Abstract

Abstract Background: TIGIT is a co-inhibitory receptor and immune checkpoint associated with T cell and natural killer (NK) cell dysfunction in cancer. Tiragolumab is an anti-TIGIT antibody with an active IgG1/kappa Fc. In a randomized double-blind phase 2 clinical trial in non-small cell lung cancer (NSCLC), tiragolumab + atezolizumab (anti-PD-L1) combination treatment demonstrated significant improvement relative to atezolizumab alone. However, the mechanisms underlying the efficacy of this combination are not well understood. Methods: In CITYSCAPE (phase 2, NCT03563716), chemotherapy-naïve patients with locally advanced/metastatic NSCLC received either placebo + 1200 mg atezolizumab or 600 mg tiragolumab + 1200 mg atezolizumab q3w IV. We collected tumor pretreatment samples and serum samples (baseline and on-treatment) from patients enrolled in the trial, which were subject to bulk RNA-seq and proteomics, respectively. In the mouse tumor model, BALB/c mice were implanted with syngeneic CT26 tumors. After tumor establishment, mice were randomized by tumor volume and then treated with control IgG2a, anti-PD-L1, anti-TIGIT IgG2a-LALAPG (Fc-inactive) ± anti-PD-L1, anti-TIGIT IgG2b ± anti-PD-L1, or anti-TIGIT IgG2a ± anti-PD-L1. Three days after treatment, CD45+ immune cells were collected from the peripheral blood and tumor from selected treatment groups and underwent single cell RNA sequencing or flow cytometry profiling. Tumor growth was also monitored to determine efficacy. Results: Here, we show that tiragolumab functions as both a conventional checkpoint inhibitor and, via Fc gamma receptor engagement, as a modulator of immunosuppressive myeloid cells and T regulatory (Treg) cells. Gene expression analysis of patient tumor samples revealed high levels of these cell subsets, were instead associated with treatment benefit in the tiragolumab + atezolizumab arm but not atezolizumab arm. Analysis of patient serum proteins suggested an association of myeloid cell activation with clinical benefit mediated by the combination therapy. In mouse tumor models, treatment with anti-PD-L1 + anti-TIGIT IgG2a (but not anti-TIGIT IgG2b or Fc-silent anti-TIGIT) led to effective tumor control in mice, suggesting a pivotal role for activating Fc receptors. Phenotypic profiling by single cell RNAseq and flow cytometry revealed that Fc-active anti-TIGIT remodels the tumor microenvironment, most prominently by inducing antigen presentation machinery in myeloid cells, counteracting of anti-PDL1-enforced CD8+ T cell exhaustion, and reducing Treg suppressive capacity. Conclusions: These findings reveal that FcR engagement is one of several distinct mechanisms by which tiragolumab unleashes antitumor immune responses, and inform further clinical development of anti-TIGIT therapies. Citation Format: Namrata S. Patil, Shyam Srivats, Yoonha Choi, Xiangnan Guan, Barzin Nabet, Lisa McGinnis, Eugene Chiang, Alexis Dunkle, Bill O’Gorman, Patrick S. Chang, Ruozhen Hu, John Silva, Joy Han, Amelia Au-Yeung, Chikara Takahashi, Nandini Molden, Pallavi Daggumati, Wendy Connolly, Melissa Johnson, Delvys Rodriguez Abreu, Byoung Chul Cho, Antoine Italiano, Ignacio Gil-Bazo, Enriqueta Felip, Ira Mellman, Raymond Meng, Sanjeev Mariathasan, Robert Johnston, David S. Shames. Anti-TIGIT antibody tiragolumab leverages myeloid cells and regulatory T cells to improve PD-L1 checkpoint blockade. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5712.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call