Your browser doesn't support javascript.
loading
Influence of TP53 Comutation on the Tumor Immune Microenvironment and Clinical Outcomes With Immune Checkpoint Inhibitors in STK11-Mutant Non-Small-Cell Lung Cancer.
Naqash, Abdul Rafeh; Floudas, Charalampos S; Aber, Etan; Maoz, Asaf; Nassar, Amin H; Adib, Elio; Choucair, Khalil; Xiu, Joanne; Baca, Yasmine; Ricciuti, Biagio; Alessi, Joao V; Awad, Mark M; Kim, Chul; Judd, Julia; Raez, Luis E; Lopes, Gilberto; Nieva, Jorge J; Borghaei, Hossein; Takebe, Naoko; Ma, Patrick C; Halmos, Balazs; Kwiatkowski, David J; Liu, Stephen V; Mamdani, Hirva.
Afiliación
  • Naqash AR; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK.
  • Floudas CS; Center for Immuno-Oncology, National Cancer Institute, NIH, Bethesda, MD.
  • Aber E; Center for Immuno-Oncology, National Cancer Institute, NIH, Bethesda, MD.
  • Maoz A; Dana Farber Cancer Institute, Boston, MA.
  • Nassar AH; Department of Hematology/Oncology, Yale New Haven Hospital, New Haven, CT.
  • Adib E; Dana Farber Cancer Institute, Boston, MA.
  • Choucair K; Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI.
  • Xiu J; Caris Life Sciences, Phoenix, AZ.
  • Baca Y; Caris Life Sciences, Phoenix, AZ.
  • Ricciuti B; Dana Farber Cancer Institute, Boston, MA.
  • Alessi JV; Dana Farber Cancer Institute, Boston, MA.
  • Awad MM; Dana Farber Cancer Institute, Boston, MA.
  • Kim C; Department of Hematology and Oncology, Georgetown University, Washington, DC.
  • Judd J; Fox Chase Cancer Center, Philadelphia, PA.
  • Raez LE; Memorial Cancer Institute//Florida Atlantic University (FAU), Miami, FL.
  • Lopes G; University of Miami Miller School of Medicine, Miami, FL.
  • Nieva JJ; University of Southern California, Los Angeles, CA.
  • Borghaei H; Fox Chase Cancer Center, Philadelphia, PA.
  • Takebe N; Developmental Therapeutics Clinic, National Cancer Institute, Bethesda, MD.
  • Ma PC; Department of Hematology/ Oncology, Penn State Cancer Institute, Hershey, PA.
  • Halmos B; Medical Oncology, Albert Einstein College of Medicine, NY.
  • Kwiatkowski DJ; Dana Farber Cancer Institute, Boston, MA.
  • Liu SV; Department of Hematology and Oncology, Georgetown University, Washington, DC.
  • Mamdani H; Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI.
JCO Precis Oncol ; 8: e2300371, 2024 Feb.
Article en En | MEDLINE | ID: mdl-38330261
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 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 = 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 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 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 = .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.
Asunto(s)

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Carcinoma de Pulmón de Células no Pequeñas / Antineoplásicos Inmunológicos / Neoplasias Pulmonares Tipo de estudio: Systematic_reviews Límite: Humans Idioma: En Revista: JCO Precis Oncol Año: 2024 Tipo del documento: Article

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Carcinoma de Pulmón de Células no Pequeñas / Antineoplásicos Inmunológicos / Neoplasias Pulmonares Tipo de estudio: Systematic_reviews Límite: Humans Idioma: En Revista: JCO Precis Oncol Año: 2024 Tipo del documento: Article