Weekly Digests
‹ Back to July

MAIT cells: linking the gut microbiome to antitumor immunity?

July 1, 2020

In patients with colon adenocarcinoma, high infiltration by mucosa-associated invariant T (MAIT) cells – which can be activated by a wide variety of bacteria and recognize epitopes presented by the MHC-like molecule using a limited set of invariant/preferential TCRs – have recently been associated with poor clinical outcomes. In a study recently published in Cell Reports Medicine (a new journal from Cell Press), Li et al. investigated the role of MAIT cells in cancer, shedding new light on how the microbiome impacts antitumor immune responses.

Based on evidence from prior studies, Li et al. investigated the hypothesis that MAIT cells can be activated by and respond to gut microbiome-associated antigens within the tumor microenvironment. To begin, they profiled samples from colorectal cancer (CRC), non-small cell lung carcinoma (NSCLC), and renal cell carcinoma (RCC) and found that MAIT cells accounted for a higher portion of total T cells in CRC compared to NSCLC or RCC. As no such difference was observed in the periphery, this suggested a tumor-specific accumulation of MAIT cells in CRC.

Comparing the profiles of MAIT cells within CRC to MAIT cells from adjacent tissue or PBMCs, the researchers identified a tumor-specific phenotype characterized by a distinct transcriptomic profile (enrichment for TCR signaling and negative apoptotic regulation pathways), high expression of CD69, CD103, CD38, and CD39, and lower expression of CD27 and CD49d compared with peripheral MAIT cells. Most tumor-infiltrating MAIT cells also expressed markers indicative of response to inflammation. Interestingly, the researchers also observed heterogeneity within tumor-infiltrating MAIT cells from CRC that was not observed in the periphery.

Among tumor-infiltrating MAIT cells, Li et al. identified a distinct cluster of CD4+ cells, some of which surprisingly co-expressed CTLA-4, Foxp3, and other markers that are typically associated with Tregs. Functional analyses revealed that unlike Tregs, CD4+Foxp3+ MAIT cells produced the pro-inflammatory cytokine TNFα, suggesting that Foxp3 expression on CD4+ MAIT cells might be a marker of activation rather than a marker of immunosuppressive function. The researchers also identified distinct signatures between CD39+ and CD39- MAIT cell populations. Further, within CD39+ MAIT cells, there was one subpopulation that expressed CD69 and CD103, both markers of tissue residency, along with CD38, and another that expressed CTLA-4, Tim3, GITR, and CD45RA.

Looking more closely at MAIT cells expressing CD39, Li et al. found that the proportion of CD39+ MAIT cells was increased in CRC, with many of these cells co-expressing CD8. The CD39+CD8+ MAIT cell population also frequently expressed markers of tissue residency and genes associated with activation and exhaustion. Further, many CD39+ MAIT cells showed evidence of increased proliferation and enhanced protection against cell death compared to CD39- MAIT cells. While CD39+ MAIT cells showed little evidence of polyfunctionality, most were able to express Granzyme A, Granzyme B, or perforin. Together, this evidence suggested that tumor-infiltrating CD39+ MAIT cells might represent a population of MAIT cells that had been activated in response to antigen stimulation.

To test the functional responses of MAIT cells, Li et al. showed that stimulation of MAIT cells with Escherichia coli induced expression of CD39. Blocking stimulation with an anti-MR1 antibody abrogated this effect, indicating that CD39 expression was induced by TCR-driven activation. Based on these results, the researchers hypothesized that the phenotypes and functions of tumor-infiltrating MAIT cells could be affected by bacteria and/or metabolites present in the tumor microenvironment in CRC. Consistent with CD39 expression as a marker of antigen stimulation, they observed that CD39 expression was higher in CRC compared to NSCLC or RCC, reflective of the abundance of bacteria typically found in the colon relative to lungs and kidneys. Furthermore, analysis of CRC tumors showed that that the bacterial load was higher in tumors than in paired adjacent tissue, as was the expression of infiltrating CD39+ MAIT cells. Further, expression of CD39 on MAIT cells was particularly enhanced in tumors with high bacterial loads compared to lower bacterial loads, suggesting that when bacteria was more abundant, MAIT cells were more likely to become activated and express CD39. Other than CD39, the expression profile of MAIT cells was fairly consistent across CRC, NSCLC, and RCC, further indicating that the unique CD39 expression on tumor-infiltrating MAIT cells in CRC occurs as the result of antigen exposure.

Between patients, bacterial loads were heterogeneous, though bacteria from the same phylum were generally correlated. Most bacterial strains were not particularly abundant, though a few, including E. coli, were notably enriched. To determine whether certain gut bacterial strains associated with CRC could affect the functions of MAIT cells, Li et al. selected a few strains of bacteroides and fusobacterium that were highly abundant in CRC. In in vitro stimulation assays, only Fusobacterium nucleatum or culture supernatents from F. nucleatum activated MAIT cells and stimulated production of IFNγ – an effect that was dependent on TCR engagement.

Exploring whether MAIT cells might impact patient clinical outcomes, Li et al. found that patients with CRC that was highly infiltrated by bacteria showed evidence of low immune and inflammatory responses. Within these patients, MAIT cells expressed markers of wound healing and tissue repair that would suggest a role in promoting epithelial growth. The gene for PyrD, an enzyme that is involved in riboflavin synthesis and supports MAIT cell development and activation, was enriched in metagenomes from CRC versus normal colon samples, suggesting that tumor-specific characteristics might play a role in bacterial infiltration and thus MAIT cell activation. Evaluating the frequency of MAIT cells across clinical stages revealed a trend, not reaching statistical significance, of higher frequency and later stage.

Overall, the results uncovered by Li et al. provide evidence that MAIT cells in CRC can be activated by and respond to microbial antigens in a TCR-dependent manner. This activation appears to be marked by the expression of CD39. Given results that the presence of MAIT cells seems to affect clinical outcomes in patients with CRC, this early research could pave the way for cancer immunotherapies that target MAIT cells or the microbiome.

by Lauren Hitchings

Meet the researcher

This week, Shamin Li and Evan Newell answered our questions.

What prompted you to tackle this research question?
SL: As my PhD focused on NKT cells, I was familiar with unconventional T cells and was interested in MAIT cells, which were still a not-so-well documented immune population, especially in the context of human malignancies. Since the research on interrelationships between cancer therapies and the microbiome was booming, I took the opportunity of our large cohort of human colorectal cancer samples to assess MAIT cell infiltration in the tumors. We were hoping that these cells recognizing bacterial metabolites could be somehow connected to bacterial infiltration in tumors, and we were able to find evidence that indicated the importance of MAIT cells in microbiota-associated cancers.
EN: I’m generally interested in how T cell antigen specificity can give rise to a multitude of different T cell populations with different functions, and then how they can work together to provide immunity to cancer and infectious disease. Although their roles are still mysterious, MAIT cells are a nice example of a highly abundant (especially in human blood and other tissues) population of T cells for which the antigen specificity is known, but roles in disease are much less understood. This was definitely encouraging when it came to Shamin’s ambition to ask more about how these cells might contribute to cancer immunity.

What was the most surprising finding of this study for you?
SL: As CD39 is a marker associated with chronic TCR activation on conventional T cells, we were intrigued by the high expression of CD39 on tumor-infiltrating MAIT cells that was only found in colorectal cancer. We hypothesized that induction of this expression could be initiated by bacterial antigen recognition, an idea that we confirmed next by in vitro stimulation assays. The observation of a dominant CD39+ MAIT cell population in colorectal cancer is a key to this work, and the presence of a Foxp3+ subset found within it could be another interesting puzzle to solve.
EN: Because MAIT cells are known to be able to functionally respond to non-specific stimulation such as cytokines, I expected that MAIT cells would not necessarily show signs of active TCR-driven responses in cancer. So, it was surprising to find that tumor-infiltrating MAIT cells expressed markers associated with T cell exhaustion, including CD39, which we are hypothesizing to predominantly require a TCR-driven signal for its expression. I was also surprised to see Foxp3-expressing CD4+ MAIT cells in human tumors. 

What was the coolest thing you’ve learned (about) recently outside of work?
SL: During my break times, I enjoy watching wildlife documentaries and reading about zoology, because I find animal life fascinating. The other day I was reading about how the auditory cortex of macaques works differently as compared to humans, and so playing a harmonic or a pitchless/noisy tune would sound the same to them. For someone who loves music, I was stunned that such a similar species to us could have such a different auditory neural response. It makes me realize how sometimes we take our perceptions for granted and reminds me that we should always take a step back to gain a different perspective.
EN: With young kids at home and in the months since the development of the COVID-19 pandemic, it has been both a source of stress and a real joy to spend more time at home than ever before. In trying to maintain their learning, I’ve particularly enjoyed reading a variety of books that I hadn’t had a chance to read before, as well as many from my own childhood. In thinking of how we might be able to improve our learning, I’m intrigued by an article I read recently suggesting that you can learn more efficiently by increasingly spacing the time between repetition. I’ll have to figure out how to implement this on my kids and the words we’ve learned – maybe there’s an iPad app for it?

References:

Shamin Li ; Yannick Simoni ; Etienne Becht ; Chiew Yee Loh ; Naisi Li ; Daniel Lachance ; Si-Lin Koo ; Teck Por Lim ; Emile Kwong Wei Tan ; Ronnie Mathew ; Andrew Nguyen ; Justin Golovato ; Julia D. Berkson ; Martin Prlic ; Bernett Lee ; Samuel S. Minot ; Niranjan Nagarajan ; Neelendu Dey ; Daniel S.W. Tan ; Iain B. Tan ; and Evan W. Newell. Human Tumor-Infiltrating MAIT Cells Display Hallmarks of Bacterial Antigen Recognition in Colorectal Cancer. Cell Reports Medicine June 23, 2020.

In the Spotlight...

Antibody targeting tumor-derived soluble NKG2D ligand sMIC reprograms NK cell homeostatic survival and function and enhances melanoma response to PDL1 blockade therapy

Basher et al. showed that B10G5, an antibody able to clear soluble human NKG2D ligand (sMIC), synergized with anti-PD-L1. Co-therapy inhibited growth of sMIC+PD-L1+ melanoma to promote survival in syngeneic sMIC+ transgenic host mice; increased numbers of activated/effector-memory, antigen-specific NKG2D+CD8+ T cells and of NK cells with elevated NKG2D and IL-2Rα expression in draining lymph nodes and spleen; and inhibited tumor angiogenesis. B10G5 reversed sMIC-induced anti-survival and anti-proliferative gene expression patterns in NK cells to enable anti-PD-L1 effects. sMIC/MIC is highly expressed in metastatic human melanoma.

Contributed by Paula Hochman

CD4+ T-cell killing of multiple myeloma cells is mediated by resident bone marrow macrophages

Using myeloma mouse model, Haabeth and Henning et al. show, with adoptive transfer or TCR-transgenic (TCR-Tg) mice, that tumor-specific CD4+ T cells elicit antitumor immune response in the bone marrow (BM) via activation of BM-resident macrophages but not neutrophils, eosinophils, or NK cells. Antitumor immunity of CD4+ T cells was dependent on IFNγ and asialo GM1 (AGM1)-expressing CD11b+ macrophages. AGM1+CD11b+ cells from tumor-challenged TCR-Tg mice showed increased MHC-II expression and tumor-specific antigen presentation. Depletion of (Ly6G/C) Gr-1+ monocyte-derived cells led to loss of antitumor immune response in the BM.

Contributed by Shishir Pant

A rational mouse model to detect on-target off-tumor CAR T cell toxicity

To address on-target, off-tumor toxicity with CAR T cell therapy, Castellarin et al. developed a model in which hHER2 was stably expressed at high (hHER2-high) or low (hHER2-low) levels on normal hepatocytes to mimic clinical situations. High-affinity (HA) and low-affinity (LA) HER2 CAR T cells (CARTs) caused lethal liver toxicity in mice with hHER-high livers, but in hHER2-low mice, LA-CARTs resulted in less liver damage and lower circulating IFNγ. LA-CARTs exhibited superior antitumor efficacy against hHER2+ xenografts compared to HA-CARTs, partly due to faster trafficking from the liver into the tumor.

Contributed by Katherine Turner

Structure-guided engineering of the affinity and specificity of CARs against Tn-glycopeptides

Using a deep mutational scan, Sharma et al. showed that the 237-antibody, which binds to Tn antigen – a cancer-specific, aberrantly O-glycosylated epitope of a murine protein (OTS8) – is affected by the peptide side chains. CAR T cells transduced with higher-affinity 237-scFv variants only slightly increased CAR T cell activation against Tn-OTS8-expressing cancer cells. Selection for 237-scFv variants against a different O-glycosylated epitope from human MUC-1 (Tn-MUC1) resulted in molecules reactive to Tn-MUC1, Tn-OTS8, and cancer cells lacking both antigens, suggesting a response against multiple Tn antigens.

Contributed by Anna Scherer

CLINICAL TRIAL: A Phase I/IIa trial of a frameshift peptide neoantigen-based vaccine for mismatch repair-deficient cancers

Microsatellite-unstable (MSI) tumors often possess identical mutations between patients, encoding conserved frameshift peptide (FSP) neoantigens in tumor suppressor genes. Kloor et al. report a clinical trial of an FSP long peptide + Montanide vaccine in 22 previously treated patients with MSI colorectal cancer. The vaccine was well tolerated in the majority of patients. All patients demonstrated FSP-specific immunity after vaccination compared to 50% before, most commonly through antibody responses, followed by CD4+ and then CD8+ T cell responses. Of the 3 patients who had tumors prior to treatment, two experienced stable disease after treatment.

Contributed by Alex Najibi

Cellular backpacks for macrophage immunotherapy

Wyatt Shields IV et al. engineered phagocytosis-resistant backpacks, a class of soft, disk-shaped, anisotropic (different strengths in different dimensions) particles containing IFNγ, that can adhere to macrophage surfaces and regulate phenotype following release. Macrophages carrying IFNγ backpacks in vitro as well as in TAMs following intratumoral (4T1 breast cancer) injection demonstrated M1 phenotype with elevated levels of iNOS, MHC-II, and CD80 expression. Treatment with macrophages carrying IFNγ backpacks safely reduced tumor growth and metastatic burden, and improved overall survival.

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.