Penpulimab, an Fc-Engineered IgG1 Anti-PD-1 Antibody, With Improved Efficacy and Low Incidence of Immune-Related Adverse Events

Zhaoliang Huang, Xinghua Pang, Tingting Zhong, Tailong Qu, Na Chen, Shun Ma, Xinrong He, Dennis Xia, Max Wang, Michelle Xia, Baiyong Li, Zhaoliang Huang, Xinghua Pang, Tingting Zhong, Tailong Qu, Na Chen, Shun Ma, Xinrong He, Dennis Xia, Max Wang, Michelle Xia, Baiyong Li

Abstract

Background: IgG4 anbibodies are deficient in stability and may contribute to tumor-associated escape from immune surveillance. We developed an IgG1 backbone anti-programmed cell death protein-1 (PD-1) antibody, penpulimab, which is designed to remove crystallizable fragment (Fc) gamma receptor (FcγR) binding that mediates antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and proinflammatory cytokine release.

Methods: Aggregation of different anti-PD-1 antibodies was tested by size exclusion chromatography, and melting temperature midpoint (Tm) and aggregation temperature onset (Tagg) were also determined. The affinity constants of penpulimab for PD-1 and human FcγRs were measured by surface plasmon resonance and biolayer interferometry. ADCC and ADCP were determined in cellular assays and antibody-dependent cytokine release (ADCR) from human macrophages was detected by ELISA. Binding kinetics of penpulimab to human PD-1 was determined by Biacore, and epitope/paratope mapping of PD-1/penpulimab was investigated using x-ray crystallography. Additionally, patients from six ongoing trials were included for analysis of immune-related adverse events (irAEs).

Results: Penpulimab demonstrated better stability and a lower level of host-cell protein residue compared with IgG4 backbone anti-PD-1 antibodies. As expected, penpulimab exhibited no apparent binding to FcγRIa, FcγRIIa_H131, FcγRIIIa_V158 and FcγRIIIa_F158, elicited no apparent ADCC and ADCP activities, and induced no remarkable IL-6 and IL-8 release by activated macrophages in vitro. Penpulimab was shown in the co-crystal study to bind to human PD-1 N-glycosylation site at N58 and had a slower off-rate from PD-1 versus nivolumab or pembrolizumab. Four hundred sixty-five patients were analyzed for irAEs. Fifteen (3.2%) patients had grade 3 or above irAEs. No death from irAEs was reported.

Conclusions: IgG1 backbone anti-PD1 antibody penpulimab has a good stability and reduced host cell protein residue, as well as potent binding to the antigen. Fc engineering has eliminated Fc-mediated effector functions of penpulimab including ADCC, ADCP and reduced ADCR, which may contribute to its more favorable safety profile.

Clinical trial registration: www.ClinicalTrials.gov, identifier: AK105-101: NCT03352531, AK105-201: NCT03722147, AK105-301: NCT03866980, AK105-202:NCT03866967, AK105-203: NCT04172571, AK105-204: NCT04172506.

Keywords: Fc engineering; IgG1 anti-PD-1 antibody; binding kinetics; immune-related adverse events; penpulimab.

Conflict of interest statement

All authors are employees of Akeso Biopharma Co., Ltd., who participated in the discovery, production and commercialization of penpulimab.

Copyright © 2022 Huang, Pang, Zhong, Qu, Chen, Ma, He, Xia, Wang, Xia and Li.

Figures

Figure 1
Figure 1
(A) Aggregates tested by size exclusion chromatography (SEC), (B) melting (Tm) and (C) aggregation temperature (Tagg) characterization using dynamic light scattering (DLS) and static light scattering (SLS), respectively, and (D) Host-cell protein (HCP) residue assay in penpulimab and other IgG4 backbone anti-PD1 antibodies.
Figure 2
Figure 2
(A) Antibody-dependent cell-mediated cytotoxicity (ADCC) activities of penpulimab and nivolumab were determined by measuring lactase dehydrogenase (LDH) release from 293T-PD1 cells. (B) Complement-dependent cytotoxicity (CDC) activities of penpulimab, penpulimab (hG1WT), a version of penpulimab with wildtype IgG1 backbone, and nivolumab were determined by measuring LDH release from CHO-K1-PD1-CTLA4 cells. (C) Antibody-dependent cellular phagocytosis (ADCP) activities of penpulimab (hG1WT), penpulimab, and nivolumab were studied by examining phagocytosis of CHO-K1-PD1 cells by murine bone marrow derived macrophages (HPMMs). Effects of Fc engineering of penpulimab on the release of inflammatory cytokines. (D) IL-6 and (E) IL-8 by HPMMs in the presence of IFN-γ. Data are expressed as mean or mean ± SEM and analyzed using one-way ANOVA. *P<0.05 and ***P<0.001 vs. isotype control; ###P<0.001 vs. negative control.
Figure 3
Figure 3
(A) PD-L1 target occupancy following intravenous administration of 1.0, 3.0 or 10.0 mg/kg penpulimab once every two weeks (Q2W) in the dose escalation study or 200 mg penpulimab Q2W in the expansion study, with 28 days per cycle. Post-infusion blood samples were collected at day 1, 2, 8, and 15 of cycle 1, and day 1 of cycle 2, 3, 5, 7, 9, 11, and 13, respectively, as indicated on the x-axis. C: cycle; D: day. Penpulimab potentiates T cell activation via PD1/PDL1 blockade. Raji-PD-L1 cells overexpressing PD-L1 were co-cultured with peripheral blood mononuclear cells (PBMCs) from a normal subject, and the production of IL-2 (B) and IFN-γ (C) was examined by ELISA. Data are shown as mean ± SEM for n = 2, and analyzed using one-way ANOVA. *P<0.05, **P<0.01 and ***P<0.001 vs. isotype control.

References

    1. Jin HT, Ahmed R, Okazaki T. Role of PD-1 in Regulating T-Cell Immunity. Curr Top Microbiol Immunol (2011) 350:17–37. doi: 10.1007/82_2010_116
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. . Safety, Activity, and Immune Correlates of Anti-PD-1 Antibody in Cancer. N Engl J Med (2012) 366:2443–54. doi: 10.1056/NEJMoa1200690
    1. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, et al. . PD-1 Blockade With Nivolumab in Relapsed or Refractory Hodgkin's Lymphoma. N Engl J Med (2015) 372:311–9. doi: 10.1056/NEJMoa1411087
    1. Homet Moreno B, Ribas A. Anti-Programmed Cell Death Protein-1/Ligand-1 Therapy in Different Cancers. Br J Cancer (2015) 112:1421–7. doi: 10.1038/bjc.2015.124
    1. Moreno-Vicente J, Willoughby JE, Taylor MC, Booth SG, English VL, Williams EL, et al. . Fc-Null Anti-PD-1 Monoclonal Antibodies Deliver Optimal Checkpoint Blockade in Diverse Immune Environments. J Immunother Cancer (2022) 10:e003735. doi: 10.1136/jitc-2021-003735
    1. Arlauckas SP, Garris CS, Kohler RH, Kitaoka M, Cuccarese MF, Yang KS, et al. . In Vivo Imaging Reveals a Tumor-Associated Macrophage-Mediated Resistance Pathway in Anti-PD-1 Therapy. Sci Transl Med (2017) 9:eaal3604. doi: 10.1126/scitranslmed.aal3604
    1. Kinder M, Greenplate AR, Strohl WR, Jordan RE, Brezski RJ. An Fc Engineering Approach That Modulates Antibody-Dependent Cytokine Release Without Altering Cell-Killing Functions. MAbs (2015) 7:494–504. doi: 10.1080/19420862.2015.1022692
    1. Pander J, Heusinkveld M, van der Straaten T, Jordanova ES, Baak-Pablo R, Gelderblom H, et al. . Activation of Tumor-Promoting Type 2 Macrophages by EGFR-Targeting Antibody Cetuximab. Clin Cancer Res (2011) 17:5668–73. doi: 10.1158/1078-0432.CCR-11-0239
    1. Dahan R, Sega E, Engelhardt J, Selby M, Korman AJ, Ravetch JV, et al. . Fcγrs Modulate the Anti-Tumor Activity of Antibodies Targeting the PD-1/PD-L1 Axis. Cancer Cell (2015) 28:285–95. doi: 10.1016/j.ccell.2015.08.004
    1. Lo Russo G, Moro M, Sommariva M, Cancila V, Boeri M, Centonze G, et al. . Antibody-Fc/FcR Interaction on Macrophages as a Mechanism for Hyperprogressive Disease in non-Small Cell Lung Cancer Subsequent to PD-1/PD-L1 Blockade. Clin Cancer Res (2019) 25:989–99. doi: 10.1158/1078-0432.CCR-18-1390
    1. Arlauckas SP, Garris CS, Kohler RH, Kitaoka M, Cuccarese MF, Yang KS, et al. . In Vivo Imaging Reveals a Tumor-Associated Macrophage-Mediated Resistance Pathway in Anti-PD-1 Therapy. Sci Transl Med (2017) 9:eaal3604. doi: 10.1126/scitranslmed.aal3604
    1. Tsukamoto H, Fujieda K, Miyashita A, Fukushima S, Ikeda T, Kubo Y, et al. . Combined Blockade of IL6 and PD-1/PD-L1 Signaling Abrogates Mutual Regulation of Their Immunosuppressive Effects in the Tumor Microenvironment. Cancer Res (2018) 78:5011–22. doi: 10.1158/0008-5472.CAN-18-0118
    1. Labrijn AF, Rispens T, Meesters J, Rose RJ, den Bleker TH, Loverix S, et al. . Species-Specific Determinants in the IgG CH3 Domain Enable Fab-Arm Exchange by Affecting the Noncovalent CH3-CH3 Interaction Strength. J Immunol (2011) 187:3238–46. doi: 10.4049/jimmunol.1003336
    1. Davies AM, Rispens T, Ooijevaar-de Heer P, Gould HJ, Jefferis R, Aalberse RC, et al. . Structural Determinants of Unique Properties of Human IgG4-Fc. J Mol Biol (2014) 426:630–44. doi: 10.1016/j.jmb.2013.10.039
    1. Bianchini R, Karagiannis SN, Jordakieva G, Jensen-Jarolim E. The Role of IgG4 in the Fine Tuning of Tolerance in IgE-Mediated Allergy and Cancer. Int J Mol Sci (2020) 21:5017. doi: 10.3390/ijms21145017
    1. Crescioli S, Correa I, Karagiannis P, Davies AM, Sutton BJ, Nestle FO, et al. . IgG4 Characteristics and Functions in Cancer Immunity. Curr Allergy Asthma Rep (2016) 16:7. doi: 10.1007/s11882-015-0580-7
    1. Wang H, Xu Q, Zhao C, Zhu Z, Zhu X, Zhou J, et al. . An Immune Evasion Mechanism With IgG4 Playing an Essential Role in Cancer and Implication for Immunotherapy. J Immunother Cancer (2020) 8:e000661. doi: 10.1136/jitc-2020-000661
    1. Tran B, Grosskopf V, Wang X, Yang J, Walker D, Jr, Yu C, et al. . Investigating Interactions Between Phospholipase B-Like 2 and Antibodies During Protein A Chromatography. J Chromatogr A (2016) 1438:31–8. doi: 10.1016/j.chroma.2016.01.047
    1. Ishikawa T, Ito T, Endo R, Nakagawa K, Sawa E, Wakamatsu K, et al. . Influence of pH on Heat-Induced Aggregation and Degradation of Therapeutic Monoclonal Antibodies. Biol Pharm Bull (2010) 33:1413–7. doi: 10.1248/bpb.33.1413
    1. Brader ML, Estey T, Bai S, Alston RW, Lucas KK, Lantz S, et al. . Examination of Thermal Unfolding and Aggregation Profiles of a Series of Developable Therapeutic Monoclonal Antibodies. Mol Pharm (2015) 12:1005–17. doi: 10.1021/mp400666b
    1. Temel DB, Landsman P, Brader ML. Orthogonal Methods for Characterizing the Unfolding of Therapeutic Monoclonal Antibodies: Differential Scanning Calorimetry, Isothermal Chemical Denaturation, and Intrinsic Fluorescence With Concomitant Static Light Scattering. Methods Enzymol (2016) 567:359–89. doi: 10.1016/bs.mie.2015.08.029
    1. Wang S, Zhang X, Wu G, Tian Z, Qian F. Optimization of High-Concentration Endostatin Formulation: Harmonization of Excipients' Contributions on Colloidal and Conformational Stabilities. Int J Pharm (2017) 530:173–86. doi: 10.1016/j.ijpharm.2017.07.057
    1. Mimoto F, Katada H, Kadono S, Igawa T, Kuramochi T, Muraoka M, et al. . Engineered Antibody Fc Variant With Selectively Enhanced Fcγriib Binding Over Both Fcγriia (R131) and Fcγriia (H131). Protein Eng Des Sel (2013) 26:589–98. doi: 10.1093/protein/gzt022
    1. Rispens T, Ooievaar-De Heer P, Vermeulen E, Schuurman J, van der Neut Kolfschoten M, Aalberse RC, et al. . Human IgG4 Binds to IgG4 and Conformationally Altered IgG1 via Fc-Fc Interactions. J Immunol (2009) 182:4275–81. doi: 10.4049/jimmunol.0804338
    1. Karagiannis P, Gilbert AE, Josephs DH, Ali N, Dodev T, Saul L, et al. . IgG4 Subclass Antibodies Impair Antitumor Immunity in Melanoma. J Clin Invest (2013) 123:1457–74. doi: 10.1172/JCI65579
    1. Dixit N, Salamat-Miller N, Salinas PA, Taylor KD, Basu SK. Residual Host Cell Protein Promotes Polysorbate 20 Degradation in a Sulfatase Drug Product Leading to Free Fatty Acid Particles. J Pharm Sci (2016) 105:1657–66. doi: 10.1016/j.xphs.2016.02.029
    1. Horita S, Nomura Y, Sato Y, Shimamura T, Iwata S, Nomura N. High-Resolution Crystal Structure of the Therapeutic Antibody Pembrolizumab Bound to the Human PD-1. Sci Rep (2016) 6:35297. doi: 10.1038/srep35297
    1. Scapin G, Yang X, Prosise WW, McCoy M, Reichert P, Johnston JM, et al. . Structure of Full-Length Human Anti-PD1 Therapeutic IgG4 Antibody Pembrolizumab. Nat Struct Mol Biol (2015) 22:953–8. doi: 10.1038/nsmb.3129
    1. Kurdi AT, Glavey SV, Bezman NA, Jhatakia A, Guerriero JL, Manier S, et al. . Antibody-Dependent Cellular Phagocytosis by Macrophages is a Novel Mechanism of Action of Elotuzumab. Mol Cancer Ther (2018) 17:1454–63. doi: 10.1158/1535-7163.MCT-17-0998
    1. Schalper KA, Carleton M, Zhou M, Chen T, Feng Y, Huang SP, et al. . Elevated Serum Interleukin-8 Is Associated With Enhanced Intratumor Neutrophils and Reduced Clinical Benefit of Immune-Checkpoint Inhibitors. Nat Med (2020) 26:688–92. doi: 10.1038/s41591-020-0856-x
    1. Yuen KC, Liu LF, Gupta V, Madireddi S, Keerthivasan S, Li C, et al. . High Systemic and Tumor-Associated IL-8 Correlates With Reduced Clinical Benefit of PD-L1 Blockade. Nat Med (2020) 26:693–8. doi: 10.1038/s41591-020-0860-1
    1. Zhang T, Song X, Xu L, Ma J, Zhang Y, Gong W, et al. . The Binding of an Anti-PD-1 Antibody to FcγrI has a Profound Impact on its Biological Functions. Cancer Immunol Immunother (2018) 67:1079–90. doi: 10.1007/s00262-018-2160-x
    1. Patnaik A, Kang SP, Rasco D, Papadopoulos KP, Elassaiss-Schaap J, Beeram M, et al. . Phase I Study of Pembrolizumab (MK-3475; Anti-PD-1 Monoclonal Antibody) in Patients With Advanced Solid Tumors. Clin Cancer Res (2015) 21:4286–93. doi: 10.1158/1078-0432.CCR-14-2607
    1. Varki A. Biological Roles of Glycans. Glycobiology (2017) 27:3–49. doi: 10.1093/glycob/cww086
    1. Okada M, Chikuma S, Kondo T, Hibino S, Machiyama H, Yokosuka T, et al. . Blockage of Core Fucosylation Reduces Cell-Surface Expression of PD-1 and Promotes Anti-Tumor Immune Responses of T Cells. Cell Rep (2017) 20:1017–28. doi: 10.1016/j.celrep.2017.07.027
    1. Chen D, Tan S, Zhang H, Wang H, He W, Shi R, et al. . The FG Loop of PD-1 Serves as a "Hotspot" for Therapeutic Monoclonal Antibodies in Tumor Immune Checkpoint Therapy. Science (2019) 14:113–24. doi: 10.1016/j.isci.2019.03.017
    1. Na Z, Yeo SP, Bharath SR, Bowler MW, Balıkçı E, Wang CI, et al. . Structural Basis for Blocking PD-1-Mediated Immune Suppression by Therapeutic Antibody Pembrolizumab. Cell Res (2017) 27:147–50. doi: 10.1038/cr.2016.77
    1. Tan S, Zhang H, Chai Y, Song H, Tong Z, Wang Q, et al. . An Unexpected N-Terminal Loop in PD-1 Dominates Binding by Nivolumab. Nat Commun (2017) 8:14369. doi: 10.1038/ncomms14369
    1. Pinho SS, Reis CA. Glycosylation in Cancer: Mechanisms and Clinical Implications. Nat Rev Cancer (2015) 15:540–55. doi: 10.1038/nrc3982
    1. Liu YC, Yen HY, Chen CY, Chen CH, Cheng PF, Juan YH, et al. . Sialylation and Fucosylation of Epidermal Growth Factor Receptor Suppress its Dimerization and Activation in Lung Cancer Cells. Proc Natl Acad Sci USA (2011) 108:11332–7. doi: 10.1073/pnas.1107385108
    1. Potapenko IO, Haakensen VD, Lüders T, Helland A, Bukholm I, Sørlie T, et al. . Glycan Gene Expression Signatures in Normal and Malignant Breast Tissue; Possible Role in Diagnosis and Progression. Mol Oncol (2010) 4:98–118. doi: 10.1016/j.molonc.2009.12.001
    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) 44:136–40. doi: 10.1053/j.seminoncol.2017.06.002
    1. Garris CS, Arlauckas SP, Kohler RH, Trefny MP, Garren S, Piot C, et al. . Successful Anti-PD-1 Cancer Immunotherapy Requires T Cell-Dendritic Cell Crosstalk Involving the Cytokines IFN-γ and IL-12. Immunity (2018) 49:1148–61.e7. doi: 10.1016/j.immuni.2018.09.024
    1. Abiko K, Matsumura N, Hamanishi J, Horikawa N, Murakami R, Yamaguchi K, et al. . IFN-γ From Lymphocytes Induces PD-L1 Expression and Promotes Progression of Ovarian Cancer. Br J Cancer (2015) 112:1501–9. doi: 10.1038/bjc.2015.101
    1. Li B, Huang Z, Pang X, Zhong T, Jin C, Chen N, et al. . 2o Penpulimab, an IgG1 Anti-PD-1 Antibody With Fc-Engineering to Eliminate Effector Functions and With Unique Epitope and Binding Properties. Ann Oncol (2021) 39:S361. doi: 10.1016/j.annonc.2021.08.280
    1. Song Y, Zhou K, Jin C, Qian Z, Hou M, Fan L, et al. . A Phase II Study of Penpulimab, an Anti-PD-1 Antibody, in Patients With Relapsed or Refractory Classic Hodgkin Lymphoma (cHL). J Clin Oncol (2021) 39:7529. doi: 10.1200/JCO.2021.39.15_suppl.7529
    1. Chen X, Hu C, Wang W, Zou Q, Li J, Lin Q, et al. . A Phase II Study of the Anti-Programmed Cell Death-1 (PD-1) Antibody Penpulimab in Patients With Metastatic Nasopharyngeal Carcinoma (NPC) Who had Progressed After Two or More Lines of Chemotherapy: Updated Results. Ann Oncol (2021) 32:S786–817. doi: 10.1016/j.annonc.2021.08.1319
    1. Khoja L, Day D, Wei-Wu Chen T, Siu LL, Hansen AR. Tumour- and Class-Specific Patterns of Immune-Related Adverse Events of Immune Checkpoint Inhibitors: A Systematic Review. Ann Oncol (2017) 28:2377–85. doi: 10.1093/annonc/mdx286
    1. FDA Nivolumab Instructions (2022). Available at: .
    1. FDA Nivolumab Instructions (2022). Available at: .

Source: PubMed

3
Tilaa