Weekly Digests
‹ Back to November

Targeting the sialoglycan-Siglec axis for cancer immunotherapy

November 16, 2022

In cancer, upregulation of sialic acid-containing glycans (hypersialylation) is a feature that has been shown to drive disease progression and immune escape through engagement with Siglec receptors and inhibition of tumor-infiltrating immune cells. In previous work, Stanczak et al. showed that genetic desialylation or treatment with sialidase could reduce tumor growth and contribute to antitumor immunity. In work recently published in Science Translational Medicine, Stanczak et al. dug further into the details of this mechanism and explored strategies to exploit it for cancer immunotherapy.

To begin, Stanczak et al. used data from The Cancer Genome Atlas (TCGA) to identify a gene signature of sialoglycan genes that positively correlated with regulation of immune function, reduced cytokine activity, and shorter patient survival in several cancer types, most notably, clear cell kidney cancer (KIRC) and squamous cell lung cancer (LUSC). This gene set was also strongly correlated with the presence of immunosuppressive and pro-tumor immune cells (including Tregs and TAMs), reduced CD4+ T cells, and CD8+ T cell dysfunction. These results were validated in patient samples, where Siglec-9 Fc binding, a measure of surface sialylated glycans, was associated with worse survival (in KIRC), and treatment with sialidase reduced T cell activation (in LUSC).

Moving to mouse models, the researchers utilized MC38 tumors with genetic knockout of GNE, the rate-limiting enzyme for sialic acid biosynthesis. As in previous work, growth of GNE-KO MC38 tumors was delayed and mouse survival was increased compared to wild-type MC38 – an effect that was dependent on CD8+ T cells and could be enhanced with anti-PD-1 and anti-CTLA-4 (ICB). In a repeat experiment in which TILs were isolated on day 7, treatment with ICB alone increased the frequency of IFNγ+ and IFNγ+TNFα+ CD8+ T cells in both WT and GNE-KO tumors, but only induced IFNγ+TNFα+IL-2+ CD8+ T cells in the GNE-KO, suggesting that desialylation could enhance T cell activation in the context of ICB. Similar results were observed using other tumor cell lines and mouse strains.

Next, the researchers evaluated therapeutic desialylation using an an antibody–sialidase fusion product, E-301, comprising a HER2-targeting antibody (trastuzumab) and two sialidase domains fused to the c-terminal Fc region of the antibody. In vitro, E-301 induced dose-dependent desialylation of EMT6-HER2 mammary carcinoma cells. Similarly, in mice bearing established EMT6-HER2 tumors, a single intraperitoneal dose induced desialylation that was most pronounced at 24 hours, and still evident at 72 hours. At this same time point, the tumors also showed a loss of Siglec ligands, further validating the results.

Looking more closely at the effects of desialylation on immune cells, the researchers implanted mice with wild-type and GNE-KO EMT6-HER2 tumors on opposing flanks. The GNE-KO tumors showed reduced sialylation, which could be reduced slightly further with E-301. Treatment with E-301 induced desialylation of Infiltrating immune cells equally on both sides. This effect was observed across a broad array of immune cells, including TAMs, DCs, PMN-MDSCs, Tregs, and CD8+ T cells, but not M-MDSCs, conventional CD4+ T cells, or NK cells.

Stanczak et al. also found that in mice bearing B16D5-HER2 melanoma tumors, treatment with E-301 induced a number of changes in immune infiltrates, with treatment increasing absolute numbers of CD45+ and CD8+ T cells, DC activation, antitumor NK cell subsets, effector CD8+ T cells, and both Th1 and Th2 CD4+ T cells, while reducing naive CD4+ T cells and exhausted CD8+ T cells. Macrophages showed particularly dramatic changes, demonstrating a shift from a more immunosuppressive phenotype to a more pro-inflammatory phenotype, with increased signs of migration, responsiveness to chemokines, and production of effector molecules and pro-inflammatory cytokines. The researchers also noted an increase in M1-like and decrease in M2-like macrophages, altering the M1:M2 ratio. Similar immune changes were observed in the E-301-treated EMT6-HER2 tumor model, and in ICB-treated GNE-KO tumors.

Looking more closely at changes in TAMs, the researchers cocultured tumor-derived TAMs with naive CD8+ T cells and found that T cells cocultured with TAMs from genetically or therapeutically desialylated tumors were more proliferative and activated. Further, when desialylated tumor cells were cocultured with bone marrow-derived macrophages (BMDMs), the BMDMs expressed higher levels of TNFα and an increase in phagocytic activity. These results are in line with sialic acid acting as a “don’t eat me” signal, and suggest that high levels of sialic acid on tumor cells promotes the polarization of TAMs towards an immunosuppressive, pro-tumorigenic phenotype.

Investigating how desialylation can repolarize macrophages, Stanczak et al. showed that among common inhibitory CD33-related Siglecs, Siglec-E was most the most broadly expressed on TAMs, and had the highest expression on anti-tumorigenic TAMs, which were increased in E-301-treated tumors. In Siglec-E-KO mice, GNE-KO tumors grew as well as wild-type, and treatment of various tumors with E-301 was ineffective. Similar results were observed in mice depleted of TAMs via treatment with an anti-CSF1R antibody and in mice with Siglec-E conditionally knocked out on CD11c+ cells – a population that included TAMs, DCs and PMN-MDSCs, but mainly represented TAMs (Siglec-E on DCs showed minimal contributions to the effects). In the Siglec-E conditional knockout model, TAMs showed reduced M2 polarization. In vitro, Siglec-E-KO TAMs did not repolarize or increase activation of cocultured T cells upon treatment with E-301.

Evaluating the potential efficacy of E-301 as an immunotherapy, the researchers showed that in mice bearing orthotopic EMT6-HER2 tumors, 4 doses of E-301 delayed tumor growth and improved survival, with 1 of the 12 mice rejecting the tumor entirely. When this mouse was rechallenged with EMT6 and EMT6-HER2 tumors on opposing flanks, both tumors were rejected, suggesting strong immune memory and induction of response beyond HER2. Similar results were observed in the B16D5-HER2 model, where treatment with E-301 reduced tumor growth dependent on CD8+ T cells and synergized with ICB. Anti-PD-1 and ICB were also more effective in Siglec-E-KO mice, with 14 out of 18 (78%) of Siglec-E-KO mice rejecting tumors, compared to 4 out of 15 (27%) wild-type mice.

Overall, these results suggest that hypersialylation on tumor cells promotes polarization of TAMs towards an immunosuppressive phenotype through interactions with Siglec-E. They also support the sialoglycan-Siglec axis as a target for immunotherapy, as therapeutic desialylation can promote the repolarization of TAMs and support strong antitumor immune responses, particularly in the setting of ICB.

Write-up and image by Lauren Hitchings.

Meet the researcher

This week, first author Michal A. Stanczak and lead author Heinz Läubli answered our questions.

Michal A. Stanczak on the left and Heinz Läubli on the right.

What was the most surprising finding of this study for you?
HL: The most surprising finding of the study was the robustness of the results across multiple experimental models. Removing sialic acids led to improved antitumor immunity, be it by genetic means or using the enzymatic approach.
MAS: For me, the simplicity of the mechanism has been most surprising. We’ve been thinking and speculating about the incredible heterogeneity of glycans and the potential for functional redundancy of Siglec receptors for a long time. And while we can’t exclude that some of that plays a role, the expression of Siglec-E specifically on TAMs turned out to be the single dominant mechanism conferring the immunosuppressive effects of tumor sialylation in our hands.

What is the outlook?
HL
and MAS: A whole range of basic and preclinical research supports tumor glycosylation as a potential target for cancer immunotherapy. The next big steps for the field will be the clinical testing of that potential. Early clinical studies delivering human sialidase to patients with cancer are currently ongoing (GLIMMER-01 study). While it remains to be seen if this will ultimately turn out to be the best approach, future investigations will further aim at the exact intracellular events in myeloid cells following therapeutic interference and will compare the efficacies of the different approaches to block sialic acid–Siglec interactions, be it by blocking Siglec receptors or directly targeting sialic acids.

What was the coolest thing you’ve learned (about) recently outside of work?
HL: The coolest thing I have learned recently is that my 9-year-old son can already play better ice hockey than me….
MAS: I recently got into making my own pasta from scratch. Took a bit of experimentation, not unlike the work in the lab, but the results are much tastier.

References:

Stanczak MA, Rodrigues Mantuano N, Kirchhammer N, Sanin DE, Jacob F, Coelho R, Everest-Dass AV, Wang J, Trefny MP, Monaco G, Bärenwaldt A, Gray MA, Petrone A, Kashyap AS, Glatz K, Kasenda B, Normington K, Broderick J, Peng L, Pearce OMT, Pearce EL, Bertozzi CR, Zippelius A, Läubli H. Targeting cancer glycosylation repolarizes tumor-associated macrophages allowing effective immune checkpoint blockade. Sci Transl Med. 2022 Nov 2. 

In the Spotlight...

LRRC15+ myofibroblasts dictate the stromal setpoint to suppress tumour immunity

Using a Dptki/kiTgfbr2fl/fl PDAC GEMM, Krishnamurty et al. provided direct genetic evidence that DPT+ universal fibroblasts give rise to LRRC15+ CAFs in a TGFBR2-dependent manner during tumorigenesis, constituting a central fibroblast axis in multiple human cancers. Selective depletion of LRRC15+ CAFs reverted this stromal compartment to a universal fibroblast-like state and significantly delayed pancreatic tumor growth in CD8+ T cell-dependent manner. LRRC15+ CAF depletion enhanced intratumoural CD8+ T cell effector function and potentiated antitumor immunity in response to anti-PD-L1 checkpoint blockade.

Contributed by Shishir Pant

A split, conditionally active mimetic of IL-2 reduces the toxicity of systemic cytokine therapy

To provide more specificity to the site of action of the broadly stimulating cytokine IL-2, Quijano-Rubio et al. re-engineered the 4-helix-bundle IL-2 mimetic Neoleukin-2/15 (a fusion of IL-2 and IL-15) into separate 3- and 1-helix fragments (split Neo-2/15), which can associate to restore activity. Targeting each fragment to tumor cell surface receptors (e.g., HER2 and EGFR separately, or both to PD-L1) using binding domains restored trans activity locally and enhanced tumor control in vivo. Split Neo-2/15 targeting of CD8 stimulated T cells in cis and slowed growth of B16 melanoma. Targeting to two surface receptors on CD19-CAR T cells enhanced tumor control and overall survival.

Contributed by Ed Fritsch

Differential expression of CCR8 in tumors versus normal tissue allows specific depletion of tumor-infiltrating T regulatory cells by GS-1811, a novel Fc-optimized anti-CCR8 antibody

Based on TCGA data for expression of surface targets on regulatory T cells, which identified CCR8 as a preferentially expressed target in tumor Tregs, Weaver and Stack et al. demonstrated that treatment of multiple murine tumors with anti-CCR8 antibodies enhanced tumor control and synergized with anti-PD-1. Activity depended on an ADCC-active Ig-Fc for Treg depletion. Gene expression and IHC analyses of treated tumors demonstrated hallmarks of a remodeled antitumor TME. A humanized anti-CCR8 mAb was active in killing Treg cells in human in vitro assays and is being tested in an ongoing clinical trial.

Contributed by Ed Fritsch

Isolating and targeting a highly active, stochastic dendritic cell subpopulation for improved immune responses

Using an assay to identify DCs that rapidly and preferentially phagocytosed 2ⲙM macroparticles (MP; bacteria-sized), Deak et al. identified a small subpopulation of CD11b+SIRPα+ cDC2-related DCs, “first responders” (FR). Coating the MP with TLR agonists led to rapid release of TNFα (stored in vesicles) and upregulation of IFNβ production from these FRs. FR stimulation was transient (over hours) and led to production of other mediators that stimulated secondary DC activity. Liposomes targeting DAP12 and PRG2, which were relatively uniquely co-expressed in FRs, demonstrated promise for in vivo delivery and enhancement of adaptive immunity.

Contributed by Ed Fritsch

A novel polymer-conjugated human IL-15 improves efficacy of CD19-targeted CAR-T cell immunotherapy

To improve the success of IL-15 supplementation for CAR T cell therapy, Hirayama and Chou et al. evaluated a novel pegylated polymer-conjugated IL-15 (NKTR-255) with improved pharmacokinetic and pharmacodynamic properties. In non-human primates, NKTR-255 enhanced T (CD8+ and CD4+) and NK cell proliferation in vitro and induced dose-dependent human CD19 CAR T proliferation, even at low target levels. In a murine lymphoma model, the addition of NKTR-255 resulted in superior human CD19 CAR T efficacy and robust and sustained persistence of functional CAR T cells, compared with CAR T cell therapy alone.

Contributed by Katherine Turner

Tetramer-aided sorting and single-cell RNA sequencing facilitate transcriptional profiling of antigen-specific CD8+ T cells

Rajasekara and Guan et al. showed that CMV pp65 tetramer-aided FACS sorting enriched even rare antigen-specific CD8+ T cells, with minimal impact on the relative clonal distribution of the total CD8+ T cell population, the phenotype distribution in individual TCR clones and their transcriptomes. Longer duration pp65 tetramer-mediated CD8+ T cell activation induced the conventional CD8+ T effector and also identified an antigen-specific response gene signature (ASR) that contained genes associated with antigen recognition and metabolic regulation. The ASR added value in predicting PFS in atezolizumab + chemotherapy-treated patients with NSCLC.

Contributed by Paula Hochman

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.