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
. 2024 Nov 20;12(11):e009743.
doi: 10.1136/jitc-2024-009743.

IL-18R supported CAR T cells targeting oncofetal tenascin C for the immunotherapy of pediatric sarcoma and brain tumors

Affiliations

IL-18R supported CAR T cells targeting oncofetal tenascin C for the immunotherapy of pediatric sarcoma and brain tumors

Elizabeth Wickman et al. J Immunother Cancer. .

Abstract

Background: Oncofetal splice variants of extracellular matrix (ECM) proteins present a unique group of target antigens for the immunotherapy of pediatric cancers. However, limited data is available if these splice variants can be targeted with T cells expressing chimeric antigen receptors (CARs).

Methods: To determine the expression of the oncofetal version of tenascin C (TNC) encoding the C domain (C.TNC) in pediatric brain and solid tumors, we used quantitative reverse transcription PCR and immunohistochemistry. Genetically modified T cells were generated from human peripheral blood mononuclear cells and evaluated in vitro and in vivo.

Results: We demonstrate that C.TNC is expressed on a protein level in pediatric tumors, including diffuse intrinsic pontine glioma, osteosarcoma, rhabdomyosarcoma, and Ewing sarcoma. We generate C.TNC-CAR T cells and establish that these recognize and kill C.TNC-positive tumor cells. However, their antitumor activity in vivo is limited. To improve the effector function of C.TNC-CAR T cells, we design a leucine zipper-based chimeric cytokine receptor that activates interleukin-18 signaling pathways (Zip18R). Expression of Zip18R in C.TNC-CAR T cells improves their ability to secrete cytokines and expand in repeat stimulation assays. C.TNC-CAR.Zip18R T cells also have significantly greater antitumor activity in vivo compared with unmodified C.TNC-CAR T cells.

Conclusions: Our study identifies the C domain of the ECM protein TNC as a promising CAR T-cell therapy for pediatric solid tumors and brain tumors. While we focus here on pediatric cancer, our work has relevance to a broad range of adult cancers that express C.TNC.

Keywords: Chimeric antigen receptor - CAR; Immunotherapy; T cell.

PubMed Disclaimer

Conflict of interest statement

Competing interests: EW, SL, JW, TIS, GK, JZ, and SG have patent applications in the fields of cell or gene therapy for cancer. MB, GK, and SG are coinventors on a patent application for the developed Zip receptor technology. SG is a member of the Scientific Advisory Board of Be Biopharma and CARGO, and the Data and Safety Monitoring Board (DSMB) of Immatics and has received honoraria from TESSA Therapeutics within the last year. The other authors declare no competing interests.

Figures

Figure 1
Figure 1. C.TNC is expressed in solid and brain tumors. (A,B) Reverse transcription quantitative PCR of (A) pediatric cell lines (n=3 bioreplicates, mean+SEM) and (B) patient-derived xenografts for C.TNC. ΔCT is relative to GAPDH. Acute lymphoblastic leukemia cell line CCRF-CEM had no C.TNC detected. (C) H-scores of primary human H3K27M+DIPG, ZFTA fusion-positive ependymoma (EPN), osteosarcoma (OS), rhabdomyosarcoma (RMS), and Ewing sarcoma (EWS) tumors. (D) Representative immunohistochemistry staining of primary FFPE human tumor samples and tonsils (negative control (neg Co)) with a C.TNC-specific antibody. 20× magnifications, 100 µM scale bar. Images were taken with an Olympus BX46 microscope and a Nikon DS-Fi3 camera at a 20× magnification and edited with Photoshop 25.5.1. C.TNC, tenascin C encoding the C domain; DIPG, diffuse intrinsic pontine glioma.
Figure 2
Figure 2. C.TNC-CAR T cells have antitumor activity in vitro and in vivo. (A) Schematic of C.TNC-CAR design. (B) Transduction efficiency of healthy donor T cells determined via flow cytometry on day 7 (n=5, mean+SEM). Graph shows the percentage of positive cells stained for each respective antibody for CAR transgene detection. (C,D) IFN-γ production measured by ELISA after 48 hours of co-culture with (C) sarcoma or (D) brain tumor cell lines at 2:1 effector to target (E:T) ratio. Negative values were plotted as zero (mean+SEM, n=3–4 for NT and C.TNC), two-way ANOVA, ***p<0.001, ****p<0.0001. (E,F) Cytotoxicity after 72 hours at a 4:1 E:T ratio determined by a luciferase-based assay (mean+SEM, n=3), two-way ANOVA, ****p<0.0001. (G) Cytotoxicity of C.TNC-negative cell line CCRF-CEM determined by a luciferase-based assay (n=4–5, mean+SEM). (H) Schematic of experiment in 5–6-week-old female NSG mice. 1×106 LM7.green fluorescent protein.firefly luciferase cells were injected intraperitoneally (i.p.), followed by 1×106 T cells injected i.p. 7 days later. (I) Flux values from weekly IVIS images (n=5 per cohort). (J) Overall survival of the mice, Mantel-Cox test, **p<0.01. (K) Schematic of experiment in 10–12-week-old male NSG mice. 1×106 DIPG007.YFP.ffLuc cells were injected intracranially (i.c.), followed by 2×106 T cells injected i.c. 7 days later. (L) Flux values from weekly IVIS images (n=5 per cohort). (M) Overall survival of the mice, Mantel-Cox test. ANOVA, analysis of variance; CAR, chimeric antigen receptor; C.TNC, tenascin C encoding the C domain; DIPG, diffuse intrinsic pontine glioma; IFN, interferon; NT, non-transduced; scFv, single chain variable fragment.
Figure 3
Figure 3. Zip18R improves CAR T-cell effector function. (A) Schematic of 1X and 2X Zip18Rs, the leucine zippers (Zip) are connected with a (G4S)3 linker. The 1X and 2X Zip18Rs retroviral vectors also encode a P2A skip sequence and mClover. TM/IC: transmembrane and intracellular. (B) Representative transduction efficiency of Ramos-Blue and Ramos-Blue KD-MyD NF-κB/AP-1 reporter cells determined via flow cytometry. (C) Absorbance values of transduced Ramos-Blue NF-κB/AP-1 reporter cell supernatant mixed with QUANTI-Blue reagent (n=3 from two separate transductions). (D) Transduction of T cells with Zip18R (n=3, mean+SEM). (E,F) Populations after (E) 7 days or (F) 14 days of cytokine starvation as determined by flow cytometry (n=3, mean+SEM). Two-way analysis of variance, *p<0.05, **p<0.01, ***p<0.001. (G) Schematic of A673 model. T cells were sorted prior to injection. 7–9-week-old male NSG mice were used. (H) Tumor caliper measurements (n=4 for tumor and CAR only, n=5 for CAR.Zip18Rs). (I) Kaplan-Meier survival curve, Mantel-Cox test, **p<0.01. (J) Tumor caliper measurements. (n=3 for tumor and CAR.2XZip18R, n=4 for CAR and CAR.1XZip18R). (K) Kaplan-Meier survival curve, Mantel-Cox test, *p<0.05. CAR, chimeric antigen receptor; NT, non-transduced; Zip18R, interleukin-18 receptor-based leucine zipper receptor.
Figure 4
Figure 4. Zip18R bolsters C.TNC-CAR T-cell effector function in vitro. (A) Transduction efficiency of primary T cells was determined via flow cytometry on day 7 post-transduction (n=4, mean+SEM). Determined via F(ab’)2 staining and mClover expression. (B) Repeat stimulation assay schematic. T cells were stimulated with LM7.green fluorescent protein.firefly luciferase at an effector to target ratio of 2:1 in the presence of IL-15 every 4 days. (C) Expansion of T cells in repeat stimulation assay. Each graph represents one donor. (D) Fold change of C.TNC-CAR and C.TNC.CAR.Zip18R T cells after stimulations 1 through 10, paired t-test, ****p<0.0001. (E–G) Quantification of (E) type 1 and (F) type 2/type 17 cytokines, and (G) chemokines 48 hours post first stimulation of the repeat stimulation assay (n=3, mean+SEM), data was log-transformed before statistical analysis, two-way analysis of variance, *p<0.05, **p<0.01, ***:p<0.001, ****p<0.0001. (H–J) Comparison of the sum of (H) type 1 cytokines, (I) type 2/type 17 cytokines, and (J) chemokines produced post first stimulation and fourth stimulation by C.TNC-CAR T cells and C.TNC-CAR.Zip18R T cells from repeat stimulation assay (n=3), unpaired t-test, *p<0.05, **p<0.01. CAR, chimeric antigen receptor; C.TNC, tenascin C encoding the C domain; GM-CSF, granulocyte-macrophage colony-stimulating factor; IFN, interferon; IL, interleukin; NT, non-transduced; TNF, tumor necrosis factor; Zip18R, interleukin-18 receptor-based leucine zipper receptor.
Figure 5
Figure 5. Zip18R signaling alters the transcriptome of C.TNC-CAR T cells. (A) Schematic of experiment. C.TNC-CAR.Zip18R T cells were collected from culture in IL-7/IL-15 and frozen for analysis or freshly collected from a co-culture assay with LM7.GFP.ffLuc (LM7.GL) tumor cells in the presence of IL-15 at 12, 24, and 48 hours post tumor cell stimulation. All four populations are present in the same culture. (B) Percentages of C.TNC-CAR+Zip18R+, C.TNC-CAR+, Zip18R+, and NT T cells at 12, 24, and 48 hours post co-culture. (C,D) Gene Set Enrichment Analysis comparing C.TNC-CAR.Zip18R T cells to C.TNC-CAR T cells at (C) baseline or (D) 48 hours post stimulation with LM7.GFP.ffLuc cells. Top 10 activated or suppressed significant (p.adjust<0.1) KEGG pathways are shown. (E–I) 1×106 NT, Zip18R, C.TNC-CAR, and C.TNC-CAR.Zip18R T cells were cultured in media or against LM7 cells at a 2:1 effector to target ratio for 48 hours and then collected for flow cytometric analysis. (E) Transduction (TDX) percentage of NT, Zip18R, C.TNC-CAR, and C.TNC-CAR.Zip18R T cells alone and after 48 hours of stimulation. CD3+ expression is shown for NT samples. (n=3, mean+SEM), two-way ANOVA, ****p<0.0001. (F–I) Expression within pure isolated populations of T cells gated on CD3+ (NT), mClover+ (Zip18R+), (G4S)3+ (C.TNC-CAR+), and (G4S)3+ mClover+ (C.TNC-CAR+Zip18R+) for (F) CD69, (G) CD28, (H) CD39, and (I) TIM3 (n=3, mean+SEM), two-way ANOVA, *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. Only showing significance values for each cohort comparing Alone versus +LM7 and C.TNC-CAR versus C.TNC-CAR.Zip18R for both conditions. All p values are reported in online supplemental SFigure 16. ANOVA, analysis of variance; CAR, chimeric antigen receptor; C.TNC, tenascin C encoding the C domain; ffLuc, firefly luciferase; GFP, green fluorescent protein; GM-CSF, granulocyte-macrophage colony-stimulating factor; IL, interleukin; KEGG, Kyoto Encylopedia of Genes and Genomes; NT, non-transduced; TNF, tumor necrosis factor; Zip18R, interleukin-18 receptor-based leucine zipper receptor.
Figure 6
Figure 6. C.TNC-CAR.Zip18R T cells have superior antitumor activity in vivo. 1×106 LM7.GFP.ffLuc cells were injected into 5–6-week-old female (Donor 1) or male (Donor 2) NSG mice. Seven days later, mice received an i.p. injection of 1×106 sorted T cells. (A,B) Tumor flux values measured by IVIS imaging. (A) Donor 1, (B) Donor 2 (n=5 per cohort per donor). (C) Combined tumor flux values from both donors at weeks 2 and 5 (n=10, mean+SEM), one-way analysis of variance, *p<0.05, **p<0.01, ***p<0.001, ****p<0.0001. (D) Kaplan-Meier survival curve, Mantel-Cox test. P values listed in the table. (E) Schematic of rechallenge experiment. 5–6-week-old male mice were used for tumor only cohort. 1×106 LM7.GFP.ffLuc cells were injected i.p. into naïve mice or C.TNC-CAR.Zip18R T-cell pretreated mice. (F) Tumor flux values measured by IVIS imaging (n=3–5). CAR, chimeric antigen receptor; C.TNC, tenascin C encoding the C domain; ffLuc, firefly luciferase; GFP, green fluorescent protein; i.p., intraperitoneal; NT, non-transduced; Zip18R, interleukin-18 receptor-based leucine zipper receptor.

References

    1. Globerson Levin A, Rivière I, Eshhar Z, et al. CAR T cells: Building on the CD19 paradigm. Eur J Immunol. 2021;51:2151–63. doi: 10.1002/eji.202049064. - DOI - PMC - PubMed
    1. Wagner J, Wickman E, DeRenzo C, et al. CAR T Cell Therapy for Solid Tumors: Bright Future or Dark Reality? Mol Ther. 2020;28:2320–39. doi: 10.1016/j.ymthe.2020.09.015. - DOI - PMC - PubMed
    1. Labanieh L, Mackall CL. CAR immune cells: design principles, resistance and the next generation. Nat New Biol. 2023;614:635–48. doi: 10.1038/s41586-023-05707-3. - DOI - PubMed
    1. Martín-Otal C, Lasarte-Cia A, Serrano D, et al. Targeting the extra domain A of fibronectin for cancer therapy with CAR-T cells. J Immunother Cancer. 2022;10:e004479. doi: 10.1136/jitc-2021-004479. - DOI - PMC - PubMed
    1. Xie YJ, Dougan M, Jailkhani N, et al. Nanobody-based CAR T cells that target the tumor microenvironment inhibit the growth of solid tumors in immunocompetent mice. Proc Natl Acad Sci U S A. 2019;116:7624–31. doi: 10.1073/pnas.1817147116. - DOI - PMC - PubMed

LinkOut - more resources