Immune profiles according to EGFR mutant subtypes and correlation with PD-1/PD-L1 inhibitor therapies in lung adenocarcinoma

Background We examined the distributions of 22 immune cell types and the responses to PD-1/PD-L1 inhibitors according to EGFR mutation profile, in three independent datasets of lung adenocarcinoma (LUAD). Methods We used CIBERSORTx to analyze the distributions of immune cells, and tumor immune dysfunction and exclusion (TIDE) or tumor mutation burden (TMB) to analyze responses to anti-PD-1/PD-L1 therapy, in two public LUAD datasets. The results were verified with a validation set that included patients treated with PD-1/PD-L1 inhibitors. Results Compared to EGFR mutants, EGFR wild-type carcinomas had higher numbers of CD8+ T cells, CD4 memory activated T cells and neutrophils, and lower numbers of resting dendritic cells and resting mast cells, in two of the datasets. In our subgroup analyses, CD8+ T cells and CD4 memory activated T cells were more numerous in EGFR rare variants than in wild-types, L858R mutants, and exon 19 deletion mutants. In our TIDE or TMB analyses, EGFR rare variants were predicted to respond better to PD-1/PD-L1 inhibitors than wild-types, L858R mutants, and exon 19 deletion mutants. In the validation set verified by immunohistochemical staining, levels of CD8+ T cells in the EGFR rare variant or wild-type groups were significantly higher than in the EGFR L858R and exon 19 deletion groups. In patients treated with PD-1/PD-L1 inhibitors, the survival rates of patients with EGFR wild-type and rare mutant carcinomas were higher than those with L858R and exon 19 deletion carcinomas. Conclusion The EGFR rare mutation form of LUAD shows a higher immune activation state compared to wild-type, L858R, and exon 19 deletion variants, indicating it as a potential target for PD-1/PD-L1 inhibitor therapy.

[1]  Y. Shang,et al.  PD-L1 expression, tumor mutational burden, and immune cell infiltration in non-small cell lung cancer patients with epithelial growth factor receptor mutations , 2022, Frontiers in Oncology.

[2]  W. Zhong,et al.  Single-cell transcriptome analysis revealed a suppressive tumor immune microenvironment in EGFR mutant lung adenocarcinoma , 2022, Journal for ImmunoTherapy of Cancer.

[3]  J. Roth,et al.  Oncogene-specific differences in tumor mutational burden, PD-L1 expression, and outcomes from immunotherapy in non-small cell lung cancer , 2021, Journal for ImmunoTherapy of Cancer.

[4]  Taicheng Zhou,et al.  Molecular Subtypes and CD4+ Memory T Cell-Based Signature Associated With Clinical Outcomes in Gastric Cancer , 2021, Frontiers in Oncology.

[5]  J. Christensen,et al.  Cytotoxic CD8+ T cells in cancer and cancer immunotherapy , 2020, British Journal of Cancer.

[6]  E. Richardson,et al.  Revisiting the role of CD4+ T cells in cancer immunotherapy—new insights into old paradigms , 2020, Cancer Gene Therapy.

[7]  Sara R. Savage,et al.  Proteogenomic Characterization Reveals Therapeutic Vulnerabilities in Lung Adenocarcinoma , 2020, Cell.

[8]  K. Horimoto,et al.  CD4+ T-cell Immunity in the Peripheral Blood Correlates with Response to Anti-PD-1 Therapy , 2019, Cancer Immunology Research.

[9]  A. Drilon,et al.  Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[10]  E. Tartour,et al.  Distinct prognostic value of circulating anti-telomerase CD4+ Th1 immunity and exhausted PD-1+/TIM-3+ T cells in lung cancer , 2019, British Journal of Cancer.

[11]  N. Schultz,et al.  EGFR mutation subtypes and response to immune checkpoint blockade treatment in non-small-cell lung cancer , 2019, Annals of oncology : official journal of the European Society for Medical Oncology.

[12]  J. Gu,et al.  PD-L1 expression and EGFR status in advanced non-small-cell lung cancer patients receiving PD-1/PD-L1 inhibitors: a meta-analysis. , 2019, Future oncology.

[13]  Ash A. Alizadeh,et al.  Determining cell-type abundance and expression from bulk tissues with digital cytometry , 2019, Nature Biotechnology.

[14]  J. Galon,et al.  Approaches to treat immune hot, altered and cold tumours with combination immunotherapies , 2019, Nature Reviews Drug Discovery.

[15]  J. Lunceford,et al.  IFN-g – related mRNA profile predicts clinical response to PD-1 blockade , 2019 .

[16]  K. Syrigos,et al.  Immune Marker Profiling and Programmed Death Ligand 1 Expression Across NSCLC Mutations , 2018, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[17]  J. Luke,et al.  T Cell–Inflamed versus Non-T Cell–Inflamed Tumors: A Conceptual Framework for Cancer Immunotherapy Drug Development and Combination Therapy Selection , 2018, Cancer Immunology Research.

[18]  X. Liu,et al.  Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response , 2018, Nature Medicine.

[19]  J. Borst,et al.  CD4+ T cell help in cancer immunology and immunotherapy , 2018, Nature Reviews Immunology.

[20]  Caicun Zhou,et al.  Lung cancer in never smokers-the East Asian experience. , 2018, Translational lung cancer research.

[21]  Peter W. Laird,et al.  Cell-of-Origin Patterns Dominate the Molecular Classification of 10,000 Tumors from 33 Types of Cancer , 2018, Cell.

[22]  B. O'Sullivan,et al.  106PThe association between PD-L1 expression, EGFR mutation and ALK translocation in a series of 982 lung cancers , 2017 .

[23]  Yi-long Wu,et al.  EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer , 2017, Oncoimmunology.

[24]  J. Lunceford,et al.  IFN-&ggr;–related mRNA profile predicts clinical response to PD-1 blockade , 2017, The Journal of clinical investigation.

[25]  F. Hirsch,et al.  PD‐L1 Expression in Lung Cancer , 2016, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[26]  S. Digumarthy,et al.  EGFR Mutations and ALK Rearrangements Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non–Small Cell Lung Cancer: A Retrospective Analysis , 2016, Clinical Cancer Research.

[27]  L. Crinò,et al.  Nivolumab versus Docetaxel in Advanced Squamous-Cell Non-Small-Cell Lung Cancer. , 2015, The New England journal of medicine.

[28]  Martin L. Miller,et al.  Mutational landscape determines sensitivity to PD-1 blockade in non–small cell lung cancer , 2015, Science.

[29]  T. Gajewski,et al.  The STING pathway and the T cell-inflamed tumor microenvironment. , 2015, Trends in immunology.

[30]  Benjamin E. Gross,et al.  The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. , 2012, Cancer discovery.

[31]  R. Kennedy,et al.  Multiple roles for CD4+ T cells in anti‐tumor immune responses , 2008, Immunological reviews.

[32]  F. Schmidt Meta-Analysis , 2008 .

[33]  E. Tartour,et al.  Prognostic Value of Tumor-Infiltrating CD4+ T-Cell Subpopulations in Head and Neck Cancers , 2006, Clinical Cancer Research.

[34]  M. F. Parry,et al.  A Retrospective Analysis , 1990 .