Influence of TP53 Comutation on the Tumor Immune Microenvironment and Clinical Outcomes With Immune Checkpoint Inhibitors in STK11-Mutant Non–Small-Cell Lung Cancer

Author:

Naqash Abdul Rafeh1ORCID,Floudas Charalampos S.2ORCID,Aber Etan2,Maoz Asaf3ORCID,Nassar Amin H.4ORCID,Adib Elio3,Choucair Khalil5ORCID,Xiu Joanne6,Baca Yasmine6,Ricciuti Biagio3ORCID,Alessi Joao V.3ORCID,Awad Mark M.3ORCID,Kim Chul7ORCID,Judd Julia8ORCID,Raez Luis E.9ORCID,Lopes Gilberto10ORCID,Nieva Jorge J.11ORCID,Borghaei Hossein8ORCID,Takebe Naoko12ORCID,Ma Patrick C.13,Halmos Balazs14ORCID,Kwiatkowski David J.3ORCID,Liu Stephen V.7ORCID,Mamdani Hirva5ORCID

Affiliation:

1. Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK

2. Center for Immuno-Oncology, National Cancer Institute, NIH, Bethesda, MD

3. Dana Farber Cancer Institute, Boston, MA

4. Department of Hematology/Oncology, Yale New Haven Hospital, New Haven, CT

5. Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI

6. Caris Life Sciences, Phoenix, AZ

7. Department of Hematology and Oncology, Georgetown University, Washington, DC

8. Fox Chase Cancer Center, Philadelphia, PA

9. Memorial Cancer Institute//Florida Atlantic University (FAU), Miami, FL

10. University of Miami Miller School of Medicine, Miami, FL

11. University of Southern California, Los Angeles, CA

12. Developmental Therapeutics Clinic, National Cancer Institute, Bethesda, MD

13. Department of Hematology/ Oncology, Penn State Cancer Institute, Hershey, PA

14. Medical Oncology, Albert Einstein College of Medicine, NY

Abstract

PURPOSE Non–small-cell lung cancer (NSCLC) with STK11mut has inferior outcomes to immune checkpoint inhibitors (ICIs). Using multiomics, we evaluated whether a subtype of STK11mut NSCLC with a uniquely inflamed tumor immune microenvironment (TIME) harboring TP53 comutations could have favorable outcomes to ICIs. PATIENTS AND METHODS NSCLC tumors (N = 16,896) were analyzed by next-generation sequencing (DNA-Seq/592 genes). A subset (n = 5,034) underwent gene expression profiling (RNA-Seq/whole transcriptome). Exome-level neoantigen load for STK11mut NSCLC was obtained from published pan-immune analysis. Tumor immune cell content was obtained from transcriptome profiles using the microenvironment cell population (MCP) counter. ICI data from POPLAR/OAK (n = 34) and the study by Rizvi et al (n = 49) were used to model progression-free survival (PFS), and a separate ICI-treated cohort (n = 53) from Dana-Farber Cancer Institute (DFCI) was used to assess time to treatment failure (TTF) and tumor RECIST response for STK11mut TP53mut versus STK11mut TP53wt NSCLC. RESULTS Overall, 12.6% of NSCLC tumors had a STK11mut with the proportions of tumor mutational burden (TMB)-high (≥10 mut/Mb), PD-L1 ≥50%, and microsatellite instability-high being 38.3%, 11.8%, and 0.72%, respectively. Unsupervised hierarchical clustering of STK11 mut (n = 463) for stimulator of interferon-gamma (STING) pathway genes identified a STING-high cluster, which was significantly enriched in TP53mut NSCLC ( P < .01). Compared with STK11mut TP53wt, tumors with STK11mut TP53mut had higher CD8+T cells and natural killer cells ( P < .01), higher TMB ( P < .001) and neoantigen load ( P < .001), and increased expression of MYC and HIF-1A ( P < .01), along with higher expression ( P < .01) of glycolysis/glutamine metabolism genes. Meta-analysis of data from OAK/POPLAR and the study by Rizvi et al showed a trend toward improved PFS in patients with STK11mut TP53mut. In the DFCI cohort, compared with the STK11mut TP53wt cohort, the STK11mut TP53mut tumors had higher objective response rates (42.9% v 16.7%; P = .04) and also had longer TTF (14.5 v 4.5 months, P adj = .054) with ICI. CONCLUSION STK11mut NSCLC with TP53 comutation is a distinct subgroup with an immunologically active TIME and metabolic reprogramming. These properties should be exploited to guide patient selection for novel ICI-based combination approaches.

Publisher

American Society of Clinical Oncology (ASCO)

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3