Abstract

KRAS (KRAS proto-oncogene, GTPase) inhibitors perform less well than other targeted drugs in vitro and fail clinical trials. To investigate a possible reason for this, we treated human and murine tumor cells with KRAS inhibitors deltarasin (targeting phosphodiesterase-δ), cysmethynil (targeting isoprenylcysteine carboxylmethyltransferase), and AA12 (targeting KRASG12C), and silenced/overexpressed mutant KRAS using custom-designed vectors. We showed that KRAS-mutant tumor cells exclusively respond to KRAS blockade in vivo, because the oncogene co-opts host myeloid cells via a C-C-motif chemokine ligand 2 (CCL2)/interleukin-1 beta (IL-1β)-mediated signaling loop for sustained tumorigenicity. Indeed, KRAS-mutant tumors did not respond to deltarasin in C-C motif chemokine receptor 2 (Ccr2) and Il1b gene-deficient mice, but were deltarasin-sensitive in wild-type and Ccr2-deficient mice adoptively transplanted with wild-type murine bone marrow. A KRAS-dependent pro-inflammatory transcriptome was prominent in human cancers with high KRAS mutation prevalence and poor predicted survival. Our findings support that in vitro cellular systems are suboptimal for anti-KRAS drug screens, as these drugs function to suppress interleukin-1 receptor 1 (IL1R1) expression and myeloid IL-1β-delivered pro-growth effects in vivo. Moreover, the findings support that IL-1β blockade might be suitable for therapy for KRAS-mutant cancers.

Highlights

  • Since its discovery, the Kirsten rat sarcoma virus (KRAS) proto-oncogene GTPase has become the holy grail of anticancer therapy [1,2]

  • Using transcriptome analyses of cell lines expressing endogenous or exogenous wild-type or mutant Kras alleles, Ccr2 (C-C motif chemokine receptor 2) and Il1b gene-deficient mice, as well as adoptive bone marrow transfer, we show that mutant KRAS established a proinflammatory C-motif chemokine ligand 2 (CCL2) (C-C motif chemokine ligand 2)/IL-1β-mediated signaling loop to host myeloid cells in vivo, which is required for KRAS-mediated tumorigenicity and, importantly, for specific KRAS inhibitor efficacy

  • We initially investigated the cellular responses of a battery of human and murine cell lines with known KRAS/Kras (KRAS proto-oncogene, GTPase) mutation status [4,15–17]

Read more

Summary

Introduction

The Kirsten rat sarcoma virus (KRAS) proto-oncogene GTPase (encoded by the human KRAS and the murine Kras genes) has become the holy grail of anticancer therapy [1,2]. Full KRAS GTPase activity and downstream signaling prerequires its integration into the cell membrane, which is facilitated by the post-translational lipidation and membrane transport of KRAS by various enzymes, such as farnesyltransferase (FT), geranylgeranytransferase (GGT), isoprenylcysteine carboxyl methyltransferase (ICMT), phosphodiesterase-δ (PDEδ), and others [3,5] To this end, therapeutic attempts to inhibit KRAS lipidation by targeting FT/GGT/ICMT were recently coupled with the development of PDEδ blockers and of allosteric and covalent inhibitors of mutated KRASG12C [6–9]. In addition to molecular structural considerations [5], the mode of KRAS oncogenic functions could be a reason for this To this end, Janes and collaborators recently reported a discordance between the in vitro and the in vivo effects of a newly developed covalent KRASG12C inhibitor [9]. Our data show that intact inflammatory tumor-to-host interactions were required for full KRAS inhibitor efficacy and imply that in vitro drug screens might not be optimal for KRAS inhibitor discovery

Cell Culture
Cellular Assays
Western Immunoblotting
Constructs
In Vivo Tumor Models and Drug Treatments
Microarrays, PCR, GSEA, and Kaplan-Meier Analyses
2.10. Immunofluorescence
2.11. Bone marrow replacement
2.12. Statistics
Mutation-Independent Effects of KRAS Inhibitors In Vitro
Specific In Vivo Effects of Deltarasin against KRAS-Mutant Tumors
Genetic KRAS Manipulation Reveals Contrasting KRAS-Dependencies In Vitro and In Vivo
Genetic
Summary oflines averaged deltarasin
A Mutant Kras Transcriptome Signature Contains Ccl2 and Il1b
CCR2+ IL-1β-Secreting Myeloid Cells Potentiate In Vivo KRAS-Dependence
Deltarasin Limits IL-1β Sensing by KRAS-Mutant Tumor
Discussion
Conclusions
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call