Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

nature reviews cancer https://doi.org/10.

1038/s41568-023-00562-w

Review article Check for updates

Innate lymphoid cells and innate-like


T cells in cancer — at the crossroads
of innate and adaptive immunity
Benjamin Ruf 1
, Tim F. Greten 1,2
& Firouzeh Korangy 1

Abstract Sections

Immunotherapies targeting conventional T cells have revolutionized Introduction

systemic treatment for many cancers, yet only a subset of patients Characteristics of ILTCs
benefit from these approaches. A better understanding of the complex and ILCs

immune microenvironment of tumours is needed to design the next The role of ILCs and ILTCs
in immunosurveillance
generation of immunotherapeutics. Innate lymphoid cells (ILCs) and
innate-like T cells (ILTCs) are abundant, tissue-resident lymphocytes Bridging adaptive and innate
immunity
that have recently been shown to have critical roles in many types of
ILC-based and ILTC-based
cancers. ILCs and ILTCs rapidly respond to changes in their surrounding cancer immunotherapy
environment and act as the first responders to bridge innate and
Conclusion
adaptive immunity. This places ILCs and ILTCs as pivotal orchestrators
of the final antitumour immune response. In this Review, we outline
hallmarks of ILCs and ILTCs and discuss their emerging role in
antitumour immunity, as well as the pathophysiological adaptations
leading to their pro-tumorigenic function. We explore the pleiotropic,
in parts redundant and sometimes opposing, mechanisms that underlie
the delicate interplay between the different subsets of ILCs and ILTCs.
Finally, we highlight their role in amplifying and complementing
conventional T cell functions and summarize immunotherapeutic
strategies for targeting ILCs and ILTCs in cancer.

Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Centre for Cancer
1

Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA. 2NCI CCR Liver
Cancer Program, National Institutes of Health, Bethesda, MD, USA. e-mail: firouzeh.korangy@nih.gov

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 351


Review article

Introduction immunity against microbial pathogens and cancer4. ILTCs share typical
Peptide-reactive, conventional CD4+ and CD8+ T cells have been major features of conventional T cells; they both develop in the thymus16 and
targets of cancer immunotherapy, improving therapeutic outcome express TCRs that upon encounter with an antigen drive their activation
across various cancer types1. Nevertheless, recent clinical studies and oligoclonal expansion12. Beyond TCR-dependent activation, ILTCs
have revealed the limitations of current adaptive immune cell-based can rapidly sense changes in their environment through the expression
approaches1. This could, however, be overcome by targeting the innate of various integrins, chemokine and cytokine receptors, enabling them
immune cell axis, namely, innate lymphoid cells (ILCs) and innate-like to promptly mount potent cytokine responses, a key feature shared with
T cells (ILTCs), in parallel2. ILCs3, including natural killer (NK) cells and innate immune cells12. Furthermore, similar to other innate immune
helper ILCs (termed ILC1s, ILC2s and ILC3s) and lymphoid tissue-inducing cells, ILTCs acquire an effector phenotype before thymic egress and
(LTi) cells, are a heterogenous cell population of the innate immune accumulate at barrier sites including the gut mucosa, lungs or liver.
system that has emerged as pivotal regulators of inflammation, tissue The development of ILCs and ILTCs is further described in Box 1, and
repair, barrier homeostasis and immune tolerance. Unconventional their tissue distribution is described in Box 2. Although ILTCs share
T cells or ILTCs (used interchangeably) are composed of three key the common features described thus far, NKT, MAIT and γδ T cells are
subsets: natural killer T (NKT) cells4, mucosal-associated invariant T characterized by distinct attributes outlined subsequently.
(MAIT) cells5 and γδ T cells6. They have pleiotropic functions and react
rapidly to non-peptide antigens via their conserved T cell receptors Natural killer T cells. iNKT cells (also known as type I NKT cells) share
(TCRs). Although the importance of NK cells7 in the context of cancer phenotypic and functional similarities with both T cells and innate
has been known for many years8, helper ILCs9 and unconventional T cells lymphocytes4. They were first described as a T cell subset expressing
have only recently gained attention as important regulators of cancer a semi-invariant TCR, composed of an invariant α-chain and a limited
immunology. ILCs as the most innate cells across the innate-adaptive number of TCR β-chains13,17,18 (Fig. 1). This semi-invariant TCR is reactive
spectrum (with degree of innateness defined as transcriptional state of to self and foreign glycolipid ligands including α-galactosylceramide
each cell type with adaptive cells on the one end and ILCs on the other (α-GalCer) presented by the MHC class I‐like molecule CD1d13.
end of the spectrum)10,11 share many characteristics with unconven- A second broad type of CD1-restricted NKT cells, type II NKT, use
tional T cells, although each cell type has distinct functional features diverse αβ TCRs that do not conform to the semi-invariant motifs
(Fig. 1). Although ILCs and ILTCs with varying degrees of innateness10,11 described earlier and are reactive to self-lipid antigen sulfatide19.
can have fundamental roles in cancer, their network of connection iNKT cells can be further categorized according to the expression
and complex interplay within the tumour microenvironment (TME) of canonical transcription factors and cytokine profiles on the basis of
are not well understood. CD4+ helper T (TH) cell subsets TH1, TH2 and TH17 (Fig. 1) and are termed
Here, we provide an overview of our current understanding of ILCs iNKT1, iNKT2 and iNKT17 throughout the text16,20,21.
and unconventional T cells in the context of cancer with an emphasis In general, the majority of iNKT cells are non-circulating, tissue-
on antitumour immunity. We provide a summary of cell type defini- resident lymphocytes, where their number varies between tissues
tions, their role in tumour immunosurveillance, how these cells can and species16. Recent work in mice suggests that there is considerable
bridge innate and adaptive immunity and current concepts of how they heterogeneity also within the iNKT fraction consisting of a circulat-
could be targeted for cancer immunotherapy. We aim to point out their ing subset with NK-like cytotoxic potential in vivo that is functionally
similarities, as well as relevant distinctions, and outline the TME factors distinct from tissue-resident iNKT cells22. Type I NKT cells are abundant
that govern ILC and ILTC antitumorigenic or pro-tumorigenic function. in laboratory mice, representing between 1% and 3% of CD3+ T cells in
With this, we discuss their potential synergistic and redundant roles, some tissues and up to 50% of hepatic T cells16. In humans, iNKT cells are
as well as crosstalk with other cells of the innate and adaptive immune less frequent23,24, accounting for less than 1% of T cells in the blood, and
system. We conclude with possible therapeutic strategies targeting type II NKT cells outnumber iNKT cells in human peripheral blood
ILCs and ILTCs to enhance anticancer immunity. mononuclear cells (PBMCs).

Characteristics of ILTCs and ILCs Mucosal-associated invariant T cells. MAIT cells are ILTCs that sense
Innate-like T cells microbial metabolites derived from vitamin B2 (riboflavin) or vitamin B9
Unconventional or ILTCs share phenotypical and functional similarities (folic acid) presented on MR1 (refs. 14,15,25,26). MAIT cells also have
of both conventional T cells and innate immune cells. Although conven- a TCR and were initially identified through their highly restricted rep-
tional, peptide-reactive T cells primarily recognize antigens presented ertoire of TCR α-chains (consisting of TRAV1-2 and thus defined as
by polymorphic major histocompatibility complex (MHC) class I and II TRAV1-2+ T cells) and limited number of TCR β-chains27,28. Beyond this
molecules, ILTCs recognize a diverse range of self-molecules and non- definition of MAIT cells (MR1-riboflavin antigen-reactive, canonical
self-molecules. This includes lipid ligands for invariant natural killer TCR+), another group of MR1-reactive T cells exists, which has recently
T (iNKT) cells, riboflavin derivatives for MAIT cells and phosphoanti- been shown to include cancer-specific (non-MAIT) T cells with the
gens for γδ T cell subsets. These are recognized through their semi- potential as pan-cancer therapeutic29.
invariant (MAIT, iNKT and some γδ T cell subsets, for example, Vγ9Vδ2+ Much like CD4+ TH cells, several MAIT cell subsets have been
T cells) or diverse (type II NKT and other γδ T cells) TCRs in an MHC- described (Fig. 1). MAIT cells sense a range of inflammatory triggers
unrestricted manner12 (Fig. 1). MHC class I-like molecules CD1d and MHC through cytokine receptors such as IL-7 receptor (IL-7R), IL-12R, IL-15R,
class I-related gene protein (MR1) present antigens to NKT cells and IL-18R and IL-23R. This can enhance TCR-mediated signals or lead to
MAIT cells, respectively13–15. They are evolutionarily conserved, suggest- MAIT cell activation in a TCR-independent, innate-like manner30,31.
ing that the ILTCs have a critical role in host physiology by mediating the Following stimulation, MAIT cells rapidly produce various effector mole­
interaction with non-self, therefore maintaining tissue homeostasis11. cules, cytokines and chemokines, including granzymes, granulysin,
Together, MAIT, γδ T and iNKT cells have emerged as crucial players in perforin, GM-CSF, interferon-γ (IFNγ), tumour necrosis factor (TNF),

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 352


Review article

Tumour Phosphoantigen
microenvironment

Key Key Key


APC
transcription cytokines cytokine
factors secreted receptors

CD1d MR1 MHC-II MHC-I


Type 1 Glycolipid Riboflavin BTN2A1– Peptide
metabolite BTN3A1 ligand T-bet • IFNγ • IL-12R
• TNF • IL-18R
• IL-15R
ILC1 CD4+ CD8+
NK iNKT1 MAIT1 γδ T CTL

Type 2
GATA3 • IL-4 • IL-33R
• IL-5 • IL-25R
• IL-9
ILC2 MAIT2 CD4+ • IL-13
iNKT2 (?) γδ T
• AREG

Type 17
RORγt • IL-17A • IL-23R
• IL-22 • IL-1RI
• TGFβR
ILC1
LTi ILC3 CD4+
iNKT17 MAIT17 γδ T

ILCs ILTCs Conventional T cells TCRVα24-Jα18 (human) TCRVγ9Vδ2


TCRVα14-Jα18 (mouse) (example)

Hours Reactivity Days TCRVα7.2-Jα33 Conventional


(human) αβ TCR
TCRVα19-Jα33 (highly variable)
Innateness Adaptiveness (mouse)

Fig. 1 | Subsets of innate lymphoid cells and innate-like T cells in the tumour phosphoantigens and butyrophilin (BTN) family members BTN3A1 and BTN2A1.
microenvironment. Innate lymphoid cells (ILCs), innate-like T cells (ILTCs) and Conventional T cells, CD4+ and CD8+ T cells, express classical αβ TCRs that are
conventional CD4+ and CD8+ T cells are displayed according to their degree of reactive to peptide ligands presented by MHC class II or MHC class I, respectively.
innateness as coined previously11. ILCs, namely, natural killer (NK) cells, ILC1s, Here, antigen-presenting cells (APCs) are used as examples for MHC, CD1d and
ILC2s, ILC3s and lymphoid tissue inducer (LTi) cells, have the highest degree MR1 expressing cells, but MR1 and CD1d are generally expressed in a broad range
of innateness and thus do no express T cell receptor (TCR). Hereby, ILCs and of both haematopoietic and non‐haematopoietic cell types. Moreover, subsets of
unconventional T cells react to environmental cues within hours, whereas adaptive ILCs and ILTCs can be categorized in accordance with major CD4+ helper T cell
immunity requires days for potent TCR-specific activation. Although ILCs do subsets (type 1, type 2 and type 17 subsets are shown; type 9 and regulatory sub­
not express a TCR, invariant natural killer T (iNKT) cells and mucosal-associated sets were omitted for brevity), sharing similar functional profiles as defined by
invariant T (MAIT) cells, subsets of ILTCs, express semi-invariant TCRs that are common transcription factor, effector molecules and cytokine receptors.
restricted to recognizing CD1d presenting glycolipids or major histocompatibility Type 1 ILCs and ILTCs express T-box expressed in T cells (T-bet) and secrete high
complex (MHC), class I-related gene protein (MR1) presenting riboflavin-derived levels of interferon-γ (IFNγ) and tumour necrosis factor (TNF). Expression of
antigens, respectively. The figure displays subsets of type I NKT cells (or iNKT cells): GATA3 represents a hallmark of type 2 subsets. These cells are characterized as
iNKT1, iNKT2 and iNKT17 cells, whereas type II NKT cells are not displayed to IL-4, IL-5, IL-9, IL-13 and amphiregulin (AREG) secreting cells. Type 17 phenotypes
improve clarity. The presence of type 2 MAIT cell subset is not clear to date. The are characterized by the expression of retinoic acid receptor-related orphan
γδ TCR cells, another ILTC subset, are more diverse and because of this only one receptor-γt (RORγt) and production of IL-17 and IL-22. CTL, cytotoxic T lymphocyte;
mechanism for activating human Vγ9Vδ2+ T cells is displayed; this is through IL-12R, IL-12 receptor; TGFβR, transforming growth factor-β receptor.

IL-17, IL-10, transforming growth factor-β (TGFβ), CC-chemokine ligand 3 γδ T cells. Like the name might suggest, γδ T cells express TCRs with
(CCL3), CCL4 and CXC-chemokine ligand 16 (CXCL16)32–34. γ-chains and δ-chains, rather than the canonical αβ TCR on conven-
MAIT cells are relatively scarce in human secondary lymphoid tional T cells. Thus, they differ in their relative dependence on classical
organs, but account for up to 10% of the T cells in circulation35. They MHC molecules. In human PBMCs, γδ T cells represent a fraction of up
are further enriched at mucosal sites and in the liver, where they are the to 10% of all T cells but can be markedly expanded at barrier sites and
most common T cell subset36. By contrast, they are scarce in common peripheral, non-lymphoid organs38. In healthy adults, circulating γδ
laboratory mouse strains C57BL/6 and BALB/c24. However, they are T cells in the peripheral blood predominantly express TCR variable
more abundant in the CAST/EiJ strain, which allowed the generation (V) genes Vγ9 and Vδ2 (therefore Vγ9Vδ2+ T cells)39,40, whereas the Vδ1
of an MAIThigh congenic (inbred) strain, a valuable tool for studying subsets (paired with various γ-chains) are more abundant in peripheral
MAIT cell biology37. tissues41. Antigen recognition and specific ligands for γδ T cells are not

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 353


Review article

Box 1

Innate lymphoid and innate-like T cells during development


Innate lymphoid cells (ILCs) and innate-like T cells (ILTCs) share and tissues can differentiate at any stage in response to their
many phenotypic and functional properties, yet fundamentally environmental signals to generate tissue-specific ILCs, that is, during
differ in their development. Both T cells and ILC lineages arise from inflammation or perturbations in tissue homeostasis also referred
common lymphoid progenitor (CLP) cells with T cell precursors to as ‘ILC poiesis’298. CLP cells as influenced by transcription factors
migrating to the thymus for further development16. Studies in mice such as nuclear factor interleukin-3-regulated (NFIL3), DNA-binding
have shown that γδ T cells develop at the fetal stage preceding protein inhibitor 2 (ID2), thymocyte selection-associated high-
mucosal-associated invariant T (MAIT) and natural killer T (NKT) mobility group box protein (TOX), T cell factor 7 (TCF7), GATA-
cell development16. ILTC development is governed by the master binding factor 3 (GATA3), zinc-finger and BTB domain-containing
regulator promyelocytic leukaemia zinc-finger protein292–294, protein 16 (ZBTB16) (in mice) and ID2, TOX, TCF7, GATA3, ROR-α and
and these cells acquire effector phenotypes that are governed ETS1 (in humans) will commit to the NK cell and ILC lineage through
by lineage-defining transcription factors that orchestrate these a series of unipotent and multipotent ILC precursors such as
specialized differentiation programmes (listed in brackets after each α-lymphoid precursor, early innate lymphoid precursor, common
subset). This lineage commitment occurs before thymic egress, helper innate lymphoid precursor and ILC precursor299. NK cells
and NKT1, MAIT1 (T-bet+), NKT2 (GATA3+), NKT17 and MAIT17 (retinoic and helper ILC1 subsets are both dependent on T-bet for their
acid receptor-related orphan receptor-γt (RORγt+) cell subsets are development, but NK cells also require eomesodermin (EOMES)300
already found in the thymus. Importantly, CD1d and MR1 expression for their development. It is important to mention that among ILC1
on thymocytes is essential for NKT and MAIT cell development. subsets, liver ILC1 subsets are distinct and have been shown to
Beyond that, these ILTCs require early-life exposure to microbiota for develop from fetal liver-derived haemopoietic stem cells via an
proper development 13,295,296 and shaping their contribution to barrier interferon-γ-dependent pathway301. Key transcription factors that
immunity and tissue homeostasis 11,297. drive ILC2 differentiation are TCF1, BCL11B and GATA3, ensuring
ILC development, although closely shared by T cells, is different effector function of ILC2 (ref. 302). ILC3 development is regulated
as it is controlled by strict transcriptional factors, which guide the by RORγt mirroring the polarization of post-TH17 cells303. Several
ILC commitment pathway. All subsets of ILCs are derived from a outstanding reviews have discussed the constantly updating field
CLP cell in the adult bone marrow. ILC precursors in circulation of ILC development in both mice and humans299,304,305.

well understood and have been an intense area of research (reviewed of other immune cell markers and therefore known as ‘lineage-negative’
elsewhere42,43). The evolutionary divergence of γδ TCR genes across cells50. On the basis of the cytokine signatures and developmental
species has hampered translation of functional studies, as the major transcription factors that mimic TH cell subsets, five main groups of
γδ T cell subsets in humans do not have orthologues in mice44. Addi- ILCs have been classified: NK cells, ILC1s, ILC2s, ILC3s and LTi cells51.
tionally, the mechanisms of indirect phosphoantigen recognition by
human Vγ9Vδ2+ T cells through the butyrophilin family of molecules do NK cells. NK cells are circulatory, cytotoxic cells whose effector func-
not apply to γδ T cells that are found in mice, as they lack the required tion is mediated by a combination of activating and inhibitory recep-
butyrophilin molecules45. Further studies are required to understand tors, leading to the secretion of pro-inflammatory cytokines such
how these cells are regulated in health and disease across species. as IFNγ and TNF7. NK cells are identified as CD3−CD56+ cells, which
Furthermore, γδ T cells can rapidly respond to stimuli in an innate- are divided into two major subsets: (1) CD56dimCD16bright cells that are
like way, indicating their potential to initiate an immune response as mainly cytotoxic and circulatory and (2) CD56brightCD16− cells that pro-
first responders. Furthermore, γδ T cells can differentiate into TH1-like, duce cytokines and are tissue-resident52. The main activating receptors
TH2-like, TH9-like, TH17-like and regulatory T (Treg)-like cells (Fig. 1). of NK cells are natural cytotoxicity receptors (NCRs): NKp46, NKp44
Shaped by the local microenvironment as sensed through cytokine and NKp30 (also known as NCR1, NCR2 and NCR3, respectively), as
receptors, they produce prototypical cytokines such as IFNγ, TNF, IL-4, well as NKG2D (also known as CD314) and CD16. Inhibitory receptors
IL-10, IL-9, IL-17 and IL-22 (refs. 46–48). include NKG2A, T cell surface glycoprotein YE1/48 (ly49) and killer
immunoglobulin-like receptors (KIRs) on mouse and human NK cells,
Innate lymphoid cells regulating their cytotoxicity, cytokine production and proliferation,
ILCs as the ‘most innate’ cells of the innate-adaptive continuum are a respectively53. Although NK cells play an important role in antitu-
heterogenous population of lymphocytes. They represent the innate mour immune responses, they can also gain a regulatory function in
counterpart of T cells and are pivotal regulators of innate immunity, inflammation, infection and cancer by suppressing T cell proliferation
inflammation and tissue repair. ILCs are largely tissue-resident cells, through the production of IL-10 and adenosine54,55.
sensing perturbations within their surrounding microenvironment and
rapidly responding by secreting cytokines, making them one of the first Helper ILCs. Helper ILC1 cells are tissue-resident, are T-bet-dependent
responders in orchestrating an effective immune response49. All ILCs are and, similar to NK cells, secrete type 1 cytokines such as IFNγ and TNF56,57.
recombination-activating gene (RAG)-independent, lacking expression ILC1s are responsive to IL-15, IL-12, IL-18, bacteria and viruses49,58–60 and

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 354


Review article

parallel the TH1 arm of the adaptive immune system. Studies show that by the expression of the chemoattractant receptor homologous mole­
in both mice and humans, ILC1s show extensive heterogeneity and differ cule (CRTH2) and CD161 and express IL1R1 in certain tissues upon
in number and function between lymphoid and mucosal sites, high- activation62.
lighting the impact of tissue localization on the phenotype and function Among ILCs, ILC3s are the most abundant subset in the intestinal
of these cells61,62. An important distinction between NK cells and ILC1 tract of both mice and humans and mirror TH17 cells70. The ILC3 subset
is that NK cells depend on eomesodermin (EOMES) and T-bet for their is defined by the expression of RORγt and KIT (also known as CD117),
development and differentiation, whereas ILC1s only require T-bet for as well as the ability to produce IL-17A and/or IL-22. They can be further
their development and differentiation63. Although ILC1s were originally divided into NCR+ ILC3s, NCR− ILC3s and LTi cells71,72. Although both
deemed to be weakly cytotoxic, several studies show that cytotoxic ILC1 NK cells and ILC1 subset have been shown to exert cytolytic function,
subsets expressing granzyme B, granzyme C and perforin can mediate in vitro studies have demonstrated that human ILC3s can also have
cytotoxicity, contributing to immunosurveillance64–66. cytotoxic activity73.
The ILC2 subset as the innate TH2 counterpart produces type 2
cytokines IL-4, IL-5, IL-9, IL-13 and amphiregulin. They also depend on The role of ILCs and ILTCs in immunosurveillance
GATA3 and retinoic acid receptor-related orphan receptor-α (RORα) At the tumour initiation stage, cancer cells produce cytokines,
for their function and respond to IL-25, thymic stromal lymphopoi- chemokines, alarmins and haematopoietic growth factors in response
etin (TSLP) and IL-33 (ref. 67). Similar to ILC1, ILC2 has tissue-specific to carcinogenesis, resulting in either pro-tumour or antitumour immu-
phenotype and function68,69. In mice, ILC2s are the most homogeneous nity74. Successful immunosurveillance of malignant cells requires the
of the other subsets and express GATA3, as well as the IL-33 receptor coordinated action of tissue-resident and circulating cells of the adap-
(IL-1 receptor-like 1 (IL1R1), also known as ST2). Human ILC2s are defined tive and innate immune system for induction of an effective antitumour

Box 2

Tissue distribution and layered immunity of innate lymphoid and


innate-like T cells
Most of our knowledge about the tissue distribution and haptens312. Hapten sensitization has been shown to induce IL-7
establishment of the innate lymphoid cell (ILC) and innate-like T cell receptor α+ ILC1 memory in the liver, such that they can migrate into
(ILTC) tissue lymphoid compartment during early development is the draining lymph nodes to generate an allergic reaction313. Lung-
derived from studies in mice, in which the cells are found to populate resident ILC2s have also shown recall responses against cytokines
many tissues and barrier sites during fetal and perinatal periods11,306. such as IL-33 (ref. 314). The role of these memory-like ILCs in cancer
Beyond that, emerging studies show that the anatomical positioning remains largely unknown.
of ILCs in various tissues during development is explained by Early-life and tissue imprinting of ILCs and ILTCs equips these
waves of ILC development, in which multipotent and unipotent ILC cells with unique features that are tailored to their tissue-specific
precursors give rise to circulating ILCs, known as layered immunity307. niche11. Such early-life signals include microbial-derived and
The circulating ILCs can differentiate into any ILC subtype depending epithelial-derived signals that can shape the compartment of
on the tissue, and most tissue-resident ILCs are seeded during unconventional T cells with lifelong consequences315. This has been
the embryonic phase. Circulating ILCs are derived from the bone demonstrated for the development of mucosal-associated invariant
marrow and can replace the different subsets of tissue-resident ILCs. T cells, as they require exposure to commensal bacteria for proper
Therefore, the progenitor cells during the embryonic phase promote intrathymic development early in life295,296. The effect of this tissue-
generation of different ILC subsets in a site-specific manner described specific functional imprint on the role of ILCs and ILTCs in the tumour
as waves or layered immunity. The different waves of the various ILC microenvironment remains unknown. Although ILTCs are considered
subsets lead to an effective and organized multilayered composition mainly tissue-resident, they have been shown to migrate from their
of ILCs. As space in the tissues is limited168,308, ILTC occupation is tissue of residence to their nearest lymph node, while retaining
temporally restricted in tissue niches, as seen for mucosal-associated the tissue of origin-specific transcriptional imprint. This emphasizes
invariant T cells in the skin295 and γδ T cells in the intestine309. their cooperative role as a functional network in shaping the adaptive
Immunological memory is typically considered a feature of immune response316.
adaptive immunity and is characterized by a rapid response to The notion that ILCs and ILTCs act as a cellular network through
previously encountered antigens through the expansion of antigen- coordinated interactions at barrier tissue or tumour sites rather than
specific individual immune cell clones310. Although the majority of individual subsets is derived from the observation that animal models
studies so far have focused on the innate function of ILCs, emerging deficient for one ILTC subset compensate with increased numbers of
evidence shows the existence of memory features in these cells311. other ILTC subsets11,122,316. Nevertheless, the distinct localization and
For example, natural killer cells have been shown to have memory stratification of ILCs and ILTCs in tissues indicate that each population
responses to viruses and other small xenobiotic elements that may contribute uniquely to tissue homeostasis and response to
elicit immune response when combined with antibodies, termed tumours.

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 355


Review article

a Tumour suppression Permissive microenvironment


IL-15 and IL-12 Dendritic cell Macrophage Monocyte

ILC2 NCR3+
ILC3 IL-12, IL-15 and IL-18 (IL-2 and IL-7)

ILC1-
like
TCR signalling Indirect tumour killing
↑ Plasticity
Dying
IL-12R IL-18R IL-15R
tumour cell ↑ Cytotoxicity
↑ Cytotoxicity
MAIT iNKT γδ T

Direct tumour killing


TCR
γδ T
TCR ligand

γδ T
ADCC NK
MAIT
CD16

FASL FAS ↑ Apoptosis


↑ MHC-I Tumour IFNγ
Perforin iNKT
cell
iNKT Granzymes ↑ Pyroptosis

↑ Fibronectin 1
NK

TRAIL
↑ Remodelling of the TME
ILC1
MAIT ILC1 ↓ Metastasis

b Tumour promotion

Type 2 immunity IL-17-dependent tumour promotion

TGFβ
MAIT IL-10
ILC2 MAIT2 NKT2 γδ T
↑ Immunosuppression ILC2
γδ T ↑ Plasticity

IL-4, IL-5 and IL-13


ILC1-
iNKT like IL-22
ILC3
↑ Plasticity ↑ Proliferation

IL-13Rα IL-4Rα ↑ Metastasis


↓ Cytotoxicity MAIT17
↓ T-bet
↓ NKG2D
NK
↓ IFNγ
↓ NKp30
IL-17R IL-17A iNKT17

Suppressive microenvironment

TGFβ γδ T

↑ Angiogenesis

Treg CAF ↑ Metastasis +/– TCR signalling


Platelets

TCRVα24-Jα18 (human) TCRVα7.2-Jα33 (human) TCRVγ9Vδ2


TCRVα14-Jα18 (mouse) TCRVα19-Jα33 (mouse) (example)

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 356


Review article

Fig. 2 | Parallel and shared pathways prime pro-tumoural and antitumoural contribute to general immunosuppression in the TME and act directly on tumour
functions of innate lymphoid and innate-like T cells. a, Regulation of cells through cognate receptors (IL-13Rα and IL-4Rα) to increase proliferation and
antitumour function. In a permissive type 1 cytokine (largely IL-12, IL-15 and IL-18)- metastasis. Similarly, type 17-polarized ILC and ILTC subsets secrete IL-17A and
primed environment, innate lymphoid cells (ILCs) and innate-like T cells (ILTCs) IL-22 to increase metastasis by acting on the stroma to promote angiogenesis or
are antitumour-polarized. Direct tumour cell-killing mechanisms are mediated directly on tumour cells to increase proliferation. Within the TME, transforming
through granzymes and perforin, as well as –FAS ligand (FASL), tumour necrosis growth factor-β (TGFβ) secreted by regulatory T (Treg) cells, platelets or cancer-
factor-related apoptosis-inducing ligand (TRAIL) or antibody-dependent cellular associated fibroblasts (CAFs) can act on ILCs and ILTCs, leading to further
cytotoxicity (ADCC). IL-15 and IL-12 have been shown to influence ILC plasticity immunosuppression (for example, enhanced secretion of IL-10) or loss of
leading to transdifferentiation of NCR+ ILC3 and ILC2 to a more cytotoxic ILC1-like cytotoxicity. In addition, TGFβ secreted by ILTCs themselves can further contribute
phenotype, leading to tumour cell death. ILTCs can also be primed for enhanced to an immunosuppressive microenvironment. One such immunosuppressive
cytotoxicity through T cell receptor (TCR) signalling, leading to indirect tumour effect of TGF-β is the conversion of cytotoxic natural killer (NK) cells into
killing through secretion of interferon-γ (IFNγ). They are primed through common non-cytotoxic ILC1s (also termed plasticity) as seen by the downregulation
cytokine receptors, IL-12 receptor (IL-12R), IL-15R and IL-18R, leading to tumour of transcription factor T-bet, NKG2D, NKp30 and pro-inflammatory IFNγ.
cell apoptosis, pyroptosis and tumour microenvironment (TME) remodelling DC, dendritic cell; iNKT, invariant natural killer T; MAIT, mucosal-associated
leading to decreased rates of metastasis. b, Pro-tumoural functions of ILCs and invariant T; MHC, major histocompatibility complex; NCR3, natural cytotoxicity
ILTCs: type 2 primed ILCs and ILTCs can secrete IL-4, IL-5 and IL-13, which can receptor (also known as NKp30); NKG2D, natural killer group 2, member D.

response75. ILCs and ILTCs as tissue-resident lymphocytes are abundant Direct tumour cell killing. ILCs and ILTCs utilize shared programmes
at many tumour sites and can sense cytokines and alarmins through to mediate direct tumour cell killing. Unlike conventional cytotoxic
cognate receptors, prompting the swift secretion of pro-inflammatory CD8+ T cells, ILCs and ILTCs do not require tumour-derived neoantigen
or tissue-protective factors appointing them as the first responders presentation on human leukocyte antigen (HLA) molecules to elicit
in the TME64,76. The functional properties of these cells in the TME are antitumour immunity82 and are therefore an attractive target for cancer
context-dependent with key-shared adaptation programmes in immunotherapy.
response to the microenvironment. Parallel pathways of activation In general, ILCs and ILTCs induce tumour cell death after inte-
by tumour-derived signals and pro-inflammatory cytokines can trigger gration of activating and inhibitory receptor signals, shifting their
antitumour transcriptional profiles in ILCs and ILTCs64. Alternatively, the effector repertoire towards cytotoxicity83,84.One such pathway is
hostile TME can highjack their potential in tissue repair and homeostasis, well studied for NK cells as they can eliminate cancer cells through
leading to their polarization towards tumour promotion3 (Fig. 2). The the so-called ‘missing-self’ mechanism. However, a unique feature
complexity in dictating pro-tumour or antitumour function explains of ILTCs is that through the expression of their (semi-invariant) TCR,
why different clinical and preclinical studies have associated ILC and ILTCs can exert TCR-dependent antitumour immunity, setting them
ILTC subsets with seemingly paradoxical outcomes. aside from ILCs84. Stimulation and activation of unconventional
Subsequently, we outline how the balance between these con- T cells with activating ligands have been shown to induce potent
served signals within the TME influences ILCs and unconventional antitumour immune responses in iNKT cells85, MAIT cells86,87 and γδ
T cells as a network of cellular mechanisms and highlight how each T cells88. For example, type I NKT cells can be activated by glycolipid
subset serves a unique purpose in the cancer niche. We discuss subse- ligand α-GalCer to mediate in vitro and in vivo antitumour effects in
quently how the presence of pro-inflammatory cytokines that affect a CD1d-restricted manner85,89. We and others have shown that mouse
all subsets of ILCs and ILTCs can lead to effective antitumour responses MAIT cells can be activated and expanded in vivo by the TCR-agonistic
(Fig. 2). Tumour cells and other cells in the TME (for example, cancer- bacterial vitamin B2-derivate 5-OP-RU (5-(2-oxopropylideneamino)-6-d-
associated fibroblasts or Treg cells)77 can secrete TGFβ, IL-17 and other ribitylaminouracil), leading to reduced tumour burden in models
cytokines that can lead to phenotypic changes in ILCs and ITLCs, induc- of lung and liver metastasis86,87. γδ T cells and iNKT cells are capable of
ing a tumour-promoting phenotype (Box 3). In Box 3, we outline the recognizing tumour cells through their TCR or by expressing activating
concept of cellular plasticity, which enables these cells to act along receptors such as NKG2D, NKp30 or NKp44 (refs. 83,84,90).
the full spectrum of the immunoregulatory axis. NK, iNKT, MAIT and γδ T cells can directly lyse target malignant cells
in vitro through the release of preformed cytolytic granules such
Antitumour function as granzyme B and perforin85,91–93. Interestingly, studies of NK-cell-
The response of ILCs and ILTCs to tumours is context-dependent, as mediated tumour cell killing in vitro show that granzyme-B-dependent
pro-inflammatory cytokines released in the TME induce a distinct cytotoxicity occurs in the early events, whereas subsequent tumour
transcriptional profile and a characteristic effector function in these cell eliminations are preferentially induced through engagement
cells. Numerous in vivo studies using genetically deficient mice or of death receptors expressed on NK cells such as FAS ligand (FASL;
depletion antibodies to NK, iNKT and γδ T cells have demonstrated the also known as CD178) and TNF-related apoptosis-inducing ligand
tumour-protective effect of NK, NKT and γδ T cells78–80. Although (TRAIL)94.
the function of these cells can generally span the continuum between TRAIL is a member of the TNF superfamily that is expressed on
tumour promotion and destruction, respective cytotoxic ILC and ILTC NK cells, ILC1, ILC3 (ref. 95), iNKT, γδ and MAIT cells and can induce
subsets are equipped with an arsenal of potent mechanisms that can apoptosis in tumour cells96,97. Intrahepatic and peripheral blood ILC3s
restrict tumour growth and spread of cancerous cells6,9,81,82. Subse- can directly kill hepatic tumour cells through increased IFNγ produc-
quently, we outline microenvironmental determinants for successful tion and TRAIL expression in vitro95. The TRAIL-dependent cytotoxic
ILC-mediated and ILTC-mediated antitumour immunity, highlighting capability of ILC1 and ILC3 cells suggests the potential of targeting
shared mechanisms and pathways that lead to direct or indirect tumour these cells as antitumour effector cells among the other cytotoxic ILC
inhibition (Fig. 2a). and ILTC subsets.

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 357


Review article

Box 3

Plasticity of innate lymphoid and innate-like T cells in the tumour


microenvironment
Specialized subsets of innate lymphoid cells (ILCs) and innate-like patients with hepatocellular carcinoma154. In patients with squamous
T cells (ILTCs) are characterized by the expression of key transcription cell carcinoma, IL-23 production by the tumour cells induced the
factors determining their effector function. An adequate response conversion of ILC1 into IL-17-producing ILC3 cells, promoting tumour
to tissue-derived stimuli requires flexibility and rapid adaptation of cell proliferation143. Pro-inflammatory IL-12 induced differentiation of
each subset, which is achieved through transdifferentiation between NKp46+ retinoic acid receptor-related orphan receptor-γt (RORγt)+
the cellular subsets and the acquisition of altered phenotypes and ILC3s into interferon-γ-producing ILC1 cells in a mouse model of
function. This plasticity phenomenon has been studied for ILCs in melanoma323. The transdifferentiation of ILC3s into ILC1 subsets was
detail and reviewed comprehensively recently317,318. Although the also recently demonstrated in human colorectal cancer samples and
classical distinction of ILCs provides a useful framework, numerous mouse models of colorectal cancer199.
studies have confirmed that their heterogeneity and complexity As unconventional T cells egress the thymus with predetermined
extend beyond the original classification (natural killer (NK) cell, memory or effector phenotypes and commitment to type 1, type 2
ILC1, ILC2 and ILC3 subsets). ILCs can change their function and or type 17 lineage (Box 1), their potential for functional plasticity in
phenotype in response to signals in their microenvironment317, the TME is more restricted in comparison to ILCs16. However, several
similar to the differentiation of naive CD4+ T cells319, macrophages studies point towards the likely ability of mucosal-associated invariant
and neutrophils320. Transcriptional analysis of ILCs has shown the T cells5, NK T cells324 and γδ T cells325 to alter their effector phenotype
presence of transitional states between major subsets both under upon microenvironmental changes. One might ask whether
homeostasis and pathological conditions321. Although most studies unconventional T cells truly shift between TH17-like and TH1-like
investigating ILC plasticity have been carried out in the context of phenotypes or whether the outgrowth of one subset versus the other
inflammatory diseases, similar pathways and cytokines can dictate explains their altered functionality in the TME. Fate-mapping studies
ILC transdifferentiation in the tumour microenvironment (TME). For in mice or the development of conditional knockout strains for these
example, in a mouse model of fibrosarcoma, transforming growth ILTC subsets will provide further insight into this challenging question.
factor-β induced the conversion of cytotoxic NK cells from the Nevertheless, in the context of many cancers, an understanding of the
surrounding non-tumour tissue into a non-cytotoxic ILC1 subset developmental relationships and degree of plasticity of ILC and ILTC
in the tumour60,322, a phenotype that was later demonstrated in subsets remain an important area for future investigations.

ILCs and ILTCs can also directly kill tumour cells in vitro and in vivo tumour cell apoptosis and pyroptosis108. The paramount role of IFNγ
through FAS (also known as CD95–FASL) engagement94,98. Unlike in mediating both ILC and ILTC antitumour immunity has been dem-
iNKT cells, NK cells mediate cytotoxicity through expression of perforin onstrated in various settings and across different in vivo models and
and granzyme, highlighting the non-redundant roles of iNKT cells98,99. tumour types86,109,110. Nevertheless, impaired IFNγ production of ILCs
Finally, the Fc regions of IgG antibodies on opsonized tumour cells and ILTCs is frequently observed in advanced cancers representing an
(antibody opsonization) bind to CD16 molecules expressed on NK important immune escape mechanism111.
cells and unconventional T cells initiating antibody-dependent cellular
cytotoxicity (ADCC)100–103. IL-15, IL-12 and IL-18 regulate ILC and ILTC antitumour function. ILC
and ILTC proliferation, maturation and antitumour function are tightly
Indirect antitumour effects through interferon-γ. IFNγ is an impor- governed by the presence of pro-inflammatory cytokines in the TME112.
tant pleiotropic cytokine with immunomodulatory effects on both The presence of IFNγ in the TME primes inflammatory myeloid cells
innate and adaptive immune responses104. In the local TME, respective such as macrophages, monocytes and dendritic cells (DCs) to produce
TH1-polarized subsets (NK cells, ILC1s, iNKT1 cells, MAIT1 cells and IL-12, IL-15 and IL-18 cytokines driving TH1-type immunity112. This in turn
γδ T cells) can produce large amounts of IFNγ as a rapid and first induces cytotoxicity in ILCs and ILTCs that express high levels of the
response to tumour growth79,105–107. Such functional redundancy likely corresponding receptors9.
allows for the robustness of defence against tumours and puts constant Although ILCs and ILTCs share the aforementioned common
pressure on cancer cells through maintained immunosurveillance. The response to a pro-inflammatory cytokine milieu, the unique features
first-responder capacities of ILTCs and ILCs are consistent with what of each subset allow for non-redundant responses in the TME. The
has been reported in in vivo infection settings, in which ILC1s were the ability for simultaneous integration of TCR-mediated and cytokine-
main early source of IFNγ in response to IL-12 in virally infected tis- induced signalling in ILTCs adds another layer of complexity to our
sue limiting viral replication49. As a result, their IFNγ secretion in the understanding of their function in the TME. Moreover, TCR ligands
tumour niche likely establishes the antitumoural cytokine milieu at (derived from bacteria or tumour cells) in synergy with cytokines can
early stages of tumour growth, creating a pro-inflammatory environ- promote distinct pro-inflammatory responses in vitro and in vivo87,113–115.
ment9. Similarly, acting directly on tumour cells, IFNγ signalling leads The response to pro-inflammatory cytokines is by no means uni-
to the upregulation of MHC-I and MHC-II, which can directly promote form across ILC and ILTC subsets. IL-12 alone or in combination with

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 358


Review article

IL-15 and IL-18 has been shown to activate NK cells9; however, IL-12 has Chronic IL-17 secretion by ILCs and ILTCs contributes to several
even more pronounced effects on helper ILC subsets, inducing IFNγ mechanisms in the TME, ranging from tumour initiation to tumour
production by ILC1 and ILC3 cells49,56,116. progression. For instance, IL-17-secreting γδ T cells accelerated
Mechanistically, binding of IL-15 on its receptor expressed on pancreatic intraepithelial neoplasia in Mist1-CreERT2;KrasG12D mice
NK, iNKT, γδ T and MAIT cells induces an intracellular signalling by direct signalling through the IL-17R on pancreatic acinar cells, which
cascade, leading to the accumulation of T-bet in the nucleus30,117,118. accelerated pancreatic intraepithelial neoplasia initiation and progres-
Consequently, IL-15-primed ILCs and ILTCs produce high levels of sion to invasive disease139. γδ T cell-derived IL-17 has also been shown
granzyme B117 and IFNγ119,120, leading to enhanced cytotoxicity towards to act directly on endothelial cells leading to enhanced angiogenesis,
cancer cells121. which subsequently led to tumour development in mouse models of
In recent years, increasingly complex in vivo, ex vivo and in vitro CMS-G4 fibrosarcoma and HPV-induced K14-HPV16 (refs. 140,141).
studies have shed light on how fine-tuned intercellular communica- Using mouse models of lung metastasis and subcutaneous tumour
tion loops shape ILC and ILTC function and phenotype in inflamma- growth, MAIT cells were found to exert tumour-promoting function
tory, infectious and neoplastic processes122. The importance of the through IL-17A leading to a lower tumour burden in MAIT cell-deficient
IL-12–IFNγ circuit is shown by the plasticity of ILC subsets in which Mr1−/− mice compared with WT mice 142. It seems possible that
in both in vivo and in vitro models, IL-12 and IL-18 mediate the trans- IL-17-secreting iNKT type 17 cells show tumour-promoting effects in
differentiation of NCR+ ILC3s into ILC1-like cells (that is, ex-RORγt+ a similar fashion4. Interestingly, studies in mouse models of HCC and
ILC3s) that secrete IFNγ and are cytotoxic58,123,124. It has been shown patients with squamous cell carcinoma show that in an IL-23-rich TME,
that RORγt+ ILC3s, when cultured with IL-2, IL-15 and IL-23, secrete IFNγ, which induces IL-17 production, ILC1s can differentiate into RORγt-
along with upregulation of T-bet, gaining ILC1 characteristics125. In the expressing NCR− ILC3 cells143,144. Additionally, human KIT+CCR6+ ILC2
context of malignancy, IL-12 (ref. 126) secreted by antigen-presenting cells can differentiate into IL-17-secreting NCR− ILC3s and upregulate
cells (APCs) amplifies IFNγ production in NK and iNKT cells127, as seen RORγt when cultured with IL-1β, IL-23 and TGF-β145, pointing to the
in 4T1 mouse models of breast cancer treated with recombinant IL-12, effect of the TME on the plasticity of these cells. ILC3 and LTi cells are
which rescued the anti-metastatic function of NK cells128. In patients major sources of IL-22, which has been shown to have a similar role to
with chromophobe renal cell carcinoma who showed prominent infil- IL-17 in the TME146,147. IL-22 protects tissue barriers against microorgan-
tration of tissue-resident CD56bright ILC1 cells, IL-15 secreted by the isms and plays an important role in intestinal homeostasis. However,
chromophobe renal cell carcinoma tumour cells activated granzyme IL-22 can also induce inflammation and result in tumour growth, as
cytotoxic ILC1 cells, emphasizing an IL-15-dependent, antitumour seen in a bacteria-induced colon cancer model146,148. Another study
function of ILC1 cells129. Given this, it is likely that ILCs and ILTCs act in showed that IL-22 can regulate cancer stem cells in CRC to promote
a synergistic manner as both initiators of an inflammatory response colon cancer149.
at early neoplastic events and signal amplifiers of an already ongoing
protective, antitumour signalling cascade. TGFβ-rich environments impair ILC and ILTC antitumour function.
TGFβ is an essential and pleiotropic cytokine produced within the
Tumour-promoting function TME of many solid tumours by tumour cells, Treg cells, fibroblasts,
Successful immune evasion is characterized by the depletion of antitu- macrophages and platelets150. Although TGF-β is a regulator of homeo-
moural immune cell subsets replaced by the accumulation of exhausted stasis, perturbations in its expression within the TME are an important
and/or immunosuppressive leukocytes. Defects in antigen presentation mechanism of tumour immune evasion and poor response to anti-
machinery, altered chemokine and cytokine milieu and metabolic alter- tumour therapy151. Within the local TME, ILCs and ILTCs can be both
ations negatively impact ILC and ILTC function in the TME. By secreting effectors and producers of TGF-β. TGF-β signalling affects NK cells
distinct combinations of effector molecules (for example, IL-4, IL-10, in vivo at a transcriptional level by silencing T-bet expression and IFNγ
IL-17 or TGF-β), ILCs and ILTCs can dampen the innate and adaptive secretion152,153. Interestingly, in primary and metastatic mouse models
immune responses against tumours. Subsequently, we highlight how of fibrosarcoma and lung metastasis, as well as in patients with HCC,
ILCs and ILTCs share a largely overlapping functional niche in tumour- TGF-β in the TME caused a shift of NK cells into less cytotoxic inter-
promoting immunity, emphasizing their unique and non-redundant mediate ILC1 and ILC1-like cells, which were unable to control tumour
functions (Fig. 2b). growth and metastasis. This demonstrated that a TGF-β-rich TME
can shape and modulate the cellular plasticity of the ILC subsets 60,154.
IL-17A-secreting ILCs and ILTCs promote tumour growth. High levels Similar mechanisms of immunosuppression by TGF-β were shown in γδ
of IL-17A in the blood of patients with cancer correlate with poor sur- T cells and iNKT cells, in which TGF-β also contributed to the induction
vival in various patient cohorts, highlighting its role in cancer develop- of IL-17-producing human Vγ9Vδ2+ T cells47 and to both the develop-
ment and progression130. In mouse models of hepatocellular carcinoma ment and peripheral responsiveness of type 17 iNKT cells155. In addition
(HCC)131, as well as breast cancer132, lung cancer133 and colorectal cancer to their response to TGF-β-signalling in the TME, ILTCs such as γδ T
(CRC)134, IL-17A deficiency has been shown to reduce tumour burden135. and MAIT cells can secrete TGF-β in vivo contributing to an immuno­
Experimental findings derived from mouse models should be inter- suppressive feedforward loop156–158. Although the relative contribution
preted with caution, as there are important differences among species. of ILC-derived and ILTC-derived TGF-β to immune evasion in cancer
For example, in humans, in some contexts, tissue-resident Vδ1+T cells remains to be determined, these findings provide further insights
have been found to preferentially produce IL-17A compared with their on how the ILC and ILTC network could act as signal amplifiers in the
circulating Vγ9Vδ2+ counterparts136, whereas in mice Vγ4+ or Vγ6+ hostile TME. Dissecting the role of TGFβ in orchestrating the function
T cells133 produce high levels of IL-17A. Furthermore, Vδ1+T cells can also and plasticity of the ILCs and ILTCs in the TME will shed light on how
be found to predominantly secrete IFNγ, as demonstrated for human differential regulation of this pivotal cytokine leads to cooperation or
studies of breast cancer and non-small-cell lung cancer (NSCLC)137,138. competition among the different innate subsets.

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 359


Review article

Type 2 immunity leads to pro-tumorigenic ILCs and ILTCs. Type 2 determinants of immune responses within tissues. In this section, we
immunity is characterized by the production of IL-4, IL-9 and IL-13 and aim to demonstrate the similarities and synergisms across the different
is responsible for maintaining homeostasis and tissue repair159. The ILC and ILTC subsets during this crosstalk.
dysregulation of type 2 immunity has been shown to promote tumour
proliferation and metastasis160. The pro-tumoural role of ILC2s is attrib- Antitumour response in a TH1 environment
uted mainly to the production of IL-4, IL-5 and IL-13 upon stimulation by ILCs and ILTCs, as a relay of potentially pro-inflammatory cells, can con-
epithelial cytokines such as IL-33, IL-25 and TSLP69,161. Mechanistically, in tribute to successful immunosurveillance through initiation and ampli-
the subcutaneous 15-12RM fibrosarcoma tumour model, iNKT-derived fication of an antitumour TH1-like immune response in a permissive
IL-13 was found to activate the IL-4R–STAT6 axis and promote tumour microenvironment64,86,87,107,169,170 (Fig. 3a).
recurrence162. Chronic stimulation of MAIT cells, by CD3 and CD28 plus ILCs and ILTCs can directly sense danger-associated molecular
IL-2 and IL-7, leads to IL-4, IL-5 and IL-13 production, which can induce patterns through cognate receptors. These appear in the TME dur-
phosphorylation of the nuclear transcription factor STAT6 (pSTAT6) ing immunogenic cell death, especially in early cancers171. Beyond
signalling in CRC cells in vitro163. that, iNKT and MAIT cells show unique features as they can sense
The dichotomy of the pro-tumour versus antitumour effect of pathogen-associated molecular patterns derived from intratumoural
TH2 signalling is organ site-dependent, stage-dependent and context- microorganisms through their TCR, if presented by CD1d or MR1 mole­
dependent for ILC2s and type 2 ILTCs164,165. Whether ILC2s and type 2 cules, respectively172. Both in vitro and in vivo, the pro-inflammatory
ILTCs can promote or suppress tumour progression relies extensively cytokines polarize ILCs and ILTCs leading to an optimal antitumour
on their tissue environment. It is clear now that our current under- response characterized by high IFNγ expression and a TH1 transcrip-
standing of these cells underestimates the vast spectrum of effec- tion factor profile6,173–175. IFNγ-signalling on macrophages and DCs in
tor functions and therefore does not yet sufficiently capture their the TME potentiates this antitumour effect through maturation of M1
heterogeneity within this group. macrophages and differentiation of DCs towards type 1 conventional
DCs (cDC1s)176. NK, γδ T and iNKT cells can recruit cDC1s177, which in turn
TCR-dependent tumour promotion. The ability of unconventional induces TH1 polarization of CD4+ helper T cells and thus leads to a CD8+
T cells to sense environmental and endogenous ligands through cytotoxic T cell response178. IFNγ as a central mediator in this circuit
their conserved TCR distinguishes them from ILCs and con­ventional then leads to improved antigen presentation capacities by the DCs
αβ T cells. TCR-derived signals are integrated with cytokine and as seen in samples of patients and mouse models. This in turn leads
chemokine signals, prompting ILTCs to have pro-tumoural or DCs to secrete more IL-12 and increases NK cell activity179. In a similar
antitumoural effector functions87,113–115. fashion, iNKT, MAIT and γδ T cells have been found to enhance NK cell
In mouse models of B16-F10 and LWT1 melanoma lung metastases, cytotoxicity in an IFNγ or CD137-dependent way86,169,180,181.
MAIT cells show tumour-promoting functions when MR1 is expressed Local conditioning of the TME cytokine milieu by ILCs and ILTCs
on tumour cells, which can be reversed by inhibitory MAIT ligands further shapes antigen-specific adaptive immunity demonstrated
Ac-6-FP or MR1 blocking antibodies142. This indicates that endoge- in vivo and in vitro87,182. Further connections to adaptive immunity
nous or microbial MAIT ligands exist in the TME, polarizing mouse with potential for an improved tumour control in human and mouse
MAIT cells towards tumour promotion in a TCR-dependent manner. exist, as ILCs and unconventional T cells can provide B cell help towards
Type II NKT cells were found to promote in vivo tumour growth in class switching of immunoglobulins, formation of germinal centres
several tumour mouse models166, and their activation by the proto- and shaping of peripheral B cell populations183–186. Although not yet
typical agonist sulfatide suppressed tumour immunosurveillance167. shown in the context of cancer, it seems likely that similar mechanisms
Functional studies on γδ T cells in mouse models of melanoma also might apply to the TME.
suggest that TCR engagement is essential for the tumour-promoting
effects of γδ T cell-produced IL-17 within the TME140. ILCs and ILTCs recruit antitumour myeloid cells
These preliminary findings on ILTCs in the tumour setting high- Emerging studies show that ILC and ILTC subsets also share similar
light how TCR signalling can shape their effector function through pathways in recruiting innate immune cells such as macrophages or
poor or null agonists and antagonists. The relative proportion of these DCs, which in turn are crucial for the induction of potent adaptive
agonistic and antagonistic signals, as well as co-stimulatory molecules, antitumour immunity75. Tumour-infiltrating ILC2s secrete IL-5 and
may vary between different primary tumour sites, tumour stages and GM-CSF recruiting eosinophils, which can subsequently secrete chemo-
microbial composition of the local TME. attractants such as CCL5, CXCL9 and CXCL10. In both mouse models of
lung cancer187 and melanoma188, this was shown to facilitate the recruit-
Bridging adaptive and innate immunity ment of CD4+ and CD8+ T cells to the tumour site. In mouse models of
Even though ILCs and ILTCs can serve as direct effectors of pro-tumoural pancreatic ductal adenocarcinoma, tumour-infiltrating ILC2s were
or antitumoural immune responses, their predominant function lies shown to activate CD103+ DCs and CD8+ T cells to induce antitumour
in their roles as orchestrators of tumour immunity. These cells bridge immunity, which was amplified using anti-programmed cell death
innate and adaptive immunity by initiating, amplifying, inhibiting or protein 1 (PD-1) blockade189.
dampening the immune response in the TME. It is increasingly clear The strong interconnectivity between ILTCs and the myeloid cell
that ILCs and ILTCs mount an innate immune response influenced by compartment is well established. For instance, MAIT cells are one of
the inflammatory signals in the TME. Thus, ILCs and ILTCs engage in the major and early producers of GM-CSF in infection settings, and
complex crosstalk with elements of the innate and adaptive immune this MAIT cell-dependent GM-CSF release in vitro and in vivo induces
system, as well as the microenvironment122,168. In turn, innate and differentiation of CCR2+ inflammatory monocytes into DCs, leading to
adaptive immunity reciprocally regulates ILCs and unconventional activation of effector T cells. This can occur both in an MR1-dependent
T cells, indicating that such cellular interaction networks are crucial and MR1-independent manner190. iNKT cells have also been shown to

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 360


Review article

bridge the innate and adaptive immune system by modulating DC T cells and type I NKT cells19,212. Expression of IL-4 on both human and
and macrophage function in both infection and tumour settings. mouse γδ T cells induced proliferation of IL-10-producing Vδ1 T cells, sup-
Tumour cells engineered to secrete GM-CSF induced the expansion pressing the function of the Vδ2 T cells165,213. Tuft cells, epithelial chem-
of CD1d-restricted T cells and maturation of DCs. This was abolished in osensory cells that reside in the epithelial lining of the intestine, express
CD1d− mice, emphasizing the role of NKT cells in interaction with DCs IL-25 under homeostasis, infection and inflammation. IL-25 derived from
and shaping the antitumour immunity191. tuft cells has been shown to further activate ILC2s to secrete IL-13, which
can mediate type 2 immune responses in the intestine214,215. IL-25 has also
ILCs and ILTCs as APCs been shown to have a pro-inflammatory role in colitis, inducing secretion
ILCs and ILTCs as part of their fundamental role in connecting the innate of IL-13 by NKT cells214,215. This reinforces the notion that through differ-
and adaptive arms of the immune system can also act as professional ent pathways, TH2 cytokines, particularly IL-13, released by ILC2s and
APCs through expression of MHC-II to modulate conventional αβ CD4+ ILTCs can have a role in mediating tumour progression. The differential
T cell responses. Changes in ILC3 expression of MHC and co-stimulatory stimulation of ILC2s by IL-33 and IL-25 is tissue-dependent, suggesting
molecules induced by inflammatory cytokines can either reinforce that the function of these cells differs between cancer types.
their regulatory role in tissue homeostasis or convert them into pro-
inflammatory ILCs inducing T cell activation192. In intestinal homeo- Type 17-dependent tumour promotion
stasis, MHC class II+ ILC3s limit inflammation through inhibition of Another multicellular programme in pro-tumour immunity, in which
microbiota-specific effector CD4+ T cell proliferation192,193; however, the combined ILC and ILTC community bridges innate and adaptive
upon intestinal inflammation, ILC3s upregulate co-stimulatory immunity, is the type 17 response, in which the canonical cytokine IL-17A
molecules to induce TH1 cell responses194. Recently, several groups furthers tumour progression (Fig. 3c). The presence of TGF-β, IL-1β, IL-6,
have shown that MHC II+ RORγt-expressing cells are responsible for IL-7 and IL-23 secreted by inflammatory macrophages, neutrophils or
generating RORγt-expressing peripheral Treg cells in the gut, promot- DCs in response to endogenous or exogenous (microbial) signals in
ing tolerance to microbiota. In human CRC tissue, intestinal ILC3s have the TME induces TH17 polarization of ILC3s, iNKT cells, γδ T cells and
been shown to gain antigen-presenting function by upregulation of MAIT cells133,216.
HLA-DR and co-stimulatory molecules IL-1β and IL-18. This is inhibited Blocking, depleting or genomically ablating these cytokines leads
by TGFβ195–198. to a reduction in the number of IL-17+ ILCs and ILTCs in mouse models
ILC3-specific deletion of MHC-II in mice promoted CRC tumour of breast cancer132,217, fibrosarcoma140,218, melanoma219 and gastric
growth and resistance to anti-PD1 immunotherapy, suggesting that cancer220,221. Notably, cellular crosstalk between IL-17A-producing
MHC-II+ ILC3s provide a protective role in CRC199. γδ T cells can also ILCs or ILTCs222 and myeloid cells is bidirectional, such that Vγ6+ T cells can
present microbial antigens and induce proliferation of naive CD4+ also mobilize peritoneal macrophages with pro-tumorigenic potential223.
T cells, similar to DC activation of αβ T cells200. In patients with gastric Microbial dysbiosis, a common feature for many cancers224, is likely
cancer, tumour-activated γδ T cells behaved as APCs, upregulating another key factor for IL-17-mediated ILC and ILTC tumour promo-
MHC-II and expressing co-stimulatory molecules to induce CD4+ and tion. In a mouse model of lung adenocarcinoma, γδ T cells were the
CD8+ αβ T cell proliferation and subsequent tumour cell death201. Taken predominant IL-17-producing cells driven by tumour-associated micro­
together, these studies underline the important role of MHC-II expres- biota133. This increased the infiltration of tumour-associated neutro-
sion on ILCs and ILTCs in direct, cell-contact-dependent regulation of phils and led to enhanced tumour progression. Similarly, ILC2s have
the adaptive immune cells in tissue homeostasis and cancer. been shown to recruit tumour-associated neutrophils to the TME in
either a CXCL1-dependent or CXCL2-dependent manner, albeit this
Type 2-mediated immune evasion study does not link these findings to microbial dysbiosis225. Another
Perturbations and inflammation in the context of cancer can induce the potential connection to the microbiome was recently demonstrated, as
release of alarmins such as IL-25, IL-33 and TSLP, which regulate type 1 IL-17A-producing MAIT cells promoted tumour growth in mouse models
and 2 immune responses202. The receptor for IL-33 and IL1R1 is present of lung metastasis in an MR1-dependent manner142. Similarly, when
on ILC2s, iNKT cells, MAIT cells and γδ T cells203–205 and can potentially MAIT cells are co-cultured with Escherichia coli antigen-exposed human
prime the cells to bridge inflammatory and danger signals in their sur- breast cancer cell lines, they show enhanced MR1-dependent IL-17A
roundings to the adaptive immune system (Fig. 3b). In several models production142,226. Comparably, IL-17A from conventional CD4+ TH17 cells
of lung metastasis, ILC2s activated by IL-33 recruit eosinophils in an and innate-like γδ17 T cells promoted tumorigenesis in bacteria-induced
IL-5-dependent manner, resulting in tumour metastasis 206. IL-5 CRC models227. ILC3s as the TH17 counterpart of the ILC family are par-
produced by iNKT cells207, γδ T cells208 and MAIT cells163 could poten- ticularly abundant at intestinal sites and have been shown to promote
tially follow a similar mechanism for the recruitment of eosinophils colonic carcinogenesis partly through IL-17-mediated sustained inflam-
and induction of type 2 immunity. mation in response to IL-23R signalling220. Nevertheless, the question of
The pro-tumorigenic role of ILC2s and TH2 type ILTC subsets is how local microbial communities within the TME shape phenotype and
mainly through release of cytokines such as IL-5 and IL-13. ILC2s secrete function of ILCs and ILTCs remains an active field of research.
IL-13 to promote accumulation of suppressive myeloid derived suppres- Finally, ILC-derived and ILTC-derived IL-17A has also been shown
sor cell (MDSC), which has been shown to regulate antitumour immu- to inhibit NK and cytotoxic CD8+ T cell function in vivo86,142,144, favour-
nity in patients with acute promyelocytic leukaemia and mouse models ing tumour progression. IL-17A production by ILC3 and γδ T cells also
of breast cancer209,210. ILC2s also produce amphiregulin, which activates promotes CXCL5 secretion by tumour cells, leading to the recruitment
Treg cells and therefore induces a suppressive microenvironment211. of immunosuppressive CXCR2+ MDSCs131,144. Further indirect CD8+ T cell
NKT cells also trigger the IL-13–MDSC axis, in which secretion of inhibition could occur through the IL-17A-dependent recruitment of
IL-13 by type II NKT cells induces TGF-β secretion by Gr1+CD11b+ MDSCs tumour-associated neutrophils132, as demonstrated for γδ T cells in
through the IL-4R–STAT6 pathway. This results in suppression of CD8+ mouse models of breast cancer132.

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 361


Review article

a ↑ CD40 Immunogenic cell death,


↑ Maturation early cancer and carcinoma in situ ↑ Antigen presentation
↑ CD80 ↑ cDC1 differentiation
↑ M1 > M2 polarization
↑ Antigen presentation ↑ GC formation
DAMPs, PAMPs, alarmins, intratumoural microbiome
↑ MHC class II Dendritic cell
Macrophage M1
IL-12 TH1-polarized IL-12 and IL-18
and
IFNγ
IL-18
ILC1 NK iNKT1 MAIT1 γδ T
↑ Maturation B cell
↑ Class switch
TCR
TH1 cytokines (IFNγ and TNF)
Tumour-specific
antibodies
↑ Leukocyte migration
↑ TH1 polarization
↑ Recruitment ↑ Effector function
↑ Cytotoxicity CD8 + Tumour Direct tumour
NK CTL cell
↑ Maturation killing ↑ T-bet
FAS FASL TCR
CD4+
Granzymes TH1
↑ TAA presentation
Perforin
↑ Apoptosis
Cancer death cell ↑ Pyroptosis

Tuft cell
b CAFs

↑ Recruitment IL-13
↑ M1 < M2 polarization IL-25
↓ Antigen presentation
Epithelial-derived alarmins (e.g., IL-33, IL-25 and TSLP)
↑ ARG1 chronic stimulation
PDL1 IL-13
↑ MMP9
↑ TGFβ

IL-13 and CD4+


Macrophage M2 ILC2 T cell
IL-4 iNKT2 MAIT2 γδ T

IL-4
B cell TH2 cytokines (IL-13, IL-4, IL-5, IL-9 and AREG)
IL-5 and
GM-CSF IL-13 IL-5, AREG
MDSC Treg
↑ Recruitment γδ1 T
IL-10
↑ Recruitment ↑ Recruitment
↓ Tumour cell killing
↓ Cytotoxicity
↓ IFNγ production CD8+
Eos NK CTL γδ2 T
Immune evasion

c
Dendritic cell Macrophage Neutrophil MDSC
CXCL5
CXCR2

↑ Recruitment
↑ Microbial dysbiosis ↑ T cell inhibition
IL-1β, IL-6, IL-7, IL-23 and TGFβ
5-OP-RU or
endogenous (?) CD8+
microbial (?) TCR CTL ↓ Tumour cell killing
ligands
Tumour IL-17 and IL-22 ↓ Cytotoxicity
ILC1 Tumour
MAIT17 NKT17 γδ T ↓ IFNγ production
cell cell
MR1 NK
Immune evasion
TH17 cytokines (IL-17A and IL-22) ↑ Tumour growth
↑ Metastasis
TAN
↑ Recruitment

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 362


Review article

Fig. 3 | The role of innate lymphoid and innate-like T cells in bridging In response, type 2-polarized ILCs and ILTCs secrete TH2 cytokines (IL-13, IL-4,
adaptive and innate immunity in cancer. a, Orchestrating an antitumour IL-5, IL-9 and amphiregulin (AREG)) with negative impact on innate and adaptive
immune response in a T helper 1 (TH1) environment. Tissue perturbations such antitumour immunity. A positive feedforward loop has been described for
as immunogenic cell death and tumour growth through released damage- tuft cells in the gut and ILC2s through secretion of IL-25 and IL-13, respectively.
associated molecular patterns (DAMPs), pathogen-associated molecular Secretion of type 2 cytokines can induce immunosuppression of antitumour
patterns (PAMPs), alarmins and the intratumoural microbiome are sensed innate and adaptive immune responses, suppressing γδ2 T cells via activation
through macrophages and dendritic cells (DCs). This leads to the secretion of γδ1 T cells, activation of regulatory T (Treg) cells, increasing myeloid-derived
of IL-12 and IL-18, fostering type 1 polarization of innate lymphoid cells (ILCs) suppressor cell (MDSC) and eosinophil (Eos) recruitment to suppress NK
and innate-like T cells (ILTCs). TH1-polarized ILCs and ILTCs then amplify this function and polarizing M2 macrophages. Increased B cell recruitment through
signalling cascade by producing large amounts of type 1 cytokines (specifically IL-5 and GM-CSF, on the contrary, can have beneficial effects on tumour control.
interferon-γ (IFNγ) and tumour necrosis factor (TNF)). These TH1 signalling c, ILCs and ILTCs as cornerstones of TH17-dependent tumour promotion. DCs,
molecules can have pleiotropic beneficial effects on various immune cells macrophages, neutrophils and MDSCs can secrete key stimulators such as IL-1β,
within the tumour immune microenvironment: IFNγ on macrophages leads IL-6, IL-7, IL-23 and TGF-β, induced through microbial dysbiosis in the tumour
to maturation, M1 > M2 polarization, increased production of IL-12 and IL-18, immune microenvironment, which is sensed through Toll-like receptors on
as well as enhanced antigen presentation capacities. Similarly, DCs show these cells. In response, ILCs and ILTCs can acquire type 17 polarization and
preferred differentiation into type 1 conventional DCs (cDC1s) leading to higher secrete large amounts of IL-17A and IL-22 that can in turn have various negative
capacities of antigen presentation and the formation of germinal centres regulatory functions on neutrophils to promote tumour growth, enhanced
(GCs), which is a crucial component of CD4+ helper T cell recruitment, effector metastatic potential and inhibition of CD8+ T cell function. TH17 cytokines can
function and TH1 polarization. On B cells, these signalling cascades can lead to directly inhibit NK cells and CTLs, leading to tumour promotion. IL-17A signalling
maturation and class switch of tumour-specific antibodies. These mechanisms on tumour cells can induce CXCL5 secretion and recruitment of MDSCs, which
lead to an increased infiltration of natural killer (NK) cells and cytotoxic CD8+ T are potent inhibitors of T cell recruitment and cytotoxicity. IL-17A in the tumour
lymphocytes (CTL) and further enhanced tumour cell-killing capacities immune microenvironment leads to an increased recruitment of tumour-
of these effector cells. On tumour cells, TH1 effector molecules can induce associated neutrophils (TANs) that can foster tumour growth and increase
apoptosis and pyroptosis. Additionally, increase in major histocompatibility metastasis and have the potential to negatively impact tumour-killing function
complex (MHC)-I-related presentation of tumour-associated antigens (TAAs) of CD8+ T cells. 5-OP-RU, 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil;
triggers enhanced direct tumour cell killing of CD8+ T cells. b, ILCs and ILTCs as CAF, cancer-associated fibroblast; CXCL5, CXC-chemokine ligand 5; CXCR2,
initiators and amplifiers of type 2-mediated immune evasion. Alarmins derived CXC-chemokine receptor 2; FASL, FAS ligand; iNKT, invariant natural killer T;
from dying cells, pre-cancerous cells, cancer-associated fibroblasts, epithelial MAIT, mucosal-associated invariant T; MMP, matrix metalloproteinase;
tuft cells, chronic stimulation through epithelial cell damage or alternatively MR1, MHC class I-related gene protein; TCR, T cell receptor; TGFβ, transforming
M2-polarized macrophages can induce type 2 polarization of ILCs and ILTCs. growth factor-β; TSLP, thymic stromal lymphopoietin.

ILC-based and ILTC-based cancer immunotherapy PD1 blockade in lymphoma models increased ADCC against
The capacity of conventional αβ T cells for antigen-directed cytotoxic- lymphoma cells232. γδ T cells have also recently been identified to
ity has been the major focus in the development of cancer immuno- contribute to immune checkpoint blockade response in patients
therapies; however, clinical research has demonstrated that a large with mismatch repair-deficient (dMMR), HLA class I-negative colon
proportion of patients do not benefit from these approaches1. It is cancer234. Similarly, in patients with melanoma, treatment with anti-
therefore necessary to explore alternative paths to engage the immune PD1 therapies induced higher numbers of circulating MAIT cells and
system in the combat against cancer. The importance of NK cells for correlated with better overall survival235. Additionally, γδ T cells may
cancer immunosurveillance has been recognized for many years8,228, be important sources of immunosuppression, as PDL1 blockade on
such that targeting them in cancer immunotherapy is ongoing in vari- γδ T cells enhanced CD4+ and CD8+ T cell infiltration in mouse mod-
ous clinical studies. In comparison, helper ILCs and unconventional els of pancreatic ductal adenocarcinoma236. In addition, alternative
T cells have only recently been recognized as important players in checkpoint inhibitors such as targeting TIGIT may provide a prudent
cancer immunotherapy, and, thus, immunotherapeutic approaches strategy to target ILCs as TIGIT is expressed on NK cells and ILC1s233.
targeting these cells are less advanced. Subsequently, we highlight Nevertheless, whether ILCs and ILTCs have prognostic or predictive
the potential of ILCs and ILTCs as new targets for cancer immuno- value for cancer immunotherapy requires further evaluation across
therapy along with future directions of harnessing their enormous various tumour types.
potential (Fig. 4).
Monoclonal antibodies to immune receptors
Immune checkpoint inhibition ILCs and ILTCs overexpress NKG2A in human tumours, leading to inhibi-
Therapeutic targeting of peptide-reactive conventional T cells through tion of effector cells237 (Fig. 4b). A humanized anti-NKG2A monoclonal
inhibition of the cytotoxic T lymphocyte protein 4 (CTLA4) and PD1– antibody238, monalizumab, has been shown to block the inhibitory
programmed death ligand 1 (PDL1) pathways has revolutionized treat- function of NKG2A and is being evaluated in several phase II trials in
ment outcomes for many types of cancers229. Similar to their adaptive patients with solid tumours239,240.
counterparts, ILCs and ILTCs in patients with cancer have also been Inhibitory KIRs negatively regulate NK cell function and are impor-
found to upregulate their cell surface expression of immune check- tant modulators of antitumour immunity241. Currently, IPH2102 and
point molecules such as PD1, CTLA4, T cell immunoglobulin and mucin lirilumab (IPH2102/BMS-986015) are monoclonal antibodies tar-
domain molecule 3 (TIM3), T cell immunoreceptor with immunoglobu- geting KIR2DL-1, KIR2DL-2 and KIR2DL-3 NK cell receptors that are
lin and ITIM domains (TIGIT) and lymphocyte activation gene 3 protein being evaluated either as monotherapy or combined with checkpoint
(LAG3)230–233 (Fig. 4a). inhibitors in acute myeloid leukaemia, multiple myeloma, chronic

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 363


Review article

a Immune checkpoint inhibition b Targeting inhibitory signalling c BiKEs and TriKEs

Tumour antigen
Antibody
BiKE TriKE

NKG2A KIRs SLAMF7


CD16
NKp46
PD1 CTLA4 TIM3 TIGIT LAG3
or PDL1

ILTC ILC ILTC


ILC ILTC NK

↓ Inhibition ↑ Cytotoxicity ↑ Cytokine ↑ Cytotoxicity ↑ Cytokine ↑ Cytotoxicity


production production

d TCR ligands e Recombinant cytokines f Adoptive therapy


CAR ILC and CAR ILTC
IL-12 IL-18 IL-15 IL-2

CAR CAR
Phosphoantigens MAIT γδ T
5-OP-RU CD1d
α-GalCer IL-12R IL-18R IL-15R IL-2R
TCR
CAR

ILC ILTC CAR CAR


NK NKT
MAIT iNKT γδ T

↑ TH1 ↑ Cytotoxicity
↑ Proliferation ↑ Cytotoxicity polarization
Endogenous TCR

Fig. 4 | Strategies to harness innate lymphoid and innate-like T cells for on the other side, ultimately NK cells and ILTCs to attack tumour cells through
cancer immunotherapy. a, Innate lymphoid cells (ILCs) and innate-like T cells CD16-mediated antibody-dependent cellular cytotoxicity. d, Stimulation of ILTCs
(ILTCs) can be targeted by blocking inhibitory signalling pathways expressed with T cell receptor (TCR) ligands can increase proliferation and cytotoxicity
on the cell surface: programmed cell death protein 1 (PD1)– programmed towards tumour cells. Proof-of-principle studies have demonstrated that
death ligand 1 (PDL1), cytotoxic T lymphocyte protein 4 (CTLA4), T cell 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil (5-OP-RU) can activate
immunoglobulin and mucin domain-containing 3 (TIM3), T cell immunoreceptor MAIT cells, phosphoantigens such as aminobisphosphonates can activate
with immunoglobulin and ITIM domains (TIGIT) and lymphocyte activation γδ T cells and α-galactosylceramide (α-GalCer) can activate invariant natural
gene 3 (LAG3). b, Anti-NKG2A and anti-killer immunoglobulin-like receptor killer T (iNKT) cells. To avoid anergy of NKT cells, antigen-presenting cells
(KIR) antibodies block inhibitory receptors on natural killer (NK) cells and such as dendritic cells can be loaded with α-GalCer and provide co-stimulatory
ILTCs. SLAMF7 can be activated with antibodies to prime antitumour immune signalling. e, Recombinant cytokines in various formulations can be used for
responses in NK cells and ILTCs. c, Bispecific or trispecific NK cell engagers (BiKEs TCR-independent stimulation of ILTCs and ILCs through cognate receptors,
or TriKEs, respectively) consist of antibody-based constructs combining distinct increasing TH1 polarization and therefore tumour cytotoxicity. f, ILCs and ILTCs
single-chain variable fragments that contain tumour-specific antigens on the can be used as platforms for adoptive cell transfer, by using chimeric antigen
one side and NK cell or ILTC receptors (such as NKp46 or IL-15R, respectively) receptor (CAR) constructs to target tumour-specific antigens.

lymphocytic leukaemia and breast and ovarian cancers with limited to maximize activation of ILTCs and NK cells249 (Fig. 4c). Given this, they
success, particularly as a monotherapy242–244. are also termed NK cell engagers. BiKEs and TriKEs that engage CD16 and
Signalling lymphocytic activation molecule family member 7 IL-15 along with tumour antigens such as CLEC12A, CD133 or epithelial
(SLAMF7) is a receptor present on ILCs and ILTCs245, which has been cell adhesion molecule have shown strong NK cell-mediated responses
shown to influence NK cell function and cytotoxicity246. Elotuzumab, against patient-derived acute myeloid leukaemia blasts250. Trifunctional
a humanized, immunostimulatory IgG1 monoclonal antibody, specifi- NK cell engagers were shown to target tumour antigen through two NK
cally activates NK cells by targeting the extracellular domain of human cell receptors, NKp46 and CD16, with high efficacy both in vitro and in
SLAMF7 and binding to CD16 on NK cells via its Fc portion. This leads a mouse model of lymphoma251,252. ILCs and ILTCs could be specifically
to an increase in NK-cell-mediated ADCC through CD69 expression, as targeted through their semi-invariant receptor to direct them to tumour
well as IFNγ and granzyme B release245,247,248. sites and to enhance their cytokine secretion or cytotoxicity.

Bi-specific and tri-specific killer cell engagers TCR ligands


Bispecific and trispecific killer engagers (BiKEs and TriKEs, respec- ILTCs can be potently stimulated and expanded in vivo and ex vivo
tively), are monoclonal antibody fragments that create a synapse through ligation of their semi-invariant TCR with prototypic ligands
between activating receptors on NK cells and tumour-specific antigens (Fig. 4d) such as glycolipids (to stimulate iNKT cells) and or riboflavin

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 364


Review article

Glossary of clinical activity (1 pathological response and 14 with stable disease


in 35 enrolled patients)257.
Most clinical applications targeting TCR stimulation of ILTCs
αβ T cells with immunosuppressive features for improved cancer immunotherapy have focused on γδ T cells and
T cells expressing a lineage-specific that are often associated with poor polyclonal Vγ9Vδ2+ T cells specifically, with modest therapeutic
heterodimeric T cell receptor consisting outcomes in cancer. efficacy88,258–260. One mechanism that was recently introduced might
of an α-chain and a β-chain (as opposed explain the lack of efficacy of this approach, as phosphoantigen-
to γδ T cell receptors). ‘Missing-self’ mechanism stimulated γδ T cells can suppress NK cell-mediated antitumour
This describes a mechanism of response to missing-self261. Taken together, TCR-stimulated ILTCs
Antibody opsonization nature killer cell activation, in which may be potent effectors of anticancer immunity, yet require a greater
Antigen-specific antibodies can bind absence or altered expression of major understanding before broader application. Optimal polarization
to foreign pathogens or transformed histocompatibility complex I molecules through co-stimulatory signals might be needed for optimized
cells and mark them for elimination by (for example, on tumour cells) leads to cytotoxicity and antitumour function.
phagocyting cells. the recognition and elimination of these
target cells. Cytokine-based immunotherapies
Degrees of innateness As ILCs and ILTCs are stimulated by many overlapping cytokines and
This term was previously coined, for Pancreatic intraepithelial share numerous cytokine receptors, immunotherapeutic approaches
example, by Gutierrez-Arcelus et al.10 neoplasia involving cytokines could collectively target subsets of ILCs and ILTCs
and Constantinides and Belkaid11, to A precursor lesion to invasive ductal triggering overlapping or synergistic mechanisms (Fig. 4e). IL-2 is
describe where an immune response adenocarcinoma of the pancreas with not only known for its role in T cell survival and expansion but also
lies on the continuum that exists well-defined histological features. induces expansion and activation of ILCs262 and ILTCs30,263,264 in different
between innate and adaptive immunity. settings. IL-2 was one of the first cytokines used to enhance antitumour
Pyroptosis function of NK cells; however, it had limited efficacy owing to adverse
Graft-versus-host disease An inflammatory pathway leading to side effects and induction of Treg cells265. IL-15 was shown to be far more
(GvHD). A potentially fatal complication cell death as mediated by activation of efficient at ex vivo expansion and activation of NK and γδ T cells266. IL-15
after allogeneic cell transfer caused by caspase 1 and inflammatory cytokines alone or in combination with IL-12 and IL-18 led to increased cytotoxic-
a detrimental reaction of the transferred such as IL-1β and IL-18. ity by upregulating perforin, granzyme and IFNγ secretion. However,
allogeneic T cells (graft) against the continuous treatment with IL-15 induces characteristics of exhaustion
(host) healthy tissue of the recipient. T-bet in NK cells267.
The transcription factor T-bet Similarly, IL-12 and IL-15 together have been shown to enhance
Lymphoid tissue-inducing (encoded by TBX21) promotes the cytotoxicity by upregulating T-bet and increasing the secretion of
(LTi) cells differentiation programmes of innate perforin, IFNγ and granzymes A and B in iNKT cells268, γδ T cells and
These cells are members of the innate and adaptive lymphocytes towards MAIT cells30,269,270. In mouse models, IL-15 stimulation induces gran-
lymphoid cell family that promote interferon-γ-producing type 1 cells. zyme C expression in helper ILC1s65, inducing cytotoxicity, potentially
generation of secondary lymphoid contributing to antitumour immunity.
structures. Toll-like receptor
Pattern-recognition receptor expressed Adoptive transfer
M2 macrophages on innate immune cells that recognizes The ability of ILCs and ILTCs to recognize tumours in an MHC-
Although defining macrophage structurally conserved molecules (also independent manner gives them a unique advantage over conventional
polarization is oversimplistic, this called pathogen-associated molecular T cells. This allows them to be used as effector cells without the risks of
is commonly used to describe patterns) derived from microorganisms. alloreactivity271,272. Therefore, designing immunotherapeutic strategies
alternatively activated macrophages targeting ILCs and ILTCs as universal ‘off-the-shelf’ or universal agents
for different types of tumours is an appealing pan-anticancer strategy.
Adoptive cell therapy is an immunotherapeutic approach, in which
derivates (to stimulate MAIT cells). γδ T cell TCRs can be indirectly lymphocytes are typically separated from the blood of patients through
activated by aminobisphosphonates or synthetic phosphoantigens. a process termed apheresis, then ex vivo expanded and reinfused back
To avoid the potential tumour-promoting functions of TH2 or TH17 into patients with cancer, also termed autologous cell transfer. Adop-
subsets, it must be established how to polarize these cells towards tive transfer of NK cells in immunotherapy takes advantage of their allo-
a TH1 phenotype. Priming potential APCs through the addition of reactivity, which is derived from a mismatch between donor NK cells
adjuvants such as Toll-like receptor ligands might be a practical strat- expressing KIR and HLA ligands on tumour cells. The majority of the
egy to maximize TH1 polarization and optimize the cytotoxicity of NK cells used for adoptive transfer use PBMC collected by apheresis;
ILTCs87,253. Eventhough glycolipid ligand α-GalCer has demonstrated however, other sources such as cord blood and bone marrow have also
promising antitumour efficacy in mouse tumour models89, repetitive been used273. These allogeneic NK cells have demonstrated safety, effi-
administration of α-GalCer induces a strong and long-lasting anergy cacy and antitumour activity in various tumours such as acute myeloid
of iNKT cells254. To further circumvent stimulation-induced anergy, the leukaemia274, colon carcinoma275 and non-Hodgkin’s lymphoma276
concept of combined adoptive transfer of iNKT cells and α-GalCer- with little toxicity such as graft-versus-host disease (GvHD). Adoptive
pulsed DCs has been applied and tested clinically in small phase I stud- iNKT cell-based therapies have also been explored in phase I and II trials,
ies255,256. In a phase II trial in patients with advanced or recurrent NSCLC, which involve pre-expansion and activation of iNKT cells before admin-
α-GalCer-pulsed APCs showed an acceptable safety profile with signs istration in patients. These approaches were tested in patients with

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 365


Review article

NSCLC, melanoma and HCC277–279. These studies showed that ex vivo immunotherapeutic strategies. ILCs specifically will be of particular
expansion of iNKT cells is feasible and well tolerated, and induction of importance, because of their high degree of innateness and therefore
TH1 responses could have antitumour potential. Although not tested most rapid responses10,49,290.
clinically, expansion of MAIT cells in vitro was shown to be feasible, as Given the complex dynamics between their cellular frequency in
they retain their function and cytotoxicity280, making them a suitable tissues and their associated cytokine milieu and receptors, ILCs and
candidate for adoptive cell transfer protocols. ILTCs can be activated by the TME to jointly either through synergy
Chimeric antigen receptors (CARs) are receptors that are geneti- or compensation elicit a robust antitumour immune response3. Their
cally engineered to target specific antigens and mediate cytotoxicity relationship with surrounding cells of the TME during carcinogenesis
against tumours281. Currently approved CAR T cell therapies are based and any conserved functional programmes (for example, expression
on αβ T cells recognizing antigen–MHC complexes and are therefore of cytokine receptors, transcription factors and effector molecules)
HLA-matched to avoid GvHD. This highlights once again that ILCs and between ILCs and ILTCs remains poorly understood. In the light of this,
ILTCs as MHC-independent cell types are strong candidates for CAR the specific mediators that shape ILC and ILTC function throughout
therapies. This notion was further substantiated by a recent longitu- carcinogenesis, and whether these cells reach a ‘tipping point’ switch-
dinal study from two patients with chronic lymphocytic leukaemia ing from antitumorigenic to pro-tumorigenic signalling remains largely
who received CD19-targeted CAR T cells (CTL019) therapy and unknown. What influence does the stage of disease, potential underly-
showed an initial response phase dominated by CD8+ CAR T cells and γδ ing chronic inflammation or tumour intrinsic factors have on ILC and
CAR T cells. This indicates that these ILTCs are likely important effector ILTC plasticity, phenotype and function? Investigating pre-cancerous
cells in this therapy282. lesions from patients and monitoring ILC and ILTC plasticity during
CAR NK, γδ CAR T and CAR NKT cells have been used in various tumour growth in mouse models of cancer will help to provide the
settings, showing both antitumour function and enhanced cytotox- necessary insight into the dynamics and hierarchies of ILCs and ILTCs
icity272. CAR NK cells having the advantage of not causing GvHD in an for targeted immunotherapeutic intervention291. The capability of ILCs
allogeneic setting have been used successfully in different haemato- and ILTCs to modify and shape the adaptive immune system under
logical malignancies and solid tumours283,284. γδ CAR T cells have shown homeostasis, as well as in tumour settings, puts them in the spotlight
superior cytotoxicity against leukaemia xenografts when compared for sharing or synergizing potential effector mechanisms and as new
with unselected CAR T cells285. In neuroblastoma cell lines express- targets for cancer immunotherapy.
ing GD2 ganglioside, transducing γδ CAR T cells with GD2-specific More studies are needed to further understand and delineate the
CAR constructs enhanced GD2-specific killing while retaining the regulatory mechanism that affects ILCs and ILTCs, as well as the clinical
innate cytotoxic capacities of γδ T cells. These cells also retained their consequences of ILC-directed and ILTC-directed therapies to approve
capacity to cross-present tumour antigens to αβ T cells, which could them as off-the-shelf or universal agents. Taking into account that
have additional antitumour effects286. Similarly, iNKT cells expanded ILC subsets are both tissue-dependent and tumour-type-dependent,
ex vivo and engineered to express GD2-targeted CAR more efficiently identification of additional markers to better distinguish ILC subsets
infiltrated tumours in xenograft models of neuroblastoma than unse- in both mice and humans will also aid in developing better targeted
lected CAR T cells287. MAIT cells are also promising candidates for therapies. Additionally, the use of genetically engineered mice and
CAR therapeutics, as they are MR1-restricted and therefore unlikely development of tools to manipulate these cells in mouse models will
to induce GVHD and are being explored in different settings271,288. be important for immunotherapeutic interventions.
Interestingly, co-expansion of iNKT cells and Vγ9Vδ2+ T cells, using Together, our understanding of ILCs and ILTC biology, as well as a
IFNγ, IL-2, anti-CD3 antibody and feeder cells, showed higher cytotoxic deeper knowledge of the mechanisms that support the innate–adaptive
activity in vitro compared with αβ CAR T cells and enhanced survival relationship, will further the development of the next generation of
in xenograft models289. immunotherapeutics. The future of immunotherapy targeting ILCs and
Although the aforementioned approaches are promising, they ILTCs will certainly generate a resounding applause from its audience.
are limited by proliferation efficiency, difficulties in activation ex vivo
before administration and short persistence of response in vivo. How- Published online: 20 April 2023
ever, as discussed earlier, ILCs and ILTCs offer many advantages such as References
lack of alloreactivity, antigen-presenting as well as immunomodulatory 1. Waldman, A. D., Fritz, J. M. & Lenardo, M. J. A guide to cancer immunotherapy: from T cell
abilities and rapid secretion of cytokines, which place these cells as the basic science to clinical practice. Nat. Rev. Immunol. 20, 651–668 (2020).
2. Sharma, P. et al. The next decade of immune checkpoint therapy. Cancer Discov. 11,
new milestones providing new and exciting opportunities for the next 838–857 (2021).
generation of adoptive cell therapy in cancer. 3. Jacquelot, N., Seillet, C., Vivier, E. & Belz, G. T. Innate lymphoid cells and cancer.
Nat. Immunol. 23, 371–379 (2022).

Conclusion 4. Crosby, C. M. & Kronenberg, M. Tissue-specific functions of invariant natural killer T cells.
Nat. Rev. Immunol. 18, 559–574 (2018).
The high frequency of ILCs and ILTCs in tissues and their pleiotropic 5. Godfrey, D. I., Koay, H. F., McCluskey, J. & Gherardin, N. A. The biology and functional
effector functions appoint them as master regulators and pivotal importance of MAIT cells. Nat. Immunol. 20, 1110–1128 (2019).
6. Silva-Santos, B., Mensurado, S. & Coffelt, S. B. γδ T cells: pleiotropic immune effectors
orchestrators of tumour immunity. As the complex and dynamic micro- with therapeutic potential in cancer. Nat. Rev. Cancer 19, 392–404 (2019).
environment of the tumour is shaped during tumorigenesis, ILCs and 7. Cozar, B. et al. Tumor-infiltrating natural killer cells. Cancer Discov. 11, 34–44 (2021).
ILTCs dance to a delicately choreographed programme as the first 8. Karre, K., Ljunggren, H. & Piontek, G. K. Selective rejection of H-2-deficient lymphoma
variants suggests alternative immune defense strategy. Nature 319, 675 (1986).
responders in the TME during tumour initiation, immunosurveillance 9. Chiossone, L., Dumas, P. Y., Vienne, M. & Vivier, E. Natural killer cells and other innate
and tumour progression64. lymphoid cells in cancer. Nat. Rev. Immunol. 18, 671–688 (2018).
It is becoming increasingly clear that unlike conventional T cells, 10. Gutierrez-Arcelus, M. et al. Lymphocyte innateness defined by transcriptional states reflects
a balance between proliferation and effector functions. Nat. Commun. 10, 687 (2019).
owing to the ability of ILCs and ILTCs to rapidly integrate and respond 11. Constantinides, M. G. & Belkaid, Y. Early-life imprinting of unconventional T cells and
to signals from their environment, they are attractive candidates for tissue homeostasis. Science 374, eabf0095 (2021).

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 366


Review article

12. Mayassi, T., Barreiro, L. B., Rossjohn, J. & Jabri, B. A multilayered immune system through 47. Caccamo, N. et al. Differentiation, phenotype, and function of interleukin-17-producing
the lens of unconventional T cells. Nature 595, 501–510 (2021). human Vγ9Vδ2 T cells. Blood 118, 129–138 (2011).
13. Bendelac, A. et al. CD1 recognition by mouse NK1+ T lymphocytes. Science 268, 48. Peters, C., Hasler, R., Wesch, D. & Kabelitz, D. Human Vδ2 T cells are a major source of
863–865 (1995). interleukin-9. Proc. Natl Acad. Sci. USA 113, 12520–12525 (2016).
14. Kjer-Nielsen, L. et al. MR1 presents microbial vitamin B metabolites to MAIT cells. Nature 49. Weizman, O. E. et al. ILC1 confer early host protection at initial sites of viral infection. Cell
491, 717–723 (2012). 171, 795–808.e12 (2017).
15. Treiner, E. et al. Selection of evolutionarily conserved mucosal-associated invariant 50. Artis, D. & Spits, H. The biology of innate lymphoid cells. Nature 517, 293–301 (2015).
T cells by MR1. Nature 422, 164–169 (2003). 51. Klose, C. S. & Artis, D. Innate lymphoid cells as regulators of immunity, inflammation and
16. Pellicci, D. G., Koay, H. F. & Berzins, S. P. Thymic development of unconventional T cells: tissue homeostasis. Nat. Immunol. 17, 765–774 (2016).
how NKT cells, MAIT cells and γδ T cells emerge. Nat. Rev. Immunol. 20, 756–770 (2020). 52. Dogra, P. et al. Tissue determinants of human NK cell development, function, and
17. Matsuda, J. L. et al. Natural killer T cells reactive to a single glycolipid exhibit a highly residence. Cell 180, 749–763.e13 (2020).
diverse T cell receptor β repertoire and small clone size. Proc. Natl Acad. Sci. USA 98, 53. Spits, H., Bernink, J. H. & Lanier, L. NK cells and type 1 innate lymphoid cells: partners
12636–12641 (2001). in host defense. Nat. Immunol. 17, 758–764 (2016).
18. Lantz, O. & Bendelac, A. An invariant T cell receptor α chain is used by a unique subset 54. Perona-Wright, G. et al. Systemic but not local infections elicit immunosuppressive IL-10
of major histocompatibility complex class I-specific CD4+ and CD4-8-T cells in mice and production by natural killer cells. Cell Host Microbe 6, 503–512 (2009).
humans. J. Exp. Med. 180, 1097–1106 (1994). 55. Neo, S. Y. et al. CD73 immune checkpoint defines regulatory NK cells within the tumor
19. Singh, A. K., Tripathi, P. & Cardell, S. L. Type II NKT cells: an elusive population with microenvironment. J. Clin. Invest. 130, 1185–1198 (2020).
immunoregulatory properties. Front. Immunol. 9, 1969 (2018). 56. Klose, C. S. N. et al. Differentiation of type 1 ILCs from a common progenitor to all
20. Georgiev, H., Ravens, I., Benarafa, C., Forster, R. & Bernhardt, G. Distinct gene expression helper-like innate lymphoid cell lineages. Cell 157, 340–356 (2014).
patterns correlate with developmental and functional traits of iNKT subsets. Nat. Commun. 57. Abt, M. C. et al. Innate immune defenses mediated by two ILC subsets are critical for
7, 13116 (2016). protection against acute Clostridium difficile infection. Cell Host Microbe 18, 27–37 (2015).
21. Lee, Y. J. et al. Lineage-specific effector signatures of invariant NKT cells are shared 58. Bernink, J. H. et al. Human type 1 innate lymphoid cells accumulate in inflamed mucosal
amongst γδ T, innate lymphoid, and Th cells. J. Immunol. 197, 1460–1470 (2016). tissues. Nat. Immunol. 14, 221–229 (2013).
22. Cui, G. et al. A circulating subset of iNKT cells mediates antitumor and antiviral immunity. 59. Fuchs, A. et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and
Sci. Immunol. 7, eabj8760 (2022). IL-15-responsive IFN-γ-producing cells. Immunity 38, 769–781 (2013).
23. Lee, Y. J. et al. Tissue-specific distribution of iNKT cells impacts their cytokine response. 60. Gao, Y. et al. Tumor immunoevasion by the conversion of effector NK cells into type 1
Immunity 43, 566–578 (2015). innate lymphoid cells. Nat. Immunol. 18, 1004–1015 (2017).
This article demonstrates how anatomic localization and systemic distribution of This article describes novel mechanisms of tumor immune escape driven by TGFβ in
iNKT1, iNKT2 and iNKT17 cells imprint their responsiveness to stimuli and cytokine the TME.
production. 61. Nussbaum, K. et al. Tissue microenvironment dictates the fate and tumor-suppressive
24. Heymann, F. & Tacke, F. Immunology in the liver — from homeostasis to disease. Nat. Rev. function of type 3 ILCs. J. Exp. Med. 214, 2331–2347 (2017).
Gastroenterol. Hepatol. 13, 88–110 (2016). 62. Meininger, I. et al. Tissue-specific features of innate lymphoid cells. Trends Immunol. 41,
25. Corbett, A. J. et al. T-cell activation by transitory neo-antigens derived from distinct 902–917 (2020).
microbial pathways. Nature 509, 361–365 (2014). 63. Diefenbach, A., Colonna, M. & Koyasu, S. Development, differentiation, and diversity of
26. Eckle, S. B. et al. Recognition of vitamin B precursors and byproducts by mucosal innate lymphoid cells. Immunity 41, 354–365 (2014).
associated invariant T cells. J. Biol. Chem. 290, 30204–30211 (2015). 64. Dadi, S. et al. Cancer immunosurveillance by tissue-resident innate lymphoid cells and
27. Reantragoon, R. et al. Antigen-loaded MR1 tetramers define T cell receptor heterogeneity innate-like T cells. Cell 164, 365–377 (2016).
in mucosal-associated invariant T cells. J. Exp. Med. 210, 2305–2320 (2013). This article shows that type 1-polarized ILTCs and type 1-like ILCs share common gene
28. Tilloy, F. et al. An invariant T cell receptor α chain defines a novel TAP-independent major expression programmes and engage in early-stage tumour-elicited immunosurveillance
histocompatibility complex class Ib-restricted α/β T cell subpopulation in mammals. in an IL-15-dependent manner.
J. Exp. Med. 189, 1907–1921 (1999). 65. Nixon, B. G. et al. Cytotoxic granzyme C-expressing ILC1s contribute to antitumor
29. Crowther, M. D. et al. Genome-wide CRISPR-Cas9 screening reveals ubiquitous T cell immunity and neonatal autoimmunity. Sci. Immunol. 7, eabi8642 (2022).
cancer targeting via the monomorphic MHC class I-related protein MR1. Nat. Immunol. 66. Moreno-Nieves, U. Y. et al. Landscape of innate lymphoid cells in human head and neck
21, 178–185 (2020). cancer reveals divergent NK cell states in the tumor microenvironment. Proc. Natl Acad.
30. Tao, H. et al. Differential controls of MAIT cell effector polarization by mTORC1/mTORC2 Sci. USA 118, e2101169118 (2021).
via integrating cytokine and costimulatory signals. Nat. Commun. 12, 2029 (2021). 67. Mjosberg, J. et al. The transcription factor GATA3 is essential for the function of human
31. Chua, W. J. et al. Polyclonal mucosa-associated invariant T cells have unique innate type 2 innate lymphoid cells. Immunity 37, 649–659 (2012).
functions in bacterial infection. Infect. Immun. 80, 3256–3267 (2012). 68. Mazzurana, L. et al. Tissue-specific transcriptional imprinting and heterogeneity in human
32. Rahimpour, A. et al. Identification of phenotypically and functionally heterogeneous innate lymphoid cells revealed by full-length single-cell RNA-sequencing. Cell Res. 31,
mouse mucosal-associated invariant T cells using MR1 tetramers. J. Exp. Med. 212, 554–568 (2021).
1095–1108 (2015). 69. Spits, H. & Mjosberg, J. Heterogeneity of type 2 innate lymphoid cells. Nat. Rev. Immunol.
33. Hinks, T. S. C. et al. Activation and in vivo evolution of the MAIT cell transcriptome in mice 22, 701–712 (2022).
and humans reveals tissue repair functionality. Cell Rep. 28, 3249–3262.e5 (2019). 70. Serafini, N. et al. Trained ILC3 responses promote intestinal defense. Science 375,
34. Leng, T. et al. TCR and inflammatory signals tune human MAIT cells to exert specific 859–863 (2022).
tissue repair and effector functions. Cell Rep. 28, 3077–3091.e5 (2019). 71. Vivier, E. et al. Innate lymphoid cells: 10 years on. Cell 174, 1054–1066 (2018).
35. Gherardin, N. A. et al. Human blood MAIT cell subsets defined using MR1 tetramers. 72. Sonnenberg, G. F. & Artis, D. Innate lymphoid cells in the initiation, regulation and
Immunol. Cell Biol. 96, 507–525 (2018). resolution of inflammation. Nat. Med. 21, 698–708 (2015).
36. Kurioka, A., Walker, L. J., Klenerman, P. & Willberg, C. B. MAIT cells: new guardians of the 73. Krabbendam, L., Heesters, B. A., Kradolfer, C. M. A., Spits, H. & Bernink, J. H. Identification
liver. Clin. Transl. Immunol. 5, e98 (2016). of human cytotoxic ILC3s. Eur. J. Immunol. 51, 811–823 (2021).
37. Cui, Y. et al. Mucosal-associated invariant T cell-rich congenic mouse strain allows 74. Fridman, W. H., Zitvogel, L., Sautes-Fridman, C. & Kroemer, G. The immune contexture in
functional evaluation. J. Clin. Invest. 125, 4171–4185 (2015). cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 14, 717–734 (2017).
38. Goodman, T. & Lefrancois, L. Expression of the γδ T-cell receptor on intestinal CD8+ 75. Hiam-Galvez, K. J., Allen, B. M. & Spitzer, M. H. Systemic immunity in cancer. Nat. Rev.
intraepithelial lymphocytes. Nature 333, 855–858 (1988). Cancer 21, 345–359 (2021).
39. Parker, C. M. et al. Evidence for extrathymic changes in the T cell receptor γ/δ repertoire. 76. Maskalenko, N. A., Zhigarev, D. & Campbell, K. S. Harnessing natural killer cells for cancer
J. Exp. Med. 171, 1597–1612 (1990). immunotherapy: dispatching the first responders. Nat. Rev. Drug Discov. 21, 559–577 (2022).
40. Vasudev, A. et al. γ/δ T cell subsets in human aging using the classical α/β T cell model. 77. Binnewies, M. et al. Understanding the tumor immune microenvironment (TIME) for
J. Leukoc. Biol. 96, 647–655 (2014). effective therapy. Nat. Med. 24, 541–550 (2018).
41. Vantourout, P. & Hayday, A. Six-of-the-best: unique contributions of γδ T cells to 78. Girardi, M. et al. Regulation of cutaneous malignancy by γδ T cells. Science 294,
immunology. Nat. Rev. Immunol. 13, 88–100 (2013). 605–609 (2001).
42. Willcox, B. E. & Willcox, C. R. γδ TCR ligands: the quest to solve a 500-million-year-old 79. Smyth, M. J., Crowe, N. Y. & Godfrey, D. I. NK cells and NKT cells collaborate in host
mystery. Nat. Immunol. 20, 121–128 (2019). protection from methylcholanthrene-induced fibrosarcoma. Int. Immunol. 13, 459–463
43. Deseke, M. & Prinz, I. Ligand recognition by the γδ TCR and discrimination between (2001).
homeostasis and stress conditions. Cell Mol. Immunol. 17, 914–924 (2020). 80. Glasner, A. et al. NKp46 receptor-mediated interferon-γ production by natural killer cells
44. Adams, E. J., Gu, S. & Luoma, A. M. Human γδ T cells: evolution and ligand recognition. increases fibronectin 1 to alter tumor architecture and control metastasis. Immunity 48,
Cell. Immunol. 296, 31–40 (2015). 107–119.e4 (2018).
45. Uldrich, A. P., Rigau, M. & Godfrey, D. I. Immune recognition of phosphoantigen-butyrophilin 81. Eckl, J. et al. Transcript signature predicts tissue NK cell content and defines renal cell
molecular complexes by γδ T cells. Immunol. Rev. 298, 74–83 (2020). carcinoma subgroups independent of TNM staging. J. Mol. Med. 90, 55–66 (2012).
46. Wesch, D., Glatzel, A. & Kabelitz, D. Differentiation of resting human peripheral blood 82. Myers, J. A. & Miller, J. S. Exploring the NK cell platform for cancer immunotherapy.
γδ T cells toward Th1- or Th2-phenotype. Cell. Immunol. 212, 110–117 (2001). Nat. Rev. Clin. Oncol. 18, 85–100 (2021).

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 367


Review article

83. Hudspeth, K., Silva-Santos, B. & Mavilio, D. Natural cytotoxicity receptors: broader 118. Townsend, M. J. et al. T-bet regulates the terminal maturation and homeostasis of NK and
expression patterns and functions in innate and adaptive immune cells. Front. Immunol. Vα14i NKT cells. Immunity 20, 477–494 (2004).
4, 69 (2013). 119. Van Acker, H. H. et al. Interleukin-15-cultured dendritic cells enhance anti-tumor γδ T cell
84. Correia, D. V., Lopes, A. & Silva-Santos, B. Tumor cell recognition by γδ T lymphocytes: functions through IL-15 secretion. Front. Immunol. 9, 658 (2018).
T-cell receptor vs. NK-cell receptors. Oncoimmunology 2, e22892 (2013). 120. Sattler, A., Dang-Heine, C., Reinke, P. & Babel, N. IL-15 dependent induction of IL-18
85. Kawano, T. et al. Antitumor cytotoxicity mediated by ligand-activated human Vα24 secretion as a feedback mechanism controlling human MAIT-cell effector functions.
NKT cells. Cancer Res. 59, 5102–5105 (1999). Eur. J. Immunol. 45, 2286–2298 (2015).
86. Petley, E. V. et al. MAIT cells regulate NK cell-mediated tumor immunity. Nat. Commun. 121. Yamaguchi, T. et al. Interleukin-15 effectively potentiates the in vitro tumor-specific
12, 4746 (2021). activity and proliferation of peripheral blood γδ T cells isolated from glioblastoma
87. Ruf, B. et al. Activating mucosal-associated invariant T cells induces a broad antitumor patients. Cancer Immunol. Immunother. 47, 97–103 (1998).
response. Cancer Immunol. Res. 9, 1024–1034 (2021). 122. Fan, X. & Rudensky, A. Y. Hallmarks of tissue-resident lymphocytes. Cell 164, 1198–1211
Together with Petley et al. (2021), these papers provide the first in vivo evidence that (2016).
MAIT cells can be targeted for cancer immunotherapy using riboflavin derivative 123. Vonarbourg, C. et al. Regulated expression of nuclear receptor RORγt confers distinct
5-OP-RU. functional fates to NK cell receptor-expressing RORγt+ innate lymphocytes. Immunity 33,
88. Wilhelm, M. et al. γδ T cells for immune therapy of patients with lymphoid malignancies. 736–751 (2010).
Blood 102, 200–206 (2003). 124. Bernink, J. H. et al. Interleukin-12 and -23 control plasticity of CD127+ group 1 and group 3
89. Kawano, T. et al. Natural killer-like nonspecific tumor cell lysis mediated by specific innate lymphoid cells in the intestinal lamina propria. Immunity 43, 146–160 (2015).
ligand-activated Vα14 NKT cells. Proc. Natl Acad. Sci. USA 95, 5690–5693 (1998). This study is one of the first to show plasticity of ILCs and differentiation of ILC3 to ILC1
90. Groh, V. et al. Broad tumor-associated expression and recognition by tumor-derived γδ in inflammatory disease in humans.
T cells of MICA and MICB. Proc. Natl Acad. Sci. USA 96, 6879–6884 (1999). 125. Cella, M. et al. Subsets of ILC3-ILC1-like cells generate a diversity spectrum of innate
91. Orange, J. S. Formation and function of the lytic NK-cell immunological synapse. lymphoid cells in human mucosal tissues. Nat. Immunol. 20, 980–991 (2019).
Nat. Rev. Immunol. 8, 713–725 (2008). This study describes the intermediate subsets of cells in the differentiation of ILC3
92. Bauer, S. et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible to ILC1 in vivo.
MICA. Science 285, 727–729 (1999). 126. Teng, M. W. et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for
93. Gherardin, N. A. et al. Enumeration, functional responses and cytotoxic capacity of immune-mediated inflammatory diseases. Nat. Med. 21, 719–729 (2015).
MAIT cells in newly diagnosed and relapsed multiple myeloma. Sci. Rep. 8, 4159 (2018). 127. Park, S. H., Kyin, T., Bendelac, A. & Carnaud, C. The contribution of NKT cells, NK cells,
94. Prager, I. et al. NK cells switch from granzyme B to death receptor-mediated cytotoxicity and other γ-chain-dependent non-T non-B cells to IL-12-mediated rejection of tumors.
during serial killing. J. Exp. Med. 216, 2113–2127 (2019). J. Immunol. 170, 1197–1201 (2003).
95. Siegler, J. J. et al. Human ILC3 exert TRAIL-mediated cytotoxicity towards cancer cells. 128. Ohs, I. et al. Restoration of natural killer cell antimetastatic activity by IL12 and
Front. Immunol. 13, 742571 (2022). checkpoint blockade. Cancer Res. 77, 7059–7071 (2017).
96. Cardoso Alves, L., Corazza, N., Micheau, O. & Krebs, P. The multifaceted role of TRAIL 129. Kansler, E. R. et al. Cytotoxic innate lymphoid cells sense cancer cell-expressed
signaling in cancer and immunity. FEBS J. 288, 5530–5554 (2021). interleukin-15 to suppress human and murine malignancies. Nat. Immunol. 23, 904–915
97. Dokouhaki, P. et al. NKG2D regulates production of soluble TRAIL by ex vivo expanded (2022).
human γδ T cells. Eur. J. Immunol. 43, 3175–3182 (2013). 130. Punt, S. et al. The correlations between IL-17 vs. Th17 cells and cancer patient survival:
98. Wingender, G., Krebs, P., Beutler, B. & Kronenberg, M. Antigen-specific cytotoxicity by a systematic review. Oncoimmunology 4, e984547 (2015).
invariant NKT cells in vivo is CD95/CD178-dependent and is correlated with antigenic 131. Ma, S. et al. IL-17A produced by γδ T cells promotes tumor growth in hepatocellular
potency. J. Immunol. 185, 2721–2729 (2010). carcinoma. Cancer Res. 74, 1969–1982 (2014).
99. Vivier, E., Ugolini, S., Blaise, D., Chabannon, C. & Brossay, L. Targeting natural killer cells 132. Coffelt, S. B. et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast
and natural killer T cells in cancer. Nat. Rev. Immunol. 12, 239–252 (2012). cancer metastasis. Nature 522, 345–348 (2015).
100. Bryceson, Y. T., March, M. E., Ljunggren, H. G. & Long, E. O. Activation, coactivation, 133. Jin, C. et al. Commensal microbiota promote lung cancer development via γδ T cells.
and costimulation of resting human natural killer cells. Immunol. Rev. 214, 73–91 Cell 176, 998–1013.e16 (2019).
(2006). This article provides evidence on how mouse γδ T cells integrate environmental cues
101. Couzi, L. et al. Antibody-dependent anti-cytomegalovirus activity of human γδ T cells from malignant tumour cells and microbiota, to promote lung inflammation and
expressing CD16 (FcγRIIIa). Blood 119, 1418–1427 (2012). tumour cell proliferation.
102. Brozova, J., Karlova, I. & Novak, J. Analysis of the phenotype and function of the 134. Chung, L. et al. Bacteroides fragilis toxin coordinates a pro-carcinogenic inflammatory
subpopulations of mucosal-associated invariant T cells. Scand. J. Immunol. 84, cascade via targeting of colonic epithelial cells. Cell Host Microbe 23, 203–214.e5 (2018).
245–251 (2016). 135. Zhao, J., Chen, X., Herjan, T. & Li, X. The role of interleukin-17 in tumor development and
103. Lo Nigro, C. et al. NK-mediated antibody-dependent cell-mediated cytotoxicity in solid progression. J. Exp. Med. 217, e20190297 (2020).
tumors: biological evidence and clinical perspectives. Ann. Transl. Med. 7, 105 (2019). 136. Lo Presti, E. et al. Squamous cell tumors recruit γδ T cells producing either IL17 or IFNγ
104. Ivashkiv, L. B. IFNγ: signalling, epigenetics and roles in immunity, metabolism, disease depending on the tumor stage. Cancer Immunol. Res. 5, 397–407 (2017).
and cancer immunotherapy. Nat. Rev. Immunol. 18, 545–558 (2018). 137. Wu, Y. et al. An innate-like Vδ1+ γδ T cell compartment in the human breast is associated
105. O’Sullivan, T. et al. Cancer immunoediting by the innate immune system in the absence with remission in triple-negative breast cancer. Sci. Transl Med. 11, eaax9364 (2019).
of adaptive immunity. J. Exp. Med. 209, 1869–1882 (2012). 138. Wu, Y. et al. A local human Vδ1 T cell population is associated with survival in non-small-
106. Martin-Fontecha, A. et al. Induced recruitment of NK cells to lymph nodes provides IFN-γ cell lung cancer. Nat. Cancer 3, 696–709 (2022).
for TH1 priming. Nat. Immunol. 5, 1260–1265 (2004). 139. McAllister, F. et al. Oncogenic Kras activates a hematopoietic-to-epithelial IL-17 signaling
107. Gao, Y. et al. γδ T cells provide an early source of interferon-γ in tumor immunity. J. Exp. Med. axis in preinvasive pancreatic neoplasia. Cancer Cell 25, 621–637 (2014).
198, 433–442 (2003). 140. Wakita, D. et al. Tumor-infiltrating IL-17-producing γδ T cells support the progression of
108. Gocher, A. M., Workman, C. J. & Vignali, D. A. A. Interferon-γ: teammate or opponent in tumor by promoting angiogenesis. Eur. J. Immunol. 40, 1927–1937 (2010).
the tumour microenvironment? Nat. Rev. Immunol. 22, 158–172 (2022). 141. Van Hede, D. et al. Human papillomavirus oncoproteins induce a reorganization of
109. Smyth, M. J. et al. Sequential production of interferon-γ by NK1.1+ T cells and natural epithelial-associated γδ T cells promoting tumor formation. Proc. Natl Acad. Sci. USA
killer cells is essential for the antimetastatic effect of α-galactosylceramide. Blood 99, 114, E9056–E9065 (2017).
1259–1266 (2002). 142. Yan, J. et al. MAIT cells promote tumor initiation, growth, and metastases via tumor MR1.
110. Ma, C. et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via Cancer Discov. 10, 124–141 (2020).
NKT cells. Science 360, eaan5931 (2018). This paper demonstrates a tumour-promoting role for MAIT cells in mouse models of
111. Wang, S. et al. Transdifferentiation of tumor infiltrating innate lymphoid cells during MR1-expressing tumours through IL-17A-dependent inhibition of NK cells.
progression of colorectal cancer. Cell Res. 30, 610–622 (2020). 143. Koh, J. et al. IL23-producing human lung cancer cells promote tumor growth via conversion
112. Briukhovetska, D. et al. Interleukins in cancer: from biology to therapy. Nat. Rev. Cancer of innate lymphoid cell 1 (ILC1) into ILC3. Clin. Cancer Res. 25, 4026–4037 (2019).
21, 481–499 (2021). 144. Liu, Y. et al. NCR– group 3 innate lymphoid cells orchestrate IL-23/IL-17 axis to promote
113. van Wilgenburg, B. et al. MAIT cells are activated during human viral infections. hepatocellular carcinoma development. EBioMedicine 41, 333–344 (2019).
Nat. Commun. 7, 11653 (2016). 145. Bernink, J. H. et al. c-Kit-positive ILC2s exhibit an ILC3-like signature that may contribute
114. Li, W. et al. Effect of IL-18 on expansion of γδ T cells stimulated by zoledronate and IL-2. to IL-17-mediated pathologies. Nat. Immunol. 20, 992–1003 (2019).
J. Immunother. 33, 287–296 (2010). This study reports the plasticity between ILC2s and IL-17-producing ILC3-like cells in
115. Kitamura, H. et al. The natural killer T (NKT) cell ligand α-galactosylceramide demonstrates human skin inflammation.
its immunopotentiating effect by inducing interleukin (IL)-12 production by dendritic cells 146. Hernandez, P., Gronke, K. & Diefenbach, A. A catch-22: interleukin-22 and cancer. Eur. J.
and IL-12 receptor expression on NKT cells. J. Exp. Med. 189, 1121–1128 (1999). Immunol. 48, 15–31 (2018).
116. Klose, C. S. et al. A T-bet gradient controls the fate and function of CCR6–RORγt+ innate 147. Vitiello, G. A. & Miller, G. Targeting the interleukin-17 immune axis for cancer immunotherapy.
lymphoid cells. Nature 494, 261–265 (2013). J. Exp. Med. 217, e2019045 (2020).
117. Wang, Y. et al. The IL-15-AKT-XBP1s signaling pathway contributes to effector functions 148. Kirchberger, S. et al. Innate lymphoid cells sustain colon cancer through production of
and survival in human NK cells. Nat. Immunol. 20, 10–17 (2019). interleukin-22 in a mouse model. J. Exp. Med. 210, 917–931 (2013).

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 368


Review article

149. Kryczek, I. et al. IL-22+CD4+ T cells promote colorectal cancer stemness via STAT3 182. Altvater, B. et al. Activated human γδ T cells induce peptide-specific CD8+ T-cell responses
transcription factor activation and induction of the methyltransferase DOT1L. Immunity to tumor-associated self-antigens. Cancer Immunol. Immunother. 61, 385–396 (2012).
40, 772–784 (2014). 183. Barral, P. et al. B cell receptor-mediated uptake of CD1d-restricted antigen augments
150. Kelly, A., Houston, S. A., Sherwood, E., Casulli, J. & Travis, M. A. Regulation of innate and antibody responses by recruiting invariant NKT cell help in vivo. Proc. Natl Acad. Sci. USA
adaptive immunity by TGFβ. Adv. Immunol. 134, 137–233 (2017). 105, 8345–8350 (2008).
151. Batlle, E. & Massague, J. Transforming growth factor-β signaling in immunity and cancer. 184. Rezende, R. M. et al. γδ T cells control humoral immune response by inducing T follicular
Immunity 50, 924–940 (2019). helper cell differentiation. Nat. Commun. 9, 3151 (2018).
152. Laouar, Y., Sutterwala, F. S., Gorelik, L. & Flavell, R. A. Transforming growth factor-β 185. Galli, G. et al. CD1d-restricted help to B cells by human invariant natural killer T lymphocytes.
controls T helper type 1 cell development through regulation of natural killer cell J. Exp. Med. 197, 1051–1057 (2003).
interferon-γ. Nat. Immunol. 6, 600–607 (2005). 186. Magri, G. et al. Innate lymphoid cells integrate stromal and immunological signals
153. Yu, J. et al. Pro- and antiinflammatory cytokine signaling: reciprocal antagonism to enhance antibody production by splenic marginal zone B cells. Nat. Immunol. 15,
regulates interferon-γ production by human natural killer cells. Immunity 24, 575–590 354–364 (2014).
(2006). 187. Ikutani, M. et al. Identification of innate IL-5-producing cells and their role in lung
154. Heinrich, B. et al. The tumour microenvironment shapes innate lymphoid cells in patients eosinophil regulation and antitumor immunity. J. Immunol. 188, 703–713 (2012).
with hepatocellular carcinoma. Gut 71, 1161–1175 (2022). 188. Jacquelot, N. et al. Blockade of the co-inhibitory molecule PD-1 unleashes ILC2-
This study shows how ILC composition and plasticity is regulated by the cytokines dependent antitumor immunity in melanoma. Nat. Immunol. 22, 851–864 (2021).
in the TME of patients with HCC. 189. Moral, J. A. et al. ILC2s amplify PD-1 blockade by activating tissue-specific cancer
155. Havenar-Daughton, C., Li, S., Benlagha, K. & Marie, J. C. Development and function of immunity. Nature 579, 130–135 (2020).
murine RORγt+ iNKT cells are under TGF-β signaling control. Blood 119, 3486–3494 This study shows the role of tumour-infiltrating ILC2s in the context of antibody-
(2012). mediated PD1 blockade to reverse PD1 inhibition and to promote antitumour T cell
156. Seo, N., Tokura, Y., Takigawa, M. & Egawa, K. Depletion of IL-10- and TGF-β-producing responses.
regulatory γδ T cells by administering a daunomycin-conjugated specific monoclonal 190. Meierovics, A. I. & Cowley, S. C. MAIT cells promote inflammatory monocyte
antibody in early tumor lesions augments the activity of CTLs and NK Cells. J. Immunol. differentiation into dendritic cells during pulmonary intracellular infection. J. Exp. Med.
163, 242–249 (1999). 213, 2793–2809 (2016).
157. Sakai, S. et al. MAIT cell-directed therapy of Mycobacterium tuberculosis infection. 191. Gillessen, S. et al. CD1d-restricted T cells regulate dendritic cell function and antitumor
Mucosal Immunol. 14, 199–208 (2021). immunity in a granulocyte-macrophage colony-stimulating factor-dependent fashion.
158. Yu, H. et al. Artificially induced MAIT cells inhibit M. bovis BCG but not M. tuberculosis Proc. Natl Acad. Sci. USA 100, 8874–8879 (2003).
during in vivo pulmonary infection. Sci. Rep. 10, 13579 (2020). 192. Hepworth, M. R. et al. Innate lymphoid cells regulate CD4+ T-cell responses to intestinal
159. Hammad, H., Debeuf, N., Aegerter, H., Brown, A. S. & Lambrecht, B. N. Emerging commensal bacteria. Nature 498, 113–117 (2013).
paradigms in type 2 immunity. Annu. Rev. Immunol. 40, 443–467 (2022). This article is the first to show the direct regulation of adaptive immune response
160. Suzuki, A., Leland, P., Joshi, B. H. & Puri, R. K. Targeting of IL-4 and IL-13 receptors for by ILC3s.
cancer therapy. Cytokine 75, 79–88 (2015). 193. Hepworth, M. R. et al. Immune tolerance. Group 3 innate lymphoid cells mediate intestinal
161. Bando, J. K., Nussbaum, J. C., Liang, H. E. & Locksley, R. M. Type 2 innate lymphoid cells selection of commensal bacteria-specific CD4+ T cells. Science 348, 1031–1035 (2015).
constitutively express arginase-I in the naive and inflamed lung. J. Leukoc. Biol. 94, 194. von Burg, N. et al. Activated group 3 innate lymphoid cells promote T-cell-mediated
877–884 (2013). immune responses. Proc. Natl Acad. Sci. USA 111, 12835–12840 (2014).
162. Terabe, M. et al. NKT cell-mediated repression of tumor immunosurveillance by IL-13 and 195. Rao, A. et al. Cytokines regulate the antigen-presenting characteristics of human
the IL-4R–STAT6 pathway. Nat. Immunol. 1, 515–520 (2000). circulating and tissue-resident intestinal ILCs. Nat. Commun. 11, 2049 (2020).
163. Kelly, J. et al. Chronically stimulated human MAIT cells are unexpectedly potent IL-13 196. Akagbosu, B. et al. Novel antigen-presenting cell imparts Treg-dependent tolerance to gut
producers. Immunol. Cell Biol. 97, 689–699 (2019). microbiota. Nature 610, 752–760 (2022).
164. Dalessandri, T., Crawford, G., Hayes, M., Castro Seoane, R. & Strid, J. IL-13 from 197. Lyu, M. et al. ILC3s select microbiota-specific regulatory T cells to establish tolerance in
intraepithelial lymphocytes regulates tissue homeostasis and protects against the gut. Nature 610, 744–751 (2022).
carcinogenesis in the skin. Nat. Commun. 7, 12080 (2016). 198. Kedmi, R. et al. A RORγt+ cell instructs gut microbiota-specific Treg cell differentiation.
165. Hao, J. et al. Regulatory role of Vγ1 γδ T cells in tumor immunity through IL-4 production. Nature 610, 737–743 (2022).
J. Immunol. 187, 4979–4986 (2011). 199. Goc, J. et al. Dysregulation of ILC3s unleashes progression and immunotherapy
166. Terabe, M. et al. A nonclassical non-Vα14Jα18 CD1d-restricted (type II) NKT cell is sufficient resistance in colon cancer. Cell 184, 5015–5030.e16 (2021).
for down-regulation of tumor immunosurveillance. J. Exp. Med. 202, 1627–1633 (2005). 200. Brandes, M., Willimann, K. & Moser, B. Professional antigen-presentation function by
167. Ambrosino, E. et al. Cross-regulation between type I and type II NKT cells in regulating human γδ T cells. Science 309, 264–268 (2005).
tumor immunity: a new immunoregulatory axis. J. Immunol. 179, 5126–5136 (2007). 201. Mao, C. et al. Tumor-activated TCRγδ+ T cells from gastric cancer patients induce the
168. Gasteiger, G. & Rudensky, A. Y. Interactions between innate and adaptive lymphocytes. antitumor immune response of TCRαβ+ T cells via their antigen-presenting cell-like
Nat. Rev. Immunol. 14, 631–639 (2014). effects. J. Immunol. Res. 2014, 593562 (2014).
169. Chang, Y. J. et al. Potent immune-modulating and anticancer effects of NKT cell 202. Roan, F., Obata-Ninomiya, K. & Ziegler, S. F. Epithelial cell-derived cytokines: more than
stimulatory glycolipids. Proc. Natl Acad. Sci. USA 104, 10299–10304 (2007). just signaling the alarm. J. Clin. Invest. 129, 1441–1451 (2019).
170. Van Acker, H. H. et al. Interleukin-15 enhances the proliferation, stimulatory phenotype, 203. Azzout, M. et al. IL-33 enhances IFNγ and TNFα production by human MAIT cells: a new
and antitumor effector functions of human γδ T cells. J. Hematol. Oncol. 9, 101 (2016). pro-Th1 effect of IL-33. Int. J. Mol. Sci. 22, 10602 (2021).
171. Kroemer, G., Galassi, C., Zitvogel, L. & Galluzzi, L. Immunogenic cell stress and death. 204. Ferhat, M. H. et al. The impact of invariant NKT cells in sterile inflammation: the possible
Nat. Immunol. 23, 487–500 (2022). contribution of the alarmin/cytokine IL-33. Front. Immunol. 9, 2308 (2018).
172. Harly, C., Robert, J., Legoux, F. & Lantz, O. γδ T, NKT, and MAIT cells during evolution: 205. Schwartz, C., O’Grady, K., Lavelle, E. C. & Fallon, P. G. Interleukin 33: an innate alarm
redundancy or specialized functions? J. Immunol. 209, 217–225 (2022). for adaptive responses beyond Th2 immunity-emerging roles in obesity, intestinal
173. Fehniger, T. A. et al. Differential cytokine and chemokine gene expression by human NK inflammation, and cancer. Eur. J. Immunol. 46, 1091–1100 (2016).
cells following activation with IL-18 or IL-15 in combination with IL-12: implications for the 206. Schuijs, M. J. et al. ILC2-driven innate immune checkpoint mechanism antagonizes
innate immune response. J. Immunol. 162, 4511–4520 (1999). NK cell antimetastatic function in the lung. Nat. Immunol. 21, 998–1009 (2020).
174. Baxevanis, C. N., Gritzapis, A. D. & Papamichail, M. In vivo antitumor activity of NKT cells 207. Sakuishi, K. et al. Invariant NKT cells biased for IL-5 production act as crucial regulators
activated by the combination of IL-12 and IL-18. J. Immunol. 171, 2953–2959 (2003). of inflammation. J. Immunol. 179, 3452–3462 (2007).
175. Lamichhane, R. et al. TCR- or cytokine-activated CD8+ mucosal-associated invariant T cells 208. de Oliveira Henriques, M. D. & Penido, C. γδ T lymphocytes coordinate eosinophil influx
are rapid polyfunctional effectors that can coordinate immune responses. Cell Rep. 28, during allergic responses. Front. Pharmacol. 3, 200 (2012).
3061–3076.e5 (2019). 209. Trabanelli, S. et al. Tumour-derived PGD2 and NKp30-B7H6 engagement drives an
176. Wculek, S. K. et al. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. immunosuppressive ILC2–MDSC axis. Nat. Commun. 8, 593 (2017).
Immunol. 20, 7–24 (2020). 210. Jovanovic, I. P. et al. Interleukin-33/ST2 axis promotes breast cancer growth and
177. Hegde, S. et al. NKT cells direct monocytes into a DC differentiation pathway. J. Leukoc. metastases by facilitating intratumoral accumulation of immunosuppressive and innate
Biol. 81, 1224–1235 (2007). lymphoid cells. Int. J. Cancer 134, 1669–1682 (2014).
178. Medina, B. D. et al. Oncogenic kinase inhibition limits Batf3-dependent dendritic cell 211. Zaiss, D. M. et al. Amphiregulin enhances regulatory T cell-suppressive function via the
development and antitumor immunity. J. Exp. Med. 216, 1359–1376 (2019). epidermal growth factor receptor. Immunity 38, 275–284 (2013).
179. Barry, K. C. et al. A natural killer-dendritic cell axis defines checkpoint therapy-responsive 212. Robertson, F. C., Berzofsky, J. A. & Terabe, M. NKT cell networks in the regulation of tumor
tumor microenvironments. Nat. Med. 24, 1178–1191 (2018). immunity. Front. Immunol. 5, 543 (2014).
180. Maniar, A. et al. Human γδ T lymphocytes induce robust NK cell-mediated antitumor 213. Mao, Y. et al. A new effect of IL-4 on human T cells: promoting regulatory Vδ1 T cells via
cytotoxicity through CD137 engagement. Blood 116, 1726–1733 (2010). IL-10 production and inhibiting function of Vδ2 T cells. Cell Mol. Immunol. 13, 217–228
181. Hayakawa, Y. et al. Critical contribution of IFN-γ and NK cells, but not perforin-mediated (2016).
cytotoxicity, to anti-metastatic effect of α-galactosylceramide. Eur. J. Immunol. 31, 214. Schneider, C., O’Leary, C. E. & Locksley, R. M. Regulation of immune responses by
1720–1727 (2001). tuft cells. Nat. Rev. Immunol. 19, 584–593 (2019).

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 369


Review article

215. Camelo, A. et al. Blocking IL-25 signalling protects against gut inflammation in a type-2 247. Veillette, A. & Guo, H. CS1, a SLAM family receptor involved in immune regulation, is a
model of colitis by suppressing nuocyte and NKT derived IL-13. J. Gastroenterol. 47, therapeutic target in multiple myeloma. Crit. Rev. Oncol. Hematol. 88, 168–177 (2013).
1198–1211 (2012). 248. Collins, S. M. et al. Elotuzumab directly enhances NK cell cytotoxicity against myeloma
216. Wu, P. et al. γδ T17 cells promote the accumulation and expansion of myeloid-derived via CS1 ligation: evidence for augmented NK cell function complementing ADCC.
suppressor cells in human colorectal cancer. Immunity 40, 785–800 (2014). Cancer Immunol. Immunother. 62, 1841–1849 (2013).
217. Kersten, K. et al. Mammary tumor-derived CCL2 enhances pro-metastatic systemic 249. Felices, M., Lenvik, T. R., Davis, Z. B., Miller, J. S. & Vallera, D. A. Generation of BiKEs and
inflammation through upregulation of IL1β in tumor-associated macrophages. TriKEs to improve NK cell-mediated targeting of tumor cells. Methods Mol. Biol. 1441,
Oncoimmunology 6, e1334744 (2017). 333–346 (2016).
218. Kimura, Y. et al. IL-17A-producing CD30+ Vδ1 T cells drive inflammation-induced cancer 250. Arvindam, U. S. et al. A trispecific killer engager molecule against CLEC12A effectively
progression. Cancer Sci. 107, 1206–1214 (2016). induces NK-cell mediated killing of AML cells. Leukemia 35, 1586–1596 (2021).
219. Carmi, Y. et al. Microenvironment-derived IL-1 and IL-17 interact in the control of lung 251. Gauthier, L. et al. Multifunctional natural killer cell engagers targeting NKp46 trigger
metastasis. J. Immunol. 186, 3462–3471 (2011). protective tumor immunity. Cell 177, 1701–1713.e16 (2019).
220. Chan, I. H. et al. Interleukin-23 is sufficient to induce rapid de novo gut tumorigenesis, 252. Demaria, O. et al. Antitumor immunity induced by antibody-based natural killer cell
independent of carcinogens, through activation of innate lymphoid cells. Mucosal engager therapeutics armed with not-α IL-2 variant. Cell Rep. Med. 3, 100783 (2022).
Immunol. 7, 842–856 (2014). 253. Ando, T., Ito, H., Ohtaki, H. & Seishima, M. Toll-like receptor agonists and
221. Rachitskaya, A. V. et al. Cutting edge: NKT cells constitutively express IL-23 receptor and α-galactosylceramide synergistically enhance the production of interferon-γ in murine
RORγt and rapidly produce IL-17 upon receptor ligation in an IL-6-independent fashion. splenocytes. Sci. Rep. 3, 2559 (2013).
J. Immunol. 180, 5167–5171 (2008). 254. Parekh, V. V. et al. Glycolipid antigen induces long-term natural killer T cell anergy in
222. Sutton, C. E., Mielke, L. A. & Mills, K. H. IL-17-producing γδ T cells and innate lymphoid mice. J. Clin. Invest. 115, 2572–2583 (2005).
cells. Eur. J. Immunol. 42, 2221–2231 (2012). 255. Kunii, N. et al. Combination therapy of in vitro-expanded natural killer T cells and
223. Rei, M. et al. Murine CD27– Vγ6+ γδ T cells producing IL-17A promote ovarian cancer growth α-galactosylceramide-pulsed antigen-presenting cells in patients with recurrent head
via mobilization of protumor small peritoneal macrophages. Proc. Natl Acad. Sci. USA 111, and neck carcinoma. Cancer Sci. 100, 1092–1098 (2009).
E3562–E3570 (2014). 256. Uchida, T. et al. Phase I study of α-galactosylceramide-pulsed antigen presenting cells
224. Elinav, E., Garrett, W. S., Trinchieri, G. & Wargo, J. The cancer microbiome. Nat. Rev. Cancer administration to the nasal submucosa in unresectable or recurrent head and neck
19, 371–376 (2019). cancer. Cancer Immunol. Immunother. 57, 337–345 (2008).
225. Xu, X. et al. Group-2 innate lymphoid cells promote HCC progression through 257. Toyoda, T. et al. Phase II study of α-galactosylceramide-pulsed antigen-presenting cells
CXCL2-neutrophil-induced immunosuppression. Hepatology 74, 2526–2543 (2021). in patients with advanced or recurrent non-small cell lung cancer. J. Immunother. Cancer
226. Zumwalde, N. A., Haag, J. D., Gould, M. N. & Gumperz, J. E. Mucosal associated invariant 8, e000316 (2020).
T cells from human breast ducts mediate a Th17-skewed response to bacterially exposed 258. Kobayashi, H. et al. Safety profile and anti-tumor effects of adoptive immunotherapy
breast carcinoma cells. Breast Cancer Res. 20, 111 (2018). using γδ T cells against advanced renal cell carcinoma: a pilot study. Cancer Immunol.
227. Housseau, F. et al. Redundant innate and adaptive sources of IL17 production drive colon Immunother. 56, 469–476 (2007).
tumorigenesis. Cancer Res. 76, 2115–2124 (2016). 259. Nicol, A. J. et al. Clinical evaluation of autologous γδ T cell-based immunotherapy for
228. Malmberg, K. J. et al. Natural killer cell-mediated immunosurveillance of human cancer. metastatic solid tumours. Br. J. Cancer 105, 778–786 (2011).
Semin. Immunol. 31, 20–29 (2017). 260. Hoeres, T., Smetak, M., Pretscher, D. & Wilhelm, M. Improving the efficiency of Vγ9Vδ2
229. Hoos, A. Development of immuno-oncology drugs — from CTLA4 to PD1 to the next T-cell immunotherapy in cancer. Front. Immunol. 9, 800 (2018).
generations. Nat. Rev. Drug Discov. 15, 235–247 (2016). 261. Walwyn-Brown, K. et al. Phosphoantigen-stimulated γδ T cells suppress natural killer-cell
230. Duan, M. et al. Activated and exhausted MAIT cells foster disease progression and responses to missing-self. Cancer Immunol. Res. 10, 558–570 (2022).
indicate poor outcome in hepatocellular carcinoma. Clin. Cancer Res. 25, 3304–3316 262. Roediger, B. et al. IL-2 is a critical regulator of group 2 innate lymphoid cell function
(2019). during pulmonary inflammation. J. Allergy Clin. Immunol. 136, 1653–1663.e57 (2015).
231. Heinrich, B. et al. Checkpoint inhibitors modulate plasticity of innate lymphoid cells in 263. Corpuz, T. M. et al. IL-2 shapes the survival and plasticity of IL-17-producing γδ T cells.
peripheral blood of patients with hepatocellular carcinoma. Front. Immunol. 13, 849958 J. Immunol. 199, 2366–2376 (2017).
(2022). 264. Krijgsman, D., Hokland, M. & Kuppen, P. J. K. The role of natural killer T cells in cancer — a
232. Rossi, C. et al. Boosting γδ T cell-mediated antibody-dependent cellular cytotoxicity by phenotypical and functional approach. Front. Immunol. 9, 367 (2018).
PD-1 blockade in follicular lymphoma. Oncoimmunology 8, 1554175 (2019). 265. Ghiringhelli, F. et al. CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a
233. Zhang, Q. et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion transforming growth factor-β-dependent manner. J. Exp. Med. 202, 1075–1085 (2005).
and elicits potent anti-tumor immunity. Nat. Immunol. 19, 723–732 (2018). 266. Viey, E. et al. Chemokine receptors expression and migration potential of tumor-
234. de Vries, N. L. et al. γδ T cells are effectors of immunotherapy in cancers with HLA class I infiltrating and peripheral-expanded Vγ9Vδ2 T cells from renal cell carcinoma patients.
defects. Nature 613, 743–750 (2023). J. Immunother. 31, 313–323 (2008).
This study suggests a contribution of PD1+ γδ T cells to the control of DNA mismatch 267. Felices, M. et al. Continuous treatment with IL-15 exhausts human NK cells via a metabolic
repair-deficient (dMMR) cancers with additional defects in antigen presentation defect. JCI Insight 3, e96219 (2018).
(HLA-I-negative) upon immune checkpoint blockade. 268. Gordy, L. E. et al. IL-15 regulates homeostasis and terminal maturation of NKT cells.
235. De Biasi, S. et al. Circulating mucosal-associated invariant T cells identify patients J. Immunol. 187, 6335–6345 (2011).
responding to anti-PD-1 therapy. Nat. Commun. 12, 1669 (2021). 269. Aehnlich, P., Carnaz Simoes, A. M., Skadborg, S. K., Holmen Olofsson, G. & Thor Straten,
236. Daley, D. et al. γδ T cells support pancreatic oncogenesis by restraining αβ T cell P. Expansion with IL-15 increases cytotoxicity of Vγ9Vδ2 T cells and is associated with
activation. Cell 166, 1485–1499.e15 (2016). higher levels of cytotoxic molecules and T-bet. Front. Immunol. 11, 1868 (2020).
237. Sivori, S. et al. NK cells and ILCs in tumor immunotherapy. Mol. Asp. Med. 80, 100870 270. Rha, M. S. et al. Human liver CD8+ MAIT cells exert TCR/MR1-independent innate-like
(2021). cytotoxicity in response to IL-15. J. Hepatol. 73, 640–650 (2020).
238. Ruggeri, L. et al. Effects of anti-NKG2A antibody administration on leukemia and normal 271. Tourret, M. et al. Human MAIT cells are devoid of alloreactive potential: prompting their
hematopoietic cells. Haematologica 101, 626–633 (2016). use as universal cells for adoptive immune therapy. J. Immunother. Cancer 9, e003123
239. Andre, P. et al. Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor (2021).
immunity by unleashing both T and NK cells. Cell 175, 1731–1743.e13 (2018). This study suggests that MAIT cells could be an attractive platform for adoptive cell
240. Segal, N. H. et al. First-in-human dose escalation of monalizumab plus durvalumab, with transfer owing to their lack of alloreactivity.
expansion in patients with metastatic microsatellite-stable colorectal cancer. J. Clin. Oncol. 272. Laskowski, T. J., Biederstadt, A. & Rezvani, K. Natural killer cells in antitumour adoptive
36, 3540–3540 (2018). cell immunotherapy. Nat. Rev. Cancer 22, 557–575 (2022).
241. Sivori, S. et al. Inhibitory receptors and checkpoints in human NK cells, implications for 273. Lupo, K. B. & Matosevic, S. Natural killer cells as allogeneic effectors in adoptive cancer
the immunotherapy of cancer. Front. Immunol. 11, 2156 (2020). immunotherapy. Cancers 11, 769 (2019).
242. Korde, N. et al. A phase II trial of pan-KIR2D blockade with IPH2101 in smoldering multiple 274. Miller, J. S. et al. Successful adoptive transfer and in vivo expansion of human
myeloma. Haematologica 99, e81–e83 (2014). haploidentical NK cells in patients with cancer. Blood 105, 3051–3057 (2005).
243. Carlsten, M. et al. Checkpoint inhibition of KIR2D with the monoclonal antibody IPH2101 275. Li, L. et al. Adoptive transfer of natural killer cells in combination with chemotherapy
induces contraction and hyporesponsiveness of NK cells in patients with myeloma. improves outcomes of patients with locally advanced colon carcinoma. Cytotherapy 20,
Clin. Cancer Res. 22, 5211–5222 (2016). 134–148 (2018).
244. Benson, D. M. Jr et al. A phase I trial of the anti-KIR antibody IPH2101 and lenalidomide 276. Yoon, D. H., Koh, Y., Park, H., Hwang, Y. K. & Kim, W. S. A phase 1 study of the combination
in patients with relapsed/refractory multiple myeloma. Clin. Cancer Res. 21, 4055–4061 of MG4101, ex vivo-expanded allogeneic NK cells and rituximab for relapsed or refractory
(2015). non-Hodgkin’s lymphoma. Blood 136, 14–15 (2020).
245. Hsi, E. D. et al. CS1, a potential new therapeutic antibody target for the treatment of 277. Motohashi, S. et al. A phase I study of in vitro expanded natural killer T cells in patients
multiple myeloma. Clin. Cancer Res. 14, 2775–2784 (2008). with advanced and recurrent non-small cell lung cancer. Clin. Cancer Res. 12, 6079–6086
246. Cruz-Munoz, M. E., Dong, Z., Shi, X., Zhang, S. & Veillette, A. Influence of CRACC, (2006).
a SLAM family receptor coupled to the adaptor EAT-2, on natural killer cell function. 278. Exley, M. A. et al. Adoptive transfer of invariant NKT cells as immunotherapy for advanced
Nat. Immunol. 10, 297–305 (2009). melanoma: a phase I clinical trial. Clin. Cancer Res. 23, 3510–3519 (2017).

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 370


Review article

279. Gao, Y. et al. Adoptive transfer of autologous invariant natural killer T cells as immunotherapy 309. Di Marco Barros, R. et al. Epithelia use butyrophilin-like molecules to shape organ-specific
for advanced hepatocellular carcinoma: a phase I clinical trial. Oncologist 26, e1919–e1930 γδ T cell compartments. Cell 167, 203–218.e17 (2016).
(2021). 310. Farber, D. L., Netea, M. G., Radbruch, A., Rajewsky, K. & Zinkernagel, R. M. Immunological
280. Parrot, T. et al. Expansion of donor-unrestricted MAIT cells with enhanced cytolytic memory: lessons from the past and a look to the future. Nat. Rev. Immunol. 16, 124–128
function suitable for TCR redirection. JCI Insight 6, e140074 (2021). (2016).
281. June, C. H. & Sadelain, M. Chimeric antigen receptor therapy. N. Engl. J. Med. 379, 64–73 311. Adams, N. M., Grassmann, S. & Sun, J. C. Clonal expansion of innate and adaptive
(2018). lymphocytes. Nat. Rev. Immunol. 20, 694–707 (2020).
282. Melenhorst, J. J. et al. Decade-long leukaemia remissions with persistence of CD4+ 312. Sun, J. C., Beilke, J. N. & Lanier, L. L. Adaptive immune features of natural killer cells.
CAR T cells. Nature 602, 503–509 (2022). Nature 457, 557–561 (2009).
283. Liu, E. et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. 313. Wang, X. et al. Memory formation and long-term maintenance of IL-7Rα+ ILC1s via a lymph
N. Engl. J. Med. 382, 545–553 (2020). node–liver axis. Nat. Commun. 9, 4854 (2018).
284. Basar, R., Daher, M. & Rezvani, K. Next-generation cell therapies: the emerging role of 314. Martinez-Gonzalez, I. et al. Allergen-experienced group 2 innate lymphoid cells acquire
CAR-NK cells. Blood Adv. 4, 5868–5876 (2020). memory-like properties and enhance allergic lung inflammation. Immunity 45, 198–208
285. Rozenbaum, M. et al. γδ CAR-T cells show CAR-directed and independent activity against (2016).
leukemia. Front. Immunol. 11, 1347 (2020). 315. Olszak, T. et al. Microbial exposure during early life has persistent effects on natural killer
286. Capsomidis, A. et al. Chimeric antigen receptor-engineered human γδ T cells: enhanced T cell function. Science 336, 489–493 (2012).
cytotoxicity with retention of cross presentation. Mol. Ther. 26, 354–365 (2018). 316. Ataide, M. A. et al. Lymphatic migration of unconventional T cells promotes site-specific
287. Heczey, A. et al. Invariant NKT cells with chimeric antigen receptor provide a novel immunity in distinct lymph nodes. Immunity 55, 1813–1828.e19 (2022).
platform for safe and effective cancer immunotherapy. Blood 124, 2824–2833 (2014). This article demonstrates how ILTCs represent functional units in different tissues
288. Dogan, M. et al. Engineering human MAIT cells with chimeric antigen receptors for where their function is imprinted by the homeostatic niche they are derived from.
cancer immunotherapy. J. Immunol. 209, 1523–1531 (2022). 317. Bal, S. M., Golebski, K. & Spits, H. Plasticity of innate lymphoid cell subsets. Nat. Rev.
289. Du, S. H. et al. Co-expansion of cytokine-induced killer cells and Vγa9Vδ2 T cells for CAR Immunol. 20, 552–565 (2020).
T-cell therapy. PLoS ONE 11, e0161820 (2016). 318. Colonna, M. Innate lymphoid cells: diversity, plasticity, and unique functions in immunity.
290. Papotto, P. H., Ribot, J. C. & Silva-Santos, B. IL-17+ γδ T cells as kick-starters of Immunity 48, 1104–1117 (2018).
inflammation. Nat. Immunol. 18, 604–611 (2017). 319. DuPage, M. & Bluestone, J. A. Harnessing the plasticity of CD4+ T cells to treat
291. Luci, C. et al. Cutaneous squamous cell carcinoma development is associated with a immune-mediated disease. Nat. Rev. Immunol. 16, 149–163 (2016).
temporal infiltration of ILC1 and NK cells with immune dysfunctions. J. Invest. Dermatol. 320. Galli, S. J., Borregaard, N. & Wynn, T. A. Phenotypic and functional plasticity of cells of
141, 2369–2379 (2021). innate immunity: macrophages, mast cells and neutrophils. Nat. Immunol. 12, 1035–1044
292. Koay, H. F. et al. A three-stage intrathymic development pathway for the mucosal- (2011).
associated invariant T cell lineage. Nat. Immunol. 17, 1300–1311 (2016). 321. Bald, T., Wagner, M., Gao, Y., Koyasu, S. & Smyth, M. J. Hide and seek: plasticity of innate
293. Savage, A. K. et al. The transcription factor PLZF directs the effector program of the lymphoid cells in cancer. Semin. Immunol. 41, 101273 (2019).
NKT cell lineage. Immunity 29, 391–403 (2008). 322. Cortez, V. S. et al. SMAD4 impedes the conversion of NK cells into ILC1-like cells by
294. Kreslavsky, T. et al. TCR-inducible PLZF transcription factor required for innate phenotype curtailing non-canonical TGF-β signaling. Nat. Immunol. 18, 995–1003 (2017).
of a subset of γδ T cells with restricted TCR diversity. Proc. Natl Acad. Sci. USA 106, 323. Eisenring, M., vom Berg, J., Kristiansen, G., Saller, E. & Becher, B. IL-12 initiates tumor
12453–12458 (2009). rejection via lymphoid tissue-inducer cells bearing the natural cytotoxicity receptor
295. Constantinides, M. G. et al. MAIT cells are imprinted by the microbiota in early life and NKp46. Nat. Immunol. 11, 1030–1038 (2010).
promote tissue repair. Science 366, eaax6624 (2019). 324. Brennan, P. J., Brigl, M. & Brenner, M. B. Invariant natural killer T cells: an innate activation
296. Legoux, F. et al. Microbial metabolites control the thymic development of mucosal- scheme linked to diverse effector functions. Nat. Rev. Immunol. 13, 101–117 (2013).
associated invariant T cells. Science 366, 494–499 (2019). 325. Lafont, V. et al. Plasticity of γδ T cells: impact on the anti-tumor response. Front. Immunol.
297. Wingender, G. et al. Intestinal microbes affect phenotypes and functions of invariant 5, 622 (2014).
natural killer T cells in mice. Gastroenterology 143, 418–428 (2012).
298. Lim, A. I. et al. Systemic human ILC precursors provide a substrate for tissue ILC Acknowledgements
differentiation. Cell 168, 1086–1100.e10 (2017). The authors thank J. Zhu (NIH) for critical reading of the manuscript and helpful suggestions
This article describes comprehensive characterization of circulating ILC precursor and comments. The authors apologize to all the scientists whose work could not be cited
cells in human peripheral blood. owing to space limitations. B.R. was supported by the International Liver Cancer Association
299. Scoville, S. D., Freud, A. G. & Caligiuri, M. A. Cellular pathways in the development of Fellowship Award 2021. T.F.G. was supported by the Intramural Research Program of the NIH,
human and murine innate lymphoid cells. Curr. Opin. Immunol. 56, 100–106 (2019). NCI (ZIA BC 011345).
300. Male, V. et al. The transcription factor E4bp4/Nfil3 controls commitment to the NK
lineage and directly regulates Eomes and Id2 expression. J. Exp. Med. 211, 635–642 Author contributions
(2014). F.K. and B.R. researched data for the article. All authors contributed substantially to discussion
301. Bai, L. et al. Liver type 1 innate lymphoid cells develop locally via an interferon-γ-dependent of the content, as well as writing, reviewing and editing the manuscript before submission.
loop. Science 371, eaba4177 (2021).
302. Ebihara, T. & Taniuchi, I. Transcription factors in the development and function of group 2 Competing interests
innate lymphoid cells. Int. J. Mol. Sci. 20, 1377 (2019). The authors declare no competing interests.
303. Cording, S. et al. Mouse models for the study of fate and function of innate lymphoid
cells. Eur. J. Immunol. 48, 1271–1280 (2018). Additional information
304. Zook, E. C. & Kee, B. L. Development of innate lymphoid cells. Nat. Immunol. 17, 775–782 Peer review information Nature Reviews Cancer thanks Hergen Spits, Pamela Ohashi and
(2016). Nicolas Jacquelot and the other, anonymous, reviewer(s) for their contribution to the peer
305. Stokic-Trtica, V., Diefenbach, A. & Klose, C. S. N. NK cell development in times of innate review of this work.
lymphoid cell diversity. Front. Immunol. 11, 813 (2020).
306. Kotas, M. E. & Locksley, R. M. Why innate lymphoid cells. Immunity 48, 1081–1090 (2018). Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in
307. Kogame, T., Egawa, G., Nomura, T. & Kabashima, K. Waves of layered immunity over published maps and institutional affiliations.
innate lymphoid cells. Front. Immunol. 13, 957711 (2022).
308. Mueller, S. N. & Mackay, L. K. Tissue-resident memory T cells: local specialists in immune This is a U.S. Government work and not under copyright protection in the US; foreign
defence. Nat. Rev. Immunol. 16, 79–89 (2016). copyright protection may apply 2023

Nature Reviews Cancer | Volume 23 | June 2023 | 351–371 371

You might also like