Preclinical characterization of dostarlimab, a therapeutic anti-PD-1 antibody with potent activity to enhance immune function in in vitro cellular assays and in vivo animal models

Sujatha Kumar, Srimoyee Ghosh, Geeta Sharma, Zebin Wang, Marilyn R Kehry, Margaret H Marino, Tamlyn Y Neben, Sharon Lu, Shouqi Luo, Simon Roberts, Sridhar Ramaswamy, Hadi Danaee, David Jenkins, Sujatha Kumar, Srimoyee Ghosh, Geeta Sharma, Zebin Wang, Marilyn R Kehry, Margaret H Marino, Tamlyn Y Neben, Sharon Lu, Shouqi Luo, Simon Roberts, Sridhar Ramaswamy, Hadi Danaee, David Jenkins

Abstract

Inhibitors of programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have dramatically changed the treatment landscape for patients with cancer. Clinical activity of anti-PD-(L)1 antibodies has resulted in increased median overall survival and durable responses in patients across selected tumor types. To date, 6 PD-1 and PD-L1, here collectively referred to as PD-(L)1, pathway inhibitors are approved by the US Food and Drug Administration for clinical use. The availability of multiple anti-PD-(L)1 antibodies provides treatment and dosing regimen choice for patients with cancer. Here, we describe the nonclinical characterization of dostarlimab (TSR-042), a humanized anti-PD-1 antibody, which binds with high affinity to human PD-1 and effectively inhibits its interaction with its ligands, PD-L1 and PD-L2. Dostarlimab enhanced effector T-cell functions, including cytokine production, in vitro. Since dostarlimab does not bind mouse PD-1, its single-agent antitumor activity was evaluated using humanized mouse models. In this model system, dostarlimab demonstrated antitumor activity as assessed by tumor growth inhibition, which was associated with increased infiltration of immune cells. Single-dose and 4-week repeat-dose toxicology studies in cynomolgus monkeys indicated that dostarlimab was well tolerated. In a clinical setting, based on data from the GARNET trial, dostarlimab (Jemperli) was approved for the treatment of adult patients with mismatch repair-deficient recurrent or advanced endometrial cancer that had progressed on or following prior treatment with a platinum-containing regimen. Taken together, these data demonstrate that dostarlimab is a potent anti-PD-1 receptor antagonist, with properties that support its continued clinical investigation in patients with cancer.

Keywords: Anti-PD-1 antibody; TSR-042; cancer; characterization; dostarlimab; immune checkpoint; solid tumors.

Conflict of interest statement

All authors are present or former employees of GlaxoSmithKline or AnaptysBio, Inc.

Figures

Figure 1.
Figure 1.
Dostarlimab binding to native PD-1 receptor expressed on human and cynomolgus monkey CD3+ T cells. Dostarlimab binds with high affinity to native human and cynomolgus monkey PD-1 expressed on the surface of CD3+ T cells. Occupancy of dostarlimab on cynomolgus monkey and human CD3+ T cells was assessed by preincubating PBMCs from either (a) cynomolgus monkey or (b) human donors with dostarlimab at varying concentrations and then incubating with either a saturating concentration (20 μg/mL) of dostarlimab or IgG4 isotype control before detection with either FITC-anti-CD3 or PE-anti-IgG4 antibodies and analysis by flow cytometry. The number of CD3+/IgG4+ cells was quantified, and percentage receptor occupancy was calculated as described in Materials and Methods. FITC: fluorescein isothiocyanate; IgG: immunoglobulin G; PBMC: peripheral blood mononuclear cells; PD-1: programmed cell death protein 1; PE: phycoerythrin
Figure 1.
Figure 1.
Dostarlimab binding to native PD-1 receptor expressed on human and cynomolgus monkey CD3+ T cells. Dostarlimab binds with high affinity to native human and cynomolgus monkey PD-1 expressed on the surface of CD3+ T cells. Occupancy of dostarlimab on cynomolgus monkey and human CD3+ T cells was assessed by preincubating PBMCs from either (a) cynomolgus monkey or (b) human donors with dostarlimab at varying concentrations and then incubating with either a saturating concentration (20 μg/mL) of dostarlimab or IgG4 isotype control before detection with either FITC-anti-CD3 or PE-anti-IgG4 antibodies and analysis by flow cytometry. The number of CD3+/IgG4+ cells was quantified, and percentage receptor occupancy was calculated as described in Materials and Methods. FITC: fluorescein isothiocyanate; IgG: immunoglobulin G; PBMC: peripheral blood mononuclear cells; PD-1: programmed cell death protein 1; PE: phycoerythrin
Figure 2.
Figure 2.
Dostarlimab disrupts binding of PD-1 to its ligands. Dostarlimab potently blocks the interaction of PD-1 with its ligands PD-L1 and PD-L2 and inhibits PD-1/PD-L1-driven signaling. (a) Dostarlimab inhibits the binding of labeled PD-L1-mFc-DyL650 and PD-L2-mFc-DyL650 to CHO-K1 cells stably expressing recombinant human PD-1. (b) Dostarlimab relieves PD-1/PD-L1-mediated repression of an NFAT-luciferase reporter downstream of TCR signaling. For a detailed description of the assay, see Materials and Methods. CHO: Chinese hamster ovary; EC50: half-maximal effective concentration; IC50: half-maximal inhibitory concentration; NFAT: nuclear factor of activated T cells response element; PD-1: programmed cell death protein 1; PD-L1(2): programmed cell death ligand 1(2); RLU: relative light units; TCR: T-cell receptor
Figure 2.
Figure 2.
Dostarlimab disrupts binding of PD-1 to its ligands. Dostarlimab potently blocks the interaction of PD-1 with its ligands PD-L1 and PD-L2 and inhibits PD-1/PD-L1-driven signaling. (a) Dostarlimab inhibits the binding of labeled PD-L1-mFc-DyL650 and PD-L2-mFc-DyL650 to CHO-K1 cells stably expressing recombinant human PD-1. (b) Dostarlimab relieves PD-1/PD-L1-mediated repression of an NFAT-luciferase reporter downstream of TCR signaling. For a detailed description of the assay, see Materials and Methods. CHO: Chinese hamster ovary; EC50: half-maximal effective concentration; IC50: half-maximal inhibitory concentration; NFAT: nuclear factor of activated T cells response element; PD-1: programmed cell death protein 1; PD-L1(2): programmed cell death ligand 1(2); RLU: relative light units; TCR: T-cell receptor
Figure 3.
Figure 3.
Functional activity of dostarlimab in primary T-cell–based assays. The functional activity of dostarlimab was evaluated in several in vitro assays, including (a) allogeneic MLR, (b) SEB activation, and (c) flu/PPD/TT activation assays. In the MLR assay, dendritic cells and allogeneic CD4+ T cells were incubated in the presence of dostarlimab or isotype control at the indicated concentrations for 48 hours, and activation of T cells was determined by quantifying the level of IL-2 secretion. Representative data from 4 donor combinations is shown in (a). PBMCs from 5 and 3 donors, respectively, were incubated as described in Materials and Methods with either SEB or flu/PPD/TT in the presence of dostarlimab or isotype control at the indicated concentrations for 72 hours and 5 days, respectively. Activation of T cells was determined by measuring the level of IL-2 or IFN-γ secretion. Results shown are the normalized mean ± SEM values across all donors. IFN: interferon; IL: interleukin; MLR: mixed lymphocyte reaction; PBMC: peripheral blood mononuclear cell; PPD: purified protein derivative from Mycobacterium tuberculosis tuberculin; SEB: staphylococcal enterotoxin B; SEM: standard error of the mean; TT: tetanus toxoid antigen
Figure 3.
Figure 3.
Functional activity of dostarlimab in primary T-cell–based assays. The functional activity of dostarlimab was evaluated in several in vitro assays, including (a) allogeneic MLR, (b) SEB activation, and (c) flu/PPD/TT activation assays. In the MLR assay, dendritic cells and allogeneic CD4+ T cells were incubated in the presence of dostarlimab or isotype control at the indicated concentrations for 48 hours, and activation of T cells was determined by quantifying the level of IL-2 secretion. Representative data from 4 donor combinations is shown in (a). PBMCs from 5 and 3 donors, respectively, were incubated as described in Materials and Methods with either SEB or flu/PPD/TT in the presence of dostarlimab or isotype control at the indicated concentrations for 72 hours and 5 days, respectively. Activation of T cells was determined by measuring the level of IL-2 or IFN-γ secretion. Results shown are the normalized mean ± SEM values across all donors. IFN: interferon; IL: interleukin; MLR: mixed lymphocyte reaction; PBMC: peripheral blood mononuclear cell; PPD: purified protein derivative from Mycobacterium tuberculosis tuberculin; SEB: staphylococcal enterotoxin B; SEM: standard error of the mean; TT: tetanus toxoid antigen
Figure 4.
Figure 4.
Antitumor and pharmacodynamic activity of dostarlimab monotherapy in a humanized mouse model of non–small cell lung cancer. (a) Study schema: dostarlimab or IgG4 isotype control (200 µg/mouse, twice weekly) was dosed to established A549 tumors (80–120 mm3) grown as xenografts in humanized NOG-EXL mice (Taconic), and (b) antitumor activity and (c) pharmacodynamic effects in the tumor. Human CD4+, CD8+, NK/NKT, and activated CD8+ (GZMB, CD8+) cells in the tumors were quantified by flow cytometry as percentage of human CD45+ cells. *p < .05 (unpaired t test). PD-1: programmed cell death protein 1; CD-8: cluster of differentiation-8; GZMB: Granzyme B; IgG: immunoglobulin G; NK: natural killer; Treg: regulatory T cells
Figure 4.
Figure 4.
Antitumor and pharmacodynamic activity of dostarlimab monotherapy in a humanized mouse model of non–small cell lung cancer. (a) Study schema: dostarlimab or IgG4 isotype control (200 µg/mouse, twice weekly) was dosed to established A549 tumors (80–120 mm3) grown as xenografts in humanized NOG-EXL mice (Taconic), and (b) antitumor activity and (c) pharmacodynamic effects in the tumor. Human CD4+, CD8+, NK/NKT, and activated CD8+ (GZMB, CD8+) cells in the tumors were quantified by flow cytometry as percentage of human CD45+ cells. *p < .05 (unpaired t test). PD-1: programmed cell death protein 1; CD-8: cluster of differentiation-8; GZMB: Granzyme B; IgG: immunoglobulin G; NK: natural killer; Treg: regulatory T cells
Figure 5.
Figure 5.
Antitumor activity of dostarlimab monotherapy in a humanized mouse model of breast cancer. (a) Study schema; dostarlimab or IgG4 isotype control (200 μg/mouse, twice weekly) was dosed to already established MDA-MB-436 tumors (80–120 mm3) grown as xenografts in humanized NOG-EXL mice (Taconic), and (b) antitumor activity. PD-1: programmed cell death protein 1; IgG: immunoglobulin G
Figure 5.
Figure 5.
Antitumor activity of dostarlimab monotherapy in a humanized mouse model of breast cancer. (a) Study schema; dostarlimab or IgG4 isotype control (200 μg/mouse, twice weekly) was dosed to already established MDA-MB-436 tumors (80–120 mm3) grown as xenografts in humanized NOG-EXL mice (Taconic), and (b) antitumor activity. PD-1: programmed cell death protein 1; IgG: immunoglobulin G
Figure 6.
Figure 6.
Concentration–time profiles of single-dose dostarlimab. Mean serum concentration–time profiles of dostarlimab after single intravenous infusion of dostarlimab to male and female cynomolgus monkeys. Concentration–time profiles were plotted through the first post-dose time point that was below the limit of quantification, if applicable. The lower limit of quantification was 31.0 ng/mL

References

    1. Ghahremanloo A, Soltani A, Modaresi SMS, Hashemy SI.. Recent advances in the clinical development of immune checkpoint blockade therapy. Cell Oncol (Dordr). 2019;42(5):609–12. doi:10.1007/s13402-019-00456-w.
    1. Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood. 2009;114(8):1545–52. doi:10.1182/blood-2009-03-206672.
    1. Chen L, Han X. Anti-PD-1/PD-L1 therapy of human cancer: past, present, and future. J Clin Invest. 2015;125(9):3384–91. doi:10.1172/JCI80011.
    1. Centanni M, Moes D, Troconiz IF, Ciccolini J, van Hasselt JGC. Clinical pharmacokinetics and pharmacodynamics of immune checkpoint inhibitors. Clin Pharmacokinet. 2019;58(7):835–57. doi:10.1007/s40262-019-00748-2.
    1. US Food and Drug Administration . Opdivo (nivolumab) label. [accessed 2020 Sept 5]. .
    1. US Food and Drug Administration . Keytruda (pembrolizumab) label. [accessed 2020 Sept 5]. .
    1. US Food and Drug Administration . Tecentriq (atezolizumab) label. [accessed 2020 Sept 5]. .
    1. US Food and Drug Administration . Bavencio (avelumab) label. [accessed 2020 Sept 5]. .
    1. US Food and Drug Administration . Imfinzi (durvalumab) label. [accessed 2020 Sept 5]. .
    1. US Food and Drug Administration . Libtayo (cemiplimab) label. [accessed 2020 Sept 5]. .
    1. Bowers PM, Horlick RA, Neben TY, Toobian RM, Tomlinson GL, Dalton JL, Jones HA, Chen A, Altobell L, Zhang X, et al. Coupling mammalian cell surface display with somatic hypermutation for the discovery and maturation of human antibodies. Proc Natl Acad Sci U S A. 2011;108(51):20455–60. doi:10.1073/pnas.1114010108.
    1. Horlick RA, Macomber JL, Bowers PM, Neben TY, Tomlinson GL, Krapf IP, Dalton JL, Verdina P, King DJ. Simultaneous surface display and secretion of proteins from mammalian cells facilitate efficient in vitro selection and maturation of antibodies. J Biol Chem. 2013;288(27):19861–69. doi:10.1074/jbc.M113.452482.
    1. Oaknin A. Preliminary safety, efficacy, and pharmacokinetic/pharmacodynamic characterization from GARNET, a phase 1/2 clinical trial of the anti-PD-1 monoclonal antibody, dostarlimab, in patients with recurrent or advanced MSI-H and MSS endometrial cancer (EC). Paper presented at: SGO Annual Meeting on Women’s Cancer; 2019 Mar 16–19; Honolulu, HI.
    1. Perez DSJ, Pikiel J, Barretina-Ginesta MP, Trigo J, Guo W, Lu S, Jenkins D, Jen KY, Danaee H, Dunlap S, et al. GARNET: preliminary safety, efficacy, pharmacokinetic, and biomarker characterization from a phase 1 clinical trial of TSR-042 (anti-PD-1 monoclonal antibody) in patients with recurrent/advanced NSCLC. Paper presented at: SITC 2018 Annual Meeting; 2018 Nov 7–11; Washington, DC.
    1. Oaknin A, Tinker AV, Gilbert L, Samouëlian V, Mathews C, Brown J, Barretina-Ginesta MP, Moreno C, Gravina A, Abdeddaim C, et al. Clinical activity and safety of the anti-programmed death 1 monoclonal antibody dostarlimab for patients with recurrent or advanced mismatch repair-deficient endometrial cancer: a nonrandomized phase 1 clinical trial. JAMA Oncol. 2020;6:1766. doi:10.1001/jamaoncol.2020.4515.
    1. US Food and Drug Administration . Jemperli (dostarlimab) label. [accessed 2020June 10]. .
    1. Chenoweth AM, Wines BD, Anania JC, Mark Hogarth P. Harnessing the immune system via FcgammaR function in immune therapy: a pathway to next-gen mAbs. Immunol Cell Biol. 2020;98(4):287–304. doi:10.1111/imcb.12326.
    1. Crescioli S, Correa I, Karagiannis P, Davies AM, Sutton BJ, Nestle FO, Karagiannis SN. IgG4 characteristics and functions in cancer immunity. Curr Allergy Asthma Rep. 2016;16(1):7. doi:10.1007/s11882-015-0580-7.
    1. Bruhns P, Iannascoli B, England P, Mancardi DA, Fernandez N, Jorieux S, Daëron M. Specificity and affinity of human Fcgamma receptors and their polymorphic variants for human IgG subclasses. Blood. 2009;113(16):3716–25. doi:10.1182/blood-2008-09-179754.
    1. Stebbings R, Findlay L, Edwards C, Eastwood D, Bird C, North D, Mistry Y, Dilger P, Liefooghe E, Cludts I, et al. “Cytokine storm” in the phase I trial of monoclonal antibody TGN1412: better understanding the causes to improve preclinical testing of immunotherapeutics. J Immunol. 2007;179(5):3325–31. doi:10.4049/jimmunol.179.5.3325.
    1. De La Rochere P, Guil-Luna S, Decaudin D, Azar G, Sidhu SS, Piaggio E. Humanized mice for the study of immuno-oncology. Trends Immunol. 2018;39(9):748–63. doi:10.1016/j.it.2018.07.001.
    1. Jiang D, England C, Ehlerding E, Graves S, Hernandez R, Cai W. Targeting programmed cell death 1 receptor (PD-1) expression in lung cancer using a humanized mouse model. J Nucl Med. 2017;58:179–86.
    1. Ito R, Takahashi T, Katano I, Kawai K, Kamisako T, Ogura T, Ida-Tanaka M, Suemizu H, Nunomura S, Ra C, et al. Establishment of a human allergy model using human IL-3/GM-CSF-transgenic NOG mice. J Immunol. 2013;191(6):2890–99. doi:10.4049/jimmunol.1203543.
    1. Dirks NL, Meibohm B. Population pharmacokinetics of therapeutic monoclonal antibodies. Clin Pharmacokinet. 2010;49(10):633–59. doi:10.2165/11535960-000000000-00000.
    1. Betts A, Keunecke A, van Steeg TJ, van der Graaf PH, Avery LB, Jones H, Berkhout J. Linear pharmacokinetic parameters for monoclonal antibodies are similar within a species and across different pharmacological targets: a comparison between human, cynomolgus monkey and hFcRn Tg32 transgenic mouse using a population-modeling approach. MAbs. 2018;10(5):751–64. doi:10.1080/19420862.2018.1462429.
    1. Fessas P, Lee H, Ikemizu S, Janowitz T. A molecular and preclinical comparison of the PD-1-targeted T-cell checkpoint inhibitors nivolumab and pembrolizumab. Semin Oncol. 2017;44(2):136–40. doi:10.1053/j.seminoncol.2017.06.002.
    1. Tan S, Zhang H, Chai Y, Song H, Tong Z, Wang Q, Qi J, Wong G, Zhu X, Liu WJ, et al. An unexpected N-terminal loop in PD-1 dominates binding by nivolumab. Nat Commun. 2017;8:14369. doi:10.1038/ncomms14369.
    1. Longoria TC, Tewari KS. Evaluation of the pharmacokinetics and metabolism of pembrolizumab in the treatment of melanoma. Expert Opin Drug Metab Toxicol. 2016;12(10):1247–53. doi:10.1080/17425255.2016.1216976.
    1. Burova E, Hermann A, Waite J, Potocky T, Lai V, Hong S, Liu M, Allbritton O, Woodruff A, Wu Q, et al. Characterization of the anti-PD-1 antibody REGN2810 and its antitumor activity in human PD-1 knock-in mice. Mol Cancer Ther. 2017;16(5):861–70. doi:10.1158/1535-7163.MCT-16-0665.
    1. Muhammed Y. The best IgG subclass for the development of therapeutic monoclonal antibody drugs and their commercial production: a review. Immunome Res. 2020;16:173.
    1. Luo W, Wang Z, Tian P, Li W. Safety and tolerability of PD-1/PD-L1 inhibitors in the treatment of non-small cell lung cancer: a meta-analysis of randomized controlled trials. J Cancer Res Clin Oncol. 2018;144(10):1851–59. doi:10.1007/s00432-018-2707-4.
    1. Weinmann SC, Pisetsky DS. Mechanisms of immune-related adverse events during the treatment of cancer with immune checkpoint inhibitors. Rheumatology (Oxford). 2019;58(suppl7):vii59–vii67. doi:10.1093/rheumatology/kez308.
    1. Sachdev JC, Patnaik A, Waypa J, Pelusi J, Beeram M, Im E, Jenkins D, McEachern K, Lu S, Guo W, et al. Safety, pharmacodynamic, and pharmacokinetic profile of TSR-042, an anti–PD–1 monoclonal antibody, in patients (pts) with advanced solid tumors. Ann Oncol. 2017;28(suppl 5):v403–v427. doi:10.1093/annonc/mdx376.050.
    1. US Food and Drug Administration . Keytruda (pembrolizumab): clinical pharmacology BLA review. [accessed 2020Feb 12]. .
    1. Bowers PM, Horlick RA, Kehry MR, Neben TY, Tomlinson GL, Altobell L, Zhang X, Macomber JL, Krapf IP, Wu BF, et al. Mammalian cell display for the discovery and optimization of antibody therapeutics. Methods. 2014;65(1):44–56. doi:10.1016/j.ymeth.2013.06.010.
    1. Abbas AK, Lichtman AH, Pillai S. Cellular and molecular immunology. 8th. Philadelphia (PA): Elsevier Saunders; 2015.

Source: PubMed

3
구독하다