Abstract 4020: Inhibition of KITin vivomodifies immune cell populations to improve the efficacy of checkpoint inhibitors in syngeneic mouse tumor models

Author(s):  
Andrew J. Garton ◽  
Lori Lopresti-Morrow ◽  
Scott Seibel ◽  
Theresa LaVallee ◽  
Richard Gedrich
2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A747-A747
Author(s):  
Andrew MacKinnon ◽  
Deepthi Bhupathi ◽  
Jason Chen ◽  
Tony Huang ◽  
Weiqun Li ◽  
...  

BackgroundTumors evade destruction by the immune system through multiple mechanisms including altering metabolism in the tumor microenvironment. Metabolic control of immune responses occurs through depletion of essential nutrients or accumulation of toxic metabolites that impair immune cell function and promote tumor growth. The secreted enzyme interleukin 4 (IL-4)-induced gene 1 (IL4I1) is an L-phenylalanine oxidase that catabolizes phenylalanine and produces phenyl-pyruvate and hydrogen peroxide. IL4I1 regulates several aspects of adaptive immunity in mice, including inhibition of cytotoxic T cells through its production of hydrogen peroxide (reviewed in1). In human tumors, IL4I1 expression is significantly elevated relative to normal tissues and is notably high in ovarian tumors and B cell lymphomas. Motivated by the hypothesis that IL4I1 is an immuno-metabolic enzyme that suppresses anti-tumor immunity, we discovered CB-668, the first known small-molecule inhibitor of IL4I1.MethodsIL4I1 enzymatic activity was measured using an HRP-coupled enzyme assay. RNA in-situ hybridization was carried out on the RNAScope platform. Syngeneic mouse tumor models were used to evaluate the anti-tumor activity of CB-668. The level of phenyl-pyruvate in tumor homogenates was measured by LC/MS.ResultsOur clinical candidate, CB-668 is a potent and selective non-competitive inhibitor of IL4I1 (IC50 = 15 nM). CB-668 has favorable in vitro ADME properties and showed low clearance and high oral bioavailability in rodents. Twice-daily oral administration of CB-668 was well-tolerated in mice and resulted in single-agent anti-tumor activity in the syngeneic mouse tumor models B16-F10, A20, and EG7. Oral CB-668 administration reduced the levels of phenyl-pyruvate in the tumor, consistent with inhibition of IL4I1 enzymatic activity. Anti-tumor activity of CB-668 was immune cell-mediated since efficacy was abrogated in CD8-depleted mice, and CB-668 treatment caused increased expression of pro-inflammatory immune genes in the tumor. Moreover, CB-668 had no direct anti-proliferative activity on tumor cells grown in vitro (IC50 > 50 µM). CB-668 also favorably combined with anti-PD-L1 therapy to reduce tumor growth in the B16-F10 tumor model.ConclusionsThese data support an immune-mediated anti-tumor effect of IL4I1 inhibition by CB-668, and suggest inhibition of IL4I1 represents a novel strategy for cancer immuno-therapy.ReferencesMolinier-Frenkel V, Prévost-Blondel A, and Castellano F. The IL4I1 Enzyme: A New Player in the Immunosuppressive Tumor Microenvironment. Cells 2019;8:1–9.


2016 ◽  
Author(s):  
Rafael Cubas ◽  
Marina Moskalenko ◽  
Jeanne Cheung ◽  
Shiuh-Ming Luoh ◽  
Erin McNamara ◽  
...  

2019 ◽  
Vol 11 (494) ◽  
pp. eaax9556
Author(s):  
Patrick M. Brunner

In mouse tumor models, the CXCR3-CXCL9 chemokine system mediates sensitivity to PD-1 blockade and could be exploited to enhance responsiveness to checkpoint inhibitors.


2018 ◽  
Author(s):  
Matthew J. Robinson ◽  
Ines Osma-Garcia ◽  
Jane Coates-Ulrichisen ◽  
Amanda Watkins ◽  
Suzanne Mosely ◽  
...  

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A251-A251
Author(s):  
Dominic Pearce ◽  
Daniel Halligan ◽  
Patrick Fadden ◽  
Chassidy Hall ◽  
Amber Blackwell ◽  
...  

BackgroundExperimental therapies that target the immune system have expanded greatly in recent years due to the success of immune checkpoint inhibitory antibodies such as ipilimumab and pembrolizumab. Preclinical development of these novel immune-oncology drugs requires the availability of well characterized mouse models to evaluate therapeutic mode of action, efficacy, and safety. Syngeneic mouse tumor models provide robust systems in which to evaluate novel immune-oncology therapies. Efficacy in these models can be measured by tumor volume changes in subcutaneous implants or by impacts on survival for orthotopic implants. Mode of action can be assessed by identifying changes in the tumor microenvironment following dosing. Multiple analytical methods can be used to track changes in immune populations and activation status from flow cytometry to immunohistochemistry to gene expression analysis.MethodsWe endeavored to characterize the functional tumor microenvironment changes for two syngeneic models following treatment with anti-PD-1 and anti-CTLA-4 antibodies The syngeneic models used for the study were both colon adenocarcinomas, MC38 and CT26. Mice bearing subcutaneous tumors were dosed intraperitoneally with either vehicle alone, anti-CTLA-4, anti-PD-1, or a combination of the two immune checkpoint inhibitors on days 1, 4, and 8. Tumors were harvested on day 9 and assessed for gene expression by microarray analysis. The gene expression results were evaluated for the relationship between treatment regimen and tumor volume change by expression level association, functional set enrichment analysis, and immune cell population gene set variation analysis.ResultsFor each of the tumor models, >10,000 genes were found to be significantly differentially expressed. Functional set enrichment analysis showed notable changes in cell cycle and mitotic markers as well as immune response markers in MC38. In contrast, CT26 showed principally changes in immune response markers. Immune cell population set analysis revealed differential impacts on numerous immune cell populations between the models which correlate with therapy induced changes in tumor growth. These include expected changes in CD8+ T-cell populations for both models but also differential changes in other populations including CD56dim NK cells, eosinophils and B cells in MC38 and neutrophils in CT26. We also conducted a genomic analysis by whole exome sequencing. Both tumor models have relatively high tumor mutational burden; CT26 TMB = 377 MB and MC38 TMB = 69/MB. These data show the value of robust bioinformatics analysis of gene expression data sets to provide insights into the mode of action and model responses to investigational immune-oncology drugs.ConclusionsN/A


Sign in / Sign up

Export Citation Format

Share Document