1. Academic Validation
  2. Discovery and pharmacological characterization of cetrelimab (JNJ-63723283), an anti-programmed cell death protein-1 (PD-1) antibody, in human cancer models

Discovery and pharmacological characterization of cetrelimab (JNJ-63723283), an anti-programmed cell death protein-1 (PD-1) antibody, in human cancer models

  • Cancer Chemother Pharmacol. 2022 Apr;89(4):515-527. doi: 10.1007/s00280-022-04415-5.
Nikki DeAngelis # 1 Catherine Ferrante # 1 Gordon Powers 2 Jocelyn Sendecki 3 Bethany Mattson 1 Darlene Pizutti 1 Kathryn Packman 1 Weirong Wang 4 Kevin Trouba 5 Rupesh Nanjunda 2 John Wheeler 2 Ray Brittingham 2 Sheng-Jiun Wu 2 Jinquan Luo 2 Matthew V Lorenzi 1 Raluca I Verona 6
Affiliations

Affiliations

  • 1 Oncology Therapeutic Area, Janssen Research & Development, Spring House, PA, USA.
  • 2 Biologics Research, Janssen Research & Development, Spring House, PA, USA.
  • 3 Translational Medicine and Early Development Statistics, Janssen Research & Development, Spring House, PA, USA.
  • 4 Biologics Development Sciences, Janssen Research & Development, Spring House, PA, USA.
  • 5 Nonclinical Safety, Janssen Research & Development, Spring House, Pennsylvania, USA.
  • 6 Oncology Therapeutic Area, Janssen Research & Development, Spring House, PA, USA. rverona@its.jnj.com.
  • # Contributed equally.
Abstract

Purpose: Preclinical characterization of cetrelimab (JNJ-63723283), a fully humanized immunoglobulin G4 kappa monoclonal antibody targeting programmed cell death protein-1 (PD-1), in human Cancer models.

Methods: Cetrelimab was generated by phage panning against human and cynomolgus monkey (cyno) PD-1 extracellular domains (ECDs) and affinity maturation. Binding to primate and rodent PD-1 ECDs, transfected and endogenous cell-surface PD-1, and inhibition of ligand binding were measured. In vitro activity was evaluated using cytomegalovirus recall, mixed lymphocyte reaction, staphylococcal enterotoxin B stimulation, and Jurkat-PD-1 nuclear factor of activated T cell reporter assays. In vivo activity was assessed using human PD-1 knock-in mice implanted with MC38 tumors and a lung patient-derived xenograft (PDX) model (LG1306) using CD34 cord-blood-humanized NSG mice. Pharmacodynamics, toxicokinetics, and safety were assessed in cynos following single and/or repeat intravenous dosing.

Results: Cetrelimab showed high affinity binding to human (1.72 nM) and cyno (0.90 nM) PD-1 and blocked binding of programmed death-ligand 1 (PD-L1; inhibitory concentration [IC] 111.7 ng/mL) and PD-L2 (IC 138.6 ng/mL). Cetrelimab dose-dependently increased T cell-mediated cytokine production and stimulated cytokine expression. Cetrelimab 10 mg/kg reduced mean MC38 tumor volume in PD-1 knock-in mice at Day 21 (P < 0.0001) versus control. In a PDX lung model, 10 mg/kg cetrelimab (every 5 days for six cycles) increased frequency of peripheral T cells and reduced (P < 0.05) mean tumor volume versus control. Activity was consistent with that of established PD-1 inhibitors. Cetrelimab dosing was well tolerated in cynos and mean drug exposure increase was dose-dependent.

Conclusion: Cetrelimab potently inhibits PD-1 in vitro and in vivo, supporting its clinical evaluation.

Keywords

Antitumor activity; Immune checkpoint inhibitor; Programmed cell death protein-1; T cell function; Toxicokinetic assessment.

Figures
Products