Weekly Digests
‹ Back to September

Tired out T cells: aberrant glycosylation drives T cell exhaustion in early tumors

September 3, 2025

Changes in glycosylation are a hallmark of cancer, and recent work has suggested that branched N-glycans on T cells may play a role in limiting their antitumor activity. In recent work, Azevedo et al. investigated the T cell glycome within tumors to better understand whether and how expression of aberrant glycans on T cells affects antitumor functions over the course of tumor development. Their results were recently published in Cancer Immunology Research.

To characterize the dynamics of N-gylcosylation in tumor-infiltrating lymphocytes, Azevedo et al. selected serrated colorectal cancer (Ser CRC) as a primary model, due to its high mutation burden and strong immune infiltration. In analyses of Ser CRC samples from patients, the researchers observed increased expression of stromal β1,6-GlcNAc branched N-glycans (by L-PHA staining) starting in pre-malignant lesions and increasing with more advanced disease. This increase also tracked with increased infiltration of T cells and CD8+ T cells, loss of granzyme B, and increased expression of MGAT5 (encoding a glycosyltransferase involved in the formation of β1,6-GlcNAc branched N-glycans). Similar results were observed based on flow cytometry of fresh colonic biopsies from patients with Ser CRC, which showed increased CD8+ T cells and a trend towards increased β1,6-GlcNAc branched N-glycans on CD8+ T cells. In pre-malignant lesions, β1,6-GlcNAc branched N-glycans negatively correlated with IFNγ and granzyme B production. Another cohort of Ser CRC patient data showed a positive correlation between MGAT5 expression and expression of CTLA-4 and TIGIT. Similarly, a cohort of patients with Lynch Syndrome showed increases in T cells and MGAT5 expression with disease progression, and MGAT5 expression correlated with increased PD-1, CTLA-4, LAG3, and TIGIT.

Next, the researchers tested whether N-glycosylation might affect T cell effector functions or exhaustion. In an MC38-OVA tumor-bearing mouse model treated with OT-I cells, the researchers observed an emergence of exhausted (PD-1+Tim3+) and terminally exhausted (PD-1+Tim3+CD39+) T cells that coincided with increasing levels of β1,6-GlcNAc branched N-glycans on CD8+ T cells. In vitro modeling of acute and chronic stimulation under conditions of normoxia or hypoxia showed that chronic activation under hypoxia (mimicking a hostile TME) induced the highest levels of PD1+Tim3+ exhausted T cells and the highest expression of β1,6-GlcNAc branched N-glycans.

To further explore the relationship between β1,6-GlcNAc branched N-glycans and exhaustion, Azevedo et al. used CRISPR/Cas9 to knock out each of 3 glycosyltransferases – Mgat1, Mgat2, and Mgat5, involved in different stages in the formation of β1,6-GlcNAc branched N-glycans – in OT-I cells. Knockout of Mgat5 most prominently decreased β1,6- GlcNAc branched N-glycans and PD-1+Tim3+ exhausted cells following activation. Expression of Tcf1 (precursor exhaustion) and Tox (late exhaustion) were also reduced, with fewer cells exhibiting a Tbet+Eomes+ exhausted phenotype. Similar results were observed under conditions of chronic stimulation and hypoxia, supporting an early role for Mgat5 and β1,6-GlcNAc branched N-glycans in inducing T cell exhaustion.

Digging deeper, the research assessed the impact of β1,6-GlcNAc branched N-glycans on T cell fitness and metabolism and found that Mgat5 KO CD8+ T cells showed increased basal and maximal respiratory capacity, increased glycolysis and glycolytic capacity, and increased CD69 expression (indicative of activation). In line with this, naive T cells isolated from Mgat5 KO mice showed increased CD69, proliferation, TCR signaling, and production of IFNγ and TNFα. Similar results were observed in mice with Mgat1 or Mgat2 knocked out in T and B cells, but to a lesser extent.

Next, Azevedo et al. explored whether Mgat5 KO CD8+ T cells might show enhanced antitumor activity. Indeed, when co-cultured with MC38-OVA cells, these cells again showed enhanced activation and reduced exhaustion, along with enhanced tumor cell killing and cytolysis, including increased degranulation (CD107a). Similar results were observed in T cells from Rag2-/- OT-I Mgat5-/- mice, which showed enhanced tumor cell killing relative to those from Rag2-/- OT-I WT mice. Additionally, overexpression of Mgat5 in CD8+ OT-I T cells resulted in increased branched N-glycans and reduced tumor cell killing, further establishing the immunoregulatory role of Mgat5.

To test the potential of Mgat5 deletion for improving antitumor efficacy in vivo, the researchers transferred Mgat5 KO OT-I cells into MC38-OVA-bearing mice and found that these cells significantly suppressed tumor growth and improved overall survival compared to transfer of WT OT-I cells. Similarly, MC38 tumor growth was reduced in Rag2-/- OT-I Mgat5-/- mice versus Rag2-/- OT-I WT mice, with a trend towards longer survival. Isolated Mgat5 KO OT-I T cells isolated from tumors exhibited higher cytotoxic capacity upon ex vivo restimulation. These also showed a trend towards decreased exhaustion (PD-1+Tim3+ CD8+ T cells) and a significant decrease in terminal exhaustion (CD39+ CD8+ T cells). RNAseq further revealed an increase in expression of genes related to survival, effector functions, and cellular respiration/oxidative phosphorylation, and a decrease in expression of genes related to stemness and memory in T cells.

Moving towards a more clinically relevant model, Azevedo et al. isolated CD3+ T cells from human PBMCs, transduced them with CD19-CARs, and used CRISPR/Cas9 to knock out MGAT5. In coculture with A549 human lung cancer cells expressing CD19, these Glyco-CAR T cells showed increased degranulation (CD107a expression) in both CD4+ and CD8+ populations, and demonstrated enhanced tumor cell killing compared to standard CAR T cells. In NSG mice bearing established A549-hCD19 tumors, treatment with Glyco-CAR T cells induced tumor growth inhibition and prolonged survived over control CAR T cells.

Together, these results show that hostile TMEs drive aberrant glycosylation of T cells, mediated in part by Mgat5 expression, driving them towards exhaustion starting in early premalignant lesions. Deletion of Mgat5 in T cells reduced the presence of β1,6-GlcNAc branched N-glycans, enhanced their metabolic fitness, and increased T cell activation and effector functions, including degranulation and cytolytic antitumor activity. When applied to human CAR T cells, MGAT5 knockout enhanced their in vitro and in vivo efficacy against solid tumors, which could translate to enhanced clinical efficacy in the future.

Write-up and image by Lauren Hitchings

Meet the researcher

This week, lead author Salomé Pinho answered our questions.

What was the most surprising finding of this study for you?
T cell therapies such as CAR T cells are not yet effective in solid tumors, due to the complexity of tumor microenvironment. We here reveal that branched N-glycosylation emerges at the surface of infiltrating T cells in pre-malignancies, imposing an exhausted and dysfunctional phenotype. We targeted Mgat5-mediated branched N-glycans on CD8+ (CAR-) T cells as a strategy to prevent exhaustion, enhancing cytotoxicity and antitumor T cell activity.
We here revealed the power of glyco-engineering CD8+ T cells in boosting the efficacy of cellular immunotherapy. We demonstrated that by removing a specific glycogene, MGAT5, the surface glycocalyx of T cells is efficiently reprogrammed, preventing T cell exhaustion and increasing cytotoxicity and tumor killing.

What is the outlook?
These findings illuminate T cell glycoengineering as an innovative strategy to unlock T cell effectiveness in cancer immunotherapy, laying foundation for proposing glycoengineering of CAR T cells as an effective strategy to create a new generation of super Glyco-CAR T cells with superior efficacy in solid tumors.

Who or what has been a major source of inspiration or motivation for you throughout your career?
A major source of inspiration throughout my career has been the transformative power of science itself — not just as a tool to improve health, but as a driver of societal progress and economic development. What continues to motivate me is the understanding that scientific discoveries only reach their full potential when pursued collaboratively, across disciplines and across borders. Witnessing how research can lead to innovative therapies that save millions of lives, new technologies, and innovations that can transform society and the economy has been reinforcing my strong belief in the power of science as a collective endeavor. This sense of purpose — working together to turn knowledge into tangible benefits — has been my guiding motivation.

References:

Azevedo CM, Xie B, Gunn WG, Peralta RM, Dantas CS, Fernandes-Mendes H, Joshi S, Dean V, Almeida P, Wilfahrt D, Mendes N, Portero JL, Poves C, Fernández-Aceñero MJ, Marcos-Pinto R, Fernandes Â, Delgoffe GM, Pinho SS. Reprogramming CD8+ T-cell branched N-glycosylation limits exhaustion, enhancing cytotoxicity and tumor killing. Cancer Immunol Res. 2025 Aug 19. 

In the Spotlight...

Engineered T cells stimulate dendritic cell recruitment and antigen spreading for potent anti-tumor immunity

Focusing on antigenic heterogeneity and antigen loss in solid tumors, Xiao, Wang, and He et al. engineered T cells to express FLT3L and XCL1 (FX). Adoptively transferred FX T cells improved DC recruitment and activation in the TME (increased IFNγ and IL-12), inducing antigen spreading and potent polyclonal T cell responses, and resulting in control and elimination of antigenic heterogeneous tumors and prevention of immune escape. XCL1 expression positively correlated with a CD8+ Tpex signature in mouse and human tumors, and with patient survival and response to ICB. FX-CAR T cells also exhibited superior tumor control in humanized mice.

Contributed by Katherine Turner

Long-range deployment of tumor-antigen-specific cytotoxic T lymphocytes inhibits lung metastasis of breast cancer

Xing, Zhou, and Wang et al. demonstrated the role of CD103+CD8+ T cells in protection against breast cancer lung metastasis. Tumor antigen-specific CD103+CD8+ T cells were primed in TDLNs, and recruited to the lungs. Extracellular vehicles from early- and late-stage tumors differentially polarized alveolar macrophages to release CCL25 and IDO1, respectively. CCL25/CCR9 signaling recruited tumor antigen-specific CD103+CD8+ T cells to the lung, whereas IDO1 impaired pulmonary CD103+CD8+ T cells to facilitate lung metastasis. IDO1 inhibition increased CD103+CD8+ T cell infiltration in the lungs and alleviated lung metastasis.

Contributed by Shishir Pant

Anchored screening identifies transcription factor blueprints underlying dendritic cell diversity and subset-specific anti-tumor immunity

Using a sequential anchored transcription factor screen, Henriques-Oliveira, Altman, and Kurochkin et al. identified two transcription factor triads: PU.1, IRF4, and PRDM1 as inducers of cells similar to type 2 conventional DCs, and SPIB, IRF8, and IKZF2 as inducers of plasmacytoid DC-like cells. Chromatin remodeling at subset-specific sites drove the lineage divergence, each displaying specific phenotypes and functional properties. In vivo reprogramming of cancer cells into distinct DC subtypes showed distinct subset-specific antitumor immunity and durable memory responses in YUMM1.7 melanoma and EO771 breast cancer models.

Contributed by Shishir Pant

First-line sacituzumab tirumotecan with tagitanlimab in advanced non-small-cell lung cancer: a phase 2 trial

Hong et al. investigated sac-TMT, a TROP2-targeted antibody–drug conjugate, in combination with tagitanlimab (anti-PD-L1) as a first-line therapy in advanced NSCLC without actionable genomic alterations. In cohorts 1A (q3w, n=40) and 1B (q2w, n=63) in the open-label trial, ORRs were 40% and 66.7%, with mPFS of 15.4 months and Not Reached, respectively. Treatment response did not differ by baseline TROP2 or PD-L1 expression, nor by histological subtype. The most common Grade 3+ AEs were hematologic (neutropenia, lymphopenia, anemia). Serious TRAEs occurred in 10.0% and 20.6% of patients, but TRAE-related discontinuation was minimal, and no deaths occurred.

Contributed by Morgan Janes

Neoadjuvant immunotherapy promotes the formation of mature tertiary lymphoid structures in a remodeled pancreatic tumor microenvironment

Sidiropoulos et al. report state-of-the-art spatial genomics and proteomic profiling of PDAC tumors and tumor-adjacent lymph nodes from patients treated with GVAX and anti-PD-1 alone or in combination with a 41BB agonist in the neoadjuvant setting. Spatial transcriptomics identified TLS-specific spatial gene expression signatures associated with improved survival in TCGA PDAC samples. TLS-adjacent stroma of pathologic responders showed ECM remodeling with decreased desmoplasia. Neoadjuvant immunotherapy induced TLS formation in diverse spatial niches with mature B cell aggregates that disseminate IgG antibodies.

Contributed by Shishir Pant

Engineered bacteria launch and control an oncolytic virus

Singer and Pabón et al. developed a Salmonella typhimurium bacterial platform that delivered non-spreading, self-replicating viral RNA, even into cell types beyond the virus’s natural tropism. S. typhimurium “encapsidating” full-length oncolytic Senecavirus A delivered i.t. into s.c. engrafted tumors cleared treated and distal tumors in athymic mice, as did i.v. treatment of immunocompetent mice (even in the presence of pre-existing circulating viral-neutralizing antibodies), without adverse effects. Additional virus engineering aimed to control viral spread and persistence and to mitigate RNA mutational escape by requiring that virion maturation depend on bacterially delivered TEV protease.

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.