TY - JOUR
T1 - Influence of TP53 Comutation on the Tumor Immune Microenvironment and Clinical Outcomes With Immune Checkpoint Inhibitors in STK11-Mutant Non–Small-Cell Lung Cancer
AU - Naqash, Abdul Rafeh
AU - Floudas, Charalampos S.
AU - Aber, Etan
AU - Maoz, Asaf
AU - Nassar, Amin H.
AU - Adib, Elio
AU - Choucair, Khalil
AU - Xiu, Joanne
AU - Baca, Yasmine
AU - Ricciuti, Biagio
AU - Alessi, Joao V.
AU - Awad, Mark M.
AU - Kim, Chul
AU - Judd, Julia
AU - Raez, Luis E.
AU - Lopes, Gilberto
AU - Nieva, Jorge J.
AU - Borghaei, Hossein
AU - Takebe, Naoko
AU - Ma, Patrick C.
AU - Halmos, Balazs
AU - Kwiatkowski, David J.
AU - Liu, Stephen V.
AU - Mamdani, Hirva
N1 - Publisher Copyright:
© 2024 by American Society of Clinical Oncology.
PY - 2024/2/1
Y1 - 2024/2/1
N2 - 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 NSCLC tumors (N 5 16,896) were analyzed by next-generation sequencing METHODS (DNA-Seq/592 genes). A subset (n 5 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 5 34) and the study by Rizvi et al (n 5 49) were used to model progression-free survival (PFS), and a separate ICI-treated cohort (n 5 53) from Dana-Farber Cancer Institute (DFCI) was used to assess time to treatment failure (TTF) and tumor RECIST response for STK11mutTP53mut versus STK11mutTP53wt 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 STK11mut (n 5 463) for stimulator of interferon-gamma (STING) pathway genes identified a STING-high cluster, which was significantly enriched in TP53mut NSCLC (P < .01). Compared with STK11mutTP53wt, tumors with STK11mutTP53mut had higher CD81T 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 STK11mutTP53mut. In the DFCI cohort, compared with the STK11mut TP53wt cohort, the STK11mutTP53mut tumors had higher objective response rates (42.9% v 16.7%; P 5 .04) and also had longer TTF (14.5 v 4.5 months, P adj 5 .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.
AB - 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 NSCLC tumors (N 5 16,896) were analyzed by next-generation sequencing METHODS (DNA-Seq/592 genes). A subset (n 5 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 5 34) and the study by Rizvi et al (n 5 49) were used to model progression-free survival (PFS), and a separate ICI-treated cohort (n 5 53) from Dana-Farber Cancer Institute (DFCI) was used to assess time to treatment failure (TTF) and tumor RECIST response for STK11mutTP53mut versus STK11mutTP53wt 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 STK11mut (n 5 463) for stimulator of interferon-gamma (STING) pathway genes identified a STING-high cluster, which was significantly enriched in TP53mut NSCLC (P < .01). Compared with STK11mutTP53wt, tumors with STK11mutTP53mut had higher CD81T 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 STK11mutTP53mut. In the DFCI cohort, compared with the STK11mut TP53wt cohort, the STK11mutTP53mut tumors had higher objective response rates (42.9% v 16.7%; P 5 .04) and also had longer TTF (14.5 v 4.5 months, P adj 5 .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.
UR - http://www.scopus.com/inward/record.url?scp=85200413840&partnerID=8YFLogxK
UR - http://www.scopus.com/inward/citedby.url?scp=85200413840&partnerID=8YFLogxK
U2 - 10.1200/PO.23.00371
DO - 10.1200/PO.23.00371
M3 - Article
C2 - 38330261
AN - SCOPUS:85200413840
SN - 2473-4284
VL - 8
JO - JCO Precision Oncology
JF - JCO Precision Oncology
M1 - e2300371
ER -