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
. 2023 Aug 7;14(1):4737.
doi: 10.1038/s41467-023-40115-1.

Antigen-dependent IL-12 signaling in CAR T cells promotes regional to systemic disease targeting

Affiliations

Antigen-dependent IL-12 signaling in CAR T cells promotes regional to systemic disease targeting

Eric Hee Jun Lee et al. Nat Commun. .

Abstract

Chimeric antigen receptor (CAR) T cell therapeutic responses are hampered by limited T cell trafficking, persistence, and durable anti-tumor activity in solid tumors. However, these challenges can be largely overcome by relatively unconstrained synthetic engineering strategies. Here, we describe CAR T cells targeting tumor-associated glycoprotein-72 (TAG72), utilizing the CD28 transmembrane domain upstream of the 4-1BB co-stimulatory domain as a driver of potent anti-tumor activity and IFNγ secretion. CAR T cell-mediated IFNγ production facilitated by IL-12 signaling is required for tumor cell killing, which is recapitulated by engineering an optimized membrane-bound IL-12 (mbIL12) molecule in CAR T cells. These T cells show improved antigen-dependent T cell proliferation and recursive tumor cell killing in vitro, with robust in vivo efficacy in human ovarian cancer xenograft models. Locoregional administration of mbIL12-engineered CAR T cells promotes durable anti-tumor responses against both regional and systemic disease in mice. Safety and efficacy of mbIL12-engineered CAR T cells is demonstrated using an immunocompetent mouse model, with beneficial effects on the immunosuppressive tumor microenvironment. Collectively, our study features a clinically-applicable strategy to improve the efficacy of locoregionally-delivered CAR T cells engineered with antigen-dependent immune-modulating cytokines in targeting regional and systemic disease.

PubMed Disclaimer

Conflict of interest statement

S.J.P. and S.J.F. are scientific advisors to and receive royalties from Mustang Bio. S.J.P. is also a scientific advisor and/or receives royalties from Imugene Ltd, Bayer, Adicet Bio, and Celularity. S.J.P., E.H.L., J.P.M., and S.J.F. are listed as co-inventors on a patent on the development of TAG72-targeted CAR-modified T cells for the treatment of TAG72-positive tumors, and S.J.P., E.H.L., and J.P.M. are listed as co-inventors on a patent on the development of membrane-bound IL12 engineered CAR T cells for the treatment of cancer, which are owned by the City of Hope. All other authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1. CD28 transmembrane in CARs containing a 4-1BB costimulatory domain enhances anti-tumor activity in vitro and in vivo.
a Illustration of a TAG72-CAR T-cell containing the humanized scFv targeting TAG72 with varying five extracellular spacer domains (EQ, dCH2, CD8h, HL, L), three transmembrane domains (CD4tm, CD8tm, CD28tm), and two intracellular costimulatory domains (4-1BB, CD28) followed by a cytolytic domain (CD3z). b Untransduced (UTD) and seven different TAG72-CAR T cells positively enriched for CD19t were evaluated by flow cytometry for Protein L to detect the scFv. c, d In vitro tumor cell killing activity relative to UTD (c) and IFNγ production by ELISA (d), of CAR T cells against tumor targets (TAG72- OVCAR8; TAG72+ OVCAR3, OV90, and OVCAR8-sTn) after 24 hr (for ELISA) or 72 hr of co-culture at an effector:target (E:T) ratio of 1:4. n = 9 from three independent experiments. Data are presented as mean values ±SEM. P values indicate differences between TAG72-dCH2(28tm)28z and TAG72-dCH2(28tm)BBz using a two-tailed Student’s t test. e Western blotting analysis of early downstream signaling mediators following CAR T-cell stimulation of indicated TAG72-CAR T cells. f Quantification of band density of phosphoprotein over their respective total protein levels. n = 2 per timepoint, representative of two independent experiments. Data are presented as mean values ±SD. g TAG72-CAR T-cell killing of OV90 cells measured by xCELLigence over 10 days (E:T = 1:20). h Schema of repetitive tumor cell challenge assay (top). TAG72-CAR T cells were co-cultured with OV90 cells (E:T = 1:2) and rechallenged with OV90 cells every two days. Remaining viable tumor cells and fold change in TAG72-CAR T cells were quantified as described in Methods prior to each tumor cell rechallenge. n = 6–9/group from at least two independent experiments. Data are presented as mean values ±SEM. P values indicate differences between 28tm28z and 28tmBBz using a two-tailed Student’s t test. i Representative bioluminescent flux imaging of intraperitoneal (i.p.) OVCAR3(eGFP/ffluc) tumor-bearing female NSG mice treated i.p. with UTD or indicated TAG72-CAR T cells. j Quantification of flux (individual mice in each group) from treated OVCAR3 tumor-bearing mice. UTD (n = 8/group); TAG72-CAR T cells (n = 10/group). Curative responses: UTD: 0/10, 4tmBBz: 0/10, 28tm28z: 0/10, 28tmBBz: 4/10. P < 0.005 comparing 28tm28z and 28tmBBz using a Multiple Mann–Whitney test.
Fig. 2
Fig. 2. Membrane-bound IL-12 engineered TAG72-CAR T cells induce higher IFNγ, T-cell expansion, and anti-tumor activity in vitro.
ac Tumor cell killing of OV90 cells by TAG72-CAR T cells (E:T = 1:20) with the addition of varying concentrations of anti-IFNγR1 blocking antibody, isotype control, and recombinant human IL-12 cytokine measured by xCELLigence over 10 days (a, b). n = 2/group at each timepoint. Data are presented as mean values ±SD. At day 10, IFNγ levels in supernatants were quantified by ELISA (c). n = 2/group, representative of two independent experiments. Data are presented as mean values ±SD. d Illustration of TAG72-CAR/mbIL12 T cell. e Flow cytometric analysis of surface or intracellular expression of mbIL12 in TAG72-CAR T cells stimulated with varying concentrations of plate-bound TAG72. n = 2/group, representative of two independent experiments. Data are presented as mean values ±SD. f Intracellular flow cytometric analysis of phosphorylated STAT3 (pSTAT3, pY705) (left) and pSTAT4 (right) in TAG72-CAR and TAG72-CAR/mbIL12 T cells stimulated with varying concentrations of plate-bound TAG72 or recombinant huIL12 (10 ng/mL). g Intracellular flow cytometric analysis of pSTAT4 in TAG72-CAR T cells co-cultured with HT1080 (TAG72−) cells transduced with mbIL12. Cells were stimulated with Immunocult CD3/CD28 per manufacturer’s recommendation. Cells were gated on CAR T cells and evaluated for pSTAT4. h TAG72-CAR/mbIL12 T cells were co-cultured with OV90 cells (E:T = 1:3) and rechallenged with OV90 cells every 2 days. The remaining viable tumor cells and TAG72-CAR T-cell proliferation were quantified as described in Methods prior to each tumor cell rechallenge. n = 3/group, representative of two independent experiments. Data are presented as mean values ±SD. P values indicate differences between TAG72-CAR and TAG72-CAR/mbIL12 using a two-tailed Student’s t test.
Fig. 3
Fig. 3. Locoregional intraperitoneal delivery of TAG72-CAR/mbIL12 T cells reduces tumor burden and increases regional and systemic CAR T-cell persistence in vivo.
a Representative bioluminescent flux imaging of i.p. OV90(eGFP/ffluc) tumor-bearing mice treated i.p. with CD19-CAR, CD19-CAR/mbIL12, TAG72-CAR or TAG72-CAR/mbIL12 T cells. b Quantification of flux (individual mice per group) from OV90(eGFP/ffluc) tumor-bearing mice treated i.p. with CD19-CAR T cells (n = 12/group), CD19-CAR/mbIL12 T cells (n = 12/group), TAG72-CAR T cells (n = 20/group) and TAG72-CAR/mbIL12 T cells (n = 17/group). Combined data are from two independent studies. P value indicates differences between TAG72-CAR and TAG72-CAR/mbIL12 using a two-tailed Student’s t test. c Representative flow cytometric analysis of TAG72-CAR T cells per µL of peritoneal ascites. d Quantification of TAG72-CAR T cells per µL of peritoneal ascites (left) at weeks 2 and 4 post-treatment. n = 2/group per timepoint. Quantification of TAG72-CAR T cells per µL of peripheral blood (right) at weeks 1, 2, and 4 post-treatment. n = 5/group per timepoint. Data are presented as mean values ±SEM. P values indicate differences between TAG72-CAR and TAG72-CAR/mbIL12 using a two-tailed Student’s t test. e Schematic for subcutaneous (s.c.) and i.p. OV90 dual-tumor model and treatment (top). Representative bioluminescent flux imaging of dual-tumor-bearing mice treated i.p. with TAG72-CAR or TAG72-CAR/mbIL12 T cells (bottom). f Quantification of flux (individual mice per group) from mice treated i.p. with TAG72-CAR T cells (n = 8/group) and TAG72-CAR/mbIL12 T cells (n = 8/group). g Quantification of subcutaneous tumor volume (individual mice per group) from mice treated i.p. with TAG72-CAR T cells. h Quantification of TAG72-CAR T cells per µL of peritoneal ascites at week 2 post-treatment. n = 2/group. i Quantification of TAG72-CAR T cells per µL of peripheral blood at days 7, 14, 21, and 28 post-treatment. n = 5/group per timepoint. P values indicate differences between TAG72-CAR and TAG72-CAR/mbIL12 using a two-tailed Student’s t test. j Immunohistochemistry of CD3+ T cells in s.c. tumors at day 12 post-treatment. Data are representative of n = 3 mice/group. Scale bar = 100 µm.
Fig. 4
Fig. 4. Locoregional intracerebroventricular delivery of HER2-CAR/mbIL12 T cells reduces tumor burden and increases regional and systemic CAR T-cell persistence in vivo.
a Schematic for intratibial (i.ti.) and intracranial (i.c.) BBM1 dual-tumor model and treatment. b Representative bioluminescent flux imaging of dual-tumor-bearing mice left untreated (no tx), or treated by intracerebroventricular (i.c.v.) injection of HER2-CAR or HER2-CAR/mbIL12 T cells. c Quantification of brain (top) or bone (bottom) flux from individual mice treated i.c.v. with HER2-CAR T cells (n = 11/group), HER2-CAR/mbIL12 T cells (n = 11/group), or no tx (n = 4/group). P < 0.05 for bone flux, and not significant (ns) for brain flux, comparing HER2-CAR and HER2-CAR/mbIL12 using a Multiple Mann–Whitney test. d, e Representative flow cytometric analysis (d) and quantification (e) of HER2-CAR T cells per µL of blood at weeks 1, 2, and 5 post-treatment. e n = 11–12/group. Data are presented as mean values ±SEM. P values indicate differences between HER2-CAR and HER2-CAR/mbIL12 using a two-tailed Student’s t test. f Immunohistochemistry of CD3+ T cells in i.ti. and i.c. tumors at day 7 post-treatment. Data are representative of n = 3 mice/group. Scale bar = 100 µm.
Fig. 5
Fig. 5. Locoregional intraperitoneal delivery of TAG72-CAR/mbIL12 T cells safely and effectively target ovarian cancer peritoneal metastasis in an immune-competent syngeneic mouse model.
a Schematic for intraperitoneal (i.p.) ID8-mSTn tumor model and treatment. b Representative bioluminescence flux imaging of tumor-bearing mice, treated by intraperitoneal (i.p.) injection of indicated T cells. c Average tumor flux and d percent weight change in indicated T-cell treatments relative to pre-treatment weight (n = 5–7/group). P values indicate differences between TAG72-CAR and TAG72-CAR/mbIL12 in tumor flux using a two-tailed Student’s t test, and between TAG72-CAR/mbIL12 and TAG72-CAR + sIL12 in percent weight change at both days 20 and 21 using a two-tailed Student’s t test. e Spleen photographs (n = 3/group) and f representative CD3 IHC in livers harvested at 7 days post treatment. Quantification of CD3+ counts per mm2: PSCA-CAR: 37.7 ± 1.7, TAG72-CAR: 54.5 ± 7.6, TAG72-CAR + sIL12: 233.5 ± 19.2, TAG72-CAR/mbIL12: 68.3 ± 5.1. Scale bar = 200 µm. g Quantification of serum levels of ALT (left) and AST (right) from mice at 6 days post treatment. n = 4–5/group. P values indicate differences between TAG72-CAR/mbIL12 and TAG72-CAR + sIL12 using a two-tailed Student’s t test. h Percent lymphocytes, monocytes, and neutrophils from complete blood count analysis collected at 7 days post treatment. n = 4/group. i, j ELISA quantification of IFNγ (i) and IL-12 (j) cytokines in mouse serum at day 18 and 20 (day 4 and 6 post treatment, respectively) post tumor injection (n = 6–7/group). All data are presented as mean values ±SEM.
Fig. 6
Fig. 6. TAG72-CAR/mbIL12 T-cell therapy induces TME modifications in ovarian cancer peritoneal metastases in an immune-competent syngeneic mouse model.
a Representative H&E and CD3 IHC in solid tumor masses collected from the upper omental region of i.p. ID8-mSTn tumor-bearing mice treated with indicated T cells. Scale bar = 100 µm. b Representative flow cytometric analysis of tumor cells (CD3-CD45- double negative) in peritoneal ascites. c Quantification of tumor cells (CD3− CD45− double negative) and d immune subsets (CD45+, CD3+, CD11b+, and NK+) as cells/mL in peritoneal ascites. e Representative flow cytometric analysis of percent CAR T cells (CD3+ CD19t+) and f quantification counts of CD4+ and CD8+ CAR T cells/mL in peritoneal ascites. g Quantification of mean fluorescent intensity (MFI) of CD137+ in CAR T cells in peritoneal ascites. h Quantification of percent CD62L+ CD44+ (Tcm) in both CAR+ and CAR− T cells in peritoneal ascites. i Quantification of myeloid cell counts (Ly6G+, Ly6C+, Ly6G−/C− double negative tumor-associated macrophages (TAM) and CD11c+ CD103+ dendritic cells (DC) as cells/mL in peritoneal ascites gated from total CD11b+ cells. Representative flow cytometric analysis of percent (j) and quantification of MFI (k) on CD103+ MHC Class II+ double positive DC in peritoneal ascites. All analyses represent data collected from ascites of ID8-mSTn tumor-bearing mice at 7 days post treatment, n = 3/group. All data are presented as mean values ±SEM. P values indicate differences between TAG72-CAR and TAG72-CAR/mbIL12 using a two-tailed Student’s t test.

Update of

References

    1. Frigault MJ, Maus MV. State of the art in CAR T cell therapy for CD19+ B cell malignancies. J. Clin. Invest. 2020;130:1586–1594. - PMC - PubMed
    1. June CH, Sadelain M. Chimeric antigen receptor therapy. N. Engl. J. Med. 2018;379:64–73. - PMC - PubMed
    1. Hong M, Clubb JD, Chen YY. Engineering CAR-T cells for next-generation cancer therapy. Cancer Cell. 2020;38:473–488. - PubMed
    1. Priceman SJ, Forman SJ, Brown CE. Smart CARs engineered for cancer immunotherapy. Curr. Opin. Oncol. 2015;27:466–474. - PMC - PubMed
    1. Schmidts A, Maus MV. Making CAR T cells a solid option for solid tumors. Front. Immunol. 2018;9:2593. - PMC - PubMed

Publication types