Skip to main content

Main menu

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Journal of Nuclear Medicine
  • SNMMI
    • JNM
    • JNMT
    • SNMMI Journals
    • SNMMI
  • Subscribe
  • My alerts
  • Log in
  • Log out
  • My Cart
Journal of Nuclear Medicine

Advanced Search

  • Home
  • Content
    • Current
    • Ahead of print
    • Past Issues
    • JNM Supplement
    • SNMMI Annual Meeting Abstracts
    • Continuing Education
    • JNM Podcasts
  • Subscriptions
    • Subscribers
    • Institutional and Non-member
    • Rates
    • Journal Claims
    • Corporate & Special Sales
  • Authors
    • Submit to JNM
    • Information for Authors
    • Assignment of Copyright
    • AQARA requirements
  • Info
    • Reviewers
    • Permissions
    • Advertisers
  • About
    • About Us
    • Editorial Board
    • Contact Information
  • More
    • Alerts
    • Feedback
    • Help
    • SNMMI Journals
  • View or Listen to JNM Podcast
  • Visit JNM on Facebook
  • Join JNM on LinkedIn
  • Follow JNM on Twitter
  • Subscribe to our RSS feeds
Research ArticleThe State of the Art

Treatment Landscape of Prostate Cancer in the Era of PSMA Radiopharmaceutical Therapy

Fabio Turco, Silke Gillessen, Ken Herrmann, Gaetano Paone and Aurelius Omlin
Journal of Nuclear Medicine May 2025, 66 (5) 665-672; DOI: https://doi.org/10.2967/jnumed.124.267730
Fabio Turco
1Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Silke Gillessen
1Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland;
2Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Ken Herrmann
3Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium, University Hospital Essen, Essen, Germany;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Gaetano Paone
2Faculty of Biosciences, Università della Svizzera Italiana, Lugano, Switzerland;
4Clinic of Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Aurelius Omlin
5Onkozentrum Zurich, University of Zurich and Tumorzentrum Hirslanden Zurich, Zurich, Switzerland
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Supplemental
  • Info & Metrics
  • PDF
Loading

Abstract

The treatment landscape of prostate cancer is quite complex because of the many therapeutic options available in different disease settings (hormonal treatments, chemotherapy, poly(adenosine diphosphate ribose) polymerase inhibitors, radiopharmaceutical therapy). Since in most cases we do not have comparative studies between these different agents, the best therapeutic sequence in patients with prostate cancer remains unsolved. In this review, we describe the different systemic therapeutic options available in each disease setting from localized disease to metastatic castration-resistant disease. We also indicate when to use each of these therapeutic options in the therapeutic sequence on the basis of the results of the available studies. A special focus of this review is the place of prostate-specific membrane antigen radiopharmaceutical therapy in the treatment algorithms.

  • radiopharmaceuticals
  • advanced prostate cancer
  • radiopharmaceutical therapy
  • systemic therapies
  • mCRPC
  • mHSPC

In the past 20 y, the landscape of advanced prostate cancer (PC) has changed dramatically. Since the publication of the TAX-324 and SWOG-9916 trials, which established docetaxel as the first survival-prolonging systemic therapy for patients with metastatic castration-resistant PC (mCRPC), a wide range of different treatment modalities has been shown to prolong overall survival (OS) in this setting (Fig. 1) (1–3). Some of these therapeutic strategies have improved OS even in earlier settings such as nonmetastatic CRPC (nmCRPC) (4–6), metastatic hormone-sensitive PC (mHSPC) (7–16), high-risk localized disease (17), and high-risk biochemical recurrence (18).

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Treatment landscape of advanced PC. dMMR = deficient mismatch repair; DT = doubling time; MSI = microsatellite instability; nmHSPC = nonmetastatic HSPC.

Since all these therapeutic options are now available for the treatment of patients with PC in different disease settings, and since in most situations there is a lack of direct comparison studies between these different therapies, the therapeutic choice for each individual patient in each specific disease setting is becoming increasingly complex. The aim of this review is to summarize the currently available evidence. Since high-level evidence to help treatment decisions is often missing, when appropriate we use in this review the voting results from the Advanced PC Consensus Conference (APCCC) of 2024 (19). A special focus of this review is the place of prostate-specific membrane antigen (PSMA) radiopharmaceutical therapy in the treatment algorithms.

HIGH-RISK LOCALIZED DISEASE

In 2021, the results of STAMPEDE arms G and J regarding patients with nonmetastatic disease were published (17). In that study, the addition of androgen deprivation therapy (ADT) for 3 y plus abiraterone with or without enzalutamide for 2 y to radiation therapy (RT) to the prostate was evaluated in patients with clinically node-positive (cN1) disease and patients with high-risk node-negative (cN0) disease (defined as having ≥2 of the following: >cT3, Gleason score >8, prostate-specific antigen [PSA] ≥40 ng/mL) compared with radiotherapy plus ADT for 3 y. The trial also enrolled patients with relapsing nonmetastatic disease with high-risk features (≤12 mo of total ADT with an interval of ≥12 mo without treatment and PSA concentration ≥4 ng/mL with a doubling time of <6 mo, or a PSA concentration ≥20 ng/mL, or nodal relapse). However, patients with relapsing disease were only 3% of the total population.

The experimental arm was shown to improve metastasis-free survival and OS (hazard ratio, 0.54 [95% CI, 0.43–0.68] and 0.6 [95% CI, 0.48–0.73], respectively). There was no difference in metastasis-free survival or OS when enzalutamide and abiraterone were administered concurrently compared with administration of abiraterone alone; thus, adverse events were more frequent (58% vs. 38%). According to the results of the study, the guidelines of the National Comprehensive Cancer Network and European Association of Urology recommend using 2 y of abiraterone when offering RT to the prostate in combination with long-term ADT for patients with cN1 or with cN0 high-risk disease according to the STAMPEDE definition (20,21). The trial used conventional imaging (CT and bone scintigraphy), and it is clear that with more sensitive imaging, in particular PSMA PET/CT, some of the patients would have had at least low-volume mHSPC.

To date, only small phase I/II trials have evaluated PSMA radiopharmaceutical therapy in patients with localized disease (22,23). Golan et al. conducted a clinical trial with 14 patients with high-risk localized PC (22). Before radical prostatectomy (RP), patients were treated with 2–3 doses of 177Lu-PSMA-I&T (7.4 GBq with a 2-wk interval between cycles). The median PSA reduction registered was 3.45 and 4.3 ng/mL after 2 and 3 177Lu-PSMA doses, respectively. In the LuTectomy trial, 20 patients received 1–2 doses of 177Lu-PSMA-617 (5 GBq with a 6-wk interval between cycles) before RP (23). The primary endpoint was tumor-absorbed radiation dose, defined as the maximum dose in grays within the prostate and the involved lymph nodes. Almost half the patients achieved a 50% PSA reduction at 6 wk after receiving 177Lu-PSMA, and the treatment was well tolerated. At restaging PSMA PET/CT after the treatment and before surgery, a partial PSMA response was achieved in 11 (55%) of 20 patients. Progressive PSMA disease with an increased SUVmax occurred in 1 (5%) of 20 patients. The remaining 8 (40%) patients had stable PSMA disease. All patients underwent RP, and no surgical complications were attributed to 177Lu-PSMA.

In these 2 trials were used 2 different molecules (177Lu-PSMA I&T vs. 177Lu-PSMA 617), different numbers of cycles (2–3 vs. 1–2 cycles), different dosages (7.4 vs. 5 GBq), and different intervals between cycles (every 2 wk vs. every 6 wk). Interestingly, in both studies, 177Lu-PSMA was administered without ADT (22,23). 177Lu-PSMA seemed to show some efficacy as a neoadjuvant treatment before RP in patients with high-risk localized disease. However, these preliminary results need to be verified in bigger, randomized trials.

HIGH-RISK BIOCHEMICAL RECURRENCE

Results from the randomized phase 3 EMBARK trial were published in 2023 (18). This trial included patients with nonmetastatic high-risk biochemical recurrence, defined as a PSA doubling time of 9 mo or less and a PSA level at least 2 ng/mL above the nadir after RT or at least 1 ng/mL above the nadir after surgery, with or without postoperative RT. Conventional imaging was used for staging. Of the participants, 25% had previously undergone RP alone, 26% received RT alone, and 49% had both RP and postoperative RT. Patients were randomized to receive 37 wk of ADT alone, enzalutamide alone, or a combination of enzalutamide and ADT. Treatment was suspended at week 37 if the PSA level was less than 0.2 ng/mL and was restarted when the PSA level was at least 5 ng/mL (if the patient had not had previous RP) or at least 2 ng/mL (if the patient had previously had RP). Patients who did not reach a PSA of 0.2 ng/mL or less at week 37 continued to receive their assigned treatments until progression or until an unacceptable adverse event occurred. Regarding metastasis-free survival, both the combination therapy and enzalutamide alone showed superior outcomes to ADT alone. EMBARK used conventional imaging, and it is clear that with more sensitive imaging, in particular PSMA PET, many of these patients have at least low-volume mHSPC. A poster presentation at the American Society of Clinical Oncology 2023 meeting on 183 EMARK-like patients staged by PSMA PET reported local relapse in 9%, pelvic nodal disease in 40%, distant lymph node metastases in 39%, and bone metastases in 23% (24). The use of androgen-receptor pathway inhibitor (ARPI) alone or in combination with ADT may change the natural history of the disease and has implications on the available treatment options at progression.

NOTEWORTHY

  • The treatment landscape in PC is becoming increasingly complex, as we have different therapeutic options available in different disease settings without, in most cases, having comparative studies between the different available drugs.

  • No single therapeutic sequence is valid for all patients. In each therapeutic choice, several factors must be considered: previous treatments; performance status; comorbidity; presence or absence of symptoms; clinical, biologic, and genomic characteristics of the disease; patient’s preferences; availability of drugs.

  • To date, the strongest evidence on the efficacy of PSMA radiopharmaceutical therapy is in the metastatic castration-resistant setting; this treatment should therefore be used only in this setting outside of clinical trials

  • Because several ongoing studies are evaluating the efficacy of PSMA radiopharmaceutical therapy in earlier settings, its use in the therapeutic algorithm is anticipated.

To our knowledge, there are no published data about the use of 177Lu-PSMA in the biochemical recurrence setting. However, there is an ongoing phase I/II trial that is evaluating 177Lu-PSMA I&T in combination with high-dose-rate brachytherapy in patients with locally recurrent PC after previous definitive RT (25).

MHSPC SETTING

For about a decade, ADT monotherapy has no longer been the standard of care in patients with mHSPC (20,21,26). Several studies have demonstrated that combination therapies are able to improve OS compared with ADT monotherapy (Figs. 2 and 3) (26). ADT plus docetaxel was the first combination shown to improve outcomes for patients with mHSPC, especially in patients with high-volume disease (7–9). Subsequently, the addition of an ARPI (abiraterone, darolutamide, enzalutamide, or apalutamide) to ADT has also been shown to improve OS compared with ADT alone (10–14). Recently, data from the ARANOTE study were presented (27). In this trial, the combination of ADT plus darolutamide improved radiographic progression-free survival (rPFS) compared with ADT alone. At a median follow-up of approximately 25 mo, OS data of this study were not yet mature. Finally, 2 phase 3 clinical trials, PEACE 1 and ARASENS, showed that triplet systemic therapy consisting of ADT plus 6 cycles of docetaxel plus abiraterone or darolutamide improves OS in comparison to doublet therapy consisting of ADT plus 6 cycles of docetaxel (15,16). We are not aware of any randomized evidence, to date, on whether systemic triplet therapy improves outcomes in comparison to doublet therapy with ADT plus an ARPI (28). At the APCCC 2024 meeting, panelists voted on questions regarding the use of systemic triplet therapy in different scenarios based on timing of metastatic presentation (de novo vs. metachronous) and volume of disease according to the CHAARTED criteria (low vs. high volume) (19). In none of the 4 subgroups analyzed (synchronous low volume, synchronous high volume, metachronous low volume, and metachronous high volume) was a consensus reached. However, in patients with high-volume disease, 94% and 84% of the panelists voted in favor of recommending triplet therapy at least for selected patients.

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Treatment options in mHSPC for high volume/risk.

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Treatment options in mHSPC for low volume/risk.

According to the evidence described, the current international guidelines recommended ADT plus ARPI with or without docetaxel as the standard-of-care systemic treatment in patients with mHSPC (20,21).

Studies on patients with mHSPC have evaluated the effect of RT to the primary (29–32). RT to the primary did not improve OS in the overall population in any of the randomized trials. For patients with low-volume synchronous mHSPC, RT to the primary in addition to ADT improved OS in a prespecified and prepowered subgroup analysis for 1 study (STAMPEDE) but not in the 2 other phase 3 trials (HORRAD and PEACE-1) (29,30,32). The only evidence regarding the effect of adding both RT to the primary tumor and an ARPI to the standard of care for patients with PC comes from PEACE-1 (32). For patients with low-volume disease, the standard of care (ADT ± docetaxel) plus abiraterone plus RT to the primary tumor improved rPFS, but not OS, in comparison to the standard of care plus abiraterone. In the PEACE-1 trial, RT to the primary reduced the occurrence of serious genitourinary events in patients independently of volume of disease. Interestingly, at the APCCC meeting there was a consensus to recommend ADT plus ARPI plus RT to the primary tumor in most patients with synchronous low-burden mHSPC (19).

Since all the studies that evaluated combination systemic therapies and RT to the primary used conventional imaging, it is unclear whether we can extrapolate the results to patients with metastatic disease diagnosed only with next-generation imaging such as PSMA PET/CT. Therefore, in daily practice, clinicians face the increasingly challenging question of which treatment to recommend for patients with synchronous oligometastatic PC.

There is no randomized trial evidence for synchronous oligometastatic HSPC suggesting a benefit of systemic therapy plus treatment of the primary plus metastasis-directed therapy for all metastases, nor is there a formal and generally accepted definition for oligometastatic disease. At the APCCC meeting, there was no consensus to recommend metastasis-directed therapy in most patients with synchronous low-burden mHSPC on conventional imaging or on next-generation imaging (19). However, there was a consensus to recommend metastasis-directed therapy for selected patients with synchronous low-burden mHSPC on next-generation imaging.

PSMA radiopharmaceutical therapy is currently not a standard of care in the mHSPC setting. Privé et al. conducted a prospective pilot study assessing the efficacy of 177Lu-PSMA-617 in 10 patients with mHSPC with a maximum of 10 metastatic lesions on PSMA PET/CT (33). These patients received 2 cycles of 177Lu-PSMA-617 (3 GBq the first cycle followed by a second cycle with 3–6 GBq after 7–9 wk) without ADT. Half the patients experienced a PSA reduction of more than 50%, with 1 patient achieving an undetectable PSA level. At 24 wk after the second cycle, 5 patients displayed at least a partial response, with 1 patient revealing a complete response, and the disease progressed in 4 patients. There are several ongoing studies evaluating 177Lu-PSMA in mHSPC both as monotherapy and in combination with the standard of care. The results of the UpFrontPSMA trial were recently published (34). This is a randomized, phase 2 trial in which patients with high-volume mHSPC were randomized to receive ADT plus 2 cycles of 177Lu-PSMA-617 7.5 GBq every 6 wk followed 6 wk later by 6 cycles of docetaxel or ADT plus 6 cycles of docetaxel. The primary endpoint was undetectable PSA (≤0.2 ng/mL) at 48 wk. Twenty-five (41%) of 61 patients in the experimental arm had undetectable PSA at 48 wk, compared with 10 (16%) of 61 patients in the ADT-plus-docetaxel group (odds ratio, 3.88; 95% CI, 1.61–9.38; P = 0.0020). Treatment with 177Lu-PSMA was generally well tolerated. Despite the fact that ADT plus docetaxel is currently no longer the standard of care, these data may suggest that 177Lu-PSMA could potentially have a role in the management of mHSPC. The randomized phase 3 PSMAddition study randomizing patients with mHSPC to ADT plus ARPI plus 6 cycles of 177Lu-PSMA-617 has completed enrollment (35). The results of this study will clarify the role of 177Lu-PSMA in this setting.

GENERAL CRPC

Castration-resistant PC refers to the condition in which PC continues to progress despite ADT.

A molecular pathologic examination of tumor tissue to identify therapy-relevant alterations (especially BRCA1/2 and other homologous recombination defects [HRR alterations], defective mismatch repair proteins, and microsatellite instability) should be performed at the latest when progression from the hormone-sensitive to the castration-resistant phase is imminent (20).

NMCRPC

In cases of PSA progression and in the absence of evidence of metastases on conventional imaging, the condition is formally classified as nmCRPC. A PSA level of at least 2 µg/L and a PSA doubling time of no more than 10 mo are associated with a poor prognosis and an increased risk of metastases (36). Three large studies (ARAMIS, PROSPER, and SPARTAN) have demonstrated a statistically significant prolongation of both rPFS and OS through the early use of an ARPI (apalutamide, darolutamide, enzalutamide) (4–6). It is important to emphasize in this context that modern imaging with PSMA PET was not used in the 3 studies mentioned. The concept of nmCRPC is an artificial one that is seen when high-risk patients are evaluated using PSMA PET, because evidence of metastases (including locoregional and distant metastases) can be found in more than 90% of patients but may not have been recognized as such on conventional imaging because of its lower accuracy (37). The use of ARPI in combination with ADT in the nmCRPC setting may change the natural history of the disease and has implications on the available treatment options at progression.

POSSIBLE SEQUENCE THERAPIES IN MCRPC

Crucial to the choice of therapy in the castration-resistant situation is prior treatment. Accordingly, different scenarios are listed and discussed separately. Supplemental Table 1 (supplemental materials are available at http://jnm.snmjournals.org) summarizes the available treatments (38).

The studies PROpel, MAGNITUDE, and TALAPRO-2 have investigated the benefit of ARPI plus poly(adenosine diphosphate ribose) polymerase (PARP) inhibitor combination therapy in the first-line mCRPC setting. (39–41). Preclinically, there is evidence of a synergistic effect between PARP inhibition and ARPI. However, clinical studies show that patients with BRCA mutations benefit most from the combination (39–41). Combination therapy has less of an effect in patients with other HRR alterations, and data for patients without HRR alterations have been controversially discussed. A significant OS benefit has yet to be demonstrated in any study at the time of writing.

It is also important to note that most patients included in the 3 studies had previously received only ADT as treatment; only 20%–30% received docetaxel in the mHSPC situation, and less than 10% received an ARPI. The use of a PARP inhibitor is associated with significant, primarily hematologic, toxicity. In all 3 studies, many patients had to interrupt therapy (≤75%), reduce the PARP inhibitor dose (≤50%), or even discontinue PARP inhibitor therapy (≤20%). Regulatory authorities interpret the data differently. The Food and Drug Administration has approved the combinations for patients with BRCA alteration (niraparib plus abiraterone and olaparib plus abiraterone) and for patients with HRR alterations including BRCA (talazoparib plus enzalutamide). Similarly to the Food and Drug Administration, the European Medicines Agency has approved the combination of niraparib plus abiraterone only for patients with BRCA alteration, whereas the other 2 combinations (olaparib plus abiraterone and talazoparib plus enzalutamide) have been approved for all patients with mCRPC regardless of mutation status when chemotherapy is not indicated.

TREATMENT OPTIONS AFTER ADT ALONE OR ADT PLUS DOCETAXEL FOR MHSPC

This situation will become less common because of the increased use of ADT-plus-ARPI combination therapies in mHSPC. In the absence of molecular alterations, an ARPI is primarily considered in this situation. A consensus was reached on this at the APCCC 2024 consensus conference (Supplemental Table 2) (19).

There is strong evidence from 2 phase III studies for abiraterone and enzalutamide (3). In these pivotal studies (PREVAIL and COU-302), only patients with asymptomatic or minimally symptomatic disease were included, and no chemotherapy pretreatment was allowed. Patients with visceral metastases or a low PSA level relative to the extent of metastasis should be closely monitored for early detection of disease progression. A valid alternative, especially in patients with unfavorable prognostic factors, could be cabazitaxel since in the TROPIC trial, cabazitaxel was shown to improve OS compared with mitoxantrone in patients with mCRPC progressing after docetaxel (3).

Data presented at the European Society for Medical Oncology meeting on the PEACE-3 trial showed a significant improvement in rPFS for the first-line mCRPC combination of 223Ra plus enzalutamide in patients with asymptomatic or mildly symptomatic bone-predominant metastases (42). About 30% of the included patients had received prior docetaxel, and very few patients (<3%) had received abiraterone for mHSPC. The study met its primary endpoint: enzalutamide plus 223Ra improved rPFS compared with placebo (hazard ratio, 0.69; 95% CI, 0.54–0.87). With 80% of events achieved, the combination therapy was shown to also improve OS in the log-rank test (0.69; 95% CI, 0.52–0.90); however, this study has not been published yet, and recommendations should therefore be given only after formal publication of the results. The use of a bone-protecting agent has been mandatory during the trial, resulting in a drastic reduction in the number of bone fractures in both arms (43). This confirms the importance, already at the start, of adding a bone-protecting agent in most patients at the start of ADT for prevention of cancer treatment–related bone loss and, more lately, in the setting of mCRPC with bone metastases to reduce the risk of skeleton-related events independently of the treatment given. The combination of enzalutamide plus 223Ra may be an option for patients with mCRPC and bone metastases who have received only ADT or ADT plus docetaxel in the mHSPC situation.

With the identification of a pathogenic BRCA alteration, data from the 3 studies (PROpel, MAGNITUDE, and TALAPRO-2) permit the use of combination therapy (ARPI plus PARP inhibitor) (39–41). The control arms of the 3 studies demonstrated that patients with a BRCA alteration respond significantly less favorably to an ARPI than does the control group without such a mutation. This finding supports the use of combination therapy. A sequential approach, meaning first an additional ARPI therapy followed by the use of a PARP inhibitor on progression, could theoretically also be possible. The data from the small, randomized phase II study BRCAAway (44) generated interesting data on this question, showing the best PFS primarily for the combined use of abiraterone and olaparib compared with sequential use. However, with a total of only 61 treated patients across 3 study arms, these data must be interpreted cautiously.

A consensus was also reached on this subject at the APCCC 2024 consensus conference (Supplemental Table 2) (19).

TREATMENT OPTIONS AFTER ADT PLUS ARPI FOR MHSPC

Today, most patients in the mHSPC setting are treated with ADT plus ARPI. A direct switch of ARPI is generally not recommended. On the basis of prospective studies and data from the control arms of randomized phase III studies, a direct ARPI switch shows very low antitumor activity (3).

On the basis of the TAX-327 study, docetaxel is currently the standard subsequent treatment in the castration-resistant phase, although in that study the patients had not previously received an ARPI (3). In the absence of molecular alterations, chemotherapy with docetaxel is primarily used in this situation. A consensus was also reached on this subject at the APCCC 2024 consensus conference (Supplemental Table 2) (19). If chemotherapy is not possible, the use of PSMA radiopharmaceutical therapy or 223Ra should be evaluated (19).

Data from the PSMAfore study, presented at the European Society for Medical Oncology 2023 meeting and subsequently published, demonstrated a significant advantage in rPFS for 177Lu-PSMA-617 (7.4 GBq intravenously every 6 wk for ≤6 cycles) compared with ARPI switching in patients who had not yet received chemotherapy and for whom chemotherapy was not urgently indicated (median rPFS, 11.6 vs. 5.59 mo) (45). To date, an OS benefit has not been demonstrated in the study, partly because many patients from the control arm were able to switch to the radiopharmaceutical therapy arm on disease progression.

At the European Society for Medical Oncology 2024 meeting, the data of the phase 3 SPLASH trial were presented. This trial enrolled PSMA PET–positive patients with mCRPC who progressed on 1 ARPI and had not received chemotherapy for mCRPC. Patients were randomized to 177Lu-PNT2002 (6.8 GBq intravenously every 8 wk for ≤4 cycles) or an ARPI change with a primary endpoint of rPFS. This trial also met the primary endpoint of improvement in rPFS (median rPFS, 9.5 vs. 6 mo) (46). Formal approval for the use of radiopharmaceutical therapy for patients with mCRPC who have not yet received chemotherapy is currently pending in many countries. It is anticipated that this treatment option will gain increasing importance in this situation over the next few years, especially with good patient selection, considering that chemotherapy remains an active treatment option after radiopharmaceutical therapy. Another phase III trial with 177Lu-PNT2002 (ECLIPSE) has compared 6 doses of 177Lu-PNT2002 (7.4 GBq every 6 wk) with an ARPI switch, and the data are expected to be presented soon.

For patients with a pathogenic BRCA alteration, there are various options available. There are strong data for monotherapy with PARP inhibitors (olaparib, rucaparib) (47,48). Currently, olaparib is approved only for patients with pathogenic BRCA alterations (germline or somatic). The 3 large, randomized studies (PROpel, MAGNITUDE, and TALAPRO-2) included few patients who had previously received an ARPI. Given that sequential use of ARPIs is associated with only low antitumor activity, the question arises as to whether a combined treatment with ARPI plus PARP inhibitor would be sensible for patients with progression under ADT and an ARPI. No consensus was reached on this at the APCCC 2024 meeting, but the overwhelming majority voted in favor of using a PARP inhibitor, whether as monotherapy or in combination with an ARPI (Supplemental Table 2) (19).

For a molecular profile associated with an increased likelihood of response to checkpoint inhibitor therapy (defective mismatch repair proteins, MSI-high), such therapy may be considered in individual cases in the mCRPC situation (49). The use can be justified by 2 studies showing efficacy independent of the cancer of origin, on which bases the Food and Drug Administration has approved pembrolizumab and dostarlimab (50,51).

TREATMENT OPTIONS AFTER ADT PLUS ARPI PLUS DOCETAXEL FOR MHSPC

A particular challenge in the future will be patients who have already received a triple-combination therapy (ADT plus ARPI plus docetaxel) in mHSPC. There is currently no strong evidence from prospective, randomized phase III studies for this specific patient group. Some of the considerations mentioned above also apply in this situation.

In the absence of molecular alterations, radiopharmaceutical therapy extrapolated from the VISION study could be given in this context (52). A prerequisite is PSMA PET imaging, which demonstrates relevant PSMA expression in all tumor manifestations. The VISION trial compared 177Lu-PSMA-617 (7.4 GBq every 6 wk for 4–6 cycles) plus the protocol-permitted standard care with standard care alone. Protocol-permitted standard care excluded chemotherapy, immunotherapy, 223Ra, and investigational drugs but allowed an ARPI switch, low-dose steroids, palliative RT, and osteoprotective therapy with bisphosphonates or denosumab. 177Lu-PSMA-617 plus standard care significantly prolonged median rPFS (8.7 vs. 3.4 mo) and median OS (15.3 vs. 11.3 mo) and has led to the approval of the radiopharmaceutical therapy in patients with mCRPC after treatment with an ARPI and 1 line of chemotherapy.

Alternatively, cabazitaxel can be used on the basis of the TROPIC study (3). A direct comparison of cabazitaxel with 177Lu-PSMA-617 in the TheraP trial showed a better response rate and tolerability for radiopharmaceutical therapy than was seen with cabazitaxel but no significant improvement in OS (53,54). Of note, the dose of 177Lu-PSMA-617 used in the TheraP trial was higher than the dose used in VISION or PSMAfore.

Patients with symptomatic progression and evidence of metabolically active bone metastases without visceral metastases, who are not fit for chemotherapy with docetaxel, might be considered for radiopharmaceutical therapy using 223Ra (55).

For patients with a pathogenic BRCA alteration, the considerations outlined above apply. The use of a PARP inhibitor could be an effective treatment option after pretreatment with ADT plus ARPI plus docetaxel. No consensus was reached on this at the APCCC 2024 meeting, but the overwhelming majority voted in favor of using a PARP inhibitor, whether as monotherapy or in combination with an ARPI (Supplemental Table 2) (19).

GENERAL CONSIDERATIONS

Currently, there is no uniform clinical, laboratory, or pathologic definition for the aggressive variant of PC. It is suspected in cases of, for example, new or extensive visceral progression, disease progression with stable or low PSA levels, or new osteolytic bone metastases. If possible, a biopsy should be performed to rule out a treatment-emergent neuroendocrine dedifferentiation or a second malignancy. The biopsy can investigate whether a dedifferentiated adenocarcinoma variant (often still showing PSA and androgen receptor expression) or a neuroendocrine differentiation (expression of synaptophysin, chromogranin A, CD56) is present. The biopsy material may also be useful for further molecular pathologic investigations. In case of diagnosis of aggressive-variant PC, an intensified treatment such as cabazitaxel plus carboplatin could be considered according to the data of the phase I/II study by Corn et al. (56).

Low platelets can be side effect of treatment (mostly chemotherapy or radiopharmaceutical therapy) but also a sign of disease progression in the sense of development of a disseminated intravascular coagulation or progressing bone marrow infiltration. Therefore, patients with thrombocytopenia have to be followed up closely and evaluated to rule out reasons other than a side effect of the treatment.

Furthermore, as recommended by international guidelines, patients with mCRPC and bone metastases should receive a bone-protecting agent (denosumab or zoledronic acid monthly) in the absence of contraindications to reduce the risk of skeleton-related events (20,21,57,58). Even in patients with hormone-sensitive disease who start long-term ADT with or without ARPI, bone health should be carefully considered, introducing calcium and vitamin D supplementation and evaluating the introduction of a bone-protecting agent to reduce ADT-related bone loss (59).

CONCLUSION

For patients with mCRPC, various treatment options are available. These include ARPI (abiraterone and enzalutamide), chemotherapy (docetaxel and cabazitaxel), PARP inhibitor (monotherapy or in combination with an ARPI), and radiopharmaceutical therapy (223Ra and 177Lu-PSMA). PSMA radiopharmaceutical therapy, in particular, might in the future be used in early disease settings (awaiting the results of ongoing studies). However, several questions remain regarding this treatment. Which patients can benefit most from this therapy? How does one choose between the various PSMA radioligand molecules (177Lu-PSMA I&T vs. 177Lu-PSMA 617)? What is the ideal dose, scheduling, and timing of these therapies? Therefore, it appears of fundamental importance to establish an even closer collaboration between oncologists and nuclear medicine physicians and, ideally, the presence of a specialist nuclear medicine physician on multidisciplinary tumor boards.

An optimal therapy sequence cannot currently be defined; the treatment sequence is based on the individual extent of the disease, existing symptoms, comorbidities, medications, and previously administered therapies. Closely monitoring patients with mCRPC is essential to not lose the opportunity to switch to another active therapy when the current one no longer works. For this reason, regular imaging should be performed, and when PSMA PET CT is used, the CT part should be done with contrast medium so as not to miss PSMA-negative progression.

DISCLOSURE

Fabio Turco receives institutional honoraria for participation in advisory boards from Bayer and receives speaker honoraria from Silvio Grasso Consulting and SAKK. Silke Gillessen (last 2 y) receives personal honoraria as an invited speaker for the Swiss Group for Clinical Cancer Research (SAKK), ESMO, and Schweizerische Gesellschaft für Medizinische Onkologie (SGMO)/Meister ConCept GmbH; receives other honoraria from University of Applied Sciences and Arts of Southern Switzerland (SUPSI); receives travel grants from Bayer, Gilead, and Intellisphere LLC; receives institutional honoraria for participating on advisory boards or in Independent Data Monitoring/Steering Committees for Astellas, AstraZeneca, Avalere Health, Bayer, Boehringer Ingelheim, Bristol-Myers Squibb, DAIICHI Sankyo, Innomedica, Ipsen, Macrogenics, MSD, and Novartis; is an invited speaker for AdMeTech Foundation, the Swiss Group for Clinical Cancer Research (SAKK), ASCO GU, Schweizerische Gesellschaft für Medizinische Onkologie (SGMO)/Meister ConCept GmbH, ESMO, PeerVoice, Pfizer, Silvio Grasso Consulting, EPG Health, and Intellisphere LLC; and has a patent for a research method for biomarker WO2009138392. Ken Herrmann receives consultant fees from Advanced Accelerator Applications (a Novartis company), Amgen, AstraZeneca, Bain Capital, Bayer, Boston Scientific, Convergent, Curium, Debiopharm, EcoR1, Fusion, GE HealthCare, Immedica, Isotopen Technologien München, Janssen, Merck, MSD, Molecular Partners, NVision, POINT Biopharma, Pentixapharm, Pfizer, Radiopharm Theranostics, Rhine Pharma, Siemens Healthineers, Sofie Biosciences, Telix, Theragnostics, and ymabs; receives research grants from Advanced Accelerator Applications (a Novartis company), Boston Scientific, and Janssen; and has stock or other ownership interests with AdvanCell, Aktis Oncology, Convergent, NVision, and Sofie Biosciences. Gaetano Paone has an advisory role (compensated, institutional) with Novartis, Bayer, and Boston Scientific and receives speaker honoraria from Silvio Grasso Consulting, Novartis, and Boston Scientific. Aurelius Omlin has an advisory role (compensated, institutional) with Abbvie, Accord, Advanced Accelerator Applications (AAA), Astra Zeneca, Astellas, Bayer, Janssen, Monrol, Merck, MSD, Myriad, Novartis, Pfizer, Roche (compensated, institutional) and with Novartis, Janssen, Bayer, MSD, AstraZeneca, Merck, Astellas (compensated); receives travel support from Astellas, Bayer, Janssen, and Sanofi Aventis; is on the Speakers Bureau (compensated, institutional) for Astellas, Bayer, and Janssen; and receives travel grants from Bayer. No other potential conflict of interest relevant to this article was reported.

Footnotes

  • Published online Feb. 27, 2025.

  • © 2025 by the Society of Nuclear Medicine and Molecular Imaging.

REFERENCES

  1. 1.↵
    1. Tannock IF,
    2. de Wit R,
    3. Berry WR,
    4. et al
    .; TAX 327 Investigators. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N Engl J Med. 2004;351:1502–1512.
    OpenUrlCrossRefPubMed
  2. 2.
    1. Petrylak DP,
    2. Tangen CM,
    3. Hussain MH,
    4. et al
    . Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N Engl J Med. 2004;351:1513–1520.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Turco F,
    2. Gillessen S,
    3. Cathomas R,
    4. Buttigliero C,
    5. Vogl UM
    . Treatment landscape for patients with castration-resistant prostate cancer: patient selection and unmet clinical needs. Res Rep Urol. 2022;14:339–350.
    OpenUrlCrossRefPubMed
  4. 4.↵
    1. Sternberg CN,
    2. Fizazi K,
    3. Saad F,
    4. et al
    .; PROSPER Investigators. Enzalutamide and survival in nonmetastatic, castration-resistant prostate cancer. N Engl J Med. 2020;382:2197–2206.
    OpenUrlCrossRefPubMed
  5. 5.
    1. Smith MR,
    2. Saad F,
    3. Chowdhury S,
    4. et al
    . Apalutamide and overall survival in prostate cancer. Eur Urol. 2021;79:150–158.
    OpenUrlPubMed
  6. 6.↵
    1. Fizazi K,
    2. Shore N,
    3. Tammela TL,
    4. et al
    .; ARAMIS Investigators. Nonmetastatic, castration-resistant prostate cancer and survival with darolutamide. N Engl J Med. 2020;383:1040–1049.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Gravis G,
    2. Fizazi K,
    3. Joly F,
    4. et al
    . Androgen-deprivation therapy alone or with docetaxel in non-castrate metastatic prostate cancer (GETUG-AFU 15): a randomised, open-label, phase 3 trial. Lancet Oncol. 2013;14:149–158.
    OpenUrlCrossRefPubMed
  8. 8.
    1. Sweeney CJ,
    2. Chen YH,
    3. Carducci M,
    4. et al
    . Chemohormonal therapy in metastatic hormone-sensitive prostate cancer. N Engl J Med. 2015;373:737–746.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. James ND,
    2. Sydes MR,
    3. Clarke NW,
    4. et al
    .; STAMPEDE Investigators. Addition of docetaxel, zoledronic acid, or both to first-line long-term hormone therapy in prostate cancer (STAMPEDE): survival results from an adaptive, multiarm, multistage, platform randomised controlled trial. Lancet. 2016;387:1163–1177.
    OpenUrlCrossRefPubMed
  10. 10.↵
    1. Fizazi K,
    2. Tran N,
    3. Fein L,
    4. et al
    .; LATITUDE Investigators. Abiraterone plus prednisone in metastatic, castration-sensitive prostate cancer. N Engl J Med. 2017;377:352–360.
    OpenUrlCrossRefPubMed
  11. 11.
    1. James ND,
    2. de Bono JS,
    3. Spears MR,
    4. et al
    .; STAMPEDE Investigators. Abiraterone for prostate cancer not previously treated with hormone therapy. N Engl J Med. 2017;377:338–351.
    OpenUrlCrossRefPubMed
  12. 12.
    1. Davis ID,
    2. Martin AJ,
    3. Stockler MR,
    4. et al
    .; ENZAMET Trial Investigators and the Australian and New Zealand Urogenital and Prostate Cancer Trials Group. Enzalutamide with standard first-line therapy in metastatic prostate cancer. N Engl J Med. 2019;381:121–131.
    OpenUrlCrossRefPubMed
  13. 13.
    1. Armstrong AJ,
    2. Szmulewitz RZ,
    3. Petrylak DP,
    4. et al
    . ARCHES: a randomized, phase III study of androgen deprivation therapy with enzalutamide or placebo in men with metastatic hormone-sensitive prostate cancer. J Clin Oncol. 2019;37:2974–2986.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Chi KN,
    2. Agarwal N,
    3. Bjartell A,
    4. et al
    .; TITAN Investigators. Apalutamide for metastatic, castration-sensitive prostate cancer. N Engl J Med. 2019;381:13–24.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Fizazi K,
    2. Foulon S,
    3. Carles J,
    4. et al
    .; PEACE-1 Investigators. Abiraterone plus prednisone added to androgen deprivation therapy and docetaxel in de novo metastatic castration-sensitive prostate cancer (PEACE-1): a multicentre, open-label, randomised, phase 3 study with a 2 × 2 factorial design. Lancet. 2022;399:1695–1707.
    OpenUrlCrossRefPubMed
  16. 16.↵
    1. Smith MR,
    2. Hussain M,
    3. Saad F,
    4. et al
    .; ARASENS Trial Investigators. Darolutamide and survival in metastatic, hormone-sensitive prostate cancer. N Engl J Med. 2022;386:1132–1142.
    OpenUrlCrossRefPubMed
  17. 17.↵
    1. Attard G,
    2. Murphy L,
    3. Clarke NW,
    4. et al
    .; Systemic Therapy in Advancing or Metastatic Prostate Cancer: Evaluation of Drug Efficacy (STAMPEDE) Investigators. Abiraterone acetate and prednisolone with or without enzalutamide for high-risk non-metastatic prostate cancer: a meta-analysis of primary results from two randomised controlled phase 3 trials of the STAMPEDE platform protocol. Lancet. 2022;399:447–460.
    OpenUrlCrossRefPubMed
  18. 18.↵
    1. Freedland SJ,
    2. de Almeida Luz M,
    3. De Giorgi U,
    4. et al
    . Improved outcomes with enzalutamide in biochemically recurrent prostate cancer. N Engl J Med. 2023;389:1453–1465.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Gillessen S,
    2. Turco F,
    3. Davis ID,
    4. et al
    . Management of patients with advanced prostate cancer. Report from the 2024 Advanced Prostate Cancer Consensus Conference (APCCC). Eur Urol. 2025;87:157–216.
    OpenUrlPubMed
  20. 20.↵
    1. Schaeffer EM,
    2. Srinivas S,
    3. Adra N,
    4. et al
    . NCCN Guidelines® insights: prostate cancer, version 3.2024. J Natl Compr Canc Netw. 2024;22:140–150.
    OpenUrlPubMed
  21. 21.↵
    1. Tilki D,
    2. van den Bergh RCN,
    3. Briers E,
    4. et al
    . EAU-EANM-ESTRO-ESUR-ISUP-SIOG guidelines on prostate cancer. Part II-2024 update: treatment of relapsing and metastatic prostate cancer. Eur Urol. 2024;86:164–182.
    OpenUrlPubMed
  22. 22.↵
    1. Golan S,
    2. Frumer M,
    3. Zohar Y,
    4. et al
    . Neoadjuvant 177Lu-PSMA-I&T radionuclide treatment in patients with high-risk prostate cancer before radical prostatectomy: a single-arm phase 1 trial. Eur Urol Oncol. 2023;6:151–159.
    OpenUrlPubMed
  23. 23.↵
    1. Eapen RS,
    2. Buteau JP,
    3. Jackson P,
    4. et al
    . Administering [177Lu]Lu-PSMA-617 prior to radical prostatectomy in men with high-risk localised prostate cancer (LuTectomy): a single-centre, single-arm, phase 1/2 study. Eur Urol. 2024;85:217–226.
    OpenUrlPubMed
  24. 24.↵
    1. Armstrong WR
    . PSMA PET findings in an “EMBARK-like” cohort of patients with high-risk non-metastatic hormone-sensitive prostate cancer: a single center post-hoc retrospective analysis [abstract]. J Clin Oncol. 2023;41(suppl):5091.
    OpenUrl
  25. 25.↵
    1. Mendez LC,
    2. Dhar A,
    3. Laidley D,
    4. et al
    . The use of lutetium-177 PSMA radioligand therapy with high dose rate brachytherapy for locally recurrent prostate cancer after previous definitive radiation therapy: a randomized, single-institution, phase I/II study (ROADSTER). BMC Cancer. 2023;23:362.
    OpenUrlPubMed
  26. 26.↵
    1. Turco F,
    2. Buttigliero C,
    3. Delcuratolo MD,
    4. et al
    . Hormonal agents in localized and advanced prostate cancer: current use and future perspectives. Clin Genitourin Cancer. 2024;22:102138.
    OpenUrlPubMed
  27. 27.↵
    1. Saad F,
    2. Vjaters E,
    3. Shore N,
    4. et al
    .; ARANOTE Study Investigators. Darolutamide in combination with androgen-deprivation therapy in patients with metastatic hormone-sensitive prostate cancer from the phase III ARANOTE trial. J Clin Oncol. 2024;42:4271–4281.
    OpenUrlPubMed
  28. 28.↵
    1. Turco F,
    2. Tucci M,
    3. Buttigliero C
    . Darolutamide in metastatic prostate cancer. N Engl J Med. 2022;386:2344.
    OpenUrl
  29. 29.↵
    1. Boevé LMS,
    2. Hulshof M,
    3. Vis AN,
    4. et al
    . Effect on survival of androgen deprivation therapy alone compared to androgen deprivation therapy combined with concurrent radiation therapy to the prostate in patients with primary bone metastatic prostate cancer in a prospective randomised clinical trial: data from the HORRAD trial. Eur Urol. 2019;75:410–418.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Parker CC,
    2. James ND,
    3. Brawley CD,
    4. et al
    .; Systemic Therapy for Advanced or Metastatic Prostate Cancer: Evaluation of Drug Efficacy (STAMPEDE) Investigators. Radiotherapy to the primary tumour for newly diagnosed, metastatic prostate cancer (STAMPEDE): a randomised controlled phase 3 trial. Lancet. 2018;392:2353–2366.
    OpenUrlCrossRefPubMed
  31. 31.
    1. Burdett S,
    2. Boevé LM,
    3. Ingleby FC,
    4. et al
    .; STOPCAP M1 Radiotherapy Collaborators. Prostate radiotherapy for metastatic hormone-sensitive prostate cancer: a STOPCAP systematic review and meta-analysis. Eur Urol. 2019;76:115–124.
    OpenUrlPubMed
  32. 32.↵
    1. Bossi A,
    2. Foulon S,
    3. Maldonado X,
    4. et al
    .; PEACE-1 Investigators. Efficacy and safety of prostate radiotherapy in de novo metastatic castration-sensitive prostate cancer (PEACE-1): a multicentre, open-label, randomised, phase 3 study with a 2 × 2 factorial design. Lancet. 2024;404:2065–2076.
    OpenUrlPubMed
  33. 33.↵
    1. Privé BM,
    2. Janssen MJR,
    3. van Oort IM,
    4. et al
    . Update to a randomized controlled trial of lutetium-177-PSMA in oligo-metastatic hormone-sensitive prostate cancer: the BULLSEYE trial. Trials. 2021;22:768.
    OpenUrlPubMed
  34. 34.↵
    1. Azad AA,
    2. Bressel M,
    3. Tan H,
    4. et al
    .; UpFrontPSMA Study Team. Sequential [177Lu]Lu-PSMA-617 and docetaxel versus docetaxel in patients with metastatic hormone-sensitive prostate cancer (UpFrontPSMA): a multicentre, open-label, randomised, phase 2 study. Lancet Oncol. 2024;25:1267–1276.
    OpenUrlPubMed
  35. 35.↵
    1. Sartor AO,
    2. Tagawa ST,
    3. Saad F,
    4. et al
    . PSMAddition: a phase 3 trial to compare treatment with 177Lu-PSMA-617 plus standard of care (SOC) versus SOC alone in patients with metastatic hormone-sensitive prostate cancer [abstract]. J Clin Oncol. 2022;40(suppl):TPS210.
    OpenUrl
  36. 36.↵
    1. Smith MR,
    2. Kabbinavar F,
    3. Saad F,
    4. et al
    . Natural history of rising serum prostate-specific antigen in men with castrate nonmetastatic prostate cancer. J Clin Oncol. 2005;23:2918–2925.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    1. Fendler WP,
    2. Weber M,
    3. Iravani A,
    4. et al
    . Prostate-specific membrane antigen ligand positron-emission tomography in men with nonmetastatic castration-resistant prostate cancer. Clin Cancer Res. 2019;25:7448–7454.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. Gillessen S,
    2. Turco F,
    3. Davis ID,
    4. et al
    . Management of patients with advanced prostate cancer. Report from the 2024 Advanced Prostate Cancer Consensus Conference (APCCC). Eur Urol. 2025;87:157–216.
    OpenUrlPubMed
  39. 39.↵
    1. Saad F,
    2. Clarke NW,
    3. Oya M,
    4. et al
    . Olaparib plus abiraterone versus placebo plus abiraterone in metastatic castration-resistant prostate cancer (PROpel): final prespecified overall survival results of a randomised, double-blind, phase 3 trial. Lancet Oncol. 2023;24:1094–1108.
    OpenUrlCrossRefPubMed
  40. 40.
    1. Chi KN,
    2. Rathkopf D,
    3. Smith MR,
    4. et al
    .; MAGNITUDE Principal Investigators. Niraparib and abiraterone acetate for metastatic castration-resistant prostate cancer. J Clin Oncol. 2023;41:3339–3351.
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. Fizazi K,
    2. Azad AA,
    3. Matsubara N,
    4. et al
    . First-line talazoparib with enzalutamide in HRR-deficient metastatic castration-resistant prostate cancer: the phase 3 TALAPRO-2 trial. Nat Med. 2024;30:257–264.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Gillessen S,
    2. Choudhury A,
    3. Saad F,
    4. et al
    . LBA1 A randomized multicenter open label phase III trial comparing enzalutamide vs a combination of radium-223 (Ra223) and enzalutamide in asymptomatic or mildly symptomatic patients with bone metastatic castration-resistant prostate cancer (mCRPC): first results of EORTC-GUCG 1333/PEACE-3 [abstract]. Ann Oncol. 2024;35(suppl 2):S1254.
    OpenUrl
  43. 43.↵
    1. Gillessen S,
    2. Choudhury A,
    3. Rodriguez-Vida A,
    4. et al
    . Decreased fracture rate by mandating bone protecting agents in the EORTC 1333/PEACEIII trial combining Ra223 with enzalutamide versus enzalutamide alone: an updated safety analysis [abstract]. J Clin Oncol. 2021(suppl);39:5002.
    OpenUrlCrossRef
  44. 44.↵
    1. Hussain MHA,
    2. Kocherginsky M,
    3. Agarwal N,
    4. et al
    . BRCAAway: a randomized phase 2 trial of abiraterone, olaparib, or abiraterone + olaparib in patients with metastatic castration-resistant prostate cancer (mCRPC) bearing homologous recombination-repair mutations (HRRm) [abstract]. J Clin Oncol. 2024;42(suppl):19.
    OpenUrlPubMed
  45. 45.↵
    1. Morris MJ,
    2. Castellano D,
    3. Herrmann K,
    4. et al
    .; PSMAfore Investigators. 177Lu-PSMA-617 versus a change of androgen receptor pathway inhibitor therapy for taxane-naive patients with progressive metastatic castration-resistant prostate cancer (PSMAfore): a phase 3, randomised, controlled trial. Lancet. 2024;404:1227–1239.
    OpenUrlPubMed
  46. 46.↵
    1. Sartor A,
    2. Jiang DM,
    3. Smoragiewicz M,
    4. et al
    . LBA65 Efficacy of 177Lu-PNT2002 in PSMA-positive mCRPC following progression on an androgen-receptor pathway inhibitor (ARPI) (SPLASH) [abstract]. Ann Oncol. 2024;35(suppl 2):A1254–A1255.
    OpenUrl
  47. 47.↵
    1. Fizazi K,
    2. Piulats JM,
    3. Reaume MN,
    4. et al
    .; TRITON3 Investigators. Rucaparib or physician’s choice in metastatic prostate cancer. N Engl J Med. 2023;388:719–732.
    OpenUrlCrossRefPubMed
  48. 48.↵
    1. de Bono J,
    2. Mateo J,
    3. Fizazi K,
    4. et al
    . Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med. 2020;382:2091–2102.
    OpenUrlCrossRefPubMed
  49. 49.↵
    1. Rebuzzi SE,
    2. Rescigno P,
    3. Catalano F,
    4. et al
    . Immune checkpoint inhibitors in advanced prostate cancer: current data and future perspectives. Cancers (Basel). 2022;14:1245.
  50. 50.↵
    1. Andre T,
    2. Berton D,
    3. Curigliano G,
    4. et al
    . Antitumor activity and safety of dostarlimab monotherapy in patients with mismatch repair deficient solid tumors: a nonrandomized controlled trial. JAMA. Netw Open. 2023;6:e2341165.
    OpenUrl
  51. 51.↵
    1. Marabelle A,
    2. Le DT,
    3. Ascierto PA,
    4. et al
    . Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol. 2020;38:1–10.
    OpenUrlCrossRefPubMed
  52. 52.↵
    1. Sartor O,
    2. de Bono J,
    3. Chi KN,
    4. et al
    .; VISION Investigators. Lutetium-177-PSMA-617 for metastatic castration-resistant prostate cancer. N Engl J Med. 2021;385:1091–1103.
    OpenUrlCrossRefPubMed
  53. 53.↵
    1. Hofman MS,
    2. Emmett L,
    3. Sandhu S,
    4. et al
    .; TheraP Trial Investigators and the Australian and New Zealand Urogenital and Prostate Cancer Trials Group. Overall survival with [177Lu]Lu-PSMA-617 versus cabazitaxel in metastatic castration-resistant prostate cancer (TheraP): secondary outcomes of a randomised, open-label, phase 2 trial. Lancet Oncol. 2024;25:99–107.
    OpenUrlPubMed
  54. 54.↵
    1. Hofman MS,
    2. Emmett L,
    3. Sandhu S,
    4. et al
    .; TheraP Trial Investigators and the Australian and New Zealand Urogenital and Prostate Cancer Trials Group. [177Lu]Lu-PSMA-617 versus cabazitaxel in patients with metastatic castration-resistant prostate cancer (TheraP): a randomised, open-label, phase 2 trial. Lancet. 2021;397:797–804.
    OpenUrlCrossRefPubMed
  55. 55.↵
    1. Parker C,
    2. Nilsson S,
    3. Heinrich D,
    4. et al
    .; ALSYMPCA Investigators. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med. 2013;369:213–223.
    OpenUrlCrossRefPubMed
  56. 56.↵
    1. Corn PG,
    2. Heath EI,
    3. Zurita A,
    4. et al
    . Cabazitaxel plus carboplatin for the treatment of men with metastatic castration-resistant prostate cancers: a randomised, open-label, phase 1-2 trial. Lancet Oncol. 2019;20:1432–1443.
    OpenUrlCrossRefPubMed
  57. 57.↵
    1. Saad F,
    2. Gleason DM,
    3. Murray R,
    4. et al
    .; Zoledronic Acid Prostate Cancer Study Group. A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma. J Natl Cancer Inst. 2002;94:1458–1468.
    OpenUrlCrossRefPubMed
  58. 58.↵
    1. Fizazi K,
    2. Carducci M,
    3. Smith M,
    4. et al
    . Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011;377:813–822.
    OpenUrlCrossRefPubMed
  59. 59.↵
    1. Coleman R,
    2. Hadji P,
    3. Body JJ,
    4. et al
    .; ESMO Guidelines Committee. Bone health in cancer: ESMO clinical practice guidelines. Ann Oncol. 2020;31:1650–1663.
    OpenUrlCrossRefPubMed
  • Received for publication December 4, 2024.
  • Accepted for publication January 29, 2025.
PreviousNext
Back to top

In this issue

Journal of Nuclear Medicine: 66 (5)
Journal of Nuclear Medicine
Vol. 66, Issue 5
May 1, 2025
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Complete Issue (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Journal of Nuclear Medicine.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Treatment Landscape of Prostate Cancer in the Era of PSMA Radiopharmaceutical Therapy
(Your Name) has sent you a message from Journal of Nuclear Medicine
(Your Name) thought you would like to see the Journal of Nuclear Medicine web site.
Citation Tools
Treatment Landscape of Prostate Cancer in the Era of PSMA Radiopharmaceutical Therapy
Fabio Turco, Silke Gillessen, Ken Herrmann, Gaetano Paone, Aurelius Omlin
Journal of Nuclear Medicine May 2025, 66 (5) 665-672; DOI: 10.2967/jnumed.124.267730

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Treatment Landscape of Prostate Cancer in the Era of PSMA Radiopharmaceutical Therapy
Fabio Turco, Silke Gillessen, Ken Herrmann, Gaetano Paone, Aurelius Omlin
Journal of Nuclear Medicine May 2025, 66 (5) 665-672; DOI: 10.2967/jnumed.124.267730
Twitter logo Facebook logo LinkedIn logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One
Bookmark this article

Jump to section

  • Article
    • Abstract
    • HIGH-RISK LOCALIZED DISEASE
    • HIGH-RISK BIOCHEMICAL RECURRENCE
    • MHSPC SETTING
    • GENERAL CRPC
    • NMCRPC
    • POSSIBLE SEQUENCE THERAPIES IN MCRPC
    • TREATMENT OPTIONS AFTER ADT ALONE OR ADT PLUS DOCETAXEL FOR MHSPC
    • TREATMENT OPTIONS AFTER ADT PLUS ARPI FOR MHSPC
    • TREATMENT OPTIONS AFTER ADT PLUS ARPI PLUS DOCETAXEL FOR MHSPC
    • GENERAL CONSIDERATIONS
    • CONCLUSION
    • DISCLOSURE
    • Footnotes
    • REFERENCES
  • Figures & Data
  • Supplemental
  • Info & Metrics
  • PDF

Related Articles

  • PubMed
  • Google Scholar

Cited By...

  • No citing articles found.
  • Google Scholar

More in this TOC Section

  • A Vision for Gastrin-Releasing Peptide Receptor Targeting for Imaging and Therapy: Perspective from Academia and Industry
  • Theranostics for Neuroblastoma: Making Molecular Radiotherapy Work Better
Show more The State of the Art

Similar Articles

Keywords

  • radiopharmaceuticals
  • advanced prostate cancer
  • radiopharmaceutical therapy
  • systemic therapies
  • mCRPC
  • mHSPC
SNMMI

© 2025 SNMMI

Powered by HighWire