Login / Signup

Selective multi-kinase inhibition sensitizes mesenchymal pancreatic cancer to immune checkpoint blockade by remodeling the tumor microenvironment.

Chiara FalcomatàStefanie BärthelSebastian A WidholzChristian SchneeweisJuan José MonteroAlbulena ToskaJonas MirThorsten KaltenbacherJeannine HeetmeyerJonathan J SwietlikJing-Yuan ChengBianca TeodorescuOliver ReichertConstantin SchmittKathrin GrabichlerAndrea ColuccioFabio BonioloChristian VeltkampMagdalena ZukowskaAngelica Arenas VargasWoo Hyun PaikMoritz JesinghausKatja SteigerRoman MareschRupert ÖllingerTim AmmonOlga BaranovMaria S RoblesJulia RechenbergerBernhard KusterFelix MeissnerMaximilian ReichertMichael FlossdorfRoland RadMarc Schmidt-SupprianGünter SchneiderDieter Saur
Published in: Nature cancer (2022)
KRAS-mutant pancreatic ductal adenocarcinoma (PDAC) is highly immunosuppressive and resistant to targeted and immunotherapies. Among the different PDAC subtypes, basal-like mesenchymal PDAC, which is driven by allelic imbalance, increased gene dosage and subsequent high expression levels of oncogenic KRAS, shows the most aggressive phenotype and strongest therapy resistance. In the present study, we performed a systematic high-throughput combination drug screen and identified a synergistic interaction between the MEK inhibitor trametinib and the multi-kinase inhibitor nintedanib, which targets KRAS-directed oncogenic signaling in mesenchymal PDAC. This combination treatment induces cell-cycle arrest and cell death, and initiates a context-dependent remodeling of the immunosuppressive cancer cell secretome. Using a combination of single-cell RNA-sequencing, CRISPR screens and immunophenotyping, we show that this combination therapy promotes intratumor infiltration of cytotoxic and effector T cells, which sensitizes mesenchymal PDAC to PD-L1 immune checkpoint inhibition. Overall, our results open new avenues to target this aggressive and therapy-refractory mesenchymal PDAC subtype.
Keyphrases