
3.7 Ongoing prospective clinical trials
In total nine studies were identified via “clinicaltrials.gov incorporating GC and RT
treatment. Additionally, five studies were included based on the recommendations of
the authors of this work. Eight and one studies incorporate GCs in the primary and
salvage PCa setting, respectively. See Table 2 and supplementary material for
synthesis of ongoing clinical trials.
3.8 Survey
Thirty experts answered R2 of the modified DELPHI survey. Half of the participating
experts reported on using GCs in clinical practice, mostly in extensive metastatic
disease (30%) and postoperative settings (27%). See Figure 3 for details.
Please see Figure 4 and Table 3 for the detailed results of the expert survey
concerning the clinical and research setting, respectively. Considering primary PCa
patients, the majority (97%) of experts were convinced that GCs could be
implemented as a dedicated feature into PCa risk group stratification systems in the
future. Consensus was reached, that additional tools for risk stratifications are
needed across NCCN risk groups (low/favorable intermediate-risk: 83%; unfavorable
intermediate-risk: 100%; high-risk: 100%) and that GCs are likely to be useful tool in
this setting (low/favorable intermediate-risk: 83%; unfavorable intermediate-risk: 90%;
14
high-risk: 93%). Experts were convinced that GCs might be applied as a predictive
biomarker and to determine optimal treatments across various risk groups, including
administration and duration of ADT, intensification of systemic therapies or addition of
radiation to elective pelvic nodes.
Considering metastatic disease, 100% of experts agreed that additional tools for
improved risk stratification are needed and 76% believed that GCs might be a useful
tool in this scenario. Relevant scenarios identified by experts were administration of
MDT and the combination of MDT and systemic therapies.
Considering the postoperative setting, 97% of experts agreed that additional tools for
improved risk stratification are needed and 89% believed that GCs might be a useful
tool in this scenario. Relevant questions identified by experts were administration of
adjuvant vs early-salvage RT, administration and duration of ADT and additional
pelvic irradiation in salvage RT.
MFS was considered as the most appropriate endpoint to evaluate the role of GCs in
clinical studies for non-metastasized PCa. Consensus that GCs might be relevant in
translational research fields was reached, in strategies to cope with intertumoral
heterogeneity (between the primary and metastatic lesions), alteration in androgen
receptor signaling and decision making for physicians.
Discussion
This work incorporated a systematic review and a modified Delphi survey to assess
the role of GC in PCa RT and to define future directions. Despite the fact that 47% of
the participants of the survey do not use GC in clinical routine, the vast majority
agreed that GC should be incorporated in RT strategies in all PCa disease stages in
the future. Differences in clinical utilization of GC is explainable by regional
differences in the distribution of facilities capable to perform GC tests and
15
reimbursement issues. However, our work shows, that the potential clinical utility is of
GCs is expected to be relevant. In line with a previous systemic review and meta-
analysis [7], our current synthesis shows that the Decipher test is the most commonly
utilized GC in PCa patients and the highest level of evidence for the Decipher GC
exists in the setting of risk stratification, outcome prediction and treatment guidance
after RP [7].
Prognostic biomarkers are helpful tools to identify patients who are at high or low risk
of recurrence and thus are candidates for treatment intensification or de-
intensification. Predictive biomarkers are warranted in order to truly guide
personalized treatments. All studies demonstrate the prognostic value of the GCs,
but due to the methodological design of the studies, no clear conclusions regarding
the predictive value of GCs can be drawn. None of the studies assessed the
predictive capacity of a GC within dedicated treatment arm of a RCT. Additionally,
only two of the studies included external validation cohorts, which underlines the
need of more high quality studies. Therefore, the results of the expert consensus may
help to guide clinical decision making, the design of future clinical trials and
translational research. Fortunately, some of the presented clinical questions are
addressed by currently ongoing clinical trials. However, we want to mention, that
these studies are designed to evaluate the prognostic capability of GCs in these new
clinical scenarios, which might form the basis for future studies addressing the
predictive capabilities. Despite the absence of prospective and externally validated
studies addressing predictive values of GC in patients treated with definitive RT, we
could identify seven studies including in total 1551 patients in which GCs were
analyzed as a prognosticator of DM and BR after definitive RT [26, 27, 29, 37-39].
See Figure 5 for details. We did not include the study by Ngyuen et al. in Figure 4,
since no individual data on the prognostic value of GCs in the cohort of patients
16
treated only with RT are presented [28]. This moderate sample size contrasts the
larger number of GC analyses in patients treated with RP (n=9792), but
demonstrates similar results with the Decipher GC score being an independent
prognosticator after both treatments. Furthermore, additional information will be
provided by forthcoming studies, validating the Decipher GC in phase III studies,
such as the NRG/RTOG 9202, 9314, 9902, 0126 and STAMPEDE trials, which have
been presented at the ASTRO 2021, ASCO GU 2022 and ESMO 2022 annual
congresses [42-44]. Thus, performing GC tests on biopsy cores yields promising
results to predict PCa aggressiveness suggesting implementation in risk and
treatment stratification after definitive RT. Confirming these results, expert consensus
was reached that GCs are a promising tool to improve PCa risk stratification in the
primary PCa setting. However, we demonstrate a lack of data for RT, pointing out the
need for additional studies, including validation of GCs in patient cohorts staged with
state-of-the-art imaging, such as PSMA-PET and multiparametric magnetic
resonance imaging (MRI), and treated with modern radiation approaches, including
stereotactic body radiotherapy (SBRT) or brachytherapy (BT). Future studies should
clarify whether the applied biopsy method (MRI-fusion vs. MRI-guided vs. no
implementation of MRI information) influences the GC results.
Extrapolating the results of Tward et al. [39] Feng et al. [14] and Ben-Salem et al.
[34], GC scores might help to stratify patients that benefit the most from concomitant
ADT, define the optimal duration of ADT, as well as identify those that may benefit
from systemic treatment intensification in definitive RT settings. Consequently,
consensus was reached by experts, that GCs are promising tools to improve
recommendations for administration of systemic therapies in the primary setting
across all NCCN risk groups including duration of ADT and addition of new hormonal
17
agents, particularly in high-risk patients. Furthermore, alterations in androgen
receptor signaling were considered to be a relevant translational research field.
Additional predicative markers are urgently needed addressing this vague clinical
issue in order to guide personalized treatment decisions. Four ongoing prospective
trials will contribute to this field of research by stratifying patients according to
genomic risk groups into intensification or de-intensification of systemic treatments
and thus optimize therapies based on GCs.
Treatment intensification in the setting of definitive RT can also be achieved by dose
escalation in all primary PCa risk stages (delivered dose to the tumor >80-90 Gy,
EQD2 α=1.6 Gy). Dose escalation improves BR-free survival and can be performed
via BT [45] or focal boosting [46]. Kishan et al. analyzed the genomic heterogeneity
of patients with Grade group 4-5 who underwent prostatectomy within the GRID
database and could identify four distinct clusters, of which one was enriched with
genes related to cell cycle and proliferation and was associated with worse DM-free
survival [47]. Furthermore, GRID analysis revealed PCa subtypes with increased
genomic radiosensitivitiy [25, 35], therefore GC scores might be utilized to identify
patients with radioresistent or radiosensitive PCa and thus guide treatment decision
in terms of (focal) dose escalation and the optimal dose in the primary setting.
However, these aspects were not considered relevant by experts, with approximately
50% agreement that GCs might be useful to identify patients who benefit from focal
dose escalation due to increased radio-resistance. On the basis of a high
fractionation sensitivity of PCa [48], moderately hypofractionated RT (MHRT) [49, 50]
and ultra-hypofractionated RT or SBRT [51, 52] have been analyzed in randomized
controlled trials and were demonstrated to have comparable relapse rates to
conventionally fractionated RT. The consideration that genomic alterations might
influence the individual α/β-value [48] and that GCs might thus be used to predict
18
whether a specific fractionation scheme is beneficial, was not considered to be
relevant by experts (≤37% agreement across NCCN risk groups). Nevertheless,
future research might provide new insights into the heterogeneity of PCa and the
linkage between the genomic signatures and radiation- and fractionation sensitivity.
For example, Dal Pra et al. reported in a congress abstract that Patients with high
PORTOS score that received 70Gy SRT dose to the fossa had better 5-year clinical
progress-free survival (94% vs. 49%, p=0.006) compared to patients that received
the 64Gy dose [53].
GCs might help to improve risk stratification for treatment escalation or de-escalation
in terms of RT to pelvic lymphatics. Prophylactic whole pelvis radiation has recently
been shown to improve BR free survival at the cost of late genitourinary toxicities
[54], thus improved patient selection to prevent overtreatment is needed. Currently, a
Phase II study adapts RT fields based on GC and includes pelvic lymphatics only in
GC high-risk patients (NCT05169970). Consensus was reached that GCs might be a
useful predictive marker to identify high-risk PCa patients who benefit the most from
elective pelvic irradiation and alleviate this controversial discussion.
Incorporating recent improvements in diagnostics, in particular PSMA-PET, might
further facilitate treatment personalization [55, 56]. Since PSMA-PET was used in
none of the identified studies, there are many open questions to what extent GC and
advanced imaging methods give complementary or redundant information addressing
outcome prediction or decision management. Expert’s answers were inconsistent
with 45% agreeing that image features might be utilized to predict GC scores in R1
and 61% agreeing that GC might be useful to predict imaging results in R2. Only one
trial analyzed the ability of GC scores to predict PET-positive extraprostatic lesions
and found a significant association with pelvic nodal disease [57]. Interestingly,
Hectors et al. extracted imaging features from multiparametric magnetic resonance
19
tomography and developed a machine-learning model, which excellently predicted a
Decipher score of 0.60 (AUC=0.84). These promising results should be validated in
larger patient cohorts. Considering the possible capability to depict intratumoral
molecular characteristics on PSMA-PET [58, 59], this imaging method should be
included for future genomic-imaging correlations.
Combination of genomic signatures and PSMA-PET-based staging of tumor
localization could be of particular interest in oligometastatic and oligorecurrent
disease stages, enabling identification of patients who benefit from metastasis- [60,
61] or primary- directed therapies [62]. Stopsack et al. reported on specific genomic
features associated with poor survival in metastatic PCa, which might be used to
intensify systemic therapies or develop targeted therapies [63]. The pooled analysis
of the STOMP and ORIOLOE trial links outcome after MDT to mutational burden in
oligometastatic PCa patients, encouraging that further research will possibly add
value to define patients with genomiclow- and high metastatic burden and guide
treatment in these stages. Consequently, consensus was reached that GCs might be
helpful as a predictive factor for progression-free survival/ prostate cancer specific
survival after MDT or systemic therapies or the combination of both in
oligometastatic, oligorecurrent or oligoprogressive patients. In contrary,
implementation of GC as a predictor for primary-directed therapies was not
considered relevant in this setting. Additionally, experts agreed that strategies to
cope with intertumoral heterogeneity between primary tumours and metastases
should be assessed in future.
A factor that should not be underestimated is decisional conflict and patient’s anxiety.
Luckily, patients with PCa have multiple treatment options in the primary,
postoperative and metastatic setting [64, 65]. Likely, the implementation of GCs as a
tool to guide treatment decision will not only be beneficial in patients aiming for RP
20
[16-22] but also for RT concepts, an opinion that is confirmed by the expert
consensus.
Further clinical trials are needed to tackle the scarcity of studies addressing the
predictive value of GC in RT and therefore accurate definition of study endpoints is
warranted. Experts reached consensus that the validated surrogate parameter MFS
is an appropriate endpoint across all NCCN risk groups. However, future research
will assess the role of MFS a surrogate end point in the era of molecular imaging [66].
We want to highlight, that most of the available prospective data on GCs still only
exists for the Decipher GC. The PORTOS signature complies with high
methodological standards since a statistical analysis of treatment interaction and an
external validation was performed in the study by Zhao et al. [31].
However, due to the lack of prospective data, external validation and its benefit on
long-term outcomes GCs are still not recommended for routine use [67].
Nevertheless, most of the presented studies included multicenter cohorts, partly from
RCTs, and potentially some of the ongoing studies will provide data to support
broader use of these assays to enable improved treatments for PCa patients.
We want to acknowledge the limitations of this work. Due to a lack in evidence for the
role of GC in the primary PCa RT setting, the discussion is mainly based on
extrapolation from data obtained from RP cohorts. However, we brought together an
internationally-recognized expert panel to optimize conclusions and to highlight future
directions in GC research for RT patients. Finally, it is important to mention that
implementation of GC in PCa is a fast-moving field and conclusions will need to be
iterated in light of rapidly evolving evidence. For example, several studies reported
their results in current congresses analyzing the role of GC in the context of primary-
definitive RT [43] or salvage RT [53] .
21
In summary, this work confirms the value of GCs and, in particular, of the Decipher
GC as a prognostic biomarker in patients undergoing RP and its predictive value for
postoperative RT. Additionally, we summarize the scarce, but promising evidence
that GCs might be equivalently useful in the setting of definitive RT. Nevertheless we
highlight that GCs currently do not comply with its great potential to function as
predictive markers and thus guide personalized treatment decisions. In this regard,
we await the highly anticipated prospective clinical trials, which will further inform the
role of GCs in the setting of RT and present an expert consensus, which can help to
design studies capable to validate GCs as predictive biomarkers and thus ultimately
guide personalized treatments. The authors want to emphasize that the development
and establishment of tumor biomarkers for PCa patients is complex. Thus, a
dedicated system for biomarker study design, conduct, analysis, and evaluation that
incorporates a hierarchal level of evidence should be applied. The presented expert
consensus might help to guide future research perspectives.
22
Data availability
The results of the systematic review are included in the manuscript. All data
generated during the DELPHI consensus are included in the manuscript and
supplementary material. The corresponding author is available for any further
questions on the data.
23
References
1. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of
Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71(3):209-
249.
2. Wenzel M, Würnschimmel C, Ruvolo CC, et al. Increasing rates of NCCN high and very high-
risk prostate cancer versus number of prostate biopsy cores. The Prostate 2021;81(12):874-881.
3. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin 2022;72(1):7-33.
4. Loeb S, Ross AE. Genomic testing for localized prostate cancer: where do we go from here?
Current opinion in urology 2017;27(5):495-499.
5. Na R, Wu Y, Ding Q, et al. Clinically available RNA profiling tests of prostate tumors: utility
and comparison. Asian journal of andrology 2016;18(4):575-579.
6. Zhao SG, Chang SL, Erho N, et al. Associations of Luminal and Basal Subtyping of Prostate
Cancer With Prognosis and Response to Androgen Deprivation Therapy. JAMA Oncology
2017;3(12):1663-1672.
7. Jairath NK, Dal Pra A, Vince R, et al. A Systematic Review of the Evidence for the Decipher
Genomic Classifier in Prostate Cancer. European Urology 2021;79(3):374-383.
8. Cuzick J, Berney DM, Fisher G, et al. Prognostic value of a cell cycle progression signature for
prostate cancer death in a conservatively managed needle biopsy cohort. Br J Cancer
2012;106(6):1095-9.
9. Moher D, Liberati A, Tetzlaff J, et al. Preferred reporting items for systematic reviews and
meta-analyses: the PRISMA statement. Ann Intern Med 2009;151(4):264-9, w64.
10. Lee DI, Shahait M, Dalela D, et al. External validation of genomic classifier-based risk-
stratification tool to identify candidates for adjuvant radiation therapy in patients with prostate
cancer. World J Urol 2021; 10.1007/s00345-020-03540-1.
11. Dalela D, Santiago-Jiménez M, Yousefi K, et al. Genomic Classifier Augments the Role of
Pathological Features in Identifying Optimal Candidates for Adjuvant Radiation Therapy in Patients
With Prostate Cancer: Development and Internal Validation of a Multivariable Prognostic Model. J
Clin Oncol 2017;35(18):1982-1990.
12. Den RB, Yousefi K, Trabulsi EJ, et al. Genomic classifier identifies men with adverse pathology
after radical prostatectomy who benefit from adjuvant radiation therapy. J Clin Oncol
2015;33(8):944-51.
13. Den RB, Feng FY, Showalter TN, et al. Genomic prostate cancer classifier predicts biochemical
failure and metastases in patients after postoperative radiation therapy. Int J Radiat Oncol Biol Phys
2014;89(5):1038-1046.
14. Feng FY, Huang H-C, Spratt DE, et al. Validation of a 22-Gene Genomic Classifier in Patients
With Recurrent Prostate Cancer: An Ancillary Study of the NRG/RTOG 9601 Randomized Clinical Trial.
JAMA Oncology 2021;7(4):544-552.
15. Pra AD, Ghadjar P, Hayoz S, et al. Validation of the Decipher genomic classifier in patients
receiving salvage radiotherapy without hormone therapy after radical prostatectomy - an ancillary
study of the SAKK 09/10 randomized clinical trial. Ann Oncol 2022; 10.1016/j.annonc.2022.05.007.
16. Gore JL, du Plessis M, Zhang J, et al. Clinical Utility of a Genomic Classifier in Men Undergoing
Radical Prostatectomy: The PRO-IMPACT Trial. Pract Radiat Oncol 2020;10(2):e82-e90.
17. Marascio J, Spratt DE, Zhang J, et al. Prospective study to define the clinical utility and benefit
of Decipher testing in men following prostatectomy. Prostate Cancer Prostatic Dis 2020;23(2):295-
302.
18. Gore JL, du Plessis M, Santiago-Jiménez M, et al. Decipher test impacts decision making
among patients considering adjuvant and salvage treatment after radical prostatectomy: Interim
results from the Multicenter Prospective PRO-IMPACT study. Cancer 2017;123(15):2850-2859.
19. Michalopoulos SN, Kella N, Payne R, et al. Influence of a genomic classifier on post-operative
treatment decisions in high-risk prostate cancer patients: results from the PRO-ACT study. Curr Med
Res Opin 2014;30(8):1547-56.
24
20. Badani KK, Thompson DJ, Brown G, et al. Effect of a genomic classifier test on clinical practice
decisions for patients with high-risk prostate cancer after surgery. BJU Int 2015;115(3):419-29.
21. 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(4):600-9.
22. Nguyen PL, Shin H, Yousefi K, et al. Impact of a Genomic Classifier of Metastatic Risk on
Postprostatectomy Treatment Recommendations by Radiation Oncologists and Urologists. Urology
2015;86(1):35-40.
23. Lobo JM, Stukenborg GJ, Trifiletti DM, et al. Reconsidering adjuvant versus salvage radiation
therapy for prostate cancer in the genomics era. J Comp Eff Res 2016;5(4):375-82.
24. Lobo JM, Trifiletti DM, Sturz VN, et al. Cost-effectiveness of the Decipher Genomic Classifier
to Guide Individualized Decisions for Early Radiation Therapy After Prostatectomy for Prostate
Cancer. Clin Genitourin Cancer 2017;15(3):e299-e309.
25. Yamoah K, Awasthi S, Mahal BA, et al. Novel Transcriptomic Interactions Between Immune
Content and Genomic Classifier Predict Lethal Outcomes in High-grade Prostate Cancer. Eur Urol
2020; 10.1016/j.eururo.2020.11.038.
26. Tosoian JJ, Birer SR, Jeffrey Karnes R, et al. Performance of clinicopathologic models in men
with high risk localized prostate cancer: impact of a 22-gene genomic classifier. Prostate Cancer
Prostatic Dis 2020;23(4):646-653.
27. Berlin A, Murgic J, Hosni A, 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(1):84-91.
28. Nguyen PL, Haddad Z, Ross AE, et al. Ability of a Genomic Classifier to Predict Metastasis and
Prostate Cancer-specific Mortality after Radiation or Surgery based on Needle Biopsy Specimens. Eur
Urol 2017;72(5):845-852.
29. Nguyen PL, Martin NE, Choeurng V, et al. Utilization of biopsy-based genomic classifier to
predict distant metastasis after definitive radiation and short-course ADT for intermediate and high-
risk prostate cancer. Prostate Cancer Prostatic Dis 2017;20(2):186-192.
30. Mahal BA, Alshalalfa M, Zhao SG, et al. Genomic and clinical characterization of stromal
infiltration markers in prostate cancer. Cancer 2020;126(7):1407-1412.
31. Zhao SG, Chang SL, Spratt DE, et al. Development and validation of a 24-gene predictor of
response to postoperative radiotherapy in prostate cancer: a matched, retrospective analysis. Lancet
Oncol 2016;17(11):1612-1620.
32. Ross AE, Den RB, Yousefi K, et al. Efficacy of post-operative radiation in a prostatectomy
cohort adjusted for clinical and genomic risk. Prostate Cancer Prostatic Dis 2016;19(3):277-82.
33. Koch MO, Cho JS, Kaimakliotis HZ, et al. Use of the cell cycle progression (CCP) score for
predicting systemic disease and response to radiation of biochemical recurrence. Cancer Biomark
2016;17(1):83-8.
34. Ben-Salem S, Hu Q, Liu Y, et al. Diversity in Androgen Receptor Action Among Treatment-
naïve Prostate Cancers Is Reflected in Treatment Response Predictions and Molecular Subtypes. Eur
Urol Open Sci 2020;22:34-44.
35. Awasthi S, Berglund A, Abraham-Miranda J, et al. Comparative Genomics Reveals Distinct
Immune-oncologic Pathways in African American Men with Prostate Cancer. Clin Cancer Res
2021;27(1):320-329.
36. Mahal BA, Yang DD, Wang NQ, et al. Clinical and Genomic Characterization of Low-Prostate-
specific Antigen, High-grade Prostate Cancer. Eur Urol 2018;74(2):146-154.
37. Freedland SJ, Gerber L, Reid J, et al. Prognostic Utility of Cell Cycle Progression Score in Men
With Prostate Cancer After Primary External Beam Radiation Therapy. International Journal of
Radiation Oncology*Biology*Physics 2013;86(5):848-853.
38. Janes JL, Boyer MJ, Bennett JP, 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. International Journal of Radiation Oncology*Biology*Physics 2022;
https://doi.org/10.1016/j.ijrobp.2022.06.101.
25
39. Tward J, Lenz L, Flake DD, II, et al. The Clinical Cell-Cycle Risk (CCR) Score Is Associated With
Metastasis After Radiation Therapy and Provides Guidance on When to Forgo Combined Androgen
Deprivation Therapy With Dose-Escalated Radiation. Int J Radiat Oncol Biol Phys 2022;113(1):66-76.
40. Ballman KV. Biomarker: Predictive or Prognostic? Journal of Clinical Oncology
2015;33(33):3968-3971.
41. Deek MP, Eecken KVd, Sutera P, et al. Long-Term Outcomes and Genetic Predictors of
Response to Metastasis-Directed Therapy Versus Observation in Oligometastatic Prostate Cancer:
Analysis of STOMP and ORIOLE Trials. Journal of Clinical Oncology 2022;40(29):3377-3382.
42. Nguyen PL, Huang HC, Davicioni E, et al. Validation of a 22-gene Genomic Classifier in the
NRG Oncology/RTOG 9202, 9413 and 9902 Phase III Randomized Trials: A Biopsy-Based Individual
Patient Meta-Analysis in High-Risk Prostate Cancer. International Journal of Radiation Oncology,
Biology, Physics 2021;111(3):S50.
43. Spratt DE, Huang H-C, Michalski JM, et al. Validation of the performance of the Decipher
biopsy genomic classifier in intermediate-risk prostate cancer on the phase III randomized trial NRG
Oncology/RTOG 0126. Journal of Clinical Oncology 2022;40(6_suppl):269-269.
44. M. Parry EG, C.D. Brawley, J.A. Proudfoot, L. Mendes1, S. Lall, A.P. Hoyle, A. Sachdeva, Y. Liu,
C.L. Amos6, M.R. Sydes, R.J. Jones, M.K. Parmar, F. Feng, C.J. Sweeney, N. Clarke, E. Davicioni, N.D.
James, L.C. Brown, G. Attard. 1358O - Clinical qualification of transcriptome signatures for advanced
prostate cancer (APC) starting androgen deprivation therapy (ADT) with or without abiraterone
acetate and prednisolone (AAP): An ancillary study of the STAMPEDE AAP trial. Annals of Oncology
(2022) 33 (suppl_7): S616-S652. 10.1016/annonc/annonc1070 2022.
45. Morris WJ, Tyldesley S, Rodda S, et al. Androgen Suppression Combined with Elective Nodal
and Dose Escalated Radiation Therapy (the ASCENDE-RT Trial): An Analysis of Survival Endpoints for a
Randomized Trial Comparing a Low-Dose-Rate Brachytherapy Boost to a Dose-Escalated External
Beam Boost for High- and Intermediate-risk Prostate Cancer. Int J Radiat Oncol Biol Phys
2017;98(2):275-285.
46. Kerkmeijer LGW, Groen VH, Pos FJ, et al. Focal Boost to the Intraprostatic Tumor in External
Beam Radiotherapy for Patients With Localized Prostate Cancer: Results From the FLAME
Randomized Phase III Trial. J Clin Oncol 2021;39(7):787-796.
47. Kishan AU, Romero T, Alshalalfa M, et al. Transcriptomic Heterogeneity of Gleason Grade
Group 5 Prostate Cancer. Eur Urol 2020;78(3):327-332.
48. Vogelius IR, Bentzen SM. Diminishing Returns From Ultrahypofractionated Radiation Therapy
for Prostate Cancer. Int J Radiat Oncol Biol Phys 2020;107(2):299-304.
49. Arcangeli G, Saracino B, Arcangeli S, et al. Moderate Hypofractionation in High-Risk, Organ-
Confined Prostate Cancer: Final Results of a Phase III Randomized Trial. J Clin Oncol
2017;35(17):1891-1897.
50. Dearnaley D, Syndikus I, Mossop H, et al. Conventional versus hypofractionated high-dose
intensity-modulated radiotherapy for prostate cancer: 5-year outcomes of the randomised, non-
inferiority, phase 3 CHHiP trial. Lancet Oncol 2016;17(8):1047-1060.
51. Widmark A, Gunnlaugsson A, Beckman L, et al. Ultra-hypofractionated versus conventionally
fractionated radiotherapy for prostate cancer: 5-year outcomes of the HYPO-RT-PC randomised, non-
inferiority, phase 3 trial. Lancet 2019;394(10196):385-395.
52. Brand DH, Tree AC, Ostler P, et al. Intensity-modulated fractionated radiotherapy versus
stereotactic body radiotherapy for prostate cancer (PACE-B): acute toxicity findings from an
international, randomised, open-label, phase 3, non-inferiority trial. Lancet Oncol 2019;20(11):1531-
1543.
53. Pra AD, Zwahlen DR, Liu VY, et al. Prognostic and Predictive Performance of a 24-Gene Post-
Operative Radiation Therapy Outcomes Score (PORTOS) in a Phase 3 Randomized Trial of Dose-
Intensified Salvage Radiotherapy after Radical Prostatectomy (SAKK 09/10). International Journal of
Radiation Oncology*Biology*Physics 2022;114(3, Supplement):S37-S38.
54. Murthy V, Maitre P, Kannan S, 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(11):1234-1242.
26
55. Emmett L, Buteau J, Papa N, et al. The Additive Diagnostic Value of Prostate-specific
Membrane Antigen Positron Emission Tomography Computed Tomography to Multiparametric
Magnetic Resonance Imaging Triage in the Diagnosis of Prostate Cancer (PRIMARY): A Prospective
Multicentre Study. Eur Urol 2021; 10.1016/j.eururo.2021.08.002.
56. Hofman MS, Lawrentschuk N, Francis RJ, et al. Prostate-specific membrane antigen PET-CT in
patients with high-risk prostate cancer before curative-intent surgery or radiotherapy (proPSMA): a
prospective, randomised, multicentre study. Lancet 2020;395(10231):1208-1216.
57. Xu MJ, Kornberg Z, Gadzinski AJ, et al. Genomic Risk Predicts Molecular Imaging-detected
Metastatic Nodal Disease in Prostate Cancer. Eur Urol Oncol 2019;2(6):685-690.
58. Woythal N, Arsenic R, Kempkensteffen C, et al. Immunohistochemical Validation of PSMA
Expression Measured by (68)Ga-PSMA PET/CT in Primary Prostate Cancer. J Nucl Med
2018;59(2):238-243.
59. Spohn SKB, Bettermann AS, Bamberg F, et al. Radiomics in prostate cancer imaging for a
personalized treatment approach - current aspects of methodology and a systematic review on
validated studies. Theranostics 2021;11(16):8027-8042.
60. Phillips R, Shi WY, Deek M, et al. Outcomes of Observation vs Stereotactic Ablative Radiation
for Oligometastatic Prostate Cancer: The ORIOLE Phase 2 Randomized Clinical Trial. JAMA Oncology
2020;6(5):650-659.
61. Werensteijn-Honingh AM, Wevers AFJ, Peters M, et al. Progression-free survival in patients
with (68)Ga-PSMA-PET-directed SBRT for lymph node oligometastases. Acta Oncol 2021;
10.1080/0284186x.2021.1955970:1-10.
62. Ali A, Hoyle A, Haran ÁM, et al. Association of Bone Metastatic Burden With Survival Benefit
From Prostate Radiotherapy in Patients With Newly Diagnosed Metastatic Prostate Cancer: A
Secondary Analysis of a Randomized Clinical Trial. JAMA Oncology 2021;7(4):555-563.
63. Stopsack KH, Nandakumar S, Wibmer AG, et al. Oncogenic Genomic Alterations, Clinical
Phenotypes, and Outcomes in Metastatic Castration-Sensitive Prostate Cancer. Clinical cancer
research : an official journal of the American Association for Cancer Research 2020;26(13):3230-
3238.
64. Vale CL, Fisher D, Kneebone A, et al. Adjuvant or early salvage radiotherapy for the treatment
of localised and locally advanced prostate cancer: a prospectively planned systematic review and
meta-analysis of aggregate data. Lancet 2020;396(10260):1422-1431.
65. Mottet N, van den Bergh RCN, Briers E, et al. EAU-EANM-ESTRO-ESUR-SIOG Guidelines on
Prostate Cancer-2020 Update. Part 1: Screening, Diagnosis, and Local Treatment with Curative Intent.
Eur Urol 2021;79(2):243-262.
66. Dess RT, Jackson WC, Spratt DE. End Point Definitions and Surrogacy in Prostate Cancer: Will
Metastasis-Free Survival Become Event-Free Survival With Advances in Molecular Imaging? Journal of
Clinical Oncology 2021;39(25):2844-2845.
67. Eggener SE, Rumble RB, Armstrong AJ, et al. Molecular Biomarkers in Localized Prostate
Cancer: ASCO Guideline. J Clin Oncol 2020;38(13):1474-1494.
27
Figure legend
Figure 1: Summarizing overview
Figure 2: PRISMA Flow diagram of the systematic database search and excluded
records. Abbreviations: GC=genomic classifier, RT=radiotherapy
28
Figure 3: Clinical applications of genomic classifiers in various prostate cancer (PCa)
stages.
29
Figure 4 Consensus answers: Bars show agreement on genomic classifiers (GC)
being a useful tool to improve risk stratification across national cancer
comprehensive network (NCCN) risk groups, recurrent and metastatic disease and in
postoperative settings or as a predictive factor for various parameters across risk
groups and disease stages. In this context predictive factor” intends to represent the
ability of genomic classifiers to identify patients who might benefit from a certain
treatment. Abbreviations: PCa=prostate cancer, ADT= androgen deprivation therapy,
RT=radiotherapy
30
Figure 5: For each study assessing definitive radiotherapy, the hazard ratio (HR) and
95% confidence intervals on multivariate analysis of genomic classifiers predicting
distant metastases and biochemical recurrence is shown. For the Decipher score
(Studies by Ngyuen et al., Berlin et al. and Toisan et al.) HR per 0.1 unit increase is
shown. For the Oncotype DX Prostate Score (Study by Janes et al.) HR per 20 unit
increase is shown. For the Cell Cycle Progression score (Studies by Freedland and
Tward et al.) HR per 1 unit increase is shown.
31
Table legend
Table 1: List of included articles on GC in prostate cancer in the setting of
radiotherapy structured after prognostic and predictive oncological endpoints,
decision making and other biomarker studies. The second column includes endpoints
and the third column identifies whether patients were primarily treated with RP or RT.
Abbreviations:
CR=clinical recurrence, GC=genomic classifier, DM=distant metastases,
ART=adjuvant radiotherapy, BR=biochemical recurrence, RP= radical prostatectomy,
PCSM=prostate cancer specific mortality, OS=overall survival, SRT= salvage
radiotherapy, ADT=androgen deprivation therapy, RT = radiotherapy, AAM = Afro-
American Men, EAM=European American Men, MDT= metastases directed therapy,
PFS= progression free survival, PCa=prostate cancer, HR=Hazard Ratio, C-
Index=concordance index, TCGA=The Cancer Genome Atlas, PORTOS=Post
32
Operative Radiation Therapy Outcome Score, CAPRA=Cancer of the Prostate Risk
Assessment
Table 2: Summary of ongoing prospective clinical trials for prostate cancer RT
applying GCs.
Table 3: Expert consensus on endpoints to be addressed in clinical studies on GC
and translational research fields. Abbreviations: PCa=prostate cancer,
NCCN=national comprehensive cancer network, GC=genomic classifier,
MFS=metastases-free survival, PCSS=prostate cancer specific survival,
PFS=progression free survival
33
Table 1:
Studies analyzing oncological endpoints - Primary localized PCa
Author
Endpoint(s)
Treatment
Cohort Details:
Number of
patients (n)/study
design/validation
C-Index/Hazard
Ratio (HR) with
95% confidence
interval
(CI)/Pinteraction (if
analyzed)
Tosoian
et al.
[26]
DM
Definitive RP
or definitive RT
+/- ADT
n=405
Multicentric
Retrospective
No dedicated
external validation
HR for DM = 1.33
(1.19 1.48)
Berlin et
al. [27]
BR, DM
Definitive RT
n=121
Prospective
registry
Monocentric
No dedicated
external validation
HR for BR = 1.36
(1.09 1.71)
HR for DM = 2.05
(1.24 4.24)
Nguyen
et al.
[28]
DM, PCSM
Definitive RP
or definitive RT
+/- ADT
n=235
Multicentric
Retrospective
No dedicated
external validation
HR for DM = 1.37
(1.06 1.78),
HR for PCSM =
1.57 (1.03 2.48)
Nguyen
et al.
[29]
DM
Definitive RT +
ADT
n=100
Retrospective
Monocentric
No dedicated
external validation
HR for DM = 1.36
(1.04 1.83)
Freedla
nd et al.
[37]
BR
Definitive RT
+/- ADT
n=141
Retrospective
Monocentric
No dedicated
external validation
HR for BR:
Per one unit
change in score:
2.11 (1.05-4.25)
Janes et
al. [38]
BR, DM,
PCSM
Definitive RT
+/- ADT
n=238
Retrospective
Multicentric
No dedicated
external validation
HR for BR:
Per 20-unit
increase: 3.62
(2.59-5.02)
HR for DM:
Per 20-unit
increase: 4.48
(2.75-7.38)
HR for PCSM:
Per 20-unit
increase: 5.36
(3.06-.9.76)
Tward
et al.
[39]
DM
Definitive
dose-
escalated RT
n=741
Retrospective
Multicentric
No dedicated
external validation
HR for DM = 2.22
(1.71 2.89)
34
Studies analyzing oncological endpoints Adjuvant RT or SRT after primary RP
First
author
Endpoint(s)
Treatment
Cohort Details:
Number of
patients (n)/study
design/validation
C-Index/Hazard
Ratio (HR) with
95% confidence
interval
(CI)/Pinteraction (if
analyzed)
Lee et
al. [10]
CR,
External
validation of
a GC based
risk-
stratification
nomogram
ART or SRT
after RP
n=350
Monocentric
Retrospective
External validation
of the [11]
nomogram
C-index = 0.84
Mahal
et al.
[30]
DM
ART or SRT
after RP
Three cohorts:
Prospective
Registry cohort
(n=5239)
retrospective
multicenter cohort
(n=1135)
TCGA cohort
(n=498)
No dedicated
external validation
HR for DM = 2.15
(1.25 3.7)
10 year MFS for
patient with high
stromal scores
24% (no ART) vs
68%, p=0.0015
(ART)
pinteraction = 0.02
Dalela
et al.
[11]
CR
ART or SRT
after RP
n= 512
Multicentric
Retrospective
No dedicated
external validation
C-index:
Decipher = 0.71,
Decipher +
clinical model
0.85
HR for CR (GC
high vs low) =
2.93 (1.58 5.55)
Den et
al. [12]
DM
ART or SRT
after RP
n=188
Bicentric
Retrospective
No dedicated
external validation
HR for clinical
metastasis. =
1.61 (1.2 2.15).
Patients with high
risk GC: ART vs
SRT HR = 0.2
(0.04 0.90)
Den et
al. [13]
BR, DM
ART or SRT
after RP
n=143
Monocentric
Retrospective
No dedicated
external validation
HR for BR = 0.75
(0.67 0.94)
HR for DM = 0.78
(0.64 0.91)
35
Zhao et
al. [31]
DM
ART or SRT
after RP
n=196 matched
training cohort
n=330 pooled
matched validation
cohort
Multicentric
Retrospective
HR for DM after
RT in the high
PORTOS Group:
0.15 (0.04 - 0.6)
pinteraction = 0.016
Ross et
al. [32]
DM
ART or SRT
after RP
n=422
Multicentric
Retrospective
No dedicated
external validation
HR for DM= 1.28
(1.08 1.52)
Studies analyzing oncological endpoints Salvage RT after primary RP
First
author
Endpoint(s)
Treatment
Cohort Details:
Number of
patients (n)/study
design/validation
C-Index/Hazard
Ratio (HR) with
95% confidence
interval
(CI)/Pinteraction (if
analyzed)
Feng et
al. [14]
DM, PCSM,
OS
SRT +- ADT
after RP
n=486
Multicentric
Prospective
No dedicated
external validation
HR for DM = 1.17
(1.05 1.32),
HR for PCSM =
1.39 (1.20
1.63),
HR for OS = 1.17
(1.06 1.29)
Koch et
al. [33]
BR
SRT after RP
n=47
Retrospective
Monocentric
No dedicated
external validation
Odds ratio for DM
or non-response
to SRT:
Per one unit
change in score:
10.4 (2.05-90.1)
Dal Pra
et al.
[15]
BR, CR
SRT after RP
n=226
Cohort from RCT
No dedicated
external validation
HR for BR:
GC continuous =
1.14 (1.04
1.25);
GC categorical
high vs low-
intermediate =
2.21 (1.41
3.47). HR for CR:
GC categorical
(high vs low-
intermediate) =
2.29 (1.32 3.98)
Studies analyzing oncological endpoints Metastasis-directed therapy in oligometastatic
PCa
Deek et
PFS
MDT (RT or
n=70
HR for PFS:
36
al. [41]
surgery) in
oligometastatic
castration
sensitive PCa
Cohort from two
prospective trials
No dedicated
external validation
low vs high
mutational
burden): 0.57 (
0.32 1.03)
MDT vs
observation in
patients with high
mutational
burden: 0.05
(0.01 0.28)
MDT vs
observation in
patients without
high mutational
burden: 0.42
(0.23 0.77)
Studies analyzing treatment decision making
First
author
Endpoint
Primary
Treatment
Gore et
al. [16]
Postoperative treatment decision
RP
Marasci
o et al.
[17]
Postoperative treatment decision
RP
Gore et
al. [18]
Postoperative treatment decision (ART or SRT)
RP
Michalo
poulos
et al.
[19]
Postoperative treatment decision in high risk
patients
RP
Badani
et al.
[20]
Postoperative treatment decision in high-risk
patients
RP
Badani
et al.
[21]
Postoperative treatment decision
RP
Nguyen
et al.
[22]
Postoperative treatment recommendations from 20
US board certificated urologist and 26 radiation
oncologist
RP
Lobo et
al. [23]
Markov Model for decision of ART vs SRT after RP
RP
Lobo et
al. [24]
Markov Model for cost effectiveness
RP
Biomarker studies
First
author
Endpoint(s)
Ben-
Salem
et al.
[34]
Androgen Receptor Activity in localized treatment naive PCa and
association with clinical risk factors, molecular markers and PCa
subtypes.
37
Yamoah
et al.
[25]
Transcriptomic interactions between tumor immune content score (ICS)
and Decipher GC
Awasthi
et al.
[35]
Differences of immune-specific genes between AAM and EAM PCa
tumor environment
Mahal
et al.
[36]
PCSM, all-cause mortality and genomic characterization of PCa
patients with low PSA and high grade PCa
Table 2:
Ongoing Trials
Trial number
Study type
Patient
characteristics
Applied GC
Treatment
decision based on
GC
NCT04513717
(NRG-GU009)
Parallel
Phase III,
randomized
NCCN high-risk
Decipher
Escalation or de-
escalation of
systemic therapy
NCT05100472
(SHORTER)
Phase II, non-
randomized
NCCN high-risk
Decipher
ADT de-escalation
NCT05050084
(NRG-GU10)
Parallel
Phase III,
randomized
NCCN
unfavorable
intermediate-
risk
Decipher
Escalation or de-
escalation of
systemic therapy
NCT04025372
(INTREPID)
Phase II,
randomized
NCCN
intermediate-
risk
Decipher
N/A (GC is required
and serves as
stratification
variable)
NCT05169970
Phase II, non-
randomized
NCCN
unfavorable
intermediate-
risk
Decipher
Inclusion of elective
pelvic lymphatics in
RT field
NCT02783950
(G-Minor)
Randomized,
parallel
assignment
RPE with pT3
or positive
margins
Decipher
Adjuvant treatment
decision (RT or
ADT)
NCT04984343
(FORT)
Phase II,
randomized
NCCN low- and
intermediate-
risk
Decipher
N/A (GC>0.6
serves as inclusion
criterion)
NCT04396808
Crossover
assignment,
NCCN low- and
intermediate-
Decipher,
Prolaris and
N/A (Impact of GC
on treatment
38
randomized
risk
Oncotype DX
decision)
NCT02723734
(VANDAAM)
Cohort
NCCN low- and
intermediate-
risk
Decipher
N/A (Impact of GC
on outcome
prediction)
NCT03495427
Cohort
NCCN low- and
intermediate-
risk
Decipher
N/A (concordance
between GC and
PSMA-PET)
(subgroup of
VANDAAM)
NCT03371719
(NRG-GU006)
Phase II,
randomized
SRT
PAM50 gene
expression
N/A (gene
expression
clustering)
NCT03770351
Cohort
NCCN low- and
intermediate-
risk
Decipher
ProstateNext
N/A (Impact of GC
on outcome
prediction
NCT03141671
Phase II,
randomized
SRT
Decipher
N/A (high risk
Decipher score as
inclusion criterion)
NCT04134260
Phase III,
randomized
SRT
Decipher
PAM50 gene
expression
N/A (Impact of GC
on outcome
prediction
39
Table 3:
Primary PCa -
NCCN
low/favorable
intermediate-risk
Which of the following endpoints do you consider
relevant to be addressed with GCs as predictors
for treatment
% of
answers
MFS
82.8%
Primary PCa - NCCN
unfavorable
intermediate-risk
Which of the following endpoints do you consider
relevant to be addressed with GCs as predictors
for treatment
MFS
96.6%
Time to distant metastases
82.1%
Primary PCa - NCCN high-
risk
Which of the following endpoints do you consider
relevant to be addressed with GCs as predictors
for treatment
MFS
96.6%
PCSS
79.3%
Time to distant metastases
93.1%
Oligometastastaticoligoprogres
sive or oligorecurrent disease
Which of the following endpoints do you consider
most relevant to be addressed with genomic
classifiers as predictors for treatment
management
PCSS
82.8%
Time to castration-resistance
75.9%
PFS
75.9%
Translationa
l research
fields
Which of the following translational research fields
do you consider as relevant to be addressed in
future research incorporating GCs?
40
Strategies to cope with intertumoral heterogeneity in
case GCs are obtained from biopsy cores in metastatic
disease (evaluation of intertumoral heterogeneity
between primary and metastases)
75.0%
Alteration in androgen signaling
75.0%
Decision making for physicians
85.7%
Article
Full-text available
There are few randomized trials to evaluate the use of PSMA-PET in the planning of post-prostatectomy radiotherapy. There are two unresolved questions 1) should we increase the dose to lesions visible on PSMA-PET 2) can we reduce dose in the case of a negative PSMA-PET. In this review, we summarize and discuss the available evidence in the literature. We found that in general, there seems to be an advantage for dose-increase, but ta large recent study from the pre-PSMA era didn’t show an advantage for dose escalation. Retrospective studies have shown that conventional doses to PSMA-PET-positive lesions seem sufficient. On the other hand, in the case of a negative PSMA-PET, there is no evidence that dose-reduction is possible. In the future, the combination of PSMA-PET with genomic classifiers could help in better identify patients who might benefit from either dose- de-or -increase. We further need to identify intraindividual references to help identify lesions with higher aggressiveness.
Article
Full-text available
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.
Article
Full-text available
Purpose: The Oncotype DX Genomic Prostate Score® (GPS™) assay has been validated as a strong prognostic indicator of adverse pathology, biochemical recurrence, distant metastasis (DM), and prostate cancer (PCa)-related death (PCD) in men with localized PCa after radical prostatectomy. However, it has yet to be tested in men undergoing external beam radiation therapy (EBRT), for whom assessing PCa progression risk could inform decisions on treatment intensity. We analyzed whether GPS results are associated with time to biochemical failure (BCF), DM, and PCD after EBRT in men with localized PCa and whether the association is modified by race. Methods and materials: We conducted a retrospective study of men with localized PCa treated with EBRT at the [ANONYMIZED FOR REVIEW] from 2000 to 2016. Study endpoints were time to BCF per the Phoenix criteria, DM, and PCD. The association of GPS results, per 20-unit increase or dichotomous variable (0-40 versus 41-100), was evaluated with each endpoint using univariable and multivariable Cox proportional hazards models. Results were then stratified by race. Results: A total of 238 patients (69% Black) met eligibility criteria. Median follow-up for patients who did not experience BCF was 7.6 years. GPS results per 20-unit increase were significantly associated with BCF (hazard ratio (HR) 3.62; 95% CI 2.59, 5.02), DM (HR 4.48; 2.75, 7.38), and PCD (HR 5.36; 3.06, 9.76) in univariable analysis. GPS results remained significant in multivariable models adjusted for baseline clinical and pathological factors, with HRs similar to the univariable analysis. There was no significant interaction between the GPS assay and race (p=0.923). HRs for BCF were similar in Black men (3.88; 2.40, 6.24) versus non-Black men (4.01; 2.42, 6.45). Conclusion: Among men treated with EBRT, the GPS assay is a strong, independent prognostic indicator of time to BCF, DM and PCD, and performs similarly in Black and non-Black men.
Article
Full-text available
Each year, the American Cancer Society estimates the numbers of new cancer cases and deaths in the United States and compiles the most recent data on population‐based cancer occurrence and outcomes. Incidence data (through 2018) were collected by the Surveillance, Epidemiology, and End Results program; the National Program of Cancer Registries; and the North American Association of Central Cancer Registries. Mortality data (through 2019) were collected by the National Center for Health Statistics. In 2022, 1,918,030 new cancer cases and 609,360 cancer deaths are projected to occur in the United States, including approximately 350 deaths per day from lung cancer, the leading cause of cancer death. Incidence during 2014 through 2018 continued a slow increase for female breast cancer (by 0.5% annually) and remained stable for prostate cancer, despite a 4% to 6% annual increase for advanced disease since 2011. Consequently, the proportion of prostate cancer diagnosed at a distant stage increased from 3.9% to 8.2% over the past decade. In contrast, lung cancer incidence continued to decline steeply for advanced disease while rates for localized‐stage increased suddenly by 4.5% annually, contributing to gains both in the proportion of localized‐stage diagnoses (from 17% in 2004 to 28% in 2018) and 3‐year relative survival (from 21% to 31%). Mortality patterns reflect incidence trends, with declines accelerating for lung cancer, slowing for breast cancer, and stabilizing for prostate cancer. In summary, progress has stagnated for breast and prostate cancers but strengthened for lung cancer, coinciding with changes in medical practice related to cancer screening and/or treatment. More targeted cancer control interventions and investment in improved early detection and treatment would facilitate reductions in cancer mortality.
Article
Full-text available
Prostate cancer (PCa) is one of the most frequently diagnosed malignancies of men in the world. Due to a variety of treatment options in different risk groups, proper diagnostic and risk stratification is pivotal in treatment of PCa. The development of precise medical imaging procedures simultaneously to improvements in big data analysis has led to the establishment of radiomics - a computer-based method of extracting and analyzing image features quantitatively. This approach bears the potential to assess and improve PCa detection, tissue characterization and clinical outcome prediction. This article gives an overview on the current aspects of methodology and systematically reviews available literature on radiomics in PCa patients, showing its potential for personalized therapy approaches. The qualitative synthesis includes all imaging modalities and focuses on validated studies, putting forward future directions.
Article
Full-text available
Background Recently, an increase in the rates of high-risk prostate cancer (PCa) was reported. We tested whether the rates of and low, intermediate, high and very high-risk PCa changed over time. We also tested whether the number of prostate biopsy cores contributed to changes rates over time. Methods Within the Surveillance, Epidemiology and End Results (SEER) database (2010–2015), annual rates of low, intermediate, high-risk according to traditional National Comprehensive Cancer Network (NCCN) and high versus very high-risk PCa according to Johns Hopkins classification were tabulated without and with adjustment for the number of prostate biopsy cores. Results In 119,574 eligible prostate cancer patients, the rates of NCCN low, intermediate, and high-risk PCa were, respectively, 29.7%, 47.8%, and 22.5%. Of high-risk patients, 39.6% and 60.4% fulfilled high and very high-risk criteria. Without adjustment for number of prostate biopsy cores, the estimated annual percentage changes (EAPC) for low, intermediate, high and very high-risk were respectively −5.5% (32.4%–24.9%, p < .01), +0.5% (47.6%–48.4%, p = .09), +4.1% (8.2%–9.9%, p < .01), and +8.9% (11.8%–16.9%, p < .01), between 2010 and 2015. After adjustment for number of prostate biopsy cores, differences in rates over time disappeared and ranged from 29.8%–29.7% for low risk, 47.9%–47.9% for intermediate risk, 8.9%–9.0% for high-risk, and 13.6%–13.6% for very high-risk PCa (all p > .05). Conclusions The rates of high and very high-risk PCa are strongly associated with the number of prostate biopsy cores, that in turn may be driven by broader use magnetic resonance imaging (MRI).
Article
Purpose/Objective(s) The SAKK 09/10 phase 3 trial showed that dose-intensified salvage radiotherapy (SRT) was not superior to conventional dose SRT for biochemically recurrent prostate cancer (PCa) after radical prostatectomy (RP). We previously reported on the validation of the 22-gene genomic classifier as prognostic in this trial but did not observe an interaction with RT dose. Herein, we examine the PORTOS classifier, an expression signature of 24 DNA damage repair and immune pathway genes, previously shown to predict benefit from postoperative radiotherapy, to test its predictive ability to SRT dose. Materials/Methods The SAKK 09/10 trial (NCT01272050) was a multicenter, randomized phase 3 trial that included PCa patients with biochemical progression after RP (PSA > 0.1 to 2 ng/mL at randomization). Patients were randomized to 64Gy vs 70Gy to the prostate bed. No patients received concurrent androgen suppression or pelvic nodal radiotherapy. After central review, the highest-grade tumors were profiled using the Decipher whole-transcriptome assay (Veracyte, San Diego, CA) to generate PORTOS. The independent prognostic ability and interaction effects were examined for the biochemical progression, clinical progression-free survival (CPFS), receipt of salvage hormone therapy, and metastasis-free survival endpoints with Cox multivariable and interaction analyses (MVA). Categorical analyses used the published threshold for PORTOS radiotherapy response (high vs low). Results Of the 226 trial patients with transcriptome data, 16% had high PORTOS radiation response compared to 84% with lower PORTOS. Patients with high PORTOS were well balanced between the randomization arms. PORTOS was not prognostic for the endpoints in MVA with the trial stratification factors. A high PORTOS, however, was predictive for improved outcomes in patients receiving dose-intensified SRT. Patients with high PORTOS that received 70Gy dose had 5-year CPFS of 94% compared to 49% with the 64Gy dose (log-rank p=0.006). In interaction analysis, PORTOS had a statistically significant interaction with RT dose for multiple endpoints: freedom from biochemical progression (interaction p=0.04), clinical progression-free survival (interaction p=0.003) and metastasis-free survival (interaction p=0.037). Conclusion In a phase 3 trial testing SRT dose intensification after RP, patients with high PORTOS had significantly better outcomes with dose-intensified treatment, suggesting that higher doses should be considered in this subgroup. Further studies are warranted to validate PORTOS as the first predictive biomarker to help personalize dose prescription in the postoperative setting.
Article
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co‐primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported. The initial STOMP and ORIOLE trial reports suggested that metastasis-directed therapy (MDT) in oligometastatic castration-sensitive prostate cancer (omCSPC) was associated with improved treatment outcomes. Here, we present long-term outcomes of MDT in omCSPC by pooling STOMP and ORIOLE and assess the ability of a high-risk mutational signature to risk stratify outcomes after MDT. The primary end point was progression-free survival (PFS) calculated using the Kaplan-Meier method. High-risk mutations were defined as pathogenic somatic mutations within ATM, BRCA1/ 2, Rb1, or TP53. The median follow-up for the whole group was 52.5 months. Median PFS was prolonged with MDT compared with observation (pooled hazard ratio [HR], 0.44; 95% CI, 0.29 to 0.66; P value < .001), with the largest benefit of MDT in patients with a high-risk mutation (HR high-risk: 0.05; HR no high-risk: 0.42; P value for interaction: .12). Within the MDT cohort, the PFS was 13.4 months in those without a high-risk mutation, compared with 7.5 months in those with a high-risk mutation (HR, 0.53; 95% CI, 0.25 to 1.11; P = .09). Long-term outcomes from the only two randomized trials in omCSPC suggest a sustained clinical benefit to MDT over observation. A high-risk mutational signature may help risk stratify treatment outcomes after MDT.
Article
Background The Decipher genomic classifier (GC) has shown to independently prognosticate outcomes in prostate cancer. The objective of this study was to validate the GC in a randomized phase 3 trial of dose-escalated salvage radiotherapy (SRT) after radical prostatectomy. Patients and Methods A clinical grade whole-transcriptome assay was performed on RP samples obtained from patients enrolled in SAKK 09/10, a phase 3 trial of 350 men with biochemical recurrence post-radical prostatectomy randomized to 64Gy vs. 70Gy without concurrent hormonal therapy or pelvic nodal radiotherapy (RT). A pre-specified statistical plan was developed to assess the impact of the GC on clinical outcomes. The primary endpoint was biochemical progression; secondary endpoints were clinical progression and time to hormone therapy. Multivariable analyses adjusted for age, T-category, Gleason score, post-radical prostatectomy persistent prostate-specific antigen (PSA), PSA at randomization, and randomization arm were conducted, accounting for competing risks. Results The analytic cohort of 226 patients was representative of the overall trial, with median follow-up of 6.3 years (IQR 6.1-7.2). GC (high vs. low-intermediate) was independently associated with biochemical progression (subdistribution hazard ratio [sHR] 2.26 [95% CI 1.42-3.60], p<0.001), clinical progression (HR 2.29 [95% CI 1.32-3.98], p=0.003), and use of hormone therapy (sHR 2.99 [95% CI 1.55-5.76], p=0.001). GC high patients had 5-year freedom from biochemical progression of 45% vs. 71% for GC low-intermediate. Dose escalation did not benefit the overall cohort, nor patients with lower vs. higher GC scores. Conclusions This study represents the first contemporary randomized controlled trial in patients treated with early SRT without concurrent hormone therapy or pelvic nodal RT that has validated the prognostic utility of the GC. Independent of standard clinicopathologic variables and RT dose, high-GC patients were more than twice as likely than lower-GC patients to experience biochemical and clinical progression and receive of salvage hormone therapy. This data confirms the clinical value of Decipher GC to personalize the use of concurrent systemic therapy in the postoperative salvage setting.
Article
269 Background: The 22-gene Decipher genomic classifier (GC) is a prognostic biomarker that has been validated in phase III trials in high-risk localized, post-prostatectomy, and metastatic and non-metastatic castration-resistant prostate cancer. Herein, we report the first validation of the biopsy GC in intermediate-risk prostate cancer from the phase III randomized trial NRG/RTOG 0126. Methods: After National Cancer Institute approval, biopsy slides were collected from the NRG biobank from RTOG 0126, a phase III randomized trial of men with intermediate-risk prostate cancer randomized to 70.2 Gy versus 79.2 Gy of radiotherapy without the use of concomitant hormone therapy. RNA was extracted from the highest grade tumor foci and processed through a quality control (QC) pipeline prior to generation of the previously locked 22-gene GC model. After GC data was generated it was linked with clinical outcomes to assess prognostic performance. The primary endpoint for this ancillary project was disease progression, defined as biochemical failure, local failure, distant metastasis or prostate cancer-specific mortality, as well as use of salvage therapy. Secondary endpoints included the previous individual endpoints, metastasis-free survival, and overall survival. Independent GC prognostic performance was assessed using cause-specific Cox or competing risk adjusted Fine-Gray multivariable models that included randomization arm and prognostic stratification factors. Death without events were treated as competing risks. Results: A total of 215 patient samples passed QC of the 449 that had suitable cDNA for expression analysis. The median follow-up was 12.8 years (range 2.4-17.7), and 61% had Gleason 3+4, 24% had Gleason 4+3, and the median PSA was 7.2 ng/mL (IQR 5.0-10.2). On multivariable analysis the 22-gene GC (per 0.1 unit) was independently prognostic for disease progression (subdistribution hazard ratio [sHR] 1.13, 95%CI (1.01-1.26), p = 0.03), biochemical failure (sHR 1.23, 95%CI 1.10-1.37, p < 0.001), distant metastasis (sHR 1.28, 95%CI 1.06-1.54, p = 0.01), and PCSM (sHR 1.45, 95%CI 1.20-1.76, p < 0.001). In patients with lower GC scores the 10-year distant metastasis rate difference between the 70.2 Gy and 79.2 Gy was 5%, as compared with 26% for higher GC patients. Conclusions: This study represents the first validation of any biopsy-based gene expression classifier in intermediate-risk prostate cancer. Decipher is independently prognostic and can identify patients that have low rates of metastatic events despite not receiving concurrent hormone therapy, and can be used to help personalize therapy in this setting. Clinical trial information: NCT00033631.
Article
Purpose/Objective(s) Decipher is a prognostic 22-gene genomic classifier (GC) prospectively validated post-prostatectomy. Herein, we validate the performance of the GC in pre-treatment biopsy samples within the context of three randomized phase III high-risk definitive radiotherapy trials. Materials/Methods Following a pre-specified and approved CTEP-CCSC analysis plan (NRG-GU-TS006), we obtained all available formalin-fixed paraffin-embedded tissue from biopsy specimens from the NRG biobank from patients enrolled on NRG/RTOG 9202, 9413, and 9902 phase III randomized trials. After central review, the highest-grade tumors were profiled on clinical-grade whole-transcriptome arrays and GC scores were obtained. The primary objective was to validate the independent prognostic ability of GC for distant metastases (DM), and secondary was prostate cancer-specific mortality (PCSM) and overall survival (OS), with Cox multivariable analyses (MVA). Results GC scores were obtained on 385 samples (n = 90 on 9202, n = 172 on 9413, and n = 123 on 9902), of which 265 passed microarray quality control (69%) and had a median follow-up of 11 years (interquartile range, 9, 13). On MVA, the GC (per 0.1 unit) was independently associated with DM (HR 1.24, 95% CI 1.11-1.39), PCSM (HR 1.27, 95% CI 1.13-1.43), and OS (HR 1.12, 95% CI 1.05-1.20) after adjusting for age, PSA, Gleason score, cT-stage, trial, and randomized treatment arm. For categorical GC, on MVA, GC score ≥ 0.45 (representing the intermediate and high GC categories) had worse DM (HR 2.18, 95% CI 1.25-3.80), PCSM (HR 2.34, 95% CI 1.31-4.16), and OS (HR 1.45, 95% CI 1.03-2.04) outcomes as compared to those with low GC. Cumulative incidence of distant-metastasis at 10-years was 29% (95% CI 20-38%) for intermediate/high GC vs 13% (95% CI 7-18%) for low GC. For the subset with GC > 0.85, the threshold for inclusion in the intensification study of NRG GU009 (PREDICT-RT), at 5-years and 10-years DM was 29% (95% CI 7-52%) and 41% (95% CI 17-66%). GC had similar prognostic ability in patients receiving short-term or long-term androgen-deprivation therapy (ADT). Conclusion This is the first validation of any gene expression biomarker on pre-treatment biopsy samples from prospective randomized trials and demonstrates an independent association of GC score with DM, PCSM, and OS. High-risk prostate cancer is a heterogeneous disease state and GC can improve risk stratification to help personalize shared decision-making. NRG-GU009/PREDICT-RT will further determine the optimal therapy based on GC score. NCT04513717
Article
Background Multiparametric magnetic resonance imaging (MRI) is validated for the detection of clinically significant prostate cancer (csPCa), although patients with negative/equivocal MRI undergo biopsy for false negative concerns. In addition, ⁶⁸Ga-PSMA-11 positron emission tomography/computed tomography (prostate-specific membrane antigen [PSMA]) may also identify csPCa accurately. Objective This trial aimed to determine whether the combination of PSMA + MRI was superior to MRI in diagnostic performance for detecting csPCa. Design, setting, and participants A prospective multicentre phase II imaging trial was conducted. A total of 296 men were enrolled with suspected prostate cancer, with no prior biopsy or MRI, recent MRI (6 mo), and planned transperineal biopsy based on clinical risk and MRI. In all, 291 men underwent MRI, pelvic-only PSMA, and systematic ± targeted biopsy. Outcome measurements and statistical analysis Sensitivity, specificity, and predictive values (negative predictive value [NPV] and positive predictive value) for csPCa were determined for MRI, PSMA, and PSMA + MRI. PSMA + MRI was defined as negative for PSMA negative Prostate Imaging Reporting and Data System (PI-RADS) 2/3 and positive for either MRI PI-RADS 4/5 or PSMA positive PI-RADS 2/3; csPCa was any International Society of Urological Pathology (ISUP) grade group ≥2 malignancy. Results and limitations Of the patients, 56% (n = 162) had csPCa; 67% had PI-RADS 3–5, 73% were PSMA positive, and 81% were combined PSMA + MRI positive. Combined PSMA + MRI improved NPV compared with MRI alone (91% vs 72%, test ratio = 1.27 [1.11–1.39], p < 0.001). Sensitivity also improved (97% vs 83%, p < 0.001); however, specificity was reduced (40% vs 53%, p = 0.011). Five csPCa cases were missed with PSMA + MRI (four ISUP 2 and one ISUP 3). Of all men, 19% (56/291) were PSMA + MRI negative (38% of PI-RADS 2/3) and could potentially have avoided biopsy, risking delayed csPCa detection in 3.1% men with csPCa (5/162) or 1.7% (5/291) overall. Conclusions PSMA + MRI improved NPV and sensitivity for csPCa in an MRI triaged population. Further randomised studies will determine whether biopsy can safely be omitted in men with a high clinical suspicion of csPCa but negative combined imaging. Patient summary The combination of magnetic resonance imaging (MRI) + prostate-specific membrane antigen positron emission tomography reduces false negatives for clinically significant prostate cancer (csPCa) compared with MRI, potentially allowing a reduction in the number of prostate biopsies required to diagnose csPCa.