This information is intended for US healthcare professionals to access current scientific information about J&J Innovative Medicine products. It is prepared by Medical Information and is not intended for promotional purposes, nor to provide medical advice.

AKEEGA® (niraparib and abiraterone acetate)

Medical Information

AKEEGA - Mechanism of Action

Last Updated: 06/11/2024

SUMMARY

  • Alternative Formats of Information
    • A summary of the information provided in this response is provided as a video that can be accessed by clicking the following link:
  • Niraparib is an orally available, highly selective poly adenosine diphosphate (ADP)-ribose polymerase inhibitor (PARPi), with potent activity against PARP-1 and PARP-2 deoxyribonucleic acid (DNA) repair polymerases.1
  • Abiraterone acetate is a prodrug that is converted in vivo to abiraterone, an androgen biosynthesis inhibitor, that selectively inhibits the enzyme 17-α hydroxylase/C17,20-lyase (CYP17). Inhibition of CYP17, which is expressed in testicular, adrenal, and prostatic tumor tissues and is required for androgen biosynthesis, may result in2:
    • Increased adrenal production of mineralocorticoids (aldosterone).3
    • Interruption of the negative feedback control of adrenocorticotropic hormone (ACTH).3,4
      • The resulting high levels of ACTH and steroid precursors upstream of CYP17 may lead to increased mineralocorticoid effects such as hypertension, hypokalemia, and fluid retention.
      • Coadministration of a corticosteroid suppresses ACTH, resulting in a reduction of steroids upstream of the CYP17 blockade that may ameliorate mineralocorticoid effects.3
  • The combination of niraparib and abiraterone acetate offers a dual-action approach to target tumors with BRCA1/2 mutations in metastatic castration-resistant prostate cancer (mCRPC).5,6 Disruption of homologous recombination repair (HRR) mechanisms by abiraterone acetate potentially sensitizes prostate cancer cells to treatment with niraparib as the combination allows for the simultaneous inhibition of two distinct mechanisms of DNA repair: androgen receptor (AR)-regulated HRR and PARP-associated base-excision repair.7,8

BACKGROUND

Role of PARP in DNA Repair and Defective Homologous Recombination Repair (HRR)

In healthy cells, damage to cellular DNA is repaired by a variety of mechanisms, including HRR and PARP-mediated base-excision repair.9 However, mutations and aberrations in genes involved in DNA repair processes can promote tumorigenesis and inappropriate cell proliferation.10 Highly prolific cancer cells are generally more prone to DNA single-strand breaks (SSBs) which evade cell death through an array of complex DNA repair pathways, including activation of PARPs.11 PARPs are a 17-member superfamily of enzymes involved in the synthesis of poly ADP-ribose (PAR) using nicotinamide adenine dinucleotide (NAD+) as a substrate.12 PAR, a signaling molecule that is synthesized in response to DNA-strand breaks, regulates maintenance of DNA integrity, gene expression, and cell division.13 PARP-1 and PARP-2 are specifically classified as DNA damage-dependent PARPs based on the ability to detect and promote the repair of DNA SSBs using base-excision repair, thereby allowing cancer cells to survive.14,15

When PARPis trap PARP-1 and PARP-2 enzymes at the site of DNA damage, this can lead to the accumulation of double-strand breaks (DSBs) which are normally repaired by HRR, an error-free mechanism that makes use of the sister chromatid as a template.14,16 Molecular defects in HRR result in faulty proteins and lead to the use of an alternative, error-prone mechanism known as non-homologous end-joining (NHEJ) repair that contributes to genomic instability and cancer.11 Additionally, DNA repair and cell survival become dependent on base excision-repair via PARP enzymes,16,17 thereby increasing susceptibility to pharmacological inhibition of DNA repair enzymes; a phenomenon known as synthetic lethality.11,16

The PARPis, as a class of drugs, exploit synthetic lethality as a therapeutic strategy for the treatment of cancers with HRR deficiencies by competing with NAD+ for the binding to the catalytic domain of PARP. Inhibiting the catalytic activity of PARP-1 induces the accumulation of unrepaired SSBs that degenerate into more lethal DSBs during DNA replication.11 PARPi-induced cytotoxic PARP-DNA complexes result in DNA damage, chromosomal instability, and death of HRR-deficient cells via apoptosis.14 Examples of HRR deficiencies include BRCA1/2 mutations and non-BRCA mutations such as ATM, BRIP1, CDK12, CHEK2, FANCA, HDAC2, and PALB2.11

Rationale for Combining a PARPi with an AR-Signaling Inhibitor

AR signaling is one of the main pathways that drive prostate cancer growth.18 The AR regulates a network of DNA repair genes, one of which is the known AR-cofactor, PARP-1.9 In addition to facilitating DNA repair, PARP-1 may support AR transcriptional activity as a potent modulator of both AR function and response to DNA damage.19 PARP-1 also has protumorigenic effects in AR-positive prostate cancer by promoting AR occupancy and function.19 PARP inhibition, therefore has antitumor effects by disrupting AR-associated oncogenic signaling. In animal studies, HRR genes interact with AR signaling to regulate DNA repair in prostate cancer, suggesting that the combination of a PARPi and abiraterone acetate may be more effective in patients with HRR mutations.20 Several studies in prostate cancer have also observed that treatment with PARPis in combination with abiraterone acetate improved radiographic progression-free survival (rPFS) compared to abiraterone acetate alone.11 These results contribute to the hypothesis of a novel type of targeted therapy involving the interplay between PARPis and AR-signaling inhibitors.

CLINICAL PHARMACOLOGY

Niraparib - Description

Niraparib tosylate monohydrate, designated chemically as 2-{4-[(3S)-piperidin-3-yl]phenyl}-2H-indazole 7-carboxamide 4-methylbenzenesulfonate hydrate, is a small-molecule selective inhibitor of the PARP-1 and PARP-2 enzymes with in vitro 50% inhibitory concentration (IC50) = 3.8 and 2.1 nM, respectively.1,12,21 Niraparib is a potent catalytic PARPi with selectivity for cancerous cells over normal cells.12

Abiraterone Acetate - Description

Abiraterone acetate, designated chemically as (3β)-17-(3-pyridinyl) androsta-5,16-dien-3-yl acetate, is the acetyl ester of abiraterone. Abiraterone acetate and its metabolite, abiraterone, are highly selective, potent, and irreversible inhibitors of CYP17 with a Kiapp of <1 nM and IC50 of 72 nM.22,23 In human microsomes, the IC50 of abiraterone for inhibition of CYP17 was approximately 10% of the IC50 of ketoconazole.24

Mechanism of Action

The combination of niraparib and abiraterone acetate offers a dual-action approach to target tumors with BRCA1/2 mutations in mCRPC.5,6 Niraparib is an orally available, highly selective PARPi with potent activity against PARP-1 and PARP-2.1 Abiraterone acetate is a prodrug that is converted in vivo to abiraterone, an androgen biosynthesis inhibitor, that selectively inhibits the CYP17 enzyme, providing antitumor effects by inhibiting the production of androgens, such as testosterone, and the activation of the AR.25,26 Inhibition of the AR reduces expression of DNA repair genes and NHEJ repair, which leads to accumulation of DNA damage, and results in a compensatory increase in PARP activity.17,27,28 Disruption of HRR mechanisms by abiraterone acetate potentially sensitizes prostate cancer cells to treatment with niraparib as the combination allows for the simultaneous inhibition of two distinct mechanisms of DNA repair: AR-regulated HRR and PARP-associated base-excision repair.7,8

Literature Search

A literature search of MEDLINE®, Embase®, BIOSIS Previews®, and Derwent Drug File (and/or other resources, including internal/external databases) was conducted on 20 May 2024.

 

References

1 Jones P, Altamura S, Boueres J, et al. Discovery of 2-{4-[(3S)-piperidin-3-yl]phenyl}-2H-indazole-7-carboxamide (MK-4827): a novel oral poly (ADP-ribose) polymerase (PARP) inhibitor efficacious in BRCA-1 and -2 mutant tumors. J Med Chem. 2009;52(22):7170-7185.  
2 O’Donnell A, Judson I, Dowsett M, et al. Hormonal impact of the 17α-hydroxylase/C(17,20)-lyase inhibitor abiraterone acetate (CB7630) in patients with prostate cancer. Br J Cancer. 2004;90(12):2317-2325.  
3 Attard G, Reid AHM, A’Hern R, et al. Selective inhibition of CYP17 with abiraterone acetate is highly active in the treatment of castration-resistant prostate cancer. J Clin Oncol. 2009;27(23):3742-3748.  
4 Attard G, Reid AH, Yap TA, et al. Phase I clinical trial of a selective inhibitor of CYP17, abiraterone acetate, confirms that castration-resistant prostate cancer commonly remains hormone driven. J Clin Oncol. 2008;26(28):4563-4571.  
5 Chi KN, Rathkopf D, Smith MR, et al. Niraparib and abiraterone acetate for metastatic castration-resistant prostate cancer. J Clin Oncol. 2023;41(18):3339-3351.  
6 Chi KN, Sandhu S, Smith MR, et al. Niraparib plus abiraterone acetate with prednisone in patients with metastatic castration-resistant prostate cancer and homologous recombination repair gene alterations: second interim analysis of the randomized phase III MAGNITUDE trial. Ann Oncol. 2023;34(9):772-782.  
7 Li L, Karanika S, Yang G, et al. Androgen receptor inhibitor-induced “BRCAness” and PARP inhibition are synthetically lethal for castration-resistant prostate cancer. Sci Signal. 2017;10(480):eaam7479.  
8 Goodwin JF, Schiewer MJ, Dean JL, et al. A hormone-DNA repair circuit governs the response to genotoxic insult. Cancer Discov. 2013;3(11):1254-1271.  
9 Lozano R, Castro E, Aragon IM, et al. Genetic aberrations in DNA repair pathways: a cornerstone of precision oncology in prostate cancer. Br J Cancer. 2021;124(3):552-563.  
10 Alhmoud JF, Woolley JF, Al Moustafa AE, et al. DNA damage/repair management in cancers. Cancers (Basel). 2020;12(4):1050.  
11 Criscuolo D, Morra F, Giannella R, et al. Identification of novel biomarkers of homologous recombination defect in DNA repair to predict sensitivity of prostate cancer cells to PARP-inhibitors. Int J Mol Sci. 2019;20(12):3100.  
12 Jones P, Wilcoxen K, Rowley M, et al. Niraparib: a poly (ADP-ribose) polymerase (PARP) inhibitor for the treatment of tumors with defective homologous recombination. J Med Chem. 2015;58(8):3302-3314.  
13 Schreiber V, Dantzer F, Ame JC, et al. Poly (ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol. 2006;7(7):517-528.  
14 Sonnenblick A, Azambuja E de, Azim HA, et al. An update on PARP inhibitors - moving to the adjuvant setting. Leukemia Res Reports. 2015;12(1):27-41.  
15 Min A, Im S. PARP inhibitors as therapeutics: beyond modulation of PARylation. Cancers (Basel). 2020;12(2):394.  
16 Teyssonneau D, Margot H, Cabart M, et al. Prostate cancer and PARP inhibitors: progress and challenges. J Hematol Oncol. 2021;14(1):51.  
17 Asim M, Tarish F, Zecchini HI, et al. Synthetic lethality between androgen receptor signalling and the PARP pathway in prostate cancer. Nat Commun. 2017;8(1):374.  
18 Aurilio G, Cimadamore A, Mazzucchelli R, et al. Androgen receptor signaling pathway in prostate cancer: from genetics to clinical applications. Cells. 2020;9(2):2653.  
19 Schiewer MJ, Goodwin JF, Han S, et al. Dual roles of PARP-1 promote cancer growth and progression. Cancer Discov. 2012;2(12):1134-1149.  
20 Saad F, Chi KN, Shore ND, et al. Niraparib with androgen receptor-axis-targeted therapy in patients with metastatic castration-resistant prostate cancer: safety and pharmacokinetic results from a phase 1b study (BEDIVERE). Cancer Chemother Pharmacol. 2021;88:25-37.  
21 Ison G, Howie LJ, Amiri-Kordestani L, et al. FDA approval summary: niraparib for the maintenance treatment of patients with recurrent ovarian cancer in response to platinum-based chemotherapy. Clin Cancer Res. 2018;24(17):4066-4071.  
22 Hille UE, Hu Q, Vock C, et al. Novel CYP17 inhibitors: synthesis, biological evaluation, structure–activity relationships and modelling of methoxy- and hydroxy-substituted methyleneimidazolyl biphenyls. Eur J Med Chem. 2009;44(7):2765-2775.  
23 Potter GA, Barrie SE, Jarman M. Novel steroidal inhibitors of human cytochrome P45017 alpha (17α-hydroxylase-C17,20-lyase): potential agents for the treatment of prostatic cancer. J Med Chem. 1995;38(13):2463-2471.  
24 Haidar S, Ehmer PB, Barassin S, et al. Effects of novel 17α-hydroxylase/C17,20-lyase (P450 17, CYP17) inhibitors on androgen biosynthesis in vitro and in vivo. J Steroid Biochem Mol Biol. 2003;84(5):555-562.  
25 Bono JD, Mateo J, Fizazi K, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med. 2020;382(22):2091-2102.  
26 Rice MA, Malhotra SV, Stoyanova T. Second-generation antiandrogens: from discovery to standard of care in castration resistant prostate cancer. Front Oncol. 2019;9:801.  
27 Polkinghorn WR, Parker JS, Lee MX, et al. Androgen receptor signaling regulates DNA repair in prostate cancers. Cancer Discov. 2013;3(11):1245-1253.  
28 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;116(29):14573-14582.