Genomic and transcriptional alterations in first-line chemotherapy exert a potentially unfavorable influence on subsequent immunotherapy in NSCLC

Background: Recent studies in non-small cell lung cancer (NSCLC) patients have demonstrated that first-line immunotherapy is associated with better therapeutic response than second-line treatment. So far, the mechanisms need to be explored. It prompted us to evaluate the association between first-line chemotherapy and subsequent immunotherapy in NSCLC as well as its underlying mechanisms at the genomic and transcriptomic level. Methods: We launched a prospective, observational clinical study, paired tumor biopsies before and after chemotherapy were collected from NSCLC patients without tyrosine kinase inhibitor (TKI)-related driver gene mutations. The analyses included genomic and transcriptional changes performed by next-generation sequencing (NGS)-based whole-exome sequencing (WES) and messager ribonucleic acid (mRNA) sequencing. Characteristic mutational alterations in 1574 genes were investigated based on mutational status, clinicopathological factors, and chemotherapy responses. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, neoantigen prediction and intratumoral heterogeneity evaluation were also performed. Results: Samples and information from 32 NSCLC patients without TKI-related driver gene mutations were obtained. We found that the total number of single nucleotide variants (SNV)/insertion-deletion (INDEL) mutations did not change significantly after chemotherapy. The tumor mutation burden (TMB) decreased significantly after chemotherapy in smoking patients and the decreased TMB correlated with a better survival of smoking patients. The change in copy number variations (CNVs) exhibited a decreasing trend during chemotherapy. Subsequent analysis at mRNA level revealed a significant decrease in the expression levels of genes related to antigen processing and presentation as well as other factors relevant for response to immunotherapy. Pathway enrichment analysis confirmed that the immune-related signaling pathways or biological processes were decreased after first-line chemotherapy. Conclusions: Our study presents an explanation for the unsatisfactory results of immunotherapy when given after chemotherapy, and suggests that first-line chemotherapy is able to influence the tumor microenvironment and decrease the efficacy of subsequent immunotherapy. The study was registered at ClinicalTrials.gov, number NCT03764917, and has completed enrolment; patients are still in follow-up.

[1]  L. Norian,et al.  Manipulating the Wnt/β-catenin signaling pathway to promote anti-tumor immune infiltration into the TME to sensitize ovarian cancer to ICB therapy. , 2020, Gynecologic oncology.

[2]  M. Maurice,et al.  Mutations and mechanisms of WNT pathway tumour suppressors in cancer , 2020, Nature reviews. Cancer.

[3]  Wei-Chung Cheng,et al.  Targeting positive feedback between BASP1 and EGFR as a therapeutic strategy for lung cancer progression , 2020, Theranostics.

[4]  C. Garbe,et al.  Distinct Mutation Patterns Reveal Melanoma Subtypes and Influence Immunotherapy Response in Advanced Melanoma Patients , 2020, Cancers.

[5]  L. Shen,et al.  Tumor copy-number alterations predict response to immune-checkpoint-blockade in gastrointestinal cancer , 2020, Journal for ImmunoTherapy of Cancer.

[6]  Gang Wu,et al.  USP7 targeting modulates anti-tumor immune response by reprogramming Tumor-associated Macrophages in Lung Cancer , 2020, Theranostics.

[7]  Hsuan-Yu Chen,et al.  DNA methylation maintains the CLDN1-EPHB6-SLUG axis to enhance chemotherapeutic efficacy and inhibit lung cancer progression , 2020, Theranostics.

[8]  Z. Lou,et al.  Identification of Deleterious NOTCH Mutation as Novel Predictor to Efficacious Immunotherapy in NSCLC , 2020, Clinical Cancer Research.

[9]  E. Winer,et al.  Tumor Mutational Burden and PTEN Alterations as Molecular Correlates of Response to PD-1/L1 Blockade in Metastatic Triple-Negative Breast Cancer , 2020, Clinical Cancer Research.

[10]  E. Sokol,et al.  Tumor Mutational Burden as a Predictive Biomarker for Response to Immune Checkpoint Inhibitors: A Review of Current Evidence. , 2019, The oncologist.

[11]  Yuan Lin,et al.  Genome landscapes of rectal cancer before and after preoperative chemoradiotherapy , 2019, Theranostics.

[12]  J. Clohessy,et al.  CAV1 - GLUT3 signaling is important for cellular energy and can be targeted by Atorvastatin in Non-Small Cell Lung Cancer , 2019, Theranostics.

[13]  Luca Mazzarella,et al.  Tumor mutational burden quantification from targeted gene panels: major advancements and challenges , 2019, Journal of Immunotherapy for Cancer.

[14]  Lihua Dong,et al.  Lung cancer in young adults aged 35 years or younger: A full-scale analysis and review , 2019, Journal of Cancer.

[15]  Chad M. Vanderbilt,et al.  JAK2, PD-L1, and PD-L2 (9p24.1) amplification in metastatic mucosal and cutaneous melanomas with durable response to immunotherapy. , 2019, Human pathology.

[16]  Jianying Zhou,et al.  Pembrolizumab versus chemotherapy for previously untreated, PD-L1-expressing, locally advanced or metastatic non-small-cell lung cancer (KEYNOTE-042): a randomised, open-label, controlled, phase 3 trial , 2019, The Lancet.

[17]  A. Zhu,et al.  Pharmacological inhibition of β-catenin/BCL9 interaction overcomes resistance to immune checkpoint blockades by modulating Treg cells , 2019, Science Advances.

[18]  M. Ladanyi,et al.  High Yield of RNA Sequencing for Targetable Kinase Fusions in Lung Adenocarcinomas with No Mitogenic Driver Alteration Detected by DNA Sequencing and Low Tumor Mutation Burden , 2019, Clinical Cancer Research.

[19]  J. Cheville,et al.  JAK2/PD-L1/PD-L2 (9p24.1) amplifications in renal cell carcinomas with sarcomatoid transformation: implications for clinical management , 2019, Modern Pathology.

[20]  Matthew D. Hellmann,et al.  First-Line Nivolumab Plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer (CheckMate 568): Outcomes by Programmed Death Ligand 1 and Tumor Mutational Burden as Biomarkers , 2019, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[21]  Chunhua Zhou,et al.  Real‐world outcomes of various regimens of recombinant human endostatin combined with chemotherapy in non‐driver gene mutation advanced non‐small cell lung cancer , 2019, Cancer medicine.

[22]  J. Luke,et al.  WNT/β-catenin Pathway Activation Correlates with Immune Exclusion across Human Cancers , 2019, Clinical Cancer Research.

[23]  John G Doench,et al.  Loss of ADAR1 in tumours overcomes resistance to immune checkpoint blockade , 2018, Nature.

[24]  Johannes G. Reiter,et al.  Minimal functional driver gene heterogeneity among untreated metastases , 2018, Science.

[25]  Chan Kwon Park,et al.  Overall survival of driver mutation-negative non-small cell lung cancer patients with COPD under chemotherapy compared to non-COPD non-small cell lung cancer patients , 2018, International journal of chronic obstructive pulmonary disease.

[26]  A. Costantini,et al.  Increased Response Rates to Salvage Chemotherapy Administered after PD-1/PD-L1 Inhibitors in Patients with Non-Small Cell Lung Cancer. , 2018, Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer.

[27]  Ahmet Zehir,et al.  Molecular Determinants of Response to Anti-Programmed Cell Death (PD)-1 and Anti-Programmed Death-Ligand 1 (PD-L1) Blockade in Patients With Non-Small-Cell Lung Cancer Profiled With Targeted Next-Generation Sequencing. , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  Joshua D. Campbell,et al.  NetSig: network-based discovery from cancer genomes , 2017, Nature Methods.

[29]  Guoping Cai,et al.  Impaired HLA Class I Antigen Processing and Presentation as a Mechanism of Acquired Resistance to Immune Checkpoint Inhibitors in Lung Cancer. , 2017, Cancer discovery.

[30]  T. Bivona,et al.  Understanding and targeting resistance mechanisms in NSCLC , 2017, Nature Reviews Cancer.

[31]  Ryan Emerson,et al.  TCR Repertoire Intratumor Heterogeneity in Localized Lung Adenocarcinomas: An Association with Predicted Neoantigen Heterogeneity and Postsurgical Recurrence. , 2017, Cancer discovery.

[32]  P. Stephens,et al.  Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers , 2017, Molecular Cancer Therapeutics.

[33]  S. Khozin,et al.  Milestone Analyses of Immune Checkpoint Inhibitors, Targeted Therapy, and Conventional Therapy in Metastatic Non–Small Cell Lung Cancer Trials: A Meta-analysis , 2017, JAMA oncology.

[34]  Lee Ann McCue,et al.  FQC Dashboard: integrates FastQC results into a web-based, interactive, and extensible FASTQ quality control tool , 2017, Bioinform..

[35]  B. Carneiro,et al.  Wnt/beta-catenin pathway: modulating anticancer immune response , 2017, Journal of Hematology & Oncology.

[36]  Nicolai J. Birkbak,et al.  Tracking the Evolution of Non‐Small‐Cell Lung Cancer , 2017, The New England journal of medicine.

[37]  Jedd D. Wolchok,et al.  T-cell invigoration to tumour burden ratio associated with anti-PD-1 response , 2017, Nature.

[38]  D. Rimm,et al.  Nuclear IRF-1 expression as a mechanism to assess “Capability” to express PD-L1 and response to PD-1 therapy in metastatic melanoma , 2017, Journal of Immunotherapy for Cancer.

[39]  Alexander V. Favorov,et al.  Transcriptional Mechanisms of Resistance to Anti–PD-1 Therapy , 2017, Clinical Cancer Research.

[40]  J. Lee,et al.  The expression profiles and regulation of PD-L1 in tumor-induced myeloid-derived suppressor cells , 2016, Oncoimmunology.

[41]  Y. Shentu,et al.  Pembrolizumab versus Chemotherapy for PD-L1-Positive Non-Small-Cell Lung Cancer. , 2016, The New England journal of medicine.

[42]  F. Hirsch,et al.  New and emerging targeted treatments in advanced non-small-cell lung cancer , 2016, The Lancet.

[43]  Keunchil Park,et al.  Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial , 2016, The Lancet.

[44]  Yu Shyr,et al.  Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy , 2016, Nature Communications.

[45]  J. Taube,et al.  Association of PD-1, PD-1 Ligands, and Other Features of the Tumor Immune Microenvironment with Response to Anti–PD-1 Therapy , 2014, Clinical Cancer Research.

[46]  Hugo Y. K. Lam,et al.  Performance comparison of exome DNA sequencing technologies , 2011, Nature Biotechnology.

[47]  W. Muller,et al.  Tenascin-C modulates tumor stroma and monocyte/macrophage recruitment but not tumor growth or metastasis in a mouse strain with spontaneous mammary cancer. , 1999, Journal of cell science.

[48]  T. Nakamaki,et al.  [Development of cardiac tamponade and emergence of arrhythmia during chemotherapy for diffuse large B-cell lymphoma]. , 2019, [Rinsho ketsueki] The Japanese journal of clinical hematology.

[49]  J. Lunceford,et al.  IFN- γ –related mRNA profile predicts clinical response to PD-1 blockade , 2017 .