Cadonilimab, a tetravalent PD-1/CTLA-4 bispecific antibody with trans-binding and enhanced target binding avidity

Xinghua Pang, Zhaoliang Huang, Tingting Zhong, Peng Zhang, Zhongmin Maxwell Wang, Michelle Xia, Baiyong Li, Xinghua Pang, Zhaoliang Huang, Tingting Zhong, Peng Zhang, Zhongmin Maxwell Wang, Michelle Xia, Baiyong Li

Abstract

Clinical studies have shown that combination therapy of antibodies targeting cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 (PD-1) significantly improves clinical benefit over PD-1 antibody alone. However, broad application of this combination has been limited by toxicities. Cadonilimab (AK104) is a symmetric tetravalent bispecific antibody with a crystallizable fragment (Fc)-null design. In addition to demonstrating biological activity similar to that of the combination of CTLA-4 and PD-1 antibodies, cadonilimab possess higher binding avidity in a high-density PD-1 and CTLA-4 setting than in a low-density PD-1 setting, while a mono-specific anti-PD-1 antibody does not demonstrate this differential activity. With no binding to Fc receptors, cadonilimab shows minimal antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis, and interleukin-6 (IL-6)/IL-8 release. These features all likely contribute to significantly lower toxicities of cadonilimab observed in the clinic. Higher binding avidity of cadonilimab in a tumor-like setting and Fc-null design may lead to better drug retention in tumors and contribute to better safety while achieving anti-tumor efficacy.

Keywords: PD-1/CTLA-4 bispecific antibody; drug retention; tumor microenvironment.

Conflict of interest statement

The authors are all employees of Akeso Biopharma outside the submitted work.

Figures

Figure 1.
Figure 1.
Tetravalent design of anti-PD1/CTLA-4 bispesific antibody cadonilimab based on co-expression of PD-1 and CTLA-4 in tumor tissue. (a) Correlation analysis of PD-1 and CTLA-4 mRNA expression levels in various tumor types from TCGA dataset using cBioportal. The X- and Y-axis represent the mRNA expression level transformed by Log(value+1). TCGA, The Cancer Genome Atlas. (b) Schematic diagram of cadonilimab tetravalent structure.
Figure 2.
Figure 2.
Cadonilimab induce IL-2 and IFN-γ production in mixed lymphocyte reaction assays. (a, b) Cadonilimab promoted activation of human peripheral blood mononuclear cells (hPBMCs), which induced a more robust secretion of IL-2 and IFN-γ in mixed culture of hPBMCs and human DCs; Compared with nivolumab plus ipilimumab, cadonilimab did not significantly improve secretion of IL-2 and IFN-γ. (c, d) Cadonilimab enhanced secretion of IL-2 and IFN-γ in mixed culture of hPBMCs and Raji-PDL1 cells. Data are shown as mean ±SEM for n = 2 and analyzed using one-way ANOVA. *P 

Figure 3.

Cadonilimab demonstrates preferential higher avidity…

Figure 3.

Cadonilimab demonstrates preferential higher avidity binding to higher density of PD-1 and CTLA-4.…

Figure 3.
Cadonilimab demonstrates preferential higher avidity binding to higher density of PD-1 and CTLA-4. In a Fortebio assay, cadonilimab showed higher binding avidity with a (a) high density of PD-1 and CTLA-4, where PD-1 (50 nM) and CTLA-4 (50 nM) were loaded onto the sensor, compared with (b) that of a relative low density of PD-1, where PD-1 (10 nM) was loaded onto the sensor. Penpulimab (parental PD-1 antibody of cadonilimab) showed similar binding avidity under these antigen conditions (a) and (b).

Figure 4.

Tetravalence binding of Cadonilimab to…

Figure 4.

Tetravalence binding of Cadonilimab to PD-1 and CTLA-4. (a) Trans-binding of Cadonilimab to…

Figure 4.
Tetravalence binding of Cadonilimab to PD-1 and CTLA-4. (a) Trans-binding of Cadonilimab to CHO-K1-CTLA-4 cells (Orange cells) which transfected with human CTLA-4, and Jurkat-PD1 cells (light blue cells) labeled with Hoechst 33342. Jurkat-PD1 cell which is a suspension lymphoblasts cell line became adherent to CHO-K1-CTLA-4 cells when co-cultured with cadonilimab, but not with nivolumab plus ipilimumab. (b) Crosslinking of cadonilimab with cells expressing PD-1 and CTLA-4 in FACS assay. AK104 or control antibodies were added to a 1:1 mix of Far red-labeled CTLA-4-expressing CHO-K1 cells and CSFE-labeled PD-1-expressing CHO-K1 cells. (c) Cadonilimab could bind to human PD-1 and CTLA-4 with similar binding activity regardless of binding PD-1 or CTLA-4 firstly. Cadonilimab was fixed onto the sensor in Fortebio assay, and sequential binding to human PD-1 (PD1-hFc) and CTLA-4 (CTLA-4-hFc) or in reverse order was performed and measured.

Figure 5.

Cadonilimab with Fc null-mutations for…

Figure 5.

Cadonilimab with Fc null-mutations for both safety and efficacy concerns. (a) Antibody-dependent cell-mediated…

Figure 5.
Cadonilimab with Fc null-mutations for both safety and efficacy concerns. (a) Antibody-dependent cell-mediated cytotoxicity (ADCC) activities were determined by measuring lactase dehydrogenase (LDH) release from 293 T-CTLA4-PD1 cells. (b) Complement-dependent cytotoxicity (CDC) activities of cadonilimab, penpulimab in hG1WT format, and ipilimumab and nivolumab were determined by measuring LDH release from CHO-K1-PD1-CTLA4 cells. (c) Antibody-dependent cellular phagocytosis (ADCP) activities of cadonilimab, penpulimab in hG1WT format plus anti-CTLA-4 mAb in hG1WT format, and nivolumab plus ipilimumab were studied by examining phagocytosis of CHO-K1-PD1-CTLA4 cells by murine bone marrow derived macrophages (MBMMs). (d, e) Cadonilimab decreased the release of inflammatory cytokines IL-8 and IL-6 from human peripheral monocyte-derived macrophages (HPMMs). Data are expressed as mean or mean ±SEM and analyzed using one-way ANOVA. *P 
Similar articles
Cited by
References
    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373:23–10. doi:10.1056/NEJMoa1504030. - DOI - PMC - PubMed
    1. Morse MA, Overman MJ, Hartman L, Khoukaz T, Brutcher E, Lenz HJ, Atasoy A, Shangguan T, Zhao H, El-Rayes B.. Safety of nivolumab plus low-dose ipilimumab in previously treated microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer. Oncologist. 2019;24:1453–61. doi:10.1634/theoncologist.2019-0129. - DOI - PMC - PubMed
    1. Gao X, McDermott DF. Ipilimumab in combination with nivolumab for the treatment of renal cell carcinoma. Expert Opin Biol Ther. 2018;18:947–57. doi:10.1080/14712598.2018.1513485. - DOI - PMC - PubMed
    1. Antonia SJ, López-Martin JA, Bendell J, Ott PA, Taylor M, Eder JP, Jäger D, Pietanza MC, Le DT, de Braud F, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17:883–95. doi:10.1016/S1470-2045(16)30098-5. - DOI - PubMed
    1. Motzer RJ, Rini BI, McDermott DF, Arén Frontera O, Hammers HJ, Carducci MA, Salman P, Escudier B, Beuselinck B, Amin A, et al. Nivolumab plus ipilimumab versus sunitinib in first-line treatment for advanced renal cell carcinoma: extended follow-up of efficacy and safety results from a randomised, controlled, phase 3 trial. Lancet Oncol. 2019;20:1370–85. doi:10.1016/S1470-2045(19)30413-9. - DOI - PMC - PubMed
Show all 30 references
Publication types
MeSH terms
Grant support
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
LinkOut - more resources
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 3.
Figure 3.
Cadonilimab demonstrates preferential higher avidity binding to higher density of PD-1 and CTLA-4. In a Fortebio assay, cadonilimab showed higher binding avidity with a (a) high density of PD-1 and CTLA-4, where PD-1 (50 nM) and CTLA-4 (50 nM) were loaded onto the sensor, compared with (b) that of a relative low density of PD-1, where PD-1 (10 nM) was loaded onto the sensor. Penpulimab (parental PD-1 antibody of cadonilimab) showed similar binding avidity under these antigen conditions (a) and (b).
Figure 4.
Figure 4.
Tetravalence binding of Cadonilimab to PD-1 and CTLA-4. (a) Trans-binding of Cadonilimab to CHO-K1-CTLA-4 cells (Orange cells) which transfected with human CTLA-4, and Jurkat-PD1 cells (light blue cells) labeled with Hoechst 33342. Jurkat-PD1 cell which is a suspension lymphoblasts cell line became adherent to CHO-K1-CTLA-4 cells when co-cultured with cadonilimab, but not with nivolumab plus ipilimumab. (b) Crosslinking of cadonilimab with cells expressing PD-1 and CTLA-4 in FACS assay. AK104 or control antibodies were added to a 1:1 mix of Far red-labeled CTLA-4-expressing CHO-K1 cells and CSFE-labeled PD-1-expressing CHO-K1 cells. (c) Cadonilimab could bind to human PD-1 and CTLA-4 with similar binding activity regardless of binding PD-1 or CTLA-4 firstly. Cadonilimab was fixed onto the sensor in Fortebio assay, and sequential binding to human PD-1 (PD1-hFc) and CTLA-4 (CTLA-4-hFc) or in reverse order was performed and measured.
Figure 5.
Figure 5.
Cadonilimab with Fc null-mutations for both safety and efficacy concerns. (a) Antibody-dependent cell-mediated cytotoxicity (ADCC) activities were determined by measuring lactase dehydrogenase (LDH) release from 293 T-CTLA4-PD1 cells. (b) Complement-dependent cytotoxicity (CDC) activities of cadonilimab, penpulimab in hG1WT format, and ipilimumab and nivolumab were determined by measuring LDH release from CHO-K1-PD1-CTLA4 cells. (c) Antibody-dependent cellular phagocytosis (ADCP) activities of cadonilimab, penpulimab in hG1WT format plus anti-CTLA-4 mAb in hG1WT format, and nivolumab plus ipilimumab were studied by examining phagocytosis of CHO-K1-PD1-CTLA4 cells by murine bone marrow derived macrophages (MBMMs). (d, e) Cadonilimab decreased the release of inflammatory cytokines IL-8 and IL-6 from human peripheral monocyte-derived macrophages (HPMMs). Data are expressed as mean or mean ±SEM and analyzed using one-way ANOVA. *P 

References

    1. Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373:23–10. doi:10.1056/NEJMoa1504030.
    1. Morse MA, Overman MJ, Hartman L, Khoukaz T, Brutcher E, Lenz HJ, Atasoy A, Shangguan T, Zhao H, El-Rayes B.. Safety of nivolumab plus low-dose ipilimumab in previously treated microsatellite instability-high/mismatch repair-deficient metastatic colorectal cancer. Oncologist. 2019;24:1453–61. doi:10.1634/theoncologist.2019-0129.
    1. Gao X, McDermott DF. Ipilimumab in combination with nivolumab for the treatment of renal cell carcinoma. Expert Opin Biol Ther. 2018;18:947–57. doi:10.1080/14712598.2018.1513485.
    1. Antonia SJ, López-Martin JA, Bendell J, Ott PA, Taylor M, Eder JP, Jäger D, Pietanza MC, Le DT, de Braud F, et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 2016;17:883–95. doi:10.1016/S1470-2045(16)30098-5.
    1. Motzer RJ, Rini BI, McDermott DF, Arén Frontera O, Hammers HJ, Carducci MA, Salman P, Escudier B, Beuselinck B, Amin A, et al. Nivolumab plus ipilimumab versus sunitinib in first-line treatment for advanced renal cell carcinoma: extended follow-up of efficacy and safety results from a randomised, controlled, phase 3 trial. Lancet Oncol. 2019;20:1370–85. doi:10.1016/S1470-2045(19)30413-9.
    1. Abbas E, Salem TZ. Back to the light side: the role of mechanotransduction in the paradoxical response to checkpoint inhibitors in cancer patients. Crit Rev Immunol. 2019;39:165–73. doi:10.1615/CritRevImmunol.2019031554.
    1. Collins M, Michot JM, Danlos FX, Mussini C, Soularue E, Mateus C, Loirat D, Buisson A, Rosa I, Lambotte O, et al. Inflammatory gastrointestinal diseases associated with PD-1 blockade antibodies. Ann Oncol. 2017;28:2860–65. doi:10.1093/annonc/mdx403.
    1. Jodai T, Yoshida C, Sato R, Kakiuchi Y, Sato N, Iyama S, Kimura T, Saruwatari K, Saeki S, Ichiyasu H, et al. A potential mechanism of the onset of acute eosinophilic pneumonia triggered by an anti-PD-1 immune checkpoint antibody in a lung cancer patient. Immun Inflamm Dis. 2019;7:3–6. doi:10.1002/iid3.238.
    1. Occhipinti M, Falcone R, Onesti CE, Marchetti P. Hyperprogressive disease and early hypereosinophilia after anti-PD-1 treatment: a case report. Drug Saf Case Rep. 2018;5:12. doi:10.1007/s40800-018-0078-z.
    1. Markman B, Tran B, Gan H, Prawira A, Coward J, Jin X, Li B, Wang M, Xia Y, Desai J. A Phase 1 study of AK104, a tetrameric bispecific antibody that targets PD-1 and CTLA-4 in patients with advanced solid tumors. J ImmunoTher Cancer. 2019;7(Suppl 1):283. doi:10.1186/s40425-019-0764-0.
    1. Wu X, Ji J, Lou H, Li Y, Feng M, Xu N, Li Y, Wang J, Huang Y, Lou G, et al. Efficacy and safety of cadonilimab, an anti-PD-1/CTLA4 bi-specific antibody, in previously treated recurrent or metastatic (R/M) cervical cancer: a multicenter, open-label, single-arm, phase II trial. Oral presentation in SGO 2022 Annual Meeting on Women’s Cancer. American, Phoenix, 2022.
    1. Ji J, Shen L, Li Z, Gao X, Ji K, Chen Y, Xu N, Liu T, Yang N, Zhong H, et al. A phase Ib/II, multicenter, open-label study of AK104, a PD-1/CTLA-4 bispecific antibody, combined with chemotherapy (chemo) as first-line therapy for advanced gastric (G) or gastroesophageal junction (GEJ) cancer. J Clin Oncol. 2022;40(4_suppl):308–308. doi:10.1200/JCO.2022.40.4_suppl.308.
    1. Li B, Xia Y, Wang Z, Zhang P. ANTI-CTLA4 AND ANTI-PD-1 BIFUNCTIONAL ANTIBODY, PHARMACEUTICAL COMPOSITION THEREOF AND USE THEREOF. US Patent 16327076. 2019.
    1. Ahmadzadeh M, Johnson LA, Heemskerk B, Wunderlich JR, Dudley ME, White DE, Rosenberg SA. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood. 2009;114:1537–44. doi:10.1182/blood-2008-12-195792.
    1. Liu R, Oldham RJ, Teal E, Beers SA, Cragg MS. Fc-bbent. Antibodies. 2020;9:64. doi:10.3390/antib9040064.
    1. Wang X, Mathieu M, Brezski RJ. IgG Fc engineering to modulate antibody effector functions. Protein Cell. 2018;9:63–73. doi:10.1007/s13238-017-0473-8.
    1. Arlauckas SP, Garris CS, Kohler RH, Kitaoka M, Cuccarese MF, Yang KS, Miller MA, Carlson JC, Freeman GJ, Anthony RM, 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. Rotz SJ, Leino D, Szabo S, Mangino JL, Turpin BK, Pressey JG. Severe cytokine release syndrome in a patient receiving PD-1-directed therapy. Pediatr Blood Cancer. 2017;64:e26642. doi:10.1002/pbc.26642.
    1. Tanaka R, Okiyama N, Okune M, Ishitsuka Y, Watanabe R, Furuta J, Ohtsuka M, Otsuka A, Maruyama H, Fujisawa Y, et al. Serum level of interleukin-6 is increased in nivolumab-associated psoriasiform dermatitis and tumor necrosis factor-α is a biomarker of nivolumab recativity. J Dermatol Sci. 2017;86:71–73. doi:10.1016/j.jdermsci.2016.12.019.
    1. Tsukamoto H, Fujieda K, Senju S, Ikeda T, Oshiumi H, Nishimura Y. Immune-suppressive effects of interleukin-6 on T-cell-mediated anti-tumor immunity. Cancer Sci. 2018;109:523–30. doi:10.1111/cas.13433.
    1. Tsukamoto H, Fujieda K, Miyashita A, Fukushima S, Ikeda T, Kubo Y, Senju S, Ihn H, Nishimura Y, Oshiumi H. 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. Schalper KA, Carleton M, Zhou M, Chen T, Feng Y, Huang SP, Walsh AM, Baxi V, Pandya D, Baradet T, 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. Dovedi SJ, Elder MJ, Yang C, Sitnikova SI, Irving L, Hansen A, Hair J, Jones DC, Hasani S, Wang B, et al. Design and efficacy of a monovalent bispecific PD-1/CTLA4 antibody that enhances CTLA4 blockade on PD-1+ activated T cells. Cancer Discov. 2021;11:1100–17. doi:10.1158/-20-1445.
    1. Berezhnoy A, Sumrow BJ, Stahl K, Shah K, Liu D, Li J, Hao SS, De Costa A, Kaul S, Bendell J, et al. Development and preliminary clinical activity of PD-1-guided CTLA-4 blocking bispecific DART molecule. Cell Rep Med. 2020;1:100163. doi:10.1016/j.xcrm.2020.100163.
    1. Huang Z, Pang X, Zhong T, Qu T, Chen N, Ma S, He X, Xia D, Wang M, Xia M, et al. Penpulimab, an Fc-engineered IgG1 anti-PD-1 antibody, with improved efficacy and low incidence of immune-related adverse events. Front Immunol. 2022;13:924542. doi:10.3389/fimmu.2022.924542.
    1. Tjulandin S, Demidov L, Moiseyenko V, Protsenko S, Semiglazova T, Odintsova S, Zukov R, Lazarev S, Makarova Y, Nechaeva M, et al. Novel PD-1 inhibitor prolgolimab: expanding non-resectable/metastatic melanoma therapy choice. Eur J Cancer. 2021;149:222–32. doi:10.1016/j.ejca.2021.02.030.
    1. Davies AM, Rispens T, Ooijevaar-de Heer P, Gould HJ, Jefferis R, Aalberse RC, Sutton BJ. 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. Yu J, Song Y, Tian W. How to select IgG subclasses in developing anti-tumor therapeutic antibodies. J Hematol Oncol. 2020;13:45. doi:10.1186/s13045-020-00876-4.
    1. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, et al. The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2:401–04. doi:10.1158/-12-0095.
    1. Coloma MJ, Morrison SL. Design and production of novel tetravalent bispecific antibodies. Nat Biotechnol. 1997;15:159–63. doi:10.1038/nbt0297-159.

Source: PubMed

3
Se inscrever