Guidelines

Prostate Cancer

6. TREATMENT

This chapter reviews the available treatment modalities, followed by separate sections addressing treatment for the various disease stages.

6.1. Estimating life expectancy and health status

6.1.1. Introduction

Evaluation of life expectancy and health status is important in clinical decision-making for early detection, diagnosis, and treatment of PCa. Prostate cancer is common in older men (median age 68) and diagnoses in men > 65 will result in a 70% increase in annual diagnosis by 2030 in Europe and the USA [433,434].

Active treatment mostly benefits patients with intermediate- or high-risk PCa and longest expected survival. In localised disease, over ten years life expectancy is considered mandatory for any benefit from local treatment and an improvement in CSS may take longer to become apparent. Older age and worse baseline health status have been associated with a smaller benefit in PCSM and life expectancy of surgery vs. AS [435]. Although in a RCT the benefit of surgery with respect to death from PCa was largest in men < 65 years of age (RR: 0.45), RP was associated with a reduced risk of metastases and use of androgen deprivation therapy (ADT) also among older men (RR: 0.68 and 0.60, respectively) [436]. External beam RT shows similar cancer control regardless of age, assuming a dose of > 72 Gy when using intensity-modulated or image-guided RT [437].

Older men have a higher incidence of PCa and may be under-treated despite the high overall mortality rates [438,439]. Of all PCa-related deaths 71% occur in men aged > 75 years [440], probably due to the higher incidence of advanced disease and death from PCa despite higher death rates from competing causes [441-443]. In the USA, only 41% of patients aged > 75 years with intermediate- and high-risk disease received curative treatment compared to 88% aged 65–74 [444].

6.1.2. Life expectancy

Life expectancy tables for European men are available online: https://ec.europa.eu/eurostat/. Survival may be variable and therefore estimates of survival must be individualised. Gait speed is a good single predictive method of life expectancy (from a standing start, at usual pace, generally over 6 meters). For men at age 75,
ten-year survival ranged from 19% < 0.4 m/s to 87%, for ≥ 1.4 m/s [445].

Figure 6.1: Predicted Median Life Expectancy by Age and Gait Speed for males* [445]*Figure reproduced with permission of the publisher, from Studenski S, et al. JAMA 2011 305(1)50.

6.1.3. Health status screening

Heterogeneity in performance increases with advancing age, so it is important to use measures other than just age or performance status (PS) when considering treatment options. The International SIOG PCa Working Group recommends that treatment for adults over 70 years of age should be based on a systematic evaluation of health status using the G8 (Geriatric 8) screening tool (see Table 5.7) [136]. This tool helps to discriminate between those who are fit and those with frailty, a syndrome of reduced ability to respond to stressors. Patients with frailty have a higher risk of mortality and negative side effects of cancer treatment [446]. Healthy patients with a G8 score > 14 or vulnerable patients with reversible impairment after resolution of their geriatric problems should receive the same treatment as younger patients. Frail patients with irreversible impairment should receive adapted treatment. Patients who are too ill should receive only palliative treatment (see Figure 5.3) [136]. Patients with a G8 score ≤ 14 should undergo a comprehensive geriatric assessment (CGA) as this score is associated with three-year mortality. A CGA is a multi-domain assessment that includes co-morbidity, nutritional status, cognitive and physical function, and social supports to determine if impairments are reversible [447]. A SR of the effect of geriatric evaluation for older cancer patients showed improved treatment tolerance and completion [448].

The Clinical Frailty Scale (CFS) is another screening tool for frailty (see Figure 5.4) [449]. Although not frequently used in the cancer setting, it is considered to be a common language for expressing degree of frailty. The scale runs from one to nine, with higher scores indicating increasing frailty. Patients with a higher CFS score have a higher 30-day mortality after surgery and are less likely to be discharged home [450].

It is important to use a validated tool to identify frailty, such as the G8 or CFS, as clinical judgement has been shown to be poorly predictive of frailty in older patients with cancer [451].

6.1.3.1. Co-morbidity

Co-morbidity is a major predictor of non-cancer-specific death in localised PCa treated with RP and is more important than age [452,453]. Ten years after watchful waiting for PCa, most men with a high co-morbidity score had died from competing causes, irrespective of age or tumour aggressiveness [452]. Measures for co-morbidity include: Cumulative Illness Score Rating-Geriatrics (CISR-G) [454,455] (Table 5.8) and Charlson Co-morbidity Index (CCI) [456].

6.1.3.2. Nutritional status

Malnutrition can be estimated from body weight during the previous three months (good nutritional status < 5% weight loss; risk of malnutrition: 5–10% weight loss; severe malnutrition: > 10% weight loss) [457].

6.1.3.3. Cognitive function

Cognitive impairment can be screened for using the mini-COG (https://mini-cog.com/) which consists of three-word recall and a clock-drawing test and can be completed within five minutes. A score of ≤ 3/5 indicates the need to refer the patient for full cognitive assessment. Patients with any form of cognitive impairment (e.g., Alzheimer’s or vascular dementia) may need a capacity assessment of their ability to make an informed decision, which is an increasingly important factor in health status assessment [458-460]. Cognitive impairment also predicts risk of delirium, which is important for patients undergoing surgery [461].

6.1.3.4. Physical function

Measures for overall physical functioning include: Karnofsky score and ECOG scores [462]. Measures for dependence in daily activities include Activities of Daily Living (ADL; basic activities) and Instrumental Activities of Daily Living (IADL; activities requiring higher cognition and judgement) [463-465].

6.1.3.5. Shared decision-making

The patient’s own values and preferences should be considered as well as the above factors. A shared decision-making process also involves anticipated changes to QoL, functional ability, and a patient’s hopes, worries and expectations about the future [466]. Particularly in older and frail patients, these aspects should be given equal importance to disease characteristics during the decision-making process [467]. Older patients may also wish to involve family members, and this is particularly important where cognitive impairment exists.

6.1.4. Conclusion

Individual life expectancy, health status, frailty, and co-morbidity, not only age, should be central in clinical decisions on screening, diagnostics, and treatment for PCa. A life expectancy of ten years is most commonly used as a threshold for benefit of local treatment. Older men may be under-treated. Patients aged 70 years of age or older who have frailty should receive a comprehensive geriatric assessment. Resolution of impairments in vulnerable men allows a similar urological approach as in fit patients.

Table 6.1: G8 screening tool (adapted from [468])


Items

Possible responses (score)

A

Has food intake declined over the past three months due to loss of appetite, digestive problems, chewing, or swallowing difficulties?

0 = severe decrease in food intake

1 = moderate decrease in food intake

2 = no decrease in food intake

B

Weight loss during the last three months?

0 = weight loss > 3 kg

1 = does not know

2 = weight loss between 1 and 3 kg

3 = no weight loss

C

Mobility?

0 = bed or chair bound

1 = able to get out of bed/chair but does not go out

2 = goes out

D

Neuropsychological problems?

0 = severe dementia or depression

1 = mild dementia

2 = no psychological problems

E

BMI? (weight in kg)/(height in m2)

0 = BMI < 19

1 = BMI 19 to < 21

2 = BMI 21 to < 23

3 = BMI > 23

F

Takes more than three prescription drugs per day?

0 = yes

1 = no

G

In comparison with other people of the same age, how does the patient consider his/her health status?

0.0 = not as good

0.5 = does not know

1.0 = as good

2.0 = better

H

Age

0 = > 85

1 = 80-85

2 = < 80

Total score

0-7

Figure 6.2: Decision tree for health status screening (men > 70 years)** [136]Mini-COGTM = Mini-COGTM cognitive test; ADLs = activities of daily living; CIRS-G = Cumulative IllnessRating Score - Geriatrics; CGA = comprehensive geriatric assessment.
* For Mini-COGTM, a cut-off points of ≤ 3/5 indicates a need to refer the patient for full evaluation of potential dementia.
**Reproduced with permission of Elsevier, from Boyle H. J., et al. Eur J Cancer 2019:116; 116 [136].

Figure 6.3: The Clinical Frailty Scale version 2.0 [449]**Permission to reproduce the CFS was granted by the copyright holder.

Table 6.2: Cumulative Illness Score Rating-Geriatrics (CISR-G)

Cumulative Illness Score Rating-Geriatrics (CISR-G)

1

Cardiac (heart only)

2

Hypertension (rating is based on severity; affected systems are rated separately)

3

Vascular (blood, blood vessels and cells, marrow, spleen, lymphatics)

4

Respiratory (lungs, bronchi, trachea below the larynx)

5

ENT (eye, ear, nose, throat, larynx)

6

Upper GI (esophagus, stomach, duodenum. Biliar and parcreatic trees; do not include diabetes)

7

Lower GI (intestines, hernias)

8

Hepatic (liver only)

9

Renal (kidneys only)

10

Other GU (ureters, bladder, urethra, prostate, genitals)

11

Musculo-Skeletal-Integumentary (muscles, bone, skin)

12

Neurological (brain, spinal cord, nerves; do not include dementia)

13

Endocrine-Metabolic (includes diabetes, diffuse infections, infections, toxicity)

14

Psychiatric/Behavioural (includes dementia, depression, anxiety, agitation, psychosis)


All body systems are scores on a 0 - 4 scale.

- 0: No problem affecting that system.

- 1: Current mild problem or past significant problem.

- 2: Moderate disability or morbidity and/or requires first line therapy.

- 3: Severe problem and/or constant and significant disability and/or hard to control chronic problems.

- 4: Extremely severe problem and/or immediate treatment required and/or organ failure and/or severe functional impairment.

Total score 0-56

6.1.5. Guidelines for evaluating health status and life expectancy

Recommendations

Strength rating

Use individual life expectancy, health status, and co-morbidity in PCa management.

Strong

Use the Geriatric-8, mini-COG and Clinical Frailty Scale tools for health status screening.

Strong

Perform a full specialist geriatric evaluation in patients with a G8 score ≤ 14.

Strong

Consider standard treatment in vulnerable patients with reversible impairments (after resolution of geriatric problems) similar to fit patients, if life expectancy is > ten years.

Weak

Offer adapted treatment or watchful waiting to patients with irreversible impairment.

Weak

Offer palliative symptom-directed therapy alone to frail patients.

Strong

6.2. Treatment modalities

6.2.1. Deferred treatment (watchful waiting/active surveillance)

As the prevalence of cancer cells in the prostate is so much higher than the risk of dying from PCa, together with the increased rate of early detection of small tumours after the introduction of PSA, there is a distinct risk of over-diagnosis and subsequent over-treatment of the disease (Chapter 3.1 Epidemiology) [8,12,469]. All available radical PCa treatment options may cause significant side effects so conservative treatment options are needed for patients with a low risk of PCa death or symptomatic progression from their PCa. Data from studies conducted on patients who did not undergo local treatment with up to 25 years of follow-up, with endpoints of OS and CSS, are available. Several series have shown a consistent CSS rate of 82–87% at ten years [470,471], and 80–95% for T1/T2 and ISUP grade group ≤ 2 PCas [472]. In three studies with data beyond 15 years, the CSS was 80%, 79% and 58% respectively [470,471,473], and two reported 20-year CSS rates of 57% and 32% [470,473]. The observed heterogeneity in outcomes is due to different inclusion criteria, with some older studies from the pre-PSA era showing worse outcomes [473]. In addition, many patients classified as ISUP grade group 1 would now be classified as ISUP grade group 2–3 based on the 2005 Gleason classification, suggesting that the above-mentioned results should be considered as minimal and current outcomes would be more favourable. Patients with well-, moderately- and poorly-differentiated tumours had 10-year CSS rates of 91%, 90% and 74%, respectively, correlating with data from a pooled analysis [472]. In screen-detected localised PCa there is also a lead-time bias, resulting in a higher rate of early detected PCa, but also an even higher risk of detecting clinically insignificant PCa that never would have caused any problems [469]. Cancer-specifc survival from untreated screen-detected PCa in patients with ISUP grade groups 1–2 is therefore likely to be even more favourable than for PCa detected of other reasons. Consequently, a high proportion of men with PSA-detected PCa are suitable for conservative management such as active surveillance (AS) or watchful waiting (WW). As the decision to choose WW is more independent from the tumour stage, and mainly dependent on patient factors/life expectancy, this approach may include patients of all ISUP grade groups (see Chapter 6.2.1.1).

The high CSS rates of localised PCa requires that a life expectancy of at least ten years should be considered mandatory for any benefit from active treatment. Co-morbidity is as important as age in predicting life expectancy in men with PCa. Increasing co-morbidity greatly increases the risk of dying from non-PCa-related causes. In an analysis of 19,639 patients aged > 65 years who were not given curative treatment, most men with a CCI score ≥ 2 had died from competing causes at ten years follow-up regardless of their age at time of diagnosis. Tumour aggressiveness had little impact on OS suggesting that patients could have been spared biopsy and diagnosis of cancer. Men with a CCI score ≤ 1 had a low risk of death at ten years, especially for well- or moderately-differentiated lesions [452]. Additionally, in the ProtecT trial (see 6.2.1.2), prostate cancer-related death was 3% at 15 years compared to death from any cause in 21.7% of patients, numbers that have been further validated in two large population-based studies from Canada and Sweden [474-476]. Estimation of competing benefits of active versus conservative treatment and death from any cause at ten and 15 years can be estimated using the PREDICT Prostate tool (available from https://prostate.predict.nhs.uk/), which was developed using registry data from the UK with external validation and is endorsed by the National Institute for Health and Care Excellence in the UK [477]. This highlights the importance of assessing co-morbidity even before considering a biopsy, but also before advising a patient with a PCa diagnosis on the optimal treatment for him.

There are two distinct strategies for conservative management that aim to reduce over-treatment: AS and WW (Table 6.2.1).

Table 6.2.1: Definitions of active surveillance and watchful waiting [478]

Definitions of active surveillance and watchful waiting


Active surveillance

Watchful waiting

Treatment intent

Curative

Palliative

Follow-up

Pre-defined schedule

Patient-specific

Assessment/markers* used

DRE, PSA, MRI at recruitment, re-biopsy

Annual (biannual) PSA and DRE if significant PSA-rise

Life expectancy

> ten years

< ten years

Aim

Minimise treatment-related toxicity without compromising survival, as the PCa is so indolent that it is unlikely to cause symptoms even with long life expectancy

Minimise treatment-related toxicity without compromising survival, as the lifespan is so limited that PCa is unlikely to cause symptoms

Eligible patients

Low- and selected intermediate-risk patients

Can apply to patients with all stages

DRE = digital rectal examination; PSA = prostate-specific antigen; MRI = magnetic resonance imaging.
*Molecular markers and/or PSMA-PET/CT (-MRI) may be used.

6.2.1.1. Watchful Waiting

Watchful waiting refers to conservative management for patients deemed unsuitable for curative treatment from the outset. The aim of WW is to balance the potential harms and benefits of early hormonal treatment, and patients are clinically ‘watched’ for the development of local or systemic progression with (imminent) disease-related complaints, at which stage they are then treated palliatively according to their symptoms in order to maintain QoL. Traditionally WW has meant waiting for symptoms of the tumour to develop and has, in some practices, not included regular follow-up in any active way. However, today we have evidence that early hormonal treatment could prolong short term survival (within a few years) for locally advanced disease, for patients with a PSAdt < twelve months, and for PSA-values over 30-50 ng/ml [479,480]. A more active follow-up of men on WW could therefore be beneficial, so that a local progression (often associated with a higher ISUP grade group), or start of metastatic spread, can be detected before they present with significant symptoms. Hormonal treatment could then be considered before symptoms emerge. The WW strategy should therefore be individualised and planned together with the patient. Biannual PSA, or annual after a period of stable disease, followed by DRE if PSA rises significantly, could then be of value, especially for men with a life expectancy > five years but unsuitable for curative treatment. There are two RCTs and one Cochrane review comparing the outcomes of WW to RP. The SPCG-4 study was a RCT from the pre-PSA era, randomising patients to either WW or RP [481]. The study found RP to provide superior CSS, OS and progression-free survival (PFS) compared to WW at a median follow-up of 23.6 years (range 3 weeks–28 years). However, the benefit in favour of RP over WW was only apparent after ten years. The PIVOT trial, a RCT conducted in the early PSA era, made a similar comparison between RP vs. WW in 731 men (50% with nonpalpable disease) but in contrast to the SPCG-4, it found little, to no, benefit of RP (cumulative incidence of all-cause death, RP vs. observation: 68% vs. 73%; RR: 0.92, 95% CI: 0.84–1.01) within a median follow-up period of 18.6 years (interquartile range, 16.6 to 20 years) [482]. Exploratory subgroup analysis showed that the borderline benefit from RP was most marked for intermediate-risk disease (RR: 0.84, 95% CI: 0.73–0.98) but there was no benefit in patients with low- or high-risk disease. Overall, no adverse effects on health related QoL (HRQoL) and psychological well-being was apparent in the first five years [483]. However, one of the criticisms of the PIVOT trial is the relatively high overall mortality rate in the WW group compared with more contemporary series. A Cochrane review performed a pooled analysis of RCTs comparing RP vs. WW [484]. Three studies were included; the previously mentioned SPCG-4 [481] and PIVOT [482] and the Veteran’s Administration Cooperative Urological Research Group (VACURG) study which was conducted in the pre-PSA era [485]. The authors found that RP compared with WW reduced time to death by any cause (HR: 0.79, 95% CI: 0.70–0.90), time to death by PCa (HR: 0.57, 95% CI: 0.44–0.73) and time to metastatic progression (HR: 0.56, 95% CI: 0.46–0.70) at 29 years’ follow-up. However, RP was associated with higher rates of urinary incontinence (RR: 3.97, 95% CI: 2.34–6.74) and ED (RR: 2.67, 95% CI: 1.63–4.38).

The overall evidence indicates that for men with asymptomatic, clinically localised PCa, and with a life expectancy of < ten years based on comorbidities and/or age, the oncological advantages of active treatment over WW are unlikely to be relevant to them. Consequently, WW should be adopted for such patients. For assistance in estimating life expectancy and health status see Section 5.4.

6.2.1.2. Active surveillance

Active surveillance aims to delay or completely avoid unnecessary treatment, and consequently unnecessary side effects, in men with clinically localised PCa, and a life expectancy of ten years or more, who do not require immediate treatment, but at the same time achieve the correct timing for curative treatment in those who eventually do [486]. Patients remain under close surveillance through structured surveillance programmes with regular follow-up consisting of PSA testing, clinical examination, MRI imaging and repeat prostate biopsies, with curative treatment being prompted by pre-defined thresholds indicative of development to potentially significant disease, which is still curable, while considering individual life expectancy.

No formal RCT is available comparing AS to curative treatment. Several cohorts have investigated AS in organ-confined disease, the findings of which were summarised in a SR [487]. A largest prospective series of men with low-risk PCa managed by AS was published [488]. Table 6.1.2 summarises the results of selective AS cohorts. It is clear that the long-term OS and CSS of patients on AS are extremely good. However, more than one-third of patients are ‘reclassified’ during follow-up, most of whom undergo curative treatment due to disease upgrading, increase in disease extent, disease stage, progression, or patient preference. There is considerable variation and heterogeneity between studies regarding patient selection and eligibility, follow-up policies (including frequency and type of imaging such as MRI imaging, type and frequency of repeat prostate biopsies, such as MRI-targeted biopsies or transperineal template biopsies, use of PSA kinetics and density, and frequency of clinical follow-up), when active treatment should be instigated (i.e., reclassification criteria) and which outcome measures should be prioritised [486]. For specific guidelines on inclusion criteria and follow-up strategies for AS, see Sections 6.2.1.2.

In the ProtecT RCT, 1,643 patients were randomised into one of three arms: active treatment with either RP or EBRT or AM with outcomes reported at ten years and 15 years [474,489]. Even though the ProtecT trial is a RCT it is not, strictly speaking, a study comparing AS to active treatment as it does not include a formal AS strategy as described above and in Sections 6.2.1.2. Active monitoring (AM), as used in the study, was a significantly less stringent surveillance strategy in terms of clinical follow-up, using PSA only, with relaxed criteria to define progression. No imaging and repeat biopsies were performed as in AS. At enrolment fifty-six percent of the patients had low-risk disease, with 90% having a PSA < 10 ng/mL, 77% ISUP grade group 1 (20% ISUP grade group 2–3), and 76% had T1c disease. The remaining patients had mainly intermediate-risk disease. The key finding was that AM was as effective as active treatment at 15 years (CSS = 96.9% in the AM-group vs. 97.8% in the RP-group and 97.1% in the EBRT-group, p=0.53), but at a cost of increased metastatic progression risk (9.4% vs. 4.7% and 5.0% respectively), as well as clinical progression at 15 years (25.9% for AM vs. 10.7% for RP/RT). Death from any cause occurred in 21.7% of the cohort, with similar numbers across treatment groups. Metastases, although rare, were more frequent than seen with comparable AS protocols [487]. A comprehensive characterisation of the ProtecT study cohort was performed after ten years, stratifying patients at baseline according to risk of progression using clinical stage, grade at diagnosis and PSA level [490]. Additionally, detailed clinico-pathological information on participants who received RP were analysed. The 15-year paper reported updated contemporary risk-stratification according to D’Amico (24.1% Intermediate risk, 9.6% high risk), CAPRA (26.4% Score 3-5, 2.5% Score 6-10) and Cambridge Prognostic Group (20.5% Group 2, 8.8% Groups 3-5). Among patients who underwent prostatectomy, 50.5% were ISUP grade group ≥2, while 28.5% had an increase in pathological stage and 32% had an increase in tumour grade. Additionally, 51% of patients who developed metastases displayed ISUP grade group 1 and 47.6% were low CAPRA risk. Over time, 61.1% of patients in the AM group received radical treatment (from 54.8% at ten years). From the ten year report the authors aimed identify prognostic markers. The results showed that treatment received, age (65–69 vs. 50–64 years), PSA, ISUP grade group at diagnosis, cT stage, risk group, number of PCa-involved biopsy cores, maximum length of tumour (median 5.0 vs. 3.0 mm), aggregate length of tumour (median 8.0 vs. 4.0 mm), and presence of perineural invasion were each associated with increased risk of disease progression (p < 0.001 for each). However, these factors could not reliably predict progression in individuals. Notably, 53% (n = 105) of patients who progressed had biopsy ISUP grade group 1 disease, although, conversely, none of the participants who received RP and subsequently progressed had pathological ISUP grade group 1 tumours. This discrepancy in progression and metastases rate between the AM arm of the ProtecT study and comparable AS protocols can, most likely, be explained by inadequate sampling by PSA testing and 10-core TRUS-guided biopsies and differences in intensity of surveillance.

It is important to note that the AM arm in ProtecT represented an intermediate approach between contemporary AS protocols and WW in terms of a monitoring strategy based almost entirely on PSA measurements alone; there was no use of MRI scan, either at recruitment or during the monitoring period, nor were there any protocol-mandated repeat prostate biopsies at regular intervals. In addition, approximately 40% of randomised patients had intermediate-risk disease (both ISUP grade group 2 and 3). Nevertheless, the ProtecT study has reinforced the role of deferred active treatment (i.e., either AS or some form of initial AM) as a feasible alternative to active curative interventions in all patients with low-grade and low-stage disease, as well as for many patients with favourable intermediate risk disease. Beyond 15 years, no RCT-data are available, as yet, although AS is likely to give more reassurance especially in younger men, based on more accurate risk stratification at recruitment and more stringent criteria regarding follow-up, imaging, repeat biopsy and reclassification. Individual life expectancy must continuously be evaluated before considering any active treatment in low-risk patients and in those with up to ten to 15 years’ individual life expectancy [490].

6.2.1.2.1. Active surveillance - inclusion criteria

Guidance regarding selection criteria for AS is limited by the lack of data from prospective RCTs. As a consequence, the Panel undertook an international collaborative study involving healthcare practitioners and patients to develop consensus statements for deferred treatment with curative intent for localised PCa, covering all domains of AS (DETECTIVE Study) [330], as well as a formal SR on the various AS protocols [491]. The criteria most often published include: ISUP grade group 1, clinical stage cT1c or cT2a, PSA < 10 ng/mL and PSA-D < 0.15 ng/mL/cc, as based on systematic biopsy schemes [487,492]. The latter threshold remains controversial [492,493]. These criteria were supported by the DETECTIVE study consensus. There was no agreement on the maximum number of systematic cores that can be involved with cancer or the maximum percentage core involvement (CI), although there was recognition that extensive disease on MRI should exclude men from AS, even though there is no firm definition on this, especially when targeted biopsies confirm ISUP grade group 1 [330]. The Movember consensus group, consisting of 27 healthcare professional and 12 lived experience participants from across the world, agreed that ISUP grade group and MRI were the most important criteria for determining eligibility to AS [494]. A SR and meta-analysis found three clinico-pathological variables which were significantly associated with reclassification, high PSA-D, > 2 positive cores (on systematic biopsies) and African-American descent [495]. A review on the risk of progression for African-American men on AS also indicated a potential increased risk of progression, but the association was not strong enough to discourage African-American men from undergoing AS, but thorough confirmatory testing is important [496]. In addition, a previous pathology consensus group suggested excluding men from AS when any of the following features were present: predominant ductal carcinoma (including pure IDC), cribriform histology, sarcomatoid carcinoma, small cell carcinoma, EPE or LVI in needle biopsy [497] and perineural invasion [498].

A multi-disciplinary consensus conference on germline testing has suggested a genetic implementation framework for the management of PCa [154]. Based on consensus, BRCA2-gene testing was recommended for AS discussions and could be performed in men with family history of prostate, breast or ovarian cancers. However, the nature of such discussions and how a positive result influences management were beyond the scope of the project. Currently, BRCA2 mutation does not exclude a patient from AS if tumour factors are otherwise favourable. Furthermore, if included in AS programmes, patients with a known BRCA2 mutation should be cautiously monitored until such time that more robust data are available.

6.2.1.2.2. Tissue-based prognostic biomarker testing for selection for AS

Biomarkers, including Oncotype Dx®, Prolaris®, Decipher®, PORTOS and ProMark® are promising (see Section 5.2.8.3). However, further data will be needed before such markers can be used in standard clinical practice [220].

6.2.1.2.3. Magnetic resonance imaging for selection for active surveillance

In men eligible for AS based upon systematic biopsy findings alone who did not have a pre-biopsy MRI, a re-biopsy within six to twelve months months (usually referred to as ‘confirmatory biopsy’) seems mandatory to exclude sampling error. A large body of literature including two RCTs and a SR, showed that adding MRI-targeted biopsy to systematic sampling at confirmatory biopsy improved detection of ISUP grade group ≥ 2 cancers and thus, patient selection for AS [121,499-504]. Adding MRI-targeted biopsy to systematic sampling at confirmatory biopsy improved upgrade detection by increments of 0-7.9 per 100 men depending on the series [499]. In a meta-analysis of 6 studies, the rate of upgrading to ISUP grade group ≥ 2 cancer increased from 20% (95% CI: 16–25%) to 27% (95% CI: 22–34%) when MRI-targeted biopsy was added to systematic biopsy [504]. The Active Surveillance MRI Study (ASIST) randomised men on AS scheduled for confirmatory biopsy to either 12-core systematic biopsy or to MRI with targeted biopsy (when indicated), combined with systematic biopsy (up to 12 cores in total). After two years of follow-up, use of MRI before confirmatory biopsy resulted in fewer failures of surveillance (19% vs. 35%, p = 0.017) and in fewer patients progressing to ISUP grade group ≥ 2 cancer (9.9% vs. 23%, p = 0.048) [502]. However, systematic biopsy retains its additional value, which argues for a combined biopsy approach [499,504]. The DETECTIVE study agreed that men eligible for AS after combined systematic- and MRI-targeted biopsy do not require a confirmatory biopsy, a recommendation further supported by the results of the MRIAS trial [330,505].

If the PCa diagnosis is made on MRI-targeted biopsy alone in order to lower the risk of over-detection of insignificant (see 5.4.1 and 5.4.2.), a confirmative systematic biopsy should be performed before definite decision of AS to rule out more widespread cancer growth in the prostate [186,189,190].

A few studies indicate that PSMA-PET-CT or PSMA-PET-MRI may have additional value to above mentioned clinico-pathological variables for risk stratification before AS [122,506]. However, so far, the studies are too small and the follow-up too short to draw any hard conclusions and for this modality to be recommended outside clinical trials.

6.2.1.2.4. Active surveillance management

Based on the DETECTIVE consensus study, the surveillance strategy should be based on serial DRE (at least once yearly), PSA (at least once, every six months) and repeated biopsy. It was also agreed that PSA progression or change in PSA kinetics alone should lead to reclassification only if accompanied by changes in histology on repeat biopsy [330]. The Movember consensus group stated that patients suitable for AS who suffer, or are at risk of, significant psychological distress should be offered more support, rather than active treatment. Furthermore, they made a number of recommendations that in some ways differ from the DETECTIVE consensus study, e.g. that routine DRE was not supported if MRI or other imaging was carried out routinely during AS, that if MRI and other parameters (PSA kinetics and density) are stable routine biopsy may be omitted and that change in clinical parameters should prompt MRI with possible biopsy rather than immediate biopsy [494]. The somewhat contradicting recommendations, made by these two different international consensus groups so close in time, clearly illustrate the lack of high-level evidence on how the strategy of AS should be planned and the urgent need of prospective randomised trials.

In 2016, the Prostate Cancer Radiological Estimation of Change in Sequential Evaluation (PRECISE) criteria were established to standardise the assessment of tumour progression on serial MRI [507]. Progression on MRI, or not, as defined by PRECISE criteria, is a strong predictor of histological upgrading [508,509]. Two independent meta-analyses assessed the value of MRI progression criteria for predicting histological progression (mostly defined as progression to ISUP grade group ≥ 2). The pooled histological progression rate was 27% in both reviews. If biopsies were triggered only by MRI progression findings, approximately two thirds of the biopsies would be avoided, at the cost of missing 40% of men with histological progression. In addition, at least half of biopsied men would have had negative findings for histological progression and thus would have undergone unnecessary biopsies. If histological progression was restricted to progression to ISUP grade group > 3, approximately 30% of histological progression would be missed and approximately 80% of the biopsies performed would be unnecessary. The use of the PRECISE criteria did not seem to change these results [510,511]. This supports maintaining protocol-mandated repeat biopsies during the course of AS.

Thus, the basis for AS protocols includes standard repeat biopsy. However, several factors have been found to be associated with low re-classification rates and long PFS: negative baseline or repeat MRI during AS [505,512-518], low PSA-D [505,513,515,518], low PSA velocity (PSAV) [519,520] or negative biopsy (i.e., no cancer at all) at confirmatory or repeat biopsy during AS [521]. Patients with stable (PRECISE 3) on repeat MRI during AS and a low PSA-D (<0.15) have a very low rate of progression and repeat biopsy may therefore be omitted [522].

A Panel SR incorporating 263 surveillance protocols showed that 78.7% of protocols mandated per-protocol confirmatory biopsies within the first two years and that 57.7% of the protocols performed repeat biopsy at least every three years for ten years after the start of AS [491]. In another review it was concluded that a negative repeat biopsy during AS was associated with a 50% decrease in the risk of future reclassification and upgrading [523]. In a single-centre AS cohort of 514 patients who underwent at least three protocol-mandated biopsies after diagnosis (the confirmatory biopsy and at least two additional surveillance biopsies), men with one negative biopsy (i.e., no cancer at all) at confirmatory or second biopsy, or men with two consecutive negative biopsies had a lower likelihood of a positive third biopsy and significantly better 10-year treatment free survival [521]. This suggests that men with repetitive negative biopsies may pursue AS with at least less frequent untriggered biopsies.

6.2.1.2.5. Active Surveillance - change in treatment

Men may remain on AS whilst they continue to consent, have a life expectancy of > ten years and the disease remains indolent. Patient anxiety about continued surveillance occurs in around 10% of patients on AS [524] and was recognised as a valid reason for active treatment [329]. An alternative for patients suitable for continuing AS would be to offer psychological support to reduce the level of anxiety [494]. A review on patient reported factor influencing the decision making, including thirteen qualitative papers and 426 men, identified a number of factors influencing the decision making when considering AS. Among the identified factors were personal risk assessment, influence of family and friends, beliefs about treatment as well as doctor and system factors, underscoring the importance of individualised, relevant, and clear information to support decision making [525]. A recent population-based cohort study from Sweden on regional differences in AS uptake and subsequent transition to radical treatment concluded that a regional tradition of a high uptake of AS was associated with a lower probability of transition to radical treatment, but not with AS failure [526]. These studies further emphasise the importance of thorough information and discussion with the patients on pros/cons of AS vs. active treatment already at the time of diagnosis for the patients to feel secure in their treatment choice and to avoid over-treatment.

A PSA change alone, including PSA-doubling time (PSA-DT, < 3 years) should not change management based on its weak link with grade progression [527,528] but rather trigger further investigation. There was clear agreement in the DETECTIVE consensus meeting as well as in the Movember consensus group that a change in PSA should lead to repeat-MRI and repeat-biopsy. It was also agreed that changes on repeat MRI during AS needed a confirmatory biopsy before considering active treatment [330,494].

The histopathology criteria required to trigger a change in management in the targeted biopsy era remain debated. MRI-targeted biopsy induces a grade shift and ISUP grade group 2–3 cancers detected by MRI-targeted biopsy have, on average, a better prognosis than those detected by systematic sampling (see Section 5.2.4.2.6.4). As an increasing number of men with favourable intermediate-risk disease are managed with AS (see section 6.2.2.1), it seems illogical to use progression to ISUP grade group 2 based on targeted biopsies as the sole criterion for reclassification. In addition, as acknowledged in the DETECTIVE consensus meeting, the number of positive cores is not an indicator of tumour volume anymore if targeted biopsies are performed [330,529]. No agreement could be reached on the pathological criteria required to trigger a change in management during the DETECTIVE consensus meeting [330]. However, based on the findings of a SR incorporating 271 reclassification protocols, patients with low-volume ISUP grade group 2 disease at recruitment, and with increased systematic core positivity (> 3 cores involvement [> 50% per core]) on repeat systematic biopsies not using MRI, should be reclassified [491]. Furthermore, in a study from the MUSIC registry over half of men with favourable intermediate-risk prostate cancer on AS remained free of treatment five years after diagnosis [530]. Their results are in concordance with the DETECTIVE and the Movember consensus statements and indicate that most men on AS will not lose their window of cure and have similar short-term oncologic outcomes as men undergoing up-front treatment and that AS is an oncologically safe option for appropriately selected men with favourable intermediate-risk prostate cancer.

The development of other comorbidities, resulting in a life expectancy of less than ten years should merit a new discussion with the patient and may result in a decision to transfer to a WW strategy.

Table 6.2.2: Active surveillance in screening-detected prostate cancer (large cohorts with longer-term follow-up)

Studies

N

Median FU (mo)

pT3 in RP patients*

10-year OS (%)

10-year CSS (%)

Adamy, et al. 2011 [489]

533-1,000

48

4/24 (17%)

90

99

Godtman, et al. 2013 [492]

439

72

-

81

99.5

Klotz, et al. 2015 [493]

993

77

-

85

98.1

Tosoian, et al. 2020 [494]

1,818

60

-

93

99.9

Carlsson, et al. 2020 [495]

2,664

52

-

94

100

Total

6,447–6,914

61.8

-

88.6

99.3

* Patients receiving active therapy following initial active surveillance.
CSS = cancer-specific survival; FU = follow-up; mo = months; n = number of patients; n.r. = not reported; OS = overall survival; RP = radical prostatectomy.

6.2.2. Radical prostatectomy

6.2.2.1. Introduction

The goal of RP by any approach is the eradication of cancer while, whenever possible, preserving pelvic organ function [531]. The procedure involves removing the entire prostate with its capsule intact and SVs, followed by vesico-urethral anastomosis. Surgical approaches have expanded from perineal and retropubic open approaches to laparoscopic and robotic-assisted techniques; anastomoses have evolved from Vest approximation sutures to continuous suture watertight anastomoses under direct vision and mapping of the anatomy of the dorsal venous complex (DVC) and cavernous nerves has led to excellent visualisation and potential for preservation of erectile function [532]. The main results from multi-centre RCTs involving RP are summarised in Table 6.1.3.

Table 6.2.3: Oncological results of radical prostatectomy in organ-confined disease in RCTs

Study

Acronym

Population

Treatment period

Median FU (mo)

Risk category

CSS (%)

Bill-Axelson, et al.

2018 [481]

SPCG-4

Pre-PSA era

1989-1999

283

Low risk and

intermediate risk

80.4

(at 23 yr.)

Wilt, et al.

2017 [482]

PIVOT

Early years of PSA testing

1994-2002

152

Low risk and

intermediate risk

95.9

91.5

(at 19.5 yr.)

Hamdy, et al.

2023 [474]

ProtecT

Screened population

1999-2009

180

Mainly low- and

intermediate risk

97

(at 15 yr.)

CSS = cancer-specific survival; FU = follow-up; mo = months; PSA = prostate-specific antigen; yr. = year.

6.2.2.2. Pre-operative preparation
6.2.2.2.1. Pre-operative patient education

As before any surgery appropriate education and patient consent is mandatory prior to RP. Peri-operative education has been shown to improve long-term patient satisfaction following RP [533]. Augmentation of standard verbal and written educational materials such as use of interactive multimedia tools [534,535] and pre-operative patient-specific 3D printed prostate models has been shown to improve patient understanding and satisfaction and should be considered to optimise patient-centred care [536].

6.2.2.3. Surgical techniques
6.2.2.3.1. Pelvic lymph node dissection

A SR has demonstrated that performing PLND during RP failed to improve oncological outcomes, including survival [536]. Moreover, two RCTs have failed to show a benefit of an extended approach vs. a limited PLND on early oncologic outcomes [537,538].

Extended PLND includes removal of the nodes overlying the external iliac artery and vein, the nodes within the obturator fossa located cranially and caudally to the obturator nerve, and the nodes medial and lateral to the internal iliac artery. With this template, the majority of patients are correctly staged [539] and as such, ePLND provides accurate information for staging and prognosis [540]

6.2.2.3.2. Lymph-node-positive patients during radical prostatectomy

Although no RCTs are available, data from prospective cohort studies comparing survival of pN+ patients (as defined following pathological examination after RP) support that RP may have a survival benefit over abandonment of RP in node-positive cases [541]. As a consequence, there is no role for performing frozen section of suspicious LNs.

6.2.2.3.3. Sentinel node biopsy analysis

The rationale for a sentinel node biopsy (SNB) is based on the concept that a sentinel node is the first to be involved by migrating tumour cells. Therefore, when this node is negative it is possible to avoid an ePLND [542]. Intraprostatic injections of indocyanine green (ICG) have been used to visualise prostate-related LNs for SNB. In a randomised comparison, Harke et al., found more cancer-containing LNs in men who underwent a PLND guided by ICG but no difference in BCR at 22.9-month follow-up [543]. A SR of 21 studies showed a sensitivity of 95.2% and NPV of 98.0% for SNB in detecting men with metastases at ePLND [544]. However, this review was hampered by widespread heterogeneity of both definitions and how SNB is performed. This prompted the development of an expert consensus report to guide further research [542].

The prospective SENTINELLE study investigated the diagnostic accuracy of sentinel lymph node biopsy-guided lymph node dissection compared to extended pelvic LN dissection in patients with intermediate- or high-risk prostate cancer. Sensitivity, specificity, NPV, and positive predictive value of SNB method in detecting patients with at least one LN metastasis were 95.4% (95% CI, 75.1-99.7), 100% (95% CI, 96.6-100), 99.2% (95% CI, 95.5-99.9), and 100% (95% CI, 80.7-100), respectively [545].

6.2.2.3.4. Prostatic anterior fat pad dissection and histologic analysis

Several multi-centre and large single-centre series have shown the presence of lymphoid tissue within the fat pad anterior to the endopelvic fascia; the prostatic anterior fat pad (PAFP) [546-552]. This lymphoid tissue is present in 5.5–10.6% of cases and contains metastatic PCa in up to 1.3% of intermediate- and high-risk patients.

When positive, the PAFP is often the only site of LN metastasis. The PAFP is therefore a rare but recognised route of spread of disease. The PAFP is always removed at RP for exposure of the endopelvic fascia and should be sent for histologic analysis as per all removed tissue.

6.2.2.3.5. Management of the dorsal venous complex

Since the description of the anatomical open RP by Walsh and Donker in the 1980s, various methods of controlling bleeding from the DVC have been proposed to optimise visualisation [553]. In the open setting, blood loss and transfusion rates have been found to be significantly reduced when ligating the DVC prior to transection [554]. However, concerns have been raised regarding the effect of prior DVC ligation on apical margin positivity and continence recovery due to the proximity of the DVC to both the prostatic apex and the urethral sphincter muscle fibres. In the robotic-assisted laparoscopic technique, due to the increased pressure of pneumoperitoneum, whether prior DVC ligation was used or not, blood loss was not found to be significantly different in one study [555]. In another study, mean blood loss was significantly less with prior DVC ligation (184 vs. 176 mL, p = 0.033), however it is debatable whether this was clinically significant [556]. The positive apical margin rate was not different, however, the latter study showed earlier return to full continence at five months post-operatively in the no prior DVC ligation group (61% vs. 40%, p < 0.01).

Ligation of the DVC can be performed with standard suture or using a vascular stapler. One study found significantly reduced blood loss (494 mL vs. 288 mL) and improved apical margin status (13% vs. 2%) when using the stapler [557].

Given the relatively small differences in outcomes, the surgeon’s choice to ligate prior to transection or not, or whether to use sutures or a stapler, will depend on their familiarity with the technique and the equipment available.

6.2.2.3.6. Nerve-sparing surgery

During prostatectomy, preservation of the neurovascular bundles (NVB) with parasympathetic nerve branches of the pelvic plexus can spare erectile function [558,559].

Extra-, inter-, and intra-fascial dissection planes can be planned, with those closer to the prostate and performed bilaterally associated with superior (early) functional outcomes [560-563]. Furthermore, many different techniques are propagated such as retrograde approach after anterior release (vs. antegrade), and athermal and traction-free handling of bundles [564-566]. Nerve-sparing (NS) surgery may be performed using clips or low bipolar energy without clear benefit favouring one technique over another regarding functional outcomes [567].

A 2021 large retrospective study of high-risk patients also found that NS did not affect BCR, risk of metastasis or of death [568]. Notably, clinical and pathological stage T3 and ISUP grade group 5 did not impact these oncological outcomes. However, as a retrospective study, it was subject to selection bias, whereby patients with unfavourable characteristics were more likely to have undergone non-nerve-sparing surgery.

A 2021 SR of 19 studies analysing the parameters used for selection of NS found that individual clinical and radiological factors were poor at predicting EPE, and consequently, the appropriateness of NS. However, nomograms that incorporated mpMRI performed better. As with all nomograms, the question remains as to where to set the cut-off point [569].

A 2022 SR of 18 comparative studies (no RCTs) of NS vs. non-nerve-sparing RP showed a RR of side-specific positive margins of 1.5, but none of them included patients with high-risk PCa [570]. There was no effect seen of NS on BCR. However, follow-up was short, and studies were subject to selection bias with mainly low-risk patients. For those patients with high-risk PCa, side-specific NS was avoided if disease was palpable or EPE was present on MRI. Indeed, a 2019 SR showed that MRI affected the decision to perform NS or not in 35% of cases without any negative impact on surgical margin rate [571] (See Section 5.3.2).

Although age and pre-operative function may remain the most important predictors for post-operative erectile function, NS has also been associated with improved continence outcomes and may therefore still be relevant for men with poor erectile function [572,573]. The association with continence may be mainly due to the dissection technique used during NS surgery, and not due to the preservation of the NVB themselves [572].

In summary, the quality of data is not adequate to permit a strong recommendation in favour of NS or non-nerve-sparing, but pre-operative risk factors for side-specific EPE such as PSA, PSA density, clinical stage, ISUP grade group, and PIRADS score, EPE and capsule contact length on MRI, should be taken into account.

6.2.2.3.7. Removal of seminal vesicles

The more aggressive forms of PCa may spread directly into the SVs. For oncological clearance, the SVs have traditionally been removed intact with the prostate specimen [574]. However, in some patients the tips of the SVs can be challenging to dissect free. Furthermore, the cavernous nerves run past the SV tips such that indiscriminate dissection of the SV tips could potentially lead to ED [575]. However, a RCT comparing nerve-sparing RP with and without a SV-sparing approach found no difference in margin status, PSA recurrence, continence or erectile function outcomes. Whilst complete SV removal should be the default, preservation of the SV tips may be considered in cases of low risk of involvement.

6.2.2.3.8. Techniques of vesico-urethral anastomosis

Following prostate removal, the bladder neck is anastomosed to the membranous urethra. The objective is to create a precisely aligned, watertight, tension-free, and stricture-free anastomosis that preserves the integrity of the intrinsic sphincter mechanism. Several methods have been described, based on the direct or indirect approach, the type of suture (i.e. barbed vs. non-barbed/monofilament), and variation in suturing technique (e.g., continuous vs. interrupted, or single-needle vs. double-needle running suture). The direct vesico-urethral anastomosis, which involves the construction of a primary end-to-end inter-mucosal anastomosis of the bladder neck to the membranous urethra by using 6 interrupted sutures placed circumferentially, has become the standard method of reconstruction for open RP [576].

The development of laparoscopic- and robotic-assisted techniques to perform RP have facilitated the introduction of new suturing techniques for the anastomosis. A SR and meta-analysis compared unidirectional barbed suture vs. conventional non-barbed suture for vesico-urethral anastomosis during robotic-assisted laparoscopic prostatectomy (RALP) [577]. The review included 3 RCTs and found significantly reduced anastomosis time, operative time and posterior reconstruction time in favour of the unidirectional barbed suture technique, but there were no differences in post-operative leak rate, length of catheterisation and continence rate. However, no definitive conclusions could be drawn due to the relatively low quality of the data. In regard to suturing technique, a SR and meta-analysis compared continuous vs. interrupted suturing for vesico-urethral anastomosis during RP [578]. The study included only one RCT with 60 patients [579]. Although the review found slight advantages for continuous suturing over interrupted suturing in terms of catheterisation time, anastomosis time and rate of extravasation, the overall quality of evidence was low and no clear recommendations were possible. A RCT [580] compared the technique of suturing using a single absorbable running suture vs. a double-needle single-knot running suture (i.e. Van Velthoven technique) in laparoscopic RP [581]. The study found slightly reduced anastomosis time with the single running suture technique, but anastomotic leak, stricture, and continence rates were similar.

Overall, although there are a variety of approaches, methods, and techniques for performing the vesico-urethral anastomosis, no clear recommendations are possible due to the lack of high-certainty evidence. In practice, the chosen method should be based on surgeon experience and individual preference [576-581].

6.2.2.3.9. Bladder neck management

Bladder neck mucosal eversion

Some surgeons perform mucosal eversion of the bladder neck as its own step in open RP with the aim of securing a mucosa-to-mucosa vesico-urethral anastomosis and avoiding anastomotic stricture. Whilst bringing bladder and urethral mucosa together by the everted bladder mucosa covering the bladder muscle layer, this step may actually delay healing of the muscle layers. An alternative is to simply ensure bladder mucosa is included in the full thickness anastomotic sutures. A non-randomised study of 211 patients with and without bladder neck mucosal eversion showed no significant difference in anastomotic stricture rate [582]. The strongest predictor of anastomotic stricture in RP is current cigarette smoking [583], but it is also 2.2 higher in open RP than RARP [584].

Bladder neck preservation

Whilst the majority of urinary continence is maintained by the external urethral sphincter at the membranous urethra (see below), a minor component is contributed by the internal lissosphincter at the bladder neck [585]. Preservation of the bladder neck has therefore been proposed to improve continence recovery post-RP. A RCT assessing continence recovery at twelve months and four years showed improved objective and subjective urinary continence in both the short- and long term without any adverse effect on oncological outcome [586]. These findings were confirmed by a SR [587]. However, concern remains regarding margin status for cancers located at the prostate base.

A SR addressing site-specific margin status found a mean base-specific positive margin rate of 4.9% with bladder neck preservation vs. only 1.9% without [585]. This study was inconclusive, but it would be sensible to exercise caution when considering bladder neck preservation if significant cancer is known to be at the prostate base. Bladder neck preservation should be performed routinely when the cancer is distant from the base. However, bladder neck preservation cannot be performed in the presence of a large median lobe or a previous transurethral resection of the prostate (TURP) [588].

6.2.2.3.10. Urethral length preservation

The membranous urethra sits immediately distal to the prostatic apex and is chiefly responsible, along with its surrounding pelvic floor support structures, for urinary continence. It consists of the external rhabdosphincter which surrounds an inner layer of smooth muscle. Using pre-operative MRI, the length of membranous urethra has been shown to vary widely.

Systematic reviews and meta-analyses found that every extra millimetre of membranous urethral length seen on MRI pre-operatively improves early return to continence post-RP [589-591]. A greater membranous urethral length as measured on preoperative MRI was an independent prognostic factor for return to urinary continence within one month after RP and remained prognostic at twelve months [591]. Therefore, it is likely that preservation of as much urethral length as possible during RP will maximise the chance of early return to continence. It may also be useful to measure urethral length pre-operatively on MRI to facilitate counselling of patients on their relative likelihood of early post-operative continence [592].

6.2.2.3.11. Cystography prior to catheter removal

Cystography may be used prior to catheter removal to check for a substantial anastomotic leak. If such a leak is found, catheter removal may then be deferred to allow further healing and sealing of the anastomosis. However, small comparative studies suggest that a cystogram to assess anastomotic leakage is not indicated as SOC before catheter removal eight to ten days after surgery [593]. If a cystogram is used, men with LUTS, large prostates, previous TURP or bladder neck reconstruction, may benefit as these factors have been associated with leakage [594,595]. Contrast-enhanced transrectal US is an alternative [596].

6.2.2.3.12. Urinary catheter

A urinary catheter is routinely placed during RP to enable bladder rest and drainage of urine while the vesicourethral anastomosis heals. Compared to a traditional catheter duration of around one week, some centres remove the transurethral catheter early (post-operative day 2–3), usually after thorough anastomosis with posterior reconstruction or in patients selected peri-operatively on the basis of anastomosis quality [597-600]. No higher complication rates were found. Although shorter catheterisation has been associated with more favourable short-term functional outcomes, no differences in long-term function were found [601]. One RCT has shown no difference in rate of UTI following indwelling catheter (IDC) removal whether prophylactic ciprofloxacin was given prior to IDC removal or not, suggesting antibiotics should not be given at catheter removal [602].

As an alternative to transurethral catheterisation, suprapubic catheter insertion during RP has been suggested. Some reports suggest less bother regarding post-operative hygiene and pain [603-607], while others did not find any differences [608,609]. No impact on long-term functional outcomes were seen.

6.2.2.3.13. Use of a pelvic drain

A pelvic drain has traditionally been used in RP for potential drainage of urine leaking from the vesico-urethral anastomosis, blood, or lymphatic fluid when a PLND has been performed. Two RCTs in the robotic-assisted laparoscopic setting have been performed [610,611]. Patients with urine leak at intra-operative anastomosis watertight testing were excluded. Both trials showed non-inferiority in complication rates when no drain was used. When the anastomosis is found to be watertight intra-operatively, it is reasonable to avoid inserting a pelvic drain. There is no evidence to guide usage of a pelvic drain in PLND.

6.2.2.4. Acute and chronic complications of radical prostatectomy

Post-operative incontinence and ED are common problems following surgery for PCa. A key consideration is whether these problems are reduced by using newer techniques such as RALP. Systematic reviews have documented complication rates after RALP [612-616], and can be compared with contemporaneous reports after radical retropubic prostatectomy (RRP) [617]. A prospective controlled non-RCT of patients undergoing RP in 14 centres using RALP or RRP showed that twelve months after RALP, 21.3% of patients were incontinent, as were 20.2% after RRP (adjusted OR: 1.08, 95% CI: 0.87–1.34) [618]. Erectile dysfunction was observed in 70.4% after RALP and 74.7% after RRP. The adjusted OR was 0.81 (95% CI: 0.66–0.98) [618].

A SR and meta-analysis of unplanned hospital visits and re-admissions post-RP analysed 60 studies with over 400,000 patients over a 20-year period up to 2020. It found an emergency room visit rate of 12% and a hospital re-admission rate of 4% at 30 days post-operatively [619].

A RCT comparing RALP and RRP reported outcomes at 12 weeks in 326 patients and functional outcomes at two years [620]. Urinary function scores did not differ significantly between RRP vs. RALP at 6 and 12 weeks post-surgery (74–50 vs. 71–10, p = 0.09; 83–80 vs. 82–50, p = 0.48), with comparable outcomes for sexual function scores (30–70 vs. 32–70, p = 0.45; 35–00 vs. 38–90, p = 0.18). In the RRP group 14 (9%) patients had post-operative complications vs. 6 (4%) in the RALP group. The intra- and peri-operative complications of RRP and RALP are listed in Table 6.1.4. Table 6.1.5 lists the Clavien-Dindo definition of surgical complications. The early use of phosphodiesterase-5 (PDE5) inhibitors in penile rehabilitation remains controversial resulting in a lack of clear recommendations (see Section 8.3.2.1).

Table 6.2.4: Intra-and peri-operative complications of retropubic RP, laparoscopic RP and RALP(adapted from [612])

Predicted probability of event

RALP (%)

Laparoscopic RP (%)

RRP (%)

Bladder neck contracture

1.0

2.1

4.9

Anastomotic leak

1.0

4.4

3.3

Infection

0.8

1.1

4.8

Organ injury

0.4

2.9

0.8

Ileus

1.1

2.4

0.3

Deep-vein thrombosis

0.6

0.2

1.4

Predicted rates of event

RALP (%)

Laparoscopic RP (%)

RRP (%)

Clavien-Dindo I

2.1

4.1

4.2

Clavien-Dindo II

3.9

7.2

17.5

Clavien-Dindo IIIa

0.5

2.3

1.8

Clavien-Dindo IIIb

0.9

3.6

2.5

Clavien-Dindo IVa

0.6

0.8

2.1

Clavien-Dindo V

< 0.1

0.2

0.2

RALP = robot-assisted laparoscopic prostatectomy; RP = radical prostatectomy; RRP = radical retropubic prostatectomy.

Table 6.2.5: Clavien-Dindo grading of surgical complications [621]

Grade

Definition

I

Any deviation from the normal post-operative course not requiring surgical, endoscopic or radiological intervention. This includes the need for certain drugs (e.g. antiemetics, antipyretics, analgesics, diuretics and electrolytes), treatment with physiotherapy and wound infections that are opened at the bedside

II

Complications requiring drug treatments other than those allowed for Grade I complications; this includes blood transfusion and total parenteral nutrition (TPN)

IIIa

Complications requiring surgical, endoscopic or radiological intervention

- intervention not under general anaesthetic

IIIb

Complications requiring surgical, endoscopic or radiological intervention

- intervention under general anaesthetic

IVa

Life-threatening complications; this includes central nervous system (CNS) complications (e.g. brain haemorrhage, ischaemic stroke, subarachnoid haemorrhage) which require intensive care,
but excludes transient ischaemic attacks (TIAs)

- single-organ dysfunction (including dialysis)

IVb

Life-threatening complications; this includes CNS complications (e.g. brain haemorrhage, ischaemic stroke, subarachnoid haemorrhage) which require intensive care, but excludes transient ischaemic attacks (TIAs)

- multi-organ dysfunction

V

Death of the patient

6.2.2.4.1. Effect of anterior and posterior reconstruction on continence

Preservation of integrity of the external urethral sphincter is critical for continence post-RP. Less clear is the effect of reconstruction of surrounding support structures to return to continence. Several small RCTs have been conducted, however, pooling analyses is hampered by variation in the definitions of incontinence and surgical approach, such as open vs. robotic and intra-peritoneal vs. extra-peritoneal. In addition, techniques used to perform both anterior suspension or reconstruction and posterior reconstruction are varied. For example, anterior suspension is performed either through periosteum of the pubis or the combination of ligated DVC and puboprostatic ligaments (PPL). Posterior reconstruction from rhabdosphincter is described to either Denonvilliers fascia posterior to bladder or to posterior bladder wall itself.

Two trials assessing posterior reconstruction in RALRP found no significant improvement in return to continence [622,623]. A third trial using posterior bladder wall for reconstruction showed only an earlier return to 1 pad per day (median 18 vs. 30 days, p = 0.024) [624]. When combining both anterior and posterior reconstruction, where for anterior reconstruction the PPL were sutured to the anterior bladder neck, another RCT found no improvement compared to a standard anastomosis with no reconstruction [625].

Four RCTs including anterior suspension have also shown conflicting results. Anterior suspension alone through the pubic periosteum, in the setting of extra-peritoneal RALRP, showed no advantage [626]. However, when combined with posterior reconstruction in RRP, one RCT showed significant improvement in return to continence at one month (7.1% vs. 26.5%, p = 0.047) and three months (15.4% vs. 45.2%, p = 0.016), but not at six months (57.9% vs. 65.4%, p = 0.609) [627]. Another anterior plus posterior reconstruction RCT using the Advanced Reconstruction of VesicoUrethral Support (ARVUS) technique and the strict definition of continence of ‘no pads‘, showed statistically significant improvement in continence at 2 weeks (43.8% vs. 11.8%), 4 weeks (62.5% vs. 14.7%), 8 weeks (68.8% vs. 20.6%), six months (75% vs. 44.1%) and twelve months (86.7% vs. 61.3%), when compared to standard posterior Rocco reconstruction [628]. Anterior suspension alone through the DVC and PPL combined without posterior construction in the setting of RRP has shown improvement in continence at one month (20% vs. 53%, p = 0.029), three months (47% vs. 73%, p = 0.034) and six months (83% vs. 100%, p = 0.02), but not at twelve months (97% vs. 100%, p = 0.313) [629]. Together, these results suggest a possible earlier return to continence, but no long-term difference.

As there is conflicting evidence on the effect of anterior and/or posterior reconstruction on return to continence post-RP, no recommendations can be made. However, no studies showed an increase in adverse oncologic outcome or complications with reconstruction.

6.2.2.4.2. Deep venous thrombosis prophylaxis

For EAU Guidelines recommendations on post-RP deep venous thrombosis prophylaxis, please see the Thromboprophylaxis Guidelines Section 3.1.6 [630]. However, these recommendations should be adapted based on national recommendations, when available.

6.2.2.4.3. Complications of extended pelvic lymph node dissection

Extended PLND increases morbidity in the treatment of PCa [540]. Overall complication rates of 19.8% vs. 8.2% were noted for ePLND vs. limited PLND, respectively, with lymphoceles (10.3% vs. 4.6%) being the most common adverse event (AE). Other authors have reported lower complication rates [631]. Briganti et al., [632] also showed more complications after extended compared to limited PLND. Twenty percent of men suffer a complication of some sort after ePLND. Thromboembolic events occur in less than 1% of cases overall, but the RR of DVT and PE associated with PLND has been found to be 7.8 and 6.3, respectively [633].

6.2.3. Radiotherapy

Intensity-modulated RT (IMRT) or volumetric modulated arc therapy (VMAT) with image-guided RT (IGRT) is currently widely recognised as the standard treatment approach for EBRT.

6.2.3.1. External beam radiation therapy
6.2.3.1.1. Technical aspects

Intensity-modulated RT and VMAT employ dynamic multi-leaf collimators, which automatically and continuously adapt to the contours of the target volume seen by each beam. Viani et al., show significantly reduced acute and late grade ≥ 2 genito-urinary (GU) and gastro-intestinal (GI) toxicity in favour of IMRT, while BCR-free rates did not differ significantly when comparing IMRT with three-dimensional conformal RT (3D-CRT) in a RCT comprising 215 patients [634]. A meta-analysis by Yu et al., (23 studies, 9,556 patients) concluded that IMRT significantly decreases the occurrence of grade 2–4 acute GI toxicity, late GI toxicity and late rectal bleeding, and achieves better PSA relapse-free survival in comparison with 3D-CRT. Intensity-modulated EBRT and 3D-CRT show comparable acute rectal toxicity, late GU toxicity and OS, while IMRT slightly increases the morbidity of acute GU toxicity [635]. Zapatero et al., found, based on 733 consecutive patients (295 IMRT vs. 438 3D-CRT), that compared with 3D-CRT, high-dose IMRT/IGRT is associated with a lower rate of late urinary complications despite a higher radiation dose [636]. In conclusion, IMRT plus IGRT remain the SOC for the treatment of PCa.

The advantage of VMAT over IMRT is shorter treatment times, generally two to three minutes in total. Both techniques allow for a more complex distribution of the dose to be delivered and provide concave isodose curves, which are particularly useful as a means of sparing the rectum. Radiotherapy treatment planning for IMRT and VMAT differs from that used in conventional EBRT, requiring a computer system capable of ‘inverse planning’ and the appropriate physics expertise. Treatment plans must conform to pre-specified dose constraints to critical organs at risk of normal tissue damage and a formal quality assurance process should be routine.

With dose escalation using IMRT/VMAT, organ movement becomes a critical issue in terms of both tumour control and treatment toxicity. Techniques will therefore combine IMRT/VMAT with some form of IGRT (usually gold marker or cone-beam CT), in which organ movement can be visualised and corrected for in real time, although the optimum means (number of applications per week) of achieving this is still unclear [637,638]. Tomotherapy is another technique for the delivery of IMRT, using a linear accelerator mounted on a ring gantry that rotates as the patient is delivered through the centre of the ring, analogous to spiral CT scanning.

The use of MR-guided adapted RT is still investigational [639]. Planning studies confirm that MR-based adaptive RT significantly reduces doses to organs at risk (OAR) and this may translate into clinical benefit [640]. Although the rates of acute GI- and GU toxicity appear low, mostly on the basis of patients treated with stereotactic RT [641], follow-up is too short for definitive conclusions [639]. The daily fraction time of up to 45 minutes [639,641], the heavy MR-workflow and the limited field size (rendering most pelvic fields too large) make its implementation not yet a routine [639]. A prospective single center RCT, the MIRAGE trial (CT-guided Stereotactic Body Radiation Therapy and MRI-guided Stereotactic Body Radiation Therapy for Prostate Cancer) demonstrates reduced acute GU and GI toxicity with MRI-guided SBRT and margin reduction from 4mm to 2mm [642]. The impact on long term toxicity, biochemical control and cost effectiveness remains undefined.

6.2.3.1.2. Dose escalation

Local control is a critical issue for the outcome of RT of PCa. It has been shown that local failure due to insufficient total dose is prognostic for death from PCa as a second wave of metastases is seen five to ten years later on [643]. Several RCTs have shown that dose escalation (range 74–80 Gy) has a significant impact on 10-year biochemical relapse as well as metastases and disease-specific mortality [644-651]. These trials have generally included patients from several risk groups, and the use of neoadjuvant/adjuvant HT has varied (see Table 6.1.6). The best evidence of an OS benefit in patients with intermediate- or high-risk PCa, but not with low-risk PCa, derives from a non-randomised but well conducted propensity-matched retrospective analysis of the U.S. National Cancer Database by Kalbasi et al., including a total of 42,481 patients [652]. If IMRT/VMAT and IGRT are used for dose escalation, rates of severe late side effects (> grade 3) for the rectum are 2–4% and for the GU tract 2–6% [646,653].

The concept of a focal boost to the dominant intraprostatic lesion (DIL) visible on MRI rather than global prostate dose escalation has been successfully validated in a RCT of 571 intermediate- and high-risk patients [653]. Patients were randomised between 77 Gy in 35 fractions of 2.2 Gy and the same dose plus a focal boost up to 18 Gy. Additional ADT was given to 65% of patients in both arms. However, the duration of the ADT was not reported. With a median follow-up of 72 months there was a moderate improvement of biochemical PFS (BPFS) (primary endpoint). In addition, focal boosting decreased local failure (HR: 0.33) and increased the rate of regional + distant MFS (HR: 0.58) [654]. No significant difference for late GU- or GI toxicity grade ≥ 2 (23% and 12% vs. 28% and 13%) was documented. For grade ≥ 3 GU-toxicity these numbers were 3.5% and 5.6% (p > 0.05). However, longer follow-up is needed to assess late GU-toxicity [654]. Of note, there was a clear decrease in biochemical failure with increasing boost dose, individually given up to 18 Gy. Systematic review of MRI-defined DIL focal boost studies using standard fractionation shows good tolerability and improved BPFS [655]. Its role when using hypofractionation and ultra-hypofractionation is under investigation.

Table 6.2.6: Randomised trials of dose escalation in localised PCa

Trial

n

PCa condition

Radiotherapy Dose

Follow-up (median)

Outcome

Results

MD Anderson

study 2011 [651]

301

T1-T3, N0, M0,

PSA ≤ 10 ng/mL

PSA 10-20 ng/mL

PSA > 20 ng/mL

70 vs.78 Gy

15 yr.

DM, DSM, FFF

All patients:

18.9% FFF at 70 Gy

12% FFF at 78 Gy

(p = 0.042)

3.4% DM at 70 Gy

1.1% DM at 78 Gy

(p = 0.018)

6.2% DSM at 70 Gy

3.2% DSM at 78 Gy

(p = 0.043)

No difference in OS

(p > 0.05)

PROG

95-09

2010 [645]

393

T1b-T2b

PSA ≤ 15 ng/mL

75% low-risk pts.

Low-risk: T1-2a,

PSA < 10 mg/mL,

GS ≤ 6

Interm-risk:

PSA 10-15 ng/mL or

GS 7 or T2b

High-risk: GS

8-10

70.2 vs.79.2 Gy including proton boost

19.8 vs. 28.8 Gy

8.9 yr.

10-yr.

ASTRO BCF

All patients:

32% BF at 70.2 Gy

17% BF at 79.2 Gy

(p < 0.0001)

Low-risk patients:

28% BF at 70.2 Gy

7% BF at 79.2 Gy

(p < 0.0001

MRC RT01 2014 [650]

843

T1b-T3a, N0, M0

PSA < 50 ng/mL neoadjuvant HT

64 vs. 74 Gy

10 yr.

BFS, OS

43% BFS at 64 Gy

55% BFS at 74 Gy

(p = 0.0003) 71%

OS both groups (p = 0.96)

Dutch rando-mised phase III trial 2014 [649]

664

T1b-T4 143 pts. with (neo) adjuvant HT

68 vs. 78 Gy

110 mo.

Freedom

biochemical

(Phoenix)

and/or clinical

failure at

10 yr.

43% FFF at 68 Gy

49% FFF at 78 Gy

(p = 0.045)

GETUG 06

2011 [648]

306

T1b-T3a, N0, M0

PSA < 50 ng/mL

70 vs. 80 Gy

61 mo.

BCF (ASTRO)

39% BF at 70 Gy

28% BF at 80 Gy

RTOG 0126

2018 [644]

1,532

T1b-T2b

ISUP grade

group 1

+ PSA 10-20

ng/mL or ISUP grade group 2/3 + PSA < 15 ng/mL

70.2 vs. 79.2 Gy

100 mo.

OS, DM, BCF (ASTRO)

75% OS at 70.2 Gy

76% OS at 79.2 Gy

6% DM at 70.2 Gy

4% DM at 79.2 Gy

(p = 0.05)

47% BCF at 70.2 Gy

31% BCF at 79.2 Gy

(p < 0.001;

Phoenix, p < 0.001)

FLAME Trial [653,654]

571

EAU risk classification:

Intermediate risk (15%)

High risk (84%)

77 Gy (35 Fx.

2.2 Gy) vs. 77 Gy

(35 Fx.)

+ focal boost

(up to 18 Gy)

ADT (65% both arms – duration unknown)

72 mo. (median)

BFS (5 yr.) DSM (5 yr.)

BFS:

92% at 77 Gy + boost

85% at 77 Gy

(p < 0.001, HR: 0.45)

DSM: p= 0.49

Focal boost in favour of:

Local control (HR: 0.33)

Distant MFS (HR: 0.58)

ADT = androgen-deprivation therapy; BF = biochemical failure; BFS = biochemical progression-free survival;DM = distant metastases; DSM = 50disease specific mortality; FFF = freedom from biochemical or clinical failure;Fx = fractions; GS = Gleason score; HT = hormone therapy; ISUP = International Society of Urological Pathology; MFS = metastasis-free survival; mo. = months; n = number of patients; OS = overall survival;PSA = prostate-specific antigen; yr. = year.

6.2.3.1.3. Hypofractionation

Fractionated RT utilises differences in the DNA repair capacity of normal and tumour tissue and slowly proliferating cells are very sensitive to an increased dose per fraction [656]. A meta-analysis of 25 studies including > 14,000 patients concluded that since PCa has a slow proliferation rate, hypofractionated RT could be more effective than conventional fractions of 1.8–2 Gy [657]. Hypofractionation (HFX) has the added advantage of being more convenient for the patient at lower cost.

Moderate HFX is defined as RT with 2.5–3.4 Gy/fx. Several studies report on moderate HFX applied in various techniques also including HT in part [658-665]. A SR concluded that studies on moderate HFX (2.5–3.4 Gy/fx) delivered with conventional 3D-CRT/IMRT have sufficient follow-up to support the safety of this therapy but long-term efficacy data are still lacking [664]. These results were confirmed by a Cochrane review on moderate HFX for clinically localised PCa [666]. Eleven studies were included (n = 8,278) with a median follow-up of 72 months showing little or no difference in PCa-specific survival (HR: 1.00). Based on 4 studies (n = 3,848), HFX probably makes little or no difference to late radiation GU toxicity (RR: 1.05) or GI toxicity (RR: 1.1), but this conclusion is based on relatively short follow-up, and ten to 15-year data will be required to confirm these findings.

Moderate HFX should only be done by experienced teams using high-quality EBRT using IGRT and IMRT/VMAT and published phase III protocols should be adhered to (Table 6.1.7).

Table 6.2.7: Major phase III randomised trials of moderate hypofractionation for primary treatment

Study/Author

n

Risk, ISUP grade, or NCCN

ADT

RT Regimen

BED, Gy

Median FU, mo

Outcome

Lee, et al. 2016 [660]

550

542

low risk

None

70 Gy/28 fx

73.8 Gy/41 fx

80

69.6

70

5 yr. DFS 86.3%

(n.s.)

5 yr. DFS 85.3%

Dearnaley, et al. CHHiP 2016 [661]

1,077/19 fx

1,074/20 fx

1,065/37 fx

15% low 73% intermediate 12% high

3-6 mo. before and during EBRT

57 Gy/19 fx

60 Gy/20 fx

74 Gy/37 fx

73.3

77.1

74

62

5 yr. BCDF 85.9% (19 fx) 90.6% (20 fx) 88.3% (37 fx)

De Vries, et al., 2020 [667]

403

392

30% ISUP grade 1

45% ISUP grade 2-3,

25% ISUP grade 4-5

None

64.6 Gy/19 fx

78 Gy/39 fx

90.4

78

89

8-yr. OS 80.8% vs. 77.6%

(p = 0.17)


8 yr. TF 24.4% vs. 26.3%

Catton, et al. 2017 [663]

608

intermediate risk

53% T1c 46% T2a-c

None

60 Gy/20 fx

77.1

72

5 yr. BCDF both arms 85%

HR: 0.96 (n.s)

598

9% ISUP grade 1

63% ISUP grade 2

28% ISUP grade 3

78 Gy/39 fx

78

ADT = androgen deprivation therapy; BCDF = biochemical or clinical disease failure; BED = biologically equivalent dose, calculated to be equivalent in 2 Gy fractions using an α/ß of 1.5 Gy; DFS = diseasefree survival;EBRT = external beam radiotherapy; FU = follow-up; fx = fractions; HR = hazard ratio; ISUP = International Society of Urological Pathology; mo. = month; n = number of patients; NCCN = National Comprehensive Cancer Network;n.s. = not significant; TF = treatment failure; yr. = year.

Ultra-HFX has been defined as RT with > 3.4 Gy per fraction [665]. It requires IGRT and (ideally) stereotactic body RT (SBRT). Table 6.1.8 provides an overview of selected studies investigating its role in treating predominantly intermediate risk localised disease. Short-term biochemical control (5-years) is comparable to conventional fractionation. However, there are concerns about high-grade GU and rectal toxicity and full long-term side effects may not yet be known [664,668]. In the HYPO-RT-PC randomised trial by Widmark et al., (n = 1,200), no difference in failure-free survival was seen for conventional or ultra-HFX but acute grade ≥ 2 GU toxicity was 23% vs. 28% (p = 0.057), favouring conventional fractionation. There were no significant differences in long-term toxicity [668]. A SR by Jackson et al., included 38 studies with 6,116 patients who received RT with < 10 fractions and ≥ 5 Gy per fraction. Five and seven-year biochemical recurrence-free survival (BRFS) rates were 95.3% and 93.7%, respectively, and estimated late grade ≥ 3 GU and GI toxicity rates were 2.0% and 1.1%, respectively [669]. The authors conclude that there is sufficient evidence to support SBRT as a standard treatment option for localised PCa, even though the median follow-up in this review was only 39 months and it included at least one trial (HYPO-RT-PC) which used 3D-CRT in 80% and IMRT/VMAT in the remainder for ultra-HFX. In their review on SBRT, Cushman et al., evaluated 14 trials, including 2,038 patients and concluded that despite a lack of long-term follow-up and the heterogeneity of the available evidence, prostate SBRT affords appropriate biochemical control with few high-grade toxicities [670]. In the Intensity-modulated fractionated RT vs. stereotactic body RT for PCa (PACE-B) trial, acute grade ≥ 2 GU or GI toxicities did not differ significantly between conventional fractionation and ultra- HFX [671]. At two years, treatment was well tolerated in both arms with no differences in RTOG ≥ Grade 2 GU or GI toxicities, but clinician scoring of urinary toxicity using CTCAE and patient reported Expanded Prostate Cancer Index Composite (EPIC)-26 urinary bother scores were both higher in the SBRT arm suggesting SBRT may increase moderate but not severe urinary symptoms post-teatment [672]. Adopting planning dose constraints to the penile bulb might minimise ED, especially in younger patients (Table 6.1.8) [673].

First results of a small (n = 30) randomised phase-II trial in intermediate-risk PCa of ‘ultra-high single dose RT’ (SDRT) with 24 Gy compared with an ultra HFX stereotactic body RT regime with 5x9 Gy, have been published [674].

Table 6.2.8: Selected trials on ultra-hypofractionation for intact localised PCa

Study

n

med FU (mo)

Risk-Group

Regimen (TD/fx)

Outcome

Widmark et al. 2019
HYPO-RT-PC [668]

1,200

60

89% intermediate

11% high

78 Gy / 39 fx, 8 wk.

42.7 Gy / 7 fx, 2.5 wk.

No SBRT

FFS at 5 yr.

84% in both arms

Brand et al. 2019

PACE-B [671]

847

variable

8% low

92% intermediate

78 Gy / 39 fx, 8 wk.

36.25 Gy / 5 fx, 1-2 wk.

SBRT

No difference in acute toxicity

Grade ≥ 2 2-year GI

3% vs. 2%, p =ns

Grade ≥ 2 2-year GU

2% vs. 3%, p =ns

FFS = failure-free survival; FU = follow-up; fx = number fractions; GI = gastro-intestinal toxicity; GU = genitourinary toxicity; mo. = months; n = number of patients; TD = total dose; SBRT = stereotactic body radiotherapy;wk. = weeks; yr. = years; ns=not significant.

6.2.3.1.4. Neoadjuvant or adjuvant hormone therapy plus radiotherapy

The combination of RT with luteinising hormone releasing hormone (LHRH) ADT has superiority compared with RT alone followed by deferred ADT on relapse, as shown by phase III RCTs [675-685] (Table 6.1.9). The main message is that for intermediate-risk disease a short duration of four to six months is optimal while a longer one, 2-3 years, is needed for high-risk patients. The largest RCT in intermediate risk disease comparing dose escalated RT with or without six months of ADT failed to demonstrate an OS advantage with a median follow-up time of 6.3 years. Six months of ADT use was associated with reduced PSA failure, fewer distant metastases and improved prostate cancer specific mortality [685].

Table 6.2.9: Selected studies of use and duration of ADT in combination with RT for PCa

Study

TNM stage

n

Trial

ADT

RT

Effect on OS

RTOG 85-31 2005 [676]

T3 or N1 M0

977

EBRT ± ADT

Orchiectomy or LHRH agonist 15% RP

65–70 Gy

Significant benefit for combined treatment

(p = 0.002) seems to be mostly caused by patients with ISUP grade group 2-5

RTOG 94-13 2007 [680]

T1c–4 N0–1

M0

1,292

ADT timing comparison

2 mo.

neoadjuvant

plus concomitant vs.

4 mo. adjuvant

suppression

Whole pelvic

RT vs.

prostate

only; 70.2 Gy

No significant difference between neoadjuvant plus concomitant vs. adjuvant androgen suppression therapy groups (interaction suspected)

RTOG 86-10 2008 [677]

T2–4 N0–1

456

EBRT ± ADT

Goserelin plus

flutamide

2 mo. before, plus

concomitant

therapy

65–70 Gy RT

No significant difference at 10 yr.

D’Amico AV, et al. 2008 [678]

T2 N0 M0 (localised unfavourable risk)

206

EBRT ± ADT

LHRH agonist plus flutamide for 6 mo.

70 Gy 3D-CRT

Significant benefit that may pertain only to men with no or minimal co-morbidity

(HR: 0.55, 95% CI: 0.34-0.90, p = 0.01)

RTOG 92-02 2008 [681]

T2c–4 N0–1 M0

1554

Short vs. prolonged ADT

LHRH agonist

given for 2 yr.

as adjuvant

after 4 mo. as

neoadjuvant

65–70 Gy

p = 0.73,

p = 0.36 overall; significant benefit

(p = 0.044) (p = 0.0061)

in subset with ISUP grade 4–5

EORTC 22961 2009 [682]

T1c–2ab N1 M0, T2c–4 N0–1 M0

970

Short vs. prolonged ADT

LHRH agonist for 6 mo. vs. 3 yr.

70 Gy 3D-CRT

Better result with 3 yr. treatment than with
6 mo. (3.8% improvement in survival at 5 yr.)

EORTC 22863 2010 [675]

T1–2 poorly differentiated and M0, or T3–4 N0–1 M0

415

EBRT ± ADT

LHRH agonist for 3 yr. (adjuvant)

70 Gy RT

Significant benefit at 10 yr. for combined treatment (HR: 0.60, 95% CI: 0.45–0.80,

p = 0.0004).

TROG 96-01 2011 [679]

T2b–4 N0 M0

802

Neoadjuvant ADT Duration

Goserelin plus flutamide 3 or 6 mo. before, plus concomitant suppression

66 Gy 3D-CRT

No significant difference in OS reported; benefit in PCa-specific survival

(HR: 0.56, 95% CI:

0.32–0.98, p = 0.04)

(10 yr.: HR: 0.84,
0.65–1.08, p = 0.18)

RTOG 99-10 2015 [683]

intermediate risk

94% T1–T2, 6% T3–4

1,579

Short vs. prolonged ADT

LHRH agonist 8 + 8 vs. 8 + 28 wk.

70.2 Gy 2D/3D

67 vs. 68%, p = 0.62, confirms 8 + 8 wk.

LHRH as a standard

PCSIII

2020 [684]

Intermediate risk

600

76 Gy alone vs.

76 Gy + ADT

vs.

70 Gy + ADT

LHRH + bicalutamide

6 mo.

4 mo. prior to RT

70 vs. 76 Gy

Significantly improved biochemical failure-free and PCa-specific survival for ADT arms, with no difference in OS.

RTOG 0815

2023 [685]

Intermediate risk

1,492

Dose escalated RT ± ADT

LHRH agonist/

antagonist + bicalutamide or flutamide

6 mo.

2 mo.prior to RT

79.2Gy (89%)

45Gy + BT boost

(11%)

No difference in OS.

Significantly improved biochemical failure-free, metastatic-free survival and PCa-specific survival for ADT arm.

ADT = androgen deprivation therapy; CI = confidence interval; EBRT = external beam radiotherapy in standardfractionation; HR = hazard ratio; ISUP = International Society of Urological Patholohy; LHRH = luteinising hormone-releasing hormone; mo. = months; n = number of patients; OS = overall survival; RP = radical prostatectomy;RT = radiotherapy; BT = brachytherapy; wk = week; yr. = year; 3D-CRT = three-dimensional conformal radiotherapy.

The question of the added value of EBRT combined with ADT has been clarified by 3 RCTs. All showed a clear benefit of adding EBRT to long-term ADT (Table 6.1.10).

Table 6.2.10: Selected studies of ADT in combination with, or without, RT for PCa

Study

TNM stage

n

Trial design

ADT

RT

Effect on OS

SPCG-7/SFUO-3 2016 [686]

T1b-2 WHO Grade 1-3, T3 N0 M0

875

ADT ± EBRT

LHRH agonist for 3 mo. Plus continuous flutamide

70 Gy 3D-CRT vs. no RT

34% (95% CI: 29-39%) vs. 17% (95% CI: 13-22% CSM at 12 (15) yr. favouring combined treatment (p < 0.0001 for 15-yr. results) NCIC CTG PR.3/MRC

PRO7/NCIC 2015 [687]

T3-4 (88%), PSA > 20 ng/mL (64%), ISUP grade group 4-5 (36%) N0 M0

1,205

ADT ± EBRT

Continuous LHRH agonist

65–70 Gy 3D-CRT vs. no RT

10-yr. OS = 49% vs. 55% favouring combined treatment HR: 0.7, p < 0.001)

Sargos, et al., 2020 [688]

T3-4 N0 M0

273

ADT ± EBRT

LHRH agonist for 3 yr.

70 Gy 3D-CRT vs. no RT

Significant reduction of clinical progression; 5-yr. OS 71.4% vs. 71.5%

ADT = androgen-deprivation therapy; CSM = cancer-specific mortality; EBRT = external beam radiotherapy;HR = hazard ratio; LHRH = luteinising hormone-releasing hormone; mo. = months; n = number of patients;OS = overall survival; PSA = prostate-specific antigen; RT = radiotherapy; 3D-CRT = three-dimensional conformal radiotherapy; yr = years.

6.2.3.1.5. Combined dose-escalated radiotherapy and androgen-deprivation therapy

The combination of ADT with various forms of RT has been extensively studied, with extremely strong evidence for the use of such combined modality therapy in several settings. The MARCAP (Individual Patient Data Meta- Analysis of Randomised Trials in Cancer of the Prostate) consortium recently conducted a meta-analysis of trials using individual patient data (IPD), and a primary endpoint of MFS, a validated surrogate for OS. Trials were eligible if they studied the use or prolongation of ADT in patients receiving definitive RT, and included 12 trials with 10,853 patients. Median follow-up was over 11 years. The use of ADT was clearly associated with significant improvements in BCR, metastatic recurrence, MFS, and OS. The benefits of ADT were independent of RT dose, age, and risk groups comparing NCCN unfavourable intermediate-risk (see Sections 4.2 and 6.2.2.3), high-risk and locally-advanced disease. There were no demonstrable benefits from the extension of duration of neoadjuvant ADT [689].

Three RCTs have shown that the benefits of ADT are independent of dose escalation, and that the use of ADT would not compensate for a lower RT dose:

1.The GICOR study shows a better biochemical DFS in high-risk patients for 3D-CRT radiation dose > 72 Gy when combined with long-term ADT [690].

2.DART01/05 GICOR shows improved OS in high-risk patients after ten years if two years of adjuvant ADT is combined with high-dose RT [691].

3.The European Organisation for Research and Treatment of Cancer (EORTC) trial 22991 shows that six months ADT improves biochemical and clinical DFS irrespective of the dose (70, 74, 78 Gy) in intermediate-risk and low-volume high-risk localised PCa patients [692].

A meta-analysis based on IPD from two RCTs (RTOG 9413 and Ottawa 0101) has compared neoadjuvant/concomitant vs. adjuvant ADT (without substratifying between favourable- and unfavourable intermediaterisk disease) in conjunction with prostate RT and reported superior PFS with adjuvant ADT, but the data heterogeneity means that this observation is hypothesis-generating only [693].

In addition, a Canadian two-arm dose-escalated (76 Gy) RCT compared neoadjuvant and concomitant with adjuvant short-term ADT in 432 patients with intermediate-risk PCa. After ten years no significant difference in OS or RT-related grade ≥ 3 GI or GU toxicity was seen [694]. Therefore both regimen in combination with dose escalation are reasonable standards.

6.2.3.2. Proton beam therapy

In theory, proton beams are an attractive alternative to photon-beam RT for PCa, as they deposit almost all their radiation dose at the end of the particle’s path in tissue (the Bragg peak), in contrast to photons which deposit radiation along their path. There is also a very sharp fall-off for proton beams beyond their deposition depth, meaning that critical normal tissues beyond this depth could be effectively spared. In contrast, photon beams continue to deposit energy until they leave the body, including an exit dose.

One RCT on dose escalation (70.2 vs. 79.2 Gy) has incorporated protons for the boost doses of either 19.8 or 28.8 Gy. This trial shows improved outcome with the higher dose but it cannot be used as evidence for the superiority of proton therapy [645]. Thus, unequivocal information showing an advantage of protons over IMRT photon therapy is still not available. Studies from the SEER database and from Harvard describing toxicity and patient-reported outcomes do not point to an inherent superiority of protons [695,696]. In terms of longer-term GI toxicity, proton therapy might even be inferior to IMRT [696].

A RCT comparing equivalent doses of proton-beam therapy with IMRT is underway. Meanwhile, proton therapy must be regarded as an experimental alternative to photon-beam therapy.

6.2.3.3. Spacer during external beam radiation therapy

Biodegradable spacer insertion involves using a liquid gel or balloon to increase the distance between the prostate and rectum and consequently reduce the amount of radiation reaching the rectum. Various materials have been used with most evidence available for CE-marked hydrogel spacers [697]. A meta-analysis including one RCT and six cohort studies using the hydrogel spacer demonstrated a 5–8% reduction in the rectal volume receiving high-dose radiation, although heterogeneity between studies is found [698]. In the final analysis of the RCT with a median follow-up of 37 months and with approximately two-thirds of patients evaluable, those treated with spacer in situ had no deterioration from baseline bowel function whilst those treated without spacer had a lower mean bowel summary score of 5.8 points which met the threshold for a minimally important difference of 4–6 points [699].

This meta-analysis highlights inconsistent reporting of procedural complications. In addition, with more widespread clinical use safety reports describe uncommon, but severe and life changing, complications including prostatic abscess, fistulae and sepsis [700]. Implantation is associated with a learning curve and should only be undertaken by teams with experience of TRUS and transperineal procedures with robust audit reporting in place [701]. Its role in the context of moderate or extreme HFX is as yet unclear.

6.2.3.4. Brachytherapy
6.2.3.4.1. Low-dose rate brachytherapy

Low-dose rate (LDR) BT uses radioactive seeds permanently implanted into the prostate. In patients declining or unsuitable for AS LDR monotherapy [702] can be offered to those with low-risk or NCCN favourable intermediate-risk and good urinary function defined as an International Prostatic Symptom Score (IPSS) < 12 and maximum flow rate > 15 mL/min on urinary flow tests [703]. The RTOG Ph3 RCT compared LDR BT +/- EBRT in participants with Gleason grade 6 and PSA < 20 or Gleason grade 7 and PSA < 10 and found that the addition of EBRT resulted in increased toxicity but no improvement in freedom from progression [704].

Patients having had a previous TURP can undergo BT without an increase in risk of urinary toxicity with due attention to dose distribution. A minimal channel TURP is recommended, leaving at least 1 cm rim of prostate tissue around the post-TURP urethral defect at the postero-lateral sides of the prostate and there should be at least a 3-month interval between TURP and BT to allow for adequate healing [705-708].

The only available RCT comparing RP and LDR BT as monotherapy was closed due to poor accrual [709]. Outcome data are available from a number of large population cohorts with mature follow-up [710-714]. The biochemical DFS for ISUP grade group 1 patients after five and ten years has been reported to range from 71% to 93% and 65% to 85%, respectively [710-714]. A significant correlation has been shown between the implanted dose and biochemical control [715]. A D90 (dose covering 90% of the prostate volume) of > 140 Gy leads to a significantly higher biochemical control rate (PSA < 1.0 ng/mL) after four years (92 vs. 68%). There is no OS benefit in adding neoadjuvant or adjuvant ADT to LDR monotherapy [716].

Low-dose rate BT can be combined with EBRT in NCCN unfavourable intermediate-risk PCa and high-risk patients. External beam RT (total dose of 78 Gy) has been compared with EBRT (total dose 46 Gy) followed by LDR BT boost (prescribed dose 115 Gy) in intermediate-risk and high-risk patients in the ASCENDE-RT randomised trial with twelve months of ADT in both arms [717,718]. The LDR boost resulted in 5-, 7-year and 10-year PSA PFS increase (89%,86% and 85% respectively, compared to 84%,75%, 70%) but with no impact on distant metastasis or OS. This improvement in biochemical control was achieved at a cost of increased late grade 3+ GU toxicity (18% compared to 8%) and 2 treatment related deaths [718,719]. Urinary toxicity was mainly in the development of urethral strictures and incontinence and great care should be taken during treatment planning.

6.2.3.4.2. High-dose rate brachytherapy

High-dose rate (HDR) BT uses a radioactive source temporarily introduced into the prostate to deliver radiation. The technical differences are outlined in Table 6.1.11. The use of the GEC (Groupe Europeen de Curietherapie)/ESTRO Guidelines is strongly recommended [720]. High-dose rate BT can be delivered in single or multiple fractions and is often combined with EBRT of at least 45 Gy, conventionally fractionated [721]. A retrospective analysis on 1641 intermediate and high-risk patients demonstrated a better distant-metastasis free survival when a HDR BT boost was added to 50 – 54 Gy EBRT. The difference mounted to 12% at ten years [722]. A SR of non-RCTs and data from population studies suggest outcomes with EBRT plus HDR BT are superior to EBRT alone [723,724].

A single-centre RCT of EBRT (55 Gy in 20 fractions) vs. EBRT (35.75 Gy in 13 fractions), followed by HDR BT (17 Gy in two fractions over 24 hours) has been reported [725]. In 218 patients with T1–3 N0M0 PCa the combination of EBRT and HDR BT showed a significant improvement in the biochemical disease-free rate (p = 0.04) at five and ten years (75% and 46% compared to 61% and 39%). However, an unexpectedly high rate of early recurrences was observed in the EBRT arm alone, even after 2 years, possibly due to a dose lower than the current standard used [725].

Supporting, but not definitive, evidence of the benefit of HDR boost is available from the TROG 03.04 RADAR trial. This multi-centre study had upfront radiation dose escalation (non-randomised) with dosing options of 66, 70, or 74 Gy EBRT, or 46 Gy EBRT plus HDR BT boost and randomised men with locally-advanced PCa to 6 or 18 months ADT. After a minimum follow-up of ten years HDR boost significantly reduced distant progression, the study primary endpoint (sub HR: 0.68, 95% CI: 0.57–0.80; p < 0.0001), when compared to EBRT alone and, independent of duration of ADT, HDR boost was associated with increased IPSS of 3 points at 18 months post-treatment resolving by three years but decreased rectal symptoms when compared to EBRT [726]. Although radiation dose escalation using BT boost provides much higher biological doses, the TROG 03.04 RADAR RCT and SRs show ADT use independently predicts better outcomes regardless of radiation dose intensification [716,726,727]. Omitting ADT may result in inferior OS and based on current evidence ADT use and duration should be in line with that used when delivering EBRT alone.

Fractionated HDR BT as monotherapy can be offered to patients with low- and intermediate-risk PCa, who should be informed that results are only available from limited series in very experienced centres. Five-year PSA control rates of 97.5% and 93.5% for low- and intermediate-risk PCa, respectively, are reported, with late grade 3+ GU toxicity rates < 5% and no, or very minimal, grade 3+ GI toxicity rates [728]. Single fraction HDR monotherapy should not be used as it has inferior biochemical control rates compared to fractionated HDR monotherapy [729].

Table 6.2.11: Difference between LDR and HDR brachytherapy


Differences in prostate brachytherapy techniques

Low dose rate (LDR)

Permanent seeds implanted

Uses Iodine-125 (I-125) (most common), Palladium-103 (103Pd-) or Cesium-131 isotopes

Radiation dose delivered over weeks and months

Acute side effects resolve over months

Radiation protection issues for patient and carers

High dose rate (HDR)

Temporary implantation

Iridium-192 (IR-192) isotope introduced through implanted needles or catheters

Radiation dose delivered in minutes

Acute side effects resolve over weeks

No radiation protection issues for patient or carers

6.2.3.5. Acute side effects of external beam radiotherapy and brachytherapy

Gastro-intestinal and urinary side effects are common during and after EBRT. In the EORTC 22991 trial, approximately 50% of patients reported acute GU toxicity of grade 1, 20% of grade 2, and 2% grade 3. In the same trial, approximately 30% of patients reported acute grade 1 GI toxicity, 10% grade 2, and less than 1% grade 3. Common toxicities included dysuria, urinary frequency, urinary retention, haematuria, diarrhoea, rectal bleeding and proctitis [730]. In addition, general side effects such as fatigue are common. It should be noted that the incidence of acute side effects is greater than that of late effects (see Section 8.2.2.1), implying that most acute effects resolve. In a RCT of conventional dose EBRT vs. EBRT and LDR BT the incidence of acute proctitis was reduced in the BT arm, but other acute toxicities were equivalent [717]. Acute toxicity of HDR BT has not been documented in a RCT, but retrospective reports confirm lower rates of GI toxicity compared with EBRT alone and grade 3 GU toxicity in 10%, or fewer, patients, but a higher incidence of urinary retention [731]. Similar findings are reported using HFX; in a pooled analysis of 864 patients treated using extreme HFX and stereotactic RT, declines in urinary and bowel domains were noted at three months which returned to baseline, or better, by six months [732].

6.2.4. Investigational therapies

6.2.4.1. Background

Besides RP, EBRT and BT, other modalities have emerged as potential therapeutic options in patients with clinically localised PCa [733-735]. These new modalities have been developed as minimally invasive procedures with the aim of providing equivalent oncological safety, reduced toxicity, and improved functional outcomes. In this section, both whole gland- and focal treatment [736,737] will be considered, looking particularly at high-intensity focused US (HIFU), cryotherapeutic ablation of the prostate (cryotherapy) and focal photodynamic therapy (PDT), as sufficient data are available to form the basis of some initial judgements. Other options such as radiofrequency ablation (RFA) and electroporation, among others, are considered to be in the early phases of evaluation [736].

6.2.4.2. Whole-gland therapies
6.2.4.2.1. Cryotherapy for whole-gland treatment

Cryotherapy uses freezing techniques to induce cell death by dehydration resulting in protein denaturation, direct rupture of cellular membranes by ice crystals and vascular stasis and microthrombi, resulting in stagnation of the microcirculation with consecutive ischaemic apoptosis [733-735]. Freezing of the prostate is ensured by the placement of 17-gauge cryo-needles under TRUS guidance, placement of thermosensors at the level of the external sphincter and rectal wall, and insertion of a urethral warmer. Two freeze-thaw cycles are used under TRUS guidance resulting in a temperature of -40°C in the mid-gland and at the neurovascular bundle. Currently, third and fourth generation cryotherapy devices are mainly used. Since its inception, cryotherapy has been used for whole-gland treatment in PCa either as a primary or salvage treatment option.

The main adverse effects of whole-gland cryosurgery are ED (18%), urinary incontinence (2–20%), urethral sloughing (0–38%), rectal pain and bleeding (3%) and recto-urethral fistula formation (0–6%) [738]. There is a lack of prospective comparative data regarding oncological outcomes of whole-gland cryosurgery as a curative treatment option for men with localised PCa, with most studies being non-comparative single-arm case series with short follow-up [738].

6.2.4.2.2. High-intensity focused ultrasound for whole-gland treatment

High-intensity focused US consists of focused US waves emitted from a transducer that cause tissue damage by mechanical and thermal effects as well as by cavitation [739]. The goal of HIFU is to heat malignant tissue above 65°C, so that it is destroyed by coagulative necrosis. High-intensity focused US is performed under general or spinal anaesthesia, with the patient lying in the lateral or supine position. Since the ultrasound energy is most often delivered from the rectal cavity, HIFU faces challenges in delivering energy to the anterior part in large prostates.

High-intensity focused US has previously been widely used for whole-gland therapy with the following adverse effects: acute urinary retention (10%), ED (23%), urethral stricture (8%), rectal pain or bleeding (11%), recto-urethral fistula (0–5%) and urinary incontinence (10%) [738]. Combining the whole-gland HIFU treatment with TUR-P reduces the rate of urethral strictures, maintains the level of incontinence, but increases the rate of ED [740].

Overall, the lack of any long-term prospective comparative studies, and data to suggest poor long-term oncological outcomes for men with high-risk localised disease [741] prevents whole-gland HIFU from being considered as a reasonable alternative to the established curative treatment options [738]. In addition, the expected improvements in functional outcome failed to materialise with 12% of patient developing incontinence and 61% developing ED [742].

6.2.4.3. Focal therapy

During the past two decades, there has been a trend towards earlier diagnosis of PCa as a result of greater public and professional awareness leading to the adoption of both formal and informal screening strategies. The effect of this has been that men are identified at an earlier stage with smaller tumours, with a greater propensity for unifocal disease potentially suitable for focal therapy [743-745]. There is also greater awareness of the risks of the consequences of treatment leading to attempts to ablate only a region of the prostate containing the tumour thereby limiting toxicity by sparing the neurovascular bundles, sphincter, and urethra [746-748]. The question remains which if any of these small unifocal tumours need treatment.

A SR included data from 5,827 patients across 72 studies and covered different energy sources including HIFU, cryotherapy, PDT, laser interstitial thermotherapy, focal BT, irreversible electroporation (IRE) and radiofrequency ablation (RFA) [749]. The review favours HIFU and PDT for their higher quality data, over 95% of pad-free incontinence and 85–90% of patients without clinically significant cancer in short-term analysis. This has to be critically analysed, because 45% of all patients with a focal approach included in this SR had an ISUP Grade Group 1 cancer. The overall quality of the evidence was low, due to the majority of studies being single-centre, non-comparative and retrospective in design, heterogeneity of definitions and approaches, follow-up strategies, outcomes, and duration of follow-up. Although the review finds high quality evidence that focal therapy has favourable functional outcomes and minimises AEs, definitive proof of oncological effectiveness of focal therapy compared to standard treatments remains unavailable.

The currently largest analysis on oncologic outcomes following focal HIFU includes 625 patients, with 70% having ISUP grade group 2/3 disease, followed for five years with an 88% failure-free survival (FFS), defined as the need for salvage treatment or systemic therapy [750]. In this study one repeated focal HIFU session was allowed and performed in 25% of all patients. Follow-up was driven by PSA and clinics, with re-biopsies performed only in 36% of patients after a significant PSA rise and suspicious MRI.

The guideline panel acknowledges the challenges for interventional RCTs [751-753]. The interim analysis and meeting reports demonstrate slow recruitment, patients declining consent and rejecting their treatment allocation into the RP group. In an attempt to overcome this propensity-matched analysis using prospective multi-centre databases have been performed for comparison of focal therapy vs. radical therapy [754,755]. Such analyses are always susceptible to unmeasured selection biases in who was selected for each treatment.

Oncological follow-up data up to 8 years can be used to counsel patients in treatment decisions [754]. Patients were managed by focal therapy had a HIFU or cryotherapy, with one retreatment, if needed. 17.1% of patients in the focal arm received a retreatment. The primary outcome was FFS defined as “need for local or systemic salvage treatment or metastasis”. Both groups included 246 patients with an average PSA of 7.9 ng/mL and 60% ISUP Grade Group 2/3 cancers. The cancer core length was 5–6 mm with 45% having bilateral cancer. The authors report similar cancer control 8 years after therapy, with FFS and BCR of 83% and 23.9% for focal therapy vs. 79% and 24.8% for RP, respectively. Similar results were demonstrated in a cohort-based analysis with a follow-up six years [755]. The use of different definitions for oncological failure in the two arms is another limitation of these studies. While any recurrence after RP was seen as failure, a second HIFU was permitted in the focal group. The current data from the HIFU Evaluation and Assessment of Treatment (HEAT) registry indicates that a repeat-HIFU does not significantly decrease urinary or erectile function [756]. However, this change of failure definition will have to be re-evaluated. It is important to note, that these results were achieved in centres with a dedicated focal program where all patients had a mpMRI with targeted and systematic biopsies or full template mapping biopsies.

The prospective HEAT registry analysed over 800 men undergoing focal HIFU for localised PCa [756]. The functional data indicate low treatment-related toxicity with less than 4% decrease in pad-free incontinence and a reduction in IIEF of 0.4 points. The impact of salvage therapies after focal therapy was investigated in small series [757,758]. If a salvage RP is necessary, the reported functional and oncological outcomes are comparable to treatment-naive patients [802,803].

One comparative RCT was conducted in a very-low risk population, for which there is currently a strong movement away from any form of active treatment. This study was comparing padeliporfin-based vascular targeted PDT vs. AS and found at a median follow-up of 24 months that less patients progressed in the PDT arm compared with the AS arm (adjusted HR: 0.34, 95% CI: 0.24–0.46), and needed less radical therapy (6% vs. 29%, p < 0.0001). Updated results were published in 2018 showing that these benefits were maintained after four years [759]. Nevertheless, limitations of the study include an unusually high observed rate of disease progression in the AS arm (58% in two years) and more patients in the AS arm chose to undergo radical therapy without a clinical indication which may have introduced confounding bias. Finally, the AS arm did not undergo any confirmatory biopsy or any MRI scanning, which is not representative of contemporary practice. A matched-pair analysis comparing focal cryo therapy to AS with 76% ISUP grade group 1 cancers failed to demonstrate any significant advantages for MFS and OS [760].

The available evidence indicates that focal therapy is associated with less AEs than whole gland or radical treatments. Many of the patients included in these trials would currently be considered to have been over treated. Robust prospective trials reporting standardised 15-year oncological outcomes [761] are needed in patients with clinically significant cancers before unrestricted recommendations in support of focal therapy for routine clinical practice can be made [736,750,761]. Currently, focal therapy using HIFU or cryotherapy should be performed within the context of a prospective registry. All other ablative modalities and treatment strategies should only be offered in well-designed prospective trial setting. In order to allow quality analysis of the collected data, the prospective registry should adhere to the EMA recommendations (Guideline on registry-based studies EMA/426390/2021), which emphasises the need for clear follow-up timelines and timely recording, completeness of core data of consecutive patients enrolled, an analysis plan in defined intervals and a data quality management. In near future the EAU will offer a quality registry within the PIONEER network.

6.2.5. General guidelines for the treatment of prostate cancer*

Recommendations

Strength rating

Offer a watchful waiting (WW) policy to asymptomatic patients with clinically localised disease and with a life expectancy < ten years (based on comorbidities and age).

Strong

No active treatment modality has shown superiority over any other active management options or deferred active treatment in terms of overall- and PCa-specific survival for clinically localised low/intermediate-risk disease.

Strong

Inform patients that all local treatments have side effects.

Strong

Surgical treatment

Inform patients that no surgical approach (open-, laparoscopic- or robotic RP) has clearly shown superiority in terms of functional or oncological results.

Weak

Consider avoiding nerve-sparing surgery when there is a risk of ipsilateral ex-tra-capsular extension (based on cT stage, ISUP grade group, magnetic reso-nance imaging, or with this information combined in a nomogram).

Weak

In patients undergoing a lymph node dissection you should perform an extended PLND.

Strong

Do not offer neoadjuvant androgen deprivation therapy before surgery.

Strong

Radiotherapeutic treatment

Offer intensity-modulated radiation therapy (IMRT) or volumetric arc radiation therapy (VMAT) plus image-guided radiation therapy (IGRT) for definitive treatment of PCa by external-beam radiation therapy.

Strong

Offer moderate hypofractionation (HFX) with IMRT/VMAT plus IGRT to the prostate to patients with localised disease (60 Gy/20 fractions in 4 weeks or 70 Gy/28 fractions in 6 weeks).

Strong

Offer low-dose rate (LDR) brachytherapy monotherapy to patients with good urinary function and NCCN favourable intermediate-risk disease.

Strong

Offer LDR or high-dose rate (HDR) brachytherapy boost combined with IMRT/VMAT plus IGRT to patients with good urinary function and NCCN unfa-vourable intermediate-risk or high-risk disease and/or locally-advanced disease.

Weak

Active therapeutic options outside surgery or radiotherapy

Offer focal therapy with HIFU or cryotherapy within a clinical trial or prospective registry.

Strong

*All recommendations are based on conventional imaging with isotope bone scan and CT/MR abdomen/pelvis.

Summary of evidence

LE

The AS strategy should be based on PSA (at least once every six months), serial DRE (at least once yearly) and repeated biopsy. Serial DRE may be omitted if MRI is stable.

3

Serial MRI can improve the detection of aggressive cancers during AS.

A progression on MRI mandates a repeat biopsy before a change in treatment strategy.

A stationary MRI does not make repeat biopsy superfluous (patients with low-risk tumour and a stable PSA-D < 0.15 may be excepted).

Recommendations

Strength rating

Base the strategy of active surveillance (AS) on a strict protocol including digital rectal examination (at least once yearly), prostate-specific antigen (PSA) (at least once every six months) and repeated biopsy every 2 to 3 years.

Strong

Patients with a low risk PCa, a stable MRI (PRECISE 3) and a stable, low PSA density
(< 0.15) may be excused from repeat biopsy.

Weak

Perform magnetic resonance imaging (MRI) and repeat biopsy if PSA is rising (PSA-doubling time < 3 years).

Strong

Base change in treatment on biopsy progression, not on progression on MRI and/or PSA.

Weak

Re-classify patients with low-volume ISUP grade group 2 disease included in AS protocols,
if repeat non-MRI-based systematic biopsies performed during monitoring reveal > 3 positive cores or maximum core involvement > 50%/core of ISUP grade group 2 disease.

Weak

6.3. Treatment by disease stages

6.3.1. Treatment of low-risk disease

6.3.1.1. Active surveillance

The main risk for men with low-risk disease is over-treatment (see sections 6.1.1, 6.2.1); AS should therefore be considered SOC for all such patients with a life expectancy > ten years based on comorbidities and age) and where curative treatment would be considered in the case of disease progression.

6.3.1.1.1. Active surveillance - inclusion criteria

Guidance regarding selection criteria for AS is limited by the lack of data from prospective RCTs. As a consequence, the Panel undertook an international collaborative study involving healthcare practitioners and patients to develop consensus statements for deferred treatment with curative intent for localised PCa, covering all domains of AS (DETECTIVE Study) [330], as well as a formal SR on the various AS protocols [491]. The criteria most often published include: ISUP grade group 1, clinical stage cT1c or cT2a, PSA < 10 ng/mL and PSA-D < 0.15 ng/mL/cc, as based on systematic biopsy schemes [487,492]. The latter threshold remains controversial [492,493]. These criteria were supported by the DETECTIVE study consensus. There was no agreement on the maximum number of systematic cores that can be involved with cancer or the maximum percentage core involvement (CI), although there was recognition that extensive disease on MRI should exclude men from AS, even though there is no firm definition on this, especially when targeted biopsies confirm ISUP grade group 1 [330]. A SR and meta-analysis found three clinico-pathological variables which were significantly associated with reclassification, high PSA-D, > 2 positive cores (on systematic biopsies) and African-American descent [495]. In addition, a previous pathology consensus group suggested excluding men from AS when any of the following features were present: predominant ductal carcinoma (including pure IDC), cribriform histology, sarcomatoid carcinoma, small cell carcinoma, EPE or LVI in needle biopsy [497] and perineural invasion [498].

A multi-disciplinary consensus conference on germline testing attempted to develop a genetic implementation framework for the management of PCa [154]. Based on consensus, BRCA2-gene testing was recommended for AS discussions and could be performed in men with family history of prostate, breast or ovarian cancers. However, the nature of such discussions and how a positive result influences management were beyond the scope of the project. Currently, BRCA2 mutation does not exclude a patient from AS if tumour factors are otherwise favourable. Furthermore, if included in AS programmes, patients with a known BRCA2 mutation should be cautiously monitored until such time that more robust data are available.

6.3.1.1.2. Tissue-based prognostic biomarker testing for selection for active surveillance

Biomarkers, including Oncotype Dx®, Prolaris®, Decipher®, PORTOS and ProMark® are promising (see Section 5.2.8.3). However, further data will be needed before such markers can be used in standard clinical practice [220].

6.3.1.1.3. Magnetic resonance imaging for selection for active surveillance

In men eligible for AS based upon systematic biopsy findings alone who did not have a pre-biopsy MRI, a re-biopsy within 6–12 months (usually referred to as ‘confirmatory biopsy’) seems mandatory to exclude sampling error. A large body of literature including two RCTs showed that adding MRI-targeted biopsy to systematic sampling at confirmatory biopsy improved detection of ISUP grade ≥ 2 cancers and thus, patient selection for AS [121,499,500,502-504]. Adding MRI-targeted biopsy to systematic sampling at confirmatory biopsy improved upgrade detection by increments of 0-7.9 per 100 men depending on the series [499]. In a meta-analysis of 6 studies, the rate of upgrading to ISUP grade group ≥ 2 cancer increased from 20% (95% CI: 16–25%) to 27% (95% CI: 22–34%) when MRI-targeted biopsy was added to systematic biopsy [504]. The Active Surveillance MRI Study (ASIST) randomised men on AS scheduled for confirmatory biopsy to either 12-core systematic biopsy or to MRI with targeted biopsy (when indicated), combined with systematic biopsy (up to 12 cores in total). After two yearsof follow-up, use of MRI before confirmatory biopsy resulted in fewer failures of surveillance (19% vs. 35%, p = 0.017) and in fewer patients progressing to ISUP grade group ≥ 2 cancer (9.9% vs. 23%, p = 0.048) [502]. However, systematic biopsy retains its additional value, which argues for a combined biopsy approach [499,504]. The DETECTIVE study agreed that men eligible for AS after combined systematic- and MRI-targeted biopsy do not require a confirmatory biopsy [330].

If the PCa diagnosis is made on MRI-targeted biopsy alone (recommended in some countries national guidelines, e.g., the Nordic countries [762] in order to lower the risk of over-detection of insignificant tumours, a confirmative systematic biopsy should be performed before definite decision of AS to rule out more widespread cancer growth in the prostate.

6.3.1.1.4. Follow-up during active surveillance

Based on the DETECTIVE consensus study, the follow-up strategy should be based on serial DRE (at least once yearly), PSA (at least once, every six months) and repeated biopsy. It was also agreed that PSA progression or change in PSA kinetics alone should lead to reclassification only if accompanied by changes in histology on repeat biopsy [330].

Yerram et al., analysed a prospectively-maintained AS cohort of 369 patients (272 with ISUP grade group 1 cancer and 97 with ISUP grade group 2 cancer) who had been selected for AS after combined systematic and MRI-targeted sampling during confirmatory biopy. At two years, systematic biopsy, MRI-targeted biopsy and combined biopsy detected grade progression in 44 patients (15.9%), 73 patients (26.4%) and 90 patients (32.5%), respectively. This suggests that both biopsy approaches retain added value, not only for confirmatory biopsy, but also during AS [763].

In 2016, the Prostate Cancer Radiological Estimation of Change in Sequential Evaluation (PRECISE) criteria were established to standardise the assessment of tumour progression on serial MRI [507]. Progression on MRI, defined using the PRECISE criteria, or not, is a strong predictor of histological upgrading [508,509]. Two independent meta-analyses assessed the value of MRI progression criteria for predicting histological progression (mostly defined as progression to ISUP grade group ≥ 2). The pooled histological progression rate was 27% in both reviews. If biopsies were triggered only by MRI progression findings, approximately two thirds of the biopsies would be avoided, at the cost of missing 40% of men with histological progression. In addition, at least half of biopsied men would have had negative findings for histological progression and thus would have undergone unnecessary biopsies. If histological progression was restricted to progression to ISUP grade group > 3, approximately 30% of histological progression would be missed and approximately 80% of the biopsies performed would be unnecessary. The use of the PRECISE criteria did not seem to change these results [510,511]. This supports maintaining protocol-mandated follow-up biopsies during the course of AS.

However, several factors have been found to be associated with low re-classification rates and long PFS; negative baseline or follow-up MRI [505,512-518], low PSA-D [505,513,515,518], low PSA velocity [519,520] or negative biopsy (i.e., no cancer at all) at confirmatory or follow-up biopsy [521]. In patients with stable (PRECISE 3) follow-up MRI, a low PSA-D may be associated with a low rate of progression [522]. Using these criteria, it might be possible, in the future, to create risk-based personalised AS biopsy schedules.

A Panel SR incorporating 263 surveillance protocols showed that 78.7% of protocols mandated per-protocol confirmatory biopsies within the first two yearsand that 57.7% of the protocols performed repeat biopsy at least every three years for ten years after the start of AS [491]. In another review it was concluded that a negative follow-up biopsy was associated with a 50% decrease in the risk of future reclassification and upgrading [523]. In a single-centre AS cohort of 514 patients who underwent at least three protocol-mandated biopsies after diagnosis (the confirmatory biopsy and at least two additional surveillance biopsies), men with one negative biopsy (i.e., no cancer at all) at confirmatory or second biopsy, or men with two consecutive negative biopsies had a lower likelihood of a positive third biopsy and significantly better 10-year treatmentfree survival [521]. This suggests that men with repetitive negative biopsies may pursue AS with at least less frequent untriggered biopsies.

6.3.1.1.5. Active Surveillance - change in treatment

Men may remain on AS whilst they continue to consent, have a life expectancy of > ten years and the disease remains indolent. Patient anxiety about continued surveillance occurs in around 10% of patients on AS [524] and was recognised as a valid reason for active treatment [330]. A thorough discussion on pros/cons of AS vs. active treatment already at the time of diagnosis is therefore of outmost importance. More common is the development of other comorbidities which may result in a decision to transfer to a WW strategy.

A PSA change alone (including PSA-DT < 3 years) should not change management based on its weak link with grade progression [527,528] but rather trigger further investigation. There was clear agreement in the DETECTIVE consensus meeting that a change in PSA should lead to repeat-MRI and repeat-biopsy. It was also agreed that changes on follow-up MRI needed a confirmatory biopsy before considering active treatment [330].

However, the histopathology criteria required to trigger a change in management in the targeted biopsy era remain debated. Magnetic resonance imaging-targeted biopsy induces a grade shift and ISUP grade group 2–3 cancers detected by MRI-targeted biopsy have, on average, a better prognosis than those detected by systematic sampling (see Section 5.2.4.2.6.4). As an increasing number of men with favourable intermediate-risk disease are managed with AS (see section 6.2.2.1), it seems illogical to use progression to ISUP grade group 2 based on targeted biopsies as the sole criterion for reclassification. In addition, as acknowledged in the DETECTIVE consensus meeting, the number of positive cores is not an indicator of tumour volume anymore if targeted biopsies are performed [330,529]. No agreement could be reached on the pathological criteria required to trigger a change in management during the DETECTIVE consensus meeting [330]. However, based on the findings of a SR incorporating 271 reclassification protocols, patients with low-volume ISUP grade group 2 disease at recruitment, and with increased systematic core positivity (> 3 cores involvement [> 50% per core]) on repeat systematic biopsies not using MRI, should be reclassified [491].

6.3.1.2. Alternatives to active surveillance

For patients with a life expectancy of < ten years (based on comorbidities and age), where curative treatment would not be an option in the case of progression, WW is SOC rather than AS.

In terms of alternatives to AS in the management of patients with low-risk disease there is some data from randomised studies. In the PIVOT trial (Section 6.1.1) which compared surgery vs. observation, only 42% of patients had low-risk disease [482]. Sub-group analysis revealed that for low-risk disease there was no statistically significant difference in all-cause mortality between surgery vs. observation (RR: 0.93, 95% CI: 0.78–1.11). In the ProtecT study (Section 6.1.1) which compared the less organised strategy of “active monitoring” (i.e. repeat PSA management only) vs. surgery vs. EBRT, 66% of patients had a D’Amico low-risk disease [474]. The study found, after 15 years follow-up, no difference between the three arms in terms of OS and CSS, but AM had higher metastatic progression (9.4%) compared with surgery (4.7%) or EBRT (5.0%). There are no robust data comparing contemporary AS protocols with either surgery or EBRT in patients with low-risk disease. Active surveillance should be considered SOC in patients with low- to intermediate risk disease and a life expectancy > ten years. Surgery and EBRT should only be considered as alternatives to AS in patients suitable for such treatments after thorough information on pros and cons of AS and active treatment, and who after such information refuse or for some other reason are deemed unfit for AS, and who accept a trade-off between toxicity and prevention of disease progression.

Other treatments such as whole-gland ablative therapy (e.g. cryotherapy or HIFU) or focal ablative therapy remain unproven in the setting of localised low-risk disease compared with AS or radical treatment options and should not be used outside a trial setting or well-designed prospective cohort setting. These treatments are discussed in detail in Section 6.1.5.

6.3.1.2.1. Androgen deprivation monotherapy

Data regarding the use of ADT monotherapy in men with low-risk localised disease may be inferred indirectly from the Early Prostate Cancer (EPC) Trial Programme which published its findings in 2006 [764]. The EPC programme comprises three large RCTs including 8,113 men with localised (cT1–2, N0/NxM0) or locally advanced (cT3–4, any N; or any T, N+, M0) PCa. The intervention was oral bicalutamide 150 mg monotherapy vs. placebo following standard care (defined as RP, radical EBRT or WW). The primary endpoints were PFS and OS. Patients were stratified according to clinical stage only; data regarding PSA and GS were not assessed. The authors found in patients with localised disease, ADT monotherapy did not improve PFS nor OS in any of the subgroups, compared with placebo. Instead, there was a statistically insignificant numerical trend towards worse OS with ADT in the WW sub-group (HR: 1.16, 95% CI: 0.99–1.37; p = 0.07). Although the trial did not directly address men with low-risk disease, it offered some evidence suggesting that otherwise asymptomatic men with localised disease should not receive ADT monotherapy. A phase 2 RCT addressed a similar approach, where patients were randomised to enzalutamide plus AS or AS alone. This study indicated that PSA progression could be delayed, and the odds of a negative biopsy increased during the median follow-up time of 1.3 years, but patients had more side effects of the treatment without showing any long-term benefits of the treatment [765]. Hence, there is no evidence supporting the use of any hormonal treatment in asymptomatic men with low-risk disease who are not eligible for any local/radical treatment; these men should simply be offered AS or WW alone.

6.3.1.3. Summary of evidence and guidelines for the management of low-risk disease*

Summary of evidence

LE

WW or AS is SOC, based on life expectancy.

2a

All active treatment options present a risk of over-treatment.

1a

Recommendations

Strength rating

Watchful Waiting

Manage patients with a life expectancy < ten years by watchful waiting.

Strong

Active surveillance (AS)

Manage patients with a life expectancy > ten years and low-risk disease by AS.

Strong

Selection of patients

Patients with cribriform or intraductal histology on biopsy should be excluded from AS.

Strong

Perform magnetic resonance imaging (MRI) before a confirmatory biopsy if no MRI has been performed before the initial biopsy.

Strong

Take both targeted biopsy (of any PI-RADS ≥ 3 lesion) and systematic biopsy if a confirmatory biopsy is performed.

Strong

If MRI is not available, per-protocol confirmatory prostate biopsies should be performed.

Weak

If a patient has had upfront MRI followed by systematic and targeted biopsies there is no need for confirmatory biopsies.

Weak

Strategy of surveillance

Repeat biopsies should be performed at least once every three years for ten years.

Weak

In case of prostate-specific antigen progression or change in digital-rectal exam-ination or MRI findings, do not progress to active treatment without a repeat biopsy.

Strong

6.3.2. Treatment of intermediate-risk disease

When managed with non-curative intent, intermediate-risk PCa is associated with 10-year and 15-year PCSM rates of 13.0% and 19.6%, respectively [841]. These estimates are based on systematic biopsies and may be overestimated in the era of MRI-targeted biopsies.

6.3.2.1. Active Surveillance

In the ProtecT trial, where 34% of the randomised patients had a D’Amico intermediate- or high-risk disease, there was no statistically significant difference in CSS at 15 years [474]. In the comprehensive characterisation of the patients in the ProtecT trial, treatment received, PSA, ISUP grade group at diagnosis, cT stage, risk group, number of PCa-involved biopsy cores, maximum length of tumour (median 5.0 vs. 3.0 mm), aggregate length of tumour (median 8.0 vs. 4.0 mm), and presence of perineural invasion were each associated with increased risk of disease progression (p < 0.001 for each). However, these factors could not reliably predict progression in individuals [487].

The outcomes of AS in intermediate-risk PCa has also been analysed in three SRs and meta-analyses, summarising available data on its oncological outcomes and comparing patients with intermediate-risk PCa to patients with low-risk disease [766-768]. The definition of AS was not strictly defined in either of the reviews: instead, the search strategies included ‘active surveillance’ as a search term, and no a priori study protocol was available. The primary outcome was the proportion of patients who remained on AS, whilst secondary outcomes included CSS, OS, and MFS in all three studies. In the first review 17 studies were included, incorporating 6,591 patients with intermediate risk disease. Sixteen studies included patients with low- and intermediate-risk disease, hence enabling comparative outcome assessment via pooled analysis. Only one study performed MRI at recruitment and during AS. There was significant clinical heterogeneity in terms of inclusion criteria for intermediate-risk disease. The results showed the proportion of patients who remained on AS was comparable between the low- and intermediate-risk groups after ten- and fifteen-years’ follow-up (OR: 0.97, 95% CI: 0.83–1.14; and OR: 0.86, 95% CI: 0.65–1.13, respectively). Cancer-specific survival was worse in the intermediate-risk group after ten years (OR: 0.47, 95% CI: 0.31–0.69) and 15 years (OR: 0.34, 95% CI: 0.2–0.58). Overall survival was not statistically significantly different at five years’ follow-up (OR: 0.84, 95% CI: 0.45–1.57) but was significantly worse in the intermediate-risk group after ten years (OR: 0.43, 95% CI: 0.35–0.53). Metastases-free survival did not significantly differ after five years (OR: 0.55, 95% CI: 0.2–1.53) but was worse in the intermediate-risk group after ten years (OR: 0.46, 95% CI: 0.28–0.77) [768]. The second review, including 25 studies and a total of 29,673 low- or intermediate-risk patients, showed similar results in terms of treatment-free survival at ten years (RR: 1.16, 95% CI: 0.99-1.36), risk of developing metastases (RR: 5.79, 95% CI: 4.61-7.29), risk of dying from PCa (RR: 3.93, 95% CI: 2.93-5.27) and risk of dying from any cause (RR: 1.44, 95% CI: 1.11-1.86) [766]. The third, most recent, review included 25 studies of which thirteen studies provided data on treatment free survival, six on CSS and seven on OS. Treatment free survival was not statistically significantly different in the intermediate risk group after 5 (RR: 0.92, 95% CI: 0.82-1.02), 10 (RR: 0.83, 95% CI: 0.55-1.23) or 15 years (RR: 0.54, 95% CI: 0.21-1.39). Cancer-specific survival was significantly lower after 15 years (RR: 0.92, 95% CI: 0.89-0.96) and OS was significantly lower after ten years (RR: 0.87, 95% CI: 0.82-0.93) in the intermediate risk group. It should be noted that many of the studies included patients with ISUP grade group 3 disease. When these studies were excluded no difference in treatment free, cancer specific or OS could be observed [767].

In a subgroup analysis of four studies comparing outcomes of patients with intermediate- and low-risk PCa of ISUP grade group ≤ 2 (n = 1,900) no statistically significant difference could be found in terms of treatment free survival or risk of developing metastases (RR: 1.03, 95% CI: 0.62-1.71 and RR: 2.09, 95% CI: 0.75-5.82, respectively). Both reviews indicate that AS in unselected intermediate-risk patients implies a higher risk of progression over time. It remains unclear whether this difference only reflects the inborne difference in outcome, that can also be seen when comparing immediate treatment of low- and intermediate-risk PCa, or if the delay in treatment caused any worsening of the outcomes in the intermediate-risk group in any way. All three reviews conclude that AS could be offered to patients with intermediate-risk disease, but they should be informed of a higher risk of progression and the latter two reviews suggests limiting the inclusion of intermediate-risk patients to those with low-volume ISUP grade group 2 disease.

A Canadian consensus group proposes that low volume ISUP grade group 2 (< 10% Gleason pattern 4 on systematic biopsies) may also be considered for AS. These recommendations have been endorsed by the ASCO [225] and the DETECTIVE study consensus [330] for those patients with a PSA < 10 ng/mL and low core positivity. The DETECTIVE Study concluded that men with favourable ISUP grade group 2 PCa (PSA < 10 ng/mL, low density, clinical stage ≤ cT2a and a low number of positive systematic cores) should also be considered for deferred treatment [330]. In this setting, re-biopsy within six to twelve months to exclude sampling error is even more relevant than in low-risk disease [492,769]. The DETECTIVE Study-related qualitative SR aimed to determine appropriate criteria for inclusion of intermediate-risk disease into AS protocols [491]. Out of 371 AS protocols included in the review, more than 50% included patients with intermediate-risk disease on the basis of PSA up to 20 ng/mL (25.3%), ISUP grade group 2 or 3 (27.7%), clinical stage cT2b/c (41.6%) and/or direct use of D’Amico risk grouping of intermediate risk or above (51.1%). The DETECTIVE study reached consensus that patients with ISUP grade group 3, or patients with intraductal or cribriform histology, should not be considered for AS. The presence of any grade 4 pattern is associated with a 3-fold increased risk of metastases compared to ISUP grade group 1, while a PSA up to 20 ng/mL might be an acceptable threshold [769-771], especially in the context of low PSA-D. In addition, it is likely that MRI and targeted biopsies will detect small foci of Gleason grade 4 cancer that might have been missed with systematic biopsy. Therefore, care must be taken when explaining this treatment strategy, especially to patients with the longest life expectancy.

There is no clear consensus on how to interpret MRI and targeted biopsies for AS but the DETECTIVE study consensus was that if targeted biopsies based upon mpMRI images are performed, the number of positive cores of the targeted biopsies are not an indicator of the extent of disease or tumour volume. Indicator of the tumour volume may be either the number of positive cores, and the length of cancer in each core, based on systematic biopsies, or the volume of the dominant lesion seen on mpMRI [330].

In summary, AS can be considered in patients with a life expectancy of more than ten years and low-volume ISUP grade group 2 (defined as ≤ 3 positive systematic cores and ≤ 50% core involvement) or another single element of intermediate-risk disease (i.e. favourable intermediate-risk disease). Patients with ISUP grade group 3 disease, or patients with intraductal or cribriform histology, should be excluded. The monitoring schedule should be diligent, given the potential higher risk of progression, development of regional or distant metastases and death of this group compared with patients with low-risk disease. During monitoring, if repeat non-MRI-based systematic biopsies reveal > 3 positive cores or maximum CI > 50%/core of ISUP grade group 2 disease, patients should be reclassified (i.e., actively treated). For patients with a life expectancy of less than ten years, and not suitable for curative treatment, WW is a valid option and should be discussed with the patient.

6.3.2.2. Radical prostatectomy

Patients with intermediate-risk PCa should be informed about the results of two RCTs (SPCG-4 and PIVOT) comparing RRP vs. WW in localised PCa. In the SPCG-4 study, death from any cause (RR: 0.71, 95% CI: 0.53–0.95), death from PCa (RR: 0.38, 95% CI: 0.23–0.62) and distant metastases (RR: 0.49, 95% CI: 0.32–0.74) were significantly reduced in intermediate-risk PCa at 18 years. In the PIVOT trial, according to a pre-planned subgroup analysis among men with intermediate-risk tumours, RP significantly reduced all-cause mortality (HR: 0.69, 95% CI: 0.49–0.98), but not death from PCa (0.50, 95% CI: 0.21–1.21) at ten years. A meta-analysis based on the findings of SPCG-4, PIVOT and ProtecT demonstrated a benefit from RP over observation with a significantly decreased risk of death of 9% and of disease progression of 43% [772]. However, no stratification by disease stages was performed. The risk of having positive LNs in intermediate-risk PCa is between 3.7–20.1% [773]. A large study only found 2.9% of LN invasion in a contemporary cohort of 6,883 patients undergoing RP and LND for intermediate risk PCa [774]. Nerve sparing surgery is discussed in Section 6.1.2.3.5.

6.3.2.3. Radiation therapy
6.3.2.3.1. Recommended IMRT/VMAT for intermediate-risk PCa

Patients suitable for ADT can be given combined IMRT/VMAT with short-term ADT (four to six months) [775-777]. The RTOG 0815 RCT demonstrated improved BFSR, metastasis free and prostate CSS with the addition of six months ADT to dose escalated RT [685]. For adjuvant RT of the pelvic lymphatics (45-50 Gy) for NCCN unfavourable intermediate risk (cN0) see Section 6.2.3.2.1 - Radiotherapy for localised high-risk PCa. For patients unsuitable (e.g., due to comorbidities) or unwilling to accept ADT (e.g., to preserve their sexual health) the recommended treatment is IMRT/VMAT (76–78 Gy or equivalent moderate HFX) or a combination of IMRT/VMAT and BT as described below (see Section 6.2.2.3.2). A secondary analysis of the PCS III trial has suggested that patients with NCCN favourable intermediate-risk disease (see Section 4.4) can safely omit ADT if their RT dose is 76 Gy, but this is based on an unplanned subgroup analysis and only 138 patients had favourable intermediate-risk disease. An individual discussion between the physician and the patient of the possible benefits and harms of omitting ADT in this group is essential [778].

6.3.2.3.2. Brachytherapy for intermediate-risk PCa

Systematic review recommends LDR BT monotherapy can be offered to patients with NCCN favourable intermediate-risk disease and good urinary function (see Section 4.4) [779]. Fractionated HDR BT as monotherapy can be offered to selected patients with intermediate-risk PCa although they should be informed that results are only available from small series in very experienced centres. Five-year PSA control rates over 90% are reported, with late grade 3+ GU toxicity rates < 5% and no, or very minimal, grade 3+ GI toxicity rates [728]. There are no direct data to inform on the use of ADT in this setting. Trimodality therapy with IMRT plus BT boost and short-term ADT can be considered for NCCN unfavourable intermediate-risk PCa (see Section 4.4) but patients should be made aware that the potential improvements in biochemical control are accompanied with an increased risk of long-term urinary problems [717,719,724].

6.3.2.4. Other options for the primary treatment of intermediate-risk PCa (experimental therapies)
6.3.2.4.1. Focal therapy

A prospective study on focal therapy using HIFU in patients with localised intermediate-risk disease was published but the data was derived from an uncontrolled single-arm case series [750]. There is a paucity of high-certainty data for either whole-gland or focal ablative therapy in the setting of intermediate-risk disease. Consequently, neither whole-gland ablative treatment nor focal treatment can be considered as standard therapy for intermediate-risk patients and, if offered, it should only be in the setting of clinical trials or prospective registries [736].

6.3.2.4.2. Androgen deprivation therapy monotherapy

Data regarding the use of ADT monotherapy for intermediate-risk disease have been inferred indirectly from the EORTC 30891 trial, which was a RCT comparing deferred ADT vs. immediate ADT in 985 patients with T0–4 N0–2 M0 disease [773]. The trial showed a small, but statistically significant, difference in OS in favour of immediate ADT monotherapy but there was no significant difference in CSS, predominantly because the risk of cancer-specific mortality was low in patients with PSA < 8 ng/mL. Consequently, the use of ADT monotherapy for this group of patients is not considered as standard, even if they are not eligible for radical treatment.

6.3.2.5. Guidelines for the treatment of intermediate-risk disease*

Recommendations

Strength rating

Watchful Waiting (WW)

Offer WW in asymptomatic patients with life expectancy < ten years (based on comorbidities and age).

Strong

Active surveillance (AS)

Offer AS to selected patients with ISUP grade group 2 disease (e.g. < 10% pattern 4, PSA < 10 ng/mL, ≤ cT2a, low disease extent on imaging and low extent of tu-mour in biopsies (≤ 3 positive cores with Gleason score 3+4 and ≤ 50% cancer involvement/core), or another single element of intermediate-risk disease with low disease extent on imaging and low biopsy extent, accepting the potential increased risk of metastatic progression.

Weak

Patients with ISUP grade group 3 disease should be excluded from AS protocols.

Strong

Re-classify patients with low-volume ISUP grade group 2 disease included in AS protocols, if repeat non-MRI-based systematic biopsies performed during moni-toring reveal > 3 positive cores or maximum CI > 50%/core of ISUP grade group 2 disease.

Weak

Radical prostatectomy (RP)

Offer RP to patients with a life expectancy of > ten years.

Strong

Radical prostatectomy can be safely delayed for at least three months.

Weak

Offer nerve-sparing surgery to patients with a low risk of extra-capsular disease on that side.

Strong

Radiotherapeutic treatment

Offer low-dose rate (LDR) brachytherapy to patients with good urinary function and NCCN favourable intermediate-risk disease.

Strong

Offer intensity-modulated radiotherapy (IMRT)/volumetric modulated arc therapy (VMAT) plus image-guided radiotherapy (IGRT), with a total dose of 76–78 Gy or moderate hypofractionation (60 Gy/20 fx in 4 weeks or 70 Gy/28 fx in 6 weeks), in combination with short-term androgen deprivation therapy (ADT) (four to six months).

Strong

Offer focal boosting to MRI-defined dominant intra-prostatic tumour when using conventionally fractionated IMRT/IGRT (1.8-2.0 Gy per fraction) ensuring that Organ at Risk constraints are not exceeded

Weak

Offer ultra-hypofractionated IMRT/IGRT or SBRT, using either 36.25 Gy (40 Gy to prostate) in 5 fx or 42.7 Gy in 7 fx delivered alternate days.

Weak

Offer LDR brachytherapy boost combined with IMRT/VMAT plus IGRT to pa-tients with good urinary function and NCCN unfavourable intermediate-risk disease, in combination with short-term ADT (four to six months).

Weak

Offer high-dose rate (HDR) brachytherapy boost combined with IMRT/VMAT plus IGRT to patients with good urinary function and NCCN unfavourable in-termediate-risk disease, in combination with short-term ADT (four to six months).

Weak

Other therapeutic options

Only offer whole-gland ablative therapy (such as cryotherapy, high-intensity focused ultrasound, etc.) or focal ablative therapy within clinical trials or registries.

Strong

Do not offer ADT monotherapy to asymptomatic men not able to receive any local treatment.

Weak

*All recommendations are based on conventional imaging with isotope bone scan and CT/MR abdomen/pelvis.

6.3.3. Treatment of high-risk localised disease

Patients with high-risk PCa are at an increased risk of PSA failure, need for secondary therapy, metastatic progression and death from PCa. Nevertheless, not all high-risk PCa patients have a uniformly poor prognosis after RP [780]. When managed with non-curative intent, high-risk PCa is associated with 10-year and 15-year PCSM rates of 28.8 and 35.5%, respectively [781]. There is no consensus regarding the optimal treatment of men with high-risk PCa.

Some evidence suggests that radical treatment for high-risk PCa can be delayed up to three months after the diagnosis without any oncological consequences [782,783]. Systematic reviews suggest that there is a higher risk of biochemical recurrence and worse pathological outcomes when definitive treatment is given beyond a 6 to 9 months delay. However, there is no conclusive data regarding stronger endpoints (CSS or OS).

6.3.3.1. Radical prostatectomy

Provided that the tumour is not fixed to the pelvic wall or there is no invasion of the urethral sphincter, RP is a standard option in selected patients with a low tumour volume. ePLND provides accurate LN staging. Pre-operative PSMA-PET/CT is more accurate for staging generally, but can miss smaller LNs (especially LNs <5mm). ePLND may also miss LN metastasis [539,784]. Risks and benefits of pelvic LN dissection vs. PSMA PET/CT should be discussed with the patient pre-operatively. Patients should be aware pre-operatively that surgery may be part of multi-modal treatment, with adjuvant or SRT or ADT. Neoadjuvant therapy using ADT with or without new generation HT or docetaxel is not indicated. (See Section 6.1.2.2.4) [785,786]. Nerve sparing management is discussed in Section 6.1.2.3.5.

At 15 years follow-up cN0 patients who undergo RP but who were found to have pN1 were reported to have an overall CSS and OS of 45% and 42%, respectively [787,788]. A SR has reported 10-year BCR-free, CSS, and OS rates ranging from 28% to 56%, 72% to 98%, and 60% to 87.6%, respectively, in pN1 patients [789]. These findings highlight that pN1 patients represent a very heterogeneous patient group and further treatment must be individualised based on risk factors (see Sections 6.2.5.2 and 6.2.5.6).

6.3.3.2. External beam radiation therapy

For high-risk localised PCa, a combined modality approach should be used consisting of IMRT/VMAT plus long-term ADT. The duration of ADT has to take into account PS, comorbidities and the number of poor prognostic factors. It is important to recognise that in several studies EBRT plus short-term ADT did not improve OS in high-risk localised PCa and long-term ADT (at least 2 to 3 years) is currently recommended for these patients [677,678,689]. Moderate HFX is an option in selected high-risk patients with localised disease. The CHHiP study included 12% high-risk patients (n = 386) but limited entry to those with a PSA < 30 ng/mL and a Roach formula risk of SV involvement < 30% [661]. Patients were ineligible if they had both T3a tumours and ISUP grade group 4 or higher.

6.3.3.2.1. Lymph node irradiation in cN0

There is no clear evidence for prophylactic irradiation of the pelvic LNs in intermediate- and high-risk disease. The long-term results of the NRG/RTOG 9413-trial which randomised intermediate-risk and high-risk localised PCa patients (1,322 cN0 patients were enrolled), showed that neoadjuvant HT plus whole pelvic RT improved PFS only compared with neoadjuvant ADT plus prostate RT and whole pelvic RT plus adjuvant ADT [790]. However, at the increased risk of ≥ grade 3 GI-toxicity.

A well-conducted single-centre RCT randomised 224 patients comparing prostate-only RT (PORT) vs. whole pelvic RT (WPRT) in localised high-risk- and locally-advanced tumours (cN0) with a risk of > 20% of positive nodes (Roach formula). With a median follow-up of 68 months there was a significant improvement of distant MFS (95.9% vs. 89.2%, HR: 0.35, p = 0.01) and DFS (89.5% vs.77.2%, p = 0.02). However, there was a significant higher rate of late GU ≥ 2 effects (17.7% vs. 7.5%, p = 0.02), the trial was relatively small in size with additional limitations and these findings are therefore insufficient to define a change in practice [791,792]. The benefits of pelvic nodal irradiation using IMRT/VMAT merit further investigation in large scale RCTs as conducted by the RTOG or the UK National Cancer Research Institute (NCRI).

6.3.3.2.2. Brachytherapy boost

In men with NCCN unfavourable intermediate- or high-risk PCa, BT boost with supplemental EBRT and HT may be considered. See Sections 6.1.3.4.1 and 6.1.3.4.2 for details on RCTs comparing EBRT alone and EBRT with LDR or HDR boost, respectively.

6.3.3.3. Options other than surgery or radiotherapy for the primary treatment of localised PCa

Currently there is a lack of evidence supporting any other treatment option apart from RP and radical RT in localised high-risk PCa. The use of ADT monotherapy was addressed by the EORTC 30891 trial [773] (see Section 6.2.4.4.2). Immediate ADT may only benefit patients with a PSA-DT < twelve months, and either a PSA > 50 ng/mL or a poorly-differentiated tumour [773,793].

6.3.3.4. Guidelines for radical and palliative treatment of high-risk localised disease*

Recommendations

Strength rating

Watchful Waiting (WW)

Offer WW to asymptomatic patients with life expectancy < ten years.

Strong

Radical prostatectomy (RP)

Offer RP to selected patients as part of potential multi-modal therapy.

Weak

Extended pelvic lymph node dissection (ePLND)

In patients undergoing a lymph node dissection you should perform an extended PLND.

Strong

Do not perform a frozen section of nodes during RP to decide whether to proceed with, or abandon, the procedure (see Section 6.2.4.1).

Strong

Radiotherapeutic treatment

Offer patients intensity-modulated radiation therapy (IMRT)/volumetric modu-lated arc therapy (VMAT) plus image-guided radiation therapy (IGRT) with 76–78 Gy in combination with long-term androgen deprivation therapy (ADT) (2 to 3 years).

Strong

Offer focal boosting to MRI-defined dominant intra-prostatic tumour when using normo-fractionated IMRT/IGRT (1.8-2.0 Gy per fraction) ensuring that Organ at Risk constraints are not exceeded.

Weak

Offer patients with good urinary function IMRT/VMAT plus IGRT with brachy-therapy boost (either high-dose rate or low-dose rate), in combination with long-term ADT (2 to 3 years).

Weak

Therapeutic options outside surgery or radiotherapy

Do not offer either whole gland or focal therapy.

Strong

Only offer ADT monotherapy to those patients unwilling or unable to receive any form of local treatment if they have a prostate-specific antigen (PSA)-doubling time < twelve months, and either a PSA > 50 ng/mL or a poorly-differentiated tumour.

Strong

*All recommendations are based on conventional imaging with isotope bone scan and CT/MR abdomen/pelvis.

6.3.4. Treatment of locally-advanced PCa

In the absence of high-level evidence, a SR could not define the most optimal treatment option [794]. Randomised controlled trials are only available for EBRT. A local treatment combined with a systemic treatment provides the best outcome, provided the patient is fit enough to receive both. The initial results of the SCPG-15 trials suggested that randomisation between surgery and EBRT is feasible, but oncologic outcomes are awaited [795].

6.3.4.1. Radical prostatectomy

Surgery for locally-advanced disease as part of a multi-modal therapy has been reported [781,796,797]. However, the comparative oncological effectiveness of RP as part of a multi-modal treatment strategy vs. upfront EBRT with ADT for locally-advanced PCa remains unknown, although a prospective phase III RCT (SPCG-15) comparing RP (with or without adjuvant or salvage EBRT) against primary EBRT and ADT among patients with locally-advanced (T3) disease is currently recruiting [798]. Data from retrospective case series demonstrated over 60% CSS at 15 years and over 75% OS at ten years [781,796,797,799-802]. For cT3b–T4 disease, PCa cohort studies showed 10-year CSS of over 87% and OS of 65% [803,804]. The indication for RP in all previously described stages assumes the absence of clinically detectable nodal involvement (cN0), based on conventional imaging. In case of suspected positive LNs during RP (initially considered cN0) the procedure should not be abandoned since RP may have a survival benefit in these patients. Intra-operative frozen section analysis is not justified in this case [541].

6.3.4.2. Treatment of cN1 M0 PCa

Lymph-node metastasised PCa is an entity where options for local therapy and systemic therapies overlap. Approximately 5% to 10% of newly diagnosed PCa patients have synchronous suspected pelvic nodal metastases on conventional imaging (CT/bone scan) without bone or visceral metastases (cN1 M0 stage). Meta-analyses have shown that PSMA-PET/CT prior to primary treatment in advanced PCa detected disease outside the prostate in 32% of cases despite prior negative conventional imaging using bone scan and pelvic CT/MRI [412]. A RCT assessing PSMA-PET/CT as staging tool in high-risk PCa confirmed these findings and showed a 32% increase in accuracy compared with conventional imaging for the detection of pelvic nodal metastases [429]. Notably, more sensitive imaging also caused a stage shift with more cases classified as miN1, but with, on average, lower nodal disease burden.

The management of cN1M0 PCa is historically based on long-term ADT combined with a local treatment. The benefit of adding local treatment has been assessed in various retrospective studies, summarised in one SR [805] including five studies only [806-810]. The results of this SR were confirmed [811]. The findings of this retrospective analysis suggested an advantage in both OS and CSS after local treatment (RT or RP) combined with ADT as compared to ADT alone. Only limited evidence exists supporting RP for cN1 patients. Moschini et al., compared the outcomes of 50 patients with cN+ with those of 252 patients with pN1, but cN0 at pre-operative staging. cN+ was not a significant predictor of CSS [812].

The addition of a brachytherapy boost to ADT plus EBRT was not associated with improved OS in a retrospective study of 1,650 cN1 patients after multivariable adjustment and propensity score matching [813].

The intensification of systemic treatment (abiraterone acetate, docetaxel, zoledronic acid) has been assessed in unplanned sub-group analyses from the STAMPEDE multi-arm RCT by stratifying for cN1 and M1 status [809,814]. The analyses were balanced for nodal involvement and for planned RT use in STAMPEDE at randomisation and at analysis. Abiraterone acetate was associated with a non-significant OS improvement (HR: 0.75, 95% CI: 0.48–1.18) in non-metastatic patients (N0/N+M0), but OS data were still immature with a low number of events. Furthermore, this was an underpowered subgroup analysis and hypothesis generating at best. Moreover, subgroup analyses were performed according to the metastatic/non-metastatic status and to the nodal status (any M) without specific data for the N1M0 population (n = 369; 20% of the overall cohort). The same would apply for the docetaxel arm in the STAMPEDE trial for which no specific subgroup analysis of newly diagnosed N1M0 PCa (n = 171, 14% of the overall cohort) was performed. However, the addition of docetaxel, zoledronic acid, or their combination, did not provide any OS benefit when stratifying by M0 and N+ status.

In the AFU-GETUG 12 trial comparing the impact of docetaxel plus estramustine in addition to ADT, 29% of included high-risk non-metastatic PCa patients had a nodal involvement (pN1) at randomisation [815]. A non-significant trend towards better relapse-free survival rates was reported in the treatment arm (HR 0.66; 86 0.43–1.01) without OS benefit. A meta-analysis of docetaxel trials in N0/N1-M0 patients concluded to an 8% 4-year survival advantage for docetaxel compared with ADT alone in terms of failure-free survival without OS benefit [816]. Two RCTs from the STAMPEDE platform protocol reported on men with de novo high-risk/locally-advanced M0 disease, or relapse after primary curative therapy with high-risk features. Thirty-nine percent of patients (n = 774) were N1 on conventional imaging [817]. Radiotherapy in addition to long-term ADT was administered in 71% of these patients. Given the MFS and OS benefits observed in the overall population (see Section 6.3.4.2), combined ADT (for 3 years) and additional abiraterone (for 2 years) should be a SOC in cN1 patients in addition to prostate- and WPRT.

Table 6.3.4.1: Selected studies assessing local treatment in (any cT) cN1 M0 prostate cancer patients

Study

n

Design

Study period/follow-up

Treatment arms

Effect on survival

Bryant, et al. 2018 [818]

648

Retrospective

(National Veterans Affairs)

2000-2015


61 mo.

ADT ± EBRT

Significant benefit for combined treatment only if PSA levels less than the median (26 ng/mL)

All-cause mortality HR: 0.50 CSS, HR: 0.38

Sarkar, et al. 2019 [819]

741

Retrospective

(National Veterans Affairs)

2000-2015


51 mo.

ADT ± local treatment (surgery or RT)

Significant benefit for RP

All cause mortality HR 0.36

CSS, HR: 0.32


No statistical difference for RP vs. RT (p > 0.1)

All-cause mortality HR: 047

CSS, HR: 0.88

Lin, et al.
2015 [807]

983 before propensity score matching

Retrospective (NCDB)

2004-2006


48 mo.

ADT ± EBRT

Significant benefit for combined treatment

5-yr OS: 73% vs. 52%

HR: 0.5

Tward, et al. 2013 [806]

1,100

Retrospective (SEER)

1988-2006


64 mo.

EBRT
(n = 397) vs. no EBRT
(n = 703)

No information on ADT)

Significant benefit for EBRT

5-yr CSS 78% vs. 71%

HR: 0.66

5-yr. OS: 68% vs. 56%,
HR: 0.70

Rusthoven,
et al. 2014 [810]

796

Retrospective (SEER)

1995-2005


61 mo.

EBRT vs. no EBRT (no information on ADT)

Significant benefit for EBRT

10-yr OS: 45% vs. 29%

HR: 0.58

Seisen, et al. 2018 [808]

1,987

Retrospective (NCDB)

2003-2011


50 mo.

ADT ± local treatment (surgery or RT)

Significant benefit for combined treatment

5-yr OS: 78.8% vs. 49.2%

HR: 0.31

No difference between RP and RT

James, et al. 2016 [809]

177

Unplanned sub-group analysis RCT

2005-2014


17 mo.

ADT ± EBRT

Significant benefit for combined treatment

5-yr OS: 93% vs. 71%

2-yr FFS: 81% vs 53%

FFS, HR: 0.48

Elumalai et al. [811]

337

Retrospective

4 centres UK

2022-2019

ADT +/- EBRT

Significant benefit for combined treatment

5-yr.OS: 87% vs. 56% HR: 0.27

5-yr. BPFS: 74.1% vs. 34.2% HR: 0.33

ADT = androgen deprivation therapy; CSS = cancer-specific survival; EBRT = external beam radiotherapy;FFS = failure-free survival; HR = hazard ratio; mo = months; n = number of patients; OS = overall survival;RP = radical prostatectomy; RT = radiotherapy; yr = year.

6.3.4.3. Options other than surgery or radiotherapy for primary treatment
6.3.4.3.1. Investigational therapies

Currently cryotherapy, HIFU or focal therapies have no place in the management of locally-advanced PCa.

6.3.4.3.2. Androgen deprivation therapy monotherapy

The deferred use of ADT as single treatment modality has been answered by the EORTC 30891 trial [773]. Nine hundred and eighty-five patients with T0–4 N0–2 M0 PCa received ADT alone, either immediately or after symptomatic progression or occurrence of serious complications. After a median follow-up of 12.8 years, the OS favoured immediate treatment (HR: 1.21, 95% CI: 1.05–1.39). Surprisingly, no different disease-free or symptom-free survival was observed, raising the question of survival benefit. In locally-advanced T3–T4 M0 HSPC unsuitable for surgery or RT, immediate ADT may only benefit patients with a PSA > 50 ng/mL and a PSA-DT < twelve months or those that are symptomatic [773,793]. The median time to start deferred treatment was 7 years. In the deferred treatment arm 25.6% of patients died without needing treatment.

6.3.4.4. Guidelines for radical- and palliative treatment of locally-advanced disease*

Recommendations

Strength rating

Radical prostatectomy (RP)

Offer RP to patients with cN0 disease as part of multi-modal therapy.

Weak

Extended pelvic lymph node dissection (ePLND)

In patients undergoing a lymph node dissection you should perform an extended PLND.

Strong

Radiotherapeutic treatments

Offer patients with cN0 disease intensity-modulated radiation therapy (IMRT)/volumetric modulated arc therapy (VMAT) plus image-guide radiation therapy in combination with long-term androgen deprivation therapy (ADT).

Strong

Offer patients with cN0 disease and good urinary function, IMRT/VMAT plus IGRT with brachytherapy boost (either high-dose rate or low-dose rate), in combination with long-term ADT.

Weak

Offer long-term ADT for at least 2 years.

Strong

Offer IMRT/VMAT plus IGRT to the prostate in combination with long-term ADT and two years of abiraterone to cN0M0 patients with ≥ 2 high-risk factors (cT3-4, Gleason ≥ 8 or PSA ≥ 40 ng/mL).

Strong

Offer IMRT/VMAT plus IGRT to the prostate plus pelvis in combination with long-term ADT and two years of abiraterone to cN1M0 patients.

Strong

Therapeutic options outside surgery or radiotherapy

Do not offer whole gland treatment or focal treatment.

Strong

*All recommendations are based on conventional imaging with isotope bone scan and CT/MR abdomen/pelvis.

6.3.5. Adjuvant treatment after radical prostatectomy

6.3.5.1. Introduction

Adjuvant treatment is by definition additional to the primary or initial therapy with the aim of decreasing the risk of relapse, despite the apparent full control following surgery. A post-operative detectable PSA is an indication of persistent prostate cells (see Section 6.3.6). All information listed below refers to patients with a post-operative undetectable PSA.

6.3.5.2. Risk factors for relapse

Patients with ISUP grade group > 2 in combination with EPE (pT3a) and particularly those with SV invasion (pT3b) and/or positive surgical margins are at high risk of progression, which can be as high as 50% after five years [820]. Irrespective of the pT stage, the number of removed nodes [821-828], tumour volume within the LNs and capsular perforation of the nodal metastases are predictors of early recurrence after RP for pN1 disease [829]. A LN density (defined as ‘the percentage of positive LNs in relation to the total number of analysed/removed LNs’) of over 20% was found to be associated with poor prognosis [830]. The number of involved nodes seems to be a major factor for predicting relapse [823,824,831]; the threshold considered is less than 3 positive nodes from an ePLND [540,823,831]. However, prospective data are needed before defining a definitive threshold value.

6.3.5.2.1. Biomarker-based risk stratification after radical prostatectomy

The Decipher® gene signature consists of a 22-gene panel representing multiple biological pathways and was developed to predict systemic progression after definitive treatment. A meta-analysis of five studies analysed the performance of the Decipher® Genomic Classifier (GC) test on men post-RP. The authors showed in multivariable analysis that Decipher® GC remained a statistically significant predictor of metastasis (HR: 1.30, 95% CI: 1.14–1.47, p < 0.001) per 0.1 unit increase in score and concluded that it can independently improve prognostication of patients post-RP within nearly all clinicopathologic, demographic, and treatment subgroups [832]. A SR of the evidence for the Decipher® GC has confirmed the clinical utility of this test in post-RP decision-making [833]. Further studies are needed to establish how to best incorporate Decipher® GC in clinical decision-making.

6.3.5.3. Immediate (adjuvant) post-operative external irradiation after RP (cN0 or pN0)

Four prospective RCTs have assessed the role of immediate post-operative RT (adjuvant RT [ART]) (undetectable PSA mostly defined as PSA < 0.1 ng/mL), demonstrating an advantage (endpoint, development of BCR) in high-risk patients (e.g., pT2 with positive surgical margins and ISUP grade group 3–5 or pT3/4 with- or without positive surgical margins and ISUP grade group 3–5) post-RP (Table 6.3.5.1). In the ARO 96-02 trial, 80% of the pT3/R1/GS 8–10 patients randomised to observation developed BCR within ten years [834]. It must be emphasised that PSA was undetectable at inclusion only in the ARO 96-02 trial which presents a major limitation interpreting these findings as patients with a detectable PSA would now be considered for salvage therapy rather than ART [834].

Table 6.3.5.1: Overview of all four randomised trials for adjuvant surgical bed radiation therapy after RP* (without ADT)

Study

n

Inclusion criteria

Randomisation

Definition of BCR PSA (ng/mL)

Median FU (mo)

Biochemical Progression-free survival

Overall survival

SWOG 8794 2009 [835]

431

pT3 cN0 ± involved SM

60-64 Gy vs. observation

> 0.4

152

10 yr.: 53% vs. 30%

(p < 0.05)

10 yr.: 74% vs. 66% Median time:

15.2 vs.

13.3 yr.,

p = 0.023

EORTC 22911 2012 [836]

1,005

pT3 ± involved SM pN0 pT2 involved SM pN0

60 Gy vs. observation

> 0.2

127

10 yr.: 60.6% vs. 41%

(p < 0.001)

81% vs. 77% n.s.

ARO 96-02 2014 [834]

388

pT3 (± involved SM) pN0 PSA post-RP undetectable

60 Gy vs. observation

> 0.05 + confirmation

112

10 yr.: 56% vs. 35%

(p = 0.0001)

10 yr.: 82% vs. 86% n.s.

FinnProstate Group 2019 [837]

250

pT2,R1/ pT3a

66.6 Gy vs. observation
(+ SRT)

> 0.4 (in 2 successive measure-

ments)

112 vs. 103 (patients alive)

10 yr.: 82% vs. 61%

p < 0.001

10 yr.: 92% vs. 87% n.s.

*See Section 6.3.5.1 for delayed (salvage) post-radical prostatectomy external irradiation.BCR = biochemical recurrence; FU = follow-up; mo = months; n = number of patients; n.s. = not significant;OS = overall survival; PSA = prostate-specific antigen; RP = radical prostatectomy; SM = surgical margin;SRT = salvage radiotherapy.

6.3.5.4. Comparison of adjuvant- and salvage radiotherapy

Two retrospective matched studies (510 and 149 patients receiving ART) failed to show an advantage for MFS [838,839]. However, both studies were underpowered for high-risk patients (pT3b/R1/ISUP grade group 4–5 PCa). In contrast to these studies, a propensity score-matched retrospective analysis of two cohorts of 366 pT3 and/or R1 patients found that compared to SRT at a PSA between 0.1 and 0.5 ng/mL, ART given at an undetectable PSA (< 0.1 ng/mL) improved all three endpoints; BCR, MFS, and OS [840].

Both approaches (ART and early SRT) together with the efficacy of adjuvant ADT are compared in three prospective RCTs: the Medical Research Council (MRC) Radiotherapy and Androgen Deprivation In Combination After Local Surgery (RADICALS) trial [841], the Trans-Tasman Oncology Group (TROG) Radiotherapy Adjuvant Versus Early Salvage (RAVES) trial [842], and the Groupe d’Etude des Tumeurs Uro- Genitales (GETUG-AFU 17) trial [843]. In addition, a pre-planned meta-analysis of all three trials has been published (Table 6.3.5.2) [844].

Two trials closed early after randomising 333/470 patients (RAVES) and 424/718 (GETUG-AFU-17) patients. RADICALS-RT included 1,396 patients with the option of subsequent inclusion in RADICALS-HT; 154/649 (24%) of patients starting in the adjuvant RT group also received neoadjuvant or adjuvant HT; 90 patients for six months/45 for 2 years/19 patients outside RADICALS-HT. From the SRT group, 61/228 (27%) received neoadjuvant or adjuvant HT for six months (n = 33) and two years (n = 13). Fifteen of these patients were treated outside the trial [841]. All men in the GETUG-AFU-17 trial (n = 424) received six months of HT. All together, 684 out of 2,153 patients received additional ADT for at least six months across both trials [844]. Radiotherapy to the pelvic lymphatics was allowed in the GETUG-AFU and in the RADICALS-RT trials.

The primary endpoint for RAVES and GETUG-AFU 17 was biochemical PFS, and for RADICALS-RT MFS. So far only PFS data has been reported, and not MFS- or OS data. With a median follow-up between 4.9 years and 6.25 years there was no statistically significant difference for biochemical PFS for both treatments in all three trials (see Table 6.2.5.2) indicating that in the majority of patients adjuvant irradiation should be avoided. Additionally, there was a significant lower rate of grade ≥ 2 GU late side effects and grade 3–4 urethral strictures in favour of early SRT; which may also be caused by the low number of patients with PSA-progression and subsequent need for early SRT at the time of analysis (40% of patients).

It is important to note that the indication for ART changed over the last ten years with the introduction of ultra-sensitive PSA-tests, favouring early SRT. Therefore many patients, randomised in these three trials (accruing 2006–2008) are not likely to benefit from ART as there is a low risk of biochemical progression (~20–30%) in, for example, pT3R0 or pT2R1-tumours. The median pre-SRT PSA in all 3 trials was 0.24 ng/mL. Therefore, patients with ‘low-risk factors’ of biochemical progression after RP should be closely followed up with ultra-sensitive assays and SRT should be discussed if needed as soon as PSA starts to rise, which has to be confirmed by a second PSA measurement (see Section 6.3). The proportion of patients with adverse pathology at RP (ISUP grade group 4–5 and pT3 with or without positive margins) in all three trials was low (between 10–20%) and therefore even the meta-analysis may be underpowered to show an outcome in favour of SRT [844]. In addition, the side-effect profile may have been impacted with a larger proportion of ART patients receiving treatment with older 3D-treatment planning techniques as compared to SRT patients (GETUG-AFU 17: ART, 69% 3D vs. 46% SRT) and patients treated more recently were more likely to undergo IMRT techniques with a proven lower rate of late side effects [634].

For these reasons, 10-year OS and MFS endpoints results should be awaited before drawing final conclusions. Due to the small number of patients with adverse pathology (ISUP grade group 4–5 and pT3) included in these three trials (between 10–20%), ART remains a recommended treatment option in highly selected patients with adverse pathology (‘high-risk patients’) i.e. ISUP grade group 4–5 and pT3 with or without positive margins [845,846]. This recommendation was supported by a published retrospective multi-centre study comparing ART and SRT in patients with high-risk features (pN1 or ISUP grade group 4–5 and pT3/4-tumours) after RP [847]. After
a median follow-up of 8.2 years of the 26,118 men included in the study, 2,104 patients died, 25.62% from PCa (n = 539) and 2,424 patients had adverse pathology compared with 23.694 who did not. After excluding men with persistent PSA after RP, ART when compared with early SRT showed a significantly lower acute mortality risk
(p = 0.02, HR: 0.33).

Table 6.3.5.2: Overview of all three randomised trials and one meta-analysis for patients treated with adjuvant vs. early salvage RT after RP

Study

n

Inclusion criteria

Randomisation

Definition of BCR PSA

(ng/mL)

Median FU (yr)

BPFS

OS or MFS

Side effects

RAVES

TROG 08.03/

ANZUP

2020 [842]

333

target

was 470

early

closed

pT3a/pT3b

any T - SM+

PSA post-RP:
< 0.1 ng/mL

64 Gy ART PSA:
< 0.1 ng/mL vs.

64 Gy early SRT

at PSA
> 0.2 ng/mL

med. pre-SRT: n.r.

> 0.4 post RT

6.1

5 yr.:

86% vs. 87%

(p > 0.05)

n.r.

LT grade

> GU:

70% vs.

54%

(p = 0.002)

RADICALS-RT

2020 [841]

1,396

pT3a/

pT3b/pT4

PSA
> 10 ng/mL
pre-RP

any T, SM+

Gleason

7-10

PSA post-RP:

< 0.2 ng/mL

52.5 Gy (20 Fx)

or 66 Gy (33 Fx) ART

early SRT

identical

at PSA > 0.1

med.pre-SRT:
0.2 ng/mL

> 0.4 or 2 at any time

4.9

5 yr.:

85% vs. 88%

(p = 0.56)

n.r.

Self-reported urinary incontinence 1 yr: 4.8 vs.

4 (p = 0.023)

urethral

stricture

grade 3/4 2 yr:

6% vs. 4%

(p = 0.02)

GETUG-AFU

17

2020 [843]

424

target was

718

early closed

pT3a/pT3b/pT4a and SM + PSA

post-RP:

< 0.1 ng/mL

66 Gy (ART) vs.

66 Gy early SRT

at PSA 0.1

both groups:

6 mo. LHRH-A

med. pre-SRT 0.24

> 0.4

6.25

5 yr:

92% vs. 90%

(p = 0.42)

n.r.

LT grade > 2 GU 27% vs. 7%

(p < 0.001)

ED: 28% vs. 8%

(p < 0.001)

ARTISTIC-Meta-analysis

2020 [844]

2,153

see above

see above

see above

4.5

5 yr.:

89% vs. 88%

p = 0.7

n.r.

n.r.

ART = adjuvant radiotherapy; BCR = biochemical recurrence; BPFS = biochemical progression-free survival; ED = erectile dysfunction; FU = follow-up; Fx = fraction; GU = genito-urinary; LHRH = luteinising hormonereleasing hormone; LT = late toxicity; mo = months; med = median; MFS = metastasis-free survival; n.r. = not reported; OS = overall survival; PSA = prostate-specific antigen; RP = radical prostatectomy; RT = radiotherapy; SRT = salvage radiotherapy; + = positive; yr = year.

6.3.5.5. Adjuvant systemic therapy in N0 disease

Adjuvant androgen ablation with bicalutamide 150 mg daily did not improve PFS in localised disease while it did for locally-advanced disease after RT. However, this never translated to an OS benefit [848]. A SR showed a possible benefit for PFS but not OS for adjuvant androgen ablation [786].

The TAX3501 trial comparing the role of leuprolide (18 months) with and without docetaxel (6 cycles) ended prematurely due to poor accrual. A phase III RCT comparing adjuvant docetaxel against surveillance after RP for locally-advanced PCa showed that adjuvant docetaxel did not confer any oncological benefit [849]. Consequently, adjuvant chemotherapy after RP should only be considered in a clinical trial [850].

6.3.5.6. Adjuvant treatment in pN1 disease
6.3.5.6.1. Adjuvant androgen ablation alone

The combination of RP and early adjuvant HT in pN+ PCa has been shown to achieve a 10-year CSS rate of 80% and has been shown to significantly improve CSS and OS in prospective RCTs [851,852]. However, these trials included mostly patients with high-volume nodal disease and multiple adverse tumour characteristics and these findings may not apply to men with less extensive nodal metastases.

6.3.5.6.2. Adjuvant radiotherapy combined with ADT in pN1 disease

In a retrospective multi-centre cohort study, maximal local control with RT to the prostatic fossa appeared to be beneficial in PCa patients with pN1 after RP, treated ‘adjuvantly‘ with continuous ADT (within six months after surgery irrespective of PSA). The beneficial impact of adjuvant RT on survival in patients with pN1 PCa was highly influenced by tumour characteristics. Men with low-volume nodal disease (< 3 LNs), ISUP grade group 2–5 and pT3–4 or R1, as well as men with 3 to 4 positive nodes were more likely to benefit from RT after surgery, while the other subgroups did not [853]. In contrast, a retrospective multi-centre study including 1,614 patients and a median follow-up of 7,02 years assessed ART + ADT. Adjuvant RT compared to SRT was associated with a decreased all-cause mortality and this reduction increased with each additional positive pelvic LN, from the first one on and the highest effect was for more than 3 positive nodes [854]. These data are in agreement with a US National Cancer Database analysis based on 5,498 patients [855]. Another US National Cancer Database study including 8,074 pN1 patients reports improved OS after ADT plus EBRT (including pelvic LNs) vs. observation and vs. ADT alone in all men with single or multiple adverse pathological features. Men without any adverse pathological features did not benefit from immediate adjuvant therapy [856].

In a SR of the literature, RT with or without ADT was associated with improved survival in men with locally-advanced disease and a higher number of positive nodes [789]. Radiotherapy to the pelvic lymphatics and the prostate fossa plus long-term ADT can be offered to patients with pN1 disease. [853,857]. However, the optimal duration of ADT is still unkown.

6.3.5.6.3. Observation of pN1 patients after radical prostatectomy and extended lymph node dissection

Several retrospective studies and a SR addressed the management of patients with pN1 PCa at RP [789,831,853,857,858]. A subset of patients with limited nodal disease (1–2 positive LNs) showed favourable oncological outcomes and did not require additional treatment.

An analysis of 209 pN1 patients with one or two positive LNs at RP showed that 37% remained metastasis-free without need of salvage treatment at a median follow-up of 60.2 months [858]. Touijer et al., reported their results of 369 pN1-positive patients (40 with and 329 without adjuvant treatment) and showed that higher pathologic grade group and > 3 positive LNs were significantly associated with an increased risk of BCR on multivariable analysis [831]. Biochemical-free survival rates in pN1 patients without adjuvant treatment ranged from 43% at four years to 28% at ten years [789]. Reported CSS rates were 78% at five years and 72% at ten years. The majority of these patients were managed with initial observation after surgery, had favourable disease characteristics, and 63% had only one positive node [789]. Initial observation followed by early salvage treatment at the time of recurrence may represent a safe option in selected patients with a low disease burden [789].

6.3.5.7. Guidelines for adjuvant treatment for pN0 and pN1 disease after radical prostatectomy*

Recommendations

Strength rating

Do not prescribe adjuvant androgen deprivation therapy (ADT) to pN0 patients.

Strong

In pN0 patients with ISUP grade group 4–5 and pT3 ± positive margins, offer adjuvant intensity-modulated radiation therapy (IMRT)/volumetric modulated arc therapy (VMAT) plus image-guided radiation therapy (IGRT).

Strong

In pN1 patients, after an extended lymph node dissection, discuss three management options, based on nodal involvement characteristics:

1. Offer adjuvant ADT;

2. Offer adjuvant ADT with additional IMRT/VMAT plus IGRT;

3. Offer observation (expectant management) to a patient after eLND and < 2 nodes and a PSA < 0.1 ng/mL.

Weak

*All recommendations are based on conventional imaging with isotope bone scan and CT/MR abdomen/pelvis.

6.3.6. Persistent PSA after radical prostatectomy

Between five and 20% of men continue to have detectable or persistent PSA after RP (when defined in the majority of studies as detectable post-RP PSA of ≥ 0.1 ng/mL within 4 to 8 weeks of surgery) [859,860]. It may result from persistent local disease, pre-existing metastases or residual benign prostate tissue.

6.3.6.1. Natural history of persistently elevated PSA after RP

Several studies have shown that persistent PSA after RP is associated with more advanced disease (such as positive surgical margins, pathologic stage ≥ T3a, positive nodal status and/or pathologic ISUP grade group > 3) and poor prognosis. Initially defined as ≥ 0.1 ng/mL, improvements in the sensitivity of PSA assays now allow for the detection of PSA at much lower levels.

Moreira et al., demonstrated that failure to achieve a PSA of less than 0.03 ng/mL within six months of surgery was associated with an increased risk of BCR and overall mortality [861,862]. However, since the majority of the published literature is based on the 0.1 ng/mL PSA cut-off, there is significantly more longterm data for this definition. Predictors of PSA persistence were higher BMI, higher pre-operative PSA and ISUP grade group ≥ 3 [862]. In patients with PSA persistence, one and 5-year BCR-free survival were 68% and 36%, compared to 95% and 72%, respectively, in men without PSA persistence [861]. Ten-year OS in patients with and without PSA persistence was 63% and 80%, respectively.

Spratt et al., confirmed that a persistently detectable PSA after RP represents one of the worst prognostic factors associated with oncological outcome [863]. Of 150 patients with a persistent PSA, 95% received RT before detectable metastasis. In a multi-variable analysis the presence of a persistently detectable PSA post-RP was associated with a 4-fold increase in the risk of developing metastasis. This was confirmed by data from Preisser et al., who showed that persistent PSA is prognostic of an increased risk of metastasis and death [864]. At 15 years after RP, MFS rates, OS and CSS rates were 53.0 vs. 93.2% (p < 0.001), 64.7 vs. 81.2% (p < 0.001) and 75.5 vs. 96.2% (p < 0.001) for persistent vs. undetectable PSA, respectively. The median follow-up was 61.8 months for patients with undetectable PSA vs. 46.4 months for patients with persistent PSA. In multivariable Cox regression models, persistent PSA represented an independent predictor for metastasis (HR: 3.59, p < 0.001), death (HR: 1.86, p < 0.001) and cancer-specific death (HR: 3.15, p < 0.001).

However, not all patients with persistent PSA after RP experience disease recurrence. Xiang et al., showed a 50% 5-year BCR-free survival in men who had a persistent PSA level > 0.1 but ≤ 0.2 ng/mL at 6–8 weeks after RP [865].

Rogers et al., assessed the clinical outcome of 160 men with a persistently detectable PSA level after RP [866]. No patient received adjuvant therapy before documented metastasis. In their study, 38% of patients had no evidence of metastases for ≥ 7 years while 32% of the patients were reported to develop metastases within 3 years. Noteworthy is that a significant proportion of patients had low-risk disease. In multi-variable analysis the PSA slope after RP (as calculated using PSA levels 3 to twelve months after surgery) and pathological ISUP grade group were significantly associated with the development of distant metastases.

6.3.6.2. Imaging in patients with persistently elevated PSA after RP

Standard imaging with bone scan and MRI has a low detection rate in men with a PSA below 2 ng/mL. However, PSMA PET/CT has been shown to identify residual cancer with positivity rates of 33%, 46%, 57%, 82%, and 97%, in men with post-RP PSA ranges of 0–0.19, 0.2–0.49, 0.5–0.99, 1–1.99, and ≥ 2 ng/mL, respectively [867-872] which can guide SRT planning [873]. Based on these post-RP PSA ranges, Schmidt-Hegemann et al., studied 129 patients who had either persistent PSA (52%) or BCR (48%) after RP, showing that men with a persistent PSA had significantly more pelvic nodal involvement on PSMA PET/CT than those developing a detectable PSA [874]. In a multi-centre retrospective study including 191 patients, 68Ga-PSMA localised biochemical persistence after RP in more than two-thirds of patients with high-risk PCa features. The obturator and presacral or mesorectal nodes were identified as high risk for residual disease [875]. Another retrospective study included 150 patients with persistent PSA after RARP who were re-staged with both 68Ga-PSMA and 18F-DCFPyL PSMA. The authors found that in the presence of persistent PSA the majority of patients already had metastatic pelvic LNs or distant metastases which would support a role of PSMA PET/ CT imaging in guiding (salvage) treatment strategies [876]. At present there is uncertainty regarding the best treatment if PSMA PET/CT shows metastatic disease outside the pelvis.

6.3.6.3. Impact of post-operative RT and/or ADT in patients with persistent PSA

The benefit of SRT in patients with persistent PSA remains unclear due to a lack of RCTs, however, it would appear that men with a persistent PSA do less well than men with BCR undergoing RT.

Preisser et al., compared oncological outcomes of patients with persistent PSA who received SRT vs. those who did not [864]. In the subgroup of patients with persistent PSA, after 1:1 propensity score matching between patients with SRT vs. no RT, OS rates at ten years after RP were 86.6 vs. 72.6% in the entire cohort (p < 0.01), 86.3 vs. 60.0% in patients with positive surgical margin (p = 0.02), 77.8 vs. 49.0% in pT3b disease (p < 0.001), 79.3 vs. 55.8% in ISUP grade group 1 disease (p < 0.01) and 87.4 vs. 50.5% in pN1 disease (p < 0.01), respectively. Moreover, CSS rates at ten years after RP were 93.7 vs. 81.6% in the entire cohort (p < 0.01), 90.8 vs. 69.7% in patients with positive surgical margin (p = 0.04), 82.7 vs. 55.3% in pT3b disease (p < 0.01), 85.4 vs. 69.7% in ISUP grade group 1 disease (p < 0.01) and 96.2 vs. 55.8% in pN1 disease (p < 0.01), for SRT vs. no RT, respectively. In multivariable models, after 1:1 propensity score matching, SRT was associated with lower risk of death (HR: 0.42, p = 0.02) and lower cancer-specific death (HR: 0.29, p = 0.03). These survival outcomes in patients with persistent PSA who underwent SRT suggest they benefit but outcomes are worse than for men experiencing BCR [877].

It is clear from a number of studies that poor outcomes are driven by the level of pre-RT PSA, the presence of ISUP grade group ≥ 4 in the RP histology and pT3b disease [878-883]. Fossati et al., suggested that only men with a persistent PSA after RP and ISUP grade group ≤ 3 benefit significantly [884], although this is not supported by Preisser et al. [864]. The current data do not allow making any clear treatment decisions.

Addition of ADT may improve PFS [879]. Choo et al., studied the addition of 2-year ADT to immediate RT to the prostate bed in patients with pT3 and/or positive surgical margins after RP [879]. Twenty-nine of the 78 included patients had persistently detectable post-operative PSA. The relapse-free rate was 85% at five years and 68% at 7 years, which was superior to the 5-year progression-free estimates of 74% and 61% in the post-operative RT arms of the EORTC and the SWOG studies, respectively, which included patients with undetectable PSA after RP [835,836]. Patients with persistently detectable post-operative PSA comprised approximately 50% and 12%, respectively, of the study cohorts in the EORTC and the SWOG studies.

In the ARO 96-02, a prospective RCT, 74 patients with PSA persistence (20%) received immediate SRT only (66 Gy per protocol [arm C]). The 10-year clinical relapse-free survival was 63% [878]. The GETUG-22 trial comparing RT with RT plus short-term ADT for post-RP PSA persistence (0.2–2.0 ng/mL) reported good tolerability of the combined treatment. The oncological endpoints are yet to be published [885].

Two SRs addressing persistent PSA confirmed a strong correlation of PSA persistence with poor oncologic outcomes [859,860]. Ploussard et al., also reported that SRT was associated with improved survival outcomes, although the available evidence is of low quality [860].

6.3.6.4. Conclusion

The available data suggest that patients with PSA persistence after RP may benefit from early aggressive multi-modality treatment, however, the lack of prospective RCTs makes firm recommendations difficult.

6.3.6.5. Recommendations for the management of persistent PSA after radical prostatectomy

Recommendations

Strength rating

Offer a prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT scan to men with a persistent prostate-specific antigen (PSA) > 0.2 ng/mL if the results will influence subsequent treatment decisions.

Weak

Treat men with no evidence of metastatic disease with salvage radiotherapy and additional hormonal therapy.

Weak

6.4. Management of PSA-only recurrence after treatment with curative intent

Follow-up will be addressed in Chapter 7 and is not discussed in this section.

6.4.1. Background

Between 27% and 53% of all patients undergoing RP or RT develop a rising PSA (PSA recurrence). Whilst metastatic progression is universally preceded by rising PSA levels, physicians must inform the patient that the natural history of PSA-only recurrence may be prolonged and that a measurable PSA may not necessarily lead to clinically apparent metastatic disease. Physicians treating patients with PSA-only recurrence face a difficult set of decisions in attempting to delay the onset of metastatic disease and death while avoiding overtreating patients whose disease may never affect their OS or QoL. It should be emphasised that the treatment recommendations for these patients should be given after discussion in a multi-disciplinary team.

6.4.1.1. PSA velocity and doubling time

Various PSA kinetics definitions have been proposed with different methods of calculation (log transformed or not) and eligible PSAs:

  • PSA velocity (PSAV): absolute annual increase in serum PSA (ng/mL/year);
  • PSA doubling time (PSA-DT): which measures the exponential increase in serum PSA over time.

Prostate-specific antigen velocity is more simple to calculate by subtracting the initial value from the final value, dividing by time. However, by ignoring middle values, not all PSA values are accurately taken into account.

Prostate-specific antigen-DT is calculated assuming an exponential rise in serum PSA. The formula takes into account the natural logarithm of 2 divided by the slope obtained from fitting a linear regression of the natural log of PSA over time [886]. However, many different PSA-DT calculations have been assessed according to the mathematical formula used and to the included PSA values (number, time period, intervals) [887]. For example, the ‘MSKCC’ method calculates a regression slope integrating all PSA values. Other methods transform PSA before calculating the slope and do not include all PSA values (different time frames and minimal intervals) [888]. O’Brien and colleagues identified more than 20 different definitions of PSAV and PSA-DT and demonstrated that obtained values could vary widely between definitions [888].

However, some rules can be considered for PSA-DT calculation [886]:

  • At least 3 PSA measurements are required;
  • A minimum time period between measurements (4 weeks) is preferable due to potential
  • statistical ‘noise’ when PSA values are obtained too close together (this statement can be
  • reconsidered in case of very active disease);
  • All included PSA values should be obtained within the past twelve months at most, to reflect the current disease activity;
  • PSA-DT is often mentioned in months, or in weeks in very active disease.

These measurements do not provide additional information compared with PSA alone [479,888-890]. In the post-local therapy relapse setting, PSA-DT has been correlated with distant progression and with poorer outcomes after salvage treatments [891,892]. Prostate-specific antigen-DT has been linked with metastasis-free- and OS in non-metastatic CRPC (nmCRPC) and identifies patients with high-risk nmCRPC who benefit from intensified therapy (PSA-DT threshold < ten months) [893].

6.4.2. Controversies in the definitions of clinically relevant PSA relapse

The PSA level that defines treatment failure depends on the primary treatment. Patients with rising PSA after RP or primary RT have different risks of subsequent symptomatic metastatic disease based on various parameters, including the PSA level. Therefore, physicians should carefully interpret BCR endpoints when comparing treatments. Clinicians should interpret a PSA rise in light of the EAU BCR risk groups (see Section 6.3.3).

After RP, the threshold that best predicts further metastases is a PSA > 0.4 ng/mL and rising [894-896]. However, with access to ultra-sensitive PSA testing, a rising PSA much below this level will be a cause for concern for patients.

After primary RT, with or without short-term hormonal manipulation, the RTOG-ASTRO Phoenix Consensus Conference definition of PSA failure (with an accuracy of > 80% for clinical failure) is ‘any PSA increase > 2 ng/mL higher than the PSA nadir value, regardless of the serum concentration of the nadir’ [897].

After HIFU or cryotherapy no endpoints have been validated against clinical progression or survival; therefore, it is not possible to give a firm recommendation of an acceptable PSA threshold after these alternative local treatments [898].

6.4.3. Natural history of biochemical recurrence

Once a PSA recurrence has been diagnosed, it is important to determine whether the recurrence has developed at local or distant sites. A SR and meta-analysis investigated the impact of BCR on clinical endpoints and concluded that patients experiencing BCR are at an increased risk of developing distant metastases, PCa-specific and overall mortality [898]. However, the effect size of BCR as a risk factor for mortality is highly variable. After primary RP its impact ranges from HR 1.03 (95% CI: 1.004–1.06) to HR 2.32 (95% CI: 1.45–3.71) [899,900]. After primary RT, OS rates are approximately 20% lower at eight to ten years follow-up even in men with minimal co-morbidity [901,902]. Still, the variability in reported effect sizes of BCR remains high and suggests that only certain patient subgroups with BCR might be at an increased risk of mortality.

The risk of subsequent metastases, PCa-specific- and overall mortality may be predicted by the initial clinical and pathologic factors (e.g., T-category, PSA, ISUP grade group) and PSA kinetics (PSA-DT and interval to PSA failure), which was further investigated by the SR [898].

For patients with BCR after RP, the following outcomes were found to be associated with significant prognostic factors:

  • distant metastatic recurrence: positive surgical margins, high RP specimen pathological ISUP grade group, high pT category, short PSA-DT, high pre-SRT PSA;
  • prostate-cancer-specific mortality: high RP specimen pathological ISUP grade group, short interval to biochemical failure as defined by investigators, short PSA-DT;
  • overall mortality: high RP specimen pathological ISUP grade group, short interval to biochemical failure, high PSA-DT.

For patients with BCR after RT, the corresponding outcomes are:

  • distant metastatic recurrence: high biopsy ISUP grade group, high cT category, short interval to biochemical failure;
  • prostate-cancer-specific mortality: short interval to biochemical failure;
  • overall mortality: high age, high biopsy ISUP grade group, short interval to biochemical failure, high initial (pre-treatment) PSA.

Based on this meta-analysis, proposal is to stratify patients into two risk categories since not all patients with BCR will have similar outcomes (see Table 6.4.1). The stratification into ‘EAU Low-Risk’ or ‘EAU High-Risk’ BCR after RP has recently been validated in a European cohort [903].

Table 6.4.1: EAU risk categories for patients developing biochemical recurrence


EAU Low Risk BCR

EAU High Risk BCR

After RP

PSA-DT > 1 yr

AND

pathological ISUP grade group < 4

PSA-DT ≤ 1 yr

OR

pathological ISUP grade group 4–5

After RT

interval to biochemical failure > 18 mo

AND

biopsy ISUP grade group < 4

interval to biochemical failure ≤ 18 mo

OR

biopsy ISUP grade group 4–5

6.4.4. The role of imaging in PSA-only recurrence

Imaging is only of value if it leads to a treatment change which results in an improved outcome. In practice, however, there are very limited data available regarding the outcome’s consequent on imaging at recurrence.

6.4.4.1. Assessment of metastases (including nodal)
6.4.4.1.1. Bone scan and abdominopelvic CT

Because BCR after RP or RT precedes clinical metastases by seven to eight years on average [825,904], the diagnostic yield of conventional imaging techniques (bone scan and abdominopelvic CT) is low in asymptomatic patients [905]. In men with PSA-only recurrence after RP the probability of a positive bone scan is < 5%, when the PSA level is < 7 ng/mL [906,907]. Only 11–14% of patients with BCR after RP have a positive CT [906]. In a series of 132 men with BCR after RP the mean PSA level and PSA velocity associated with a positive CT were 27.4 ng/mL and 1.8 ng/mL/month, respectively [908].

6.4.4.1.2. Choline PET/CT

In two different meta-analyses the combined sensitivities and specificities of choline PET/CT for all sites of recurrence in patients with BCR were 86–89% and 89–93%, respectively [909,910]. Choline PET/CT may detect multiple bone metastases in patients showing a single metastasis on bone scan [911] and may be positive for bone metastases in up to 15% of patients with BCR after RP and negative bone scan [912]. The specificity of choline PET/CT is also higher than bone scan, with fewer false positive and indeterminate findings [416]. Detection of LN metastases using choline PET/CT remains limited by the relatively poor sensitivity of the technique (see Section 5.3.2.3). Choline PET/CT sensitivity is strongly dependent on the PSA level and kinetics [424,913,914]. In patients with BCR after RP, PET/CT detection rates are only 5–24% when the PSA level is < 1 ng/mL but rise to 67–100% when the PSA level is > 5 ng/mL. Despite its limitations, choline PET/CT may change medical management in 18–48% of patients with BCR after primary treatment [915-917].

Choline PET/CT should only be recommended in patients fit enough for curative loco-regional salvage treatment.

6.4.4.1.3. Fluoride PET/CT

18F-NaF PET/CT has a higher sensitivity than bone scan in detecting bone metastases [918]. However, 18F -NaF PET/CT is limited by a relative lack of specificity and by the fact that it does not assess soft-tissue metastases [919].

6.4.4.1.4. Fluciclovine PET/CT

18F-Fluciclovine PET/CT has been approved in the U.S. and Europe and it is therefore one of the PCa-specific radiotracers widely commercially available [919-922].

18F-Fluciclovine PET/CT has a slightly higher sensitivity than choline PET/CT in detecting the site of relapse in BCR [923]. In a multi-centre trial evaluating 596 patients with BCR in a mixed population, fluciclovine PET/CT showed an overall detection rate of 67.7%; lesions could be visualised either at local level (38.7%) or in pelvic LNs (32.6%) [924]. As for choline PET/CT, fluciclovine PET/CT sensitivity is dependent on the PSA level, with a sensitivity likely inferior to 50% at PSA level < 1 ng/mL.

In a prospective RCT evaluating the impact of 18F-fluciclovine PET/CT on SRT management decisions in patients with recurrence post-prostatectomy, in 28 of 79 (35.4%) patients overall radiotherapeutic management changed following 18F-fluciclovine PET/CT [925]. 18F-Fluciclovine PET/CT had a significantly higher positivity rate than conventional imaging (abdominopelvic CT or MRI plus bone scan) for whole body (79.7% vs. 13.9%, p < 0.001), prostate bed (69.6% vs. 5.1%, p < 0.001), and pelvic LNs (38.0% vs. 10.1%, p < 0.001) [925]. However, as yet, no data demonstrating that these changes translate into a survival benefit are available.

6.4.4.1.5. Prostate-specific membrane antigen based PET/CT

PSMA PET/CT has shown good potential in patients with BCR, although most studies are limited by their retrospective design. Reported predictors of 68Ga-PSMA PET in the recurrence setting were updated based on a high-volume series (see Table 6.4.2) [867]. High sensitivity (75%) and specificity (99%) were observed on per-lesion analysis.

Table 6.4.2: PSMA-positivity separated by PSA level category 

PSA (ng/mL)

68Ga-PMSA PET positivity

< 0.2

33% (CI: 16–51)

02–0.49

45% (CI: 39–52)

0.5–0.99

59% (CI: 50–68)

1.0–1.99

75% (CI: 66–84)

2.0+

95% (CI: 92–97)

PSA = prostate-specific antigen; 68Ga-PSMA PET = Gallium-68 prostate-specific membrane antigen positronemission tomography.

PSMA PET/CT seems substantially more sensitive than choline PET/CT, especially for PSA levels < 1 ng/mL [926,927]. In a study of 314 patients with BCR after treatment and a median PSA level of 0.83 ng/mL, 68Ga-PSMA PET/CT was positive in 197 patients (67%) [928]. In another prospective multi-centre trial including 635 patients with BCR after RP (41%), RT (27%), or both (32%), PPV for 68Ga-PSMA PET/CT was 0.84 (95% CI: 0.75–0.90) by histopathologic validation (primary endpoint, n = 87) and 0.92 (95% CI: 0.75–0.90) by a composite reference standard. Detection rates significantly increased with PSA value [929].

A prospective multi-centre, multi-reader, open-label, phase II/III trial (OSPREY) evaluated the diagnostic performance of 18F-DCFPyL in patients with presumptive radiologic evidence of recurrent or metastatic PCa on conventional imaging [837]. Median sensitivity and median PPV were 95.8% (95% CI: 87.8%–99.0%) and 81.9% (95% CI: 73.7%–90.2%), respectively.

Another prospective study evaluated the diagnostic performance of 18F-DCFPyL in 208 men with BCR after RP or RT. The primary endpoint, the correct localisation rate was achieved, demonstrating positive findings on 18F-DCFPyL PET/CT in the setting of negative standard conventional imaging [930]. At present there are no conclusive data about comparison of such tracers [931].

A prospective, open label, cross-over study, the PYTHON trial, has compared the per-patient detection rates (DR) of 18F-DCFPyL versus 18F-fluoromethylcholine PET/CT, in biochemical recurrence (BCR) setting. A total of 201 high-risk PCa patients with first BCR after radical prostatectomy or radiation therapy have completed the study. The per-patient DR was significantly higher for 18F-DCFPyL compared to 18F-fluoromethylcholine PET/CT (58% (117/201 patients) vs. 40% (81/201 patients), p < 0.0001). DR increased with higher PSA values for both tracers (PSA ≤ 0.5 ng/ml: 26/74 (35%) vs. 22/74 (30%); PSA 0.5 to ≤ 1.0 ng/ml: 17/31 (55%) vs. 10/31 (32%); PSA 1.01 to < 2.0 ng/ml: 13/19 (68%) vs. 6/19 (32%);PSA > 2.0: 50/57 (88%) vs. 39/57 (68%) for 18FDCFPyL- and 18F-fluoromethylcholine -PET/CT, respectively) [932]. Comparable results were found in a phase III trial of 18F-PSMA-1007 versus 18F-Fluorocholine PET/CT for the localisation of prostate cancer biochemical recurrence. In this prospective, randomised, crossover multi-centre study, the overall correct detection rates were significantly higher for 18F-PSMA-1007 than for 18F-fluorocholine when undetermined findings were considered positive for malignancy (0.82 vs. 0.65; p < 0.0001) [933].

6.4.4.1.6. Whole-body and axial MRI

Whole body MRI has not been widely evaluated in BCR because of its limited value in the detection of early metastatic involvement in normal-sized LNs [414,427,934]. In a prospective series of 68 patients with BCR, the diagnostic performance of DW-MRI was significantly lower than that of 68Ga-PSMA PET/CT and 18NaF PET/CT for diagnosing bone metastases [935].

6.4.4.2. Assessment of local recurrences
6.4.4.2.1. Local recurrence after radical prostatectomy

Because the sensitivity of anastomotic biopsies is low, especially for PSA levels < 1 ng/mL [905], SRT is usually decided on the basis of BCR without histological proof of local recurrence. The dose delivered to the prostatic fossa tends to be uniform since it has not been demonstrated that a focal dose escalation at the site of recurrence improves the outcome. Therefore, most patients undergo SRT without local imaging.

Magnetic resonance imaging can detect local recurrences in the prostatic bed. The PSA threshold for MRI positivity seems between 0.3 and 0.5 ng/mL; PSA kinetics also influence the MRI positivity, even at low PSA values [936]. Two single-centre studies found that a negative MRI was an independent predictor of failure of SRT [937,938]. Conversely, a small (≤0.4 cc) relapse located at the vesico-urethral anastomosis is associated with excellent prognosis at salvage RT [939]. The Prostate Imaging for Recurrence Reporting (PI-RR) system has been recently launched to standardise MRI interpretation in the context of BCR after RP or RT [940]. Initial assessment suggests good reproducibility of the score [941].

Choline PET/CT is less sensitive for local relapse than MRI but detects more regional and distant metastases [942].

The detection rates of 68Ga-PSMA PET/CT in patients with BCR after RP increase with the PSA level [943]. PSMA PET/CT studies showed that a substantial part of recurrences after RP were located outside the prostatic fossa, even at low PSA levels [868,944]. Combining 68Ga-PSMA PET and MRI may improve the detection of local recurrences, as compared to 68Ga-PSMA PET/CT alone [945-947].

The EMPIRE-1, a single-centre, open-label, phase II/III RCT included 365 patients with detectable PSA after RP, but negative results on conventional imaging. They were randomised to RT directed by conventional imaging alone or to conventional imaging plus 18F-fluciclovine-PET/CT; patients with M1 disease in the PET/CT group (n = 4) were excluded Patients with cN1 were irradiated to the pelvic nodes, but without a boost to the metastases. After a median follow-up of 3.5 years, the PET/CT group was significantly associated with longer event-free survival (HR: 2.04, 95% CI: 1.06–3.93, p = 0.0327) [948].

6.4.4.2.2. Local recurrence after radiation therapy

In patients with BCR after RT, biopsy status is a major predictor of outcome, provided the biopsies are obtained 18–24 months after initial treatment. Given the morbidity of local salvage options it is necessary to obtain histological proof of the local recurrence before treating the patient [905].

MRI has yielded excellent results in identifying local recurrence and can be used for biopsy targeting and guiding local salvage treatment [905,949,950], even if it slightly underestimates the volume of the local recurrence [951]. Prostate-specific membrane antigen PET/CT can also detect local recurrences after RT [867] and concordance between PSMA PET/CT and MRI is highly suggestive of cancer recurrence [952].

6.4.4.3. Summary of evidence of imaging in case of biochemical recurrence

In patients with BCR imaging can detect both local recurrences and distant metastases, however, the sensitivity of detection depends on the PSA level. After RP, PSMA PET/CT is the imaging modality with the highest sensitivity at low PSA levels (< 0.5 ng/mL) and may help distinguishing patients with recurrences confined to the prostatic fossa from those with distant metastases which may impact the design and use of post-RP SRT. After RT, MRI has shown excellent results at detecting local recurrences and guiding prostate biopsy. Given the substantial morbidity of post-RT local salvage treatments, distant metastases must be ruled out in patients with local recurrences and who are fit for these salvage therapies. Choline-,fluciclovine- or PSMA-PET/CT can be used to detect metastases in these patients but for this indication PSMA PET/CT seems the most sensitive technique.

6.4.4.4. Summary of evidence and guidelines for imaging in patients with biochemical recurrence

Recommendations

Strength rating

Prostate-specific antigen (PSA) recurrence after radical prostatectomy

Perform prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) if the PSA level is > 0.2 ng/mL and if the results will influence subsequent treatment decisions (EAU BCR risk groups).

Weak

In case PSMA PET/CT is not available, and the PSA level is ≥ 1 ng/mL, perform fluciclovine PET/CT or choline PET/CT imaging if the results will influence subsequent treatment decisions.

Weak

PSA recurrence after radiotherapy

Perform prostate magnetic resonance imaging to localise abnormal areas and guide biopsies in patients fit for local salvage therapy.

Weak

Perform PSMA PET/CT (if available) or fluciclovine PET/CT or choline PET/CT in patients fit for curative salvage treatment.

Strong

6.4.5. Treatment of PSA-only recurrences

The timing and treatment modality for PSA-only recurrences after RP or RT remain a matter of controversy based on the limited evidence.

6.4.5.1. Treatment of PSA-only recurrences after radical prostatectomy
6.4.5.1.1. Salvage radiotherapy for PSA-only recurrence after radical prostatectomy (cTxcN0M0, withoutPET/CT)

Early SRT provides the possibility of cure for patients with an increasing PSA after RP. Boorjian et al., reported a 75% reduced risk of systemic progression with SRT when comparing 856 SRT patients with 1,801 non-SRT patients [953]. The RAVES and RADICAL trials assessing SRT in post-RP patients with PSA levels exceeding 0.1–0.2 ng/mL showed 5-year freedom from BCR and BCR-free survival rates of 88% [841,954]. Tilki et al. demonstrated the results of a matched pair analysis of 1832 patients with BCR, 32.9% (n = 603) received SRT without ADT, 1229 (67,1%) had a observational strategy. The median follow-up was 95.9 months. Median total SRT dose was 70.2 Gy. After 1:1 propensity score matching, at fiveteen years after RP, MFS and OS rates were 84.3 versus 76.9% (p < 0.001) and 85.3 versus 74.4% (p = 0.04) for SRT and noRT, respectively [955].

The PSA level at BCR was shown to be prognostic [953]. More than 60% of patients who are treated before the PSA level rises to > 0.5 ng/mL will achieve an undetectable PSA level [956-958], corresponding to a ~80% chance of being progression-free five years later [959]. A retrospective analysis of 635 patients who were followed after RP and experienced BCR and/or local recurrence and either received no salvage treatment (n = 397) or SRT alone (n = 160) within two years of BCR showed that SRT was associated with a 3-fold increase in PCa-specific survival relative to those who received no salvage treatment (p < 0.001). Salvage RT has been shown to be effective mainly in patients with a short PSA-DT [960].

In a retrospective multi-centre study including 25,551 patients with at most one high-risk factor after RP (ISUP grade group 4-5 or pT3/4), initiating sRT above a PSA level of 0.25 ng/mL was associated with increased ACM-risk. After a median follow-up of six years, patients who received sRT at a PSA level >0.25 ng/mL had a significantly higher ACM-risk (AHR, 1.49; 95% CI, 1.11 to 2.00; P =.008) compared with men who received sRT when the PSA was ≤0.25 mg/mL [961]. For an overview of SRT see Table 6.4.3.

The EAU BCR definitions have been externally validated and may be helpful for individualised treatment decisions [898,903]. Despite the indication for salvage RT, a ‘wait and see‘ strategy remains an option for the EAU BCR ‘Low-Risk’ group [898,903].

Although biochemical progression is now widely accepted as a surrogate marker of PCa recurrence; metastatic disease, disease-specific and OS are more meaningful endpoints to support clinical decision-making. A SR and meta-analysis on the impact of BCR after RP reports SRT to be favourable for OS and PCSM. In particular SRT should be initiated in patients with rapid PSA kinetics after RP and with a PSA cut-off of 0.4 ng/mL [898]. An international multi-institutional analysis of pooled data from RCTs has suggested that MFS is the most valid surrogate endpoint with respect to impact on OS [962,963]. Table 6.4.4 summarises results of recent studies on clinical endpoints after SRT.

Table 6.4.3: Selected studies of post-prostatectomy salvage radiotherapy, stratified by pre-salvage radiotherapy PSA level* (cTxcN0M0, without PET/CT)

Study

n

Median FU (mo)

pre-SRT PSA (ng/mL) median

RT dose
ADT

bNED/PFS (year)

5-yr. results

Bartkowiak, et al. 2018 [964]

464

71

0.31

66.6 Gy

54% (5.9)

73% vs. 56%; PSA

< 0.2 vs. > 0.2 ng/mL
p < 0.0001

Stish, et al.

2016 [956]

1,106

107

0.6

68 Gy

16% ADT

50% (5)

36% (10)

44% vs. 58%; PSA

< 0.5 vs. > 0.5 ng/mL
p < 0.001

Tendulkar,
et al. 2016 [965]

2,460

60

0.5

66 Gy

16% ADT

56% (5)

Pre-SRT PSA

71% 0.01–0.2 ng/mL

63% 0.21–0.5 ng/mL

54% 0.51–1.0 ng/mL

43% 1.01–2.0 ng/mL

37% > 2.0 ng/mL
p < 0.001

Tilki et al. 2023 [961]

25,551

SRT at:

PSA <0.25

N=1,556

PSA>0.25

N=1.677

No RT: N=21,645

72

Not given

Med.

68.4 Gy

SRT+ADT:1489

ART:N= 673

ADT: 208


Not given

ACM (six years):

 

HR 1.49 of higher risk when SRT at start was >0.25

(p=0.008)

*Androgen deprivation therapy can influence the outcome ‘biochemically no evidence of disease (bNED)’ or ‘progression-free survival’. To facilitate comparisons, 5-year bNED/PFS read-outs from Kaplan-Meier plots are included.
ADT = androgen deprivation therapy; bNED = biochemically no evidence of disease; FU = follow up; mo = months; n = number of patients; PFS = progression-free survival; PSA = prostate-specific antigen; SRT = salvage radiotherapy; yr = year.

Table 6.4.4: Selected studies reporting clinical endpoints after SRT (cTxcN0M0, without PET/CT)(the majority of included patients did not receive ADT)

Study

n

Median FU (mo)

Regimen

Outcome

Bartkowiak, et al. 2018 [964]

464

71

66.6 (59.4-72) Gy no ADT

5.9 yr. OS

post-SRT PSA < 0.1 ng/mL 98%

post-SRT PSA > 0.1 ng/mL 92%

p = 0.005

Jackson,

et al. 2014 [966]

448

64

68.4 Gy no ADT

5 yr. DM

post-SRT PSA < 0.1 ng/mL 5%

post-SRT PSA > 0.1 ng/mL 29%

p < 0.0001

5 yr. DSM

post-SRT PSA < 0.1 ng/mL 2%

post-SRT PSA > 0.1 ng/mL 7%

p < 0.0001

OS

post-SRT PSA < 0.1 ng/mL 97%

post-SRT PSA > 0.1 ng/mL 90%

p < 0.0001

Stish,

et al. 2016 [956]

1,106

107

68 (64.8-70.2) Gy 39% 2D treatment planning

incl. 16% ADT

5 and 8.9 yr. DM

SRT: PSA < 0.5 ng/mL 7% and 12%

SRT: PSA > 0.5 ng/mL 14% and 23%

p < 0.001

5 and 8.9 yr. DSM

SRT: PSA < 0.5 ng/mL < 1% and 6%

SRT: PSA > 0.5 ng/mL 5% and 10%

p = 0.02 5 and 8.9 yr. OS

SRT: PSA < 0.5 ng/mL 94% and 86%

SRT: PSA > 0.5 ng/mL 91% and 78%

p = 0.14

Tendulkar,

et al. 2016 [965]

2,460

60

66 (64.8-68.4) Gy incl. 16% ADT

10-yr. DM (19% all patients)

Pre-SRT PSA

9% 0.01–0.2 ng/mL

15% 0.21–0.5 ng/mL

19% 0.51–1.0 ng/mL

20% 1.01–2.0 ng/mL

37% > 2.0 ng/mL

p < 0.001

ADT = androgen deprivation therapy; DM = distant metastasis; DSM = disease specific mortality; FU = follow up; mo. = month; n = number of patients; OS = overall survival; PSA = prostate specific antigen; SRT = salvage radiotherapy.

6.4.5.1.2. Salvage radiotherapy combined with androgen deprivation therapy (cTxcN0, without PET/CT)

Data from RTOG 9601 suggest both CSS and OS benefit when adding two years of bicalutamide (150 mg o.d.) to SRT [967]. According to GETUG-AFU 16 also 6-months treatment with a LHRH-analogue can significantly improve 10-year BCR, biochemical PFS and, modestly, MFS. However, SRT combined with either goserelin or placebo showed similar DSS and OS rates [968]. In addition, Pollack et al., reported on the results of a randomised 3-arm phase III trial (NRG Oncology/RTOG 0534 SPPORT) adding six months treatment with a LHRH-analogue to SRT of the prostate bed (PBRT) (group 2) compared with PBRT alone (group 1) or the former combination with PBRT-RT and pelvic LN RT (PLNRT) (group 3) [969]. The primary endpoint was freedom from progression (FFP) after five years. However, using the phoenix-definition of biochemical progression (nadir + 2 ng/mL used for definitive RT), and not the criterion of nadir + 0.2, as is used commonly (but without clear evidence) will have resulted in a later diagnosis of progression in the SPPORT trial.

With a median follow-up of 8.2 years of the surviving patients FFP increased significant for group 3 (87.4%) compared with group 2 (81.3%) (p = 0.0027) and group 1 (70.9%) (p < 0.0001) The difference between group 2 and group 1 was also significant (p < 0.0001). Distant metastasis incidence rates were lowest in group 3 and were lower compared with group 1 (PBRT only, HR: 0.52) similar to the rate of PCa deaths (HR: 0.51). No significant difference was seen for OS. There was a significantly higher risk of both acute- and late side effects in group 3. Therefore, the role of additional PLNRT remains unclear and should be further proven in RCTs including PSMA PET-CT [970]. Table 6.4.5 provides an overview of these three RCTs.

These RCTs support adding ADT to SRT. However, when interpreting these data it has to be kept in mind that RTOG 9601 used outdated radiation dosages (< 66 Gy) and technique. The question with respect to the patient risk profile, whether to offer combination treatment or not, and the optimal combination (LHRH or bicalutamide) remains, as yet, unsolved. The EAU BCR risk classification may offer guidance in this respect [898].

One of these RCTs reports improved OS (RTOG 96-01) and the other (GETUG-AFU 16) improved MFS but due to methodological discrepancies and also related to follow-up and risk, it is, as yet, not evident which patients should receive ADT, which type of ADT, and for how long. Men at high risk of further progression (e.g., with a PSA ≥ 0.7 ng/mL and GS ≥ 8) may benefit from SRT combined with two years of ADT; for those at lower risk (e.g., PSA < 0.7 ng/mL and GS = 8) SRT combined with six months of ADT may be sufficient. Men with a low-risk profile (PSA < 0.5 ng/mL and GS < 8) may receive SRT alone. In a sub-analysis of men with a PSA of 0.61 to 1.5 (n = 253) there was an OS benefit associated with antiandrogen assignment (HR: 0.61, 95% CI: 0.39–0.94) [971]. In those receiving early SRT (PSA 0.6 ng/mL, n = 389), there was no improvement in OS (HR: 1.16, 95% CI: 0.79–1.70), with increased other-cause mortality (sub-distribution HR: 1.94, 95% CI: 1.17–3.20, p = 0.01) and increased odds of late grades 3–5 cardiac and neurologic toxic side effects (OR: 3.57, 95% CI: 1.09–15.97, p = 0.05). These results suggest that pre-SRT PSA level may be a prognostic biomarker for outcomes of anti-androgen treatment with SRT. In patients receiving late SRT (PSA > 0.6 ng/mL), HT was associated with improved outcomes. In men receiving early SRT (PSA < 0.6 ng/mL), long-term anti-androgen treatment was not associated with improved OS [971].

A SR addressing the benefit from combining HT with SRT suggested risk stratification of patients based on the pre-SRT PSA (< 0.5, 0.6–1, > 1 ng/mL), margin status and ISUP grade as a framework to individualise treatment [972].

Table 6.4.5: Randomised controlled trials comparing salvage radiotherapy combined with androgen deprivation therapy vs. salvage radiotherapy alone

Study

n

Risk groups

Median FU (mo)

Regimen

Outcome

GETUG-AFU 16 2019 [968]

369 RT + ADT

374 RT

ISUP grade

< 2/3 89%


ISUP grade
> 4 11%

cN0

112

66 Gy PBRT + 6 mo. LHRH analogue


66 Gy BPRT

10-yr.

PFS: RT + ADT, 64%

PFS: RT, 49%

p < 0.0001

MFS: RT + ADT, 75%

MFS: RT, 69%

p = 0.034

RTOG 9601
2017 [967]

384 RT + ADT

376 RT

pT2 R1, pT3 cN0

156

64.8 Gy PBRT + bicalutamide 24 mo.


64.8 Gy PBRT + placebo

12-yr.

cumulative DM

RT + ADT: 14%

RT + placebo: 23%

p = 0.005

OS

RT + ADT: 76%

RT + placebo: 71%

p = 0.04

DSM

RT + ADT: 5.8%

RT + placebo: 13.4%

p < 0.001

NRG Oncology/RTOG 0534 SPPORT [969]

564 PBRT + alone


578 PBRT + ADT


574 PBRT +

PLNRT + ADT

pT2 or pT3

ISUP < 5

Pre SRT
PSA: 0.1-2.0

survivors:

8.2 years

64.8–70.2 Gy PBRT


64.8–70.2 Gy PBRT +

6 mo. LHRH analogue


64.8–70.2 Gy PBRT + 45 Gy PLNRT +

6 mo. LHRH analogue

5-yr. FFP (primary endpoint)

70.9% Group 1

81.3% Group 2

87.4% Group 3

Comparisons :

G 3 vs. G 1:

p < 0.0001

G 2 vs. G 1

p < 0.0001

G 3 vs. G 2

p < 0.0027

ADT = androgen deprivation therapy; DM = distant metastasis; DSM = disease specific mortality; PFS = progression free survival; FFP = Freedom From Progression; FU = follow-up; LHRH = luteinising hormone-releasing hormone; MFS = metastasis-free survival; OS = overall survival; PFS = progression-free survival; mo = months; n = number of patients; RT = radiotherapy; yr = year, PBRT = prostate bed radiotherapy; PLNRT = pelvic lymph node radiotherapy.

6.4.5.1.2.1. Target volume, dose, toxicity

There have been various attempts to define common outlines for ‘clinical target volumes‘ for pN0 PCa [973,974] and for organs at risk of normal tissue complications [973]. However, given the variations of techniques and dose-constraints, a satisfactory consensus has not yet been achieved. A benefit in biochemical PFS but not MFS has been reported in patients receiving whole pelvis SRT (± ADT) but the advantages must be weighed against possible side effects [970]. This is supported by data from the SPPORTTrial (NRG Oncology/RTOG 0534 SPPORT) but it remains controversial [969].

The optimal SRT dose has not been well defined. It should be at least 64 Gy to the prostatic fossa (± the base of the SVs, depending on the pathological stage after RP) [846,957,975]. In a SR, the pre- SRT PSA level and SRT dose both correlated with BCR, showing that relapse-free survival decreased by 2.4% per 0.1 ng/mL PSA and improved by 2.6% per Gy, suggesting that the treatment dose above 70 Gy should be administered at the lowest possible PSA level [976]. The combination of pT stage, margin status and ISUP grade group and the PSA at SRT seems to define the risk of biochemical progression, metastasis and overall mortality [838,977,978]. In a study on 894 node-negative PCa patients, doses ranging from 64 to > 74 Gy were assigned to twelve risk groups defined by their pre-SRT PSA classes < 0.1, 0.1–0.2, 0.2–0.4, and > 0.4 ng/mL and ISUP grade group < 1 vs. 2/3 vs. > 4 [979]. The updated Stephenson nomograms incorporate the SRT and ADT doses as predictive factors for biochemical failure and distant metastasis [965].

Two RCT’s were published (Table 6.4.6). Intensity-modulated radiation therapy plus IGRT was used in 57% of the patients in the SAKK-trial [980] and in all patients of a Chinese trial [981]. No patient had a PSMA PET/CT before randomisation. The primary endpoint in both trials was ‘freedom from biochemical progression’, which was not significantly improved with higher doses. However, in the Chinese trial a subgroup analysis showed a significant improvement of this endpoint for patients with Gleason 8-10 tumours (79.7% vs. 55%, p = 0.049) [981]. In this trial, patients were treated with ART or SRT and the number of patients was relatively small (n = 144). At this time it seems difficult to draw final conclusions about the optimal total RT-dose and longer follow-up should be awaited.

Table 6.4.6: Randomised trials investigating dose escalation for SRT without ADT and without PET-CT

Trial

n

PCa condition

Radiotherapy

Dose

Follow-up (median)

Outcome

Results

SAKK 09/10

trial, 2021

[846]

350

pT2a-3b

R0 – R1

pN0 or cN0

PSA post op

undetectable

(< 0.1 ng/mL)

or persistent

(> 0.1 ng/mL < 0.4 ng/mL)

64 Gy vs.70 Gy


No ADT allowed


VMAT+ IGRT: 57%

3-D planning: 43%

6.2 yr.

Primary endpoint:

FFBP

6 yr. FFBP:

62% vs. 61%

OS: no difference


Late side effects:

GI grade 2:

7.3% vs. 20%

GI grade 3:

4.2% vs. 2.3%

p for ≥ grade 2/3:

0.009

Phase-III-Trial

Qi X, et al.,

2020 [981]

144

ART: 33%

SRT: 67%

pT2-4

R0-R1

pN0 or cN0

Med. PSA pre-RT: 0.2 ng/mL

66 Gy vs.

72 Gy

All patients

VMAT+ IGRT

No ADT allowed


High risk

(pT3-4, GS: 8-10,

PSA>20 ng/mL):

whole pelvis RT:

126 (87.5%)

49 mo.

Primary endpoint:

FFBP

4 yr. FFBP:

75.9% vs. 82.6%

(p > 0.05)

High risk (GS: 8–10):

55.7% vs. 79.7%

p < 0.049)


Late side effects:

GI + GU grade 2

p > 0.05

No grade 3

ADT = androgen deprivation therapy; ART = adjuvant radiotherapy; FFBP = freedom from biochemical failure; GI = gastro-intestinal; GU = genito-urinary; Gy = Gray; IGRT = image guided radiotherapy; mo = month; n = number of patients; PSA = prostate-specific antigen; RT = radiotherapy; SRT = y = year; vs. = versus; VMAT = volumetric arc radiation therapy.

Salvage RT is associated with toxicity. In one report on 464 SRT patients receiving median 66.6 (max. 72) Gy, acute grade 2 toxicity was recorded in 4.7% for both the GI and GU tract. Two men had late grade 3 reactions of the GI tract, but overall, severe GU tract toxicity was not observed. Late grade 2 complications occurred in 4.7% (GI tract) and 4.1% (GU tract), respectively, and 4.5% of the patients developed moderate urethral stricture [964].

In a RCT on dose escalation for SRT (n = 350), acute grade 2 and 3 GU toxicity was observed in 13.0% and 0.6%, respectively, with 64 Gy and in 16.6% and 1.7%, respectively, with 70 Gy. Gastro-intestinal tract grades 2 and 3 toxicity occurred in 16.0% and 0.6%, respectively, with 64 Gy, and in 15.4% and 2.3%, respectively, with 70 Gy [982,983]. Late grade 2 and 3 GI toxicity was significantly increased with higher doses but without significant differences in QoL. In this study, however, the rectal wall dose constraints were rather permissive and in 44% of the patients outdated 3-D-techniques were used [980].

With dose escalation over 72 Gy and/or up to a median of 76 Gy, the rate of severe side effects, especially GU symptoms, clearly increases, even with newer planning and treatment techniques [984,985]. In particular, when compared with 3D-CRT, IMRT was associated with a reduction in grade 2 GI toxicity from 10.2 to 1.9% (p = 0.02) but no effect on the relatively high level of GU toxicity was shown (5-year, 3D-CRT 15.8% vs. IMRT 16.8%) [984]. However, in a RCT comparing 66 Gy and 72 Gy with all patients having IMRT plus IGRT (n = 144), no significant differences for GI and GU-toxicity was demonstrated [981]. After a median salvage IMRT dose of 76 Gy however, the 5-year risk of grade 2–3 toxicity rose to 22% for GU and 8% for GI symptoms, respectively [985]. Doses of at least 64 Gy and up to 72 Gy in patients without PET/CT can be recommended [964,982].

6.4.5.1.2.2. Salvage radiotherapy with or without ADT (cTx cN0/1) with PET/CT

In a prospective multi-centre study of 323 patients with BCR, PSMA PET/CT changed the management intent in 62% of patients as compared to conventional staging. This was due to a significant reduction in the number of men in whom the site of disease recurrence was unknown (77% vs. 19%, p < 0.001) and a significant increase in the number of men with metastatic disease (11% vs. 57%) [986].

A prospective study in a subgroup of 119 BCR patients with low PSA (< 0.5 ng/mL) reported a change in the intended treatment in 30.2% of patients [868]; however, no data exist on the impact on final outcome.

Another prospective study in 272 patients with early biochemical recurrent PCa after RP showed that 68Ga-PSMA PET/CT may tailor further therapy decisions (e.g., local vs. systemic treatment) at low PSA values (0.2–1 ng/mL) [870].

A single-centre study retrospectively assessed 164 men who underwent imaging with PSMA PET/CT for a rising PSA after RP with PSA levels < 0.5 ng/mL. In men with a negative PSMA PET/CT who received SRT, 85% (23 out of 27) demonstrated a treatment response compared to a further PSA increase in 65% of those not treated (22 out of 34). In the 36/99 men with disease confined to the prostate fossa on PSMA, 83% (29 out of 36) responded to SRT [987]. Thus, PSMA PET/CT might stratify men into a group with high response (negative findings or recurrence confined to the prostate) and poor response (positive nodes or distant disease) to SRT.

A multi-centre retrospective study evaluated patients who underwent SRT for BCR after RP, without any signs of distant metastatic disease on PET/CT. After case-controle matching, two cohorts (n = 108 patients each), with and without PSMA PET/CT prior to SRT were analysed. In the cohort without PSMA PET/CT, 23 patients (21%) had BCR at one year after SRT vs. nine patients (8%) who underwent restaging with PSMA PET/C prior to SRT (p = 0.007). PSMA-PET/CT was found to be associated with an improved oncological outcome in patients with BCR after RP, receiving SRT to the prostatic fossa [988]. It is worth mentioning that in this study the median biologically effective radiation dose administered in the PSMA-cohort was significantly higher than in the historical cohort (70 Gy vs. 66 Gy, respectively, p < 0.001). However, in the SAKK 09/10 randomised phase-III-trial (all patients without PET-CT before SRT) the biochemical progression rate after SRT between patients who underwent 64 Gy or 70 Gy to the prostate bed, without HT for BCR, did not differ significantly [980]. Therefore, it is questionable whether this difference in administered radiation dose influenced the outcome in both cohorts. As there are no prospective phase III data including PET-CT before SRT (in particular not for PCa-specific survival or OS) these results have to be confirmed before a recommendation can be provided.

A single-centre open-label, phase II/III RCT (EMPIRE-1) evaluated the role of 18F-fluciclovine-PET/CT compared with conventional imaging for SRT. Three hundred and sixty five patients with detectable PSA after RP but negative results on conventional imaging, were randomised to RT directed by conventional imaging alone or to conventional imaging plus PET/CT; patients with M1 disease in the PET/CT group (n = 4) were excluded. Patients with cN1 were irradiated to the pelvic lymphatics but without a boost to the metastasis. Median follow 103 up was 3.5 years. In adjusted analyses, the study group was significantly associated with event-free survival (HR: 2.04, 95% CI: 1.06–3.93, p = 0.0327) [948].

6.4.5.1.2.3. Nodal-directed therapy for rcN1 (with PET/CT)

Radiolabelled PSMA PET/CT is increasingly used as a diagnostic tool to assess metastatic disease burden in patients with BCR following prior definitive therapy. A review including 30 studies and 4,476 patients showed overall estimates of positivity in a restaging setting of 38% in pelvic LNs and 13% in extra-pelvic LN metastases [867]. The percentage positivity of PSMA PET/CT was proven to increase with higher PSA values [867]. Results of this review demonstrated a high sensitivity and specificity of 68Ga-PSMA in advanced PCa, with a per-lesion-analysed sensitivity and specificity of 75% and 99%, respectively.

A large retrospective international study included patients with LN-recurrent PCa (cN1 and M1a) and PSA progression following multi-modality treatment (surgery and post-operative RT) [989]. The aim of the study was to compare standard of care (SOC) with nodal metastasis-directed therapy (MDT). The nodal MDT-group showed significantly better CSS than the SOC control group (5-year survival 98.6% vs. 95.7%, p < 0.01, respectively) [989].

Another retrospective study compared SBRT with elective nodal irradiation (ENRT) in nodal oligo-recurrent PCa (n = 506 patients, 365 of which with N1 pelvic recurrence). With a median follow-up of 36 months, ENRT (n = 197) was associated with a significant reduction of nodal recurrences (p < 0.001), compared with SABR (n = 309) of 2% vs. 18%, respectively. In multi-variable analysis, patients with one LN at recurrence had longer adjusted MFS after ENRT (HR: 0.50, 95% CI: 0.30–0.85, p = 0.009). The tendency to relapse was higher for pelvic- than extra-pelvic nodes (p < 0.001) [990]. For patients presenting with two or more (extra)pelvic LNs, adjusted MFS was not significantly different (HR: 0.92, 95% CI: 0.54– 1.59, p = 0.8). In these situations, SABR should be used in highly selected patients in prospective cohorts or clinical trials only, before any recommendations can be made. For MDT in M1 patients see Section 6.4.7.

6.4.5.1.3. Salvage lymph node dissection

The surgical management of recurrent nodal metastases in the pelvis has been the topic of several retrospective analyses [991-993] and a SR [994]. The reported five-year BCR-free survival rates ranged from 6% to 31%. Five-year OS was approximately 84% [994]. Biochemical recurrence rates were found to be dependent on PSA at salvage surgery and location and number of positive nodes [995]. Addition of RT to the lymphatic template after salvage LN dissection may improve the BCR rate [996]. In a multi-centre retrospective study long-term outcomes of 189 patients who underwent salvage LN dissection were reported to be worse than previously described in studies with shorter follow-up [997]. Biochemical recurrence (BCR)-free survival at ten years was 11%. Patients with a PSA response after salvage LN dissection and patients receiving ADT within six months from salvage LN dissection had a lower risk of death from PCa [997]. The majority of the patients (81%) had received a choline PET and median PSA at salvage LN dissection was 2.5 ng/mL. In a cohort study including patients treated with salvage LN dissection via PSMA--radioguided surgery (PSMA-RGS), two-year BCR-free survival rate was 32% [998]. In multi-variable analyses, higher pre-operative PSA, higher number of PSMA-avid lesions, multiple (pelvic plus retroperitoneal), and retroperitoneal localisation of lesions at pre-operative imaging were independent predictors of BCR after PSMA-RGS. High-level evidence for the oncological value of salvage LN dissection (including adjuvant RT of the LNs) is still lacking [994].

6.4.5.2. Management of PSA failures after radiation therapy

Therapeutic options in these patients are ADT or salvage local procedures, as well as a ‘wait and see’ approach, based on EAU BCR risk categories at relapse. A SR and meta-analysis included studies comparing the efficacy and toxicity of salvage RP, salvage HIFU, salvage cryotherapy, SBRT, salvage LDR BT, and salvage HDR BT in the management of locally recurrent PCa after primary radical EBRT [999]. The outcomes were BCR-free survival at two and five years. No significant differences with regards to recurrence-free survival (RFS) between these modalities was found. Five-year RFS ranged from 50% after cryotherapy to 60% after HDR BT and SBRT. The authors reported that severe GU toxicity exceeded 21% for HIFU and RP, whereas it ranged from 4.2% to 8.1% with re-irradiation. Differences in severe GI toxicity also appeared to favour re-irradiation, particularly HDR BT [999]. Due to the methodological limitations of this review (the majority of the included studies were uncontrolled single-arm case series and there was considerable heterogeneity in the definitions of core outcomes) the available evidence for these treatment options is of low quality and strong recommendations regarding the choice of any of these techniques cannot be made. The following is an overview of the most important findings for each of these techniques.

6.4.5.2.1. Salvage radical prostatectomy

Salvage RP after RT is associated with a higher likelihood of AEs compared to primary surgery because of the risk of fibrosis and poor wound healing due to radiation [1000].

6.4.5.2.1.1. Oncological outcomes

In a SR of the literature, Chade, et al., showed that SRP provided five and ten years BCR-free survival estimates ranging from 47–82% and from 28–53%, respectively. The ten-year CSS and OS rates ranged from 70–83% and from 54–89%, respectively. The pre-SRP PSA value and prostate biopsy ISUP grade group were the strongest predictors of the presence of organ-confined disease, progression, and CSS [1001]. In a multi-centre analysis including 414 patients, five-year BCR-free survival, CSS and OS were 56.7%, 97.7% and 92.1%, respectively [1002]. Pathological T stage ≥ T3b (OR: 2.348) and GS (up to OR 7.183 for GS > 8) were independent predictors for BCR (see Table 6.4.7).

Table 6.4.7: Oncological results of selected salvage radical prostatectomy case series

Study

n

Median FU (mo)

Pathologic Organ-confined (%)

PSM (%)

Lymph-node involvement (%)

BCR-free probability (%)

CSS (%)

Time probability

Chade, et al. 2011 [1003]

404

55

55

25

16

37

83

10 yr.

Mandel, et al. 2016 [1004]

55

36

50

27

22

49

89

5 yr.

Ogaya-Pinies,
et al. 2018 [1005]

96

14

50

17

8

85*

-

14 mo.

Marra, et al. 2021 [1002]

414

36

46

30

16

57

98

5 yr.

*Percentage of patients without BCR.
BCR = biochemical recurrence; CSS = cancer-specific survival; FU = follow-up; mo = months; n = number of patients; PSM = positive surgical margin; yr. = year.

6.4.5.2.1.2. Morbidity

Compared to primary open RP, SRP is associated with a higher risk of later anastomotic stricture (47 vs. 5.8%), urinary retention (25.3% vs. 3.5%), urinary fistula (4.1% vs. 0.06%), abscess (3.2% vs. 0.7%) and rectal injury (9.2 vs. 0.6%) [1006]. A series, these complications appear to be less common [1000,1001,1004].

Functional outcomes are also worse compared to primary surgery, with urinary incontinence ranging from 21% to 90% and ED in nearly all patients [1001,1004].

6.4.5.2.1.3. Summary of salvage radical prostatectomy

In general, SRP should be considered only in patients with low co-morbidity, a life expectancy of at least ten years, a pre-SRP PSA < 10 ng/mL and initial biopsy ISUP grade group ≤ 2/3, no LN involvement or evidence of distant metastatic disease pre-SRP, and those whose initial clinical staging was T1 or T2 [1001].

6.4.5.2.2. Salvage cryoablation of the prostate
6.4.5.2.2.1. Oncological outcomes

Salvage cryoablation of the prostate (SCAP) has been proposed as an alternative to salvage RP, as it has a potentially lower risk of morbidity and equal efficacy.

In a SR a total of 32 studies assessed SCAP, recruiting a total of 5,513 patients. The overwhelming majority of patients (93%) received whole-gland SCAP. The adjusted pooled analysis for two-year BCR-free survival for SCAP was 67.49% (95% CI: 61.68–72.81%), and for five-year BCR-free survival was 50.25% (95% CI: 44.10–56.40%). However, the certainty of the evidence was low. Table 6.3.8 summarises the results of a selection of the largest series on SCAP to date in relation to oncological outcomes (BCR only) [999] (Table 6.4.8).

Table 6.4.8: Oncological results of selected salvage cryoablation of the prostate case series, including at least 250 patients

Study

n

Median FU (mo)

Time point of outcome measurement (yr)

BCR-free probability

Definition of failure

Ginsburg, et al. 2017 [1007]

898

19.0

5

71.3%

Phoenix criteria

Spiess, et al. 2010 [1008]

450

40.8

3.4

39.6%

PSA > 0.5 ng/mL

Li, et al. 2015 [1009]

486

18.2

5

63.8%

Phoenix criteria

Kovac, et al. 2016 [1010]

486

18.2

5

75.5%
(nadir PSA < 0.4 ng/mL);

22.1%
(nadir PSA > 0.4 ng/mL)

Phoenix criteria

Ahmad, et al. 2013 [1011]

283

23.9

3

67.0%
(nadir PSA < 1 ng/mL);

14.0%
(nadir PSA > 1 ng/mL)

Phoenix criteria

Pisters, et al. 2008 [1012]

279

21.6

5

58.9% (ASTRO)

54.5% (Phoenix)

ASTRO and Phoenix criteria

ASTRO = American Society for Therapeutic Radiology and Oncology; BCR = biochemical recurrence;FU = follow-up; mo. = months; n = number of patients; PSA = prostate-specific antigen; yr. = year.

6.4.5.2.3. Salvage re-irradiation
6.4.5.2.3.1. Salvage brachytherapy for radiotherapy failure

Carefully selected patients with a good PS, primary localised PCa, good urinary function and histologically proven local recurrence are candidates for salvage BT using either HDR or LDR.

In a SR a total of sixteen studies (four prospective) and 32 studies (two prospective) assessed salvage HDR and LDR BT, respectively, with the majority (> 85%) receiving whole-gland BT rather than focal treatment [999]. The adjusted pooled analysis for two-year BCR-free survival for HDR was 77% (95% CI: 70–83%) and for LDR was 81% (95% CI:74–86%). The five-year BCR-free survival for HDR was 60% (95% CI: 52–67%) and for LDR was 56% (95% CI: 48–63%). As noted above, BT techniques are associated with lower rates of severe GU toxicity when compared to RP or HIFU, at 8% for HDR (95% CI: 5.1–11%) and 8.1% for LDR (95% CI: 4.3–13%). Rates of severe GI toxicity are reported to be very low at 0% for HDR (95% CI: 0–0.2%) and 1.5% for LDR (95% CI: 0.2–3.4%). High-dose-rate or LDR BT are effective treatment options with an acceptable toxicity profile. However, the published series are small and likely under-report toxicity. Consequently, this treatment should be offered in experienced centres ideally within randomised clinical trials or prospective registry studies (see Table 6.4.9).

Table 6.4.9: Treatment-related toxicity and BCR-free probability in selected salvage brachytherapy studies including at least 100 patients.

Study

Study design

n and BT type

Median FU (mo)

Treatment toxicity

BCR-free probability

Lopez, et al. 2019 [1013]

multi-centre retrospective

75 HDR

44 LDR

52

23.5% late G3+ GU

5 yr 71%
(95% CI: 65.9-75.9%)

Crook, et al. 2019 [1014]

multi-centre prospective

100 LDR

54

14% late G3 combined GI/GU

n.r.

Smith, et al. 2020 [1015]

single-centre retrospective

108 LDR

76

15.7%/2.8% late G3 GU/GI

5 yr. 63.1%

10 yr. 52%

Lyczek, et al. 2009 [1016]

single-centre retrospective

115 HDR

n.r.

12.2%/0.9%

late G3+ GU/GI

60% at 40 mo.

BT = brachytherapy; CI = confidence interval; G = grade; GI = gastro-intestinal; GU = genito-urinary; HDR = high-dose rate; LDR = low-dose rate; mo = months; n = number of patients; n.r. = not reported; yr = year.

6.4.5.2.3.2. Salvage stereotactic ablative body radiotherapy for radiotherapy failure

6.4.5.2.3.2.1.Oncological outcomes and morbidity

Stereotactic ablative body radiotherapy (CyberKnife® or linac-based treatment) is a potentially viable new option to treat local recurrence after RT. Carefully selected patients with good IPSS-score, without obstruction, good PS and histologically proven localised local recurrence are potential candidates for SABR. In a metaanalysis and SR five mostly retrospective studies including 206 patients were treated with CyberKnife® or linac-based treatment showing two-year RFS estimates (61.6%, 95% CI: 52.6–69.9%) [999]. In a retrospective multi-centre study (n = 100) the median pre-salvage PSA was 4.3 ng/mL with 34% of patients having received ADT for twelve months (median). All recurrences were biopsy proven. Patients were treated with the CyberKnife® with a single dose of 6 Gy in six daily fractions (total dose 36 Gy). With a median followup of 30 months the estimated three-year second BCR-free survival was 55% [1017].

In a smaller retrospective series including 50 men with histologically proven local recurrence with a median pre-salvage PSA of 3.9 ng/mL only 15% had received additional ADT. The estimated five-year second BCR-free survival was 60% (median follow-up of 44 months) which is an outcome comparable to series treating patients with RP, HIFU or BT [1018]. Table 6.4.10 summarises the results of the two larger SABR series addressing oncological outcomes and morbidity.

Table 6.4.10: Treatment-related toxicity and BCR-free survival in selected SABR studies

Study

Study design

n and RT-type

Median FU (mo)

Fractionation

(SD/TD)

ADT

Treatment toxicity

BCR-free survival

Bergamin, et al. 2020 [1019]

single-centre prospective

25

LINAC

based

25

SD 6-6.2

TD 36-38 Gy

0/25

2 yr. late

G1 GI 8%

G2 GU 4%

2 yr. 80%

Fuller, et al. 2020 [1018]

single-centre retrospective

50

Cyber Knife

44

SD 6.8 Gy

TD 34 Gy

7/50

5 yr: 8% late G3+ GU

5 yr. 60%

Pasquier, et al. 2020 [1017]

multi-centre retrospective

100

Cyber Knife

30

SD 6 Gy

TD 36 Gy

34/100

median 12 mo.

3 yr. grade 2+ GU 20.8%

GI 1%

3 yr. 55%

BCR = biochemical recurrence; FU = follow-up; mo = months; n = number of patients; RT-type = type of radio-therapy; SD = single dose; TD = total dose; yr = year.

6.4.5.2.3.2.2.Morbidity

In a retrospective single-centre study with 50 consecutive patients chronic significant toxicity was only seen for the GU domain with five-year grade 2+ and grade 3+ GU rates of 17% and 8%, respectively. No GI toxicity > grade 1 was seen. Of note, of the fifteen patients who were sexually potent pre-salvage SBRT, twelve subsequently lost potency [1018]. In a retrospective French (GETUG) multi-centre series (n = 100) the three-year late grade 2+ GU and GI toxicity was 20.8% (95% CI: 13–29%) and 1% (95% CI: 0.1–5.1%), respectively [1017].

6.4.5.2.3.2.3.Summary of salvage stereotactic ablative body radiotherapy

Despite the encouraging results so far the number of patients treated with SABR is relatively limited. In view of the rates of higher grade 2+ GU side effects, SABR should only be offered to selected patients, in experienced centres as part of a clinical trial or well-designed prospective study.

6.4.5.2.4. Salvage high-intensity focused ultrasound
6.4.5.2.4.1. Oncological outcomes

Salvage HIFU has emerged as an alternative thermal ablation option for radiation-recurrent PCa. Being relatively newer than SCAP the data for salvage HIFU are even more limited. A SR and metaanalysis included 20 studies (n = 1,783) assessing salvage HIFU [999]. The overwhelming majority of patients (86%) received whole-gland salvage HIFU. The adjusted pooled analysis for two-year BCR-free survival for salvage HIFU was 54.14% (95% CI: 47.77–60.38%) and for five-year BCR-free survival 52.72% (95% CI: 42.66– 62.56%). However, the certainty of the evidence was low. Table 6.4.11 summarises the results of a selection of the largest series on salvage HIFU to date in relation to oncological outcomes (BCR only).

Table 6.4.11: Oncological results of selected salvage cryoablation of the prostate case series, including at least 250 patients

Study

n

Median FU (mo)

Time point of outcome measurement (yr)

BCR-free probability

Definition of failure

Crouzet, et al.

2017 [1020]

418

39.6

5

49.0%

Phoenix criteria

Murat, et al.

2009 [1021]

167

Mean 18.1

3

25.0% (high-risk)

53.0% (low-risk)*

Phoenix criteria or positive biopsy or initiation of post-HIFU salvage therapy

Kanthabalan, et al. 2017 [1022]

150

35.0

3

48.0%

Phoenix criteria

Jones, et al.

2018 [1023]

100

12.0

1

50.0%

Nadir PSA > 0.5 ng/mL or positive biopsy

*Results stratified by pre-EBRT D’Amico risk groups.
BCR = biochemical recurrence; FU = follow-up; mo = months; n = number of patients; yr = year.

6.4.5.2.4.2. Morbidity

The main adverse effects and complications relating to salvage HIFU include urinary incontinence, urinary retention due to bladder outflow obstruction, rectourethral fistula and ED. The SR and meta-analysis showed an adjusted pooled analysis for severe GU toxicity for salvage HIFU of 22.66% (95% CI: 16.98–28.85%) [999]. The certainty of the evidence was low. Table 6.4.12 summarises the results of a selection of the largest series on salvage HIFU to date in relation to GU outcomes. 

Table 6.4.12: Peri-operative morbidity, erectile function and urinary incontinence in selected salvage HIFU case series, including at least 100 patients

Study

n

Time point of outcome measurement (yr)

Incontinence* (%)

Obstruction/

retention (%)

Rectourethral

fistula (%)

ED (%)

Crouzet, et al.

2017 [1020]

418

Median 39.6

42.3

18.0

2.3

n.r.

Murat, et al.

2009 [1021]

167

Median 18.1

49.5

7.8

3.0

n.r.

Kanthabalan, et al. 2017 [1022]

150

24

12.5

8.0

2.0

41.7

Jones, et al.

2018 [1023]

100

12

42.0

49.0

5.0

74.0

*Incontinence was heterogeneously defined; figures represent at least 1 pad usage. ED = erectile dysfunction; n.r. = not reported; n = number of patients.

6.4.5.2.4.3. Summary of salvage high-intensity focused ultrasound

There is a lack of high-certainty data which prohibits any recommendations regarding the indications for salvage HIFU in routine clinical practice. There is also a risk of significant morbidity associated with its use in the salvage setting. Consequently, salvage HIFU should only be performed in selected patients in experienced centres as part of a clinical trial or well-designed prospective cohort study.

6.4.6. Hormonal therapy for relapsing patients

The Panel conducted a SR including studies published from 2000 onwards [1024]. Conflicting results were found on the clinical effectiveness of HT after previous curative therapy of the primary tumour. Some studies reported a favourable effect of HT, including the only RCT addressing the research question of this review (86% vs. 79% advantage in OS in the early HT group) [1025]. Other studies did not find any differences between early vs. delayed, or no, HT. One study found an unfavourable effect of HT [1026]. This may be the result of selecting clinically unfavourable cases for (early) HT and more intensive diagnostic workup and follow-up in these patients.

The studied population is highly heterogeneous regarding their tumour biology and therefore clinical course. Predictive factors for poor outcomes were short PSA-DT, high ISUP grade group, high PSA, increased age and comorbidities. In some studies, such as the Boorjian, et al., study [1027], high-risk patients, mainly defined by a high ISUP grade group and a short PSA-DT (most often less than six months) seem to benefit most from (early) HT, especially men with a long life expectancy.

Non-steroidal anti-androgens have been claimed to be inferior compared to castration, but this difference was not seen in M0 patients [960]. One of the included RCTs suggested that intermittent HT is not inferior to continuous HT in terms of OS and CSS [1028]. A small advantage was found in some QoL domains but not overall QoL outcomes. An important limitation of this RCT is the lack of any stratifying criteria such as PSA-DT or initial risk factors. Based on the lack of definitive efficacy and the undoubtedly associated significant side effects, patients with recurrence after primary curative therapy should not receive standard HT since only a minority of them will progress to metastases or PCa-related death. The objective of HT should be to improve OS, postpone distant metastases, and improve QoL. Biochemical response to only HT holds no clinical benefit for a patient. For older patients and those with comorbidities the side effects of HT may even decrease life expectancy; in particular cardiovascular risk factors need to be considered [1029,1030]. Early HT should be reserved for those at the highest risk of disease progression defined mainly by a short PSA-DT at relapse
(< 6–12 months) or a high initial ISUP grade group (> 2/3) and a long life expectancy.

A three-arm randomised phase III trial (EMBARK) looked at patients with prostate cancer who had high-risk biochemical recurrence defined as a PSA-DT of ≤9 months and a PSA level of ≥2 ng/ml above the nadir after radiation therapy or ≥1 ng/ml after radical prostatectomy with or without postoperative radiation therapy [1031]. Patients were randomly assigned 1:1:1 to receive enzalutamide daily plus leuprolide every 12 weeks (combination group), placebo plus leuprolide (leuprolide-alone group), or enzalutamide monotherapy (monotherapy group). The primary end point was MFS, in the combination group as compared with the leuprolide- alone group. The MFS in the monotherapy group as compared with the leuprolide-alone group was a key secondary endpoint. A total of 1068 patients were randomised. After a median follow-up of 60.7 months, the five year - MFS was 87.3% (95% CI, 83.0 - 90.6) in the combination group, 71.4% (95% CI, 65.7 - 76.3) in the leuprolide-alone group, and 80.0% (95% CI, 75.0 - 84.1) in the monotherapy group. The combination of enzalutamide plus leuprolide was superior to leuprolide alone with regards to the MFS (HR 0.42; 95% CI, 0.30 - 0.61; P<0.001). Enzalutamide monotherapy also showed a superior MFS compared to leuprolide alone (HR 0.63; 95% CI, 0.46 - 0.87; P = 0.005). These results led to the FDA approval for enzalutamide alone or in combination with ADT for patients with high-risk biochemical recurrence in November 2023 [1032].

At the time of the MFS analysis, OS data were immature with 12% deaths in the overall population.

Also, an intermittent treatment approach can be considered. Enzalutamide treatment can be suspended if PSA is undetectable (< 0.2 ng/mL) after 36 weeks of therapy. Treatment may be reinitiated when PSA has increased to ≥ 2.0 ng/mL for patients who had prior radical prostatectomy or ≥ 5.0 ng/mL for patients who had prior primary radiation therapy. There were no new safety signals. Of note, at a median follow-up of five years, the overall percentage of patients who had fractures was 14% [1033].

6.4.7. Observation

In unselected relapsing patients the median actuarial time to the development of metastasis will be 8 years and the median time from metastasis to death will be a further five years [825]. For patients with EAU Low-Risk BCR features (see Section 6.3.3), unfit patients with a life expectancy of less than ten years or patients unwilling to undergo salvage treatment, active follow-up may represent a viable option.

6.4.8. Guidelines for second-line therapy after treatment with curative intent

Local salvage treatment

Strength rating

Recommendations for biochemical recurrence (BCR) after radical prostatectomy

Offer early salvage intensity-modulated radiotherapy/volumetric arc radiation therapy plus image-guided radiotherapy to men with two consecutive PSA rises.

Strong

A negative positron emission tomography/computed tomography (PET/CT) scan should not delay salvage radiotherapy (SRT), if otherwise indicated.

Strong

Offer monitoring, including prostate-specific antigen (PSA), to EAU Low-Risk BCR patients.

Weak

Do not wait for a PSA threshold before starting treatment. Once the decision for SRT has been made, SRT (at least 64 Gy) should be given as soon as possible.

Strong

Offer hormonal therapy in addition to SRT to men with BCR.

Weak

Recommendations for BCR after radiotherapy

Offer monitoring, including PSA to EAU Low-Risk BCR patients.

Weak

Only offer salvage radical prostatectomy (RP), brachytherapy, stereotactic body radiotherapy, high-intensity focused ultrasound, or cryosurgical ablation to highly selected patients with biopsy-proven local recurrence within a clinical trial setting or well-designed prospective cohort study undertaken in experienced centres.

Strong

Recommendations for systemic salvage treatment

Do not offer androgen deprivation therapy to M0 patients with a PSA-doubling time > twelve months.

Strong

Offer enzalutamide with or without androgen deprivation therapy to M0 patients with high-risk BCR, defined as a PSA doubling time of ≤9 months and a PSA level of ≥2ng/ml above nadir after radiation therapy or ≥1 ng/ml after radical prostatectomy with or without postoperative radiation therapy.

Strong

Recommendations for follow-up after radical prostatectomy or radiotherapy

Routinely follow-up asymptomatic patients by obtaining at least a disease-specific history and serum prostate-specific antigen (PSA) measurement.

Strong

At recurrence, only perform imaging if the result will affect treatment planning.

Strong

6.5. Systemic treatments for prostate cancer

6.5.1. Hormonal therapy

6.5.1.1. Different types of hormonal therapy

Androgen deprivation can be achieved by suppressing the secretion of testicular androgens in different ways. This can be combined with inhibiting the action of circulating androgens at the level of their receptor which has been known as complete (or maximal or total) androgen blockade (CAB) using the old-fashioned antiandrogens [1034].

6.5.1.2. Castration level

The castration level of testosterone is < 50 ng/dL (1.7 nmol/L), which was defined more than 40 years ago when testosterone testing was less sensitive. Current methods have shown that the mean value after surgical castration is 15 ng/dL [1035]. Therefore, a more appropriate level should be defined as < 20 ng/dL ( < 0.7 nmol/L). This definition is important as better results are repeatedly observed with lower testosterone levels compared to 50 ng/dL [1036-1038]. However, the castrate level considered by the regulatory authorities and in clinical trials addressing castration in PCa is still the historical < 50 ng/dL (1.7 nmol/L).

6.5.1.3. Bilateral orchiectomy

Bilateral orchiectomy or subcapsular pulpectomy is still considered the primary treatment modality for ADT. It is a simple, cheap and virtually complication-free surgical procedure. It is easily performed under local anaesthesia, and it is the quickest way to achieve a castration level which is usually reached within less than twelve hours. It is irreversible and therefore does not allow for intermittent treatment [1039].

6.5.1.4. Oestrogens

Treatment with oestrogens results in testosterone suppression and is not associated with bone loss [1040]. Early studies tested oral diethylstilboestrol (DES) at several doses. Due to severe side effects, especially thromboembolic complications, even at lower doses these drugs are not considered as standard first-line treatment [1041,1042]. Oestrogen patches are under investigation [1043].

6.5.1.5. Luteinising-hormone-releasing hormone agonists

Long-acting LHRH agonists are currently the main forms of ADT. These synthetic analogues of LHRH are delivered as depot injections on a 1-, 2-, 3-, 6-monthly, or yearly basis. The first injection induces a transient rise in luteinising hormone (LH) and follicle-stimulating hormone (FSH) leading to the ‘testosterone surge’ or ‘flare-up’ phenomenon which starts two to three days after administration and lasts for about one week. This may lead to detrimental clinical effects (the clinical flare) such as increased bone pain, acute bladder outlet obstruction, obstructive renal failure, spinal cord compression, and cardiovascular death due to hypercoagulation status [1044]. Patients at risk are usually those with high-volume symptomatic bony disease. Concomitant therapy with an anti-androgen decreases the incidence of clinical flare but does not completely remove the risk. Anti-androgen therapy is usually continued for four weeks but neither the timing nor the duration of anti-androgen therapy are based on strong evidence. In addition, the long-term impact of preventing ‘flare up’ is unknown [1045].

Chronic exposure to LHRH agonists results in the down-regulation of LHRH-receptors, suppressing LH and FSH secretion and therefore testosterone production. A castration level is usually obtained within two to four weeks [1046]. Although there is no formal direct comparison between the various compounds, they are considered to be equally effective [1047]. So far, no survival difference between LHRH agonists and orchiectomy has been reported due to the lack of high-quality trials [1048].The different products have practical differences that need to be considered in everyday practice, including the storage temperature, whether a drug is ready for immediate use or requires reconstitution, and whether a drug is given by subcutaneous or intramuscular injection.

6.5.1.6. Luteinising-hormone-releasing hormone antagonists

Luteinising-hormone-releasing hormone antagonists immediately bind to LHRH receptors, leading to a rapid decrease in LH, FSH and testosterone levels without any flare. The practical shortcoming of these compounds is the lack of a long-acting depot formulation with, so far, only monthly formulations being available. Degarelix is a LHRH antagonist. The standard dosage is 240 mg in the first month followed by monthly injections of 80 mg. Most patients achieve a castrate level at day three [1046]. A phase III RCT compared degarelix to monthly leuprorelin following up patients for twelve months, suggesting a better PSA PFS for degarelix 240/80 mg compared to monthly leuprorelin [1049]. A SR did not show a major difference between agonists and degarelix and highlighted the paucity of on-treatment data beyond twelve months as well as the lack of survival data [1050]. Its definitive superiority over the LHRH analogues remains to be proven. Short-term follow-up data from a meta-analysis indicate that the use of LHRH antagonist is associated with significantly lower overall mortality and cardiovascular events as compared with agonists. On the other hand, other adverse effects such as decreased libido, hot flushes, ED, weight gain, and injection site reactions are seen less often with the agonists [1051,1052].

Relugolix is an oral LHRH antagonist. It was compared to the LHRH agonist leuprolide in a randomised phase III trial [1053]. The primary endpoint was sustained testosterone suppression to castrate levels through 48 weeks. There was a significant difference of 7.9 percentage points (95% CI: 4.1–11.8) showing non-inferiority and superiority of relugolix. The incidence of major adverse cardiovascular events was significantly lower with relugolix (prespecified safety analysis). Relugolix has been approved by the FDA [1054] and EMA [1055] for hormone sensitive PCa.

6.5.1.7. Anti-androgens

These oral compounds are classified according to their chemical structure as:

  • steroidal, e.g., cyproterone acetate (CPA), megestrol acetate and medroxyprogesterone
  • acetate;
  • non-steroidal or pure, e.g., nilutamide, flutamide and bicalutamide.

Both classes compete with androgens at the receptor level. This leads to an unchanged or slightly elevated testosterone level. Conversely, steroidal anti-androgens have progestational properties leading to central inhibition by crossing the blood-brain barrier.

6.5.1.7.1. Steroidal anti-androgens

These compounds are synthetic derivatives of hydroxyprogesterone. Their main pharmacological side effects are secondary to castration (gynaecomastia is quite rare) whilst the non-pharmacological side effects are cardiovascular toxicity (4–40% for CPA) and hepatotoxicity.

Cyproterone acetate was the first licensed anti-androgen but the least studied. Its most effective dose as monotherapy is still unknown. Although CPA has a relatively long half-life (31–41 hours), it is usually administered in two or three fractionated doses of 100 mg each. In one RCT CPA showed a poorer OS when compared with LHRH analogues [1056]. An underpowered RCT comparing CPA monotherapy with flutamide in M1b PCa did not show any difference in DSS and OS at a median follow-up of 8.6 years [1057]. Other CPA monotherapy studies suffer from methodological limitations preventing firm conclusions.

6.5.1.7.2. Non-steroidal anti-androgens

Non-steroidal anti-androgen monotherapy with e.g. nilutamide, flutamide or bicalutamide does not suppress testosterone secretion and it is claimed that libido, overall physical performance and bone mineral density (BMD) are frequently preserved [1058]. Non-androgen-related pharmacological side effects differ between agents. Bicalutamide shows a more favourable safety and tolerability profile than flutamide and nilutamide [1059]. The dosage licensed for use in CAB is 50 mg/day, and 150 mg/day for monotherapy. The androgen pharmacological side effects are mainly gynaecomastia (70%) and breast pain (68%). However, non-steroidal anti-androgen monotherapy offers clear bone protection compared with LHRH analogues and probably LHRH antagonists [1058,1060]. All three agents share the potential for liver toxicity (occasionally fatal), requiring regular monitoring of patients’ liver enzymes.

6.5.1.7.3. New androgen receptor pathway inhibitors (ARPis)

Once on ADT the development of castration-resistance (CRPC) is only a matter of time. It is considered to be mediated through two main overlapping mechanisms: androgen-receptor (AR)-independent and AR-dependent mechanisms (see Section 6.5 - Castrate-resistant PCa). In CRPC, the intracellular androgen level is increased compared to androgen sensitive cells and an over-expression of the AR has been observed, suggesting an adaptive mechanism [1061]. This has led to the development of several new compounds targeting the androgen axis. In mCRPC, abiraterone acetate and enzalutamide have been approved. In addition to ADT (sustained castration), abiraterone acetate, apalutamide and enzalutamide have been approved for the treatment of metastatic hormone sensitive PCa (mHSPC) by the FDA and the EMA. For the updated approval status see EMA and FDA websites [1032,1062-1065]. Finally, apalutamide, darolutamide and enzalutamide have been approved for non-metastatic CRPC (nmCRPC) at high risk of further metastases [1066-1070].

6.5.1.7.3.1. Abiraterone acetate

Abiraterone acetate is a CYP17 inhibitor (a combination of 17α-hydrolase and 17,20-lyase inhibition). By blocking CYP17, abiraterone acetate significantly decreases the intracellular testosterone level by suppressing its synthesis at the adrenal level and inside the cancer cells (intracrine mechanism). This compound must be used together with prednisone/prednisolone to prevent drug-induced hyperaldosteronism [1062,1064].

6.5.1.7.3.2. Apalutamide, darolutamide, enzalutamide (alphabetical order)

These agents are novel non-steroidal anti-androgens with a higher affinity for the AR receptor than traditional non-steroidal anti-androgens. In addition, while previous non-steroidal anti-androgens still allow transfer of ARs to the nucleus and would act as partial agonists, all three agents also block AR transfer and therefore suppress any possible agonist-like activity [1065-1067]. Darolutamide has structurally unique properties In particular, in preclinical studies, it was shown not to cross the blood-brain barrier [1071,1072].

6.5.2. Cytotoxic drug treatment

6.5.2.1. Taxanes

Paclitaxel derivatives promote the assembly of microtubules and inhibit the subsequent depolymeization, impairing the tubulin dynamics that foster the mitotic spindle assembly during interphase in mitosis [1073]. Docetaxel binds ß-tubulin dimers in a 1:1 stoichiometric ratio, exhibiting a stronger dynamic instability using its inhibitory effect in tubulin depolymerization [1074]. It also activates NF-kB causing apoptosis via a mitochondria-dependent pathway [1075]. Docetaxel shows significant activity against prostatetumours. Cabazitaxel also works by binding to the microtubules. This prevents cellular mitosis and stabilises the tumour cells. As a result, the cells do not divide. In addition, it inhibits androgen receptors by binding to the microtubules and microtubule-associated motor protein dynein. As a consequence, androgen receptor nuclear translocation is prevented [1073]. Common side-effects include peripheral neuropathy, myalgias, neutropenia and arthralgia.

6.5.3. Non-hormonal non-cytotoxic drug treatments

6.5.3.1. Poly (ADP-ribose) polymerase inhibitirs (PARPi)

Poly (ADP-ribose) polymerase inhibitors (PARPi) block the enzyme poly ADP-ribose polymerase (PARP) and were developed aiming to selectively target cancer cells harbouring BRCA mutations and other mutations inducing homologous recombination deficiency and high level of replication pressure with a sensitivity to PARPi treatment. Due to the oncogenic loss of some DNA repair effectors and incomplete DNA repair repertoire, some cancer cells are addicted to certain DNA repair pathways such as Poly (ADP-ribose) polymerase (PARP)-related single-strand break repair pathway. The interaction between BRCA and PARP is a form of synthetic lethal effect which means the simultaneously functional loss of two genes leads to cell death, while a defect in any single gene only has a limited effect on cell viability [1076]. The therapeutic indications for PCa are discussed in Sections 6.5.8.1.

6.5.3.2. AKT inhibitors

AKT inhibitors are small molecules which are designed to target and bind to all three isoforms of AKT, which is a key component of the PI3K/AKT pathway. In clinical trials, ipatasertib, an oral, highly specific, AKT inhibitor was used and showed significant activity when combined with abiraterone acetate in patients with loss of the tumour suppressor protein PTEN on immunohistochemistry within the tumour [1077,1078]. Available data can be found in table 6.5.2. Currently, there are no approved AKT inhibitors.

6.5.3.3. Immune checkpoint inhibitors

Checkpoint inhibitors target the molecules CTLA4, programmed cell death protein 1 (PD-1), and programmed death-ligand 1 (PD-L1). For advanced PCa patients that are microsatellite instability-high/deficient mismatch repair (MSI-H/dMMR), the PD-1 inhibitor pembrolizumab has been approved by the FDA but not by the EMA. The label is tumour agnostic [1079,1080]. See also Section 6.6.2.1

6.5.3.4. Radiopharmaceutical therapy

Radiopharmaceutical therapy (RPT) is based on the delivery of radioactive isotopes to tumour-associated targets. The mechanism of action for RPT is radiation-induced killing of cells. Radionuclides with different emission properties are used to deliver radiation. The most commonly used radionuclides are represented by β-particles (e.g., 177Lu) or α-particles (e.g., 223Ra, 225Ac). 177Lu is increasingly used because of its optimal imaging range (100–200 keV), favourable half time (6.6 days) and appropriate β-particle energy for therapy. The short path of the β-particles (0.05–0.08 mm) results in minimal toxic effects in adjacent healthy tissue. These properties enable such radionuclides to be used as theranostics (i.e., the same radionuclide may be used for both diagnostic and therapeutic purposes). However, an essential requirement prior to any RPT is to assess the targeting of the agent, mainly using PET techniques which show the tumour expression and the extent of cancer [1081]. 177Lu has been approved by the FDA for the treatment of adult patients with PSMA-positive mCRPC who have been treated with ARPI and taxane-based chemotherapy [1082,1083]. Clinical details are discussed in Section 6.6.8.

6.6. Management of Metastatic prostate cancer

6.6.1. Introduction

All prospective data available rely on the definition of M1 disease based on CT scan or MRI and bone scintigraphy. The influence on treatment and outcome of newer, more accurate, imaging has not been assessed yet.

6.6.2. Prognostic factors

Median survival of patients with newly diagnosed metastases (synchronous mHSPC) is approximately 50 months with ADT alone, however, it is highly variable since the M1 population is heterogeneous [1084]. Several prognostic factors for survival have been suggested including the number and location of bone metastases, presence of visceral metastases, ISUP grade group, PS status and initial PSA and alkaline phosphatase level, but only few have been validated [1085-1088].

‘Volume‘ of disease as a potential predictor was introduced by CHAARTED (Chemo-hormonal Therapy versus Androgen Ablation Randomised Trial for Extensive Disease in Prostate Cancer) [1088-1090] (see table 6.4.1) and subsequently, in STAMPEDE, was shown to be predictive in an adequately powered subgroup analysis for benefit of addition of prostate RT to ADT in the subgroup of patients with low volume/burden disease [1091] (See table 6.4.1).

‘Metachronous’ metastatic disease (after radical local treament of the primary tumour) vs. synchronous (or de novo) metastatic disease has also been shown to have generally a better prognosis [1092].

Based on a large SWOG 9346 cohort, the PSA level after seven months of ADT was used to create three prognostic groups (see Table 6.4.2) [1093]. A PSA ≤ 0.2 ng/mL at seven months has been confirmed as a prognostic marker for men receiving ADT for metastatic disease in the CHAARTED study independent of the addition of docetaxel [1094]. Similarly reaching PSA levels of ≤ 0.1ng/ml after six months were shown to be correlated with long-term outcomes in the LATITUDE study [1095]. Also for patients treated with ADT and apalutamide a deep PSA decline defined by ≥ 90% from baseline or to PSA≤ 0.2 ng/ml at a landmark of three months was associated with longer OS [1096] for patients.

Table 6.6.2.1 Definition of high- and low-volume in CHAARTED and high- and low-risk in LATITUDE [1088-1090high- and low-risk in LATITUDE [1070]


High

Low

CHAARTED (volume)

> 4 Bone metastases including > 1 outside vertebral column or pelvis

AND/OR

Visceral metastasis*

Not high

LATITUDE

(risk)

> 2 high-risk features of:

> 3 Bone metastasis

Visceral metastasis

> ISUP grade 4

Not high

*Lymph nodes are not considered as visceral metastases.

Table 6.6.2.2: Prognostic factors based on the SWOG 9346 study [1093]

PSA after 7 months after start of ADT

Median survival on ADT monotherapy

< 0.2 ng/mL

75 months

0.2 < 4 ng/mL

44 months

> 4 ng/mL

13 months

6.6.3. First-line hormonal treatment

Primary ADT has been the SOC for over 50 years [1034]. There is no high-level evidence in favour of a specific type of ADT for oncological outcomes, neither for orchiectomy nor for a LHRH agonist or antagonist. The level of testosterone is reduced much faster with orchiectomy and LHRH antagonist, therefore patients with impending spinal cord compression or other potential impending complications from the cancer should be treated with either a bilateral orchidectomy or LHRH antagonists as the preferred options.

There is a suggestion in some studies and a SR and meta-analysis that cardiovascular side effects are less frequent in patients treated with LHRH antagonists than patients treated with LHRH agonists [1053,1097-1099]; therefore, patients with pre-existing cardiovascular disease or other cardiovascular risk factors might be considered to be treated with antagonists if a chemical castration is chosen.

6.6.3.1. Non-steroidal anti-androgen monotherapy

Based on a Cochrane review comparing older generation non-steroidal anti-androgen (NSAA) monotherapy to ADT (either medical or surgical), NSAA was considered to be less effective in terms of OS, clinical progression, treatment failure and treatment discontinuation due to AEs [1100] and is generally not recommended also because ADT-based combination treatments have become SOC.

6.6.3.2. Intermittent versus continuous androgen deprivation therapy

Three independent reviews [1101-1103] and two meta-analyses [1104,1105] looked at the clinical efficacy of intermittent androgen deprivation (IAD) therapy. All of these reviews included eight RCTs of which only three were conducted in patients with exclusively M1 disease.

So far, the SWOG 9346 is the largest trial addressing IAD in M1b patients [1106]. Of 3,040 screened patients, only 1,535 patients met the inclusion criteria. This highlights that only about 50% of M1b patients can be expected to be candidates for IAD, i.e. the best PSA responders. This was a non-inferiority trial leading to inconclusive results: the actual upper limit was above the pre-specified 90% upper limit of 1.2 (HR: 1.1, CI: 0.99–1.23), the pre-specified non-inferiority limit was not achieved, and the results did not show a significant inferiority for any treatment arm. However, based on this study inferior survival with IAD cannot be completely ruled out even in this highly selected subgroup. The use of intermittent ADT has been superseeded as continuous ADT based combination therapy has become SOC.

6.6.3.3. Early versus deferred androgen deprivation therapy

Early treatment before the onset of symptoms is recommended in the majority of patients with metastatic hormone-sensitive disease despite lack of randomised phase III data in this specific setting and specifically not with the combination therapies that are standard nowadays.

A Cochrane analysis from 2019 about the topic concluded that early ADT probably extends time to death of any cause and time to death from PCa [1107]. Since the analysis included only a very limited number of metastatic patients, the benefit of early ADT in this setting remains unproven. All of the trials testing the combination therapies in the metastatic hormone-sensitive setting also included asymptomatic patients.

The only candidates with metastatic disease who may possibly be considered for deferred treatment are asymptomatic patients with a strong wish to avoid treatment-related side effects. The risk of developing symptoms, and even dying from PCa, without receiving the benefit from ADT with deferred treatment has been highlighted [1108,1109], but in the era before next generation imaging was used.

Patients with deferred treatment for advanced PCa must be amenable to close follow-up. Another potential exception are patients with recurrent oligometastatic disease who have a strong wish to postpone the start of ADT (see Section 6.4.7).

6.6.4. Combination therapies

All of the following combination therapies have been studied with continuous ADT, not intermittent ADT.

6.6.4.1.‘Combined’ androgen blockade with older generation NSAA (bicalutamide, flutamide, nilutamide)

Systematic reviews have shown that CAB using a NSAA appears to provide a small survival advantage (< 5%) vs. monotherapy (surgical castration or LHRH agonists) [1110,1111] but this minimal survival advantage must be balanced against the increased side effects. In addition, the newer combination therapies (see Tables 6.4.3, 6.4.4, 6.4.5) are more effective as shown specifically for enzalutamide which was tested against NSAA in a phase III trial [1112]. More recently another trial has demonstrated a significant OS benefit for the addition of rezvilutamide vs. addition of bicalutamide to ADT in patients with high-volume mHSPC [1113]. Therefore combination with NSAAs should only be considered if other combination therapies are not available.

6.6.4.2. Androgen deprivation combined with other agents
6.6.4.2.1. Androgen deprivation therapy combined with chemotherapy

Three large RCTs were conducted [775,1088,1114]. All trials compared ADT alone as the SOC with ADT combined with immediate docetaxel (75 mg/sqm, every three weeks within three months of ADT initiation). The primary objective in all three studies was to assess OS. The key findings are summarised in Table 6.4.3.

Table 6.6.3: Key findings - Hormonal treatment combined with chemotherapy


STAMPEDE [775,1115]

GETUG-AFU 15 [1114]

CHAARTED [1088,1089]


ADT

ADT + Docetaxel
(6 cycles) + P

ADT

ADT + Docetaxel
(9 cycles)

ADT

ADT + Docetaxel
(6 cycles)

N

1,184

592

193

192

393

397

Newly diagnosed M+

58%

59%

75%

67%

73%

73%

Key inclusion criteria

Patients scheduled for long-term ADT

- newly diagnosed M1 or N+ situations

- locally advanced (at least two of cT3 cT4, ISUP grade > 4, PSA > 40 ng/mL)

- relapsing locally treated disease with a PSA > 4 ng/mL and a PSA-DT < 6 mo.


or PSA > 20 ng/mL,

or nodal

or metastatic relapse

Metastatic disease Karnofsky score > 70%

Metastatic disease
ECOG PS 0, 1 or 2

Primary objective

OS

OS

OS

Median follow up (mo)

43; 78.2 (update M1)

50

54 (update)

HR (95% CI)

0.78 (0.66-0.93)

1.01 (0.75-1.36)

0.72 (0.59-0.89)

M1 only

N

1,086

-

-

HR (95% CI)

0.81 (0.69-0.95)

-

-

ADT = androgen deprivation therapy; CI = confidence interval; ECOG = Eastern Cooperative Oncology Group; HR = hazard ratio; ISUP = International Society for Urological Pathology; mo = month; n = number of patients; OS = overall survival; P = prednisone; PSA-DT = prostate-specific antigen-doubling time.

In the GETUG 15 trial, all patients had M1 PCa, either de novo or after a primary treatment [1114]. They were stratified based on previous treatment and Glass risk factors [1085]. In the CHAARTED trial the same inclusion criteria applied, and patients were stratified according to disease volume (see Table 6.4.1) [1088].

STAMPEDE is a multi-arm multi-stage trial in which the reference arm (ADT monotherapy) included 1,184 patients. One of the experimental arms was docetaxel combined with ADT (n = 593), another was docetaxel combined with zoledronic acid (n = 593). Patients were included with either M1 or N1 or having two of the following three criteria: T3/4, PSA ≥ 40 ng/mL or ISUP grade group 4–5. Also relapsed patients after local treatment were included if they met one of the following criteria: PSA ≥ 4 ng/mL with a PSA-DT < six months or a PSA ≥ 20 ng/mL, N1 or M1. No stratification was used regarding metastatic disease volume (high/low volume) [775]. In all three trials toxicity was mainly haematological with around 12–15% grade 3–4 neutropenia, and 6–12% grade 3–4 febrile neutropenia. The use of granulocyte colony-stimulating factor receptor (GCSF) was shown to be beneficial in reducing febrile neutropenia. Primary or secondary prophylaxis with GCSF should be based on available guidelines [1116,1117].

Docetaxel in all three trials was used at the standard dose of 75 mg/sqm every three weeks, six cycles in CHAARTED and STAMPEDE and up to nine cycles in GETUG-AFU-15. In subgroup analyses from GETUG-AFU 15 and CHAARTED the beneficial effect of the addition of docetaxel to ADT was most evident in men with de novo metastatic high-volume disease [1089,1090], while it was in the same range whatever the volume in the post-hoc analysis from STAMPEDE [1115]. The effect of adding docetaxel was less apparent in men who had prior local radical treatment although the numbers were small and the event rates lower. A SR and meta-analysis which included these three trials showed that the addition of docetaxel to SOC improved survival [1117]. The HR of 0.77 (95% CI: 0.68–0.87, p < 0.0001) translates into an absolute improvement in 4-year survival of 9% (95% CI: 5–14). In a SR and meta-analysis of individual participant data from the the three trials it has been shown that there is no meaningful beneficial effect of addition of docetaxel to ADT for patients with metachronous low volume disease. Interestingly the largest absolute improvement at five years was observed for the patients with high volume and clinical stage four disease [1118].

Based on these data, upfront docetaxel combined with ADT was considered as a standard in men presenting with metastases at first presentation, provided they are fit enough to receive docetaxel [1117]. More recently two large Phase III studies have now shown an OS benefit by adding an ARPI to ADT and docetaxel. Therefore adding docetaxel alone to ADT should only be considered if no ARPI is available or all available ones are contraindicated (see Section 6.4.4.2.3).

6.6.4.2.2. Combination with an ARPI alone (abiraterone, apalutamide, enzalutamide)

In two large RCTs (STAMPEDE, LATITUDE) the addition of abiraterone acetate (1000 mg daily) plus prednisone (5 mg daily) to ADT in men with mHSPC was studied [814,1070,1119] (see table 6.4.4). The primary objective of both trials was an improvement in OS. Both trials showed a significant OS benefit. In LATITUDE with only de novo high-risk metastatic patients included, the HR reached 0.62 (0.51–0.76) [1070]. The HR in STAMPEDE was very similar with 0.63 (0.52–0.76) in the total patient population (metastatic and non-metastatic) and a HR of 0.61 in the subgroup of metastatic patients [814]. While only high-risk patients were included in the LATITUDE trial a post-hoc analysis from STAMPEDE showed the same benefit whatever the risk or the volume category was [1120].

All secondary objectives such as PFS, time to radiographic progression, time to pain, or time to chemotherapy were in favour of the combination. No difference in treatment-related deaths was observed with the combination of ADT plus AAP compared to ADT monotherapy (HR: 1.37 [0.82–2.29]). However, twice as many patients discontinued treatment due to toxicity in the combination arms in STAMPEDE (20%) compared to LATITUDE (12%) [1119]. Based on these data upfront AAP combined with ADT should be considered as a standard in men presenting with metastases at first presentation, provided they are fit enough to receive the drug.

In three large RCTs (TITAN, ARCHES and ENZAMET) the addition of AR antagonists to ADT in men with mHSPC was tested [1068,1069,1112]. In ARCHES the primary endpoint was radiographic PFS (rPFS). In the primary analysis rPFS was significantly improved for the combination of enzalutamide and ADT with a HR of 0.39 (0.3–0.5). Approximately 36% of the patients had low-volume disease; around 25% had prior local therapy and 18% of the patients had received prior docetaxel. In the final prespecified analysis the key secondary enpoint OS was significantly improved with a HR of 0.66 (0.53-0.81) and a significant benefit for rPFS was maintained with a HR of 0.63 (0,52–0.76) [1121]. In ENZAMET the primary endpoint was OS. The addition of enzalutamide to ADT in the first analysis improved OS with a HR of 0.67 (0.52–0.86) compared to ADT plus a non-steroidal antiandrogen. Approximately half of the patients had concomitant docetaxel; about 40% had prior local therapy and about half of the patients had low-volume disease [1069]. In a planned later analysis with a median follow-up of 68 months the OS benefit of adding enzalutamide was maintained with a HR of 0.7 (0.58-0.84) [1122] In the TITAN trial, ADT plus apalutamide was used and rPFS and OS were co-primary endpoints. In the primary analysis rPFS was significantly improved by the addition of apalutamide with a HR of 0.48 (0.39–0.6); OS at 24 months was improved for the combination with a HR of 0.67 (0.51–0.89). In the final analysis the HR for OS was 0.65 (0.53–0.79) without adjustment for cross-over. In this trial 16% of patients had prior local therapy, 37% had low-volume disease and 11% received prior docetaxel [1068,1123]. In the more recently published CHART trial, ADT plus rezvilutamide was tested vs. ADT plus bicalutamide in patients with high-volume de novo metastatic disease. Ninety percent of the patients were recruited in China. Overall survival and rPFS were co-primary endpoints. At the pre-planned interim analysis rezvilutamide significantly improved rPFS compared with bicalutamide with a HR of 0.44 (0.33–0.58) and OS with a HR of 0.58 (0.44–0.77) [1113].

In summary, the addition of the new AR antagonists significantly improves clinical outcomes with no convincing evidence of differences between subgroups. The majority of patients had de novo metastatic disease and the evidence is most compelling in this situation. In the trials with the new AR antagonists, a proportion of patients had metachronous disease (see Table 6.4.5); in the subgroup analyses the effect seemed to be consistent and therefore, a combination should also be offered for men progressing after radical local therapy [1122,1124,1125].

Table 6.6.4: Results from the STAMPEDE arm G and LATITUDE studies


STAMPEDE [814]

LATITUDE [1070]


ADT

ADT + AA + P

ADT + placebo

ADT + AA + P

N

957

960

597

602

Newly diagnosed N+

20%

19%

0

0

Newly diagnosed M+

50%

48%

100%

100%

Key inclusion criteria

Patients scheduled for long-term ADT

- newly diagnosed M1 or N+ situations

- locally advanced (at least two of cT3 cT4, ISUP grade > 4, PSA > 40 ng/mL)

- relapsing locally treated disease with a PSA > 4 ng/mL and a PSA-DT < 6 mo.


or PSA > 20 ng/mL

or nodal

or metastatic relapse

Newly diagnosed M1 disease and 2 out of the 3 risk factors: ISUP grade > 4, > 3 bone lesions, measurable visceral metastasis

Primary objective

OS


OS

Radiographic PFS

Median follow up (mo)

40

30.4

3-yr. OS

83% (ADT + AA + P)

76% (ADT)

66% (ADT + AA + P)

49% (ADT + placebo)

HR (95% CI)

0.63 (0.52-0.76)

0.62 (0.51-0.76)

M1 only

N

1,002

1,199

3-yr. OS

NA

66% (ADT + AA + P)

49% (ADT + placebo)

HR (95% CI)

0.61 (0.49-0.75)

0.62 (0.51-0.76)

HR

FFS (biological, radiological, clinical or death): 0.29 (0.25-0.34)

rPFS:

0.49 (0.39-0.53)

AA = abiraterone acetate; ADT = androgen deprivation therapy; CI = confidence interval; FFS = failure-freesurvival; HR = hazard ratio; ISUP = International Society of Urological Pathology; mo = month; n = number of patients; NA = not available; OS = overall survival; P = prednisone; rPFS = radiographic progression-free survival; PSA = prostate-specific antigen; yr. = year.

Table 6.6.5: Results from the ENZAMET and TITAN studies with OS as primary endpoint


ENZAMET [1122]

TITAN [1068,1123]


ADT+ older antagonist
± docetaxel (SOC)

ADT + enzalutamide ± docetaxel

ADT + placebo

ADT + apalutamide

N

562

563

527

525

Newly diagnosed M+

72.1%

72.5%

83.7%

78.3%

Low volume

47%

48%

36%

38%

Primary objective

OS


OS

rPFS

Median follow up (mo)

34

30.4

3-yr. OS

3-yr survival:

80% (ADT + enzalutamide)

72% (SOC)

2-yr survival:

84% (ADT + apalutamide)

74% (ADT + placebo)

HR (95% CI) for OS

0.67 (0.52-0.86)

0.67 (0.51-0.89)

ADT = androgen deprivation therapy; CI = confidence interval; HR = hazard ratio; mo = month; n = number ofpatients; OS = overall survival; SOC = standard of care; rPFS = radiographic progression-free survival; yr = year.

Table 6.6.6: Results from the ARCHES and CHART studies 


ARCHES [1069,1121]

CHART [1113]


ADT ± docetaxel

ADT + enzalutamide ± docetaxel

ADT +
bicalutamide

ADT + rezvilutamide

N

576

574

328

326

Newly diagnosed M+

63%

70%

100%

100%

Low volume

35%

38%

0%

0%

Use of early docetaxel

18% (previous)

18% (previous)

0%

0%

Primary objective

rPFS


OS

rPFS

Median follow up (mo)

44.6

29.3

Median rPFS (mo.)

38.9

49.8

23.5

Not reached

HR (95% CI) for rPFS

HR: 0.63 (0.52–0.76)

HR: 0.46 (0.36–0.60)

Median OS

Not reached

Not reached

Not reached

Not reached

HR (95% CI) for OS

0.66 (0.53–0.81): Main secondary endpoint

0.58 (0.44–0.77)

HR = hazard ratio; mo = month; n = number of patients; OS = overall survival; rPFS = radiographic progression-free survival; yr = year. 

6.6.4.2.3. Combination with docetaxel and an ARPI

The addition of abiraterone to ADT and docetaxel has been reported to have a benefit in rPFS and in OS in the PEACE-1 trial [1126,1127]. The trial has a 2x2 factorial design and participants with de novo (synchronous) metastatic PCa were randomised to SOC; at the beginning of the trial ADT, later ADT plus docetaxel for 6 cycles if chemotherapy-fit) vs. SOC plus radiotherapy vs. SOC plus abiraterone vs. SOC plus radiotherapy plus abiraterone. Co-primary endpoints were rPFS and OS, both were statistically significantly improved in the total population. Also in the group of patients who received ADT plus docetaxel as SOC (n = 710) both rPFS and OS were increased with a HR: 0.5 (0.34–0.71) and 0.75 (0.59–0.95), respectively. Of note; in this population about 35% had low-volume disease. Toxicity was modestly increased, mostly hypertension.

In the ARASENS Phase III trial all patients received ADT and docetaxel for 6 cycles as SOC plus darolutamide or placebo [1128]. 1,306 metastatic patients were included, 14 % of them with relapsed disease after radical local treatment (metachronous). Primary endpoint was OS and this was statistically significanty increased by the addition of darolutamide with a HR of 0.68 (0.57–0.8).

Interestingly, in this trial the occurrence of AEs was similar in both arms. In both trials docetaxel and the ARPI have been given concomitantly. Of the included patients 77% had high volume and 70% high-risk disease. In an unplanned subgroup analysis the beneficial effect of adding darolutamide versus placebo for OS was seen in the patients with high-volume (HR 0.69; 0.57-0.82), with high-risk (HR 0.71; 0.58-0.86) and in low-risk disease (HR 0.62; 0.42-0.9), for the small subgroup of patients with low-volume disease the results were suggestive of an OS benefit (HR 0.68; 0.41-1.13) [1129]

Also in ENZAMET, TITAN and ARCHES there were patients who received docetaxel as a part of SOC, thus not all concomitantly, but the percentage of patients receiving docetaxel in these trials was much lower [1068,1069,1112].

There are also SRs and network meta-analysis for systemic triplet therapies and they confirm that the triplets are more effective than ADT and docetaxel alone [1130], in one analysis looking into subgroups statistically significant for patients with high volume disease and de novo disease [1131].

6.6.5. Treatment selection and patient selection

There have been several network meta-analyses of the published data concluding that combination therapy is more efficient than ADT alone, but none of the doublet combination therapies has been convincingly proven to be superior over another [1132-1137]. In a SR and meta-analysis looking at association between age and efficacy of combination therapy patients seemed to profit from combination therapy irrespective of age [1138]. As a consequence, patients should be offered combination treatment unless there are clear contra-indications or they present with asymptomatic disease and a very short life expectancy (based on non-cancer comorbidities).

Since the data of the above mentioned Phase III trials of the triplet therapies have been reported, docetaxel as sole addition to ADT is not longer a valid option in the majority of patients if an androgen receptor pathway inhibitor (ARPI) is available and there are no contra-indications to use one. From subgroup analysis of all the above-mentioned RCTs we know that probably all subgroups (high vs. low volume/risk and synchronous vs. metachronous) can profit from the addition of an ARPI to ADT. Therefore, in view of the current data the recommendation is using ADT plus ARPI as the sole additional therapy or the triplet with an ARPI plus docetaxel. Formally the question what the added value of adding docetaxel to ADT plus an ARPI has not been evaluated, but since triplet therapy seems not to add a lot of unexpected overlapping toxicities, the data should be discussed with patients who are fit for chemotherapy and an ARPI, realising that most of the toxicity is caused by adding the chemotherapy. There is more evidence for using the triplet in synchronous disease and the OS benefit in PEACE-1 seemed to be driven mostly by the high volume patients at the time point of the analysis for the publication, in ARASENS only few patients had low volume disease.

Of interest in some SRs and meta-analysis the authors found no significant difference for OS and/or PFS using the systemic triplet therapy compared to adding an ARPI alone to ADT [1135,1139-1141]. In contrast one meta-analysis suggested a benefit of systemic triplet therapy versus ADT and ARPI and another meta-analysis showed a benefit in patients with high volume disease [1136,1142]. In summary, the choice will most likely be driven by fitness for docetaxel, the nature of the disease (low/high volume; synchronous/metachronous), patient preference, the specific side effects, availability, logistics and cost.

6.6.6. Treatment of the primary tumour in newly diagnosed metastatic disease

The first reported trial evaluating prostate RT in men with metastatic castration-sensitive disease was the HORRAD trial. Four hundred and thirty-two patients were randomised to ADT alone or ADT plus IMRT with IGRT to the prostate. Overall survival was not significantly different (HR: 0.9 [0.7–1.14]), median time to PSA progression was significantly improved in the RT arm (HR: 0.78 [0.63–0.97]) [1143]. The STAMPEDE trial evaluated 2,061 men with metastatic castration-sensitive PCa (mCSPC) who were randomised to ADT alone vs. ADT plus RT to the prostate. This trial confirmed that RT to the primary tumour did not improve OS in unselected patients [1091]. However, following the results from CHAARTED and prior to analysing the data, the original screening investigations were retrieved, and patients categorised as low- or high volume. In the low-volume subgroup (n = 819) there was a significant OS benefit by the addition of prostate RT. This was confirmed by the latest analysis of long-term follow-up (median follow-up of 61 months [HR: 0.64 for OS benefit in the low-volume group]) [1144].

A secondary, not pre-planned analysis of the STAMPEDE trial confirmed the benefit of prostate RT in patients with ≤ 3 bone metastases, but also showed a benefit in patients with M1a disease [1145]. No evidence of difference in time to symptomatic local events was found with median follow-up of over five years [1144].The dose used in these indications should be equivalent of up to 72 Gy in 2 Gy fractions.

Therefore, RT of the prostate only in patients with low-volume metastatic disease should be considered. Of note, only 18% of these patients had additional docetaxel and no patients had additional AAP, so no clear recommendation can be made about triple combinations. In addition, it is not clear if these data can be extrapolated to RP as local treatment as results of ongoing trials are awaited.

In a SR and meta-analysis including the above two RCTs, the authors found that, overall, there was no evidence that the addition of prostate RT to ADT improved survival in unselected patients (HR: 0.92, 95% CI: 0.81–1.04, p = 0.195) [1146]. However, there was a clear difference in the effect of metastatic burden on survival with an absolute improvement of 7% in three-year survival in men who had four or fewer bone metastases.

6.6.7. Metastasis-directed therapy in M1-patients

In patients relapsing after a local treatment, a metastases-targeting therapy has been proposed, with the aim to delay systemic treatment. In a retrospective analysis on 211 patients treated with MDT, Milenkovic et al. aimed at defining prognostic factors for MFS, palliative ADT-free (pADT) survival and cause-specific survival (CSS). With a median follow-up of 42 months after MDT, patients with cN1 only had significantly superior 5-years MFS, pADT and CSS when compared to patients with M1 disease (p<0.02). Of interest, 23% of patients were free of biochemical recurrence at five years [1147]. There are two randomised phase II trials testing metastasis-directed therapy (MDT) using surgery ± SABR vs. surveillance [1148] or SABR vs. surveillance in men with oligo-recurrent PCa [1149]. Oligo-recurrence was defined as < 3 lesions on choline-PET/CT only [1148] or conventional imaging with MRI/CT and/or bone scan [1149]. The sample size was small with 62 and 54 patients, respectively, and a substantial proportion of them had nodal disease only [1148]. Androgen deprivation therapy-free survival was the primary endpoint in one study which was longer with MDT than with surveillance [1148]. The primary endpoint in the ORIOLE trial was progression after six months which was significantly lower with SBRT than with surveillance (19% vs. 61%, p = 0.005) [1149].

Recently the combined results of STOMP and ORIOLE confirmed the significant improvement in PFS in favour of MDT (HR: 0.44, p < 0.001) [1150].

A phase II trial assessed the biochemical response after 18F-DCFPyL PET/MRI and subsequent MDT. Overall biochemical response rate, defined as ≥ 50% PSA decline, was 60%, including 22% of patients with complete biochemical response [1151].

Currently there are no data to suggest an improvement in OS. Two comprehensive reviews highlighted MDT (SABR) as a promising therapeutic approach that must still be considered as investigational until the results of the ongoing RCT are available [1152,1153]. Thus far, the toxicity of MDT appears to be low, but this also needs to be confirmed [1154,1155].

6.6.8. Guidelines for the first-line treatment of hormone-sensitive metastatic disease*

Recommendations

Strength rating

Offer immediate systemic treatment with androgen deprivation therapy (ADT) to palliate symptoms and reduce the risk for potentially serious sequelae of advanced disease (spinal cord compression, pathological fractures, ureteral obstruction) to M1 symptomatic patients.

Strong

Offer short-term administration of an older generation androgen receptor (AR) antagonist to M1 patients starting LHRH agonist to reduce the risk of the ‘flare-up’ phenomenon.

Weak

At the start of ADT offer luteinising hormone-releasing hormone (LHRH) antagonists or orchiectomy to patients with impending clinical complications such as spinal cord compression or bladder outlet obstruction.

Strong

Do not offer AR antagonist monotherapy to patients with M1 disease.

Strong

Do not offer ADT monotherapy to patients whose first presentation is M1 disease if they have no contra-indications for combination therapy and have a sufficient life expectancy to benefit from combination therapy (≥ 1 year) and are willing to accept the increased risk of side effects.

Strong

Offer ADT combined with abiraterone acetate plus prednisone or apalutamide or enzalutamide to patients with M1 disease who are fit for the regimen.

Strong

Offer docetaxel only in combination with ADT plus abiraterone or darolutamide to patients with M1 disease who are fit for docetaxel.

Strong

Offer ADT combined with prostate radiotherapy (using doses up to the equivalent of 72 Gy in 2 Gy fractions) to patients whose first presentation is M1 disease and who have low volume of disease by CHAARTED criteria.

Strong

Do not offer ADT combined with surgery to M1 patients outside of clinical trials.

Strong

Only offer metastasis-directed therapy to M1 patients within a clinical trial setting or a well-designed prospective cohort study.

Strong

*All the following statements are based on metastatic disease defined by bone scintigraphy and CT scan/MRI.

6.7. Treatment: Castration-resistant PCa (CRPC)

6.7.1. Definition of CRPC

Castrate serum testosterone < 50 ng/dL or 1.7 nmol/L plus either:

a.Biochemical progression: Three consecutive rises in PSA at least one week apart resulting in two 50% increases over the nadir, and a PSA > 2 ng/mL or

b.Radiological progression: The appearance of new lesions: either two or more new bone lesions on bone scan or a soft tissue lesion using RECIST (Response Evaluation Criteria in Solid Tumours) [1156]. Symptomatic progression alone must be questioned and subject to further investigation. It is not sufficient to diagnose CRPC.

6.7.2. Management of mCRPC - general aspects

Selection of treatment for mCRPC is multifactorial and in general dependent on:

  • previous treatment for mHSPC and for non-mHSPC;
  • previous treatment for nmCRPC and mCRPC;
  • quality of response and pace of progression on previous treatment;
  • known cross resistance between androgen receptor pathway inhibitor (ARPI);
  • co-medication and known drug interactions (see approved summary of product characteristics);
  • known genetic alterations and microsatellite instability–high (MSI-H)/mismatch repair–deficient (dMMR) status;
  • known histological variants and DNA repair deficiency (to consider platinum or targeted therapy like PARPi);
  • local approval status of drugs and reimbursement situation;
  • available clinical trials;
  • The patient and his comorbidities.
6.7.2.1. Molecular diagnostics

All metastatic patients should be offered somatic genomic testing for homologous repair and MMR defects early on, preferably before firstline mCRPC treatment is established. Testing should preferably be performed on metastatic carcinoma tissue but testing on primary tumour may also be performed. Alternatively, but still less common, genetic testing on circulating tumour DNA (ctDNA) is an option and has been used in some trials. One test, the FoundationOne® Liquid CDx, has been FDA approved [1157]. Defective MMR assessment can be performed by IHC for MMR proteins (MSH2, MSH6, MLH1 and PMS2) and/or by next generation sequencing (NGS) assays [1158]. Germline testing for BRCA1/2, ATM and MMR is recommended for high-risk- and particularly for metastatic PCa if clinically indicated.

Molecular diagnostics should be performed by a certified (accredited) institution using a standard NGS multiplication procedure (minimum depth of coverage of 200 X). The genes and respective exons should be listed; not only DNA for mutations but RNA needs to be examined for fusions and protein expression to obtain all clinically relevant information. A critical asset is the decision support helping to rate the mutations according to their clinical relevance [1159,1160].

Level 1 evidence for the use of PARP-inhibitors has been reported [248,1161,1162]. Microsatellite instability (MSI)-high (or MMR deficiency) is rare in PCa, but for those patients, pembrolizumab has been approved by the FDA and could be a valuable additional treatment option [1080,1163]. Germline molecular testing is discussed in Section 5.1.6 - Genetic testing for inherited PCa. Recommendations for germline testing are provided in Section 5.1.7.

6.7.3. Treatment decisions and sequence of available options

Approved agents for the treatment of mCRPC in Europe are docetaxel, abiraterone/prednisolone (AAP), enzalutamide, cabazitaxel, olaparib, niraparib/AAP, talazoparib/enzalutamide, radium-223 and lutetium (177Lu) vipivotide tetraxetan. Regarding CRPC, darolutamide and apalutamide have been approved only for nmCRPC. In general, sequencing of ARPIs like abiraterone and enzalutamide is not recommended particularly if the time of response to ADT and to the first ARPI was short (≤ six to twelve months) and high-risk features of rapid progression are present (see detailed discussion in Section 6.7.7) [1164,1165].

The use of chemotherapy with docetaxel and subsequent cabazitaxel in the treatment sequence is recommended and should be applied early enough when the patient is still fit for chemotherapy. This is supported by high-level evidence [1164].

6.7.4. Non-metastatic CRPC

Frequent PSA testing in men treated with ADT has resulted in earlier detection of biochemical progression. Of these men approximately one-third will develop bone metastases within two years, detected by conventional imaging [893].

In men with CRPC and no detectable clinical metastases using bone scan and CT-scan, baseline PSA level, PSA velocity and PSA-DT have been associated with time to first bone metastasis, bone MFS and OS [893,1166]. These factors may be used when deciding which patients should be evaluated for metastatic disease. A consensus statement by the PCa Radiographic Assessments for Detection of Advanced Recurrence (RADAR) group suggested a bone scan and a CT scan when the PSA reached 2 ng/mL and if this was negative, it should be repeated when the PSA reached 5 ng/mL, and again after every doubling of the PSA based on PSA testing every three months in asymptomatic men [1167]. Symptomatic patients should undergo relevant investigations regardless of PSA level. With more sensitive imaging techniques like PSMA PET/CT or whole-body MRI, more patients are diagnosed with early mCRPC [1168]. It remains unclear if the use of PSMA PET/CT in this setting improves outcome.

Three large phase III RCTs, PROSPER [1169] , SPARTAN [1170] and ARAMIS [1171] , evaluated MFS as the primary endpoint in patients with nmCRPC (M0 CRPC) treated with enzalutamide (PROSPER) vs. placebo or apalutamide (SPARTAN) vs. placebo or darolutamide (ARAMIS) vs. placebo ,respectively (see Table 6.7.1). The M0 status was established by CT and bone scans. Only patients at high risk for the development of metastasis with a short PSA-DT of ≤ ten months were included. Patient characteristics in the trials revealed that about two-thirds of participants had a PSA-DT of < six months. All trials showed a significant MFS benefit. All three trials showed a survival benefit after a follow-up of more than 30 months. In view of the long-term treatment with these AR targeting agents in asymptomatic patients, potential AEs need to be taken into consideration and the patient informed accordingly.

Table 6.7.1: Randomised phase III controlled trials – nmCRPC

Study

Intervention

Comparison

Selection criteria

Main outcomes

ARAMIS

2019, 2020

[1171,1172]

ADT + darolutamide

ADT + placebo

nmCRPC;

baseline

PSA > 2 ng/mL

PSA-DT < 10 mo.

59% reduction of distant progression or death

Median MFS: darolutamide 40.4 vs placebo 18.4 mo;

31% reduction in risk of death

HR = 0.69 (95% CI: 0.53–0.88)

p = 0.003

PROSPER

2018, 2020

[1169,1173]

ADT + enzalutamide

ADT + placebo

nmCRPC;

baseline
PSA > 2 ng/mL

PSA-DT < 10 mo.

71% reduction of distant progression or death

Median MFS: enzalutamide 36.6 vs placebo 14.7 months;

27% reduction in risk of death

HR = 0.73 (95% CI: 0.61–0.89)

p = 0.001

SPARTAN

2018, 2021

[1170,1174]

ADT + apalutamide

ADT + placebo

nmCRPC;

baseline
PSA > 2 ng/mL

PSA-DT < 10 mo.

72% reduction of distant progression or death

Median MFS: apalutamide 
40.5 vs placebo 16.2 months;

22% reduction in risk of death

HR = 0.78 (95% CI: 0.64–0.96)

p = 0.0161

ADT = androgen-deprivation therapy; CI = confidence interval; HR = hazard ratio; MFS = metastasis-freesurvival; nmCRPC = non-metastatic castrate-resistent prostate cancer; PSA-DT = prostate-specific antigen doubling time.

6.7.5. Metastatic CRPC

The remainder of this section focuses on the management of men with proven mCRPC on conventional imaging.

6.7.5.1. Conventional androgen deprivation in CRPC

Eventually men with PCa will show evidence of disease progression despite castration. Two trials have shown only a marginal survival benefit for patients remaining on LHRH analogues during second- and thirdline therapies [1175,1176]. However, in the absence of prospective data, the modest potential benefits of continuing castration outweigh the minimal risk of treatment. In addition, all subsequent treatments have been studied in men with ongoing androgen suppression, therefore, it should be continued in these patients.

6.7.6. First-line treatment of metastatic CRPC

6.7.6.1. Abiraterone

Abiraterone was evaluated in 1,088 chemo-naive, asymptomatic or mildly symptomatic mCRPC patients in the phase III COU-AA-302 trial. Patients were randomised to abiraterone acetate or placebo, both combined with prednisone [1177]. Patients with visceral metastases were excluded. The main stratification factors were ECOG PS 0 or 1 and asymptomatic or mildly symptomatic disease. Overall survival and rPFS were the co-primary endpoints. After a median follow-up of 22.2 months there was significant improvement of rPFS (median 16.5 vs. 8.2 months, HR: 0.52, p < 0.001) and the trial was unblinded. At the final analysis with a median follow-up of 49.2 months, the OS endpoint was significantly positive (34.7 vs. 30.3 months, HR: 0.81, 95% CI: 0.70–0.93, p = 0.0033) [1178]. Adverse events related to mineralocorticoid excess and liver function abnormalities were more frequent with abiraterone, but mostly grade 1–2. Subset analysis of this trial showed the drug to be equally effective in an elderly population (> 75 years) [1179].

6.7.6.2. Enzalutamide

A randomised phase III trial (PREVAIL) included a similar patient population and compared enzalutamide and placebo [1180]. Men with visceral metastases were eligible but the numbers included were small. Corticosteroids were allowed but not mandatory. PREVAIL was conducted in a chemo-naive mCRPC population of 1,717 men and showed a significant improvement in both co-primary endpoints, rPFS (HR: 0.186, CI: 0.15–0.23, p < 0.0001), and OS (HR: 0.706, CI: 0.6–0.84, p < 0.001). A ≥ 50% decrease in PSA was seen in 78% of patients. The most common clinically relevant AEs were fatigue and hypertension. Enzalutamide was equally effective and well tolerated in men > 75 years [1181] as well as in those with or without visceral metastases [1182]. However, for men with liver metastases, there seemed to be no discernible benefit [1182,1183].

Enzalutamide has also been compared with bicalutamide 50 mg/day in a randomised double-blind phase II study (TERRAIN) showing a significant improvement in PFS (15.7 months vs. 5.8 months, HR: 0.44, p < 0.0001) in favour of enzalutamide [1183]. With extended follow-up and final analysis the benefit in OS and rPFS were confirmed [1184].

6.7.6.3. Docetaxel

A statistically significant improvement in median survival of 2.0–2.9 months has been shown with docetaxel based chemotherapy compared to mitoxantrone plus prednisone [1185,1186]. The standard first-line chemotherapy is docetaxel 75 mg/m2, three-weekly doses combined with prednisone 5 mg twice a day (BID), up to ten cycles. Prednisone can be omitted if there are contra-indications or no major symptoms. The following independent prognostic factors: visceral metastases, pain, anaemia (Hb < 13 g/dL), bone scan progression, and prior estramustine may help stratify the response to docetaxel. Patients can be categorised into three risk groups: low risk (0 or 1 factor), intermediate (2 factors) and high risk (3 or 4 factors), and show three significantly different median OS estimates of 25.7, 18.7 and 12.8 months, respectively [1187].

Age by itself is not a contra-indication to docetaxel [1188] but attention must be paid to careful monitoring and comorbidities as discussed in Section 5.4 - Estimating life expectancy and health status [1189]. In men with mCRPC who are thought to be unable to tolerate the standard dose and schedule, docetaxel 50 mg/m2 every two weeks seems to be well tolerated with less grade 3–4 AEs and a prolonged time to treatment failure [1190].

6.7.6.4. Sipuleucel-T

In 2010 a phase III trial of sipuleucel-T showed a survival benefit in 512 asymptomatic or minimally symptomatic mCRPC patients [1191]. After a median follow-up of 34 months, the median survival was 25.8 months in the sipuleucel-T group compared to 21.7 months in the placebo group, with a HR of 0.78 (p = 0.03). No PSA decline was observed and PFS was similar in both arms. The overall tolerance was very good, with more cytokine-related AEs grade 1–2 in the sipuleucel-T group, but the same grade 3–4 AEs in both arms. Sipuleucel-T is not available in Europe.

6.7.6.5. Ipatasertib

The AKT inhibitor ipatasertib in combination with AAP was studied in asymptomatic or mildly symptomatic patients with and without PTEN loss by IHC and previously untreated for mCRPC. The randomised phase III trial (IPAtential) showed a significant benefit for the first endpoint rPFS in the PTEN loss (IHC) 18.5 vs. 16.5 mo; p = 0.0335, HR: 0.77, 95% CI: 0.61–0,98) but not in the intention to treat (ITT) population. The OS results are still pending. Side effects of the AKT inhibitor ipatasertib include rash and diarrhoea [1077]. Grade 3 or higher AEs occurred nearly double as often in the combination group and the discontinuation rate due to AEs was four times higher. This combination is still investigational [1192].

6.7.6.6. Combinations with PARP inhibitors

Based on the suggestion that there is a synergistic antitumour effect when combining an ARPI with a PARP inhibitor, several such combination trials were conducted in first-line mCRPC patients with different trial designs, different patient selection and conflicting results.

Abiraterone/prednisone plus olaparib

A randomised double-blind, phase III trial (PROpel) of AAP plus olaparib (300 mg twice daily) or placebo in patients with mCRPC in the first-line setting was conducted [1193,1194]. Patients (n=796) were randomly assigned 1:1 to study treatment regardless of homologous recombination repair gene mutation (HRRm) status which was retrospectively evaluated and determined by tumour tissue and circulating tumour DNA tests. The primary end point was imaging-based PFS (ibPFS) by investigator assessment. The result was significantly positive in favour of the combination with ibPFS of 24.8 vs. 16.6 mo (HR 0.66; 95% CI: 0.54 to 0.81; p = 0.001). In the prespecified final analyses the key secondary endpoint OS had only 47.9% maturity and did not meet the prespecified 2-sided boundary for significance (HR 0.95% CI: 0.81, 0.67-1.0, p= 0,054).The subgroup of patients with positive HRRm status showed a rPFS HR of 0.50 (CI: 0.34 to 0.73). The BRCA mutated patients (11% of the ITT population) had an even larger benefit for rPFS (HR 0.24; 95% CI: 0.12, 0.45) and the OS HR in these patients was 0.30 (95% CI: 0.15, 0.59), suggesting that the improvement in rPFS observed in the ITT population was primarily driven by patients with a BRCA mutation [1195].

The most common AEs in patients receiving olaparib plus AAP were anaemia (48%; ≥G3 15%; at least one blood transfusion in 18%; multiple transfusions 12% [1195]. The most common adverse reactions with olaparib plus abiraterone were anaemia (48%), fatigue (38%), nausea (30%), diarrhoea (19%), decreased appetite (16%), lymphopenia (14%), dizziness (14%), and abdominal pain (13%); 18% of patients required at least one blood transfusion and 12% required multiple transfusions. The combination of olaparib plus AAP was approved by the EMA for the treatment of adult patients with mCRPC in whom chemotherapy is not clinically indicated [1240]. In the US, the FDA has approved olaparib with AAP for mCRPC patients with deleterious or suspected deleterious BRCA-mutations as determined by an FDA-approved companion diagnostic test [1197].

Abiraterone/prednisone plus niraparib

In a randomised, double-blind, phase III trial (MAGNITUDE) AAP plus niraparib 200 mg once/daily or placebo, was evaluated [1198]. The study prospectively included 2 cohorts, an HRR-negative and an HRR-positive cohort. The HRR-negative cohort was closed early for futility after enrolling 200 patients. In the overall HRR-positive cohort, the addition of Niraparib to AAP resulted in a significant improvement in the first endpoint rPFS compared to AAP plus placebo (HR = 0.73; 95% CI 0.56-0.96; p = 0.0217) and the median rPFS was 16.5 vs. 13.7 months in favour of the combination. In particular, the 113 patients with BRCA 1/ 2 mutations [1199] who received AAP plus niraparib [1199] derived a major rPFS benefit (19.5 versus 10.9 months; HR = 0.55 [95% CI 0.39-0.78]; nominal p = 0.0007). The OS data is still immature. The most common side effects with Niraparib plus AAP in the ITT population were anemia (46.2%), fatigue (26.4%), hypertension (31.6%) and constipation (30.7%). The combination of niraparib plus AAP in a dual-action tablet has been approved by the EMA and the FDA for patients with mCRPC and BRCA 1/2 mutations in whom chemotherapy is not clinically indicated [1200].

Enzalutamide plus Talazoparib

A randomised double-blind, phase III trial (TALAPRO-2) of the PARP inhibitor talazoparib (0.5mg daily) plus enzalutamide versus enzalutamide/placebo showed a significantly better median rPFS (first endpoint) in favour of the combintion regardless of homologous recombination repair pathway status [1202].

The median was not yet reached for the combination as compared to 21.9 mo in the control arm (95% CI 16.6-25.1). The HR for rPFS was 0.63 (0.51-0.78) with p<0.0001. For the subgroups of patients with HRR mutations the benefit of the combination was much more pronounced. The HRR gene-mutated population showed a median rPFS of 27.9 (16.6–not reached) for the talazoparib combination versus 16.4 (10.9–24.6) for the placebo group (0.46; 95% CI: 0.30–0.70; p=0.0003 ) and 0.70 (0.54–0.89; p=0.0039) in patients with a status of non-deficient or unknown. In an exploratory analysis, the HR for rPFS in patients with BRCA-mutated mCRPC was 0.23 (0.10–0.53; p=0.0002) and, in patients with non-BRCAm HRR gene-mutated mCRPC, it was 0.66 (0.39–1.12; p=0.12) in favour of the talazoparib combination.[1202] The OS data are still immature. The expected clinical benefit in the subgroups needs to be weighed against the potential burden of side effects.

The most common treatment-emergent adverse events with the addition of talazoparib were anaemia, neutropenia, and fatigue; the most common grade 3–4 event was anaemia (46%), which improved after dose reduction, however, 39% required a blood transfusion, including 22% who required multiple transfusions, 8% discontinued treatment due to anemia and 2 patients on the combination were diagnosed with myelodysplastic syndrome/acute myeloid leukemia [1202]. In TALAPRO-2 also an HRR-deficient-only cohort (cohort 2; N = 230) was recruited. The primary analysis for the combined HRR-deficient population (N = 399) met the rPFS endpoint with a HR 0.45 (95% CI, 0.33 to 0.61; P < 0.0001; median not reached at the time of the analysis for the talazoparib group versus 13.8 months for the placebo group). Also for this cohort data for OS are immature but favor talazoparib (HR 0.69; 95% confidence interval, 0.46 to 1.03; P = 0.07) [1203].

The FDA approved talazoparib with enzalutamide only for HRR gene-mutated mCRPC [1201]. The European commission approved talazoparib with enzalutamide for the treatment of patients with mCRPC (with or without gene mutations) in whom chemotherapy is not clinically indicated.

Table 6.7.2: Randomised phase III controlled trials - first-line treatment of mCRPC

Study

Intervention

Comparison

Selection criteria

Main outcomes

DOCETAXEL

SWOG 99-16

2004 [1204]

docetaxel/EMP,

every 3 weeks,

60 mg/m2, EMP

3 x 280 mg/day

mitoxantrone,

every 3 weeks,

12 mg/m2 prednisone 5 mg BID


OS: 17.52 vs. 15.6 mo. (p = 0.02, HR: 0.80; 95% CI: 0.67-0.97) PFS: 6.3 vs. 3.2 mo.

(p < 0.001)

TAX 327

2004, 2008

[1185,1186]

docetaxel, every

3 weeks, 75 mg/m2

prednisone 5 mg BID

or

docetaxel, weekly, 30 mg/m2

prednisone 5 mg

BID

mitoxantrone,

every 3 weeks,

12 mg/m2, Prednisone 5 mg BID


OS: 19.2 for 3 weekly vs. 17.8 mo. 4-weekly and 16.3 in the control group.

(p = 0.004, HR: 0.79, 95% CI: 0.67-0.93)

ABIRATERONE

COU-AA-302

2013, 2014, 2015 [1177,1178,1205]

abiraterone +

prednisone

placebo + prednisone

- No previous docetaxel.

- ECOG 0-1.

- PSA or radiographic progression.

- No or mild symptoms.

- No visceral metastases.

OS: 34.7 vs. 30.3 mo. (HR: 0.81, p = 0.0033). FU: 49.2 mo. rPFS: 16.5 vs. 8.3 mo.

(p < 0.0001)

ENZALUTAMIDE

PREVAIL

2014 [1180]

enzalutamide

placebo

- No previous docetaxel.

- ECOG 0-1.

- PSA or radiographic progression.

- No or mild symptoms.

- 10% had visceral mets.

OS: 32.4 vs. 30.2 mo. (p < 0.001). FU: 22 mo. (p < 0.001 HR: 0.71, 95% CI: 0.60-0.84) rPFS: 20.0 mo. vs.

5.4 mo. HR: 0.186 (95% CI: 0.15-0.23)

p < 0.0001)

SIPULEUCEL-T

IMPACT 2010 [1191]

sipuleucel-T

placebo

- Some with previous docetaxel.

- ECOG 0-1.

- Asymptomatic or minimally symptomatic.

OS: 25.8 vs. 21.7 mo. (p = 0.03 HR: 0.78, 95% CI: 0.61-0.98).
FU: 34.1 mo. PFS: 3.7 vs. 3.6 mo. (no difference)

2006 [1206]

sipuleucel-T

placebo

- ECOG 0-1.

- No visceral met.

- No corticosteroids.

OS: 25.9 vs. 21.4 mo. (p = 0.1). FU: 36 mo. PFS: 11.7 vs. 10.0 wk.

IPATASERTIB

IPAtential150

2021 [1192]

ipatasertib (400 mg/d) + abiraterone (1000 mg/d) + prednisone
(5 mg bid)

abiraterone + prednisolone + placebo

Previously untreated for mCRPC, asymptomatic/mildly symptomatic, with and without PTEN loss by IHC

rPFS in PTEN loss (IHC) population: 
18.5 vs. 16.5 mo.

(p = 0.0335, HR: 0.77 95% CI: 0.61-0.98)

COMBINATIONS

PROpel [1193,1194]

olaparib (300mg BID)

+ abiraterone (1000 mg/d) + prednisone (5 mg BID)

placebo + abiraterone + prednisone

-ECOG 0-1

- regardless of HRRm (retrospective testing)

- prior taxane for mHSPC allowed

ibPFS in ITT population:

24.8 vs. 16.6 mo; 
HR: 0.66; 95% CI: 0.54–0.81;

(p = 0.001)

ibPFS in BRCA+:

HR 0.24; 95% CI: 0.12- 0.45





OS in ITT population: 42.1 vs. 34.7 mo; 
HR 0.81; 95% CI:

0.67-1.0;

(p= 0.054)

OS in BRCA+: HR 0.29; 95% CI: 0.15- 0.56

MAGNITUDE [1199,1207]

niraparib 200 mg/d + abiraterone (1000 mg/d plus prednisone 5 mg BID)

placebo + abiraterone (1,000 mg/d plus prednisone 5 mg BID)

- ECOG 0-1

- AAP ≤ 4mo allowed for mCRPC;

- HRR-biomarker positive cohort

- prior docetaxel for mHSPC allowed

- prior ARPI for mHSPC allowed

- prior ARPI for mCRPC allowed

rPFS (central review) in HRR+:

16.5 vs. 13.7 mo

HR = 0.73; 95% CI: 0.56-0.96; (p = 0.022)

rPFS (central review) in BRCA 1/ 2+:

16.5 vs. 13.7 mo

rPFS 19.5 versus 10.9 months; HR= 0.55; 95% CI 0.39-0.78; (nominal p= 0.0007)

TALAPRO-2 [1202]

talazoparib (0.5mg/d) + enzalutamide 160mg/d

enzalutamide + placebo

- ECOG 0-1

- All-comers: HHR deficient and HRR non-deficient or unknown

-prior AAP or docetaxel allowed for mHSPC

rPFS in ITT: NR (27.5-NR) vs. 21.9 mo;

HR 0.63; 95% CI: 0.51-0.78 (p<0.0001);

rPFS in BRCA+:

HR 0·23; 95%

CI: 0.10–0.53; p=0·0002

BID = twice a day; CI = confidence interval; ECOG = Eastern Cooperative Oncology Group; EMP = estramustine; FU = follow-up; HR = hazard ratio; mets. = metastases; mo = month; ib (imaging based); (r)PFS = (radiographic) progression-free survival; OS = overall survival; IHC = immunohistochemistry ; HRRm = homologour recombination repair genes mutation; BRCA+ = BRCA gene mutated; ITT = intention to treat; BICR = blinded independent central review.

6.7.7. Second-line treatment for mCRPC

All patients who receive treatment for mCRPC will eventually progress. All treatment options in this setting are presented in Table 6.7.3. High-level evidence exists for second-line treatments after first-line treatment for mCRPC with docetaxel or with ARPI. There is a paucity of high-level data with regards to the sequence of treatments in case of pretreatment with ARPI and/or docetaxel for mHSPC.

6.7.7.1. Cabazitaxel

Cabazitaxel is a taxane with activity in docetaxel-resistant cancers. It was studied in a large prospective, randomised, phase III trial (TROPIC) comparing cabazitaxel plus prednisone vs. mitoxantrone plus prednisone in 755 patients with mCRPC, who had progressed after or during docetaxel-based chemotherapy [1208]. Patients received a maximum of ten cycles of cabazitaxel (25 mg/m2) or mitoxantrone (12 mg/m2) plus prednisone (10 mg/day). Overall survival was the primary endpoint which was significantly longer with cabazitaxel (median: 15.1 vs. 12.7 months, p < 0.0001). There was also a significant improvement in PFS (median: 2.8 vs. 1.4 months, p < 0.0001), objective RECIST response (14.4% vs. 4.4%, p < 0.005), and PSA response rate (39.2% vs. 17.8%, p < 0.0002). Treatment-associated WHO grade 3–4 AEs developed significantly more often in the cabazitaxel arm, particularly haematological (68.2% vs. 47.3%, p < 0.0002) but also non-haematological (57.4 vs. 39.8%, p < 0.0002) toxicity. In two post-marketing randomised phase III trials, cabazitaxel was shown not to be superior to docetaxel in the first-line setting; in the second-line setting in terms of OS, 20 mg/m2 cabazitaxel was not inferior to 25 mg/m2, but less toxic. Therefore, the lower dose should be preferred [1209,1210]. Cabazitaxel should preferably be given with prophylactic granulocyte colonystimulating factor (G-CSF) and should be administered by physicians with expertise in handling neutropenia and sepsis [1211].

6.7.7.2. Abiraterone acetate after docetaxel

Positive results of the large phase III trial (COU-AA-301) were reported after a median follow-up of 12.8 months [1212] and confirmed by the final analysis [1213]. A total of 1,195 patients with mCRPC were randomised 2:1 to AAP or placebo plus prednisone. All patients had progressive disease based on the Prostate Cancer Clinical Trials Working Group 2 (PCWG2) criteria after docetaxel therapy (with a maximum of two previous chemotherapeutic regimens). The primary endpoint was OS, with a planned HR of 0.8 in favour of AAP. After a median follow-up of 20.2 months, the median survival in the AAP group was 15.8 months compared to 11.2 months in the placebo arm (HR: 0.74, p < 0.0001). The benefit was observed in all subgroups and all the secondary objectives were in favour of AAP (PSA, radiologic tissue response, time to PSA or objective progression). The incidence of the most common grade 3–4 AEs did not differ significantly between arms, but mineralocorticoid-related side effects were more frequent in the AAP group, mainly grade 1–2 (fluid retention, oedema and hypokalaemia).

6.7.7.3. Enzalutamide after docetaxel

The planned interim analysis of the AFFIRM study was published in 2012 [1214]. This trial randomised 1,199 patients with mCRPC in a 2:1 fashion to enzalutamide or placebo. The patients had progressed after docetaxel treatment, according to the PCWG2 criteria. Corticosteroids were not mandatory, but could be prescribed, and were received by about 30% of the patients. The primary endpoint was OS, with an expected HR benefit of 0.76 in favour of enzalutamide. After a median follow-up of 14.4 months, the median survival in the enzalutamide group was 18.4 months compared to 13.6 months in the placebo arm (HR: 0.63, p < 0.001). This led to the recommendation to halt and unblind the study. The benefit was observed irrespective of age, baseline pain intensity, and type of progression. In the final analysis with longer follow-up the OS results were confirmed despite crossover and extensive post-progression therapies [1160]. Enzalutamide was active also in patients with visceral metastases.

All the secondary objectives were in favour of enzalutamide (PSA, soft tissue response, QoL, time to PSA, or objective progression). No difference in terms of side effects was observed in the two groups, with a lower incidence of grade 3–4 AEs in the enzalutamide arm. There was a 0.6% incidence of seizures in the enzalutamide group compared to none in the placebo arm.

6.7.7.4. Radium-223 after ARPI or both ARPI and docetaxel

The only bone-specific drug that is associated with a survival benefit is the α-emitter radium-223. In a large phase III trial (ALSYMPCA) 921 patients with symptomatic mCRPC, who failed or were unfit for docetaxel, were randomised to six injections of 50 kBq/kg radium-223 or placebo plus SOC. The primary endpoint was OS. Radium-223 significantly improved median OS by 3.6 months (HR: 0.70, p < 0.001) and was also associated with prolonged time to first skeletal event, improvement in pain scores and improvement in QoL [1215]. The associated toxicity was mild and, apart from slightly more haematologic toxicity and diarrhoea with radium-223, which did not differ significantly from that in the placebo arm [1215]. Radium-223 was effective and safe whether or not patients were docetaxel pre-treated [1216]. Due to safety concerns, use of radium-223 was restricted to after docetaxel and at least one AR targeted agent [1217]. In particular, the use of radium-223 in combination with AAP showed significant safety risks related to fractures and more deaths. This was most striking in patients without the concurrent use of bone health agents [1218] so that radium-223 should always be used together with bone health agents (see chapter 6.7.11.2)

6.7.7.5. Rucaparib after ARPI [1219]

In a 2:1 randomised, controlled, phase III trial (TRITON-3) 405 mCRPC patients were included. Patients were selected for a BRCA1, BRCA2, or ATM alteration and disease progression after treatment with an ARPI for mCRPC. Treatment was as follows: rucaparib 600 mg twice daily or a physician’s choice control, either second line docetaxel or the ARPI which had not been given previously. The first endpoint rPFS in the intention-to-treat group was significantly better with rucaparib (median, 10.2 months and 6.4 months, respectively; HR 0.61; 95% CI, 0.47 to 0.80; p<0.001). The small ATM subgroup did not derive abenefit. An interim analysis revealed OS to be immature. The study designed allowed for cross-over and 60% of patients received a PARP inhibitor at progression (47% rucaparib). With regards to the control arms, the median rPFS was longer with rucaparib than with docetaxel (11.2 months vs. 8.3 months; hazard ratio, 0.53; 95% CI, 0.37 to 0.77) and it was also longer than with an ARPI (11.2 months vs. 4.5 months; hazard ratio, 0.38; 95% CI, 0.25 to 0.58). The most frequent adverse events with rucaparib were fatigue, nausea and anemia, including 24% Grade ≥3 anemia and 29% of patients on rucaparib required at least one blood transfusion [1220]. Rucaparib has been approved by the FDA.

6.7.7.6. Olaparib after ARPI (see chapter 6.7.8.3 PARP inhibitors for mCRPC)

6.7.8. Treatment after docetaxel and one line of hormonal treatment for mCRPC

6.7.8.1. General considerations

For men progressing quickly on AR targeted therapy (< twelve months) it is now clear that cabazitaxel is the treatment supported by the best data. The CARD trial, an open label randomised phase III trial, evaluated cabazitaxel after docetaxel and one line of ARPI (either AAP or enzalutamide) [1164]. It included patients progressing in less than twelve months on previous abiraterone or enzalutamide for mCRPC. Cabazitaxel more than doubled rPFS vs. another ARPI and reduced the risk of death by 36% vs. ARPI. The rPFS with cabazitaxel remained superior regardless of the ARPI sequence and if docetaxel was given before, or after, the first ARPI.

The choice of further treatment after docetaxel and one line of HT for mCRPC is open for patients who have a > twelve months response to first-line abiraterone or enzalutamide for mCRPC [1184]. Either second-line chemotherapy (cabazitaxel), radium-223 (if bone-only metastases), 177Lu–PSMA-617 radioligand therapy [1221,1222] and PARP inhibitors (if BRCA mutation) are valuable options.

Men previously treated with at least one ARPI or both an ARPI and docetaxel and whose tumours demonstrated homozygous deletions or deleterious mutations in DNA-repair genes showed an 88% response rate to olaparib [1223] and in another confirmatory trial a composite response of 54.3% (95% CI: 39.0–69.1) in the 400 mg cohort and in 18 of 46 (39.1%; 25.1–54.6) evaluable patients in the 300 mg cohort [1224]. See also section ‘Second-line management’). In general, subsequent treatments in unselected patients are expected to have less benefit than with earlier use [1225,1226] and there is evidence of cross-resistance between enzalutamide and abiraterone [1227,1228].

6.7.8.2. Radiopharmaceuticals
6.7.8.2.1. Introduction

Historically, several radiopharmaceuticals including phosphorous-32, strontium-89, yttrium-90, samarium-153, and rhenium-186 were developed for the treatment of bone pain secondary to metastases from PCa [1229]. They proved effective in a palliation setting, by relieving pain and improving QoL, especially in the setting of diffuse bone metastases. However, they never gained widespread adoption. The first radioisotope to demonstrate a survival benefit was radium-223 (see Section 6.7.7.4).

6.7.8.2.2. PSMA-based therapy

The increasing use of PSMA PET as a diagnostic tracer and the realisation that this allowed identification of a greater number of metastatic deposits led to attempts to treat cancer by replacing the imaging isotope with a therapeutic isotope which accumulates where the tumour is demonstrated (theranostics) [1230]. Therefore, after identification of the target, usually with diagnostic 68Gallium-labelled PSMA, therapeutic radiopharmaceuticals labelled with β(lutetium-177 or yttrium-90) or α(actinium-225)-emitting isotopes could be used to treat metastatic PCa.

The PSMA therapeutic radiopharmaceutical supported by the most robust data is 177Lu-PSMA-617. The first patient was treated in 2014 and early clinical studies evaluating the safety and efficacy of 177Lu-PSMA therapy have demonstrated promising results, despite the fact that a significant proportion of men had already progressed on multiple therapies [1231]. The early data were based on single-centre experience [1232]. Data from uncontrolled prospective phase II trials reported high response rates with low toxic effects [1233,1234]. Positive signals are also coming from a randomised pase II trial (TheraP) [1235].

In TheraPpatients for whom cabazitaxel was considered the next appropriate standard treatment after docetaxel and who were highly selected by 68Ga-PSMA-11 and 18FDG PET-CT scans, were randomised to receive 177Lu-PSMA-617 (6.0–8.5 GBq intravenously, every six weeks, for up to six cycles) or cabazitaxel (20 mg/m2 for up to ten cycles). The primary endpoint was a reduction of at least 50% in PSA. The first endpoint was met (66% vs. 37% for 177Lu–PSMA-617 vs. cabazitaxel, respectively, by intention to treat; difference 29% (95% CI: 16–42; p < 0.0001; and 66% vs. 44% by treatment received; difference 23% [9–37]; p = 0.0016) [1235]. At 36 months follow-up, the secondary endpoint OS was similar in those patients randomly assigned to 177Lu-PSMA versus cabazitaxel (19.1 vs. 19.6 months, difference -0.5, 95% CI -3.7 to + 2.7; 177Lu-PSMA vs. cabazitaxel, respectively), HR: 0.97, 95% CI: 0.7-1.4, p=0.99) [1236].

An open-label phase III trial (VISION) compared 177Lutetium Vipivotid tetraxetan (177Lu-PSMA-617 radioligand therapy) with protocol-permitted SOC (i.e., excluded chemotherapy, immunotherapy, radium-223 and investigational drugs) in mCRPC patients, with PSMA expressing metastases on PET/CT, previously treated with at least one ARPI and one (around 53%) or two taxanes. Imaging-based PFS and OS were the alternate primary endpoints. More than 800 patients were randomised. 177Lu-PSMA-617 plus SOC significantly prolonged both imaging-based PFS and OS, as compared with SOC alone (see Table 6.6.3). Grade 3 or above AEs were higher with 177Lu-PSMA-617 than without (52.7% vs. 38.0%), but QoL was not adversely affected. 177Lu–PSMA-617 has shown to be an additional treatment option in this mCRPC population [1237].

A SR and updated meta-analysis, investigated the proportion of patients with any or more than 50% PSA decrease, and OS. The review, including 69 articles and a total of 4,157 patients, showed that patients treated with 177Lu–PSMA 617 had a significantly higher response to therapy compared to controls, based on ≥ 50% PSA decrease (OR = 5.33, 95% CI: 1.24–22.90, p < 0.05). Meta-analysis revealed an OS of 0.26 according to pooled HRs for any PSA decline, which was significant after 177Lu–PSMA-617 therapy (95% CI: 0.18–0.37, p < 0.00001) and an OS of 0.52 for ≥ 50% PSA decrease, also significant after radioligand (RLT) (95% CI: 0.40–0.67, p < 0.00001) [1238].

There is an increasing interest for PSMA-targeted alpha therapy (225Ac-PSMA) due to the ability to deliver potent higher local radiation more selectively to cancer cells than PSMA-targeted beta therapy, while minimising unwanted damage to the surrounding normal tissues. Additionally, the intensive radiation to cancer cells results in more effective DNA strand breakage and reduces the development of treatment resistance. A meta-analysis, including nine studies with 263 patients, investigated the therapeutic effects of 225Ac-PSMA RLT in patients with metastatic CRPC, pre-treated with chemotherapy, 177Lu-PSMA and/or radium-223. The pooled proportions of patients with more than 50% PSA decline and any PSA decline were 60.99% (95% CI: 54.92%– 66.83%) and 83.57% (95% CI: 78.62%–87.77%), respectively. The estimated mean PFS and mean OS were 9.15 months (95% CI: 6.69–11.03 months) and 11.77 months (95% CI: 9.51–13.49 months), respectively. These findings suggests that 225Ac-PSMA RLT may be an effective treatment option for patients with mCRPC [1239]. Despite the encouraging therapeutic response and survival of patients who received 225Ac-PSMA RLT, major AEs like xerostomia and severe haematotoxicity have to be considered as possible reasons for dose reduction or discontinuation of the therapy.

6.7.8.3. PARP inhibitors for mCRPC

So far, two PARP inhibitors as monotherapy, olaparib and rucaparib, are licenced by the FDA (EMA only approved olaparib) and several other PARP inhibitors are under investigation or were approved only in combination with an ARPI (see chapter 6.7.6.6; e.g., talazoparib, niraparib).

A randomised phase III trial (PROfound) compared the PARP inhibitor olaparib to an alternative ARPI in mCRPC with alterations in ≥ 1 of any qualifying gene with a role in HRR and progression on an ARPI. Most patients were heavily pre-treated with 1–2 chemotherapies and up to 2 ARPIs [248,1162]. Radiographic PFS by blinded independent central review in the BRCA1/2 or ATM mutated population (Cohort A) was the first endpoint and significantly favoured olaparib (HR: 0.49, 95% CI: 0.38–0.63). The final results for OS demonstrated a significant improvement among men with BRCA1/2 or ATM mutations (Cohort A) (p = 0.0175; HR: 0.69, 95% CI: 0.50– 0.97). This was not significant in men with any (other) HRR alteration (Cohort B) (HR: 0.96, 95% CI: 0.63–1.49). Of note, patients in the physician’s choice of enzalutamide/abiraterone-arm who progressed, 66% (n = 86/131) crossed over to olaparib.

The most common AEs were anaemia (46.1% vs. 15.4%), nausea (41.4% vs. 19.2%), decreased appetite (30.1% vs. 17.7%) and fatigue (26.2% vs. 20.8%) for olaparib vs. enzalutamide/abiraterone. Among patients receiving olaparib 16.4% discontinued treatment secondary to an AEs, compared to 8.5% of patients receiving enzalutamide/abiraterone. Interestingly, 4.3% of patients receiving olaparib had a pulmonary embolism, compared to 0.8% among those receiving enzalutamide/abiraterone, none of which were fatal. There were no reports of myelodysplastic syndrome or acute myeloid leukaemia. This is the first trial to show a benefit for genetic testing and precision medicine in mCRPC.

The olaparib approval by the FDA is for patients with deleterious or suspected deleterious germline- or somatic HRR gene-mutated mCRPC, who have progressed following prior treatment with enzalutamide or abiraterone. The EMA approved olaparib for patients with BRCA1 and BRCA2 alterations [1240]. The recommended olaparib dose is 600 mg daily (300 mg taken orally twice daily), with or without food.

Rucaparib has been approved by the FDA for patients with deleterious BRCA mutations (germline and/or somatic) who have been treated with ARPI and a taxane-based chemotherapy [1241]. Approval was based on the results of the single-arm TRITON2 trial (NCT02952534). The confirmed ORR per independent radiology review in 62 patients with deleterious BRCA mutations was 43.5% (95% CI: 31–57) [1242]. Rucaparib second line after ARPI was studied in the TRITON 3 trial and is discussed in chapter 6.7.7.5

The combination of ARPI plus a PARP inhibitor in first-line mCRPC was studied in several RCTs including AAP plus Olaparib [1193], AAP plus Niraparib [1198] and Enzalutamide plus Talazoparib [1202]. See Table 6.7.2.

6.7.8.4. Sequencing treatment
6.7.8.4.1. ARPI -> ARPI (chemotherapy-naïve mCRPC patients)

The use of sequential ARPIs in mCRPC showed limited benefit in retrospective series as well as in one prospective trial [1245-1252]. In particular in patients who had a short response to the first ARPI for mCRPC (< twelve months), this sequence should be avoided because of known cross resistance and the availability of chemotherapy and PARP inhibitors (if a relevant mutation is present).

In highly selected patients treated for more than 24 weeks with AAP, the sequence with enzalutamide showed some activity with a median rPFS of 8.1 months (95% CI: 6.1–8.3) and an unconfirmed PSA response rate of 27% [1253]. In case the patient is unfit for chemotherapy and a PARP inhibitor, best supportive care should be considered in case no other appropriate treatment option is available (clinical trial or immunotherapy if MSI-high). An ARPI-ARPI sequence should never be the preferred option but might be considered in such patients if the PS still allows for active treatment and the potential side effects seem manageable.

First prospective cross-over data on an ARPI-ARPI sequence [1245] and a SR and meta-analysis suggest that for the endpoints PFS and PSA PFS, but not for OS, abiraterone followed by enzalutamide is the preferred choice [1254].

6.7.8.4.2. ARPI -> PARP inhibitor

This sequence in patients with deleterious or suspected deleterious germline or somatic HRR gene-mutated mCRPC is supported by data from the randomised phase III PROfound trial studying olaparib [1162] and TRITON 3 studying rucaparib [1219]. A subgroup of patients in pROfound was pre-treated with one or two ARPIs and no chemotherapy (35%).

The ARPI-PARP inhibitor sequence versus ARPI-ARPI or ARPI-docetaxel in patients with BRCA 1/ 2 (and ATM) altered tumours was studied in TRITON-3 and showed a significant rPFS benefit in favour of the PARP inhibitor following the first ARPI. These data underscore the importance of early genomic testing in mCRPC patients. (see also chapter 6.7.7.5)

6.7.8.4.3. Docetaxel for mHSPC -> docetaxel rechallenge

There is limited evidence for second- or third-line use of docetaxel after treatment with docetaxel for mHSPC. Docetaxel seems to be less active than ARPI at progression to mCRPC following docetaxel for mHSPC [1255].

6.7.8.4.4. ARPI -> docetaxel or docetaxel -> ARPI followed by PARP inhibitor

Both olaparib and rucaparib are active in biomarker-selected mCRPC patients after ARPI and docetaxel in either sequence [1162,1241].

6.7.8.4.5. ARPI before or after docetaxel

There is level 1 evidence for both sequences (see Table 6.7.3).

6.7.8.4.6. ARPI –> docetaxel -> cabazitaxel or docetaxel –> ARPI -> cabazitaxel

Both third-line treatment sequences are supported by level 1 evidence. Of note, there is high-level evidence favouring cabazitaxel vs. a second ARPI after docetaxel and one ARPI in particular in patients progressing ≤ twelve months on a prior ARPI. CARD is the first prospective randomised phase III trial addressing this question (see Table 6.7.3) [1164].

Table 6.7.3: Randomised controlled phase II/III - second-line/third-line trials in mCRPC

Study

Intervention

Comparison

Selection criteria

Main outcomes

ABIRATERONE

COU-AA-301

2012 [1213]

abiraterone + prednisone HR

placebo + prednisone

- Previous docetaxel.

- ECOG 0–2.

- PSA or radiographic progression.

OS: 15.8 vs. 11.2 mo.

(p < 0.0001, HR: 0.74, 95%

CI: 0.64–0.86; p < 0.0001).

FU: 20.2 mo.

rPFS: no change

COU-AA-301

2011 [1212]




OS: 14.8 vs. 10.9 mo.

(p < 0.001 HR: 0.65; 95%

CI: 0.54–0.77).

FU: 12.8 mo.

rPFS: 5.6 vs. 3.6 mo.

Radium-223

ALSYMPCA

2013 [1256]

radium-223

placebo

- Previous or no previous docetaxel.

- ECOG 0–2.

- Two or more symptomatic bone metastases.

- No visceral metastases.

OS: 14.9 vs. 11.3 mo.

(p = 0.002, HR: 0.61; 95%

CI: 0.46–0.81).

All secondary endpoints show a benefit over best SOC.

CABAZITAXEL

TROPIC

2013 [1208]

cabazitaxel + prednisone

mitoxantrone + prednisone

- Previous docetaxel.

- ECOG 0–2.

OS: 318/378 vs. 346/377 events

(OR: 2.11; 95% CI: 1.33–3.33).

FU: 25.5 months OS > 2 yr 27% vs. 16% PFS: -

TROPIC

2010 [1204]




OS: 15.1 vs. 12.7 mo.

(p < 0.0001, HR: 0.70;

95% CI: 0.59–0.83). FU: 12.8 mo.

PFS: 2.8 vs. 1.4 mo.

(p < 0.0001, HR: 0.74,

95% CI: 0.64-0.86)

CARD

2019 [1164]

cabazitaxel

(25 mg/m2 Q3W)

+ prednisone

+ G-CSF

ARTA: abiraterone + prednisone

OR

enzalutamide

- Previous docetaxel.

- Progression ≤ 12 mo. on prior alternative ARPI (either before or after docetaxel)

OS: 13.6 vs. 11.0 mo.

(p = 0.008, HR: 0.64, 95%

CI: 0.46–0.89).

rPFS 8.0 vs. 3.7 mo.

(p < 0.001, HR: 0.54,

95% CI: 0.40–0.73).

FU: 9.2 mo.

ENZALUTAMIDE

AFFIRM

2012 [1214]

enzalutamide

placebo

- Previous docetaxel.

- ECOG 0–2.

OS: 18.4 vs. 13.6 mo.

(p < 0.001, HR: 0.63; 95%

CI: 0.53–0.75).

FU: 14.4 mo.

rPFS: 8.3 vs. 2.9 mo.

(HR: 0.40; 95% CI: 0.35–0.47, p < 0.0001).

PARP inhibitor

PROfound

2020 [248,1162,1224]

olaparib

abiraterone + prednisolone or enzalutamide;

cross-over allowed at progression

- Previous ARPI,

- alterations in HRR genes

rPFS: 7.39 vs. 3.55 mo.

(p < 0.0001, HR: 0.34; 95%
CI: 0.25–0.47), conf. ORR
33.3% vs. 2.3% (OR 20.86, 95% CI: 4.18–379.18).

OS: 19.1 mo vs. 14.7 mo.

(in pts with BRCA1/2, ATM alterations)

(p = 0.0175; HR 0.69, 95%

CI: 0.5–0.97).

TRITON-3 [1219]

rucaparib

(600 mg BID)

docetaxel or abiraterone acetate or enzalutamide

- EOCG 0-1

- Previous one ARPI

- BRCA 1/ 2 or ATM alteration

rPFS: ITT

10.2 mo vs. 6.4 mo,

HR 0.61; 95% CI, 0.47 to 0.80;

(P<0.001 for both comparisons)

Radioligand therapy

VISION

2021 [1237]

177Lu-PSMA-617

SOC

SOC alone

- Previous at least 1 ARPI and one or two taxane regimens;

- Mandatory: PSMA-positive gallium-68 (68Ga)–labelled PSMA-PET scan

Imaging-based PFS:

8.7 vs. 3.4 mo.

(p < 0.001; HR 0.40; 99.2%

CI: 0.29–0.57)

OS: 15.3 vs. 11.3 mo.

(p < 0.001; HR 0.62; 95%

CI: 0.5–0.74)

TheraP

2021 [1235,1236]

177Lu-PSMA-617

(8.5 GBq

i.v.q 6-weekly, decreasing

0.5 GBq/cycle; up to 6 cycles)

cabazitaxel

(20 mg/m2

i.v.q 3-weekly, up to 10 cycles)

- Post docetaxel,

- Suitable for cabaziaxel

First endpoint PSA reduction of > 50%:

66 vs. 37 PSA responses; 66% vs. 37% by ITT; difference 29%

(95% CI: 16–42; p < 0.0001; and 66% vs. 44% by treatment received; difference 23% [9–37]; p = 0.0016).

Secondary endpoint OS:

19.1 vs. 19.6 mo (177Lu-PSMA vs. cabazitaxel).

HR: 0.97, 95% CI: 0.7-1.4

(p=0.99)

*Only studies reporting survival outcomes as primary endpoints have been included.
ARPI = androgen receptor pathway inhibitor; CI = confidence interval; ECOG = Eastern Cooperative Oncology Group; FU = follow-up; GBq = gigabecquerel; HR = hazard ratio; Lu = lutetium; mo = months OS = overall survival; OR = odds ratio; ORR = objective response rate; PSA = prostate-specific antigen; PSMA = prostatespecific membrane antigen; (r)PFS = (radiographic) progression-free survival; SOC = standard of care; yr = year; HRR= homologous recombination repair.

6.7.8.5. Platinum chemotherapy

Cisplatin or carboplatin as monotherapy or combinations have shown limited activity in unselected patients in the pre-docetaxel era [1257]. The combination of cabazitaxel and carboplatin was evaluated in pre-treated mCRPC patients in a randomised phase I/II trial. The combination improved the median PFS from 4.5 months (95% CI: 3.5–5.7) to 7.3 months (95% CI: 5.5–8.2; HR: 0.69, 95% CI: 0.50–0.95, p = 0.018) and the combination was well tolerated [1258]. On a histopathological and molecular level, there is preliminary evidence that platinum adds efficacy in patients with aggressive variant PCa molecular signatures including TP53, RB1, and PTEN [1259].

Patients with mCRPC and alterations in DDR genes are more sensitive to platinum chemotherapy than unselected patients [1260], also after progression on PARP inhibitors. Interestingly, in contemporary retrospective series, unselected patients as well as patients without DDR gene alterations also showed a 50% PSA decline when treated with platinum in up to 36% of patients. In view of the excellent tolerability of e.g., carboplatin monotherapy, platinum could be offered to patients with far advanced mCRPC harbouring DDR gene aberrations after having progressed on standard treatment options. Prospective controlled trials are ongoing.

6.7.9. Monitoring of treatment

Baseline examinations should include a medical history, clinical examination as well as baseline blood tests (PSA, total testosterone level, full blood count, renal function, baseline liver function tests, alkaline phosphatase), bone scan and CT of chest, abdomen and pelvis [1261,1262]. The use of choline or PSMA PET/CT scans for progressing CRPC is unclear and most likely not as beneficial as for patients with BCR or hormone-naive disease. Flares, PSMA upregulation and discordant results compared with PSA response or progression on ARPI have been described [1263]. Prostate-specific antigen alone is not reliable enough [1264] for monitoring disease activity in advanced CRPC since visceral metastases may develop in men without rising PSA [1265]. Instead, the PCWG2 recommends a combination of bone scintigraphy and CT scans, PSA measurements and clinical benefit in assessing men with CRPC [1266]. A majority of experts at the 2015 Advanced Prostate Cancer Consensus Conference (APCCC) suggested regular review and repeating blood profile every two to three months with bone scintigraphy and CT scans at least every six months, even in the absence of a clinical indication [1261]. This reflects that the agents with a proven OS benefit all have potential toxicity and considerable cost and patients with no objective benefit should have their treatment modified. The APCCC participants stressed that such treatments should not be stopped for PSA progression alone. Instead, at least two of the three criteria (PSA progression, radiographic progression and clinical deterioration) should be fulfilled to stop treatment. For trial purposes, the updated PCWG3 put more weight on the importance of documenting progression in existing lesions and introduced the concept of no longer ‘clinically benefiting‘ to underscore the distinction between first evidence of progression and the clinical need to terminate or change treatment [1267]. These recommendations also seem valid for clinical practice outside trials.

6.7.10. When to change treatment

The timing of mCRPC treatment change remains a matter of debate in mCRPC although it is clearly advisable to start or change treatment immediately in men with symptomatic progressing metastatic disease. Preferably, any treatment change should precede development of de novo symptoms or worsening of existing symptoms. Although, the number of effective treatments is increasing, head-to-head comparisons are still rare, as are prospective data assessing the sequencing of available agents. Therefore it is not clear how to select the most appropriate ‘second-line‘ treatment, in particular in patients without HRR alterations or other biomarkers. A positive example, however, is the CARD trial which clearly established cabazitaxel as the better third-line treatment in docetaxel pre-treated patients after one ARPI compared to the use of a second ARPI [1164].

The ECOG PS has been used to stratify patients. Generally men with a PS of 0–1 are likely to tolerate treatments and those with a PS of > 2 are less likely to benefit. However, it is important that treatment decisions are individualised, in particular when symptoms related to disease progression are impacting on PS. In such cases, a trial of active life-prolonging agents to establish if a given treatment will improve the PS may be appropriate. Sequencing of treatment is discussed in the summary papers published following the 2019 and 2022 APCCC Conferences [1268,1269].

6.7.11. Symptomatic management in metastatic castration-resistant prostate cancer

Castration-resistant PCa is usually a debilitating disease often affecting the elderly male. A multidisciplinary approach is required with input from urologists, medical oncologists, radiation oncologists, nurses, psychologists and social workers [1268,1270]. Critical issues of palliation must be addressed when considering additional systemic treatment, including management of pain, constipation, anorexia, nausea, fatigue and depression.

6.7.11.1. Common complications due to bone metastases

Most patients with CRPC have painful bone metastases. External beam RT is highly effective, even as a single fraction [1271,1272]. A single infusion of a third generation bisphosphonate could be considered when RT is not available [1273]. Common complications due to bone metastases include vertebral collapse or deformity, pathological fractures and spinal cord compression. Cementation can be an effective treatment for painful spinal fracture whatever its origin, clearly improving both pain and QoL [1274]. It is important to offer standard palliative surgery, which can be effective for managing osteoblastic metastases [1275,1276]. Impending spinal cord compression is an emergency. It must be recognised early and patients should be educated to recognise the warning signs. Once suspected, high-dose corticosteroids must be given and MRI performed as soon as possible. A systematic neurosurgery or orthopaedic surgeon consultation should be planned to discuss a possible decompression, followed by EBRT [1277]. Otherwise, EBRT with, or without, systemic therapy, is the treatment of choice.

6.7.11.2. Preventing skeletal-related events
6.7.11.2.1. Bisphosphonates

Zoledronic acid has been evaluated in mCRPC to reduce skeletal-related events (SRE). This study was conducted when no active anti-cancer treatments, but for docetaxel, were available. Six hundred and forty three patients who had CRPC with bone metastases were randomised to receive zoledronic acid, 4 or 8 mg every three weeks for fiveteen consecutive months, or placebo [1278]. The 8 mg dose was poorly tolerated and reduced to 4 mg but did not show a significant benefit. However, at fiveteen and 24 months of follow-up, patients treated with 4 mg zoledronic acid had fewer SREs compared to the placebo group (44 vs. 33%, p = 0.021) and in particular fewer pathological fractures (13.1 vs. 22.1%, p = 0.015). Furthermore, the time to first SRE was longer in the zoledronic acid group. No survival benefit has been seen in any prospective trial with bisphosphonates.

6.7.11.2.2. RANK ligand inhibitors

Denosumab is a fully human monoclonal antibody directed against RANKL (receptor activator of nuclear factor κ-B ligand), a key mediator of osteoclast formation, function, and survival. In M0 CRPC, denosumab has been associated with increased bone-MFS compared to placebo (median benefit: 4.2 months, HR: 0.85, p = 0.028) [1271]. This benefit did not translate into a survival difference (43.9 compared to 44.8 months, respectively) and neither the FDA or the EMA have approved denosumab for this indication [1279].

The efficacy and safety of denosumab (n = 950) compared with zoledronic acid (n = 951) in patients with mCRPC was assessed in a phase III trial. Denosumab was superior to zoledronic acid in delaying or preventing SREs as shown by time to first on-study SRE (pathological fracture, radiation or surgery to bone, or spinal cord compression) of 20.7 vs. 17.1 months, respectively (HR: 0.82, p = 0.008). Both urinary N-telopeptide and bone-specific alkaline phosphatase were significantly suppressed in the denosumab arm compared with the zoledronic acid arm (p < 0.0001 for both). However, these findings were not associated with any survival benefit and in a post-hoc re-evaluation of endpoints, denosumab showed identical results when comparing SREs and symptomatic skeletal events [1280].

The potential toxicity (e.g., osteonecrosis of the jaw, hypocalcaemia) of these drugs must always be kept in mind (5–8.2% in M0 CRPC and mCRPC, respectively) [1281,1282]. Patients should have a dental examination before starting therapy as the risk of jaw necrosis is increased by several risk factors including a history of trauma, dental surgery or dental infection [1283]. Also, the risk for osteonecrosis of the jaw increased numerically with the duration of use in a pivotal trial [1284] (one year vs. two years with denosumab), but this was not statistically significant when compared to zoledronic acid [1279]. According to the EMA, hypocalcaemia is a concern in patients treated with denosumab and zoledronic acid. Hypocalcaemia must be avoided by adequate intake of calcium and vitamin D before initiating therapy [1285]. Hypocalcaemia should be identified and prevented during treatment with bone protective agents (risk of severe hypocalcaemia is 8% and 5% for denosumab and zoledronic acid, respectively) [1282]. Serum calcium should be measured in patients starting therapy and monitored during treatment, especially during the first weeks and in patients with risk factors for hypocalcaemia or on other medication affecting serum calcium. Daily calcium (> 500 mg) and vitamin D (> 400 IU equivalent) are recommended in all patients, unless in case of hypercalcaemia [1282,1286,1287].

6.7.12. Summary of evidence and guidelines for life-prolonging treatments of castrate-resistant disease

Summary of evidence

LE

Treatment for mCRPC will be influenced by which treatments patients have already been exposed to.

4

Recommendations

Strength rating

Ensure that testosterone levels are confirmed to be < 50 ng/dL before diagnosing castrate-resistant PCa (CRPC).

Strong

Counsel, manage and treat patients with metastatic CRPC (mCRPC) in a multidisciplinary team.

Strong

Treat patients with mCRPC with life-prolonging agents.

Strong

Offer mCRPC patients somatic and/or germline molecular testing as well as testing for mismatch repair deficiencies or microsatellite instability.

Strong

6.7.13. Guidelines for systematic treatments of castrate-resistant disease

Summary statement for mCRPC first line combination therapy:

The combination of ARPI plus PARP inhibitors showed a significant rPFS benefit in RCTs for unselected patients. However, this benefit is mainly driven by HRR- and even more pronounced by BRCA 1/2- altered patients. So far, no clear OS benefit was seen, and the side effects of PARP inhibitors add substantial toxicity to ARPI monotherapy. Therefore, no recommendation is given for patients without HRR or BRCA 1/2 -mutations and the data will be re-evaluated after longer follow-up.

Recommendations

Strength rating

Base the choice of treatment on the performance status (PS), symptoms, comorbidities, location and extent of disease, genomic profile, patient preference, and on previous treatment for hormone-sensitive metastatic PCa (mHSPC) (alphabetical order: abiraterone, cabazitaxel, docetaxel, enzalutamide, 177lutetium-PSMA-617-radioligand therapy, radium-223, sipuleucel-T, and for patients with DNA homologous recombination repair (HRR) alterations olaparib, olaparib/abiraterone, niraparib/abiraterone, rucaparib, talazoparib/enzalutamide).

Strong

Avoid sequencing of androgen receptor targeted agents.

Weak

Offer chemotherapy to patients previously treated with abiraterone or enzalutamide.

Strong

Offer patients with metastatic castrate-resistant PCa (mCRPC) who are candidates for cytotoxic therapy and are chemotherapy naiive docetaxel with 75 mg/m2 every three weeks.

Strong

Offer patients previously untreated for mCRPC and harbouring an HRR or BRCA mutation abiraterone in combination with olaparib if the patient is fit for both agents.

Strong

Offer patients previously untreated for mCRPC and harbouring a BRCA mutation abiraterone in combination with niraparib if the patient is fit for both agents.

Strong

Offer patients previously untreated for mCRPC and harbouring an HRR-mutation enzalutamide in combination with talazoparib if the patient is fit for both agents.

Strong

Offer poly(ADP-ribose) polymerase (PARP) inhibitors to pre-treated mCRPC patients with relevant DNA repair gene mutations.

Strong

Offer patients with mCRPC and progression following docetaxel chemotherapy further life-prolonging treatment options, which include abiraterone, cabazitaxel, enzalutamide, radium-223 and olaparib in case of DNA homologous recombination repair (HRR) alterations.

Strong

Base further treatment decisions of mCRPC on PS, previous treatments, symptoms, comorbidities, genomic profile, extent of disease and patient preference.

Strong

Offer abiraterone or enzalutamide to patients previously treated with one or two lines of chemotherapy.

Strong

Offer cabazitaxel to patients previously treated with docetaxel.

Strong

Offer cabazitaxel to patients previously treated with docetaxel and progressing within twelve months of treatment with abiraterone or enzalutamide.

Strong

Offer 177Lu-PSMA-617 to pre-treated mCRPC patients with one or more metastatic lesions, highly expressing PSMA (exceeding the uptake in the liver) on the diagnostic radiolabelled PSMA PET/CT scan.

Strong

6.7.14. Guideline for non-metastatic castrate-resistant disease

Recommendation

Strength rating

Offer apalutamide, darolutamide or enzalutamide to patients with M0 CRPC and a high risk of developing metastasis (PSA-DT < ten months) to prolong time to metastases and overall survival.

Strong

6.7.15. Guidelines for supportive care of castrate-resistant disease

These recommendations are in addition to appropriate systemic therapy.

Recommendations

Strength rating

Offer bone protective agents to patients with metastatic prostate cancer and skeletal metastases to prevent osseous complications.

Strong

Monitor serum calcium and offer calcium and vitamin D supplementation when prescribing either denosumab or bisphosphonates.

Strong

Treat painful bone metastases early on with palliative measures such as intensity-modulated radiation therapy/volumetric arc radiation therapy plus image-guided radiation therapy and adequate use of analgesics.

Strong

In patients with spinal cord compression start immediate high-dose corticosteroids and assess for spinal surgery followed by irradiation. Offer radiation therapy alone if surgery is not appropriate.

Strong

Figure 6.4: Treatment non-metastasized (M0) – asymptomatic disease

* Rule of thumb: Life expectancy ten years.
** Recommendation based on clinical staging using digital rectal examination, not imaging.
*** Recommendation based on staging using combination of bone scan and CT.
**** See text, dependent on GG and (biopsy) volume.

1EBRT: IMRT/VMAT + IGRT of the prostate.

 (light green) = weak recommendation.

ADT = androgen deprivation therapy; EBRT =external beam radiotherapy; ECE = extracapsular extension; ePLND = extended pelvic lymph node dissection; GG = grade group; HDR = high-dose rate; IDC = intraducal carcinoma; IGRT = image-guided radiotherapy; IMRT = intensity-modulated radiotherapy; LDR = low-dose rate; VMAT = volumetric modulated arc therapy.

Figure 6.5: Treatment of metastasized (M1*) – disease, M+HSPC

* Based on staging using combination of bone scan and CT.
** Alphabetical order.
***not for low volume, metachronous disease.
1EBRT: IMRT/VMAT + IGRT of the prostate (equivalent of up to 72 Gy in 2 Gy fractions).

 (light green) = weak recommendation.

EBRT = external beam radiotherapy; IGRT = image-guided radiotherapy; IMRT = intensity-modulated radiotherapy.

#Note: Please be aware that the various options in the following flowcharts present a generalised approach only, and cannot take the management of individual patients into account, nor the availability of resources.