ArticlePDF Available

Activated and Exhausted MAIT Cells Foster Disease Progression and Indicate Poor Outcome in Hepatocellular Carcinoma

Authors:

Abstract and Figures

Purpose: Innate immunity is an indispensable arm of tumor immune surveillance, and the liver is an organ with a predominance of innate immunity, where mucosal-associated invariant T (MAIT) cells are enriched. However, little is known about the phenotype, functions, and immunomodulatory role of MAIT cells in hepatocellular carcinoma (HCC). Experimental Design: The distribution, phenotype, and function of MAIT cells in patients with HCC were evaluated by both flow cytometry (FCM) and in vitro bioassays. Transcriptomic analysis of MAIT cells was also performed. Prognostic significance of tumor-infiltrating MAIT cells was validated in four independent cohorts of patients with HCC. Results: Despite their fewer densities in HCC tumor than normal liver, MAIT cells were significantly enriched in the HCC microenvironment compared with other mucosa-associated organs. Tumor-derived MAIT cells displayed a typical CCR7⁻CD45RA⁻CD45RO⁺CD95⁺ effector memory phenotype with lower costimulatory and effector capabilities. Tumor-educated MAIT cells significantly upregulated inhibitory molecules like PD-1, CTLA-4, TIM-3, secreted significantly less IFNγ and IL17, and produced minimal granzyme B and perforin while shifting to produce tumor-promoting cytokines like IL8. Transcriptome sequencing confirmed that tumor-derived MAIT cells were reprogrammed toward a tumor-promoting direction by downregulating genes enriched in pathways of cytokine secretion and cytolysis effector function like NFKB1 and STAT5B and by upregulating genes like IL8, CXCL12, and HAVCR2 (TIM-3). High infiltration of MAIT cells in HCC significantly correlated with an unfavorable clinical outcome, revealed by FCM, qRT-PCR, and multiplex IHC analyses, respectively. Conclusions: HCC-infiltrating MAIT cells were functionally impaired and even reprogrammed to shift away from antitumor immunity and toward a tumor-promoting direction.
Content may be subject to copyright.
Translational Cancer Mechanisms and Therapy
Activated and Exhausted MAIT Cells Foster
Disease Progression and Indicate Poor Outcome in
Hepatocellular Carcinoma
Meng Duan
1
, Shyamal Goswami
2
, Jie-Yi Shi
1
, Lin-Jie Wu
3
, Xiao-Ying Wang
1
, Jia-Qiang Ma
2
,
Zhao Zhang
1
, Yang Shi
4
, Li-Jie Ma
1
, Shu Zhang
1
, Rui-Bin Xi
3,5
,Ya Cao
6
, Jian Zhou
1,7,8
,
Jia Fan
1,7,8
, Xiao-Ming Zhang
2
, and Qiang Gao
1,8
Abstract
Purpose: Innate immunity is an indispensable arm of
tumor immune surveillance, and the liver is an organ with
a predominance of innate immunity, where mucosal-associ-
ated invariant T (MAIT) cells are enriched. However, little is
known about the phenotype, functions, and immunomodu-
latory role of MAIT cells in hepatocellular carcinoma (HCC).
Experimental Design: The distribution, phenotype, and
function of MAIT cells in patients with HCC were evaluated
by both ow cytometry (FCM) and in vitro bioassays. Tran-
scriptomic analysis of MAIT cells was also performed. Prog-
nostic signicance of tumor-inltrating MAIT cells was vali-
dated in four independent cohorts of patients with HCC.
Results: Despite their fewer densities in HCC tumor
than normal liver, MAIT cells were signicantly enriched
in the HCC microenvironment compared with other mucosa-
associated organs. Tumor-derived MAIT cells displayed a
typical CCR7
CD45RA
CD45RO
þ
CD95
þ
effector memory
phenotype with lower costimulatory and effector capabilities.
Tumor-educated MAIT cells signicantly upregulated inhibitory
molecules like PD-1, CTLA-4, TIM-3, secreted signicantly less
IFNgand IL17, andproduced minimalgranzyme B and perforin
while shifting to produce tumor-promoting cytokines like IL8.
Transcriptome sequencing conrmed that tumor-derived MAIT
cells were reprogrammed toward a tumor-promoting direction
by downregulating genes enriched in pathways of cytokine
secretionand cytolysis effector function like NFKB1and STAT5B
and by upregulating genes like IL8, CXCL12,andHAVCR2
(TIM-3). High inltration of MAIT cells in HCC signicantly
correlated with an unfavorable clinical outcome, revealed by
FCM, qRT-PCR, and multiplex IHC analyses, respectively.
Conclusions: HCC-inltrating MAIT cells were functionally
impaired and even reprogrammed to shift away from antitu-
mor immunity and toward a tumor-promoting direction.
See related commentary by Carbone, p. 3199
Introduction
Hepatocellular carcinoma (HCC) is the fth most common
cancer in men and the seventh among women in the world (1).
HCC represents a typicalinammation/immune-related tumor that
usually develops in an inamed brotic or cirrhotic liver (2). The
recent breakthrough in HCC immunotherapy targeting immune
checkpoint PD-1/PD-L1 has substantially improved the patient
survival (3, 4). A deeper understanding of interactions between
immune cells and cancer cells within the HCC microenvironment
may reveal diverse therapeutic approaches for HCC (5, 6). In
addition to adaptive immune system specialized in recognizing
tumor antigen, innate immunity is another indispensable arm of
immune system, directly or indirectly participating in tumor con-
trol (7, 8). Indeed, the liver is an organ with a predominance of
diverse range of innate lymphocytes, among which mucosal-
associated invariant T (MAIT) cells are of paramount importance.
MAIT cells are a specialized innate-like T-cell subset, accounting
for 10% of circulating CD4
T cells in adults, featured with the
expression of the semiinvariant T-cell antigen receptor (TCR,
va7.2-Ja33) that detects microbial vitamin B metabolites presented
by major histocompatibility complex class Irelated protein 1
(MR1; refs. 9, 10). Both the TCR expressed by MAIT cells and the
antigen-presenting molecule MR1 are evolutionarily conserved
among mammals, indicating a strong selective pressure to maintain
1
Department of Liver Surgery and Transplantation, Liver Cancer Institute,
Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer
Invasion (Ministry of Education), Fudan University, Shanghai, China.
2
Key
Laboratory of Molecular Virology & Immunology, Institut Pasteur of
Shanghai, Chinese Academy of Sciences, Shanghai, China.
3
School of
Mathematical Sciences, Peking University, Beijing, China.
4
Peking-Tsinghua
Center for Life Sciences, Academy for Advanced Interdisciplinary Studies,
Peking University, Beijing, China.
5
Center for Statistical Sciences, Peking
University, Beijing, China.
6
Cancer Research Institute, Xiangya School of
Medicine, Central South University, Hunan, China.
7
Institute of Biomedical
Sciences, Fudan University, Shanghai, China.
8
State Key Laboratory of
Genetic Engineering, Fudan University, Shanghai, China.
Note: Supplementary data for this article are available at Clinical Cancer
Research Online (http://clincancerres.aacrjournals.org/).
M. Duan, S. Goswami, J.-Y. Shi contributed equally to this article.
Corresponding Authors: Qiang Gao, Liver Cancer Institute, Zhong Shan
Hospital and Shanghai Medical School, Fudan University, 180 Fenglin
Road, Shanghai 200032, China. Phone: 8621-6403-7181; Fax: 8621-6403-
7181; E-mail: gao.qiang@zs-hospital.sh.cn; Jia Fan, E-mail: fan.jia@zs-
hospital.sh.cn; and Xiao-Ming Zhang, Key Laboratory of Molecular Virology
& Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences.
E-mail: xmzhang@ips.ac.cn
Clin Cancer Res 2019;25:330416
doi: 10.1158/1078-0432.CCR-18-3040
2019 American Association for Cancer Research.
Clinical
Cancer
Research
Clin Cancer Res; 25(11) June 1, 2019
3304
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
immunity mediated by MAIT cells (11). Several subsets of
MAIT cells have been dened, most of which, if not all, are
CD161
high
IL17-secreting CD8
þ
T-cell subset (9, 12). After being acti-
vated by microbial antigen bound with MR1, MAIT cells are licensed
to kill targets by secreting IFNg, granzyme, and perforin (13).
Intriguingly, contrary to the name, MAIT cells are most enriched
in normal human livers, comprising up to 50% of all intrahepatic
T cells (14). It has been reported that hepatic MAIT cells are highly
activated within the liver and may be protective against a range of
bacteria, fungi, and viruses along with the large phagocytic Kupf-
fer cell population (8). Overall, a reduction and dysfunction in
MAIT cells were observed in blood and livers from patients with
chronic hepatitis B virus (HBV) or hepatitis B virus (HCV) infec-
tions and nonvirus chronic liver diseases (11, 1517). As HCC is
usually complicated with those chronic liver diseases, dysfunction
of MAIT cells in HCC could be expected.
It seems plausible that MAIT cells may have a protective role in
the immune system, considering the highly conserved TCR and
the cytokine secretion pattern. Previous studies investigated MAIT
cells mainly in the infectious or autoimmune diseases, showing
that the inltration and function of the subset were substantially
impaired (11). The only data regarding the role of MAIT cells in
human cancer comes from the colorectal cancer studies, indicat-
ing that activated MAIT cells accumulated in tumors correlated
with inferior patient survival (1820). Although liver served as an
immunologic organ where MAIT cells are mostly enriched, the
function and immunomodulatory role of the MAIT cells in HCC
have yet not been fully claried. In this study, we aimed to
determine the distribution, phenotype, function, and clinical
relevance of MAIT cells in human HCC.
Materials and Methods
Patients and samples
For ow cytometry (FCM) analysis, fresh paired tumor/peritu-
mor tissues and peripheral blood samples were obtained from a
cohort of 50 patients with HCC who underwent surgical resection
between 2014 and 2015. For analyzing TCRva7.2-Ja33 mRNA
expression, fresh frozen tumor/peritumor tissues were obtained
from another cohort of 207 patients with HCC who received
curative operation between 2009 and 2013. For tissue microarrays
(TMA), archival tissues were obtained from two independent
cohorts of 224 and 360 patients with HCC who received curative
surgery in 2007 and in 2006, respectively. Patient information
and clinicopathologic features of all the cohorts are summarized
in Supplementary Table S1. Informed consent was obtained from
each patient prior to receive the sample. This study was conducted
in accordance with ethical principles that have their origin in the
Declaration of Helsinki, and the ethical standards of the Research
Ethics Committee of Zhongshan Hospital (Shanghai, China).
Mononuclear cell isolation and FCM
Mononuclear cells from freshly resected liver tissues and peri-
pheral blood were isolated and stained for FCM as described
previously (ref. 21; details in Supplementary Materials and
Methods and Supplementary Table S2).
Immunouorescence
Immunouorescence on frozen section was performed accord-
ing to a two-step way method and scanned by TCS SP5 (Leica
Biosystems). Multiplex IHC on parafn-embedded TMAs was
performed using Opal Tyramide Signal Amplication (TSA)
based staining regents (PerkinElmer). Detailed information is
provided in Supplementary Materials and Methods.
MAIT cell coculture and cytokine detection
Information of MAIT cell activation, coculture, and cytokine
detection are described in Supplementary Materials and Methods.
RNA isolation, RT-PCR, and RNA sequencing
Total RNA was extracted using TRIzol Reagent (Invitrogen)
according to the manufacturer's instructions. The TCRva7.2-Ja33
mRNA levels were determined by real-time RT-PCR as described
previously (22). RNA sequencing (RNA-seq) was carried out as
described by Picelli and colleagues with minor modica-
tions (23). Detailed information is described in Supplementary
Materials and Methods.
Statistical analysis
Statistical analyses were performed using GraphPad Prism 7.03
(GraphPad Software). The experimental data were shown as mean
SD. Cutoff values for patient grouping in all the cohorts were
dened by lowest tertile (33rd percentile) of MAIT cell's frequen-
cy/density. A two-tailed P<0.05 was considered signicant.
Detailed information is provided in Supplementary Materials
and Methods.
Results
MAIT cell inltration is signicantly decreased in HCC
We rst used FCM analysis to determine MAIT cell distribution
in blood and tissue of patients with HCC and healthy donors.
Consistent with the previous report (9), we conrmed that CD4
cells constituted >95% of total MAIT cells, irrespective of speci-
mens from normal liver, HCC tissues, or peripheral blood (Fig. 1A
and B; Supplementary Fig. S1A).
Even in peripheral blood, the frequency of CD4
MAIT cells
among total CD3
þ
T cells was signicantly decreased in patients
Translational Relevance
Hepatocellular carcinoma (HCC) represents a typical
inammation and immune-related cancer. The mucosal-
associat ed invariant T (MAIT) cell, an innate immune cell subset,
is enriched in the liver where innate immunity is dominant.
Here, we demonstrate that although tumor-inltrating MAIT
cells display a typical effector memory phenotype, their effector
functions and cytotoxic capability are signicantly impaired.
Upregulation of PD-1, CTLA-4, and TIM-3, a common feature
of T-cell exhaustion, is evidenced in HCC-derived MAIT cells.
MAIT cells are found to be activated within tumor milieu and
reprogrammed to produce a signicantamountofprotumor
cytokines. A high density of tumor-inltrating MAIT cells signi-
cantly and independently correlated with dismal clinical
outcomes in patients with HCC. Thus, strategies to modulate
the functional activities of MAIT cells may provide a new avenue
for antitumor therapy in HCC. Extended understanding of
interactions between innate immunity and the HCC microen-
vironment may further provide new clues for more effective
immune therapy.
MAIT Cells in HCC
www.aacrjournals.org Clin Cancer Res; 25(11) June 1, 2019 3305
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
with HCC compared with healthy donors (mean: 0.9% vs. 2.1%,
P<0.01; Fig. 1C). Trend continues in tumor tissues, CD4
MAIT
cell frequency within total CD3
þ
T-cell population was signi-
cantly decreased in HCC tumor compared with either paired
peritumor (mean: 4.2% vs. 16.5%, P<0.001; n¼50) or normal
liver (mean: 4.2% vs. 21.6%, P<0.001; n¼20; Fig. 1C).
Next, immunouorescence imaging conrmed our FCM nd-
ings (Fig. 1D), reecting a decreased density of CD161
þ
TCRva7.2
þ
MAIT cells in the tumor center compared with peri-
tumor (mean: 7.2 vs. 17.4 cells/mm
2
,P¼0.014; n¼8; Fig. 1E).
Moreover, using TCRva7.2 and IL18R as the surrogate for MAIT
cell detection, the results also showed a reduction of
TCRva7.2
þ
IL18R
þ
MAIT cells in tumor as compared with peri-
tumor (Supplementary Fig. S1B). These results indicated that
either absolute or relative number of MAIT cells was signicantly
lower in the blood and tumor samples of patients with HCC.
We further checked the relationship between MAIT cell inl-
tration and clinical features and identied MAIT cell frequency
was lower in patients with liver cirrhosis in FCM cohort (mean:
3.46% vs. 6.90%, P¼0.053; n¼50; Supplementary Table S1). In
addition, in a larger qRT-PCR cohort (n¼209), TCRva7.2-Ja33
mRNA relative expression level was also decreased in patients with
liver cirrhosis (mean: 0.014 vs. 0.069, P¼0.057), or chronic HBV
infection (mean: 0.061 vs. 0.125, P¼0.075; Supplementary
Table S1). The above results suggest that liver cirrhosis is a
potential factor associated with the decrease of MAIT cell
inltration.
Tumor-inltrating MAIT cells display a typical effector memory
phenotype
In patients with HCC and healthy donors, we determined the
phenotypic features of MAIT cells, gated on MR1-5-OP-RU
þ
after
CD3
þ
CD4
CD161
þ
TCRva7.2
þ
, to exclude contaminations
from CD161
int
Va7.2
þ
T CD4
þ
mainstream T cells (24), by
comparing with conventional T-cellrelated costimulatory/mem-
ory/activation molecules. Our data indicated that most, if not all,
MAIT cells displayed a CCR7
CD45RA
effector memory phe-
notype that may harbor immediate effector function (Supple-
mentary Fig. S2A and S2B). Likewise, MAIT cells expressed high
levels of CD45RO and CD95 in both patients with HCC and
healthy donors, irrespective of in the liver, tumor, and blood
(Fig. 2A; Supplementary Fig. S2C). Of note, the expression of
Figure 1.
MAIT cell denition, analyzing strategy, and distribution in patients with HCC by FCM and immunouorescence imaging. A, MAIT cell staining and analyzing
strategy by FCM in peripheral blood and tissues of patients with HCC. B, MAIT cell subset composition in tissues and peripheral blood of patients with HCC and
healthy donors. HD, healthy donor; PBMC, peripheral blood mononuclear cells. C, CD4
MAIT cell frequency in total T cells in peripheral blood and tissues of
patients with HCC and healthy donors detected by FCM analysis (,P<0.01; ,P<0.001 by MannWhitney Utest or one-way ANOVA and Tukey multiple
comparison tests). HD, healthy donor; PBMC, peripheral blood mononuclear cells. D, Representative staining for TCRva7.2 and CD161 on frozen sections, scanned
by Leica SP5 under 63 objective. E, Summary of density information of CD161
þ
and TCRva7.2
þ
MAIT cells in paired peritumor and tumor tissues (P¼0.018 by
MannWhitney Utest). Lines and error bars are presented as the mean SD.
Duan et al.
Clin Cancer Res; 25(11) June 1, 2019 Clinical Cancer Research3306
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
costimulatory molecules CD28 and CD127 on MAIT cells was
signicantly decreased in HCC tissues compared with either
peritumor or normal liver tissues (for CD28 mean: 92.6% vs.
97.5% or 98.3%, P¼0.010; for CD127 mean: 92.8% vs. 98.8% or
98.1%, P¼0.031; Fig. 2B), whereas in peripheral blood, no
noticeable expression difference was found between HCC and
healthy donors (Supplementary Fig. S2D). In addition, MAIT cells
expressed signicantly higher level of activating markers CD38
and HLA-DR in tumor compared with its counterpart in peritu-
mor or normal liver tissues (for CD38 mean: 14.6% vs. 8.69% or
4.54%, P¼0.044; for HLA-DR mean: 15.9% vs. 6.98% or 3.44%,
P¼0.014; Fig. 2C), whereas no obvious differences of these two
markers were seen on peripheral MAIT cells between HCC
patients and healthy donors (Supplementary Fig. S2E). Elevated
HLA-DR and CD38 expression could be resulted from chronic
infections that coexists and alternatively indicates a common
exhausted T-cell phenotype (25, 26). Furthermore, we checked
a group of ten T-cell and NK cellrelated activating and inhibitory
molecules (27) and found that two effector functionrelated
molecules CD160 (mean: 39.0% vs. 72.3% or 96.3%, P<
0.001) and KLRG1 (mean: 76.5% vs. 82.7% or 95.7%, P¼
0.019; Fig. 2D; Supplementary Fig. S2G) had signicantly lower
expression on HCC-inltrated MAIT cells, whereas CD160 was
also lower on circulating MAIT cells in patients with HCC (Sup-
plementary Fig. S2F). Collectively, these data showed that tumor
MAIT cells displayed a typical CCR7
CD45RA
CD45RO
þ
CD95
þ
effector memory phenotype with activated status and potentially
decreased effector capabilities.
Figure 2.
Expression of costimulatory and activation receptors on intrahepatic and peripheral blood MAIT cells. A, Representative plots of CD45RO and CD95 expression
on tissue and blood MAIT cells (gated on CD161
þ
TCRva7.2
þ
MR1-tet
þ
). Expression of costimulatory receptors (B), activation receptors (C), and inhibiting
receptors (D) on tissue MAIT cells was also detected by FCM (gated on CD161
þ
TCRva7.2
þ
MR1-tet
þ
). Representative overlays for tumor, peritumor, normal liver,
and isotype control and total summary data are shown (,P<0.05; ,P<0.01; ,P<0.001 by one-way ANOVA and Tukey multiple compari son tests). Lines
and error bars are presented as the mean SD.
MAIT Cells in HCC
www.aacrjournals.org Clin Cancer Res; 25(11) June 1, 2019 3307
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
Chemokine receptor expression prole in tumor-inltrating
MAIT cells
Because MAIT cells are accounting almost half of the intrahe-
patic T cells supposed to be mediated by CCR6 and CXCR6, and
evidenced a decrease expression of gut-homing receptor
CCR9 (9, 15). Herein, to further support MAIT trafcking, we
screened the 18 classic chemokine receptors both in patients with
HCC and healthy donors. Compared with healthy donors, the
expression of all the three receptors, CCR6 (MFI mean: 568.8 vs.
876 or 817.8, P¼0.042), CXCR6 (MFI mean: 104.9 vs. 115.1 or
177.6, P¼0.002), and CCR9 (MFI mean: 80.4 vs. 134.9 or 168.9,
P¼0.016), on MAIT cells was signicantly downregulated in the
patients with HCC, particularly in the tumor center (Fig. 3A;
Supplementary Fig. S3A), indicating a possible mechanism relat-
ed to low inltration of MAIT cells in HCC.
Scenario goes different while comparing CXCR3 expression
with chronic liver diseases, where a positive expression of CXCR3
was detected on MAIT cells (15) and mild expression of CXCR3
was detected in patients with HCC (data not shown). CCR2 is
related to IL17-secreting T cells, and is signicantly upregulated in
CD161
þ
T cells (28). In our study, there is a tendency of lower
expression of CCR2 in tumor tissues compared with normal livers
(Fig. 3B; Supplementary Fig. S3B). Previous study indicated
heterogeneous levels of CXCR4 expression on MAIT cells (15),
and here, the same phenomenon is reected (Fig. 3B). In a normal
liver, MAIT cells show a high expression of CCR5 (15), and we
found that HCC-inltrating MAIT cells maintained high expres-
sion of this marker (Fig. 3B). Taken together, these results showed
a selective downregulation of certain chemokine receptors may
affect the trafcking and residing capacity of tumor-inltrating
MAIT cells during the progression of hepatocarcinogenesis.
Apoptosis is unlikely involved in the impaired inltration of
MAIT cells in HCC
In addition to aberrant chemotaxis, apoptosis could also result
in less immune cells in tumor tissues. Bcl-2 family proteins are
Figure 3.
Expression of chemokine receptors and immune checkpoint molecules on circulating and intrahepatic MAIT cells. Aand B, Expressi on of chemokine receptors on
circulating and intrahepatic MAIT cells detected by FCM (gated on CD161
þ
TCRva7.2
þ
MR1-tet
þ
). Representative overlay plots for HCC peripheral blood
mononuclear cells (PBMC) and healthy donor (HD) PBMCs as well as for tumor, peritumor, normal liver, and isotype control are shown. Cand D, Representative
FCM overlay plots for PD-1
þ
MAIT cells in tumor, peritumor, and normal liver gated on total MAIT cells and its summary data (gated on CD4
CD161
þ
TCRva7.2
þ
).
HD, healthy donor; PBMC, peripheral blood mononuclear cells. E, Summary information for the expression of CTLA-4 and TIM-3 on peritumor and tumor-derived
MAIT cells detected by FCM (gated on CD161
þ
TCRva7.2
þ
MR1-tet
þ
). F, Representative FCM overlay plots show PD-1
þ
MAIT cells (black line) had simultaneously
higher CTLA-4 and TIM-3 expression than their PD-1
counterpart (light gray). Lines and error bars are presen ted as the mean SD (,P<0.05; ,P<0.01 by
MannWhitney Utest or one-way ANOVA and Tukey multiple comparison tests).
Duan et al.
Clin Cancer Res; 25(11) June 1, 2019 Clinical Cancer Research3308
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
central regulators of cell apoptosis (29). Therefore, we compared
the expression of apoptosis-related Bcl-2 family molecules in
MAIT cells between HCC and normal liver tissues. However,
neither proapoptotic molecules like BAX and BID, nor the anti-
apoptotic molecule Bcl-2 showed any differences between
patients with HCC and healthy donors (Supplementary Fig.
S3C). This indicated that apoptosis may not be involved in
reduction of MAIT cell frequency in the tumor tissues.
HCC-inltrated MAIT cells express higher levels of immune
checkpoints
In HCC, several mechanisms are involved in maintaining
immunosuppressive microenvironment, including the upregula-
tion of immune checkpoints (30). In this study, signicant
upregulation of PD-1 on MAIT cells was detected in tumors
compared with peritumor or normal livers in FCM cohort
(mean: 8.8% vs. 4.3% or 3.6%, P<0.001 and P¼0.015,
respectively; Fig. 3C and D). In peripheral blood, patients with
HCC harbored signicantly higher PD-1
þ
MAIT cells than healthy
donors (mean: 1.3% vs. 0.6%, P¼0.044; Fig. 3D). Likewise, MAIT
cell expression of another two immune inhibitory molecules,
CTLA-4 (mean: 6.9% vs. 1.6% or 1.2%, P¼0.006) and TIM-3
(mean: 3.2% vs. 1.3% or 0.23%, P¼0.024), was also signicantly
increased in the tumor tissues than normal liver tissues in FCM
cohort, whereas mild expression was detected in their peripheral
blood counterparts (Fig. 3E; Supplementary Fig. S4A and S4B).
Moreover, PD-1
high
MAIT cells had simultaneously higher CTLA-4
and TIM-3 expression (Fig. 3F). To validate these ndings, we
cocultured MAIT cells sorted from healthy blood with HCC cell
lines using Transwell system. FCM data showed that after cocul-
ture with HCC cells for 48 hours, signicant upregulation of PD-1,
CTLA-4, and TIM-3 on MAIT cells was observed in either contact
(P¼0.0010.014) or noncontact manners (P¼0.0010.009;
Supplementary Fig. S4CS4F). These results indicated that HCC-
inltrated MAIT cells were educated by tumor cells to be func-
tionally exhausted.
HCC-inltrated MAIT cells aberrantly produce tumor-
promoting cytokines
Previous studies have indicated that MAIT cells had the ability
to produce both Th1- and Th17-type cytokines after in vitro
stimulation with phorbol 12-myristate 13-acetate (PMA) and
ionomycin or anti-CD3 and anti-CD28 (9, 15, 31). In our study,
we showed that the intrinsic IFNg- and IL17-secreting ability of
tumor-derived MAIT cells was signicantly inhibited compared
with their counterparts in peritumor and normal liver tissues (for
IFNgmean: 47.8% vs. 83.1% or 90.2%, P<0.001; for IL17 mean:
5.88% vs. 9.87% or 14.6%, P¼0.028; Fig. 4A and B), after
stimulating with PMA and ionomycin for 5 hours. Consistently,
MAIT cells from peripheral blood of patients with HCC secreted
signicantly less IFNgand IL17 compared with healthy donors
(for IFNgmean: 72.9% vs. 90.2%, P¼0.013; for IL17 mean: 4.3%
vs. 14.6%, P<0.001; Supplementary Fig. S5A). Intriguingly, a
signicant upregulation of IL8, which has a crucial role in pro-
moting tumor angiogenesis and progression, was detected in
tumor-derived MAIT cells compared with peritumor and healthy
donors [for tissue mean: 0.73% vs. 0.36% or 0.02%, P¼0.017;
for peripheral blood mononuclear cell (PBMC), 0.79% vs. 0.02%,
P¼0.028; Supplementary Fig. S5B and S5C]. By contrast, IL4,
IL10, and IL22 were variable and no differences were observed
among MAIT cells from different sources (Supplementary
Fig. S5BS5D). Collectively, these results indicated that HCC
microenvironment inhibited the inherent cytokine-secreting
potential of MAIT cells, and promoted the secretion of IL8 by
MAIT cells.
HCC-derived MAIT cells produce minimal granzyme B and
perforin
Besides Th1/Th17 cytokine secretion, effector function of MAIT
cells depends on degranulation to kill sensitized targets (11).
Previous study has indicated that resting human MAIT cells are
featured by a lack of granzyme B and low perforin expression, but
with high expression of granzyme A and granzyme K (13). In this
study, we rst determined the expression of granzymes and
perforin on resting MAIT cells. After recovery from liver tissues,
immune cells were stained with uorescence dyeconjugated
antibodies following a FOXP3 staining protocol. Under ex vivo
condition, neither granzyme B nor perforin was detected in MAIT
cells (Fig. 4C; Supplementary Fig. S5E). Different from granzyme
B and perforin, the secretion of which is tightly controlled in vivo,
both noncytotoxic molecules granzyme A and granzyme K had
high expression in intrahepatic MAIT cells, with a relatively lower
expression of granzyme K in HCC-derived MAIT cells (Fig. 4C;
Supplementary Fig. S5E).
MAIT cells can be activated in a MR1-dependent manner or
through IL12 and IL18 stimulation in a TCR-independent manner
in vitro (31). MAIT cells can also be efciently activated after
cocultured with nonprofessional antigen-presenting cells (APC)
pretreated with Escherichia coli stimulation, leading to substantial
secretion of granzyme B and perforin (31). Following this strategy,
MAIT cells were successfully activated to produce granzyme B and
perforin after coculture with E. colipretreated THP-1 cells for 5
hours (Fig. 4D). However, the frequencies of MAIT cells expres-
sing granzyme B (mean: 1.56% vs. 11.8% or 16.3%, P<0.001)
and perforin (mean: 36.0% vs. 46.3% or 62.7%, P¼0.002) were
signicantly lower in tumor compared with peritumor or normal
liver, where MAIT cells from normal liver produced almost 10-
and 2-fold more granzyme B and perforin than HCC tissue,
respectively (Fig. 4E). Furthermore, signicantly increased apo-
ptosis and impaired proliferation of HCC cells were observed after
in vitro coculture with MAIT cells derived from PBMCs of healthy
donors or peritumor liver tissues, with markedly upregulated
secretion of GM-CSF, TNFa, IFNg, and MIP1ain the coculture
supernatant (Supplementary Figs. S6 and S7). However, coculture
supernatant from tumor-inltrating MAIT cells and HCC cells
signicantly promoted proliferation and invasion, and inhibited
apoptosis of HCC cells in vitro (Supplementary Fig. S7). These
results indicated that normal MAIT cells could induce
apoptosis of HCC cells, whereas HCC-inltrated MAIT cells lose
tumor-killing ability, promoted proliferation, and invasion of
HCC cells.
Molecular characterization of intrahepatic MAIT cells by
RNA-seq
Our above data revealed obvious phonotypic differences
between HCC and liver-derived MAIT cells. We further deter-
mined their global gene expression differences by sorting MAIT
cells from paired tumor and peritumor liver tissues in 5 patients
with HCC, as well as from 5 normal liver of healthy donors, for
RNA-seq. RNA-seq analysis detected more than 6,000 signicant-
ly differentially expressed genes in tumor-derived MAIT cells
compared with peritumor or normal liver tissues, suggesting an
MAIT Cells in HCC
www.aacrjournals.org Clin Cancer Res; 25(11) June 1, 2019 3309
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
Figure 4.
Cytokine secretion and degranulation prole of intrahepatic and peripheral MAIT cells. A, IFNg- and IL17- secreting proles of intrahepatic MAIT cells after PMA,
ionomycin, and BFA (PIB) stimulation for 5 hours and (B) its summary data (gated on CD4
CD161
þ
TCRva7.2
þ
). C, Representative FCM overlay plots for perforin
and granzymes secreted by peripheral and intrahepatic MAIT cells under still condition (gated on CD161
þ
TCRva7.2
þ
MR1-tet
þ
). HD, healthy donor. D, Bacterial
stimulation led to degranulation and changes in cytotoxic prole of MAIT cells. Sorted MAIT cells from normal liver, peritumor, and tumor tissues were cocultured
with THP-1 cells, pretreated with or without E. coli, and analyzed by FCM to determine perforin and granzyme B secretion in respective tissues. E, Cumulative
data showing reduced frequency of perforin- and granzyme Bexpressin g MAIT cells (gated on CD161
þ
TCRva7.2
þ
MR1-tet
þ
). GrB, granzyme B. Lines and error
bars are presented as the mean SD (,P<0.05; ,P<0.01; ,P<0.001 by one-way ANOVA and Tukey multiple comparison tests).
Duan et al.
Clin Cancer Res; 25(11) June 1, 2019 Clinical Cancer Research3310
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
Figure 5.
Gene expression prole of intrahepatic MAIT cells based on RNA-seq data. A, Visualization of 15 tissues by rst two principal components (comp) of principle
component analysis computed on gene expression matrix. B, The Venn diagrams of different ially expressed genes for each pair of groups (P for peritumor,
T for tumor, and HDL for normal liver tissues). DEGs were identied by negative binomial generalized linear model (nbGLM) with |log
2
(fold-change)| >1 and
P<0.01. C, Hierarchical clustering for all DEGs in B.D, Enrichment analysis of up- and downregulated DEGs uniquely altered in tumor MAIT cells. Dot size and
color represent the number of genes and Pvalues, respectively. E, Representative expression plot of differentially expressed genes involving cytokine secretion,
tumor promotion, and metabolism in MAIT cells between tumor an d other tissues. |log
2
(fold-change)| >1, P<0.01, by nbGLM.
MAIT Cells in HCC
www.aacrjournals.org Clin Cancer Res; 25(11) June 1, 2019 3311
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
aberrant gene expression prole in MAIT cells within HCC micro-
environment (Fig. 5AC; Supplementary Table S3).
Next, we aimed to determine the biological pathways uniquely
altered in tumor MAIT cells based on those down- and upregu-
lated genes. The downregulated genes, like NFKB1,STAT5B, and
TGFB1 (3234), were enriched in pathways of cytokine secreting
and cytolysis effector function, consistent with our ndings that
effector function of tumor MAIT cells were severely impaired
(Fig. 5D and E; Supplementary Table S3), whereas signicantly
upregulated genes in tumor MAIT cells, like APOE and ALDH1A2,
were enriched in pathways involved in aberrant glucose and
cholesterol metabolism (Fig. 5CE; Supplementary Table S3).
Moreover, 114 differentially expressed genes were shared when
comparing either of tumor, peritumor, and normal liver tissues,
and Gene Ontology analysis indicated that those genes were
mainly involved in metabolism, supporting the notion that
immune cells may undergo metabolic reprogramming in tumor
milieu (Fig. 5B; Supplementary Fig. S8A and S8B; Supplementary
Table S4; refs. 35, 36).
Genes aberrantly upregulated in tumor MAIT cells, like IL8,
CXCL12, and HAVCR2 (TIM-3), foster HCC development
(37, 38). Specially, IL8, an important proinammatory and
angiogenic factor (39), was one of the most upregulated genes
in tumor-derived MAIT cells, which was consistent with our
previous cytokine secretion result. On the basis of The Caner
Genome Atlas HCC survival data, we showed that patients with
higher expression of IL8 were signicantly correlated with reduced
survival (P¼0.012, by log rank test; Supplementary Fig. S8C).
Thus, RNA-seq results clearly indicated an inhibited cytolytic
effector function and induced tumor-promoting potential of
MAIT cells in the HCC microenvironment.
MAIT inltration correlates with unfavorable outcomes in HCC
On the basis of the unique cytokine secretion pattern, MAIT
cells were reported to have a protective role in constraining
bacterial infection. Although in the tumor context, different
conclusions have been made (1820, 40). In this study,
we evaluated the relationship of tumor-inltrating MAIT cell
density with HCC patient prognosis by FCM, qRT-PCR, and
immunostaining.
The 2-year overall survival (OS) and relapse-free survival (RFS)
rates for FCM cohort (n¼50) were 82.3% and 56.6%. To
determine the relationship between MAIT cell inltration and
survival, patients were divided into MAIT-high and MAIT-low
groups according to MAIT cell percentage in CD3
þ
T cells in
tumors using lowest tertile of MAIT percentage as the cutoff. Log-
rank test showed that patients with high MAIT cell inltration had
signicantly poor RFS (P¼0.031) and relatively lower OS (P¼
0.118) compared with low-inltration group (Fig. 6A).
TCRva-Ja33 mRNA expression level was previously used to
dene MAIT cell inltration (22, 41). In our study, qRT-PCR
data showed that TCRva-Ja33 mRNA expression was
signicantly decreased in tumor compared with peritumor
tissues in a cohort of 207 patients (0.066 vs. 0.222, P<
0.001; Fig. 6B). When stratifying the patients using lowest
tertile of TCRva7.2 mRNA expression level as cutoff, patients
with high TCRva-Ja33 mRNA level showed signicantly
dismal survival (P¼0.036) and increased recurrence (P¼
0.034; Fig. 6C). Furthermore, TCRva7.2-Ja33 mRNA expres-
sion level was revealed to be an independent prognostic
factor for both OS [HR 1.92; 95% condence interval (CI),
1.163.24; P¼0.041] and RFS (HR 1.89; 95% CI, 1.142.88;
P¼0.046) in addition to the factors like tumor size,
differentiation, capsule, and vascular invasion (Cox propor-
tional hazards regression; Supplementary Table S5).
We further detected absolute number of MAIT cells by TSA
method on HCC TMA (n¼224). MAIT cells were mainly enriched
in portal area (Fig. 6D), the density of which signicantly
decreased in tumor compartment than peritumor tissues (mean
number: 4.1 vs. 5.8 cells/core, P<0.001; Fig. 6E). Similarly, in
tumor, when stratifying the patients using lowest tertile of MAIT
cell density as cutoff (1.5 MAIT cells/core), patients with high
MAIT cell inltration signicantly correlated with unfavorable
outcomes (for OS: P¼0.045; for RFS: P¼0.119; Fig. 6F), and
was conrmed as an independent index for OS (Supplementary
Table S6). Then, in the validation cohort, using same cutoff,
we conrmed that high inltration of MAIT cells was associated
with poor OS (P¼0.005) and RFS (P¼0.01; Supplementary
Fig. S9; Supplementary Table S6). Collectively, survival analysis
of all four cohorts demonstrated that high density of tumor-
inltrating MAIT cells indicated dismal clinical outcomes in
patients with HCC.
Discussion
Innate immunity plays an important role in antitumor immune
responses, among which MAIT cells are a population of innate-
type T cells preferentially enriched in human liver, indicating its
pivotal role in liver immunology. Importantly, this may be the
rst report of such rigorous evaluation of the distribution, phe-
notype, function, and clinical relevance of circulating and inl-
trating MAIT cells in patients with HCC. We found that overall
signicant decrease of MAIT cells in tumor and peripheral blood
of patients with HCC signify a systemic dysregulation occurred in
disease state, which was different from the conventional healthy
donors. Of note, tumor-inltrating MAIT cells displayed an acti-
vating and exhausted phenotype with impaired effector capabil-
ity, and even shifted to produce tumor-promoting cytokines.
Interestingly, our prime ndings further reveal that the high
density of tumor-inltrating MAIT cells signicantly correlated
with unfavorable clinical outcomes, and we could possibly infer
that MAIT cells are reprogrammed within the tumor microenvi-
ronment and may contribute to HCC development.
Reduction of circulating MAIT cell frequency has been reported
in various kinds of bacterial infections and viral infections,
including HBV and HCV, as well as in patients with colon
cancer (16, 1820, 42). Similar to our FCM analysis, we observed
a signicant decrease of circulating MAIT cell frequency in patients
with HCC. This reduction may be attributed to tumor-associated
factors or chronic infectious conditions, considering that majority
of patients were HBV
þ
, which couldn't be ruled out. However,
whether MAIT cell inltration in tumor tissues was decreased or
increased compared with the nontumor counterparts remains
controversial. An increased MAIT cell inltration in colon cancer
and a decreased inltration in colorectal hepatic metastases were
observed, which were compared with normal colon and normal
liver respectively (19, 43). Our ow cytometric, IHC, and qRT-
PCR data collectively demonstrated that MAIT cell inltration was
signicantly lower in HCC tumor than of peritumor tissues. Being
aware of the abundance of intrahepatic MAIT cells, we wondered
to compare the absolute number or frequency of inltrating MAIT
cells aided with multiplex IHC. Our results showed that the
Duan et al.
Clin Cancer Res; 25(11) June 1, 2019 Clinical Cancer Research3312
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
Figure 6.
Higher inltration of MAIT cells correlates with unfavorable clinical outcomes. A, KaplanMeier curves for OS and RFS according to MAIT cell frequency in the
FCM cohort (n¼50). B, Relative expression of TCRva7.2 in tumor and peritumor tissues (n¼209, paired ttest). C, KaplanMeier curves for OS and RFS
according to TCRva7.2 mRNA expression level in the qRT-PCR cohort. D, Representative images of MAIT cell distribution in peritumor tissues. The subset was
dened as CD3
þ
MDR-1
þ
IL-18R
þ
cells using the TSA method, and immunouorescence images were scanned at 20on the Vectra Automated Imaging System.
E, MAIT cell absolute number signicantly decreased in tumor compared with peritumor (mean number: 4.1 vs. 5.8 cells/core, P<0.001; n¼224, by paired ttest).
F, KaplanMeier curves for OS and RFS according to MAIT cell density in the TMA training cohort. Lines and error bars are presented as the mean SD.
MAIT Cells in HCC
www.aacrjournals.org Clin Cancer Res; 25(11) June 1, 2019 3313
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
densities of MAIT cells in HCC and paired liver tissues were 7.2
versus 17.4 cells/mm
2
, whereas MAIT cells in colon cancer and
healthy colon mucosa were reported to be 6.1 versus 2.6 cells/
mm
2
(19). Alternatively, when evaluating MAIT cell frequency
among CD3
þ
T cells, similar trend was observed (4.2% and 3.2%
for HCC and colon cancer, respectively; ref. 18). Obviously, MAIT
cell inltration in HCC slightly surpassed that in colon cancer.
Hence, we assumed that MAIT cells were relatively accumulated in
HCC although less than the peritumor liver tissues, similar to the
ndings that MAIT cells heavily inltrated colorectal hepatic
metastases, but to a lesser extent than the liver (19, 43). A selective
downregulation of chemokine receptors in HCC-inltrating MAIT
cells observed in our study could be a possible explanation for the
less inltration.
In both the patients with HCC and healthy donors, MAIT cells
displayed a typical CCR7
CD45RA
CD45RO
þ
CD95
þ
effector
memory phenotype (9, 44). MAIT cells are known to respond
against external stimuli reecting with the release of cytokines in
the surroundings and have some important implications in
disease regulation. However, HCC-inltrating MAIT cells' effector
function was found to be severely impaired, and even could
produce tumor-promoting cytokine like IL8. First, both IFNgand
IL17 secreted by tumor-derived MAIT cells were signicantly
lower than the counterparts in peritumor and healthy liver tissues.
Meanwhile, our observations also detect an obvious decline of
CD160 and CD127 expression in tumor-derived MAIT cells.
Generally, MAIT cells are known to express higher level of CD160
and CD127 both in peripheral blood and liver tissues, which are
necessary for Th1 and Th17 cytokine production (14, 45). Thus, it
is possible that suppressed IFNgand IL17 secretion of intratumor
MAIT cells could be ascribed to CD160 and CD127 downregula-
tion. Second, MAIT cells' effector function mainly relied on
granzyme B and perforin, which are key molecules necessary for
the efcient cytotoxic activity (46, 47). Our data demonstrated
that HCC-inltrating MAIT cells produced signicantly less gran-
zyme B and perforin than control, indicating the cytotoxic poten-
tial of intratumor MAIT cells was substantially inhibited, assum-
ing a consistent local microenvironmental impact induced the
transition. Third, we found that PD-1, CTLA-4, and TIM-3 expres-
sion was markedly upregulated in HCC-inltrating MAIT cells,
together with a high expression of the activating markers, CD38
and HLA-DR, which share common features of exhausted T cells,
was thought to be negative immune regulator in the tumor
milieu (26, 48). Finally, a signicant upregulation of IL8, known
for tumor-promoting factor (49), was detected in HCC-derived
MAIT cells. Additional in vitro coculture experiments conrmed
the tumor-promoting function mediated by intratumor MAIT
cells as compared with peritumor or circulating MAIT cells.
Altogether, similar to tumor-associated macrophages and neu-
trophils, we postulated that tumor-inltrating MAIT cells were
reprogrammed to a tumor-promoting direction. As such, cocul-
ture with HCC cells could lead to a signicant upregulation of PD-
1, CTLA-4, and TIM-3 on MAIT cells. RNA-seq analysis of inl-
trating MAIT cells further validated that genes related to cell
activation, cytokine secretion, and metabolism were rerouted to
favor a tumor-promoting function in HCC.
Consistent with their tumor-promoting function, we found
that high levels of tumor-inltrating MAIT cells signicantly and
independently correlated with dismal clinical outcomes as estab-
lished in four independent cohorts of patients with HCC. To date,
the prognostic value of MAIT cells has only been reported in colon
cancer, where high densities of tumor-inltrating MAIT cells were
also associated with poor survival and serum CEA level positively
correlated with MAIT cell inltration (18). Nonetheless, our
results were mainly derived from patients with HBV-related HCC.
It will be important to validate the prognostic value of MAIT cells
among patients with HCC with other etiologies like in HCV or
fatty liver.
In summary, our ndings showed that HCC-inltrating
MAIT cells were skewed toward a tumor-promoting direction
and were detrimental to patient prognosis. Soluble factors
derived from HCC cells or direct contact with HCC cells can
activate MAIT cells within the tumor milieu, markedly suppress
their cytotoxic capability, and induce them to produce signif-
icant amount of protumor cytokines. These reprogrammed
MAIT cells shifted away from antitumor toward tumor-
suppressive and proangiogenic pathways. Strategies that mod-
ulate the function of MAIT cells may provide a new avenue for
antitumor therapy in HCC.
Transcript Proling
The RNA-seq data in this paper have been submitted to the National Center
for Biotechnology Information (NCBI) Gene Expression Omnibus (GEO;
accession GSE117627).
Disclosure of Potential Conicts of Interest
No potential conicts of interest were disclosed.
Authors' Contributions
Conception and design: M. Duan, S. Goswami, J. Fan, X.-M. Zhang, Q. Gao
Development of methodology: M. Duan, S. Goswami, J.-Y. Shi, Q. Gao
Acquisition of data (provided animals, acquired and managed patients,
provided facilities, etc.): M. Duan, S. Goswami, J.-Y. Shi, J.-Q. Ma, L.-J. Ma,
R.-B. Xi, Q. Gao
Analysis and interpretation of data (e.g., statistical analysis, biostatistics,
computational analysis): M. Duan, S. Goswami, L.-J. Wu, J.-Q. Ma, Z. Zhang,
Y. Shi, Q. Gao
Writing, review, and/or revision of the manuscript: M. Duan, S. Goswami,
X.-Y. Wang, J. Fan, X.-M. Zhang, Q. Gao
Administrative, technical, or material support (i.e., reporting or organizing
data, constructing databases): M. Duan, J.-Y. Shi, Y. Cao, J. Zhou, J. Fan, Q. Gao
Study supervision: M. Duan, X.-Y. Wang, S. Zhang, Y. Cao, J. Zhou, J. Fan,
X.-M. Zhang, Q. Gao
Acknowledgments
The MR1 tetramer technology was developed jointly by Dr. James
McCluskey, Dr. Jamie Rossjohn, and Dr. David Fairlie, and the material
was produced by the NIH. The authors thank Dr. Xiong Ma from the
Department of Gastroenterology and Hepatology, Renji Hospital, Shanghai
Jiaotong University (Shangai, China) for the MR1 tetramer as a generous gift.
This work was supported by the Strategic Priority Research Program (grant
number XDB29030302), Interdisciplinary Innovation Team, Frontier
Science Key Research Project (grant number QYZDB-SSW-SMC036), Chinese
Academy of Sciences (to X.-M. Zhang), National Natural Science Foundation
of China (grant numbers 81772556 and 81572367 to X.-Y. Wang; grant
numbers 81522036 and 81572292 to Q. Gao; and grant number 81872321
to J.-Y. Shi), and National Program for Special Support of Eminent
Professionals (to Q. Gao).
The costs of publication of this article were defrayed in part by the
payment of page charges. This article must therefore be hereby marked
advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate
this fact.
Received September 19, 2018; revised December 25, 2018; accepted February
1, 2019; published rst February 5, 2019.
Duan et al.
Clin Cancer Res; 25(11) June 1, 2019 Clinical Cancer Research3314
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
References
1. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer
statistics, 2012. CA Cancer J Clin 2015;65:87108.
2. Bruix J, Reig M, Sherman M. Evidence-based diagnosis, staging, and
treatment of patients with hepatocellular carcinoma. Gastroenterology
2016;150:83553.
3. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, et al.
Nivolumab in patients with advanced hepatocellular carcinoma (Check-
Mate 040): an open-label, non-comparative, phase 1/2 dose escalation and
expansion trial. Lancet 2017;389:2492502.
4. Beatty GL, Gladney WL. Immune escape mechanisms as a guide for cancer
immunotherapy. Clin Cancer Res 2015;21:68792.
5. Littman DR. Releasing the brakes on cancer immunotherapy. Cell 2015;
162:118690.
6. Sia D, Jiao Y, Martinez-Quetglas I, Kuchuk O, Villacorta-Martin C,
de Moura MC, et al. Identication of an immune-specic class of hepa-
tocellular carcinoma, based on molecular features. Gastroenterology 2017;
153:81226.
7. Woo SR, Corrales L, Gajewski TF. Innate immune recognition of cancer.
Annu Rev Immunol 2015;33:44574.
8. Jenne CN, Kubes P. Immune surveillance by the liver. Nat Immunol 2013;
14:9961006.
9. Dusseaux M, Martin E, Serriari N, Peguillet I, Premel V, Louis D, et al.
Human MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-
17-secreting T cells. Blood 2011;117:12509.
10. Kjer -Nielsen L, Patel O, Corbett AJ, Le Nours J, Meehan B, Liu L, et al. MR1
presents microbial vitamin B metabolites to MAIT cells. Nature 2012;491:
71723.
11. Kurio kaA, Walker LJ, Klenerman P, Willb ergCB. MAIT cells: new guardi ans
of the liver. Clin Transl Immunology 2016;5:e98.
12. Reantragoon R, Corbett AJ, Sakala IG, Gherardin NA, Furness JB, Chen Z,
et al. Antigen-loaded MR1 tetramers dene T cell receptor heterogeneity in
mucosal-associated invariant T cells. J Exp Med 2013;210:230520.
13. Kurioka A, Ussher JE, Cosgrove C, Clough C, Fergusson JR, Smith K, et al.
MAIT cells are licensed through granzyme exchange to kill bacterially
sensitized targets. Mucosal Immunol 2015;8:42940.
14. Tang XZ, Jo J, Tan AT, Sandalova E, Chia A, Tan KC, et al. IL-7 licenses
activation of human liver intrasinusoidal mucosal-associated invariant T
cells. J Immunol 2013;190:314252.
15. Jeffery HC, van Wilgenburg B, Kurioka A, Parekh K, Stirling K, Roberts S,
et al. Biliary epithelium and liver B cells exposed to bacteria activate
intrahepatic MAIT cells through MR1. J Hepatol 2016;64:111827.
16. Eberhard JM, Kummer S, Hartjen P, H
ufner A, Diedrich T, Degen O, et al.
Reduced CD161þMAIT cell frequencies in HCV and HIV/HCV co-infec-
tion: is the liver the heart of the matter? J Hepatol 2016;65:12613.
17. Walke r LJ, Marrinan E, Muenchhoff M, Ferguson J, Kloverpris H, Cheroutre
H, et al. CD8alphaalpha expression marks terminally differentiated human
CD8þT cells expanded in chronic viral infection. Front Immunol 2013;4:
223.
18. Ling L, Lin Y, Zheng W, Hong S, Tang X, Zhao P, et al. Circulating and
tumor-inltrating mucosal associated invariant T (MAIT) cells in colorec tal
cancer patients. Sci Rep 2016;6:20358.
19. Zabijak L, Attencourt C, Guignant C, Chatelain D, Marcelo P, Marolleau J-P,
et al. Increased tumor inltration by mucosal-associated invariant T cells
correlates with poor survival in colorectal cancer patients. Cancer Immunol
Immunother 2015;64:16018.
20. Sundstr
om P, Ahlmanner F, Ak
eus P, Sundquist M, Als
en S, Yrlid U, et al.
Human mucosa-associated invariant T cells accumulate in colon adeno-
carcinomas but produce reduced amounts of IFN-g. J Immunol 2015;195:
347281.
21. Shi JY, Gao Q, Wang ZC, Zhou J, Wang XY, Min ZH, et al. Margin-inltrating
CD20(þ) B cells display an atypical memory phenotype and correlate with
favorable prognosis in hepatocellular carcinoma. Clin Cancer Res 2013;19:
59946005.
22. Lepore M, Kalinichenko A, Colone A, Paleja B, Singhal A, Tschumi A, et al.
Parallel T-cell cloning and deep sequencing of human MAIT cells reveal
stable oligoclonal TCRbeta repertoire. Nat Commun 2014;5:3866.
23. Picelli S, Bjorklund AK, Reinius B, Sagasser S, Winberg G, Sandberg R. Tn5
transposase and tagmentation procedures for massively scaled sequencing
projects. Genome Res 2014;24:203340.
24. Gherardin NA, Souter MN, Koay HF, Mangas KM, Seemann T, Stinear TP,
et al. Human blood MAIT cell subsets dened using MR1 tetramers.
Immunol Cell Biol 2018;96:50725.
25. Yong YK, Saeidi A, Tan HY, Rosmawati M, Enstrom PF, Batran RA, et al.
Hyper-expression of PD-1 is associated with the levels of exhausted and
dysfunctional phenotypes of circulating CD161(þþ)TCR iValpha7.2(þ)
mucosal-associated invariant T cells in chronic hepatitis B virus infection.
Front Immunol 2018;9:472.
26. Baitsch L, Baumgaertner P, Devevre E, Raghav SK, Legat A, Barba L, et al.
Exhaustion of tumor-specic CD8(þ) T cells in metastases from melanoma
patients. J Clin Invest 2011;121:235060.
27. Henson SM, Akbar AN. KLRG1more than a marker for T cell senescence.
Age 2009;31:28591.
28. Billerbeck E, Kang YH, Walker L, Lockstone H, Grafmueller S, Fleming V,
et al. Analysis of CD161 expression on human CD8þT cells denes a
distinct functional subset with tissue-homing properties. Proc Natl Acad Sci
U S A 2010;107:300611.
29. Reed JC. Proapoptotic multidomain Bcl-2/Bax-family proteins: mechan-
isms, physiological roles, and therapeutic opportunities. Cell Death Differ
2006;13:137886.
30. Hato T, Goyal L, Greten TF, Duda DG, Zhu AX. Immune checkpoint
blockade in hepatocellular carcinoma: current progress and future direc-
tions. Hepatology 2014;60:177682.
31. Ussher JE, Bilton M, Attwod E, Shadwell J, Richardson R, de Lara C, et al.
CD161þþ CD8þT cells, including the MAIT cell subset, are specically
activated by IL-12þIL-18 in a TCR-independent manner. Eur J Immunol
2014;44:195203.
32. Lougar is V, Patrizi O, Baronio M, Tabellini G, Tampella G, Damiati E, et al.
NFKB1 regulates human NK cell maturation and effector functions.
Clin Immunol 2017;175:99108.
33. Luedde T, Schwabe RF. NF-kappaB in the liverlinking injury, brosis and
hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2011;8:10818.
34. Smith AJ, Humphries SE. Cytokine and cytokine receptor gene polymorp h-
isms and their functionality. Cytokine Growth Factor Rev 2009;20:4359.
35. Biswas SK. Metabolic reprogramming of immune cells in cancer progres-
sion. Immunity 2015;43:43549.
36. Wu X, Thi VLD, Liu P, Takacs CN, Xiang K, Andrus L, et al. Pan-genotype
hepatitis E virus replication in stem cellderived hepatocellular systems.
Gastroenterology 2018;154:66374. e7.
37. Kim H-D, Song G-W, Park S, Jung MK, Kim MH, Kang HJ, et al. Association
between expression level of PD1 by tumor-inltrating CD8þT cells and
features of hepatocellular carcinoma. Gastroenterology 2018;155:
193650.
38. Liu H, Liu Y, Liu W, Zhang W, Xu J. EZH2-mediated loss of miR-622
determines CXCR4 activation in hepatocellular carcinoma. Nat Commun
2015;6:8494.
39. Park SY, Han J, Kim JB, Yang MG, Kim YJ, Lim HJ, et al. Interleukin-8 is
related to poor chemotherapeutic response and tumourigenicity in hepa-
tocellular carcinoma. Eur J Cancer 2014;50:34150.
40. Zheng C, Zheng L, Yoo J-K, Guo H, Zhang Y, Guo X, et al. Landscape of
inltrating T cells in liver cancer revealed by single-cell sequencing. Cell
2017;169:134256.
41. Peterfalvi A, Gomori E, Magyarlaki T, Pal J, Banati M, Javorhazy A, et al.
Invariant Va7. 2-Ja33 TCR is expressed in human kidney and brain tumors
indicating inltration by mucosal-associated invariant T (MAIT) cells.
Int Immunol 2008;20:151725.
42. Salou M, Franciszkiewicz K, Lantz O. MAIT cells in infectious diseases.
Curr Opin Immunol 2017;48:714.
43. Shaler CR, Tun-Abraham ME, Skaro AI, Khazaie K, Corbett AJ, Mele T, et al.
Mucosa-associated invariant T cells inltrate hepatic metastases in patients
with colorectal carcinoma but are rendered dysfunctional within and
adjacent to tumor microenvironment. Cancer Immunol Immunother
2017;66:156375.
44. Takata H, Takiguchi M. Three memory subsets of human CD8þT cells
differently expressing three cytolytic effector molecules. J Immunol 2006;
177:433040.
45. Tu TC, Brown NK, Kim TJ, Wroblewska J, Yang X, Guo X, et al. CD160 is
essential for NK-mediated IFN-gamma production. J Exp Med 2015;212:
41529.
www.aacrjournals.org Clin Cancer Res; 25(11) June 1, 2019 3315
MAIT Cells in HCC
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
46. Makedonas G, Banerjee PP, Pandey R, Hersperger AR, Sanborn KB, Hardy
GA, et al. Rapid up-regulation and granule-independent transport of
perforin to the immunological synapse dene a novel mechanism of
antigen-specic CD8þT cell cytotoxic activity. J Immunol 2009;182:
55609.
47. Harari A, Bellutti Enders F, Cellerai C, Bart PA, Pantaleo G. Distinct
proles of cytotoxic granules in memory CD8 T cells correlate with
function, differentiation stage, and antigen exposure. J Virol 2009;83:
286271.
48. Sakuishi K, Apetoh L, Sullivan JM, Blazar BR, Kuchroo VK, Anderson AC.
Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore
anti-tumor immunity. J Exp Med 2010;207:218794.
49. Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting
chronic inammation: a magic bullet? Science 2013;339:28691.
Clin Cancer Res; 25(11) June 1, 2019 Clinical Cancer Research3316
Duan et al.
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
2019;25:3304-3316. Published OnlineFirst February 5, 2019.Clin Cancer Res
Meng Duan, Shyamal Goswami, Jie-Yi Shi, et al.
and Indicate Poor Outcome in Hepatocellular Carcinoma
Activated and Exhausted MAIT Cells Foster Disease Progression
Updated version
10.1158/1078-0432.CCR-18-3040doi:
Access the most recent version of this article at:
Material
Supplementary
http://clincancerres.aacrjournals.org/content/suppl/2019/02/08/1078-0432.CCR-18-3040.DC1
Access the most recent supplemental material at:
Cited articles
http://clincancerres.aacrjournals.org/content/25/11/3304.full#ref-list-1
This article cites 49 articles, 14 of which you can access for free at:
Citing articles
http://clincancerres.aacrjournals.org/content/25/11/3304.full#related-urls
This article has been cited by 9 HighWire-hosted articles. Access the articles at:
E-mail alerts related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.org
To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at
Permissions
Rightslink site.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
.http://clincancerres.aacrjournals.org/content/25/11/3304
To request permission to re-use all or part of this article, use this link
on January 10, 2022. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from
Published OnlineFirst February 5, 2019; DOI: 10.1158/1078-0432.CCR-18-3040
... In colorectal cancer (CRC) patients, circulating MAIT cells may decline, accompanied by an activation pro le and an accumulation near or inside cancerous lesions [27,28] . Cervica [29] , lung [30,31] , gastric [32] , esophageal [33] , and hepatic [34][35] cancers demonstrate altered MAIT homeostasis. MAIT cells have been shown to exhibit diverse functional trends in different studies; thus, their role in these conditions remains unclear. ...
... Altered phenotype and cytokine production patterns in MAIT cells have been reported in different cancer types [27][28][29][30][31][32][33][34][35][36][37][38] . Multiple tumor types, including colorectal [27,28] , cervical [29] , gastric [32] , hepatocellular carcinomas [34,35] , multiple myeloma [39] , and acute myeloid leukemia [23] , have shown depleted circulating MAIT cells. ...
... Altered phenotype and cytokine production patterns in MAIT cells have been reported in different cancer types [27][28][29][30][31][32][33][34][35][36][37][38] . Multiple tumor types, including colorectal [27,28] , cervical [29] , gastric [32] , hepatocellular carcinomas [34,35] , multiple myeloma [39] , and acute myeloid leukemia [23] , have shown depleted circulating MAIT cells. In the present study also, the rate of circulating MAIT cells was found to be reduced in AML samples, and the frequency of MAIT cells was associated with the cytogenetic pro le. ...
Preprint
Full-text available
Acute myeloid leukemia (AML) is an aggressive cancer characterized by significantly low mucosa-associated invariant T (MAIT) cells. Though the reasons for their decline and their functional implications in AML are yet to be explored, their levels have been reported to be associated with the prognosis of the disease. This study aimed to characterize the frequency, phenotype, and function of circulating MAIT cells during AML progression. Our results showed that the levels of circulating MAIT cells in patients with AML (AML-MAIT cells) were lower compared to healthy donors, and they were found to express high levels of HLA-DR, PD-1, and TIM-3, indicating that the AML-MAIT cells exhibited an activated and exhausted phenotype. AML-MAIT cells expressed higher KLRG1 and CD57 levels, indicating that circulating AML-MAIT cells displayed a senescent phenotype. The observation that MAIT cells in patients with AML had both senescent and pro-apoptotic phenotypes may underlie the decrease in circulating AML-MAIT cells. Additionally, circulating AML-MAIT cells produced less IFN-γ, TNF-α, and granzyme B, suggesting dysfunctional anti-tumor immunity. Moreover, we found AML patients with adverse cytogenetic have fewer MAIT cells than that with intermediate/favorable cytogenetic. In conclusion, circulating MAIT cells are decreased due to exhausted and senescent phenotypes and are functionally impaired in patients with AML. Therefore, enhancing circulating MAIT cells might be an attractive therapeutic strategy for patients with AML.
... As discussed above, the signals received by MAIT cells, such as cytokines, cell-cell interactions, and metabolic cues, can sway them towards either promoting tumor growth or exerting anti-tumor effects ( Figure 1B). Understanding these signals and the conditions under which they operate is crucial for harnessing the therapeutic potential of MAIT cells in cancer (25,56,67,68,70,73,87). ...
... This state of exhaustion is characterized by a reduced capacity to proliferate, produce cytokines, and mediate cytotoxicity, alongside an increased expression of inhibitory receptors such as PD-1, CTLA-4, and TIM-3. Such phenotypic changes indicate that MAIT cells may contribute to an immunosuppressive tumor microenvironment, potentially aiding in cancer progression (25,56,67,70,87). ...
... As MAIT cells encounter an ongoing barrage of signals -from cancer cell antigens presented by MR1 to inflammatory cytokines and chemokines -they initially respond robustly. However, this sustained activation can lead to a state of overstimulation, may lead to the previously mentioned functional dysregulation (25,56,67,70,87). ...
Article
Full-text available
Mucosal-associated invariant T (MAIT) cells play diverse roles in cancer, infectious diseases, and immunotherapy. This review explores their intricate involvement in cancer, from early detection to their dual functions in promoting inflammation and mediating anti-tumor responses. Within the solid tumor microenvironment (TME), MAIT cells can acquire an ‘exhausted’ state and secrete tumor-promoting cytokines. On the other hand, MAIT cells are highly cytotoxic, and there is evidence that they may have an anti-tumor immune response. The frequency of MAIT cells and their subsets has also been shown to have prognostic value in several cancer types. Recent innovative approaches, such as programming MAIT cells with chimeric antigen receptors (CARs), provide a novel and exciting approach to utilizing these cells in cell-based cancer immunotherapy. Because MAIT cells have a restricted T cell receptor (TCR) and recognize a common antigen, this also mitigates potential graft-versus-host disease (GVHD) and opens the possibility of using allogeneic MAIT cells as off-the-shelf cell therapies in cancer. Additionally, we outline the interactions of MAIT cells with the microbiome and their critical role in infectious diseases and how this may impact the tumor responses of these cells. Understanding these complex roles can lead to novel therapeutic strategies harnessing the targeting capabilities of MAIT cells.
... However, most studies paid attention to few subsets of immune cell but not all immune cell types. Immune cells like MAIT and central-memory, which play important roles in immune system(16), also play pivotal roles in lots of cancer types (17,18). ...
... They can be activated by TCR-dependent and TCR-independent mechanisms and exhibit rapid, innate-like effector responses (30). Previous study reported that MAIT were functionally impaired and even reprogrammed to shift away from antitumor immunity and toward a tumor-promoting direction (17). However, in other studies, MAIT cells were also found to be abundant in healthy liver tissue, but diminished in number in the tumor site, and reduced MAIT cell frequencies appeared to correlate with poor prognosis in these patients, which was consistent with our ndings(18, 31). ...
Preprint
Full-text available
Background The aim of this study was to determine whether differences in the cellular composition of the immune infiltrate in HCC influence survival and identify predictors for immunotherapy efficacy in hepatocellular carcinoma. Methods A total of 362 patients from TCGA cohort and 204 patients from ICGC with HCC were included in the study. Two immune features were selected out of 24 immune features to construct immunotypes based on the Cox regression model. Hub genes of DEGs were identified by STRING and Cyto-scape. The role of hub genes on immunotherapy efficacy prediction was evaluated by Kaplan–Meier survival analysis in immunotherapy cohorts. The effects of LCK on HCC cell proliferation and migration were evaluated by CCK8, trans-well and wound healing assays. Results Eight immune cell subsets were associated with HCC prognosis. Two immune cells (MAIT and central-memory) were selected to construct 3 immunotypes which could predict overall survival in the TCGA cohort (X² = 24.13, P < 0.0001) and ICGC cohort (validation cohort, X² = 10.51, P = 0.005). GO and GSEA analysis showed up-regulated immune-related pathway in Cluster3, and Cluster3 showed significantly higher immune checkpoint molecules (PD-L1, PD-1, CTLA-4, PD-L2, LAG3 and TIM3) expression. Three hub genes (CCR5, CCR7 and LCK) were identified based on the differential expression genes between Cluster3 and Cluster1. CCR5, CCR7 and LCK were efficient predictors for immune infiltration, especially CTL, and immunotherapy efficacy. We also verified that LCK conferred proliferation and metastasis of HCC cells and immunotherapy resistance of HCC patients. Conclusion Immune cell abundance and immunotypes could effectively predict prognosis of HCC. Furthermore, CCR5, CCR7 and LCK were identified as predictors for immunotherapy efficacy in hepatocellular carcinoma.
... The number of mucosal-associated invariant T (MAIT) cells and natural killer cells increased from Pro-T to Pro-Meta, suggesting that innate immunity may play a role in constructing the TME of the Pro-Meta group. Duan et al. found that tumor-educated MAIT cells can promote tumors by upregulating inhibitory factors, such as CTLA-4 [12]. Three types of macrophages, MMP9 + , TREM2 + , and MARCO + , were more abundant in the Pro-Meta group (Fig. 1B). ...
Article
Full-text available
Background Patients with primary multifocal hepatocellular carcinoma (HCC) have a poor prognosis and often experience a high rate of treatment failure. Multifocal HCC is mainly caused by intrahepatic metastasis (IM), and though portal vein tumor thrombosis (PVTT) is considered a hallmark of IM, the molecular mechanism by which primary HCC cells invade the portal veins remains unclear. Therefore, it is necessary to recognize the early signs of metastasis of HCC to arrange better treatment for patients. Results To determine the differential molecular features between primary HCC with and without phenotype of metastasis, we used the CIBERSORTx software to deconvolute cell types from bulk RNA-Seq based on a single-cell transcriptomic dataset. According to the relative abundance of tumorigenic and metastatic hepatoma cells, VEGFA⁺ macrophages, effector memory T cells, and natural killer cells, HCC samples were divided into five groups: Pro-T, Mix, Pro-Meta, NKC, and MemT, and the transcriptomic and genomic features of the first three groups were analyzed. We found that the Pro-T group appeared to retain native hepatic metabolic activity, whereas the Pro-Meta group underwent dedifferentiation. Genes highly expressed in the group Pro-Meta often signify a worse outcome. Conclusions The HCC cohort can be well-typed and prognosis predicted according to tumor microenvironment components. Primary hepatocellular carcinoma may have obtained corresponding molecular features before metastasis occurred.
... MAITs are an overabundant T cell subtype in the healthy human liver. The role of MAIT cells in cancer is not well defined, as increased MAIT cell numbers have been correlated with a poor prognosis in HCC [93]. In contrast, higher MAIT cell infiltration was correlated with a favorable prognosis within a cohort of CCA patients [94]. ...
Article
Full-text available
Simple Summary Liver cancer is a deadly disease, in which hepatocellular carcinoma and cholangiocarcinoma are the most common types. Despite numerous advances, treatment options still remain poor for liver cancer patients. Tumor development and progression as well as response to treatment are highly modulated by the cellular and stromal components that together with tumor cells generate the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) are one of the principal immune cell components of the liver TME, and they play a crucial role in sustaining tumorigenesis. This review summarizes recent findings on TAM biology, and it analyses the importance of the development of macrophage-targeting strategies for the treatment of liver cancer. Abstract Liver cancer is one of the leading causes of cancer-related mortality. Hepatocellular carcinoma and cholangiocarcinoma are the most common types, and despite numerous advances, therapeutic options still remain poor for these cancer patients. Tumor development and progression strictly depend on a supportive tumor microenvironment (TME). Tumor-associated macrophages (TAMs) are the most abundant immune cells population within a tumorigenic liver; they sustain cancer cells’ growth and invasiveness, and their presence is correlated with a poor prognosis. Furthermore, TAM cross-talk with cells and components of the TME promotes immunosuppression, a desmoplastic response, and angiogenesis. In this review, we summarize the latest advances in understanding TAM heterogeneity and function, with a particular focus on TAM modulation of the TME. We also discuss the potential of targeting macrophage subpopulations and how this is now being exploited in current clinical trials for the treatment of liver cancer.
Article
Full-text available
Background Liver cancer (LC) is a prevalent malignancy and a leading cause of cancer-related mortality worldwide. Extensive research has been conducted to enhance patient outcomes and develop effective prevention strategies, ranging from molecular mechanisms to clinical interventions. Single-cell sequencing, as a novel bioanalysis technology, has significantly contributed to the understanding of the global cognition and dynamic changes in liver cancer. However, there is a lack of bibliometric analysis in this specific research area. Therefore, the objective of this study is to provide a comprehensive overview of the knowledge structure and research hotspots in the field of single-cell sequencing in liver cancer research through the use of bibliometrics. Method Publications related to the application of single-cell sequencing technology to liver cancer research as of December 31, 2023, were searched on the web of science core collection (WoSCC) database. VOSviewers, CiteSpace, and R package “bibliometrix” were used to conduct this bibliometric analysis. Results A total of 331 publications from 34 countries, primarily led by China and the United States, were included in this study. The research focuses on the application of single cell sequencing technology to liver cancer, and the number of related publications has been increasing year by year. The main research institutions involved in this field are Fudan University, Sun Yat-Sen University, and the Chinese Academy of Sciences. Frontiers in Immunology and Nature Communications is the most popular journal in this field, while Cell is the most frequently co-cited journal. These publications are authored by 2799 individuals, with Fan Jia and Zhou Jian having the most published papers, and Llovet Jm being the most frequently co-cited author. The use of single cell sequencing to explore the immune microenvironment of liver cancer, as well as its implications in immunotherapy and chemotherapy, remains the central focus of this field. The emerging research hotspots are characterized by keywords such as 'Gene-Expression', 'Prognosis', 'Tumor Heterogeneity', 'Immunoregulation', and 'Tumor Immune Microenvironment'. Conclusion This is the first bibliometric study that comprehensively summarizes the research trends and developments on the application of single cell sequencing in liver cancer. The study identifies recent research frontiers and hot directions, providing a valuable reference for researchers exploring the landscape of liver cancer, understanding the composition of the immune microenvironment, and utilizing single-cell sequencing technology to guide and enhance the prognosis of liver cancer patients.
Article
Primary liver cancer is a solid malignancy with a high mortality rate. The success of immunotherapy has shown great promise in improving patient care and highlights a crucial need to understand the complexity of the liver tumor immune microenvironment (TIME). Recent advances in single-cell and spatial omics technologies, coupled with the development of systems biology approaches, are rapidly transforming the landscape of tumor immunology. Here we review the cellular landscape of liver TIME from single-cell and spatial perspectives. We also discuss the cellular interaction networks within the tumor cell community in regulating immune responses. We further highlight the challenges and opportunities with implications for biomarker discovery, patient stratification, and combination immunotherapies.
Article
Full-text available
Metabolic dysfunction-associated steatohepatitis (MASH) is the progressed version of metabolic dysfunction-associated steatotic liver disease (MASLD) characterized by inflammation and fibrosis, but also a pathophysiological “hub” that favors the emergence of liver malignancies. Current research efforts aim to identify risk factors, discover disease biomarkers, and aid patient stratification in the context of MASH-induced hepatocellular carcinoma (HCC), the most prevalent cancer among MASLD patients. To investigate the tumorigenic transition in MASH-induced HCC, researchers predominantly exploit preclinical animal-based MASH models and studies based on archived human biopsies and clinical trials. Recapitulating the immune response during tumor development and progression is vital to obtain mechanistic insights into MASH-induced HCC. Notably, the advanced complexity behind MASLD and MASH pathogenesis shifted the research focus towards innate immunity, a fundamental element of the hepatic immune niche that is usually altered robustly in the course of liver disease. During the last few years, however, there has been an increasing interest for deciphering the role of adaptive immunity in MASH-induced HCC, particularly regarding the functions of the various T cell populations. To effectively understand the specific role of T cells in MASH-induced HCC development, scientists should urgently fill the current knowledge gaps in this field. Pinpointing the metabolic signature, sketching the immune landscape, and characterizing the cellular interactions and dynamics of the specific T cells within the MASH-HCC liver are essential to unravel the mechanisms that adaptive immunity exploits to enable the emergence and progression of this cancer. To this end, our review aims to summarize the current state of research regarding the T cell functions linked to MASH-induced HCC.
Article
Full-text available
Mucosal-associated invariant T (MAIT) cells are a subpopulation of unconventional T cells widely involved in chronic liver diseases. However, the potential role and regulating factors of MAIT cells in alveolar echinococcosis (AE), a zoonotic parasitic disease by Echinococcus multilocularis (E. multilocularis) larvae chronically parasitizing liver organs, has not yet been studied. Blood samples (n=29) and liver specimens (n=10) from AE patients were enrolled. The frequency, phenotype, and function of MAIT cells in peripheral blood and liver tissues of AE patients were detected by flow cytometry. The morphology and fibrosis of liver tissue were examined by histopathology and immunohistochemistry. The correlation between peripheral MAIT cell frequency and serologic markers was assessed by collecting clinicopathologic characteristics of AE patients. And the effect of in vitro stimulation with E. multilocularis antigen (Emp) on MAIT cells. In this study, MAIT cells are decreased in peripheral blood and increased in the close-to-lesion liver tissues, especially in areas of fibrosis. Circulating MAIT exhibited activation and exhaustion phenotypes, and intrahepatic MAIT cells showed increased activation phenotypes with increased IFN-γ and IL-17A, and high expression of CXCR5 chemokine receptor. Furthermore, the frequency of circulating MAIT cells was correlated with the size of the lesions and liver function in patients with AE. After excision of the lesion site, circulating MAIT cells returned to normal levels, and the serum cytokines IL-8, IL-12, and IL-18, associated with MAIT cell activation and apoptosis, were altered. Our results demonstrate the status of MAIT cell distribution, functional phenotype, and migration in peripheral blood and tissues of AE patients, highlighting their potential as biomarkers and therapeutic targets.
Article
Full-text available
Mucosal-associated invariant T (MAIT) cells, defined as CD161⁺⁺TCR iVα7.2⁺ T cells, play an important role in the innate defense against bacterial infections, and their functionality is impaired in chronic viral infections. Here, we investigated the frequency and functional role of MAIT cells in chronic hepatitis B virus (HBV) infection. The peripheral CD3⁺CD161⁺⁺TCR iVα7.2⁺ MAIT cells in chronic HBV-infected patients and healthy controls were phenotypically characterized based on CD57, PD-1, TIM-3, and CTLA-4, as well as HLA-DR and CD38 expression. The frequency of MAIT cells was significantly decreased among chronic HBV-infected individuals as compared to controls. Expression of CD57, PD-1, CTLA-4, as well as HLA-DR and CD38 on MAIT cells was significantly elevated in chronic HBV-infected individuals relative to controls. The percentage of T cell receptor (TCR) iVα7.2⁺ CD161⁺ MAIT cells did not correlate with HBV viral load but inversely with HLA-DR on CD4⁺ T cells and MAIT cells and with CD57 on CD8⁺ T cells suggesting that decrease of MAIT cells may not be attributed to direct infection by HBV but driven by HBV-induced chronic immune activation. The percentage and expression levels of PD-1 as well as CTLA-4 on MAIT cells inversely correlated with plasma HBV-DNA levels, which may suggest either a role for MAIT cells in the control of HBV infection or the effect of HBV replication in the liver on MAIT cell phenotype. We report that decrease of TCR iVα7.2⁺ MAIT cells in the peripheral blood and their functions were seemingly impaired in chronic HBV-infected patients likely because of the increased expression of PD-1.
Article
Full-text available
Mucosal-associated invariant T (MAIT) cells represent up to 10% of circulating human T-cells. They are usually defined using combinations of non-lineage-specific (surrogate) markers such as anti-TRAV1-2, CD161, IL-18Rα and CD26. The development of MR1-Ag tetramers now permits the specific identification of MAIT cells based on TCR specificity. Here, we compare these approaches for identifying MAIT cells and show that surrogate markers are not always accurate in identifying these cells, particularly the CD4⁺ fraction. Moreover, while all MAIT cell subsets produced comparable levels of IFNγ, TNF and IL-17A, the CD4⁺ population produced more IL-2 than the other subsets. In a human ontogeny study, we show that the frequencies of most MR1 tetramer⁺ MAIT cells, with the exception of CD4⁺ MAIT cells, increased from birth to about 25 years-of-age and declined thereafter. We also demonstrate a positive association between the frequency of MAIT cells and other unconventional T cells including Natural Killer T (NKT) cells and Vδ2⁺ γδ T cells. Accordingly, this study demonstrates that MAIT cells are phenotypically and functionally diverse, that surrogate markers may not reliably identify all of these cells, and that their numbers are regulated in an age-dependent manner and correlate with NKT and Vδ2⁺ γδ T cells.
Article
Full-text available
Background & aims: The 4 genotypes of hepatitis E virus (HEV) that infect humans (genotypes 1-4) vary in geographical distribution, transmission, and pathogenesis. Little is known about the properties of HEV or its hosts that contribute to these variations. Primary isolates grow poorly in cell culture; most studies have relied on variants adapted to cancer cell lines, which likely alter virus biology. We investigated the infection and replication of primary isolates of HEV in hepatocyte-like cells (HLCs) derived from human embryonic and induced pluripotent stem cells. Methods: Using a cell culture-adapted genotype 3 strain and primary isolates of genotypes 1-4, we compared viral replication kinetics, sensitivity to drugs, and ability of HEV to activate the innate immune response. We studied HLCs using quantitative reverse-transcriptase PCR and immunofluorescence assay and ELISAs. We used an embryonic stem cell line that can be induced to express the CRISPR-Cas9 machinery to disrupt the peptidylprolyl isomerase A gene (PPIA), encoding cyclophilin A (CYPA)-a protein reported to inhibit replication of cell culture-adapted HEV. We further modified this line to rescue expression of CYPA before terminal differentiation to HLCs and performed HEV infection studies. Results: HLCs were permissive for infection by non-adapted, primary isolates of HEV genotypes 1-4. HEV infection of HLCs induced a replication-dependent type III interferon response. Replication of primary HEV isolates, unlike the cell culture-adapted strain, was not affected by disruption of PPIA or exposure to the CYPA inhibitor cyclosporine A. Conclusions: Cell culture adaptations alter the replicative capacities of HEV. HLCs offer an improved, physiologically relevant, and genetically tractable system for studying the replication of primary HEV isolates. HLCs could provide a model to aid development of HEV drugs and a system to guide personalized regimens, especially for patients with chronic hepatitis E who have developed resistance to ribavirin.
Article
Full-text available
Mucosa-associated invariant T (MAIT) cells are innate-like T lymphocytes that are unusually abundant in the human liver, a common site of colorectal carcinoma (CRC) metastasis. However, whether they contribute to immune surveillance against colorectal liver metastasis (CRLM) is essentially unexplored. In addition, whether MAIT cell functions can be impacted by chemotherapy is unclear. These are important questions given MAIT cells’ potent immunomodulatory and inflammatory properties. Herein, we examined the frequencies and functions of peripheral blood, healthy liver tissue, tumor-margin and tumor-infiltrating MAIT cells in 21 CRLM patients who received no chemotherapy, FOLFOX, or a combination of FOLFOX and Avastin before they underwent liver resection. We found that MAIT cells, defined as CD3ε+Vα7.2+CD161++ or CD3ε+MR1 tetramer+ cells, were present within both healthy and tumor-afflicted hepatic tissues. Paired and grouped analyses of samples revealed the physical proximity of MAIT cells to metastatic lesions to drastically influence their functional competence. Accordingly, unlike those residing in the healthy liver compartment, tumor-infiltrating MAIT cells failed to produce IFN-γ in response to a panel of TCR and cytokine receptor ligands, and tumor-margin MAIT cells were only partially active. Furthermore, chemotherapy did not account for intratumoral MAIT cell insufficiencies. Our findings demonstrate for the first time that CRLM-penetrating MAIT cells exhibit wide-ranging functional impairments, which are dictated by their physical location but not by preoperative chemotherapy. Therefore, we propose that MAIT cells may provide an attractive therapeutic target in CRC and that their ligands may be combined with chemotherapeutic agents to treat CRLM.
Article
Full-text available
Background For patients with advanced hepatocellular carcinoma, sorafenib is the only approved drug worldwide, and outcomes remain poor. We aimed to assess the safety and efficacy of nivolumab, a programmed cell death protein-1 (PD-1) immune checkpoint inhibitor, in patients with advanced hepatocellular carcinoma with or without chronic viral hepatitis. Methods We did a phase 1/2, open-label, non-comparative, dose escalation and expansion trial (CheckMate 040) of nivolumab in adults (≥18 years) with histologically confirmed advanced hepatocellular carcinoma with or without hepatitis C or B (HCV or HBV) infection. Previous sorafenib treatment was allowed. A dose-escalation phase was conducted at seven hospitals or academic centres in four countries or territories (USA, Spain, Hong Kong, and Singapore) and a dose-expansion phase was conducted at an additional 39 sites in 11 countries (Canada, UK, Germany, Italy, Japan, South Korea, Taiwan). At screening, eligible patients had Child-Pugh scores of 7 or less (Child-Pugh A or B7) for the dose-escalation phase and 6 or less (Child-Pugh A) for the dose-expansion phase, and an Eastern Cooperative Oncology Group performance status of 1 or less. Patients with HBV infection had to be receiving effective antiviral therapy (viral load <100 IU/mL); antiviral therapy was not required for patients with HCV infection. We excluded patients previously treated with an agent targeting T-cell costimulation or checkpoint pathways. Patients received intravenous nivolumab 0·1–10 mg/kg every 2 weeks in the dose-escalation phase (3+3 design). Nivolumab 3 mg/kg was given every 2 weeks in the dose-expansion phase to patients in four cohorts: sorafenib untreated or intolerant without viral hepatitis, sorafenib progressor without viral hepatitis, HCV infected, and HBV infected. Primary endpoints were safety and tolerability for the escalation phase and objective response rate (Response Evaluation Criteria In Solid Tumors version 1.1) for the expansion phase. This study is registered with ClinicalTrials.gov, number NCT01658878. Findings Between Nov 26, 2012, and Aug 8, 2016, 262 eligible patients were treated (48 patients in the dose-escalation phase and 214 in the dose-expansion phase). 202 (77%) of 262 patients have completed treatment and follow-up is ongoing. During dose escalation, nivolumab showed a manageable safety profile, including acceptable tolerability. In this phase, 46 (96%) of 48 patients discontinued treatment, 42 (88%) due to disease progression. Incidence of treatment-related adverse events did not seem to be associated with dose and no maximum tolerated dose was reached. 12 (25%) of 48 patients had grade 3/4 treatment-related adverse events. Three (6%) patients had treatment-related serious adverse events (pemphigoid, adrenal insufficiency, liver disorder). 30 (63%) of 48 patients in the dose-escalation phase died (not determined to be related to nivolumab therapy). Nivolumab 3 mg/kg was chosen for dose expansion. The objective response rate was 20% (95% CI 15–26) in patients treated with nivolumab 3 mg/kg in the dose-expansion phase and 15% (95% CI 6–28) in the dose-escalation phase. Interpretation Nivolumab had a manageable safety profile and no new signals were observed in patients with advanced hepatocellular carcinoma. Durable objective responses show the potential of nivolumab for treatment of advanced hepatocellular carcinoma. Funding Bristol-Myers Squibb.
Article
Background & aims: T-cell exhaustion, or an impaired capacity to secrete cytokines and proliferate with overexpression of immune checkpoint receptors, occurs during chronic viral infections but has also been observed in tumors, including hepatocellular carcinomas (HCCs). We investigated features of exhaustion in CD8+ T cells isolated from HCC specimens. Methods: We obtained HCC specimens, along with adjacent non-tumor tissues and blood samples, from 90 patients who underwent surgical resection at Asan Medical Center (in Seoul, Korea) from April 2016 through April 2018. Intrahepatic lymphocytes and tumor-infiltrating T cells were analyzed by flow cytometry. Tumor-infiltrating CD8+ T cells were sorted by flow cytometry into populations based on expression level of programmed cell death 1 (PDCD1 or PD1): PD1-high, PD1-intermediate, and PD1-negative. Sorted cells were analyzed by RNA-seq. Proliferation and production of interferon gamma (IFNG) and tumor necrosis factor (TNF) by CD8+ T cells were measured in response to anti-CD3 and antibodies against immune checkpoint receptors including PD1, hepatitis A virus cellular receptor 2 (HAVCR2 or TIM3), lymphocyte activating 3 (LAG3), or isotype control. Tumor-associated antigen-specific CD8+ T cells were identified using HLA-A*0201 dextramers. PDL1 expression on tumor tissue was assessed by immunohistochemistry. Results: PD1-high, PD1-intermediate, and PD1-negative CD8+ T cells from HCCs had distinct gene expression profiles. PD1-high cells expressed higher levels of genes that regulate T-cell exhaustion than PD1-intermediate cells. PD1-high cells expressed TIM3 and LAG3, and a low proportion was TCF1+, TBEThigh/eomesoderminlow, and CD127+. PD1-high cells produced the lowest amounts of IFNG and TNF upon anti-CD3 stimulation. Differences in proportions of PD1-high CD8+ T cells led to the identification of 2 subgroups of HCCs; HCCs with a larger proportion of PD1-high cells were more aggressive than HCCs with a smaller proportion. HCCs with a larger proportion of PD1-high cells had higher levels of predictive biomarkers of response to anti-PD1 therapy. Incubation of CD8+ T cells from HCCs with high proportions of PD1-high cells with antibodies against PD1 and TIM3 or LAG3 further restored proliferation and production of IFNG and TNF in response to anti-CD3. Conclusions: We found HCC specimens to contain CD8+ T cells that express different levels of PD1. HCCs with high proportions of PD1-high CD8+ T cells express TIM3 and/or LAG3 and produce low levels of IFNG and TNF in response to anti-CD3. Incubation of these cells with antibodies against PD1 and TIM3 or LAG3 further restore proliferation and production of cytokines; HCCs with high proportions of PD1-high CD8+ T cells might be more susceptible to combined immune checkpoint blockade-based therapies.
Article
In humans, MAIT cells represent the most abundant T cell subset reacting against bacteria. Their frequency in the blood is decreased in a large variety of infectious diseases of either bacterial or viral origin. MAIT cells accumulate at the site of bacterial infection and are protective in experimental infection models. Recent epidemiological evidence supports an implication of MAIT cells in protecting against tuberculosis. MAIT cells can be activated either through direct recognition of microbial ligands or by inflammatory cytokines such as IL-12 and IL-18. MAIT cells secrete IFN-γ, IL-17 and/or other effector molecules according to the context of triggering. MAIT cells can kill bacterially infected epithelial cells in vitro. Herein, we summarize and discuss the data suggesting a role for MAIT cells in infectious diseases.
Article
Systematic interrogation of tumor-infiltrating lymphocytes is key to the development of immunotherapies and the prediction of their clinical responses in cancers. Here, we perform deep single-cell RNA sequencing on 5,063 single T cells isolated from peripheral blood, tumor, and adjacent normal tissues from six hepatocellular carcinoma patients. The transcriptional profiles of these individual cells, coupled with assembled T cell receptor (TCR) sequences, enable us to identify 11 T cell subsets based on their molecular and functional properties and delineate their developmental trajectory. Specific subsets such as exhausted CD8⁺ T cells and Tregs are preferentially enriched and potentially clonally expanded in hepatocellular carcinoma (HCC), and we identified signature genes for each subset. One of the genes, layilin, is upregulated on activated CD8⁺ T cells and Tregs and represses the CD8⁺ T cell functions in vitro. This compendium of transcriptome data provides valuable insights and a rich resource for understanding the immune landscape in cancers.
Article
Background and Aims Agents that induce an immune response against tumors by altering T-cell regulation have increased survival times of patients with advanced-stage tumors, such as melanoma or lung cancer. We aimed to characterize molecular features of immune cells that infiltrate hepatocellular carcinomas (HCCs) to determine whether these types of agents might be effective against liver tumors. Methods We analyzed HCC samples from 956 patients. We separated gene expression profiles from tumor, stromal, and immune cells using a non-negative matrix factorization algorithm. We then analyzed the gene expression pattern of inflammatory cells in HCC tumors samples. We correlated expression patterns with the presence of immune cell infiltrates and immune regulatory molecules, determined by pathology and immunohistochemical analyses, in a training set of 228 HCC samples. We validated the correlation in a validation set of 728 tumor samples. Using data from 190 tumors in the Cancer Genome Atlas, we correlated immune cell gene expression profiles with numbers of chromosomal aberrations (based on single-nucleotide polymorphism array) and mutations (exome sequence data). Results We found approximately 25% of HCCs to have markers of an inflammatory response, with high expression levels of the CD274 molecule (PD-L1) and programmed cell death 1 (PD-1), markers of cytolytic activity, and fewer chromosomal aberrations. We called this group of tumors the Immune class. It contained 2 subtypes, characterized by markers of an adaptive T-cell response or exhausted immune response. The exhausted immune response subclass expressed many genes regulated by transforming growth factor beta 1 (TGFB) that mediate immunosuppression. We did not observe any differences in numbers of mutations or expression of tumor antigens between the immune-specific class and other HCCs. Conclusions In an analysis of HCC samples from 956 patients, we found almost 25% to express markers of an inflammatory response. We identified 2 subclasses, characterized by adaptive or exhausted immune responses. These findings indicate that some HCCs might be susceptible to therapeutic agents designed to block the regulatory pathways in T cells, such as PD-L1, PD-1, or TGFB inhibitors.