Briere et al. evaluated the impact of the KRASG12C inhibitor MRTX849 on immune signaling and response to ICB. In immunocompromised xenograft models, MRTX849 decreased tumor-intrinsic expression of immunosuppressive factors and increased MHC class I proteins. In KRASG12C syngeneic and GEM models, MRTX849 increased intratumoral M1 macrophages, dendritic cells, CD4+, and CD8+ T cells, and decreased MDSCs, thus reversing the immunosuppressive microenvironment. MRTX849 plus anti-PD-1 therapy led to durable tumor regression and protection against rechallenge in a syngeneic model and prolonged survival in KRASG12C Msh2fl/fl lung GEM.

Contributed by Shishir Pant

ABSTRACT: KRASG12C inhibitors, including MRTX849, are promising treatment options for KRAS-mutant non-small cell lung cancer (NSCLC). PD-1 inhibitors are approved in NSCLC; however, strategies to enhance checkpoint inhibitor therapy (CIT) are needed. KRASG12C mutations are smoking-associated transversion mutations associated with high tumor mutation burden (TMB), PD-L1 positivity and an immunosuppressive tumor microenvironment. To evaluate the potential of MRTX849 to augment CIT, its impact on immune signaling and response to CIT was evaluated. In human tumor xenograft models, MRTX849 increased MHC class I protein expression and decreased RNA and/or plasma protein levels of immunosuppressive factors. In a KRASG12C-mutant CT26 syngeneic mouse model, MRTX849 decreased intratumoral myeloid-derived suppressor cells (MDSCs) and increased M1-polarized macrophages, dendritic cells, CD4+ and CD8+ T cells. Similar results were observed in lung KrasG12C-mutant syngeneic and a genetically engineered mouse (GEM) model. In the CT26 KrasG12C model, MRTX849 demonstrated marked tumor regression when tumors were established in immune-competent BALB/c mice; however, the effect was diminished when tumors were grown in T-cell deficient nu/nu mice. Tumors progressed following anti-PD-1 or MRTX849 single agent treatment in immune-competent mice; however, combination treatment demonstrated durable, complete responses (CRs). Tumors did not re-establish in the same mice that exhibited durable CRs when re-challenged with tumor cell inoculum, demonstrating these mice developed adaptive anti-tumor immunity. In a GEM model, treatment with MRTX849 plus anti-PD-1 led to increased progression-free survival compared to either single agent alone. These data demonstrate KRAS inhibition reverses an immunosuppressive tumor microenvironment and sensitizes tumors to CIT through multiple mechanisms.

Author Info: (1) Research, Mirati Therapeutics (United States). (2) cancer center, New York University School of Medicine. (3) Research, Mirati Therapeutics (United States). (4) Research - Biol

Author Info: (1) Research, Mirati Therapeutics (United States). (2) cancer center, New York University School of Medicine. (3) Research, Mirati Therapeutics (United States). (4) Research - Biology, Mirati Therapeutics (United States). (5) Research, Mirati Therapeutics (United States). (6) Research, Mirati Therapeutics (United States). (7) The University of Texas MD Anderson Cancer Center. (8) Perlmutter Cancer Center, New York University Langone Medical Center. (9) Research, Mirati Therapeutics (United States). (10) Research, Mirati Therapeutics (United States). (11) Bioinformatics, Monoceros biosystems. (12) Core, Monoceros Biosystems LLC. (13) Bioinformatics, Monoceros Biosystems LLC. (14) Perlmutter Cancer Center, New York University Langone Medical Center. (15) Research, Mirati Therapeutics (United States). (16) Mirati Therapeutics, Inc. olsonp@mirati.com.