Skip to main content
Erschienen in: Strahlentherapie und Onkologie 11/2020

Open Access 12.05.2020 | Original Article

Efficacy of repeated PSMA PET-directed radiotherapy for oligorecurrent prostate cancer after initial curative therapy

verfasst von: PD Dr. med. Christoph Henkenberens, Ann-Kathrin Oehus, Thorsten Derlin, Frank Bengel, Tobias L. Ross, Markus A. Kuczyk, Stefan Janssen, Hans Christiansen, Christoph A. J. von Klot

Erschienen in: Strahlentherapie und Onkologie | Ausgabe 11/2020

Abstract

Purpose

To assess the outcome of prostate cancer (PCa) patients diagnosed with oligorecurrent disease and treated with a first and a second PSMA (prostate-specific membrane antigen ligand) PET(positron-emission tomography)-directed radiotherapy (RT).

Patients and methods

Thirty-two patients with oligorecurrent relapse after curative therapy received a first PSMA PET-directed RT of all metastases. After biochemical progression, all patients received a second PSMA PET-directed RT of all metastases. The main outcome parameters were biochemical progression-free survival (bPFS) and androgen deprivation therapy-free survival (ADT-FS). The intervals of BPFS were analyzed separately as follows: the interval from the last day of PSMA PET-directed RT to the first biochemical progression was defined as bPFS_1 and the interval from second PSMA PET-directed RT to further biochemical progression was defined as bPFS_2.

Results

The median follow-up duration was 39.5 months (18–60). One out of 32 (3.1%) patients died after 47 months of progressive metastatic prostate cancer (mPCa). All patients showed biochemical responses after the first PSMA PET-directed RT and the median prostate-specific antigen (PSA) level before RT was 1.70 ng/mL (0.2–3.8), which decreased significantly to a median PSA nadir level of 0.39 ng/mL (range <0.07–3.8; p = 0.004). The median PSA level at biochemical progression after the first PSMA PET-directed RT was 2.9 ng/mL (range 0.12–12.80; p = 0.24). Furthermore, the PSA level after the second PSMA PET-directed RT at the last follow-up (0.52 ng/mL, range <0.07–154.0) was not significantly different (p = 0.36) from the median PSA level (1.70 ng/mL, range 0.2–3.8) before the first PSMA PET-directed RT. The median bPFS_1 was 16.0 months after the first PSMA PET-directed RT (95% CI 11.9–19.2) and the median bPFS_2 was significantly shorter at 8.0 months (95% CI 6.3–17.7) after the second PSMA PET-directed RT (p = 0.03; 95% CI 1.9–8.3). Multivariate analysis revealed no significant parameter for bPFS_1, whereas extrapelvic disease was the only significant parameter (p = 0.02, OR 2.3; 95% CI 0.81-4.19) in multivariate analysis for bPFS_2. The median ADT-FS was 31.0 months (95% CI 20.1–41.8) and multivariate analysis showed that patients with bone metastases, compared to patients with only lymph node metastases at first PSMA PET-directed RT, had a significantly higher chance (p = 0.007, OR 4.51; 95% CI 1.8–13.47) of needing ADT at the last follow-up visit.

Conclusion

If patients are followed up closely, including PSMA PET scans, a second PSMA PET-directed RT represents a viable treatment option for well-informed and well-selected patients.
Hinweise

Availability of data and material

All data and material are available upon justified request.

Introduction

The cornerstone of treatment for non-castrated metastatic prostate cancer (mPCa) is androgen deprivation therapy (ADT), which has remained unchanged over the past years [1]. The negative impact of ADT on quality of life (QoL) [1] has resulted in a search for alternatives for well-selected patients using personalized treatment concepts on the basis of prostate-specific membrane antigen ligand positron-emission tomography (PSMA PET) [25]. There is increasing evidence that patients with a limited number of metastases have a better prognosis than patients with widespread metastatic disease, and data outside large prospective trials suggest that metastasis-directed therapies (MDTs) for mPCa patients with a so-called “oligometastatic status,” based on a generally accepted imaging-based cut-off of five metastases, improve the clinical outcome [2, 6]. The recent introduction of PSMA ligand PET has substantially improved the diagnostic accuracy of staging at low prostate-specific antigen (PSA) levels [711], allowing refined and well-monitored individualized radio-oncological treatment concepts that aim to improve PSA kinetics, prolong progression-free survival, defer the initiation of ADT, and potentially cure the patient [25, 12]. The STOMP [13] and POPSTAR trials [14], as well as the data published by Kneebone et al. [15], demonstrated that MDT alone might delay ADT for a relevant period. However, approximately half of the patients will develop oligoprogressive disease after MDT [13], making these patients amenable to repeated MDT and further improving the PSA kinetics and delaying the initiation of ADT [16]. Data on the feasibility and clinical outcome of a second MDT guided by PSMA PET imaging after previous PSMA PET-directed radiotherapy (RT) are very limited.
Thus, in the current study, we assessed the outcomes of patients with mPCa diagnosed with oligorecurrent disease after initial curative therapy who were treated with a second PSMA PET-directed radiotherapy (RT).

Patients and methods

Between June 2014 and February 2019, 32 hormone-naïve patients with oligorecurrent PCa after primary curative therapy received a first PSMA PET-directed RT as MDT for all metastases, and after oligoprogression, the patients received a definitive second PSMA PET-directed RT for all metastases. This retrospective study was approved by the local ethics committee (no. 3661-2017), and all cases were discussed and approved for RT by the multidisciplinary uro-oncologic board. Oligometastatic disease was defined as ≤5 visceral, bone, or lymph node metastases. The patients’ characteristics are summarized in Table 1.
Table 1
Patient characteristics
Characteristics
Median (range); n (%)
Age at PCa diagnosis (years)
63 (53–74)
Initial PSA (ng/ml)
9.8 (3.7–47.4)
Primary therapy
RPE alone
11 (34.4)
RPE + aRT
9 (28.1)
RPE + sRT
10 (31.2)
EBRT + temporary ADT
2 (6.3)
cT1c
2 (6.3)
pT2c
13 (40.6)
pT3a
4 (12.5)
pT3b
12 (37.5)
pT4
1 (3.1)
Initial N stage
N0
28 (87.5)
N1
4 (12.5)
Surgical margins
R0
28 (93.3)
R1
2 (6.7)
Initial risk group
High
19 (59.4)
Very high
13 (40.6)
Initial PSA (ng/ml)
9.8 (3.7–47.4)
PSA nadir after definitive therapy (ng/ml)
0.07 (<0.07–3.0)
Interval (months) from definitive therapy to PSMA PET
47 (12–168)
PSA level at PSMA-PET imaging (ng/ml)
1.70 (0.2–4.8)
Patients with ADT at PSMA-ligand PET imaging
0 (%)
Median PSA-dt at time of PSMA-PET imaging (months)
8.7 (3.1–17.2)
<6
7 (21.9)
>6, <12
10 (31.2)
>12
15 (46.9)
ADT androgen deprivation therapy, aRT adjuvant radiotherapy, dt doubling time, EBRT external beam radiation therapy, PCa prostate cancer, PSMA-PET prostate-specific membrane antigen positron-emission tomography, PSA prostate-specific antigen, RP radical prostatectomy, sRT salvage radiotherapy

PET imaging

Each patient received PET imaging with a 68gallium-labeled PSMA ligand [17] and imaging was performed according to the joint EANM and SNMMI guidelines [18]. PSMA PET scans were acquired in conjunction with low-dose computed tomography (CT) on a dedicated PET/CT system (Siemens Biograph mCT 128 Flow; Siemens, Knoxville, TN, USA) equipped with an extended field-of-view lutetium oxyorthosilicate PET component, a 128-slice spiral CT component, and a magnetically powered table optimized for continuous scanning. No intravenous contrast material was administered. All patients gave written informed consent before PSMA PET/CT. A positive visual assessment of increased focal tracer uptake higher than the surrounding background activity was used as the criterion for malignancy [9].

Radiotherapy treatment

Patients with lymph node metastases or relapse in the prostatic fossa were treated with conventionally fractionated RT (CF-RT) and patients with bone metastases were treated with slightly hypofractionated RT (HF-RT). In the case of lymph node metastases, the clinical target volume (CTV) encompassed the lymph drainage vessel to the next bifurcation without the whole ipsilateral lymphatic drainage vessel; the dose was 50.0 Gy (single dose 2.0 Gy), followed by a sequential CF-RT boost of 10.0 Gy (single dose 2.0 Gy) to the lymph node metastases. Relapse in the prostate bed was treated with CF-RT doses of 70.0–74.0 Gy (single dose of 2.0 Gy). Bone metastases were treated with HF-RT and a single dose of 2.5 Gy up to a dose of 45.0 Gy. The planning target volume (PTV) for lymph node metastases, bone metastases, and local relapse in the prostatic fossa included the CTV plus a 10-mm safety margin in all directions to account for setup errors. Image guidance was conducted at least twice a week with megavoltage cone-beam CT. Visceral metastases were treated with image-guided stereotactic body radiation therapy (SBRT) and a total dose of 37.5 Gy (single dose 12.5 Gy) to the encompassing 67% PTV isodose. The PTV included the internal target volume (ITV) plus a 4-mm safety margin in all directions to account for setup errors.

Follow-up and endpoints

All patients had periodic urologic follow-up evaluations, which included PSA measurements every 3 months. Biochemically progressive disease after RT was defined as two consecutive increases in PSA levels from the nadir PSA level or a PSA level above baseline. Biochemical nonresponse was defined as the first PSA level 3 months after RT that was ≥10% of the baseline PSA level at the time of PSMA PET/CT scan before RT. Patients who met the criteria of biochemical progression had a second PSMA PET/CT for restaging purposes, which was used for the second PSMA PET-directed RT to irradiate all new metastases. Patients with extensive disease (≥6 metastases) received systemic therapy according to the urologist’s choice. In general, outcomes were defined from the last day of RT. The outcome parameters were biochemical progression-free survival (bPFS), ADT-free survival (ADT-FS), overall survival (OS), and toxicity. The intervals of BPFS were analyzed separately as follows: the interval from the last day of first PSMA PET-directed RT to the first biochemical progression was defined as bPFS_1 and the interval from second PSMA PET-directed RT to further biochemical progression was defined as bPFS_2. RT-associated toxicity was analyzed using the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) v4.0 [19]. To assess the local failure rate we performed a coregistration of the PSMA PET/CT scans with the RT treatment plans. Focally increased tracer uptake higher than the surrounding background within the PTV was classified as infield relapse.

Statistical analysis

For statistical analysis, SPSS Statistics v25.0 (IBM, Armonk, New York, USA) was used. We used the paired Student’s t-test to compare pre-RT with post-RT parametric parameters and the Wilcoxon signed-rank test when the data were not normally distributed. The time-to-event data were calculated using the Kaplan–Meier method. Factors for RT treatment failure were analyzed with the log rank test in univariate analyses and significant factors were further assessed with multivariate analyses to identify independent variables for bRFS and ADT-FS. P-values <0.05 were considered statistically significant. Graphical presentations of the patterns of progression were created using a free software for statistical computing and graphics (R version 3.0.3).

Results

PSMA PET staging and therapy for metastases

A total of 59 PSMA ligand-positive metastases were detected and treated with RT. Of these, 66.1% (39/59) were pelvic lymph node metastases, 5.1% (3/59) were periaortic lymph node metastases, 22.0% (13/59) were bone metastases, 1.7% (1/59) were visceral metastases, and 5.1% (3/59) were local relapses in the prostatic fossa. A total of 53.1% of patients (17/32) had only lymph node metastases, 31.3% (10/32) of patients had only bone metastases, 12.5% (4/32) of patients had lymph node and bone metastases, and 2.6% (1/32) of patients had visceral metastases.

Patterns of progression and patient outcomes

Table 2 summarizes the results of PSMA PET scans and first PSMA PET-directed RT, as well as the results from PSMA PET restaging after biochemical progression and a consecutive second PSMA PET-directed RT. Analysis of the RT treatment plans and the PSMA PET/CT scans for all metastases at first (n = 59) and second (n = 60) PSMA PET-directed RT resulted in a cumulative infield relapse rate of 3.4% (4/119) and in a cumulative outfield relapse rate of 96.6% (115/119). The four infield relapses occurred in the right iliac lymph nodes (25%, 1/4), in a rib metastasis (25%, 1/4), and in pelvic bone metastases (50%, 2/4).
Table 2
Results of PSMA ligand PET staging prior to PSMA ligand-based radiotherapy and PSMA ligand PET restaging prior to second PSMA ligand-based radiotherapy
 
PSMA ligand-based RT
Second PSMA ligand-based RT
 
N (%)
N (%), p-value
No. of PSMA ligand-positive lesions
59 (100)
60 (100); 0.90
Total no. of LNs
42 (69.2)
36 (60); 0.51
…Pelvic LNs
39 (36.2)
15 (25); 0.03
…Periaortic/interaortocaval LNs
3 (27.6)
19 (31.7); 0.09
Total no. of bone metastases
13 (24.3)
23 (38.3); 0.04
…Pelvic bone
7 (13.0)
10 (16.7); 0.37
…Spinal bone
4 (5.4)
11 (18.3); 0.14
…Other
2 (5.9)
2 (3.3); 0.64
Prostatic fossa
3 (6.5)
0 (0); 0.32
Total no. of visceral metastases
1 (1.7)
1 (1.7); 0.91
Concurrent ADT at radiotherapy
0 (0.0)
3 (9.4); 0.32
Other local therapies (e.g., surgery, RFA)
0 (0.0)
0 (0.0)
 
Median (range)
Median (range); p-value
PSA before RT
1.71 (0.21–3.8)
2.90 (0.12–22.80); 0.23
PSA-dt (months)
8.7 (3.1–17.2)
5.8 (1.6–8.2); 0.05
No. irradiated metastases
2 (1–4)
2 (1–5); 0.90
ADT androgen deprivation therapy, dt doubling-time, LNs lymph node metastases, PCa prostate cancer, PSMA-PET prostate-specific membrane antigen positron-emission tomography, PSA prostate-specific antigen, RFA radiofrequency ablation, sRT salvage radiotherapy, SBRT stereotactic body radiation therapy, SIB simultaneous integrated boost
The median follow-up time was 39.5 months (18–60). One out of 32 (3.1%) patients died after 47 months of progressive mPCa. All patients showed biochemical responses after the first PSMA PET-directed RT: the median PSA level before first RT was 1.70 ng/mL (0.2–3.8), which decreased significantly to a median PSA nadir level of 0.39 ng/mL (range <0.07–3.8; p = 0.004). The median PSA level at biochemical progression after the first PSMA PET-directed RT was 2.9 ng/mL (range 0.12–12.80; p = 0.24). Furthermore, the PSA level after the second PSMA PET-directed RT at the last follow-up (0.52 ng/mL, range <0.07–154.0) was not significantly different (p = 0.36) from the median PSA level (1.70 ng/mL, range 0.2–3.8) before the first PSMA PET-directed RT. Additionally, 12.5% (4/32) of patients did not show a biochemical response after second the PSMA PET-directed RT and were classified as nonresponders. Fig. 1 gives a schematic overview of first and second progressions following the first and second PSMA PET-directed RT, respectively. Figs. 2 and 3 show the initial distribution of metastases at the time of the first PSMA PET-directed RT and the location of new metastases at the time of the second PSMA PET-directed RT, as well as at the time of further progression after the second PSMA PET-directed RT.
The median bPFS_1 was 16.0 months after first PSMA PET-directed RT (95% CI 11.9–19.2; Fig. 4a) and the median bPFS_2 was significantly shorter at 8.0 months (95% CI 6.3–17.7; Fig. 4b) after second PSMA PET-directed RT (p = 0.03; 95% CI 1.9–8.3). None of the parameters significant for bPFS_1 in the univariate analyses reached significance in multivariate analyses. Extrapelvic disease was the only significant parameter (p = 0.02, OR 2.3; 95% CI 0.81–4.19) in multivariate analysis for bPFS_2. Table 3 shows the detailed results of uni- and multivariate analyses for bPFS_1 and bPFS_2.
Table 3
Results of uni- and multivariate analyses for first and second biochemical progression-free survival (bPFS_1, bPFS_2)
 
bPFS_1
bPFS_2
Univariate analysis
Multivariable analysis
Univariate analysis
Multivariable analysis
p-value
p-value, OR (95% CI)
p-value
p-value, OR (95% CI)
Initial T stage (≤T2 vs. ≥T3)
0.91
0.96
Initial N stage (N0 vs. N1)
0.39
0.67
Initial PSA level in ng/ml (≤20 vs. >20)
0.81
0.82
Initial PSA level in ng/ml (≤10 vs. >10)
0.92
0.54
PSA nadir after RP (≤0.07 ng/mL vs. >0.07 ng/mL)
0.33
0.39
Gleason score (≤7 vs. ≥8)
0.18
0.22
No. of removed LN at RP (≤15 vs. >15)
0.63
0.34
Initial risk group (high risk vs. very high risk)
0.32
0.54
PSA-dt (months; ≤6, >6)
0.02
0.12, 1.92 (1.02–8.52)
0.19
PSA dt (months, ≤12, >12)
0.05
0.23, 1.76 (0.34–8.71)
0.46
No. of irradiated metastases (1 vs. >1)
0.73
0.19
No. of irradiated metastases (≤2 vs. ≥3)
0.23
0.05
0.56, 0.73 (0.29–1.28)
No. of irradiated metastases (≤3 vs. ≥4)
0.24
0.20
Type of metastases (LNs vs. bone)
0.45
0.04
0.19, 0.64 (0.23–1.39)
Extrapelvic disease (LNs and/or bone)
0.92
0.02
0.02, 2.3 (0.81–4.19)
dt doubling time, LN lymph node, PSA prostate-specific antigen, RP radical prostatectomy
At the last follow-up, 28.1% (9/32) of patients did not need ADT. The median ADT-FS was 31.0 months (95% CI 20.1–41.8; Fig. 5) and multivariate analysis showed that patients with bone metastases, compared to patients with only lymph node metastases at the first PSMA PET-directed RT, had a significantly higher chance (p = 0.007, OR 4.51; 95% CI 1.8–13.47) of needing ADT at the last follow-up visit. Furthermore, patients with oligoprogression outside the pelvis, compared to patients with oligoprogression limited to the pelvis at the second PSMA PET-directed RT, also had a significantly higher risk of needing ADT at the last follow-up visit (p = 0.03, OR 3.2; 95% CI 1.2–15.10). Table 4 shows the detailed results of uni- and multivariate analyses for ADT-FS.
Table 4
Results of uni- and multivariate analyses for androgen deprivation therapy-free survival (ADT-FS)
 
Univariate analysis
Multivariate analysis
p-value
p-value
OR, (95% CI)
Initial T stage (≤T2 vs. ≥T3)
0.19
Initial N stage (N0 vs. N1)
0.66
Initial PSA level in ng/ml (≤10 vs. >10)
0.47
Initial PSA level in ng/ml (≤20 vs. >20)
0.50
PSA nadir after RP (≤0.07 ng/mL vs. >0.07 ng/mL)
0.97
Gleason score (≤7 vs. ≥8)
0.10
No. of removed LNs at RP (≤15 vs. >15)
0.36
Initial risk group (high risk vs. very high risk)
0.75
PSA-dt (months; ≤6, >6) at first PSMA ligand-based RT
0.01
0.74
1.25, 0.34–4.60
PSA-dt (months; ≤12, >12) at first PSMA ligand-based RT
0.13
PSA-dt (months; ≤6, >6) at second PSMA ligand-based RT
0.27
PSA-dt (months; ≤12, >12) at second PSMA ligand-based RT
0.87
No. of irradiated metastases (1 vs. >1) at first PSMA ligand-based RT
0.81
No. of irradiated metastases (no. of irradiated metastases [≤2 vs. ≥3] at first PSMA ligand-based RT)
0.27
No. of irradiated metastases (no. of irradiated metastases [≤3 vs. ≥4] at first PSMA ligand-based RT)
0.05
0.59
1.68, 0.35–6.02
No. of irradiated metastases (1 vs. >1) at second PSMA ligand-based RT
0.52
No. of irradiated metastases (≤2 vs. ≥3) at second PSMA ligand-based RT
0.92
No. of irradiated metastases (≤3 vs. ≥4) at second PSMA ligand-based RT
0.36
Type of metastases at first PSMA ligand-based RT (lymph node vs. bone)
<0.001
0.007
4.51, 1.8–13.47
Extrapelvic disease at first PSMA ligand-based RT (lymph node vs. bone)
0.31
Type of metastases at second PSMA ligand-based RT (LN vs. bone)
0.82
Extrapelvic disease at second PSMA ligand-based RT (LN and/or bone)
0.01
0.03
3.2, 1.2–15.10
dt doubling time, LN lymph node, PSA prostate-specific antigen, PSMA prostate-specific membrane antigen, RP radical prostatectomy

Toxicity

Acute grade III toxicity was not observed; 3.1% (1/32) of patients developed grade II acute genitourinary toxicity. Grade II acute gastrointestinal toxicity occurred in 12.5% (4/32) of patients and grade I acute gastrointestinal toxicity occurred in 9.4% (3/32) of patients. Late grade III gastrointestinal toxicity occurred in 3.1% (1/32) of patients and grade II genitourinary toxicity occurred in 3.1% (1/78) of patients.

Discussion

The implementation of PSMA ligand PET imaging has substantially improved the diagnostic accuracy of detecting metastatic PCa at low PSA levels [7, 8, 20]. Although large randomized prospective studies are lacking, MDT is considered a viable treatment option for well-selected patients with oligorecurrent PCa [21]. Furthermore, there is still controversy about the optimal timing to initiate palliative ADT for asymptomatic metastatic patients because of the lack of prospective trials in the PSA era [1]. Furthermore, ADT offers no curative potential [1] and significantly impairs QoL in a relevant number of patients [22].
Smaller prospective trials with heterogeneous patient cohorts, including one with choline PET imaging [13], one with PSMA ligand PET imaging [14], and one with sodium fluoride (NA-F) PET imaging [15], showed encouraging results for MDT in oligometastatic prostate cancer. The STOMP [13] and POPSTAR trials [14], as well as the data published by Kneebone et al. [15], demonstrated that MDT alone might delay ADT for a relevant period. However, patients with MDT alone develop biochemical progression earlier than patients with MDT plus ADT [1315]. To the best of the authors’ knowledge, there are no published data comparing MDT alone versus MDT plus ADT with regard to hard endpoints such as prostate carcinoma-specific survival. In our opinion, patients must be well informed that ADT is the standard of care and any type of MDT is an individual treatment concept outside the current guidelines, although the optimal timing of initiation of ADT at low PSA levels remains unknown [1]. Nevertheless, approximately half of the patients will develop oligoprogressive disease after MDT alone [7], making these patients amenable to a second MDT and further delaying the start of ADT [13, 23, 24]. Additionally, due to the lack of data from prospective trials, the current guidelines do not reflect the diagnostic accuracy of PSMA ligand PET staging at low PSA levels to select patients who should or should not receive ADT [1].
To the best of our knowledge, a second RT for oligorecurrent PCa on the basis of PSMA ligand PET staging and restaging has never been reported. We showed that 28.1% (9/32) of patients did not need ADT at the last follow-up, resulting in a median ADT-FS of 31.0 (95% CI 20.1–41.8) months, which is approximately the same as the duration that other authors have reported [13, 23, 24]. None of the published data [13, 23, 24] included PSMA ligand PET staging for MDT that led to MDT at higher PSA levels, particularly increasing the likelihood that patients had an underestimated extent of lymph node metastases [11, 22]. In addition, the initiation of ADT based on the urologist’s choice at low PSA levels might be a confounder for ADT-FS and, therefore, hamper the comparison of these retrospective data.
We found that the type of irradiated metastases at oligorecurrence was the most important clinical parameter for ADT-FS. Patients with bone metastases had a significantly higher risk (p = 0.007, OR 4.51; 95% CI 1.8–13.47) of needing ADT at their last follow-up compared to patients with only lymph node metastases. Population-based data supported our findings because patients with bone metastases have a worse prognosis and lower cancer-specific survival (CSS) than those with lymph node metastases [25, 26]. A recently published SEER database analysis suggested that patients with stage M1a tumors receive significantly greater clinical benefits from local therapies to the prostate than patients with stage M1b tumors [27]. Although the median number of irradiated metastases per patient at the first PSMA PET-directed RT and at the second PSMA PET-directed RT was the same (n = 2; p = 0.90), the median BPFS_2 after the second PSMA PET-directed RT was significantly shorter than the median BPFS_1 after the first PSMA PE-directed RT (8.0 months vs. 16.0 months, p = 0.03; 95% CI 1.9–8.3). Moreover, the shorter BPFS after the second PSMA PET-directed RT was associated with a more widespread pattern of metastases (increase in bone metastases and extrapelvic lymph node metastases), which is a possible indicator for an evolving tumor biology. Until now, the evolutionary history of metastatic prostate cancer of monoclonal versus polyclonal cell seeding leading to a linear versus branching pattern of metastatic spread [28] at low PSA levels remains unknown. Many genomic and nongenomic biomarkers have been investigated in mPC [29, 30], but none of these markers are available outside of dedicated study protocols for clinical routine. On the other hand, exploratory analyses of patients with limited tumor burden according to the CHAARTED criteria revealed no OS benefit for escalated systemic therapy using either the combination of ADT + docetaxel [31] or ADT + enzalutamide [32] compared to ADT alone, indicating a different biology. With regard to PSA kinetics as a biomarker, we found that the median PSA-dt at the second PSMA PET-directed RT was significantly shorter than the median PSA-dt at the first PSMA PET-directed RT (5.8 versus 8.7 months, p = 0.05), indicating a biologically more aggressive disease at oligoprogression after the first PSMA PET-directed RT. Furthermore, a PSA-dt <6 months, compared to a PSA-dt >6 months, showed a trend towards significance (p = 0.12) in multivariate analyses for a worse BPFS_1. Ost el. showed that patients with non-castrate mPCA and one metastasis plus a PSA-dt >3 months had a 5-year CSS rate of 100%, whereas patients with a PSA-dt <3 months plus >1 metastasis had a 5-year CSS rate of only 8% [33]. There is controversy about the radiation dose, field size, and elective node irradiation when PSMA ligand PET is used for MDT of oligorecurrent mPCa. Data from the choline PET era confirmed that choline PET underestimated the extent of lymph node metastases [34], which is reflected by the fact that approximately two out of three patients treated with SBRT for pelvic lymph node metastases relapsed with lymph node metastases [25, 35], leading to a higher relapse rate than that after elective node irradiation (ENI), although the relapse rate concerning bone and visceral metastases seems to be comparable between SBRT and ENI [36]. The extent of lymph node metastases can be assessed more precisely by PSMA PET than by choline PET [7]. Therefore, we do not recommend ENI because the initiation of systemic therapies according to high- and low-burden disease and the corresponding prognosis do not depend on lymph node metastases, but on bone and visceral metastases [1]. In addition, other retrospective data from the PSMA PET era assessing the pattern of progression of lesion-directed RT without ENI of contralateral lymphatic drainage showed a significant increase in retroperitoneal and osseous metastases and only a very small number of contralateral pelvic recurrences [2]. These results may indicate that ENI may have been a necessary compensator for the poor detection rate of lymph node metastases of choline PET scans.
Some limitations of this study should be acknowledged. The retrospective nature has inherent limitations and might have introduced selection bias, although the presented cohort had a strict follow-up schedule and staging was performed only with PSMA ligand PET. Therefore, fewer metastases should be missed with this method than with conventional imaging and choline PET [11, 20, 35], leading to well-selected patients [37]. Moreover, the second PSMA ligand PET for restaging purposes allows assessment of the metabolic response after RT, leading to a reliable discrimination of new metastases and successfully irradiated metastases [38]. In addition, the study included a selected cohort with only high-risk and very high-risk patients. Therefore, caution should be taken when generalizing the observed results for patients with intermediate- or low-risk oligorecurrent PCa. Additionally, the sample size of 32 patients limited the statistical power, although the observed clinical results are robust and contribute significantly to the discussion of a second PSMA PET-directed MDT after curative primary therapy in a quickly changing clinical field.

Conclusion

Repeated PMSA PET-directed RT for oligorecurrent prostate cancer postponed ADT without significant toxicities. If patients are followed up closely, including PSMA PET scans, repeated PSMA PET-directed RT represents a viable treatment option for well-informed and well-selected patients.

Compliance with ethical guidelines

Conflict of interest

C. Henkenberens, A.-K. Oehus, T. Derlin, F. Bengel, T.L. Ross, M.A. Kuczyk, S. Janssen, H. Christiansen, and C.A.J. von Klot declare that they have no competing interests.

Ethical standards

This retrospective analysis was approved by the local ethics committee.
Open Access This 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/​.

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.

Strahlentherapie und Onkologie

Print-Titel

•Übersichten, Originalien, Kasuistiken

•Kommentierte Literatur aus der Radioonkologie, Strahlenbiologie und -physik

Literatur
1.
Zurück zum Zitat Cornford P, Bellmunt J, Bolla M, Briers E, De Santis M, Gross T, Henry AM, Joniau S, Lam TB, Mason MD, van der Poel HG, van der Kwast TH, Rouvière O, Wiegel T, Mottet N (2017) EAU-ESTRO-SIOG guidelines on prostate cancer. Part II: Treatment of relapsing, metastatic, and castration-resistant prostate cancer. Eur Urol 71:630–642PubMed Cornford P, Bellmunt J, Bolla M, Briers E, De Santis M, Gross T, Henry AM, Joniau S, Lam TB, Mason MD, van der Poel HG, van der Kwast TH, Rouvière O, Wiegel T, Mottet N (2017) EAU-ESTRO-SIOG guidelines on prostate cancer. Part II: Treatment of relapsing, metastatic, and castration-resistant prostate cancer. Eur Urol 71:630–642PubMed
2.
Zurück zum Zitat Soldatov A, von Klot CAJ, Walacides D, Derlin T, Bengel FM, Ross TL, Wester HJ, Derlin K, Kuczyk MA, Christiansen H, Henkenberens C (2019) Patterns of progression after 68ga-PSMA-Ligand PET/CT-guided radiation therapy for recurrent prostate cancer. Int J Radiat Oncol Biol Phys 103:95–104PubMed Soldatov A, von Klot CAJ, Walacides D, Derlin T, Bengel FM, Ross TL, Wester HJ, Derlin K, Kuczyk MA, Christiansen H, Henkenberens C (2019) Patterns of progression after 68ga-PSMA-Ligand PET/CT-guided radiation therapy for recurrent prostate cancer. Int J Radiat Oncol Biol Phys 103:95–104PubMed
3.
Zurück zum Zitat Kroeze SGC, Henkenberens C, Schmidt-Hegemann NS, Vogel MME, Kirste S, Becker J, Burger IA, Derlin T, Bartenstein P, Eiber M, Mix M, la Fougère C, Christiansen H, Belka C, Combs SE, Grosu AL, Müller AC, Guckenberger M (2020) Prostate-specific membrane antigen positron emission tomography-detected oligorecurrent prostate cancer treated with metastases-directed radiotherapy: Role of addition and duration of androgen deprivation. Eur Urol Focus. https://doi.org/10.1016/j.euf.2019.08.012CrossRef Kroeze SGC, Henkenberens C, Schmidt-Hegemann NS, Vogel MME, Kirste S, Becker J, Burger IA, Derlin T, Bartenstein P, Eiber M, Mix M, la Fougère C, Christiansen H, Belka C, Combs SE, Grosu AL, Müller AC, Guckenberger M (2020) Prostate-specific membrane antigen positron emission tomography-detected oligorecurrent prostate cancer treated with metastases-directed radiotherapy: Role of addition and duration of androgen deprivation. Eur Urol Focus. https://​doi.​org/​10.​1016/​j.​euf.​2019.​08.​012CrossRef
4.
Zurück zum Zitat Walacides D, Meier A, Knöchelmann AC, Meinecke D, Derlin T, Bengel FM, Ross TL, Wester HJ, Derlin K, Kuczyk MA, von Klot CAJ, Christiansen H, Henkenberens C (2019) Comparison of 68Ga-PSMA ligand PET/CT versus conventional cross-sectional imaging for target volume delineation for metastasis-directed radiotherapy for metachronous lymph node metastases from prostate cancer. Strahlenther Onkol 195:420–429PubMed Walacides D, Meier A, Knöchelmann AC, Meinecke D, Derlin T, Bengel FM, Ross TL, Wester HJ, Derlin K, Kuczyk MA, von Klot CAJ, Christiansen H, Henkenberens C (2019) Comparison of 68Ga-PSMA ligand PET/CT versus conventional cross-sectional imaging for target volume delineation for metastasis-directed radiotherapy for metachronous lymph node metastases from prostate cancer. Strahlenther Onkol 195:420–429PubMed
5.
Zurück zum Zitat Henkenberens C, Derlin T, Bengel FM, Ross TL, Wester HJ, Hueper K, Kuczyk MA, Christiansen H, von Klot CA (2018) Patterns of relapse as determined by 68Ga-PSMA ligand PET/CT after radical prostatectomy: Importance for tailoring and individualizing treatment. Strahlenther Onkol 194:303–310PubMed Henkenberens C, Derlin T, Bengel FM, Ross TL, Wester HJ, Hueper K, Kuczyk MA, Christiansen H, von Klot CA (2018) Patterns of relapse as determined by 68Ga-PSMA ligand PET/CT after radical prostatectomy: Importance for tailoring and individualizing treatment. Strahlenther Onkol 194:303–310PubMed
6.
Zurück zum Zitat Triggiani L, Alongi F, Buglione M, Detti B, Santoni R, Bruni A, Maranzano E, Lohr F, D’Angelillo R, Magli A, Bonetta A, Mazzola R, Pasinetti N, Francolini G, Ingrosso G, Trippa F, Fersino S, Borghetti P, Ghirardelli P, Magrini SM (2017) Efficacy of stereotactic body radiotherapy in oligorecurrent and in oligoprogressive prostate cancer: new evidence from a multicentric study. Br J Cancer 116:1520–1525PubMedPubMedCentral Triggiani L, Alongi F, Buglione M, Detti B, Santoni R, Bruni A, Maranzano E, Lohr F, D’Angelillo R, Magli A, Bonetta A, Mazzola R, Pasinetti N, Francolini G, Ingrosso G, Trippa F, Fersino S, Borghetti P, Ghirardelli P, Magrini SM (2017) Efficacy of stereotactic body radiotherapy in oligorecurrent and in oligoprogressive prostate cancer: new evidence from a multicentric study. Br J Cancer 116:1520–1525PubMedPubMedCentral
7.
Zurück zum Zitat Calais J, Czernin J, Cao M, Kishan AU, Hegde JV, Shaverdian N, Sandler K, Chu FI, King CR, Steinberg ML, Rauscher I, Schmidt-Hegemann NS, Poeppel T, Hetkamp P, Ceci F, Herrmann K, Fendler WP, Eiber M, Nickols NG (2018) 68ga-PSMA-11 PET/CT mapping of prostate cancer biochemical recurrence after radical prostatectomy in 270 patients with a PSA level of less than 1.0 ng/mL: impact on salvage radiotherapy planning. J Nucl Med 59:230–237PubMedPubMedCentral Calais J, Czernin J, Cao M, Kishan AU, Hegde JV, Shaverdian N, Sandler K, Chu FI, King CR, Steinberg ML, Rauscher I, Schmidt-Hegemann NS, Poeppel T, Hetkamp P, Ceci F, Herrmann K, Fendler WP, Eiber M, Nickols NG (2018) 68ga-PSMA-11 PET/CT mapping of prostate cancer biochemical recurrence after radical prostatectomy in 270 patients with a PSA level of less than 1.0 ng/mL: impact on salvage radiotherapy planning. J Nucl Med 59:230–237PubMedPubMedCentral
8.
Zurück zum Zitat McCarthy M, Francis R, Tang C, Watts J, Campbell A (2019) A multicenter prospective clinical trial of 68gallium PSMA HBED-CC PET-CT restaging in biochemically relapsed prostate carcinoma: Oligometastatic rate and distribution compared with standard imaging. Int J Radiat Oncol Biol Phys 104:801–808PubMed McCarthy M, Francis R, Tang C, Watts J, Campbell A (2019) A multicenter prospective clinical trial of 68gallium PSMA HBED-CC PET-CT restaging in biochemically relapsed prostate carcinoma: Oligometastatic rate and distribution compared with standard imaging. Int J Radiat Oncol Biol Phys 104:801–808PubMed
9.
Zurück zum Zitat Schmuck S, Mamach M, Wilke F, von Klot CA, Henkenberens C, Thackeray JT, Sohns JM, Geworski L, Ross TL, Wester HJ, Christiansen H, Bengel FM, Derlin T (2017) Multiple time-point 68ga-PSMA I&T PET/CT for characterization of primary prostate cancer: value of early dynamic and delayed imaging. Clin Nucl Med 42:e286–e293PubMed Schmuck S, Mamach M, Wilke F, von Klot CA, Henkenberens C, Thackeray JT, Sohns JM, Geworski L, Ross TL, Wester HJ, Christiansen H, Bengel FM, Derlin T (2017) Multiple time-point 68ga-PSMA I&T PET/CT for characterization of primary prostate cancer: value of early dynamic and delayed imaging. Clin Nucl Med 42:e286–e293PubMed
10.
Zurück zum Zitat Schmuck S, Nordlohne S, von Klot CA, Henkenberens C, Sohns JM, Christiansen H, Wester HJ, Ross TL, Bengel FM, Derlin T (2017) Comparison of standard and delayed imaging to improve the detection rate of [68Ga]PSMA I&T PET/CT in patients with biochemical recurrence or prostate-specific antigen persistence after primary therapy for prostate cancer. Eur J Nucl Med Mol Imaging 44:960–968PubMed Schmuck S, Nordlohne S, von Klot CA, Henkenberens C, Sohns JM, Christiansen H, Wester HJ, Ross TL, Bengel FM, Derlin T (2017) Comparison of standard and delayed imaging to improve the detection rate of [68Ga]PSMA I&T PET/CT in patients with biochemical recurrence or prostate-specific antigen persistence after primary therapy for prostate cancer. Eur J Nucl Med Mol Imaging 44:960–968PubMed
11.
Zurück zum Zitat Schmuck S, von Klot CA, Henkenberens C, Sohns JM, Christiansen H, Wester HJ, Ross TL, Bengel FM, Derlin T (2017) Initial experience with volumetric 68ga-PSMA I&T PET/CT for assessment of whole-body tumor burden as a quantitative imaging biomarker in patients with prostate cancer. J Nucl Med 58:1962–1968PubMed Schmuck S, von Klot CA, Henkenberens C, Sohns JM, Christiansen H, Wester HJ, Ross TL, Bengel FM, Derlin T (2017) Initial experience with volumetric 68ga-PSMA I&T PET/CT for assessment of whole-body tumor burden as a quantitative imaging biomarker in patients with prostate cancer. J Nucl Med 58:1962–1968PubMed
12.
Zurück zum Zitat Tosoian JJ, Gorin MA, Ross AE, Pienta KJ, Tran PT, Schaeffer EM (2017) Oligometastatic prostate cancer: definitions, clinical outcomes, and treatment considerations. Nat Rev Urol 14:15–25PubMed Tosoian JJ, Gorin MA, Ross AE, Pienta KJ, Tran PT, Schaeffer EM (2017) Oligometastatic prostate cancer: definitions, clinical outcomes, and treatment considerations. Nat Rev Urol 14:15–25PubMed
13.
Zurück zum Zitat Ost P, Reynders D, Decaestecker K, Fonteyne V, Lumen N, De Bruycker A, Lambert B, Delrue L, Bultijnck R, Claeys T, Goetghebeur E, Villeirs G, De Man K, Ameye F, Billiet I, Joniau S, Vanhaverbeke F, De Meerleer G (2018) Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence: A prospective, randomized, multicenter phase II trial. J Clin Oncol 36:446–453PubMed Ost P, Reynders D, Decaestecker K, Fonteyne V, Lumen N, De Bruycker A, Lambert B, Delrue L, Bultijnck R, Claeys T, Goetghebeur E, Villeirs G, De Man K, Ameye F, Billiet I, Joniau S, Vanhaverbeke F, De Meerleer G (2018) Surveillance or metastasis-directed therapy for oligometastatic prostate cancer recurrence: A prospective, randomized, multicenter phase II trial. J Clin Oncol 36:446–453PubMed
14.
Zurück zum Zitat Siva S, Bressel M, Murphy DG, Shaw M, Chander S, Violet J, Tai KH, Udovicich C, Lim A, Selbie L, Hofman MS, Kron T, Moon D, Goad J, Lawrentschuk N, Foroudi F (2018) Stereotactic Abative Body Radiotherapy (SABR) for oligometastatic prostate cancer: a prospective clinical trial. Eur Urol 74:455–462PubMed Siva S, Bressel M, Murphy DG, Shaw M, Chander S, Violet J, Tai KH, Udovicich C, Lim A, Selbie L, Hofman MS, Kron T, Moon D, Goad J, Lawrentschuk N, Foroudi F (2018) Stereotactic Abative Body Radiotherapy (SABR) for oligometastatic prostate cancer: a prospective clinical trial. Eur Urol 74:455–462PubMed
15.
Zurück zum Zitat Kneebone A, Hruby G, Ainsworth H, Byrne K, Brown C, Guo L, Guminski A, Eade T (2018) Stereotactic body radiotherapy for oligometastatic prostate cancer detected via prostate-specific membrane antigen positron emission tomography. Eur Urol Oncol 1:531–537PubMed Kneebone A, Hruby G, Ainsworth H, Byrne K, Brown C, Guo L, Guminski A, Eade T (2018) Stereotactic body radiotherapy for oligometastatic prostate cancer detected via prostate-specific membrane antigen positron emission tomography. Eur Urol Oncol 1:531–537PubMed
17.
Zurück zum Zitat Eder M, Schäfer M, Bauder-Wüst U, Hull WE, Wängler C, Mier W, Haberkorn U, Eisenhut M (2012) 68Ga-complex lipophilicity and the targeting property of a urea-based PSMA inhibitor for PET imaging. Bioconjug Chem 23:688–697PubMed Eder M, Schäfer M, Bauder-Wüst U, Hull WE, Wängler C, Mier W, Haberkorn U, Eisenhut M (2012) 68Ga-complex lipophilicity and the targeting property of a urea-based PSMA inhibitor for PET imaging. Bioconjug Chem 23:688–697PubMed
18.
Zurück zum Zitat Fendler WP, Eiber M, Beheshti M, Bomanji J, Ceci F, Cho S, Giesel F, Haberkorn U, Hope TA, Kopka K, Krause BJ, Mottaghy FM, Schöder H, Sunderland J, Wan S, Wester HJ, Fanti S, Herrmann K (2017) 68Ga-PSMA PET/CT: Joint EANM and SNMMI procedure guideline for prostate cancer imaging: version 1.0. Eur J Nucl Med Mol Imaging 44:1014–1024PubMed Fendler WP, Eiber M, Beheshti M, Bomanji J, Ceci F, Cho S, Giesel F, Haberkorn U, Hope TA, Kopka K, Krause BJ, Mottaghy FM, Schöder H, Sunderland J, Wan S, Wester HJ, Fanti S, Herrmann K (2017) 68Ga-PSMA PET/CT: Joint EANM and SNMMI procedure guideline for prostate cancer imaging: version 1.0. Eur J Nucl Med Mol Imaging 44:1014–1024PubMed
20.
Zurück zum Zitat Afshar-Oromieh A, Holland-Letz T, Giesel FL, Kratochwil C, Mier W, Haufe S, Debus N, Eder M, Eisenhut M, Schäfer M, Neels O, Hohenfellner M, Kopka K, Kauczor HU, Debus J, Haberkorn U (2017) Diagnostic performance of 68Ga-PSMA-11 (HBED-CC) PET/CT in patients with recurrent prostate cancer: evaluation in 1007 patients. Eur J Nucl Med Mol Imaging 44:1258–1268PubMedPubMedCentral Afshar-Oromieh A, Holland-Letz T, Giesel FL, Kratochwil C, Mier W, Haufe S, Debus N, Eder M, Eisenhut M, Schäfer M, Neels O, Hohenfellner M, Kopka K, Kauczor HU, Debus J, Haberkorn U (2017) Diagnostic performance of 68Ga-PSMA-11 (HBED-CC) PET/CT in patients with recurrent prostate cancer: evaluation in 1007 patients. Eur J Nucl Med Mol Imaging 44:1258–1268PubMedPubMedCentral
21.
Zurück zum Zitat Gillessen S, Attard G, Beer TM, Beltran H, Bossi A, Bristow R, Carver B, Castellano D, Chung BH, Clarke N, Daugaard G, Davis ID, de Bono J, Borges Dos Reis R, Drake CG, Eeles R, Efstathiou E, Evans CP, Fanti S, Feng F, Fizazi K, Frydenberg M, Gleave M, Halabi S, Heidenreich A, Higano CS, James N, Kantoff P, Kellokumpu-Lehtinen PL, Khauli RB, Kramer G, Logothetis C, Maluf F, Morgans AK, Morris MJ, Mottet N, Murthy V, Oh W, Ost P, Padhani AR, Parker C, Pritchard CC, Roach M, Rubin MA, Ryan C, Saad F, Sartor O, Scher H, Sella A, Shore N, Smith M, Soule H, Sternberg CN, Suzuki H, Sweeney C, Sydes MR, Tannock I, Tombal B, Valdagni R, Wiegel T, Omlin A (2018) Management of patients with advanced prostate cancer: the report of the advanced prostate cancer consensus conference APCCC 2017. Eur Urol 73:178–211PubMed Gillessen S, Attard G, Beer TM, Beltran H, Bossi A, Bristow R, Carver B, Castellano D, Chung BH, Clarke N, Daugaard G, Davis ID, de Bono J, Borges Dos Reis R, Drake CG, Eeles R, Efstathiou E, Evans CP, Fanti S, Feng F, Fizazi K, Frydenberg M, Gleave M, Halabi S, Heidenreich A, Higano CS, James N, Kantoff P, Kellokumpu-Lehtinen PL, Khauli RB, Kramer G, Logothetis C, Maluf F, Morgans AK, Morris MJ, Mottet N, Murthy V, Oh W, Ost P, Padhani AR, Parker C, Pritchard CC, Roach M, Rubin MA, Ryan C, Saad F, Sartor O, Scher H, Sella A, Shore N, Smith M, Soule H, Sternberg CN, Suzuki H, Sweeney C, Sydes MR, Tannock I, Tombal B, Valdagni R, Wiegel T, Omlin A (2018) Management of patients with advanced prostate cancer: the report of the advanced prostate cancer consensus conference APCCC 2017. Eur Urol 73:178–211PubMed
22.
Zurück zum Zitat Morgans AK, Stockler MR (2019) Patient-reported outcomes in metastatic castration-sensitive prostate cancer in the adjuvant setting. Eur Urol Focus 5:144–146PubMedPubMedCentral Morgans AK, Stockler MR (2019) Patient-reported outcomes in metastatic castration-sensitive prostate cancer in the adjuvant setting. Eur Urol Focus 5:144–146PubMedPubMedCentral
23.
Zurück zum Zitat Gomez-Iturriaga A, Casquero Ocio F, Ost P, Fernandez I, Rodeño E, Llarena R, Garcia-Olaverri J, Ortiz de Zarate R, Cacicedo J, Ahtamon A, Bilbao P (2019) Outcomes after a first and/or second salvage treatment in patients with oligometastatic prostate cancer recurrence detected by (18-F) choline PET-CT. Eur J Cancer Care (Engl) 28:e1309325 Gomez-Iturriaga A, Casquero Ocio F, Ost P, Fernandez I, Rodeño E, Llarena R, Garcia-Olaverri J, Ortiz de Zarate R, Cacicedo J, Ahtamon A, Bilbao P (2019) Outcomes after a first and/or second salvage treatment in patients with oligometastatic prostate cancer recurrence detected by (18-F) choline PET-CT. Eur J Cancer Care (Engl) 28:e1309325
24.
Zurück zum Zitat Berkovic P, De Meerleer G, Delrue L, Lambert B, Fonteyne V, Lumen N, Decaestecker K, Villeirs G, Vuye P, Ost P (2013) Salvage stereotactic body radiotherapy for patients with limited prostate cancer metastases: deferring androgen deprivation therapy. Clin Genitourin Cancer 11:27–32PubMed Berkovic P, De Meerleer G, Delrue L, Lambert B, Fonteyne V, Lumen N, Decaestecker K, Villeirs G, Vuye P, Ost P (2013) Salvage stereotactic body radiotherapy for patients with limited prostate cancer metastases: deferring androgen deprivation therapy. Clin Genitourin Cancer 11:27–32PubMed
25.
Zurück zum Zitat Nørgaard M, Jensen AØ, Jacobsen JB, Cetin K, Fryzek JP, Sørensen HT (2010) Skeletal related events, bone metastasis and survival of prostate cancer: a population based cohort study in Denmark (1999 to 2007). J Urol 184:162–167PubMed Nørgaard M, Jensen AØ, Jacobsen JB, Cetin K, Fryzek JP, Sørensen HT (2010) Skeletal related events, bone metastasis and survival of prostate cancer: a population based cohort study in Denmark (1999 to 2007). J Urol 184:162–167PubMed
26.
Zurück zum Zitat Gandaglia G, Karakiewicz PI, Briganti A, Passoni NM, Schiffmann J, Trudeau V, Graefen M, Montorsi F, Sun M (2015) Impact of the site of metastases on survival in patients with metastatic prostate cancer. Eur Urol 68:325–334PubMed Gandaglia G, Karakiewicz PI, Briganti A, Passoni NM, Schiffmann J, Trudeau V, Graefen M, Montorsi F, Sun M (2015) Impact of the site of metastases on survival in patients with metastatic prostate cancer. Eur Urol 68:325–334PubMed
27.
Zurück zum Zitat Pompe RS, Tilki D, Preisser F, Leyh-Bannurah SR, Bandini M, Marchioni M, Gild P, Tian Z, Fossati N, Cindolo L, Shariat SF, Huland H, Graefen M, Briganti A, Karakiewicz PI (2018) Survival benefit of local versus no local treatment for metastatic prostate cancer-Impact of baseline PSA and metastatic substages. Prostate 78:753–757PubMed Pompe RS, Tilki D, Preisser F, Leyh-Bannurah SR, Bandini M, Marchioni M, Gild P, Tian Z, Fossati N, Cindolo L, Shariat SF, Huland H, Graefen M, Briganti A, Karakiewicz PI (2018) Survival benefit of local versus no local treatment for metastatic prostate cancer-Impact of baseline PSA and metastatic substages. Prostate 78:753–757PubMed
28.
Zurück zum Zitat Gundem G, Van Loo P, Kremeyer B, Alexandrov LB, Tubio JMC, Papaemmanuil E, Brewer DS, Kallio HML, Högnäs G, Annala M, Kivinummi K, Goody V, Latimer C, O’Meara S, Dawson KJ, Isaacs W, Emmert-Buck MR, Nykter M, Foster C, Kote-Jarai Z, Easton D, Whitaker HC, ICGC Prostate Group (2015) The evolutionary history of lethal metastatic prostate cancer. Nature 520:353–357PubMedPubMedCentral Gundem G, Van Loo P, Kremeyer B, Alexandrov LB, Tubio JMC, Papaemmanuil E, Brewer DS, Kallio HML, Högnäs G, Annala M, Kivinummi K, Goody V, Latimer C, O’Meara S, Dawson KJ, Isaacs W, Emmert-Buck MR, Nykter M, Foster C, Kote-Jarai Z, Easton D, Whitaker HC, ICGC Prostate Group (2015) The evolutionary history of lethal metastatic prostate cancer. Nature 520:353–357PubMedPubMedCentral
29.
Zurück zum Zitat Cucchiara V, Cooperberg MR, Dall’Era M, Lin DW, Montorsi F, Schalken JA, Evans CP (2018) Genomic markers in prostate cancer decision making. Eur Urol 73:572–582PubMed Cucchiara V, Cooperberg MR, Dall’Era M, Lin DW, Montorsi F, Schalken JA, Evans CP (2018) Genomic markers in prostate cancer decision making. Eur Urol 73:572–582PubMed
31.
Zurück zum Zitat Kyriakopoulos CE, Chen YH, Carducci MA, Liu G, Jarrard DF, Hahn NM, Shevrin DH, Dreicer R, Hussain M, Eisenberger M, Kohli M, Plimack ER, Vogelzang NJ, Picus J, Cooney MM, Garcia JA, DiPaola RS, Sweeney CJ (2018) Chemohormonal therapy in metastatic hormone-sensitive prostate cancer: long-term survival analysis of the randomized phase III E3805 CHAARTED trial. J Clin Oncol 36:1080–1087PubMedPubMedCentral Kyriakopoulos CE, Chen YH, Carducci MA, Liu G, Jarrard DF, Hahn NM, Shevrin DH, Dreicer R, Hussain M, Eisenberger M, Kohli M, Plimack ER, Vogelzang NJ, Picus J, Cooney MM, Garcia JA, DiPaola RS, Sweeney CJ (2018) Chemohormonal therapy in metastatic hormone-sensitive prostate cancer: long-term survival analysis of the randomized phase III E3805 CHAARTED trial. J Clin Oncol 36:1080–1087PubMedPubMedCentral
32.
Zurück zum Zitat Davis ID, Martin AJ, Stockler MR, Begbie S, Chi KN, Chowdhury S, Coskinas X, Frydenberg M, Hague WE, Horvath LG, Joshua AM, Lawrence NJ, Marx G, McCaffrey J, McDermott R, McJannett M, North SA, Parnis F, Parulekar W, Pook DW, Reaume MN, Sandhu SK, Tan A, Tan TH, Thomson A, Tu E, Vera-Badillo F, Williams SG, Yip S, Zhang AY, Zielinski RR, Sweeney CJ (2019) Enzalutamide with standard first-line therapy in metastatic prostate cancer. N Engl J Med 381:121–131PubMed Davis ID, Martin AJ, Stockler MR, Begbie S, Chi KN, Chowdhury S, Coskinas X, Frydenberg M, Hague WE, Horvath LG, Joshua AM, Lawrence NJ, Marx G, McCaffrey J, McDermott R, McJannett M, North SA, Parnis F, Parulekar W, Pook DW, Reaume MN, Sandhu SK, Tan A, Tan TH, Thomson A, Tu E, Vera-Badillo F, Williams SG, Yip S, Zhang AY, Zielinski RR, Sweeney CJ (2019) Enzalutamide with standard first-line therapy in metastatic prostate cancer. N Engl J Med 381:121–131PubMed
33.
Zurück zum Zitat Ost P, Decaestecker K, Lambert B, Fonteyne V, Delrue L, Lumen N, Ameye F, De Meerleer G (2014) Prognostic factors influencing prostate cancer-specific survival in non-castrate patients with metastatic prostate cancer. Prostate 74:297–305PubMed Ost P, Decaestecker K, Lambert B, Fonteyne V, Delrue L, Lumen N, Ameye F, De Meerleer G (2014) Prognostic factors influencing prostate cancer-specific survival in non-castrate patients with metastatic prostate cancer. Prostate 74:297–305PubMed
35.
Zurück zum Zitat van Leeuwen PJ, Stricker P, Hruby G, Kneebone A, Ting F, Thompson B, Nguyen Q, Ho B, Emmett L (2016) (68) Ga-PSMA has a high detection rate of prostate cancer recurrence outside the prostatic fossa in patients being considered for salvage radiation treatment. BJU Int 117:732–739PubMed van Leeuwen PJ, Stricker P, Hruby G, Kneebone A, Ting F, Thompson B, Nguyen Q, Ho B, Emmett L (2016) (68) Ga-PSMA has a high detection rate of prostate cancer recurrence outside the prostatic fossa in patients being considered for salvage radiation treatment. BJU Int 117:732–739PubMed
36.
Zurück zum Zitat De Bleser E, Jereczek-Fossa BA, Pasquier D, Zilli T, Van As N, Siva S, Fodor A, Dirix P, Gomez-Iturriaga A, Trippa F, Detti B, Ingrosso G, Triggiani L, Bruni A, Alongi F, Reynders D, De Meerleer G, Surgo A, Loukili K, Miralbell R, Silva P, Chander S, Di Muzio NG, Maranzano E, Francolini G, Lancia A, Tree A, Deantoni CL, Ponti E, Marvaso G, Goetghebeur E, Ost P (2019) Metastasis-directed therapy in treating nodal oligorecurrent prostate cancer: a multi-institutional analysis comparing the outcome and toxicity of stereotactic body radiotherapy and elective nodal radiotherapy. Eur Urol 76:732–739PubMed De Bleser E, Jereczek-Fossa BA, Pasquier D, Zilli T, Van As N, Siva S, Fodor A, Dirix P, Gomez-Iturriaga A, Trippa F, Detti B, Ingrosso G, Triggiani L, Bruni A, Alongi F, Reynders D, De Meerleer G, Surgo A, Loukili K, Miralbell R, Silva P, Chander S, Di Muzio NG, Maranzano E, Francolini G, Lancia A, Tree A, Deantoni CL, Ponti E, Marvaso G, Goetghebeur E, Ost P (2019) Metastasis-directed therapy in treating nodal oligorecurrent prostate cancer: a multi-institutional analysis comparing the outcome and toxicity of stereotactic body radiotherapy and elective nodal radiotherapy. Eur Urol 76:732–739PubMed
37.
Zurück zum Zitat Frenzel T, Tienken M, Abel M, Berliner C, Klutmann S, Beyersdorff D, Schwarz R, Krüll A, Bannas P (2018) The impact of [68Ga]PSMA I&T PET/CT on radiotherapy planning in patients with prostate cancer. Strahlenther Onkol 194:646–654PubMed Frenzel T, Tienken M, Abel M, Berliner C, Klutmann S, Beyersdorff D, Schwarz R, Krüll A, Bannas P (2018) The impact of [68Ga]PSMA I&T PET/CT on radiotherapy planning in patients with prostate cancer. Strahlenther Onkol 194:646–654PubMed
38.
Zurück zum Zitat Baumann R, Koncz M, Luetzen U, Krause F, Dunst J (2018) Oligometastases in prostate cancer : Metabolic response in follow-up PSMA-PET-CTs after hypofractionated IGRT. Strahlenther Onkol 194:318–324PubMed Baumann R, Koncz M, Luetzen U, Krause F, Dunst J (2018) Oligometastases in prostate cancer : Metabolic response in follow-up PSMA-PET-CTs after hypofractionated IGRT. Strahlenther Onkol 194:318–324PubMed
Metadaten
Titel
Efficacy of repeated PSMA PET-directed radiotherapy for oligorecurrent prostate cancer after initial curative therapy
verfasst von
PD Dr. med. Christoph Henkenberens
Ann-Kathrin Oehus
Thorsten Derlin
Frank Bengel
Tobias L. Ross
Markus A. Kuczyk
Stefan Janssen
Hans Christiansen
Christoph A. J. von Klot
Publikationsdatum
12.05.2020
Verlag
Springer Berlin Heidelberg
Erschienen in
Strahlentherapie und Onkologie / Ausgabe 11/2020
Print ISSN: 0179-7158
Elektronische ISSN: 1439-099X
DOI
https://doi.org/10.1007/s00066-020-01629-5

Weitere Artikel der Ausgabe 11/2020

Strahlentherapie und Onkologie 11/2020 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.