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
. 2025 Jun 4;17(801):eadp8166.
doi: 10.1126/scitranslmed.adp8166. Epub 2025 Jun 4.

IFN-γ-resistant CD28 CAR T cells demonstrate increased survival, efficacy, and durability in multiple murine tumor models

Affiliations

IFN-γ-resistant CD28 CAR T cells demonstrate increased survival, efficacy, and durability in multiple murine tumor models

Stefanie R Bailey et al. Sci Transl Med. .

Abstract

Interferon-γ (IFN-γ) plays complex and, sometimes, contradictory roles in cancer, which can affect patient responses to treatments such as immunotherapies. We recently demonstrated that IFN-γ production by chimeric antigen receptor (CAR) T cells is not required for efficacy in hematologic tumor models, whereas IFN-γ receptor (IFN-γR) signaling in solid tumor cells facilitates CAR T cell adhesion and antigen-specific cytotoxicity. Here, we show that IFN-γ induces apoptosis of CAR T cells bearing a CD28 intracellular signaling domain, which can be reduced through targeting of IFN-γ or IFN-γR. In hematologic malignancies, knockout of IFN-γR (IFN-γRKO) in CAR T cells increased their persistence without compromising efficacy. In xenograft and syngeneic solid tumor models, IFN-γR knockout CAR T cells displayed more potent tumor control, prolonged survival, and improved T cell memory that conferred protection from tumor rechallenge. RNA sequencing of tumor-infiltrating IFN-γRKO CAR T cells derived from tumor-bearing mice revealed increased cell death in tumor cells. Collectively, these data show that inhibition of IFN-γ signaling can increase the expansion and antitumor activity of CD28-based CAR T cells in liquid and solid tumors.

PubMed Disclaimer

Conflict of interest statement

Competing interests:

S.R.B. and M.V.M. are inventors on patents related to this work held by MGH (PCT/US2020/065733; Provisional patent MGH023–056). M.V.M. is an inventor on additional patents related to adoptive cell therapies held by MGH and University of Pennsylvania (some licensed to Novartis). I.S. is currently employed at Arsenal Bio. M.W. received an honoraria from Pierre Fabre and BMS. M.V.M. holds equity in 2SeventyBio, A2Bio, Affyimmune, Cargo, Century Therapeutics, GBMnewco, Neximmune, Oncternal, and TCR2; serves on the Board of Directors of 2Seventy Bio; and has served as a consultant for multiple companies involved in cell therapies. M.V.M. receives research support from Kite Pharma and Moderna Therapeutics, which is unrelated to this project. M.V.M.’s interests were reviewed and are managed by Massachusetts General Hospital, and Mass General Brigham in accordance with their conflict-of-interest policies.

Figures

Fig. 1.
Fig. 1.. Knockout of IFNγ or IFNγ receptor drives greater CD28 CAR-T cell expansion in vitro.
(A) Schematic diagram of genetic constructs used to generate human chimeric antigen receptor (CAR)-T cells with a CD19 single-chain fragment variable (scFv), CD28 costimulatory domain, and CRISPR/Cas9 editing of T cell receptor A constant region locus (TRAC; CARCD19), TRAC and IFNG (IFNγKO CARCD19), or TRAC and IFNGR (IFNγRKO CARCD19). (B) Expression of CD3 (Allophycocyanin, APC; left), IFNγR (Alexa Fluor 647, AF647; middle) and phosphoSTAT1 (pSTAT1) signaling (AF647; right) following exposure to recombinant human IFNγ on resting CAR-T cells and donor-matched untransduced (UTD) T cells) with representative histograms from 3 biological replicates. (C) IFNγ (Brilliant Violet 510, BV510) expression on CAR-T cells was assessed by flow cytometry following four-hour activation with phorbol myristate acetate (PMA) and ionomycin; (n=3 biological replicates). Y-axis is an empty channel. (D) CD4 and CD8 expression on CAR-T cells and UTD T cells were determined using flow cytometry; n=6 biological replicates. Data are shown as mean ± SEM with P values determined by two-way ANOVA with Tukey’s multiple comparisons test. (E) CAR-T cells were activated overnight with CD19 antigen and secreted cytokines were measured by Luminex; bubble plot represents average cytokine expression (ng/ml) from 3 biological replicates. (F) Expansion of CD28 CAR-T cells in response to Nalm6 cells was monitored using real-time Incucyte imaging. Average expansion (±SEM) was measured over 72 hours. Data represents 4 biological replicates which were analyzed by one-way ANOVA with Tukey’s multiple comparisons analysis at 24, 48, and 72 hours. Significance of IFNγKO CARCD19 vs. CARCD19 (pink *) or IFNγRKO CARCD19 vs. CARCD19 (turquoise *,**) is shown. (G) Schematic diagram of constructs used to generate control, IFNγKO, and IFNγRKO CAR-T cells with a CD19 scFv and 4–1BB costimulatory domain. (H) Representative histograms of CD3 (APC; left) and IFNγR (AF647; middle) expression on resting CAR-T cells and donor-matched UTD T cells measured by flow cytometry alongside IFNγ following a four-hour activation with PMA and Ionomycin (BV510; right). Data was analyzed from 3 biological replicates. (I) Expansion of 4–1BB CAR-T cells in response to Nalm6 cells was monitored using real-time Incucyte imaging. Average expansion (±SEM) was measured over 72 hours. Data represents 3 biological replicates which were analyzed by one-way ANOVA. *P<0.05, **P<0.01, ****P<0.0001; ns, not significant.
Fig. 2.
Fig. 2.. IFNγKO and IFNγRKO CARCD19 maintain anti-tumor activity against hematologic malignancies and display greater persistence in vivo.
(A) CAR-T cells were cultured with Nalm6 cells (1:1) and tumor cytolysis was measured by Incucyte. At 3 and 6 days, cells were collected, counted, and CAR-T cells were re-plated with fresh Nalm6 cells (1:1) before being returned to the Incucyte. Data represents mean (±SEM) for 2 biological replicates. (B to F) NSG mice bearing intravenously administered CBGGFP+ JeKo-1 tumor cells (1e6 IV) were left untreated (tumor only; TO) or treated with 1e6 CARCD19, IFNγKO CARCD19, or IFNγRKO CARCD19 (1e6 IV). Data was obtained from 4–5 mice/group per donor (2 donors total). (B) Schematic timeline of in vivo CAR-T cell treatment. Tumor burden was assessed by bioluminescent imaging for each CAR-T cell group and tumor only control over 75 days post-CAR-T cell infusion (C) and graphed by flux (D). (E) Overall survival was measured for each CAR-T cell treatment cohort and represented by Kaplan-Meier survival curve. (F) Mice were bled 14 days post-CAR-T cell infusion and T cell persistence was determined using flow cytometry. Data are shown as mean ± SEM with P values determined by one-way ANOVA with Tukey’s multiple comparisons analysis (D, F) or Mantel-Cox test (E). **P<0.01, ****P<0.0001; ns, not significant.
Fig. 3.
Fig. 3.. Deletion of IFNγ or IFNγR protects CD19 CAR-T cells from cell death.
(A) CAR-T cells were activated overnight with plate-bound CD19 antigen prior to RNA sequencing using NanoString and graphed as volcano plots. Proliferation (blue dots) and cell death (pink dots) genes are highlighted in CARCD19 (purple shading) compared to either IFNγKO CARCD19 (pink shading) or IFNγRKO CARCD19 (turquoise shading). Three adjusted P value cutoffs are shown as a dotted bottom line (P<0.05), dashed middle line (P<0.01), and a dot-dash top line (P<0.001). Data was derived from 3 biological replicates. (B) Proliferation in response to CD19+ Nalm6 cells was assessed by Cell Trace Violet at 6 days post-activation. Data are represented as histograms and pie charts showing the percentage of cells (white numbers) for each number of cell divisions (0=gray, 1=red, 2=turquoise, 3=purple, 4=orange) derived from 3 biological replicates. (C and D) Annexin V (C) and cleaved Caspase 3 (D) expression were determined by flow cytometry for CAR-T cell groups at rest (no activation; NA) and 48 hours post-activation (CD19); n=3 biological replicates. (E) Annexin V expression on CAR-T cells activated with plate-bound CD19 antigen was assessed using Incucyte real-time analysis over a 72-hour period in 3–6 biological replicates]. Significance at 72 hours was noted for IFNγRKO CARCD19 vs. CARCD19 (turquoise *). (F and G) CAR-T cells were activated overnight with CD19 antigen in the absence or presence of exogenous recombinant human IFNγ prior to RNA sequencing using NanoString. Data is presented as bar graphs (F) and volcano plots with adjusted P value cutoffs shown as a dotted bottom line (P<0.05) and dashed top line (P<0.01) (G); n=3 biological replicates. Interferon signaling (purple dots) and cell death (magenta dots)-related genes are highlighted. (H) Wildtype CD19-specific CAR-T cells with no CRISPR/Cas9 editing were activated with CD19 in the absence or presence of blocking antibodies to IFNγ (αIFNγ) or IFNγR (αIFNγR) and Annexin V expression was assessed by Incucyte; n=2 biological replicates. Data are shown as mean ± SEM with P values determined by one-way ANOVA with Tukey’s multiple comparisons analysis (C, D, E, F, H). *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001; ns, not significant.
Fig. 4.
Fig. 4.. Loss of IFNγ signaling drives greater survival in mesothelin-specific CD28 CAR-T cells.
(A) Schematic diagram of CARMESO, IFNγKO CARMESO, and IFNγRKO CARMESO constructs with the anti-mesothelin SS1 scFv and CD28 costimulatory domain generated from healthy donors. (B) Representative histograms depicting expression of CD3 (APC), IFNγR (AF647), and IFNγ (BV510) for mesothelin-specific CAR-T cell constructs. (C) CD4 and CD8 frequencies were determined by flow cytometry for 6 biological replicates. Data are shown as mean ± SEM with P values analyzed by one-way ANOVA with Tukey’s multiple comparisons test. (D) Fold change expansion of CAR-T cells in response to mesothelin was monitored using real-time Incucyte imaging over a 72-hour period (left) and quantitative flow cytometry (right) from 3–4 biological replicates. Data are shown as mean ± SEM with P values analyzed by one-way ANOVA with Tukey’s multiple comparison test. Significance noted for IFNγRKO CARMESO vs. CARMESO (turquoise **). (E) Memory markers CD45RA (Peridinin chlorophyll protein-Cyanine5.5, PerCP/Cy5.5) and CD62L (Fluorescin isothiocyanate, FITC) were assessed on resting CAR-T cells and shown by representative flow cytometry plots and pie charts for overall averages for 6 biological replicates. Data is shown as naïve (CD45RA+CD62L+), central memory (CM; CD45RA-CD62L+), effector memory (EM; CD45RA-CD62L-), and effector (EFF; CD45RA+CD62L-). (F) CAR-T cells were activated with mesothelin, and exhaustion markers were measured by flow cytometry with pre- (PRE) and post-activation (POST) graphed. Data are shown as mean ± SEM from 3 biological replicates. (G) Proliferation of CAR-T cells in response to mesothelin was assessed using Cell Trace Violet and displayed as a representative donor and average pie chart (top) and collective averages (bottom) showing mean ± SEMt; n=3 biological replicates. Pie charts show the percentage of cells (white numbers) for each number of cell divisions (0=gray, 1=red, 2=turquoise, 3=purple, 4=orange, 5=blue, 6=green). (H) Annexin V on CAR-T cells exposed to mesothelin was assessed using Incucyte real-time analysis and displayed as fold change of average green area compared to timepoint zero; n=4 biological replicates. Data are shown as mean ± SEM with P values determined by one-way ANOVA with Tukey’s multiple comparisons analysis (IFNγRKO CARMESO vs. CARMESO P=.0856). *P<0.05, **P<0.01, ***P<0.001.
Fig. 5.
Fig. 5.. IFNγRKO CARMESO exhibit greater anti-tumor activity and survival against pancreatic cancer.
(A) CAR-T cells and donor-matched UTD T cells were activated with mesothelin antigen overnight. Granzyme B and perforin expression were examined using flow cytometry with representative histograms shown for 3 biological replicates. (B) CARMESO, IFNγKO CARMESO, and IFNγRKO CARMESO were mixed with mesothelin+ AsPC-1, BxPC-3, or Capan2 cells at a 1:1 ratio and tumor cytolysis was assessed by Incucyte. Statistical significance was determined by one-way ANOVA with Tukey’s multiple comparisons test for 2–3 biological replicates and is shown as ± SEM. P values shown are CARMESO (purple *) or IFNγRKO CARMESO (turquoise *) compared to IFNγKO CARMESO. (C) NSG mice bearing subcutaneous AsPC-1 tumor cells (1.5e6 SC) were left untreated (tumor only; TO) or treated with 3e6 CARMESO, IFNγKO CARMESO, or IFNγRKO CARMESO (IV). (D) Mice were bled weekly through day 35 post-CAR-T cell infusion and CAR-T cell persistence was determined using flow cytometry. Data are shown as mean ± SEM with P values determined by one-way ANOVA with Tukey’s multiple comparisons test. (E) Tumor burden was assessed by weekly caliper measurements. Data are shown as mean ± SEM and analyzed by one-way ANOVA with Tukey’s multiple comparison test. Statistical significance was assessed on days 56 (IFNγRKO CARMESO (turquoise *) or CARMESO (purple *) vs. TO), 77 (IFNγRKO CARMESO (turquoise *) vs. IFNγKO CARMESO), and 97 (IFNγRKO CARMESO (turquoise *) vs. CARMESO). (F) Overall survival of mice treated with CAR-T cells was assessed by Mantel-Cox test. For C-F, two biological replicates were used at 4–5 mice/group for each donor. *P<0.05, **P<0.01.
Fig. 6.
Fig. 6.. Pancreatic tumors from IFNγRKO CARMESO-treated mice exhibit greater interferon signaling and apoptosis.
(A) Schematic diagram representing mouse NSG mice bearing subcutaneous AsPC-1 tumor cells (1.5e6 administered SC) which were left untreated (tumor only; TO) or treated with 3e6 CARMESO, IFNγKO CARMESO, or IFNγRKO CARMESO by IV. Tumors were collected 14 days post-CAR-T cell infusion, processed, and sorted into CAR-T cells and AsPC-1 tumor cells. (B) Representative flow cytometry plots of the GFP+ tumor and PE-TexasRed+ CAR-T cells isolated from the tumors of AsPC-1-bearing mice. (C and D) CARMESO and IFNγRKO CARMESO from before infusion (PRE) and post-infusion from the tumor (POST) (C) and tumor cells from mice receiving no treatment (TO) or treated with CARMESO or IFNγRKO CARMESO (D) were isolated and analyzed by NanoString. Z-scores for all genes in each group are displayed as a heatmap. (E) Interferon gene signatures are shown for mesothelin-specific CAR-T cells. Gene counts were normalized. Purple arrows indicate gene signatures which were expressed more in CARMESO, turquoise arrows indicate gene signatures expressed higher in the IFNγRKO CARMESO. (F) Cell death gene signatures were assessed by NanoString in CARMESO and IFNγRKO CARMESO T cells. Gene counts for anti-apoptotic (green arrow) and pro-apoptotic (red arrow) were normalized. (G and H) Interferon (G) and pro-apoptotic (H) gene signatures are shown by the scatterplot for CAR-T cell constructs pre and post infusion. Gene counts were normalized, and data are shown as mean ± SEM with P values determined by one-way ANOVA with Tukey’s multiple comparisons test. (I) Interferon gene counts from tumors receiving no treatment (TO) or treated with CARMESO or IFNγRKO CARMESO were normalized with purple arrows indicating gene signatures which were expressed more in CARMESO-treated tumors and turquoise arrows indicating gene signatures expressed higher in IFNγRKO CARMESO-treated mice. (J) Cell death gene signatures were assessed by NanoString in tumors from untreated or CARMESO or IFNγRKO CARMESO-treated mice. Gene counts for anti-apoptotic (green arrow) and pro-apoptotic (red arrow) were normalized. (K and L) Interferon (K) and pro-apoptotic (L) gene signatures in tumors are shown by scatterplot for non-treated or CAR-T cell-treated mice. Gene counts were normalized, and data are shown as mean ± SEM with P values determined by one-way ANOVA with Tukey’s multiple comparisons test. For this experiment, data was collected from 2 mice per group for each donor, with two donors total. Data shown in C to L are from 1 representative donor. *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001; ns, not significant.
Fig. 7.
Fig. 7.. EGFR-targeting IFNγRKO CAR-T cells confer protection from tumor rechallenge.
(A) Schematic diagram of control and IFNγRKO CAR-T cells targeting EGFR generated from healthy donors. (B) Schematic of in vivo treatment protocol. NSG mice bearing subcutaneous AsPC-1 tumor cells (1.5e6 SC) were left untreated (tumor only; TO) or treated with 3e6 CAREGFR or IFNγRKO CAREGFR (IV) and tumor burden was measured by caliper weekly. Seventy days post-CAR-T cell infusion, mice displaying curative responses (3 mice/group) were re-challenged with 1.5e6 AsPC-1 tumor cells in the flank opposite of the initial tumor injection. Tumor burden was assessed using weekly caliper measurements. (C and D) Tumor size (C) and overall survival (D) were assessed in tumor only and EGFR-specific CAR-T cell-treated mice over a 200-day period. Mice were rechallenged on day 70. Significance between tumor only and both treated groups is shown at day 56, with IFNγRKO CARMESO vs CARMESO shown at day 143. Data are shown as mean ± SEM with P values determined by one-way ANOVA with Tukey’s multiple comparisons test (C; day 56), unpaired student’s t-test (C; day 143), or Mantel-Cox (D). 5 mice/group with 3 mice/group for re-challenge, taken from 2 donors. *P<0.05, **P<0.01,***P<0.001.
Fig. 8.
Fig. 8.. IFNγRKO CAR-T cells display increased efficacy and survival in a syngeneic model of triple-negative breast cancer.
(A) Schematic diagram of treatment protocol. Briefly, T cells were isolated from CD45.2+ C57BL/6 wildtype (WT) or Ifngr KO (IFNγRKO) mice and retrovirally transduced to express control (CARFITC) or B7H3-targeted (CARB7H3) CAR constructs containing a CD28 costimulatory domain prior to in vitro and in vivo assessment. (B and C) Transduction efficiency (B) and memory (C) were assessed by flow cytometry. Naïve (CD44-CD62L+), central memory (CM; CD44+CD62L+), and effector memory (EM; CD44+CD62L-) frequencies were determined for WT CAR and IFNγRKO CAR. Naïve cells were <2% for each group, making it not clearly seen on the graph. Data are shown as mean ± SEM with P values determined by unpaired t-test for 3 biological replicates. (D to G) 3e5 triple-negative breast cancer cells overexpressing B7H3 (E0771 B7H3) were injected into the mammary fat pad of CD45.1+ C57BL/6 mice. Tumor-bearing mice were conditioned with 100µg/g cyclophosphamide prior to intravenous (IV) administration of 5e6 CD8+ CAR-T cells and tumor size was monitored by caliper throughout the experiment and graphed as averages (D) and individual mice (E). On day 23, tumors were measured by caliper (F), collected, and frequencies of CAR-T cells were determined by flow cytometry (D). For in vivo experiment, data are derived from 5 mice per group. Data are shown as mean ± SEM with P values determined by two-way ANOVA (D), one-way ANOVA with Tukey’s multiple comparison test (F), and unpaired t-test (G). *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001; ns, not significant.

References

    1. Laetsch TW, Maude SL, Rives S, Hiramatsu H, Bittencourt H, Bader P, Baruchel A, Boyer M, De Moerloose B, Qayed M, Buechner J, Pulsipher MA, Myers GD, Stefanski HE, Martin PL, Nemecek E, Peters C, Yanik G, Khaw SL, Davis KL, Krueger J, Balduzzi A, Boissel N, Tiwari R, O’Donovan D, Grupp SA, Three-Year Update of Tisagenlecleucel in Pediatric and Young Adult Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia in the ELIANA Trial. J Clin Oncol 41, 1664–1669 (2023); published online EpubMar 20 ( 10.1200/JCO.22.00642). - DOI - PMC - PubMed
    1. Cappell KM, Sherry RM, Yang JC, Goff SL, Vanasse DA, McIntyre L, Rosenberg SA, Kochenderfer JN, Long-Term Follow-Up of Anti-CD19 Chimeric Antigen Receptor T-Cell Therapy. J Clin Oncol 38, 3805–3815 (2020); published online EpubNov 10 ( 10.1200/JCO.20.01467). - DOI - PMC - PubMed
    1. Maude SL, Laetsch TW, Buechner J, Rives S, Boyer M, Bittencourt H, Bader P, Verneris MR, Stefanski HE, Myers GD, Qayed M, De Moerloose B, Hiramatsu H, Schlis K, Davis KL, Martin PL, Nemecek ER, Yanik GA, Peters C, ..., S. A. Grupp, Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. N Engl J Med 378, 439–448 (2018); published online EpubFeb 1 ( 10.1056/NEJMoa1709866). - DOI - PMC - PubMed
    1. Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, Gao P, Bandyopadhyay S, Sun H, Zhao Z, Lundh S, Pruteanu-Malinici I, Nobles CL, Maji S, Frey NV, Gill SI, Loren AW, Tian L, Kulikovskaya I, ..., June CH, Decade-long leukaemia remissions with persistence of CD4(+) CAR T cells. Nature 602, 503–509 (2022); published online EpubFeb ( 10.1038/s41586-021-04390-6). - DOI - PMC - PubMed
    1. Wittibschlager V, Bacher U, Seipel K, Porret N, Wiedemann G, Haslebacher C, Hoffmann M, Daskalakis M, Akhoundova D, Pabst T, CAR T-Cell Persistence Correlates with Improved Outcome in Patients with B-Cell Lymphoma. Int J Mol Sci 24, (2023); published online EpubMar 16 ( 10.3390/ijms24065688). - DOI - PMC - PubMed

LinkOut - more resources