» Articles » PMID: 32606052

Development and Validation of a Genomic Mutation Signature to Predict Response to PD-1 Inhibitors in Non-squamous NSCLC: a Multicohort Study

Overview
Date 2020 Jul 2
PMID 32606052
Citations 32
Authors
Affiliations
Soon will be listed here.
Abstract

Background: Genetic variations of some driver genes in non-small cell lung cancer (NSCLC) had shown potential impact on immune microenvironment and associated with response or resistance to programmed cell death protein 1 (PD-1) blockade immunotherapy. We therefore undertook an exploratory analysis to develop a genomic mutation signature (GMS) and predict the response to anti-PD-(L)1 therapy.

Methods: In this multicohort analysis, 316 patients with non-squamous NSCLC treated with anti-PD-(L)1 from three independent cohorts were included in our study. Tumor samples from the patients were molecularly profiled by MSK-IMPACT or whole exome sequencing. We developed a risk model named GMS based on the MSK training cohort (n=123). The predictive model was first validated in the separate internal MSK cohort (n=82) and then validated in an external cohort containing 111 patients from previously published clinical trials.

Results: A GMS risk model consisting of eight genes (, , , , , , , and ) was generated to classify patients into high and low GMS groups in the training cohort. Patients with high GMS in the training cohort had longer progression-free survival (hazard ratio (HR) 0.41, 0.28-0.61, p0.0001) and overall survival (HR 0.53, 0.32-0.89, p0.0275) compared with low GMS. We noted equivalent findings in the internal validation cohort and in the external validation cohort. The GMS was demonstrated as an independent predictive factor for anti-PD-(L)1 therapy comparing with tumor mutational burden. Meanwhile, GMS showed undifferentiated predictive value in patients with different clinicopathological features. Notably, both GMS and PD-L1 were independent predictors and demonstrated poorly correlated; inclusion of PD-L1 with GMS further improved the predictive capacity for PD-1 blockade immunotherapy.

Conclusions: Our study highlights the potential predictive value of GMS for immunotherapeutic benefit in non-squamous NSCLC. Besides, the combination of GMS and PD-L1 may serve as an optimal partner in guiding treatment decisions for anti-PD-(L)1 based therapy.

Citing Articles

The modification role and tumor association with a methyltransferase: KMT2C.

Jiao Y, Lv Y, Liu M, Liu Y, Han M, Xiong X Front Immunol. 2024; 15:1444923.

PMID: 39165358 PMC: 11333232. DOI: 10.3389/fimmu.2024.1444923.


Genomic correlates of the response to first-line PD-1 blockade plus chemotherapy in patients with advanced non-small-cell lung cancer.

Jiang T, Chen J, Wang H, Wu F, Chen X, Su C Chin Med J (Engl). 2024; .

PMID: 39164816 PMC: 11407809. DOI: 10.1097/CM9.0000000000003094.


Multiple omics integrative analysis identifies GARS1 as a novel prognostic and immunological biomarker: from pan-cancer to bladder cancer.

Liu W, Wei C, He Q, Chen Z, Zhuang W, Guo Y Sci Rep. 2024; 14(1):19025.

PMID: 39152248 PMC: 11329754. DOI: 10.1038/s41598-024-70041-1.


Integrating knowledge graphs into machine learning models for survival prediction and biomarker discovery in patients with non-small-cell lung cancer.

Fang C, Arango Argoty G, Kagiampakis I, Khalid M, Jacob E, Bulusu K J Transl Med. 2024; 22(1):726.

PMID: 39103897 PMC: 11302075. DOI: 10.1186/s12967-024-05509-9.


Single-cell sequencing reveals novel proliferative cell type: a key player in renal cell carcinoma prognosis and therapeutic response.

Ye B, Ji H, Zhu M, Wang A, Tang J, Liang Y Clin Exp Med. 2024; 24(1):167.

PMID: 39052149 PMC: 11272756. DOI: 10.1007/s10238-024-01424-x.


References
1.
Dong Z, Zhong W, Zhang X, Su J, Xie Z, Liu S . Potential Predictive Value of and Mutation Status for Response to PD-1 Blockade Immunotherapy in Lung Adenocarcinoma. Clin Cancer Res. 2017; 23(12):3012-3024. DOI: 10.1158/1078-0432.CCR-16-2554. View

2.
Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky J, Desrichard A . Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med. 2014; 371(23):2189-2199. PMC: 4315319. DOI: 10.1056/NEJMoa1406498. View

3.
Yuan Y, Liu L, Chen H, Wang Y, Xu Y, Mao H . Comprehensive Characterization of Molecular Differences in Cancer between Male and Female Patients. Cancer Cell. 2016; 29(5):711-722. PMC: 4864951. DOI: 10.1016/j.ccell.2016.04.001. View

4.
Dong Z, Wu S, Liao R, Huang S, Wu Y . Potential biomarker for checkpoint blockade immunotherapy and treatment strategy. Tumour Biol. 2016; 37(4):4251-61. DOI: 10.1007/s13277-016-4812-9. View

5.
Topalian S, Hodi F, Brahmer J, Gettinger S, Smith D, Mcdermott D . Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012; 366(26):2443-54. PMC: 3544539. DOI: 10.1056/NEJMoa1200690. View