Improved understanding of NSCLC immunotherapy response mechanisms from single-cell analysis
Kamila Justyna Bienkowska,Christopher Jon Hanley
DOI: https://doi.org/10.21037/tlcr-23-428
2023-08-29
Translational Lung Cancer Research
Abstract:Kamila Justyna Bienkowska, Christopher Jon Hanley School of Cancer Sciences, University of Southampton, Southampton, UK Comment on: Hu J, Zhang L, Xia H, et al . Tumor microenvironment remodeling after neoadjuvant immunotherapy in non-small cell lung cancer revealed by single-cell RNA sequencing. Genome Med 2023;15:14. Keywords: Non-small cell lung cancer (NSCLC); immunotherapy; single-cell RNA sequencing Submitted Jun 30, 2023. Accepted for publication Aug 01, 2023. Published online Aug 08, 2023. doi: 10.21037/tlcr-23-428 Lung cancer is the leading cause of cancer-related deaths worldwide. In recent years, immune checkpoint blockade (ICB) has been widely used for the treatment of non-small cell lung cancer (NSCLC)—the predominant lung cancer subtype. However, ICB response rates remain limited. For example, analysis of multiple NSCLC trials using nivolumab showed this treatment increased 4-year overall survival rates from 5% to 14% but only achieved complete or partial responses in 16% of cases (1). Further understanding of how the tumour microenvironment changes in response to ICB, specifically what cell populations and/or molecular mechanisms drive response and resistance, is required to improve ICB efficacy through predictive biomarkers and novel combination therapies. In a recent study published in Genome Medicine (2), Hu et al. used a multi-modal approach to investigate changes in the NSCLC multicellular ecosystem that underpin response to chemotherapy and ICB combination treatment in NSCLC. This study utilised bulk tissue and single cell (sc)RNA-sequencing, immunostaining and in silico modelling to investigate mechanisms associated with major pathological response (MPR). ScRNA-sequencing was performed on 3 treatment naive (TN) tumour biopsies; and 12 surgically resected tumour specimens following neo-adjuvant chemotherapy and ICB; a subset of these samples was also analysed by multi-marker immunostaining. Pathological response varied within the 12 post-treatment samples, with MPR achieved in 4 cases (leaving 8 exhibiting non-MPR). An independent TN cohort (n=21) was also examined with bulk RNA-sequencing, enabling the evaluation of molecular signatures' potential as predictive biomarkers. Despite this relatively small sample size, the study revealed multiple cell subpopulations and molecular signatures that varied between pathological response groups, providing valuable insight into key drivers of response to combined chemotherapy and ICB in NSCLC ( Figure 1 ). Furthermore, many of these findings may also have implications for other solid cancers, as key findings were validated in cohorts of melanoma patients treated with ICBs. The principle aim of ICB is to overcome CD8 + T-cell suppression and initiate cytotoxic clearance of tumour cells (3). In NSCLC, ICB treatments most commonly target the interaction between the PD-1 receptor and ligands PD-L1/L2. Nivolumab (anti-PD-1) was the first immune checkpoint inhibitor approved for use in lung cancer, soon followed by pembrolizumab, cemiplimab, sintilimab, toripalimab and camrelizumab (the latter three were used in Hu et al. 's study). Anti-PD-L1 drugs atezolizumab and durvalumab have also been approved for use in NSCLC. Depending on national regulatory approvals, these drugs are used in a variety of clinical settings: including first-line neo-adjuvant therapy in combination with chemotherapy (as investigated in Hu et al. 's study); adjuvant treatment following surgery and chemotherapy; second-line monotherapy and as consolidation therapy in patients unable to undergo surgery (4-7). Given that the efficacy of PD-1 blockade is predicated on establishing an adaptive immune response against the tumour, it is not surprising that Hu et al. found significant enrichment of T-cells and B-cells within the stromal compartment of MPR samples compared to both TN and non-MPR samples. However, closer examination of the phenotypic heterogeneity within these immune microenvironments, enabled by scRNA-seq analysis, demonstrated that specific immune cell subpopulations were variably abundant between these sample groups and also highlighted novel epithelial phenotypes associated with treatment response. Ten clusters of epithelial cells were identified in Hu et al. 's scRNA-seq analysis. Non-malignant (diploid) clusters (representing AT1, AT2, club, basal and ciliated cells), were generally enriched in the MPR group. This was suggested to represent alveolar regeneration following ablation of the malignant epithelium in patients who positively responded to therapy. Notably, CX3CL1 and genes associated with antigen presentation via major histocompatibility complex (MHC)-Class II were found to be up-regulated in the epithelial compartment of MPR samples. MHC-Class II machinery is typically expressed by profess -Abstract Truncated-
oncology,respiratory system