Next Article in Journal
Radiological Assessment and Therapeutic Evaluation in Hepatocellular Carcinoma: Differentiation and Treatment Response with Japanese Guidelines
Previous Article in Journal
Evolution in the Surgical Management of Gastric Cancer Peritoneal Metastases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Status of Neoadjuvant Treatment Before Surgery in High-Risk Localized Prostate Cancer

by
Juan Gómez Rivas
1,2,3,
Luis Enrique Ortega Polledo
1,*,
Irene De La Parra Sánchez
1,
Beatriz Gutiérrez Hidalgo
1,
Javier Martín Monterrubio
1,
María Jesús Marugán Álvarez
1,
Bhaskar K. Somani
4,
Dmitry Enikeev
5,6,7,8,
Javier Puente Vázquez
9,
Noelia Sanmamed Salgado
10,
María Isabel Galante Romo
1,2 and
Jesús Moreno Sierra
1,2,3
1
Urology Department, Hospital Clínico San Carlos, 28040 Madrid, Spain
2
Health Research Institute, Hospital Clínico San Carlos, 28040 Madrid, Spain
3
Urology, Surgery Department, Universidad Complutense de Madrid, 28040 Madrid, Spain
4
Department of Urology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
5
Urology Department, Vienna Medical University, 1090 Wien, Austria
6
Institute for Urology and Reproductive Health, Sechenov University, 119435 Moscow, Russia
7
Medicine Dean and Assoc. Deans, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
8
Urology Department, Rabin Medical Center, Petach Tikva 4941492, Israel
9
Medical Oncology Department, Hospital Clínico San Carlos, 28040 Madrid, Spain
10
Radiation Oncology Department, Hospital Clínico San Carlos, 28040 Madrid, Spain
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(1), 99; https://doi.org/10.3390/cancers17010099
Submission received: 20 December 2024 / Revised: 24 December 2024 / Accepted: 25 December 2024 / Published: 31 December 2024
(This article belongs to the Section Cancer Therapy)

Simple Summary

The likelihood of a biochemical recurrence, metastatic progression and cancer-related mortality after the initial treatment in patients diagnosed with localized high-risk (HR) prostate cancer (PCa) is higher when compared with patients with low (LR) or intermediate-risk (IR) disease. The use of conventional androgen deprivation therapy (ADT) as a neoadjuvant therapy prior to radical prostatectomy (RP) has already been explored by several studies, showing a reduction in positive surgical margins and tumor volume but failing to demonstrate any survival advantages. However, novel agents such as androgen receptor-targeted agents (ARTAs) have exhibited significant survival benefits in both metastatic and non-metastatic castration-resistant PCa, as well as metastatic hormone-sensitive PCa. The significance of the pathological response following neoadjuvant hormonal therapy remains uncertain. In this narrative review, we cover the current status of neoadjuvant therapies before surgery in high-risk localized PCa.

Abstract

Localized high-risk (HR) prostate cancer (PCa) is a heterogeneous disease whose likelihood of a biochemical recurrence, metastatic progression and cancer-related mortality after initial treatment is higher when compared with patients with low (LR) or intermediate-risk (IR) disease. In the past, neoadjuvant therapy has shown an improvement in postoperative oncological variables but failed to demonstrate any survival advantages. With the promising results from novel treatments in metastatic and non-metastatic castration resistant PCa settings, new evidence has appeared in the literature in the neoadjuvant setting. Background/Objectives: To describe the current evidence for different neoadjuvant treatments before a radical prostatectomy in high-risk prostate cancer. Methods: We performed a comprehensive English literature search for original and review articles through January–August 2024, using Pubmed, Medline and ClinicalTrials.gov databases, as well as a comprehensive review of different international guidelines, searching the following terms: “neoadjuvant ADT prostate cancer”, “neoadjuvant ADT”, “prostate cancer surgery” and “neoadjuvant high-risk prostate cancer”. We included 61 papers for the final review. Results and Discussion: Neoadjuvant therapy is not recommended in daily practice by any international guideline. The National Comprehensive Cancer Network (NCCN) guidelines strongly discourage the use of ADT as a neoadjuvant therapy outside of clinical trials. ADT + ARTAs show promising data in phase-II trials, including favorable pCR, MRD, PSA relapse and salvage therapy rates. Clinical trials on chemotherapy, 177Lu-PSMA, genomic-targeted therapies and markers of response leave room for further evidence acquisition due to their encouraging results. Conclusions: Currently, no phase III data supports systemic neoadjuvant therapy before RP. Phase II studies show promising data for ADT with second-generation agents, including favorable pCR, MRD, PSA relapse and salvage therapy rates.

1. Introduction

Patients diagnosed with localized high-risk (HR) prostate cancer (PCa) face an elevated likelihood of a biochemical recurrence, metastatic progression and cancer-related mortality after initial treatment, when compared with patients with low (LR) or intermediate-risk (IR) disease [1,2]. The exploration of neoadjuvant therapies before RP for HR PCa is gaining significant momentum, as highlighted by a range of ongoing clinical trials and recent studies. These trials aim to determine whether preoperative treatment can enhance surgical outcomes, reduce tumor burdens and improve long-term survival rates for patients with localized HR PCa.
Several studies have explored the use of conventional androgen deprivation therapy (ADT) as a neoadjuvant therapy to be used prior to a radical prostatectomy (RP) [3,4,5,6,7,8,9,10,11,12,13,14,15]. These studies have shown a reduction in positive surgical margins and tumor volume but have not demonstrated any survival advantages. However, novel agents such as androgen receptor-targeted agents (ARTAs) have exhibited significant survival benefits in both metastatic and non-metastatic castration-resistant PCa, as well as in metastatic hormone-sensitive PCa [16,17,18]. The significance of the pathological response following neoadjuvant hormonal therapy remains uncertain, primarily due to the challenging nature of assessing prostate tissue after hormonal treatment [19]. Pathological evaluations are hindered by the effect of hormonal therapy on the prostate tissue. To overcome these challenges, longitudinal imaging techniques can be employed to identify early responders. These imaging methods can assess the impact of neoadjuvant hormonal therapy on the local tumor, as well as potential micrometastases.

2. Materials and Methods

In this narrative review, we cover the current status of neoadjuvant therapies before surgery in HR localized PCa. We performed a comprehensive English literature search for original and review articles through January–August 2024, using Pubmed, Medline and ClinicalTrials.gov databases, as well as a comprehensive review of The American Urological Association (AUA) guidelines, European Association of Urology (EAU) guidelines, European Society for Medical Oncology (ESMO) clinical practice guidelines and National Comprehensive Cancer Network (NCCN) guidelines. We searched for the following terms: neoadjuvant ADT prostate cancer, neoadjuvant ADT, prostate cancer surgery and neoadjuvant high-risk prostate cancer.
The combination of terms found 1314 related articles; the final number of selected papers for this manuscript was 61, including only prospective studies, systematic reviews and meta-analysis and published or ongoing RCTs (phases I, II or III) that included at least 1 arm of HR localized PCa with neoadjuvant treatment prior to RP (Figure 1).

3. Results and Discussion

3.1. Current Guideline Recommendations

Currently, there is insufficient evidence to create a consensus on neoadjuvant therapy with ADT in HR PCa. The European Association of Urology (EAU) guidelines do not recommend neoadjuvant therapy with ADT prior to RP as a standard clinical practice, based on a systematic review and meta-analysis conducted in 2021 [2,20]. The American Urological Association (AUA) does not include neoadjuvant ADT among its recommendations. However, it is mentioned as a possible option in the future as part of a multimodal treatment to intensify the management of HR patients with localized disease, although the existing evidence is insufficient [21]. The European Society for Medical Oncology (ESMO) clinical practice guidelines make no mention of neoadjuvant therapies prior to surgery [22]. In contrast, the National Comprehensive Cancer Network (NCCN) guidelines strongly discourage the use of ADT as a neoadjuvant therapy outside of clinical trials due to a lack of evidence regarding survival outcomes [23].

3.2. Trials on Neoadjuvant Treatment with ADT (1st Generation Anti-Androgens)

ADT can be achieved either with LHRH agonists, LHRH antagonists, anti-androgen treatments or combining a LHRH agonist with an anti-androgen treatment [3,4]. Since the 1990s, several trials have been conducted to study the effect of ADT prior to RP using different pharmacological schemes. The first studies demonstrated significant changes in pathological findings, such as a reduction in pathological stage and a reduction in positive surgical margins or seminal vesicle involvement. However, these studies do not report on medium or long-term oncological outcomes. Additionally, these studies included patients with LR and IR PCa, which may have underestimated the results [4,5].
During the first decade of the 21st century, a new series of trials was conducted to evaluate the duration of neoadjuvant therapies, comparing different strategies (3-month vs. 6-month and 3-month vs. 8-month). These studies also showed significant differences in pathological findings, especially in those undergoing longer ADT schemes. Nonetheless, despite some of these studies evaluating overall survival or disease-free survival (defined by clinical or biochemical progression), they did not find significant differences among the groups [5] (Table 1). Retrospective studies and non-randomized studies specifically targeting HR PCa patients offer similar results on local disease control and local disease control with a slight increase in morbidity, due to ADT-related toxicity (mainly hot flushes and gynecomastia) [5].

3.3. Trials on Neoadjuvant Chemotherapy

ADT before surgery in men with localized PCa has shown a histological benefit but no significant improvements in their biochemical progression-free survival [6,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26]. In contrast, treatment with Docetaxel has shown an increase in median overall survival in patients with mCRPC [27,28]. Combining both concepts (neoadjuvant therapy in localized PCa and chemotherapy treatment), the CALGB (Alliance) group conducted a study to assess the effect of a neoadjuvant treatment with Docetaxel prior to surgery in patients with high-risk localized PCa [29] by randomizing 788 patients with clinically localized, HR PCa into 2 groups: neoadjuvant chemohormonal therapy (CHT) (391 patients, neoadjuvant arm: Docetaxel with ADT (Leuprolide or Goserelin)) or RP alone (397 patients, surgery arm), with a median follow-up of 6.1 years. The three-year biochemical-PFS (BPFS) (0.89 vs. 0.84, CI 95% for the difference, −0.01 to 0.16), 5-year BPFS (0.81 vs. 0.74, CI 95% for the difference, −0.01 to 0.11; p = 0.11) and PCa Cancer-Specific Mortality (CSM) (HR 0.69; 95% CI, 0.32 to 1.07) showed no differences between the treatment groups. However, the overall BPFS (HR 0.69; 95% CI, 0.48 to 0.99), MFS (HR 0.70; 95% CI, 0.32 to 0.95) and OS (HR 0.61; 95% CI, 0.40 to 0.94) were all superior in the neoadjuvant arm.
Previously, several phase I and phase II studies showed that neoadjuvant chemotherapy with Docetaxel prior to RP was well tolerated and led to a reduction in clinical staging [30,31,32]. To assess whether neoadjuvant treatments with Docetaxel and ADT in patients with HR localized PCa improved outcomes compared to those treated exclusively with surgery, a multicenter, randomized phase III clinical trial was designed and conducted. The eligibility criteria were stage cT1–3a PCa (digital rectal examination), a PSA <100 ng/mL, a negative bone scan and no distant disease on abdominal–pelvic computerized tomography (CT) or magnetic resonance imaging (MRI). The definition of HR was determined using the Kattan 1998 nomogram [33], considering patients at a HR to be those with a <60% probability of being biochemical progression-free in the first 5 years. Later, all patients with a Gleason score of 8–10 were also included.
The patients were randomized into two arms: surgery alone or a neoadjuvant therapy with Docetaxel followed by RP. Patients in the neoadjuvant arm received Docetaxel every 3 weeks for 6 cycles, with prior oral dexamethasone and ADT for 18–24 weeks. The primary endpoint was established as 3-year biochemical progression-free survival. The study results demonstrated the adequate tolerability of this neoadjuvant therapy, but do not support the routine use of neoadjuvant chemotherapy in this population due to the lack of significant improvements in 3-year biochemical progression-free survival. These data might be affected by the high percentage of patients (48%) who received early salvage radiotherapy with or without ADT before reaching the primary endpoint (biochemical failure) at 3 years. However, the authors remain confident that in the long term, a more relevant impact will be observed in both primary and secondary endpoints: metastasis-free survival, cancer-specific survival and overall survival.

3.4. Trials on Neoadjuvant Treatment with ADT and ARTAs

Since the advent of ARTAs, the management of HR PCa, for both locally advanced and metastatic stages of the disease, has changed. However, there is still a lack of evidence to recommend the use of these drugs as a neoadjuvant therapy before RP [2,3,4,5,6,7,8,9,10]. Currently, the results of some completed phase I, II single-arm studies and randomized controlled trials (RCT) are published, as well as ongoing trials evaluating different strategies both for ADT and ARTAs in a neoadjuvant setting (Table 2).
Bastos et al. [34] conducted a phase II RCT with two arms, including ADT with Abiraterone vs. ADT with Abiraterone + Apalutamide, but their results regarding the oncological outcomes are yet to be published.
Taplin et al. [35,36,37], have conducted two phase II RCTs including IR and HR localized PCa. They compared Enzalutamide + Abiraterone + Prednisone + ADT (ELAP) to Enzalutamide + ADT (EL) (NCT02268175) [34], while also comparing Apalutamide + Abiraterone + Prednisone + Leuprolide (AAPL) with Abiraterone + Prednisone + Leuprolide (APL) (NCT02903368) [35,36]. In the first trial, they recruited 75 patients (50 ELAP, 25 EL), and they reported a favorable pathologic complete response (pCR) with a trend toward improved pathological outcomes with ELAP. In the second study, they recruited 118 patients from four different institutions. AAPL presented similar results in pCR and minimal residual disease (MRD) rates compared to the APL scheme. Their results showed that the addition of Apalutamide did not improve pathologic outcomes.
Fleshner et al. [38] led a multicenter, open-label, phase II RCT in patients with D’Amico HR localized PCa treated with the neoadjuvant Abiraterone + Leuprolide with or without Cabazitaxel [37]. The primary outcome was to determine the pCR or MRD rates. The secondary outcomes included surgical margins, lymph node invasion (LNI), pathologic stage, 12-month biochemical relapse-free survival (BRFS) rates and safety profile. They enrolled 70 patients (Cabazitaxel arm: 37; No Cabazitaxel arm: 33). The pCR/MRD rates were similar in both arms. In contrast, the patients with pCR/MRD had a superior 12-month BRFS (96% vs. 62%, p = 0.03). They concluded that the neoadjuvant addition of Cabazitaxel to Abiraterone + Leuprolide prior to RP did not improve the pCR nor MRD in HR localized PCa.
Schweizer et al. [39] conducted an open-label, single-arm phase II trial with HR or very high-risk (VHR) PCa treated with Abiraterone + Apalutamide + Degarelix + Leuprolide + Indomethacin for 12 weeks prior to RP [38]. The primary objective was to determine the pCR rate. The secondary objectives included the minimal residual burden (MRB) and the elucidation of molecular features of resistance. The study recruited 20 patients for its analysis, showing a 5% pCR rate and 30% MRB rate, for an overall pathologic response rate of 35%. Additionally, 90% of the patients were at a ypT3 stage at RP and 35% were N+. They concluded that indomethacin did not add significant benefit to the treatment and that their results contribute to existing literature that suggests that neoadjuvant therapies may have favorable clinical and pathological outcomes.
Lee et al. [40,41] conducted an open-label, single-site phase II trial with IR and HR non-metastatic PCa, using Apalutamide as a neoadjuvant monotherapy for 12 weeks prior to RP. They included 30 patients (20 HR patients), but they did not report pCR in this trial. They observed that biochemical non-responders had a reduced AR activity in pre-treatment biopsies and a reduced androgen response, as well as significantly upregulated innate immune-related pathways like allograft rejection, inflammatory responses and the complement cascade (p < 0.05). The results of the self-reported Health-Related Quality of Life outcomes have already been published; the overall outcomes were maintained, but fatigue and erectile dysfunction (ED) were observed [40,41].
The interim analysis for a phase II RCT on neoadjuvant Apalutamide/Abiraterone acetate with Prednisone and the feasibility of performing a nerve-sparing radical prostatectomy in men with HR PCa (NCT02949284) [42] has been published and the early results indicate that receiving neoadjuvant therapy before RP resulted in a reduction in tumor volume and the trends support an increased potency preservation without an adverse effect on positive surgical margin rates.
The ARNEO trial is a double-blind, placebo-controlled trial that evaluates Degarelix +/− Apalutamide as a neoadjuvant therapy before RP in HR PCa [43]. No statistically significant differences were observed in the BCR between the Apalutamide arm and the placebo arm. The primary study endpoint of MRD was not associated with improved BRF, but BFRS was improved in patients with organ-confined disease (ypT2) compared to ypT3-T4. Patients treated with neoadjuvant Degarelix + Apalutamide had improved metastatic disease-free survival compared to Degarelix alone and RP alone (standard of care cohort).
Finally, the PROTEUS trial (NCT03767244) [44], is a large phase III study aiming to determine if Apalutamide + ADT before and after RP is better than placebo + ADT in HR localize PCa or locally advanced PCa, comparing pCR rate and metastasis-free survival.
While these trials represent a significant step forward in the quest to improve outcomes for HR PCa, it is crucial to note that neoadjuvant approaches before prostatectomy are not yet considered standard care.
A summary of ongoing trials on neoadjuvant ADT and ARTAs are included in Table 3 and Table 4.
Table 2. Completed trials on neoadjuvant ADT and ARTAs. pCR: Pathological Complete Response, MRB: Minimal Residual Burden; MRD: Minimal Residual Disease; RCB: Residual Cancer Burden; OS: overall survival; BPFS: Biochemical progression-free survival.
Table 2. Completed trials on neoadjuvant ADT and ARTAs. pCR: Pathological Complete Response, MRB: Minimal Residual Burden; MRD: Minimal Residual Disease; RCB: Residual Cancer Burden; OS: overall survival; BPFS: Biochemical progression-free survival.
Author and RCTPhaseInclusion CriteriaOnly High-Risk Localized PCaNeoadjuvant Therapy (Drugs)Results
Bastos et al. [34]
NCT02789878
II
Randomized
Single masking
T3 (DRE)
or Gleason ≥ 8
or PSA > 20
YesGoserelin + Abiraterone
vs.
Goserelin + Abiraterone + Apalutamide
NYR
Taplin et al. [35]
NCT02268175
II
Randomized
Open-label
Intermediate Risk: Gleason 4 + 3
High-Risk: Gleason 8–10 or PSA > 20 or cT3 (MRI)
NoEnzalutamide + Abiraterone + Prednisone + Leuprolide (ELAP)
vs.
Enzalutamide + Leuprolide (EL)
pCR ELAP: 30%
EL: 16%
(p = 0.26)
Taplin et al. [36,37]
NCT02903368
II, Randomized
Open-label
Gleason ≥ 7 (4 + 3)
Or
Gleason 7 (3 + 4) and 1 of the following:
-
PSA > 20
-
≥cT3 (MRI)
YesArm 1A: Abiraterone + Apalutamide + Leuprolide + Prednisone (AAPL)
vs.
Arm 1B: Abiraterone + Prednisone + Leuprolide + (APL)
Arm 2A: Abiraterone + Apalutamide + Leuprolide + Prednisone (AAPL) 12 months
vs.
Arm 2B: observation
Part 1: pCR
1A: 22%
1B: 20%
(p = 0.4)
Part 2: BPFS:
2A: 81%
2B: 72%
HR: 0.81, 90% CI, 0.43–1.49)
Fleshner et al. [38]
NCT02543255
II
Randomized
Open-label
cT2c-cT3 (DRE+/−imaging)
or Gleason ≥ 8
or PSA > 20
YesArm A: Abiraterone + Prednisone + Leuprolide + Cabazitaxel
vs.
Arm B: Abiraterone + Prednisone + Leuprolide
pCR/MRB
Arm A: 43.2%
Arm B: 45.5%
(p = 0.85)
pCR
Arm A: 5.4%
Arm B: 9.1%
(p = 0.66)
Schweizer et al. [39]
NCT02849990
II
Single-arm
Open-label
High-risk or very high-risk (NCCN Guidelines)NoAbiraterone + Apalutamide + Degarelix + Leuprolide + IndomethacinpCR 5%
MRD 30%
PSA relapse:
14% of pCR group/46% of non-pCR group.
Lee et al. [40,41]
NCT03124433
II
Single-arm
Open-label
D’Amico Intermediate Risk and High-Risk non-metastaticNoApalutamidepCR: 0%
Median Cancer Burden Reduction: 41.7%.
Devos et al. [43]
NCT03080116
II
Randomized
Open-label
Intermediate or High-risk PCaNoArm 1: Degarelix + Apalutamide
vs.
Arm 2: Degarelix + Placebo
MRD:
Arm 1: 38%
Arm 2: 91%
CI 95%
(p = 0.002)
PTEN loss: less MRD (11% vs. 43%) (p = 0.002).
Higher RCB (1.6 vs. 0.4 cm3) (p = 0.0001)

3.5. Trials on Neoadjuvant Treatment with Lu-177

Lutetium-177-PSMA is a radioligand that selectively binds to cancer cells expressing PSMA, inducing cellular damage through the generation of free radicals [55]. There is evidence of its benefits to overall survival in patients with metastatic castration-resistant PCa [56].
Clinical trials like LUNAR [57] have explored its utility in oligorecurrent PCa. Based on the evidence from different therapies already used in other stages of the disease, such as Lu-177, the idea of utilizing them in a neoadjuvant setting for localized PCa has arisen. Golan et al. [58] published the safety profile of Lu-177 at this stage, with a single-arm feasibility study, including patients with localized HR PCa or pelvic lymph node involvement with high PSMA expression in the prostate on previously conducted 68Ga-PSMA PET-CT. The patients received LuPSMA (two or three doses) intravenously at 2-week intervals, and surgery (robot-assisted RP (RARP) with ilio-obturator lymphadenectomy (LND)) was performed 4 weeks after the last dose of LuPSMA. Of the 14 included patients, RARP with LND was performed in 13 cases, all without intraoperative incidents. The PSA decreased by 17% after two doses of LuPSMA and 34% after three doses, although the impact of this decrease is still unknown. Immediate postoperative complications did not increase, and at 3 months post-surgery, 12 patients (92%) required one pad or fewer. In other clinical trials, a higher toxicity associated with LuPSMA was reported, likely due to the higher number of doses used in patients with metastatic PCa and extensive bone involvement. Up to 10% of these patients suffered bone marrow lesions, depicting a different and non-comparable scenario regarding toxicities [56,57,58,59].
The study concluded that LuPSMA administration as a neoadjuvant treatment, followed by RARP, appears to be safe from a surgical perspective. While the oncological results are still pending, post-surgical continence does not seem to be affected by LuPSMA. The study’s limitations are evident due to the small number of patients included. However, by demonstrating the safety of LuPSMA prior to surgery, there is now space for broader clinical trials.
LuTectomy is a prospective phase I/II study of Lutetium-177-PSMA-617 as a neoadjuvant therapy prior to RP [60], for HR PCa (ISUP grade group 3–5, PSA > 20 ng/mL, ≥cT2, N1), M0 and high uptake (SUVmax > 20) on PSMA-PET-CT. It included 20 patients, who received a 5 GBq dose of 177Lu-PSMA-617 (Cohort A: 10 patients, Cohort B: 10 patients), followed six weeks later by RP + pelvic LND. The primary outcome was dosimetry (tumor radiation absorbed dose (TRAD)).
The TRAD after cycle 1 for all lesions was 35.5 Gy (Interquartile Range (IQR) 19.5–50.1), with a 19.6 Gy (IQR 11.3–48.4) delivered to the prostate. Five patients received radiation to their lymph nodes. Nine patients (45%) achieved a PSA decline of >50%. The AEs related to 177Lu-PSMA-617 were grade 1 fatigue (40%), nausea (35%), dry mouth (30%) and thrombocytopenia (20%). No grade 3/4 toxicities or Clavien 3–5 complications occurred.
The final results of the trial showed that 177Lu-PSMA-617 before RP is safe and effective as a neoadjuvant therapy, and its dose delivery appears to be high and targeted, but variable. The biochemical, imaging and pathological responses were encouraging.

3.6. Trials on Neoadjuvant Treatments Based on Baseline Genomic Alterations

There are several trials evaluating neoadjuvant treatments based on different genomic alterations. Non-hormonal treatments are being investigated as immunotherapies that can improve anti-tumor immune responses and help to control distant micrometastasis [61].
Recently, B7-H3 was explored as a target for therapy development in PCa as it is highly expressed in this type of cancer. B7-H3 comes from a family of coregulatory molecules, in which PD-L1 and PD-L2 are included. Enoblituzumab, a humanized B7-H3 targeting antibody, was tested in a single-arm study perform by Shenderov et al. [61]. A total of 32 patients with HR localized PCa were treated over 6 weeks with Enoblituzumab prior to RP. This treatment was well tolerated (12% of cases had adverse effects at a grade of 3, with no grade 4 cases) and 66% of patients achieved an undetectable PSA one year after RP.
The genomic umbrella neoadjuvant study (GUNS) is a multi-arm study assessing targeted therapies in patients with selected genomic alterations with HR PCa [62]. Initially, they received 8 weeks of treatment with LHRH agonists and Apalutamide. After that, different target therapies in different subprotocols were added. These subprotocols were as follows: Abiraterone/Prednisone for no targetable actionable aberration; presence of TMPRSS2-ERG fusion, CHD1 loss or SPOP mutations: (~50% expected prevalence in study population), Docetaxel for the loss of tumor suppressor genes—PTEN, TP53 or TB loss (~40%, bad prognosis), PARPi (Niraparib) for DNA damage response alterations (e.g., BRCA1/2, ATM, FANCONI, CDK12) in 6–8% and PD-L1 (Atezolizumab) for hypermutations, microsatellite instability (MSI), Lynch syndrome or CDK12 of less than 5%. The primary endpoint of the study was pCR.
Another ongoing study is the NePtune trial [63], a phase II single-arm study assessing the role of neoadjuvant PARP inhibition with Olaparib followed by RP in patients with HR PCa that present with BRCA 1/2 gene alterations. The treatment duration of Olaparib prior to RP is 6 months. The primary endpoints are as follows: pCR and MRD.

3.7. Molecular and Pathological Response Markers in Neoadjuvant Therapy

There is increasing interest in knowing the molecular markers that might help with predicting treatment responses, given the heterogeneity of HR PCa. While neoadjuvant therapy may show benefit in some patients treated with RP, inducing pCR or MRD, there is a growing interest in the underlying biology and molecular markers of treatment responses, which remain unclear.
Although anatomopathological response is a marker for estimating long-term survival in several tumor types, it remains unclear regarding PCa. Mckay et al. [36] performed a multicenter phase II study to assess the impact of pathological responses on recurrence rates in patients treated with intense neoadjuvant treatments prior to RP. They found that PTEN-loss and ERG + expression were associated with larger residual tumors and a higher residual cancer burden (RCB). The presence of intraductal carcinoma was found to be another marker of a higher T stage, residual tumor size or residual cancer.
In the ARNEO study [43], the investigators introduced the role of PET-PSMA as biomarker to assess responses to neoadjuvant hormonal treatment in patients treated with Degarelix with or without Apalutamide prior to RP. They concluded that PET-PSMA estimated tumor volumes, finding that the PSMA SUVmax after neoadjuvant therapy was lower in patients with minimum residual disease. Another crucial value was the PSA, showing that a Nadir after therapy of < 0.3 ng/mL was associated with a lower RCB. Like in the other studies mentioned before, a PTEN loss was associated with a higher RCB.
Tewari et al. [64] analyzed molecular features in patients treated with intense neoadjuvant therapy prior to RP (with Enzalutamide, ADT, Abiraterone and Prednisone or ADT with Apalutamide and Abiraterone), comparing patients with exceptional responses (ERs) and the ones that had non-responses (NRs). An ER was understood as complete response or with <5 mm residual tumor after RP. No differences were seen between groups in the tumor mutational burden or in the proportion of genome alterations. They found that recurrent mutations in SPOP were seen exclusively in the ER group, and that TP53 mutations, PTEN loss, PHLPP1 loss and androgen receptor deletions were exclusively in the NR group. More upregulated genes were identified in ER (136 genes) compared with the NR group (21 genes). The presence of TMPRSS2-ERG fusions were more frequent in the NR group of their cohort, and an increase in TGF-beta signaling was seen in the NR group.
These findings may help while accurately choosing a tailor-made neoadjuvant therapy. More trials will be needed to better characterize the molecular alterations of these tumors and help to predict the pathology response after a neoadjuvant treatment. An ideal marker should help us identify non-responders prior to RP in order to avoid any delay in a definitive treatment, but there is little evidence regarding the clinical impact and molecular determinants, and the impact of delaying definitive treatment in this setting.
Ongoing and future studies focusing on neoadjuvant therapies should incorporate translational endpoints. This approach will aid in identifying early resistance mechanisms and developing innovative biomarkers. Ultimately, these advancements will enable personalized neoadjuvant hormonal treatments for patients diagnosed with high-risk PCa.

4. Conclusions

Currently, no phase III data supports the use of systemic neoadjuvant therapies before RP. Phase II studies show promising data for ADT with second-generation agents, including a favorable pCR and MRD, along with PSA relapse and salvage therapy rates. Ongoing and future studies focusing on neoadjuvant therapies should incorporate translational endpoints, helping to identify early resistance mechanisms and develop innovative biomarkers. Eventually, this will enable us to tailor neoadjuvant hormonal treatments for patients diagnosed with HR PCa.

Author Contributions

Conceptualization, J.G.R.; methodology, J.G.R., L.E.O.P., I.D.L.P.S., B.G.H., J.M.M. and M.J.M.Á.; validation, J.G.R., L.E.O.P., B.K.S., D.E., J.P.V., N.S.S., M.I.G.R. and J.M.S.; formal analysis, J.G.R., L.E.O.P., J.P.V. and N.S.S.; investigation, J.G.R., L.E.O.P., I.D.L.P.S., B.G.H., J.M.M. and M.J.M.Á.; data curation, J.G.R., L.E.O.P., I.D.L.P.S., B.G.H., J.M.M. and M.J.M.Á.; writing—original draft preparation, J.G.R., L.E.O.P., I.D.L.P.S., B.G.H., J.M.M. and M.J.M.Á.; writing—review and editing, J.G.R., L.E.O.P., J.P.V. and N.S.S.; supervision, J.G.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Würnschimmel, C.; Wenzel, M.; Wang, N.; Tian, Z.; Karakiewicz, P.I.; Graefen, M.; Huland, H.; Tilki, D. Radical prostatectomy for localized prostate cancer: 20-year oncological outcomes from a German high-volume center. Urol. Oncol. Semin. Orig. Investig. 2021, 39, 830.e17–830.e26. [Google Scholar] [CrossRef] [PubMed]
  2. EAU Guidelines. Edn. Presented at the EAU Annual Congress Milan 2023; EAU Guidelines Office: Arnhem, The Netherlands, 2023; ISBN 978-94-92671-19-6. [Google Scholar]
  3. Gómez Rivas, J.; Fernandez, L.; Abad-Lopez, P.; Moreno-Sierra, J. Androgen deprivation therapy in localized prostate cancer. Current status and future trends. Actas Urol. Esp. 2023, 47, 398–407. [Google Scholar] [CrossRef] [PubMed]
  4. Ashrafi, A.N.; Yip, W.; Aron, M. Neoadjuvant Therapy in High-Risk Prostate Cancer. Indian J. Urol. 2020, 36, 251–261. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  5. Kumar, S.; Shelley, M.; Harrison, C.; Coles, B.; Wilt, T.J.; Mason, M.D. Neo-adjuvant and adjuvant hormone therapy for localised and locally advanced prostate cancer. Cochrane Database Syst. Rev. 2006, 2006, CD006019. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  6. Dalkin, B.L.; Ahmann, F.R.; Nagle, R.; Johnson, C.S. Randomized study of neoadjuvant testicular androgen ablation therapy before radical prostatectomy in men with clinically localized prostate cancer. J Urol. 1996, 155, 1357–1360. [Google Scholar] [CrossRef] [PubMed]
  7. Goldenberg, S.L.; Klotz, L.H.; Srigley, J.; Jewett, M.A.; Mador, D.; Fradet, Y.; Barkin, J.; Chin, J.; Paquin, J.M.; Bullock, M.J.; et al. Randomized, prospective, controlled study comparing radical prostatectomy alone and neoadjuvant androgen withdrawal in the treatment of localized prostate cancer. Canadian Urologic Oncology Group. J. Urol. 1996, 156, 873–877. [Google Scholar] [CrossRef] [PubMed]
  8. Labrie, F.; Cusan, L.; Gomez, J.L.; Diamond, P.; Suburu, R.; Lemay, M.; Tetu, B.; Fradet, Y.; Bélanger, A.; Candas, B. Neoadjuvant hormonal therapy: The Canadian experience. Urology 1997, 49 (Suppl. S3A), 56–64. [Google Scholar] [CrossRef] [PubMed]
  9. Fair, W.R.; Rabbani, F.; Bastar, A.; Betancourt, J. Neoadjuvant Hormone Therapy Before Radical Prostatectomy: Update on the Memorial Sloan-Kettering Cancer Center Trials. Mol Urol. 1999, 3, 253–260. [Google Scholar] [PubMed]
  10. Schulman, C.C.; Debruyne, F.M.; Forster, G.; Selvaggi, F.P.; Zlotta, A.R.; Witjes, W.P. 4-Year follow-up results of a European prospective randomized study on neoadjuvant hormonal therapy prior to radical prostatectomy in T2-3N0M0 prostate cancer. European Study Group on Neoadjuvant Treatment of Prostate Cancer. Eur. Urol. 2000, 38, 706–713. [Google Scholar] [CrossRef] [PubMed]
  11. Aus, G.; Abrahamsson, P.A.; Ahlgren, G.; Hugosson, J.; Lundberg, S.; Schain, M.; Schelin, S.; Pedersen, K. Three-month neoadjuvant hormonal therapy before radical prostatectomy: A 7-year follow-up of a randomized controlled trial. BJU Int. 2002, 90, 561–566. [Google Scholar] [CrossRef] [PubMed]
  12. Soloway, M.S.; Pareek, K.; Sharifi, R.; Wajsman, Z.; McLeod, D.; Wood, D.P., Jr.; Puras-Baez, A. Lupron Depot Neoadjuvant Prostate Cancer Study Group. Neoadjuvant androgen ablation before radical prostatectomy in cT2bNxMo prostate cancer: 5-year results. J. Urol. 2002, 167, 112–116. [Google Scholar] [CrossRef] [PubMed]
  13. Selli, C.; Montironi, R.; Bono, A.; Pagano, F.; Zattoni, F.; Manganelli, A.; Selvaggi, F.P.; Comeri, G.; Fiaccavento, G.; Guazzieri, S.; et al. Effects of complete androgen blockade for 12 and 24 weeks on the pathological stage and resection margin status of prostate cancer. J. Clin. Pathol. 2002, 55, 508–513. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  14. Klotz, L.H.; Goldenberg, S.L.; Jewett, M.A.; Fradet, Y.; Nam, R.; Barkin, J.; Chin, J.; Chatterjee, S.; Canadian Uro-Oncology Group. Long-term followup of a randomized trial of 0 versus 3 months of neoadjuvant androgen ablation before radical prostatectomy. J. Urol. 2003, 170, 791–794. [Google Scholar] [CrossRef] [PubMed]
  15. Prezioso, D.; Lotti, T.; Polito, M.; Montironi, R. Neoadjuvant hormone treatment with leuprolide acetate depot 3.75 mg and cyproterone acetate, before radical prostatectomy: A randomized study. Urol. Int. 2004, 72, 189–195. [Google Scholar] [CrossRef] [PubMed]
  16. Fizazi, K.; Tran, N.; Fein, L.; Matsubara, N.; Rodriguez-Antolin, A.; Alekseev, B.Y.; Özgüroğlu, M.; Ye, D.; Feyerabend, S.; Protheroe, A.; et al. Abiraterone plus Prednisone in Metastatic, Castration-Sensitive Prostate Cancer. N. Engl. J. Med. 2017, 377, 352–360. [Google Scholar] [CrossRef] [PubMed]
  17. Chi, K.N.; Agarwal, N.; Bjartell, A.; Chung, B.H.; Pereira de Santana Gomes, A.J.; Given, R.; Juárez Soto, Á.; Merseburger, A.S.; Özgüroğlu, M.; Uemura, H.; et al. Apalutamide for Metastatic, Castration-Sensitive Prostate Cancer. N. Engl. J. Med. 2019, 381, 13–24. [Google Scholar] [CrossRef] [PubMed]
  18. Davis, I.D.; Martin, A.J.; Stockler, M.R.; Begbie, S.; Chi, K.N.; Chowdhury, S.; Coskinas, X.; Frydenberg, M.; Hague, W.E.; Horvath, L.G.; 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. [Google Scholar] [CrossRef] [PubMed]
  19. Gleave, M.E.; Goldenberg, S.L.; Chin, J.L.; Warner, J.; Saad, F.; Klotz, L.H.; Jewett, M.; Kassabian, V.; Chetner, M.; Dupont, C.; et al. Randomized comparative study of 3 versus 8-month neoadjuvant hormonal therapy before radical prostatectomy: Biochemical and pathological effects. J. Urol. 2001, 166, 500–506; discussion 506–507. [Google Scholar] [CrossRef] [PubMed]
  20. Liu, W.; Yao, Y.; Liu, X.; Liu, Y.; Zhang, G.M. Neoadjuvant hormone therapy for patients with high-risk prostate cancer: A systematic review and meta-analysis. Asian J. Androl. 2021, 23, 429–436. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  21. Eastham, J.A.; Auffenberg, G.B.; Barocas, D.A.; Chou, R.; Crispino, T.; Davis, J.W.; Eggener, S.; Horwitz, E.M.; Kane, C.J.; Kirkby, E.; et al. Clinically Localized Prostate Cancer: AUA/ASTRO Guideline, Part I: Introduction, Risk Assessment, Staging, and Risk-Based Management. J. Urol. 2022, 208, 10–18. [Google Scholar] [CrossRef] [PubMed]
  22. Parker, C.; Castro, E.; Fizazi, K.; Heidenreich, A.; Ost, P.; Procopio, G.; Tombal, B.; Gillessen, S.; ESMO Guidelines Committee. Prostate cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2020, 31, 1119–1134. [Google Scholar] [CrossRef] [PubMed]
  23. Schaeffer, E.M.; Srinivas, S.; Adra, N.; An, Y.; Barocas, D.; Bitting, R.; Bryce, A.; Chapin, B.; Cheng, H.H.; D’Amico, A.V.; et al. Prostate Cancer, Version 4.2023, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Canc. Netw. 2023, 21, 1067–1096. [Google Scholar] [CrossRef] [PubMed]
  24. Yee, D.S.; Lowrance, W.T.; Eastham, J.A.; Maschino, A.C.; Cronin, A.M.; Rabbani, F. Long-term follow-up of 3-month neoadjuvant hormone therapy before radical prostatectomy in a randomized trial. BJU Int. 2010, 105, 185–190. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  25. Klotz, L.H.; Goldenberg, S.L.; Jewett, M.; Barkin, J.; Chetner, M.; Fradet, Y.; Chin, J.; Laplante, S. CUOG randomized trial of neoadjuvant androgen ablation before radical prostatectomy: 36-month post-treatment PSA results. Canadian Urologic Oncology Group. Urology 1999, 53, 757–763. [Google Scholar] [CrossRef] [PubMed]
  26. Witjes, W.P.; Schulman, C.C.; Debruyne, F.M. Preliminary results of a prospective randomized study comparing radical prostatectomy versus radical prostatectomy associated with neoadjuvant hormonal combination therapy in T2-3 N0 M0 prostatic carcinoma. The European Study Group on Neoadjuvant Treatment of Prostate Cancer. Urology 1997, 49 (Suppl. 3A), 65–69. [Google Scholar] [CrossRef] [PubMed]
  27. Petrylak, D.P.; Tangen, C.M.; Hussain, M.H.; Lara, P.N., Jr.; Jones, J.A.; Taplin, M.E.; Burch, P.A.; Berry, D.; Moinpour, C.; Kohli, M.; et al. Docetaxel and estramustine compared with mitoxantrone and prednisone for advanced refractory prostate cancer. N. Engl. J. Med. 2004, 351, 1513–1520. [Google Scholar] [CrossRef] [PubMed]
  28. Tannock, I.F.; de Wit, R.; Berry, W.R.; Horti, J.; Pluzanska, A.; Chi, K.N.; Oudard, S.; Théodore, C.; James, N.D.; Turesson, I.; et al. Docetaxel plus prednisone or mitoxantrone plus prednisone for advanced prostate cancer. N. Engl. J. Med. 2004, 351, 1502–1512. [Google Scholar] [CrossRef] [PubMed]
  29. Eastham, J.A.; Heller, G.; Halabi, S.; Monk, J.P., 3rd; Beltran, H.; Gleave, M.; Evans, C.P.; Clinton, S.K.; Szmulewitz, R.Z.; Coleman, J.; et al. Cancer and Leukemia Group B 90203 (Alliance): Radical Prostatectomy with or Without Neoadjuvant Chemohormonal Therapy in Localized, High-Risk Prostate Cancer. J. Clin. Oncol. 2020, 38, 3042–3050. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  30. Oh, W.K.; Hagmann, E.; Manola, J.; George, D.J.; Gilligan, T.D.; Jacobson, J.O.; Smith, M.R.; Kaufman, D.S.; Kantoff, P.W. A phase I study of estramustine, weekly docetaxel, and carboplatin chemotherapy in patients with hormone-refractory prostate cancer. Clin. Cancer Res. 2005, 11, 284–289. [Google Scholar] [CrossRef] [PubMed]
  31. Hussain, M.; Smith, D.C.; El-Rayes, B.F.; Du, W.; Vaishampayan, U.; Fontana, J.; Sakr, W.; Wood, D. Neoadjuvant docetaxel and estramustine chemotherapy in high-risk/locallyadvanced prostate cancer. Urology 2003, 61, 774–780. [Google Scholar] [CrossRef] [PubMed]
  32. Dreicer, R.; Magi-Galluzzi, C.; Zhou, M.; Rothaermel, J.; Reuther, A.; Ulchaker, J.; Zippe, C.; Fergany, A.; Klein, E.A. Phase II trial of neoadjuvant docetaxel before radical prostatectomy for locally advanced prostate cancer. Urology 2004, 63, 1138–1142. [Google Scholar] [CrossRef] [PubMed]
  33. Kattan, M.W.; Eastham, J.A.; Stapleton, A.M.; Wheeler, T.M.; Scardino, P.T. A preoperative nomogram for disease recurrence following radical prostatectomy for prostate cancer. J. Natl. Cancer Inst. 1998, 90, 766–771. [Google Scholar] [CrossRef] [PubMed]
  34. Bastos, D. Neoadjuvant Androgen Deprivation Therapy Plus Abiraterone With or Without Apalutamide for High-Risk Prostate Cancer. Available online: https://clinicaltrials.gov/study/NCT02789878 (accessed on 20 August 2024).
  35. McKay, R.R.; Ye, H.; Xie, W.; Lis, R.; Calagua, C.; Zhang, Z.; Trinh, Q.D.; Chang, S.L.; Harshman, L.C.; Ross, A.E.; et al. Evaluation of Intense Androgen Deprivation Before Prostatectomy: A Randomized Phase II Trial of Enzalutamide and Leuprolide With or Without Abiraterone. J. Clin. Oncol. 2019, 37, 923–931. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  36. McKay, R.R.; Xie, W.; Ye, H.; Fennessy, F.M.; Zhang, Z.; Lis, R.; Calagua, C.; Rathkopf, D.; Laudone, V.P.; Bubley, G.J.; et al. Results of a Randomized Phase II Trial of Intense Androgen Deprivation Therapy prior to Radical Prostatectomy in Men with High-Risk Localized Prostate Cancer. J. Urol. 2021, 206, 80–87. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  37. McKay, R.R.; Xie, W.; Yang, X.; Acosta, A.; Rathkopf, D.; Laudone, V.P.; Bubley, G.J.; Einstein, D.J.; Chang, P.; Wagner, A.A.; et al. Postradical prostatectomy prostate-specific antigen outcomes after 6 versus 18 months of perioperative androgen-deprivation therapy in men with localized, unfavorable intermediate-risk or high-risk prostate cancer: Results of part 2 of a randomized phase 2 trial. Cancer 2024, 130, 1629–1641. [Google Scholar] [CrossRef] [PubMed]
  38. Fleshner, N.E.; Sayyid, R.K.; Hansen, A.R.; Chin, J.L.K.; Fernandes, R.; Winquist, E.; van der Kwast, T.; Sweet, J.; Lajkosz, K.; Kenk, M.; et al. Neoadjuvant Cabazitaxel plus Abiraterone/Leuprolide Acetate in Patients with High-Risk Prostate Cancer: ACDC-RP Phase II Trial. Clin. Cancer Res. 2023, 29, 3867–3874. [Google Scholar] [CrossRef] [PubMed]
  39. Graham, L.S.; True, L.D.; Gulati, R.; Schade, G.R.; Wright, J.; Grivas, P.; Yezefski, T.; Nega, K.; Alexander, K.; Hou, W.M.; et al. Targeting backdoor androgen synthesis through AKR1C3 inhibition: A presurgical hormonal ablative neoadjuvant trial in high-risk localized prostate cancer. Prostate 2021, 81, 418–426. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  40. Lee, L.S.; Sim, A.Y.L.; Ong, C.W.; Yang, X.; Ng, C.C.Y.; Liu, W.; Rajasegaran, V.; Lim, A.M.S.; Aslim, E.J.; Ngo, N.T.; et al. NEAR trial: A single-arm phase II trial of neoadjuvant apalutamide monotherapy and radical prostatectomy in intermediate- and high-risk prostate cancer. Prostate Cancer Prostatic Dis. 2022, 25, 741–748, Erratum in Prostate Cancer Prostatic Dis. 2022, 25, 803. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  41. Yang, X.; Allen, J.C.; Aslim, E.J.; Tay, K.J.; Yuen, S.P.J.; Kanesvaran, R.; Chua, M.L.K.; Chong, T.W.; Ho, S.S.H.; Lee, L.S. Patient-reported outcomes of a phase II neoadjuvant apalutamide (ARN-509) and radical prostatectomy in treatment of intermediate- to high-risk prostate cancer (NEAR) trial. Int. J. Urol. 2022, 29, 1322–1330. [Google Scholar] [CrossRef] [PubMed]
  42. Ghodoussipour, S. Androgen Receptor Antagonist ARN-509 with or Without Abiraterone Acetate, Gonadotropin-Releasing Hormone Analog, and Prednisone in Treating Patients with High-Risk Prostate Cancer Undergoing Surgery. Available online: https://clinicaltrials.gov/study/NCT02949284?term=NCT02949284&rank=1 (accessed on 20 August 2024).
  43. Devos, G.; Tosco, L.; Baldewijns, M.; Gevaert, T.; Goffin, K.; Petit, V.; Mai, C.; Laenen, A.; Raskin, Y.; Van Haute, C.; et al. ARNEO: A Randomized Phase II Trial of Neoadjuvant Degarelix with or Without Apalutamide Prior to Radical Prostatectomy for High-risk Prostate Cancer. Eur. Urol. 2023, 83, 508–518. [Google Scholar] [CrossRef] [PubMed]
  44. Janssen Research & Development, LLC. A Study of Apalutamide in Participants with High-Risk, Localized or Locally Advanced Prostate Cancer Who Are Candidates for Radical Prostatectomy (PROTEUS). Available online: https://clinicaltrials.gov/study/NCT03767244 (accessed on 20 August 2024).
  45. Szmulewitz, R. A Study of Enzalutamide Plus the Glucocorticoid Receptor Antagonist Relacorilant Versus Placebo for Patients with High-Risk Localized Prostate Cancer. Available online: https://clinicaltrials.gov/study/NCT05726292 (accessed on 20 August 2024).
  46. Ravi, P. A Phase 1/2 Study of Darolutamide and Abemaciclib in High-Risk Prostate Cancer. Available online: https://clinicaltrials.gov/study/NCT05617885 (accessed on 20 August 2024).
  47. Reiter, R. An Open-label, Neoadjuvant Phase 2 Study Comparing the Effects of AR Inhibition with and Without SRC or MEK Inhibition on the Development of EMT in Prostate Cancer. Available online: https://clinicaltrials.gov/study/NCT01990196 (accessed on 20 August 2024).
  48. Zeng, H. Neoadjuvant Therapy of Abiraterone Plus ADT for Intraductal Carcinoma of the Prostate. Available online: https://clinicaltrials.gov/study/NCT04736108 (accessed on 20 August 2024).
  49. Zhang, Y. An Evaluation of the Efficacy and Safety of Apalutamide as Neoadjuvant Endocrine Therapy in High-Risk and Oligometastatic Prostate Cancer. Available online: https://clinicaltrials.gov/study/NCT04997252 (accessed on 20 August 2024).
  50. Hongqian, G. Neoadjuvant Therapy of Abiraterone Plus ADT for High Risk Prostate Cancer. Available online: https://clinicaltrials.gov/study/NCT04356430 (accessed on 20 August 2024).
  51. Hongqian, G. Efficacy and Safety of Darolutamide Combined with ADT in High-Risk/Very High-Risk Localized Prostate Cancer. Available online: https://clinicaltrials.gov/study/NCT05249712 (accessed on 20 August 2024).
  52. Hongqian, G. A Prospective, Multi-Arm, Multi-Center Clinical Trial on Neoadjuvant Intense Endocrine Therapy for High Risk and Locally Advanced Prostate Cancer. Available online: https://clinicaltrials.gov/study/NCT05406999 (accessed on 20 August 2024).
  53. Kopp, R.P. Precision Neoadjuvant Therapy for High Risk Localized Prostate Cancer with PTEN Loss. Available online: https://clinicaltrials.gov/study/NCT05593497 (accessed on 20 August 2024).
  54. Zhu, Y. Fluzoparib and Abiraterone in the preSurgery Treatment of Prostate Cancer: FAST Trial. Available online: https://clinicaltrials.gov/study/NCT05223582 (accessed on 20 August 2024).
  55. Yong, K.J.; Milenic, D.E.; Baidoo, K.E.; Brechbiel, M.W. Mechanisms of Cell Killing Response from Low Linear Energy Transfer (LET) Radiation Originating from 177Lu Radioimmunotherapy Targeting Disseminated Intraperitoneal Tumor Xenografts. Int. J. Mol. Sci. 2016, 17, 736. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  56. Sartor, O.; de Bono, J.; Chi, K.N.; Fizazi, K.; Herrmann, K.; Rahbar, K.; Tagawa, S.T.; Nordquist, L.T.; Vaishampayan, N.; El-Haddad, G.; et al. Lutetium-177-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer. N. Engl. J. Med. 2021, 385, 1091–1103. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Ma, T.M.; Czernin, J.; Felix, C.; Alano, R.; Wilhalme, H.; Valle, L.; Steinberg, M.L.; Dahlbom, M.; Reiter, R.E.; Rettig, M.B.; et al. LUNAR: A randomized Phase 2 study of 177 Lutetium-PSMA Neoadjuvant to Ablative Radiotherapy for Oligorecurrent Prostate Cancer (clinical trial protocol). BJU Int. 2023, 132, 65–74. [Google Scholar] [CrossRef] [PubMed]
  58. Golan, S.; Frumer, M.; Zohar, Y.; Rosenbaum, E.; Yakimov, M.; Kedar, D.; Margel, D.; Baniel, J.; Steinmetz, A.P.; Groshar, D.; 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. [Google Scholar] [CrossRef] [PubMed]
  59. Hofman, M.S.; Emmett, L.; Sandhu, S.; Iravani, A.; Joshua, A.M.; Goh, J.C.; Pattison, D.A.; Tan, T.H.; Kirkwood, I.D.; Ng, S.; et al. [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. [Google Scholar] [CrossRef] [PubMed]
  60. Eapen, R.S.; Buteau, J.P.; Jackson, P.; Mitchell, C.; Oon, S.F.; Alghazo, O.; McIntosh, L.; Dhiantravan, N.; Scalzo, M.J.; O’Brien, J.; 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. [Google Scholar] [CrossRef] [PubMed]
  61. Shenderov, E.; De Marzo, A.M.; Lotan, T.L.; Wang, H.; Chan, S.; Lim, S.J.; Ji, H.; Allaf, M.E.; Chapman, C.; Moore, P.A.; et al. Neoadjuvant enoblituzumab in localized prostate cancer: A single-arm, phase 2 trial. Nat. Med. 2023, 29, 888–897. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. Gleave, M.E. The Genomic Umbrella Neoadjuvant Study (GUNS). Vancouver Prostate Centre, Vancouver, Canada. Available online: https://clinicaltrials.gov/study/NCT04812366 (accessed on 20 August 2024).
  63. McKay, R. Study of Neoadyuvant PARP Inhibition Followed by Radical Prostatectomy in Patients with Unfavorable Intermediate-Risk or High-Risk Prostate Cancer with BRCA ½ Gene Alterations (NePtune). Available online: https://clinicaltrials.ucsd.edu/trial/NCT05498272 (accessed on 20 August 2024).
  64. Tewari, A.K.; Cheung, A.T.M.; Crowdis, J.; Conway, J.R.; Camp, S.Y.; Wankowicz, S.A.; Livitz, D.G.; Park, J.; Lis, R.T.; Bosma-Moody, A.; et al. Molecular features of exceptional response to neoadjuvant anti-androgen therapy in high-risk localized prostate cancer. Cell Rep. 2021, 36, 109665. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Screening and inclusion diagram. HR PCa: high-risk prostate cancer.
Figure 1. Screening and inclusion diagram. HR PCa: high-risk prostate cancer.
Cancers 17 00099 g001
Table 1. Trials on neoadjuvant ADT (1st generation anti-androgens). OS: overall survival; PFS: progression-free survival; NR: not reported.
Table 1. Trials on neoadjuvant ADT (1st generation anti-androgens). OS: overall survival; PFS: progression-free survival; NR: not reported.
AuthornClinical StageNeoadjuvant Therapy (NADT)
(Months)
Positive Margins (%)
NADT vs. Only Surgery
Seminal Invasion (%)
NADT vs. Only Surgery
pN+ (%)
NADT vs. Only Surgery
OS (%)
NADT vs. Only Surgery
PFS (%)
NADT vs. Only Surgery
Follow Up (Years)
Dalkin et al. [6]
1996
56T1c–T2bGoserelin (3)18% vs 14%--NRNRNR
Goldenberg et al. [7]
1996
213T1b–T2cCyproterone (3)28% vs. 65%28% vs. 14%7% vs. 3%NRNRNR
Labrie et al. [8]
1997
161T2–T3Leuprolide + Flutamide (3)7.8% vs. 33.8%NRNRNRNRNR
Fair et al. [9]
1999
140T1–T2Goserelin + Flutamide
(3)
19% vs. 37%NRNRNRNR2.9
Schulman et al. [10]
2000
402T2–T3Goserelin + Flutamide (3)13% vs. 37%NRNR96% vs. 96% 74% vs. 67%4
Aus et al. [11]
2002
126T1b–T3aTriptorelin + Cyproterone (3)23.6% vs. 45.5%NRNR83% vs. 86%49.8% vs. 51.5%6.85
Soloway et al. [12]
2002
303T2bLeuprolide + Flutamide (3)18% vs. 48%15% vs. 22%6% vs. 6%NR64.8% vs. 67.6%5
Selli et al. [13]
2002
431T2–T3Goserelin + Bicalutamide
(3 or 6 months)
28% (3 months)/23% (6 months) vs. 53%11% (3 months)/11% (6 months) vs. 11% 8% (3 months)/4% (6 months) vs. 12%NRNRNR
Klotz et al. [14]
2003
213T1b–T2Cyproterone (3)28% vs. 65%NRNR93% vs. 95%60.2% vs. 68.2%6
Prezioso et al. [15]
2004
183T1a–T2bLeuprolide + Cyproterone (3)39% vs. 60%NR3% vs. 11%NRNRNR
Yee et al. [24]
2010
148T1b–T3Goserelin + Flutamide (3)19% vs. 38%4% vs. 6%1% vs. 3%86% vs. 92%80% vs. 78%8
Table 3. Ongoing randomized trials on neoadjuvant ADT and ARTAs. ADT: Androgen Deprivation Therapy; ARN-509 (Androgen Receptor Antagonist ARN-509).
Table 3. Ongoing randomized trials on neoadjuvant ADT and ARTAs. ADT: Androgen Deprivation Therapy; ARN-509 (Androgen Receptor Antagonist ARN-509).
Author and RCTPhaseInclusion CriteriaOnly High-Risk Localized PCaNeoadjuvant Therapy (Drugs)Status
Ghodoussipour et al. [42]
NCT02949284
II
Randomized
Open-label
≤T3 and Gleason ≥ 8 (4 + 3) and PSA >20 and >1 coreYesARN-509
vs.
Abiraterone + Prednisone + ARN-509 + GnRH Analogue
vs.
None
Recruiting
Janssen Research & Development, LLC [44]
NCT03767244
III
Randomized
Double blind
High-risk localized or locally advancedYesApalutamide + ADT
vs.
Placebo + ADT
Active, not recruiting
Szmulewitz et al. [45]
NCT05726292
II
Randomized
Double-blind
≥cT3 or ISUP ≥ 4 or PSA >20YesEnzalutamide + Relacorilant + ADT
vs.
Placebo + ADT
Recruiting
Ravi et al. [46]
NCT05617885
II Randomized
Open-label
Gleason ≥ 8
Or
Gleason 7 (4 + 3) and 1 of the following:
-
PSA > 20
-
≥cT3
-
EPE on
-
biopsy
YesAbemaciclib + Darolutamide + ADT
vs.
Darolutamide + ADT
Active, not recruiting
Reiter et al. [47]
NCT01990196
II
Randomized
Open-label
≥cT3a or Gleason ≥ 7 (4 + 3) or any Gleason 5 or PSA > 20YesDegarelix + Enzalutamide
vs.
Degarelix + Enzalutamide + MEK-inhibition (Trametinib)
vs.
Degarelix + Enzalutamide + SRC-inhibition (Dasatinib)
Active, not recruiting
Table 4. Ongoing single-arm and non-randomized trials on neoadjuvant ADT and ARTAs.
Table 4. Ongoing single-arm and non-randomized trials on neoadjuvant ADT and ARTAs.
Author and RCTPhaseInclusion CriteriaOnly High-Risk Localized PCaNeoadjuvant Therapy (Drugs)Status
Zeng et al. [48]
NCT04736108
II
Single-arm
Non-randomized
Intraductal carcinoma +
≥cT3a
or Gleason ≥ 8
or PSA > 20
YesAbiraterone + Goserelin + PrednisoneNot yet recruiting
Zhang et al. [49]
NCT04997252
II
Single-arm
Non-randomized
High-risk localized PCa or Oligometastatic PCaNoApalutamide + LHRH agonistRecruiting
Hongqian et al. [50]
NCT04356430
II
Single-arm
Non-randomized
≥cT3 (PSMA PET/CT or mpMRI)
or Gleason ≥ 8
or PSA > 20
YesAbiraterone + Goserelin + PrednisoneUnknown
Hongqian et al. [51]
NCT05249712
II
Single-arm
Non-randomized
≥cT3 (PSMA PET/CT or mpMRI)
or Gleason ≥ 8
or PSA > 20 or N1
NoDarolutamide + ADTRecruiting
Hongqian et al. [52]
NCT05406999
II
Non-randomized
Open-label
≥cT3 (PSMA PET/CT or mpMRI)
or Gleason ≥ 8
or PSA > 20 or N1
NoADT alone
vs. ADT + Abiraterone
vs. ADT + Enzalutamide
vs. ADT + Apalutamide
vs. ADT + Darolutamide
vs. ADT + Rizvilutamide
vs. ADT + PARP inhibitor + Abiraterone
vs. ADT + PARP inhibitor
Recruiting
Kopp et al. [53]
NCT05593497
II
Single-arm
Open-label
Gleason > 7 (>ISUP-3) or
PSA > 20 (<T3 or Gleason < 7 must have 5-year PF probability ≤50% estimated by MSKCC pre-radical prostatectomy nomogram)
+ ≤10% PTEN staining
YesLeuprolide + Abiraterone + CapivasertibRecruiting
Zhu et al. [54]
NCT05223582
II
Single-arm
Open-label
High-risk or very high-risk (NCCN Guidelines)NoAbiraterone + Fluzoparib + Prednisone + ADTActive, not recruiting
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gómez Rivas, J.; Ortega Polledo, L.E.; De La Parra Sánchez, I.; Gutiérrez Hidalgo, B.; Martín Monterrubio, J.; Marugán Álvarez, M.J.; Somani, B.K.; Enikeev, D.; Puente Vázquez, J.; Sanmamed Salgado, N.; et al. Current Status of Neoadjuvant Treatment Before Surgery in High-Risk Localized Prostate Cancer. Cancers 2025, 17, 99. https://doi.org/10.3390/cancers17010099

AMA Style

Gómez Rivas J, Ortega Polledo LE, De La Parra Sánchez I, Gutiérrez Hidalgo B, Martín Monterrubio J, Marugán Álvarez MJ, Somani BK, Enikeev D, Puente Vázquez J, Sanmamed Salgado N, et al. Current Status of Neoadjuvant Treatment Before Surgery in High-Risk Localized Prostate Cancer. Cancers. 2025; 17(1):99. https://doi.org/10.3390/cancers17010099

Chicago/Turabian Style

Gómez Rivas, Juan, Luis Enrique Ortega Polledo, Irene De La Parra Sánchez, Beatriz Gutiérrez Hidalgo, Javier Martín Monterrubio, María Jesús Marugán Álvarez, Bhaskar K. Somani, Dmitry Enikeev, Javier Puente Vázquez, Noelia Sanmamed Salgado, and et al. 2025. "Current Status of Neoadjuvant Treatment Before Surgery in High-Risk Localized Prostate Cancer" Cancers 17, no. 1: 99. https://doi.org/10.3390/cancers17010099

APA Style

Gómez Rivas, J., Ortega Polledo, L. E., De La Parra Sánchez, I., Gutiérrez Hidalgo, B., Martín Monterrubio, J., Marugán Álvarez, M. J., Somani, B. K., Enikeev, D., Puente Vázquez, J., Sanmamed Salgado, N., Galante Romo, M. I., & Moreno Sierra, J. (2025). Current Status of Neoadjuvant Treatment Before Surgery in High-Risk Localized Prostate Cancer. Cancers, 17(1), 99. https://doi.org/10.3390/cancers17010099

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop