Prostaatcarcinoom

Initiatief: NVU Aantal modules: 89

Eerstelijnsbehandeling CRPC

Uitgangsvraag

Wat is de aanbevolen eerstelijnsbehandeling bij patiënten met een hormoonresistent prostaatcarcinoom (CRPC)?

 

De uitgangsvraag omvat de volgende deelvragen:

  1. Wat is de plaats van ‘androgen receptor signaling inhibitors’ (ARSI) als aanvullende behandeloptie bij ADT gebruik?
    1. Bij patiënten met niet-gemetastaseerde CRPC
    2. Bij patiënten met gemetastaseerde CRPC
  2. Wat is de plaats van chemotherapie als aanvullende behandeloptie bij ADT gebruik?
  3. Wat is de plaats van radioligand therapie als aanvullende behandeloptie bij ADT gebruik?

Aanbeveling

Zet de testosterononderdrukking voort bij patiënten die voldoen aan de criteria voor nmCRPC met een oplopend PSA. Geef geen aanvullende behandeling.

Overweeg om een PSMA PET scan te maken om metastasen aan te tonen.

Indien er hiermee beperkte metastasen worden aangetoond, overweeg een verder expectatief beleid.

Start een vervolgbehandeling wanneer metastasen aangetoond zijn middels CT- en skeletscan.

 

Geef bij een progressief mCRPC bij een patiënt die ‘chemo-fit’ is,  docetaxel 75 mg/m2 plus prednison 5 mg bid iedere 3 weken maximaal 10 kuren.

Overweeg als behandeloptie voor behandeling van asymptomatisch of licht symptomatisch mCRPC -  abiraterone 1000 mg plus prednison 5 mg bid of enzalutamide 160 mg.

 

Geef bij patiënten met een progressief mCRPC die niet ‘chemo-fit’ zijn of om andere redenen niet aan chemotherapie toekomen: abiraterone 1000 mg plus prednison 5 mg bid

of enzalutamide 160 mg. Overweeg radium-223 (6 injecties gegeven met intervallen van 4 weken) bij beperkte lymfekliermetastasering en afwezigheid van viscerale metastasen en contra-indicaties voor ARSI en chemotherapie.

 

Bespreek de patiënt in een MDO.

Bespreek de keuze van de therapie en de beste timing van het starten van de therapie met de patiënt.

Overwegingen

Indien er sprake is van een biochemische of radiologische progressie onder testosteronverlagende therapie, dan dient aangetoond te worden of er sprake is van CRPC naar de geldende criteria. Een belangrijk discussiepunt is de beste timing van de start van een volgende palliatieve behandeling en de keuze van de meest aangewezen therapie. De beschikbaarheid van gerandomiseerde studies die een keuze voor een volgende behandeling onderbouwen zijn zeer beperkt. Bespreking binnen het multidisciplinaire team is nu een vereiste. Een aanvullende behandeling met een klassieke androgeen receptor blokker zoals bicalutamide wordt als obsoleet beschouwd.

 

nmCRPC is een nieuwe entiteit en wordt gedefinieerd als een oplopend serum PSA bij een patiënt met een adequaat onderdrukt serum testosteron, terwijl er middels een CT van thorax en abdomen en skeletscan, geen metastasen aantoonbaar zijn. Er zijn drie redenen waarom deze nieuwe indicatie niet of nauwelijks van belang is voor de Nederlandse praktijk. Ten eerste zijn Nederlandse urologen en radiotherapeuten behoudend met het starten van testosteron-deprivatie en zetten deze veelal pas in op het moment dat er metastasen zijn aangetoond. Ten tweede is in Nederland de PSMA-PET scan breed beschikbaar. Een studie heeft aangetoond dat meer dan 90% van de patiënten die voldoen aan de criteria voor nmCRPC, op PSMA PET scan wel aantoonbare metastasen hebben en 55% afstand metastasen (M1) (Fendler, 2019). Hierdoor komt de entiteit nmCRPC nauwelijks voor in Nederland. Een derde punt van reserve is dat de studies metastasen-vrije overleving als primair eindpunt hadden. Hierdoor konden deze middelen niet volgens PASKWIL criteria worden beoordeeld.

 

Bij patiënten met een mCRPC kan er feitelijk voor drie verschillende klassen behandelingen   gekozen worden, namelijk een behandeling met chemotherapie (docetaxel, cabazitaxel), een behandeling met hormonale middelen (abiraterone of enzalutamide), of een behandeling met radionucliden (radium-223). Al deze behandelingen worden vergoed, zijn beschikbaar en er is een wisselende hoeveelheid ervaring mee.

Abiraterone en enzalutamide zijn geregistreerd voor behandeling van patiënten met een asymptomatisch of gering symptomatisch mCRPC. Er zijn geen gegevens wat de optimale keuze is tussen deze twee middelen. Ook radium-223 kan gegeven worden in eerstelijns behandeling, aangezien latere behandeling met chemotherapie nog steeds mogelijk is, omdat invloed op beenmergfunctie minimaal is. Echter, in 2018 heeft PRAC, de geneesmiddelen veiligheidscommissie van EMA, geadviseerd dat radium-223 gebruik beperkt moet worden tot patiënten die tenminste twee eerdere behandelingen hebben gehad en patiënten voor wie geen andere behandelopties meer zijn. Dit naar aanleiding van de ERA223 studie waarin een hoger percentage fracturen en sterfte werd gevonden in de met radium-223 en abiraterone behandelde studiearm.

Verschillen in bijwerkingen, geneesmiddeleninteracties en co morbiditeit kunnen een rol spelen bij de keuze tussen de beschikbare middelen. Het lijkt voor de hand te liggen om bij een fitte patiënt en een uitgebreide ziekte met een agressief beloop, primair te kiezen voor chemotherapie. Daarbij moet meegewogen worden dat het niet zeker is of in een latere fase van het ziekte beloop patiënten nog fit genoeg zijn voor chemotherapie.

De voorkeur van de patiënt moet bij het vaststellen van het behandelbeleid zeker gewicht in de schaal leggen. Het is vaak behulpzaam om met een patiënt niet alleen de verschillende behandelingsmethoden te bespreken, maar ook uit te leggen dat de beste resultaten bereikt worden als een patiënt uiteindelijk met middelen uit alle drie deze methoden behandeld wordt. Een eerste keuze voor een bepaalde behandeling sluit latere behandeling met andere middelen niet uit en er zijn ook geen harde aanwijzingen dat een eerste keuze automatisch latere keuzen onmogelijk maakt.

Patiënten met bij diagnose gemetastaseerde ziekte worden veelal in het vroege, hormoon gevoelige stadium met chemotherapie of een ARSI behandeld, direct na het starten van een primaire hormonale behandeling (androgeen deprivatie therapie). Er zijn geen gegevens over de optimale behandelkeuze indien er na deze vroege behandeling progressie optreedt (er mCRPC wordt vastgesteld) en er een indicatie bestaat voor een vervolgbehandeling. Het lijkt het beste deze vragen bij een langdurig therapievrij interval te benaderen alsof er een eerstelijns keuze gemaakt moet worden, terwijl er bij progressie onder therapie of een heel kort therapievrij interval een “tweedelijns keuze” gemaakt moet worden. In het bepalen van de sequentie van de behandelingen is te adviseren de verschillende klassen middelen te alterneren. Dit geldt in het bijzonder voor de ARSI’s abiraterone en enzalutamide. Deze middelen kennen een belangrijke kruisresistentie en de kans op een repons op een tweede ARSI, direct na progressie op een eerste ARSI is minimaal (Khalaf, 2019).

 

Voor- en nadelen van de interventie en de kwaliteit van het bewijs

Op het moment van schrijven van de richtlijn, zijn er vier eerste lijn behandelopties voor patiënten met progressief mCRPC en een indicatie en mogelijkheid voor een behandeling (docetaxel, abiraterone, enzalutamide en radium-223). Alle vier behandelmogelijkheden hebben een vergelijkbaar overlevingsvoordeel laten zien als eerste lijn behandeling. Daarnaast hebben meerdere secundaire eindpunten in de studies, waaronder kwaliteit van leven, een gunstig effect laten zien van deze middelen. De bewijskracht van de studies naar een meerwaarde van de middelen is sterk. Er zitten echter belangrijke verschillen in de kwaliteit en bewijskracht van de studies wat betreft secondaire eindpunten. Daarmee zijn docetaxel en de twee ARSI’s te overwegen als eerste lijn mCRPC behandeling. Ook voor radium-223 is aangetoond dat deze een waarde heeft als eerste lijn behandeling, echter dit op basis van een subgroep analyse. Bovendien heeft de EMA geadviseerd deze behandeling niet als eerste lijn mCRPC therapie toe te passen. Een algemeen voorbehoud bij de conclusies van de studies is dat deze inmiddels ouder zijn en patiënten in cross-over niet met een van de nieuwere therapieën zijn behandeld. Daarnaast zijn er weinig gegevens waarop de voorkeur voor een eerste lijn behandeloptie, boven een ander, gebaseerd kan zijn.

 

Waarden en voorkeuren van patiënten

De behandeling van mCRPC is palliatief. De genoemde middelen hebben alle bijwerkingen en de overlevingswinst die met de middelen mediaan wordt bereikt, is beperkt. Daarmee is kwaliteit van leven van de patiënt belangrijk om in de beslissingen mee te nemen. Een belangrijk aspect hiervan is dat de voorkeur van de patiënt, na verkrijgen van complete informatie, zwaar weegt in de beslissing. Deze dient te worden geïncorporeerd in de medische overwegingen wat betreft of, en met welk middel, patiënt als eerste lijn mCRPC therapie zal worden behandeld. Aangezien de overlevingswinsten, maar ook de voordelen op secundaire eindpunten van de verschillende middelen elkaar niet veel ontlopen, tellen intensiviteit van de behandeling en te verwachten bijwerkingen zwaar in de keuze voor een eerste lijn behandeling. Zo kan een relatief jonge patiënt, met weinig comorbiditeit, docetaxel als eerste lijn behandeling verkiezen en daarmee alle vervolgopties openhouden, terwijl een oudere patiënt met veel comorbiditeit, afziet van chemotherapie en een ARSI prefereert.

 

Kosten (middelenbeslag)

Er zijn grote verschillen in de prijs tussen de eerste lijn mCRPC behandelopties. Op het moment van schrijven van deze richtlijn, zijn docetaxel en abiraterone behandeling veruit het goedkoopste. Het is een maatschappelijke discussie of deze verschillen in kosten de veelal beperkte overlevingswinst rechtvaardigen. Het is ook een plicht voor de behandelaar, om deze middelen alleen voor te schrijven binnen de indicatie en kritisch te blijven op de werkzaamheid en de behandeling niet onnodig lang voort te zetten. Per september 2022 is het patent op abiraterone vervallen en zijn er meerdere aanbieders van een generiek product. Daarmee is abiraterone op dit moment de goedkoopste behandeloptie. Daarom, is het vanuit het perspectief van  maatschappelijke verantwoordelijkheid wenselijk om te kiezen voor abiraterone wanneer een ARSI geïndiceerd is, als er medisch-inhoudelijk geen argumenten zijn om voor enzalutamide te kiezen. 

 

Aanvaardbaarheid, haalbaarheid en implementatie

De genoemde middelen vallen onder vergoede zorg en zijn voorbehouden aan behandelaars die geschoold zijn om te werken met deze middelen en patiënten categorie. Er zijn geen bijzondere omstandigheden nodig voor behandeling met docetaxel en ARSI’s en worden door alle oncologische centra aangeboden in Nederland. Daarmee zijn deze behandelingen breed beschikbaar. Uitzondering hierop is radium-223, welke alleen kan worden toegediend door centra met een afdeling nucleaire geneeskunde met de vereiste vergunningen.

 

Rationale van de aanbeveling: weging van argumenten voor en tegen de diagnostische procedure

 

  1. Wat is de plaats van ‘androgen receptor signaling inhibitors’ (ARSI) als aanvullende behandeloptie bij ADT?
    1. Bij patiënten met niet-gemetastaseerde CRPC

nmCRPC is een in Nederland weinig voorkomende entiteit. Daarnaast hadden de studies niet overall survival als primair eindpunt. Daarmee is het advies om patiënten te vervolgen tot aantoonbare metastasen (CT, skelet scan) en dan vervolgbehandeling te overwegen.

 

    1. Bij patiënten met gemetastaseerde CRPC

Voor patiënten met gemetastaseerd CRPC en een indicatie voor een vervolgbehandeling is een ARSI een van de  behandelopties. De aangetoonde overlevingswinst van deze middelen is vergelijkbaar met die van docetaxel. De keuze zal veelal op een ARSI vallen, wanneer een patiënt in het hormoongevoelige stadium met docetaxel is behandeld. Abiraterone en enzalutamide hebben een vergelijkbare overlevingswinst laten zien. Keuze voor abiraterone of enzalutamide kan worden gebaseerd op het bijwerkingen profiel of de combinatie van abiraterone met prednison wenselijk is. Daarnaast zijn de lage kosten van abiraterone ook een belangrijk argument om voor dit middel te kiezen.

 

  1. Wat is de plaats van chemotherapie als aanvullende behandeloptie bij ADT?

Docetaxel chemotherapie heeft als eerste lijn behandeling een gelijke overlevingswinst laten zien als abiraterone en enzalutamide. Docetaxel is een intensievere behandeling dan ARSI, wat betreft ziekenhuisbezoeken en bijwerkingen. Toch zal de keuze op docetaxel vallen, wanneer een patiënt eerder met een ARSI is behandeld in het hormoongevoelige stadium. Daarnaast wordt gekozen voor docetaxel wanneer de ziekte een snel en agressief beloop heeft. Er is echter geen bewijs dat docetaxel in deze patiënten effectiever is of sneller werkt dan de ARSI’s.

 

  1. Wat is de plaats van radioligand therapie als aanvullende behandeloptie bij ADT?

Effectiviteit van radium-223 als eerste lijn behandeling van mCRPC is aangetoond in een subgroep analyse. Hiermee is de bewijslast van effectiviteit als eerste lijn behandeling minder dan van de andere opties. Daarnaast zijn er door de geneesmiddelenveiligheidscommissie van de EMA, adviezen uitgegaan voor plaatsbepaling van dit middel binnen de behandelopties voor mCRPC. Radium-223 wordt geadviseerd in te zetten als derde lijn behandeling en in de eerste lijn alleen  voor patiënten voor wie geen verdere behandelopties meer zijn.

Onderbouwing

Hormoonresistent prostaatkanker is het stadium van de ziekte waarin testosteronsynthese onderdrukking in de testikels, geen remming van ziekte meer bewerkstelligt. Sedert de introductie van docetaxel in 2004, zijn er meerdere behandelmogelijkheden voor CRPC bijgekomen waarvan een overlevingswinst is aangetoond. De overlevingswinst van deze middelen is echter beperkt tot 3-4 maanden. Een belangrijke vraag blijft de optimale sequentie van deze behandelingen. Gerandomiseerde studies beantwoorden deze vraag veelal niet. Deze module behandelt de keuze voor een eerste lijn behandeling.

Sub question 1a ARSI nmCRPC

 

1. Survival

High GRADE

ARSI, combined with ADT, versus ADT alone or placebo results in an increased survival (metastasis free survival, progression free survival, overall survival, and long term survival) in patients with non metastatic castration resistant prostate cancer (nmCRPC).

 

Sources: Fizazi, 2019; Hussain, 2018; Small, 2019; Smith 2018; Smith, 2019 and Sternberg, 2020

 

2. Toxicity

High GRADE

ARSI, combined with ADT, versus ADT alone or placebo results in comparable adverse events any grade in patients with nmCRPC.

ARSI, combined with ADT, versus ADT alone or placebo results in higher adverse events grade 3 in patients with nmCRPC.

 

Sources: Sopeña Sutil, 2021; Smith, 2021; Sternberg, 2021

 

3. Costs

No GRADE

The outcome costs was not assessed by the included studies.

 

4. Quality of life

High GRADE

ARSI, combined with ADT, versus ADT alone or placebo results in a comparable quality of life at follow up in patients with nmCRPC.

 

Sources: Fizazi, 2019; Hussain, 2018 and Smith, 2018    

 

Sub question 1b ARSI mCRPC

 

1. Survival

High GRADE

ARSI versus placebo results in an increased survival (overall survival, progression free survival and long term survival) in patients with metastatic castration resistant prostate cancer (mCRPC).

 

Sources: Beer, 2014; Morris, 2015 and Ryan, 2015

 

2. Toxicity

High GRADE

At the end of the study period (ca. 2 year follow up), ARSI versus placebo results in comparable adverse events any grade and adverse events grade 3 in patients with mCRPC.

 

At long term (ca. 4 year follow up), ARSI results in comparable adverse events any grade but higher adverse events grade 3 in patients with mCRPC.

 

Sources: Beer, 2014; Ryan, 2013, Ryan, 2015

 

3. Costs

No GRADE

The outcome costs was not assessed by the included studies in patients with mCRPC.

 

4. Quality of life

High GRADE

ARSI versus placebo results in an improved quality of life in patients with mCRPC.

 

Sources: Beer, 2014 and Ryan, 2013

 

Sub question 3. Radioligand therapy

 

1. Survival

Moderate GRADE

Radioligand therapy versus placebo probably results in an increased survival (overall survival, progression free survival and death related to prostate cancer) in patients with metastatic castration resistant prostate cancer (mCRPC).

 

Sources: Parker, 2013 and Parker, 2018

 

2. Toxicity

High GRADE

Radioligand therapy versus placebo results in comparable adverse events of any grade in patients with mCRPC.

 

Sources: Parker, 2013 and Parker, 2018

 

3. Costs

No GRADE

The outcome costs was not assessed by the included studies.

 

4. Quality of life

Moderate GRADE

Radioligand therapy versus placebo probably results in a not clinically relevant change in quality of life at follow up compared to baseline in patients with mCRPC.

 

Sources: Parker, 2013 and Parker, 2018

1. ARSI

 

Sub question 1a ARSI nmCRPC

 

Description of studies


Three trials were included for sub question 1a: the ARAMIS trial with one publication (Fizazi, 2019), the PROSPER trial with 2 publications (Hussain, 2018 and Sternberg, 2020) and the SPARTAN trial with 3 publications (Small, 2019; Smith, 2018 and Smith, 2021). An overview of study characteristics can be found in Table 1.

 

Table 1. Overview study characteristics ARAMIS, PROSPER and SPARTAN

 

The ARAMIS trial

The ARAMIS trial (Fizazi, 2019) assessed the effect of darolutamide versus placebo in patients with non-metastatic castration-resistant prostate cancer (nmCRPC). The

trial was conducted in 409 centers in 36 countries worldwide. The primary endpoint was metastasis free survival, whereas secondary outcomes were overall survival, time to pain progression, time to chemotherapy and time to a symptomatic skeletal event. The median follow up time was 18 months. Patients were randomly assigned in a 2:1 ratio to receive the intervention (twice daily 600mg darolutamide) or matched placebo and ADT. Patients discontinued therapy because of protocol-defined progression, adverse events or withdrawal of consent. Patients that took a prohibited therapy (described in protocol) had to leave the trial but were followed for survival. A total of 1509 patients (955 darolutamide and 554 placebo) were assessed for the intention-to-treat analysis. Groups were comparable at baseline.

 

The PROSPER trial

The PROSPER trial (Hussain, 2018 and Sternberg, 2020) investigated the effect of enzalutamide versus placebo in patients with nmCRPC. The trial was conducted in more than 300 sites in 32 countries worldwide. The primary endpoint was metastasis free survival, whereas secondary outcomes were overall survival, time to PSA progression, PSA response rate, time to first use of a subsequent antineoplastic therapy, time to first use of chemotherapy, time to pain progression, health-related quality of life and adverse events. Median follow-up time was 23 months for the primary analysis and 48 months for the long term (third interim) analysis. Patients were randomly assigned in a 2:1 ratio to receive the intervention (160 mg enzalutamide once daily) or placebo once daily, next to continuation of ADT (either with a gonadotropin-releasing hormone agonist or antagonist or with previous bilateral orchiectomy). After the primary analysis, patients were unblinded and given the opportunity to switch to the enzalutamide group for the open-label extension. A total of 87/114 (76%) decided to switch. At the long term follow up (ca 4 years) 31% of enzalutamide treated patients had discontinued treatment because of disease progression, adverse events or other reasons. 1401 patients (933 enzalutamide and 468 placebo) were randomized and evaluated in an intention to treat analysis. Groups were comparable at baseline.

 

The SPARTAN trial

The SPARTAN trial (Small, 2019; Smith, 2018 and Smith, 2021) examined the effect of apalutamide versus placebo in patients with nmCRPC. The trial was conducted in 332 centers in 26 countries in North America, Europe, and the Asia–Pacific region. The primary endpoint was metastasis free survival, whereas secondary outcomes were time to metastasis, progression-free survival, time to symptomatic progression, overall survival and time to initiation of chemotherapy. The median follow up time was 20 months for the first interim analysis (Smith, 2018) and 52 months for the long term analysis (Smith, 2021). Patients were randomly assigned in a 2:1 ratio to receive apalutamide (240 mg per day) or matched placebo and ADT. After the primary analysis, patients in the placebo group were unblinded and given the opportunity to cross over to the open label apalutamide group. After unblinding, 19% patients of the placebo group without disease progression decided to switch to apalutamide. At the long term follow up, 70% of the intervention group and 100% of placebo group had discontinued treatment because of progressive disease, adverse events or other reasons. A total of 1207 patients (806 intervention and 401 control) were assessed in an intention-to-treat analysis. Groups were comparable at baseline.

 

Results

 

1. Survival

An overview of study characteristics and corresponding hazard ratios for all survival outcomes can be found in Table 2.

 

Table 2 Overview survival outcomes reported by ARAMIS, PROSPER and SPARTAN

 

Interim analysis

Long term analysis

 

1.1 Survival overall (HR, 95%CI)

1.2 Metastasis free survival (HR, 95% CI)

1.3 Progression free survival (HR, 95% CI)

1.4 Death related to prostate cancer (HR, 95% CI)

1.5 Long term survival (HR, 95% CI)

ARAMIS trial (Fizazi, 2019)

0.71 (0.51-0.99)

0.41 (0.34-0.49)

0.13 (0.11-0.16)

NR

NR

PROSPER trial (Hussain, 2018; Sternberg, 2020)

0.80 (0.58-1.09)

0.29 (0.24-0.35)

0.07 (0.05-0.08)

0.81 (0.72-0.91)

0.73 (0.61-0.89)

SPARTAN trial (Small, 2019; Smith 2018 and 2019)

0.70 (0.47-1.04)

0.27 (0.22-0.34)

0.29 (0.24-0.36)

NR

0.78 (0.64-0.96)

Results pooled

0.74 (0.60-0.90)

0.32 (0.29-0.36)

0.14 (0.13-0.16)

0.81 (0.72-0.91)

0.76 (0.66-0.87)

Sources: Fizazi, 2019; Hussain, 2018; Small, 2019; Smith 2018; Smith, 2019 and Sternberg, 2020

 

Interim analysis

 

1.1         Overall survival

Overall survival at first interim follow up was reported by three trials (ARAMIS, PROSPER and SPARTAN). The outcome was defined as the number of patients still alive at the end of the trial period. The outcome was assessed at the first interim follow up at 18, 23 and 20 months follow up respectively.

 

In the ARAMIS trial, overall survival was reported in 877/955 (91.8%) in the treatment group versus 496/554 (89.5%) in the placebo group (HR (95%CI): 0.71 (0.50-0.99)) (Fizazi, 2019).

In the PROSPER trial, overall survival was reported in 830/933 (89%) in the treatment group versus 402/468 (87%) in the placebo group (HR (95%CI): 0.80 (0.58-1.09)) (Hussain, 2018).

In the SPARTAN trial, overall survival was reported with HR (95%CI): 0.70 (0.47-1.04) at the first interim analysis at 20 month-follow up. For this analysis, absolute survival numbers were not given (Smith, 2018).

The pooled overall survival (using the first interim analysis numbers of SPARTAN) was estimated at HR (95%): 0.74 (0.60-0.90) favouring ARSI (Figure 1). The risk reduction in overall death was estimated at 26% due to ARSI.

 

Figure 1 Overall survival

 

N.B. Follow up at median 18, 20 and 23 months for ARAMIS, PROSPER and SPARTAN respectively.

Sources: Fizazi, 2019; Hussain, 2018 and Smith, 2018

 

1.2          Metastases free survival

Metastases free survival at first interim follow up was reported by three trials (ARAMIS, PROSPER and SPARTAN). The outcome was defined as the median number of months before metastases were diagnosed. The outcome was assessed at the first interim follow up at 18, 23 and 20 months follow up respectively.

 

In the ARAMIS trial, median metastases free survival was reported as 40.4 months in the treatment group versus 18.4 months in the placebo group (HR (95%CI): 0.41 (0.34-0.50) favouring treatment) (Fizazi, 2019).

In the PROSPER trial, median metastases free survival was reported as 36.6 months in the treatment group versus 14.7 months in the placebo group (HR (95%CI): 0.29 (0.24-0.35) favouring treatment) (Hussain, 2018).

In the SPARTAN trial, median metastases free survival was reported as 40.5 months in the treatment group versus 16.2 months in the placebo group (HR (95%CI): 0.27 (0.22-0.34) favouring treatment) (Smith, 2018).

The pooled metastases free survival was estimated at HR (95%CI): 0.32 (0.29-0.36) favouring ARSI (Figure 2). The risk reduction in metastases was estimated at 68% due to ARSI.

 

Figure 2 Metastases free survival

N.B. Follow up at median 18, 20 and 23 months for ARAMIS, PROSPER and SPARTAN respectively.

Sources: Fizazi, 2019; Hussain, 2018 and Smith, 2018

 

1.3         Overall survival at the long term follow up

Two trials (PROSPER and SPARTAN) reported an extended follow-up of 48 and 52 months, respectively, and established overall survival of the intervention and placebo arms. The outcome was defined as the number of patients still alive at the long term follow up and the median number of months still alive. The outcome was measured at the long term open label extension at a median follow up of 48 and 52 months respectively.  (Rodriguez-Vida, 2022; Smith, 2021; Tombal, 2022; Wenzel, 2022)

 

In the PROSPER trial, 645/933 patients (69%) were still alive in the treatment group compared to 290/468 (62%) in the placebo group. Median survival was reported as 67 months in the treatment group compared to 56 months in the placebo group (HR (95%CI): 0.73 (0.61-0.89) favouring treatment).

In the SPARTAN trial, at the long term follow up, 532/806 patients (66%) were still alive in the treatment group compared to 247/401 patients (62%) in the placebo group. Median survival was reported as 73.9 months in the treatment group versus 59.9 months in the placebo group (HR (95%): 0.78 (0.64-0.96) favouring treatment).

The pooled overall survival was estimated at HR (95%CI): 0.76 (0.66-0.87) (figure 5). The reduction in death was estimated at 24% due to ARSI treatment.

 

Figure 5 Long term survival

N.B. Follow up median follow up at 48 and 52 months for PROSPER and SPARTAN

Sources: Smith, 2021 and Sternberg, 2020

 

2. Toxicity

Data on toxicity was reported by three trials (ARAMIS, PROSPER and SPARTAN). Data was separately assessed for any event, grade ≥3 events and events leading to death. An overview can be found in table 3. In apalutamide treated patients, grade 3 and higher rash and hypertension, in enzalutamide treated patients  grade 3 and higher fatigue and hypertension and in darolutamide treated patients fatigue was more common than in placebo treated patients

 

Table 3 Toxicity overview reported by ARAMIS, PROSPER and SPARTAN

1At the end of the trial period, after planned inclusion of app. N=1500 (ARAMIS), N=1440 (PROSPER) and N=1200 (SPARTAN) and before reveal of treatment and crossover allowance, median follow up 18 (ARAMIS), 23 (PROSPER) and 20 months (SPARTAN).

2At the long term follow up, after crossover allowance, intention-to-treat analysis, median follow up 48 (PROSPER) and 52 months (SPARTAN)

Sources: Sopeña Sutil (2021) for interim analysis and Smith, 2021 and Sternberg, 2021 for long term analysis

 

Interim analysis

 

Adverse events
Adverse events of any grade were common among treatment groups (varying 87% to 96%) and not significantly higher in the treatment groups compared to placebo (3 trials, n=4104, RR (95%CI) 1.08 (0.98-1.18)). However ≥ Grade 3 adverse events were more common in the treatment groups (3 trials, n=4104, RR (95%CI) 1.31 (1.18-1.45)). Any adverse event leading to death was not significantly higher in the treatment groups (3 trials, n=4104, RR (95%CI) 2.67 (0.79-9.02)) (Sopeña Sutil, 2021).

 

Long term analysis

 

Adverse events at long term assessment
Adverse events at the long term analysis was reported by two studies: PROSPER (Sternberg, 2020) and SPARTAN (Smith, 2021). The outcome was assessed at a median follow up of 48 months (PROSPER) and 52 months (SPARTAN).

Adverse events at any grade were not significantly higher at the long term follow up (2 trials, n=2596, RR (95%CI) 1.09 (0.97-1.23)). The number of patients with ≥ Grade 3 adverse events was significantly higher in the treatment group (RR (95%CI) 1.64 (1.43-1.89) and adverse events leading to death was higher in the treatment group 7.38 (3.00-18.20).

In the PROSPER, <1% of patients in the control group and 2% of patients in the enzalutamide treated group died of a cardiovascular event. According to the PROSPER research group itself, none of the cardiovascular events  in the treatment group could have been related to the treatment medication (Sternberg, 2020).

 

4. Quality of life

 

Interim analysis

 

All three trials reported Health related Quality of Life (HRQoL) as FACT-P, EORTC-5D-3L and EORTC-QLQ-PR25 questionnaires scores, however all three trials used different ways of reporting. One trial reported mean FACT-P scores and various EORTC questionnaires scores at 16 weeks follow up compared to baseline (ARAMIS, Fizazi, 2019), one trial reported mean time in months to a decrease of the FACT-P score of at least 10 points (PROSPER, Hussain, 2018) and one trial reported the change in total FACT-P score from baseline to 29 months (SPARTAN, Smith, 2018). Variation in health related Quality of life reporting strategies, makes comparison of the trials challenging.

 

ARAMIS reported similar mean FACT-P and various EORTC-scores at 16w follow up in the intervention and control groups, suggesting no difference in HRQoL. PROSPER reported mean time to degradation in HRQoL questionnaires scores of 11.1 months in both groups, also suggesting no difference in HRQoL. SPARTAN reported a reduction in FACT-P score from baseline of 0.99 and 3.29 at the 29m follow up in the intervention and control arms of the study, respectively. With a threshold of 10 points for a clinical significant change, also HRQoL assessment in SPARTAN suggests no difference in HRQoL between intervention and control ars. With that, all three trials reported comparable quality of life scores at follow up (Fizazi, 2019; Hussain, 2018 and Smith, 2018).

 

Level of evidence of the literature

The level of evidence (GRADE method) is determined per comparison and outcome measure and is based on results from RCTs and therefore starts at level ‘high’.

 

The level of evidence regarding the outcome measure survival was not further downgraded.

 

The level of evidence regarding the outcome measure toxicity was not further downgraded.

 

The outcome costs was not assessed by the included studies.

 

The level of evidence regarding the outcome measure quality of life was not further downgraded.

 

Conclusions

 

1. Survival

High GRADE

ARSI, combined with ADT, versus ADT alone or placebo results in an increased survival (metastasis free survival, progression free survival, overall survival, and long term survival) in patients with non metastatic castration resistant prostate cancer (nmCRPC).

 

Sources: Fizazi, 2019; Hussain, 2018; Small, 2019; Smith 2018; Smith, 2019 and Sternberg, 2020

 

2. Toxicity

High GRADE

ARSI, combined with ADT, versus ADT alone or placebo results in comparable adverse events any grade in patients with nmCRPC.

ARSI, combined with ADT, versus ADT alone or placebo results in higher adverse events grade 3 in patients with nmCRPC.

 

Sources: Sopeña Sutil, 2021; Smith, 2021; Sternberg, 2021

 

3. Costs

No GRADE

The outcome costs was not assessed by the included studies.

 

4. Quality of life

High GRADE

ARSI, combined with ADT, versus ADT alone or placebo results in a comparable quality of life at follow up in patients with nmCRPC.

 

Sources: Fizazi, 2019; Hussain, 2018 and Smith, 2018    

 

Sub question 1b ARSI mCRPC

 

Description of studies

 

Two trials were included for sub question 1b: the COU-AA-302-study with five publications (Basch, 2013; Morris, 2015; Rathkopf, 2014; Ryan, 2013 and Ryan, 2015) and the PREVAIL trial with two publications (Beer, 2014 and Loriot, 2015). Since publication of the previous version of this module (2016), no new publications appeared.

 

Table 5 Overview study characteristics COU-AA-302 and PREVAIL

 

 

Trial years

No. of participants (intervention, control)

Country/setting, no. of participating centres

Intervention, control

Postbaseline subsequent cytotoxic therapy

The COU-AA-302 study

2010-2012

1088 (546, 542)

151, 12 countries

Abiraterone-prednisone, prednisone alone

Intervention: 252/546 (46%)

Control: 339/542 (63%)*

The PREVAIL study

2010-2012

1717 (872, 845)

Ca 180 centres worldwide, not exactly reported

Enzalutamide, placebo

Intervention: 382/872 (44%)

Control: 642/845 (76%)

*Docetaxel and Cabazitaxel added

 

The COU-AA-302 study

The COU-AA-302 study assessed the effect of abiraterone plus prednisone versus prednisone alone in chemotherapy-naïve patients with metastatic castration-resistant prostate cancer (mCRPC). The study is a phase-3 RCT conducted in 151 centers in 12 countries (in collaboration with US and European agencies). The primary end point was radiographic progression of prostate cancer at a bone scan or computed tomography (CT) or magnetic resonance imaging (MRI) or death by any cause. Three interim analyses for overall survival (OS) were planned at approximately 15%, 40% and 55% of expected deaths. At the first interim analysis (December 2010) median follow up time was 8.3 months, at the second interim analysis (December 2011) median follow up time was 27.2 months and median follow up time at the third interim analysis (May 2012) was not reported. Patients were randomly assigned in a 1:1 ratio to receive abiraterone acetate 1,000 mg daily + prednisone 5 mg or placebo + prednisone. Patients discontinued therapy because of radiographic or clinical progression (protocol-defined), adverse events or withdrawal of consent. A total of 1088 patients (546 Abiraterone-prednisone and 542 placebo-prednisone) were successfully randomized (Morris, 2015).

 

The PREVAIL study

The PREVAIL study investigated the effect of enzalutamide versus placebo in chemotherapy-naïve patients with metastatic castration-resistant prostate cancer (mCRPC). The phase-3 RCT was conducted in approximately 180 centers worldwide (exact locations not mentioned). The primary end points were radiographic progression-free survival and overall survival. At the planned interim analysis (May 2012) for survival after 516 deaths of planned enrollment of 1680 patients, median follow up was approximately 22 months. Patients were randomly assigned in a 1:1 ratio to receive either enzalutamide (160mg daily) or placebo capsules. Treatment discontinuation was the result of death, withdrawal of consent, disease progression (predefined), adverse events or protocol violation. A total of 1717 participants were successfully randomized over 872 enzalutamide and 845 placebo group.

 

Results

 

1. Survival

An overview of study characteristics and corresponding hazard ratios for all survival outcomes can be found in table 6.

 

Table 6 Overview survival outcomes reported by COU-AA-302 and PREVAIL

 

Interim analysis

Long term analysis

 

 

1.1 Survival overall (HR, 95%CI)

1.2 Progression free survival (HR, 95% CI)

1.3 Death related to prostate cancer (RR, 95% CI)

1.4 Long term survival (HR, 95% CI)

The COU-AA-302 study

0.75 (0.61 to 0.93)

0.53 (0.45 to 0.62)

NR

0.81 (0.70 to 0.93)

The PREVAIL study

0.71 (0.60 to 0.84)

0.19 (0.15 to 0.23)

No long term follow up performed.

No long term follow up performed.

Results pooled

0.73 (0.64 to 0.83)

Not pooled, different definitions and follow up time

NR

0.81 (0.70 to 0.93)

Sources: Beer, 2014; Morris, 2015 and Ryan, 2015

 

Interim analysis

 

1.1         Overall survival

Overall survival at first interim follow up was reported by two trials (COU-AA-302 and PREVAIL). The outcome was defined as the number of deaths (out of total number of patients) at the end of the trial period. The outcome was assessed at the first interim follow up at 27 months (COU-AA-302) and 22 months (PREVAIL).

 

The COU-AA-302 reported 147 deaths in the abiraterone (total number still alive not reported) + prednisone group versus 186 deaths in the prednisolone alone group (HR (95%CI): 0.75 (0.61-0.93) (Morris, 2015). The PREVAIL study reported 241/872 (28%) of patients died in the enzalutamide group versus 299/845 (35%) of patients died in the placebo group (HR (95%CI): 0.71 (0.60-0.84). The pooled overall survival was estimated at HR (95%CI): 0.73 (0.64-0.83) favouring ARSI. The risk reduction in death was estimated at 27% due to ARSI.

 

Figure 6 Overall survival at first interim analysis at 27 and 22 months for COU-AA-302 and PREVAIL respectively

 

1.2          Progression free survival

Progression free survival at first interim follow up was reported by two trials (COU-AA-302 and PREVAIL). COU-AA-302 defined the outcome as “time from random assignment to the first occurrence of either progression by bone scan, progression by computed tomography (CT) or magnetic resonance imaging as defined by modified RECIST (version 1.0), or death resulting from any cause (COU-AA-302)”. PREVAIL defined the outcome as “the rate of radiographic progression-free survival at the 12months follow up”. Because of different study definitions it was decided not to pool data.

 

The COU-AA-302 estimated the median time to disease progression at the first interim follow up at 16.5 months abiraterone plus prednisone group versus 8.3 months in the placebo group (HR (95%CI): 0.53 (0.45-0.62) (follow up 27 months). The PREVAIL study reported the rate of radiographic progression-free survival at the 12 months follow up, which was 65% in the enzalutamide group versus 14% in the placebo group (HR (95%CI): 0.19 (0.15-0.23).

 

Table 7 Progression-free survival reported by COU-AA-302 and PREVAIL

 

Progression free survival treatment group

Progression free survival control group

Hazard ratio (95%CI)

COU-AA-302

16.5 months1

8.3 months1

0.53 (0.45-0.62)

PREVAIL

65%2

14%2

0.19 (0.15-0.23)

1Median follow up time 27 months. 2Median follow up time 12 months.

Sources: Beer, 2014 (PREVAIL) and Morris, 2015 (COU-AA-203)

 

Long term analysis

 

1.3          Death related to prostate cancer at long term follow up

The two included studies did not report this outcome measure.

 

1.4          Long term survival

Long term survival was reported by one study (COU-AA-302). COU-AA-302 defined the outcome as the risk of death in the treatment group compared to the placebo group at a follow up of a median of 49.2 months.

 

In the COU-AA-302 trial, 352/546 (65%) deaths occurred in the abiraterone acetate group versus 387/542(71%) in the placebo group (HR (95%CI): 0.81 (0.70–0.93). Median overall survival was 34.7 months in the treatment group versus 30.3 months in the placebo group. The reduction in death due to ARSI was estimated at 19%.

 

Table 8 Long term survival at long term follow up reported by COU-AA-302

 

Overall death treatment group

Overall death control group

Hazard ratio (95%CI)

COU-AA-302

352/546 (65%)1

387/542 (71%)1

0.81 (0.70–0.93)

2 Median follow up time 49 months.

Source: Ryan, 2015 (COU-AA-302)

 

2 Toxicity

 

Both trials (COU-AA-302 and PREVAIL) reported on toxicity. COU-AA-302 reported adverse events at long term follow up at a median follow up time of 49 months. An overview can be found in table 9. In abiraterone treated patients, grade 3 and higher cardiac disorders and increased liver enzymes were more common than in control patients, in enzalutamide treated patients, grade 3 and higher hypertension was more common than in control patients.

 

Table 9 Toxicity overview reported in COU-AA-302 and PREVAIL

1At the end of the trial period, after planned enrolment of n=1000 and median follow up time 22 months for COU-AA-302 and after planned enrolment of n=1680 and median follow up time 22 months for PREVAIL

2At the long term follow up, after crossover allowance, intention-to-treat analysis, median follow up 49 months for COU-AA-302

Source: Beer, 2014; Ryan, 2013, Ryan, 2015

 

Interim analysis

 

Adverse events

Adverse events at any grade were common and occurred in 99% and 97% in the treatment groups (abiraterone and enzalutamide respectively) versus 97% and 93% in the placebo groups (RR (95%CI): 1.03 (1.01 to 1.05). Adverse events ≥grade 3 occurred in 48% and 43% in the abiraterone and enzalutamide group respectively versus 43% and 37% in the placebo groups ((RR (95%CI): 1.08 (0.93 to 1.25). Adverse events leading to death occurred in 4% and 4% in the abiraterone and enzalutamide group respectively versus 2% and 4% in the placebo groups (HR (95%CI): 1.26 (0.86 to 1.86) (Beer, 2014 and Ryan, 2013). The COU-AA-302 study reported the most common adverse events leading to death were general disorders, including disease progression, a decline in physical health, and infections including

pneumonia and respiratory tract infection.

 

Long term analysis

 

Adverse events at long term analysis

The COU-AA-302 study reported on adverse events in the long term analysis at a median follow up of 49.2 months. Adverse events at any grade occurred in 100% of treated patients and 97% of placebo patients. Adverse events ≥ grade 3 occurred in 54% of treatment group patients versus 44% of placebo group patients (RR (95%CI): 1.22 (1.08-1.39). Adverse events leading to death occurred in 4% of treatment group patients versus 2% of placebo group patients (RR (95%CI): 1.59 (0.85 to 3.01). The COU-AA-302 study reported three patients (1%) in the treatment group had general physical health deterioration as a sign of clinical progression resulting in death versus three cases (1%) in the placebo group (Ryan, 2015)

 

3. Costs

 

Prices (November 2022) for Enzalutamide and Abiraterone are reported at 113,62 and 113,57 euro per day, respectively, while prices for abiraterone have decreased significantly since the patent has expired in September 2022  (source: Zorginstituut Nederland)

  

4. Quality of life

Quality of life was measured with the FACT-P score by both studies (COU-AA-302 and PREVAIL). The outcome measure was defined as the number of months until a decline of 10 points or more occurs of the FACT-P total score in both studies (Beer, 2014 (supplementary data) and Ryan, 2013). An overview can be found in table 10.

 

Table 10 Quality of life reported in COU-AA-203 and PREVAIL

 

COU-AA-302

PREVAIL

Pooled results

 

Abiraterone + prednisone (n=546)

N (%)

Placebo + prednisone (n=542)

N (%)

Enzalutamide (n=872)

N (%)

Placebo (n=845)

N (%)

HR (95%CI)

Quality of life deterioration, measured with FACT-P

12.7 mo.

8.3 mo.

11.3 mo.

5.6 mo.

0.69 (0.56-0.86)

Sources: Beer, 2014 and Ryan, 2013

 

The COU-AA-302 reported a median time of 12.7 months until quality of life decline occurred in the treatment group versus 8.3 months in the placebo group. The PREVAIL reported a median time of 11.3 months versus 5.6 months until quality of life decline occurred. The pooled results were HR (95%CI): 0.69 (0.56-0.86) favouring treatment.

 

Level of evidence of the literature

The level of evidence (GRADE method) is determined per comparison and outcome measure and is based on results from RCTs and therefore starts at level ‘high’.

 

The level of evidence regarding the outcome measure survival was not further downgraded.

 

The level of evidence regarding the outcome measure toxicity was not further downgraded.

 

The outcome costs was not assessed by the included studies.

 

The level of evidence regarding the outcome measure quality of life was not further downgraded.

 

Conclusions

 

1. Survival

High GRADE

ARSI versus placebo results in an increased survival (overall survival, progression free survival and long term survival) in patients with metastatic castration resistant prostate cancer (mCRPC).

 

Sources: Beer, 2014; Morris, 2015 and Ryan, 2015

 

2. Toxicity

High GRADE

At the end of the study period (ca. 2 year follow up), ARSI versus placebo results in comparable adverse events any grade and adverse events grade 3 in patients with mCRPC.

 

At long term (ca. 4 year follow up), ARSI results in comparable adverse events any grade but higher adverse events grade 3 in patients with mCRPC.

 

Sources: Beer, 2014; Ryan, 2013, Ryan, 2015

 

3. Costs

No GRADE

The outcome costs was not assessed by the included studies in patients with mCRPC.

 

4. Quality of life

High GRADE

ARSI versus placebo results in an improved quality of life in patients with mCRPC.

 

Sources: Beer, 2014 and Ryan, 2013

 

Sub question 2. Chemotherapy

 

For this sub question, no studies were found. All studies that were found lacked a control group without chemotherapy treatment or ADT alone. More information can be found in the Table of excluded studies. Therefore, no systematic literature analysis to this question was performed.

 

In short: Docetaxel was the first therapeutic intervention that showed an overall survival benefit over a drug that has not shown to improve overall survival (mitoxantrone) (Tannock et al NEJM, 2004). Six years later, the TROPIC study was published which showed that cabazitaxel treated patients, previously treated with docetaxel, had a survival benefit over treatment with mitoxantrone (de Bono, Lancet, 2010). The 2019 CARD study showed that patients previously treated with docetaxel and an ARSI, derived a longer overall survival when treated with cabazitaxel than patients treated with an ARSI after progressing on another ARSI (de Wit, NEJM, 2019).

 

Sub question 3. Radioligand therapy

 

Description of studies

One trial was included for sub question 3 radioligand therapy: the ALSYMPCA trial with six publications (Hoskin, 2014; Nome, 2015; Parker, 2013 and Parker, 2016 & Sartor, 2014 and Sartor, 2017). An overview of the study can be found in table 11.

 

Table 11 Overview study characteristics ALSYMPCA

 

 

Trial years

No. of participants (intervention, control)

Country/setting, no. of participating centres

Intervention, control

Postbaseline subsequent cytotoxic therapy

The ALSYMPCA trial

2008-2011

921 (614, 307)

136 centres, 19 countries

Ra-223, placebo

NR

 

The ALSYMPCA trial

The ALSYMPCA trial assessed the effect of radium-223 (ra-223) on bone metastases in patients with mCRPC. The trial was conducted in 136 study centers in 19 countries. Patients with skeletal metastases were included, but no visceral involvement (Nome, 2015). A single enlarged lymph node was allowed. The primary end point was overall survival, whereas time to the first skeletal event and various biochemical end points were also assessed (Parker, 2013). The first interim analysis occurred when approximately 50% of deaths had occurred (i.e. 320 deaths, n=901) (Parker, 2013). The second interim analysis took place at the end of the trial period when the intended 900 participants were included. At this moment 528 deaths had occurred (n=921), before planned early discontinuation and allowance for crossover for ethical reasons on the basis of the first interim analysis (Parker, 2013). Afterwards several post-hoc assessments at n=921 were performed (Hoskin, 2014, Parker, 2013 and Sartor, 2017).

Patients were stratified according to previous use or nonuse of docetaxel, baseline alkaline phosphatase level and current use or nonuse of bisphosphonate. Subsequently, they were randomized in a 2:1 ratio to receive up to six intravenous injections of ra-223 or placebo. At the time of the first analysis, approximately 58% had received all six injections in the ra-223 group and 47% in the placebo group. The dose was 50 kBq per kilogram of body weight. One injection was administered every four weeks. Besides this, patients received routine care provided at the treatment center such as radiation therapy, glucocorticoids, antiandrogens, ketoconazole, or estrogens. Chemotherapy and hemibody external radiotherapy was not allowed during the trial period. Planned follow up was 3 years. Patients discontinued treatment because of adverse events, clinical progression, crossover or withdrawal. Groups were comparable at baseline.

 

1. Survival

An overview of study characteristics and corresponding hazard ratios for all survival outcomes can be found in table 12.

 

Table 12 Overview survival outcomes ALSYMPCA

 

Interim analysis

Long term analysis

 

 

1.1 Survival overall (HR, 95%CI)

1.2 Progression free survival (HR, 95% CI)

1.3 Death related to prostate cancer (RR, 95% CI)

1.4 Long term survival (HR, 95% CI)

ALSYMPCA

0.70 (0.58-0.83)

0.66 (0.52-0.83)

NR

NR

Sources: Parker, 2013 and Parker, 2018

 

Interim analysis

 

1.1         Overall survival at first interim follow up

Overall survival at first interim follow up was defined as the time from randomization until the date of death, regardless of the cause (Parker, 2013). The outcome was assessed at the end of the trial period when n=921 patients were included. At this moment, the median number of injections was six in the ra-223 group and five in the placebo group. The median follow up time in months was not reported.

 

Table 12 Overall survival at interim analysis

 

Overall survival treatment group

Overall survival control group

Hazard ratio (95%CI)

ALSYMPCA

14.9 months

11.3 months

0.70 (0.58-0.83)

Sources: Parker, 2013

 

The study reported an overall survival of median 14.9 months in the treatment group versus 11.2 months in the control group (HR (95%CI): 0.70 (0.58-0.83)). It is estimated ra-223 prolonged survival with app. 4 months.

 

1.2         Progression free survival

Parker (2013) defined the outcome as time since randomization until first skeletal event occurred. The outcome was assessed at the end of the trial period (see above).

 

 Table 13 Progression free survival at first interim follow up when app. 50% of deaths had occurred n=809

 

PFS toch? treatment group

PFS control group

Hazard ratio (95%CI)

ALSYMPCA

15.6 months

9.8 months

0.66 (0.52-0.83)

Sources: Parker, 2013

 

The study reported time until first symptomatic event of median 15.6 months in the intervention group versus 9.8 months in the placebo group (HR (95%CI): 0.66 (0.52-0.83)). It is estimated ra-223 prolonged time until symptomatic skeletal event with app. 6 months.

 

Long term analysis

Not reported.

 

1.4         Long term survival

Not reported.

 

2             Toxicity

ALSYMPCA reported on toxicity. The authors reported on adverse events (any grade, grade 3-4 and serious adverse events). They did not report on adverse events leading to death. Median follow up time at interim analysis is not reported. Median follow up at long term analysis was 13 months (0-36) in the ra-223 group and 9 months (0-36) in the placebo group. An overview can be found in table 14. Grade 3 and higher adverse events were infrequent in the trial and no significant differences between the intervention and control arm were observed.

 

Table 14 Toxicity overview reported in ALSYMPCA

ALSYMPCA

 

 

Radium-223 (n=600)

N (%)

Placebo (n=301)

N (%)

RR (95% CI)

Interim analysis1

Adverse event

Any grade

≥ Grade 3

Leading to death

558 (93)

339 (56)

NR

290 (96)

188 (62)

NR

0.97 (0.93-1.00)

0.90 (0.81-1.01)

NR

Long term analysis2 – at the long term follow up, after crossover allowance

Adverse event

Any grade

≥ Grade 3

Leading to death

564 (94)

350 (58)

98 (16)

292 (97)

194 (65)

68 (23)

0.96 (0.94-1.00)

0.91 (0.81-1.01)

0.72 (0.54-0.95)

1At the end of the trial period, after planned inclusion of 900 participants and before reveal of treatment and crossover allowance, median follow up in months not reported, n=901

2At the long term follow up, after crossover allowance, intention-to-treat analysis, median follow up 13 months in ra-223 and 9 months in placebo group, n=901

Sources: Parker, 2013 and Parker, 2018

 

Interim analysis

Adverse events

Adverse events were defined as any adverse event for which medical treatment was required, and determined at the interim analysis when the intended study population was admitted (n=901) (Parker, 2013).

Adverse events at any grade were common among both groups and not significantly different among groups (RR (95%CI): 0.97 (0.93-1.00). ≥ Grade 3 events were also not significantly different among groups ( RR (95%CI): 0.90 (0.81-1.01)). The authors did not report adverse events leading to death, however they did report one grade 5 hematologic adverse event was possibly related to the study drug: thrombocytopenia in a

patient in the radium-223 group, who died from pneumonia with hypoxemia without bleeding.

 

Long term analysis

 

Adverse events at long term analysis

Parker (2018) assessed the toxicity after extended follow up (median 13 and 9 months in intervention and control group respectively). They performed intention-to-treat (all patients were analyzed to the assigned groups), although data of the radium-223 + crossover group can be found in the article. A total of 901 (of 921) participants entered the safety data analysis, of which 195 (33%) in the intervention group and 134 (45%) in the placebo group did not proceed for 3-year follow up. Of patients that did proceed in the 3-year follow up, 355 (88%) in the ra-223 group and 153 (92%) in the placebo group discontinued during the three-year follow up because of death (70% in treatment group), patient request (7% in treatment group), disease progression (3% in treatment group), lost to follow up (<2%) or adverse events (<1%). These numbers were comparable in the placebo group. In total, 48 (12%) of ra-223 patients completed the three-year follow up versus 12 (7%) in the placebo group.

 

Adverse events at any grade were non-significantly different among groups (RR (95%CI): 0.96 (0.94-1.00) and ≥ grade 3 events were also non-significantly different (RR (95%CI): 0.91 (0.81-1.01)). The number of adverse events leading to death, however, was significantly higher in the placebo group (RR (95%CI): 0.72 (0.54-0.95). According to Parker (2018) two deaths were possibly related to treatment: one patient who received two injections died of myocardial infarction or bowel ischemia at 8w after the first injection, and one patient who received one injection died of general health deterioration and multiple organ failure at 4w after the injection.

 

3.            Costs

Not reported.

 

4.            Quality of life

ALSYMPCA assessed quality of life with the FACT-P scale (0-156 with higher scores indicating better quality of life).

 

The study reported a mean reduction at 16 weeks follow up compared to baseline of 2.6 points in the treatment group and 6.8 points in the placebo group, which are both not clinically significant. An increase of ≥10 points was regarded as a meaningful change. In the intervention group, 25% had a meaningful change versus 16% of the placebo group. This result was statistically significant in favour of the intervention but not clinically different (≥10 points).

 

Table 15 Change in Quality of life at 16 weeks as reported in ALSYMPCA

 

ALSYMPCA

Pooled results

 

Radium-223 (n=614)

Placebo (n=307)

HR (95%CI)

Quality of life measured with FACT-P

-2.7

25% ≥10 points change

-6.8

16% ≥10 points change

Not reported, p=0.006

Sources: Parker, 2013

 

Level of evidence of the literature

The level of evidence (GRADE method) is determined per comparison and outcome measure and is based on results from RCTs and therefore starts at level ‘high’.

 

The level of evidence regarding the outcome measure survival was downgraded by one level because of risk of bias (no clear reporting of the median follow up time at the first and second interim analysis and no clear reporting of long term survival outcome measures).

 

The level of evidence regarding the outcome measure toxicity was not further downgraded.

 

The outcome costs was not assessed by the included studies.

 

The level of evidence regarding the outcome measure quality of life was downgraded by one level because of unclear outcome measure reporting (effect measures are missing).

 

Conclusions

 

1. Survival

Moderate GRADE

Radioligand therapy versus placebo probably results in an increased survival (overall survival, progression free survival and death related to prostate cancer) in patients with metastatic castration resistant prostate cancer (mCRPC).

 

Sources: Parker, 2013 and Parker, 2018

 

2. Toxicity

High GRADE

Radioligand therapy versus placebo results in comparable adverse events of any grade in patients with mCRPC.

 

Sources: Parker, 2013 and Parker, 2018

 

3. Costs

No GRADE

The outcome costs was not assessed by the included studies.

 

4. Quality of life

Moderate GRADE

Radioligand therapy versus placebo probably results in a not clinically relevant change in quality of life at follow up compared to baseline in patients with mCRPC.

 

Sources: Parker, 2013 and Parker, 2018

A systematic review of the literature was performed to answer the following question: What is the effect of ARSI, in chemotherapy-naïve patients with non-metastatic and metastatic castration-resistant prostate cancer (nmCRPC and mCRPC)?

 

1. ARSI

 

1a ARSI nmCRPC

 

Research question: What is the effect of ARSI + ADT versus ADT alone in patients with non-metastatic castration-resistant prostate cancer (nmCRPC) as a first line treatment?

 

P: Chemotherapy-naïve patients with nmCRPC

I:  ARSI (abiraterone, enzalutamide, apalutamide, darolutamide) combined with ADT

C: ADT alone          

O: Survival, toxicity, costs, quality of life

 

1b ARSI mCRPC

 

Research question: What is the effect of ARSI + ADT versus ADT alone in patients with metastatic castration-resistant prostate cancer (mCRPC) as a first line treatment?

 

P: Chemotherapy-naïve patients with mCRPC

I: ARSI (abiraterone, enzalutamide, apalutamide) combined with ADT

C: ADT alone          

O: Survival, toxicity, costs, quality of life

 

2. Chemotherapy

 

Research question: What is the effect of chemotherapy + ADT versus ADT alone in patients with metastatic castration-resistant prostate cancer (mCRPC) as a first line treatment?

 

P: Chemotherapy-naïve patients with mCRPC

I: Chemotherapy combined with ADT

C: ADT alone          

O: Survival, toxicity, costs, quality of life

 

3. Radioligand therapy

 

Research question: What is the effect of radioligand therapy + ADT versus ADT alone in patients with metastatic castration-resistant prostate cancer (mCRPC) as a first line treatment?

 

P: Chemotherapy-naïve patients with mCRPC

I: Radioligand combined with ADT

C: ADT alone          

O:  Survival, toxicity, costs, quality of life

 

Relevant outcome measures

The guideline development group considered survival (progression free survival, metastases free survival and overall survival) as a critical outcome measure for decision making; and toxicity, costs and quality of life as important outcome measures for decision making.

 

The working group defined the outcome measures as follows:

- Survival was defined as overall survival (regardless of cause), metastasis-free survival (in non-metastatic patients) and progression-free survival.
- Toxicity was defined as adverse events that were more frequent in the treatment arm than in the control arm of the registration trials and could potentially be related to treatment.

- Costs was defined as reported by ‘Zorginstituut Nederland’ (if reported).
- Quality of life was defined as measured with a validated questionnaire such as FACT-P or EQ-5D and reported in the registration trials.

 

The working group defined the limits of clinical relevance for the crucial outcome measure ‘overall survival’; an overall survival benefit >12 weeks OR a hazard ratio ≤0.7 due to treatment, which is in line with the current PASKWIL criteria used by the ‘NVMO commissie ter Beoordeling van Oncologische Middelen (cieBOM) to evaluate palliative treatments. For the other outcomes (survival, toxicity, quality of life), the GRADE default limits ≤0.8HR/RR≥1.25 were used, unless different limits for clinical relevance were given by the included studies.

Search and select (Methods)

On April 16th, 2021 the Medline (via OVID) and Embase (via Embase.com) databases were searched using relevant search terms for systematic reviews (SRs) and randomized controlled trials (RCTs) published from 2014 onwards. The new literature (2014-2021) is added to the literature from the original module published in 2016. The detailed search strategies are depicted under the tab Methods.The literature search yielded 1172 unique hits. Studies were selected based on the following criteria:

- Systematic reviews (SRs), (network) meta analyses, or Randomized Controlled Trials (RCTs).
- Involving chemotherapy-naïve patients with non-metastatic and metastatic castration resistant prostate cancer.

- Comparing ARSI, chemotherapy or targeted therapy with ADT versus ADT alone or ADT + placebo.
- Assessing survival, toxicity, costs or quality of life in the selected studies.
- In case of multiple publications, the most recent publication was selected, or the publication which addressed an study outcome of specific interest.

 

For sub question 1 ARSI, 22 studies were initially selected, 17 were excluded and 5 studies were included. For sub question 1a ARSI nmCRPC 3 studies were included and sub question 1b ARSI mCRPC 2 studies were included.

For sub question 2 chemotherapy, 7 studies were initially selected and none were included.

For sub question 3 radioligand, 11 studies were initially selected and one was included.

 

See ‘Table of excluded studies’ for reasons of exclusion under tab Methods.

 

Results

6 studies were included in the analysis of the literature. Important study characteristics and results are summarized in the evidence tables. The assessment of the risk of bias is summarized in the risk of bias tables.

  1. 1 - Basch E, Autio K, Ryan CJ, Mulders P, Shore N, Kheoh T, Fizazi K, Logothetis CJ, Rathkopf D, Smith MR, Mainwaring PN, Hao Y, Griffin T, Li S, Meyers ML, Molina A, Cleeland C. Abiraterone acetate plus prednisone versus prednisone alone in chemotherapy-naive men with metastatic castration-resistant prostate cancer: patient-reported outcome results of a randomised phase 3 trial. Lancet Oncol. 2013 Nov;14(12):1193-9.
  2. 2 - Beer TM, Armstrong AJ, Rathkopf DE, Loriot Y, Sternberg CN, Higano CS, Iversen P, Bhattacharya S, Carles J, Chowdhury S, Davis ID, de Bono JS, Evans CP, Fizazi K, Joshua AM, Kim CS, Kimura G, Mainwaring P, Mansbach H, Miller K, Noonberg SB, Perabo F, Phung D, Saad F, Scher HI, Taplin ME, Venner PM, Tombal B; PREVAIL Investigators. Enzalutamide in metastatic prostate cancer before chemotherapy. N Engl J Med. 2014 Jul 31;371(5):424-33
  3. 3 - Di Nunno V, Mollica V, Santoni M, Gatto L, Schiavina R, Fiorentino M, Brunocilla E, Ardizzoni A, Massari F. New Hormonal Agents in Patients With Nonmetastatic Castration-Resistant Prostate Cancer: Meta-Analysis of Efficacy and Safety Outcomes. Clin Genitourin Cancer. 2019 Oct;17(5):e871-e877
  4. 4 - Fendler, Wolfgang P., et al. "Prostate-Specific Membrane Antigen Ligand Positron Emission Tomography in Men with Nonmetastatic Castration-Resistant Prostate CancerDisease Burden by PSMA-PET in nmCRPC." Clinical Cancer Research 25.24 (2019): 7448-7454.
  5. 5 - Fizazi K, Shore N, Tammela TL, Ulys A, Vjaters E, Polyakov S, Jievaltas M, Luz M, Alekseev B, Kuss I, Kappeler C, Snapir A, Sarapohja T, Smith MR; ARAMIS Investigators. Darolutamide in Nonmetastatic, Castration-Resistant Prostate Cancer. N Engl J Med. 2019 Mar 28;380(13):1235-1246.
  6. 6 - Hird AE, Magee DE, Bhindi B, Ye XY, Chandrasekar T, Goldberg H, Klotz L, Fleshner N, Satkunasivam R, Klaassen Z, Wallis CJD. A Systematic Review and Network Meta-analysis of Novel Androgen Receptor Inhibitors in Non-metastatic Castration-resistant Prostate Cancer. Clin Genitourin Cancer. 2020 Oct;18(5):343-350.
  7. 7 - Hoskin P, Sartor O, O'Sullivan JM, Johannessen DC, Helle SI, Logue J, Bottomley D, Nilsson S, Vogelzang NJ, Fang F, Wahba M, Aksnes AK, Parker C. Efficacy and safety of radium-223 dichloride in patients with castration-resistant prostate cancer and symptomatic bone metastases, with or without previous docetaxel use: a prespecified subgroup analysis from the randomised, double-blind, phase 3 ALSYMPCA trial. Lancet Oncol. 2014 Nov;15(12):1397-406.
  8. 8 - Hussain M, Fizazi K, Saad F, Rathenborg P, Shore N, Ferreira U, Ivashchenko P, Demirhan E, Modelska K, Phung D, Krivoshik A, Sternberg CN. Enzalutamide in Men with Nonmetastatic, Castration-Resistant Prostate Cancer. N Engl J Med. 2018 Jun 28;378(26):2465-2474.
  9. 9 - Khalaf DJ, Annala M, Taavitsainen S, Finch DL, Oja C, Vergidis J, Zulfiqar M, Sunderland K, Azad AA, Kollmannsberger CK, Eigl BJ, Noonan K, Wadhwa D, Attwell A, Keith B, Ellard SL, Le L, Gleave ME, Wyatt AW, Chi KN. Optimal sequencing of enzalutamide and abiraterone acetate plus prednisone in metastatic castration-resistant prostate cancer: a multicentre, randomised, open-label, phase 2, crossover trial. Lancet Oncol. 2019 Dec;20(12):1730-1739.
  10. 10 - Loriot Y, Miller K, Sternberg CN, Fizazi K, De Bono JS, Chowdhury S, Higano CS, Noonberg S, Holmstrom S, Mansbach H, Perabo FG, Phung D, Ivanescu C, Skaltsa K, Beer TM, Tombal B. Effect of enzalutamide on health-related quality of life, pain, and skeletal-related events in asymptomatic and minimally symptomatic, chemotherapy-naive patients with metastatic castration-resistant prostate cancer (PREVAIL): results from a randomised, phase 3 trial. Lancet Oncol. 2015 May;16(5):509-21.
  11. 11 - Morris MJ, Molina A, Small EJ, de Bono JS, Logothetis CJ, Fizazi K, de Souza P, Kantoff PW, Higano CS, Li J, Kheoh T, Larson SM, Matheny SL, Naini V, Burzykowski T, Griffin TW, Scher HI, Ryan CJ. Radiographic progression-free survival as a response biomarker in metastatic castration-resistant prostate cancer: COU-AA-302 results. J Clin Oncol. 2015 Apr 20;33(12):1356-63.
  12. 12 - Nome R, Hernes E, Bogsrud TV, Bjøro T, Fosså SD. Changes in prostate-specific antigen, markers of bone metabolism and bone scans after treatment with radium-223. Scand J Urol. 2015 Jun;49(3):211-7.
  13. 13 - Parker C, Nilsson S, Heinrich D, Helle SI, O'Sullivan JM, Fosså SD, Chodacki A, Wiechno P, Logue J, Seke M, Widmark A, Johannessen DC, Hoskin P, Bottomley D, James ND, Solberg A, Syndikus I, Kliment J, Wedel S, Boehmer S, Dall'Oglio M, Franzén L, Coleman R, Vogelzang NJ, O'Bryan-Tear CG, Staudacher K, Garcia-Vargas J, Shan M, Bruland ØS, Sartor O; ALSYMPCA Investigators. Alpha emitter radium-223 and survival in metastatic prostate cancer. N Engl J Med. 2013 Jul 18;369(3):213-23.
  14. 14 - Parker CC, Coleman RE, Sartor O, Vogelzang NJ, Bottomley D, Heinrich D, Helle SI, O'Sullivan JM, Fosså SD, Chodacki A, Wiechno P, Logue J, Seke M, Widmark A, Johannessen DC, Hoskin P, James ND, Solberg A, Syndikus I, Kliment J, Wedel S, Boehmer S, Dall'Oglio M, Franzén L, Bruland ØS, Petrenciuc O, Staudacher K, Li R, Nilsson S. Three-year Safety of Radium-223 Dichloride in Patients with Castration-resistant Prostate Cancer and Symptomatic Bone Metastases from Phase 3 Randomized Alpharadin in Symptomatic Prostate Cancer Trial. Eur Urol. 2018 Mar;73(3):427-435.
  15. 15 - Rathkopf DE, Smith MR, de Bono JS, Logothetis CJ, Shore ND, de Souza P, Fizazi K, Mulders PF, Mainwaring P, Hainsworth JD, Beer TM, North S, Fradet Y, Van Poppel H, Carles J, Flaig TW, Efstathiou E, Yu EY, Higano CS, Taplin ME, Griffin TW, Todd MB, Yu MK, Park YC, Kheoh T, Small EJ, Scher HI, Molina A, Ryan CJ, Saad F. Updated interim efficacy analysis and long-term safety of abiraterone acetate in metastatic castration-resistant prostate cancer patients without prior chemotherapy (COU-AA-302). Eur Urol. 2014 Nov;66(5):815-25.
  16. 16 - Rodriguez-Vida A, Rodríguez-Alonso A, Useros-Rodríguez E, et al. Impact of New Systemic Therapies in Overall Survival in Non-Metastatic Castration Resistant Prostate Cancer: Systematic Review and Meta-Analysis. Clin Genitourin Cancer. 2022;20(2):197.e1-197.e10.
  17. 17 - Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, Fizazi K, Mainwaring P, Piulats JM, Ng S, Carles J, Mulders PF, Basch E, Small EJ, Saad F, Schrijvers D, Van Poppel H, Mukherjee SD, Suttmann H, Gerritsen WR, Flaig TW, George DJ, Yu EY, Efstathiou E, Pantuck A, Winquist E, Higano CS, Taplin ME, Park Y, Kheoh T, Griffin T, Scher HI, Rathkopf DE; COU-AA-302 Investigators. Abiraterone in metastatic prostate cancer without previous chemotherapy. N Engl J Med. 2013 Jan 10;368(2):138-48
  18. 18 - Ryan CJ, Smith MR, Fizazi K, Saad F, Mulders PF, Sternberg CN, Miller K, Logothetis CJ, Shore ND, Small EJ, Carles J, Flaig TW, Taplin ME, Higano CS, de Souza P, de Bono JS, Griffin TW, De Porre P, Yu MK, Park YC, Li J, Kheoh T, Naini V, Molina A, Rathkopf DE; COU-AA-302 Investigators. Abiraterone acetate plus prednisone versus placebo plus prednisone in chemotherapy-naive men with metastatic castration-resistant prostate cancer (COU-AA-302): final overall survival analysis of a randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2015 Feb;16(2):152-60.
  19. 19 - Sartor O, Coleman R, Nilsson S, Heinrich D, Helle SI, O'Sullivan JM, Fosså SD, Chodacki A, Wiechno P, Logue J, Widmark A, Johannessen DC, Hoskin P, James ND, Solberg A, Syndikus I, Vogelzang NJ, O'Bryan-Tear CG, Shan M, Bruland ØS, Parker C. Effect of radium-223 dichloride on symptomatic skeletal events in patients with castration-resistant prostate cancer and bone metastases: results from a phase 3, double-blind, randomised trial. Lancet Oncol. 2014 Jun;15(7):738-46.
  20. 20 - Sartor O, Coleman RE, Nilsson S, Heinrich D, Helle SI, O'Sullivan JM, Vogelzang NJ, Bruland Ø, Kobina S, Wilhelm S, Xu L, Shan M, Kattan MW, Parker C. An exploratory analysis of alkaline phosphatase, lactate dehydrogenase, and prostate-specific antigen dynamics in the phase 3 ALSYMPCA trial with radium-223. Ann Oncol. 2017 May 1;28(5):1090-1097
  21. 21 - Small EJ, Saad F, Chowdhury S, Oudard S, Hadaschik BA, Graff JN, Olmos D, Mainwaring PN, Lee JY, Uemura H, De Porre P, Smith AA, Zhang K, Lopez-Gitlitz A, Smith MR. Apalutamide and overall survival in non-metastatic castration-resistant prostate cancer. Ann Oncol. 2019 Nov 1;30(11):1813-1820.
  22. 22 - Smith MR, Saad F, Chowdhury S, Oudard S, Hadaschik BA, Graff JN, Olmos D, Mainwaring PN, Lee JY, Uemura H, Lopez-Gitlitz A, Trudel GC, Espina BM, Shu Y, Park YC, Rackoff WR, Yu MK, Small EJ; SPARTAN Investigators. Apalutamide Treatment and Metastasis-free Survival in Prostate Cancer. N Engl J Med. 2018 Apr 12;378(15):1408-1418.
  23. 23 - Smith MR, Saad F, Chowdhury S, Oudard S, Hadaschik BA, Graff JN, Olmos D, Mainwaring PN, Lee JY, Uemura H, De Porre P, Smith AA, Brookman-May SD, Li S, Zhang K, Rooney B, Lopez-Gitlitz A, Small EJ. Apalutamide and Overall Survival in Prostate Cancer. Eur Urol. 2021 Jan;79(1):150-158.
  24. 24 - Sopeña Sutil R, Silva Ruiz J, Garcia Gomez B, Romero-Otero J, Garcia-Gonzalez L, Duarte Ojeda JM, de Velasco G, Castellano Gauna D, Rodriguez Antolin A. Seizures and Neuropsychiatric Toxicity in Patients with Non-Metastatic CRPC Treated with New Antiandrogens: Systematic Review and Meta-Analysis. Oncol Res Treat. 2021;44(4):154-163.
  25. 25 - Sternberg CN, Fizazi K, Saad F, Shore ND, De Giorgi U, Penson DF, Ferreira U, Efstathiou E, Madziarska K, Kolinsky MP, Cubero DIG, Noerby B, Zohren F, Lin X, Modelska K, Sugg J, Steinberg J, Hussain M; PROSPER Investigators. Enzalutamide and Survival in Nonmetastatic, Castration-Resistant Prostate Cancer. N Engl J Med. 2020 Jun 4;382(23):2197-2206.
  26. 26 - Tombal B, Sternberg CN, Hussain M, et al. Matching-adjusted indirect treatment comparison of the efficacy of enzalutamide versus apalutamide for the treatment of nonmetastatic castration-resistant prostate cancer. ESMO Open. 2022;7(3):100510.
  27. 27 - Wenzel M, Nocera L, Collà Ruvolo C, et al. Overall survival and adverse events after treatment with darolutamide vs. apalutamide vs. enzalutamide for high-risk non-metastatic castration-resistant prostate cancer: a systematic review and network meta-analysis [published correction appears in Prostate Cancer Prostatic Dis. 2023 Mar 10;:]. Prostate Cancer Prostatic Dis. 2022;25(2):139-148.

1a ARSI nmCRPC

Research question: What is the effect of ARSI + ADT versus ADT alone in patients with non-metastatic castration-resistant prostate cancer (nmCRPC)?

Study reference

Study characteristics

Patient characteristics 

Intervention (I)

Comparison / control (C)

 

Follow-up

Outcome measures and effect size

Comments

ARAMIS

 

Publication:

Fizazi, 2019

Type of study:

RCT

 

Setting and country: 409 centers in 36 countries worldwide

 

Funding and conflicts of interest: Funded by Bayer

HealthCare and Orion Pharma. Authors were funded by the sponsor.

Inclusion criteria:

- ≥ 18 y/o

- castration-resistant prostate cancer (CRPC) (confirmed histology and cytologically)
- baseline PSA >2ng/mm

- PSA doubling <10months

- ECOG 0-1 (out of 5)

- no metastases at baseline radionuclide body scan

- pelvic lymph nodes <cm was allowed

 

Exclusion criteria:

- patients with detectable metastases or history of metastases

- previous seizures or conditions predisposing to seizures

 

N total at baseline:

Intervention: 955

Control: 554

 

Important prognostic factors:

Well balanced because of randomization

Describe intervention (treatment/procedure/test):

Darolutamide 600mg twice daily with food.

 

 

 

Describe control (treatment/procedure/test):

Placebo twice daily with food.

 

Length of follow-up:

17.9 months.

Loss-to-follow-up:

Intervention: no losses to follow up

Control: no losses to follow up.

 

Incomplete outcome data:

Intervention: 1 incomplete outcome data

Reasons: no data of one person on adverse events.

Control: no incomplete outcome data.

 

 

Outcome measures and effect size (include 95%CI and p-value if available):

 

1. Survival

 

Interim analysis


1.1 Overall survival

I: 877/955 (91,8%)
C: 496/554 (89,5%)
HR (95%CI): 0.71 (0.50 to 0.99), P=0.045

 

1.2 Metastases free survival

I: 40.4 months
C: 18.4 months

HR (95%CI): 0.41 (0.34 to 0.50), P<0.001

 

1.3 Progression free survival

I: 33.2 months

C: 7.3 months

HR (95%CI): 0.13 (0.11 to 0.16), P<0.001

 

Long term analysis

 

1.4 Death related to prostate cancer (long term)

Not reported

 

1.5 Overall survival (long term)

Not reported

 

2. Toxicity

 

Interim analysis

 

Adverse event, grade 3-4

I: 236 (24.7)

C: 108 (19.5)

RR (95%CI): 1.31 (1.18 to 1.45)

 

Long term analysis

 

2.2 Long term adverse events

No long term follow up performed.

 

3. Costs

Not reported

 

4. Quality of life

Measured with FACT-P at 16w follow up. Higher scores indicate a better health-related quality of life, ranging from 1–156; a positive difference favours darolutamide.

I: 112.9 (111.8 to 114.0)

C: 111.6 (110.5 to 112.7)

MD (95%CI): 1.3 (0.4 to 2.1)

Minimal important difference: 10.

Authors conclusion: “Clear benefit for darolutamide compared to placebo for metastasis-free survival. No clinically relevant difference in safety data, quality of life outcomes were similar.”

 

 

PROSPER

 

Publications: Hussain, 2018 and Sternberg 2020

Type of study:

RCT

 

Setting and country: 300 sites in 32 countries worldwide

 

Funding and conflicts of interest: Funded by Pfizer and Astellas

Pharma. Authors were funded by the sponsor for editorial assistance. The authors made agreements with the sponsor regarding data confidentiality.

Inclusion criteria:

- ≥ 18 y/o

- CRPC (confirmed pathologically)

- rising PSA despite castration-

associated testosterone levels

- ADT + gonadotropin-releasing

hormone agonist or antagonist or bilateral orchiectomy

- 3 rising PSA levels 1w apart

- baseline PSA >2ng/ml

- PSA doubling time <10months

- Eastern

Cooperative Oncology Group performance-status

score of 0 or 1

 

Exclusion criteria:

- previous metastatic disease on CT or MRI or bone scanning

- suspected brain metastases, active leptomeningeal

disease or history of seizures

 

N total at baseline:

Intervention: 933

Control: 468

 

Important prognostic factors:

Well balanced because of randomization.

 

Describe intervention (treatment/procedure/test):

Enzalutamide (at

a dose of 160 mg) once daily.

Describe intervention (treatment/procedure/test):

Placebo once daily.

Length of follow-up:

23 months (median) until first interim analysis.

 

Loss-to-follow-up:

No losses to follow up reported.

 

Incomplete outcome data:

I: 3 for outcome adverse events

C: 3 for outcome adverse events

Reasons: not reported.

 

Outcome measures and effect size (include 95%CI and p-value if available):

 

1. Survival

 

Interim analysis


1.1 Overall survival

I: 830/933 (89%)

C: 402/468 (87%)

HR (95%CI): 0.80 (0.58 to 1.09), p=0.15

(Hussain, 2018)

 

1.2 Metastases free survival

I: 36.6 months

C: 14.7 months

HR (95%CI): 0.29 (0.24 to 0.35), p<0.001

(Hussain, 2018)

 

1.3 Progression free survival

I: 37.2 months

C: 3.9 months

HR (95%CI): 0.07 (0.05 to 0.08), p<0.001

(Hussain, 2018)

 

Long term analysis

 

1.4 Death related to prostate cancer

I: 178 (19%)

C: 42 (9%)

HR (95%CI): 0.81 (0.72 to 0.91)

(Sternberg, 2021)

 

1.5 Overall survival (long term)

No. of persons still alive

I: 645/933 (69.1%)

C: 290/468 (62.0%)

HR (95%CI):

(Sternberg, 2021)

 

Median months survival.

I: 67.0 months (64.0–NR)

C: 56.3 months (54.4–63.0)

HR (95%CI): 0.73 (0.61 to 0.89)

(Sternberg, 2021)

 

2. Toxicity

 

Interim analysis

 

Adverse event, grade 3-4

I: 292 (31)

C: 109 (23)

RR (95%CI): 1.34 (1.11 to 1.62)

(Hussain, 2018)

 

Long term analysis

 

Adverse event, grade 3-4, long term

I:  446 (48)

C: 126 (27)

RR (95%CI): 1.77 (1.50 to 2.08)

(Sternberg, 2021)

 

3. Costs

Not reported

 

4. Quality of life

“Scores on the Functional Assessment of Cancer Therapy–Prostate (FACT-P) scale range from 0 to 156, with higher scores indicating more

favourable health-related quality of life. Degradation in the FACT-P score was defined as a decrease of at least 10 points from baseline in the

global score for each patient.”

I: 11.1 months until degradation

C: 11.1 months until degradation

RR (95%CI): comparable degradation time

Authors conclusion:

Hussain (2018): Enzalutamide treatment resulted in a significant delay in the development of metastases, and increase in overall survival, without difference in quality of life. Adverse events were more common in the treatment group.

 

Sternberg (2021): Enzalutamide treatment is associated with more adverse events, especially cardiovascular events, although median treatment time was longer (33.9 months vs. 14.2 months) and cardiovascular events are common in the group of men older than 70 years old. Enzalutamide treatment leads to a significantly longer overall survival and adverse rate event is similar to the reported safety profile of the medicine.

SPARTAN

 

Publications: Smith 2018, Small 2019 and Smith 2021

Type of study:

RCT

 

Setting and country: 332 centers in 26 countries worldwide

 

Funding and conflicts of interest: funded by Janssen Research and Development. Authors were funded by the sponsor for editorial assistance. The authors made agreements with the sponsor regarding data confidentiality.

Inclusion criteria:

- ≥ 18 y/o

- CRPC (confirmed histology and cytologically)

- high risk for development of metastasis

- PSA

doubling time <10months during continuous ADT

- continuing ADT during trial

 

Exclusion criteria:

- Distant bone metastases at scan

- Local or regional nodal disease

- Malignant pelvic lymph nodes <2cm from short axis and below aortic bifurcation

 

N total at baseline:

Intervention: 806

Control: 401

 

Important prognostic factors:

Well balanced because of randomization

Describe intervention (treatment/procedure/test):

Apalutamide

(240 mg per day) taken orally daily, until protocol defined progression, adverse events or withdrawal of consent occurred.

Describe  control (treatment/procedure/test):

Placebo (matched) until protocol defined progression, adverse events or withdrawal of consent occurred.

Length of follow-up:

20.3 months (median) until first interim analysis.

 

Loss-to-follow-up:

Intervention: 3

Control: 3

Reasons: 6 patients underwent randomization but never received treatment or placebo.

 

Incomplete outcome data:

Intervention: 3 on outcome adverse events

Control: 3 on outcome adverse events

Reasons: 6 patients underwent randomization but never received treatment or placebo. These patients are not known for the outcome adverse events, but are included in the analysis about survival.

 

Outcome measures and effect size (include 95%CI and p-value if available):

 

1. Survival

 

Interim analysis


1.1 Overall survival

I: 628/806 (78%)

C: 294/401 (73%)

HR (95%CI): 0.76 (0.60 to 0.96), P=0.0197

(Small, 2019)

 

1.2 Metastases free survival

I: 40.5 months

C: 16.2 months

HR (95%CI): 0.27 (0.22 to 0.34), p<0.001

(Smith, 2018)

 

1.3 Progression free survival

I: 40.5 months

C: 14.7 months

HR (95%CI): 0.29 (0.24 to 0.36), p<0.001

(Smith, 2018)

 

Long term analysis

 

1.4 Death related to prostate cancer (long term)

Not reported

 

1.5 Overall survival (long term)

No. of persons still alive

I: 532/806 (66%)

C: 247/401 (62%)

HR (95%): 0.78 (0.64 to 0.96), p = 0.016

(Smith, 2021)

 

No. of months median survival

I: 73.9

C: 59.9

HR (95%): 0.78 (0.64 to 0.96), p = 0.016

(Smith, 2021)

 

2. Toxicity

 

Interim analysis

 

Adverse event, grade 3-4

I: 362 (45.1)

C: 136 (34.2)

RR (95%CI): 1.32 (1.13 to 1.54)

(Smith, 2018)

 

Long term analysis

 

Adverse event, grade 3-4

I:  449 (56)

C: 145 (36)

RR (95%CI): 1.53 (1.33 to 1.77)

(Smith, 2021)

 

3. Costs

Not reported

 

4. Quality of life

Scores on the Functional Assessment of Cancer Therapy–Prostate (FACT-P) questionnaire range from 0 to 156, with higher scores indicating more favourable health-related quality of life.

I: −0.99±0.98

C: −3.29±1.97

HR (95%CI): not estimated. Both groups did not differ clinically significant from baseline. (Smith, 2018)

Authors conclusion:

Smith (2018): Survival was significantly longer with apalutamide than with placebo in patients with nmCRPC.

Small (2019): Apalutamide may prolong survival in patients with high-risk nmCRPC.

Smith (2021): Apalutamide improves overall survival and lengthens time before initiation of cytotoxic chemotherapy in patients with nmCRPC. With longer follow up, safety profile was similar to that shown in earlier studies.

 

1b ARSI mCRPC

Research question: What is the effect of ARSI + ADT versus ADT alone in patients with metastatic castration-resistant prostate cancer (mCRPC)?

Study reference

Study characteristics

Patient characteristics 

Intervention (I)

Comparison / control (C)

 

Follow-up

Outcome measures and effect size

Comments

COU-AA-302

 

Publications: Basch, 2013; Morris, 2015; Rathkopf, 2014; Ryan, 2013 and Ryan, 2015

Type of study: RCT

 

Setting and country: 151 centers in 12 countries

 

Funding and conflicts of interest: data assessors were independent. Source of funding was Ortho Biotech Oncology Research & Development (unit of Cougar Biotechnology).

Inclusion criteria:

- age >18yo,

- metastatic, histologically or cytologically confirmed

adenocarcinoma of the prostate

- previous ADT therapy

- furthermore see article Ryan (2013).

 

Exclusion criteria:

- visceral metastases

- previous therapy with ketoconazole

lasting more than 7 days

 

N total at baseline:

Intervention: 546

Control: 542

 

Important prognostic factors:

Groups were comparable at baseline.

Describe intervention (treatment/procedure/test):

Abiraterone at a dose of 1 g (administered as four 250-mg tablets) + prednisone 5 mg orally twice daily

 

 

 

Describe control (treatment/procedure/test):

four placebo tablets once daily at least 1 hour before and 2 hours after a meal + prednisone 5 mg orally twice daily

 

Length of follow-up:

bone scanning were performed every 8 weeks during the first 24 weeks and every 12 weeks thereafter. Patient-reported outcomes were assessed at baseline and at every visit with the use of the BPISF. FACT-P questionnaires were completed every third visit. The median follow-up duration for all patients was 22.2 months (Ryan, 2013). Long term median follow up of 49.2 months (Ryan, 2015).

 

Loss-to-follow-up:

Intervention, N (%): 4

Reasons (describe): did not receive study drug.

 

Control, N (%): 2

Reasons (describe): did not receive study drug.

These were not included in toxicity analysis, but were included in the survival analysis.

 

Incomplete outcome data:

Intervention, N (%): 0

 

Control, N (%): 0

 

 

Outcome measures and effect size (include 95%CI and p-value if available):

 

1. Survival

 

Interim analysis


1.1 Overall survival

Number of deaths regardless of cause during trial period.

I: 147 deaths

C: 186 deaths

HR (95%CI): 0.75 (0.61-0.93) (Morris, 2015)

 

1.2 Progression free survival

Time from random assignment until first occurrence of progression by bone scan at median follow up of 27 months.

I: 16.5 months

C: 8.3 months

HR (95%CI): 0.53 (0.45-0.62)

(Beer, 2014 and Morris, 2015)

 

Long term analysis

 

1.3 Death related to prostate cancer

Not reported.

 

1.4 Long term survival

Overall death over long term follow up of median 49 months.

I: 352/546 (65%)

C: 387/542 (71%)

HR (95%CI): 0.81 (0.70–0.93)

 

2. Toxicity

 

Interim analysis

 

Adverse event, grade 3-4

I: 258 (48)

C: 225 (42)

RR (95%CI): NR

 

Long term analysis

Adverse event, grade 3-4

I: 290 (54%)

C: 236 (44%)

RR (95%CI): NR

 

3. Costs

Not reported

 

4. Quality of life

Number of months until a decline of >10 points occurred in FACT-P score

I: 12.7 mo.

C: 8.3 mo.

HR (95%CI): 0.78 (0.66–0.92)

Authors’ conclusion: “In summary, the results show benefit from the use of abiraterone in patients with asymptomatic or mildly symptomatic metastatic castration-resistant prostate cancer who have not received previous chemotherapy. These findings include increased rates of radiographic progression-free survival and overall survival, as well as clinically meaningful secondary end points, such as delays in the use of opiates for pain and chemotherapy and patient-reported outcomes related to health-related quality of life.”

 

PREVAIL

 

Publications:

Beer, 2014 and Loriot, 2015

Type of study: RCT

 

Setting and country: 180 centers worldwide (exact locations not mentioned)

 

Funding and conflicts of interest: Funded by Medivation and Astellas Pharma. Disclosures found in the Appendix. “Dr. Armstrong reports grant support from Medivation/Astellas, Janssen, Sanofi Aventis, Dendreon, and Bayer during the conduct of the study, and personal fees from Medivation/Astellas, Janssen, Dendreon, Sanofi Aventis, and Bayer outside the submitted work.”

 

Inclusion criteria:

- histologically confirmed adenocarcinoma of the prostate

- documented metastases and PSA progression

- despite receiving LHRH therapy or orchiectomy with serum level testosterone (<1.73 nmol/L) or less.

- Concomitant ADT.

- Additional inclusion criteria in article of Beer (2014).

 

Exclusion criteria:

- cytotoxic chemotherapy before

- visceral metastases

- heart failure

N total at baseline:

Intervention: 832

Control: 801

 

Important prognostic factors:

Groups were comparable at baseline.

Describe intervention (treatment/procedure/test):

oral enzalutamide (at a dose of 160 mg) once daily

 

 

 

Describe  control (treatment/procedure/test):

placebo once daily with or without food

 

Length of follow-up:

Imaging was performed at the time of screening, at weeks 9, 17, and 25, and every 12 weeks thereafter. Total follow up was median 12 months for radiographic progression and median 22 months for overall survival outcomes.

 

Loss-to-follow-up:

Intervention, N (%): 1

Reasons (describe): lost to follow up.

 

Control, N (%): 0

 

Incomplete outcome data:

Intervention, N (%): 0

Reasons (describe): all participants except for one lost to follow in the intervention group were included in the safety (toxicity) analysis.

 

Control, N (%): 0

Reasons (describe): all participants were included in safety analysis.

 

 

Outcome measures and effect size (include 95%CI and p-value if available):

 

1. Survival

 

Interim analysis


1.1 Overall survival

Number of deaths regardless of cause during trial period.

I: 241/872 (28%)

C: 299/845 (35%)

HR (95%CI): 0.71 (0.60-0.84)

 

1.2 Progression free survival

Rate of radiographic progression at 12 months follow up.

I: 65%

C: 14%

HR (95%CI): 0.19 (0.15-0.23)

 

Long term analysis

No long term follow up performed.

 

2. Toxicity

 

Interim analysis

 

Adverse event, grade 3-4

I: 374 (43)

C: 313 (37)

RR (95%CI) 1.08 (0.93-1.25)

 

Long term analysis

No long term follow up performed.

 

3. Costs

Not reported.

 

4. Quality of life

Number of months until decline of 10 points or more occurred on FACT-P total score.

I: 11.3 mo.

C: 5.6 mo.

HR (95%CI): 0.63, confidence interval not reported. P<0.001

Authors’ conclusion: “In conclusion, in men with minimally symptomatic or asymptomatic metastatic prostate cancer who had not received chemotherapy, enzalutamide, an oral therapy with an excellent side-effect profile, significantly delayed radiographic disease progression or death, the need for cytotoxic chemotherapy, and the deterioration in quality of life and significantly improved overall survival.”

 

 

2 Chemotherapy

No evidence tables.

 

3 Radioligand therapy
Research question: What is the effect of radioligand therapy + ADT versus ADT alone in patients with metastatic castration-resistant prostate cancer (mCRPC)?

 

Study reference

Study characteristics

Patient characteristics

Intervention (I)

Comparison / control (C)

 

Follow-up

Outcome measures and effect size

Comments

ALSYMPCA

 

Publications: Hoskin, 2014; Nome, 2015; Parker, 2013 and Parker, 2016 & Sartor, 2014 and Sartor, 2017.

Type of study: RCT

 

Setting and country: 136 study centers in 19 countries

 

Funding and conflicts of interest: Funded by Algeta and Bayer HealthCare Pharmaceuticals. Conflicts of interest and disclosure forms are attached to online version of the article (Parker, 2013). No potential conflicts of interest relevant to this article.

Inclusion criteria:

- histologically confirmed, progressive

castration-resistant prostate cancer

- with two or

more bone metastases detected on skeletal scintigraphy

- no known visceral metastases

- receiving the best standard of care

- had received

docetaxel

- more see article Parker (2013).

 

Exclusion criteria:

- received

chemotherapy within the previous 4 weeks

- had

not recovered from adverse events due to chemotherapy.

- more see Parker (2013)

 

N total at baseline:

Intervention: 614

Control: 307

 

Important prognostic factors:

Groups comparable at baseline (see Parker, 2013)

 

Describe intervention (treatment/procedure/test):

six intravenous injections of radium-223 (at a dose of 50 kBq per kilogram of body weight), administered every four weeks. Randomized 2:1 ratio.

 

 

 

Describe control (treatment/procedure/test):

A matching placebo injection, six injections administered every four weeks.

 

Length of follow-up:

3 years

 

Loss-to-follow-up:

Intervention, N (%): 13 (2)

Reasons (describe): withdrawal before first injection, not included in safety (toxicity) analysis

 

Control, N (%): 5 (2)

Reasons (describe): withdrawal before first injection, not included in safety (toxicity) analysis

 

Incomplete outcome data:

Intervention, N (%): 2 (0.2)

Reasons (describe): “Two patients received no treatment and had missing dates of withdrawal.”

 

Control, N (%): 0

 

 

Outcome measures and effect size (include 95%CI and p-value if available):

 

1. Survival

 

Interim analysis


1.1 Overall survival

The time from randomization until the date of death, regardless of the cause.

I: 14.9 mo.

C: 11.3 mo.

HR (95%CI): 0.70 (0.58-0.83)

 

1.2 Progression free survival

Time since randomization until first skeletal event.

I: 15.6 mo.

C: 9.8 mo.

HR (95%CI): 0.66 (0.52-0.83)

 

Long term analysis

 

1.3 Death related to prostate cancer

Not reported.

 

1.4 Overall survival long term

Not reported.

 

2. Toxicity

 

Interim analysis

 

Adverse event, grade 3-4

I: 339 (56)

C: 188 (62)

RR (95%CI): 0.90 (0.81-1.01)

 

Long term analysis

 

Adverse event, grade 3-4

I: 350 (58)

C: 194 (65)

RR (95%CI): 0.91 (0.81-1.01)

 

3. Costs

Not reported.

 

4. Quality of life

Mean reduction on FACT-P score (0-156 with higher scores indicating better QoL) from baseline to 16 week follow up.

I: -2.7

C: -6.8

HR (95%CI): not reported, p<0.001

Authors’ conclusion: “This updated final long-term safety follow-up ALSYMPCA

analysis shows that radium-223 is well tolerated in CRPC

patients with symptomatic bone metastases, with minimal

nonhematologic AEs, a low incidence of myelosuppression

with long-term preservation of hematopoietic function, and no new safety signals.” (Parker, 2018)

 

Risk of bias tables

 

1a ARSI nmCRPC

 

Research question: What is the effect of ARSI + ADT versus ADT alone in patients with non-metastatic castration-resistant prostate cancer (nmCRPC)?

Study reference

 

(first author, publication year)

Was the allocation sequence adequately generated?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Was the allocation adequately concealed?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Blinding: Was knowledge of the allocated

interventions adequately prevented?

 

Were patients blinded?

 

Were healthcare providers blinded?

 

Were data collectors blinded?

 

Were outcome assessors blinded?

 

Were data analysts blinded?

Was loss to follow-up (missing outcome data) infrequent?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Are reports of the study free of selective outcome reporting?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Was the study apparently free of other problems that could put it at a risk of bias?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Overall risk of bias

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

ARAMIS

Publication:

Fizazi, 2019

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Low risk

 

LOW

 

SPARTAN

Publications: Smith 2018, Small 2019 and Smith 2021

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Low risk

 

LOW

 

PROSPER

Publications: Hussain, 2018 and Sternberg 2020

Low risk

 

Unclear risk

Low risk

 

Low risk

 

Low risk

 

Low risk

 

LOW

 

Data derives from systematic review and meta-analysis by Sopeña Sutil e.a. (2021)

 

1b ARSI mCRPC

 

Research question: What is the effect of ARSI + ADT versus ADT alone in patients with metastatic castration-resistant prostate cancer (mCRPC)?

Study reference

 

(first author, publication year)

Was the allocation sequence adequately generated?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Was the allocation adequately concealed?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Blinding: Was knowledge of the allocated

interventions adequately prevented?

 

Were patients blinded?

 

Were healthcare providers blinded?

 

Were data collectors blinded?

 

Were outcome assessors blinded?

 

Were data analysts blinded?

Was loss to follow-up (missing outcome data) infrequent?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Are reports of the study free of selective outcome reporting?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Was the study apparently free of other problems that could put it at a risk of bias?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Overall risk of bias

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

COU-AA-302

Publications: Basch, 2013; Morris, 2015; Rathkopf, 2014; Ryan, 2013 and Ryan, 2015

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Low risk

 

LOW

 

PREVAIL

Publications:

Beer, 2014 and Loriot, 2015

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Low risk

 

LOW

 

N.B. Also based on previous module’s assessment in 2016

 

2 Chemotherapy

No  risk of bias tables.

 

3 Radioligand therapy

 

Research question: What is the effect of radioligand therapy + ADT versus ADT alone in patients with metastatic castration-resistant prostate cancer (mCRPC)?

Study reference

 

(first author, publication year)

Was the allocation sequence adequately generated?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Was the allocation adequately concealed?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Blinding: Was knowledge of the allocated

interventions adequately prevented?

 

Were patients blinded?

 

Were healthcare providers blinded?

 

Were data collectors blinded?

 

Were outcome assessors blinded?

 

Were data analysts blinded?

Was loss to follow-up (missing outcome data) infrequent?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Are reports of the study free of selective outcome reporting?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Was the study apparently free of other problems that could put it at a risk of bias?

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

Overall risk of bias

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

 

ALSYMPCA

Publications: Hoskin, 2014; Nome, 2015; Parker, 2013 and Parker, 2016 & Sartor, 2014 and Sartor, 2017

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Low risk

 

Unclear reporting, results are not reported in table of supplementary table, only in text and the total number of included patients per group for this outcome measure was not reported.

LOW

 

N.B. Also based on previous module’s assessment in 2016

 

Table of excluded studies

1. ARSI

Reference

Reason for exclusion

Chi, 2015

Wrong population, patients progressing after abiraterone or enzalutamide.

Chowdhury, 2020

Wrong analysis, matching-adjusted indirect comparison (MAIC) by weighting patients from SPARTAN study to baseline characteristics of PROSPER.

de Bono, 2011 (CIE-BOM literature)

Wrong population, patients progressing after chemotherapy. N.B. COU-AA-301 trial.

Fang, 2017

Wrong comparison, abiraterone and enzalutamide were compared.

Gracia, 2016

Wrong comparison; comparison of hypertension, neurological and psychiatric adverse events on enzalutamide and abiraterone acetate plus prednisone. Only abstract available.

Kang, 2017

Wrong comparison, no control group with ADT, but with placebo or prednisone?

Kumar, 2020

A more recent meta-analysis and more extensive publication is available by Hird (2020).

Meunier, 2021

Wrong population, black population.

Mori, 2020

A more recent version of this meta-analysis is available by Wenzel (2021), however the study by Wenzel did not consider different follow up times. Therefore, the meta-analysis of Hird (2020) was chosen.

Roviello, 2016 "Role of the novel…"

More recent SRs are available. This study uses the trials: AFFIRM, COU-AA-301, COU-AA-302, ELM-PC 4, ELM-PC 5, PREVAIL. The study included both patients naive to chemotherapy and patients progressing after chemotherapy. Subgroup analyses are unclear.

Roviello, 2016 "Incidence and relative.."

Wrong comparison, CYP-17 inhibitors and prednisone versus placebo and prednisone

Roviello, 2020

More recent studies and SRs available. Study uses the JADAD-5-item scale for assessment, while others use the Cochrane tool or GRADE assessment. The more extensive meta-analysis of Hird (2020) was chosen.

Scher, 2012 (CIE-BOM literature)

Wrong population, patients progressing after chemotherapy. N.B. AFFIRM trial.

Ternov, 2021

Wrong comparison, enzalutamide versus AAP for mCRPC.

Wang, 2021

Wrong population, men with mCRPC in real world practice, inclusion of cohort studies with patients with mCRPC. Comparison abiraterone versus enzalutamide

Wenzel, 2021

Wrong analysis. The study is an extension of Mori (2020) and included the most recent publications of SPARTAN and PROSPER, however the study did not analyse for different follow up times between studies. The goal of the study was to compare the three medicines.

Zhang, 2017

Wrong comparison, indirect comparison between abiraterone acetate and enzalutamide

 

2. Chemotherapy

Reference

Reason for exclusion

Berthold, 2008

Wrong comparison: no control group without chemotherapy or ADT alone

de Bono, 2010

Wrong comparison: no control group without chemotherapy, patients progressing after docetaxel treatment

de Bono, 2020

Wrong comparison: no control group without chemotherapy, patients progressing after enzalutamide or abiraterone treatment

Leal, 2019

Wrong comparison: only two studies from 1995/1996 with control arm endocrine therapy only

Petrylak, 2004

Wrong comparison: no control group without chemotherapy or ADT alone

Song, 2018

Wrong comparison: no control arm with ADT only

Tannock, 2004

Wrong comparison: no control group without chemotherapy or ADT alone

2. Radioligand therapy

Reference

Reason for exclusion

de Bono, 2016 (CIE-BOM)

Wrong population; patients progressing after chemotherapy. N.B. VISION trial.

Dizdarevic, 2020

20 articles about prostate cancer (of which many articles about 1 RCT (ALSYMPCA) and the others about phase I and II studies)

McGann, 2015

 

Nilsson, 2007

Wrong population; patients progressing after chemotherapy. N.B. Included in module of 2016.

Nilsson, 2013

Wrong population; patients progressing after chemotherapy. N.B. Included in module of 2016.

Sadaghiani, 2021

Does not comply to PICO

Satapathy, 2021

 

Sartor, 2021 (CIE-BOM)

Wrong population; patients progressing after chemotherapy. N.B. VISION trial.

Terrisse, 2020

Control group no ADT. Reports overall survival and symptomatic skeletal event-free survival for alpha-emitting and beta-emitting radio-isotopes.

von Eyben, 2020

No control group with ADT? No RCTs?

Autorisatiedatum en geldigheid

Laatst beoordeeld  : 22-12-2023

Laatst geautoriseerd  : 22-12-2023

Geplande herbeoordeling  :

Bij het opstellen van de module heeft de werkgroep een inschatting gemaakt over de maximale termijn waarop herbeoordeling moet plaatsvinden en eventuele aandachtspunten geformuleerd die van belang zijn bij een toekomstige herziening (update). De geldigheid van de richtlijnmodule komt eerder te vervallen indien nieuwe ontwikkelingen aanleiding zijn een herzieningstraject te starten.

Initiatief en autorisatie

Initiatief:
  • Nederlandse Vereniging voor Urologie
Geautoriseerd door:
  • Nederlandse Internisten Vereniging
  • Nederlandse Vereniging voor Urologie
  • ProstaatKankerStichting

Algemene gegevens

De richtlijnontwikkeling werd ondersteund door het Kennisinstituut van de Federatie Medisch Specialisten (www.kennisinstituut.nl) en werd gefinancierd uit de Stichting Kwaliteitsgelden Medisch Specialisten (SKMS). Patiënten participatie bij deze richtlijn werd medegefinancierd uit de Stichting Kwaliteitsgelden Patiënten Consumenten (SKPC) binnen het programma KIDZ. De financier heeft geen enkele invloed gehad op de inhoud van de richtlijn.

Samenstelling werkgroep

Werkgroep

  • Prof. dr. R.J.A. van Moorselaar, uroloog, werkzaam in het Amsterdam Universitair Medische Centra, locatie VUmc, NVU, voorzitter
  • Prof. dr. J.O. Barentsz, radioloog, werkzaam in het RadboudUMC, NVvR
  • Drs. R.J. van Alphen, internist-oncoloog werkzaam in het ETZ
  • Drs. R. van der Giessen, patiënten vertegenwoordiger, Prostaatkankerstichting
  • Prof. dr. L. Incrocci, radiotherapeut-oncoloog, werkzaam in het Erasmus MC, NVRO
  • Dr. M.J.R. Janssen, nucleair geneeskundige, werkzaam in het RadboudUMC, NVvN
  • Dr. G.J.L.H. van Leenders, patholoog, werkzaam in het Erasmus Medisch Centrum, NVvP
  • Dr. I.M. van Oort, uroloog, werkzaam bij het RadboudUMC, NVU
  • Dr. I. Schoots, radioloog, werkzaam bij het Erasmus MC, NVvR
  • Dr. D.M. Somford, uroloog, werkzaam bij CWZ Nijmegen, NVU
  • Dr. Y. Reisman, uroloog, seksuoloog NVVS, Flare-Health, Amstelveen, NVVS
  • Drs. I. Zantingh, gz-psycholoog – seksuoloog NVVS, werkzaam bij Antoni van Leeuwenhoek, NVVS
  • C.N. Tillier, verpleegkundig specialist urologie, werkzaam bij Antoni van Leeuwenhoek, V&VN
  • Drs. H.A.M. Vanhauten, werkzaam bij het UMCG, radiotherapeut-oncoloog. NVRO
  • Dr. E. Vegt, nucleair geneeskundige, werkzaam in het Erasmus MC, NVNG
  • Dr. M. Tascilar, internist-oncoloog, werkzaam in Isala, NIV
  • Dr. A. Bergman, internist-oncoloog werkzaam bij Antoni van Leeuwenhoek, NIV

Met ondersteuning van:

  • Dr. I. Mostovaya, senior adviseur, Kennisinstituut van de Federatie Medisch Specialisten
  • Dr. J. Boschman, senior adviseur, Kennisinstituut van de Federatie Medisch Specialisten
  • Dr. M. van Son, adviseur, Kennisinstituut van de Federatie Medisch Specialisten

Belangenverklaringen

De KNMG-code ter voorkoming van oneigenlijke beïnvloeding door belangenverstrengeling is gevolgd. Alle werkgroepleden hebben schriftelijk verklaard of zij in de laatste drie jaar directe financiële belangen (betrekking bij een commercieel bedrijf, persoonlijke financiële belangen, onderzoeksfinanciering) of indirecte belangen (persoonlijke relaties, reputatiemanagement, kennisvalorisatie) hebben gehad. Een overzicht van de belangen van werkgroepleden en het oordeel over het omgaan met eventuele belangen vindt u in onderstaande tabel. De ondertekende belangenverklaringen zijn op te vragen bij het secretariaat van het Kennisinstituut van de Federatie Medisch Specialisten.

 

Werkgroeplid

Functie

Nevenfuncties

Gemelde belangen

Ondernomen actie

* Van Moorselaar

Uroloog, Amsterdam UMC

Advisory board: Astellas, AstraZeneca Bayer, Janssen, Sanofi-Genzyme: Betaald

Incoming president European Board of Urology: onbetaald

Board member European School of Urology: onbetaald

geen

Geen actie.

In transparantieregister diverse keren honoraria genoemd voor Astellas, AstraZeneca Bayer, Janssen, Sanofi-Genzyme (2018-2020). Bij herziening RL 2017 zat Jeroen ook al in deze adviescommisies (alleen AstraZeneca is erbij gekomen)

Reisman

Directeur en Seksuoloog Flare-Health, Amstelveen (0.5 fte)

Hoofdopleider Seksuologie, RINO Amsterdam (zzp contract)

Past-President European Society for Sexual Medicine (ESSM) - deelnemen aan het bestuur

 

Bestuurlid en Past-President Multidisciplinary Joint Committee for Sexual Medicine of the European Union for Medical Specialists (UEMS)

 

Co-director ESSM School and Advanced Course of Sexual Medicine

 

Voorzitter network workinggroup HPV information and education van European Cancer Organisation (ECCO)

 

Honorary Professor of Urology, Federal State Institute of Urology, Moscow

 

Visiting Professor of Andrology, Shanghai Jiao Tong University, China

 

Allemaal vrijwillig

Adviseur Besins Health Care

Spreker: Lundbeck, Lilly, Coloplast en Berlin-Chemie

 

Expertise op gebied van Onco-Seksuologie

Editor "Cancer, Intimacy and Sexuality"

Geen actie.

Tillier

Verpleegkundig Specialist Urologie NKI-AVL, Amsterdam

EAUN Chair Elect (onbetaald)

Consultant IPSEN bij het schrijven van informiatie boekje voor patienten over hormonale behandeling bij prostaatkanker

Geen.

Geen actie.

Schoots

Radioloog, Radiologie & Nucleair Geneeskunde, Erasmus MC

(0.8 fte betaald)

Research appointment, Radiologie, AVL-NKI (0.2 fte betaald)

Advies functie - Quantib NV: radiologische software als hulp voor radiologische beoordeling van prostaat MRI - (betaald - aan werkgever Erasmus MC)

Geen actie. De UV's die we updaten hebben geen betrekking op de prostaat MRI.

Van der Giessen

Lid Kwaliteits Groep Prostaatkanker Stichting (vrijwilliger)

Lid Overleggroep NWO (Erasmus- vrijwilliger))

Geen.

Geen.

Geen actie.

Vegt

Nucleair geneeskundige - Erasmus MC Rotterdam

Geen.

Deelname aan de studie "ROTOR registry'' naar uitkomsten van Ra-223-therapie bj prostaatcarnicoom, gefinancieerd door Bayer.

ROTOR registry heeft mogelijk enige betrekking op UV1 hormoongevoelig prostaatcarcinoom. Werkgroeplid is niet als trekker/meelezer betrokken bij deze UV.

Van Oort

Oncologisch Uroloog, Radboudumc Nijmegen

Geen.

adviseurschap bij

Astellas, Sanofi, Janssen, Roche, Bayer

 

gefinancieerd onderzoek door: Astellas, Janssen, Bayer

 

Radboudumc heeft de SelectMdx urinetest ontwikkeld, eigendom ligt nu extern

Geen actie.

In transparantieregister diverse keren honoraria genoemd voor Astellas, Bayer, Janssen, Sanofi-Genzyme (2018-2020). Bij herziening RL 2017 zat Inge ook in deze adviescommisies. Onderzoek gefinancierd door de industrie (was bij 2017 werkgroep ook zo).

Incrocci

Radiotherapeut-oncoloog, Erasmus MC, Rotterdam

Geen.

Geen.

Geen actie.

Barentsz

Hoogleraar Radiologie Radboudumc

Geen.

onbetaald adviseur van SPL Medical en Soteria Medical

Geen actie.

Bergman

Internist-Oncoloog, Nederlands Kanker Instituut, Antoni van Leeuwenhoek

'Bezoldigde sprekers/organisatie vergoedingen van Bayer, Astellas, Sanofi

Participatie Advisory boards: Astelas, Jansen, Bayer, Sanofi.

 

Financiering Investigator Initiated Studies door: Astellas, Sanofi, Bayer, Amgen

Participatie industry sponsored Studies van: Merck, Astellas, Jansen, Bayer, Astra Zeneca.

 

Bestuurslid Dutch Uro Oncology Study Group.

Geen actie.

Vergoedingen industrie en onderzoek gefinancierd door industrie

Vanhauten

Radiotherapeut oncoloog UMCG

Bestuurslid Prostaatcentrum Noord-Nederland, onbetaald.

Enkele malen per jaar lezing/panellid Astellas, Jansen, Prevents waarvoor sprekersvergoeding.

Geen.

Geen actie.

Tascilar

Internist-Oncoloog

Commissie COM (NVMO), aanvragen van off label indicaties voor oncologische middelen, vacatiegelden

Lid cieBOM, vacatiegelden

Bestuurslid DRCG, onbetaald

lid Raad van Advies Prostaatkankerstichting, onbetaald

DUOS werkgroep gemetastaseerd prostaatcarcinoom, onbetaald

Geen.

Geen actie.

Mensink

VrÍjwilliger, Belangenbehartiger Kwliteit van Zorg bij Prostaatkankerstichting

Geen.

Geen.

Geen actie.

Janssen

nucleair geneeskundige Radboudumc

Voorzitter onderwijscommissie NVNG (onbetaald)

lid onderwijscommissie NVvR (onbetaald

lid commissie wetenschappelijke ontmoetingen NVNG

docent landelijk differentiantenonderwijs nucleaire radiologie (onkostenvergoeding)

PROPER-ABX studie, vergelijkende studie waarin Axumin en PSMA-1007 woren vergeleken qua diagnostische waarbij bij patienten met biochemische recidief prostaatcarcinoom, gefinancieerd door ABX.

Geen actie. De UV's die we updaten hebben geen betrekking op Axumin en PSMA-1007.

Van Alphen

internist oncoloog bij ETZ te Tilburg fulltime

bestuurslid NVMO 3 uur per week (betaald aan het MSB)

Bestuurslid NVMO vacatie gelden 3 uur per week aan MSB

NIV platform kwaliteit 4 avonden per jaar vacatie -> MSB

Richtlijnen commissie NVMO 2 uur per maand vacatie -> MSB

Richtlijnen commissie NIV 1 uur per maand vacatie -> MSB

Commissie kwaliteit NVMO: 1 uur per maand vacatie MSB

Inval vanuit NIV voor module hormoonsensitief prostaat carcinoom.

ja, Studies met Pfizer, Bayer, Astellas, eigenlijk alle firma's die met prostaatkanker werken.

Ik ken geen oncoloog in een groter perifeer ziekenhuis of academie die hier nee kan invullen. Hooguit neutraal als je voor alle firma's studies hebt lopen.

Geen actie???

Zantingh

gz-psycholoog / seksuoloog NVVS

Antoni van Leeuwenhoek

Centrum kwaliteit van leven

 

docent cursus Seksuologie in de GZ opleiding

RINO Amsterdam

 

eigenaar / praktijkhouder

Seksuologie Praktijk Utrecht

onbetaald

lid mediacommissie NVVS

 

onbetaald

voorzitter NVVS Special Interest Group onco-seksuologie

Geen.

Geen actie.

Van Leenders

Patholoog, Erasmus MC, Rotterdam

Bestuurslid Nederlandse Expertgroep Urologische Pathologie (NEUP), European Network of Uropathology (ENUP), International Society of Urological Pathology (ISUP)

 

Wetenschappelijke raad ProstaatKankerStichting (PKS) en stichting Egidius

 

Lidmaatschap EAU guideline committee prostate cancer, en International Collaboration on Cancer Reporting (ICCR) prostate cancer

Geen

Geen actie.

Somford

Uroloog CWZ  en Prosper (1.0 FTE)

 

Uroloog Radboudumc (detachering)

Voorzitter Werkgroep Oncologische Urologie (WOU) van de NVU

 

Voorzitter Clinical Audit Board (CAB) Multidisciplinaire Kwaliteitstregistratie Prostaakanker (DICA)

 

Voorzitter Lokale Toetsingscommissie CWZ

 

Lid Wetenschappelijke Commissie (NVU)

 

Associate Editor Frontiers in Oncology

Research Grants (CWZ): KWF, Besins Health Care

 

Contracted research (CWZ): Janssen, Eli Lilly, Astellas, Blue Earth Diagnostics, Bayer, SPL Medical, QED Therapeutics

 

Advisory Boards:

Astellas, Janssen, MSD, Bayer

 

Consultancy:

Patient+

 

Lectures:

Bayer, Janssen

 

Geen actie. Research grants hebben gene betrekking op herziene uitgangsvragen.

Inbreng patiëntenperspectief

Er werd aandacht besteed aan het patiëntenperspectief door een afgevaardigde van een patiëntenvereniging, de ProstaatKankerStichting, in de werkgroep te laten participeren.

Implementatie

In de verschillende fasen van het ontwikkelproces is rekening gehouden met de implementatie van de richtlijnmodule en de praktische uitvoerbaarheid van de aanbevelingen. Daarbij is uitdrukkelijk gelet op factoren die de invoering van de module in de praktijk kunnen bevorderen of belemmeren. De implementatietabel is te vinden bij de aanverwante producten.

Werkwijze

AGREE

Deze module is opgesteld conform de eisen vermeld in het rapport Medisch Specialistische Richtlijnen 2.0 van de adviescommissie Richtlijnen van de Raad Kwaliteit. Dit rapport is gebaseerd op het AGREE II instrument (Appraisal of Guidelines for Research & Evaluation II; Brouwers, 2010), dat een internationaal breed geaccepteerd instrument is. Voor een stap-voor-stap beschrijving hoe een evidence-based module tot stand komt wordt verwezen naar het stappenplan Ontwikkeling van Medisch Specialistische Richtlijnen van het Kennisinstituut van de Federatie Medisch Specialisten.

 

Knelpuntenanalyse

Uit de inventarisatie van de knelpunten door werkgroep/commissie Prostaatcarcinoom van de NVU (2017) bleek dat er een noodzaak was voor (revisie) van deze richtlijnmodule.

 

Uitgangsvraag en uitkomstmaten

Op basis van de uitkomsten van de knelpuntenanalyse is door de werkgroepleden en de adviseur een uitgangsvraag opgesteld. Vervolgens inventariseerde de werkgroep welke uitkomstmaten voor de patiënt relevant zijn, waarbij zowel naar gewenste als ongewenste effecten werd gekeken. De werkgroep waardeerde deze uitkomstmaten volgens hun relatieve belang bij de besluitvorming rondom aanbevelingen, als kritiek, belangrijk (maar niet kritiek) en onbelangrijk. Tevens definieerde de werkgroep tenminste voor de kritieke uitkomstmaten welke verschillen zij klinisch (patiënt) relevant vonden.

 

Strategie voor zoeken en selecteren van literatuur

Aan de hand van specifieke zoektermen werd gezocht naar gepubliceerde wetenschappelijke studies in (verschillende) elektronische databases. Tevens werd aanvullend gezocht naar studies aan de hand van de literatuurlijsten van de geselecteerde artikelen. In eerste instantie werd gezocht naar studies met de hoogste mate van bewijs. De werkgroepleden selecteerden de via de zoekactie gevonden artikelen op basis van vooraf opgestelde selectiecriteria. De geselecteerde artikelen werden gebruikt om de uitgangsvraag te beantwoorden. De geselecteerde databases waarin is gezocht en de gehanteerde selectiecriteria zijn te vinden in de module met desbetreffende uitgangsvraag. De zoekstrategie is opvraagbaar bij de Richtlijnendatabase, zie het tabblad Zoekverantwoording voor verdere details.

 

Kwaliteitsbeoordeling individuele studies

Individuele studies werden systematisch beoordeeld, op basis van op voorhand opgestelde methodologische kwaliteitscriteria, om zo het risico op vertekende studieresultaten (risk of bias) te kunnen inschatten. Deze beoordelingen kunt u vinden in de Risk of Bias (RoB) tabellen. De gebruikte RoB instrumenten zijn gevalideerde instrumenten die worden aanbevolen door de Cochrane Collaboration:

•          AMSTAR – voor systematische reviews.

 

Samenvatten van de literatuur

De relevante onderzoeksgegevens van alle geselecteerde artikelen werden overzichtelijk weergegeven in evidence-tabellen. De belangrijkste bevindingen uit de literatuur werden beschreven in de samenvatting van de literatuur. Indien van toepassing: Bij een voldoende aantal studies en overeenkomstigheid (homogeniteit) tussen de studies werden de gegevens ook kwantitatief samengevat (meta-analyse) met behulp van Review Manager 5.

 

Beoordelen van de kracht van het wetenschappelijke bewijs

De kracht van het wetenschappelijke bewijs werd bepaald volgens de GRADE-methode. GRADE staat voor: Grading Recommendations Assessment, Development and Evaluation (zie http://www.gradeworkinggroup.org/).

 

GRADE onderscheidt vier gradaties voor de kwaliteit van het wetenschappelijk bewijs: hoog, redelijk, laag en zeer laag. Deze gradaties verwijzen naar de mate van zekerheid die er bestaat over de literatuurconclusie (Schünemann, 2013).

 

GRADE

Definitie

Hoog

  • er is hoge zekerheid dat het ware effect van behandeling dichtbij het geschatte effect van behandeling ligt zoals vermeld in de literatuurconclusie;
  • het is zeer onwaarschijnlijk dat de literatuurconclusie verandert wanneer er  resultaten van nieuw grootschalig onderzoek aan de literatuuranalyse worden toegevoegd.

Redelijk

  • er is redelijke zekerheid dat het ware effect van behandeling dichtbij het geschatte effect van behandeling ligt zoals vermeld in de literatuurconclusie;
  • het is mogelijk dat de conclusie verandert wanneer er resultaten van nieuw grootschalig onderzoek aan de literatuuranalyse worden toegevoegd.

Laag

  • er is lage zekerheid dat het ware effect van behandeling dichtbij het geschatte effect van behandeling ligt zoals vermeld in de literatuurconclusie;
  • er is een reële kans dat de conclusie verandert wanneer er resultaten van nieuw grootschalig onderzoek aan de literatuuranalyse worden toegevoegd.

Zeer laag

  • er is zeer lage zekerheid dat het ware effect van behandeling dichtbij het geschatte effect van behandeling ligt zoals vermeld in de literatuurconclusie;
  • de literatuurconclusie is zeer onzeker.

 

Formuleren van de conclusies

Voor elke relevante uitkomstmaat werd het wetenschappelijk bewijs samengevat in een of meerdere literatuurconclusies waarbij het niveau van bewijs werd bepaald volgens de GRADE-methodiek. De werkgroepleden maakten de balans op van elke interventie (overall conclusie). Bij het opmaken van de balans werden de gunstige en ongunstige effecten voor de patiënt afgewogen. De overall bewijskracht wordt bepaald door de laagste bewijskracht gevonden bij een van de kritieke uitkomstmaten. Bij complexe besluitvorming waarin naast de conclusies uit de systematische literatuuranalyse vele aanvullende argumenten (overwegingen) een rol spelen, werd afgezien van een overall conclusie. In dat geval werden de gunstige en ongunstige effecten van de interventies samen met alle aanvullende argumenten gewogen onder het kopje Overwegingen.

 

Overwegingen (van bewijs naar aanbeveling)

Om te komen tot een aanbeveling zijn naast (de kwaliteit van) het wetenschappelijke bewijs ook andere aspecten belangrijk en worden meegewogen, zoals de expertise van de werkgroepleden, de waarden en voorkeuren van de patiënt, kosten, beschikbaarheid van voorzieningen en organisatorische zaken. Deze aspecten worden, voor zover geen onderdeel van de literatuursamenvatting, vermeld en beoordeeld (gewogen) onder het kopje Overwegingen.

 

Formuleren van aanbevelingen

De aanbevelingen geven antwoord op de uitgangsvraag en zijn gebaseerd op het beschikbare wetenschappelijke bewijs en de belangrijkste overwegingen, en een weging van de gunstige en ongunstige effecten van de relevante interventies. De kracht van het wetenschappelijk bewijs en het gewicht dat door de werkgroep wordt toegekend aan de overwegingen, bepalen samen de sterkte van de aanbeveling. Conform de GRADE-methodiek sluit een lage bewijskracht van conclusies in de systematische literatuuranalyse een sterke aanbeveling niet a priori uit, en zijn bij een hoge bewijskracht ook zwakke aanbevelingen mogelijk. De sterkte van de aanbeveling wordt altijd bepaald door weging van alle relevante argumenten tezamen.

 

Randvoorwaarden (Organisatie van zorg)

Bij de ontwikkeling van de module is expliciet rekening gehouden met de organisatie van zorg: alle aspecten die een randvoorwaarde zijn voor het verlenen van zorg (zoals coördinatie, communicatie, (financiële) middelen, menskracht en infrastructuur). Randvoorwaarden die relevant zijn voor het beantwoorden van een specifieke uitgangsvraag maken onderdeel uit van de overwegingen bij de bewuste uitgangsvraag, randvoorwaarden die van invloed zijn op de implementatie van de aanbeveling zijn opgenomen in de implementatietabel.

 

Kennislacunes

Tijdens de ontwikkeling van deze module is systematisch gezocht naar onderzoek waarvan de resultaten bijdragen aan een antwoord op de uitgangsvraag. Er is nagegaan of (aanvullend) wetenschappelijk onderzoek gewenst is om de uitgangsvraag te kunnen beantwoorden. Mocht dit bij deze module het geval zijn, dan is er een aanbeveling voor het doen van onderzoek opgenomen in de bijlage Kennislacunes. Deze bijlage is te vinden onder de aanverwante producten.

 

Commentaar- en autorisatiefase

De conceptmodule werd aan de betrokken (wetenschappelijke) verenigingen, instanties en (patiënt) organisaties voorgelegd ter commentaar. De commentaren werden verzameld en besproken met de werkgroep. Naar aanleiding van de commentaren werd de conceptmodule aangepast en definitief vastgesteld door de werkgroep. De definitieve module werd aan de deelnemende (wetenschappelijke) verenigingen en (patiënt) organisaties voorgelegd voor autorisatie en door hen geautoriseerd dan wel geaccordeerd. De commentaartabel is op te vragen bij het Kennisinstituut via secretariaat@kennisinstituut.nl

 

Literatuur

Brouwers MC, Kho ME, Browman GP, et al. AGREE Next Steps Consortium. AGREE II: advancing guideline development, reporting and evaluation in health care. CMAJ. 2010;182(18):E839-42. doi: 10.1503/cmaj.090449. Epub 2010 Jul 5. Review. PubMed PMID: 20603348.

Medisch Specialistische Richtlijnen 2.0. Adviescommissie Richtlijnen van de Raad Kwalitieit. http://richtlijnendatabase.nl/over_deze_site/over_richtlijnontwikkeling.html. 2012.

Schünemann H, Brożek J, Guyatt G, et al. GRADE handbook for grading quality of evidence and strength of recommendations. Updated October 2013. The GRADE Working Group. Available from http://gdt.guidelinedevelopment.org/central_prod/_design/client/handbook/handbook.html. 2013.

Schünemann HJ, Oxman AD, Brozek J, et al. Grading quality of evidence and strength of recommendations for diagnostic tests and strategies. BMJ. 2008;336(7653):1106-10. doi: 10.1136/bmj.39500.677199.AE. Erratum in: BMJ. 2008;336(7654). doi: 10.1136/bmj.a139. PubMed PMID: 18483053.

Ontwikkeling van Medisch Specialistische Richtlijnen: stappenplan. Kennisinstituut van de Federatie Medisch Specialisten.

Zoekverantwoording

Zoekacties zijn opvraagbaar. Neem hiervoor contact op met de Richtlijnendatabase.

Volgende:
Follow-up