Xaluritamig, a STEAP1 × CD3 XmAb 2+1 Immune Therapy for Metastatic Castration-Resistant Prostate Cancer: Results from Dose Exploration in a First-in-Human Study

William K Kelly, Daniel C Danila, Chia-Chi Lin, Jae-Lyun Lee, Nobuaki Matsubara, Patrick J Ward, Andrew J Armstrong, David Pook, Miso Kim, Tanya B Dorff, Stefanie Fischer, Yung-Chang Lin, Lisa G Horvath, Christopher Sumey, Zhao Yang, Gabor Jurida, Kristen M Smith, Jamie N Connarn, Hweixian L Penny, Julia Stieglmaier, Leonard J Appleman, William K Kelly, Daniel C Danila, Chia-Chi Lin, Jae-Lyun Lee, Nobuaki Matsubara, Patrick J Ward, Andrew J Armstrong, David Pook, Miso Kim, Tanya B Dorff, Stefanie Fischer, Yung-Chang Lin, Lisa G Horvath, Christopher Sumey, Zhao Yang, Gabor Jurida, Kristen M Smith, Jamie N Connarn, Hweixian L Penny, Julia Stieglmaier, Leonard J Appleman

Abstract

Xaluritamig (AMG 509) is a six-transmembrane epithelial antigen of the prostate 1 (STEAP1)-targeted T-cell engager designed to facilitate lysis of STEAP1-expressing cancer cells, such as those in advanced prostate cancer. This first-in-human study reports monotherapy dose exploration for patients with metastatic castration-resistant prostate cancer (mCRPC), primarily taxane pretreated. Ninety-seven patients received ≥1 intravenous dose ranging from 0.001 to 2.0 mg weekly or every 2 weeks. MTD was identified as 1.5 mg i.v. weekly via a 3-step dose. The most common treatment-related adverse events were cytokine release syndrome (CRS; 72%), fatigue (45%), and myalgia (34%). CRS occurred primarily during cycle 1 and improved with premedication and step dosing. Prostate-specific antigen (PSA) and RECIST responses across cohorts were encouraging [49% PSA50; 24% objective response rate (ORR)], with greater frequency at target doses ≥0.75 mg (59% PSA50; 41% ORR). Xaluritamig is a novel immunotherapy for prostate cancer that has shown encouraging results supporting further development.

Significance: Xaluritamig demonstrated encouraging responses (PSA and RECIST) compared with historical established treatments for patients with late-line mCRPC. This study provides proof of concept for T-cell engagers as a potential treatment for prostate cancer, validates STEAP1 as a target, and supports further clinical investigation of xaluritamig in prostate cancer. See related commentary by Hage Chehade et al., p. 20. See related article by Nolan-Stevaux et al., p. 90. This article is featured in Selected Articles from This Issue, p. 5.

©2023 The Authors; Published by the American Association for Cancer Research.

Figures

Figure 1.
Figure 1.
A, Frequency and highest-grade AEs occurring in ≥20% of patients treated with xaluritamig across all cohorts, TEAE vs. TRAEs (defined by the investigator as having reasonable possibility of being caused by xaluritamig). B, Incidence and grade of CRS (32) by cycle and dose schedule. Cohort 10 was excluded (0.1–1.0 mg), as dosing schedule was adjusted for the remaining patients after initial patients with a 10-fold dose increase in 1 step experienced DLTs. ALT, alanine aminotransferase; AST, aspartate aminotransferase.
Figure 2.
Figure 2.
A, Mean (SD) xaluritamig serum concentration vs. time profile by individual cohorts for C1. Black dotted lines represent the lower end of the minimum predicted efficacious exposure based on EC90 for xaluritamig-mediated cell killing in vitro (74 ng/mL) and the upper end of the minimum predicted efficacious exposure based on IC50 for xaluritamig-mediated mouse tumor growth inhibition in vivo (259 ng/mL). Cohort 7c was dosed weekly for C1 and then switched to every 2 weeks starting C2 and beyond, which is not captured in this analysis. At the time of the data cut, there were 94 patients with at least one postbaseline xaluritamig concentration. T-cell pharmacodynamic biomarker response through the first infusion period is dose dependent. Peripheral lymphocyte margination (B), CD8+CD69+ activated T cells as a percentage of total CD8+ T cells (C), and induction of secreted cytokines at the indicated time points after infusion (D). Lines represent median fold change or difference of population percentage from C1D1 predose values ± median average deviation. Transparent points depict individual observations. Sample sizes within each priming dose group at each time point are shown as a strip annotation across the top of each figure. EC, effective concentration; IC, inhibitory concentration.
Figure 3.
Figure 3.
Clinical activity of xaluritamig in evaluable patients. A, Best PSA percentage change from baseline. Asterisk indicates confirmed PSA responders, and dashed lines indicate PSA50 and PSA90 declines. B, Best percentage change in size of tumor target lesions. Dashed line indicates 30% reduction in tumor SLD from baseline. C, Example of patient showing response by PSA and radiographic assessments: CT scan and PSA curve over time of a heavily pretreated 65-year-old patient with stage IV prostate adenocarcinoma. Patient was enrolled into Cohort 11 (3-step 1.5 mg target dose of xaluritamig). CT scans showed three target lesions (two liver, one lymph node) and multiple nontarget lesions in the liver as well as two lymph nodes during screening. Patient achieved 99% PSA decline from baseline on C7D1 and PR (37.3% reduction of target lesions) after 2 cycles, which was confirmed at 16 weeks and maintained after 24 weeks. AEs occurred during C1 of treatment and included recurrent CRS, tinea faciei (both grade 1), rash, and worsening of back pain (both grade 2). During further treatment cycles, rash (grade 1), myalgia, and hyperkalemia (both grade 2) were reported. Patient remains on treatment at the time of publication. Red arrows indicate sites of tumor. D, Time on treatment for patients in high-dose cohorts. PSA and RECIST responses [RECIST evaluable (gray bars) and non–RECIST evaluable (white bars)] are presented for patients in high-dose cohorts. Patients whose treatment was ongoing are noted by an arrowhead. Double parallel lines (//) represent patients who have extended beyond 48 weeks: one patient is ongoing treatment at 90 weeks, one is ongoing treatment at 84 weeks, and one ended treatment at 58 weeks. NE, not evaluable; PD, progressive disease; SD, stable disease; SLD, sum of longest diameters.

References

    1. Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin 2023;73:17–48.
    1. Cancer Fact Sheets, Prostate . Lyon (France), Geneva (Switzerland): International Agency for Research on Cancer, World Health Organization; 2020 [cited 2023 Jul 3]. Available from: .
    1. Wadosky KM, Koochekpour S. Molecular mechanisms underlying resistance to androgen deprivation therapy in prostate cancer. Oncotarget 2016;7:64447–70.
    1. Cancer Stat Facts: Prostate Cancer; [about 4 screens]. [cited 2023 Jul 3]. Available from: .
    1. Turco F, Gillessen S, Cathomas R, Buttigliero C, Vogl UM. Treatment landscape for patients with castration-resistant prostate cancer: patient selection and unmet clinical needs. Res Rep Urol 2022;14:339–50.
    1. Gillessen S, Bossi A, Davis ID, de Bono J, Fizazi K, James ND, et al. . Management of patients with advanced prostate cancer-metastatic and/or castration-resistant prostate cancer: Report of the Advanced Prostate Cancer Consensus Conference (APCCC) 2022. Eur J Cancer 2023;185:178–215.
    1. Sorrentino C, Di Carlo E. Molecular targeted therapies in metastatic prostate cancer: recent advances and future challenges. Cancers (Basel) 2023;15:2885.
    1. Powers E, Karachaliou GS, Kao C, Harrison MR, Hoimes CJ, George DJ, et al. . Novel therapies are changing treatment paradigms in metastatic prostate cancer. J Hematol Oncol 2020;13:144.
    1. Merck Provides Update on Phase 3 Trials KEYNOTE-641 and KEYNOTE-789. 2023 Feb 28 [cited 2023 Jul 20] . Available from: .
    1. Powles T, Yuen KC, Gillessen S, Kadel EE, Rathkopf D, Matsubara N, et al. . Atezolizumab with enzalutamide versus enzalutamide alone in metastatic castration-resistant prostate cancer: a randomized phase 3 trial. Nat Med 2022;28:144–53.
    1. Antonarakis ES, Park SH, Goh JC, Shin SJ, Lee JL, Mehra N, et al. . Pembrolizumab plus olaparib for patients with previously treated and biomarker-unselected metastatic castration-resistant prostate cancer: the randomized, open-label, phase III KEYLYNK-010 trial. J Clin Oncol 2023;41:3839–50.
    1. Zhou S, Liu M, Ren F, Meng X, Yu J. The landscape of bispecific T cell engager in cancer treatment. Biomark Res 2021;9:38.
    1. Atallah-Yunes SA, Robertson MJ, Davé UP, Ghione P, Perna F. Novel immune-based treatments for diffuse large B-cell lymphoma: the post-CAR T cell era. Front Immunol 2022;13:901365.
    1. Kantarjian H, Stein A, Gökbuget N, Fielding AK, Schuh AC, Ribera J-M, et al. . Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med 2017;376:836–47.
    1. Nathan P, Hassel JC, Rutkowski P, Baurain J-F, Butler MO, Schlaak M, et al. . Overall survival benefit with tebentafusp in metastatic uveal melanoma. N Engl J Med 2021;385:1196–206.
    1. Ravi G, Costa LJ. Bispecific T-cell engagers for treatment of multiple myeloma. Am J Hematol 2023;98:S13–21.
    1. Tucker MD, Zhu J, Marin D, Gupta RT, Gupta S, Berry WR, et al. . Pembrolizumab in men with heavily treated metastatic castrate-resistant prostate cancer. Cancer Med 2019;8:4644–55.
    1. Lim EA, Schweizer MT, Chi KN, Aggarwal R, Agarwal N, Gulley J, et al. . Phase 1 study of safety and preliminary clinical activity of JNJ-63898081, a PSMA and CD3 bispecific antibody, for metastatic castration-resistant prostate cancer. Clin Genitourin Cancer 2023;21:366–75.
    1. De Bono JS, Fong L, Beer TM, Gao X, Geynisman DM, Burris HA III, et al. . Results of an ongoing phase 1/2a dose escalation study of HPN424, a tri-specific half-life extended PSMA-targeting T-cell engager, in patients with metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol 2021;39:5013–13.
    1. Tran B, Horvath L, Dorff T, Rettig M, Lolkema MP, Machiels JP, et al. . Results from a phase I study of AMG 160, a half-life extended (HLE), PSMA-targeted, bispecific T-cell engager (BiTE®) immune therapy for metastatic castration-resistant prostate cancer (mCRPC). Ann Oncol 2020;31 Suppl 4:S507. Abstract nr 609O.
    1. Xu M, Evans L, Bizzaro CL, Quaglia F, Verrillo CE, Li L, et al. . STEAP1–4 (six-transmembrane epithelial antigen of the prostate 1–4) and their clinical implications for prostate cancer. Cancers (Basel) 2022;14:4034.
    1. Nolan-Stevaux O. AMG 509: a novel, humanized, half-life extended, bispecific STEAP1 × CD3 T cell recruiting XmAb® 2+1 antibody [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27–28 and Jun 22–24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr DDT02-03).
    1. Danila DC, Szmulewitz RZ, Vaishampayan U, Higano CS, Baron AD, Gilbert HN, et al. . Phase I study of DSTP3086S, an antibody-drug conjugate targeting six-transmembrane epithelial antigen of prostate 1, in meta­static castration-resistant prostate cancer. J Clin Oncol 2019;37:3518–27.
    1. Bhatia V, Kamat NV, Pariva TE, Wu LT, Tsao A, Sasaki K, et al. . Targeting advanced prostate cancer with STEAP1 chimeric antigen receptor T cell and tumor-localized IL-12 immunotherapy. Nat Commun 2023;14:2041.
    1. Lee Lab, Human Biology and Clinical Research Divisions, Cancer Consortium. Boost and attack approach for metastatic prostate cancer therapy! Fred Hutch Cancer Center . Available from: .
    1. Nolan-Stevaux O, Li C, Liang L, Zhan J, Estrada J, Osgood T, et al. . AMG 509 (xaluritamig), an anti-STEAP1 XmAb 2+1 T-cell redirecting immune therapy with avidity-dependent activity against prostate cancer. Cancer Discov 2024;14:90–103.
    1. Ball K, Dovedi SJ, Vajjah P, Phipps A. Strategies for clinical dose optimization of T cell-engaging therapies in oncology. MAbs 2023;15:2181016.
    1. Project Optimus. Reforming the dose optimization and dose selection paradigm in oncology; [about 4 screens]. [cited 2023 Jul 24]. Available from: .
    1. The Human Protein Atlas. STEAP1; [about 5 screens]. [cited 2023 Jul 24]. Available from: .
    1. Dickinson MJ, Carlo-Stella C, Morschhauser F, Bachy E, Corradini P, Iacoboni G, et al. . Glofitamab for relapsed or refractory diffuse large B-cell lymphoma. N Engl J Med 2022;387:2220–31.
    1. Neuenschwander B, Branson M, Gsponer T. Critical aspects of the Bayesian approach to phase I cancer trials. Stat Med 2008;27:2420–39.
    1. Scher HI, Morris MJ, Stadler WM, Higano C, Basch E, Fizazi K, et al. . Trial design and objectives for castration-resistant prostate cancer: updated recommendations from the Prostate Cancer Clinical Trials Working Group 3. J Clin Oncol 2016;34:1402–18.
    1. Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. . Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014;124:188–95.
    1. Coiffier B, Altman A, Pui CH, Younes A, Cairo MS. Guidelines for the management of pediatric and adult tumor lysis syndrome: an evidence-based review. J Clin Oncol 2008;26:2767–78.
    1. Kuznetsova A, Brockhoff PB, Christensen RHB. lmerTest Package: tests in linear mixed effects models. J Stat Softw 2017;82:1–26.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc Series B Methodol 1995;57:289–300.

Source: PubMed

3
Abonnere