You are on page 1of 10

TICB 1612 No.

of Pages 10

Trends in Cell Biology

Review

The Roles of CD8+ T Cell Subsets in


Antitumor Immunity
Michael St. Paul1,2 and Pamela S. Ohashi1,2,*

Effector CD8+ T cells are typically thought to be a homogenous group of Highlights


cytotoxic cells that produce interferon-(IFN) γ. However, recent findings have There are multiple subsets of CD8+
challenged this notion because multiple subsets of CD8+ T cells have been de- T cells, not all of which have cytotoxic
function and produce IFN-γ.
scribed, each with distinct effector functions and cytotoxic potential. These
subsets, referred to as the Tc subsets, have also been detected in tumor micro- A variety of Tc subsets have been de-
environments (TMEs), where they potentially influence the antitumor response tected within the TME and, depending
and patient outcomes. In this review, we highlight the prevalence and roles of on the subset, can be either positively
or negatively correlated with prognosis.
Tc subsets in the TME. We also discuss their therapeutic applications in the con-
text of adoptive immunotherapy to treat cancer. In the context of adoptive immunother-
apy to treat cancer, the degree of tumor
control is greatly influenced by the sub-
CD8+ T Cells: More Than Just IFN-γ and Granzyme set of T cells transferred.
The immune system comprises multiple cell subtypes, which cooperate to defend the body from
pathogens and tumors. T cells have an important role in both orchestrating the overall immune
response and directly killing damaged cells. Typically, these functions are mediated by CD4+
and CD8+ T cells, respectively. Several different subsets of effector CD4+ T cells have been iden-
tified, each with distinct cytokine profiles, surface markers, transcriptomes, and roles in diseases
[1]. However, CD8+ T cells are typically regarded as being a uniform population of cells that secret
large amounts of IFN-γ and the protease granzyme B, which act synergistically to kill infected or
tumorigenic cells. As a result, this outlook does not fully encompass the diversity within the CD8+
effector T cell pool. In addition to regulatory [2] and follicular CD8+ T cell populations [3], multiple
types of effector CD8+ Tc subsets mirroring the CD4+ T helper subsets have been identified. Each
CD8+ Tc subset is functionally distinct and potentially has differential roles during antitumor im-
mune responses. In this review, we provide an overview of the differential roles of each subset,
before focusing on the antitumor immune responses they elicit.

The CD8+ Tc Subsets


Differentiation towards a specific CD8+ Tc lineage in vitro is mediated by the presence of a defined
cocktail of polarizing cytokines present during the initial activation of a naïve T cell. These cytokines
invoke signaling cascades leading to the expression of specific transcription factors, which facilitate
lineage commitment and adoption of distinct effector phenotypes. Each Tc subset expresses a
diverse profile of cytokines and has unexpected differences in their cytotoxic potential, with
some subsets showing poor cytolytic ability. To date, several CD8+ Tc subsets have been identi-
fied, including the conventional IFN-γ-producing Tc1s, interleukin (IL)-4 producing Tc2s, IL-9 1
Princess Margaret Cancer Center,
producing Tc9s, IL-17 producing Tc17s, and IL-22 producing Tc22s (Figure 1). (See Table 1.) University Health Network, Toronto, ON,
M5G 2C1, Canada
2
Tc1 Cells: The Typical Cytotoxic T Cell Department of Immunology, University
As typical cytotoxic CD8+ T cells, Tc1s demonstrate exceptional cytotoxic activity and efficiently of Toronto, Toronto, ON, M5S 1C1,
Canada
kill tumor cells and cells harboring intracellular pathogens. Induction of Tc1s is mediated by the
cytokine IL-12 [4], typically produced by antigen-presenting cells (APCs), including macrophages
and dendritic cells, upon exposure to pathogen-derived maturation stimuli. Functionally, Tc1 cells *Correspondence:
are defined by their high levels of perforin, granzyme B, IFN-γ, and tumor necrosis factor (TNF-α) pohashi@uhnresearch.ca (P.S. Ohashi).

Trends in Cell Biology, Month 2020, Vol. xx, No. xx https://doi.org/10.1016/j.tcb.2020.06.003 1


© 2020 Elsevier Ltd. All rights reserved.
Trends in Cell Biology

Trends in Cell Biology

Figure 1. The CD8+ Tc Subsets. Upon initial exposure to antigen in the presence of polarizing cytokines, naïve CD8+ T
cells can differentiate into a variety of different Tc subsets, each producing distinct cytokine profiles. Abbreviations: IFN,
interferon; IL, interleukin; TGF, transforming growth factor; TNF, tumor necrosis factor

in conjunction with the low expression of cytokines associated with other Tc lineages, namely IL-4,
IL-9, and IL-17 [5]. In addition to their cytokine profile, Tc1 cells can also be distinguished from
other Tc subsets by their elevated expression of IL18R on the cell surface [6,7].

Polarization of Tc1 cells is mediated by several key transcription factors, including signal trans-
ducer and activator of transcription (STAT) 4, T-bet, and eomesodermin (EOMES). Being directly
downstream of the IL-12 receptor, STAT4 has an early role in upregulating T-bet, which together
promote the production of IFN-γ [8]. This process is supported by EOMES, which has been
shown to cooperate with T-bet to further enhance IFN-γ production [9]. In fact, this synergy
between T-bet and EOMES is critical for the effector properties of Tc1 cells, as demonstrated
through gene knockout studies, which show a diminished ability to clear viral infections in CD8+
T cells lacking both of these transcription factors [10].

Tc2 Cells
CD8+ Tc2 cells were the first additional Tc subset identified following Tc1. They are known for
their production of type II cytokines, such as IL-4, IL-5, and IL-13, in conjunction with diminished
production of IFN-γ [5,11–13]. Tc2 cells also express high levels of granzyme B and demonstrate

Table 1. The CD8+ Tc Subsets


Subset Effector cytokines Transcription factors Surface markers Cytotoxic Refs
Tc1 IFN-γ, TNF-α T-bet, EOMES, IL18R Yes [6,8]
STAT4
Tc2 IL-4, IL-5, IL-13 GATA3, STAT6 CRTH2 Yes [14,15,17]
Tc9 IL-9 IRF4, STAT6 – No [18,19]
Tc17 IL-17A, IL-17F, RORyT, IRF4, IL23R, CD38, CD86, No [7,23,26,28]
IL-21, IL-22 STAT3 CD101, CD161, CCR6,
41BBlo
Tc22 IL-22, TNF-α, IL-2 AhR – Yes [7]

2 Trends in Cell Biology, Month 2020, Vol. xx, No. xx


Trends in Cell Biology

robust cytotoxic abilities, comparable with those of Tc1 cells [14]. The polarization of Tc2 cells is
driven by the cytokine IL-4, which activates the transcription factors STAT6 and GATA3 to pro-
mote the expression of genes essential for the Tc2 lineage [15]. Given that these cells secrete
an abundance of type II cytokines, which can promote IgE production and the recruitment of
pathogenic cells, such as eosinophils, Tc2s have an important role in mediating allergy
responses, particularly in the respiratory tract [16,17].

Tc9 Cells
One of the more recently described Tc subsets are CD8+ Tc9 cells, which produce IL-9 but rel-
atively little IFN-γ [18,19]. However, given the controversies surrounding the notion that T helper
(Th)9 cells are a lineage distinct from Th2 cells [20], it remains to be fully determined whether the
Tc9 lineage is distinct from Tc2 cells. Nevertheless, the polarizing conditions for Tc9 cells have
been defined and include the combined actions of the cytokines IL-4 and TGF-β, mediated by
the transcription factors STAT6 and interferon regulatory factor 4 (IRF4) [18]. Surprisingly,
unlike Tc1 and Tc2 cells, Tc9 cells demonstrate poor cytotoxic function owing, in part, to their
limited production of granzyme B [18]. The biological relevance of Tc9 cells and their lack of
cytotoxicity is currently unclear. However, Tc9 cells have been detected in the small intestine
[21] as well as in atopic dermatitis lesions [18] and in the blood of patients with asthma [22],
where they are thought to promote disease progression.

Tc17 Cells
IL-17-producing CD8+ Tc17 cells are a distinct subset of T cells known for the high production of
IL-17A, IL-17F, and IL-22, in conjunction with their low expression of IFN-γ [10,23,24]. The com-
bination of IL-6 and TGF-β drive the differentiation of Tc17 cells, which is further enhanced by the
addition of IL-1β, IL-21, and/or IL-23 [25,26]. IL-6 signaling leads to activation of the transcription
factors STAT3 and RORγt, which are essential for the differentiation towards the Tc17 lineage
[23,27]. Similar to Tc9 cells, Tc17s also express low levels of granzyme B and have poor cytolytic
function [23]. Several surface markers for Tc17 cells have been identified at the mRNA or protein
level including, IL-23R, CD38, CD86, CD101, CD103, and CCR6 [7,23,26,28]. The biological
relevance of Tc17 cells is more defined than many of the other newer subsets and these cells
have been detected in human skin tissue under steady-state conditions [29,30] as well as within
the tumor micro-environment (TME) [31], and are thought to be protective against certain fungal
pathogens [32]. Moreover, deleterious roles of Tc17 cells have been identified in the context of
autoimmune disorders, including multiple sclerosis [33], and in responses to graft versus host
disease [34,35]. In addition to acting as effector cells, evidence suggests that Tc17 can also
differentiate into long-lived memory T cells (Box 1).

Box 1. Memory Tc Subsets


Memory CD8+ T cells serve as a long-lived reservoir of antigen-specific T cells poised to rapidly respond upon re-
encountering antigen. Part of this response includes robust IFN-γ production, suggesting that many memory CD8+ T cells
are derived from the Tc1 lineage [40]. However, whether memory cells originate from other Tc subsets is not clear. There is
some evidence to suggest that Tc17 cells can differentiate into a long-lived population that can persist for over a year
[32,41,42]. Moreover, these memory Tc17 cells undergo robust proliferation and IL-17 production upon re-exposure to
antigen and mediate protection against fungal challenge [32,42]. Metabolically, Tc17 cells demonstrate increased maximal
oxidative respiration capacity, which is a feature typically attributed to memory T cells [41]. Interestingly, in one study, it
was found that memory Tc17 cells were CD62L–, while memory Tc1 cells were CD62L+ and resembled central memory T
cells [32]. This suggests that memory Tc17 cells do not always adopt a central memory phenotype. In support of this
hypothesis, it was found that Tc17 cells express higher levels of the tissue-resident memory T cell (TRM) marker CD103
relative to Tc1 cells [26]. Indeed, tissue-resident Tc17 cells have been detected in the skin of healthy patients and are thought
to have a role in responses against commensal microorganisms [30]. Taken together, these studies provide early evidence
that subsets other than Tc1 cells can differentiate into long-lived memory populations.

Trends in Cell Biology, Month 2020, Vol. xx, No. xx 3


Trends in Cell Biology

Tc22 Cells
Tc22 cells are a subset of CD8+ T cells that primarily produce IL-22 with little production of the
other lineage-defining cytokines, including IL-17. Analogous to Th22 cells, it is controversial as
to whether Tc22 cells can be classified as a distinct T cell lineage, given that Th17 and Tc17
cells can also produce IL-22 [36]. Tc22s have been detected in inflamed lesions, such as atopic
dermatitis and psoriasis, where they are thought to contribute to disease pathology [36,37], in
response to viral infections, including HIV [38], and have been found to infiltrate squamous cell
carcinoma [39]. Recent research found that IL-6 in conjunction with TNF-α and an aryl-
hydrocarbon receptor (AhR) agonist were the driving factors of Tc22 differentiation [7]. Through
gene knockout studies, it was demonstrated that the AhR transcription factor was critical for
commitment towards the Tc22 lineage. Functionally, Tc22s expressed granzyme B, were highly
cytolytic, and provided exceptional antitumor activity in adoptive transfer studies. Although the
biological significance of Tc22s is still relatively undefined, they can be detected in human ovarian
cancer patients, where they can comprise up to 35% of CD8+ T cells expanded from tumors [7].

Tc Subsets in the TME


The TME is a hive of immune activity where different cell types act to either promote or inhibit the
growth of a tumor. A variety of immune cells can be detected in the TME, including CD8+ T cells
and their subsets. The most frequent subset described in the literature is the classical IFNγ+ Tc1
cell, which has been detected in human and mouse tumor-infiltrating lymphocytes (TIL) isolated

Table 2. Frequency of CD8+ Tc Subsets Infiltrating the TME


Subset Tumor (species) Frequency Frequency of Tc1 Refs
(% of total CD8+ TILs) (% of total CD8+ TILs)
Tc2 Cervical (human) 67% b1% [48]
Breast (mouse) ~50% ~50% [80,81]
Lung (human) 2.2% 64% [82,83]
Chronic lymphocytic leukemia (human) 4.2% 48% [84]
Lymphoma (human) 2–4% 0–2.5% [85]
Breast (human) ~10%a ~16%a [49,50]
Tc17 Liver (human) 1–5% Not reported [51]
Head + neck (mouse) 10% Not reported [31]
Melanoma (mouse) 4% Not reported [31]
Prostate (mouse) 30% Not reported [31]
Sarcoma (mouse) 7% Not reported [31]
a a
Breast (human) 1–29% 6–66% [50]
Ovarian (human) 0–20% 1–99% [7,86]
Gastric (human) ~10% Not reported [52,87]
Nasopharyngeal (human) ~1% Not reported [88]
Bile duct (human) ~0.5% ~45% [89]
Head + neck (human) ~2%b Not reported [53]
b
Gallbladder (human) ~2% Not reported [90]
Tc22 Ovarian (human) 0–35% 1–99% [7]
Squamous cell carcinoma (human) ~3% ~60% [39]
Gastric (human) 0–18% Not reported [52]

a
Tumor-draining lymph node.
b
Peripheral blood.

4 Trends in Cell Biology, Month 2020, Vol. xx, No. xx


Trends in Cell Biology

from various cancers, including melanoma, ovarian, breast, and lung (Table 2). The presence of
Tc1s tends to correlate with a more favorable prognosis [43], likely due to their exceptional cyto-
toxic potential in conjunction with IFN-γ [44]. In the TME, IFN-γ synergizes with activation signals
to upregulate MHC-I on APCs, thereby enhancing antigen presentation and the activation of naïve
T cells in the tumor and draining lymph nodes [45]. Moreover, IFN-γ can also help to reprogram
the suppressive cells in the TME, such as regulatory T cells (Tregs), to disarm one of the weapons
that tumors use to evade killing [46]. Furthermore, IFN-γ also acts directly on tumor cells to upreg-
ulate MHC-I, thereby increasing their susceptibility to CD8+ T cell-dependent cytotoxicity [45]. In
fact, the direct effects of IFN-γ on tumor cells are critical to the antitumor response, as exemplified
by the fact that patients with melanoma who have tumors with loss-of-function mutations in sig-
naling molecules downstream of the IFN-γ receptor are resistant to immunotherapy, potentially
due to alterations in immunoediting [47].

Despite the literature being predominantly Tc1 centric, other Tc subsets have been detected as
part of the CD8+ TILs. Surprisingly, in some patients, subsets other than Tc1 cells comprise a
greater percentage of the CD8+ TILs (Table 2). For instance, Sheu and colleagues analyzed the
cytokine expression pattern of CD8+ TILs from eight patients with cervical cancer [48]. Using
IFN-γ and IL-5 as surrogate markers for Tc1 and Tc2 cells, respectively, they found that 67%
of the CD8+ T cells infiltrating the tumor were Tc2 cells, compared with b1% of Tc1 cells.
However, the clinical relevance and prognostic indicator of increased Tc2 cells within the TME
is currently unclear. In patients with breast cancer, some evidence suggests that the Tc2:Tc1
ratio in the tumor training lymph node (TDLN) does not correlate with tumor size [49]. Instead,
as demonstrated by Faghih et al. it may correlate with stage, because patients with stage III
breast cancer had nearly twice as many Tc2 cells in the TDLN compared with patients with
stage II cancer, while the percentage of Tc1 cells remained constant [50]. Given that tumor
stage negatively correlates with patient survival, Tc2 cells may also negatively correlate with
survival in breast cancer, and this possibility should be further investigated.

Multiple studies have also detected Tc17s in several mouse and human tumors, such as in hepa-
tocellular carcinoma, melanoma, prostate cancer, and gastric cancer [31,51,52]. In one study by
Zhuang and colleagues, the amount of Tc17s in the TILs of patients with gastric cancer was sig-
nificantly higher in stage III and IV tumors compared with those in stage I and II [52]. Importantly,
the patients with high Tc17s in their TIL had a significant decrease in overall survival relative to
those with low amounts of Tc17s. Similarly, it has been shown that patients with head and
neck cancer with high levels of circulating Tc17s have a significantly worse overall survival com-
pared with patients with low levels of Tc17s [53]. Although the mechanisms behind this are not
clear, it is possible this poor prognosis is mediated in part by IL-17. In this context, IL-17 can
directly act on tumor cells or other cell types within the TME to promote the production of
inhibitory and angiogenic factors, such as vascular endothelial growth factor (VEGF), in addition
to facilitating the recruitment of cell types that can be protumorigenic, including neutrophils [54]
and myeloid derived suppressor cells (MDSCs) [55]. Indeed, a meta-analysis of human cancers
correlated increased IL-17 levels with poor overall survival for multiple cancers, including hepato-
cellular carcinoma and lung cancer [56,57]. However, in some cases, such as in esophageal
cancer, it can also be a positive prognostic factor [56]. Therefore, although current findings sug-
gest that Tc17 cells in the tumor have a limited capacity to control tumor growth, there may be a
small subset of tumors in which Tc17 and IL-17 production can be beneficial.

Being relatively novel subsets there is less information as to the presence and significance of Tc9
and Tc22 cells within the TME. Nevertheless, Tc22 cells have been found in patients with gastric
cancer [52] and those with transplant-associated squamous cell carcinoma [39]. Recently, Tc22s

Trends in Cell Biology, Month 2020, Vol. xx, No. xx 5


Trends in Cell Biology

were also found in ~30% of patients with ovarian cancer, where they comprised up to 35% of
expanded CD8+ TILs and, in some patients, outnumbered either Tc1 or Tc17 subsets [7].
Importantly, patients with increased amounts of IL-22-producing CD8+ T cells had a significantly
longer recurrence-free survival, suggesting a potential prognostic significance for these cells in
ovarian cancer. Together, this and other research clearly demonstrates that subsets other than
Tc1 cells can be found within the TME and have a role in the antitumor response and prognosis.

Role of the TME in Tc Polarization


It is as yet unclear whether the Tc subsets detected within the TME are polarized elsewhere and
recruited to the TME, or are polarized upon entering the TME due to factors present in this envi-
ronment. In support of the latter hypothesis, there is evidence to suggest that tertiary lymphoid
structures can arise within TME that actively recruit and activate naïve T cells [58–60]. Once in
the tumor, the naïve T cells can be influenced by cytokines typically found in high levels in the
TME, such as TGF-β, to facilitate commitment to the Tc9 and Tc17 lineages. Moreover, the
naïve T cells may interact with APCs within the TME and nearby tertiary lymphoid structures,
many of which are dysfunctional and produce altered cytokine profiles and co-stimulatory
molecules that can influence the lineage commitment of these CD8+ T cells. Indeed, one study
found that macrophages isolated from human hepatocellular carcinoma tumors, but not from
non-tumor tissue, gave rise to Tc17 cells when cultured with CD8+ T cells in vitro in the absence
of exogenous polarizing cytokines [51].This was shown to occur primarily by IL-1β and IL-23
secreted by these tumor-associated macrophages, which help drive commitment to the Tc17
lineage. As for Tc1 cells, there are conflicting results because APCs isolated from TME can either
enhance [61] or inhibit [62] Tc1 differentiation and IFN-γ production. However, whether direct
T cell APC interactions within the TME are important relative to antigen presentation in the
lymph node remains to be clarified. Whether APC cells can induce other Tc subsets within the
TME is also not clear; however, evidence has shown that certain APC populations can preferen-
tially induce Tc9 [63] and Tc22 [64] subsets. It is also possible that existing Tc subsets can be
converted from one subset to another upon exposure to factors within the TME. Indeed, plasticity
has been demonstrated, for example, in Tc9 and Tc17 cells because they can switch to a Tc1
phenotype upon transfer into tumor-bearing mice [19,24]. Taken together, it is clear that the
importance of the TME in shaping the CD8+ T cell response is beginning to be unraveled;
however, future studies should aim to better understand the mechanisms involved and the
impact of these subsets on antitumor responses.

Adoptive Immunotherapy with Polarized T Cells


Harnessing the power of the immune system to treat cancer is a promising therapeutic approach.
Many types of immunotherapies are being actively explored, and successful strategies include
the transfer of tumor-specific T cells including those expressing transgenic T cell receptors
(TCR) or chimeric antigen receptors (CAR) as well as TIL therapy, which utilizes T cells isolated
from the patient’s own tumor [65–67]. Central to these strategies is the activation and expansion
of tumor-specific T cells in vitro before adoptive transfer. Identifying ways to enhance this process
is an area of active investigation, with one approach being to polarize the CD8+ T cells into a Tc
subset with robust antitumor properties. Before the identification of the more novel subsets,
Tc1 cells were the ‘one to beat’ in this regard because they continuously outperformed Tc2
cells in multiple mouse adoptive transfer tumor models [14,68,69]. This is unexpected, given
that in vitro both Tc1 and Tc2 cells have similar degrees of cytotoxicity, which has a substantial
role in their in vivo antitumor effects [70]. It is likely that there are other factors responsible for
the diminished performance of Tc2, such as their production of IL-4. Although IL-4 can act as
a growth factor to enhance T cell proliferation [71], IL-4 has been shown to have protumorigenic
effects, which may serve to dampen a Tc2-mediated response. For instance, IL-4 has a well-

6 Trends in Cell Biology, Month 2020, Vol. xx, No. xx


Trends in Cell Biology

established role in facilitating the commitment of macrophages towards the M2 phenotype, char-
acterized, in part, by their secretion of anti-inflammatory molecules that inhibit T cell function [72].

IFN-γ is largely dispensable for the antitumor effects of Tc2 cells [73]. This is in contrast to Tc1 and
other Tc subsets, because their antitumor functions are dependent on their ability to produce IFN-γ
upon entering the TME. For instance, a study by Garcia-Hernandez and colleagues demonstrated
that Tc17 cells transferred into B16 tumor-bearing mice could inhibit tumor growth, albeit much
less than that observed with Tc1 cells. This Tc17-dependent immunity was shown to be depen-
dent, in part, on their ability to produce IFN-γ [74]. This suggests that Tc17 cells demonstrate
some degree of plasticity (Box 2). Additionally, the antitumor effects of Tc17 are context dependent
and treatment success is determined by the model used. For instance, minimal antitumor activity
was observed following the adoptive transfer of Tc17 cells into mice bearing established tumors
[7,74,75]. However, when Tc17 cells were administered in conjunction with lymphodepletion,
tumor antigen vaccination, and/or supportive in vivo IL-2 treatments, robust antitumor effects
were noted [24,76]. This is likely due to the intrinsic stem-like properties attributed to Tc17 cells be-
cause they express high levels of the transcription factor TCF7, which is associated with increased
longevity and self-renewal capabilities [77]. As a result, it is likely that Tc17 cells persist and undergo
a robust proliferative burst upon exposure to the in vivo supportive treatments, thereby facilitating
an antitumor response. This also might occur for Tc9 cells, based on recent findings that polarized
Tc9 cells alone demonstrate poor antitumor functions in treating well-established B16 tumors [7].
However, when Tc9s were administered in conjunction with supportive in vivo treatments (IL-2 in-
jections, tumor antigen vaccination +/– lymphodepletion), Tc9s displayed exceptional results in
treating well-established melanoma tumors [19,78]. As such, it is tempting to speculate that Tc9
cells function similar to Tc17 cells in that they can persist and undergo robust proliferative burst
upon encountering these supportive treatments, but lack satisfactory antitumor functions on
their own.

Another aspect that can influence the antitumor response of Tc subsets is their metabolic state.
Indeed, recent reports have linked increased T cell bioenergetics and mitochondrial metabolism
with improved antitumor responses [79]. Along these lines, it was shown that Tc22 cells demon-
strate robust antitumor properties, performing at least as well as, if not better than, Tc1 cells in a
B16 melanoma tumor model [7]. Mechanistically, an increase was observed in mitochondrial
proteins and ATP in Tc22 cells relative to either Tc1 or Tc17 cells, suggesting potential metabolic
differences between each subset. In addition to Tc22 cells, it has also been reported that choles-
terol metabolism in Tc9 cells also influences their antitumor activity [78]. Taken together, it is clear
that, in the context of immune therapy, each subset has different functional and metabolic
properties associated with their lineage, which should be taken into consideration when designing
future immunotherapeutic approaches.

Box 2. Plasticity within the Tc Subsets


Plasticity within the CD8+ T cell subsets has been observed in multiple instances. Following their adoptive transfer into
tumor-bearing mice, in vitro polarized Tc17 cells [24] and Tc9 cells [19] convert into a Tc1 phenotype. Moreover, a study
by Flores-Santibáñez and colleagues found that adoptively transferred Tc17 cells retained their Tc17 phenotype in the
TDLN, although a degree of plasticity towards the Tc1 phenotype was observed in Tc17 cells that had infiltrated the tumor
[77]. However, stable commitment towards the Tc17 lineage has been observed in other models, such as in the context of
vaccination [42]. There is also evidence to suggest that tumor infiltrating Tc22 cells may retain their Tc22 phenotype and IL-
22 production despite a prolonged ex vivo expansion in IL-2 [7]. However, it is also possible that some degree of plasticity
exists between Tc17 and Tc22 cells, given that both these subsets are polarized with IL-6 and that Tc17 cells also produce
IL-22. In summary, these studies suggest that Tc lineages have the potential to remain stable but that in vivo signals
facilitate the conversion towards another Tc lineage, such as Tc1. Future studies should aim to identify these factors to
better understand the mechanisms in maintaining commitment to a Tc lineage.

Trends in Cell Biology, Month 2020, Vol. xx, No. xx 7


Trends in Cell Biology

Concluding Remarks Outstanding Questions


Overall, it is clear that there is more to CD8+ T cells than their cytolytic capacity and ability to pro- What are the specific roles of these
duce IFN-γ, and that referring to CD8+ T cells as simply cytotoxic T cells is a misnomer. Indeed, subsets in the TME and what are their
implications for prognosis?
multiple CD8+ Tc subsets have been identified, each with distinct effector functions, including
noncytolytic subsets. These subsets have been detected in the TME and, in many instances, What surface markers can be used to
their biological significance is largely unknown. It is clear that, even for the same cancer type, precisely identify each Tc subset?
there are large discrepancies between different patients as to the relative frequencies of each
Will the presence of certain subsets
Tc subset within the TME. It is likely that this composition of Tc subsets will influence not only influence the response to immunological
prognosis and survival, but also response rates to novel therapies targeting the immune system. checkpoint blockade and/or serve as a
Further research will be needed to understand the exact function of these subsets in the antitu- biomarker for response?
mor response as well as the factors within the TME that can influence their differentiation (see
Will polarizing transferred CD8+ T cells
Outstanding Questions). Novel treatment strategies can then be developed to selectively target to different subsets enhance adoptive
specific Tc subsets to enhance cancer treatments and prognosis. immunotherapy clinically?

Acknowledgments
P.S.O. holds a Canada Research Chair in Autoimmunity and Tumor Immunity. Figures created with BioRender.

References
1. Zhu, J. et al. (2010) Differentiation of effector CD4 T cell populations. 18. Visekruna, A. et al. (2012) Tc9 cells, a new subset of CD8(+) T cells,
Annu. Rev. Immunol. 28, 445–489 support Th2-mediated airway inflammation. Eur. J. Immunol. 43,
2. Yu, Y. et al. (2018) Recent advances in CD8+ regulatory T cell re- 606–618
search. Oncol. Lett. 15, 8187–8194 19. Lu, Y. et al. (2014) Tumor-specific IL-9-producing CD8+ Tc9 cells
3. Yu, D. and Ye, L. (2018) A portrait of CXCR5+ follicular cytotoxic are superior effector than type-I cytotoxic Tc1 cells for adoptive
CD8+ T cells. Trends Immunol. 39, 965–979 immunotherapy of cancers. Proc. Natl. Acad. Sci. U. S. A. 111,
4. Croft, M. et al. (1994) Generation of polarized antigen-specific 2265–2270
CD8 effector populations: reciprocal action of interleukin (IL)-4 20. Zhao, P. et al. (2013) IL-9 and Th9 cells: progress and challenges.
and IL-12 in promoting type 2 versus type 1 cytokine profiles. Int. Immunol. 25, 547–551
J. Exp. Med. 180, 1715–1728 21. Chang, S.Y. et al. (2013) Circulatory antigen processing by
5. Mittrücker, H.-W. et al. (2014) Heterogeneity in the differentiation mucosal dendritic cells controls CD8+ T cell activation. Immunity
and function of CD8+ T cells. Arch. Immunol. Ther. Exp. (Warsz) 38, 153–165
62, 449–458 22. Wang, W. et al. (2016) Increased circulating IL-9-producing
6. Chan, W.L. et al. (2001) Human IL-18 receptor and ST2L are CD8+T cells are associated with eosinophilia and high FeNO
stable and selective markers for the respective Type 1 and in allergic asthmatics. Exp. Ther. Med. 12, 4055–4060
Type 2 circulating lymphocytes. J. Immunol. 167, 1238–1244 23. Huber, M. et al. (2009) A Th17-like developmental process leads
7. St. Paul, M. et al. (2020) IL6 Induces an IL22 + CD8+ T-cell subset to CD8(+) Tc17 cells with reduced cytotoxic activity. Eur.
with potent antitumor function. Cancer Immunol. Res. 8, 321–333 J. Immunol. 39, 1716–1725
8. Yang, Y. et al. (2007) Identification of a distant T-bet enhancer 24. Hinrichs, C.S. et al. (2009) Type 17 CD8+ T cells display
responsive to IL-12/Stat4 and IFNgamma/Stat1 signals. Blood enhanced antitumor immunity. Blood 114, 596–599
110, 2494–2500 25. Liu, S.-J. et al. (2007) Induction of a distinct CD8 Tnc17 subset
9. Pearce, E.L. et al. (2003) Control of effector CD8+ T cell function by transforming growth factor-β and interleukin-6. J. Leukoc.
by the transcription factor Eomesodermin. Science 302, Biol. 82, 354–360
1041–1043 26. Yen, H.-R. et al. (2009) Tc17 CD8 T cells: functional plasticity
10. Intlekofer, A.M. et al. (2008) Anomalous type 17 response to viral and subset diversity. J. Immunol. 183, 7161–7168
infection by CD8+ T cells lacking T-bet and eomesodermin. Science 27. El-Behi, M. et al. (2014) Committed Tc17 cells are phenotypically
(80-. ) 321, 408–411 and functionally resistant to the effects of IL-27. Eur. J. Immunol.
11. Mingari, M.C. et al. (1984) Frequent coexpression of cytolytic 44, 3003–3014
activity and lymphokine production among human T lymphocytes. 28. Singh, S.P. et al. (2008) Human T cells that are able to produce
Production of B cell growth factor and interleukin 2 by T8+ and T4+ IL-17 express the chemokine receptor CCR6. J. Immunol. 180,
cytolytic clones. Eur. J. Immunol. 14, 1066–1069 214–221
12. Salgame, P. et al. (1991) Differing lymphokine profiles of func- 29. Linehan, J.L. et al. (2018) Non-classical immunity controls microbi-
tional subsets of human CD4 and CD8 T cell clones. Science ota impact on skin immunity and tissue repair. Cell 172, 784–796
(80-. ) 254, 279–282 30. Naik, S. et al. (2015) Commensal-dendritic-cell interaction
13. Seder, R.A. et al. (1992) CD8+ T cells can be primed in vitro to specifies a unique protective skin immune signature. Nature
produce IL-4. J. Immunol. 148, 1652–1656 520, 104–108
14. Kemp, R.A. and Ronchese, F. (2001) Tumor-specific Tc1, 31. Kryczek, I. et al. (2007) Cutting edge: Th17 and regulatory T cell
but not Tc2, cells deliver protective antitumor immunity. dynamics and the regulation by IL-2 in the tumor microenvironment.
J. Immunol. 167, 6497–6502 J. Immunol. 178, 6730–6733
15. Pai, S.-Y. et al. (2004) GATA-3 deficiency abrogates the devel- 32. Nanjappa, S.G. et al. (2012) Tc17 cells mediate vaccine immu-
opment and maintenance of T helper type 2 cells. Proc. Natl. nity against lethal fungal pneumonia in immune deficient hosts
Acad. Sci. U. S. A. 101, 1993–1998 lacking CD4+ T cells. PLoS Pathog. 8, e1002771
16. Schaller, M.A. et al. (2005) CD8+ T cell contributions to allergen 33. Huber, M. et al. (2013) IL-17A secretion by CD8+ T cells supports
induced pulmonary inflammation and airway hyperreactivity. Th17-mediated autoimmune encephalomyelitis. J. Clin. Invest.
Eur. J. Immunol. 35, 2061–2070 123, 247–260
17. Hilvering, B. et al. (2018) Synergistic activation of pro- 34. Furlan, S.N. et al. (2016) Systems analysis uncovers inflamma-
inflammatory type-2 CD8+ T lymphocytes by lipid mediators in tory Th/Tc17-driven modules during acute GVHD in monkey
severe eosinophilic asthma. Mucosal Immunol. 11, 1408–1419 and human T cells. Blood 128, 2568–2579

8 Trends in Cell Biology, Month 2020, Vol. xx, No. xx


Trends in Cell Biology

35. Gartlan, K.H. et al. (2015) Tc17 cells are a proinflammatory, plas- 60. Cabrita, R. et al. (2020) Tertiary lymphoid structures improve im-
tic lineage of pathogenic CD8+ T cells that induce GVHD without munotherapy and survival in melanoma. Nature 577, 561–565
antileukemic effects. Blood 126, 1609–1621 61. Fujita, H. et al. (2012) Langerhans cells from human cutaneous
36. Res, P.C.M. et al. (2010) Overrepresentation of IL-17A and IL-22 squamous cell carcinoma induce strong type 1 immunity.
producing CD8 T cells in lesional skin suggests their involvement J. Invest. Dermatol. 132, 1645–1655
in the pathogenesis of psoriasis. PLoS One 5, e14108 62. McDonnell, A.M. et al. (2015) Tumor-infiltrating dendritic cells
37. Nograles, K.E. et al. (2009) IL-22-producing “T22” T cells exhibit defective cross-presentation of tumor antigens, but is
account for upregulated IL-22 in atopic dermatitis despite reversed by chemotherapy. Eur. J. Immunol. 45, 49–59
reduced IL-17-producing TH17 T cells. J. Allergy Clin. Immunol. 63. Chang, S.Y. et al. (2013) Intestinal dendritic cells survey circulatory
123, 1244–1252 antigens prior to induction of CD8(+) T cells. Immunity 38, 1–20
38. Oliveira, L.M.S. et al. (2015) Increased frequency of circulating 64. Fujita, H. et al. (2009) Human Langerhans cells induce distinct IL-
Tc22/Th22 cells and polyfunctional CD38− T cells in HIV- 22-producing CD4+ T cells lacking IL-17 production. Proc. Natl.
exposed uninfected subjects. Sci. Rep. 5, 13883 Acad. Sci. U. S. A. 106, 21795–21800
39. Zhang, S. et al. (2013) Increased Tc22 and Treg/CD8 ratio con- 65. Lim, W.A. and June, C.H. (2017) The principles of engineering
tribute to aggressive growth of transplant associated squamous immune cells to treat cancer. Cell 168, 724–740
cell carcinoma. PLoS One 8, e62154 66. Rosenberg, S.A. et al. (2008) Adoptive cell transfer: a clinical path
40. Samji, T. and Khanna, K.M. (2017) Understanding memory to effective cancer immunotherapy. Nat. Rev. Cancer 8, 299–308
CD8+ T cells. Immunol. Lett. 185, 32–39 67. Tran, E. et al. (2017) “Final common pathway” of human cancer
41. Flores-Santibáñez, F. et al. (2018) In vitro-generated Tc17 cells immunotherapy: targeting random somatic mutations. Nat.
present a memory phenotype and serve as a reservoir of Tc1 Immunol. 18, 255–262
cells in vivo. Front. Immunol. 9, 1–12 68. Nishimura, F. et al. (2006) Adoptive transfer of type 1 CTL mediates
42. Nanjappa, S.G. et al. (2017) Antifungal Tc17 cells are durable effective anti-central nervous system tumor response: critical roles
and stable, persisting as long-lasting vaccine memory without of IFN-inducible protein-10. Cancer Res. 66, 4478–8447
plasticity towards IFNγ cells. PLoS Pathog. 13, 1–25 69. Dobrzanski, M.J. et al. (2006) CD8-mediated type 1 antitumor
43. Fridman, W.H. et al. (2012) The immune contexture in human responses selectively modulate endogenous differentiated and
tumours: impact on clinical outcome. Nat. Rev. Cancer 12, nondifferentiated T cell localization, activation, and function in
298–306 progressive breast cancer. J. Immunol. 177, 8191–8201
44. Ayers, M. et al. (2017) IFN-γ-related mRNA profile predicts clini- 70. Dobrzanski, M.J. et al. (2004) Effector cell-derived lymphotoxin
cal response to PD-1 blockade. J. Clin. Invest. 127, 2930–2940 alpha and Fas ligand, but not perforin, promote Tc1 and Tc2
45. Zaidi, M.R. and Merlino, G. (2011) The two faces of interferon-y effector cell-mediated tumor therapy in established pulmonary
in cancer. Clin. Cancer Res. 17, 6118–6124 metastases. Cancer Res. 64, 406–414
46. Overacre-Delgoffe, A.E. et al. (2017) Interferon-γ drives Treg 71. Morris, S.C. et al. (2009) Endogenously produced IL-4
fragility to promote anti-tumor immunity. Cell 169, 1130–1141 nonredundantly stimulates CD8+ T cell proliferation. J. Immunol.
47. Shin, D.S. et al. (2017) Primary resistance to PD-1 blockade 182, 1429–1438
mediated by JAK1/2 mutations. Cancer Discov. 7, 188–201 72. Huber, S. et al. (2010) Alternatively activated macrophages inhibit
48. Sheu, B-C.C. et al. (2001) Predominant Th2/Tc2 polarity of T-cell proliferation by Stat6-dependent expression of PD-L2.
tumor-infiltrating lymphocytes in human cervical cancer. Blood 116, 3311–3320
J. Immunol. 167, 2972–2978 73. Dobrzanski, M.J. et al. (2000) Type 1 and Type 2 CD8+ effector T
49. Ehi, K. et al. (2008) Analysis of T-helper type 1 and 2 cells and cell subpopulations promote long-term tumor immunity and pro-
T-cytotoxic type 1 and 2 cells of sentinel lymph nodes in breast tection to progressively growing tumor. J. Immunol. 164,
cancer. Oncol. Rep. 19, 601–607 916–925
50. Faghih, Z. et al. (2013) IL-17 and IL-4 producing CD8+ T cells in 74. Garcia-Hernandez, M.D.L.L. et al. (2010) Adoptive transfer of
tumor draining lymph nodes of breast cancer patients: positive tumor-specific Tc17 effector T cells controls the growth of B16
association with tumor progression. Iran. J. Immunol. 10, melanoma in mice. J. Immunol. 184, 4215–4227
193–204 75. Tajima, M. et al. (2011) IL-17/IFN-γ double producing CD8+ T
51. Kuang, D.-M. et al. (2010) Tumor-activated monocytes promote (Tc17/IFN-γ) cells: a novel cytotoxic T-cell subset converted
expansion of IL-17-producing CD8+ T cells in hepatocellular car- from Tc17 cells by IL-12. Int. Immunol. 23, 751–759
cinoma patients. J. Immunol. 185, 1544–1549 76. Bowers, J.S. et al. (2015) Dendritic cells in irradiated mice trigger
52. Zhuang, Y. et al. (2012) CD8+ T cells that produce interleukin-17 the functional plasticity and antitumor activity of adoptively trans-
regulate myeloid-derived suppressor cells and are associated ferred Tc17 cells via IL12 signaling. Clin. Cancer Res. 21,
with survival time of patients with gastric cancer. Gastroenterology 2546–2557
143, 951–962 77. Flores-Santibáñez, F. et al. (2015) CD73-mediated adenosine
53. Lee, M.-H. et al. (2018) Interleukin 17 and peripheral IL-17- production promotes stem cell-like properties in mouse Tc17
expressing T cells are negatively correlated with the overall survival cells. Immunology 146, 582–594
of head and neck cancer patients. Oncotarget 9, 9825–9837 78. Ma, X. et al. (2018) Cholesterol negatively regulates IL-9-producing
54. Benevides, L. et al. (2015) IL17 promotes mammary tumor progres- CD8+ T cell differentiation and antitumor activity. J. Exp. Med. 215,
sion by changing the behavior of tumor cells and eliciting tumorigenic 1555–1569
neutrophils recruitment. Cancer Res. 75, 3788–3799 79. Scharping, N.E. et al. (2016) The tumor microenvironment represses
55. He, D. et al. (2010) IL-17 promotes tumor development through T cell mitochondrial biogenesis to drive intratumoral T cell metabolic
the induction of tumor promoting microenvironments at tumor insufficiency and dysfunction. Immunity 45, 374–388
sites and myeloid-derived suppressor cells. J. Immunol. 184, 80. Karyampudi, L. et al. (2014) Accumulation of memory precursor
2281–2288 cd8 t cells in regressing tumors following combination therapy
56. Wang, S. et al. (2017) Prognostic role of intratumoral IL-17A with vaccine and anti-pd-1 antibody. Cancer Res. 74, 2974–2985
expression by immunohistochemistry in solid tumors: a 81. Reome, J.B. et al. (2004) Type 1 and type 2 tumor infiltrating
meta-analysis. Oncotarget 8, 66382–66391 effector cell subpopulations in progressive breast cancer. Clin.
57. Wang, X.-F. et al. (2017) The prognostic value of interleukin-17 in Immunol. 111, 69–81
lung cancer: a systematic review with meta-analysis based on 82. Ito, N. et al. (1999) Lung carcinoma: analysis of T helper type 1
Chinese patients. PLoS One 12, e0185168 and 2 cells and T cytotoxic type 1 and 2 cells by intracellular
58. Goc, J. et al. (2014) Dendritic cells in tumor-associated tertiary cytokine detection with flow cytometry. Cancer 85, 2359–2367
lymphoid structures signal a Th1 cytotoxic immune contex- 83. Ito, N. et al. (2005) Prognostic significance of T helper 1 and 2
ture and license the positive prognostic value of infiltrating and T cytotoxic 1 and 2 cells in patients with non-small cell
CD8+ T cells. Cancer Res. 74, 705–715 lung cancer. Anticancer Res. 25, 2027–2031
59. Thompson, E.D. et al. (2010) Tumor masses support naive T cell 84. Podhorecka, M. et al. (2002) T type 1 / type 2 subsets balance in
infiltration, activation, and differentiation into effectors. J. Exp. B-cell chronic lymphocytic leukemia — the three-color flow
Med. 207, 1791–1804 cytometry analysis. Leuk. Res. 26, 657–660

Trends in Cell Biology, Month 2020, Vol. xx, No. xx 9


Trends in Cell Biology

85. Anichini, A. et al. (2019) Skewed T-cell differentiation in patients 88. Li, J. et al. (2011) Distribution, characterization, and induction of
with indolent non-Hodgkin lymphoma reversed by ex vivo T-cell CD8+regulatory T cells and IL-17-producing CD8+T cells in
culture with gammac cytokines. Blood 107, 602–610 nasopharyngeal carcinoma. J. Transl. Med. 9, 189
86. Kryczek, I. et al. (2009) Phenotype, distribution, generation, and 89. Chellappa, S. et al. (2017) CD8 + T cells that coexpress RORγt
functional and clinical relevance of Th17 cells in the human tumor and T-bet are functionally impaired and expand in patients with
environments. Blood 114, 1141–1149 distal bile duct cancer. J. Immunol. 198, 1729–1739
87. Saito, H. et al. (2015) Clinical relevance of the number of 90. Sudam Patil, R. et al. (2016) IL17 producing γδT cells induce
interleukin-17-producing CD 8+ T cells in patients with gastric angiogenesis and are associated with poor survival in gallbladder
cancer. Surg. Today 45, 1429–1435 cancer patients. Int. J. Cancer 139, 869–881

10 Trends in Cell Biology, Month 2020, Vol. xx, No. xx

You might also like