Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Review
. 2023 Mar 1;31(3):631-646.
doi: 10.1016/j.ymthe.2022.11.019. Epub 2022 Dec 5.

Mucosal-associated invariant T cells for cancer immunotherapy

Affiliations
Review

Mucosal-associated invariant T cells for cancer immunotherapy

Yan-Ruide Li et al. Mol Ther. .

Abstract

Human mucosal-associated invariant T (MAIT) cells are characterized by their expression of an invariant TCR α chain Vα7.2-Jα33/Jα20/Jα12 paired with a restricted TCR β chain. MAIT cells recognize microbial peptides presented by the highly conserved MHC class I-like molecule MR1 and bridge the innate and acquired immune systems to mediate augmented immune responses. Upon activation, MAIT cells rapidly proliferate, produce a variety of cytokines and cytotoxic molecules, and trigger efficient antitumor immunity. Administration of a representative MAIT cell ligand 5-OP-RU effectively activates MAIT cells and enhances their antitumor capacity. In this review, we introduce MAIT cell biology and their importance in antitumor immunity, summarize the current development of peripheral blood mononuclear cell-derived and stem cell-derived MAIT cell products for cancer treatment, and discuss the potential of genetic engineering of MAIT cells for off-the-shelf cancer immunotherapy.

Keywords: CAR engineering; GvHD; MAIT cell; allogeneic cell therapy; cancer immunotherapy; chimeric antigen receptor engineering; combination therapy; graft-versus-host disease; mucosal-associated invariant T cell; off-the-shelf; stem cell engineering.

PubMed Disclaimer

Conflict of interest statement

Declaration of interests Y.-R.L. and L.Y. are inventors on patents relating to this article filed by UCLA. L.Y. is a scientific adviser to AlzChem and Amberstone Biosciences, and a co-founder, stockholder, and advisory board member of Appia Bio. None of the declared companies contributed to or directed any of the research reported in this article.

Figures

None
Graphical abstract
Figure 1
Figure 1
Recognition, activation, and tumor cell killing mechanism of MAIT cells (A) The main T cell subsets and their antigen recognition. Conventional αβ CD4+ and CD8+ T cells recognize peptide antigens presented through the MHC-II and MHC-I molecules on the antigen-presenting cells (APCs), respectively. The invariant natural killer T (iNKT) cells recognize glycolipid antigens presented by MHC class I-like molecule, CD1d. Gamma delta T (γδ T) cell TCR recognizes different ligands presented by diverse receptors including MHC, CD1, and other surface-bound proteins. MAIT cells express invariant αβ TCR, which binds to riboflavin (vitamin B2) biosynthesis derivatives presented by MR1 on APCs. Ag, antigen. (B) TCR-dependent and -independent MAIT cell activation. MAIT TCR-dependent activation requires riboflavin (vitamin B2) biosynthesis derivatives to be presented on MR1 to a TCR in conjunction with co-stimulation. In addition, MAIT cells can be activated by cytokines such as IL-12 and IL-18 in a TCR-independent pathway. Activated MAIT cells proliferate and secret various cytokines, chemokines, and cytotoxic molecules. (C) Tumor cell killing mechanism by MAIT cells. MAIT cells can attack tumors through multiple mechanisms, including direct killing of MR1+ tumor cells through MR1/TCR recognition, killing of MR1 tumor cells through NK pathway, adjuvant effects on boosting dendritic cell (DC) and cytotoxic T lymphocyte (CTL) antitumor activities, and inhibition of TAMs and MDSCs.
Figure 2
Figure 2
Generation and genetic engineering of human peripheral blood mononuclear cell-derived MAIT (PBMC-MAIT) cells (A) Generation of MAIT or CAR-engineered MAIT (CAR-MAIT) cells from human PBMCs. MACS-enriched ex vivo MAIT cells can be co-cultured with either irradiated MR1-tetramer-negative PBMCs or latex bead-based artificial APCs. (B) CAR/TCR/NK triple tumor cell killing mechanism. CAR-MAIT cells target tumor cells through CAR ligand/CAR, MR1/5-OP-RU/TCR, and NK ligands/NK activating receptors. The multiple tumor targeting mechanisms grant MAIT cells a stronger antitumor capacity and an enhanced capacity to counteract tumor antigen escape. (C) Multiple genetic engineering approaches could be incorporated into the generation of PBMC-MAIT cells, such as arming CARs to enhance antitumor efficacy, overexpressing cytokines (i.e., IL-2 and IL-15) to boost immune reaction, ablating HLA-I and HLA-II to reduce host T cell-mediated alloresponse, transducing HLA-E or CD47 to reduce host NK cell-mediated alloresponse, knocking out checkpoints (i.e., PD-1 and CTLA-4) to reduce the immunosuppression, and depleting CD52 to grant cells resistance to the T cell depletion preconditioning treatment.
Figure 3
Figure 3
Generation and genetic engineering of human stem cell-derived MAIT cells (A) Cloning of human MAIT TCR genes. Single human MAIT cells are sorted from healthy donor PBMCs using flow cytometry. The sorted single MAIT cells are subjected to TCR cloning using a single-cell TCR sequencing and cloning technology. A pair of MAIT TCR α- and β-chain genes are selected based on the functional analysis such as cytokine secretion capacity and activation marker expression. (B) Generating MAIT cells from hematopoietic stem cells (HSCs). Human CD34+ HSCs from cord blood or peripheral blood stem cells are transduced with MAIT TCR and then cultured in an in vitro HSC differentiation system, such as OP9-DL1, artificial thymic organoid (ATO), or feeder-free culture. At the end of culture, MAIT cells with transduced MAIT TCR are generated. MAIT cells can be engineered with CARs to enhance their antitumor capacity. Gene editing such as CRISPR-Cas9 can be incorporated into HSCs or MAIT cells to achieve specific gene modifications. (C) Generating MAIT cells from induced pluripotent stem cells (iPSCs). Cord blood MAIT cells are transduced with Sendai viral vectors carrying KLF4, OCT3/4, SOX2, and c-MYC. MAIT-iPSC cell lines are established after passing the standardized pluripotency tests. MAIT-iPSCs are cultured on OP9 feeder cells and differentiate into CD34+/CD43+ lymphocyte precursors. CD34+/CD43+ precursors are then cultured on OP9/DL1 feeder cell layers for 30 days to form MAIT-iPSC-derived MAIT-like (reMAIT) cells.

References

    1. Amini A., Pang D., Hackstein C.-P., Klenerman P. MAIT cells in barrier tissues: lessons from immediate neighbors. Front. Immunol. 2020;11:584521. doi: 10.3389/fimmu.2020.584521. - DOI - PMC - PubMed
    1. Dusseaux M., Martin E., Serriari N., Péguillet I., Premel V., Louis D., Milder M., Le Bourhis L., Soudais C., Treiner E., Lantz O. Human MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-17–secreting T cells. Blood. 2011;117:1250–1259. doi: 10.1182/blood-2010-08-303339. - DOI - PubMed
    1. Legoux F., Salou M., Lantz O. MAIT cell development and functions: the microbial connection. Immunity. 2020;53:710–723. doi: 10.1016/j.immuni.2020.09.009. - DOI - PubMed
    1. Treiner E., Duban L., Bahram S., Radosavljevic M., Wanner V., Tilloy F., Affaticati P., Gilfillan S., Lantz O. Selection of evolutionarily conserved mucosal-associated invariant T cells by MR1. Nature. 2003;422:164–169. doi: 10.1038/nature01433. - DOI - PubMed
    1. Tilloy F., Treiner E., Park S.H., Garcia C., Lemonnier F., de la Salle H., Bendelac A., Bonneville M., Lantz O. An invariant T cell receptor alpha chain defines a novel TAP-independent major histocompatibility complex class Ib-restricted alpha/beta T cell subpopulation in mammals. J. Exp. Med. 1999;189:1907–1921. doi: 10.1084/jem.189.12.1907. - DOI - PMC - PubMed

Publication types

MeSH terms

Substances