Weekly Digests
‹ Back to June

​​Dimming IFN signaling to brighten CAR-T persistence and efficacy

June 18, 2025

While CAR T cell therapy has shown great success in hematological cancers, relapses may occur, and the therapy has so far shown limited efficacy in solid tumors. Based on their previous data, which showed that genetic deletion of IFNγ in CD19 CAR T cells results in fewer toxicities and better expansion, Bailey, Takei, Escobar, et al. assessed the effects of IFNγ in CAR T cell activity, with the goal of improving CAR T cell efficacy. Their results were recently published in Science Translational Medicine.

To study the effects of IFNγ in CAR T cells, several CD19-targeting CAR-T products were created, including CARCD19 (control), IFNγ-KO CARCD19 (lacking IFNγ production), and IFNγR-KO CARCD19 (cannot respond to IFNγ) T cells. In vitro, activation and cytokine production in response to antigen were similar for all CAR-T, except for IFNγ in the IFNγ-KO CARCD19. However, IFNγR-KO CARCD19 and IFNγ-KO CARCD19 expanded more after antigen exposure. These results were limited to CAR-T with a CD28 costimulatory domain, as in versions with 4-1BB, the loss of IFNγ signaling did not improve expansion.

The three CAR-T versions exhibited similar cytotoxic capacity in vitro. In NSG mice bearing JeKo-1 mantle cell lymphoma, the three CAR-T products induced comparable antitumor activity and survival. However, the IFNγ-KO CARCD19 and IFNγR-KO CARCD19 cells had a higher peripheral expansion after 14 days than the controls.

RNAseq analysis of in vitro-activated CAR-T showed that while all had similar upregulation of highly expressed genes related to antigen exposure, the IFNγ-KO CARCD19 and IFNγR-KO CARCD19 had a decrease in IFN signaling-related genes. Despite the role of IFNγ on checkpoint molecule expression, no differences in checkpoint molecule expression were detected between the three CAR T cell subtypes.

While gene expression suggested better proliferative potential of the IFNγ-KO CARCD19 and the IFNγR-KO CARCD19 cells, in vitro experiments showed similar proliferation. The researchers hypothesized that the increased persistence might be due to protection from IFNγ-induced cell death. Indeed, control CAR T cells upregulated cell death-related genes, increased annexin V, and cleaved caspase 3 after antigen stimulation, and live cell imaging confirmed reduced cell death in IFNγ-KO CARCD19 and IFNγR-KO CARCD19.

Next, the researchers hypothesized that deleting IFNγR in CAR T cells may improve their efficacy in solid tumors. To test this, they developed similar CAR constructs as previously, but targeting mesothelin (CARMESO). As with the CARCD19, all subsets showed similar activation and functionality. The IFNγR-KO CARMESO cells expanded more in response to mesothelin, and both IFNγ-KO CARMESO and IFNγR-KO CARMESO had increased expansion when exposed to mesothelin-expressing pancreatic cancer cell lines in vitro. Similar to the findings with CARCD19, no differences were detected in terms of differentiation, immune checkpoint expression, or proliferation. However, the CAR T cells lacking IFN signaling had reduced apoptotic cell death, suggesting the effects of blocking IFN signaling in CAR T cells (with a CD28 costimulatory domain) were independent of the target antigen.

In vitro, the IFNγ-KO CARMESO had lower antitumor activity, while the IFNγR-KO CARMESO had similar activity as the control CAR-T. The lower activity of the IFNγ-lacking CAR-T may be related to previous findings that IFNγ production by CAR T cells is necessary for antitumor efficacy in solid tumors. Confirming this notion, NSG mice with AsPC-1 pancreatic tumors treated with IFNγ-KO CARMESO had reduced tumor control and survival, while those receiving IFNγR-KO CARMESO exhibited longer tumor control and survival compared to controls, focusing the research efforts on the receptor knockouts.

When the tumor microenvironment was assessed two weeks after CAR-T infusion (IFNγR-KO CARMESO or control), the researchers found that mice that received the IFNγR-KO CARMESO had a 2-fold increase in the CAR T cell population and 50% reduction in tumor cells compared to controls. RNAseq analysis of isolated CAR T cells showed that while the cells had similar in vitro differentiation pre-infusion, the tumor-isolated IFNγR-KO CARMESO exhibited higher levels of CD45RO, GZMA, and GZMB than controls. Among the tumor-isolated IFNγR-KO CARMESO, there were fewer Tregs and more CD8+ T cells than among control CARMESO. Confirming the in vitro data, the IFNγR-KO CARMESO upregulated anti-apoptotic genes, whereas the controls exhibited overexpression of apoptosis- and cell death-related genes. Analysis of the tumor cells showed an increase in IFN-related genes and upregulation of immune checkpoint proteins, tumor suppressor genes, and pro-apoptotic markers in tumors treated with IFNγR-KO CARMESO, suggesting that inhibition of IFNγ uptake by CAR-T drives IFN signaling and cell death in tumor cells.

To determine whether IFNγR-KO CAR T cells could persist and protect from tumor rechallenge, the researchers moved to EGFR-targeting CAR-T, as the tumor model expresses higher levels of EGFR than mesothelin. Mice were treated 2 weeks after tumor inoculation, and control and IFNγR-KO CAREGFR T cells showed similar antitumor activity and mouse survival, with several mice in both groups having complete responses. However, when the cured mice were rechallenged with tumor cells, the ones who originally received control CAR-T were not protected, while those who had received IFNγR-KO CAREGFR had a small tumor relapse that was followed by rapid clearance of the tumor, resulting in long-term survival.

While these results are promising, NSG mice lack an endogenous tumor microenvironment, and IFNγ can affect various cell subsets. Therefore, the researchers assessed if similar results could be obtained in a syngeneic mouse model. T cells from CD45.2+ WT or ifngr-KO C57Bl/6 mice were obtained and transduced to express a B7H3-targeting CAR with a CD28 costimulatory domain. Further, a triple-negative breast cancer cell line overexpressing B7H3 was generated and injected into the mammary fat pads of CD45.1+ C67BL/6 mice. Mice were conditioned with cyclophosphamide, and two days later, infused intravenously with 5x106 CD8+ CAR-T. IFNγR-KO CARB7H3 showed increased antitumor activity and better persistence (measured at day 23) than their WT counterparts.

Together, these data reveal that the deletion of IFNγR from CD28 CAR-T can protect against IFN-induced cell death without affecting efficacy, thereby increasing functional persistence. Further, due to lack of competition for IFNγ, more IFN signaling in tumors aids the antitumor effects of IFNγR-KO CAR T cells in hematological, and importantly, solid tumors in animal models, suggesting this modification in CAR T cell design has potential for further clinical development.

Write-up by Maartje Wouters, image by Lauren Hitchings

References:

Bailey SR, Takei HN, Escobar G, Kann MC, Bouffard AA, Kienka T, Supper VM, Armstrong A, Salas-Benito D, Phillips MK, Birocchi F, Vatsa S, Silva H, Scarfò I, Wehrli M, Grauwet K, Darnell EP, Graham CE, Leick MB, Korell F, Berger TR, Maus MV. IFN-γ-resistant CD28 CAR T cells demonstrate increased survival, efficacy, and durability in multiple murine tumor models. Sci Transl Med. 2025 Jun 4.

In the Spotlight...

An Il12 mRNA-LNP adjuvant enhances mRNA vaccine-induced CD8 T cell responses

Focused on improving long-term and cross-variant protective memory for mRNA vaccines, Aunins and Phan et al. first found that endogenous IL-12 was dispensable for generating CD8+ T cell responses to lipid nanoparticle (LNP)-encapsulated mRNA vaccines. However, as an adjuvant, co-administration of il12mRNA-LNPs with OVA mRNA-LNPs resulted in significantly expanded circulating effector and memory CD8+ T cells, including tissue-resident CD8+ T cells in crucial barrier sites. LNP-IL-12 enhanced CD8+ T cell responses to SARS-CoV-2 and influenza virus antigens, and improved protection against Listeria monocytogenes-OVA and B16-OVA melanoma.

Contributed by Katherine Turner

CSF1R+ myeloid-monocytic cells drive CAR-T cell resistance in aggressive B cell lymphoma

Stahl, Gödel, and Balke-Want et al. performed multimodal analysis of pre- and post-CAR T cell-treated tissue from patients with relapsed or refractory aggressive B cell lymphoma (B-NHL) and identified a distinct CSF1R+CD14+CD68+ lymphoma-associated myeloid-monocytic (LAMM) cell population associated with poor outcomes in both human and murine B-NHL. Cell–cell communication analysis inferred that direct LAMM–T cell interactions via the PGE2–EP2/EP4 axis impaired CAR T cell functions. Anti-CD19 CAR T cell therapy with CSF1R blockade exhibited synergistic effects and improved survival in an autochthonous lymphoma mouse model.

Contributed by Shishir Pant

Methotrexate exerts antitumor immune activity and improves the clinical efficacy of immunotherapy in patients with solid tumors

Yang et al. found that methotrexate (MTX) competitively inhibited the membrane cGAMP hydrolase ENPP1, which both enhanced immune cell activation by preventing loss of exogenous cGAMP, and inhibited downstream accumulation of suppressive adenosine. In addition, MTX activated STING in cancer cells by inducing DNA damage. MTX synergized with radiation (IR) and ICB in vivo, increasing infiltration of CD8+ and CD11c+ cells into cold tumors. Addition of MTX to IR/ICB in an open-label trial enhanced responses vs. historical controls, where response was correlated with baseline ENPP1/cGAS abundance and post-treatment increases in IFN pathway cytokines.

Contributed by Morgan Janes

Secretion of a VEGF-blocking scFv enhances CAR T-cell potency

Analyzing scRNAseq data from CAR T cell infusion products, Supper and Donner et al. found that VEGFR1 was expressed in 41BB.z CAR T cells and associated negatively with treatment response. In vitro, activation increased VEGFR1 expression on 41BB.z, but not 28.z CAR T cells; activated CAR T cells also secreted VEGF. To block VEGF signaling, CAR T cells were engineered to secrete an anti-VEGF scFv. This modification (cf. a nonspecific scFv) improved CAR T cell expansion in target cell coculture, and tumor infiltration and antitumor efficacy in vivo. VEGF and endothelial cells were diminished around the engineered CAR T cells in tumors.

Contributed by Alex Najibi

Splenic fibroblastic reticular cells orchestrate dendritic cell maturation and facilitate CD8+ T cell priming and protective memory

Alexandre et al. found that fibroblastic reticular cells (FRCs), which support the architecture of the white pulp in the spleen, also supported virus-specific CD8+ T cell responses during LCMV infection by coordinating early viral replication and an inflammatory milieu for DC maturation and optimal T cell activation. FRCs also supported T cell clustering with cDC1s in the T cell zone (prior to migration to the infected marginal zone), leading to the generation of virus-specific effector T cells and the generation and maintenance of memory T cells. Without FRCs, T cells clustered with cDC1 and virus-infected cells in the marginal zone, and T cell responses were suboptimal.

Contributed by Lauren Hitchings

Sustained macrophage reprogramming is required for CD8+ T cell-dependent long-term tumor eradication

Jardim and Bica et al. demonstrated that i.t. myeloid cell treatment (MCT), a combination of TLR3 and CD40 agonists, effectively reprogrammed TAMs to adopt an antitumor phenotype (iNOS+ and CD38+) and induced tumor control in an orthotopic E0771 mouse breast cancer model. MCT-activated TAMs were transient and required secondary injections for sustained phenotypic and functional maintenance. MCT-induced TAMs promoted antigen presentation and effector CD8+ T cell recruitment, ultimately resulting in long-term tumor eradication. ROS and TNFα acted as potent antitumor effector molecules of MCT-activated TAMs in vivo.

Contributed by Shishir Pant

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.