Abstract

Immune checkpoint inhibitors (ICIs) can elicit durable antitumor responses in patients with non-small cell lung cancer (NSCLC), but only 20% to 25% of patients respond to treatment. As important genes in the DNA damage response pathway, comutation in the tumor protein p53 (TP53) and ataxia-telangiectasia mutated (ATM) genes may be associated with genomic instability and hypermutation. However, the prevalence of TP53 and ATM comutation and its association with response to ICIs are not fully understood. To examine the prevalence of the TP53 and ATM comutation, the potential mechanism, and its association with response to ICIs among patients with NSCLC. This multiple-cohort study included patients with NSCLC from the Geneplus Institute, the Cancer Genome Atlas (TCGA), and the Memorial Sloan Kettering Cancer Center (MSKCC) databases and from the POPLAR and OAK randomized controlled trials. Samples in the Geneplus cohort were collected and analyzed from April 30, 2015, through February 28, 2019. Data from TCGA, the MSKCC, and the POPLAR and OAK cohorts were obtained on January 1, 2019, and analyzed from January 1 to April 10, 2019. Next-generation sequencing assays were performed on tumor samples by the Geneplus Institute. Genomic, transcriptomic, and clinical data were obtained from TCGA and MSKCC databases. Comprehensive genetic profiling was performed to determine the prevalence of TP53 and ATM comutation and its association with prognosis and response to ICIs. The main outcomes were TP53 and ATM comutation frequency, overall survival (OS), progression-free survival, gene set enrichment analysis, and immune profile in NSCLC. Patients with NSCLC analyzed in this study included 2020 patients in the Geneplus cohort (mean [SD] age, 59.5 [10.5] years; 1168 [57.8%] men), 1031 patients in TCGA cohort (mean [SD] age, 66.2 [9.5] years; 579 [56.2%] men), 1527 patients in the MSKCC cohort (662 [43.4%] men), 350 patients in the MSKCC cohort who were treated with ICIs (mean [SD] age, 61.4 [13.8] years; 170 [48.6%] men), and 853 patients in the POPLAR and OAK cohort (mean [SD] age, 63.0 [9.1] years; 527 [61.8%] men). Sites of TP53 and ATM comutation were found scattered throughout the genes, and no significant difference was observed in the frequency of TP53 and ATM comutation within the histologic subtypes and driver genes. In 5 independent cohorts of patients with NSCLC, TP53 and ATM comutation was associated with a significantly higher tumor mutation burden compared with the sole mutation and with no mutation (TCGA, MSKCC, Geneplus, and POPLAR and OAK cohort). Among patients treated with ICIs in the MSKCC cohort, TP53 and ATM comutation was associated with better OS than a single mutation and no mutation among patients with any cancer (median OS: TP53 and ATM comutation, not reached; TP53 mutation alone, 14.0 months; ATM mutation alone, 40.0 months; no mutation, 22.0 months; P = .001; NSCLC median OS: TP53 and ATM comutation, not reached; TP53 mutation alone, 11.0 months; ATM mutation alone, 16.0 months; no mutation, 14.0 months; P = .24). Similar results were found in the POPLAR and OAK cohort in which the disease control benefit rate, progression-free survival, and OS were all greater in patients with the TP53 and ATM comutation compared with the other 3 groups (median progression-free survival: TP53 and ATM comutation, 10.4 months; TP53 mutation, 1.6 months; ATM mutation, 3.5 months; no mutation, 2.8 months; P = .01; median OS: TP53 and ATM comutation, 22.1 months; TP53 mutation, 8.3 months; ATM mutation, 15.8 months; no mutation, 15.3 months; P = .002). This study's findings suggest that the TP53 and ATM comutation occurs in a subgroup of patients with NSCLC and is associated with an increased tumor mutation burden and response to ICIs. This suggests that TP53 and ATM comutation may have implications as a biomarker for guiding ICI treatment.

Highlights

  • Recent developments in immune checkpoint inhibitors (ICIs) have improved the survival in a multitude of advanced malignant neoplasms, including non–small cell lung cancer (NSCLC).[1,2,3,4,5,6] most patients receiving ICIs do not derive a benefit

  • Patients with NSCLC analyzed in this study included 2020 patients in the Geneplus cohort, 1031 patients in the Cancer Genome Atlas (TCGA) cohort, 1527 patients in the Memorial Sloan Kettering Cancer Center (MSKCC) cohort (662 [43.4%] men), 350 patients in the MSKCC cohort who were treated with ICIs, and 853 patients in the POPLAR and OAK cohort

  • This study’s findings suggest that the tumor protein p53 (TP53) and ataxia-telangiectasia mutated (ATM) comutation occurs in a subgroup of patients with NSCLC and is associated with an increased tumor mutation burden and response to ICIs

Read more

Summary

Introduction

Recent developments in immune checkpoint inhibitors (ICIs) have improved the survival in a multitude of advanced malignant neoplasms, including non–small cell lung cancer (NSCLC).[1,2,3,4,5,6] most patients receiving ICIs do not derive a benefit. An important aspect of immunotherapy is how to identify and develop predictive biomarkers of ICI response.[7,8] The commonly used clinically applicable predictive biomarker has been programmed cell death 1 ligand 1 (PD-L1), known as cluster of differentiation 274 or CD274 (OMIM 605402) determined with immunohistochemistry. The sensitivity and specificity of PD-L1 expression are modest,[9] which has prompted the search for additional tools.[10,11] Mutation of mismatch repair (MMR) genes is known contribute to damage to the DNA damage response (DDR) pathway, which is associated with an increase in the tumor mutation burden (TMB),[12] including catalytic subunit of DNA polymerase epsilon (POLE) (OMIM 174762) gene; DNA polymerase δ 1, catalytic subunit (POLD1) (OMIM 174761); breast cancer susceptibility gene 1 (BRCA1) (OMIM 113705); and breast cancer susceptibility gene 2 (BRCA2) (OMIM 600185), which are associated with the efficacy of ICIs in treating lung cancer, but the frequency of occurrence among patients with lung cancer is low.[13,14,15]

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call