NK cells limit the synergistic anti-tumor effect of PD-1 inhibition and βγ-biased IL-2.

Int J Biol Macromol

Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell The

Published: September 2025


Category Ranking

98%

Total Visits

921

Avg Visit Duration

2 minutes

Citations

20

Article Abstract

Despite its potential as a cancer immunotherapy, wild-type IL-2 is limited by dose-limiting toxicities, including vascular leak syndrome, and its strong activation of regulatory T cells (Tregs), which dampens anti-tumor immunity. These drawbacks are largely driven by IL-2's binding to IL-2Rα, and avoiding this interaction can reduce IL-2-associated toxicities, although it cannot completely eliminate them. To overcome these limitations, βγ-biased IL-2 variants (Non-α-IL-2) have been developed to selectively activate effector T and NK cells. However, the clinical efficacy of these agents remains limited. Disappointing outcomes have been observed both in monotherapy and in combination with PD-1 inhibitors. This study investigates the anti-tumor efficacy of Non-α-IL-2, a long-acting, non-IL-2Rα-binding IL-2 variant conjugated to an anti-human serum albumin single-domain antibody, in a colon cancer mouse model. We found that the depletion of NK cells enhances the anti-tumor effect of Non-α-IL-2 combined with PD-1 inhibitors. Analysis of the tumor microenvironment revealed that depletion of NK cells increased CD8 T cell activation and infiltration. T cell subset analysis revealed that the number of exhausted CD8 T cells increased upon NK cell depletion. Moreover, we found that Ulbp1, a ligand of NK activating receptor NKG2D is highly expressed in exhausted CD8 T cells whereas MHC-I, which inhibits NK cell activation, is downregulated in exhausted CD8 T cells. Our results provide a possible explanation for the limited synergy between βγ-biased IL-2 and PD-1 blockade: βγ-biased IL-2-activated NK cells eliminate exhausted CD8 T cells, which are largely tumor-antigen specific T cells, in the TME. These findings underscore the importance of designing next-generation IL-2 variants that, when used in combination with PD-1 inhibitors, minimize NK cell activation to maximize CD8 T cell anti-tumor responses.

Download full-text PDF

Source
http://dx.doi.org/10.1016/j.ijbiomac.2025.147437DOI Listing

Publication Analysis

Top Keywords

exhausted cd8
16
cd8 cells
16
βγ-biased il-2
12
pd-1 inhibitors
12
cell activation
12
cells
11
il-2 variants
8
combination pd-1
8
depletion cells
8
cells increased
8

Similar Publications

NK cells limit the synergistic anti-tumor effect of PD-1 inhibition and βγ-biased IL-2.

Int J Biol Macromol

September 2025

Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, China; Institute of Cell The

Despite its potential as a cancer immunotherapy, wild-type IL-2 is limited by dose-limiting toxicities, including vascular leak syndrome, and its strong activation of regulatory T cells (Tregs), which dampens anti-tumor immunity. These drawbacks are largely driven by IL-2's binding to IL-2Rα, and avoiding this interaction can reduce IL-2-associated toxicities, although it cannot completely eliminate them. To overcome these limitations, βγ-biased IL-2 variants (Non-α-IL-2) have been developed to selectively activate effector T and NK cells.

View Article and Find Full Text PDF

Background: Immunotherapy (IO) combined with tyrosine kinase inhibitors (TKI) are now first-line therapy for advanced renal cell carcinoma (RCC), though reliable predictive biomarkers remain elusive. Recent evidence demonstrates that karyopherin α2 subunit (KPNA2), a nuclear transport regulator, plays key roles in tumorigenesis and therapy resistance.

Methods: Two cohorts were analyzed: an institutional cohort of metastatic RCC patients (ZS-MRCC) and the phase III JAVELIN Renal 101 trial cohort.

View Article and Find Full Text PDF

Tissue microenvironment characteristics associated with elevated risk of colorectal cancer (CRC) in Lynch syndrome (LS) are poorly characterized. We applied the multimodal single cell sequencing platform ExCITE-seq to define the colonic cellular composition and transcriptome of LS carriers with and without a history of CRC compared with general population controls. Our analysis revealed widespread remodeling in LS that included striking expansion of epithelial stem and progenitor cells, and loss of fibroblast populations.

View Article and Find Full Text PDF

Tissue microenvironment characteristics associated with elevated risk of colorectal cancer (CRC) in Lynch syndrome (LS) are poorly characterized. We applied the multimodal single cell sequencing platform ExCITE-seq to define the colonic cellular composition and transcriptome of LS carriers with and without a history of CRC compared with general population controls. Our analysis revealed widespread remodeling in LS that included striking expansion of epithelial stem and progenitor cells, and loss of fibroblast populations.

View Article and Find Full Text PDF

Although checkpoint immunotherapy has primarily focused on CD8⁺ T cells, emerging evidence highlights an important role for cytotoxic CD4⁺ T cells in mediating therapeutic responses. However, research on the functional properties of cytotoxic CD4⁺ T cells in the context of immunotherapy is still at an early stage and remains insufficiently defined. Utilizing single-cell RNA-sequencing datasets obtained from metastatic melanoma patients treated with checkpoint inhibitors targeting PD-1 and/or CTLA-4, we performed transcriptomic profiling of conventional CD4⁺ T cells, excluding proliferative and regulatory (FOXP3⁺) subsets, and compared responders and non-responders as distinct groups.

View Article and Find Full Text PDF