Raadpleeg ook literatuurlijst niet-toxische middelen, voeding en behandelingen specifiek bij prostaatkanker van arts-bioloog drs. Engelbert Valstar. 

Als donateur kunt u ook korting krijgen bij verschillende bedrijven, waaronder bij Medpro voor o.a. prostasol  een veel gebruikt natuurlijk middel bij prostaatkanker als alternatief voor hormoontherapie.


5 juli 2023: Bron: The Lancet, Published:June 04, 2023

Als patiënten met uitgezaaide castratieresistente prostaatkanker naast enzalutamide ook dagelijks de parpremmer Talazoparib (Talzenne) krijgen dan verbeterde de progressievrije ziekte aanzienlijk ongeacht de mutatiestatus bij de patiënten. 

In Talapro-2, een gerandomiseerde fase III-studie evalueerden de auteurs de resultaten van enzalutamide met of zonder talazoparib bij totaal 805 patiënten met uitgezaaide castratieresistente prostaatkanker in de eerste lijn.

Radiografische progressievrije overleving, objectief responspercentage, volledig responspercentage, tijd tot prostaatspecifieke antigeenprogressie en tijd tot cytotoxische chemotherapie waren allemaal significant superieur in de combinatiegroep, ongeacht de HRR = homologe recombinatie reparatie (HRR) genveranderingsstatus (mutatiestatus). Bijwerkingen kwamen iets vaker voor in de combinatiegroep, met name bloedarmoede.

Uit het abstract vertaald:

  • Tussen 7 januari 2019 en 17 september 2020 werden 805 patiënten ingeschreven en willekeurig toegewezen (402 patiënten aan de talazoparib-groep en 403 patiënten aan de placebogroep).
  • De mediane follow-up voor rPFS (= radiografisch progressievrije ziekte) was 24,9 maanden (IQR 21,9-30,2) voor de talazoparib-groep en 24,6 maanden (14,4-30,2) voor de placebogroep.
  • Bij de geplande primaire analyse was de mediane rPFS nog niet bereikt (95% BI 27,5 maanden-niet bereikt) voor talazoparib plus enzalutamide en 21,9 maanden (16,6-25,1) voor placebo plus enzalutamide (hazard ratio 0· 63; 95%-BI 0,51-0,78; p<0,0001).
  • In de talazoparib-groep waren de meest voorkomende tijdens de behandeling optredende bijwerkingen anemie, neutropenie en vermoeidheid; de meest voorkomende ernstige bijwerking van graad 3-4 was bloedarmoede (185 [46%] van de 398 patiënten), die wel verbeterde na dosisverlaging, en slechts 33 (8%) van de 398 patiënten stopten met talazoparib vanwege anemie.
  • Aan de behandeling gerelateerde sterfgevallen kwamen voor bij geen enkele patiënt in de talazoparib-groep en bij twee patiënten (<1%) in de placebogroep.

Voor het volledige studierapport moet worden betaald. Hier het abstract van de studie:

Talazoparib plus enzalutamide in men with first-line metastatic castration-resistant prostate cancer (TALAPRO-2): a randomised, placebo-controlled, phase 3 trial

Summary

Background

Co-inhibition of poly(ADP-ribose) polymerase (PARP) and androgen receptor activity might result in antitumour efficacy irrespective of alterations in DNA damage repair genes involved in homologous recombination repair (HRR). We aimed to compare the efficacy and safety of talazoparib (a PARP inhibitor) plus enzalutamide (an androgen receptor blocker) versus enzalutamide alone in patients with metastatic castration-resistant prostate cancer (mCRPC).

Methods

TALAPRO-2 is a randomised, double-blind, phase 3 trial of talazoparib plus enzalutamide versus placebo plus enzalutamide as first-line therapy in men (age ≥18 years [≥20 years in Japan]) with asymptomatic or mildly symptomatic mCRPC receiving ongoing androgen deprivation therapy. Patients were enrolled from 223 hospitals, cancer centres, and medical centres in 26 countries in North America, Europe, Israel, South America, South Africa, and the Asia-Pacific region. Patients were prospectively assessed for HRR gene alterations in tumour tissue and randomly assigned (1:1) to talazoparib 0·5 mg or placebo, plus enzalutamide 160 mg, administered orally once daily. Randomisation was stratified by HRR gene alteration status (deficient vs non-deficient or unknown) and previous treatment with life-prolonging therapy (docetaxel or abiraterone, or both: yes vs no) in the castration-sensitive setting. The sponsor, patients, and investigators were masked to talazoparib or placebo, while enzalutamide was open-label. The primary endpoint was radiographic progression-free survival (rPFS) by blinded independent central review, evaluated in the intention-to-treat population. Safety was evaluated in all patients who received at least one dose of study drug. This study is registered with ClinicalTrials.gov (NCT03395197) and is ongoing.

Findings

Between Jan 7, 2019, and Sept 17, 2020, 805 patients were enrolled and randomly assigned (402 to the talazoparib group and 403 to the placebo group). Median follow-up for rPFS was 24·9 months (IQR 21·9–30·2) for the talazoparib group and 24·6 months (14·4–30·2) for the placebo group. At the planned primary analysis, median rPFS was not reached (95% CI 27·5 months–not reached) for talazoparib plus enzalutamide and 21·9 months (16·6–25·1) for placebo plus enzalutamide (hazard ratio 0·63; 95% CI 0·51–0·78; p<0·0001). In the talazoparib group, the most common treatment-emergent adverse events were anaemia, neutropenia, and fatigue; the most common grade 3–4 event was anaemia (185 [46%] of 398 patients), which improved after dose reduction, and only 33 (8%) of 398 patients discontinued talazoparib due to anaemia. Treatment-related deaths occurred in no patients in the talazoparib group and two patients (<1%) in the placebo group.

Interpretation

Talazoparib plus enzalutamide resulted in clinically meaningful and statistically significant improvement in rPFS versus standard of care enzalutamide as first-line treatment for patients with mCRPC. Final overall survival data and additional long-term safety follow-up will further clarify the clinical benefit of the treatment combination in patients with and without tumour HRR gene alterations.

Funding

Pfizer.


References

  1. 1.
    • Sung H 
    • Ferlay J 
    • Siegel RL 
    • et al.
    Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries.
    CA Cancer J Clin. 2021; 71209-249
  2. 2.
    • Siegel RL 
    • Miller KD 
    • Wagle NS 
    • Jemal A
    Cancer statistics, 2023.
    CA Cancer J Clin. 2023; 7317-48
  3. 3.
    • Chung JH 
    • Dewal N 
    • Sokol E 
    • et al.
    Prospective comprehensive genomic profiling of primary and metastatic prostate tumors.
    JCO Precis Oncol. 2019; 3 (PO.18.00283.)
  4. 4.
    • Shen Y 
    • Rehman FL 
    • Feng Y 
    • et al.
    BMN 673, a novel and highly potent PARP1/2 inhibitor for the treatment of human cancers with DNA repair deficiency.
    Clin Cancer Res. 2013; 195003-5015
  5. 5.
    • Farmer H 
    • McCabe N 
    • Lord CJ 
    • et al.
    Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy.
    Nature. 2005; 434917-921
  6. 6.
    • Bryant HE 
    • Schultz N 
    • Thomas HD 
    • et al.
    Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase.
    Nature. 2005; 434913-917
  7. 7.
    • Lord CJ 
    • Ashworth A
    PARP inhibitors: synthetic lethality in the clinic.
    Science. 2017; 3551152-1158
  8. 8.
    • Tran C 
    • Ouk S 
    • Clegg NJ 
    • et al.
    Development of a second-generation antiandrogen for treatment of advanced prostate cancer.
    Science. 2009; 324787-790
  9. 9.
    • Murai J 
    • Huang SY 
    • Renaud A 
    • et al.
    Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib.
    Mol Cancer Ther. 2014; 13433-443
  10. 10.
    • de Bono JS 
    • Mehra N 
    • Scagliotti GV 
    • et al.
    Talazoparib monotherapy in metastatic castration-resistant prostate cancer with DNA repair alterations (TALAPRO-1): an open-label, phase 2 trial.
    Lancet Oncol. 2021; 221250-1264
  11. 11.
    • de Bono J 
    • Mateo J 
    • Fizazi K 
    • et al.
    Olaparib for metastatic castration-resistant prostate cancer.
    N Engl J Med. 2020; 3822091-2102
  12. 12.
    • Fizazi K 
    • Piulats JM 
    • Reaume MN 
    • et al.
    Rucaparib or physician's choice in metastatic prostate cancer.
    N Engl J Med. 2023; 388719-732
  13. 13.
    • Rao A 
    • Moka N 
    • Hamstra DA 
    • Ryan CJ
    Co-inhibition of androgen receptor and PARP as a novel treatment paradigm in prostate cancer—where are we now?.
    Cancers (Basel). 2022; 14801
  14. 14.
    • Asim M 
    • Tarish F 
    • Zecchini HI 
    • et al.
    Synthetic lethality between androgen receptor signalling and the PARP pathway in prostate cancer.
    Nat Commun. 2017; 8374
  15. 15.
    • Polkinghorn WR 
    • Parker JS 
    • Lee MX 
    • et al.
    Androgen receptor signaling regulates DNA repair in prostate cancers.
    Cancer Discov. 2013; 31245-1253
  16. 16.
    • Gui B 
    • Gui F 
    • Takai T 
    • et al.
    Selective targeting of PARP-2 inhibits androgen receptor signaling and prostate cancer growth through disruption of FOXA1 function.
    Proc Natl Acad Sci USA. 2019; 11614573-14582
  17. 17.
    • Schiewer MJ 
    • Goodwin JF 
    • Han S 
    • et al.
    Dual roles of PARP-1 promote cancer growth and progression.
    Cancer Discov. 2012; 21134-1149
  18. 18.
    • Agarwal N 
    • Azad A 
    • Shore ND 
    • et al.
    Talazoparib plus enzalutamide in metastatic castration-resistant prostate cancer: TALAPRO-2 phase III study design.
    Future Oncol. 2022; 18425-436
  19. 19.
    • Agarwal N 
    • Shore ND 
    • Dunshee C 
    • et al.
    Clinical and safety outcomes of TALAPRO-2: a two-part phase III study of talazoparib (TALA) in combination with enzalutamide (ENZA) in metastatic castration-resistant prostate cancer (mCRPC).
    J Clin Oncol. 2019; 375076
  20. 20.
    • US Department of Health and Human Services
    Common terminology criteria for adverse events (CTCAE) version 4.03.
  21. 21.
    • Beer TM 
    • Armstrong AJ 
    • Rathkopf D 
    • et al.
    Enzalutamide in men with chemotherapy-naïve metastatic castration-resistant prostate cancer: extended analysis of the phase 3 PREVAIL study.
    Eur Urol. 2017; 71151-154
  22. 22.
    • Abida W 
    • Armenia J 
    • Gopalan A 
    • et al.
    Prospective genomic profiling of prostate cancer across disease states reveals germline and somatic alterations that may affect clinical decision making.
    JCO Precis Oncol. 2017; 1 (PO.17.00029.)
  23. 23.
    • Armenia J 
    • Wankowicz SAM 
    • Liu D 
    • et al.
    The long tail of oncogenic drivers in prostate cancer.
    Nat Genet. 2018; 50645-651
  24. 24.
    • Robinson D 
    • Van Allen EM 
    • Wu YM 
    • et al.
    Integrative clinical genomics of advanced prostate cancer.
    Cell. 2015; 1611215-1228
  25. 25.
    • Clarke NW 
    • Armstrong AJ 
    • Thiery-Vuillemin A 
    • et al.
    Abiraterone and olaparib for metastatic castration-resistant prostate cancer.
    NEJM Evid. 2022; 1EVIDoa2200043
  26. 26.
    • Chi KN 
    • Rathkopf D 
    • Smith MR 
    • et al.
    Niraparib and abiraterone acetate for metastatic castration-resistant prostate cancer.
    J Clin Oncol. 2023; (published online March 23.)
  27. 27.
    • Zandarashvili L 
    • Langelier MF 
    • Velagapudi UK 
    • et al.
    Structural basis for allosteric PARP-1 retention on DNA breaks.
    Science. 2020; 368eaax6367
  28. 28.
    • Beer TM 
    • Armstrong AJ 
    • Rathkopf DE 
    • et al.
    Enzalutamide in metastatic prostate cancer before chemotherapy.
    N Engl J Med. 2014; 371424-433
  29. 29.
    • Mehra N 
    • Fizazi K 
    • de Bono JS 
    • et al.
    Talazoparib, a poly (ADP-ribose) polymerase inhibitor, for metastatic castration-resistant prostate cancer and DNA damage response alterations: TALAPRO-1 safety analyses.
    Oncologist. 2022; 27e783-e795

Linked Articles






Gerelateerde artikelen
 

Gerelateerde artikelen

Cabozantinib in combinatie >> Olaparib geeft betere resultaten >> Rupacarib (Rubraca) geeft >> Talazoparib toegevoegd aan >> Talazoparib, een PARP remmer, >> Parpremmers bij prostaatkanker, >>