A phase I dose-escalation study of enzalutamide in combination with the AKT inhibitor AZD5363 (capivasertib) in patients with metastatic castration-resistant prostate cancer

M P Kolinsky, P Rescigno, D Bianchini, Z Zafeiriou, N Mehra, J Mateo, V Michalarea, R Riisnaes, M Crespo, I Figueiredo, S Miranda, D Nava Rodrigues, P Flohr, N Tunariu, U Banerji, R Ruddle, A Sharp, J Welti, M Lambros, S Carreira, F I Raynaud, K E Swales, S Plymate, J Luo, H Tovey, N Porta, R Slade, L Leonard, E Hall, J S de Bono, M P Kolinsky, P Rescigno, D Bianchini, Z Zafeiriou, N Mehra, J Mateo, V Michalarea, R Riisnaes, M Crespo, I Figueiredo, S Miranda, D Nava Rodrigues, P Flohr, N Tunariu, U Banerji, R Ruddle, A Sharp, J Welti, M Lambros, S Carreira, F I Raynaud, K E Swales, S Plymate, J Luo, H Tovey, N Porta, R Slade, L Leonard, E Hall, J S de Bono

Abstract

Background: Activation of the PI3K/AKT/mTOR pathway through loss of phosphatase and tensin homolog (PTEN) occurs in approximately 50% of patients with metastatic castration-resistant prostate cancer (mCRPC). Recent evidence suggests that combined inhibition of the androgen receptor (AR) and AKT may be beneficial in mCRPC with PTEN loss.

Patients and methods: mCRPC patients who previously failed abiraterone and/or enzalutamide, received escalating doses of AZD5363 (capivasertib) starting at 320 mg twice daily (b.i.d.) given 4 days on and 3 days off, in combination with enzalutamide 160 mg daily. The co-primary endpoints were safety/tolerability and determining the maximum tolerated dose and recommended phase II dose; pharmacokinetics, antitumour activity, and exploratory biomarker analysis were also evaluated.

Results: Sixteen patients were enrolled, 15 received study treatment and 13 were assessable for dose-limiting toxicities (DLTs). Patients were treated at 320, 400, and 480 mg b.i.d. dose levels of capivasertib. The recommended phase II dose identified for capivasertib was 400 mg b.i.d. with 1/6 patients experiencing a DLT (maculopapular rash) at this level. The most common grade ≥3 adverse events were hyperglycemia (26.7%) and rash (20%). Concomitant administration of enzalutamide significantly decreased plasma exposure of capivasertib, though this did not appear to impact pharmacodynamics. Three patients met the criteria for response (defined as prostate-specific antigen decline ≥50%, circulating tumour cell conversion, and/or radiological response). Responses were seen in patients with PTEN loss or activating mutations in AKT, low or absent AR-V7 expression, as well as those with an increase in phosphorylated extracellular signal-regulated kinase (pERK) in post-exposure samples.

Conclusions: The combination of capivasertib and enzalutamide is tolerable and has antitumour activity, with all responding patients harbouring aberrations in the PI3K/AKT/mTOR pathway.

Clinical trial number: NCT02525068.

Keywords: AKT inhibitor; AZD5363; biomarkers; capivasertib; enzalutamide; prostate cancer.

Copyright © 2020 The Author(s). Published by Elsevier Ltd.. All rights reserved.

Figures

Figure 1
Figure 1
Percent change in prostate-specific antigen (PSA) at 12 weeks relative to baseline PSA. Each bar represents an individual patient. Light colour indicates the patient previously received treatment with both abiraterone and enzalutamide; dark colour indicates prior treatment with only abiraterone and not enzalutamide. Patients indicated with × discontinued before 12 weeks but safety follow-up results are available; in these patients, the percent change of PSA at discontinuation relative to baseline is presented. The patient indicated with + also met response criteria for RECIST and circulating tumour cell (CTC) conversion. Patients indicated with a dot discontinued treatment before 12 weeks with no post-treatment PSA values obtained. Dose level refers to the dosage of capivasertib the patient received in mg. Phosphate and tensin homolog (PTEN) status refers to immunohistochemistry (IHC) expression with N representing normal and L representing loss. ARV7 status refers to pre-treatment tumour biopsy baseline AR-V7 expression by IHC with + indicating an H-score of >10 and − indicating ≤10. Phosphorylated extracellular signal-related kinases (pERK) refers to increased expression by IHC on post-treatment tumour biopsy samples relative to baseline indicated by +, whereas − indicates no increase. NGS refers to next-generation sequencing with + representing known or likely deleterious mutations in PI3K/AKT/mTOR pathway genes and − representing an absence of such mutations. NA indicates not available. Patients meeting response criteria assessed by PSA, soft tissue objective response by RECIST, CTC conversions, and overall [indicated by (-r) respectively] are indicated by (Yes), with non-responders indicated by (No), and (N/E) indicating non-evaluable. † indicates non-confirmed CTC conversions. Reasons for discontinuation included progressive disease (PD), patient choice (PC), and AE.
Supplementary Figure S1
Supplementary Figure S1
Supplementary Figure S2
Supplementary Figure S2
Supplementary Figure S3a
Supplementary Figure S3a
Supplementary Figure S3b
Supplementary Figure S3b
Supplementary Figure S4
Supplementary Figure S4
Supplementary Figure S5
Supplementary Figure S5
Supplementary Figure S6
Supplementary Figure S6
Supplementary Figure S7
Supplementary Figure S7
Supplementary Figure S8
Supplementary Figure S8

References

    1. Watson P.A., Arora V.K., Sawyers C.L. Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer. Nat Rev Cancer. 2015;15(12):701–711.
    1. de Bono J.S., Logothetis C.J., Molina A. Abiraterone and increased survival in metastatic prostate cancer. N Engl J Med. 2011;364(21):1995–2005.
    1. Scher H.I., Fizazi K., Saad F. Increased survival with enzalutamide in prostate cancer after chemotherapy. N Engl J Med. 2012;367(13):1187–1197.
    1. Sarker D., Reid A.H., Yap T.A. Targeting the PI3K/AKT pathway for the treatment of prostate cancer. Clin Cancer Res. 2009;15(15):4799–4805.
    1. Robinson D., Van Allen E.M., Wu Y.M. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161(5):1215–1228.
    1. Carver B.S., Chapinski C., Wongvipat J. Reciprocal feedback regulation of PI3K and androgen receptor signaling in PTEN-deficient prostate cancer. Cancer Cell. 2011;19(5):575–586.
    1. Toren P., Kim S., Cordonnier T. Combination AZD5363 with enzalutamide significantly delays enzalutamide-resistant prostate cancer in preclinical models. Eur Urol. 2015;67(6):986–990.
    1. Marques R.B., Aghai A., de Ridder C.M. High Efficacy of combination therapy using PI3K/AKT inhibitors with androgen deprivation in prostate cancer preclinical models. Eur Urol. 2015;67(6):1177–1185.
    1. Banerji U., Dean E.J., Pérez-Fidalgo J.A. A phase I open-label study to identify a dosing regimen of the pan-AKT inhibitor AZD5363 for evaluation in solid tumors and in PIK3CA-mutated breast and gynecologic cancers. Clin Cancer Res. 2018;24(9):2050–2059.
    1. Tamura K., Hashimoto J., Tanabe Y. Safety and tolerability of AZD5363 in Japanese patients with advanced solid tumors. Cancer Chemother Pharmacol. 2016;77(4):787–795.
    1. Oken M.M., Creech R.H., Tormey D.C. Toxicity and response criteria of the Eastern Cooperative Oncology Group. Am J Clin Oncol. 1982;5(6):649–655.
    1. Storer B.E. Design and analysis of phase I clinical trials. Biometrics. 1989;45(3):925–937.
    1. Antonarakis E.S., Lu C., Wang H. AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371(11):1028–1038.
    1. Mateo J., Carreira S., Sandhu S. DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med. 2015;373(18):1697–1708.
    1. Nava Rodrigues D., Rescigno P., Liu D. Immunogenomic analyses associate immunological alterations with mismatch repair defects in prostate cancer. J Clin Invest. 2018;128(10):4441–4453.
    1. Graff J.N., Alumkal J.J., Drake C.G. Early evidence of anti-PD-1 activity in enzalutamide-resistant prostate cancer. Oncotarget. 2016;7(33):52810–52817.
    1. Mulholland D.J., Tran L.M., Li Y. Cell autonomous role of PTEN in regulating castration-resistant prostate cancer growth. Cancer Cell. 2011;19(6):792–804.
    1. Bitting R.L., Armstrong A.J. Targeting the PI3K/Akt/mTOR pathway in castration-resistant prostate cancer. Endocr Relat Cancer. 2013;20(3):R83–R99.
    1. Ferraldeschi R., Nava Rodrigues D., Riisnaes R. PTEN protein loss and clinical outcome from castration-resistant prostate cancer treated with abiraterone acetate. Eur Urol. 2015;67(4):795–802.
    1. Turner N.C., Alarcón E., Armstrong A.C. BEECH: a dose-finding run-in followed by a randomised phase 2 study assessing the efficacy of AKT inhibitor capivasertib (AZD5363) combined with paclitaxel in patients with oestrogen receptor-positive advanced or metastatic breast cancer, and in a PIK3CA mutant sub-population. Ann Oncol. 2019;30(5):774–780.
    1. Antonarakis E.S., Lu C., Luber B. Androgen receptor splice variant 7 and efficacy of taxane chemotherapy in patients with metastatic castration-resistant prostate cancer. JAMA Oncol. 2015;1(5):582–591.
    1. McKay M.M., Morrison D.K. Integrating signals from RTKs to ERK/MAPK. Oncogene. 2007;26(22):3113–3121.
    1. Rozengurt E. Mitogenic signaling pathways induced by G protein-coupled receptors. J Cell Physiol. 2007;213(3):589–602.
    1. Zimmermann S., Moelling K. Phosphorylation and regulation of Raf by AKT (protein kinase B) Science. 1999;286(5445):1741–1744.
    1. Dhillon A.S., Meikle S., Yazici Z. Regulation of Raf-1 activation and signalling by dephosphorylation. EMBO J. 2002;21(1-2):64–71.
    1. Guan K.L., Figueroa C., Brtva T.R. Negative regulation of the serine/threonine kinase B-Raf by AKT. J Biol Chem. 2000;275(35):27354–27359.
    1. Cheung M., Sharma A., Madhunapantula S.V. AKT3 and mutant V600E B-Raf cooperate to promote early melanoma development. Cancer Res. 2008;68(9):3429–3439.
    1. de Bono J.S., De Giorgi U., Rodrigues D.N. Randomized phase II study evaluating AKT blockade with ipatasertib, in combination with abiraterone, in patients with metastatic prostate cancer with and without PTEN loss. Clin Cancer Res. 2018;25(3):928–936.
    1. Yates J.W., Dudley P., Cheng J. Validation of a predictive modeling approach to demonstrate the relative efficacy of three different schedules of the AKT inhibitor AZD5363. Cancer Chemother Pharmacol. 2015;76(2):343–356.

Source: PubMed

3
購読する