Michela Capello, Angelica Sette, Theo Plantinga, Craig J Thalhauser, Vanessa M Spires, Kristina B Nürmberger, Jordan M Blum, Brandon W Higgs, Patricia Garrido Castro, Christina Yu, Carol Costa Sa, Sina Fellermeier-Kopf, Saskia M Burm, Kristin Strumane, Aras Toker, Andrea Imle, Bruna de Andrade Pereira, Alexander Muik, Tahamtan Ahmadi, Özlem Türeci, Mark Fereshteh, Ugur Sahin, Maria Jure-Kunkel, Nora Pencheva
{"title":"Acasunlimab是一种fc惰性PD-L1×4-1BB双特异性抗体,与PD-1阻断联合,通过互补的免疫调节作用增强抗肿瘤免疫。","authors":"Michela Capello, Angelica Sette, Theo Plantinga, Craig J Thalhauser, Vanessa M Spires, Kristina B Nürmberger, Jordan M Blum, Brandon W Higgs, Patricia Garrido Castro, Christina Yu, Carol Costa Sa, Sina Fellermeier-Kopf, Saskia M Burm, Kristin Strumane, Aras Toker, Andrea Imle, Bruna de Andrade Pereira, Alexander Muik, Tahamtan Ahmadi, Özlem Türeci, Mark Fereshteh, Ugur Sahin, Maria Jure-Kunkel, Nora Pencheva","doi":"10.1136/jitc-2024-011377","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Next-generation cancer immunotherapies aim to improve patient outcomes by combining inhibitory signal blockade with targeted T-cell costimulation in tumor and lymphoid tissues. Acasunlimab (DuoBody-PD-L1×4-1BB) is an investigational, bispecific antibody designed to elicit an antitumor immune response via conditional 4-1BB activation strictly dependent on simultaneous programmed death-ligand 1 (PD-L1) binding. Since 4-1BB is coexpressed with programmed cell death protein-1 (PD-1) on CD8<sup>+</sup> T cells, PD-1 blockade and simultaneous costimulation through 4-1BB may synergistically enhance T-cell effector functions. We hypothesized that combining acasunlimab with PD-1 blockade to fully disrupt PD-1 interactions with both PD-L1 and PD-L2 would amplify the depth and duration of antitumor immunity.</p><p><strong>Methods: </strong>The effect of acasunlimab and pembrolizumab combination was analyzed in vitro using functional immune cell assays, including mixed-lymphocyte reactions and antigen-specific T-cell proliferation and cytotoxicity assays. The antitumor activity of the combination was tested in vivo in (1) MC38, MB49, Pan02, and B16F10 syngeneic tumor models using acasunlimab and anti-PD-1 mouse-surrogate antibodies; and (2) triple knock-in mice expressing the human targets using an acasunlimab chimeric antibody (chi-acasunlimab) and pembrolizumab. The mechanism of action of the combination was investigated in the MC38 syngeneic model through immunohistochemistry, flow cytometry, and bulk RNA sequencing.</p><p><strong>Results: </strong>The combination reinvigorated dysfunctional T cells in vitro, while also potentiating T-cell expansion, interleukin (IL)-2 and interferon gamma secretion and cytotoxic activity. In vivo, the combination of chi-acasunlimab and pembrolizumab or mouse-surrogate antibodies potentiated antitumor activity and survival in the humanized knock-in and multiple syngeneic mouse models, leading to durable complete tumor regressions in the MC38 model consistent with therapeutic synergy. Mechanistically, the combination enhanced clonal expansion of tumor-specific CD8<sup>+</sup> T cells in tumor-draining lymph nodes and increased the density of proliferating and cytotoxic CD8<sup>+</sup> T cells in the tumor microenvironment. It also potentiated the IL-2 signaling pathway, increasing the proportion of granzyme B (GZMB<sup>+</sup>) stem-like CD8<sup>+</sup> T cells thought to have superior effector function.</p><p><strong>Conclusion: </strong>These preclinical results demonstrate that conditional 4-1BB stimulation combined with complete PD-1 blockade enhances antitumor immunity through complementary mechanisms. The acasunlimab and pembrolizumab combination is being evaluated in Phase 2 (NCT05117242) and pivotal Phase 3 (NCT06635824) trials in patients with metastatic non-small cell lung cancer after checkpoint inhibitor therapy failure.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 4","pages":""},"PeriodicalIF":10.6000,"publicationDate":"2025-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11987116/pdf/","citationCount":"0","resultStr":"{\"title\":\"Acasunlimab, an Fc-inert PD-L1×4-1BB bispecific antibody, combined with PD-1 blockade potentiates antitumor immunity via complementary immune modulatory effects.\",\"authors\":\"Michela Capello, Angelica Sette, Theo Plantinga, Craig J Thalhauser, Vanessa M Spires, Kristina B Nürmberger, Jordan M Blum, Brandon W Higgs, Patricia Garrido Castro, Christina Yu, Carol Costa Sa, Sina Fellermeier-Kopf, Saskia M Burm, Kristin Strumane, Aras Toker, Andrea Imle, Bruna de Andrade Pereira, Alexander Muik, Tahamtan Ahmadi, Özlem Türeci, Mark Fereshteh, Ugur Sahin, Maria Jure-Kunkel, Nora Pencheva\",\"doi\":\"10.1136/jitc-2024-011377\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><strong>Background: </strong>Next-generation cancer immunotherapies aim to improve patient outcomes by combining inhibitory signal blockade with targeted T-cell costimulation in tumor and lymphoid tissues. Acasunlimab (DuoBody-PD-L1×4-1BB) is an investigational, bispecific antibody designed to elicit an antitumor immune response via conditional 4-1BB activation strictly dependent on simultaneous programmed death-ligand 1 (PD-L1) binding. Since 4-1BB is coexpressed with programmed cell death protein-1 (PD-1) on CD8<sup>+</sup> T cells, PD-1 blockade and simultaneous costimulation through 4-1BB may synergistically enhance T-cell effector functions. We hypothesized that combining acasunlimab with PD-1 blockade to fully disrupt PD-1 interactions with both PD-L1 and PD-L2 would amplify the depth and duration of antitumor immunity.</p><p><strong>Methods: </strong>The effect of acasunlimab and pembrolizumab combination was analyzed in vitro using functional immune cell assays, including mixed-lymphocyte reactions and antigen-specific T-cell proliferation and cytotoxicity assays. The antitumor activity of the combination was tested in vivo in (1) MC38, MB49, Pan02, and B16F10 syngeneic tumor models using acasunlimab and anti-PD-1 mouse-surrogate antibodies; and (2) triple knock-in mice expressing the human targets using an acasunlimab chimeric antibody (chi-acasunlimab) and pembrolizumab. The mechanism of action of the combination was investigated in the MC38 syngeneic model through immunohistochemistry, flow cytometry, and bulk RNA sequencing.</p><p><strong>Results: </strong>The combination reinvigorated dysfunctional T cells in vitro, while also potentiating T-cell expansion, interleukin (IL)-2 and interferon gamma secretion and cytotoxic activity. In vivo, the combination of chi-acasunlimab and pembrolizumab or mouse-surrogate antibodies potentiated antitumor activity and survival in the humanized knock-in and multiple syngeneic mouse models, leading to durable complete tumor regressions in the MC38 model consistent with therapeutic synergy. Mechanistically, the combination enhanced clonal expansion of tumor-specific CD8<sup>+</sup> T cells in tumor-draining lymph nodes and increased the density of proliferating and cytotoxic CD8<sup>+</sup> T cells in the tumor microenvironment. It also potentiated the IL-2 signaling pathway, increasing the proportion of granzyme B (GZMB<sup>+</sup>) stem-like CD8<sup>+</sup> T cells thought to have superior effector function.</p><p><strong>Conclusion: </strong>These preclinical results demonstrate that conditional 4-1BB stimulation combined with complete PD-1 blockade enhances antitumor immunity through complementary mechanisms. The acasunlimab and pembrolizumab combination is being evaluated in Phase 2 (NCT05117242) and pivotal Phase 3 (NCT06635824) trials in patients with metastatic non-small cell lung cancer after checkpoint inhibitor therapy failure.</p>\",\"PeriodicalId\":14820,\"journal\":{\"name\":\"Journal for Immunotherapy of Cancer\",\"volume\":\"13 4\",\"pages\":\"\"},\"PeriodicalIF\":10.6000,\"publicationDate\":\"2025-04-10\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11987116/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Journal for Immunotherapy of Cancer\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1136/jitc-2024-011377\",\"RegionNum\":1,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q1\",\"JCRName\":\"IMMUNOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal for Immunotherapy of Cancer","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1136/jitc-2024-011377","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
Acasunlimab, an Fc-inert PD-L1×4-1BB bispecific antibody, combined with PD-1 blockade potentiates antitumor immunity via complementary immune modulatory effects.
Background: Next-generation cancer immunotherapies aim to improve patient outcomes by combining inhibitory signal blockade with targeted T-cell costimulation in tumor and lymphoid tissues. Acasunlimab (DuoBody-PD-L1×4-1BB) is an investigational, bispecific antibody designed to elicit an antitumor immune response via conditional 4-1BB activation strictly dependent on simultaneous programmed death-ligand 1 (PD-L1) binding. Since 4-1BB is coexpressed with programmed cell death protein-1 (PD-1) on CD8+ T cells, PD-1 blockade and simultaneous costimulation through 4-1BB may synergistically enhance T-cell effector functions. We hypothesized that combining acasunlimab with PD-1 blockade to fully disrupt PD-1 interactions with both PD-L1 and PD-L2 would amplify the depth and duration of antitumor immunity.
Methods: The effect of acasunlimab and pembrolizumab combination was analyzed in vitro using functional immune cell assays, including mixed-lymphocyte reactions and antigen-specific T-cell proliferation and cytotoxicity assays. The antitumor activity of the combination was tested in vivo in (1) MC38, MB49, Pan02, and B16F10 syngeneic tumor models using acasunlimab and anti-PD-1 mouse-surrogate antibodies; and (2) triple knock-in mice expressing the human targets using an acasunlimab chimeric antibody (chi-acasunlimab) and pembrolizumab. The mechanism of action of the combination was investigated in the MC38 syngeneic model through immunohistochemistry, flow cytometry, and bulk RNA sequencing.
Results: The combination reinvigorated dysfunctional T cells in vitro, while also potentiating T-cell expansion, interleukin (IL)-2 and interferon gamma secretion and cytotoxic activity. In vivo, the combination of chi-acasunlimab and pembrolizumab or mouse-surrogate antibodies potentiated antitumor activity and survival in the humanized knock-in and multiple syngeneic mouse models, leading to durable complete tumor regressions in the MC38 model consistent with therapeutic synergy. Mechanistically, the combination enhanced clonal expansion of tumor-specific CD8+ T cells in tumor-draining lymph nodes and increased the density of proliferating and cytotoxic CD8+ T cells in the tumor microenvironment. It also potentiated the IL-2 signaling pathway, increasing the proportion of granzyme B (GZMB+) stem-like CD8+ T cells thought to have superior effector function.
Conclusion: These preclinical results demonstrate that conditional 4-1BB stimulation combined with complete PD-1 blockade enhances antitumor immunity through complementary mechanisms. The acasunlimab and pembrolizumab combination is being evaluated in Phase 2 (NCT05117242) and pivotal Phase 3 (NCT06635824) trials in patients with metastatic non-small cell lung cancer after checkpoint inhibitor therapy failure.
期刊介绍:
The Journal for ImmunoTherapy of Cancer (JITC) is a peer-reviewed publication that promotes scientific exchange and deepens knowledge in the constantly evolving fields of tumor immunology and cancer immunotherapy. With an open access format, JITC encourages widespread access to its findings. The journal covers a wide range of topics, spanning from basic science to translational and clinical research. Key areas of interest include tumor-host interactions, the intricate tumor microenvironment, animal models, the identification of predictive and prognostic immune biomarkers, groundbreaking pharmaceutical and cellular therapies, innovative vaccines, combination immune-based treatments, and the study of immune-related toxicity.