Darolutamide Potentiates the Antitumor Efficacy of a PSMA-targeted Thorium-227 Conjugate by a Dual Mode of Action in Prostate Cancer Models

Stefanie Hammer, Andreas Schlicker, Sabine Zitzmann-Kolbe, Simon Baumgart, Urs B Hagemann, Arne Scholz, Bernard Haendler, Pascale Lejeune, Jenny Karlsson, Christine Ellingsen, Hartwig Hennekes, Carsten H Nielsen, Mark U Juul, Dominik Mumberg, Christoph A Schatz, Stefanie Hammer, Andreas Schlicker, Sabine Zitzmann-Kolbe, Simon Baumgart, Urs B Hagemann, Arne Scholz, Bernard Haendler, Pascale Lejeune, Jenny Karlsson, Christine Ellingsen, Hartwig Hennekes, Carsten H Nielsen, Mark U Juul, Dominik Mumberg, Christoph A Schatz

Abstract

Purpose: Androgen receptor (AR) inhibitors are well established in the treatment of castration-resistant prostate cancer and have recently shown efficacy also in castration-sensitive prostate cancer. Although most patients respond well to initial therapy, resistance eventually develops, and thus, more effective therapeutic approaches are needed. Prostate-specific membrane antigen (PSMA) is highly expressed in prostate cancer and presents an attractive target for radionuclide therapy. Here, we evaluated the efficacy and explored the mode of action of the PSMA-targeted thorium-227 conjugate (PSMA-TTC) BAY 2315497, an antibody-based targeted alpha-therapy, in combination with the AR inhibitor darolutamide.

Experimental design: The in vitro and in vivo antitumor efficacy and mode of action of the combination treatment were investigated in preclinical cell line-derived and patient-derived prostate cancer xenograft models with different levels of PSMA expression.

Results: Darolutamide induced the expression of PSMA in androgen-sensitive VCaP and LNCaP cells in vitro, and the efficacy of darolutamide in combination with PSMA-TTC was synergistic in these cells. In vivo, the combination treatment showed synergistic antitumor efficacy in the low PSMA-expressing VCaP and in the high PSMA-expressing ST1273 prostate cancer models, and enhanced efficacy in the enzalutamide-resistant KUCaP-1 model. The treatments were well tolerated. Mode-of-action studies revealed that darolutamide induced PSMA expression, resulting in higher tumor uptake of PSMA-TTC, and consequently, higher antitumor efficacy, and impaired PSMA-TTC-mediated induction of DNA damage repair genes, potentially contributing to increased DNA damage.

Conclusions: These results provide a strong rationale to investigate PSMA-TTC in combination with AR inhibitors in patients with prostate cancer.

©2021 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
Darolutamide and PSMA-TTC combination shows synergistic antitumor efficacy in vitro. Isobolograms for the in vitro combination effect of PSMA-TTC and darolutamide on the proliferation of VCaP (A) and LNCaP (B) prostate cancer cells. PSMA (FOLH1) expression in VCaP (C) and LNCaP (D) cells treated with 5 kBq/mL PSMA-TTC and/or 2 μmol/L darolutamide as determined by qRT-PCR 48 hours after treatment (n = 2). The expression is presented as fold change compared with untreated cells. Cell surface PSMA expression as determined by flow cytometry in darolutamide or enzalutamide-treated VCaP (E) and LNCaP (F) cells. DMSO served as a baseline control and is depicted with a dashed line. CI, combination index; DMSO, dimethyl sulfoxide.
Figure 2.
Figure 2.
Darolutamide induces PSMA expression and shows synergistic antitumor efficacy with PSMA-TTC in the VCaP prostate cancer model. Male CB17-Scid mice were treated with vehicle, a single dose of 150 or 300 kBq/kg PSMA-TTC (intravenous, total protein dose 0.14 mg/kg, treatment day indicated with a green arrow) and/or 100 mg/kg darolutamide [(twice daily)× 23, orally, treatment period indicated with a blue bar]. A, Growth curves of VCaP tumors (n = 11–12). B, VCaP tumor weight at the end of the study (n = 11–12). C,PSMA RNA expression in VCaP tumors as determined by qRT-PCR (n = 2–4). D, PSMA protein expression in VCaP tumors as determined by immunoblotting (n = 4–7). E, Thorium-227 accumulation in VCaP tumors 66 hours after dosing (n = 4). F, Expression of phosphorylated Chk2 (pChk2) in VCaP tumors as determined by immunoblotting (n = 3–6). Signals from pChk2 were normalized to total Chk2. Statistical analyses were performed using linear models followed by Dunnett or Sidak method. *, P < 0.05; **, P < 0.01; ***, P < 0.001 in comparison with vehicle. ##, P < 0.01; ###, P < 0.001 in comparison with the corresponding darolutamide monotherapy. †, P < 0.05; ††, P < 0.01; †††, P < 0.001 in comparison with the corresponding PSMA-TTC monotherapy.
Figure 3.
Figure 3.
PSMA-TTC and darolutamide show synergistic antitumor efficacy in the ST1273 prostate cancer PDX model. A, Growth curves of ST1273 tumors in female NMRI nude mice (n = 9–10) treated with vehicle, 250 kBq/kg PSMA-TTC (total protein dose 0.14 mg/kg, intravenous, treatment day indicated with a green arrow) and/or 15, 30, or 100 mg/kg darolutamide [(twice daily)× 21, orally; treatment period indicated with a blue bar]. B, Tumor volumes of individual ST1273 tumors shown in A on day 33. C, Growth curves of ST1273 tumors in female NMRI nude mice (n = 10) treated with vehicle, 75, 125, or 250 kBq/kg PSMA-TTC (intravenous, total protein dose 0.14 mg/kg; treatment day indicated with a green arrow) and/or 100 mg/kg darolutamide [(twice daily)× 21, orally; treatment period indicated with a blue bar]. For the generation of the mean tumor volume curves in A and C, the last measured tumor volume values of mice which were euthanized due to large tumor size were included into the calculation of the mean value in the graph until n ≧ 8 (last study day shown in graph). D, Tumor volumes of individual ST1273 tumors shown in C on day 19. Statistical analyses were performed on day 33 or day 19 for the studies shown in A and C, respectively, using the estimated linear model followed by Sidak method. **, P < 0.01; ***, P < 0.001 in comparison with vehicle. #, P < 0.05; ##, P < 0.01; ###, P < 0.001 in comparison with the corresponding darolutamide monotherapy. †, P < 0.05; ††, P < 0.01; †††, P < 0.001 in comparison with the corresponding PSMA-TTC monotherapy.
Figure 4.
Figure 4.
Darolutamide shows enhanced antitumor efficacy in combination with PSMA-TTC in the KUCaP-1 prostate cancer PDX model. Male CB17-Scid mice (n = 10 mice/group) were treated with vehicle, 150 kBq/kg PSMA-TTC every 2 weeks (Q2W) × 2, intravenous, total protein dose 0.43 mg/kg, treatment days indicated with green arrows), 200 mg/kg darolutamide (once daily, orally, treatment period indicated with a blue bar) or their combination. A, Growth curves of KUCaP-1 PDX tumors. B, KUCaP-1 tumor weights at the end of the study. Statistical analyses were performed using linear models followed by Sidak method. *, P < 0.05; **, P < 0.01; ***, P < 0.001 in comparison with vehicle on day 33; ###, P < 0.001 in comparison with darolutamide monotherapy on day 33.
Figure 5.
Figure 5.
Darolutamide impairs the PSMA-TTC–mediated induction of DNA repair genes in the ST1273 PDX prostate cancer model. A, Enrichment plot of androgen-dependent genes after treatment with 250 kBq/kg PSMA-TTC alone (FDR = 0.004) or in combination with 100 mg/kg (twice daily) darolutamide (FDR = 0.0014). B, Enrichment plot of DNA repair genes after treatment with 250 kBq/kg PSMA-TTC alone (FDR = 0.0026) or in combination with 100 mg/kg (twice daily) darolutamide (FDR = 0.29). RNA-seq analysis of tumor samples was performed 72 hours after treatment. Data were compared with the untreated group and hallmark data sets from the Molecular Signatures Database v7.1. C, Expression of the differentially expressed genes included in the DNA repair hallmark gene set used in A. Red and blue colors indicate higher or lower expression of the selected genes compared with mean expression, respectively, after treatment with 250 kBq/kg PSMA-TTC alone or in combination with 100 mg/kg darolutamide in comparison with untreated control. D, γH2AX expression as determined by IHC in untreated ST1273 tumors and in ST1273 tumors treated with 250 kBq/kg PSMA-TTC alone or in combination with 100 mg/kg darolutamide (n = 3). IHC analysis was performed 336 hours after treatment. Scale bars indicate 100 μm. E, γH2AX expression in tumor tissues shown in D, quantified with HSA software. *, P < 0.05; **, P < 0.01 in comparison with untreated control. FDR, false discovery rate.

References

    1. Mattiuzzi C, Lippi G. Current cancer epidemiology. J Epidemiol Glob Health 2019;9:217–22.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin 2020;70:7–30.
    1. 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.
    1. Nevedomskaya E, Baumgart SJ, Haendler B. Recent advances in prostate cancer treatment and drug discovery. Int J Mol Sci 2018;19:1359.
    1. Swami U, McFarland TR, Nussenzveig R, Agarwal N. Advanced prostate cancer: treatment advances and future directions. Trends Cancer 2020;6:702–15.
    1. Yap TA, Smith AD, Ferraldeschi R, Al-Lazikani B, Workman P, de Bono JS. Drug discovery in advanced prostate cancer: translating biology into therapy. Nat Rev Drug Discov 2016;15:699–718.
    1. Moilanen AM, Riikonen R, Oksala R, Ravanti L, Aho E, Wohlfahrt G, et al. . Discovery of ODM-201, a new-generation androgen receptor inhibitor targeting resistance mechanisms to androgen signaling-directed prostate cancer therapies. Sci Rep 2015;5:12007.
    1. Fizazi K, Shore N, Tammela TL, Ulys A, Vjaters E, Polyakov S, et al. . Darolutamide in nonmetastatic, castration-resistant prostate cancer. N Engl J Med 2019;380:1235–46.
    1. Hussain M, Fizazi K, Saad F, Rathenborg P, Shore N, Ferreira U, et al. . Enzalutamide in men with nonmetastatic, castration-resistant prostate cancer. N Engl J Med 2018;378:2465–74.
    1. Silver DA, Pellicer I, Fair WR, Heston WD, Cordon-Cardo C. Prostate-specific membrane antigen expression in normal and malignant human tissues. Clin Cancer Res 1997;3:81–5.
    1. Ruigrok EAM, van Weerden WM, Nonnekens J, de Jong M. The future of PSMA-targeted radionuclide therapy: an overview of recent preclinical research. Pharmaceutics 2019;11:560.
    1. Czerwinska M, Bilewicz A, Kruszewski M, Wegierek-Ciuk A, Lankoff A. Targeted radionuclide therapy of prostate cancer-from basic research to clinical perspectives. Molecules 2020;25:1743.
    1. Hammer S, Hagemann UB, Zitzmann-Kolbe S, Larsen A, Ellingsen C, Geraudie S, et al. . Preclinical efficacy of a PSMA-targeted thorium-227 conjugate (PSMA-TTC), a targeted alpha therapy for prostate cancer. Clin Cancer Res 2020;26:1985–96.
    1. Wick M, Quinn M, Mangold A, Gamez L, Diaz A, Vaught T, et al. . Establishment and characterization of a hormone dependent, PSA/PSMA positive prostate PDX model. Eur J Cancer 2016;69:S113.
    1. Yoshida T, Kinoshita H, Segawa T, Nakamura E, Inoue T, Shimizu Y, et al. . Antiandrogen bicalutamide promotes tumor growth in a novel androgen-dependent prostate cancer xenograft model derived from a bicalutamide-treated patient. Cancer Res 2005;65:9611–6.
    1. Chou TC. Theoretical basis, experimental design, and computerized simulation of synergism and antagonism in drug combination studies. Pharmacol Rev 2006;58:621–81.
    1. Reddy N, Ong GL, Behr TM, Sharkey RM, Goldenberg DM, Mattes MJ. Rapid blood clearance of mouse IgG2a and human IgG1 in many nude and nu/+ mouse strains is due to low IgG2a serum concentrations. Cancer Immunol Immunother 1998;46:25–33.
    1. Hagemann UB, Ellingsen C, Schuhmacher J, Kristian A, Mobergslien A, Cruciani V, et al. . Mesothelin-targeted thorium-227 conjugate (MSLN-TTC): preclinical evaluation of a new targeted alpha therapy for mesothelin-positive cancers. Clin Cancer Res 2019;25:4723–34.
    1. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R, et al. . New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228–47.
    1. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 2011;12:323.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014;15:550.
    1. R Core Team. R: A language and environment for statistical computing. R Foundation for Statistical Computing. Available from: .
    1. Korotkevich G, Sukhov V, Sergushichev A. Fast gene set enrichment analysis; 2019. Available from: .
    1. Liberzon A, Birger C, Thorvaldsdottir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 2015;1:417–25.
    1. Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, et al. . Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci U S A 2005;102:15545–50.
    1. Wright GL Jr, Grob BM, Haley C, Grossman K, Newhall K, Petrylak D, et al. . Upregulation of prostate-specific membrane antigen after androgen-deprivation therapy. Urology 1996;48:326–34.
    1. Meller B, Bremmer F, Sahlmann CO, Hijazi S, Bouter C, Trojan L, et al. . Alterations in androgen deprivation enhanced prostate-specific membrane antigen (PSMA) expression in prostate cancer cells as a target for diagnostics and therapy. EJNMMI Res 2015;5:66.
    1. Kranzbuhler B, Salemi S, Umbricht CA, Muller C, Burger IA, Sulser T, et al. . Pharmacological upregulation of prostate-specific membrane antigen (PSMA) expression in prostate cancer cells. Prostate 2018;78:758–65.
    1. Sugawara T, Baumgart SJ, Nevedomskaya E, Reichert K, Steuber H, Lejeune P, et al. . Darolutamide is a potent androgen receptor antagonist with strong efficacy in prostate cancer models. Int J Cancer 2019;145:1382–94.
    1. Sumanasuriya S, De Bono J. Treatment of advanced prostate cancer-a review of current therapies and future promise. Cold Spring Harb Perspect Med 2018;8:a030635.
    1. Miyahira AK, Pienta KJ, Morris MJ, Bander NH, Baum RP, Fendler WP, et al. . Meeting report from the Prostate Cancer Foundation PSMA-directed radionuclide scientific working group. Prostate 2018;78:775–89.
    1. Hofman MS, Violet J, Hicks RJ, Ferdinandus J, Thang SP, Akhurst T, et al. . [177Lu]-PSMA-617 radionuclide treatment in patients with metastatic castration-resistant prostate cancer (LuPSMA trial): a single-centre, single-arm, phase 2 study. Lancet Oncol 2018;19:825–33.
    1. Kratochwil C, Bruchertseifer F, Giesel FL, Weis M, Verburg FA, Mottaghy F, et al. . 225Ac-PSMA-617 for PSMA targeting alpha-radiation therapy of patients with metastatic castration-resistant prostate cancer. J Nucl Med 2016;57:1941–4.
    1. Sathekge M, Bruchertseifer F, Knoesen O, Reyneke F, Lawal I, Lengana T, et al. . (225)Ac-PSMA-617 in chemotherapy-naive patients with advanced prostate cancer: a pilot study. Eur J Nucl Med Mol Imaging 2019;46:129–38.
    1. Jones W, Griffiths K, Barata PC, Paller CJ. PSMA theranostics: review of the current status of PSMA-targeted imaging and radioligand therapy. Cancers 2020;12:1367.
    1. Kratochwil C, Bruchertseifer F, Rathke H, Bronzel M, Apostolidis C, Weichert W, et al. . Targeted alpha-therapy of metastatic castration-resistant prostate cancer with (225)Ac-PSMA-617: dosimetry estimate and empiric dose finding. J Nucl Med 2017;58:1624–31.
    1. Rupp NJ, Umbricht CA, Pizzuto DA, Lenggenhager D, Topfer A, Muller J, et al. . First clinico-pathological evidence of a non PSMA-related uptake mechanism for (68)Ga-PSMA-11 in salivary glands. J Nucl Med 2019;60:1270–6.
    1. Tagawa ST, Milowsky MI, Morris M, Vallabhajosula S, Christos P, Akhtar NH, et al. . Phase II study of Lutetium-177-labeled anti-prostate-specific membrane antigen monoclonal antibody J591 for metastatic castration-resistant prostate cancer. Clin Cancer Res 2013;19:5182–91.
    1. Hagemann UB, Wickstroem K, Hammer S, Bjerke RM, Zitzmann-Kolbe S, Ryan OB, et al. . Advances in precision oncology: targeted thorium-227 conjugates as a new modality in targeted alpha therapy. Cancer Biother Radiopharm 2020;35:497–510.
    1. Luckerath K, Wei L, Fendler WP, Evans-Axelsson S, Stuparu AD, Slavik R, et al. . Preclinical evaluation of PSMA expression in response to androgen receptor blockade for theranostics in prostate cancer. EJNMMI Res 2018;8:96.
    1. Evans MJ, Smith-Jones PM, Wongvipat J, Navarro V, Kim S, Bander NH, et al. . Noninvasive measurement of androgen receptor signaling with a positron-emitting radiopharmaceutical that targets prostate-specific membrane antigen. Proc Natl Acad Sci U S A 2011;108:9578–82.
    1. Hope TA, Aggarwal R, Chee B, Tao D, Greene KL, Cooperberg MR, et al. . Impact of (68)Ga-PSMA-11 PET on management in patients with biochemically recurrent prostate cancer. J Nucl Med 2017;58:1956–61.
    1. Aggarwal R, Wei X, Kim W, Small EJ, Ryan CJ, Carroll P, et al. . Heterogeneous flare in prostate-specific membrane antigen positron emission tomography tracer uptake with initiation of androgen pathway blockade in metastatic prostate cancer. Eur Urol Oncol 2018;1:78–82.
    1. Emmett L, Yin C, Crumbaker M, Hruby G, Kneebone A, Epstein R, et al. . Rapid modulation of PSMA expression by androgen deprivation: serial (68)Ga-PSMA-11 PET in men with hormone-sensitive and castrate-resistant prostate cancer commencing androgen blockade. J Nucl Med 2019;60:950–4.
    1. Rosar F, Dewes S, Ries M, Schaefer A, Khreish F, Maus S, et al. . New insights in the paradigm of upregulation of tumoral PSMA expression by androgen receptor blockade: enzalutamide induces PSMA upregulation in castration-resistant prostate cancer even in patients having previously progressed on enzalutamide. Eur J Nucl Med Mol Imaging 2020;47:687–94.
    1. Current K, Meyer C, Magyar CE, Mona CE, Almajano J, Slavik R, et al. . Investigating PSMA-targeted radioligand therapy efficacy as a function of cellular PSMA levels and intratumoral PSMA heterogeneity. Clin Cancer Res 2020;26:2946–55.
    1. Li L, Karanika S, Yang G, Wang J, Park S, Broom BM, et al. . Androgen receptor inhibitor-induced "BRCAness" and PARP inhibition are synthetically lethal for castration-resistant prostate cancer. Sci Signal 2017;10:eaam7479.
    1. Polkinghorn WR, Parker JS, Lee MX, Kass EM, Spratt DE, Iaquinta PJ, et al. . Androgen receptor signaling regulates DNA repair in prostate cancers. Cancer Discov 2013;3:1245–53.
    1. Thompson TC, Li L, Broom BM. Combining enzalutamide with PARP inhibitors: pharmaceutically induced BRCAness. Oncotarget 2017;8:93315–6.
    1. Goodwin JF, Schiewer MJ, Dean JL, Schrecengost RS, de Leeuw R, Han S, et al. . A hormone-DNA repair circuit governs the response to genotoxic insult. Cancer Discov 2013;3:1254–71.
    1. Brechbiel MW. Targeted alpha-therapy: past, present, future? Dalton Trans 2007;43:4918–28.
    1. Fizazi K, Shore N, Tammela TL, Ulys A, Vjaters E, Polyakov S, et al. . Nonmetastatic, castration-resistant prostate cancer and survival with darolutamide. N Engl J Med 2020;383:1040–9.

Source: PubMed

3
Abonnere