Skip to main content
Erschienen in: memo - Magazine of European Medical Oncology 1/2024

Open Access 29.12.2023 | short review

Metastasis-directed therapy: new standard or too early to change paradigm?

verfasst von: Jakob Klemm, Pawel Rajwa, Marcin Miszczyk, Stephan Brönimann, Ekaterina Laukhtina, Ichiro Tsuboi, Akihiro Matsukawa, Mehdi Kardoust Parizi, Pierre I. Karakiewicz, Shahrokh F. Shariat

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 1/2024

Summary

Metastasis-directed therapy (MDT) is an emerging treatment strategy for patients with oligometastatic prostate cancer (PCa), particularly for oligorecurrent disease. This review aims to summarize findings from several prospective trials in the setting of oligorecurrent PCa. We found that MDT is feasible, has high tolerability, and is effective in terms of local control of treated lesions and of deferring disease progression in well-selected patients. Selecting patients for MDT requires thoughtful consideration of factors such as the castration status, the number of detected metastases, and the imaging modality used for metastasis detection. Notably, the studies included in this review varied in terms of these factors, complicating the comparability of their results. Despite the existence of several prospective clinical trials in the field, there is an absence of high-level evidence attributable to the lack of phase 3 clinical trials. As a result, current guidelines recommend the administration of MDT exclusively within the context of clinical trials. Despite this, retrospective series indicate that MDT is already frequently utilized outside of clinical trials.
Hinweise
The authors Jakob Klemm and Pawel Rajwa contributed equally to the manuscript.

Publisher’s Note

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

Introduction

Metastasis-directed therapy (MDT) is a promising treatment option for oligometastatic prostate cancer (PC), a condition that occupies the clinical spectrum between non-metastatic and widespread systemic disease [1]. Characterized by a limited volume of metastatic cells and locations (predominantly 1–5 metastatic lesions), this specific stage of cancer can occur in both hormone-sensitive (mHSPC) and castration-resistant (mCRPC) forms of metastatic prostate cancer (mPCa), presenting either synchronously or metachronously. Recent advancements in imaging modalities have brought oligometastatic PC into the spotlight, suggesting potential advantages of MDT [2]. Initial evidence from phase 2 trials indicates that MDT, used alone, could improve progression-free survival (PFS) in patients with oligorecurrent PC (OPC; [2, 3]). However, guidelines recommend using MDT only as an investigational approach within clinical trials [4].
In the setting of newly diagnosed metastases (mHSPC), since data on the efficacy of MDT are sparse, current guidelines continue to endorse the combination of androgen deprivation therapy (ADT) with androgen receptor signaling inhibitors (ARSI; [4]), which have been proven to improve overall survival (OS) and PFS [58].
Against this backdrop, it is hypothesized that MDT could postpone disease progression and delay systemic treatment in patients with OPC after local therapy with curative intent such as RP or RT. Given the rapid evolution of evidence and several ongoing clinical trials investigating MDT, we aimed to summarize evidence derived from prospective trials deploying MDT in the setting of OPC following RT or RP.

Evidence acquisition

We searched the PubMed database up to 1 October 2023 using pre-defined search criteria as follows: (prospective) AND ((metastatic) OR (oligometastatic)) AND (prostate cancer) AND (prostatectomy) AND ((radiotherapy) OR (radiation therapy) OR (metastasis directed) OR (MDT) OR (radiosurgery) OR (metastasectomy)).
We identified eight publications reporting on prospective clinical trials investigating MDT only in the setting of oligorecurrent prostate cancer since January 2018 (Table 1). Furthermore, we conducted a search on https://​clinicaltrials.​gov for active phase 3 clinical trials that are utilizing MDT for OPC (Table 2).
Table 1
Clinical trials employing MDT alone for oligorecurrent prostate cancer
Study
Type
Setting
Intervention
Inclusion
Imaging
N
(study arm/control arm)
Median FU
Evaluated clinical outcomes
STOMP (2018) [3]
RCT
100% mHSPC
SBRT or metastasectomy vs. observation
1–3 extracranial metastases, asymptomatic biochemical recurrence following curative treatment (no ADT, no local relapse, serum testosterone levels >50 ng/mL); ECOG ≤ 1
Choline PET
31/31
36
ADT-free survival, PSA progression, local progression
ORIOLE (2020) [2]
RCT
100% mHSPC
SBRT vs. observation
1–3 asymptomatic metastases after primary curative treatment, and no ADT within 6 months of enrolment
Conventional imaging
36/18
19
Progression-free survival, biochemical progression-free survival, distant metastases-free survival
Pan (2022) [14]
nCT
Non-metastatic PC patients based on conventional imaging who experienced early PSA (prostate-specific antigen) progression on ADT
SBRT vs. ADT in control group
1–5 bone or lymph node metastases and early PSA progression on ADT following curative treatment; ECOG ≤ 1, life expectancy > 12 months
PSMA-PET
29/18
21
Metastasis-free survival
POPSTAR (2018) [9]
SA
67% mHSPC, 33% mCRPC
SRS
1–3 bone or lymph node metastases following curative treatment; ECOG ≤ 2
NaF-PET
33
24
Local progression-free survival, distant progression-free survival, ADT-free survival, PSA response
Kneebone (2018) [10]
SA
100% mHSPC
SRS or SBRT
1–3 lymph node or bone metastases following biochemical recurrence after primary curative treatment
PSMA-PET
57
16
Progression-free survival, biochemical progression-free survival, distant metastases-free survival
Glicksman (2022) [11]
SA
100% mHSPC
SBRT or metastasectomy
1–6 metastases following biochemical recurrence after RP and postoperative RT +/− ADT
PSMA-PET
74
24
Biochemical response, PSA progression-free survival, ADT-free survival, salvage treatment-free survival, CRPC-free survival
OLI‑P (2022) [12, 13]
SA
100% mHSPC
SBRT or conventional RT
1–5 bone or lymph node metastases following local curative therapy and PSA ≤ 10 ng/mL; Life expectancy ≥ 5 years
PSMA-PET
63
37
PSA progression-free time, time to start systemic therapy, progression-free survival, overall survival. local progression-free time, time to the first tumor-related clinical event
RCT randomized clinical trial, nCT multi-armed clinical trial, SA single-arm clinical trial, SBRT stereotactic body radiotherapy, SRS stereotactic radiosurgery, RT radiotherapy, ADT androgen deprivation therapy, ECOG Eastern Cooperative Oncology Group Performance Status Scale, PSA prostate-specific antigen, RP radical prostatectomy, PET positron emission tomography, PSMA prostate-specific membrane antigen, NaF sodium fluoride, FU follow-up, MDT metastasis-directed therapy (MDT), SOC standard of care, mHSPC metastatic hormone-sensitive prostate cancer, mCRPC metastatic castration-resistant prostate cancer
Table 2
Ongoing clinical phase 3 trials employing MDT
Study
Setting
Intervention
Planned cohort size
Primary endpoint
NCT05352178 (SPARKLE)
Oligorecurrent disease
MDT alone vs. MDT + 1 month of ADT vs. MDT + 6 months of ADT
873
Poly-metastatic-free survival (PMFS)
NCT04302454 (ADOPT)
Oligometastatic disease
MDT alone vs. MDT + ADT
280
Metastases progression-free survival (MPFS)
NCT04787744 (VA STARPORT)
Oligorecurrent disease
SOC systemic therapy + MDT vs. SOC systemic therapy
464
Radiographic progression-free survival (rPFS)
NCT04423211
Oligorecurrent disease
MDT + SOC treatment vs. SOC treatment alone
804
Progression-free survival (PFS)
NCT04983095 (METRO)
Oligometastatic disease
MDT + SOC treatment vs. SOC treatment alone
114
Failure-free survival
NCT04115007 (PRESTO)
Oligometastatic disease
MDT + SOC treatment vs. SOC treatment alone
350
Castration-resistant prostate cancer-free survival
ADT antiandrogen therapy, MDT metastasis-directed therapy, SOC standard of care

Single-arm prospective trials

The POPSTAR trial [9], published in 2018, applied stereotactic radiosurgery (SRS) to 33 OPC patients, consisting of 67% mHSPC and 33% mCRPC, each presenting 1–3 bone or lymph node metastases identified through conventional imaging and sodium fluoride positron emission tomography (PET) scans. Lymph node metastases were exclusive in 36.4% of patients. Aside from confirming feasibility (97% completed full treatment) and tolerability (a single grade 3 Common Terminology Criteria of Adverse Events [CTCAE] adverse event noted), a 24-month median follow-up exposed 1‑ and 2‑year local and disease PFS rates of 97% and 58%, and 93% and 39%, respectively. A 2-year ADT-free survival rate of 48% was observed in mHSPC patients.
Similarly, Kneebone et al. [10] investigated SRS or stereotactic body radiotherapy (SBRT) to treat all lesions observed in 57 OPC patients, who harbored 1–3 lymph node or bone metastases detected via a prostate-specific membrane antigen (PSMA) PET scanning. All participants had mHSPC, with 65% displaying only nodal metastases. The primary endpoint was a biochemical failure, determined by a post-SBRT or SRS PSA level of nadir +0.2 ng/mL. Within a 16-month median follow-up, the median biochemical disease-free survival (bDFS) was 11 months, with a 31.9% bDFS rate observed at 15 months. Notably, the study reported no in-field failures, and no toxicities of grade ≥ 3 (according to CTCAE) were observed.
In 2022, Glicksman et al. [11] studied 74 hormone-sensitive patients with biochemical recurrence after RP and postoperative RT with or without ADT; all had 1–6 PSMA PET-detected metastases but no evidence of metastases on conventional imaging (i.e., computed tomography [CT] and/or bone scan). Lymph node metastases only were observed in 86.5% of the patients. Most patients received SBRT (87%), while a small fraction (13%) underwent metastasectomy. The primary endpoint was a ≥ 50% PSA decline following MDT. Over a median follow-up of 24 months, half of the patients exhibited a biochemical response (51%), and the median biochemical PFS and ADT-free survival were 21 and 45 months, respectively. One patient experienced grade 3 toxicity (intraoperative ureteric injury).
Finally, the 2022 OLI-P-trial [12, 13] reported results for local ablative RT (aRT) in patients with 1–5 PSMA PET-detected metastases after curative treatment with a life expectancy of ≥ 5 years. All participants were hormone sensitive and 68.3% of the patients had only nodal disease. With 63 participants who met the inclusion criteria, no treatment-related toxicities were observed 2 years after aRT, meeting its primary endpoint of grade ≥ 2 toxicity in less than 15% (p < 0.001). Only one instance of grade 3 toxicity (bacterial cystitis) was reported within the 37-month median follow-up. The median PSA progression-free survival was 13 months, with 13% of the 47 patients experiencing PSA progression resulting in ADT initiation before reaching the PSA recurrence definition. A subsequent publication [13] reported a 3-year local PFS rate of 93.5%, with distant progression observed in 52% of the patients over a 41-month follow-up within the same study cohort.

Prospective randomized clinical trials and multi-arm clinical trials

Published in 2018, STOMP [3] was the first RCT to assess MDT alone in OPC. The study randomized 62 patients with 1–3 extracranial metastases (diagnosed after RP or RT via choline PET) into two groups: MDT via SBRT/metastasectomy or observation. All patients were hormone-sensitive, and 54.8% had only nodal metastases. The primary endpoint was ADT-free survival. Over a 36-month median follow-up, the MDT group had a median ADT-free survival of 21 months versus 13 months in the observation group (hazard ratio [HR]: 0.6, 80% confidence interval (CI): 0.40–0.90, log-rank p = 0.1). The MDT group had no symptomatic or local progression compared to three and six cases, respectively, in the control group.
The 2020 ORIOLE trial [2] randomly assigned OPC patients with 1–3 asymptomatic metastases and no ADT within the last 6 months to either SBRT or observation (2:1 ratio). All 54 patients were hormone sensitive and diagnosed via conventional imaging, with 58% having only nodal disease. The primary endpoint, progression at 6 months (progression of PSA, progression on conventional imaging, PCa-related symptoms, ADT initiation, or death), occurred in 19% (SBRT) versus 61% (observation) of patients (p = 0.005). Within a 19-month median follow-up, median PFS was not reached in the SBRT group versus 6 months in observation (HR: 0.3, 95% CI: 0.11–0.81, p = 0.002). With SBRT a 98.6% local control rate was achieved at 6 months. Although PSMA PET was conducted in treatment planning, the team was blinded to findings, leading to 16% of 36 SBRT-treated patients harboring supposedly untreated lesions. More patients with untreated lesions experienced progression (38% vs. 5%, p = 0.03), and their median PFS was 11.8 months versus not reached in patients without untreated lesions (HR: 0.26, 95% CI: 0.09–0.76, p = 0.006). No grade 3 or higher adverse events were reported in either group including the SBRT group.
In 2022, Pan et al. [14] evaluated metastasis-free survival (MFS) efficacy and toxicity in non-metastatic PC patients based on conventional imaging who experienced early PSA progression on ADT after RP or RT. All patients underwent PSMA and fluorodeoxyglucose (FDG) PET imaging. The three-armed trial recommended SBRT for patients with ≤ 5 nonvisceral metastases (SBRT group), while those without detectable metastases or refusing SBRT continued ADT (N/M and ADT groups). Out of 74 screened, 67 met the inclusion criteria: 47 with N+/M+ disease (29 in SBRT group, 18 in ADT group), and 20 with N/M disease. Lymph node metastases only were found in 34% of the SBRT group and 50% of the ADT group. Over a 21-month median follow-up, the ADT group’s MFS was shorter than that for the SBRT group (11 months vs. not reached; HR: 4.69, 95% CI: 4.04–40.3, p < 0.001). Similarly, the N/M group’s median MFS was not reached, indicating no significant difference between the SBRT group and the N/M group (p = 0.3). Multivariable analysis revealed SBRT as the sole MFS prognostic factor for MFS in N+/M+ patients (HR: 0.10, 95% CI: 0.03–0.32, p < 0.001). No grade ≥ 3 toxicities occurred in the SBRT group.

Discussion

The efficacy of MDT in enhancing local control is supported across the results of all referenced clinical trials. These trials also highlight the favorable tolerability and feasibility of MDT. However, a crucial consideration is that MDT primarily serves to postpone disease progression, rather than halt it entirely, thereby helping to delay the need for systemic treatment. Furthermore, the positive response rates to MDT observed might also support the hypothesis of tumor cell seeding by the metastases themselves rather than from the primary tumor, suggesting a potential mechanism behind the efficacy of localized treatments in oligometastatic settings [15]. While these outcomes are indeed promising, the limitations inherent to the studies without evidence on long-term oncologic outcome improvements (OS, CSS, or any other improved surrogate endpoint). Given that patients with oligorecurrent PC typically exhibit long median OS rates under systemic treatment [16], the emphasis on quality of life and potential treatment side effects becomes increasingly significant. Consequently, even if MDT does not markedly enhance OS, it could still play a role in the PC treatment landscape. This is due to its ability to delay systemic treatment, potentially mitigating the side effects associated with it and resulting in extended periods of high-quality life. On the other hand, MDT could also be used for therapy intensification. At present, the body of evidence primarily comprises phase 2 trials with relatively small sample sizes. Discrepancies in endpoints and definitions across studies further confound the interpretation of outcomes. Furthermore, the use of varied imaging modalities for diagnosis could potentially skew these outcomes, given that patients diagnosed via conventional imaging may be “under”-staged compared to those diagnosed using PSMA-PET [17].
In this brief review, we did not include studies examining MDT in conjunction with systemic treatment, or those conducted in alternative contexts such as primary synchronous OPC. However, recent findings from a phase 2 RCT suggest MDT + ADT may surpass ADT alone in oligometastatic prostate cancer patients with five or fewer metastases, particularly in terms of median PFS (not reached vs. 16 months, HR: 0.25, p < 0.001; [18]). This underscores that the applicability of MDT may not be confined solely to oligorecurrent PC, and combination therapies could offer enhanced efficacy. Although MDT has shown the capability of delaying systemic treatment, antiandrogenic combination therapy remains the standard of care in the oligometastatic setting [4]. Considering the side effects and the lifelong administration of this therapy, it could be argued that combining MDT with ADT, which might increase efficacy, would potentially allow for a pause in antiandrogenic therapy in cases of good and stable PSA response. However, further prospective studies are needed to clarify this question, and the definitive role of MDT in systemic PCa treatment requires robust evidence from large-scale phase 3 RCTs (the currently registered phase 3 trials are presented in Table 2). As per current guidelines [4], the application of MDT should be limited to the context of clinical trials until such evidence becomes available. Nevertheless, in clinical practice, MDT is widely used in different forms [1923]. This is largely due to the rising use of PSMA PET, which uncovers a previously unidentified stage of disease—PSMA PET-avid lesions undetectable with conventional imaging—a scenario that current guidelines do not specifically address. In this context, clinicians frequently resort to MDT as a means to impact disease progression and delay the onset of ADT-related toxicities, with a minimal risk of severe AEs. The ESTRO-ACROP consensus statement [24], despite its low level of evidence, provides valuable guidance for clinical decision-making in determining the appropriate setting and site for MDT application.

Conclusion

Metastasis-directed therapy (MDT) presents a promising therapeutic avenue for treating oligorecurrent prostate cancer following local therapy with curative intent. However, until substantial evidence from large phase 3 RCTs emerges, the application of MDT should remain within the realm of clinical trials. Furthermore, when considering MDT, it is crucial to determine the metastatic load as reliably as possible in advance via prostate-specific membrane antigen–positron emission tomography–computed tomography (PSMA-PET-CT), to ensure the treatment encompasses all metastases.
Take-home message
Metastasis-directed therapy (MDT) provides adequate local disease control and is safe and well tolerated by patients. Additionally, it can postpone disease progression, thereby delaying the need for systemic treatment. However, the current lack of level 1 evidence has led to guidelines not recommending its use outside of clinical trials. Nevertheless, it is being adopted in clinical practice, particularly for patients with prostate-specific membrane antigen–positron emission tomography (PSMA PET)-avid lesions and normal conventional imaging—a disease stage for which there are currently no clear guideline recommendations. Therefore, it is crucial that patients deciding on MDT for oligometastatic disease are thoroughly informed about the current lack of level 1 evidence and the current guideline recommendations.

Conflict of interest

J. Klemm, P. Rajwa, M. Miszczyk, S. Brönimann, E. Laukhtina, I. Tsuboi, A. Matsukawa, M.K. Parizi, P.I. Karakiewicz and S.F. Shariat declare that they have no competing interests.
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/​.

Publisher’s Note

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

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Rao A, Vapiwala N, Schaeffer EM, Ryan CJ. Oligometastatic Prostate Cancer: A Shrinking Subset or an Opportunity for Cure? Am Soc Clin Oncol Educ Book. 2019; 309–20. Rao A, Vapiwala N, Schaeffer EM, Ryan CJ. Oligometastatic Prostate Cancer: A Shrinking Subset or an Opportunity for Cure? Am Soc Clin Oncol Educ Book. 2019; 309–20.
2.
Zurück zum Zitat Phillips R, Shi WY, Deek M, Radwan N, Lim SJ, Antonarakis ES, et al. Outcomes of Observation vs Stereotactic Ablative Radiation for Oligometastatic Prostate Cancer: The ORIOLE Phase 2 Randomized Clinical Trial. Jama Oncol. 2020;6:650–9.CrossRefPubMedPubMedCentral Phillips R, Shi WY, Deek M, Radwan N, Lim SJ, Antonarakis ES, et al. Outcomes of Observation vs Stereotactic Ablative Radiation for Oligometastatic Prostate Cancer: The ORIOLE Phase 2 Randomized Clinical Trial. Jama Oncol. 2020;6:650–9.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Ost P, Reynders D, Decaestecker K, Fonteyne V, Lumen N, Bruycker AD, et al. Surveillance or Metastasis-Directed Therapy for Oligometastatic Prostate Cancer Recurrence: A Prospective, Randomized, Multicenter Phase II Trial. J Clin Oncol. 2018;36:446–53.CrossRefPubMed Ost P, Reynders D, Decaestecker K, Fonteyne V, Lumen N, Bruycker AD, et al. Surveillance or Metastasis-Directed Therapy for Oligometastatic Prostate Cancer Recurrence: A Prospective, Randomized, Multicenter Phase II Trial. J Clin Oncol. 2018;36:446–53.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;79:263–82.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;79:263–82.CrossRefPubMed
5.
Zurück zum Zitat Fizazi K, Tran N, Fein L, Matsubara N, Rodriguez-Antolin A, Alekseev BY, et al. Abiraterone plus prednisone in metastatic, castration-sensitive prostate cancer. N Engl J Med. 2017;377:352–60.CrossRefPubMed Fizazi K, Tran N, Fein L, Matsubara N, Rodriguez-Antolin A, Alekseev BY, et al. Abiraterone plus prednisone in metastatic, castration-sensitive prostate cancer. N Engl J Med. 2017;377:352–60.CrossRefPubMed
6.
Zurück zum Zitat Davis ID, Martin AJ, Stockler MR, Begbie S, Chi KN, Chowdhury S, et al. Enzalutamide with standard first-line therapy in metastatic prostate cancer. N Engl J Med. 2019;381:121–31.CrossRefPubMed Davis ID, Martin AJ, Stockler MR, Begbie S, Chi KN, Chowdhury S, et al. Enzalutamide with standard first-line therapy in metastatic prostate cancer. N Engl J Med. 2019;381:121–31.CrossRefPubMed
7.
Zurück zum Zitat Chi KN, Agarwal N, Bjartell A, Chung BH, Pereira de Santana Gomes AJ, Given R, et al. Apalutamide for metastatic, castration-sensitive prostate cancer. N Engl J Med. 2019;381:13–24.CrossRefPubMed Chi KN, Agarwal N, Bjartell A, Chung BH, Pereira de Santana Gomes AJ, Given R, et al. Apalutamide for metastatic, castration-sensitive prostate cancer. N Engl J Med. 2019;381:13–24.CrossRefPubMed
8.
Zurück zum Zitat Smith MR, Hussain M, Saad F, Fizazi K, Sternberg CN, Crawford ED, et al. Darolutamide and Survival in Metastatic, Hormone-Sensitive Prostate Cancer. N Engl J Med. 2022;386:1132–42.CrossRefPubMedPubMedCentral Smith MR, Hussain M, Saad F, Fizazi K, Sternberg CN, Crawford ED, et al. Darolutamide and Survival in Metastatic, Hormone-Sensitive Prostate Cancer. N Engl J Med. 2022;386:1132–42.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Siva S, Bressel M, Murphy DG, Shaw M, Chander S, Violet J, et al. Stereotactic Abative Body Radiotherapy (SABR) for Oligometastatic Prostate Cancer: A Prospective Clinical Trial. Eur Urol. 2018;74:455–62.CrossRefPubMed Siva S, Bressel M, Murphy DG, Shaw M, Chander S, Violet J, et al. Stereotactic Abative Body Radiotherapy (SABR) for Oligometastatic Prostate Cancer: A Prospective Clinical Trial. Eur Urol. 2018;74:455–62.CrossRefPubMed
10.
Zurück zum Zitat Kneebone A, Hruby G, Ainsworth H, Byrne K, Brown C, Guo L, et al. Stereotactic Body Radiotherapy for Oligometastatic Prostate Cancer Detected via Prostate-specific Membrane Antigen Positron Emission Tomography. Eur Urol Oncol. 2018;1:531–7. Kneebone A, Hruby G, Ainsworth H, Byrne K, Brown C, Guo L, et al. Stereotactic Body Radiotherapy for Oligometastatic Prostate Cancer Detected via Prostate-specific Membrane Antigen Positron Emission Tomography. Eur Urol Oncol. 2018;1:531–7.
11.
Zurück zum Zitat Glicksman RM, Ramotar M, Metser U, Chung PW, Liu Z, Vines D, et al. Extended Results and Independent Validation of a Phase 2 Trial of Metastasis-Directed Therapy for Molecularly Defined Oligometastatic Prostate Cancer. Int J Radiat Oncol Biol Phys. 2022;114:693–704.CrossRefPubMed Glicksman RM, Ramotar M, Metser U, Chung PW, Liu Z, Vines D, et al. Extended Results and Independent Validation of a Phase 2 Trial of Metastasis-Directed Therapy for Molecularly Defined Oligometastatic Prostate Cancer. Int J Radiat Oncol Biol Phys. 2022;114:693–704.CrossRefPubMed
12.
Zurück zum Zitat Hölscher T, Baumann M, Kotzerke J, Zöphel K, Paulsen F, Müller A‑C, et al. Toxicity and Efficacy of Local Ablative, Image-guided Radiotherapy in Gallium-68 Prostate-specific Membrane Antigen Targeted Positron Emission Tomography—staged, Castration-sensitive Oligometastatic Prostate Cancer: The OLI‑P Phase 2 Clinical Trial. European Urology Oncology. 2022;5:44–51. Hölscher T, Baumann M, Kotzerke J, Zöphel K, Paulsen F, Müller A‑C, et al. Toxicity and Efficacy of Local Ablative, Image-guided Radiotherapy in Gallium-68 Prostate-specific Membrane Antigen Targeted Positron Emission Tomography—staged, Castration-sensitive Oligometastatic Prostate Cancer: The OLI‑P Phase 2 Clinical Trial. European Urology Oncology. 2022;5:44–51.
13.
Zurück zum Zitat Hölscher T, Baumann M, Kotzerke J, Zöphel K, Paulsen F, Müller AC, et al. Local Control after Locally Ablative, Image-Guided Radiotherapy of Oligometastases Identified by Gallium-68-PSMA-Positron Emission Tomography in Castration-Sensitive Prostate Cancer Patients (OLI-P). Cancers (basel). 2022;14. Hölscher T, Baumann M, Kotzerke J, Zöphel K, Paulsen F, Müller AC, et al. Local Control after Locally Ablative, Image-Guided Radiotherapy of Oligometastases Identified by Gallium-68-PSMA-Positron Emission Tomography in Castration-Sensitive Prostate Cancer Patients (OLI-P). Cancers (basel). 2022;14.
14.
Zurück zum Zitat Pan J, Wei Y, Zhang T, Liu C, Hu X, Zhao J, et al. Stereotactic Radiotherapy for Lesions Detected via (68)Ga-Prostate-specific Membrane Antigen and (18)F-Fluorodexyglucose Positron Emission Tomography/Computed Tomography in Patients with Nonmetastatic Prostate Cancer with Early Prostate-specific Antigen Progression on Androgen Deprivation Therapy: A Prospective Single-center Study. Eur Urol Oncol. 2022;5:420–7.CrossRefPubMed Pan J, Wei Y, Zhang T, Liu C, Hu X, Zhao J, et al. Stereotactic Radiotherapy for Lesions Detected via (68)Ga-Prostate-specific Membrane Antigen and (18)F-Fluorodexyglucose Positron Emission Tomography/Computed Tomography in Patients with Nonmetastatic Prostate Cancer with Early Prostate-specific Antigen Progression on Androgen Deprivation Therapy: A Prospective Single-center Study. Eur Urol Oncol. 2022;5:420–7.CrossRefPubMed
15.
Zurück zum Zitat Kim M‑Y, Oskarsson T, Acharyya S, Nguyen DX, Zhang XHF, Norton L, et al. Tumor Self-Seeding by Circulating Cancer Cells. Cell. 2009;139:1315–26.CrossRefPubMedPubMedCentral Kim M‑Y, Oskarsson T, Acharyya S, Nguyen DX, Zhang XHF, Norton L, et al. Tumor Self-Seeding by Circulating Cancer Cells. Cell. 2009;139:1315–26.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Sweeney CJ, Chen Y‑H, Carducci M, Liu G, Jarrard DF, Eisenberger M, et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. N Engl J Med. 2015;373:737–46.CrossRefPubMedPubMedCentral Sweeney CJ, Chen Y‑H, Carducci M, Liu G, Jarrard DF, Eisenberger M, et al. Chemohormonal Therapy in Metastatic Hormone-Sensitive Prostate Cancer. N Engl J Med. 2015;373:737–46.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Hofman MS, Lawrentschuk N, Francis RJ, Tang C, Vela I, Thomas P, et al. Prostate-specific membrane antigen PET-CT in patients with high-risk prostate cancer before curative-intent surgery or radiotherapy (proPSMA): a prospective, randomised, multicentre study. Lancet. 2020;395:1208–16.CrossRefPubMed Hofman MS, Lawrentschuk N, Francis RJ, Tang C, Vela I, Thomas P, et al. Prostate-specific membrane antigen PET-CT in patients with high-risk prostate cancer before curative-intent surgery or radiotherapy (proPSMA): a prospective, randomised, multicentre study. Lancet. 2020;395:1208–16.CrossRefPubMed
18.
Zurück zum Zitat Tang C, Sherry AD, Haymaker C, Bathala T, Liu S, Fellman B, et al. Addition of Metastasis-Directed Therapy to Intermittent Hormone Therapy for Oligometastatic Prostate Cancer: The EXTEND Phase 2 Randomized Clinical Trial. JAMA Oncol. 2023;9:825–34.CrossRefPubMed Tang C, Sherry AD, Haymaker C, Bathala T, Liu S, Fellman B, et al. Addition of Metastasis-Directed Therapy to Intermittent Hormone Therapy for Oligometastatic Prostate Cancer: The EXTEND Phase 2 Randomized Clinical Trial. JAMA Oncol. 2023;9:825–34.CrossRefPubMed
19.
Zurück zum Zitat Milenkovic U, Kuijk J, Roussel E, Devos G, Van den Broeck T, Van Eecke H, et al. Predictors of Recurrence After Metastasis-directed Therapy in Oligorecurrent Prostate Cancer Following Radical Prostatectomy. Eur Urol Oncol. 2023. Milenkovic U, Kuijk J, Roussel E, Devos G, Van den Broeck T, Van Eecke H, et al. Predictors of Recurrence After Metastasis-directed Therapy in Oligorecurrent Prostate Cancer Following Radical Prostatectomy. Eur Urol Oncol. 2023.
20.
Zurück zum Zitat De Bleser E, Jereczek-Fossa BA, Pasquier D, Zilli T, Van As N, Siva S, et al. 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. 2019;76:732–9.CrossRefPubMed De Bleser E, Jereczek-Fossa BA, Pasquier D, Zilli T, Van As N, Siva S, et al. 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. 2019;76:732–9.CrossRefPubMed
21.
Zurück zum Zitat Steuber T, Jilg C, Tennstedt P, De Bruycker A, Tilki D, Decaestecker K, et al. Standard of Care Versus Metastases-directed Therapy for PET-detected Nodal Oligorecurrent Prostate Cancer Following Multimodality Treatment: A Multi-institutional Case-control Study. Eur Urol Focus. 2019;5:1007–13.CrossRefPubMed Steuber T, Jilg C, Tennstedt P, De Bruycker A, Tilki D, Decaestecker K, et al. Standard of Care Versus Metastases-directed Therapy for PET-detected Nodal Oligorecurrent Prostate Cancer Following Multimodality Treatment: A Multi-institutional Case-control Study. Eur Urol Focus. 2019;5:1007–13.CrossRefPubMed
22.
Zurück zum Zitat Maurer T, Graefen M, van der Poel H, Hamdy F, Briganti A, Eiber M, et al. Prostate-Specific Membrane Antigen-Guided Surgery. J Nucl Med. 2020;61:6–12.CrossRefPubMed Maurer T, Graefen M, van der Poel H, Hamdy F, Briganti A, Eiber M, et al. Prostate-Specific Membrane Antigen-Guided Surgery. J Nucl Med. 2020;61:6–12.CrossRefPubMed
23.
Zurück zum Zitat Maurer T, Robu S, Schottelius M, Schwamborn K, Rauscher I, van den Berg NS, et al. (99m)Technetium-based Prostate-specific Membrane Antigen-radioguided Surgery in Recurrent Prostate Cancer. Eur Urol. 2019;75:659–66.CrossRefPubMed Maurer T, Robu S, Schottelius M, Schwamborn K, Rauscher I, van den Berg NS, et al. (99m)Technetium-based Prostate-specific Membrane Antigen-radioguided Surgery in Recurrent Prostate Cancer. Eur Urol. 2019;75:659–66.CrossRefPubMed
24.
Zurück zum Zitat Zilli T, Achard V, Dal Pra A, Schmidt-Hegemann N, Jereczek-Fossa BA, Lancia A, et al. Recommendations for radiation therapy in oligometastatic prostate cancer: An ESTRO-ACROP Delphi consensus. Radiother Oncol. 2022;176:199–207.CrossRefPubMed Zilli T, Achard V, Dal Pra A, Schmidt-Hegemann N, Jereczek-Fossa BA, Lancia A, et al. Recommendations for radiation therapy in oligometastatic prostate cancer: An ESTRO-ACROP Delphi consensus. Radiother Oncol. 2022;176:199–207.CrossRefPubMed
Metadaten
Titel
Metastasis-directed therapy: new standard or too early to change paradigm?
verfasst von
Jakob Klemm
Pawel Rajwa
Marcin Miszczyk
Stephan Brönimann
Ekaterina Laukhtina
Ichiro Tsuboi
Akihiro Matsukawa
Mehdi Kardoust Parizi
Pierre I. Karakiewicz
Shahrokh F. Shariat
Publikationsdatum
29.12.2023
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 1/2024
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-023-00945-7

Weitere Artikel der Ausgabe 1/2024

memo - Magazine of European Medical Oncology 1/2024 Zur Ausgabe