A risk score combining co-expression modules related to myeloid cells and alternative splicing associates with response to PD-1/PD-L1 blockade in non-small cell lung cancer

Background Comprehensive analysis of transcriptomic profiles of non-small cell lung cancer (NSCLC) may provide novel evidence for biomarkers associated with response to PD-1/PD-L1 immune checkpoint blockade (ICB). Methods We utilized weighted gene co-expression network analysis (WGCNA) to analyze transcriptomic data from two NSCLC datasets from Gene Expression Omnibus (GSE135222 and GSE126044) that involved patients received ICB treatment. We evaluated the correlation of co-expression modules with ICB responsiveness and functionally annotated ICB-related modules using pathway enrichment analysis, single-cell RNA sequencing, flow cytometry and alternative splicing analysis. We built a risk score using Lasso-COX regression based on hub genes from ICB-related modules. We investigated the alteration of tumor microenvironment between high- and low- risk groups and the association of the risk score with previously established predictive biomarkers. Results Our results identified a black with positive correlation and a blue module with negative correlation to ICB responsiveness. The black module was enriched in pathway of T cell activation and antigen processing and presentation, and the genes assigned to it were consistently expressed on myeloid cells. We observed decreased alternative splicing events in samples with high signature scores of the blue module. The Lasso-COX analysis screened out three genes (EVI2B, DHX9, HNRNPM) and constructed a risk score from the hub genes of the two modules. We validated the predictive value of the risk score for poor response to ICB therapy in an in-house NSCLC cohort and a pan-cancer cohort from the KM-plotter database. The low-risk group had more immune-infiltrated microenvironment, with higher frequencies of precursor exhausted CD8+ T cells, tissue-resident CD8+ T cells, plasmacytoid dendritic cells and type 1 conventional dendritic cells, and a lower frequency of terminal exhausted CD8+ T cells, which may explain its superior response to ICB therapy. The significant correlation of the risk score to gene signature of tertiary lymphoid structure also implicated the possible mechanism of this predictive biomarker. Conclusions Our study identified two co-expression modules related to ICB responsiveness in NSCLC and developed a risk score accordingly, which could potentially serve as a predictive biomarker for ICB response.

[1]  L. Paz-Ares,et al.  At the crossroads of immunotherapy for oncogene-addicted subsets of NSCLC , 2023, Nature Reviews Clinical Oncology.

[2]  Caicun Zhou,et al.  Non‐small cell lung cancer in China , 2022, Cancer communications.

[3]  I. Wistuba,et al.  Predictive Biomarkers for Immunotherapy in Lung Cancer: Perspective from the IASLC Pathology Committee. , 2022, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[4]  Binzhi Qian,et al.  Macrophage diversity in cancer revisited in the era of single-cell omics. , 2022, Trends in immunology.

[5]  B. Nakken,et al.  Tetraspanin CD53 controls T cell immunity through regulation of CD45RO stability, mobility, and function. , 2022, Cell reports.

[6]  B. Győrffy,et al.  Transcriptomic datasets of cancer patients treated with immune-checkpoint inhibitors: a systematic review , 2022, Journal of Translational Medicine.

[7]  G. Zhu,et al.  The CD8α–PILRα interaction maintains CD8+ T cell quiescence , 2022, Science.

[8]  Joe Y. Chang,et al.  Non-Small Cell Lung Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. , 2022, Journal of the National Comprehensive Cancer Network : JNCCN.

[9]  A. Saliba,et al.  Type 1 conventional dendritic cells maintain and guide the differentiation of precursors of exhausted T cells in distinct cellular niches. , 2022, Immunity.

[10]  Shugeng Gao,et al.  Worldwide burden and epidemiological trends of tracheal, bronchus, and lung cancer: A population-based study , 2022, EBioMedicine.

[11]  C. Sautès-Fridman,et al.  C1q+ macrophages: passengers or drivers of cancer progression. , 2022, Trends in cancer.

[12]  B. Nabet,et al.  Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer. , 2022, Cancer cell.

[13]  P. Laurent-Puig,et al.  Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. , 2022, Immunity.

[14]  M. Hellmann,et al.  First-Line Immunotherapy for Non-Small-Cell Lung Cancer. , 2022, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[15]  Xueda Hu,et al.  Temporal single-cell tracing reveals clonal revival and expansion of precursor exhausted T cells during anti-PD-1 therapy in lung cancer , 2021, Nature Cancer.

[16]  Xueda Hu,et al.  Pan-cancer single-cell landscape of tumor-infiltrating T cells , 2021, Science.

[17]  S. Keith,et al.  Association Between Sex and Immune Checkpoint Inhibitor Outcomes for Patients With Melanoma , 2021, JAMA network open.

[18]  Yi-long Wu,et al.  Prediction of unfavourable response to checkpoint blockade in lung cancer patients through an integrated tumour-immune expression score , 2021, Translational oncology.

[19]  G. Giaccone,et al.  Sex-based differences in response to anti-PD-1 or PD-L1 treatment in patients with non-small-cell lung cancer expressing high PD-L1 levels. A systematic review and meta-analysis of randomized clinical trials , 2021, ESMO open.

[20]  V. Swarup,et al.  Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer’s disease , 2021, Nature Genetics.

[21]  Xiaochen Bo,et al.  clusterProfiler 4.0: A universal enrichment tool for interpreting omics data , 2021, Innovation.

[22]  J Zhang,et al.  Single-cell profiling of tumor heterogeneity and the microenvironment in advanced non-small cell lung cancer , 2021, Nature Communications.

[23]  I. Wistuba,et al.  PD-L1 as a biomarker of response to immune-checkpoint inhibitors , 2021, Nature Reviews Clinical Oncology.

[24]  A. Jemal,et al.  Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries , 2021, CA: a cancer journal for clinicians.

[25]  Xueda Hu,et al.  A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells , 2021, Cell.

[26]  P. Van Loo,et al.  Meta-analysis of tumor- and T cell-intrinsic mechanisms of sensitization to checkpoint inhibition , 2021, Cell.

[27]  A. Goodman,et al.  The Challenges of Tumor Mutational Burden as an Immunotherapy Biomarker. , 2020, Cancer cell.

[28]  Raphael Gottardo,et al.  Integrated analysis of multimodal single-cell data , 2020, Cell.

[29]  T. Owonikoko,et al.  Efficacy and safety of immune checkpoint blockade in self‐identified Black patients with advanced non–small cell lung cancer , 2020, Cancer.

[30]  Young-Joon Kim,et al.  Genome-wide identification of differentially methylated promoters and enhancers associated with response to anti-PD-1 therapy in non-small cell lung cancer , 2020, Experimental & Molecular Medicine.

[31]  Xiaole Shirley Liu,et al.  TIMER2.0 for analysis of tumor-infiltrating immune cells , 2020, Nucleic Acids Res..

[32]  J. Valcárcel,et al.  Roles and mechanisms of alternative splicing in cancer — implications for care , 2020, Nature Reviews Clinical Oncology.

[33]  G. Mills,et al.  Sex-associated molecular differences for cancer immunotherapy , 2020, Nature Communications.

[34]  C. Klein,et al.  Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy , 2020, Science Translational Medicine.

[35]  A. Kallies,et al.  Precursor exhausted T cells: key to successful immunotherapy? , 2019, Nature Reviews Immunology.

[36]  J. Stebbing,et al.  A meta-analysis comparing responses of Asian versus non-Asian cancer patients to PD-1 and PD-L1 inhibitor-based therapy , 2020, Oncoimmunology.

[37]  D. Schadendorf,et al.  Tertiary lymphoid structures improve immunotherapy and survival in melanoma , 2020, Nature.

[38]  Jung Kyoon Choi,et al.  DNA methylation loss promotes immune evasion of tumours with high mutation and copy number load , 2019, Nature Communications.

[39]  A. Abernethy,et al.  Real‐world progression, treatment, and survival outcomes during rapid adoption of immunotherapy for advanced non–small cell lung cancer , 2019, Cancer.

[40]  D. Baltimore,et al.  Alternative mRNA splicing in cancer immunotherapy , 2019, Nature Reviews Immunology.

[41]  G. Freeman,et al.  Intratumoral Activity of the CXCR3 Chemokine System Is Required for the Efficacy of Anti-PD-1 Therapy. , 2019, Immunity.

[42]  E. Wherry,et al.  CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. , 2019, Annual review of immunology.

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

[44]  J. Lunceford,et al.  T-Cell-Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated With Pembrolizumab Across 20 Cancers: KEYNOTE-028. , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[45]  Fan Zhang,et al.  Fast, sensitive, and accurate integration of single cell data with Harmony , 2018, bioRxiv.

[46]  K. Hiom,et al.  DHX9 helicase promotes R-loop formation in cells with impaired RNA splicing , 2018, Nature Communications.

[47]  Yassen Assenov,et al.  Maftools: efficient and comprehensive analysis of somatic variants in cancer , 2018, Genome research.

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

[49]  Marieke E. Ijsselsteijn,et al.  Molecular and pharmacological modulators of the tumor immune contexture revealed by deconvolution of RNA-seq data , 2018, bioRxiv.

[50]  Steven J. M. Jones,et al.  Comprehensive Analysis of Alternative Splicing Across Tumors from 8,705 Patients , 2018, Cancer cell.

[51]  P. Carmeliet,et al.  Phenotype molding of stromal cells in the lung tumor microenvironment , 2018, Nature Medicine.

[52]  R. V. van Lier,et al.  Tissue-resident memory T cells at the center of immunity to solid tumors , 2018, Nature Immunology.

[53]  A. Gupta,et al.  Interferon Gamma Messenger RNA Signature in Tumor Biopsies Predicts Outcomes in Patients with Non–Small Cell Lung Carcinoma or Urothelial Cancer Treated with Durvalumab , 2018, Clinical Cancer Research.

[54]  Matthew A. Wyczalkowski,et al.  Systematic Analysis of Splice-Site-Creating Mutations in Cancer , 2018, Cell reports.

[55]  R. Weinberg,et al.  Understanding the tumor immune microenvironment (TIME) for effective therapy , 2018, Nature Medicine.

[56]  R. Bourgon,et al.  TGF-β attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells , 2018, Nature.

[57]  P. Hegde,et al.  MA 05.09 Pre-Existing Immunity Measured by Teff Gene Expression in Tumor Tissue is Associated with Atezolizumad Efficacy in NSCLC , 2017 .

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

[59]  A. Butte,et al.  xCell: digitally portraying the tissue cellular heterogeneity landscape , 2017, bioRxiv.

[60]  Jun S. Liu,et al.  Comprehensive analyses of tumor immunity: implications for cancer immunotherapy , 2016, Genome Biology.

[61]  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.

[62]  Robert Brown,et al.  TCGASpliceSeq a compendium of alternative mRNA splicing in cancer , 2015, Nucleic Acids Res..

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

[64]  Yan Song,et al.  Targeting PD1–PDL1 immune checkpoint in plasmacytoid dendritic cell interactions with T cells, natural killer cells and multiple myeloma cells , 2015, Leukemia.

[65]  R. Emerson,et al.  PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.

[66]  Chonghui Cheng,et al.  Cell type-restricted activity of hnRNPM promotes breast cancer metastasis via regulating alternative splicing , 2014, Genes & development.

[67]  H. Choy,et al.  hnRNP M facilitates exon 7 inclusion of SMN2 pre-mRNA in spinal muscular atrophy by targeting an enhancer on exon 7. , 2014, Biochimica et biophysica acta.

[68]  G. Getz,et al.  Inferring tumour purity and stromal and immune cell admixture from expression data , 2013, Nature Communications.

[69]  Justin Guinney,et al.  GSVA: gene set variation analysis for microarray and RNA-Seq data , 2013, BMC Bioinformatics.

[70]  D. Rotin,et al.  LAPTM5 Protein Is a Positive Regulator of Proinflammatory Signaling Pathways in Macrophages* , 2012, The Journal of Biological Chemistry.

[71]  Peter Langfelder,et al.  Fast R Functions for Robust Correlations and Hierarchical Clustering. , 2012, Journal of statistical software.

[72]  Trevor Hastie,et al.  Regularization Paths for Generalized Linear Models via Coordinate Descent. , 2010, Journal of statistical software.

[73]  S. Horvath,et al.  WGCNA: an R package for weighted correlation network analysis , 2008, BMC Bioinformatics.

[74]  G. Eele The Challenges , 2006 .

[75]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[76]  Liquid Biopsies Non-small-cell lung cancer , 2015, Nature Reviews Disease Primers.

[77]  L. Schwartz,et al.  New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). , 2009, European journal of cancer.

[78]  Andrew E. Jaffe,et al.  Bioinformatics Applications Note Gene Expression the Sva Package for Removing Batch Effects and Other Unwanted Variation in High-throughput Experiments , 2022 .