Weekly Digests
‹ Back to April

Unraveling the role of soluble CTLA-4

April 16, 2025

CTLA-4 is a well established immune checkpoint with a key a role in limiting autoimmunity, but it exists in both membrane-bound (mCTLA-4) and soluble (sCTLA-4) forms, and the different sources, kinetics, and functions of sCTLA-4 in particular are not fully understood. In research recently published in Immunity, Osaki and Sakaguchi explored numerous aspects of sCTLA-4, identified Tregs as its primary source, and uncovered its role in suppressing type 1 immunity while allowing for maintenance of type 2 immunity in settings of chronic inflammation.

To begin, the researchers evaluated expression of sCTLA-4 in normal mice, and found that compared to conventional CD4+ T cells, Tregs showed increased expression of sCTLA-4, which was further increased in activated/effector Tregs, outside of lymphoid organs, and in chronic inflammatory settings, including colitis and tumors. TCR activation of Tregs initially reduced sCTLA-4 expression and instead increased mCTLA-4 expression, but over time, sCTLA-4 expression was restored and increased to above baseline, staying elevated for at least 3 weeks.

Investigating the role of sCTLA-4, the researchers generated a mouse model that was deficient in sCTLA-4, but maintained mCTLA-4 (S-M+). While these mice grew and survived normally over 30 weeks, they showed enlarged ectopic lymphoid structures in the adipose tissues, increased leukocyte infiltration into lungs at 30 weeks, and higher concentrations of serum antibodies (including IgEs), IL-6, and TNFα than wild-type S+M+ mice. There was also an increase in effector memory CD8+ and CD4+ T cells (including those secreting IFNγ), follicular helper T cells (including those secreting IL-4), IL-17+ CD4+ T cells, and germinal center B cells, and a decrease in Tregs. Macrophages expressed higher levels of M1 markers and lower levels of M2 markers. DCs were predominantly cDC1/classical conventional DCs, which are known to contribute to Th1 and Th17 induction. These results suggested that sCTLA-4 deficiency supported cDC1 differentiation to evoke type 1 immune responses, marked by Th1, Th17, Tfh, and Tc1 differentiation in CD4+ T cells, an M1-like phenotype in macrophages, increased antibody production, and a high potential for autoimmunity.

Next, the researchers generated a mouse model that expressed sCTLA-4 (at relatively high levels), but not mCTLA-4 (S+M-). While S-M- mice quickly developed fatal systemic immunity, S+M- mice were less debilitated and lived longer, but still showed signs of autoimmunity. While both S+M- and S-M- mice showed an increase in activated conventional CD4+ and CD8+ T cells relative to wild-type mice, Th1 and Th17 cells were lower in the mesenteric LNs of S+M- mice compared to S-M- mice. Further, S+M- mice showed increased concentrations in the serum of Th2 cytokines IL-4, IL-5, IL-10, M-CSF, but lower concentrations of IL-6 and IL-17A compared to S-M- mice. These results suggested that the presence of sCTLA-4 alleviated severe autoimmunity by reducing type 1 immunity, without disrupting type 2 immunity.

Osaki and Sakaguchi hypothesized that sCTLA-4 exerts immunosuppressive effects through its interactions with immune cells expressing CD80/CD86, such as CD11b+ cells (monocytes/macrophages/granulocytes), CD11c+ DCs, or B220+ B cells. Evaluating these immune populations in 3-week-old S+M- and S+M+ (wild-type) mice, the researchers found that while S-M- mice showed a notable increase in the proportion of CD80+CD11b+ cells among splenic CD11b+ cells (compared to S+M+ mice), there was no significant increase in S+M- mice. Cytokine analysis showed that splenic CD11b+ cells in S+M- mice expressed high levels of IL-4, while S-M- mice expressed high levels of IL-6. Further, macrophages from S+M- mice showed increased production of IL-10, expression of M2 markers, and genes related to tissue remodeling, and decreased genes related to the Th17 pathway compared to those from S-M- mice, which expressed higher levels of M1 markers.

Despite these phenotypic differences in macrophages from S+M- or S-M- mice, the expression levels of cytokine receptors were similar. Therefore, the researchers evaluated whether their differences could be attributed to differential exposure to Th2 cytokines. They found that treating macrophages with IL-4 and/or IL-10 increased CD206 and M2 marker expression and suppressed M1 marker expression, mimicking observations in S+M- mice. However, direct treatment of macrophages with sCTLA-4 did not have the same effect, suggesting that sCTLA-4 induces cytokine production by another immune cell type. They further found that in S+M- mice (compared to S-M- mice), DCs were more polarized towards cDC2s, and the frequencies of IL-4- and/or IL-5-producing circulating CD4+ T cells were increased, which appeared to drive the activation and maturation of eosinophils, increasing their expression of siglec-F and IL-4. Additionally, peritoneal macrophages expressed higher levels of eotaxins and CCL22, which were also increased in serum.

Investigating whether sCTLA-4 facilitates Th2 differentiation through interactions with CD80/CD86, the researchers evaluated the effects of a CTLA-4-Ig (positive control), sCTLA-4, and a mutant sCTLA-4 that lacks the binding domain for CD80/CD86 (Y139 sCTLA-4). In in vitro activation and differentiation assays, CTLA-4-Ig and sCTLA-4 both inhibited the proliferation of naive T cells and differentiation towards Th1, while Y139A sCTLA-4 did not. When IL-4 was added to induce Th2 conditions, sCTLA-4 allowed for Th2 differentiation, even at concentrations that suppressed Th1 differentiation. Similar results were observed with blockade of CD80, CD86, or both, with or without the addition of IL-4. These results suggest that sCTLA-4 blocks CD80/CD86 on APCs, limiting their capacity to activate T cells and induce Th1 differentiation, but not IL-4-induced Th2 differentiation.

Finally, Osaki and Sakaguchi examined the functions of sCTLA-4 in various murine disease models. They found that in an induced colitis model, both sCTLA-4 and mCTLA-4 protected mice from damage by inhibiting Th1/Th17 cell differentiation and expansion. Similarly, in an experimental asthma model, sCTLA-4 contributed to protection against more severe bronchoconstriction and peribronchiolar cellular infiltration, and in a wound healing model, sCTLA-4 contributed to proper wound healing. In murine cancer models. sCTLA-4 contributed to faster tumor growth and reduced infiltration of CD8+ T cells, particularly cytolytic CD8+ T cells, dependent on binding to CD80/CD86. It also contributed to lower IFNγ and a higher ratio of M2-like macrophages.

Overall, these results show that sCTLA-4 is produced predominantly by Tregs and plays a role in limiting autoimmunity though blocking CD80/86 on APCs, thus limiting the induction of type 1 immunity, while still allowing for the induction of type 2 immunity. However, in cancer settings, sCTLA-4 contributed to a less favorable TME and faster tumor growth, suggesting that it also limits antitumor immunity and could serve as a potential target, along with mCTLA-4, for cancer immunotherapy.

Write-up and image by Lauren Hitchings

References:

Osaki M, Sakaguchi S. Soluble CTLA-4 regulates immune homeostasis and promotes resolution of inflammation by suppressing type 1 but allowing type 2 immunity. Immunity. 2025 Apr 8.

In the Spotlight...

Cancer cell-derived arginine fuels polyamine biosynthesis in tumor-associated macrophages to promote immune evasion

Zhu et al. found that serum arginine increased during breast cancer progression and originated primarily from cancer epithelial cells, which expressed the arginine biosynthetic enzyme ASS1. Macrophage polyamines downstream of arginine signaling regulated TAM suppressive activity, and Ass1 KD in breast cancer cells inhibited tumor growth only in the presence of macrophages. Activity of spermine, a potent suppressive polyamine, was critically dependent on p53 signaling, which promoted demethylation of key genes, including PPARG by the enzyme TDG. In vivo, TDG KD inhibited TAM polarization and increased CD8+ T cell infiltration to promote tumor control.

Contributed by Morgan Janes

Spatial transcriptomics reveals tryptophan metabolism restricting maturation of intratumoral tertiary lymphoid structures

Tang et al. used single-cell spatial transcriptomics to comprehensively profile tertiary lymphoid structures (TLSs) under various maturation stages in hepatocellular carcinoma (HCC). Immature TLSs were classified as conforming or deviating, exhibiting distinct molecular features and response to ICB therapy. Conforming TLSs, like mature TLSs, were associated with response to ICB. Tumor cell-driven tryptophan metabolism in the TME influenced the deviation of TLS maturation. Inhibiting tryptophan metabolism promoted intratumoral TLS maturation, enhanced tumor control, and synergized with anti-PD-1 therapy in an orthotopic HCC mouse model.

Contributed by Shishir Pant

DDX54 downregulation enhances anti-PD1 therapy in immune-desert lung tumors with high tumor mutational burden

Gonga and Lee et al. focused on mechanisms of ICI resistance in immune-desert lung tumors that lack T cell infiltration, despite having a high mutational burden (TMB-H), by analyzing gene regulatory networks in TMB-H lung tumors from TCGA. DDX54 was identified as a master regulator of immune-desert phenotypes via induction of immunosuppressive oncogenic Myc and Wnt pathways, EMT, cancer stemness, and CD38 and CD47 expression. In TMB-H syngeneic mouse lung models, DDX54 knockdown combined with anti-PD-1 increased immune cell infiltration (T cells, NK cells, M1 monocytes, and dendritic cells) and improved sensitivity to anti-PD-1.

Contributed by Katherine Turner

Anti-CTLA4 therapy leads to early expansion of a peripheral Th17 populaton and inducton of Th1 cytokines

Nakazawa et al. studied the peripheral immune profile of a cohort of patients with advanced solid tumors; 54 patients received anti-PD-(L)1 alone and 50 received anti-PD-(L)1 + anti-CTLA-4. The addition of anti-CTLA-4 was associated with a greater increase in peripheral blood CD4+ TH cell subsets, including Treg and Th17 cells, a higher Th17:Treg ratio, and an unexpected increase in Th17 cell expression of TBET, the key regulator of TH1 cell development. The plasma of recipients of combination therapy exhibited an increase in TH1-associated cytokines, particularly the IFNγ-inducible cytokines iTAC, MIG, and IP-10, but not in Th17, Th2, Treg, or myeloid cell cytokines.

Contributed by Paula Hochman

The local microenvironment suppresses the synergy between irradiation and anti-PD1 therapy in breast-to- brain metastasis

Wischnewski et al. demonstrated that CD8+ T cells infiltrated breast cancer brain metastases (BC-BrM), but failed to elicit an effective antitumor immune response, in contrast to genetically identical extracranial tumors. Brain irradiation transiently elevated the lymphoid-to-myeloid cell ratio in the BrM TME, and while it did not synergize with anti-PD-1 in the BrM, this combination did exhibit synergy in extracranial tumors, suggesting an immunosuppressive role of the brain-specific TME. Transcriptional and functional analyses identified neutrophils and TREM2-expressing macrophages as key mediators of local T cell suppression within the brain.

Contributed by Shishir Pant

The response to anti–PD-1 and anti–LAG-3 checkpoint blockade is associated with regulatory T cell reprogramming

Rolig and Peng et al. identified mouse tumor models with high (LAG3hi) or low (LAG3lo) frequencies of LAG3+ lymphocytes. While LAG3lo mice were sensitive to anti-PD-1, LAG3hi mice were anti-PD-1-resistant, but showed increased sensitivity with the addition of anti-LAG3; this effect was dependent on CD8+ T cells and was enhanced with the depletion of CD4+ T cells. Further, in LAG3hi mice, responses correlated with Treg phenotype plasticity, with improved responses in mice with expanded populations of unstable Tregs. A similar correlation was observed in anti-PD-1 + anti-LAG3-treated patients with metastatic melanoma.

Contributed by Lauren Hitchings

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.