Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2021 Sep;70(9):2701-2719.
doi: 10.1007/s00262-021-03001-7. Epub 2021 Jul 9.

Anti-tumor effects of RTX-240: an engineered red blood cell expressing 4-1BB ligand and interleukin-15

Affiliations

Anti-tumor effects of RTX-240: an engineered red blood cell expressing 4-1BB ligand and interleukin-15

Shannon L McArdel et al. Cancer Immunol Immunother. 2021 Sep.

Abstract

Recombinant agonists that activate co-stimulatory and cytokine receptors have shown limited clinical anticancer utility, potentially due to narrow therapeutic windows, the need for coordinated activation of co-stimulatory and cytokine pathways and the failure of agonistic antibodies to recapitulate signaling by endogenous ligands. RTX-240 is a genetically engineered red blood cell expressing 4-1BBL and IL-15/IL-15Rα fusion (IL-15TP). RTX-240 is designed to potently and simultaneously stimulate the 4-1BB and IL-15 pathways, thereby activating and expanding T cells and NK cells, while potentially offering an improved safety profile through restricted biodistribution. We assessed the ability of RTX-240 to expand and activate T cells and NK cells and evaluated the in vivo efficacy, pharmacodynamics and tolerability using murine models. Treatment of PBMCs with RTX-240 induced T cell and NK cell activation and proliferation. In vivo studies using mRBC-240, a mouse surrogate for RTX-240, revealed biodistribution predominantly to the red pulp of the spleen, leading to CD8 + T cell and NK cell expansion. mRBC-240 was efficacious in a B16-F10 melanoma model and led to increased NK cell infiltration into the lungs. mRBC-240 significantly inhibited CT26 tumor growth, in association with an increase in tumor-infiltrating proliferating and cytotoxic CD8 + T cells. mRBC-240 was tolerated and showed no evidence of hepatic injury at the highest feasible dose, compared with a 4-1BB agonistic antibody. RTX-240 promotes T cell and NK cell activity in preclinical models and shows efficacy and an improved safety profile. Based on these data, RTX-240 is now being evaluated in a clinical trial.

Keywords: CD8-Positive T-Lymphocytes; Cell engineering; Immunotherapy; Investigational therapies; Natural killer cells.

PubMed Disclaimer

Figures

Fig. 1
Fig. 1
4-1BBL and IL-15TP on RTX-240 stimulate receptor-specific activation in reporter cells. a Representative flow cytometry plots of RTX-240 stained for 4-1BBL and IL-15TP. b Fold change in NFkB activation in 4-1BB/NFkB reporter HEK293 cells incubated with engineered RBCs or 4-1BB agonistic antibody + cross-linking antibody (10 nM and 1 nM agonist with 25 nM and 2.5 nM cross-linker, respectively) compared with media alone. c Fold change in JAK/STAT activation by IL-15 in reporter HEK-Blue-IL-2 cells incubated with engineered RBCs or recombinant (r) IL-15 (threefold dilutions from 100 pg/mL), compared with media alone. Bars indicate SD of 2–3 technical replicates. IL-15TP, trans-presented interleukin 15; RBC, red blood cell; SD, standard deviation
Fig. 2
Fig. 2
Co-stimulation of human T cells in vitro. PBMCs were labeled with CTFR and incubated with anti-CD3 (0.5 μg/mL) and the indicated treatments for 5 days, then analyzed by flow cytometry for: a CD8 + T cell number, b CD8 + T cell proliferation (percentage of cells that went through at least one division) and Granzyme B expression on CD8 + T cells c. Bars indicate SD of 3 biological replicates. Flow plots are representative data from one donor. CTFR, CellTrace Far Red dye; GzmB, Granzyme B; IL-15TP, trans-presented interleukin 15; PBMC, peripheral blood mononuclear cell; rIL-15, recombinant IL-15; SD, standard deviation
Fig. 3
Fig. 3
Direct activation of CD8 + T cells and NK cells in vitro. (ai) PBMCs were labeled with CTFR and incubated with the indicated treatments for 8 days, then analyzed by flow cytometry for: a memory CD8 + T cell numbers; b memory CD8 + T cell proliferation (percentage of cells that went through at least one division); c CD8 + effector memory differentiation; d NK cell numbers; e NK cell proliferation (percentage of cells that went through at least one division); fi expression of the following molecules on NK cells: f TRAIL, g NKp44, h Granzyme B and i 4-1BB. j Purified NK cells were incubated in vitro overnight with the indicated treatments, then incubated with labeled K562 target cells for 4 h. Target cell killing was measured by flow cytometry. Bars indicate ai SD of 3 biological replicates j or SD of 3 technical replicates. Flow plots are representative data from one donor. CTFR, CellTrace Far Red dye; GzmB, Granzyme B; IL-15TP, trans-presented interleukin-15; NK, natural killer; PBMC, peripheral blood mononuclear cell; rIL-15, recombinant IL-15; SD, standard deviation
Fig. 3
Fig. 3
Direct activation of CD8 + T cells and NK cells in vitro. (ai) PBMCs were labeled with CTFR and incubated with the indicated treatments for 8 days, then analyzed by flow cytometry for: a memory CD8 + T cell numbers; b memory CD8 + T cell proliferation (percentage of cells that went through at least one division); c CD8 + effector memory differentiation; d NK cell numbers; e NK cell proliferation (percentage of cells that went through at least one division); fi expression of the following molecules on NK cells: f TRAIL, g NKp44, h Granzyme B and i 4-1BB. j Purified NK cells were incubated in vitro overnight with the indicated treatments, then incubated with labeled K562 target cells for 4 h. Target cell killing was measured by flow cytometry. Bars indicate ai SD of 3 biological replicates j or SD of 3 technical replicates. Flow plots are representative data from one donor. CTFR, CellTrace Far Red dye; GzmB, Granzyme B; IL-15TP, trans-presented interleukin-15; NK, natural killer; PBMC, peripheral blood mononuclear cell; rIL-15, recombinant IL-15; SD, standard deviation
Fig. 3
Fig. 3
Direct activation of CD8 + T cells and NK cells in vitro. (ai) PBMCs were labeled with CTFR and incubated with the indicated treatments for 8 days, then analyzed by flow cytometry for: a memory CD8 + T cell numbers; b memory CD8 + T cell proliferation (percentage of cells that went through at least one division); c CD8 + effector memory differentiation; d NK cell numbers; e NK cell proliferation (percentage of cells that went through at least one division); fi expression of the following molecules on NK cells: f TRAIL, g NKp44, h Granzyme B and i 4-1BB. j Purified NK cells were incubated in vitro overnight with the indicated treatments, then incubated with labeled K562 target cells for 4 h. Target cell killing was measured by flow cytometry. Bars indicate ai SD of 3 biological replicates j or SD of 3 technical replicates. Flow plots are representative data from one donor. CTFR, CellTrace Far Red dye; GzmB, Granzyme B; IL-15TP, trans-presented interleukin-15; NK, natural killer; PBMC, peripheral blood mononuclear cell; rIL-15, recombinant IL-15; SD, standard deviation
Fig. 4
Fig. 4
mRBC-240 biodistribution, PK and PD. a Representative images of mRBC-240 in different tissues (tumor, spleen, liver, draining lymph nodes, lung, kidney, and heart). mRBC-240 cells were labeled with CTFR prior to injection (red channel). Blood vessels were identified by CD31 staining (green channel) and nucleated cells were detected using Hoechst staining (blue channel). b Representative images of mRBC-240 in the red pulp of the spleen. c mRBC-240 biodistribution: quantification of the density (cells per mm2) of mRBC-240 and mRBC-CTRL in different organs 24 h following a single IV dose (n = 5). Data presented ± SEM **p < 0.01, ***p < 0.001, comparing mRBC-240 versus mRBC-CTRL. All comparisons were analyzed by t test. d PK of mRBC-240: concentration of mRBC-240 in the blood was assessed. Blood was collected 4 h post the first dose and each day thereafter and the percentage of Cy5-labeled mRBC-CTRL (1 × 109) or mRBC-240 (1 × 109 or 1 × 108) in circulation was assessed by flow cytometry (n = 4 mice per group). e PD of mRBC-240 in the spleen of non-tumor-bearing mice: counts of total NK cells, CD8 + T cells and IFNγ + CD8 + T cells were analyzed by flow cytometry in the spleen on day 14 (n = 4 mice per group). f PD of mRBC-240 in the spleen of tumor-bearing mice (B16-F10): frequencies of NK cells and CD8 + T cells were analyzed by flow cytometry in the spleen on day 12 (n = 5 mice per group). Bars indicate SD of biological replicates. Comparisons were analyzed by a one-way ANOVA or unpaired T test and compared with mRBC-CTRL group and showing as **p < 0.01, ***p < 0.001, ****p < 0.0001. CTFR, CellTrace Far Red dye; IFNγ, interferon-γ; IV, intravenous; mRBC, mouse red blood cell; NK, natural killer; PD, pharmacodynamics; PK, pharmacokinetics; SD, standard deviation; SEM, standard error of the mean
Fig. 4
Fig. 4
mRBC-240 biodistribution, PK and PD. a Representative images of mRBC-240 in different tissues (tumor, spleen, liver, draining lymph nodes, lung, kidney, and heart). mRBC-240 cells were labeled with CTFR prior to injection (red channel). Blood vessels were identified by CD31 staining (green channel) and nucleated cells were detected using Hoechst staining (blue channel). b Representative images of mRBC-240 in the red pulp of the spleen. c mRBC-240 biodistribution: quantification of the density (cells per mm2) of mRBC-240 and mRBC-CTRL in different organs 24 h following a single IV dose (n = 5). Data presented ± SEM **p < 0.01, ***p < 0.001, comparing mRBC-240 versus mRBC-CTRL. All comparisons were analyzed by t test. d PK of mRBC-240: concentration of mRBC-240 in the blood was assessed. Blood was collected 4 h post the first dose and each day thereafter and the percentage of Cy5-labeled mRBC-CTRL (1 × 109) or mRBC-240 (1 × 109 or 1 × 108) in circulation was assessed by flow cytometry (n = 4 mice per group). e PD of mRBC-240 in the spleen of non-tumor-bearing mice: counts of total NK cells, CD8 + T cells and IFNγ + CD8 + T cells were analyzed by flow cytometry in the spleen on day 14 (n = 4 mice per group). f PD of mRBC-240 in the spleen of tumor-bearing mice (B16-F10): frequencies of NK cells and CD8 + T cells were analyzed by flow cytometry in the spleen on day 12 (n = 5 mice per group). Bars indicate SD of biological replicates. Comparisons were analyzed by a one-way ANOVA or unpaired T test and compared with mRBC-CTRL group and showing as **p < 0.01, ***p < 0.001, ****p < 0.0001. CTFR, CellTrace Far Red dye; IFNγ, interferon-γ; IV, intravenous; mRBC, mouse red blood cell; NK, natural killer; PD, pharmacodynamics; PK, pharmacokinetics; SD, standard deviation; SEM, standard error of the mean
Fig. 5
Fig. 5
mRBC-240 promotes tumor control in B16-F10 and CT26 models. a Lung metastases enumeration in C57BL/6 mice inoculated IV with B16-F10 tumor cells. Analysis performed on day 14 (n = 8 mice/group). The frequency of b NK cells (NK1.1 +) and c terminally differentiated NK cells (NK1.1 + CD11b + CD27-KLRG1 +) and d CD8 + T cells in the lung were analyzed by flow cytometry. Data presented ± SD and comparisons analyzed by one-way ANOVA *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. e Tumor growth curves for CT26 tumor model in BALB/c mice. Data presented ± SEM ****p < 0.0001, comparisons were analyzed by two-way ANOVA. The numbers of f tumor-infiltrating CD8 + T cells, g proliferating (Ki67 +) CD8 + T cells and h activated (CD44 +) CD8 + T cells as well as i tumor-infiltrating NK cells were analyzed by flow cytometry on day 11 (n = 8 mice per group). Data presented ± SD *p < 0.05. Comparisons were analyzed by unpaired T test. IL-15TP, trans-presented interleukin 15; IV, intravenous; mRBC, mouse red blood cell; NK, natural killer; rIL-15, recombinant IL-15; SD, standard deviation; SEM, standard error of the mean
Fig. 5
Fig. 5
mRBC-240 promotes tumor control in B16-F10 and CT26 models. a Lung metastases enumeration in C57BL/6 mice inoculated IV with B16-F10 tumor cells. Analysis performed on day 14 (n = 8 mice/group). The frequency of b NK cells (NK1.1 +) and c terminally differentiated NK cells (NK1.1 + CD11b + CD27-KLRG1 +) and d CD8 + T cells in the lung were analyzed by flow cytometry. Data presented ± SD and comparisons analyzed by one-way ANOVA *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. e Tumor growth curves for CT26 tumor model in BALB/c mice. Data presented ± SEM ****p < 0.0001, comparisons were analyzed by two-way ANOVA. The numbers of f tumor-infiltrating CD8 + T cells, g proliferating (Ki67 +) CD8 + T cells and h activated (CD44 +) CD8 + T cells as well as i tumor-infiltrating NK cells were analyzed by flow cytometry on day 11 (n = 8 mice per group). Data presented ± SD *p < 0.05. Comparisons were analyzed by unpaired T test. IL-15TP, trans-presented interleukin 15; IV, intravenous; mRBC, mouse red blood cell; NK, natural killer; rIL-15, recombinant IL-15; SD, standard deviation; SEM, standard error of the mean
Fig. 6
Fig. 6
mRBC-240 is tolerated in mice. a Quantification of liver macrophages (F4/80 +). b Representative images of IHC staining of macrophages (F4/80) from selected mice. c Quantification of liver-infiltrating CD8 + T cells and d liver-infiltrating cytotoxic CD8 + T cells (CD8 + /Eomes + /KLRG1 +). Quantification was performed on liver samples of C57BL/6 wild-type mice by flow cytometry on day 18 following 4 doses of either mRBC-240 (1 × 109, 3 × 108, or 1 × 108), mRBC-CTRL, 4-1BB agonistic antibody (10 mg/kg or 2.5 mg/kg) (n = 8 mice/group) or PBS. e ALT liver enzyme levels (U/L) in serum on day 18. f Inflammation scoring performed on H&E stained liver sections. g Representative images of H&E staining of liver sections from selected mice. All comparisons were analyzed by a one-way ANOVA and compared with control groups and showing as *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001. ALT, alanine aminotransferase; IHC, immunohistochemistry; H&E, hematoxylin and eosin; mRBC, mouse red blood cell; PBS, phosphate-buffered saline

References

    1. Darvin P, Toor SM, Nair VS, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;50:1–11. doi: 10.1038/s12276-018-0191-1. - DOI - PMC - PubMed
    1. Chavez JC, Bachmeier C, Kharfan-Dabaja MA. CAR T-cell therapy for B-cell lymphomas: clinical trial results of available products. Ther Adv Hematol. 2019;10:1–20. doi: 10.1177/2040620719841581. - DOI - PMC - PubMed
    1. Alexander W. The Checkpoint immunotherapy revolution. Pharm Ther. 2016;43:185–191. - PMC - PubMed
    1. Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ. Toxicity and management in CAR T-cell therapy. Mol Ther. 2016;3:16011. doi: 10.1038/mto.2016.11. - DOI - PMC - PubMed
    1. Li Y, Sun R (2018) Tumor immunotherapy: New aspects of natural killer cells. Chin J Cancer Res 30:173–196. 10.21147/j.issn.1000-9604.2018.02.02 - PMC - PubMed

MeSH terms