Weekly Digests
‹ Back to October

A type 2 cytokine fusion with type 1 effects – using IL-4 to enhance antitumor immunity

October 9, 2024

Interleukin 4 (IL-4), a cytokine typically associated with humoral (type 2) immune responses, has recently been shown to promote survival in T and B cells. Studying whether IL-4 could be exploited to enhance cancer immunotherapy, Feng and Bai et al. developed a fusion protein of IL-4 and a mutant IgG2a antibody (Fc–IL-4), and tested its effects across various settings. Their results, recently published in Nature, showed that Fc–IL-4 induced direct metabolic reprogramming in terminally exhausted CD8+ T cells, reinvigorating their functionality and enhancing antitumor immunity, especially in combination with existing immunotherapies.

To begin, the researchers evaluated Fc–IL-4 in the context of mice bearing B16F10 tumors (expressing gp100) and treated with adoptive cell transfer (ACT) of PMEL T cells (targeting gp100). Here, the addition of Fc–IL-4 increased both PMEL and endogenous T cells in tumors, and enhanced their polyfunctionality and production of granzyme B. Among PMEL T cells, the PD-1+Tim3+ (TTE) subset was enriched. These effects were restricted to tumor antigen-specific T cells, as they were not observed for OT-1 cells transferred to non-OVA-expressing tumors.

Investigating changes to immune cells in tumors, the researchers performed scRNAseq and sorted tumor antigen-specific Thy1.1+CD8+ TILs from tumors from ACT- and Fc–IL-4-treated mice. Several of the clusters that emerged resembled TTE cells, with high expression of co-inhibitory molecules, while 1 showed attenuated expression of these markers. After Fc–IL-4 treatment, the more TTE-like clusters increased, while the less exhausted cluster was limited. Expression of Ifng, Gzmb, Prf1, and Nkg7 were also higher in treated samples, suggesting enhanced cytotoxic activity.

Next, the researchers evaluated whether Fc–IL-4 could enhance antitumor immunity, and found that across numerous murine tumor models, the addition of Fc–IL-4 to ACT significantly improved antitumor responses, leading to durable cures and protection from rechallenge. Similar results were observed using a human Fc–IL-4 in xenograft mouse models treated with human CAR T cells, including a model of recurrent leukemia. In some models, Fc–IL-4 showed antitumor activity on its own, suggesting that it supports endogenous antitumor immune responses as well. Lymphodepletion was not required for antitumor efficacy, and no overt toxicities associated with Fc–IL-4 were observed. Fc–IL-4 also enhanced the antitumor efficacy of immune checkpoint blockade (ICB; anti-PD-1 + anti-CTLA-4) in an MC38 tumor model, where it led to clearance of established tumors and protection from rechallenge.

Selective depletion studies revealed that CD8+ T cells were essential to the antitumor efficacy of Fc–IL-4, while progenitor exhausted T cells were dispensable. By transferring different T cell subsets for ACT, the researchers found that only mice treated with PD-1+TIM-3+ CD8+ TTE cells showed enhanced enrichment and effector functions in response to Fc–IL-4, suggesting Fc–IL-4 acts mainly on this subset. In line with this, CD8+ TTE cells expressed the highest levels of IL-4Rɑ among all evaluated subsets, and IL-4Rɑ-KO CD8+ TTE cells failed to respond to Fc–IL-4, suggesting that Fc–IL-4 acts through direct signaling on CD8+ TTE cells. The use of FTY720 to block T cell egress from lymph nodes had little impact on the effects of Fc–IL-4, suggesting that it acts primarily on cells within tumors, rather than on cells recruited from the periphery.

The researchers found that while neutralizing IL-4 in tumor models showed little impact, the addition of Fc–IL-4 promoted survival in CD8+ TTE cells through upregulation of Bcl-2 and mitigation of apoptosis. Based on previous research showing that IL-4 promoted glycolysis in B cells, the researchers next evaluated whether the same might be true in T cells. Indeed, they found that in ex vivo-induced CD8+ TTE cells, Fc–IL-4 increased Glut1 expression, glucose uptake capacity, and extracellular lactate, and upon TCR stimulation, enhanced the extracellular acidification rate (ECAR); oxidative phosphorylation was not impacted. Fc–IL-4 also induced alterations in 41 metabolites, including upregulation of 3 associated with glycolysis. Analysis of metabolic genes in PMEL T cells treated with Fc-IL-4 showed enrichment for cells characterized by the glycolysis–gluconeogenesis gene module. Further, blockade of glycolysis abrogated the effects of Fc–IL-4 on CD8+ TTE cells, suggesting that promoting glycolysis is critical to treatment-enhanced antitumor immunity.

Digging deeper into the molecular mechanisms behind the changes induced by Fc–IL-4, Feng and Bai et al. performed ATACseq and transcriptome co-profiling, revealing distinct molecular profiles between treated and untreated CD8+ TTE cells, with treated cells showing changes in cytotoxicity and survival markers. Differential motif analysis of ATACseq data showed that motifs for Stat6 transcription factor binding were most enhanced, while motifs for Foxo1 transcription factor (a negative regulator of mTOR) binding were most reduced. In line with this, analysis of signaling pathways regulated by differentially expressed genes revealed that Fc–IL-4 was associated with significant upregulation of mTOR, eIF4, p70S6K, PI3K/AKT, JAK–STAT signaling, NF-κB activation, and glycolysis. Upstream regulator analysis also predicted upregulation of NF-κB, Myc, Pi3k, Akt1, and Stat6, which were found to promote glycolysis. Knockout of STAT6 partially attenuated the efficacy of Fc-IL-4 in an ACT model, and the addition of blockades for AKT or mTOR fully abrogated its benefits, pointing to a role for both STAT6 and PI3K-AKT-mTOR signaling in Fc–IL-4-mediated antitumor immunity.

To identify enzymes involved in Fc–IL-4-induced glycolysis, the researchers found that LDHA showed the most pronounced upregulation after treatment, dependent on STAT6 and PI3K-AKT-mTOR signaling, and played a critical role in Fc-IL-4-induced responses. Given that LDHA facilitates NAD+ production for glycolysis, the researchers evaluated NAD+ levels. While NAD+ was generally reduced in CD8+ TTE cells compared to progenitor exhausted T cells, treatment with Fc–IL-4 increased NAD+ in ex vivo-stimulated CD8+ TTE cells, dependent on LDHA. Increases in metabolites involved in nicotinate and nicotinamide metabolic pathways were also observed after treatment, and supplementing cells with nicotinamide riboside (a NAD+ precursor) showed similar results to Fc–IL-4, suggesting that treatment increased NAD+ generation.

Together, these results suggest that Fc–IL-4 can direct enrich and enhance the antitumor immune functions of tumor-infiltrating antigen-specific CD8+ TTE cells by binding directly to IL-4Rɑ, triggering STAT6 and PI3K-AKT-mTOR signaling. This, in turn, mediates increased glucose uptake and NAD+ concentration, which enhances glycolysis, dependent on LADH, and improves the survival and effector functions of CD8+ TTE cells. In mice, these effects enhanced antitumor immunity alone and in combination with existing ACT and ICB immunotherapies, suggesting strong potential for future use.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, co-first author Bing Feng answered our questions.

From left to right: Weilin Li, a PhD student; Tom Enbar, PhD student; Li Tang, Associate Professor of Bioengineering at EPFL; Lucia Bonati, PhD student.


What was the most surprising finding of this study for you?
Unlike a type 1 immune response, which has traditionally been the target of cancer therapies like immune checkpoint inhibitors and CAR T cells, type 2 responses are mobilized to fight parasitic immune threats, like worms. Until now, researchers thought that type 2 immune factors were not useful in fighting cancer, and could even promote tumor growth. In this study, surprisingly, we found IL-4, a type 2 immune cytokine, could enhance current immunotherapies, which are all type 1-centric, leading to enduring antitumor immunity and long-term survival in animal models. Our results show that type 1 and type 2 immunity can be thought of in terms of synergy, like yin and yang. This discovery represents a major paradigm change in the field of cancer immunotherapy, from type 1 centric to a synergy of type 1 and type 2 immune responses, therefore is critical for designing next-generation cancer immunotherapy with improved patient responses.

Specifically, we show that IL-4 signals can potently enhance IFN-γ secretion and boost cytotoxicity of functionally impaired CD8+ T cells, particularly those in a terminally exhausted state. Another unexpected part is that STAT6, not STAT5, was crucial for IL-4's role of promoting CD8+ T cell effector function, despite IL-4 being a γc cytokine typically associated with STAT5.

What is the outlook?
Our findings suggest that IL-4 could synergize with existing cancer immunotherapies, including CAR T cell and immune checkpoint blockade (ICB) therapy. We next aim to translate this discovery to clinically applicable therapies. Two major strategies are under development. One is to use combinatory therapies involving Fc-IL-4 (a long-circulating cytokine) in combination with CAR T cells or ICBs, and the other is to develop IL-4-secreting CAR T cells. We have accumulated successful prior experience in clinical studies in IL-10-expressing CAR T cells (Nat. Biotech. 2024; Nat. Immunol. 2021), which are being tested in several ongoing first-in-human investigator-initiated clinical trials (ClinicalTrials.gov IDs: NCT06393335, NCT05715606, NCT05747157, NCT06120166, NCT06277011). The Preliminary findings have been reported in AACR 2024, EHA 2024, ASGCT 2024, SOHO 2024, etc. We are confident that the new discovery of the role of IL-4 in cancer will likely make some impacts in the clinic.

Moving forward, we are also interested in investigating other type 2 immune factors, including various cytokines and immune cells, to further develop next-generation cancer immunotherapies.

What was the coolest thing you’ve learned (about) recently outside of work?
The coolest thing is that I found my soulmate and life partner recently, and we are getting married soon…

References:

Feng B, Bai Z, Zhou X, Zhao Y, Xie YQ, Huang X, Liu Y, Enbar T, Li R, Wang Y, Gao M, Bonati L, Peng MW, Li W, Tao B, Charmoy M, Held W, Melenhorst JJ, Fan R, Guo Y, Tang L. The type 2 cytokine Fc-IL-4 revitalizes exhausted CD8+ T cells against cancer. Nature. 2024 Sep 25.

In the Spotlight...

Crosslinking of Ly6a metabolically reprograms CD8 T cells for cancer immunotherapy

Maliah et al. showed that mice receiving chronic UVB exposure prior to s.c. or i.v. injection of melanoma cells exhibited enhanced growth of local tumors, but not metastases; reduced skin-draining LN, but not splenic CD8+ T cell antitumor activity; and resistance to anti-PD-1 treatment. UVB-induced immunosuppression was mediated by a Ly6ahi T cell subset that was shown to be upregulated by type I IFNs secreted by DCs in the inflammatory TME. Anti-Ly6a Abs signaled T cells via cMyc to prevent loss of mitochondrial metabolism and boost the T cell response to tumor cells, even in mice resistant to anti-PD-1 treatment.

Contributed by Paula Hochman

Spatial analysis reveals targetable macrophage-mediated mechanisms of immune evasion in hepatocellular carcinoma minimal residual disease

Using post-chemoembolization residual human hepatocellular carcinoma (HCC), Lemaitre, Adeniji, and Suresh et al. identified a spatial neighborhood of PD-L1+ M2-like macrophages and stem-like tumor cells that correlated with CD8+ T cell exhaustion and poor survival. Spatial transcriptomics showed that macrophage-derived TGFβ1 mediated the persistence of stem-like tumor cells. In a transgenic mouse model of HCC, recurrences arose from stem-like minimal residual disease. Combined blockade of PD-L1 and TGFβ excluded immunosuppressive macrophages, recruited activated CD8+ T cells, and eliminated residual stem-like tumor cells in mouse models of HCC.

Contributed by Shishir Pant

Preclinical Evaluation of AZD6422, an Armored Chimeric Antigen Receptor T Cell Targeting CLDN18.2 in Gastric, Pancreatic, and Esophageal Cancer

Barrett et al designed a CLDN18.2 CAR T cell product incorporating a CLDN18.2 binding domain (selected on the basis of cytotoxicity, despite its low affinity), a dnTGFβ receptor, and a shortened expansion process to enhance efficacy in solid tumors. A CAR containing CD28 was safe and effective in a mouse xenograft model, while 4-1BB led to severe on-target/off-tumor toxicity. TGFβ armoring promoted complete tumor regression and sustained IFNγ responses, and short manufacturing promoted a TCM phenotype and improved cytotoxicity upon serial restimulation. The final product demonstrated efficacy across six PDX models with varying CLDN18.2 and TGFβ levels.

Contributed by Morgan Janes

Engineered allogeneic T cells decoupling T-cell-receptor and CD3 signalling enhance the antitumour activity of bispecific antibodies

Kapetanovic et al. used targeted genomic mutagenesis of the FGxGT motif in the TRAJ region to develop Allogeneic-Engineered Decoupled (AED) T cells, for which TCR–antigen binding was functionally decoupled from CD3 signaling. AED T cells remained unresponsive to cognate antigen stimulation. When coadministered with the bispecific CD3/CD19 antibody, blinatumomab, primary AED T cells recognized and cleared human CD19+ tumor cells in a xenograft mouse model, without signs of alloreactivity, despite the presence of cognate antigen. AED T cells were compatible with other bispecific T cell engagers.

Contributed by Ute Burkhardt

Deficiency of metabolic regulator PKM2 activates the pentose phosphate pathway and generates TCF1+ progenitor CD8+ T cells to improve immunotherapy

Markowitz et al. studied metabolic mechanisms regulating TCF1high progenitor CD8+ T cells, which are critical for immunotherapy efficacy. By comparing metabolic pathway genes in tumors that respond differentially to PD-1-targeted therapies, they found that knockdown of glycolytic pyruvate kinase muscle 2 (PKM2) in CD8+ T cells resulted in decreased glycolytic flux, increased pentose phosphate pathway (PPP) activity, and enrichment of a TCF1high progenitor exhausted-like phenotype with increased responsiveness to PD-1 blockade in vivo. A small molecule PPP agonist phenocopied the effects of PKM2 KO, and enhanced the efficacy of T cells in a patient-derived human organoid system.

Contributed by Katherine Turner

Spatial single-cell protein landscape reveals vimentin(high) macrophages as immune-suppressive in the microenvironment of hepatocellular carcinoma

Qiu et al. established a detailed spatial map of single-cell phenotypes and multicellular neighborhoods of tumor tissues from 401 patients with hepatocellular carcinoma. The spatial single-cell analysis identified three spatial patterns (SP-HI, -LI, and -PF) with distinct prognostic, genomic, and molecular features. An integrated analysis combining CODEX, scRNAseq, and spatial transcriptomics datasets identified that co-localization of VIMhigh macrophages and Tregs correlated with tumor progression in HCC. In vitro functional studies showed that VIMhigh macrophages secreted IL-1β to promote the immune-suppressive function of Tregs.

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.