Skip to main content
Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging 1/2022

Open Access 19.08.2022 | Original Article

The maximum standardized uptake value in patients with recurrent or persistent prostate cancer after radical prostatectomy and PSMA-PET-guided salvage radiotherapy—a multicenter retrospective analysis

verfasst von: Simon K. B. Spohn, Andrea Farolfi, Sarah Schandeler, Marco M. E. Vogel, Juri Ruf, Michael Mix, Simon Kirste, Francesco Ceci, Stefano Fanti, Helena Lanzafame, Francesca Serani, Christian Gratzke, August Sigle, Stephanie E. Combs, Denise Bernhardt, Juergen E. Gschwend, Josef A. Buchner, Christian Trapp, Claus Belka, Peter Bartenstein, Lena Unterrainer, Marcus Unterrainer, Matthias Eiber, Stephan G. Nekolla, Kilian Schiller, Anca L. Grosu, Nina-Sophie Schmidt-Hegemann, Constantinos Zamboglou, Jan C. Peeken

Erschienen in: European Journal of Nuclear Medicine and Molecular Imaging | Ausgabe 1/2022

Abstract

Purpose

This study aims to evaluate the association of the maximum standardized uptake value (SUVmax) in positron-emission tomography targeting prostate-specific membrane antigen (PSMA-PET) prior to salvage radiotherapy (sRT) on biochemical recurrence free survival (BRFS) in a large multicenter cohort.

Methods

Patients who underwent 68 Ga-PSMA11-PET prior to sRT were enrolled in four high-volume centers in this retrospective multicenter study. Only patients with PET-positive local recurrence (LR) and/or nodal recurrence (NR) within the pelvis were included. Patients were treated with intensity-modulated-sRT to the prostatic fossa and elective lymphatics in case of nodal disease. Dose escalation was delivered to PET-positive LR and NR. Androgen deprivation therapy was administered at the discretion of the treating physician. LR and NR were manually delineated and SUVmax was extracted for LR and NR. Cox-regression was performed to analyze the impact of clinical parameters and the SUVmax-derived values on BRFS.

Results

Two hundred thirty-five patients with a median follow-up (FU) of 24 months were included in the final cohort. Two-year and 4-year BRFS for all patients were 68% and 56%. The presence of LR was associated with favorable BRFS (p = 0.016). Presence of NR was associated with unfavorable BRFS (p = 0.007). While there was a trend for SUVmax values ≥ median (p = 0.071), SUVmax values ≥ 75% quartile in LR were significantly associated with unfavorable BRFS (p = 0.022, HR: 2.1, 95%CI 1.1–4.6). SUVmax value in NR was not significantly associated with BRFS. SUVmax in LR stayed significant in multivariate analysis (p = 0.030). Sensitivity analysis with patients for who had a FU of > 12 months (n = 197) confirmed these results.

Conclusion

The non-invasive biomarker SUVmax can prognosticate outcome in patients undergoing sRT and recurrence confined to the prostatic fossa in PSMA-PET. Its addition might contribute to improve risk stratification of patients with recurrent PCa and to guide personalized treatment decisions in terms of treatment intensification or de-intensification.
This article is part of the Topical Collection on Oncology—Genitourinary.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s00259-022-05931-5.
This article is part of the Topical Collection on Oncology - Genitourinary.
Simon K. B. Spohn, and Andrea Farolfi have shared first authorship.
Constantinos Zamboglou and Jan C. Peeken have shared last authorship.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Up to 50% of patients with localized prostate cancer (PCa) undergoing radical prostatectomy (RPE) experience biochemical relapse within the first 5 years after treatment [13]. Early salvage radiation therapy (sRT) with or without androgen deprivation therapy (ADT) is recommended as the only curative treatment option [4]. However, recurrence rate after sRT is represented by heterogeneous patterns and influenced by clinico-pathological features such as pre-treatment prostate-specific antigen values (PSA), International Society of Urological Pathology Grade (ISUP), extracapsular extension (ECE), and seminal vesicle infiltration (SVI) and surgical margins with progression-free survival rates vary between 20 and 70% after 5 years [2, 5]. Therefore, additional prognostic markers are needed to improve risk stratification and guide personalized treatment approaches such as dose escalation, adaption of RT fields, or intensification of systemic treatments. Implementation of positron emission tomography (PET) targeting the prostate-specific membrane antigen (PSMA), a cell-surface transmembrane protein over-expressed in PCa cells [6], improves detection rates of recurrent PCa lesions even at low PSA levels and outside the prostatic fossa [7, 8]. These findings have high impact on the management of salvage treatments with alteration in approximately half of patients [911]. While retrospective evidence supports putative improvements of biochemical recurrence free survival rates due to PSMA-PET-guided sRT [12, 13], results of prospective trials are pending ([14], NCT04794777, PATRON–NCT04557501). Emerging data suggest that the maximum standardized uptake value (SUVmax) can be used as a biomarker to prognosticate clinically significant PCa [15], Gleason-Score (GS) [16, 17], and distant metastases [18] in primary PCa, but no data exist on the applicability of this PSMA-PET feature in patients with relapse undergoing sRT. In search of new, non-invasive biomarkers for personalized risk stratification, this retrospective multicenter study aims to evaluate the impact of the SUVmax on biochemical recurrence free survival (BRFS) in patients with recurrent or persistent PCa cancer after RPE and 68 Ga-PSMA11-guided salvage radiotherapy.

Methods

Patients and treatment

This multicenter study collected from high-volume centers in Germany (University Medical Centre Freiburg, Klinikum Rechts der Isar Technical University Munich (TUM), University Hospital of the Ludwig-Maximillian’s-University Munich (LMU)) and Italy (IRCCS Azienda Ospedaliero-Universitaria di Bologna). The study received institutional review board approval from all participating institutions (Freiburg No.: 15/18; TUM:466/16 S; Bologna: 385/2021/Oss/AOUBo, LMU: 17–765). The centers collected data from patients who received radical surgery and underwent 68 Ga-PSMA11-PET due to PSA persistence (PSA after surgery ≥ 0.1 ng/ml) or recurrence (PSA ≥ 0.2 as nadir after surgery) and were subsequently treated with PSMA-PET-guided sRT. Treatment decisions were taken locally at the discretion of the treating physicians according to standards of care at the time of treatment [4] and based on PSMA-PET/CT findings. RT to the prostatic fossa was not omitted in case of nodal recurrence (NR) only. See supplementary Table 1 for details on salvage RT concepts. ADT was administered at the discretion of the treating physician. Patients were excluded if distant metastases (lymph nodes above the iliac bifurcation, bone metastases, or visceral metastases) were present in PSMA PET/CT and if ADT was given prior to PSMA PET/CT scans. Two hundred and fifty-one patients with local recurrence (LR) and/or nodal recurrence (NR) treated with sRT between 2014 and 2020 met initial inclusion criteria. Sixteen patients were excluded due to equivocal PET findings not suitable for accurate contouring of PET-lesions, resulting in two hundred and thirty-five patients in the final cohort. Additionally, a subgroup of patients with follow-up time > 12 months was created (n = 197). See Fig. 1 for a consort flow diagram.

68Ga-PSMA11 PET and images analysis

68 Ga-PSMA11 was synthesized according to good manufacture practice in all centers and in accordance with international procedural guidelines [19]. PET/CT images were acquired approximately 60 min after tracer injection (approximately 1.8–2.2 MBq 68 Ga-PSMA11 per kg bodyweight) in all centers and for the PSMA PET/CT contrast-enhanced or unenhanced CTs using a slice thickness of 2 mm 120 kVp, 100–400 mAs, and dose modulations were performed for attenuation correction. The following scanners were used: Freiburg: 16-slice Gemini TF Big Bore, 64-slice Gemini TF or Vereos, all Philips Healthcare, USA; TUM: Biograph mCT/128 slice CT, Siemens Healthineers, Germany; LMU: Biograph 64 and Biograph mCT (Siemens Healthineers, Germany) or Discovery 690 (GE Healthcare, USA); Bologna: Discovery MI or Discovery 710 (both GE Healthcare, USA). All scanners fulfilled the requirements indicated in the European Association of Nuclear Medicine (EANM) imaging guidelines and obtained EANM Research Ltd. (EARL1) accreditation during acquisition.
The following reconstruction algorithms were used: Freiburg: Gemini TF 64 and Gemini TF BigBore: LOR-based ordered-subset iterative time-of-flight algorithm using spherical coordinates (BLOB-OS-TF) with 3 iterations and 33 subsets and a relaxation parameter for smoothing. Vereos: BLOB-OS-TF with 3 iterations and 9 subsets without smoothing [20]; TUM: The reconstruction algorithm included point-spread-function and time-of-flight with 3 iterations and 21 subsets; LMU: Biograph 64: TrueX (3 iterations, 21 subsets) with Gaussian post-reconstruction smoothing (2 mm full width at half-maximum). Biograph mCT: TrueX (3 iterations, 21 subsets). Discovery 690: VUE Point Fx algorithm with 2 iterations and 36 subsets; Bologna: Discovery MI: time-of-flight, 8 subsets and 4 iterations; Discovery 710: time-of-flight, 18 subsets and 3 iterations.
All systems resulted in a PET image with a voxel size of 2 × 2 × 2 mm3. Images were normalized to decay corrected injected activity per kg body weight (SUV g/ml).
All PSMA-PET images were locally reviewed prior to data sharing by two nuclear medicine physicians with experience on PCa imaging and according to reporting international guidelines [21, 22]. Disagreements were resolved by consensus.

Image processing

Image analysis was performed with 3D Slicer v4.10.0 [23]. Considering the local nuclear medicine report, PSMA-PET-positive LR and NR lesions were manually contoured by one reader (SS) with > 3 years’ experience in PSMA-PET segmentation using a window level from SUVmin-max: 0–5 based on previous windowing recommendations in primary PCa patients [24]. Under consideration of CT images and available PSMA-PET/CT results, any focal uptake higher than adjacent background in more than one slice was considered to represent PCa. Equivocal or small findings limited to one slice were not segmented. SUVmax was extracted for each lesion separately. Since segmentation of LR adjacent to the bladder wall can be challenging, an inter-observer variability analysis was performed by a second experienced reader (CZ) in a subset of 15 cases.

Data collection and follow-up

Data collection included age at sRT, International Society of Urologic Pathology Grading (ISUP), pathological T-, N-stage and status of surgical margins after RPE, PSA prior to sRT, site of recurrence (local, nodal or both), and administration and duration of ADT and sRT doses. Follow-up assessments included serum PSA testing at regular intervals based on the institutional clinical praxis.

Statistical analysis

The primary study endpoint was BRFS, defined as serum PSA > 0.2 ng/ml above the post-sRT nadir without initiation of additional salvage therapies or death of any cause. Descriptive statistics were performed with Excel 2016 (Microsoft Cooperation, USA) and GraphPad Prism v8.4.2 (GraphPad Software Inc, USA). Uni- and multivariate Cox-regression was performed with SPSS v27.0 (IBM, USA) to assess the impact of the different variables on BRFS.
Variables were dichotomized: ISUP < 3 and ≥ 3, pathological T stage < pT3 and ≥ pT3, pathological N stage pN + and pN-, positive surgical margin vs negative surgical margin, pre-sRT PSA < 0.5 ng/ml and > 0.5 ng/ml, presence and absence of local recurrence, presence and absence of nodal recurrence, administration or omission of ADT, RT to the pelvics, and dose delivered to the prostatic fossa/local recurrence (< and ≥ 72 Gy (α/β = 1.6 Gy)). Due to missing established threshold values, SUVmax values dichotomized < median and ≥ median as well as < 75% quartile (third quartile) and ≥ 75% quartile of the values of the cohort. Kaplan–Meier survival curves compared by log-rank test (GraphPad Prism v8.4.2, GraphPad Software Inc, USA) were used for analysis of the respective parameters. Thresholds of median and 75% quartile SUVmax values were applied separately for LR and NR lesions. The respective threshold of the whole cohort was applied for subgroup analysis. Time-dependent receiver-operating-characteristics (ROC) analysis was performed using R v4.1.2 [25]. Maximally selected rank statistic optimized for the log-rank test using R v4.1.2 [25] was performed to determine an optimal cut-off value for SUVmax.

Results

Patient characteristics

Two hundred thirty-five patients (Freiburg n = 39, TUM n = 56, LMU n = 64, Bologna n = 76) were included in the final analysis. Ninety-seven patients had LR only, 95 patients LN only, and 43 patients LR and NR in PSMA-PET. Fifty-one percent of patients received ADT. Fifty-nine percent of these patients received ADT over a duration of ≤ 12 months. Median follow-up was 24 months (IQR 16–41 months). No patient died during FU. See Table 1 for details.
Table 1
Patient characteristics
Cohort
All
 > 12-month follow-up
Number
235
 
197
 
Age (range)
71 (65–75)
 
71 (65–75)
 
ISUP grade
  1 + 2
45
19%
34
17%
  3–5
185
79%
159
81%
  n/a
5
2%
4
2%
pT-stage
  2a–c
75
32%
61
31%
  3–4
134
57%
112
57%
  n/a
26
11%
24
12%
Resection stage
  R0
98
42%
80
41%
  R1
73
31%
66
34%
  n/a
64
27%
51
26%
pN-stage
  pN0
130
55%
108
55%
  pN1
51
22%
42
21%
  n/a
54
23%
47
24%
PSA prior sRT
   < 0.5 ng/ml
66
28%
57
29%
   ≥ 0.5 ng/ml
164
70%
137
70%
  n/a
5
2%
3
2%
LR in PSMA-PET
140
60%
117
59%
NR in PSMA-PET
138
59%
114
58%
LR only in PSMA-PET
97
41%
83
42%
NR only in PSMA-PET
95
40%
80
41%
LR and NR in PSMA-PET
43
18%
34
17%
RT to the prostatic fossa/local recurrence(α/β = 1.6 Gy)
   < 70 Gy
162
69%
139
71%
   ≥ 70 Gy
33
14%
24
12
   ≥ 72 Gy
38
16%
32
16%
  n/a
2
1%
2
1%
RT to elective pelvics
154
60%
125
63%
n/a
31
13%
24
12%
ADT
120
51%
99
50%
  Of which > 12 months
49
41%
44
44%
  Of which ≤ 12 months FU
71
59%
55
56%
Abbreviation: ISUP, International Society of Urological Pathology; pT, pathological T stage; pN, pathological nodal stage; n/a, not available; PSA, prostate-specific antigen; sRT, salvage radiotherapy; LR, local recurrence; NR, nodal recurrence; PSMA-PET, positron-emission tomography targeting prostate-specific membrane antigen; ADT, androgen deprivation therapy
Median SUVmax for LR and NR was 7.6 (IQR 5.3–12.8) and 7.8 (IQR 4.3–17.5), respectively. The inter-observer analysis of 15 patients revealed significantly different volumes of manually segmented PET-positive LR lesions (median 2.7 ml (IQR 1.8–7.3 ml) vs 1.2 ml (IQR 0.5–4.1 ml), p < 0.001) but no significant differences between SUVmax values. See supplementary Fig. 1 for details.

Cox-regression

Two-year BRFS for all patients was 68%. For patients with LR and NR only, 2-year BRFS was 80% and 65%, respectively. See Fig. 2 for Kaplan–Meier-curves. In univariate analysis, established clinical and histopathological parameters of RPE were not significantly associated with BRFS. In univariate analysis, presence of LR (p = 0.016, HR 0.5 95%CI 0.3–0.9) and ADT (p =  < 0.001, HR 0.4 95%CI 0.2–0.7) was associated with more favorable BRFS and the presence of NR with unfavorable BRFS (p = 0.007, HR 2.1 (95%CI 1.2–3.5). In LR lesions, there was a trend for association of values ≥ median SUVmax (7.8) and unfavorable BRFS (p = 0.071), while SUVmax values ≥ 75% quartile (12.8) were significantly associated with unfavorable BRFS (p = 0.022, HR 2.3) (95%CI 1.1–4.6). In NR lesions, no significant association of values ≥ median or 75% of SUVmax (17.5) was observed. To further assess whether SUVmax values in LR are associated with BRFS, we performed a subgroup analysis with patients who only had LR (n = 97). This analysis showed again no significant association of classical clinical and histopathological parameters with BRFS, but a trend for association of values ≥ median SUVmax with unfavorable BRFS (p = 0.05, HR 2.6, 95%CI 1.0–6.9), while SUVmax values ≥ 75% quartile were significantly associated with unfavorable BRFS (p = 0.001, HR 4.6, 95%CI 1.9–11.5). ADT was not associated with BRFS in this cohort. See Table 2 and Fig. 3 for details.
Table 2
Univariate and multivariate Cox-regression
Endpoint: BRFS
Cohort
All (n = 235)
Only local recurrence (n = 97)
All > 12 months follow-up (n = 197)
Only local recurrence > 12 months follow-up (n = 83)
Variable
n = 235
p-value
HR (95%CI)
p-value
HR (95%CI)
p-value
HR (95%CI)
p-value
HR (95%CI)
Univariate
ISUP
0.139
1.7 (0.8–3.5)
0.198
2.3 (0.7–7.7)
0.089
2.0 (0.9–4.4)
0.311
1.9 (0.5–6.7)
pT-stage
0.104
1.6 (0.9–2.7)
0.179
2.0 (0.7–5.6)
0.053
1.8 (1.0–3.3)
0.150
2.3 (0.7–7.0)
pN-stage
0.476
1.2 (0.7–2.1)
0.799
0.8 (0.2–3.6)
0.157
1.5 (0.9–2.7)
0.578
0.7 (0.1–2.9)
positive margin
0.153
0.7 (0.4–1.2)
0.814
1.1 (0.4–3.1)
0.359
0.8 (0.4–1.4)
0.738
0.8 (0.3–2.5)
Pre-sRT PSA
0.103
1.6 (0.9–3.0)
0.404
1.6 (0.5–4.8)
0.226
1.5 (0.8–2.6)
0.335
1.9 (0.5–6.5)
ADT
 < 0.001
0.4 (0.2–0.7)
0.623
0.8 (0.3–2.0)
 < 0.001
0.4 (0.2–0.7)
0.593
0.8 (0.3–2.1)
LR
0.016
0.5 (0.3–0.9)
-
-
0.012
0.5 (0.3–0.9)
-
-
NR
0.007
2.1 (1.2–3.5)
-
-
0.005
2.2 (1.3–3.9)
-
-
RT to elective pelvics
0.107
1.7 (0.9–3.5)
0.945
1.0 (0.3–3.1)
0.125
1.7 (0.9–3.4)
0.916
1.1 (0.4–3.2)
Dose to LR ≥ 72 Gy
0.426
0.8 (0.4–1.5)
0.072
0.2 (0.0–1.2)
0.361
0.7 (0.3–1.5)
0.086
0.2 (0.0–1.3)
SUVmax in LR > median
0.071
2.0 (0.9–4.1)
0.050
2.6 (1.0–6.9)
0.087
2.0 (0.9–4.4)
0.150
2.1 (0.8–5.6)
SUVmax in LR > 75% IQR
0.022
2.3 (1.1–4.6)
 < 0.001
4.6 (1.9–11.5)
0.049
2.1 (1.0–4.4)
0.005
3.9 (1.5–10.1)
SUVmax in NR > median
0.640
1.1 (0.6–2.0)
-
-
0.786
1.1 (0.6–2.0)
-
-
SUVmax in NR > 75% IQR
0.895
1.0 (0.5–1.8)
-
-
0.799
0.9 (0.5–1.8)
-
-
Multivariate
SUVmax LR 75% IQR
0.022
2.3 (1.1–4.6)
-
-
0.049
2.1 (1.0–4.4)
-
-
ADT
ns
 
-
-
ns
 
-
-
LR
ns
 
-
-
ns
 
-
-
NR
ns
 
-
-
ns
 
-
-
Abbreviations: BRFS, biochemical recurrence free survival; ADT, androgen deprivation therapy; ISUP, International Society of Urological Pathology Grading; pT, pathological T-stage; pN, pathological nodal stage; sRT, salvage radiotherapy; PSA, prostate-specific antigen; LR, presence of local recurrence; NR, presence of nodal recurrence; Gy, gray; SUVmax, maximal standardized uptake value; HR, hazard ratio; 95%CI, 95% confidence interval; ns, non-significant bold indicates statistical significance
To analyze robustness and consider the confounding factor of short-term ADT, a sensitivity analysis with patients with a FU of > 12 months (n = 197, median FU 27 months IQR 20–43) was performed confirming the results showing no significant association of clinical and histopathological parameters with BRFS, presence of LR being significantly associated with favorable BRFS (p = 0.012, HR 0.5 95%CI 0.3–0.9), and presence of NR (p = 0.005, HR 2.2 95%CI 1.3–3.9) and SUVmax values ≥ 75% quartile in LR being associated with unfavorable BRFS (p = 0.041, HR 2.2 95%CI 1.0–4.6). Sensitivity analysis of patients with LR only and > 12 months FU (n = 83) showed a strong association of SUVmax values ≥ 75% in LR quartile with unfavorable BRFS (p = 0.005, HR 3.9 95%CI 1.5–10.1). See Table 2 and Fig. 3.
In multivariate analysis, SUVmax values in LR ≥ 75% quartile stayed significantly associated with unfavorable BRFS in the cohorts including all patients (p = 0.022) and in patients with > 12 months FU (p = 0.041). Presence of LR or NR in PET and administration of ADT was not significantly associated with BRFS in multivariate analysis. See Table 2.

Time-dependent ROC

Time-dependent ROC analysis of SUVmax values of patients with > 12 months FU and > 12 months FU and LR only yielded a concordance index (C-index) of 0.66 (95%CI 0.54–0.78) and 0.71 (95%CI 0.57–0.85), respectively. Prediction improved at 18 months FU. SUVmax cut-off values determined by the maximally selected rank statistic were 11.8 for all patients and 13.0 for patients with > 12 months FU in all patients and in patients with LR only, respectively. See Fig. 4 for details and supplementary Table 2 for further analyses.

Discussion

SRT is the last curative option for patients with recurrent or persistent PCa after surgery, but heterogeneous responses demonstrate the need for improved and differentiated risk stratification and subsequently appropriate adaptions in disease management. While implementation of PSMA-PET led to relevant improvements in disease localization, with results of prospective trials pending, it is not yet clear whether the sole spatial information of tumor burden and the accompanying changes in treatment management have a relevant impact on progression rates. Considering that PSMA-PET-positive findings might only represent the “tip of the iceberg,” the additional biological information provided by this molecular imaging bears great potential. To our knowledge, this is the first large multicenter retrospective study to evaluate the potential of SUVmax values in PSMA-PET as a new biomarker in patients with PCa persistence/recurrence. The findings from this study suggest that SUVmax values may significantly contribute to identify patients who are at higher risk for progression after sRT and therefore might benefit from treatment intensification, guiding personalized treatment approaches.
In contrary to prospective trials investigating sRT, most of the patients included in our study are likely to be at advanced recurrent disease stages with 70% having a PSA ≥ 0.5 ng/ml prior to sRT and 59% having nodal recurrence. Therefore, the comparison with data of recently published randomized controlled trials [2628] is limited. Nevertheless, sRT in fossa-confined patients yielded BRFS in 80% after 2 years. Considering slightly different definitions of endpoints, these results are comparable to freedom from biochemical failure in approximately 50–70% for patients with a pre-sRT PSA between 0.2 and 2.0 ng/ml in a large retrospective study of conventionally staged patients [5, 29]. BRFS rates for patients with node-positive PSMA-PET findings dropped dramatically to 65% after 2 years, which is in line with findings from a prospective trial, reporting 3-year BRFS rates of 45% with PSMA-PET-positive disease outside the prostatic fossa [30]. Our results thereby confirm that PSMA-PET findings are highly prognostic for BRFS.
In our retrospective analysis, presence of PSMA-PET-positive local and nodal recurrence positive on PSMA-PET were prognosticators for BRFS after sRT, whereas classical pathological and clinical parameters were not. Since Emmett et al. prospectively demonstrated the prognostic value of PET-positive findings, it is likely that they dominate established parameters in our cohort, since we included only patients with PSMA-PET-positive lesions. The improved BRFS associated with the presence of LR (HR 0.5, p = 0.016) is explainable by the dose escalation of PET-positive LRs yielding sufficient RT dose coverage, which was previously reported to be beneficial [10].
Despite favorable outcomes for patients with LR only, still nearly one-third of patients suffered from progression after sRT. In multivariate analysis, our results demonstrate a significant association of SUVmax ≥ 75% quartile (HR 2.3, p = 0.022) with unfavorable BRFS in this subgroup. Sensitivity analysis confirmed these results with a HR of 3.9 in patients with LR only and FU of > 12 months. These findings are in line with the biological understanding of PSMA, with high PSMA-expression being associated with more aggressive disease [31] and SUVmax correlating with PSMA-expression [32]. Therefore, patients with LR and high SUVmax values might represent a subgroup with more aggressive PCa, potentially suffering from micro metastases outside the prostatic bed at the time of imaging and therefore benefiting from intensified treatments. Cox-regression and ROC analysis furthermore suggest that SUVmax in LR might be a valuable prognosticator in patients with both LRs and NR. However, these results need to be interpreted carefully, since ADT is administered more often and for a longer period of time in patients with NR (44% in our cohort).
Whether extraction of additional radiomic features from PSMA-PET images enables identification of additional prognosticators [33] needs to be evaluated in future studies. However, implementation of SUVmax offers great potential in this scenario, since it is easily and non-invasively determinable with minimal resources and without additional costs and is not affected by interobserver variability [34]. AUC values of the time-dependent ROC analysis showed the best discrimination in patients with > 12 months FU and LR only. In an exploratory analysis, we calculated SUVmax cut-off value for optimal discrimination. In all patients, the optimized cut-off value was slightly lower than the 75% quartile (11.8 vs 12.8). In the subgroup with FU > 12 months, the cut-off value was, however, more similar (13.0). Thus, a SUVmax threshold of approximately 13.0 should be validated in future studies in PSMA-PET imaging. This being said, the role of SUVmax in these patient subgroups needs to be evaluated in future studies including new tracers to validate putative cut-off values and design studies, which evaluate treatment intensification such as extension of RT fields to elective nodes or intensified systemic treatments. To define optimal RT fields, patterns of metastases need to be vigorously analyzed. Intensification of systemic treatments in sRT is currently investigated by the FORMULA-059 RCT (NCT03141671). Keeping in mind that sRT should be initiated at low PSA levels, implementation of SUVmax into risk stratification might even be relevant in this scenario, with approximately 50% and 65% of patients having PET-positive findings at PSA values < 0.2 ng/ml and between 0.2 and 0.5 ng/ml [30]. Furthermore, administration of ADT was associated with favorable BRFS in the entire cohort in univariate analysis, but not in multivariate analysis. Additionally, ADT was not associated with BRFS in patients with LR only. Despite RCTs demonstrated a benefit of adding short-term ADT to sRT [35], our results show that these findings cannot directly be transferred into the PSMA-PET era. Our findings suggest that patients with recurrence confined to the prostatic fossa in PSMA-PET might not benefit from systemic, but rather from local treatment intensification. Considering recent results of the multicenter retrospective SPIDER 01 (Abstract OC-0607), we therefore evaluated the effect of doses ≥ 72 Gy to the PSMA-PET-defined local recurrence. We could not identify a statistical significant difference but a trend for favorable BRFS in patients with LR only, who received dose escalation ≥ 72 Gy (p = 0.086). However, this analysis is hampered by the relatively small number of patients receiving dose escalation and short follow-up. Future studies are needed to evaluate this aspect in depth.
Interestingly presence of NR was associated with significantly unfavorable BRFS (HR 2.1) but not SUVmax values in NR, suggesting that the additional biologic information provided by SUVmax values does not contribute to this patient subgroup, who already suffer from relevantly poorer prognosis. These patients might benefit from systemic treatment intensification, since despite dose escalation to PET-positive nodes, it is likely that sRT might not cover non-visible tumor spread.
Our study has some limitations. First, due to its retrospective design, protocols for PSMA-PET scans, sRT, and follow-up varied between centers and are prone to selection bias. Since the tracer kinetics depends on the time between injection and image acquisition, we want to point out that all scans were acquired in line with recent guidelines in all centers, but use of different PET scanners might affect comparability. Second, it is known that different reconstruction methods such as time-of-flight and point-spread-function have implication to SUV values. Most importantly, SUV values might be underestimated when taken from small lesions, i.e., sub-centimeter. In order to quantify this aspect, we analyzed SUVmax values with respect to lesion volume. We found a tendency of smaller SUVmax values for lesions < 1 ml, which could only be found in a minority of patients (n = 14). Thus, we are convinced that this limitation is very limited towards the overall results. Furthermore, there are many PSMA-ligands on the market and to date both 68 Ga-PSMA-11 and 18F-DCFPyL received FDA approval, with different pharmacokinetics and pharmacodynamics as well as isotopes, raising questions about the reproducibility of SUV values. In our study, we employed only 68 Ga-PSMA-11, in order to generate more standardized data [36]. Therefore, the different reconstruction algorithms between the different institutions pose the risk for deviation. In order to limit the inter-institution variability, we only employed PET/CT systems with EARL1 accreditations. However, this heterogeneity can also be regarded as a strength of our retrospective analysis as it makes the data more applicable in clinical routine when different scanners and reconstructions are used between different centers. Third, no central review of PET images was performed with potential differences in interpretation between centers. Fourth, the median FU is relatively short with a median FU of 24 months. Lastly pathological data from RPE was missing in up to 27% of patients, likely contributing to inferior Cox-regression results.

Conclusion

Our study is the first to demonstrate that the SUVmax value is a promising new non-invasive biomarker to prognosticate outcome in patients undergoing sRT and recurrence confined to the prostatic fossa ± nodal recurrences in PSMA-PET. Its addition might contribute to improve risk stratification of patients with recurrent PCa and to guide personalized treatment decisions.

Declarations

Competing interests

ME reports prior consulting activities for BED, Novartis, Telix, Progenics, Bayer, Point Biopharma, and Janssen and a patent application for rhPSMA. All other authors declare no conflicts of interest regarding this study.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Radiologie

Kombi-Abonnement

Mit e.Med Radiologie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Radiologie, den Premium-Inhalten der radiologischen Fachzeitschriften, inklusive einer gedruckten Radiologie-Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Stephenson AJ, Scardino PT, Kattan MW, Pisansky TM, Slawin KM, Klein EA, et al. Predicting the outcome of salvage radiation therapy for recurrent prostate cancer after radical prostatectomy. J Clin Oncol. 2007;25(15):2035–41.CrossRefPubMed Stephenson AJ, Scardino PT, Kattan MW, Pisansky TM, Slawin KM, Klein EA, et al. Predicting the outcome of salvage radiation therapy for recurrent prostate cancer after radical prostatectomy. J Clin Oncol. 2007;25(15):2035–41.CrossRefPubMed
2.
Zurück zum Zitat Stephenson AJ, Shariat SF, Zelefsky MJ, Kattan MW, Butler EB, Teh BS, et al. Salvage radiotherapy for recurrent prostate cancer after radical prostatectomy. JAMA. 2004;291(11):1325–32.CrossRefPubMed Stephenson AJ, Shariat SF, Zelefsky MJ, Kattan MW, Butler EB, Teh BS, et al. Salvage radiotherapy for recurrent prostate cancer after radical prostatectomy. JAMA. 2004;291(11):1325–32.CrossRefPubMed
3.
Zurück zum Zitat Wiegel T, Bartkowiak D, Bottke D, Bronner C, Steiner U, Siegmann A, et al. Adjuvant radiotherapy versus wait-and-see after radical prostatectomy: 10-year follow-up of the ARO 96–02/AUO AP 09/95 trial. Eur Urol. 2014;66(2):243–50.CrossRefPubMed Wiegel T, Bartkowiak D, Bottke D, Bronner C, Steiner U, Siegmann A, et al. Adjuvant radiotherapy versus wait-and-see after radical prostatectomy: 10-year follow-up of the ARO 96–02/AUO AP 09/95 trial. Eur Urol. 2014;66(2):243–50.CrossRefPubMed
4.
Zurück zum Zitat Cornford P, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer. Part II-2020 update: treatment of relapsing and metastatic prostate cancer. Eur Urol. 2021;79(2):263–82.CrossRefPubMed Cornford P, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, et al. EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer. Part II-2020 update: treatment of relapsing and metastatic prostate cancer. Eur Urol. 2021;79(2):263–82.CrossRefPubMed
5.
Zurück zum Zitat Tendulkar RD, Agrawal S, Gao T, Efstathiou JA, Pisansky TM, Michalski JM, et al. Contemporary update of a multi-institutional predictive nomogram for salvage radiotherapy after radical prostatectomy. J Clin Oncol. 2016;34(30):3648–54.CrossRefPubMed Tendulkar RD, Agrawal S, Gao T, Efstathiou JA, Pisansky TM, Michalski JM, et al. Contemporary update of a multi-institutional predictive nomogram for salvage radiotherapy after radical prostatectomy. J Clin Oncol. 2016;34(30):3648–54.CrossRefPubMed
6.
Zurück zum Zitat Banerjee SR, Pullambhatla M, Byun Y, Nimmagadda S, Green G, Fox JJ, et al. 68Ga-labeled inhibitors of prostate-specific membrane antigen (PSMA) for imaging prostate cancer. J Med Chem. 2010;53(14):5333–41.CrossRefPubMedPubMedCentral Banerjee SR, Pullambhatla M, Byun Y, Nimmagadda S, Green G, Fox JJ, et al. 68Ga-labeled inhibitors of prostate-specific membrane antigen (PSMA) for imaging prostate cancer. J Med Chem. 2010;53(14):5333–41.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Fendler WP, Calais J, Eiber M, Flavell RR, Mishoe A, Feng FY, et al. Assessment of 68Ga-PSMA-11 PET accuracy in localizing recurrent prostate cancer: A prospective single-arm clinical trial. JAMA Oncol. 2019;5(6):856–63.CrossRefPubMedPubMedCentral Fendler WP, Calais J, Eiber M, Flavell RR, Mishoe A, Feng FY, et al. Assessment of 68Ga-PSMA-11 PET accuracy in localizing recurrent prostate cancer: A prospective single-arm clinical trial. JAMA Oncol. 2019;5(6):856–63.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Farolfi A, Ceci F, Castellucci P, Graziani T, Siepe G, Lambertini A, et al. (68)Ga-PSMA-11 PET/CT in prostate cancer patients with biochemical recurrence after radical prostatectomy and PSA < 0.5 ng/ml. Efficacy and impact on treatment strategy. Eur J Nucl Med Mol Imaging. 2019;46(1):11–9.CrossRefPubMed Farolfi A, Ceci F, Castellucci P, Graziani T, Siepe G, Lambertini A, et al. (68)Ga-PSMA-11 PET/CT in prostate cancer patients with biochemical recurrence after radical prostatectomy and PSA < 0.5 ng/ml. Efficacy and impact on treatment strategy. Eur J Nucl Med Mol Imaging. 2019;46(1):11–9.CrossRefPubMed
9.
Zurück zum Zitat Schmidt-Hegemann N-S, Eze C, Li M, Rogowski P, Schaefer C, Stief C, et al. Impact of 68Ga-PSMA PET/CT on the radiotherapeutic approach to prostate cancer in comparison to CT: A retrospective analysis. J Nucl Med. 2019;60(7):963–70.CrossRefPubMedPubMedCentral Schmidt-Hegemann N-S, Eze C, Li M, Rogowski P, Schaefer C, Stief C, et al. Impact of 68Ga-PSMA PET/CT on the radiotherapeutic approach to prostate cancer in comparison to CT: A retrospective analysis. J Nucl Med. 2019;60(7):963–70.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Schmidt-Hegemann N-S, Zamboglou C, Thamm R, Eze C, Kirste S, Spohn S, et al. A multi-institutional analysis of prostate cancer patients with or without 68Ga-PSMA PET/CT prior to salvage radiotherapy of the prostatic fossa. Front Oncol. 2021;11:723536. Published 2021 Oct 1. https://doi.org/10.3389/fonc.2021.723536 Schmidt-Hegemann N-S, Zamboglou C, Thamm R, Eze C, Kirste S, Spohn S, et al. A multi-institutional analysis of prostate cancer patients with or without 68Ga-PSMA PET/CT prior to salvage radiotherapy of the prostatic fossa. Front Oncol. 2021;11:723536. Published 2021 Oct 1. https://​doi.​org/​10.​3389/​fonc.​2021.​723536
11.
Zurück zum Zitat Han S, Woo S, Kim YJ, Suh CH. Impact of (68)Ga-PSMA PET on the management of patients with prostate cancer: A systematic review and meta-analysis. Eur Urol. 2018;74(2):179–90.CrossRefPubMed Han S, Woo S, Kim YJ, Suh CH. Impact of (68)Ga-PSMA PET on the management of patients with prostate cancer: A systematic review and meta-analysis. Eur Urol. 2018;74(2):179–90.CrossRefPubMed
12.
Zurück zum Zitat Schmidt Hegemann NS, Rogowski P, Eze C, Schäfer C, Stief C, Lang S, et al. Outcome after 68Ga-PSMA-11 versus choline PET-based salvage radiotherapy in patients with biochemical recurrence of prostate cancer: A matched-pair analysis. Cancers (Basel). 2020;12(11):3395. Published 2020 Nov 16. https://doi.org/10.3390/cancers12113395 Schmidt Hegemann NS, Rogowski P, Eze C, Schäfer C, Stief C, Lang S, et al. Outcome after 68Ga-PSMA-11 versus choline PET-based salvage radiotherapy in patients with biochemical recurrence of prostate cancer: A matched-pair analysis. Cancers (Basel). 2020;12(11):3395. Published 2020 Nov 16. https://​doi.​org/​10.​3390/​cancers12113395
13.
Zurück zum Zitat Schmidt-Hegemann N-S, Stief C, Kim T-H, Eze C, Kirste S, Strouthos I, et al. Outcome after PSMA PET/CT based salvage radiotherapy in patients with biochemical recurrence after radical prostatectomy: A bi-institutional retrospective analysis. J Nucl Med. 2019;60(2):227-233. https://doi.org/10.2967/jnumed.118.212563 Schmidt-Hegemann N-S, Stief C, Kim T-H, Eze C, Kirste S, Strouthos I, et al. Outcome after PSMA PET/CT based salvage radiotherapy in patients with biochemical recurrence after radical prostatectomy: A bi-institutional retrospective analysis. J Nucl Med. 2019;60(2):227-233. https://​doi.​org/​10.​2967/​jnumed.​118.​212563
14.
Zurück zum Zitat Calais J, Czernin J, Fendler WP, Elashoff D, Nickols NG. Randomized prospective phase III trial of (68)Ga-PSMA-11 PET/CT molecular imaging for prostate cancer salvage radiotherapy planning [PSMA-SRT]. BMC Cancer. 2019;19(1):18.CrossRefPubMedPubMedCentral Calais J, Czernin J, Fendler WP, Elashoff D, Nickols NG. Randomized prospective phase III trial of (68)Ga-PSMA-11 PET/CT molecular imaging for prostate cancer salvage radiotherapy planning [PSMA-SRT]. BMC Cancer. 2019;19(1):18.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Jiao J, Kang F, Zhang J, Quan Z, Wen W, Zhao X, et al. Establishment and prospective validation of an SUV(max) cutoff value to discriminate clinically significant prostate cancer from benign prostate diseases in patients with suspected prostate cancer by (68)Ga-PSMA PET/CT: A real-world study. Theranostics. 2021;11(17):8396–411.CrossRefPubMedPubMedCentral Jiao J, Kang F, Zhang J, Quan Z, Wen W, Zhao X, et al. Establishment and prospective validation of an SUV(max) cutoff value to discriminate clinically significant prostate cancer from benign prostate diseases in patients with suspected prostate cancer by (68)Ga-PSMA PET/CT: A real-world study. Theranostics. 2021;11(17):8396–411.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Roberts MJ, Morton A, Donato P, Kyle S, Pattison DA, Thomas P, et al. (68)Ga-PSMA PET/CT tumour intensity pre-operatively predicts adverse pathological outcomes and progression-free survival in localised prostate cancer. Eur J Nucl Med Mol Imaging. 2021;48(2):477–82.CrossRefPubMed Roberts MJ, Morton A, Donato P, Kyle S, Pattison DA, Thomas P, et al. (68)Ga-PSMA PET/CT tumour intensity pre-operatively predicts adverse pathological outcomes and progression-free survival in localised prostate cancer. Eur J Nucl Med Mol Imaging. 2021;48(2):477–82.CrossRefPubMed
17.
Zurück zum Zitat Uprimny C, Kroiss AS, Decristoforo C, Fritz J, von Guggenberg E, Kendler D, et al. (68)Ga-PSMA-11 PET/CT in primary staging of prostate cancer: PSA and Gleason score predict the intensity of tracer accumulation in the primary tumour. Eur J Nucl Med Mol Imaging. 2017;44(6):941–9.CrossRefPubMed Uprimny C, Kroiss AS, Decristoforo C, Fritz J, von Guggenberg E, Kendler D, et al. (68)Ga-PSMA-11 PET/CT in primary staging of prostate cancer: PSA and Gleason score predict the intensity of tracer accumulation in the primary tumour. Eur J Nucl Med Mol Imaging. 2017;44(6):941–9.CrossRefPubMed
18.
Zurück zum Zitat Wang Z, Zheng A, Li Y, Dong W, Liu X, Yuan W, et al. 18F-PSMA-1007 PET/CT performance on risk stratification discrimination and distant metastases prediction in newly diagnosed prostate cancer. Front Oncol. 2021;11:759053. Published 2021 Oct 28. https://doi.org/10.3389/fonc.2021.759053 Wang Z, Zheng A, Li Y, Dong W, Liu X, Yuan W, et al. 18F-PSMA-1007 PET/CT performance on risk stratification discrimination and distant metastases prediction in newly diagnosed prostate cancer. Front Oncol. 2021;11:759053. Published 2021 Oct 28. https://​doi.​org/​10.​3389/​fonc.​2021.​759053
19.
Zurück zum Zitat Fendler WP, Eiber M, Beheshti M, Bomanji J, Ceci F, Cho S, et al. (68)Ga-PSMA PET/CT: Joint EANM and SNMMI procedure guideline for prostate cancer imaging: Version 1.0. Eur J Nucl Med Mol Imaging. 2017;44(6):1014–24.CrossRefPubMed Fendler WP, Eiber M, Beheshti M, Bomanji J, Ceci F, Cho S, et al. (68)Ga-PSMA PET/CT: Joint EANM and SNMMI procedure guideline for prostate cancer imaging: Version 1.0. Eur J Nucl Med Mol Imaging. 2017;44(6):1014–24.CrossRefPubMed
20.
Zurück zum Zitat Zamboglou C, Carles M, Fechter T, Kiefer S, Reichel K, Fassbender TF, et al. Radiomic features from PSMA PET for non-invasive intraprostatic tumor discrimination and characterization in patients with intermediate- and high-risk prostate cancer - a comparison study with histology reference. Theranostics. 2019;9(9):2595–605.CrossRefPubMedPubMedCentral Zamboglou C, Carles M, Fechter T, Kiefer S, Reichel K, Fassbender TF, et al. Radiomic features from PSMA PET for non-invasive intraprostatic tumor discrimination and characterization in patients with intermediate- and high-risk prostate cancer - a comparison study with histology reference. Theranostics. 2019;9(9):2595–605.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Ceci F, Oprea-Lager DE, Emmett L, Adam JA, Bomanji J, Czernin J, et al. E-PSMA: the EANM standardized reporting guidelines v1.0 for PSMA-PET. Eur J Nucl Med Mol Imaging. 2021;48(5):1626–38.CrossRefPubMedPubMedCentral Ceci F, Oprea-Lager DE, Emmett L, Adam JA, Bomanji J, Czernin J, et al. E-PSMA: the EANM standardized reporting guidelines v1.0 for PSMA-PET. Eur J Nucl Med Mol Imaging. 2021;48(5):1626–38.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Eiber M, Herrmann K, Calais J, Hadaschik B, Giesel FL, Hartenbach M, et al. Prostate cancer molecular imaging standardized evaluation (PROMISE): proposed miTNM classification for the interpretation of PSMA-ligand PET/CT. J Nucl Med. 2018;59(3):469–78.CrossRefPubMed Eiber M, Herrmann K, Calais J, Hadaschik B, Giesel FL, Hartenbach M, et al. Prostate cancer molecular imaging standardized evaluation (PROMISE): proposed miTNM classification for the interpretation of PSMA-ligand PET/CT. J Nucl Med. 2018;59(3):469–78.CrossRefPubMed
23.
Zurück zum Zitat Fedorov A, Beichel R, Kalpathy-Cramer J, Finet J, Fillion-Robin J-C, Pujol S, et al. 3D Slicer as an image computing platform for the Quantitative Imaging Network. Magn Reson Imaging. 2012;30(9):1323–41.CrossRefPubMedPubMedCentral Fedorov A, Beichel R, Kalpathy-Cramer J, Finet J, Fillion-Robin J-C, Pujol S, et al. 3D Slicer as an image computing platform for the Quantitative Imaging Network. Magn Reson Imaging. 2012;30(9):1323–41.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Zamboglou C, Fassbender TF, Steffan L, Schiller F, Fechter T, Carles M, et al. Validation of different PSMA-PET/CT-based contouring techniques for intraprostatic tumor definition using histopathology as standard of reference. Radiother Oncol. 2019;141:208–13.CrossRefPubMed Zamboglou C, Fassbender TF, Steffan L, Schiller F, Fechter T, Carles M, et al. Validation of different PSMA-PET/CT-based contouring techniques for intraprostatic tumor definition using histopathology as standard of reference. Radiother Oncol. 2019;141:208–13.CrossRefPubMed
25.
Zurück zum Zitat Team RC. R: a language and environment for statistical computing. Vienna: R Foundation for Statistical Computing; 2021. Team RC. R: a language and environment for statistical computing. Vienna: R Foundation for Statistical Computing; 2021.
26.
Zurück zum Zitat Kneebone A, Fraser-Browne C, Duchesne GM, Fisher R, Frydenberg M, Herschtal A, et al. Adjuvant radiotherapy versus early salvage radiotherapy following radical prostatectomy (TROG 08.03/ANZUP RAVES): a randomised, controlled, phase 3, non-inferiority trial. Lancet Oncol. 2020;21(10):1331–40.CrossRefPubMed Kneebone A, Fraser-Browne C, Duchesne GM, Fisher R, Frydenberg M, Herschtal A, et al. Adjuvant radiotherapy versus early salvage radiotherapy following radical prostatectomy (TROG 08.03/ANZUP RAVES): a randomised, controlled, phase 3, non-inferiority trial. Lancet Oncol. 2020;21(10):1331–40.CrossRefPubMed
27.
Zurück zum Zitat Parker CC, Clarke NW, Cook AD, Kynaston HG, Petersen PM, Catton C, et al. Timing of radiotherapy after radical prostatectomy (RADICALS-RT): a randomised, controlled phase 3 trial. Lancet. 2020;396(10260):1413–21.CrossRefPubMed Parker CC, Clarke NW, Cook AD, Kynaston HG, Petersen PM, Catton C, et al. Timing of radiotherapy after radical prostatectomy (RADICALS-RT): a randomised, controlled phase 3 trial. Lancet. 2020;396(10260):1413–21.CrossRefPubMed
28.
Zurück zum Zitat Sargos P, Chabaud S, Latorzeff I, Magné N, Benyoucef A, Supiot S, et al. Adjuvant radiotherapy versus early salvage radiotherapy plus short-term androgen deprivation therapy in men with localised prostate cancer after radical prostatectomy (GETUG-AFU 17): A randomised, phase 3 trial. Lancet Oncol. 2020;21(10):1341–52.CrossRefPubMed Sargos P, Chabaud S, Latorzeff I, Magné N, Benyoucef A, Supiot S, et al. Adjuvant radiotherapy versus early salvage radiotherapy plus short-term androgen deprivation therapy in men with localised prostate cancer after radical prostatectomy (GETUG-AFU 17): A randomised, phase 3 trial. Lancet Oncol. 2020;21(10):1341–52.CrossRefPubMed
29.
Zurück zum Zitat Pisansky TM, Agrawal S, Hamstra DA, Koontz BF, Liauw SL, Efstathiou JA, et al. Salvage radiation therapy dose response for biochemical failure of prostate cancer after prostatectomy-a multi-institutional observational study. Int J Radiat Oncol Biol Phys. 2016;96(5):1046–53.CrossRefPubMed Pisansky TM, Agrawal S, Hamstra DA, Koontz BF, Liauw SL, Efstathiou JA, et al. Salvage radiation therapy dose response for biochemical failure of prostate cancer after prostatectomy-a multi-institutional observational study. Int J Radiat Oncol Biol Phys. 2016;96(5):1046–53.CrossRefPubMed
30.
Zurück zum Zitat Emmett L, Tang R, Nandurkar R, Hruby G, Roach P, Watts JA, et al. 3-year freedom from progression after 68Ga-PSMA PET/CT–triaged management in men with biochemical recurrence after radical prostatectomy: Results of a prospective multicenter trial. J Nucl Med. 2020;61(6):866–72.CrossRefPubMed Emmett L, Tang R, Nandurkar R, Hruby G, Roach P, Watts JA, et al. 3-year freedom from progression after 68Ga-PSMA PET/CT–triaged management in men with biochemical recurrence after radical prostatectomy: Results of a prospective multicenter trial. J Nucl Med. 2020;61(6):866–72.CrossRefPubMed
31.
Zurück zum Zitat Hupe MC, Philippi C, Roth D, Kümpers C, Ribbat-Idel J, Becker F, et al. Expression of prostate-specific membrane antigen (PSMA) on biopsies is an independent risk stratifier of prostate cancer patients at time of initial diagnosis. Front Oncol. 2018;8:623. Published 2018 Dec 20. https://doi.org/10.3389/fonc.2018.00623 Hupe MC, Philippi C, Roth D, Kümpers C, Ribbat-Idel J, Becker F, et al. Expression of prostate-specific membrane antigen (PSMA) on biopsies is an independent risk stratifier of prostate cancer patients at time of initial diagnosis. Front Oncol. 2018;8:623. Published 2018 Dec 20. https://​doi.​org/​10.​3389/​fonc.​2018.​00623
32.
Zurück zum Zitat Woythal N, Arsenic R, Kempkensteffen C, Miller K, Janssen JC, Huang K, et al. Immunohistochemical validation of PSMA expression measured by (68)Ga-PSMA PET/CT in primary prostate cancer. J Nucl Med. 2018;59(2):238–43.CrossRefPubMed Woythal N, Arsenic R, Kempkensteffen C, Miller K, Janssen JC, Huang K, et al. Immunohistochemical validation of PSMA expression measured by (68)Ga-PSMA PET/CT in primary prostate cancer. J Nucl Med. 2018;59(2):238–43.CrossRefPubMed
33.
Zurück zum Zitat Spohn SKB, Bettermann AS, Bamberg F, Benndorf M, Mix M, Nicolay NH, 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):8027–42.CrossRefPubMedPubMedCentral Spohn SKB, Bettermann AS, Bamberg F, Benndorf M, Mix M, Nicolay NH, 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):8027–42.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Miksch J, Bottke D, Krohn T, Thamm R, Bartkowiak D, Solbach C, et al. Interobserver variability, detection rate, and lesion patterns of 68Ga-PSMA-11-PET/CT in early-stage biochemical recurrence of prostate cancer after radical prostatectomy. Eur J Nucl Med Mol Imaging. 2020;47(10):2339–47.CrossRefPubMedPubMedCentral Miksch J, Bottke D, Krohn T, Thamm R, Bartkowiak D, Solbach C, et al. Interobserver variability, detection rate, and lesion patterns of 68Ga-PSMA-11-PET/CT in early-stage biochemical recurrence of prostate cancer after radical prostatectomy. Eur J Nucl Med Mol Imaging. 2020;47(10):2339–47.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Carrie C, Magné N, Burban-Provost P, Sargos P, Latorzeff I, Lagrange JL, et al. Short-term androgen deprivation therapy combined with radiotherapy as salvage treatment after radical prostatectomy for prostate cancer (GETUG-AFU 16): A 112-month follow-up of a phase 3, randomised trial. Lancet Oncol. 2019;20(12):1740–9.CrossRefPubMed Carrie C, Magné N, Burban-Provost P, Sargos P, Latorzeff I, Lagrange JL, et al. Short-term androgen deprivation therapy combined with radiotherapy as salvage treatment after radical prostatectomy for prostate cancer (GETUG-AFU 16): A 112-month follow-up of a phase 3, randomised trial. Lancet Oncol. 2019;20(12):1740–9.CrossRefPubMed
36.
Zurück zum Zitat Munk OL, Tolbod LP, Hansen SB, Bogsrud TV. Point-spread function reconstructed PET images of sub-centimeter lesions are not quantitative. EJNMMI Phys. 2017;4(1):5.CrossRefPubMedPubMedCentral Munk OL, Tolbod LP, Hansen SB, Bogsrud TV. Point-spread function reconstructed PET images of sub-centimeter lesions are not quantitative. EJNMMI Phys. 2017;4(1):5.CrossRefPubMedPubMedCentral
Metadaten
Titel
The maximum standardized uptake value in patients with recurrent or persistent prostate cancer after radical prostatectomy and PSMA-PET-guided salvage radiotherapy—a multicenter retrospective analysis
verfasst von
Simon K. B. Spohn
Andrea Farolfi
Sarah Schandeler
Marco M. E. Vogel
Juri Ruf
Michael Mix
Simon Kirste
Francesco Ceci
Stefano Fanti
Helena Lanzafame
Francesca Serani
Christian Gratzke
August Sigle
Stephanie E. Combs
Denise Bernhardt
Juergen E. Gschwend
Josef A. Buchner
Christian Trapp
Claus Belka
Peter Bartenstein
Lena Unterrainer
Marcus Unterrainer
Matthias Eiber
Stephan G. Nekolla
Kilian Schiller
Anca L. Grosu
Nina-Sophie Schmidt-Hegemann
Constantinos Zamboglou
Jan C. Peeken
Publikationsdatum
19.08.2022
Verlag
Springer Berlin Heidelberg
Erschienen in
European Journal of Nuclear Medicine and Molecular Imaging / Ausgabe 1/2022
Print ISSN: 1619-7070
Elektronische ISSN: 1619-7089
DOI
https://doi.org/10.1007/s00259-022-05931-5

Weitere Artikel der Ausgabe 1/2022

European Journal of Nuclear Medicine and Molecular Imaging 1/2022 Zur Ausgabe