Dose escalation results from a first-in-human, phase 1 study of glucocorticoid-induced TNF receptor-related protein agonist AMG 228 in patients with advanced solid tumors

Ben Tran, Richard D Carvajal, Aurelien Marabelle, Sandip Pravin Patel, Patricia M LoRusso, Erik Rasmussen, Gloria Juan, Vijay V Upreti, Courtney Beers, Gataree Ngarmchamnanrith, Patrick Schöffski, Ben Tran, Richard D Carvajal, Aurelien Marabelle, Sandip Pravin Patel, Patricia M LoRusso, Erik Rasmussen, Gloria Juan, Vijay V Upreti, Courtney Beers, Gataree Ngarmchamnanrith, Patrick Schöffski

Abstract

Background: This open-label, first-in-human, phase 1 study evaluated the safety, pharmacokinetics, pharmacodynamics, and maximum tolerated dose (MTD) of AMG 228, an agonistic human IgG1 monoclonal antibody targeting glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR), in patients with refractory advanced solid tumors.

Methods: AMG 228 was administered intravenously every 3 weeks (Q3W). Dose escalation was in two stages: single-patient cohorts (3, 9, 30, and 90 mg), followed by "rolling six" design (n = 2-6; 180, 360, 600, 900, and 1200 mg). Primary endpoints included incidence of dose-limiting toxicities (DLTs), AEs, and pharmacokinetics. Additional endpoints were objective response and pharmacodynamic response.

Results: Thirty patients received AMG 228, which was well tolerated up to the maximum planned dose (1200 mg). No DLTs occurred; the MTD was not reached. The most common treatment-related AEs were fatigue (13%), infusion-related reaction (7%), pyrexia (7%), decreased appetite (7%), and hypophosphatemia (7%). Two patients had binding anti-AMG 228 antibodies (one at baseline); no neutralizing antibodies were detected. AMG 228 exhibited target-mediated drug disposition, and serum exposure was approximately dose proportional at 180-1200 mg and greater than dose proportional at 3-1200 mg. Doses > 360 mg Q3W achieved serum trough coverage for 95% in vitro GITR occupancy. Despite GITR coverage in peripheral blood and tumor biopsies, there was no evidence of T-cell activation or anti-tumor activity.

Conclusions: In patients with advanced solid tumors, AMG 228 Q3W was tolerable up to the highest tested dose (1200 mg), exhibited favorable pharmacokinetics, and provided target coverage indicating a pharmacokinetic profile appropriate for longer intervals. However, there was no evidence of T-cell activation or anti-tumor activity with AMG 228 monotherapy.

Trial registration: ClinicalTrials.gov, NCT02437916 .

Keywords: Agonistic antibody; Antibodies, monoclonal; Clinical trial, phase 1; Dose, maximum tolerated; Glucocorticoid-induced TNFR-related protein.

Conflict of interest statement

Ethics approval and consent to participate

All procedures were approved by the following institutional review boards or ethics committees: Commissie Medische Ethiek van de Universitaire Ziekenhuizen KU Leuven; Ethikkommision; Comité de Protection des Personnes Ile de France; Columbia University IRB; UCSD Human Research Protections Program; WIRB; and Melbourne Health Human Research Ethics Committee.

Consent for publication

All patients provided written, informed consent.

Competing interests

BT has received a grant, consulting fees, and honoraria from Amgen Inc. RDC is a consultant for AstraZeneca, Bristol-Myers Squibb, Castle Biosciences, Foundation Medicine, Immunocore, Incyte, Merck, Novartis, and Roche/Genentech, has served on clinical advisory boards for Aura Biosciences and Rgenix, and has served on a scientific advisory board for Chimeron. SPP has received research support from Amgen Inc. ER, GJ, VVU, and GN are employees of and own stock in Amgen Inc. At the time of this study, CB was an employee of Amgen Inc. AM, PML, and PS have no conflicts to disclose.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Figures

Fig. 1
Fig. 1
Mean (± SD) pharmacokinetic profile of AMG 228 following intravenous administration of AMG 228 every 3 weeks. Blood samples were collected predose, at the end of infusion, and postdose over the 3-week dosing interval during treatment cycles
Fig. 2
Fig. 2
Best change from baseline in the sum of longest diameters of target lesions. CRC, colorectal cancer; HirPD, immune-related progressive disease; irSD, immune-related stable disease. aPatient experienced clinical progression with new brain metastasis (off schedule scan) after the primary lesions showed decreases in size. bBecause patient’s only postbaseline scan was an abdominal CT. CRC, colorectal cancer; NSCLC, non–small-cell lung cancer; SCCHN, squamous cell carcinoma of the head and neck; TCC, transitional carcinoma of the bladder
Fig. 3
Fig. 3
Pharmacodynamic assessment of target coverage and T cell activation. a flow cytometry analysis of GITR expression among peripheral blood Tregs pretreatment and posttreatment. b immunohistochemistry assessment of GITR expression and T cell activation in tumor biopsies pretreatment and posttreatment. c immunohistochemistry evidence of immune modulation following treatment (cycle 3, day 43) versus screening in a colorectal cancer biopsy. aLines not visible for 7 patients due to lack of GITR+ cells at pretreatment

References

    1. Nocentini G, Giunchi L, Ronchetti S, Krausz LT, Bartoli A, Moraca R, Migliorati G, Riccardi C. A new member of the tumor necrosis factor/nerve growth factor receptor family inhibits T cell receptor-induced apoptosis. Proc Natl Acad Sci U S A. 1997;94:6216–6221. doi: 10.1073/pnas.94.12.6216.
    1. Kwon B, Yu KY, Ni J, Yu GL, Jang IK, Kim YJ, Xing L, Liu D, Wang SX, Kwon BS. Identification of a novel activation-inducible protein of the tumor necrosis factor receptor superfamily and its ligand. J Biol Chem. 1999;274:6056–6061. doi: 10.1074/jbc.274.10.6056.
    1. Gurney AL, Marsters SA, Huang RM, Pitti RM, Mark DT, Baldwin DT, Gray AM, Dowd AD, Brush AD, Heldens AD, Schow AD, Goddard AD, Wood WI, Baker KP, Godowski PJ, Ashkenazi A. Identification of a new member of the tumor necrosis factor family and its receptor, a human ortholog of mouse GITR. Curr Biol. 1999;9:215–218. doi: 10.1016/S0960-9822(99)80093-1.
    1. Stephens GL, McHugh RS, Whitters MJ, Young DA, Luxenberg D, Carreno BM, Collins M, Shevach EM. Engagement of glucocorticoid-induced TNFR family-related receptor on effector T cells by its ligand mediates resistance to suppression by CD4+CD25+ T cells. J Immunol. 2004;173:5008–5020. doi: 10.4049/jimmunol.173.8.5008.
    1. Esparza EM, Arch RH. Glucocorticoid-induced TNF receptor functions as a costimulatory receptor that promotes survival in early phases of T cell activation. J Immunol. 2005;174:7869–7874. doi: 10.4049/jimmunol.174.12.7869.
    1. Tone M, Tone Y, Adams E, Yates SF, Frewin MR, Cobbold SP, Waldmann H. Mouse glucocorticoid-induced tumor necrosis factor receptor ligand is costimulatory for T cells. Proc Natl Acad Sci U S A. 2003;100:15059–15064. doi: 10.1073/pnas.2334901100.
    1. Ko K, Yamazaki S, Nakamura K, Nishioka T, Hirota K, Yamaguchi T, Shimizu J, Nomura T, Chiba T, Sakaguchi S. Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+CD25+CD4+ regulatory T cells. J Exp Med. 2005;202:885–891. doi: 10.1084/jem.20050940.
    1. Coe D, Begom S, Addey C, White M, Dyson J, Chai JG. Depletion of regulatory T cells by anti-GITR mAb as a novel mechanism for cancer immunotherapy. Cancer Immunol Immunother. 2010;59:1367–1377. doi: 10.1007/s00262-010-0866-5.
    1. Mahne AE, Mauze S, Joyce-Shaikh B, Xia J, Bowman EP, Beebe AM, Cua DJ, Jain R. Dual roles for regulatory T-cell depletion and costimulatory signaling in agonistic GITR targeting for tumor immunotherapy. Cancer Res. 2017;77:1108–1118. doi: 10.1158/0008-5472.CAN-16-0797.
    1. Sukumar S, Wilson DC, Yu Y, Wong J, Naravula S, Ermakov G, Riener R, Bhagwat B, Necheva AS, Grein J, Churakova T, Mangadu R, Georgiev P, Manfra D, Pinheiro EM, Sriram V, Bailey WJ, Herzyk D, McClanahan TK, Willingham A, Beebe AM, Sadekova S. Characterization of MK-4166, a clinical agonistic antibody that targets human GITR and inhibits the generation and suppressive effects of T regulatory cells. Cancer Res. 2017;77:4378–4388. doi: 10.1158/0008-5472.CAN-16-1439.
    1. Knee DA, Hewes B, Brogdon JL. Rationale for anti-GITR cancer immunotherapy. Eur J Cancer. 2016;67:1–10. doi: 10.1016/j.ejca.2016.06.028.
    1. Skolnik JM, Barrett JS, Jayaraman B, Patel D, Adamson PC. Shortening the timeline of pediatric phase I trials: the rolling six design. J Clin Oncol. 2008;26:190–195. doi: 10.1200/JCO.2007.12.7712.
    1. Neuenschwander B, Branson M, Gsponer T. Critical aspects of the Bayesian approach to phase I cancer trials. Stat Med. 2008;27:2420–2439. doi: 10.1002/sim.3230.
    1. Wolchok JD, Hoos A, O'Day S, Weber JS, Hamid O, Lebbe C, Maio M, Binder M, Bohnsack O, Nichol G, Humphrey R, Hodi FS. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. Clin Cancer Res. 2009;15:7412–7420. doi: 10.1158/1078-0432.CCR-09-1624.
    1. Koon HB, Shepard DR, Merghoub T, Schaer DA, Sirard CA, Wolchok JD. First-in-human phase 1 single-dose study of TRX-518, an anti-human glucocorticoid-induced tumor necrosis factor receptor (GITR) monoclonal antibody in adults with advanced solid tumors. J Clin Oncol. 2016;34:abstr 3017. doi: 10.1200/JCO.2016.34.15_suppl.3017.
    1. Siu LL, Steeghs N, Meniawy T, Joerger M, Spratlin JL, Rottey S, Nagrial A, Cooper A, Meier R, Guan X, Phillips P, Bajaj G, Gokemeijer J, Korman AJ, Aung KL, Carlino MS. Preliminary results of a phase I/IIa study of BMS-986156 (glucocorticoid-induced tumor necrosis factor receptor-related gene [GITR] agonist), alone and in combination with nivolumab in pts with advanced solid tumors. J Clin Oncol. 2017;35:104. doi: 10.1200/JCO.2017.35.15_suppl.104.
    1. Bulliard Y, Jolicoeur R, Windman M, Rue SM, Ettenberg S, Knee DA, Wilson NS, Dranoff G, Brogdon JL. Activating fc gamma receptors contribute to the antitumor activities of immunoregulatory receptor-targeting antibodies. J Exp Med. 2013;210:1685–1693. doi: 10.1084/jem.20130573.
    1. Romano E, Kusio-Kobialka M, Foukas PG, Baumgaertner P, Meyer C, Ballabeni P, Michielin O, Weide B, Romero P, Speiser DE. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A. 2015;112:6140–6145. doi: 10.1073/pnas.1417320112.
    1. Ascierto PA, Del Vecchio M, Robert C, Mackiewicz A, Chiarion-Sileni V, Arance A, Lebbe C, Bastholt L, Hamid O, Rutkowski P, McNeil C, Garbe C, Loquai C, Dreno B, Thomas L, Grob JJ, Liszkay G, Nyakas M, Gutzmer R, Pikiel J, Grange F, Hoeller C, Ferraresi V, Smylie M, Schadendorf D, Mortier L, Svane IM, Hennicken D, Qureshi A, Maio M. Ipilimumab 10 mg/kg versus ipilimumab 3 mg/kg in patients with unresectable or metastatic melanoma: a randomised, double-blind, multicentre, phase 3 trial. Lancet Oncol. 2017;18:611–622. doi: 10.1016/S1470-2045(17)30231-0.
    1. Zappasodi R, Li Y, Abu-Akeel M, Qi J, Wong P, Sirard C, Postow M, Schaer DA, Newman W, Koon H, Velcheti V, Callahan MK, Wolchok JD, Merghoub T. Intratumor and peripheral Treg modulation as a pharmacodynamic biomarker of the GITR agonist antibody TRX-518 in the first-in-human trial. Cancer Res. 2017;77:abstr CT108. doi: 10.1158/1538-7445.AM2017-CT018.
    1. Marin-Acevedo JA, Dholaria B, Soyano AE, Knutson KL, Chumsri S, Lou Y. Next generation of immune checkpoint therapy in cancer: new developments and challenges. J Hematol Oncol. 2018;11:39. doi: 10.1186/s13045-018-0582-8.
    1. Curti BD, Kovacsovics-Bankowski M, Morris N, Walker E, Chisholm L, Floyd K, Walker J, Gonzalez I, Meeuwsen T, Fox BA, Moudgil T, Miller W, Haley D, Coffey T, Fisher B, Delanty-Miller L, Rymarchyk N, Kelly T, Crocenzi T, Bernstein E, Sanborn R, Urba WJ, Weinberg AD. OX40 is a potent immune-stimulating target in late-stage cancer patients. Cancer Res. 2013;73:7189–7198. doi: 10.1158/0008-5472.CAN-12-4174.

Source: PubMed

3
Suscribir