Weekly Digests
‹ Back to December

IL-9-powered CAR-T for improved solid tumor responses

December 3, 2025

Overcoming immunosuppression in the tumor microenvironment (TME) is a major challenge in CAR T cell research to improve therapeutic efficacy towards solid tumors. To address some of the CAR-T functionality challenges, Castelli et al. engineered CAR T cells with an authentic IL-9 receptor (IL-9R) to enable IL-9 responsiveness. These CAR-T were assessed in various syngenic murine and human xenograft solid tumor models in a recent publication in Immunity.

The researchers first engineered mouse T cells with a mesothelin-targeting CAR (A03 CAR) and the wild-type IL-9R (CAR-IL9R T cells). Treatment of these cells in vitro with IL-9 resulted in a dose-dependent phosphorylation of STAT1, STAT3, and STAT5. CAR-IL9R T cells showed improved killing capacity, with increased production of IFNγ, TNFα, and IL-10. Furthermore, IL-9 treatment led to a shift toward a stem-like memory phenotype in the CAR-T.

To study in vivo effects, C57/BL6 mice were subcutaneously (s.c.) injected with pancreatic ductal adenocarcinoma (PDAC) PDA7940b cells. T cells from CD45.1+ B6.SJL-PtprcaPepcb/BoyJ mice were used to engineer CAR-IL-9R T cells, which were injected intravenously (i.v.). IL-9 was delivered intratumorally (i.t.) by injection of a replication-deficient adenovirus encoding murine IL-9 (Ad-mIL9) for local production of IL-9. Treatment with CAR-IL9R and Ad-mIL9 improved overall survival (OS) and reduced tumor progression. Tumors and spleens collected on day 5 or 6 of treatment had higher CAR T cell tumor infiltration than controls. This observation suggested improved persistence and trafficking, as no differences in systemic CAR-T cell expansion were detected in the blood. Furthermore, no significant changes in other immune populations were detected in the tumor.

Moving to a human model system, NSG mice were implanted with the human PDAC line AsPC-1. Primary T cells from healthy donors were transduced with a human mesothelin-directed CAR construct (M5 CAR) and IL-9R. Mice were treated i.v. with the CAR-T and received an i.t. injection of Ad-hIL9. Low doses of CAR-IL9R T cells (0.03E6) combined with Ad-hIL9 resulted in improved antitumor efficacy compared to higher doses of control CAR-T (1E6). When mice that were in complete remission after this treatment were rechallenged with AsPC-1 in the contralateral flank on day 90 after treatment, tumors were rejected.

In the human PDAC model system, peripheral CAR-IL9R T cells expanded, persisted, and showed increased cytokine production for up to 10 weeks after treatment. Tumors had higher infiltration of both CD8+ and CD4+ CAR T cells, with a higher proportion of early memory T cells. There was also a decrease in the percentage of PD-1+TIM3+ T cells, and an increase in PD-1-TIM3- and PD-1-LAG3- T cells.

Using a continuous antigen exposure (CAE) assay, in which CAR-IL9R T cells were cocultured with AsPC-1 cells every other day for 10 days, the researchers investigated the effects of repeated antigen stimulation in the presence or absence of IL-9. IL-9-treated CAR-IL9R T showed the best tumor killing capacity, which was antigen-specific and correlated with high production of IFNγ. While control CAR T cells downregulated surface CAR expression, IL-9-treated cells continued to proliferate, and retained CAR expression.

For mechanistic assessment, CAR-IL9R T cells from three donors were subjected to the CAE assay (with or without IL-9), harvested at day 10, and subjected to scRNAseq. Ingenuity pathway analysis revealed the Th1 pathway, IL-20 family signaling, cholesterol biosynthesis, and IL-10 signaling as top-enriched pathways in the IL-9-treated CAR-T.

Based on these results, the researchers investigated the role IL-10 plays in the phenotype and function of CAR-IL9R T cells. NSG mice were engrafted with PC3-PSMA prostate tumor cells and treated i.t. with Ad-hIL9, and i.v. with CAR-T (targeting PSMA, coexpressing a dominant negative TGF-βRII and IL-9R). In this model, the CAR-IL9R T cells plus IL-9 treatment also resulted in reduced tumor progression and improved OS, with increased numbers of peripheral CAR-T compared to controls. IL-10 neutralization did not affect antitumor efficacy or blood CAR T cell counts, suggesting that IL-10 was not a major driver of the enhanced functionality.

The researchers then performed clustering analysis on the scRNAseq data. In the IL-9-treated group, a higher proportion of cells were found in effector clusters, with an increase in the CD4+ activated and GNLY++ effector clusters, while the group without IL-9 treatment had more CAR-T in the CD8+ exhausted and GZMA++ cytotoxic clusters. Differential gene expression analysis showed that IL-9 treatment in CD8+ CAR-IL9R T cells increased transcription factors, chemokine-related genes, markers of naive/memory CD8+ T cells, and cytotoxicity-related genes. In CD4+ CAR-IL9R T cells, IL-9 treatment increased expression of chemokine-related genes, markers of naive/memory T cells, proliferation-associated genes, and cytotoxicity-related genes. Gene set enrichment analysis revealed increases in expression of genes related to interleukin signaling pathways in CD4+ and CD8+ CAR-T, and within the CD4+ population, there was an increase in Th1 and Th9 cells.

Assessing CAR T cell trajectories over pseudotimes identified 6 trajectories, of which, four differed between CAR T cells treated with IL-9 and those exposed to mock conditions. In CD8+ T cells, IL-9 enhanced progression from naive to central memory and effector states, while progression to exhausted/dysfunctional states was reduced. In CD4+ T cells, trajectories toward proliferative states were promoted. These data were confirmed by RNA velocity analysis, which also showed a pattern of movement away from terminal states, and toward less differentiated phenotypes and more proliferative effector states.

Investigation of the regulatory interactions between transcription factors and genes showed a strong enrichment in STAT4 activity in the CAR-IL9R T after IL-9 treatment, which has not previously been linked to IL-9 signaling. The researchers validated this finding at the protein level, showing robust phosphorylation of STAT4. STAT4 deletion using CRISPR-Cas9 reduced IFNγ and TNFα production in IL-9-treated CAR-IL9R T cells. These cells had a slight increase in memory markers, and a limited reduction of in vitro cytotoxicity, suggesting STAT4 may contribute to the IL-9-mediated effector functions.

Therefore, IL-9 signaling in CAR-T in the TME positively impacts T cell fate, resulting in less differentiated states and reduced T cell exhaustion. Given that this modification improved antitumor activity, CAR T cell expansion, and persistence in solid tumor models, it might help overcome some of the major obstacles to clinical CAR-T treatment of solid tumors.

Write-up by Maartje Wouters, image by Ute Burkhardt

Meet the researcher

This week, first author Sofía Castelli answered our questions.

First author Sofía Castelli

What was the most surprising finding of this study for you?
The most surprising finding for me was how dramatically IL-9 signaling amplified CAR T cell potency. We found that very low doses of IL-9-engineered CAR T cells were sufficient to clear tumors in vivo, which was far beyond what we anticipated. Even more striking, these cells were not only more effective, but also far more persistent, remaining in circulation long after conventional CAR T cells contracted. Our in vitro dysfunction model mirrored this behavior; IL-9 signaling essentially reprogrammed CAR T cells toward effector, memory, and proliferative states, rather than exhaustion. This level of fate redirection was completely unexpected.

What is the outlook?
A key next step is to test IL-9-signaling CAR T cells across additional solid tumor models and in more clinically relevant systems, such as humanized or patient-derived models, before we can eventually move this therapy into the clinic. It will also be important to further investigate the molecular pathways that underlie the improved phenotype. From an engineering perspective, developing tunable or inducible IL-9 circuits and exploring combinations with complementary therapies could further enhance efficacy. Together, these studies will help define the broader applicability of IL-9-signaling CAR T cells and their potential for clinical translation.

If you could go back in time and give your early-career self one piece of advice for navigating a scientific career, what would it be?
I would tell my early-career self not to be discouraged by uncertainty or by experiments that don’t work as planned. Those moments are a normal part of research and often lead to better questions. I’d also remind myself to step back occasionally and keep the broader scientific goals in view, rather than getting lost in day-to-day setbacks. Some of the most meaningful progress comes from exploring ideas that fall slightly outside the obvious or expected, so staying open to those directions is important.

References:

Castelli S, Wilson WV, Uslu U, Finck AV, Rommel PC, Assenmacher CA, Atoche SJ, Siurala M, Aznar MA, Young RM, June CH. IL-9 signaling redirects CAR T cell fate toward CD8+ memory and CD4+ cycling states, enhancing antitumor efficacy. Immunity. 2025 Nov 21.

In the Spotlight...

EVOLVE platform, a trispecific T cell engager with integrated CD2 costimulation, for the treatment of solid and hematologic tumors

Sergeeva and Jin et al. developed the EVOLVE trispecific T cell engager (TCE) platform, comprising a tumor antigen-targeting domain, an affinity-attenuated CD3 agonist (optimized to increase T cell viability and effector function), and an LFA-3 extracellular domain fragment (to induce CD2 costimulation) in an IgG1 format, with Fc chain effector-inactivating mutations. EVOLVE TCEs exhibited greater tumor-killing potency than CD3 affinity-matched bispecifics in vitro, and induced greater, more durable antitumor activity and lower tumor antigen-dependent cytokine release than control (higher and lower affinity) CD3 TCEs in mouse xenograft models.

Contributed by Paula Hochman

MHC-II-restricted neoantigen vaccine reverses immune microenvironment and overcomes resistance to immune-checkpoint inhibitors in cold tumors

Song, Lu, and Shi et al. demonstrated that an MHC-II restricted neoantigen vaccine (M44) increased inflammatory signaling within the TME, enhanced CD4+ and CD8+ T cell infiltration, and reduced tumor growth in B16 tumors, while showing signs of T cell exhaustion. Vaccination increased the inferred interaction between TIGIT on T cells and its ligand PVR on myeloid cells, impairing the function and proliferation of Th1 and effector and memory CD8+ T cells. M44 vaccine plus TIGIT antibody inhibited tumor growth, enhanced the helper and cytotoxic functions of antigen-specific CD4+ T cells, and increased effector and memory CD8+ T cells.

Contributed by Shishir Pant

Target antigen-displaying extracellular vesicles boost CAR T cell efficacy in cell and mouse models of neuroblastoma

Investigating neuroblastoma, Giudice et al. found that in both mice and patient samples, tumor-derived extracellular vesicles (EVs) displayed CAR T cell target antigens GPC2 and GD2, with minimal expression of PD-L1, and could activate respective CAR T cells. To utilize this therapeutically, the researchers engineered GPC2+ synthetic EVs as CAR T cell enhancers, and armed them with either albumin-binding domains or GD2-binding domains. Serial infusion of these SyntEVs coated GPC2lo tumor cells with GPC2 (circumventing antigen downregulation), enhanced CAR T cell persistence, and increased tumor control.

Contributed by Lauren Hitchings

Targeting orthotopic and metastatic pancreatic cancer with allogeneic stem cell-engineered mesothelin-redirected CAR-NKT cells

Li et al. generated CAR-NKT cells by transducing cord blood HSPCs with an anti-mesothelin CAR, the iNKT TCR, and sIL-15, prior to a 6-week NKT differentiation protocol with high purity and yield. Compared to CAR T cells, CAR-NKT had reduced HLA expression, displayed an activated and memory phenotype, and killed both mesothelin-positive (via CAR) and -negative (via NK receptor) pancreatic cell lines. CAR-NKT also exhibited superior tumor distribution and efficacy in orthotopic and metastatic pancreatic tumor models, effectively treated mixed-antigen tumors, and showed lower risk for GvHD and CRS than CAR T.

Contributed by Alex Najibi

Thrombospondin-1-CD47 signaling contributes to the development of T cell exhaustion in cancer

Weng et al. focused on unexplored mechanisms that contribute to T cell exhaustion. They observed that CD47, upregulated on human and murine tumor-infiltrating exhausted CD8+ T cells, bound matrix protein thrombospondin (TSP-1) to promote exhaustion. Mechanistically, TSP-1–CD47 binding activated calcineurin-NFAT signaling, resulting in TOX induction, inhibitory receptor expression (PD-1, TIM-3 and LAG-3) and decreased effector levels (TNFα and IFNγ), inducing CD8+ T cell exhaustion and tumor progression. Disrupting the TSP-1–CD47 interaction prevented T cell exhaustion, restored T cell function, and enhanced anti-PD-1 therapy.

Contributed by Katherine Turner

Tumour-reactive heterotypic CD8 T cell clusters from clinical samples

Ibanez-Molero and Veldman et al. isolated heterotypic clusters of CD8+ T cells with tumor cells and antigen-presenting cells (APCs) from melanoma metastases, revealing stable conjugates normally excluded by single-cell gating. These clustered T cells showed enriched tumor-reactive and exhausted signatures, increased TCR clonality, and binding to specific APC and tumor subtypes expressing synapse-forming ligands. Cluster-derived T cells produced higher IFNγ/TNF and showed greater cytotoxicity against autologous melanoma ex vivo. In matched PDX models, adoptive transfer of cluster-derived T cells showed higher infiltration, activation, and tumor control.

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.