Durable blockade of PD-1 signaling links preclinical efficacy of sintilimab to its clinical benefit

Jie Wang, Keke Fei, Hua Jing, Zhihai Wu, Weiwei Wu, Shuaixiang Zhou, Haiqing Ni, Bingliang Chen, Yan Xiong, Yanpeng Liu, Bo Peng, Dechao Yu, Haiping Jiang, Junjian Liu, Jie Wang, Keke Fei, Hua Jing, Zhihai Wu, Weiwei Wu, Shuaixiang Zhou, Haiqing Ni, Bingliang Chen, Yan Xiong, Yanpeng Liu, Bo Peng, Dechao Yu, Haiping Jiang, Junjian Liu

Abstract

Blockade of immune checkpoint pathways by programmed cell death protein 1 (PD-1) antibodies has demonstrated broad clinical efficacy against a variety of malignancies. Sintilimab, a highly selective, fully human monoclonal antibody (mAb), blocks the interaction of PD-1 and its ligands and has demonstrated clinical benefit in various clinical studies. Here, we evaluated the affinity of sintilimab to human PD-1 by surface plasmon resonance and mesoscale discovery and evaluated PD-1 receptor occupancy and anti-tumor efficacy of sintilimab in a humanized NOD/Shi-scid-IL2rgamma (null) (NOG) mouse model. We also assessed the receptor occupancy and immunogenicity of sintilimab from clinical studies in humans (9 patients with advanced solid tumor and 381 patients from 4 clinical studies, respectively). Sintilimab bound to human PD-1 with greater affinity than nivolumab (Opdivo®, MDX-1106) and pembrolizumab (Keytruda®, MK-3475). The high affinity of sintilimab is explained by its distinct structural binding mode to PD-1. The pharmacokinetic behavior of sintilimab did not show any significant differences compared to the other two anti-PD-1 mAbs. In the humanized NOG mouse model, sintilimab showed superior PD-1 occupancy on circulating T cells and a stronger anti-tumor effect against NCI-H292 tumors. The strong anti-tumor response correlated with increased interferon-γ-secreting, tumor-specific CD8+ T cells, but not with CD4+ Tregs in tumor tissue. Pharmacodynamics testing indicated a sustained mean occupancy of ≥95% of PD-1 molecules on circulating T cells in patients following sintilimab infusion, regardless of infusion dose. Sintilimab infusion was associated with 0.52% (2/381 patients) of anti-drug antibodies and 0.26% (1/381 patients) neutralizing antibodies. These data validate sintilimab as a novel, safe, and efficacious anti-PD-1 mAb for cancer immunotherapy.

Keywords: PD-1; antibody; cancer immunotherapy; immunogenicity; receptor occupancy; sintilimab.

Figures

Figure 1.
Figure 1.
Higher binding affinity of sintilimab to human PD-1 compared with MDX-1106 and MK-3475. (a) Binding affinity and kinetics of anti-PD-1 mAbs to human PD-1 determined by surface plasma resonance. Sintilimab, MDX-1106 and MK-3475 were performed with human PD-1 in multi-cycle kinetics and raw data were fitted using 1:1 binding model. (b) Meso Scale Discovery analyses of the binding between human PD-1 and anti-PD-1 mAbs. (c) Individual residue of sintilimab forming the hydrophobic core was mutated to alanine. SPR analysis reveals residue mutations impair sintilimab binding to the hPD-1 protein.
Figure 2.
Figure 2.
Sintilimab showed in vitro and in vivo higher levels of PD-1 receptor occupancy. Human PBMC were stimulated to express PD-1 before incubation with sintilimab, MDX-1106 or MK-3475. Flow cytometry results showing proportions of CD3+ T cells that bind with different anti-PD-1 mAbs (a) and the mean fluorescence intensity of PD-1 (b). Data are expressed as the means ± SE of three independent experiments. (c) The effects of anti-PD-1 mAbs on mixed lymphocyte reaction (MLR) response. CD4+ T cells isolated from human PBMC were co-cultured with mature monocyte-derived dendritic cells at a ratio of 10:1 in the presence of different concentrations of anti-PD-1 mAbs. Twelve hours later, unbound mAbs was removed. Cells were co-cultured for 4 more days and the concentration of IL-2 in cultural supernatant was detected by Cisbio kit. In NOG mice reconstituted with human immune cells, PD-1 receptor occupancy on circulating CD3+ T cells 24 h (d) and 72 h (e) after anti-PD-1 mAbs intraperitoneal injection at doses of 1, 3 and 10 mg/kg (n ≥ 3 mice/group). (f) Mean (± SE) serum concentration-time profiles following a single IV administration of 10 mg/kg sintilimab, MDX-1106 or MK-3475 to hPD-1 knock-in mice (n = 3 animals per group).
Figure 3.
Figure 3.
Better tumor control and T cell response of sintilimab treatment in NCI-H292 tumor mouse model reconstituted with human immune cells. Six days after human PBMC intravenous implantation, NCI-H292 tumor cells were implanted subcutaneously into the right flanks of NOG mice. (a) Tumor growth of NCI-H292 tumor-bearing mice treated with human IgG (10 mg/kg), sintilimab (0.1, 1 and 10 mg/kg), MDX-1106 (10 mg/kg) or MK-3475 (10 mg/kg). (b) Tumor weights at the end of the study. Three weeks after NCH-H292 cells implantation, tumors were collected and total counts of CD3+ T cells per gram tumor (c), CD8+ T cells per gram tumor (d), CD8/Treg ratios (e), IFN-γ producing tumor-specific CD8+ cells per gram tumor (f) were quantified (n ≥ 5 mice/group).
Figure 4.
Figure 4.
PD-1 receptor occupancy on circulating CD3+ T cells in patients with advanced solid tumors following infusion with sintilimab. Sintilimab was administered at doses of 1, 3 or 10 mg/kg on study day 0. Blood was collected on day 0, 1, 7, 13 and 27.

References

    1. Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubata T, Yagita H, Honjo T.. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol. 1996;8:765–72. doi:10.1093/intimm/8.5.765.
    1. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. Embo J. 1992;11:3887–95.
    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. Chen L, Flies DB. Molecular mechanisms of T cell co-stimulation and co-inhibition. Nat Rev Immunol. 2013;13:227–42. doi:10.1038/nri3405.
    1. Shi Y, Su H, Song Y, Jiang W, Sun X, Qian W, Zhang W, Gao Y, Jin Z, Zhou J, et al. Safety and activity of sintilimab in patients with relapsed or refractory classical Hodgkin lymphoma (ORIENT-1): a multicentre, single-arm, phase 2 trial. Lancet Haematol. 2019;6:e12–e9. doi:10.1016/S2352-3026(18)30192-3.
    1. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–67. doi:10.1038/nature14011.
    1. Tan DSW. Changing the natural history of non-small-cell lung cancer through upfront programmed death protein 1/programmed death-ligand 1 blockade. J Clin Oncol. 2017;35:2735–36. doi:10.1200/JCO.2017.73.6215.
    1. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372:311–19. doi:10.1056/NEJMoa1411087.
    1. Zhang S, Zhang M, Wu W, Yuan Z, Tsun A, Wu M, Chen B, Li J, Miao X, Miao X, et al. Preclinical characterization of sintilimab, a fully human anti-PD-1 therapeutic monoclonal antibody for cancer. AntibodyTherapeutics. 2018;1:65–73.
    1. Wang C, Thudium KB, Han M, Wang XT, Huang H, Feingersh D, Garcia C, Wu Y, Kuhne M, Srinivasan M, et al. In vitro characterization of the anti-PD-1 antibody nivolumab, BMS-936558, and in vivo toxicology in non-human primates. Cancer Immunol Res. 2014;2:846–56. doi:10.1158/2326-6066.CIR-14-0040.
    1. Homet Moreno B, Ribas A. Anti-programmed cell death protein-1/ligand-1 therapy in different cancers. Br J Cancer. 2015;112:1421–27. doi:10.1038/bjc.2015.124.
    1. Armand P, Engert A, Younes A, Fanale M, Santoro A, Zinzani PL, Timmerman JM, Collins GP, Ramchandren R, Cohen JB, et al. Nivolumab for relapsed/refractory classic hodgkin lymphoma after failure of autologous hematopoietic cell transplantation: extended follow-up of the multicohort single-arm phase II checkmate 205 trial. J Clin Oncol. 2018;36:1428–39. doi:10.1200/JCO.2017.76.0793.
    1. Chen R, Zinzani PL, Fanale MA, Armand P, Johnson NA, Brice P, Radford J, Ribrag V, Molin D, Vassilakopoulos TP, et al. Phase II study of the efficacy and safety of pembrolizumab for relapsed/refractory classic Hodgkin lymphoma. J Clin Oncol. 2017;35:2125–32. doi:10.1200/JCO.2016.72.1316.
    1. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, Stankevich E, Pons A, Salay TM, McMiller TL, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol. 2010;28:3167–75. doi:10.1200/JCO.2009.26.7609.
    1. Talotta R, Rucci F, Canti G, Scaglione F. Pros and cons of the immunogenicity of monoclonal antibodies in cancer treatment: a lesson from autoimmune diseases. Immunotherapy. 2019;11:241–54. doi:10.2217/imt-2018-0081.
    1. Sathish JG, Sethu S, Bielsky MC, de Haan L, French NS, Govindappa K, Green J, Griffiths CEM, Holgate S, Jones D, et al. Challenges and approaches for the development of safer immunomodulatory biologics. Nat Rev Drug Discov. 2013;12:306–24. doi:10.1038/nrd3974.
    1. Schellekens H, Jiskoot W. Eprex-associated pure red cell aplasia and leachates. Nat Biotechnol. 2006;24:613–14. doi:10.1038/nbt0606-613.
    1. Agrawal S, Statkevich P, Bajaj G, Feng Y, Saeger S, Desai DD, Park J-S, Waxman IM, Roy A, Gupta M. Evaluation of immunogenicity of nivolumab monotherapy and its clinical relevance in patients with metastatic solid tumors. J Clin Pharmacol. 2017;57:394–400. doi:10.1002/jcph.818.
    1. van Vugt M, de Greef R, Freshwater T, Mangin E, van Aarle F,AK. Immunogenicity of pembrolizumab (pembro) in patients (pts) with advanced melanoma (MEL) and non-small cell lung cancer (NSCLC): pooled results from KEYNOTE-001, 002, 006, and 010. J Clin Oncol. 2016;34:3063. doi:10.1200/JCO.2016.34.15_suppl.3063.
    1. Berezov A, Zhang HT, Greene MI, Murali R. Disabling erbB receptors with rationally designed exocyclic mimetics of antibodies: structure-function analysis. J Med Chem. 2001;44:2565–74. doi:10.1021/jm000527m.
    1. Copeland RA, Pompliano DL, Meek TD. Drug-target residence time and its implications for lead optimization. Nat Rev Drug Discov. 2006;5:730–39. doi:10.1038/nrd2082.
    1. Dong H, Strome SE, Salomao DR, Tamura H, Hirano F, Flies DB, Roche PC, Lu J, Zhu G, Tamada K, et al. Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002;8:793–800. doi:10.1038/nm730.
    1. Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, Rietz C, Flies DB, Lau JS, Zhu G, et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005;65:1089–96.
    1. Rongvaux A, Takizawa H, Strowig T, Willinger T, Eynon EE, Flavell RA, Manz MG. Human hemato-lymphoid system mice: current use and future potential for medicine. Annu Rev Immunol. 2013;31:635–74. doi:10.1146/annurev-immunol-032712-095921.
    1. Shay T, Jojic V, Zuk O, Rothamel K, Puyraimond-Zemmour D, Feng T, Wakamatsu E, Benoist C, Koller D, Regev A. Conservation and divergence in the transcriptional programs of the human and mouse immune systems. Proc Natl Acad Sci U S A. 2013;110:2946–51. doi:10.1073/pnas.1222738110.
    1. Spitzer MH, Carmi Y, Reticker-Flynn NE, Kwek SS, Madhireddy D, Martins MM, Gherardini PF, Prestwood TR, Chabon J, Bendall SC, et al. Systemic immunity is required for effective cancer immunotherapy. Cell. 2017;168:487–502 e15. doi:10.1016/j.cell.2016.12.022.
    1. Hoofring SA, Lopez R, Hock MB, Kaliyaperumal A, Patel SK, Swanson SJ, Chirmule N, Starcevic M. Immunogenicity testing strategy and bioanalytical assays for antibody-drug conjugates. Bioanalysis. 2013;5:1041–55. doi:10.4155/bio.13.10.
    1. Sauerborn M, van Dongen W. Practical considerations for the pharmacokinetic and immunogenic assessment of antibody-drug conjugates. BioDrugs. 2014;28:383–91. doi:10.1007/s40259-014-0096-z.
    1. Johnson CB, Win SY. Combination therapy with PD-1/PD-L1 blockade: an overview of ongoing clinical trials. Oncoimmunology. 2018;7:e1408744. doi:10.1080/2162402X.2017.1408744.
    1. Gotwals P, Cameron S, Cipolletta D, Cremasco V, Crystal A, Hewes B, Mueller B, Quaratino S, Sabatos-Peyton C, Petruzzelli L, et al. Prospects for combining targeted and conventional cancer therapy with immunotherapy. Nat Rev Cancer. 2017;17:286–301. doi:10.1038/nrc.2017.17.
    1. Shankar G, Devanarayan V, Amaravadi L, Barrett YC, Bowsher R, Finco-Kent D, Fiscella M, Gorovits B, Kirschner S, Moxness M, et al. Recommendations for the validation of immunoassays used for detection of host antibodies against biotechnology products. J Pharm Biomed Anal. 2008;48:1267–81. doi:10.1016/j.jpba.2008.09.020.
    1. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER), Center for Biologics Evaluation and Research Immunogenicity testing of therapeutic protein products -developing and validating assays for anti-drug antibody detection: guidance for industry. 2019.

Source: PubMed

3
Abonner